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.

  • Perspective
  • Published:

Tumour heterogeneity and the evolutionary trade-offs of cancer

Abstract

Tumours vary in gene expression programmes and genetic alterations. Understanding this diversity and its biological meaning requires a theoretical framework, which could in turn guide the development of more accurate prognosis and therapy. Here, we review the theory of multi-task evolution of cancer, which is based upon the premise that tumours evolve in the host and face selection trade-offs between multiple biological functions. This theory can help identify the major biological tasks that cancer cells perform and the trade-offs between these tasks. It introduces the concept of specialist tumours, which focus on one task, and generalist tumours, which perform several tasks. Specialist tumours are suggested to be sensitive to therapy targeting their main task. Driver mutations tune gene expression towards specific tasks in a tissue-dependent manner and thus help to determine whether a tumour is specialist or generalist. We discuss potential applications of the theory of multi-task evolution to interpret the spatial organization of tumours and intratumour heterogeneity.

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: Evolutionary trade-offs constrain optimal traits (gene expression, morphology) to the geometry of polyhedra.
Fig. 2: ParTI infers tasks and trade-offs from data.
Fig. 3: Spatial gradients and maximization of combined tissue performance creates continua in gene expression.

Similar content being viewed by others

References

  1. Cancer Genome Atlas Research Network et al. Integrated genomic characterization of papillary thyroid carcinoma. Cell 159, 676–690 (2014).

    Google Scholar 

  2. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 490, 61–70 (2012).

    Google Scholar 

  3. Curtis, C. et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature 486, 346–352 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Eirew, P. et al. Dynamics of genomic clones in breast cancer patient xenografts at single-cell resolution. Nature 518, 422–426 (2015).

    CAS  PubMed  Google Scholar 

  5. 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 

  6. Kim, C. et al. Chemoresistance evolution in triple-negative breast cancer delineated by single-cell sequencing. Cell 173, 879–893.e13 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Li, H. et al. Reference component analysis of single-cell transcriptomes elucidates cellular heterogeneity in human colorectal tumors. Nat. Genet. 49, 708–718 (2017).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  10. Navin, N. et al. Tumour evolution inferred by single-cell sequencing. Nature 472, 90–94 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Venteicher, A. S. et al. Decoupling genetics, lineages, and microenvironment in IDH-mutant gliomas by single-cell RNA-seq. Science 355, eaai8478 (2017).

    PubMed  PubMed Central  Google Scholar 

  13. Hu, Z., Sun, R. & Curtis, C. A population genetics perspective on the determinants of intra-tumor heterogeneity. Biochim. Biophys. Acta Rev. Cancer 1867, 109–126 (2017).

    CAS  PubMed  Google Scholar 

  14. Lipinski, K. A. et al. Cancer evolution and the limits of predictability in precision cancer medicine. Trends Cancer 2, 49–63 (2016).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Nagpal, K. et al. Development and validation of a deep learning algorithm for improving Gleason scoring of prostate cancer. NPJ Digit. Med. 2, 48 (2019).

    PubMed  PubMed Central  Google Scholar 

  17. Maley, C. C. et al. Genetic clonal diversity predicts progression to esophageal adenocarcinoma. Nat. Genet. 38, 468–473 (2006).

    CAS  PubMed  Google Scholar 

  18. Maley, C. C., Koelble, K., Natrajan, R., Aktipis, A. & Yuan, Y. An ecological measure of immune-cancer colocalization as a prognostic factor for breast cancer. Breast Cancer Res. 17, 131 (2015).

    PubMed  PubMed Central  Google Scholar 

  19. Mobadersany, P. et al. Predicting cancer outcomes from histology and genomics using convolutional networks. Proc. Natl Acad. Sci. USA 115, E2970–E2979 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Robertson, S., Azizpour, H., Smith, K. & Hartman, J. Digital image analysis in breast pathology — from image processing techniques to artificial intelligence. Transl. Res. 194, 19–35 (2018).

    PubMed  Google Scholar 

  21. Sørlie, T. et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc. Natl Acad. Sci. USA 98, 10869–10874 (2001).

    PubMed  PubMed Central  Google Scholar 

  22. Beerenwinkel, N., Greenman, C. D. & Lageren, J. Computational cancer biology: an evolutionary perspective. PLoS Comput. Biol. 12, e1004717 (2016).

    PubMed  PubMed Central  Google Scholar 

  23. Gatenby, R. A. & Brown, J. Mutations, evolution and the central role of a self-defined fitness function in the initiation and progression of cancer. Biochim. Biophys. Acta Rev. Cancer 1867, 162–166 (2017).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  26. Sottoriva, A., Barnes, C. P. & Graham, T. A. Catch my drift? Making sense of genomic intra-tumour heterogeneity. Biochim. Biophys. Acta Rev. Cancer 1867, 95–100 (2017).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Schwartz, R. & Schäffer, A. A. The evolution of tumour phylogenetics: principles and practice. Nat. Rev. Genet. 18, 213–229 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. 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 

  30. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Gillespie, J. H. Population Genetics: A Concise Guide (Johns Hopkins University Press, 2004).

  32. McDonald, T. O., Chakrabarti, S. & Michor, F. Currently available bulk sequencing data do not necessarily support a model of neutral tumor evolution. Nat. Genet. 50, 1620–1623 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. 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 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Waclaw, B. et al. A spatial model predicts that dispersal and cell turnover limit intratumour heterogeneity. Nature 525, 261–264 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Alon, U. An Introduction to Systems Biology: Design Principles of Biological Circuits (CRC Press, 2019).

