1932

Abstract

Next-generation sequencing has allowed identification of millions of somatic mutations and epigenetic changes in cancer cells. A key challenge in interpreting cancer genomes and epigenomes is distinguishing which genetic and epigenetic changes are drivers of cancer development. Frequency-based and function-based approaches have been developed to identify candidate drivers; we discuss the advantages and drawbacks of these methods as well as their latest refinements. We focus particularly on identification of the types of drivers most likely to be missed, such as genes affected by copy number alterations, mutations in noncoding regions, dysregulation of microRNA, epigenetic changes, and mutations in chromatin modifiers.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-pathol-012414-040312
2015-01-24
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/pathol/10/1/annurev-pathol-012414-040312.html?itemId=/content/journals/10.1146/annurev-pathol-012414-040312&mimeType=html&fmt=ahah

Literature Cited

  1. 1. Cancer Genome Atlas Res. Netw 2008. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455:72161061–68 [Google Scholar]
  2. Futreal PA, Coin L, Marshall M, Down T, Hubbard T. 2.  et al. 2004. A census of human cancer genes. Nat. Rev. Cancer 4:3177–83 [Google Scholar]
  3. 3. Int. Cancer Genome Consort 2010. International network of cancer genome projects. Nature 464:7291993–98 [Google Scholar]
  4. Stratton MR, Campbell PJ, Futreal PA. 4.  2009. The cancer genome. Nature 458:7239719–24 [Google Scholar]
  5. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr, Kinzler KW. 5.  2013. Cancer genome landscapes. Science 339:61271546–58 [Google Scholar]
  6. Forbes SA, Tang G, Bindal N, Bamford S, Dawson E. 6.  et al. 2010. COSMIC (the Catalogue of Somatic Mutations in Cancer): a resource to investigate acquired mutations in human cancer. Nucleic Acids Res. 38:Database IssueD652–57 [Google Scholar]
  7. Garay JP, Gray JW. 7.  2012. Omics and therapy—a basis for precision medicine. Mol. Oncol. 6:2128–39 [Google Scholar]
  8. Gonzalez-Perez A, Mustonen V, Reva B, Ritchie GRS, Creixell P. 8.  et al. 2013. Computational approaches to identify functional genetic variants in cancer genomes. Nat. Methods 10:8723–29 [Google Scholar]
  9. Gnad F, Baucom A, Mukhyala K, Manning G, Zhang Z. 9.  2013. Assessment of computational methods for predicting the effects of missense mutations in human cancers. BMC Genomics 14:Suppl. 3S7 [Google Scholar]
  10. Zhang J, Liu J, Sun J, Chen C, Foltz G, Lin B. 10.  2014. Identifying driver mutations from sequencing data of heterogeneous tumors in the era of personalized genome sequencing. Brief. Bioinform. 15:2244–55 [Google Scholar]
  11. Kern SE, Winter JM. 11.  2006. Elegance, silence and nonsense in the mutations literature for solid tumors. Cancer Biol. Ther. 5:4349–59 [Google Scholar]
  12. Ruark E, Snape K, Humburg P, Loveday C, Bajrami I. 12.  et al. 2013. Mosaic PPM1D mutations are associated with predisposition to breast and ovarian cancer. Nature 493:7432406–10 [Google Scholar]
  13. Venables JP, Klinck R, Koh C, Gervais-Bird J, Bramard A. 13.  et al. 2009. Cancer-associated regulation of alternative splicing. Nat. Struct. Mol. Biol. 16:6670–76 [Google Scholar]
  14. Slack GW, Gascoyne RD. 14.  2011. MYC and aggressive B-cell lymphomas. Adv. Anat. Pathol. 18:3219–28 [Google Scholar]
  15. Pakneshan S, Salajegheh A, Smith RA, Lam AK. 15.  2013. Clinicopathological relevance of BRAF mutations in human cancer. Pathology 45:4346–56 [Google Scholar]
  16. Ciriello G, Miller ML, Aksoy BA, Senbabaoglu Y, Schultz N, Sander C. 16.  2013. Emerging landscape of oncogenic signatures across human cancers. Nat. Genet. 45:101127–33 [Google Scholar]
  17. Bunting SF, Nussenzweig A. 17.  2013. End-joining, translocations and cancer. Nat. Rev. Cancer 13:7443–54 [Google Scholar]
  18. Mitelman F, Johansson B, Mertens F. 18.  2007. The impact of translocations and gene fusions on cancer causation. Nat. Rev. Cancer 7:4233–45 [Google Scholar]
  19. Khurana E, Fu Y, Colonna V, Mu XJ, Kang HM. 19.  et al. 2013. Integrative annotation of variants from 1092 humans: application to cancer genomics. Science 342:61541235587 [Google Scholar]
  20. Hofmann W, Schlag PM. 20.  2000. BRCA1 and BRCA2—breast cancer susceptibility genes. J. Cancer Res. Clin. Oncol. 126:9487–96 [Google Scholar]
  21. Hamilton SR. 21.  1992. The adenoma-adenocarcinoma sequence in the large bowel: variations on a theme. J. Cell. Biochem. 50:Suppl. 16G:41–46 [Google Scholar]
