Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Non-coding driver mutations in human cancer

Abstract

Tumour formation involves random mutagenic events and positive evolutionary selection acting on a subset of such events, referred to as driver mutations. A decade of careful surveying of tumour DNA using exome-based analyses has revealed a multitude of protein-coding somatic driver mutations, some of which are clinically actionable. Today, a transition towards whole-genome analysis is well under way, technically enabling the discovery of potential driver mutations occurring outside protein-coding sequences. Mutations are abundant in this vast non-coding space, which is more than 50 times larger than the coding exome, but reliable identification of selection signals in non-coding DNA remains a challenge. In this Review, we discuss recent findings in the field, where the emerging landscape is one in which non-coding driver mutations appear to be relatively infrequent. Nevertheless, we highlight several notable discoveries. We consider possible reasons for the relative absence of non-coding driver events, as well as the difficulties associated with detecting signals of positive selection in non-coding DNA.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Mechanisms by which non-coding mutations can contribute to tumorigenesis.
Fig. 2: Sources of heterogeneity in genomic mutation rate for consideration when assessing signals of selection in non-coding DNA.
Fig. 3: Local vulnerability to ultraviolet mutagenesis at ETS-binding sites is a characteristic feature of strong mutation hotspots in melanoma.
Fig. 4: Top recurrently mutated protein-coding and non-coding elements in PCAWG.

Similar content being viewed by others

References

  1. Huang, F. W. et al. Highly recurrent TERT promoter mutations in human melanoma. Science 339, 957–959 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Horn, S. et al. TERT promoter mutations in familial and sporadic melanoma. Science 339, 959–961 (2013). Together with Huang et al. (2013), this paper establishes non-coding somatic promoter mutations as a mechanism for oncogene activation.

    Article  CAS  PubMed  Google Scholar 

  3. Garraway, L. A. & Lander, E. S. Lessons from the cancer genome. Cell 153, 17–37 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. The ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium. Pan-cancer analysis of whole genomes. Nature 578, 82–93 (2020).

    Article  CAS  Google Scholar 

  5. Rheinbay, E. et al. Analyses of non-coding somatic drivers in 2,658 cancer whole genomes. Nature 578, 102–111 (2020). As the largest and most ambitious pan-cancer study of non-coding somatic mutations to date, this large consortium effort confirmed high prevalence of TERT promoter mutations in human cancer and also suggested additional lower-frequency candidates.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ciriello, G. et al. Emerging landscape of oncogenic signatures across human cancers. Nat. Genet. 45, 1127–1133 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Corona, R. I. et al. Non-coding somatic mutations converge on the PAX8 pathway in ovarian cancer. Nat. Commun. 11, 2020 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hnisz, D. et al. Activation of proto-oncogenes by disruption of chromosome neighborhoods. Science 351, 1454–1458 (2016). This paper establishes disrupted chromatin domain structure as a mechanism for oncogene activation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Schuster, S. L. & Hsieh, A. C. The untranslated regions of mRNAs in cancer. Trends Cancer 5, 245–262 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Shuai, S. et al. The U1 spliceosomal RNA is recurrently mutated in multiple cancers. Nature 574, 712–716 (2019).

    Article  CAS  PubMed  Google Scholar 

  11. Suzuki, H. et al. Recurrent noncoding U1 snRNA mutations drive cryptic splicing in SHH medulloblastoma. Nature 574, 707–711 (2019). Together with Shuai et al. (2019), this paper reports a rare functional somatic mutation in a non-coding RNA.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Belkadi, A. et al. Whole-genome sequencing is more powerful than whole-exome sequencing for detecting exome variants. Proc. Natl Acad. Sci. USA 112, 5473–5478 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhang, X. & Meyerson, M. Illuminating the noncoding genome in cancer. Nat. Cancer 1, 864–872 (2020).

