DOI QR코드

DOI QR Code

Mutational Analysis of Extranodal NK/T-Cell Lymphoma Using Targeted Sequencing with a Comprehensive Cancer Panel

  • Choi, Seungkyu (Department of Pathology, Dankook University College of Medicine) ;
  • Go, Jai Hyang (Department of Pathology, Dankook University College of Medicine) ;
  • Kim, Eun Kyung (Department of Pathology, Eulji Medical Center, Eulji University School of Medicine) ;
  • Lee, Hojung (Department of Pathology, Eulji Medical Center, Eulji University School of Medicine) ;
  • Lee, Won Mi (Department of Pathology, Eulji Medical Center, Eulji University School of Medicine) ;
  • Cho, Chun-Sung (Department of Neurosurgery, Dankook University College of Medicine) ;
  • Han, Kyudong (Department of Nanobiomedical Science, BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University)
  • Received : 2016.08.01
  • Accepted : 2016.08.11
  • Published : 2016.09.30

Abstract

Extranodal natural killer (NK)/T-cell lymphoma, nasal type (NKTCL), is a malignant disorder of cytotoxic lymphocytes of NK or T cells. It is an aggressive neoplasm with a very poor prognosis. Although extranodal NKTCL reportedly has a strong association with Epstein-Barr virus, the molecular pathogenesis of NKTCL has been unexplored. The recent technological advancements in next-generation sequencing (NGS) have made DNA sequencing cost- and time-effective, with more reliable results. Using the Ion Proton Comprehensive Cancer Panel, we sequenced 409 cancer-related genes to identify somatic mutations in five NKTCL tissue samples. The sequencing analysis detected 25 mutations in 21 genes. Among them, KMT2D, a histone modification-related gene, was the most frequently mutated gene (four of the five cases). This result was consistent with recent NGS studies that have suggested KMT2D as a novel driver gene in NKTCL. Mutations were also found in ARID1A, a chromatin remodeling gene, and TP53, which also recurred in recent NGS studies. We also found mutations in 18 novel candidate genes, with molecular functions that were potentially implicated in cancer development. We suggest that these genes may result in multiple oncogenic events and may be used as potential bio-markers of NKTCL in the future.