  22. Zeichner SB, Raj N, Cusnir M, Francavilla M, Hirzel A. 22.  2012. A de novo germline APC mutation (3927del5) in a patient with familial adenomatous polyposis: case report and literature review. Clin. Med. Insights Oncol. 6:315–23 [Google Scholar]
  23. Garcia-Casado Z, Romero I, Fernandez-Serra A, Rubio L, Llopis F. 23.  et al. 2011. A de novo complete BRCA1 gene deletion identified in a Spanish woman with early bilateral breast cancer. BMC Med. Genet. 12:134 [Google Scholar]
  24. Gonzalez KD, Buzin CH, Noltner KA, Gu D, Li W. 24.  et al. 2009. High frequency of de novo mutations in Li–Fraumeni syndrome. J. Med. Genet. 46:10689–93 [Google Scholar]
  25. Bourdeaut F, Lequin D, Brugières L, Reynaud S, Dufour C. 25.  et al. 2011. Frequent hSNF5/INI1 germline mutations in patients with rhabdoid tumor. Clin. Cancer Res. 17:131–38 [Google Scholar]
  26. Knudson AG Jr. 26.  1971. Mutation and cancer: statistical study of retinoblastoma. PNAS 68:4820–23 [Google Scholar]
  27. Akhurst RJ, Derynck R. 27.  2001. TGF-β signaling in cancer—a double-edged sword. Trends Cell Biol. 11:11S44–51 [Google Scholar]
  28. Armitage P, Doll R. 28.  1954. The age distribution of cancer and a multi-stage theory of carcinogenesis. Br. J. Cancer 8:11–12 [Google Scholar]
  29. Kandoth C, McLellan MD, Vandin F, Ye K, Niu B. 29.  et al. 2013. Mutational landscape and significance across 12 major cancer types. Nature 502:7471333–39 [Google Scholar]
  30. Tamborero D, Gonzalez-Perez A, Lopez-Bigas N. 30.  2013. OncodriveCLUST: exploiting the positional clustering of somatic mutations to identify cancer genes. Bioinformatics 29:182238–44 [Google Scholar]
  31. Ryslik GA, Cheng Y, Cheung KH, Modis Y, Zhao H. 31.  2013. Utilizing protein structure to identify non-random somatic mutations. BMC Bioinform. 14:190 [Google Scholar]
  32. Reimand J, Bader GD. 32.  2013. Systematic analysis of somatic mutations in phosphorylation signaling predicts novel cancer drivers. Mol. Syst. Biol. 9:637 [Google Scholar]
  33. Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K. 33.  et al. 2013. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499:7457214–18 [Google Scholar]
  34. Getz G, Höfling H, Mesirov JP, Golub TR, Meyerson M. 34.  et al. 2007. Comment on “The consensus coding sequences of human breast and colorectal cancers.”. Science 317:58441500 [Google Scholar]
  35. Gonzalez-Perez A, Lopez-Bigas N. 35.  2012. Functional impact bias reveals cancer drivers. Nucleic Acids Res. 40:21e169 [Google Scholar]
  36. Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ. 36.  et al. 2008. An integrated genomic analysis of human glioblastoma multiforme. Science 321:58971807–12 [Google Scholar]
  37. Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C. 37.  et al. 2012. The landscape of cancer genes and mutational processes in breast cancer. Nature 486:7403400–4 [Google Scholar]
  38. Evans P, Avey S, Kong Y, Krauthammer M. 38.  2013. Adjusting for background mutation frequency biases improves the identification of cancer driver genes. IEEE Trans. NanoBiosci. 12:3150–57 [Google Scholar]
  39. Hodis E, Watson IR, Kryukov GV, Arold ST, Imielinski M. 39.  et al. 2012. A landscape of driver mutations in melanoma. Cell 150:2251–63 [Google Scholar]
  40. Jeon S, Lambert PF. 40.  1995. Integration of human papillomavirus type 16 DNA into the human genome leads to increased stability of E6 and E7 mRNAs: implications for cervical carcinogenesis. PNAS 92:51654–58 [Google Scholar]
  41. Eick D, Piechaczyk M, Henglein B, Blanchard JM, Traub B. 41.  et al. 1985. Aberrant c-myc RNAs of Burkitt's lymphoma cells have longer half-lives. EMBO J. 4:13B3717–25 [Google Scholar]
  42. Wiestner A, Tehrani M, Chiorazzi M, Wright G, Gibellini F. 42.  et al. 2007. Point mutations and genomic deletions in CCND1 create stable truncated cyclin D1 mRNAs that are associated with increased proliferation rate and shorter survival. Blood 109:114599–606 [Google Scholar]
  43. Hodgkinson A, Chen Y, Eyre-Walker A. 43.  2012. The large-scale distribution of somatic mutations in cancer genomes. Hum. Mutat. 33:1136–43 [Google Scholar]
  44. Pleasance ED, Cheetham RK, Stephens PJ, McBride DJ, Humphray SJ. 44.  et al. 2010. A comprehensive catalogue of somatic mutations from a human cancer genome. Nature 463:7278191–96 [Google Scholar]
  45. Lohr JG, Stojanov P, Lawrence MS, Auclair D, Chapuy B. 45.  et al. 2012. Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing. PNAS 109:103879–84 [Google Scholar]
  46. 46. Cancer Genome Atlas Res. Netw 2012. Comprehensive genomic characterization of squamous cell lung cancers. Nature 489:7417519–25 [Google Scholar]
  47. 47. Cancer Genome Atlas Res. Netw 2012. Comprehensive molecular characterization of human colon and rectal cancer. Nature 487:7407330–37 [Google Scholar]