    Article  PubMed  Google Scholar 

  14. Khurana, E. et al. Role of non-coding sequence variants in cancer. Nat. Rev. Genet. 17, 93–108 (2016).

    Article  CAS  PubMed  Google Scholar 

  15. Stratton, M. R., Campbell, P. J. & Futreal, P. A. The cancer genome. Nature 458, 719–724 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Van den Eynden, J., Basu, S. & Larsson, E. Somatic mutation patterns in hemizygous genomic regions unveil purifying selection during tumor evolution. PLoS Genet. 12, e1006506 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Martincorena, I. et al. Universal patterns of selection in cancer and somatic tissues. Cell 171, 1029–1041.e21 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. McFarland, C. D., Korolev, K. S., Kryukov, G. V., Sunyaev, S. R. & Mirny, L. A. Impact of deleterious passenger mutations on cancer progression. Proc. Natl Acad. Sci. USA 110, 2910–2915 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Supek, F. & Lehner, B. Scales and mechanisms of somatic mutation rate variation across the human genome. DNA Repair 81, 102647 (2019).

    Article  CAS  PubMed  Google Scholar 

  20. Schuster-Bockler, B. & Lehner, B. Chromatin organization is a major influence on regional mutation rates in human cancer cells. Nature 488, 504–507 (2012).

    Article  PubMed  CAS  Google Scholar 

  21. Supek, F. & Lehner, B. Differential DNA mismatch repair underlies mutation rate variation across the human genome. Nature 521, 81–84 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Zheng, C. L. et al. Transcription restores DNA repair to heterochromatin, determining regional mutation rates in cancer genomes. Cell Rep. 9, 1228–1234 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Polak, P. et al. Cell-of-origin chromatin organization shapes the mutational landscape of cancer. Nature 518, 360–364 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Alexandrov, L. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013). This paper established the concept of mutational signatures, which are helpful when detecting signals of selection in mutation data and are useful for studying mutational processes active in tumours.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ikehata, H. & Ono, T. The mechanisms of UV mutagenesis. J. Radiat. Res. 52, 115–125 (2011).

    Article  CAS  PubMed  Google Scholar 

  26. Lawrence, M. S. et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499, 214–218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gonzalez-Perez, A., Sabarinathan, R. & Lopez-Bigas, N. Local determinants of the mutational landscape of the human genome. Cell 177, 101–114 (2019).

    Article  CAS  PubMed  Google Scholar 

  28. Frigola, J. et al. Reduced mutation rate in exons due to differential mismatch repair. Nat. Genet. 49, 1684–1692 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Weinhold, N., Jacobsen, A., Schultz, N., Sander, C. & Lee, W. Genome-wide analysis of noncoding regulatory mutations in cancer. Nat. Genet. 46, 1160–1165 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Fredriksson, N. J., Ny, L., Nilsson, J. A. & Larsson, E. Systematic analysis of noncoding somatic mutations and gene expression alterations across 14 tumor types. Nat. Genet. 46, 1258–1263 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. Denisova, E. et al. Frequent DPH3 promoter mutations in skin cancers. Oncotarget 6, 35922–35930 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Araya, C. L. et al. Identification of significantly mutated regions across cancer types highlights a rich landscape of functional molecular alterations. Nat. Genet. 48, 117–125 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Colebatch, A. J. et al. Clustered somatic mutations are frequent in transcription factor binding motifs within proximal promoter regions in melanoma and other cutaneous malignancies. Oncotarget 7, 66569–66585 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Fredriksson, N. J. et al. Recurrent promoter mutations in melanoma are defined by an extended context-specific mutational signature. PLoS Genet. 13, e1006773 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Mao, P. et al. ETS transcription factors induce a unique UV damage signature that drives recurrent mutagenesis in melanoma. Nat. Commun. 9, 2626 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Elliott, K. et al. Elevated pyrimidine dimer formation at distinct genomic bases underlies promoter mutation hotspots in UV-exposed cancers. PLoS Genet. 14, e1007849 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Sabarinathan, R., Mularoni, L., Deu-Pons, J., Gonzalez-Perez, A. & Lopez-Bigas, N. Nucleotide excision repair is impaired by binding of transcription factors to DNA. Nature 532, 264–267 (2016).

    Article  CAS  PubMed  Google Scholar 

  38. Perera, D. et al. Differential DNA repair underlies mutation hotspots at active promoters in cancer genomes. Nature 532, 259–263 (2016).

