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:

Genetic and non-genetic clonal diversity in cancer evolution

Abstract

The observation and analysis of intra-tumour heterogeneity (ITH), particularly in genomic studies, has advanced our understanding of the evolutionary forces that shape cancer growth and development. However, only a subset of the variation observed in a single tumour will have an impact on cancer evolution, highlighting the need to distinguish between functional and non-functional ITH. Emerging studies highlight a role for the cancer epigenome, transcriptome and immune microenvironment in functional ITH. Here, we consider the importance of both genetic and non-genetic ITH and their role in tumour evolution, and present the rationale for a broad research focus beyond the cancer genome. Systems-biology analytical approaches will be necessary to outline the scale and importance of functional ITH. By allowing a deeper understanding of tumour evolution this will, in time, encourage development of novel therapies and improve outcomes for patients.

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

Access options

Buy this article

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

Fig. 1: Functional and non-functional intra-tumour heterogeneity in tumour evolution.
Fig. 2: Methods of assessing tumour evolution and clonal frequency inference.
Fig. 3: Methods of assessing tumour evolution: dN/dS.
Fig. 4: Tumour evolution may be incorrectly classified using an exclusively genomic approach.
Fig. 5: Comparison of cancer genes defined by the COSMIC Cancer Gene Census and by a systematic pan-cancer whole-exome sequencing mutation-based approach.
Fig. 6: Prognostic impact of intra-tumour heterogeneity.

Similar content being viewed by others

References

  1. Darwin, C. On the Origin of Species by Means of Natural Selection, or the Preservation of Favored Races in the Struggle for Life (J. Murray, 1859).

  2. Nowell, P. C. The clonal evolution of tumor cell populations. Science 194, 23–28 (1976).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Williams, M. J., Werner, B., Graham, T. A. & Sottoriva, A. Functional versus non-functional intratumor heterogeneity in cancer. Mol. Cell. Oncol. 3, e1162897 (2016).

    PubMed  PubMed Central  Google Scholar 

  5. Turajlic, S., Sottoriva, A., Graham, T. & Swanton, C. Resolving genetic heterogeneity in cancer. Nat. Rev. Genet. 20, 404–416 (2019).

    CAS  PubMed  Google Scholar 

  6. Marusyk, A., Janiszewska, M. & Polyak, K. Intratumor heterogeneity: the Rosetta Stone of therapy resistance. Cancer Cell 37, 471–484 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. McGranahan, N., Burrell, R. A., Endesfelder, D., Novelli, M. R. & Swanton, C. Cancer chromosomal instability: therapeutic and diagnostic challenges. EMBO Rep. 13, 528–538 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Coons, S. W., Johnson, P. C. & Shapiro, J. R. Cytogenetic and flow cytometry DNA analysis of regional heterogeneity in a low grade human glioma. Cancer Res. 55, 1569–1577 (1995).

    CAS  PubMed  Google Scholar 

  10. Teixeira, M. R., Pandis, N., Bardi, G., Andersen, J. A. & Heim, S. Karyotypic comparisons of multiple tumorous and macroscopically normal surrounding tissue samples from patients with breast cancer. Cancer Res. 56, 855–859 (1996).

    CAS  PubMed  Google Scholar 

  11. Navin, N. et al. Inferring tumor progression from genomic heterogeneity. Genome Res. 20, 68–80 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Ohta, T. Slightly deleterious mutant substitutions in evolution. Nature 246, 96–98 (1973).

    CAS  PubMed  Google Scholar 

  14. Ling, S. et al. Extremely high genetic diversity in a single tumor points to prevalence of non-Darwinian cell evolution. Proc. Natl. Acad. Sci. 112, E6496–E6505 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Sottoriva, A. et al. A big bang model of human colorectal tumor growth. Nat. Genet. 47, 209–216 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Williams, M. J., Werner, B., Barnes, C. P., Graham, T. A. & Sottoriva, A. Identification of neutral tumor evolution across cancer types. Nat. Genet. 48, 238–244 (2016). This study demonstrates that a subset of tumours evolve without clear evidence of subclonal selection.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Sun, R. et al. Between-region genetic divergence reflects the mode and tempo of tumor evolution. Nat. Genet. 49, 1015–1024 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Cross, W. et al. The evolutionary landscape of colorectal tumorigenesis. Nat. Ecol. Evol. 2, 1661–1672 (2018).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Gao, R. et al. Punctuated copy number evolution and clonal stasis in triple-negative breast cancer. Nat. Genet. 48, 1119–1130 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Notta, F. et al. A renewed model of pancreatic cancer evolution based on genomic rearrangement patterns. Nature 538, 378–382 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Field, M. G. et al. Punctuated evolution of canonical genomic aberrations in uveal melanoma. Nat. Commun. 9, 116 (2018).