  48. Hua X, Xu H, Yang Y, Zhu J, Liu P, Lu Y. 48.  2013. DrGaP: a powerful tool for identifying driver genes and pathways in cancer sequencing studies. Am. J. Hum. Genet. 93:3439–51 [Google Scholar]
  49. Eshleman JR, Markowitz SD. 49.  1995. Microsatellite instability in inherited and sporadic neoplasms. Curr. Opin. Oncol. 7:183–89 [Google Scholar]
  50. Alhopuro P, Sammalkorpi H, Niittymäki I, Biström M, Raitila A. 50.  et al. 2012. Candidate driver genes in microsatellite-unstable colorectal cancer. Int. J. Cancer 130:71558–66 [Google Scholar]
  51. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SAJR, Behjati S. 51.  et al. 2013. Signatures of mutational processes in human cancer. Nature 500:7463415–21 [Google Scholar]
  52. Lada AG, Dhar A, Boissy RJ, Hirano M, Rubel AA. 52.  et al. 2012. AID/APOBEC cytosine deaminase induces genome-wide kataegis. Biol. Direct. 7:47 discussion 47 [Google Scholar]
  53. Nik-Zainal S, Alexandrov LB, Wedge DC, Van Loo P, Greenman CD. 53.  et al. 2012. Mutational processes molding the genomes of 21 breast cancers. Cell 149:5979–93 [Google Scholar]
  54. Roberts SA, Lawrence MS, Klimczak LJ, Grimm SA, Fargo D. 54.  et al. 2013. An APOBEC cytidine deaminase mutagenesis pattern is widespread in human cancers. Nat. Genet. 45:9970–76 [Google Scholar]
  55. Burns MB, Temiz NA, Harris RS. 55.  2013. Evidence for APOBEC3B mutagenesis in multiple human cancers. Nat. Genet. 45:9977–83 [Google Scholar]
  56. Wood LD, Parsons DW, Jones S, Lin J, Sjöblom T. 56.  et al. 2007. The genomic landscapes of human breast and colorectal cancers. Science 318:58531108–13 [Google Scholar]
  57. 57. Cancer Genome Atlas Res. Netw 2011. Integrated genomic analyses of ovarian carcinoma. Nature 474:7353609–15 [Google Scholar]
  58. Wong CC, Martincorena I, Rust AG, Rashid M, Alifrangis C. 58.  et al. 2014. Inactivating CUX1 mutations promote tumorigenesis. Nat. Genet. 46:133–38 [Google Scholar]
  59. Fröhling S, Scholl C, Levine RL, Loriaux M, Boggon TJ. 59.  et al. 2007. Identification of driver and passenger mutations of FLT3 by high-throughput DNA sequence analysis and functional assessment of candidate alleles. Cancer Cell 12:6501–13 [Google Scholar]
  60. Lin J, Gan CM, Zhang X, Jones S, Sjöblom T. 60.  et al. 2007. A multidimensional analysis of genes mutated in breast and colorectal cancers. Genome Res. 17:91304–18 [Google Scholar]
  61. Shah SP, Roth A, Goya R, Oloumi A, Ha G. 61.  et al. 2012. The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature 486:7403395–99 [Google Scholar]
  62. Wendl MC, Wallis JW, Lin L, Kandoth C, Mardis ER. 62.  et al. 2011. PathScan: a tool for discerning mutational significance in groups of putative cancer genes. Bioinformatics 27:121595–602 [Google Scholar]
  63. Dees ND, Zhang Q, Kandoth C, Wendl MC, Schierding W. 63.  et al. 2012. MuSiC: identifying mutational significance in cancer genomes. Genome Res. 22:81589–98 [Google Scholar]
  64. Babaei S, Hulsman M, Reinders M, de Ridder J. 64.  2013. Detecting recurrent gene mutation in interaction network context using multi-scale graph diffusion. BMC Bioinform. 14:29 [Google Scholar]
  65. Cerami E, Demir E, Schultz N, Taylor BS, Sander C. 65.  2010. Automated network analysis identifies core pathways in glioblastoma. PLOS ONE 5:2e8918 [Google Scholar]
  66. Brosh R, Rotter V. 66.  2009. When mutants gain new powers: news from the mutant p53 field. Nat. Rev. Cancer 9:10701–13 [Google Scholar]
  67. Vandin F, Upfal E, Raphael BJ. 67.  2012. De novo discovery of mutated driver pathways in cancer. Genome Res. 22:2375–85 [Google Scholar]
  68. Miller CA, Settle SH, Sulman EP, Aldape KD, Milosavljevic A. 68.  2011. Discovering functional modules by identifying recurrent and mutually exclusive mutational patterns in tumors. BMC Med. Genomics 4:34 [Google Scholar]
  69. Leiserson MDM, Blokh D, Sharan R, Raphael BJ. 69.  2013. Simultaneous identification of multiple driver pathways in cancer. PLOS Comput. Biol. 9:5e1003054 [Google Scholar]
  70. Yeang CH, McCormick F, Levine A. 70.  2008. Combinatorial patterns of somatic gene mutations in cancer. FASEB J. 22:82605–22 [Google Scholar]
  71. Peña-Llopis S, Christie A, Xie XJ, Brugarolas J. 71.  2013. Cooperation and antagonism among cancer genes: the renal cancer paradigm. Cancer Res. 73:144173–79 [Google Scholar]
  72. Kaminker JS, Zhang Y, Watanabe C, Zhang Z. 72.  2007. CanPredict: a computational tool for predicting cancer-associated missense mutations. Nucleic Acids Res. 35:Web Server IssueW595–98 [Google Scholar]