    Article  CAS  PubMed  Google Scholar 

  39. Teng, G. & Papavasiliou, F. N. Immunoglobulin somatic hypermutation. Annu. Rev. Genet. 41, 107–120 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Migliazza, A. et al. Frequent somatic hypermutation of the 5′ noncoding region of the BCL6 gene in B-cell lymphoma. Proc. Natl Acad. Sci. USA 92, 12520–12524 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Pasqualucci, L. et al. Hypermutation of multiple proto-oncogenes in B-cell diffuse large-cell lymphomas. Nature 412, 341–346 (2001).

    Article  CAS  PubMed  Google Scholar 

  42. Harris, R. S. & Liddament, M. T. Retroviral restriction by APOBEC proteins. Nat. Rev. Immunol. 4, 868–877 (2004).

    Article  CAS  PubMed  Google Scholar 

  43. Roberts, S. A. et al. Clustered mutations in yeast and in human cancers can arise from damaged long single-strand DNA regions. Mol. Cell 46, 424–435 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Nik-Zainal, S. et al. Landscape of somatic mutations in 560 breast cancer whole-genome sequences. Nature 534, 47–54 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Buisson, R. et al. Passenger hotspot mutations in cancer driven by APOBEC3A and mesoscale genomic features. Science 364, eaaw2872 (2019).

  46. Wu, S. et al. Whole-genome sequencing identifies ADGRG6 enhancer mutations and FRS2 duplications as angiogenesis-related drivers in bladder cancer. Nat. Commun. 10, 720 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sun, Y. & Ma, L. New insights into long non-coding RNA MALAT1 in cancer and metastasis. Cancers 11 (2), 216 (2019).

  48. Klec, C., Prinz, F. & Pichler, M. Involvement of the long noncoding RNA NEAT1 in carcinogenesis. Mol. Oncol. 13, 46–60 (2019).

    Article  CAS  PubMed  Google Scholar 

  49. Wedge, D. C. et al. Sequencing of prostate cancers identifies new cancer genes, routes of progression and drug targets. Nat. Genet. 50, 682–692 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Fujimoto, A. et al. Whole-genome mutational landscape and characterization of noncoding and structural mutations in liver cancer. Nat. Genet. 48, 500–509 (2016).

    Article  CAS  PubMed  Google Scholar 

  51. Li, S., Shuch, B. M. & Gerstein, M. B. Whole-genome analysis of papillary kidney cancer finds significant noncoding alterations. PLoS Genet. 13, e1006685 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Imielinski, M., Guo, G. & Meyerson, M. Insertions and deletions target lineage-defining genes in human cancers. Cell 168, 460–472 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. [No authors listed] Cancer genome complexity made simple. Cancer Discov. 10, 480 (2020).

    Article  Google Scholar 

  54. Cieslik, M. & Chinnaiyan, A. M. Global genomics project unravels cancer’s complexity at unprecedented scale. Nature 578, 39–40 (2020).

    Article  CAS  PubMed  Google Scholar 

  55. Li, B. S. et al. MicroRNA-25 promotes gastric cancer migration, invasion and proliferation by directly targeting transducer of ERBB2, 1 and correlates with poor survival. Oncogene 34, 2556–2565 (2015).

    Article  CAS  PubMed  Google Scholar 

  56. Arthur, S. E. et al. Genome-wide discovery of somatic regulatory variants in diffuse large B-cell lymphoma. Nat. Commun. 9, 4001 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Hornshoj, H. et al. Pan-cancer screen for mutations in non-coding elements with conservation and cancer specificity reveals correlations with expression and survival. NPJ Genom. Med. 3, 1 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Urbanek-Trzeciak, M. O. et al. Pan-cancer analysis of somatic mutations in miRNA genes. EBioMedicine 61, 103051 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Wang, W., Wei, Z., Lam, T. W. & Wang, J. Next generation sequencing has lower sequence coverage and poorer SNP-detection capability in the regulatory regions. Sci. Rep. 1, 55 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Rheinbay, E. et al. Recurrent and functional regulatory mutations in breast cancer. Nature 547, 55–60 (2017). This paper identifies recurrent functional promoter mutations in a cancer-relevant gene through promoter capture sequencing, highlighting the limitations of regular WGS.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Li, K. et al. Noncoding variants connect enhancer dysregulation with nuclear receptor signaling in hematopoietic malignancies. Cancer Discov. 10, 724–745 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Dabney, J. & Meyer, M. Length and GC-biases during sequencing library amplification: a comparison of various polymerase-buffer systems with ancient and modern DNA sequencing libraries. Biotechniques 52, 87–94 (2012).