    PubMed  PubMed Central  Google Scholar 

  23. Reiter, J. G. et al. An analysis of genetic heterogeneity in untreated cancers. Nat. Rev. Cancer 19, 639–650 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Papaemmanuil, E. et al. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood 122, 3616–3627 quiz 3699 (2013).

  25. Brastianos, P. K. et al. Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets. Cancer Discov. 5, 1164–1177 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Landau, D. A. et al. Mutations driving CLL and their evolution in progression and relapse. Nature 526, 525–530 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Gerstung, M. et al. The evolutionary history of 2,658 cancers. Nature 578, 122–128 (2020). This study, part of the PCAWG, provides an overview of evolutionary patterns across cancer types, identifying different driver events that typically occur early or late in cancer.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Nik-Zainal, S. et al. The life history of 21 breast cancers. Cell 149, 994–1007 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Kessler, D. A. & Levine, H. Large population solution of the stochastic Luria–Delbrück evolution model. Proc. Natl Acad. Sci. USA 110, 11682–11687 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Caravagna, G. et al. Subclonal reconstruction of tumors by using machine learning and population genetics. Nat. Genet. 52, 898–907 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Litchfield, K. et al. Representative sequencing: unbiased sampling of solid tumor tissue. Cell Rep. 31, 107550 (2020).

    CAS  PubMed  Google Scholar 

  33. Durinck, S. et al. Temporal dissection of tumorigenesis in primary cancers. Cancer Discov. 1, 137–143 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  36. López, S. et al. Interplay between whole-genome doubling and the accumulation of deleterious alterations in cancer evolution. Nat. Genet. 52, 283–293 (2020).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  38. Tarabichi, M. et al. Neutral tumor evolution? Nat. Genet. 50, 1630–1633 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Nam, A. S., Chaligne, R. & Landau, D. A. Integrating genetic and non-genetic determinants of cancer evolution by single-cell multi-omics. Nat. Rev. Genet. 22, 3–18 (2020).

    PubMed  PubMed Central  Google Scholar 

  40. Laks, E. et al. Clonal decomposition and DNA replication states defined by scaled single-cell genome sequencing. Cell 179, 1207–1221.e22 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Cross, W. et al. Stabilising selection causes grossly altered but stable karyotypes in metastatic colorectal cancer. bioRxiv https://doi.org/10.1101/2020.03.26.007138 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  46. Watkins, T. B. K. et al. Pervasive chromosomal instability and karyotype order in tumour evolution. Nature 587, 126–132 (2020). This multi-region, pan-cancer analysis of tumour karyotype uncovers parallel evolution of events within different subclones in one-third of tumours sampled, and identifies the important role of chromosomal instability in generating subclonal diversity in cancer.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Bolhaqueiro, A. C. F. et al. Ongoing chromosomal instability and karyotype evolution in human colorectal cancer organoids. Nat. Genet. 51, 824–834 (2019).

    CAS  PubMed  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  49. Clarke, T. L. et al. Histone lysine methylation dynamics control EGFR DNA copy-number amplification. Cancer Discov. 10, 306–325 (2020).

    CAS  PubMed  Google Scholar 

  50. Chen, H. et al. A pan-cancer analysis of enhancer expression in nearly 9000 patient samples. Cell 173, 386–399.e12 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Gröbner, S. N. et al. The landscape of genomic alterations across childhood cancers. Nature 555, 321–327 (2018).

    PubMed  Google Scholar 

  52. Kahles, A. et al. Comprehensive analysis of alternative splicing across tumors from 8,705 patients. Cancer Cell 34, 211–224.e6 (2018).

