Выявление мутаций генов эпигенетической регуляции генома IDH1/2, DNMT3A, ASXL1 и их сочетания с мутациями FLT3, NPM1, RUNX1 у пациентов с острыми миелоидными лейкозами

Е.В. Белоцерковская1,2, Е.К. Зайкова1,2, А.В. Петухов1,2,3, О.Н. Демидов2, К.А. Левчук1, И.Г. Будаева1, Д.В. Зайцев1, Ю.Д. Роговая1, А.А. Шатилова1, К.В. Богданов1, Ю.В. Миролюбова1, Т.С. Никулина1, А.Ю. Зарицкий1, Л.Л. Гиршова1

1 ФГБУ «НМИЦ им. В.А. Алмазова» Минздрава России, ул. Аккуратова, д. 2, Санкт-Петербург, Российская Федерация, 197341

2 ФГБУН «Институт цитологии РАН», Тихорецкий пр-т, д. 4, Санкт-Петербург, Российская Федерация, 194064

3 НТУ «Сириус», Олимпийский пр-т, д. 1, Сочи, Российская Федерация, 354340

Для переписки: Екатерина Васильевна Белоцерковская, канд. биол. наук, ул. Аккуратова, д. 2, Санкт-Петербург, Российская Федерация, 197341; e-mail: belotserkovskaya.ev@gmail.com

Для цитирования: Белоцерковская Е.В., Зайкова Е.К., Петухов А.В. и др. Выявление мутаций генов эпигенетической регуляции генома IDH1/2, DNMT3A, ASXL1 и их сочетания с мутациями FLT3, NPM1, RUNX1 у пациентов с острыми миелоидными лейкозами. Клиническая онкогематология. 2021;14(1):13–21.

DOI: 10.21320/2500-2139-2021-14-1-13-21


РЕФЕРАТ

Цель. Выявление мутаций генов IDH1/IDH2, DNMT3A и ASXL1, ответственных за эпигенетическую регуляцию генома, при впервые диагностированных острых миелоидных лейкозах (ОМЛ) у взрослых и их сочетания с мутациями генов FLT3, NPM1, RUNX1.

Материалы и методы. В исследование включено 56 пациентов с впервые выявленным ОМЛ, проходивших лечение в ФГБУ «НМИЦ им. В.А. Алмазова» Минздрава России. Среди них было 34 мужчины и 22 женщины в возрасте 18–76 лет (медиана 46 лет). Мутационный статус генов эпигенетической регуляции IDH1, IDH2, DNMT3A и ASXL1 определяли методом секвенирования по Сэнгеру. Молекулярно-генетический анализ генов FLT3, NPM1, RUNX1-RUNX1T1 выполняли с использованием коммерческих наборов.

Результаты. Мутации генов эпигенетической регуляции обнаружены у 14 (25 %) из 56 пациентов. Распространенность мутаций не была связана с группами риска (= 0,072). Мутации IDH1/2 выявлены у 15,6 % пациентов и статистически значимо чаще обнаруживались одновременно с мутациями NPM1 (62,5 %; = 0,01) по сравнению с пациентами с диким типом IDH1/2. У большинства пациентов мутации IDH1/2 были связаны с нормальным кариотипом (= 0,002). Мутация DNMT3A (R882) определена у 4 (7,1 %) из 56 пациентов анализируемой группы. У 6 (11,1 %) пациентов были идентифицированы мутации ASXL1, которые сочетались мутациями с RUNX1-RUNX1T1 и FLT3-ITD.

Заключение. Мутации генов эпигенетической регуляции часто обнаруживаются у пациентов с ОМЛ и могут сочетаться с нарушениями в генах NPM1, FLT3 и RUNX1.

Ключевые слова: острые миелоидные лейкозы, гены эпигенетической регуляции IDH1, IDH2, DNMT3A и ASXL1, эпигенетические факторы.

Получено: 20 августа 2020 г.

Принято в печать: 2 декабря 2020 г.