Keywords

References

  1. William BM, Armitage JO. International analysis of the frequency and outcomes of NK/T-cell lymphomas. Best Pract Res Clin Haematol 2013;26:23-32. https://doi.org/10.1016/j.beha.2013.04.003
  2. Vose J, Armitage J, Weisenburger D; International T-Cell Lymphoma Project. International peripheral T-cell and natural killer/T-cell lymphoma study: pathology findings and clinical outcomes. J Clin Oncol 2008;26:4124-4130. https://doi.org/10.1200/JCO.2008.16.4558
  3. Kim TM, Lee SY, Jeon YK, Ryoo BY, Cho GJ, Hong YS, et al. Clinical heterogeneity of extranodal NK/T-cell lymphoma, nasal type: a national survey of the Korean Cancer Study Group. Ann Oncol 2008;19:1477-1484. https://doi.org/10.1093/annonc/mdn147
  4. Zhang J, Chiodini R, Badr A, Zhang G. The impact of next-generation sequencing on genomics. J Genet Genomics 2011;38:95-109. https://doi.org/10.1016/j.jgg.2011.02.003
  5. Morin RD, Gascoyne RD. Newly identified mechanisms in B-cell non-Hodgkin lymphomas uncovered by next-generation sequencing. Semin Hematol 2013;50:303-313. https://doi.org/10.1053/j.seminhematol.2013.09.004
  6. Vaque JP, Martinez N, Batlle-Lopez A, Perez C, Montes-Moreno S, Sanchez-Beato M, et al. B-cell lymphoma mutations: improving diagnostics and enabling targeted therapies. Haematologica 2014;99:222-231. https://doi.org/10.3324/haematol.2013.096248
  7. Morin RD, Mungall K, Pleasance E, Mungall AJ, Goya R, Huff RD, et al. Mutational and structural analysis of diffuse large B-cell lymphoma using whole-genome sequencing. Blood 2013;122:1256-1265. https://doi.org/10.1182/blood-2013-02-483727
  8. Lohr JG, Stojanov P, Lawrence MS, Auclair D, Chapuy B, Sougnez C, et al. Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by wholeexome sequencing. Proc Natl Acad Sci U S A 2012;109: 3879-3884. https://doi.org/10.1073/pnas.1121343109
  9. Bea S, Valdes-Mas R, Navarro A, Salaverria I, Martin-Garcia D, Jares P, et al. Landscape of somatic mutations and clonal evolution in mantle cell lymphoma. Proc Natl Acad Sci U S A 2013;110:18250-18255. https://doi.org/10.1073/pnas.1314608110
  10. Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, et al. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th ed. Lyon: International Agency for Research on Cancer, 2008.
  11. Han JY, Kim SH, Lee YS, Lee SY, Hwang JA, Kim JY, et al. Comparison of targeted next-generation sequencing with conventional sequencing for predicting the responsiveness to epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) therapy in never-smokers with lung adenocarcinoma. Lung Cancer 2014;85:161-167. https://doi.org/10.1016/j.lungcan.2014.04.009
  12. Zhang W, Meehan J, Su Z, Ng HW, Shu M, Luo H, et al. Whole genome sequencing of 35 individuals provides insights into the genetic architecture of Korean population. BMC Bioinformatics 2014;15 Suppl 11:S6.
  13. Richards CS, Bale S, Bellissimo DB, Das S, Grody WW, Hegde MR, et al. ACMG recommendations for standards for interpretation and reporting of sequence variations: Revisions 2007. Genet Med 2008;10:294-300. https://doi.org/10.1097/GIM.0b013e31816b5cae
  14. Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet 2000;25:25-29. https://doi.org/10.1038/75556
  15. Van Allen EM, Wagle N, Levy MA. Clinical analysis and interpretation of cancer genome data. J Clin Oncol 2013;31:1825-1833. https://doi.org/10.1200/JCO.2013.48.7215
  16. Raphael BJ, Dobson JR, Oesper L, Vandin F. Identifying driver mutations in sequenced cancer genomes: computational approaches to enable precision medicine. Genome Med 2014;6:5. https://doi.org/10.1186/gm524
  17. Sukhai MA, Craddock KJ, Thomas M, Hansen AR, Zhang T, Siu L, et al. A classification system for clinical relevance of somatic variants identified in molecular profiling of cancer. Genet Med 2016;18:128-136. https://doi.org/10.1038/gim.2015.47
  18. Bennett NC, Farah CS. Next-generation sequencing in clinical oncology: next steps towards clinical validation. Cancers (Basel) 2014;6:2296-2312. https://doi.org/10.3390/cancers6042296
  19. Dienstmann R, Dong F, Borger D, Dias-Santagata D, Ellisen LW, Le LP, et al. Standardized decision support in next generation sequencing reports of somatic cancer variants. Mol Oncol 2014;8:859-873. https://doi.org/10.1016/j.molonc.2014.03.021
  20. Lee S, Park HY, Kang SY, Kim SJ, Hwang J, Lee S, et al. Genetic alterations of JAK/STAT cascade and histone modification in extranodal NK/T-cell lymphoma nasal type. Oncotarget 2015;6:17764-17776. https://doi.org/10.18632/oncotarget.3776
  21. Jiang L, Gu ZH, Yan ZX, Zhao X, Xie YY, Zhang ZG, et al. Exome sequencing identifies somatic mutations of DDX3X in natural killer/T-cell lymphoma. Nat Genet 2015;47:1061-1066. https://doi.org/10.1038/ng.3358
  22. Dobashi A, Tsuyama N, Asaka R, Togashi Y, Ueda K, Sakata S, et al. Frequent BCOR aberrations in extranodal NK/T-Cell lymphoma, nasal type. Genes Chromosomes Cancer 2016;55:460-471. https://doi.org/10.1002/gcc.22348