    CAS  PubMed  Google Scholar 

  53. Shiraishi, Y. et al. A comprehensive characterization of cis-acting splicing-associated variants in human cancer. Genome Res. 28, 1111–1125 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Demircioğlu, D. et al. A pan-cancer transcriptome analysis reveals pervasive regulation through alternative promoters. Cell 178, 1465–1477.e17 (2019).

    PubMed  Google Scholar 

  55. Calabrese, C. et al. Genomic basis for RNA alterations in cancer. Nature 578, 129–136 (2020). This pan-cancer study of paired whole genomes and transcriptomes illustrates the variety of transcriptomic alterations in cancer, and underlines the influence of copy number events and non-coding mutations on gene expression.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Zhang, Y. et al. High-coverage whole-genome analysis of 1220 cancers reveals hundreds of genes deregulated by rearrangement-mediated cis-regulatory alterations. Nat. Commun. 11, 736 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Baysal, B. E., Sharma, S., Hashemikhabir, S. & Janga, S. C. RNA editing in pathogenesis of cancer. Cancer Res. 77, 3733–3739 (2017).

    CAS  PubMed  Google Scholar 

  58. Chen, L. et al. Recoding RNA editing of AZIN1 predisposes to hepatocellular carcinoma. Nat. Med. 19, 209–216 (2013).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Sondka, Z. et al. The COSMIC Cancer Gene Census: describing genetic dysfunction across all human cancers. Nat. Rev. Cancer 18, 696–705 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Carter, S. L., Eklund, A. C., Kohane, I. S., Harris, L. N. & Szallasi, Z. A signature of chromosomal instability inferred from gene expression profiles predicts clinical outcome in multiple human cancers. Nat. Genet. 38, 1043–1048 (2006).

    CAS  PubMed  Google Scholar 

  62. Biswas, D. et al. A clonal expression biomarker associates with lung cancer mortality. Nat. Med. 25, 1540–1548 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Salami, S. S., Tomlins, S. A. & Palapattu, G. S. Transcriptomic heterogeneity in multifocal prostate cancer. JCI Insight 3, e123468 (2018).

    PubMed Central  Google Scholar 

  64. Thomsen, M. B. H. et al. Comprehensive multiregional analysis of molecular heterogeneity in bladder cancer. Sci. Rep. 7, 11702 (2017).

    PubMed  PubMed Central  Google Scholar 

  65. Aran, D., Sirota, M. & Butte, A. J. Systematic pan-cancer analysis of tumour purity. Nat. Commun. 6, 8971 (2015).

    CAS  PubMed  Google Scholar 

  66. Wang, F. et al. Integrated transcriptomic–genomic tool Texomer profiles cancer tissues. Nat. Methods 16, 401–404 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Teschendorff, A. E. & Enver, T. Single-cell entropy for accurate estimation of differentiation potency from a cell’s transcriptome. Nat. Commun. 8, 15599 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Marjanovic, N. D. et al. Emergence of a high-plasticity cell state during lung cancer evolution. Cancer Cell 38, 229–246 (2020). This study of a mouse model of lung adenocarcinoma illustrates that highly plastic stem-like cells with diverse transcriptional states drive resistance to therapy and poor clinical outcome.

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Lafave, L. M. et al. Epigenomic state transitions characterize tumor progression in mouse lung adenocarcinoma. Cancer Cell 38, 212–228.e13 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Izzo, F. et al. DNA methylation disruption reshapes the hematopoietic differentiation landscape. Nat. Genet. 52, 378–387 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Raphael, B. J. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell 32, 185–203.e13 (2017).

    Google Scholar 

  72. McDonald, O. G. et al. Epigenomic reprogramming during pancreatic cancer progression links anabolic glucose metabolism to distant metastasis. Nat. Genet. 49, 367–376 (2017). This study of pancreatic cancer is an example of the important potential role of non-genetic variation in cancer evolution.

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Pastore, A. et al. Corrupted coordination of epigenetic modifications leads to diverging chromatin states and transcriptional heterogeneity in CLL. Nat. Commun. 10, 1874 (2019).