  73. Torkamani A, Schork NJ. 73.  2008. Prediction of cancer driver mutations in protein kinases. Cancer Res. 68:61675–82 [Google Scholar]
  74. D'Antonio M, Ciccarelli FD. 74.  2013. Integrated analysis of recurrent properties of cancer genes to identify novel drivers. Genome Biol. 14:5R52 [Google Scholar]
  75. Lee W, Jiang Z, Liu J, Haverty PM, Guan Y. 75.  et al. 2010. The mutation spectrum revealed by paired genome sequences from a lung cancer patient. Nature 465:7297473–77 [Google Scholar]
  76. Rambaldi D, Giorgi FM, Capuani F, Ciliberto A, Ciccarelli FD. 76.  2008. Low duplicability and network fragility of cancer genes. Trends Genet. 24:9427–30 [Google Scholar]
  77. Jonsson PF, Bates PA. 77.  2006. Global topological features of cancer proteins in the human interactome. Bioinformatics 22:182291–97 [Google Scholar]
  78. Li BQ, You J, Chen L, Zhang J, Zhang N. 78.  et al. 2013. Identification of lung-cancer-related genes with the shortest path approach in a protein-protein interaction network. BioMed. Res. Int. 2013:267375 [Google Scholar]
  79. Ciccarelli FD. 79.  2010. The (r)evolution of cancer genetics. BMC Biol. 8:74 [Google Scholar]
  80. Reimand J, Wagih O, Bader GD. 80.  2013. The mutational landscape of phosphorylation signaling in cancer. Sci. Rep. 3:2651 [Google Scholar]
  81. Gonzalez-Perez A, Lopez-Bigas N. 81.  2011. Improving the assessment of the outcome of nonsynonymous SNVs with a consensus deleteriousness score. Am. J. Hum. Genet. 88:4440–49 [Google Scholar]
  82. Mao Y, Chen H, Liang H, Meric-Bernstam F, Mills GB, Chen K. 82.  2013. CanDrA: cancer-specific driver missense mutation annotation with optimized features. PLOS ONE 8:10e77945 [Google Scholar]
  83. Tamborero D, Gonzalez-Perez A, Perez-Llamas C, Deu-Pons J, Kandoth C. 83.  et al. 2013. Comprehensive identification of mutational cancer driver genes across 12 tumor types. Sci. Rep. 3:2650 [Google Scholar]
  84. Gonzalez-Perez A, Perez-Llamas C, Deu-Pons J, Tamborero D, Schroeder MP. 84.  et al. 2013. IntOGen-mutations identifies cancer drivers across tumor types. Nat. Methods 10:111081–82 [Google Scholar]
  85. Hanahan D, Weinberg RA. 85.  2011. Hallmarks of cancer: the next generation. Cell 144:5646–74 [Google Scholar]
  86. Burkholder B, Huang RY, Burgess R, Luo S, Jones VS. 86.  et al. 2014. Tumor-induced perturbations of cytokines and immune cell networks. Biochim. Biophys. Acta 1845:2182–201 [Google Scholar]
  87. Jacks T, Fazeli A, Schmitt EM, Bronson RT, Goodell MA, Weinberg RA. 87.  1992. Effects of an Rb mutation in the mouse. Nature 359:6393295–300 [Google Scholar]
  88. Jacks T, Remington L, Williams BO, Schmitt EM, Halachmi S. 88.  et al. 1994. Tumor spectrum analysis in p53-mutant mice. Curr. Biol. 4:11–7 [Google Scholar]
  89. Malkin D, Li FP, Strong LC, Fraumeni JF Jr, Nelson CE. 89.  et al. 1990. Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms. Science 250:49851233–38 [Google Scholar]
  90. Cheon DJ, Orsulic S. 90.  2011. Mouse models of cancer. Annu. Rev. Pathol. Mech. Dis. 6:95–119 [Google Scholar]
  91. Watson IR, Takahashi K, Futreal PA, Chin L. 91.  2013. Emerging patterns of somatic mutations in cancer. Nat. Rev. Genet. 14:10703–18 [Google Scholar]
  92. Okuno K, Ohta S, Kato H, Taga T, Sugita K, Takeuchi Y. 92.  2010. Expression of neural stem cell markers in malignant rhabdoid tumor cell lines. Oncol. Rep. 23:2485–92 [Google Scholar]
  93. Garraway LA, Widlund HR, Rubin MA, Getz G, Berger AJ. 93.  et al. 2005. Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma. Nature 436:7047117–22 [Google Scholar]
  94. Stepanenko AA, Vassetzky YS, Kavsan VM. 94.  2013. Antagonistic functional duality of cancer genes. Gene 529:2199–207 [Google Scholar]
  95. Boehm JS, Hahn WC. 95.  2011. Towards systematic functional characterization of cancer genomes. Nat. Rev. Genet. 12:7487–98 [Google Scholar]
  96. Heyer J, Kwong LN, Lowe SW, Chin L. 96.  2010. Non-germline genetically engineered mouse models for translational cancer research. Nat. Rev. Cancer 10:7470–80 [Google Scholar]
  97. Bozic I, Antal T, Ohtsuki H, Carter H, Kim D. 97.  et al. 2010. Accumulation of driver and passenger mutations during tumor progression. PNAS 107:4318545–50 [Google Scholar]
  98. Zack TI, Schumacher SE, Carter SL, Cherniack AD, Saksena G. 98.  et al. 2013. Pan-cancer patterns of somatic copy number alteration. Nat. Genet. 45:101134–40 [Google Scholar]
  99. Hagerstrand D, Tong A, Schumacher SE, Ilic N, Shen RR. 99.  et al. 2013. Systematic interrogation of 3q26 identifies TLOC1 and SKIL as cancer drivers. Cancer Discov. 3:91044–57 [Google Scholar]