    Article  CAS  PubMed  Google Scholar 

  63. Payne, J. L. & Wagner, A. Mechanisms of mutational robustness in transcriptional regulation. Front. Genet. 6, 322 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Bell, R. J. et al. Cancer. The transcription factor GABP selectively binds and activates the mutant TERT promoter in cancer. Science 348, 1036–1039 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Kircher, M. et al. Saturation mutagenesis of twenty disease-associated regulatory elements at single base-pair resolution. Nat. Commun. 10, 3583 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Jung, I. et al. A compendium of promoter-centered long-range chromatin interactions in the human genome. Nat. Genet. 51, 1442–1449 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Kim, K. et al. Chromatin structure-based prediction of recurrent noncoding mutations in cancer. Nat. Genet. 48, 1321–1326 (2016).

    Article  CAS  PubMed  Google Scholar 

  68. Zhou, S. et al. Noncoding mutations target cis-regulatory elements of the FOXA1 plexus in prostate cancer. Nat. Commun. 11, 441 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Castro-Giner, F., Ratcliffe, P. & Tomlinson, I. The mini-driver model of polygenic cancer evolution. Nat. Rev. Cancer 15, 680–685 (2015).

    Article  CAS  PubMed  Google Scholar 

  70. Sonawane, A. R. et al. Understanding tissue-specific gene regulation. Cell Rep. 21, 1077–1088 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Tiong, K. L. & Yeang, C. H. Explaining cancer type specific mutations with transcriptomic and epigenomic features in normal tissues. Sci. Rep. 8, 11456 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Mansour, M. R. et al. Oncogene regulation. An oncogenic super-enhancer formed through somatic mutation of a noncoding intergenic element. Science 346, 1373–1377 (2014). This paper uncovers an intriguing mechanism for oncogene activation involving de novo formation of a super-enhancer through short somatic indels.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Hu, S. et al. Whole-genome noncoding sequence analysis in T-cell acute lymphoblastic leukemia identifies oncogene enhancer mutations. Blood 129, 3264–3268 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Liu, Y. et al. Discovery of regulatory noncoding variants in individual cancer genomes by using cis-X. Nat. Genet. 52, 811–818 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Puente, X. S. et al. Non-coding recurrent mutations in chronic lymphocytic leukaemia. Nature 526, 519–524 (2015).

    Article  CAS  PubMed  Google Scholar 

  76. Liu, E. M. et al. Identification of cancer drivers at CTCF insulators in 1,962 whole genomes. Cell Syst. 8, 446–455.e8 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Guo, Y. A. et al. Mutation hotspots at CTCF binding sites coupled to chromosomal instability in gastrointestinal cancers. Nat. Commun. 9, 1520 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Katainen, R. et al. CTCF/cohesin-binding sites are frequently mutated in cancer. Nat. Genet. 47, 818–821 (2015).

    Article  CAS  PubMed  Google Scholar 

  79. Poulos, R. C. et al. Functional mutations form at CTCF-cohesin binding sites in melanoma due to uneven nucleotide excision repair across the motif. Cell Rep. 17, 2865–2872 (2016).

    Article  CAS  PubMed  Google Scholar 

  80. Kaiser, V. B., Taylor, M. S. & Semple, C. A. Mutational biases drive elevated rates of substitution at regulatory sites across cancer types. PLoS Genet. 12, e1006207 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Zhu, H. et al. Candidate cancer driver mutations in distal regulatory elements and long-range chromatin interaction networks. Mol. Cell 77, 1307–1321.e10 (2020).

