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:

Illuminating the noncoding genome in cancer

Abstract

Understanding the mechanisms underlying tumorigenesis requires comprehensive annotation of the cancer genome. The majority of the human genome consists of noncoding regions, harboring functional elements that regulate the expression of protein-coding genes, including proto-oncogenes or tumor-suppressor genes. Technologies such as whole-genome and long-read sequencing provide powerful means to identify alterations in the noncoding genome of cancer cells, paving the way for the study of their functional relevance. Recent analyses of noncoding alterations have revealed additional mechanisms underlying tumor development, progression and response to therapies, and uncovered new targets for drug development. Here we highlight recent findings about noncoding alterations, review established and emerging methods and models to study these alterations, and discuss the outlook for noncoding alterations as therapeutic targets.

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: Functional elements in the noncoding genome.
Fig. 2: Genomic alterations of functional noncoding elements in cancer.
Fig. 3: Methods to detect and study somatic alterations in the noncoding genome.
Fig. 4: Therapeutic implications of studying noncoding alterations in cancer genomes and epigenomes.

Similar content being viewed by others

References

  1. Boyle, A. P. et al. High-resolution mapping and characterization of open chromatin across the genome. Cell 132, 311–322 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Lieberman-Aiden, E. et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326, 289–293 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Rao, S. S. P. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  CAS  Google Scholar 

  6. Moore, J. E. et al. Expanded encyclopaedias of DNA elements in the human and mouse genomes. Nature 583, 699–710 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Kundaje, A. et al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Thurman, R. E. et al. The accessible chromatin landscape of the human genome. Nature 489, 75–82 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Djebali, S. et al. Landscape of transcription in human cells. Nature 489, 101–108 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bartel, D. P. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116, 281–297 (2004).

    Article  CAS  PubMed  Google Scholar 

  11. Rinn, J. L. et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 129, 1311–1323 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhao, J., Sun, B. K., Erwin, J. A., Song, J.-J. & Lee, J. T. Polycomb proteins targeted by a short repeat RNA to the mouse X chromosome. Science 322, 750–756 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Nora, E. P. et al. Spatial partitioning of the regulatory landscape of the X-inactivation centre. Nature 485, 381–385 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Dixon, J. R. et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 485, 376–380 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Dowen, J. M. et al. Control of cell identity genes occurs in insulated neighborhoods in mammalian chromosomes. Cell 159, 374–387 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Hnisz, D., Day, D. S. & Young, R. A. Insulated neighborhoods: structural and functional units of mammalian gene control. Cell 167, 1188–1200 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Bailey, S. D. et al. ZNF143 provides sequence specificity to secure chromatin interactions at gene promoters. Nat. Commun. 2, 6186 (2015).

    Article  PubMed  CAS  Google Scholar 

  18. Weintraub, A. S. et al. YY1 is a structural regulator of enhancer-promoter loops. Cell 171, 1573–1588.e28 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Gu, B. et al. Transcription-coupled changes in nuclear mobility of mammalian cis-regulatory elements. Science 359, 1050–1055 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Boija, A. et al. Transcription factors activate genes through the phase-separation capacity of their activation domains. Cell 175, 1842–1855.e16 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Hnisz, D. et al. Super-enhancers in the control of cell identity and disease. Cell 155, 934–947 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Lovén, J. et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153, 320–334 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Sabari, B. R. et al. Coactivator condensation at super-enhancers links phase separation and gene control. Science 361, eaar3958 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bailey, M. H. et al. Comprehensive characterization of cancer driver genes and mutations. Cell 173, 371–385.e18 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  Google Scholar 

  28. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. 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 

  30. Horn, S. et al. TERT promoter mutations in familial and sporadic melanoma. Science 339, 959–961 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. 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 

  32. Bell, R. J. A. 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 

  33. Rheinbay, E. et al. Analyses of non-coding somatic drivers in 2,658 cancer whole genomes. Nature 578, 102–111 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. 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 

  36. Mazrooei, P. et al. Cistrome partitioning reveals convergence of somatic mutations and risk variants on master transcription regulators in primary prostate tumors. cancer cell 36, 674–689.e6 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. 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 