  100. Rueda OM, Diaz-Uriarte R. 100.  2009. Detection of recurrent copy number alterations in the genome: taking among-subject heterogeneity seriously. BMC Bioinform. 10:308 [Google Scholar]
  101. Bignell GR, Greenman CD, Davies H, Butler AP, Edkins S. 101.  et al. 2010. Signatures of mutation and selection in the cancer genome. Nature 463:7283893–98 [Google Scholar]
  102. Beroukhim R, Getz G, Nghiemphu L, Barretina J, Hsueh T. 102.  et al. 2007. Assessing the significance of chromosomal aberrations in cancer: methodology and application to glioma. PNAS 104:5020007–12 [Google Scholar]
  103. Akavia UD, Litvin O, Kim J, Sanchez-Garcia F, Kotliar D. 103.  et al. 2010. An integrated approach to uncover drivers of cancer. Cell 143:61005–17 [Google Scholar]
  104. Stephens PJ, Greenman CD, Fu B, Yang F, Bignell GR. 104.  et al. 2011. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 144:127–40 [Google Scholar]
  105. Liu P, Erez A, Nagamani SCS, Dhar SU, Kołodziejska KE. 105.  et al. 2011. Chromosome catastrophes involve replication mechanisms generating complex genomic rearrangements. Cell 146:6889–903 [Google Scholar]
  106. Baca SC, Prandi D, Lawrence MS, Mosquera JM, Romanel A. 106.  et al. 2013. Punctuated evolution of prostate cancer genomes. Cell 153:3666–77 [Google Scholar]
  107. Zhang CZ, Leibowitz ML, Pellman D. 107.  2013. Chromothripsis and beyond: rapid genome evolution from complex chromosomal rearrangements. Genes Dev. 27:232513–30 [Google Scholar]
  108. Malhotra A, Lindberg M, Faust GG, Leibowitz ML, Clark RA. 108.  et al. 2013. Breakpoint profiling of 64 cancer genomes reveals numerous complex rearrangements spawned by homology-independent mechanisms. Genome Res. 23:5762–76 [Google Scholar]
  109. Huang FW, Hodis E, Xu MJ, Kryukov GV, Chin L, Garraway LA. 109.  2013. Highly recurrent TERT promoter mutations in human melanoma. Science 339:6122957–59 [Google Scholar]
  110. Maurano MT, Humbert R, Rynes E, Thurman RE, Haugen E. 110.  et al. 2012. Systematic localization of common disease-associated variation in regulatory DNA. Science 337:60991190–95 [Google Scholar]
  111. Ward LD, Kellis M. 111.  2012. Interpreting noncoding genetic variation in complex traits and human disease. Nat. Biotechnol. 30:111095–106 [Google Scholar]
  112. Horn S, Figl A, Rachakonda PS, Fischer C, Sucker A. 112.  et al. 2013. TERT promoter mutations in familial and sporadic melanoma. Science 339:6122959–61 [Google Scholar]
  113. Wu RC, Ayhan A, Maeda D, Kim KR, Clarke BA. 113.  et al. 2013. Frequent somatic mutations of the telomerase reverse transcriptase promoter in ovarian clear cell carcinoma but not in other major types of gynecologic malignancies. J. Pathol. 232:4473–81 [Google Scholar]
  114. Wu S, Huang P, Li C, Huang Y, Li X. 114.  et al. 2013. Telomerase reverse transcriptase gene promoter mutations help discern the origin of urogenital tumors: a genomic and molecular study. Eur. Urol. 65:2274–77 [Google Scholar]
  115. Lindsey JC, Schwalbe EC, Potluri S, Bailey S, Williamson D, Clifford SC. 115.  2013. TERT promoter mutation and aberrant hypermethylation are associated with elevated expression in medulloblastoma and characterise the majority of non-infant SHH subgroup tumours. Acta Neuropathol. 127:2307–9 [Google Scholar]
  116. Liu T, Wang N, Cao J, Sofiadis A, Dinets A. 116.  et al. 2013. The age- and shorter telomere–dependent TERT promoter mutation in follicular thyroid cell–derived carcinomas. Oncogene 334978–84
  117. Goutagny S, Nault JC, Mallet M, Henin D, Zucman Rossi J, Kalamarides M. 117.  2014. High incidence of activating TERT promoter mutations in meningiomas undergoing malignant progression. Brain Pathol. 24:2184–89 [Google Scholar]
  118. Qu Y, Shi L, Wang D, Zhang B, Yang Q. 118.  et al. 2014. Low frequency of TERT promoter mutations in a large cohort of gallbladder and gastric cancers. Int. J. Cancer 134:122993–94 [Google Scholar]
  119. 119. ENCODE Project Consortium 2004. The ENCODE (ENCyclopedia Of DNA Elements) Project. Science 306:5696636–40 [Google Scholar]
  120. Boyle AP, Hong EL, Hariharan M, Cheng Y, Schaub MA. 120.  et al. 2012. Annotation of functional variation in personal genomes using RegulomeDB. Genome Res. 22:91790–97 [Google Scholar]
  121. Whitfield TW, Wang J, Collins PJ, Partridge EC, Aldred SF. 121.  et al. 2012. Functional analysis of transcription factor binding sites in human promoters. Genome Biol. 13:9R50 [Google Scholar]
  122. Bryne JC, Valen E, Tang ME, Marstrand T, Winther O. 122.  et al. 2008. JASPAR, the open access database of transcription factor-binding profiles: new content and tools in the 2008 update. Nucleic Acids Res. 36:Database IssueD102–6 [Google Scholar]