Читать статью в PDF

Статистика Plumx русский

ЛИТЕРАТУРА

  1. Wang M, Yang C, Zang L, et al. Molecular mutations and their cooccurrences in cytogenetically normal Acute Myeloid Leukemia. Stem Cells Int. 2017;2017:1–11. doi: 10.1155/2017/6962379.
  2. 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(4):424–47. doi: 1182/blood-2016-08-733196.
  3. Gambacorta V, Gnani D, Vago L, et al. Epigenetic Therapies for Acute Myeloid Leukemia and Their Immune-Related Effects. Front Cell Dev Biol. 2019;7:207. doi: 10.3389/fcell.2019.00207.
  4. Santini Hypomethylating agents in the treatment of acute myeloid leukemia: A guide to optimal use. Crit Rev Oncol Hemat. 2009;140:1–7. doi: 10.1016/j.critrevonc.2019.05.013.
  5. Kim Enasidenib: First Global Approval. Drugs. 2017;77(15):1705–11. doi: 10.1007/s40265-017-0813-2.
  6. Liu X, Gong Y. Isocitrate dehydrogenase inhibitors in acute myeloid leukemia. Biomark Res. 2019;7(1):22. doi: 10.1186/s40364-019-0173-z.
  7. Cai SF, Levine RL. Genetic and epigenetic determinants of AML pathogenesis. Semin Hematol. 2018;56(2):84–9. doi: 10.1053/j.seminhematol.2018.08.001.
  8. Steensma DP, Bejar R, Jaiswal S, et al. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood. 2015;126(1):9–16. doi: 10.1182/blood-2015-03-631747.
  9. 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. doi: 10.1056/nejmoa1409405.
  10. Bowman RL, Busque L, Levine RL. Clonal Hematopoiesis and Evolution to Hematopoietic Malignancies. Cell Stem Cell. 2018;22(2):157–70. doi: 10.1016/j.stem.2018.01.011.
  11. Jaiswal S, Natarajan P, Silver AJ, et al. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N Engl J Med. 2017;377(2):111–21. doi: 10.1056/nejmoa1701719.
  12. Buscarlet M, Provost S, Zada YF, et al. DNMT3A and TET2 dominate clonal hematopoiesis and demonstrate benign phenotypes and different genetic predispositions. Blood. 2017;130(6):753–62. doi: 10.1182/blood-2017-04-777029.
  13. Yuan X, Peng L, Zeng W, et al. DNMT3A R882 Mutations Predict a Poor Prognosis in AML. Medicine. 2016;95(18):e3519. doi: 10.1097/md.0000000000003519.
  14. Marcucci G, Maharry K, Wu Y, et al. IDH1 and IDH2 Gene Mutations Identify Novel Molecular Subsets Within De Novo Cytogenetically Normal Acute Myeloid Leukemia: A Cancer and Leukemia Group B Study. J Clin Oncol. 2010;28(14):2348–55. doi: 10.1200/JCO.2009.27.3730.
  15. Schnittger S, Eder C, Jeromin S, et al. ASXL1 exon 12 mutations frequent in AML with intermediate risk karyotype and are independently associated with an adverse outcome. Leukemia. 2013;27(1):82–91. doi: 1038/leu.2012.262.
  16. Pratcorona M, Abbas S, Sanders MA, et al. Acquired mutations in ASXL1 in acute myeloid leukemia: prevalence and prognostic value. Haematologica. 2012;97(3):388. doi: 10.3324/haematol.2011.051532.
  17. Wagner K, Damm F, Gohring G, et al. Impact of IDH1 R132 mutations and an IDH1 single nucleotide polymorphism in cytogenetically normal acute myeloid leukemia: SNP rs11554137 is an adverse prognostic factor. J Clin Oncol. 2010;28(14):2356–64. doi: 10.1200/jco.2009.27.6899.
  18. Dinardo CD, Ravandi F, Agresta S, et al. Characteristics, clinical outcome, and prognostic significance of IDH mutations in AML. Am J Hematol. 2015;90(8):732–6. doi: 10.1002/ajh.24072.
  19. Brunetti L, Gundry MC, Goodell MA. DNMT3A in Leukemia. Cold Spring Harb Perspect Med. 2017;7(2):a030320. doi: 10.1101/cshperspect.a030320.
  20. Park SH, Choi JC, Kim SY, et al. Incidence and Prognostic Impact of DNMT3A Mutations in Korean Normal Karyotype Acute Myeloid Leukemia Patients. BioMed Res Int. 2015;2015:1–11. doi: 10.1155/2015/723682.