    PubMed  PubMed Central  Google Scholar 

  74. Hua, X. et al. Genetic and epigenetic intratumor heterogeneity impacts prognosis of lung adenocarcinoma. Nat. Commun. 11, 2459 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Zhu, B. et al. The genomic and epigenomic evolutionary history of papillary renal cell carcinomas. Nat. Commun. 11, 3096 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Mertins, P. et al. Proteogenomics connects somatic mutations to signalling in breast cancer. Nature 534, 55–62 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Zhang, H. et al. Integrated proteogenomic characterization of human high-grade serous ovarian cancer. Cell 166, 755–765 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Mun, D.-G. et al. Proteogenomic characterization of human early-onset gastric cancer. Cancer Cell 35, 111–124.e10 (2019).

    CAS  PubMed  Google Scholar 

  79. Chen, Y. J. et al. Proteogenomics of non-smoking lung cancer in east Asia delineates molecular signatures of pathogenesis and progression. Cell 182, 226–244.e17 (2020).

    CAS  PubMed  Google Scholar 

  80. Gillette, M. A. et al. Proteogenomic characterization reveals therapeutic vulnerabilities in lung adenocarcinoma. Cell 182, 200–225.e35 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Xu, J.-Y. et al. Integrative proteomic characterization of human lung adenocarcinoma. Cell 182, 245–261.e17 (2020).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  83. Marty, R. et al. MHC-I genotype restricts the oncogenic mutational landscape. Cell 171, 1272–1283.e15 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Marty Pyke, R. et al. Evolutionary pressure against MHC class II binding cancer mutations. Cell 175, 416–428.e13 (2018).

    PubMed  Google Scholar 

  85. DuPage, M., Mazumdar, C., Schmidt, L. M., Cheung, A. F. & Jacks, T. Expression of tumour-specific antigens underlies cancer immunoediting. Nature 482, 405–409 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Rooney, M. S., Shukla, S. A., Wu, C. J., Getz, G. & Hacohen, N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell 160, 48–61 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Anagnostou, V. et al. Evolution of neoantigen landscape during immune checkpoint blockade in non-small cell lung cancer. Cancer Discov. 7, 264–276 (2017).

    CAS  PubMed  Google Scholar 

  89. Rosenthal, R. et al. Neoantigen-directed immune escape in lung cancer evolution. Nature 567, 479–485 (2019). This work highlights the role of immune editing in shaping early cancer evolution by negative selection, as well as the diversity of mechanisms of immune evasion.

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Jiménez-Sánchez, A. et al. Unraveling tumor–immune heterogeneity in advanced ovarian cancer uncovers immunogenic effect of chemotherapy. Nat. Genet. 52, 582–593 (2020).

    PubMed  PubMed Central  Google Scholar 

  91. Davoli, T., Uno, H., Wooten, E. C. & Elledge, S. J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 355, eaaf8399 (2017).

    PubMed  PubMed Central  Google Scholar 

  92. McGranahan, N. et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 351, 1463–1470 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Joshi, K. et al. Spatial heterogeneity of the T cell receptor repertoire reflects the mutational landscape in lung cancer. Nat. Med. 25, 1549–1559 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Roh, W. et al. Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance. Sci. Transl. Med. 9, eaah3560 (2017).

    PubMed  PubMed Central  Google Scholar 

  95. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Snyder, A. et al. Contribution of systemic and somatic factors to clinical response and resistance to PD-L1 blockade in urothelial cancer: an exploratory multi-omic analysis. PLOS Med. 14, e1002309 (2017).

    PubMed  PubMed Central  Google Scholar 

  97. Wolf, Y. et al. UVB-induced tumor heterogeneity diminishes immune response in melanoma. Cell 179, 219–235.e21 (2019). In this study, a mouse model of melanoma illustrates that increased clonal diversity of a developing tumour is associated with evasion of the anticancer immune response.

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Gejman, R. S. et al. Rejection of immunogenic tumor clones is limited by clonal fraction. eLife 7, e41090 (2018).

    PubMed  PubMed Central  Google Scholar 

  99. Maire, C. L. et al. Glioma escape signature and clonal development under immune pressure. J. Clin. Invest. 130, 5257–5271 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Zhang, A. W. et al. Interfaces of malignant and immunologic clonal dynamics in ovarian cancer. Cell 173, 1755–1769.e22 (2018).