  123. Hoffman MM, Birney E. 123.  2010. An effective model for natural selection in promoters. Genome Res. 20:5685–92 [Google Scholar]
  124. Pleasance ED, Stephens PJ, O'Meara S, McBride DJ, Meynert A. 124.  et al. 2010. A small-cell lung cancer genome with complex signatures of tobacco exposure. Nature 463:7278184–90 [Google Scholar]
  125. 125. 1000 Genomes Proj. Consort 2012. An integrated map of genetic variation from 1,092 human genomes. Nature 491:742256–65 [Google Scholar]
  126. Venter JC, Adams MD, Myers EW, Li PW, Mural RJ. 126.  et al. 2001. The sequence of the human genome. Science 291:55071304–51 [Google Scholar]
  127. Cowper-Sal·lari R, Zhang X, Wright JB, Bailey SD, Cole MD. 127.  et al. 2012. Breast cancer risk-associated SNPs modulate the affinity of chromatin for FOXA1 and alter gene expression. Nat. Genet. 44:111191–98 [Google Scholar]
  128. Michalova E, Vojtesek B, Hrstka R. 128.  2013. Impaired pre-mRNA processing and altered architecture of 3′ untranslated regions contribute to the development of human disorders. Int. J. Mol. Sci. 14:815681–94 [Google Scholar]
  129. Xu N, Chen CY, Shyu AB. 129.  1997. Modulation of the fate of cytoplasmic mRNA by AU-rich elements: key sequence features controlling mRNA deadenylation and decay. Mol. Cell. Biol. 17:84611–21 [Google Scholar]
  130. Esquela-Kerscher A, Slack FJ. 130.  2006. Oncomirs—microRNAs with a role in cancer. Nat. Rev. Cancer 6:4259–69 [Google Scholar]
  131. Mayr C, Hemann MT, Bartel DP. 131.  2007. Disrupting the pairing between let-7 and Hmga2 enhances oncogenic transformation. Science 315:58181576–79 [Google Scholar]
  132. Zhou L, Yuan Q, Yang M. 132.  2012. A functional germline variant in the P53 polyadenylation signal and risk of esophageal squamous cell carcinoma. Gene 506:2295–97 [Google Scholar]
  133. Stacey SN, Sulem P, Jonasdottir A, Masson G, Gudmundsson J. 133.  et al. 2011. A germline variant in the TP53 polyadenylation signal confers cancer susceptibility. Nat. Genet. 43:111098–103 [Google Scholar]
  134. Lee YS, Dutta A. 134.  2009. MicroRNAs in cancer. Annu. Rev. Pathol. Mech. Dis. 4:199–227 [Google Scholar]
  135. Shahrouki P, Larsson E. 135.  2012. The non-coding oncogene: a case of missing DNA evidence?. Front. Genet. 3:170 [Google Scholar]
  136. Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E. 136.  et al. 2004. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. PNAS 101:92999–3004 [Google Scholar]
  137. Zhang L, Huang J, Yang N, Greshock J, Megraw MS. 137.  et al. 2006. MicroRNAs exhibit high frequency genomic alterations in human cancer. PNAS 103:249136–41 [Google Scholar]
  138. Thomson JM, Newman M, Parker JS, Morin-Kensicki EM, Wright T, Hammond SM. 138.  2006. Extensive post-transcriptional regulation of microRNAs and its implications for cancer. Genes Dev. 20:162202–7 [Google Scholar]
  139. Lujambio A, Ropero S, Ballestar E, Fraga MF, Cerrato C. 139.  et al. 2007. Genetic unmasking of an epigenetically silenced microRNA in human cancer cells. Cancer Res. 67:41424–29 [Google Scholar]
  140. Lehmann U, Hasemeier B, Christgen M, Müller M, Römermann D. 140.  et al. 2008. Epigenetic inactivation of microRNA gene hsa-mir-9-1 in human breast cancer. J. Pathol. 214:117–24 [Google Scholar]
  141. Anglesio MS, Wang Y, Yang W, Senz J, Wan A. 141.  et al. 2013. Cancer-associated somatic DICER1 hotspot mutations cause defective miRNA processing and reverse-strand expression bias to predominantly mature 3p strands through loss of 5p strand cleavage. J. Pathol. 229:3400–9 [Google Scholar]
  142. Muralidhar B, Goldstein LD, Ng G, Winder DM, Palmer RD. 142.  et al. 2007. Global microRNA profiles in cervical squamous cell carcinoma depend on Drosha expression levels. J. Pathol. 212:4368–77 [Google Scholar]
  143. Crosby ME, Kulshreshtha R, Ivan M, Glazer PM. 143.  2009. MicroRNA regulation of DNA repair gene expression in hypoxic stress. Cancer Res. 69:31221–29 [Google Scholar]
  144. Donohoe DR, Bultman SJ. 144.  2012. Metaboloepigenetics: interrelationships between energy metabolism and epigenetic control of gene expression. J. Cell. Physiol. 227:93169–77 [Google Scholar]
  145. Baylin SB, Jones PA. 145.  2011. A decade of exploring the cancer epigenome—biological and translational implications. Nat. Rev. Cancer 11:10726–34 [Google Scholar]
  146. Sproul D, Kitchen RR, Nestor CE, Dixon JM, Sims AH. 146.  et al. 2012. Tissue of origin determines cancer-associated CpG island promoter hypermethylation patterns. Genome Biol. 13:10R84 [Google Scholar]
  147. Schuebel KE, Chen W, Cope L, Glöckner SC, Suzuki H. 147.  et al. 2007. Comparing the DNA hypermethylome with gene mutations in human colorectal cancer. PLOS Genet. 3:9e157 [Google Scholar]