  23. Dubuc AM, Remke M, Korshunov A, Northcott PA, Zhan SH, Mendez-Lago M, et al. Aberrant patterns of H3K4 and H3K27 histone lysine methylation occur across subgroups in medulloblastoma. Acta Neuropathol 2013;125:373-384. https://doi.org/10.1007/s00401-012-1070-9
  24. Zang ZJ, Cutcutache I, Poon SL, Zhang SL, McPherson JR, Tao J, et al. Exome sequencing of gastric adenocarcinoma identifies recurrent somatic mutations in cell adhesion and chromatin remodeling genes. Nat Genet 2012;44:570-574. https://doi.org/10.1038/ng.2246
  25. Jiang Y, Redmond D, Nie K, Eng KW, Clozel T, Martin P, et al. Deep sequencing reveals clonal evolution patterns and mutation events associated with relapse in B-cell lymphomas. Genome Biol 2014;15:432.
  26. Wang K, Nahas MK, Yelensky R, Otto GA, Lipson D, He J, et al. Novel chromatin modifying gene alterations and significant survival association of ATM and P53 in mantle cell lymphoma. Blood 2014;124:3033.
  27. Li H, Kaminski MS, Li Y, Yildiz M, Ouillette P, Jones S, et al. Mutations in linker histone genes HIST1H1 B, C, D, and E; OCT2 (POU2F2); IRF8; and ARID1A underlying the pathogenesis of follicular lymphoma. Blood 2014;123:1487-1498. https://doi.org/10.1182/blood-2013-05-500264
  28. Wiegand KC, Shah SP, Al-Agha OM, Zhao Y, Tse K, Zeng T, et al. ARID1A mutations in endometriosis-associated ovarian carcinomas. N Engl J Med 2010;363:1532-1543. https://doi.org/10.1056/NEJMoa1008433
  29. Giulino-Roth L, Wang K, MacDonald TY, Mathew S, Tam Y, Cronin MT, et al. Targeted genomic sequencing of pediatric Burkitt lymphoma identifies recurrent alterations in antiapoptotic and chromatin-remodeling genes. Blood 2012;120:5181-5184. https://doi.org/10.1182/blood-2012-06-437624
  30. Wu RC, Wang TL, Shih IM. The emerging roles of ARID1A in tumor suppression. Cancer Biol Ther 2014;15:655-664. https://doi.org/10.4161/cbt.28411
  31. Hoshida Y, Hongyo T, Jia X, He Y, Hasui K, Dong Z, et al. Analysis of p53, K-ras, c-kit, and beta-catenin gene mutations in sinonasal NK/T cell lymphoma in northeast district of China. Cancer Sci 2003;94:297-301. https://doi.org/10.1111/j.1349-7006.2003.tb01436.x
  32. Hongyo T, Hoshida Y, Nakatsuka S, Syaifudin M, Kojya S, Yang WI, et al. p53, K-ras, c-kit and beta-catenin gene mutations in sinonasal NK/T-cell lymphoma in Korea and Japan. Oncol Rep 2005;13:265-271.
  33. Quintanilla-Martinez L, Kremer M, Keller G, Nathrath M, Gamboa-Dominguez A, Meneses A, et al. p53 mutations in nasal natural killer/T-cell lymphoma from Mexico: association with large cell morphology and advanced disease. Am J Pathol 2001;159:2095-2105. https://doi.org/10.1016/S0002-9440(10)63061-1
  34. Bar-Eli M, Ahuja H, Gonzalez-Cadavid N, Foti A, Cline MJ. Analysis of N-RAS exon-1 mutations in myelodysplastic syndromes by polymerase chain reaction and direct sequencing. Blood 1989;73:281-283.
  35. Meyerson M, Gabriel S, Getz G. Advances in understanding cancer genomes through second-generation sequencing. Nat Rev Genet 2010;11:685-696. https://doi.org/10.1038/nrg2841
  36. MacConaill LE. Existing and emerging technologies for tumor genomic profiling. J Clin Oncol 2013;31:1815-1824. https://doi.org/10.1200/JCO.2012.46.5948
  37. Coppo P, Gouilleux-Gruart V, Huang Y, Bouhlal H, Bouamar H, Bouchet S, et al. STAT3 transcription factor is constitutively activated and is oncogenic in nasal-type NK/T-cell lymphoma. Leukemia 2009;23:1667-1678. https://doi.org/10.1038/leu.2009.91
  38. Koo GC, Tan SY, Tang T, Poon SL, Allen GE, Tan L, et al. Janus kinase 3-activating mutations identified in natural killer/T-cell lymphoma. Cancer Discov 2012;2:591-597. https://doi.org/10.1158/2159-8290.CD-12-0028
  39. Kucuk C, Jiang B, Hu X, Zhang W, Chan JK, Xiao W, et al. Activating mutations of STAT5B and STAT3 in lymphomas derived from ${\gamma}{\delta}-T$ or NK cells. Nat Commun 2015;6:6025. https://doi.org/10.1038/ncomms7025
  40. Chen YW, Guo T, Shen L, Wong KY, Tao Q, Choi WW, et al. Receptor-type tyrosine-protein phosphatase $\kappa$ directly targets STAT3 activation for tumor suppression in nasal NK/T-cell lymphoma. Blood 2015;125:1589-1600. https://doi.org/10.1182/blood-2014-07-588970
  41. Huang Y, de Leval L, Gaulard P. Molecular underpinning of extranodal NK/T-cell lymphoma. Best Pract Res Clin Haematol 2013;26:57-74. https://doi.org/10.1016/j.beha.2013.04.006
  42. Schmitt C, Sako N, Bagot M, Huang Y, Gaulard P, Bensussan A. Extranodal NK/T-cell lymphoma: toward the identification of clinical molecular targets. J Biomed Biotechnol 2011;2011:790871.
  43. Huang Y, de Reynies A, de Leval L, Ghazi B, Martin-Garcia N, Travert M, et al. Gene expression profiling identifies emerging oncogenic pathways operating in extranodal NK/T-cell lymphoma, nasal type. Blood 2010;115:1226-1237. https://doi.org/10.1182/blood-2009-05-221275

Cited by

  1. EBV-negative aggressive NK-cell leukemia/lymphoma: a clinical and pathological study from a single institution vol.30, pp.8, 2017, https://doi.org/10.1038/modpathol.2017.37