  38. Cho, S. W. et al. Promoter of lncRNA gene PVT1 is a tumor-suppressor DNA boundary element. Cell 173, 1398–1412.e22 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Schaub, M. A., Boyle, A. P., Kundaje, A., Batzoglou, S. & Snyder, M. Linking disease associations with regulatory information in the human genome. Genome Res. 22, 1748–1759 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zhang, X., Bailey, S. D. & Lupien, M. Laying a solid foundation for Manhattan—‘setting the functional basis for the post-GWAS era’. Trends Genet. 30, 140–149 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Cowper-Sal-lari, R. et al. Breast cancer risk–associated SNPs modulate the affinity of chromatin for FOXA1 and alter gene expression. Nat. Genet. 44, 1191–1198 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Pomerantz, M. M. et al. The 8q24 cancer risk variant rs6983267 shows long-range interaction with MYC in colorectal cancer. Nat. Genet. 41, 882–884 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wright, J. B., Brown, S. J. & Cole, M. D. Upregulation of c-MYC in cis through a large chromatin loop linked to a cancer risk-associated single-nucleotide polymorphism in colorectal cancer cells. Mol. Cell. Biol. 30, 1411–1420 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Gao, P. et al. Biology and clinical implications of the 19q13 aggressive prostate cancer susceptibility locus. Cell 174, 576–589.e18 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hua, J. T. et al. Risk SNP-mediated promoter-enhancer switching drives prostate cancer through lncRNA PCAT19. Cell 174, 564–575.e18 (2018).

    Article  CAS  PubMed  Google Scholar 

  46. Zhang, X., Cowper-Sal lari, R., Bailey, S. D., Moore, J. H. & Lupien, M. Integrative functional genomics identifies an enhancer looping to the SOX9 gene disrupted by the 17q24.3 prostate cancer risk locus. Genome Res. 22, 1437–1446 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhang, X. et al. Identification of focally amplified lineage-specific super-enhancers in human epithelial cancers. Nat. Genet. 48, 176–182 (2016).

    Article  CAS  PubMed  Google Scholar 

  48. Shi, J. et al. Role of SWI/SNF in acute leukemia maintenance and enhancer-mediated Myc regulation. Genes Dev. 27, 2648–2662 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Viswanathan, S. R. et al. Structural alterations driving castration-resistant prostate cancer revealed by linked-read genome sequencing. Cell 174, 433–447.e19 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Herranz, D. et al. A NOTCH1-driven MYC enhancer promotes T cell development, transformation and acute lymphoblastic leukemia. Nat. Med. 20, 1130–1137 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhang, X. et al. Somatic superenhancer duplications and hotspot mutations lead to oncogenic activation of the KLF5 transcription factor. Cancer Discov. 8, 108–125 (2018).

    Article  CAS  PubMed  Google Scholar 

  52. Takeda, D. Y. et al. A somatically acquired enhancer of the androgen receptor is a noncoding driver in advanced prostate cancer. Cell 174, 422–432.e13 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Quigley, D. A. et al. Genomic hallmarks and structural variation in metastatic prostate cancer. cell 174, 758–769.e9 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Morton, A. R. et al. Functional enhancers shape extrachromosomal oncogene amplifications. Cell 179, 1330–1341.e13 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Wu, S. et al. Circular ecDNA promotes accessible chromatin and high oncogene expression. Nature 575, 699–703 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Battey, J. et al. The human c-myc oncogene: structural consequences of translocation into the IgH locus in Burkitt lymphoma. Cell 34, 779–787 (1983).

    Article  CAS  PubMed  Google Scholar 

  57. Shima, E. A. et al. Gene encoding the alpha chain of the T-cell receptor is moved immediately downstream of c-myc in a chromosomal 8;14 translocation in a cell line from a human T-cell leukemia. Proc. Natl. Acad. Sci. USA 83, 3439–3443 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Tomlins, S. A. et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310, 644–648 (2005).

    Article  CAS  PubMed  Google Scholar 

  59. Kron, K. J. et al. TMPRSS2-ERG fusion co-opts master transcription factors and activates NOTCH signaling in primary prostate cancer. Nat. Genet. 49, 1336–1345 (2017).