  148. Nguyen CT, Weisenberger DJ, Velicescu M, Gonzales FA, Lin JCY. 148.  et al. 2002. Histone H3–lysine 9 methylation is associated with aberrant gene silencing in cancer cells and is rapidly reversed by 5-aza-2′-deoxycytidine. Cancer Res. 62:226456–61 [Google Scholar]
  149. Fahrner JA, Eguchi S, Herman JG, Baylin SB. 149.  2002. Dependence of histone modifications and gene expression on DNA hypermethylation in cancer. Cancer Res. 62:247213–18 [Google Scholar]
  150. Kondo Y, Shen L, Cheng AS, Ahmed S, Boumber Y. 150.  et al. 2008. Gene silencing in cancer by histone H3 lysine 27 trimethylation independent of promoter DNA methylation. Nat. Genet. 40:6741–50 [Google Scholar]
  151. Fraga MF, Ballestar E, Villar-Garea A, Boix-Chornet M, Espada J. 151.  et al. 2005. Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nat. Genet. 37:4391–400 [Google Scholar]
  152. Fu Y, He C. 152.  2012. Nucleic acid modifications with epigenetic significance. Curr. Opin. Chem. Biol. 16:5–6516–24 [Google Scholar]
  153. Tan M, Luo H, Lee S, Jin F, Yang JS. 153.  et al. 2011. Identification of 67 histone marks and histone lysine crotonylation as a new type of histone modification. Cell 146:61016–28 [Google Scholar]
  154. Beck S, Bernstein BE, Campbell RM, Costello JF, Dhanak D. 154.  et al. 2012. A blueprint for an international cancer epigenome consortium. A report from the AACR Cancer Epigenome Task Force. Cancer Res. 72:246319–24 [Google Scholar]
  155. Frühwald MC, O'Dorisio MS, Dai Z, Tanner SM, Balster DA. 155.  et al. 2001. Aberrant promoter methylation of previously unidentified target genes is a common abnormality in medulloblastomas—implications for tumor biology and potential clinical utility. Oncogene 20:365033–42 [Google Scholar]
  156. Anderton JA, Lindsey JC, Lusher ME, Gilbertson RJ, Bailey S. 156.  et al. 2008. Global analysis of the medulloblastoma epigenome identifies disease-subgroup-specific inactivation of COL1A2. Neuro-Oncology 10:6981–94 [Google Scholar]
  157. Hosoya K, Yamashita S, Ando T, Nakajima T, Itoh F, Ushijima T. 157.  2009. Adenomatous polyposis coli 1A is likely to be methylated as a passenger in human gastric carcinogenesis. Cancer Lett. 285:2182–89 [Google Scholar]
  158. Kalari S, Pfeifer GP. 158.  2010. Identification of driver and passenger DNA methylation in cancer by epigenomic analysis. Adv. Genet. 70:277–308 [Google Scholar]
  159. Feinberg AP, Ohlsson R, Henikoff S. 159.  2006. The epigenetic progenitor origin of human cancer. Nat. Rev. Genet. 7:121–33 [Google Scholar]
  160. Fertig EJ, Markovic A, Danilova LV, Gaykalova DA, Cope L. 160.  et al. 2013. Preferential activation of the Hedgehog pathway by epigenetic modulations in HPV negative HNSCC identified with meta-pathway analysis. PLOS ONE 8:11e78127 [Google Scholar]
  161. Herman JG, Latif F, Weng Y, Lerman MI, Zbar B. 161.  et al. 1994. Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma. PNAS 91:219700–4 [Google Scholar]
  162. Konermann S, Brigham MD, Trevino AE, Hsu PD, Heidenreich M. 162.  et al. 2013. Optical control of mammalian endogenous transcription and epigenetic states. Nature 500:7463472–76 [Google Scholar]
  163. Maeder ML, Angstman JF, Richardson ME, Linder SJ, Cascio VM. 163.  et al. 2013. Targeted DNA demethylation and activation of endogenous genes using programmable TALE-TET1 fusion proteins. Nat. Biotechnol. 31:121137–42 [Google Scholar]
  164. Mendenhall EM, Williamson KE, Reyon D, Zou JY, Ram O. 164.  et al. 2013. Locus-specific editing of histone modifications at endogenous enhancers. Nat. Biotechnol. 31:121133–36 [Google Scholar]
  165. Feltus FA, Lee EK, Costello JF, Plass C, Vertino PM. 165.  2003. Predicting aberrant CpG island methylation. PNAS 100:2112253–58 [Google Scholar]
  166. Schlesinger Y, Straussman R, Keshet I, Farkash S, Hecht M. 166.  et al. 2007. Polycomb-mediated methylation on Lys27 of histone H3 pre-marks genes for de novo methylation in cancer. Nat. Genet. 39:2232–36 [Google Scholar]
  167. Viré E, Brenner C, Deplus R, Blanchon L, Fraga M. 167.  et al. 2006. The Polycomb group protein EZH2 directly controls DNA methylation. Nature 439:7078871–74 [Google Scholar]
  168. Estécio MRH, Gallegos J, Vallot C, Castoro RJ, Chung W. 168.  et al. 2010. Genome architecture marked by retrotransposons modulates predisposition to DNA methylation in cancer. Genome Res. 20:101369–82 [Google Scholar]
  169. McCabe MT, Lee EK, Vertino PM. 169.  2009. A multifactorial signature of DNA sequence and polycomb binding predicts aberrant CpG island methylation. Cancer Res. 69:1282–91 [Google Scholar]