    CAS  PubMed  Google Scholar 

  101. Angelova, M. et al. Evolution of metastases in space and time under immune selection. Cell 175, 751–765.e16 (2018).

    CAS  PubMed  Google Scholar 

  102. Pennycuick, A. et al. Immune surveillance in clinical regression of pre-invasive squamous cell lung cancer. Cancer Discov. 10, 1489–1499 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Zapata, L. et al. Negative selection in tumor genome evolution acts on essential cellular functions and the immunopeptidome. Genome Biol. 19, 67 (2018).

    PubMed  PubMed Central  Google Scholar 

  104. Eynden, J. Van Den, Jiménez-sánchez, A., Miller, M. L. & Larsson, E. Lack of detectable neoantigen depletion signals in the untreated cancer genome. Nat. Genet. 51, 1741–1748 (2019).

    PubMed  PubMed Central  Google Scholar 

  105. Zapata, L. et al. dN/dS dynamics quantify tumour immunogenicity and predict response to immunotherapy. bioRxiv https://doi.org/10.1101/2020.07.21.215038 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  106. Ghorani, E. et al. The T cell differentiation landscape is shaped by tumour mutations in lung cancer. Nat. Cancer 1, 546–561 (2020).

    PubMed  PubMed Central  Google Scholar 

  107. Keren, L. et al. A structured tumor-immune microenvironment in triple negative breast cancer revealed by multiplexed ion beam imaging. Cell 174, 1373–1387.e19 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Abduljabbar, K. et al. Geospatial immune variability illuminates differential evolution of lung adenocarcinoma. Nat. Med. 26, 1054–1062 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Wang, M. et al. Role of tumor microenvironment in tumorigenesis. J. Cancer 8, 761–773 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Korenchan, D. E. & Flavell, R. R. Spatiotemporal pH heterogeneity as a promoter of cancer progression and therapeutic resistance. Cancers 11, 1026 (2019).

    CAS  PubMed Central  Google Scholar 

  111. Lloyd, M. C. et al. Darwinian dynamics of intratumoral heterogeneity: not solely random mutations but also variable environmental selection forces. Cancer Res. 76, 3136–3144 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Hoefflin, R. et al. Spatial niche formation but not malignant progression is a driving force for intratumoural heterogeneity. Nat. Commun. 7, 11845 (2016).

    Google Scholar 

  113. Lujambio, A. et al. Non-cell-autonomous tumor suppression by p53. Cell 153, 449–460 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Peinado, H. et al. Pre-metastatic niches: organ-specific homes for metastases. Nat. Rev. Cancer 17, 302–317 (2017).

    CAS  PubMed  Google Scholar 

  115. Montagner, M. et al. Crosstalk with lung epithelial cells regulates Sfrp2-mediated latency in breast cancer dissemination. Nat. Cell Biol. 22, 289–296 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Marusyk, A. et al. Non-cell-autonomous driving of tumour growth supports sub-clonal heterogeneity. Nature 514, 54–58 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Schürch, C. M. et al. Coordinated cellular neighborhoods orchestrate antitumoral immunity at the colorectal cancer invasive front. Cell 182, 1341–1359.e19 (2020).

    PubMed  PubMed Central  Google Scholar 

  118. Wagner, J. et al. A single-cell atlas of the tumor and immune ecosystem of human breast cancer. Cell 177, 1330–1345.e18 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Ali, H. R. et al. Imaging mass cytometry and multiplatform genomics define the phenogenomic landscape of breast cancer. Nat. Cancer 1, 163–175 (2020).

    PubMed  Google Scholar 

  120. Jackson, H. W. et al. The single-cell pathology landscape of breast cancer. Nature 578, 615–620 (2020).

    CAS  PubMed  Google Scholar 

  121. Birkbak, N. J. & McGranahan, N. Cancer genome evolutionary trajectories in metastasis. Cancer Cell 37, 8–19 (2020).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Yates, L. R. et al. Genomic evolution of breast cancer metastasis and relapse. Cancer Cell 32, 169–184.e7 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Hu, Z., Li, Z., Ma, Z. & Curtis, C. Multi-cancer analysis of clonality and the timing of systemic spread in paired primary tumors and metastases. Nat. Genet. 52, 701–708 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Reiter, J. G. et al. Lymph node metastases develop through a wider evolutionary bottleneck than distant metastases. Nat. Genet. 52, 692–700 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. El-Kebir, M., Satas, G. & Raphael, B. J. Inferring parsimonious migration histories for metastatic cancers. Nat. Genet. 50, 718–726 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Reiter, J. G. et al. Minimal functional driver gene heterogeneity among untreated metastases. Science 361, 1033–1037 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Rabbie, R. et al. Multi-site clonality analysis uncovers pervasive heterogeneity across melanoma metastases. Nat. Commun. 11, 4306 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03004755 (2019).