  21. Chotirat S, Thongnoppakhun W, Promsuwicha O, et al. Molecular alterations of isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) metabolic genes and additional genetic mutations in newly diagnosed acute myeloid leukemia patients. J Hematol Oncol. 2012;5(1):5. doi: 10.1186/1756-8722-5-5.
  22. Petrova L, Vrbacky F, Lanska M, et al. IDH1 and IDH2 mutations in patients with acute myeloid leukemia: Suitable targets for minimal residual disease monitoring? Clin Biochem. 2018;61:34–9. doi: 10.1016/j.clinbiochem.2018.08.012.
  23. Waitkus MS, Diplas BH, Yan H. Biological Role and Therapeutic Potential of IDH Mutations in Cancer. Cancer Cell. 2018;34(2):186–95. doi: 10.1016/j.ccell.2018.04.011.
  24. Clark O, Yen K, Mellinghoff IK. Molecular Pathways: Isocitrate Dehydrogenase Mutations in Cancer. Clin Cancer Res. 2016;22(8):1837–42. doi: 1158/1078-0432.CCR-13-1333.
  25. Yan H, Parsons DW, Jin G, et al. IDH1 and IDH2 Mutations in Gliomas. N Engl J Med 2009;360(8):765–73. doi: 10.1056/NEJMoa0808710.
  26. Parsons DW, Jones S, Zhang X, et al. An Integrated Genomic Analysis of Human Glioblastoma Multiforme. 2008;321(5897):1807–12. doi: 10.1126/science.1164382.
  27. Whitehall VLJ, Dumenil TD, McKeone DM, et al. Isocitrate dehydrogenase 1 R132C mutation occurs exclusively in microsatellite stable colorectal cancers with the CpG island methylator phenotype. Epigenetics. 2014;9(11):1454–60. doi: 10.4161/15592294.2014.971624.
  28. Mardis ER, Ding L, Dooling DJ, et al. Recurring Mutations Found by Sequencing an Acute Myeloid Leukemia Genome. N Engl J Med. 2009;361(11):1058–66. doi: 10.1056/NEJMoa0903840.
  29. Green CL, Evans CM, Zhao L, et al. The prognostic significance of IDH2 mutations in AML depends on the location of the mutation. Blood. 2011;118(2):409–12. doi: 10.1182/blood-2010-12-322479.
  30. Berenstein R, Blau IW, Kar A, et al. Comparative examination of various PCR-based methods for DNMT3A and IDH1/2 mutations identification in acute myeloid leukemia. J Exp Clin Cancer Res. 2014;33(1):44. doi: 10.1186/1756-9966-33-44.
  31. Mizuta S, Yamane N, Komai T, et al. Investigation of screening method for DNMT3A mutations by high‐resolution melting analysis in acute myeloid leukemia. Int J Lab Hematol. 2019;41(5):593–600. doi: 10.1111/ijlh.13056.
  32. МотыкоЕ.В., Блау О.В., Полушкина Л.Б. и др. Прогностическое значение генетических мутаций у больных острыми миелоидными лейкозами: результаты совместного исследования гематологических клиник Санкт-Петербурга (Россия) и клиники Шарите (Германия). Клиническая онкогематология. 2019;12(2):211–9. doi: 10.21320/2500-2139-2019-12-2-211-219.
    [Motyko EV, Blau OV, Polushkina LB, et al. Prognostic Value of Genetic Mutations in Patients with Acute Myeloid Leukemias: Results of a Cooperative Study of Hematology Clinics of Saint Petersburg (Russia) and Charite Clinic (Germany). Clinical oncohematology. 2019;12(2):211–9. doi: 10.21320/2500-2139-2019-12-2-211-219. (In Russ)]
  33. ElNahass YH, Badawy RH, ElRefaey FA, et al. IDH Mutations in AML Patients; A higher Association with Intermediate Risk Cytogenetics. Asian Pacif J Cancer Prev. 2020;21(3):721–5. doi: 10.31557/APJCP.2020.21.3.721.
  34. Ferret Y, Boissel N, Helevaut N, et al. Clinical Relevance Of IDH1/2 Mutant Allele Burden During Follow-Up In Acute Myeloid Leukemia. A Study By The French ALFA Group. Haematologica. 2018;103(5):822–9. doi: 10.3324/haematol.2017.183525.
  35. Brambati C, Galbiati S, Xue E, et al. Droplet digital polymerase chain reaction for DNMT3A and IDH1/2 mutations to improve early detection of acute myeloid leukemia relapse after allogeneic hematopoietic stem cell transplantation. Haematologica. 2016;101(4):e157–e161. doi: 10.3324/haematol.2015.135467.