  170. Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB, Issa JP. 170.  1999. CpG island methylator phenotype in colorectal cancer. PNAS 96:158681–86 [Google Scholar]
  171. Shen H, Laird PW. 171.  2013. Interplay between the cancer genome and epigenome. Cell 153:138–55 [Google Scholar]
  172. Richly H, Aloia L, Di Croce L. 172.  2011. Roles of the Polycomb group proteins in stem cells and cancer. Cell Death Dis. 2:e204 [Google Scholar]
  173. De Carvalho DD, Sharma S, You JS, Su SF, Taberlay PC. 173.  et al. 2012. DNA methylation screening identifies driver epigenetic events of cancer cell survival. Cancer Cell 21:5655–67 [Google Scholar]
  174. Hon GC, Hawkins RD, Caballero OL, Lo C, Lister R. 174.  et al. 2012. Global DNA hypomethylation coupled to repressive chromatin domain formation and gene silencing in breast cancer. Genome Res. 22:2246–58 [Google Scholar]
  175. Younesy H, Möller T, Heravi-Moussavi A, Cheng JB, Costello JF. 175.  et al. 2014. ALEA: a toolbox for allele-specific epigenomics analysis. Bioinformatics 30:81172–74 [Google Scholar]
  176. Moulton T, Crenshaw T, Hao Y, Moosikasuwan J, Lin N. 176.  et al. 1994. Epigenetic lesions at the H19 locus in Wilms' tumour patients. Nat. Genet. 7:3440–47 [Google Scholar]
  177. Esteller M, Catasus L, Matias-Guiu X, Mutter GL, Prat J. 177.  et al. 1999. hMLH1 promoter hypermethylation is an early event in human endometrial tumorigenesis. Am. J. Pathol. 155:51767–72 [Google Scholar]
  178. Khan DH, Gonzalez C, Cooper C, Sun JM, Chen HY. 178.  et al. 2013. RNA-dependent dynamic histone acetylation regulates MCL1 alternative splicing. Nucleic Acids Res. 42:31656–70 [Google Scholar]
  179. Khan DH, Jahan S, Davie JR. 179.  2012. Pre-mRNA splicing: role of epigenetics and implications in disease. Adv. Biol. Regul. 52:3377–88 [Google Scholar]
  180. Laird PW, Jackson-Grusby L, Fazeli A, Dickinson SL, Jung WE. 180.  et al. 1995. Suppression of intestinal neoplasia by DNA hypomethylation. Cell 81:2197–205 [Google Scholar]
  181. Yamada Y, Jackson-Grusby L, Linhart H, Meissner A, Eden A. 181.  et al. 2005. Opposing effects of DNA hypomethylation on intestinal and liver carcinogenesis. PNAS 102:3813580–85 [Google Scholar]
  182. Gaudet F, Hodgson JG, Eden A, Jackson-Grusby L, Dausman J. 182.  et al. 2003. Induction of tumors in mice by genomic hypomethylation. Science 300:5618489–92 [Google Scholar]
  183. Gonzalez-Perez A, Jene-Sanz A, Lopez-Bigas N. 183.  2013. The mutational landscape of chromatin regulatory factors across 4,623 tumor samples. Genome Biol. 14:9r106 [Google Scholar]
  184. Roberts CWM, Biegel JA. 184.  2009. The role of SMARCB1/INI1 in development of rhabdoid tumor. Cancer Biol. Ther. 8:5412–16 [Google Scholar]
  185. Shain AH, Pollack JR. 185.  2013. The spectrum of SWI/SNF mutations, ubiquitous in human cancers. PLOS ONE 8:1e55119 [Google Scholar]
  186. 186. Cancer Genome Atlas Res. Netw 2013. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 368:222059–74 [Google Scholar]
  187. Roberts CW, Galusha SA, McMenamin ME, Fletcher CD, Orkin SH. 187.  2000. Haploinsufficiency of Snf5 (integrase interactor 1) predisposes to malignant rhabdoid tumors in mice. PNAS 97:2513796–800 [Google Scholar]
  188. Béguelin W, Popovic R, Teater M, Jiang Y, Bunting KL. 188.  et al. 2013. EZH2 is required for germinal center formation and somatic EZH2 mutations promote lymphoid transformation. Cancer Cell 23:5677–92 [Google Scholar]
  189. Schwartzentruber J, Korshunov A, Liu XY, Jones DTW, Pfaff E. 189.  et al. 2012. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature 482:7384226–31 [Google Scholar]
  190. Losman JA, Kaelin WG Jr. 190.  2013. What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer. Genes Dev. 27:8836–52 [Google Scholar]
  191. Yan J, Ng SB, Tay JLS, Lin B, Koh TL. 191.  et al. 2013. EZH2 overexpression in natural killer/T-cell lymphoma confers growth advantage independently of histone methyltransferase activity. Blood 121:224512–20 [Google Scholar]
  192. Yap DB, Chu J, Berg T, Schapira M, Cheng SW. 192.  et al. 2011. Somatic mutations at EZH2 Y641 act dominantly through a mechanism of selectively altered PRC2 catalytic activity, to increase H3K27 trimethylation. Blood 117:82451–59 [Google Scholar]
  193. Ernst T, Chase AJ, Score J, Hidalgo-Curtis CE, Bryant C. 193.  et al. 2010. Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat. Genet. 42:8722–26 [Google Scholar]
  194. Hirst M, Marra MA. 194.  2009. Epigenetics and human disease. Int. J. Biochem. Cell Biol. 41:1136–46 [Google Scholar]
/content/journals/10.1146/annurev-pathol-012414-040312
Loading
  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error