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:

Clonal expansion in non-cancer tissues

Abstract

Cancer is a clonal disorder derived from a single ancestor cell and its progenies that are positively selected by acquisition of ‘driver mutations’. However, the evolution of positively selected clones does not necessarily imply the presence of cancer. On the contrary, it has become clear that expansion of these clones in phenotypically normal or non-cancer tissues is commonly seen in association with ageing and/or in response to environmental insults and chronic inflammation. Recent studies have reported expansion of clones harbouring mutations in cancer driver genes in the blood, skin, oesophagus, bronchus, liver, endometrium and bladder, where the expansion could be so extensive that tissues undergo remodelling of an almost entire tissue. The presence of common cancer driver mutations in normal tissues suggests a strong link to cancer development, providing an opportunity to understand early carcinogenic processes. Nevertheless, some driver mutations are unique to normal tissues or have a mutation frequency that is much higher in normal tissue than in cancer, indicating that the respective clones may not necessarily be destined for evolution to cancer but even negatively selected for carcinogenesis depending on the mutated gene. Moreover, tissues that are remodelled by genetically altered clones might define functionalities of aged tissues or modified inflammatory processes. In this Review, we provide an overview of major findings on clonal expansion in phenotypically normal or non-cancer tissues and discuss their biological significance not only in cancer development but also in ageing and inflammatory diseases.

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: Clonal evolution in normal tissues.
Fig. 2: Clonal expansion in normal tissues in various organs.
Fig. 3: Biological significance of positively selected clones in normal tissues.

Similar content being viewed by others

References

  1. Greenman, C. et al. Patterns of somatic mutation in human cancer genomes. Nature 446, 153–158 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Greaves, M. & Maley, C. C. Clonal evolution in cancer. Nature 481, 306–313 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. McGranahan, N. & Swanton, C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell 168, 613–628 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. 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 

  6. 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 

  7. 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 

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

    Article  CAS  Google Scholar 

  9. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Alexandrov, L. B. et al. The repertoire of mutational signatures in human cancer. Nature 578, 94–101 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Burrell, R. A., McGranahan, N., Bartek, J. & Swanton, C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature 501, 338–345 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Yachida, S. et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 467, 1114–1117 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Turajlic, S. et al. Tracking cancer evolution reveals constrained routes to metastases: TRACERx renal. Cell 173, 581–594.e12 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mitchell, T. J. et al. Timing the landmark events in the evolution of clear cell renal cell cancer: TRACERx renal. Cell 173, 611–623.e17 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Suzuki, H. et al. Mutational landscape and clonal architecture in grade II and III gliomas. Nat. Genet. 47, 458–468 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. Mazor, T. et al. DNA methylation and somatic mutations converge on the cell cycle and define similar evolutionary histories in brain tumors. Cancer Cell 28, 307–317 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Nikbakht, H. et al. Spatial and temporal homogeneity of driver mutations in diffuse intrinsic pontine glioma. Nat. Commun. 7, 11185 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhang, J. et al. Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing. Science 346, 256–259 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. de Bruin, E. C. et al. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science 346, 251–256 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Jamal-Hanjani, M. et al. Tracking the evolution of non-small-cell lung cancer. N. Engl. J. Med. 376, 2109–2121 (2017).

    Article  CAS  PubMed  Google Scholar 

  22. Sottoriva, A. et al. A Big Bang model of human colorectal tumor growth. Nat. Genet. 47, 209–216 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Uchi, R. et al. Integrated multiregional analysis proposing a new model of colorectal cancer evolution. PLoS Genet. 12, e1005778 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Roerink, S. F. et al. Intra-tumour diversification in colorectal cancer at the single-cell level. Nature 556, 457–462 (2018).

    Article  CAS  PubMed  Google Scholar 

  25. Saito, T. et al. A temporal shift of the evolutionary principle shaping intratumor heterogeneity in colorectal cancer. Nat. Commun. 9, 2884 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Gundem, G. et al. The evolutionary history of lethal metastatic prostate cancer. Nature 520, 353–357 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Cooper, C. S. et al. Analysis of the genetic phylogeny of multifocal prostate cancer identifies multiple independent clonal expansions in neoplastic and morphologically normal prostate tissue. Nat. Genet. 47, 367–372 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Shain, A. H. et al. The genetic evolution of melanoma from precursor lesions. N. Engl. J. Med. 373, 1926–1936 (2015).

    Article  PubMed  CAS  Google Scholar 

  29. Harbst, K. et al. Multiregion whole-exome sequencing uncovers the genetic evolution and mutational heterogeneity of early-stage metastatic melanoma. Cancer Res. 76, 4765–4774 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Bashashati, A. et al. Distinct evolutionary trajectories of primary high-grade serous ovarian cancers revealed through spatial mutational profiling. J. Pathol. 231, 21–34 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Eckert, M. A. et al. Genomics of ovarian cancer progression reveals diverse metastatic trajectories including intraepithelial metastasis to the fallopian tube. Cancer Discov. 6, 1342–1351 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hao, J. J. et al. Spatial intratumoral heterogeneity and temporal clonal evolution in esophageal squamous cell carcinoma. Nat. Genet. 48, 1500–1507 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Murugaesu, N. et al. Tracking the genomic evolution of esophageal adenocarcinoma through neoadjuvant chemotherapy. Cancer Discov. 5, 821–831 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Bolli, N. et al. Heterogeneity of genomic evolution and mutational profiles in multiple myeloma. Nat. Commun. 5, 2997 (2014).

    Article  PubMed  CAS  Google Scholar 

  35. Yates, L. R. et al. Subclonal diversification of primary breast cancer revealed by multiregion sequencing. Nat. Med. 21, 751–759 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Xue, R. et al. Variable intra-tumor genomic heterogeneity of multiple lesions in patients with hepatocellular carcinoma. Gastroenterology 150, 998–1008 (2016).

    Article  PubMed  Google Scholar 

  37. Genovese, G. et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 371, 2477–2487 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Jaiswal, S. et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 371, 2488–2498 (2014). Together with Genovese et al. (2014), this landmark study describes age-related clonal haematopoiesis having common mutations in myeloid neoplasms, using unbiased sequencing and its relationship not only to haematological malignancy but also to cardiovascular diseases.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Yoshizato, T. et al. Somatic mutations and clonal hematopoiesis in aplastic anemia. N. Engl. J. Med. 373, 35–47 (2015). This study describes clonal haematopoiesis in patients with aplastic anaemia and its link to the clinical leukaemia risk and response to immunosuppressive therapy.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Martincorena, I. et al. High burden and pervasive positive selection of somatic mutations in normal human skin. Science 348, 880–886 (2015). This study shows clonal expansion in sun-exposed skin by targeted sequencing of common drivers in skin cancers, and is the first study demonstrating clonal expansion in normal solid tissues.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Ross-Innes, C. S. et al. Whole-genome sequencing provides new insights into the clonal architecture of Barrett’s esophagus and esophageal adenocarcinoma. Nat. Genet. 47, 1038–1046 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Stachler, M. D. et al. Paired exome analysis of Barrett’s esophagus and adenocarcinoma. Nat. Genet. 47, 1047–1055 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Huang, K. K. et al. Genomic and epigenomic profiling of high-risk intestinal metaplasia reveals molecular determinants of progression to gastric cancer. Cancer Cell 33, 137–150.e5 (2018).

    Article  CAS  PubMed  Google Scholar 

  44. Martincorena, I. et al. Somatic mutant clones colonize the human esophagus with age. Science 362, 911–917 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Yokoyama, A. et al. Age-related remodelling of oesophageal epithelia by mutated cancer drivers. Nature 565, 312–317 (2019). This study shows extensive remodelling of the oesophageal epithelium with ageing, which is substantially accelerated by heavy smoking and alcohol drinking, and comprehensively describes driver mutations and their history.

    Article  CAS  PubMed  Google Scholar 

  46. Kim, S. K. et al. Comprehensive analysis of genetic aberrations linked to tumorigenesis in regenerative nodules of liver cirrhosis. J. Gastroenterol. 54, 628–640 (2019).

    Article  CAS  PubMed  Google Scholar 

  47. Brunner, S. F. et al. Somatic mutations and clonal dynamics in healthy and cirrhotic human liver. Nature 574, 538–542 (2019). This study shows that clonal expansion in cirrhotic liver does not necessarily accompany driver mutations.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Kakiuchi, N. et al. Frequent mutations that converge on the NFKBIZ pathway in ulcerative colitis. Nature 577, 260–265 (2020). This study describes crypt expansion throughout life in the healthy colon, positive selection in ulcerative colitis and negative selection of positively selected clones during carcinogenesis.

    Article  CAS  PubMed  Google Scholar 

  49. Nanki, K. et al. Somatic inflammatory gene mutations in human ulcerative colitis epithelium. Nature 577, 254–259 (2020). This study describes positive selection of clones in ulcerative colitis.

    Article  CAS  PubMed  Google Scholar 

  50. Moore, L. et al. The mutational landscape of normal human endometrial epithelium. Nature 580, 640–646 (2020). This study describes positive selection in endometrial glands in which the majority of glands acquire driver mutations in older people.

    Article  CAS  PubMed  Google Scholar 

  51. Yoshida, K. et al. Tobacco smoking and somatic mutations in human bronchial epithelium. Nature 578, 266–272 (2020). This study describes a mutational landscape of the bronchial epithelium according to past and current history of tobacco smoking. Once remodelled by driver-mutated clones, bronchial epithelium clones can be replaced by mitotically quiescent cells after smoking cessation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Fearon, E. R., Burke, P. J., Schiffer, C. A., Zehnbauer, B. A. & Vogelstein, B. Differentiation of leukemia cells to polymorphonuclear leukocytes in patients with acute nonlymphocytic leukemia. N. Engl. J. Med. 315, 15–24 (1986).

    Article  CAS  PubMed  Google Scholar 

  53. Fialkow, P. J. et al. Clonal development, stem-cell differentiation, and clinical remissions in acute nonlymphocytic leukemia. N. Engl. J. Med. 317, 468–473 (1987).

    Article  CAS  PubMed  Google Scholar 

  54. Fey, M. F. et al. Clonality and X-inactivation patterns in hematopoietic cell populations detected by the highly informative M27 β DNA probe. Blood 83, 931–938 (1994).

    Article  CAS  PubMed  Google Scholar 

  55. Busque, L. et al. Nonrandom X-inactivation patterns in normal females: lyonization ratios vary with age. Blood 88, 59–65 (1996).

    Article  CAS  PubMed  Google Scholar 

  56. van Kamp, H., Landegent, J. E., Jansen, R. P., Willemze, R. & Fibbe, W. E. Clonal hematopoiesis in patients with acquired aplastic anemia. Blood 78, 3209–3214 (1991).

    Article  PubMed  Google Scholar 

  57. Taylor, R. W. et al. Mitochondrial DNA mutations in human colonic crypt stem cells. J. Clin. Invest. 112, 1351–1360 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Greaves, L. C. et al. Mitochondrial DNA mutations are established in human colonic stem cells, and mutated clones expand by crypt fission. Proc. Natl Acad. Sci. USA 103, 714–719 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Baker, A. M. et al. Quantification of crypt and stem cell evolution in the normal and neoplastic human colon. Cell Rep. 8, 940–947 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. McDonald, S. A. et al. Mechanisms of field cancerization in the human stomach: the expansion and spread of mutated gastric stem cells. Gastroenterology 134, 500–510 (2008).

    Article  CAS  PubMed  Google Scholar 

  61. Gutierrez-Gonzalez, L. et al. The clonal origins of dysplasia from intestinal metaplasia in the human stomach. Gastroenterology 140, 1251–1260.e1–6 (2011).

    Article  CAS  PubMed  Google Scholar 

  62. Gutierrez-Gonzalez, L. et al. Analysis of the clonal architecture of the human small intestinal epithelium establishes a common stem cell for all lineages and reveals a mechanism for the fixation and spread of mutations. J. Pathol. 217, 489–496 (2009).

    Article  CAS  PubMed  Google Scholar 

  63. Fellous, T. G. et al. A methodological approach to tracing cell lineage in human epithelial tissues. Stem Cell 27, 1410–1420 (2009).

    Article  CAS  Google Scholar 

  64. Lin, W. R. et al. The histogenesis of regenerative nodules in human liver cirrhosis. Hepatology 51, 1017–1026 (2010).

    Article  CAS  PubMed  Google Scholar 

  65. Cereser, B. et al. Analysis of clonal expansions through the normal and premalignant human breast epithelium reveals the presence of luminal stem cells. J. Pathol. 244, 61–70 (2018).

    Article  CAS  PubMed  Google Scholar 

  66. Gaisa, N. T. et al. The human urothelium consists of multiple clonal units, each maintained by a stem cell. J. Pathol. 225, 163–171 (2011).

    Article  CAS  PubMed  Google Scholar 

  67. Gaisa, N. T. et al. Clonal architecture of human prostatic epithelium in benign and malignant conditions. J. Pathol. 225, 172–180 (2011).

    Article  CAS  PubMed  Google Scholar 

  68. Moad, M. et al. Multipotent basal stem cells, maintained in localized proximal niches, support directed long-ranging epithelial flows in human prostates. Cell Rep. 20, 1609–1622 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Mitani, K. et al. Somatic mosaicism in chronic myeloid leukemia in remission. Blood 128, 2863–2866 (2016).

    Article  CAS  PubMed  Google Scholar 

  70. Ziegler, A. et al. Sunburn and p53 in the onset of skin cancer. Nature 372, 773–776 (1994).

    Article  CAS  PubMed  Google Scholar 

  71. Nakazawa, H. et al. UV and skin cancer: specific p53 gene mutation in normal skin as a biologically relevant exposure measurement. Proc. Natl Acad. Sci. USA 91, 360–364 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Jonason, A. S. et al. Frequent clones of p53-mutated keratinocytes in normal human skin. Proc. Natl Acad. Sci. USA 93, 14025–14029 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Ling, G. et al. Persistent p53 mutations in single cells from normal human skin. Am. J. Pathol. 159, 1247–1253 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Tian, D. et al. Multifocal accumulation of p53 protein in esophageal carcinoma: evidence for field cancerization. Int. J. Cancer 78, 568–575 (1998).

    Article  CAS  PubMed  Google Scholar 

  75. Katada, C. et al. Alcohol consumption and multiple dysplastic lesions increase risk of squamous cell carcinoma in the esophagus, head, and neck. Gastroenterology 151, 860–869.e7 (2016).

    Article  CAS  PubMed  Google Scholar 

  76. Maley, C. C. et al. The combination of genetic instability and clonal expansion predicts progression to esophageal adenocarcinoma. Cancer Res. 64, 7629–7633 (2004).

    Article  CAS  PubMed  Google Scholar 

  77. Van der Vorst, S., Dekairelle, A. F., Weynand, B., Hamoir, M. & Gala, J. L. Assessment of p53 functional activity in tumor cells and histologically normal mucosa from patients with head and neck squamous cell carcinoma. Head Neck 34, 1542–1550 (2012).

    Article  PubMed  Google Scholar 

  78. Franklin, W. A. et al. Widely dispersed p53 mutation in respiratory epithelium. A novel mechanism for field carcinogenesis. J. Clin. Invest. 100, 2133–2137 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Leedham, S. J. et al. Clonality, founder mutations, and field cancerization in human ulcerative colitis-associated neoplasia. Gastroenterology 136, 542–550.e6 (2009).

    Article  PubMed  Google Scholar 

  80. Galandiuk, S. et al. Field cancerization in the intestinal epithelium of patients with Crohn’s ileocolitis. Gastroenterology 142, 855–864.e8 (2012).

    Article  PubMed  Google Scholar 

  81. Shimizu, T. et al. Accumulation of somatic mutations in TP53 in gastric epithelium with Helicobacter pylori infection. Gastroenterology 147, 407–417.e3 (2014).

    Article  CAS  PubMed  Google Scholar 

  82. Metzker, M. L. Sequencing technologies — the next generation. Nat. Rev. Genet. 11, 31–46 (2010).

    Article  CAS  PubMed  Google Scholar 

  83. Meyerson, M., Gabriel, S. & Getz, G. Advances in understanding cancer genomes through second-generation sequencing. Nat. Rev. Genet. 11, 685–696 (2010).

    Article  CAS  PubMed  Google Scholar 

  84. Chatterjee, N. & Walker, G. C. Mechanisms of DNA damage, repair, and mutagenesis. Env. Mol. Mutagen. 58, 235–263 (2017).

    Article  CAS  Google Scholar 

  85. Welch, J. S. et al. The origin and evolution of mutations in acute myeloid leukemia. Cell 150, 264–278 (2012). This study shows for the first time that normal blood progenitor cells accumulate somatic mutations in an age-dependent manner.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Lodato, M. A. et al. Aging and neurodegeneration are associated with increased mutations in single human neurons. Science 359, 555–559 (2018).

    Article  CAS  PubMed  Google Scholar 

  87. Lee-Six, H. et al. The landscape of somatic mutation in normal colorectal epithelial cells. Nature 574, 532–537 (2019). This study described the mutational signatures and landscape in colon crypts.

    Article  CAS  PubMed  Google Scholar 

  88. Franco, I. et al. Somatic mutagenesis in satellite cells associates with human skeletal muscle aging. Nat. Commun. 9, 800 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Blokzijl, F. et al. Tissue-specific mutation accumulation in human adult stem cells during life. Nature 538, 260–264 (2016). This study measures the mutational load in normal cells from small intestine, colon and liver cells, using organoid culture.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Osorio, F. G. et al. Somatic mutations reveal lineage relationships and age-related mutagenesis in human hematopoiesis. Cell Rep. 25, 2308–2316.e4 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Kuijk, E. et al. Early divergence of mutational processes in human fetal tissues. Sci. Adv. 5, eaaw1271 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Hasaart, K. A. L. et al. Mutation accumulation and developmental lineages in normal and Down syndrome human fetal haematopoiesis. Sci. Rep. 10, 12991 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Pfeifer, G. P. Mutagenesis at methylated CpG sequences. Curr. Top. Microbiol. Immunol. 301, 259–281 (2006).

    CAS  PubMed  Google Scholar 

  94. Harris, R. S., Petersen-Mahrt, S. K. & Neuberger, M. S. RNA editing enzyme APOBEC1 and some of its homologs can act as DNA mutators. Mol. Cell 10, 1247–1253 (2002).

    Article  CAS  PubMed  Google Scholar 

  95. Taylor, B. J. et al. DNA deaminases induce break-associated mutation showers with implication of APOBEC3B and 3A in breast cancer kataegis. eLife 2, e00534 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  96. Burns, M. B. et al. APOBEC3B is an enzymatic source of mutation in breast cancer. Nature 494, 366–370 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Pfeifer, G. P., You, Y. H. & Besaratinia, A. Mutations induced by ultraviolet light. Mutat. Res. 571, 19–31 (2005).

    Article  CAS  PubMed  Google Scholar 

  98. Pfeifer, G. P. et al. Tobacco smoke carcinogens, DNA damage and p53 mutations in smoking-associated cancers. Oncogene 21, 7435–7451 (2002).

    Article  CAS  PubMed  Google Scholar 

  99. Chang, J. et al. Genomic analysis of oesophageal squamous-cell carcinoma identifies alcohol drinking-related mutation signature and genomic alterations. Nat. Commun. 8, 15290 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Tomita-Mitchell, A. et al. Mismatch repair deficient human cells: spontaneous and MNNG-induced mutational spectra in the HPRT gene. Mutat. Res. 450, 125–138 (2000).

    Article  CAS  PubMed  Google Scholar 

  101. Poon, S. L. et al. Genome-wide mutational signatures of aristolochic acid and its application as a screening tool. Sci. Transl. Med. 5, 197ra101 (2013).

    Article  PubMed  CAS  Google Scholar 

  102. Letouze, E. et al. Mutational signatures reveal the dynamic interplay of risk factors and cellular processes during liver tumorigenesis. Nat. Commun. 8, 1315 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Maura, F. et al. A practical guide for mutational signature analysis in hematological malignancies. Nat. Commun. 10, 2969 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Pleguezuelos-Manzano, C. et al. Mutational signature in colorectal cancer caused by genotoxic pks+ E. coli. Nature 580, 269–273 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Lawson, A. R. J. et al. Extensive heterogeneity in somatic mutation and selection in the human bladder. Science 370, 75–82 (2020).

    Article  CAS  PubMed  Google Scholar 

  106. Li, Y. et al. Patterns of somatic structural variation in human cancer genomes. Nature 578, 112–121 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Busque, L. et al. Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis. Nat. Genet. 44, 1179–1181 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Xie, M. et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat. Med. 20, 1472–1478 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. McKerrell, T. et al. Leukemia-associated somatic mutations drive distinct patterns of age-related clonal hemopoiesis. Cell Rep. 10, 1239–1245 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Young, A. L., Challen, G. A., Birmann, B. M. & Druley, T. E. Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults. Nat. Commun. 7, 12484 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Papaemmanuil, E. et al. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood 122, 3616–3627 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Haferlach, T. et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia 28, 241–247 (2014).

    Article  CAS  PubMed  Google Scholar 

  113. Papaemmanuil, E. et al. Genomic classification and prognosis in acute myeloid leukemia. N. Engl. J. Med. 374, 2209–2221 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Grinfeld, J. et al. Classification and personalized prognosis in myeloproliferative neoplasms. N. Engl. J. Med. 379, 1416–1430 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Tyner, J. W. et al. Functional genomic landscape of acute myeloid leukaemia. Nature 562, 526–531 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Laurie, C. C. et al. Detectable clonal mosaicism from birth to old age and its relationship to cancer. Nat. Genet. 44, 642–650 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Jacobs, K. B. et al. Detectable clonal mosaicism and its relationship to aging and cancer. Nat. Genet. 44, 651–658 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Loh, P. R. et al. Insights into clonal haematopoiesis from 8,342 mosaic chromosomal alterations. Nature 559, 350–355 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Suda, K. et al. Clonal expansion and diversification of cancer-associated mutations in endometriosis and normal endometrium. Cell Rep. 24, 1777–1789 (2018). This study shows that endometriosis may derive from endometrial glands that acquire driver mutations by retrograde menstruation.

    Article  CAS  PubMed  Google Scholar 

  120. Li, R. et al. Macroscopic somatic clonal expansion in morphologically normal human urothelium. Science 370, 82–89 (2020).

    Article  CAS  PubMed  Google Scholar 

  121. Olafsson, S. et al. Somatic evolution in non-neoplastic IBD-affected colon. Cell 182, 672–684.e11 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Zhu, M. et al. Somatic mutations increase hepatic clonal fitness and regeneration in chronic liver disease. Cell 177, 608–621.e12 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Inman, G. J. et al. The genomic landscape of cutaneous SCC reveals drivers and a novel azathioprine associated mutational signature. Nat. Commun. 9, 3667 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  124. Bonilla, X. et al. Genomic analysis identifies new drivers and progression pathways in skin basal cell carcinoma. Nat. Genet. 48, 398–406 (2016).

    Article  CAS  PubMed  Google Scholar 

  125. Besaratinia, A., Kim, S. I., Bates, S. E. & Pfeifer, G. P. Riboflavin activated by ultraviolet A1 irradiation induces oxidative DNA damage-mediated mutations inhibited by vitamin C. Proc. Natl Acad. Sci. USA 104, 5953–5958 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Yizhak, K. et al. RNA sequence analysis reveals macroscopic somatic clonal expansion across normal tissues. Science 364, eaaw0726 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Totoki, Y. et al. Trans-ancestry mutational landscape of hepatocellular carcinoma genomes. Nat. Genet. 46, 1267–1273 (2014).

    Article  CAS  PubMed  Google Scholar 

  128. Alberg, A. J., Brock, M. V., Ford, J. G., Samet, J. M. & Spivack, S. D. Epidemiology of lung cancer: diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest 143, e1S–e29S (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Tang, J. et al. The genomic landscapes of individual melanocytes from human skin. Nature 586, 600–605 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Kang, H. & Shibata, D. Direct measurements of human colon crypt stem cell niche genetic fidelity: the role of chance in non-Darwinian mutation selection. Front. Oncol. 3, 264 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  131. Nguyen, H. et al. Deficient Pms2, ERCC1, Ku86, CcOI in field defects during progression to colon cancer. J. Vis. Exp. 41, e1931 (2010).

    Google Scholar 

  132. Buc, E. et al. High prevalence of mucosa-associated E. coli producing cyclomodulin and genotoxin in colon cancer. PLoS ONE 8, e56964 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Salk, J. J. et al. Clonal expansions in ulcerative colitis identify patients with neoplasia. Proc. Natl Acad. Sci. USA 106, 20871–20876 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Stachler, M. D. et al. Detection of mutations in Barrett’s esophagus before progression to high-grade dysplasia or adenocarcinoma. Gastroenterology 155, 156–167 (2018).

    Article  CAS  PubMed  Google Scholar 

  135. Fujimoto, A. et al. Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat. Genet. 44, 760–764 (2012).

    Article  CAS  PubMed  Google Scholar 

  136. Schulze, K. et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat. Genet. 47, 505–511 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Cancer Genome Atlas Research Network. Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell 169, 1327–1341.e23 (2017).

    Article  CAS  Google Scholar 

  138. Kensler, T. W. et al. Transforming cancer prevention through precision medicine and immune-oncology. Cancer Prev. Res. 9, 2–10 (2016).

    Article  CAS  Google Scholar 

  139. Curtius, K., Wright, N. A. & Graham, T. A. An evolutionary perspective on field cancerization. Nat. Rev. Cancer 18, 19–32 (2018).

    Article  CAS  PubMed  Google Scholar 

  140. Hori, M. et al. Cancer incidence and incidence rates in Japan in 2009: a study of 32 population-based cancer registries for the Monitoring of Cancer Incidence in Japan (MCIJ) project. Jpn. J. Clin. Oncol. 45, 884–891 (2015).

    Article  PubMed  Google Scholar 

  141. Statistical Research and Applications Branch, National Cancer Institute. DevCan: Probability of Developing or Dying of Cancer Software. Version 6.7.7 (National Cancer Institute, 2019).

  142. Abelson, S. et al. Prediction of acute myeloid leukaemia risk in healthy individuals. Nature 559, 400–404 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Desai, P. et al. Somatic mutations precede acute myeloid leukemia years before diagnosis. Nat. Med. 24, 1015–1023 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Akbari, M. R. et al. PPM1D mutations in circulating white blood cells and the risk for ovarian cancer. J. Natl Cancer Inst. 106, djt323 (2014).

    Article  PubMed  CAS  Google Scholar 

  145. Wong, T. N. et al. Cellular stressors contribute to the expansion of hematopoietic clones of varying leukemic potential. Nat. Commun. 9, 455 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  146. Sun, X. et al. Arid1a has context-dependent oncogenic and tumor suppressor functions in liver cancer. Cancer Cell 32, 574–589.e6 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Alcolea, M. P. et al. Differentiation imbalance in single oesophageal progenitor cells causes clonal immortalization and field change. Nat. Cell Biol. 16, 615–622 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Demehri, S., Turkoz, A. & Kopan, R. Epidermal Notch1 loss promotes skin tumorigenesis by impacting the stromal microenvironment. Cancer Cell 16, 55–66 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Kagoya, Y. et al. JAK2V617F+ myeloproliferative neoplasm clones evoke paracrine DNA damage to adjacent normal cells through secretion of lipocalin-2. Blood 124, 2996–3006 (2014).

    Article  CAS  PubMed  Google Scholar 

  150. Jaiswal, S. et al. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N. Engl. J. Med. 377, 111–121 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  151. Fuster, J. J. et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science 355, 842–847 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Qian, Y. et al. The adaptor Act1 is required for interleukin 17-dependent signaling associated with autoimmune and inflammatory disease. Nat. Immunol. 8, 247–256 (2007).

    Article  CAS  PubMed  Google Scholar 

  153. Lee, J. S. et al. Interleukin-23-independent IL-17 production regulates intestinal epithelial permeability. Immunity 43, 727–738 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Reikvam, D. H. et al. Epithelial–microbial crosstalk in polymeric Ig receptor deficient mice. Eur. J. Immunol. 42, 2959–2970 (2012).

    Article  CAS  PubMed  Google Scholar 

  155. Joseph, C. G. et al. Association of the autoimmune disease scleroderma with an immunologic response to cancer. Science 343, 152–157 (2014).

    Article  CAS  PubMed  Google Scholar 

  156. Olcaydu, D. et al. A common JAK2 haplotype confers susceptibility to myeloproliferative neoplasms. Nat. Genet. 41, 450–454 (2009).

    Article  CAS  PubMed  Google Scholar 

  157. Kilpivaara, O. et al. A germline JAK2 SNP is associated with predisposition to the development of JAK2(V617F)-positive myeloproliferative neoplasms. Nat. Genet. 41, 455–459 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Jones, A. V. et al. JAK2 haplotype is a major risk factor for the development of myeloproliferative neoplasms. Nat. Genet. 41, 446–449 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. 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 

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

    Article  CAS  PubMed  Google Scholar 

  161. Wu, J. & Izpisua Belmonte, J. C. Stem cells: a renaissance in human biology research. Cell 165, 1572–1585 (2016).

    Article  CAS  PubMed  Google Scholar 

  162. Atlasi, Y. & Stunnenberg, H. G. The interplay of epigenetic marks during stem cell differentiation and development. Nat. Rev. Genet. 18, 643–658 (2017).

    Article  CAS  PubMed  Google Scholar 

  163. Coorens, T. H. H. et al. Embryonal precursors of Wilms tumor. Science 366, 1247–1251 (2019). This study describes that H19 locus hypermethylation, epi-mutation, can drive clonal expansion during nephrogenesis, which can contribute to the development of Wilms tumour.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Khurana, E. et al. Integrative annotation of variants from 1092 humans: application to cancer genomics. Science 342, 1235587 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  165. 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 

  166. 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 

  167. 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 

  168. Melton, C., Reuter, J. A., Spacek, D. V. & Snyder, M. Recurrent somatic mutations in regulatory regions of human cancer genomes. Nat. Genet. 47, 710–716 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  170. 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 

  171. 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 

  172. 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 

  173. Pickar-Oliver, A. & Gersbach, C. A. The next generation of CRISPR–Cas technologies and applications. Nat. Rev. Mol. Cell Biol. 20, 490–507 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Kretzschmar, K. & Clevers, H. Organoids: modeling development and the stem cell niche in a dish. Dev. Cell 38, 590–600 (2016).

    Article  CAS  PubMed  Google Scholar 

  175. Drost, J. & Clevers, H. Organoids in cancer research. Nat. Rev. Cancer 18, 407–418 (2018).

    Article  CAS  PubMed  Google Scholar 

  176. Burgess, D. J. Spatial transcriptomics coming of age. Nat. Rev. Genet. 20, 317 (2019).

    Article  CAS  PubMed  Google Scholar 

  177. Stuart, T. & Satija, R. Integrative single-cell analysis. Nat. Rev. Genet. 20, 257–272 (2019).

    Article  CAS  PubMed  Google Scholar 

  178. Schatz, M. C. Nanopore sequencing meets epigenetics. Nat. Methods 14, 347–348 (2017).

    Article  CAS  PubMed  Google Scholar 

  179. Ho, S. S., Urban, A. E. & Mills, R. E. Structural variation in the sequencing era. Nat. Rev. Genet. 21, 171–189 (2020).

    Article  CAS  PubMed  Google Scholar 

  180. Jaiswal, S. & Ebert, B. L. Clonal hematopoiesis in human aging and disease. Science 366, eaan4673 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Freed, D., Stevens, E. L. & Pevsner, J. Somatic mosaicism in the human genome. Genes 5, 1064–1094 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  182. Linder, D. & Gartler, S. M. Distribution of glucose-6-phosphate dehydrogenase electrophoretic variants in different tissues of heterozygotes. Am. J. Hum. Genet. 17, 212–220 (1965).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Behjati, S. et al. Genome sequencing of normal cells reveals developmental lineages and mutational processes. Nature 513, 422–425 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Ju, Y. S. et al. Somatic mutations reveal asymmetric cellular dynamics in the early human embryo. Nature 543, 714–718 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Hou, Y. et al. Single-cell exome sequencing and monoclonal evolution of a JAK2-negative myeloproliferative neoplasm. Cell 148, 873–885 (2012).

    Article  CAS  PubMed  Google Scholar 

  186. Nicholson, A. M. et al. Fixation and spread of somatic mutations in adult human colonic epithelium. Cell Stem Cell 22, 909–918.e8 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Seishi Ogawa.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Cancer thanks R. van Boxtel 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.

Glossary

Driver mutations

Those mutations that confer selective advantage for their target cells to allow clonal outgrowth, whereas those mutations acquired prior to a driver mutation are also inherited by driver-mutated and clonally expanded cells despite conferring no selective roles and are called passenger mutations.

Positively selected clones

Clones that outcompete surrounding cells by acquisition of driver mutations that confer selective advantages to the cells.

Lyonized cells

Those cells undergoing inactivation of one of the two X chromosome copies in females.

Skewed chromosome X inactivation

In the presence of a clone, inactivation of one of the two chromosomes in early development of female organisms (lyonization) may be skewed from the 1:1 ratio that is expected for a random process, which can be used to infer the presence of clonal evolution in the sample of interest.

Genetic drift

A change in the frequency of an existing gene variant in a population due to random sampling of cells or organisms.

Allele dropout

The preferential amplification of one of a pair of heterozygous alleles frequently observed in amplification from a single cell or a small number of cells, in which the other allele is totally under-represented or fails to be detected.

Ratio of non-synonymous to synonymous mutations

(dN/dS). The ratio of the number of non-synonymous substitutions per non-synonymous site to the number of synonymous substitutions per synonymous site, frequently used as an indicator of selective pressure acting on a protein coding gene.

Variant allele frequency

The frequency of the mutated allele, correlating with the clonal cell fraction within a sample, that is calculated by dividing the number of mutant reads by the total number of reads that encompass the mutated position in sequencing analysis.

Loss of heterozygosity

An allelic state in which one of the two parental alleles is lost with or without copy number loss, providing a common mechanism of biallelic inactivation of tumour suppressor genes or duplication of a gain-of-function mutation accompanied by loss of wild-type alleles.

Parity

The number of times a female has given birth.

Chromothripsis

A mutational process by which clustered chromosomal rearrangements occur in a single event affecting one or a few chromosomes.

Negative selection

The selective removal of defective/disadvantageous alleles or cells having such alleles from a population of species or cells.

Field carcinogenesis

A biological process in which large areas of tissues or organs are affected by carcinogenic alterations.

Epi-mutations

Heritable changes without any accompanying actual base pair changes in primary DNA sequences, which are typically represented by DNA methylation, histone modifications and other mechanisms.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kakiuchi, N., Ogawa, S. Clonal expansion in non-cancer tissues. Nat Rev Cancer 21, 239–256 (2021). https://doi.org/10.1038/s41568-021-00335-3

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-021-00335-3

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