  36. Patel KP, Ravandi F, Ma D, et al. Acute myeloid leukemia with IDH1 or IDH2 mutation: frequency and clinicopathologic features. Am J Clin Pathol. 2011;135(1):35–45. doi: 10.1309/AJCPD7NR2RMNQDVF.
  37. Zou Y, Bai HX, Wang Z, Yang L. Comparison of immunohistochemistry and DNA sequencing for the detection of IDH1 mutations in gliomas. Neuro Oncol. 2015;17(3):477–8. doi: 10.1093/neuonc/nou351.
  38. Petiti J, Rosso V, Croce E, et al. Highly Sensitive Detection of IDH2 Mutations in Acute Myeloid Leukemia. J Clin Med. 2020;9(1):271. doi: 10.3390/jcm9010271.
  39. Aref S, Kamel AS, Abdel AMF, et al. Prevalence and clinical effect of IDH1 and IDH2 mutations among cytogenetically normal acute myeloid leukemia patients. Clin Lymphoma Myel Leuk. 2015;15(9):550–5. doi: 10.1016/j.clml.2015.05.009.
  40. 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):3717–23. doi: 10.1200/jco.2010.28.2285.
  41. Xu Q, Li Y, Lv N, et al. Correlation between isocitrate dehydrogenase gene aberrations and prognosis of patients with acute myeloid leukemia: A systematic review and meta-analysis. Clin Cancer Res. 2017;23(15):4511–22. doi: 10.1158/1078-0432.ccr-16-2628.
  42. Montalban-Bravo G, DiNardo CD. The role of IDH mutations in acute myeloid leukemia. Future Oncol. 2018;10(14):979–93. doi: 10.2217/fon-2017-0523.
  43. Amatangelo MD, Quek L, Shih A, et al. Enasidenib induces acute myeloid leukemia cell differentiation to promote clinical response. Blood. 2017;130(6):732–42. doi: 10.1182/blood-2017-04-779447.
  44. Okano M, Xie S, Li E. Cloning and characterization of a family of novel mammalian DNA (cytosine-5) methyltransferases. Nat Genet. 1998;19(3):219–20. doi: 10.1038/890.
  45. Ley TJ, Ding L, Walter MJ, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363(25):2424–33. doi: 10.1056/NEJMoa1005143.
  46. Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059–74. doi: 1056/NEJMoa1301689.
  47. Блау О.В. Мутации генов при острых миелоидных лейкозах. Клиническая онкогематология. 2016;9(3):245–56. doi: 10.21320/2500-2139-2016-9-3-245-256.
    [Blau OV. Genetic Mutations in Acute Myeloid Leukemia. Clinical oncohematology. 2016;9(3):245–56. doi: 10.21320/2500-2139-2016-9-3-245-256. (In Russ)]
  48. Guryanova OA, Shank K, Spitzer B, et al. DNMT3A mutations promote anthracycline resistance in acute myeloid leukemia via impaired nucleosome remodeling. Nat Med. 2016;22(12):1488–95. doi: 10.1038/nm.4210.
  49. Hou HA, Kuo YY, Liu CY, et al. DNMT3A mutations in acute myeloid leukemia: stability during disease evolution and clinical implications. Blood. 2012;119(2):559–68. doi: 10.1182/blood-2011-07-369934.
  50. Ploen GG, Nederby L, Guldberg P, et al. Persistence of DNMT3A mutations at long-term remission in adult patients with AML. Br J Haematol. 2014;167(4):478–86. doi: 10.1111/bjh.13062.
  51. Rothenberg-Thurley M, Amler S, Goerlich D, et al. Persistence of pre-leukemic clones during first remission and risk of relapse in acute myeloid leukemia. Leukemia. 2018;32(7):1598–608. doi: 10.1038/s41375-018-0034-z.
  52. 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):2072–83. doi: 10.1200/jco.2014.59.2022.
  53. Gaidzik VI, Schlenk RF, Paschka P, et al. Clinical impact of DNMT3A mutations in younger adult patients with acute myeloid leukemia: Results of the AML Study Group (AMLSG). Blood. 2013;121(23):4769–77. doi: 10.1182/blood-2012-10-461624.
  54. Elsayed GM, Fahmi AEA, Shafiket NF, et al. Study of DNA methyl transferase 3A mutation in acute myeloid leukemic patients. Egypt J Med Hum Genet. 2018;19(4):315–9. doi: 10.1016/j.ejmhg.2018.05.005.
  55. Berenstein R, Blau IW, Suckert N, et al. Quantitative detection of DNMT3A R882H mutation in acute myeloid leukemia. J Exp Clin Cancer Res. 2015;34(1):55. doi: 10.1186/s13046-015-0173-2.
  56. Young AL, Challen GA, Birmann BM, et al. Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults. Nat Commun. 2016;7(1):12484. doi: 10.1038/ncomms12484.
  57. Asada S, Fujino T, Goyama S, et al. The role of ASXL1 in hematopoiesis and myeloid malignancies. Cell Mol Life Sci. 2019;76(13):2511–23 doi: 10.1007/s00018-019-03084-7.
  58. Chou WC, Huang HH, Hou HA, et al. Distinct clinical and biological features of de novo acute myeloid leukemia with additional sex comb-like 1 (ASXL1) mutations. Blood. 2010;116(20):4086–94. doi: 10.1182/blood-2010-05-283291.
  59. Molenaar RJ, Thota S, Nagata Y, et al. Clinical and biological implications of ancestral and non-ancestral IDH1 and IDH2 mutations in myeloid neoplasms. Leukemia. 2015;29(11):2134–42. doi: 10.1038/leu.2015.91.
  60. Asada S, Kitamura T. Aberrant histone modifications induced by mutant ASXL1 in myeloid neoplasms. Int J Hematol. 2019;110(2):179–86. doi: 10.1007/s12185-018-2563-7.
  61. Shivarov V, Ivanova M, Naumova E. Rapid Detection of DNMT3A R882 Mutations in Hematologic Malignancies Using a Novel Bead-Based Suspension Assay with BNA(NC) Probes. PLoS ONE. 2014;9(6):e99769. doi: 10.1371/journal.pone.0099769.
  62. Gelsi-Boyer V, Trouplin V, Adelaide J, et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2009;145(6):788–800. doi: 10.1111/j.1365-2141.2009.07697.x.
  63. Abbas S, Lugthart S, Kavelaars F, et al. Acquired mutations in the genes encoding IDH1 and IDH2 both are recurrent aberrations in acute myeloid leukemia: prevalence and prognostic value. Blood. 2010;116(12):2122–6. doi: 10.1182/blood-2009-11-250878.
  64. Dunlap JB, Leonard J, Rosenberg M, et al. The combination of NPM1, DNMT3A, and IDH1/2 mutations leads to inferior overall survival in AML. Am J Hematol. 2019;94(8):913–20. doi: 10.1002/ajh.25517.
  65. Virijevic M, Karan-Djurasevic T, Marjanovic I, et al. Somatic mutations of isocitrate dehydrogenases 1 and 2 are prognostic and follow-up markers in patients with acute myeloid leukaemia with normal karyotype. Radiol Oncol. 2016;50(4):385–93. doi: 10.1515/raon-2016-0044.
  66. Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374(23):2209–21. doi: 10.1056/NEJMoa1516192.
  67. Boddu P, Takahashi K, Pemmaraju N, et al. Influence of IDH on FLT3ITD status in newly diagnosed AML. Leukemia. 2017;31(11):2526– doi: 10.1038/leu.2017.244.
  68. Yan X-J, Xu J, Gu Z-H, et al. Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nat Genet. 2011;43(4):309–15. doi: 10.1038/ng.788.
  69. Abdel-Wahab O, Adli M, Saunders L, et al. ASXL1 Mutations Promote Myeloid Transformation Through Inhibition of PRC2-Mediated Gene Repression. Blood. 2011;118(21):405. doi: 10.1182/blood.v118.21.405.405.
  70. Inoue D, Matsumoto M, Nagase R. Truncation mutants of ASXL1 observed in myeloid malignancies are expressed at detectable protein levels. Exp Hematol. 2016;44(3):172–6.e1. doi: 10.1016/j.exphem.2015.11.011.
  71. Gelsi-Boyer V, Brecqueville M, Devillier R, et al. Mutations in ASXL1 are associated with poor prognosis across the spectrum of malignant myeloid diseases. J Hematol Oncol. 2012;5(1):12. doi: 10.1186/1756-8722-5-12.
  72. Paschka P, Schlenk RF, Gaidzik VI. ASXL1 mutations in younger adult patients with acute myeloid leukemia: a study by the German Austrian Acute Myeloid Leukemia Study Group. Haematologica. 2015;100(3):324–30. doi: 10.3324/haematol.2014.114157.