    Article  CAS  PubMed  Google Scholar 

  82. Bailey, S. D. et al. Noncoding somatic and inherited single-nucleotide variants converge to promote ESR1 expression in breast cancer. Nat. Genet. 48, 1260–1266 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Tate, J. G. et al. COSMIC: the catalogue of somatic mutations in cancer. Nucleic Acids Res. 47, D941–D947 (2019).

    Article  CAS  PubMed  Google Scholar 

  84. Vinagre, J. et al. Frequency of TERT promoter mutations in human cancers. Nat. Commun. 4, 2185 (2013).

    Article  PubMed  CAS  Google Scholar 

  85. Guo, Y. A., Chang, M. M. & Skanderup, A. J. MutSpot: detection of non-coding mutation hotspots in cancer genomes. NPJ Genom. Med. 5, 26 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  86. Canver, M. C. et al. BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis. Nature 527, 192–197 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Rajagopal, N. et al. High-throughput mapping of regulatory DNA. Nat. Biotechnol. 34, 167–174 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Diao, Y. et al. A new class of temporarily phenotypic enhancers identified by CRISPR/Cas9-mediated genetic screening. Genome Res. 26, 397–405 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Diao, Y. et al. A tiling-deletion-based genetic screen for cis-regulatory element identification in mammalian cells. Nat. Methods 14, 629–635 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Fulco, C. P. et al. Systematic mapping of functional enhancer-promoter connections with CRISPR interference. Science 354, 769–773 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Korkmaz, G. et al. Functional genetic screens for enhancer elements in the human genome using CRISPR-Cas9. Nat. Biotechnol. 34, 192–198 (2016).

    Article  CAS  PubMed  Google Scholar 

  92. Sanjana, N. E. et al. High-resolution interrogation of functional elements in the noncoding genome. Science 353, 1545–1549 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Xie, S., Duan, J., Li, B., Zhou, P. & Hon, G. C. Multiplexed engineering and analysis of combinatorial enhancer activity in single cells. Mol. Cell 66, 285–299.e5 (2017).

    Article  CAS  PubMed  Google Scholar 

  94. Shuai, S. et al. Combined burden and functional impact tests for cancer driver discovery using DriverPower. Nat. Commun. 11, 734 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Lanzos, A. et al. Discovery of cancer driver long noncoding RNAs across 1112 tumour genomes: new candidates and distinguishing features. Sci. Rep. 7, 41544 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Lochovsky, L., Zhang, J., Fu, Y., Khurana, E. & Gerstein, M. LARVA: an integrative framework for large-scale analysis of recurrent variants in noncoding annotations. Nucleic Acids Res. 43, 8123–8134 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Lochovsky, L., Zhang, J. & Gerstein, M. MOAT: efficient detection of highly mutated regions with the mutations overburdening annotations tool. Bioinformatics 34, 1031–1033 (2018).

    Article  CAS  PubMed  Google Scholar 

  98. Soltis, A. R., Dalgard, C. L., Pollard, H. B. & Wilkerson, M. D. MutEnricher: a flexible toolset for somatic mutation enrichment analysis of tumor whole genomes. BMC Bioinformatics 21, 338 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  99. Juul, M. et al. ncdDetect2: improved models of the site-specific mutation rate in cancer and driver detection with robust significance evaluation. Bioinformatics 35, 189–199 (2019).

    Article  CAS  PubMed  Google Scholar 

  100. Mularoni, L., Sabarinathan, R., Deu-Pons, J., Gonzalez-Perez, A. & Lopez-Bigas, N. OncodriveFML: a general framework to identify coding and non-coding regions with cancer driver mutations. Genome Biol. 17, 128 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Umer, H. M. et al. A significant regulatory mutation burden at a high-affinity position of the CTCF motif in gastrointestinal cancers. Hum. Mutat. 37, 904–913 (2016).

    Article  CAS  PubMed  Google Scholar 

  102. Guilhamon, P. & Lupien, M. SMuRF: a novel tool to identify regulatory elements enriched for somatic point mutations. BMC Bioinformatics 19, 454 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Lawrence, M. S. et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 505, 495–501 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Hayflick, L. & Moorhead, P. S. The serial cultivation of human diploid cell strains. Exp. Cell Res. 25, 585–621 (1961).