    Article  CAS  PubMed  Google Scholar 

  60. Bandopadhayay, P. et al. MYB-QKI rearrangements in angiocentric glioma drive tumorigenicity through a tripartite mechanism. Nat. Genet. 48, 273–282 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Drier, Y. et al. An oncogenic MYB feedback loop drives alternate cell fates in adenoid cystic carcinoma. Nat. Genet. 48, 265–272 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Gröschel, S. et al. A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in leukemia. Cell 157, 369–381 (2014).

    Article  PubMed  CAS  Google Scholar 

  63. Northcott, P. A. et al. Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma. Nature 511, 428–434 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Peifer, M. et al. Telomerase activation by genomic rearrangements in high-risk neuroblastoma. Nature 526, 700–704 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. 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 

  66. Hnisz, D. et al. Activation of proto-oncogenes by disruption of chromosome neighborhoods. Science 351, 1454–1458 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  68. 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 

  69. Weischenfeldt, J. et al. Pan-cancer analysis of somatic copy-number alterations implicates IRS4 and IGF2 in enhancer hijacking. Nat. Genet. 49, 65–74 (2017).

    Article  CAS  PubMed  Google Scholar 

  70. Iwai, Y. et al. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc. Natl. Acad. Sci. USA 99, 12293–12297 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Brahmer, J. R. et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 366, 2455–2465 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Topalian, S. L. et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Kataoka, K. et al. Aberrant PD-L1 expression through 3′-UTR disruption in multiple cancers. Nature 534, 402–406 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  75. Suzuki, H. et al. Recurrent noncoding U1 snRNA mutations drive cryptic splicing in SHH medulloblastoma. Nature 574, 707–711 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Kandoth, C. et al. Mutational landscape and significance across 12 major cancer types. Nature 502, 333–339 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. 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 

  78. Vogelstein, B. et al. Cancer genome landscapes. Science 339, 1546–1558 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Zack, T. I. et al. Pan-cancer patterns of somatic copy number alteration. Nat. Genet. 45, 1134–1140 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Adams, E. J. et al. FOXA1 mutations alter pioneering activity, differentiation and prostate cancer phenotypes. Nature 571, 408–412 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Parolia, A. et al. Distinct structural classes of activating FOXA1 alterations in advanced prostate cancer. Nature 571, 413–418 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Nabet, B. et al. Deregulation of the Ras-Erk signaling axis modulates the enhancer landscape. Cell Reports 12, 1300–1313 (2015).

    Article  CAS  PubMed  Google Scholar 

  83. Liu, F. et al. EGFR Mutation promotes glioblastoma through epigenome and transcription factor network remodeling. Mol. Cell 60, 307–318 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Supek, F., Miñana, B., Valcárcel, J., Gabaldón, T. & Lehner, B. Synonymous mutations frequently act as driver mutations in human cancers. Cell 156, 1324–1335 (2014).

    Article  CAS  PubMed  Google Scholar 

  85. Flavahan, W. A. et al. Insulator dysfunction and oncogene activation in IDH mutant gliomas. Nature 529, 110–114 (2016).

    Article  CAS  PubMed  Google Scholar 

  86. Flavahan, W. A. et al. Altered chromosomal topology drives oncogenic programs in SDH-deficient GISTs. Nature 575, 229–233 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Stephens, P. J. et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 144, 27–40 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Baca, S. C. et al. Punctuated evolution of prostate cancer genomes. Cell 153, 666–677 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Cortés-Ciriano, I. et al. Comprehensive analysis of chromothripsis in 2,658 human cancers using whole-genome sequencing. Nat. Genet. 52, 331–341 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Eid, J. et al. Real-time DNA sequencing from single polymerase molecules. Science 323, 133–138 (2009).

    Article  CAS  PubMed  Google Scholar 

  91. Derrington, I. M. et al. Nanopore DNA sequencing with MspA. Proc. Natl. Acad. Sci. USA 107, 16060–16065 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Zheng, G. X. Y. et al. Haplotyping germline and cancer genomes with high-throughput linked-read sequencing. Nat. Biotechnol. 34, 303–311 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Tang, A. D. et al. Full-length transcript characterization of SF3B1 mutation in chronic lymphocytic leukemia reveals downregulation of retained introns. Nat. Commun. 11, 1438 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Abraham, B. J. et al. Small genomic insertions form enhancers that misregulate oncogenes. Nat. Commun. 8, 14385 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Corces, M. R. et al. The chromatin accessibility landscape of primary human cancers. Science 362, eaav1898 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Burton, J. N. et al. Chromosome-scale scaffolding of de novo genome assemblies based on chromatin interactions. Nat. Biotechnol. 31, 1119–1125 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Harewood, L. et al. Hi-C as a tool for precise detection and characterisation of chromosomal rearrangements and copy number variation in human tumours. Genome Biol. 18, 125 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Dixon, J. R. et al. Integrative detection and analysis of structural variation in cancer genomes. Nat. Genet. 50, 1388–1398 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Tourdot, R.W. & Zhang, C.-Z. Determination of complete chromosomal haplotypes by bulk DNA sequencing. Preprint at bioRxiv https://doi.org/10.1101/629337 (2020).

  100. Nagano, T. et al. Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature 502, 59–64 (2013).

    Article  CAS  PubMed  Google Scholar 

  101. Ramani, V. et al. Massively multiplex single-cell Hi-C. Nat. Methods 14, 263–266 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Gaj, T., Gersbach, C. A. & Barbas, C. F. III. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 31, 397–405 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Pulecio, J., Verma, N., Mejía-Ramírez, E., Huangfu, D. & Raya, A. CRISPR/Cas9-Based Engineering of the Epigenome. Cell Stem Cell 21, 431–447 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Thakore, P. I. et al. Highly specific epigenome editing by CRISPR-Cas9 repressors for silencing of distal regulatory elements. Nat. Methods 12, 1143–1149 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Qi, L. S. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Gilbert, L. A. et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 442–451 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Akıncılar, S. C. et al. Long-range chromatin interactions drive mutant TERT Promoter activation. Cancer Discov. 6, 1276–1291 (2016).

    Article  PubMed  Google Scholar 

  108. Choi, P. S. & Meyerson, M. Targeted genomic rearrangements using CRISPR/Cas technology. Nat. Commun. 5, 3728 (2014).

    Article  CAS  PubMed  Google Scholar 

  109. Joung, J. et al. Genome-scale activation screen identifies a lncRNA locus regulating a gene neighbourhood. Nature 548, 343–346 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Liu, S. J. et al. CRISPRi-based genome-scale identification of functional long noncoding RNA loci in human cells. Science 355, aah7111 (2017).

    Article  PubMed  CAS  Google Scholar 

  111. Sur, I. K. et al. Mice lacking a Myc enhancer that includes human SNP rs6983267 are resistant to intestinal tumors. Science 338, 1360–1363 (2012).

    Article  CAS  PubMed  Google Scholar 

  112. Ulitsky, I., Shkumatava, A., Jan, C. H., Sive, H. & Bartel, D. P. Conserved function of lincRNAs in vertebrate embryonic development despite rapid sequence evolution. Cell 147, 1537–1550 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Yue, F. et al. A comparative encyclopedia of DNA elements in the mouse genome. Nature 515, 355–364 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Tuveson, D. & Clevers, H. Cancer modeling meets human organoid technology. Science 364, 952–955 (2019).

    Article  CAS  PubMed  Google Scholar 

  115. Sato, T. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009).

    Article  CAS  PubMed  Google Scholar 

  116. Boj, S. F. et al. Organoid models of human and mouse ductal pancreatic cancer. Cell 160, 324–338 (2015).

    Article  CAS  PubMed  Google Scholar 

  117. Gao, D. et al. Organoid cultures derived from patients with advanced prostate cancer. Cell 159, 176–187 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Artegiani, B. et al. Fast and efficient generation of knock-in human organoids using homology-independent CRISPR-Cas9 precision genome editing. Nat. Cell Biol. 22, 321–331 (2020).

    Article  CAS  PubMed  Google Scholar 

  119. Neal, J. T. et al. Organoid modeling of the tumor immune microenvironment. Cell 175, 1972–1988.e16 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Ben-David, U. et al. Genetic and transcriptional evolution alters cancer cell line drug response. Nature 560, 325–330 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Barretina, J. et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483, 603–607 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Ghandi, M. et al. Next-generation characterization of the Cancer Cell Line Encyclopedia. Nature 569, 503–508 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Garnett, M. J. et al. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature 483, 570–575 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Gabay, M., Li, Y. & Felsher, D. W. MYC activation is a hallmark of cancer initiation and maintenance. Cold Spring Harb. Perspect. Med. 4, a014241 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Shankar, G. M. et al. Rapid intraoperative molecular characterization of glioma. JAMA Oncol. 1, 662–667 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  126. Shen, S. Y. et al. Sensitive tumour detection and classification using plasma cell-free DNA methylomes. Nature 563, 579–583 (2018).

    Article  CAS  PubMed  Google Scholar 

  127. Snyder, M. W., Kircher, M., Hill, A. J., Daza, R. M. & Shendure, J. Cell-free DNA comprises an in vivo nucleosome footprint that informs its tissues-of-origin. Cell 164, 57–68 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Rathert, P. et al. Transcriptional plasticity promotes primary and acquired resistance to BET inhibition. Nature 525, 543–547 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Kwiatkowski, N. et al. Targeting transcription regulation in cancer with a covalent CDK7 inhibitor. Nature 511, 616–620 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Bradner, J. E., Hnisz, D. & Young, R. A. Transcriptional addiction in cancer. Cell 168, 629–643 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Knutson, S. K. et al. A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells. Nat. Chem. Biol. 8, 890–896 (2012).

    Article  CAS  PubMed  Google Scholar 

  132. Sakamoto, K. M. et al. Protacs: chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation. Proc. Natl. Acad. Sci. USA 98, 8554–8559 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Winter, G. E. et al. Selective Target protein degradation via phthalimide conjugation. Science 348, 1376–1381 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Rees, H. A. & Liu, D. R. Base editing: precision chemistry on the genome and transcriptome of living cells. Nat. Rev. Genet. 19, 770–788 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Li, X. et al. Programmable base editing of mutated TERT promoter inhibits brain tumour growth. Nat. Cell Biol. 22, 282–288 (2020).

    Article  CAS  PubMed  Google Scholar 

  136. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. 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 

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

    Article  CAS  PubMed  Google Scholar 

  140. 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 

  141. Imielinski, M., Guo, G. & Meyerson, M. Insertions and deletions target lineage-defining genes in human cancers. Cell 168, 460–472.e14 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Durkin, S. G. & Glover, T. W. Chromosome fragile sites. Annu. Rev. Genet. 41, 169–192 (2007).

    Article  CAS  PubMed  Google Scholar 

  143. Glover, T. W., Wilson, T. E. & Arlt, M. F. Fragile sites in cancer: more than meets the eye. Nat. Rev. Cancer 17, 489–501 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. George, J. et al. Comprehensive genomic profiles of small cell lung cancer. Nature 524, 47–53 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Huyghe, J. R. et al. Discovery of common and rare genetic risk variants for colorectal cancer. Nat. Genet. 51, 76–87 (2019).

    Article  CAS  PubMed  Google Scholar 

  146. Kilpivaara, O. et al. A germline JAK2 SNP is associated with predisposition to the development of JAK2V617F-positive myeloproliferative neoplasms. Nat. Genet. 41, 455–459 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Carter, H. et al. Interaction landscape of inherited polymorphisms with somatic events in cancer. Cancer Discov. 7, 410–423 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Flavahan, W. A., Gaskell, E. & Bernstein, B. E. Epigenetic plasticity and the hallmarks of cancer. Science 357, eaal2380 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  149. Mack, S. C. et al. Epigenomic alterations define lethal CIMP-positive ependymomas of infancy. Nature 506, 445–450 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Priestley, P. et al. Pan-cancer whole-genome analyses of metastatic solid tumours. Nature 575, 210–216 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank G. Ha from the Fred Hutchinson Cancer Research Center for discussions. We acknowledge support from the US National Cancer Institute: R35CA197568 (M.M.) and R00CA215244 (X.Z.).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Matthew Meyerson.

Ethics declarations

Competing interests

M.M. receives commercial research grants from Bayer, Novo Ventures, Ono and Janssen; receives royalties from LabCorp; and serves as a consultant and advisory board member for OrigiMed.

Additional information

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, X., Meyerson, M. Illuminating the noncoding genome in cancer. Nat Cancer 1, 864–872 (2020). https://doi.org/10.1038/s43018-020-00114-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-020-00114-3

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