  130. Parikh, A. R. et al. Liquid versus tissue biopsy for detecting acquired resistance and tumor heterogeneity in gastrointestinal cancers. Nat. Med. 25, 1415–1421 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Noorani, A. et al. Genomic evidence supports a clonal diaspora model for metastases of esophageal adenocarcinoma. Nat. Genet. 52, 74–83 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Lo, H. C. et al. Resistance to natural killer cell immunosurveillance confers a selective advantage to polyclonal metastasis. Nat. Cancer 1, 709–722 (2020).

    PubMed  Google Scholar 

  133. Landau, D. A. et al. Evolution and impact of subclonal mutations in chronic lymphocytic leukemia. Cell 152, 714–726 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Andor, N. et al. Pan-cancer analysis of the extent and consequences of intratumor heterogeneity. Nat. Med. 22, 105–113 (2016).

    CAS  PubMed  Google Scholar 

  135. Rye, I. H. et al. Intratumor heterogeneity defines treatment-resistant HER2+ breast tumors. Mol. Oncol. 12, 1838–1855 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Turajlic, S. et al. Deterministic evolutionary trajectories influence primary tumor growth: TRACERx renal. Cell 173, 595–610.e11 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Cerrano, M. et al. Prognostic impact of clonal diversity in acute myeloid leukemia (AML) treated with intensive chemotherapy (IC). Blood 134, 2700 (2019).

    Google Scholar 

  138. Iacobuzio-Donahue, C. A., Litchfield, K. & Swanton, C. Intratumor heterogeneity reflects clinical disease course. Nat. Cancer 1, 3–6 (2020).

    PubMed  Google Scholar 

  139. Martinez, P. et al. Dynamic clonal equilibrium and predetermined cancer risk in Barrett’s oesophagus. Nat. Commun. 7, 12158 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Wei, S. C., Duffy, C. R. & Allison, J. P. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 8, 1069–1086 (2018).

    PubMed  Google Scholar 

  141. Birkbak, N. J. et al. Paradoxical relationship between chromosomal instability and survival outcome in cancer. Cancer Res. 71, 3447–3452 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Jamal-Hanjani, M. et al. Extreme chromosomal instability forecasts improved outcome in ER-negative breast cancer: a prospective validation cohort study from the TACT trial. Ann. Oncol. 26, 1340–1346 (2015).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  144. Moore, L. et al. The mutational landscape of normal human endometrial epithelium. Nature 580, 640–646 (2020).

    CAS  PubMed  Google Scholar 

  145. Killcoyne, S. et al. Genomic copy number predicts esophageal cancer years before transformation. Nat. Med. 26, 1726–1732 (2020).

    PubMed  PubMed Central  Google Scholar 

  146. Noble, R. & Burley, J. T. When, why and how tumour clonal diversity predicts survival. Evol. Appl. 13, 1558–1568 (2020).

    PubMed  PubMed Central  Google Scholar 

  147. Riaz, N. et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell 171, 934–949.e16 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Pearson, A. et al. High-level clonal FGFR amplification and response to FGFR inhibition in a translational clinical trial. Cancer Discov. 6, 838–851 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Russo, M. et al. Adaptive mutability of colorectal cancers in response to targeted therapies. Science 366, 1473–1480 (2019).

    CAS  PubMed  Google Scholar 

  150. Mok, T. S. et al. Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer. N. Engl. J. Med. 376, 629–640 (2017).

    CAS  PubMed  Google Scholar 

  151. Oldrini, B. et al. MGMT genomic rearrangements contribute to chemotherapy resistance in gliomas. Nat. Commun. 11, 3883 (2020).

    PubMed  PubMed Central  Google Scholar 

  152. Bell, C. C. et al. Targeting enhancer switching overcomes non-genetic drug resistance in acute myeloid leukaemia. Nat. Commun. 10, 2723 (2019).

    PubMed  PubMed Central  Google Scholar 

  153. Fong, C. Y. et al. BET inhibitor resistance emerges from leukaemia stem cells. Nature 525, 538–542 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Hinohara, K. et al. KDM5 histone demethylase activity links cellular transcriptomic heterogeneity to therapeutic resistance. Cancer Cell 34, 939–953.e9 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. McGranahan, N. & Swanton, C. Neoantigen quality, not quantity. Sci. Transl. Med. 11, eaax7918 (2019).

    PubMed  Google Scholar 

  156. Rambow, F. et al. Toward minimal residual disease-directed therapy in melanoma. Cell 174, 843–855.e19 (2018). This work highlights the role played by stem-like cancer cells in non-genetic mechanisms of resistance to cancer therapy.

    CAS  PubMed  Google Scholar 

  157. Pleasance, E. et al. Pan-cancer analysis of advanced patient tumors reveals interactions between therapy and genomic landscapes. Nat. Cancer 1, 452–468 (2020).

    PubMed  Google Scholar 

  158. Gao, J. et al. Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell 167, 397–404.e9 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Sahin, U. et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 547, 222–226 (2017).

    CAS  PubMed  Google Scholar 

  160. Siravegna, G. et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat. Med. 21, 795–801 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Xue, Y. et al. An approach to suppress the evolution of resistance in BRAF(V600E)-mutant cancer. Nat. Med. 23, 929–937 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Gopal, P. et al. Clonal selection confers distinct evolutionary trajectories in BRAF-driven cancers. Nat. Commun. 10, 5143 (2019).

    PubMed  PubMed Central  Google Scholar 

  163. Acar, A. et al. Exploiting evolutionary steering to induce collateral drug sensitivity in cancer. Nat. Commun. 11, 1923 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Algazi, A. et al. Abstract CT013: SWOG S1320: improved progression-free survival with continuous compared to intermittent dosing with dabrafenib and trametinib in patients with BRAF mutated melanoma. Cancer Res. 80, CT013 LP–CT013 (2020).

    Google Scholar 

Download references

Acknowledgements

The authors thank C. Bailey, A. Frankell, E. Colliver, C.-M. Ruiz and J. Demeulemeester for their thoughtful review of the manuscript. N.M is a Sir Henry Dale Fellow, jointly funded by the Wellcome Trust and the Royal Society (Grant Number 211179/Z/18/Z), and also receives funding from Cancer Research UK Lung Cancer Centre of Excellence, Rosetrees, and the NIHR BRC at University College London Hospitals.

Author information

Authors and Affiliations

Authors

Contributions

J.R.M.B. and N.M. both researched data for the article and made a substantial contribution to discussion of content, writing, reviewing and editing the article.

Corresponding author

Correspondence to Nicholas McGranahan.

Ethics declarations

Competing interests

The authors declare no competing interests. N.M. has received consultancy fees and has stock options in Achilles Therapeutics. N.M. holds European patents relating to targeting neoantigens (PCT/EP2016/ 059401), identifying patient response to immune checkpoint blockade (PCT/ EP2016/071471), determining HLA LOH (PCT/GB2018/052004), predicting survival rates of patients with cancer (PCT/GB2020/050221). J.R.M.B. declares no competing interests.

Additional information

Peer review information

Nature Reviews Cancer thanks M. Gerstung 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.

Supplementary information

Glossary

Intra-tumour heterogeneity

(ITH). Variation within the same tumour; this can be non-functional, a result of neutral evolution, or functional, leading to selection that shapes ongoing tumour evolution.

Chromosomal instability

(CIN). A defect in which cells can gain, lose or rearrange parts of chromosomes or whole chromosomes during cell division; this is a source of variation in cancer.

Chromothripsis

A mutational process in which large numbers of clustered structural rearrangements occur in single or multiple chromosomes.

Molecular time

An estimate of the timing of an event, from the first cell division following fertilization to a cell division that occurred only recently before sampling.

Enhancer

A short genomic region that influences the expression of another gene in cis.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Black, J.R.M., McGranahan, N. Genetic and non-genetic clonal diversity in cancer evolution. Nat Rev Cancer 21, 379–392 (2021). https://doi.org/10.1038/s41568-021-00336-2

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-021-00336-2

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