    Article  CAS  PubMed  Google Scholar 

  105. Kim, N. W. et al. Specific association of human telomerase activity with immortal cells and cancer. Science 266, 2011–2015 (1994).

    Article  CAS  PubMed  Google Scholar 

  106. Meyerson, M. et al. hEST2, the putative human telomerase catalytic subunit gene, is up-regulated in tumor cells and during immortalization. Cell 90, 785–795 (1997).

    Article  CAS  PubMed  Google Scholar 

  107. Bouaoun, L. et al. TP53 variations in human cancers: new lessons from the IARC TP53 database and genomics data. Hum. Mutat. 37, 865–876 (2016).

    Article  CAS  PubMed  Google Scholar 

  108. Bell, R. J. et al. Understanding TERT promoter mutations: a common path to immortality. Mol. Cancer Res. 14, 315–323 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Killela, P. J. et al. TERT promoter mutations occur frequently in gliomas and a subset of tumors derived from cells with low rates of self-renewal. Proc. Natl Acad. Sci. USA 110, 6021–6026 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

E.L. is supported by grants from the Knut and Alice Wallenberg Foundation, the Swedish Medical Research Council and the Swedish Cancer Society.

Author information

Authors and Affiliations

Authors

Contributions

K.E. and E.L. both researched data for the article and made a substantial contribution to discussion of content, writing, reviewing and editing the article.

Corresponding author

Correspondence to Erik Larsson.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Cancer thanks J. Wong, A. Gonzalez-Perez and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

ActiveDriverWGS: https://cran.r-project.org/web/packages/ActiveDriverWGS/vignettes/ActiveDriverWGSR.html

CNCDriver: https://github.com/khuranalab/CNCDriver

COSMIC Cancer Gene Census: https://cancer.sanger.ac.uk/census

Driver Power: https://github.com/smshuai/DriverPower

ExinAtor: https://github.com/alanzos/ExInAtor/

fishHook: https://github.com/mskilab/fishHook

ICGC: https://dcc.icgc.org

LARVA: http://larva.gersteinlab.org/

MOAT: http://moat.gersteinlab.org/

MutEnricher: https://github.com/asoltis/MutEnricher

MutSigCV: https://software.broadinstitute.org/cancer/cga/mutsig

MutSpot: https://github.com/skandlab/MutSpot

ncdDetect2: http://moma.ki.au.dk/ncddetect/

ncDriver: http://moma.ki.au.dk/ncDriver/

OncodriveFML: http://bbglab.irbbarcelona.org/oncodrivefml/home

regDriver: https://github.com/husensofteng/regDriver

SMuRF: https://github.com/LupienLab/SMURF

TCGA: https://gdc.cancer.gov

UCSC Genome Browser: https://genome.ucsc.edu/

Glossary

Point mutations

Single-base changes in the genome.

Indels

Small insertions or deletions of DNA bases.

Enhancer

A distal regulatory region bound by transcription factors that aids recruitment of polymerases.

Passenger mutations

DNA alterations that do not provide a growth advantage.

Structural variants

Genomic alterations, such as gene duplications, translocations or inversions, affecting a large chromosomal segment, typically greater than 1 kb.

Neutral selection

Neither positive nor negative selection, that is, lacking influence on cell fitness.

Heterochromatin

DNA in a tightly packed and condensed form, marked by H3K9 methylation.

Cyclobutane pyrimidine dimer

The principal ultraviolet-induced DNA damage lesion, resulting from dimerization of neighbouring pyrimidines.

MicroRNA

A small regulatory RNA (around 22 bp) that can alter gene expression through mRNA silencing.

Topologically associating domain

A genomic segment showing a high degree of within-region physical interaction.

COSMIC Cancer Gene Census

A list of approximately 700 genes known to be mutated and have functional roles in cancer, compiled from decades of research.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Elliott, K., Larsson, E. Non-coding driver mutations in human cancer. Nat Rev Cancer 21, 500–509 (2021). https://doi.org/10.1038/s41568-021-00371-z

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-021-00371-z

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer