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:

Cancers make their own luck: theories of cancer origins

Abstract

Cancer has been a leading cause of death for decades. This dismal statistic has increased efforts to prevent the disease or to detect it early, when treatment is less invasive, relatively inexpensive and more likely to cure. But precisely how tissues are transformed continues to provoke controversy and debate, hindering cancer prevention and early intervention strategies. Various theories of cancer origins have emerged, including the suggestion that it is ‘bad luck’: the inevitable consequence of random mutations in proliferating stem cells. In this Review, we discuss the principal theories of cancer origins and the relative importance of the factors that underpin them. The body of available evidence suggests that developing and ageing tissues ‘walk a tightrope’, retaining adequate levels of cell plasticity to generate and maintain tissues while avoiding overstepping into transformation. Rather than viewing cancer as ‘bad luck’, understanding the complex choreography of cell intrinsic and extrinsic factors that characterize transformation holds promise to discover effective new ways to prevent, detect and stop cancer before it becomes incurable.

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: Major causes of disease-related death.
Fig. 2: Organ sites of common human cancers.
Fig. 3: Principal theories of cancer origins.
Fig. 4: Origins of childhood cancer.
Fig. 5: Origins of adult cancer.

References

  1. Centers for Disease Control. Leading Causes of Death 1900–1998 https://www.cdc.gov/nchs/data/dvs/lead1900_98.pdf (2000).

  2. Office for National Statistics. Causes of Death Over 100 Years https://www.ons.gov.uk/peoplepopulationandcommunity/birthsdeathsandmarriages/deaths/articles/causesofdeathover100years/2017-09-18 (2017).

  3. Institute of Medicine (US) Committee for the Study of the Future of Public Health. The Future of Public Health (National Academies Press (US), 1988).

  4. Koch, R. Die aetiologie der milzbrand-krankheit, begründet auf die entwicklungsgeschichte des bacillus anthracis [German]. Beitr. Z. Biol. Pflanz. 1, 277–308 (1876).

    Google Scholar 

  5. Smith, P. W., Watkins, K. & Hewlett, A. Infection control through the ages. Am. J. Infect. Control. 40, 35–42 (2012).

    Article  PubMed  Google Scholar 

  6. Ehrlich, P. & Bertheim, A. Über das salzsaure 3.3′-Diamino-4.4′-dioxy-arsenobenzol und seine nächsten Verwandten [German]. Ber. der Dtsch. chemischen Ges. 45, 756–766 (1912).

    Article  CAS  Google Scholar 

  7. Fleming, A. On the antibacterial action of cultures of a penicillium, with special reference to their use in the isolation of B. influenzae. Br. J. Exp. Pathol. 10, 226–236 (1929).

    CAS  PubMed Central  Google Scholar 

  8. Plotkin, S. History of vaccination. Proc. Natl Acad. Sci. USA 111, 12283–12287 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Voysey, M. et al. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet 397, 99–111 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. King, M. L. & Sullivan, M. M. The similarity of the effect of podophyllin and colchicine and their use in the treatment of condylomata acuminata. Science 104, 244–245 (1946).

    Article  CAS  PubMed  Google Scholar 

  11. Pappenheimer, A. M. & Vance, M. The effects of intravenous injections of dichloroethylsulfide in rabbits, with special reference to its leucotoxic action. J. Exp. Med. 31, 71–94 (1920).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Sawyers, C. Targeted cancer therapy. Nature 432, 294–297 (2004).

    Article  CAS  PubMed  Google Scholar 

  13. Cable, J. et al. Frontiers in cancer immunotherapy — a symposium report. Ann. N. Y. Acad. Sci. 1489, 30–47 (2021).

    Article  CAS  PubMed  Google Scholar 

  14. Waldman, A. D., Fritz, J. M. & Lenardo, M. J. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat. Rev. Immunol. 20, 651–668 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Sung, H. et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71, 209–249 (2021). This work presents global statistics of cancer incidence and mortality, emphasizing the human health emergency posed by cancer and the need for earlier intervention.

    Article  PubMed  Google Scholar 

  16. McPhail, S. et al. Risk factors and prognostic implications of diagnosis of cancer within 30 days after an emergency hospital admission (emergency presentation): an International Cancer Benchmarking Partnership (ICBP) population-based study. Lancet Oncol. 23, 587–600 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Danaei, G., vander Hoorn, S., Lopez, A. D., Murray, C. J. L. & Ezzati, M. Causes of cancer in the world: comparative risk assessment of nine behavioural and environmental risk factors. Lancet 366, 1784–1793 (2005).

    Article  PubMed  Google Scholar 

  18. National Health Services. NHS Long Term Plan for Cancer https://www.longtermplan.nhs.uk/areas-of-work/cancer/ (2019).

  19. WHO. WHO report on cancer: setting priorities, investing wisely and providing care for all. World Health Organization https://www.who.int/publications/i/item/9789240001299 (2022).

  20. Crosby, D. et al. Early detection of cancer. Science 375, aay9040 (2022).

    Article  Google Scholar 

  21. Fitzgerald, R. C. et al. Cytosponge-trefoil factor 3 versus usual care to identify Barrett’s oesophagus in a primary care setting: a multicentre, pragmatic, randomised controlled trial. Lancet 396, 333–344 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Gredner, T., Mons, U., Niedermaier, T., Brenner, H. & Soerjomataram, I. Impact of tobacco control policies implementation on future lung cancer incidence in Europe: an international, population-based modeling study. Lancet Reg. Health Eur. 4, 100074 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Health and Safety Executive. Mesothelioma Statistics for Great Britain, 2023 https://www.hse.gov.uk/statistics/causdis/mesothelioma/mesothelioma.pdf (2023).

  24. Chang, M. H. et al. Decreased incidence of hepatocellular carcinoma in hepatitis B vaccinees: a 20-year follow-up study. J. Natl Cancer Inst. 101, 1348–1355 (2009).

    Article  CAS  PubMed  Google Scholar 

  25. Roden, R. B. S. & Stern, P. L. Opportunities and challenges for human papillomavirus vaccination in cancer. Nat. Rev. Cancer 18, 240–254 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. GBD 2019 Cancer Risk Factors Collaborators. The global burden of cancer attributable to risk factors, 2010–19: a systematic analysis for the Global Burden of Disease Study 2019. Lancet 400, 563–591 (2022).

    Article  Google Scholar 

  27. Force, L. M. et al. The global burden of childhood and adolescent cancer in 2017: an analysis of the Global Burden of Disease Study 2017. Lancet Oncol. 20, 1211–1225 (2019).

    Article  Google Scholar 

  28. Johnston, W. T. et al. Childhood cancer: estimating regional and global incidence. Cancer Epidemiol. 71, 101662 (2021).

    Article  CAS  PubMed  Google Scholar 

  29. Nordling, C. O. A new theory on the cancer-inducing mechanism. Br. J. Cancer 7, 68–72 (1953).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Boveri, T. concerning the origin of malignant tumors. J. Cell Sci. 7, 68–72 (1929).

    Google Scholar 

  31. Knudson, A. G. Mutation and cancer: statistical study of retinoblastoma. Proc. Natl Acad. Sci. USA 68, 820–823 (1971).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Armitage, P. & Doll, R. The age distribution of cancer and a multi-stage theory of carcinogenesis. Br. J. Cancer 8, 1–12 (1954). Together with Nordling (1953), Boveri (1929) and Knudson (1971), this study details the mutation theory of cancer in which malignant transformation is proposed to arise as a consequence of several changes in DNA that confer proliferative and invasive cell properties.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wiley Online Library. Complete pathologic maturation and regression of stage IVS neuroblastoma without treatment. Cancer 62, 818–925 (1967).

    Google Scholar 

  34. Huggins, C. Endocrine-induced regression of cancers. Science 156, 1050–1054 (1967).

    Article  CAS  PubMed  Google Scholar 

  35. Mintz, B. & Illmensee, K. Normal genetically mosaic mice produced from malignant teratocarcinoma cells. Proc. Natl Acad. Sci. USA 72, 3585–3589 (1975).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Committee on Chemical Environmental Mutagens, Board on Toxicology and Environmental Health Hazards, Commission on Life Sciences & National Research Council. Identifying and Estimating the Genetic Impact of Chemical Mutagens https://nap.nationalacademies.org/read/19435/chapter/1 (National Academy Press, 1983).

  37. Riva, L. et al. The mutational signature profile of known and suspected human carcinogens in mice. Nat. Genet. 52, 1189–1197 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Soto, A. M. & Sonnenschein, C. The tissue organization field theory of cancer: a testable replacement for the somatic mutation theory. BioEssays 33, 332–340 (2011). This work details the tissue organization field theory of cancer in which cell extrinsic properties of cells are identified as key regulators of transformation.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Tomasetti, C. & Vogelstein, B. Cancer etiology. Variation in cancer risk among tissues can be explained by the number of stem cell divisions. Science 347, 78–81 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zhu, L. et al. Multi-organ mapping of cancer risk. Cell 166, 1132–1146.e7 (2016). This work uses Cre-conditional mouse models for lifelong lineage tracing and cancer induction studies and demonstrates that cancer in multiple organs results from the mutation of stem cells in a cancer-susceptible state that varies with age, damage and site, underpinning the ground state theory of cancer.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Tomasetti, C., Li, L. & Vogelstein, B. Stem cell divisions, somatic mutations, cancer etiology, and cancer prevention. Science 355, 1330–1334 (2017). Together with Tomasetti and Vogelstein (2015), this study describes the ‘bad luck’ theory of cancer in which transformation is proposed to result from the chance mutation of proliferating stem cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Davey Smith, G., Relton, C. L. & Brennan, P. Chance, choice and cause in cancer aetiology: individual and population perspectives. Int. J. Epidemiol. 45, 605–613 (2016).

    Article  PubMed  Google Scholar 

  43. Abascal, F. et al. Somatic mutation landscapes at single-molecule resolution. Nature 593, 405–410 (2021).

    Article  CAS  PubMed  Google Scholar 

  44. Lopez-Bigas, N. & Gonzalez-Perez, A. Are carcinogens direct mutagens? Nat. Genet. 52, 1137–1138 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. Boshart, M. et al. A new type of papillomavirus DNA, its presence in genital cancer biopsies and in cell lines derived from cervical cancer. EMBO J. 3, 1151–1157 (1984).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Koutsky, L. A. et al. A controlled trial of a human papillomavirus type 16 vaccine. N. Engl. J. Med. 347, 1645–1651 (2002).

    Article  CAS  PubMed  Google Scholar 

  47. Zhu, L. et al. Prominin 1 marks intestinal stem cells that are susceptible to neoplastic transformation. Nature 457, 603–607 (2009).

    Article  CAS  PubMed  Google Scholar 

  48. Kretzschmar, K. & Watt, F. M. Lineage tracing. Cell 148, 33–45 (2012).

    Article  CAS  PubMed  Google Scholar 

  49. Cairns, J. Mutation selection and the natural history of cancer. Nature 255, 197–200 (1975).

    Article  CAS  PubMed  Google Scholar 

  50. Casás-Selves, M. & Degregori, J. How cancer shapes evolution, and how evolution shapes cancer. Evolution 4, 624–634 (2011).

    PubMed  PubMed Central  Google Scholar 

  51. Campisi, J. Aging, tumor suppression and cancer: high wire-act! Mech. Ageing Dev. 126, 51–58 (2005).

    Article  CAS  PubMed  Google Scholar 

  52. Porter, S. N. et al. Fetal and neonatal hematopoietic progenitors are functionally and transcriptionally resistant to Flt3-ITD mutations. eLife 5, e18882 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Giambra, V. et al. Epigenetic restoration of fetal-like IGF1 signaling inhibits leukemia stem cell activity. Cell Stem Cell 23, 714–726.e7 (2018).

    Article  CAS  PubMed  Google Scholar 

  54. Bae, T. et al. Different mutational rates and mechanisms in human cells at pregastrulation and neurogenesis. Science 359, 550–555 (2018).

    Article  CAS  PubMed  Google Scholar 

  55. 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 

  56. Hill, W. et al. Lung adenocarcinoma promotion by air pollutants. Nature 616, 159–167 (2023). This combination of human epidemiological and tissue studies as well as mouse models demonstrates that principles of the ground state theory of cancer operate in determining lung adenocarcinoma risk.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Visvader, J. E. Cells of origin in cancer. Nature 469, 314–322 (2011).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  59. Zhang, M., Lee, A. V. & Rosen, J. M. The cellular origin and evolution of breast cancer. Cold Spring Harb. Perspect. Med 7, a027128 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Wang, X. et al. A luminal epithelial stem cell that is a cell of origin for prostate cancer. Nature 461, 495–500 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Kim, C. F. B. et al. Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell 121, 823–835 (2005).

    Article  CAS  PubMed  Google Scholar 

  62. Barker, N. et al. Crypt stem cells as the cells-of-origin of intestinal cancer. Nature 457, 608–611 (2009).

    Article  CAS  PubMed  Google Scholar 

  63. Barker, N. et al. Lgr5+ve stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell 6, 25–36 (2010).

    Article  CAS  PubMed  Google Scholar 

  64. Colom, B. et al. Mutant clones in normal epithelium outcompete and eliminate emerging tumours. Nature 598, 510–514 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Hope, K. J., Jin, L. & Dick, J. E. Acute myeloid leukemia originates from a hierarchy of leukemic stem cell classes that differ in self-renewal capacity. Nat. Immunol. 5, 738–743 (2004).

    Article  CAS  PubMed  Google Scholar 

  66. Johnson, R. A. et al. Cross-species genomics matches driver mutations and cell compartments to model ependymoma. Nature 466, 632–636 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Gibson, P. et al. Subtypes of medulloblastoma have distinct developmental origins. Nature 468, 1095–1099 (2010). Together with Johnson et al. (2010), this study demonstrates that neural lineages in the embryonic brain are susceptible to different oncogenic mutations, resulting in the development of the various subtypes of childhood brain tumours observed in the clinic.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Huntly, B. J. P. et al. MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine hematopoietic progenitors. Cancer Cell 6, 587–596 (2004).

    Article  CAS  PubMed  Google Scholar 

  69. Cozzio, A. et al. Similar MLL-associated leukemias arising from self-renewing stem cells and short-lived myeloid progenitors. Genes Dev. 17, 3029 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Kreso, A. & Dick, J. E. Evolution of the cancer stem cell model. Cell Stem Cell 14, 275–291 (2014).

    Article  CAS  PubMed  Google Scholar 

  71. Magee, J. A., Piskounova, E. & Morrison, S. J. Cancer stem cells: impact, heterogeneity, and uncertainty. Cancer Cell 21, 283–296 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Schneider, G., Schmidt-Supprian, M., Rad, R. & Saur, D. Tissue-specific tumorigenesis: context matters. Nat. Rev. Cancer 17, 239–253 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Haigis, K. M., Cichowski, K. & Elledge, S. J. Tissue-specificity in cancer: the rule, not the exception. Science 363, 1150–1151 (2019).

    Article  CAS  PubMed  Google Scholar 

  74. Hendrikse, L. D. et al. Failure of human rhombic lip differentiation underlies medulloblastoma formation. Nature 609, 1021–1028 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Vladoiu, M. C. et al. Childhood cerebellar tumours mirror conserved fetal transcriptional programs. Nature 572, 67–73 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Taylor, M. D. et al. Radial glia cells are candidate stem cells of ependymoma. Cancer Cell 8, 323–335 (2005).

    Article  CAS  PubMed  Google Scholar 

  77. Hovestadt, V. et al. Resolving medulloblastoma cellular architecture by single-cell genomics. Nature 572, 74–79 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Patmore, D. M. et al. DDX3X suppresses the susceptibility of hindbrain lineages to medulloblastoma. Dev. Cell 54, 455–470.e5 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Filbin, M. G. et al. Developmental and oncogenic programs in H3K27M gliomas dissected by single-cell RNA-seq. Science 360, 331–335 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Nagaraja, S. et al. Transcriptional dependencies in diffuse intrinsic pontine glioma. Cancer Cell 31, 635–652.e6 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Pathania, M. et al. H3.3K27M cooperates with Trp53 loss and PDGFRA gain in mouse embryonic neural progenitor cells to induce invasive high-grade gliomas. Cancer Cell 32, 684–700.e9 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Funato, K., Major, T., Lewis, P. W., Allis, C. D. & Tabar, V. Use of human embryonic stem cells to model pediatric gliomas with H3.3K27M histone mutation. Science 346, 1529–1533 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Silveira, A. B. et al. H3.3 K27M depletion increases differentiation and extends latency of diffuse intrinsic pontine glioma growth in vivo. Acta Neuropathol. 137, 637–655 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Riggi, N. et al. EWS-FLI-1 expression triggers a Ewing’s sarcoma initiation program in primary human mesenchymal stem cells. Cancer Res. 68, 2176–2185 (2008).

    Article  CAS  PubMed  Google Scholar 

  85. Sole, A. et al. Unraveling Ewing sarcoma tumorigenesis originating from patient-derived mesenchymal stem cells. Cancer Res. 81, 4994–5006 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Krivtsov, A. V. et al. Cell of origin determines clinically relevant subtypes of MLL-rearranged AML. Leukemia 27, 852–860 (2013).

    Article  CAS  PubMed  Google Scholar 

  87. Custers, L. et al. Somatic mutations and single-cell transcriptomes reveal the root of malignant rhabdoid tumours. Nat. Commun. 12, 1407 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Coorens, T. H. H. et al. Embryonal precursors of wilms tumor. Science 366, 1247–1251 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Abarrategi, A. et al. Osteosarcoma: cells-of-origin, cancer stem cells, and targeted therapies. Stem Cell Int. 2016, 3631764 (2016).

    Google Scholar 

  90. Jansky, S. et al. Single-cell transcriptomic analyses provide insights into the developmental origins of neuroblastoma. Nat. Genet. 53, 683–693 (2021).

    Article  CAS  PubMed  Google Scholar 

  91. White, R. M. et al. DHODH modulates transcriptional elongation in the neural crest and melanoma. Nature 471, 518–522 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Kaufman, C. K. et al. A zebrafish melanoma model reveals emergence of neural crest identity during melanoma initiation. Science 351, aad2197 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  93. Baggiolini, A. et al. Developmental chromatin programs determine oncogenic competence in melanoma. Science 373, eabc1048 (2022).

    Article  Google Scholar 

  94. Horvath, S. & Raj, K. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat. Rev. Genet. 19, 371–384 (2018).

    Article  CAS  PubMed  Google Scholar 

  95. Booth, L. N. & Brunet, A. The aging epigenome. Mol. Cell 62, 728–744 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Widschwendter, M. et al. Epigenome-based cancer risk prediction: rationale, opportunities and challenges. Nat. Rev. Clin. Oncol. 15, 292–309 (2018).

    Article  PubMed  Google Scholar 

  97. Xie, W. et al. DNA methylation patterns separate senescence from transformation potential and indicate cancer risk. Cancer Cell 33, 309–321.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Ohnishi, K. et al. Premature termination of reprogramming in vivo leads to cancer development through altered epigenetic regulation. Cell 156, 663–677 (2014). This study using mouse models shows that epigenetic reprogramming can drive tumorigenesis but this transformation is reversible, demonstrating that irreversible genetic change is not required for tumour formation.

    Article  CAS  PubMed  Google Scholar 

  99. Chen, X., Pappo, A. & Dyer, M. A. Pediatric solid tumor genomics and developmental pliancy. Oncogene 34, 5207–5215 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Gilbertson, R. J. Mapping cancer origins. Cell 145, 25–29 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. White, M. C. et al. Age and cancer risk: a potentially modifiable relationship. Am. J. Prev. Med. 46, S7–S15 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  102. Morales Berstein, F. et al. Assessing the causal role of epigenetic clocks in the development of multiple cancers: a Mendelian randomization study. eLife 11, e75374 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  103. Rodrigues, C. P., Shvedunova, M. & Akhtar, A. Epigenetic regulators as the gatekeepers of hematopoiesis. Trends Genet. 37, 125–142 (2021).

    Article  CAS  Google Scholar 

  104. Sun, D. et al. Epigenomic profiling of young and aged HSCs reveals concerted changes during aging that reinforce self-renewal. Cell Stem Cell 14, 673–688 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Johnson, K. C., Houseman, E. A., King, J. E. & Christensen, B. C. Normal breast tissue DNA methylation differences at regulatory elements are associated with the cancer risk factor age. Breast Cancer Res. 19, 81 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  106. Nacev, B. A. et al. The expanding landscape of ‘oncohistone’ mutations in human cancers. Nature 567, 473–478 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. 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 

  108. Jaiswal, S. et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 371, 2488–2498 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  109. 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  Google Scholar 

  110. Kar, S. P. et al. Genome-wide analyses of 200,453 individuals yield new insights into the causes and consequences of clonal hematopoiesis. Nat. Genet. 54, 1155–1166 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Yang, L., Rau, R. & Goodell, M. A. DNMT3A in haematological malignancies. Nat. Rev. Cancer 15, 152–165 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Reed, S. C., Croessmann, S. & Park, B. H. CHIP happens: clonal hematopoiesis of indeterminate potential and its relationship to solid tumors. Clin. Cancer Res. 29, 1403–1411 (2023).

    Article  CAS  PubMed  Google Scholar 

  113. Frigola, J. et al. Epigenetic remodeling in colorectal cancer results in coordinate gene suppression across an entire chromosome band. Nat. Genet. 38, 540–549 (2006).

    Article  CAS  PubMed  Google Scholar 

  114. Campbell, P. J. et al. Pan-cancer analysis of whole genomes. Nature 578, 82–93 (2020).

    Article  Google Scholar 

  115. Shen, X., Song, S., Li, C. & Zhang, J. Synonymous mutations in representative yeast genes are mostly strongly non-neutral. Nature 606, 725–731 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Kruglyak, L. et al. Insufficient evidence for non-neutrality of synonymous mutations. Nature 616, E8–E9 (2023).

    Article  CAS  PubMed  Google Scholar 

  117. 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 

  118. Benisty, H., Weber, M., Hernandez-Alias, X., Schaefer, M. H. & Serrano, L. Mutation bias within oncogene families is related to proliferation-specific codon usage. Proc. Natl Acad. Sci. USA 117, 30848–30856 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Blokzijl, F. et al. Tissue-specific mutation accumulation in human adult stem cells during life. Nature 538, 260–264 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Martincorena, I. et al. High burden and pervasive positive selection of somatic mutations in normal human skin. Science 348, 880–886 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. 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 

  122. Yokoyama, A. et al. Age-related remodelling of oesophageal epithelia by mutated cancer drivers. Nature 565, 312–317 (2019).

    Article  CAS  PubMed  Google Scholar 

  123. 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 

  124. Zhu, M. et al. Somatic mutations increase hepatic clonal fitness and regeneration in chronic liver disease. Cell 177, 608–621.e12 (2019). Together with Martincorena et al. (2015), Martincorena et al. (2018), Yokoyama et al. (2019) and Yizhak et al. (2019), this study describes the accumulation of large clones harbouring potentially oncogenic mutations in otherwise apparently normal ageing human tissues.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Hill, J. Cautions Against the Immoderate Use of Snuff: Founded on the Known Qualities of the Tobacco Plant: and the Effects It Must Produce When This Way Taken into the Body: and Enforced by Instances of Persons Who Have Perished Miserably of Diseases, Occasioned, or Rendered Incurable by Its Use http://resource.nlm.nih.gov/2166041R (1761).

  126. Song, M. & Giovannucei, E. L. Cancer risk: accuracy of literature. J. Natl Cancer Inst. 66, 1784 (1981).

    Google Scholar 

  127. O’callaghan, M. Cancer risk: accuracy of literature. Science 347, 729 (2015).

    Article  PubMed  Google Scholar 

  128. Wild, C. et al. Cancer risk: role of chance overstated. Science 347, 728 (2015).

    Article  CAS  PubMed  Google Scholar 

  129. Potter, J. D. & Prentice, R. L. Cancer risk: tumors excluded. Science 347, 727 (2015).

    Article  CAS  PubMed  Google Scholar 

  130. Gotay, C., Dummer, T. & Spinelli, J. Cancer risk: prevention is crucial. Science 347, 729 (2015).

    Article  Google Scholar 

  131. Ashford, N. A. et al. Cancer risk: role of environment. Science 347, 729–731 (2015).

    Article  Google Scholar 

  132. Wu, S., Powers, S., Zhu, W. & Hannun, Y. A. Substantial contribution of extrinsic risk factors to cancer development. Nature 529, 43–47 (2016).

    Article  CAS  PubMed  Google Scholar 

  133. Song, M., Vogelstein, B., Giovannucci, E. L., Willett, W. C. & Tomasetti, C. Cancer prevention: molecular and epidemiologic consensus. Science 361, 1317–1318 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Jones, D. L. & Wagers, A. J. No place like home: anatomy and function of the stem cell niche. Nat. Rev. Mol. Cell Biol. 9, 11–21 (2008).

    Article  CAS  PubMed  Google Scholar 

  135. Plaks, V., Kong, N. & Werb, Z. The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 16, 225–238 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Calabrese, C. et al. A perivascular niche for brain tumor stem cells. Cancer Cell 11, 69–82 (2007).

    Article  CAS  PubMed  Google Scholar 

  137. Gilbertson, R. J. & Rich, J. N. Making a tumour’s bed: glioblastoma stem cells and the vascular niche. Nat. Rev. Cancer 7, 733–736 (2007).

    Article  CAS  PubMed  Google Scholar 

  138. Hambardzumyan, D. et al. PI3K pathway regulates survival of cancer stem cells residing in the perivascular niche following radiation in medulloblastoma in vivo. Genes Dev. 22, 436–448 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Taniguchi, S. et al. Tumor-initiating cells establish an IL-33–TGF-β; niche signaling loop to promote cancer progression. Science 369, eaay1813 (2020).

    Article  CAS  PubMed  Google Scholar 

  140. Winkler, J., Abisoye-Ogunniyan, A., Metcalf, K. J. & Werb, Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat. Commun. 11, 5120 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Bissell, M. J. & Hines, W. C. Why don’t we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nat. Med. 17, 320–329 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Watt, F. M. & Huck, W. T. S. Role of the extracellular matrix in regulating stem cell fate. Nat. Rev. Mol. Cell Biol. 14, 467–473 (2013).

    Article  CAS  PubMed  Google Scholar 

  143. Frantz, C., Stewart, K. M. & Weaver, V. M. The extracellular matrix at a glance. J. Cell Sci. 123, 4195–4200 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Jones, C. E. et al. Stromal PTEN regulates extracellular matrix organization in the mammary gland. Neoplasia 21, 132–145 (2019).

    Article  CAS  PubMed  Google Scholar 

  145. Trimboli, A. J. et al. Pten in stromal fibroblasts suppresses mammary epithelial tumours. Nature 461, 1084–1091 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Stoker, A. W., Hatier, C. & Bissell, M. J. The embryonic environment strongly attenuates v-src oncogenesis in mesenchymal and epithelial tissues, but not in endothelia. J. Cell Biol. 111, 217–228 (1990).

    Article  CAS  PubMed  Google Scholar 

  147. Gonzalez, H., Hagerling, C. & Werb, Z. Roles of the immune system in cancer: from tumor initiation to metastatic progression. Genes. Dev. 32, 1267–1284 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Saluzzo, S. et al. Delayed antiretroviral therapy in HIV-infected individuals leads to irreversible depletion of skin- and mucosa-resident memory T cells. Immunity 54, 2842–2858.e5 (2021).

    Article  CAS  PubMed  Google Scholar 

  149. Gentles, A. J. et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 21, 938–945 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Teng, M. W. L., Swann, J. B., Koebel, C. M., Schreiber, R. D. & Smyth, M. J. Immune-mediated dormancy: an equilibrium with cancer. J. Leukoc. Biol. 84, 988–993 (2008).

    Article  CAS  PubMed  Google Scholar 

  151. Kim, H.-J. & Cantor, H. CD4 T-cell subsets and tumor immunity: the helpful and the not-so-helpful. Cancer Immunol. Res. 2, 91–98 (2014).

    Article  CAS  PubMed  Google Scholar 

  152. McGranahan, N. et al. Allele-specific HLA loss and immune escape in lung cancer evolution. Cell 171, 1259–1271.e11 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Chowell, D. et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 359, 582–587 (2018).

    Article  CAS  PubMed  Google Scholar 

  154. Mantovani, A., Marchesi, F., Malesci, A., Laghi, L. & Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 14, 399–416 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Böttcher, J. P. et al. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell 172, 1022–1037.e14 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  156. Agudo, J. et al. GFP-specific CD8 T cells enable targeted cell depletion and visualization of T-cell interactions. Nat. Biotechnol. 33, 1287–1292 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Agudo, J. et al. Quiescent tissue stem cells evade immune surveillance. Immunity 48, 271–285.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Parkin, D. M. The global health burden of infection-associated cancers in the year 2002. Int. J. Cancer 118, 3030–3044 (2006).

    Article  CAS  PubMed  Google Scholar 

  159. Plummer, M. et al. Global burden of cancers attributable to infections in 2012: a synthetic analysis. Lancet Glob. Health 4, e609–e616 (2016).

    Article  PubMed  Google Scholar 

  160. Zapatka, M. et al. The landscape of viral associations in human cancers. Nat. Genet. 52, 320–330 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Thrift, A. P., Wenker, T. N. & El-Serag, H. B. Global burden of gastric cancer: epidemiological trends, risk factors, screening and prevention. Nat. Rev. Clin. Oncol. 20, 338–349 (2023).

    Article  PubMed  Google Scholar 

  162. Jiang, Y., Han, Q., Zhao, H. & Zhang, J. The mechanisms of HBV-induced hepatocellular carcinoma. J. Hepatocell. Carcinoma 8, 435 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  163. Abhik, S. & S, R. E. Mechanisms of B-cell oncogenesis induced by Epstein–Barr virus. J. Virol. 93, e00238-19 (2019).

    Article  Google Scholar 

  164. Saha, A., Jha, H. C., Upadhyay, S. K. & Robertson, E. S. Epigenetic silencing of tumor suppressor genes during in vitro Epstein–Barr virus infection. Proc. Natl Acad. Sci. USA 112, E5199–E5207 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Martinez-Zapien, D. et al. Structure of the E6/E6AP/p53 complex required for HPV-mediated degradation of p53. Nature 529, 541–545 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Sung, W.-K. et al. Genome-wide survey of recurrent HBV integration in hepatocellular carcinoma. Nat. Genet. 44, 765–769 (2012).

    Article  CAS  PubMed  Google Scholar 

  167. Roberts, S. A. et al. An APOBEC cytidine deaminase mutagenesis pattern is widespread in human cancers. Nat. Genet. 45, 970–976 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Elinav, E., Garrett, W. S., Trinchieri, G. & Wargo, J. The cancer microbiome. Nat. Rev. Cancer 19, 371–376 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Rubinstein, M. R. et al. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin. Cell Host Microbe 14, 195–206 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Kostic, A. D. et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe 14, 207–215 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Schiffman, M. et al. Carcinogenic human papillomavirus infection. Nat. Rev. Dis. Prim. 2, 16086 (2016).

    Article  PubMed  Google Scholar 

  172. Yang, E. J. et al. Microanatomy of the cervical and anorectal squamocolumnar junctions: a proposed model for anatomical differences in HPV-related cancer risk. Mod. Pathol. 28, 994–1000 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Budhwani, M., Lukowski, S. W., Porceddu, S. V., Frazer, I. H. & Chandra, J. Dysregulation of stemness pathways in hpv mediated cervical malignant transformation identifies potential oncotherapy targets. Front. Cell Infect. Microbiol. 10, 307 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Burns, M. B., Temiz, N. A. & Harris, R. S. Evidence for APOBEC3B mutagenesis in multiple human cancers. Nat. Genet. 45, 977–983 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Alexandrov, L. B. et al. Clock-like mutational processes in human somatic cells. Nat. Genet. 47, 1402–1407 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Roswall, N. & Weiderpass, E. Alcohol as a risk factor for cancer: existing evidence in a global perspective. J. Prev. Med. Public Health 48, 1–9 (2015).

    PubMed  PubMed Central  Google Scholar 

  178. Bagnardi, V. et al. Alcohol consumption and site-specific cancer risk: a comprehensive dose–response meta-analysis. Br. J. Cancer 112, 580–593 (2015).

    Article  CAS  PubMed  Google Scholar 

  179. Rumgay, H. et al. Global burden of cancer in 2020 attributable to alcohol consumption: a population-based study. Lancet Oncol. 22, 1071–1080 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  180. Garaycoechea, J. I. et al. Alcohol and endogenous aldehydes damage chromosomes and mutate stem cells. Nature 553, 171–177 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Ahmad, A. S., Ormiston-Smith, N. & Sasieni, P. D. Trends in the lifetime risk of developing cancer in Great Britain: comparison of risk for those born from 1930 to 1960. Br. J. Cancer 112, 943–947 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. van Velthoven, C. T. J. & Rando, T. A. Stem cell quiescence: dynamism, restraint, and cellular idling. Cell Stem Cell 24, 213–225 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  183. Berenblum, I. & Shubik, P. A new, quantitative, approach to the study of the stages of chemical carcinogenesis in the mouse’s skin. Br. J. Cancer 1, 383–391 (1947).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Coussens, L. M. & Werb, Z. Inflammation and cancer. Nature 420, 860–867 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Walter, D. et al. Exit from dormancy provokes DNA-damage-induced attrition in haematopoietic stem cells. Nature 520, 549–552 (2015).

    Article  CAS  PubMed  Google Scholar 

  186. Insinga, A. et al. DNA damage in stem cells activates p21, inhibits p53, and induces symmetric self-renewing divisions. Proc. Natl Acad. Sci. USA 110, 3931–3936 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Behrens, A., van Deursen, J. M., Rudolph, K. L. & Schumacher, B. Impact of genomic damage and ageing on stem cell function. Nat. Cell Biol. 16, 201–207 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Tian, H. et al. A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable. Nature 478, 255–259 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Amcheslavsky, A., Jiang, J. & Ip, Y. T. Tissue damage-induced intestinal stem cell division in Drosophila. Cell Stem Cell 4, 49–61 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Lin, B. et al. Modulating cell fate as a therapeutic strategy. Cell Stem Cell 23, 329–341 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Ito, M. et al. Stem cells in the hair follicle bulge contribute to wound repair but not to homeostasis of the epidermis. Nat. Med. 11, 1351–1354 (2005).

    Article  CAS  PubMed  Google Scholar 

  192. Tata, P. R. et al. Dedifferentiation of committed epithelial cells into stem cells in vivo. Nature 503, 218–223 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Alonso-Curbelo, D. et al. A gene–environment-induced epigenetic program initiates tumorigenesis. Nature 590, 642–648 (2021). This study uses a combination of mutation and organ damage in the pancreas to demonstrate how gene–environment interactions can rapidly produce gene-regulatory programmes that dictate early neoplastic commitment.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Panciera, T. et al. Reprogramming normal cells into tumour precursors requires ECM stiffness and oncogene-mediated changes of cell mechanical properties. Nat. Mater. 19, 797–806 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Hiraiwa, K. & van Eeden, S. F. Contribution of lung macrophages to the inflammatory responses induced by exposure to air pollutants. Mediators Inflamm. 2013, 619523 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  196. Poplawski, G. H. D. et al. Injured adult neurons regress to an embryonic transcriptional growth state. Nature 581, 77–82 (2020).

    Article  CAS  PubMed  Google Scholar 

  197. Fazilaty, H. et al. Tracing colonic embryonic transcriptional profiles and their reactivation upon intestinal damage. Cell Rep. 36, 109484 (2021).

    Article  CAS  PubMed  Google Scholar 

  198. Vercauteren Drubbel, A. et al. Reactivation of the Hedgehog pathway in esophageal progenitors turns on an embryonic-like program to initiate columnar metaplasia. Cell Stem Cell 28, 1411–1427.e7 (2021).

    Article  CAS  PubMed  Google Scholar 

  199. Miao, Q. et al. SOX11 and SOX4 drive the reactivation of an embryonic gene program during murine wound repair. Nat. Commun. 10, 4042 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  200. Gurtner, G. C., Werner, S., Barrandon, Y. & Longaker, M. T. Wound repair and regeneration. Nature 453, 314–321 (2008).

    Article  CAS  PubMed  Google Scholar 

  201. Rolfe, K. J. & Grobbelaar, A. O. A review of fetal scarless healing. ISRN Dermatol. 2012, 698034 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  202. Sadek, H. & Olson, E. N. Toward the goal of human heart regeneration. Cell Stem Cell 26, 7–16 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Ganesh, K. & Massagué, J. Targeting metastatic cancer. Nat. Med. 27, 34–44 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Dillekås, H., Rogers, M. S. & Straume, O. Are 90% of deaths from cancer caused by metastases? Cancer Med. 8, 5574–5576 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  205. Massagué, J. & Ganesh, K. Metastasis-initiating cells and ecosystems. Cancer Discov. 11, 971–994 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  206. Massagué, J. & Obenauf, A. C. Metastatic colonization by circulating tumour cells. Nature 529, 298–306 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  207. Yang, J. et al. Guidelines and definitions for research on epithelial–mesenchymal transition. Nat. Rev. Mol. Cell Biol. 21, 341–352 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  208. Nájera, G. S. & Weijer, C. J. The evolution of gastrulation morphologies. Development 150, dev200885 (2023).

    Article  PubMed Central  Google Scholar 

  209. Podsypanina, K. et al. Seeding and propagation of untransformed mouse mammary cells in the lung. Science 321, 1841–1844 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Rahrmann, E. P. et al. The NALCN channel regulates metastasis and nonmalignant cell dissemination. Nat. Genet. 54, 1827–1838 (2022). Metastasis is the single biggest risk factor for cancer death: this study identifies NALCN as a regulator of cell shedding from solid tissues independent of cancer, divorcing this process from tumorigenesis and unmasking a potential new target for antimetastatic therapies.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Waddington, C. H. The Strategy of the Genes: A Discussion of Some Aspects of Theoretical Biology (Routledge, 2014).

  212. Lee, A. et al. BOADICEA: a comprehensive breast cancer risk prediction model incorporating genetic and nongenetic risk factors. Genet. Med. 21, 1708–1718 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  213. Jamshidi, A. et al. Evaluation of cell-free DNA approaches for multi-cancer early detection. Cancer Cell 40, 1537–1549.e12 (2022).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors are grateful to B. Simons for helpful discussions during writing of the manuscript. R.J.G is supported by Major Centre Core and Children’s Brain Tumour Centre of Excellence grants from Cancer Research UK, The Brain Tumour Charity, and P01CA96832 and U54CA243125 from the US National Cancer Institute.

Author information

Authors and Affiliations

Authors

Contributions

All authors researched data for the article and contributed substantially to discussion of the content. R.J.G. and A.J. wrote the article. All authors reviewed and/or edited the manuscript before submission.

Corresponding author

Correspondence to Richard J. Gilbertson.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Cancer thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

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

Related links

World Cancer Research Fund: https://www.wcrf.org/cancer-trends/worldwide-cancer-data/

Glossary

Apolipoprotein B mRNA-editing enzyme catalytic polypeptide

(APOBEC). An enzyme that edits mRNA species by deaminating cytosine to uracil.

Barrett oesophagus

A precursor condition for oesophageal cancer in which there is an abnormal (metaplastic) change in the mucosal cells lining the lower portion of the oesophagus, from stratified squamous epithelium to simple columnar epithelium.

Blastocysts

Clusters of dividing cells made by a fertilized egg, comprising the early stage of an embryo.

Developmental regulators

Genes that play an important role in the control of normal tissue development.

Dysplasia

The presence of abnormal cells within a tissue that may represent the precursor of malignant change.

Ependymoma

The third most common brain tumour of children arising from radial glia throughout the neural axis.

Internal tandem duplication

Duplication of sections of DNA adjacent to the original sequence.

Medulloblastoma

The most common malignant brain tumour to affect children, arising in the hindbrain from progenitor cells of the upper or lower rhombic lips.

Melanoblast

A neural crest-derived precursor cell of melanocytes, the cells that make pigment in the skin.

Metaplasia

The emergence of new cell types or disproportionate numbers of normal cell types.

Reprogramming factors

Transcription factors including OCT3 and OCT4, SOX2, MYC and KLF4 that can convert a differentiated somatic cell state into a pluripotent embryonic-like state.

Rhombomere

A transiently divided segment of the developing neural tube within the hindbrain.

Telomerase reverse transcriptase

(TERT). Part of a distinct subgroup of RNA-dependent polymerases that lengthen telomeres (the ends of DNA strands).

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jassim, A., Rahrmann, E.P., Simons, B.D. et al. Cancers make their own luck: theories of cancer origins. Nat Rev Cancer 23, 710–724 (2023). https://doi.org/10.1038/s41568-023-00602-5

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-023-00602-5

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