  37. Shoval, O. et al. Evolutionary trade-offs, pareto optimality, and the geometry of phenotype space. Science 336, 1157–1160 (2012).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  39. Aktipis, C. A., Boddy, A. M., Gatenby, R. A., Brown, J. S. & Maley, C. C. Life history trade-offs in cancer evolution. Nat. Rev. Cancer 13, 883–892 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Gallaher, J. A., Brown, J. S. & Anderson, A. R. A. The impact of proliferation-migration tradeoffs on phenotypic evolution in cancer. Sci. Rep. 9, 1–10 (2019).

    Google Scholar 

  41. Gillies, R. J., Brown, J. S., Anderson, A. R. A. & Gatenby, R. A. Eco-evolutionary causes and consequences of temporal changes in intratumoural blood flow. Nat. Rev. Cancer 18, 576–585 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Hatzikirou, H., Basanta, D., Simon, M., Schaller, K. & Deutsch, A. ‘Go or grow’: the key to the emergence of invasion in tumour progression? Math. Med. Biol. 29, 49–65 (2012).

    CAS  PubMed  Google Scholar 

  43. Broxterman, H. J. et al. Induction by verapamil of a rapid increase in ATP consumption in multidrug-resistant tumor cells. FASEB J. 2, 2278–2282 (1988).

    CAS  PubMed  Google Scholar 

  44. Jerby, L. et al. Metabolic associations of reduced proliferation and oxidative stress in advanced breast cancer. Cancer Res. 72, 5712–5720 (2012).

    CAS  PubMed  Google Scholar 

  45. Gade, T. P. F. et al. Ischemia induces quiescence and autophagy dependence in hepatocellular carcinoma. Radiology 283, 702–710 (2017).

    PubMed  Google Scholar 

  46. Wang, X. et al. Exit from quiescence displays a memory of cell growth and division. Nat. Commun. 8, 321 (2017).

    PubMed  PubMed Central  Google Scholar 

  47. Evdokimova, V., Tognon, C., Ng, T. & Sorensen, P. H. Reduced proliferation and enhanced migration: two sides of the same coin? Molecular mechanisms of metastatic progression by YB-1. Cell Cycle 8, 2901–2906 (2009).

    CAS  PubMed  Google Scholar 

  48. Tsai, J. H. & Yang, J. Epithelial-mesenchymal plasticity in carcinoma metastasis. Genes Dev. 27, 2192–2206 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Orlando, P. A., Gatenby, R. A. & Brown, J. S. Tumor evolution in space: the effects of competition colonization tradeoffs on tumor invasion dynamics. Front. Oncol. 3, 45 (2013).

    PubMed  PubMed Central  Google Scholar 

  50. Adler, M. et al. Continuum of gene-expression profiles provides spatial division of labor within a differentiated cell type article continuum of gene-expression profiles provides spatial division of labor within a differentiated cell type. Cell Syst. 8, 43–52.e5 (2019).

    CAS  PubMed  Google Scholar 

  51. Hart, Y. et al. Inferring biological tasks using Pareto analysis of high-dimensional data. Nat. Methods 12, 233–235 (2015).

    CAS  PubMed  Google Scholar 

  52. Korem, Y. et al. Geometry of the gene expression space of individual cells. PLoS Comput. Biol. 11, e1004224 (2015).

    PubMed  PubMed Central  Google Scholar 

  53. Szekely, P., Sheftel, H., Mayo, A. & Alon, U. Evolutionary tradeoffs between economy and effectiveness in biological homeostasis systems. PLoS Comput. Biol. 9, e1003163 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Tendler, A., Mayo, A. & Alon, U. Evolutionary tradeoffs, Pareto optimality and the morphology of ammonite shells. BMC Syst. Biol. 9, 12 (2015).

    PubMed  PubMed Central  Google Scholar 

  55. Szekely, P., Korem, Y., Moran, U., Mayo, A. & Alon, U. The mass-longevity triangle: pareto optimality and the geometry of life-history trait space. PLoS Comput. Biol. 11, 1–19 (2015).

    Google Scholar 

  56. Hausser, J. et al. Tumor diversity and the trade-off between universal cancer tasks. Nat. Commun. 10, 5423 (2019).

    PubMed  PubMed Central  Google Scholar 

  57. Trink, A. et al. Geometry of gene expression space of Wilms’ tumors from human patients. Neoplasia 20, 871–881 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Cutler, A. A. & Breiman, L. Archetypial analysis. Technometrics 36, 338–347 (1994).

    Google Scholar 

  59. Mørup, M. & Hansen, L. K. Archetypal analysis for machine learning and data mining. Neurocomputing 80, 54–63 (2012).

    Google Scholar 

  60. Aubert, O. et al. Archetype analysis identifies distinct profiles in renal transplant recipients with transplant glomerulopathy associated with allograft survival. J. Am. Soc. Nephrol. 30, 625–639 (2019).

    PubMed  PubMed Central  Google Scholar 

  61. Dai, Z. et al. Identification of cancer-associated metabolic vulnerabilities by modeling multi-objective optimality in metabolism. Cell Commun. Signal. 17, 124 (2019).

    PubMed  PubMed Central  Google Scholar 

  62. Thøgersen, J. C., Mørup, M., Damkiær, S., Molin, S. & Jelsbak, L. Archetypal analysis of diverse Pseudomonas aeruginosa transcriptomes reveals adaptation in cystic fibrosis airways. BMC Bioinformatics 14, 279 (2013).

    PubMed  PubMed Central  Google Scholar 

  63. van Dijk, D. et al. Recovering gene interactions from single-cell data using data diffusion. Cell 174, 716–729.e27 (2018).

    PubMed  PubMed Central  Google Scholar 

  64. Wei, S. C. et al. Negative co-stimulation constrains T cell differentiation by imposing boundaries on possible cell states. Immunity 50, 1084–1098.e10 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Arnold, S. J. Morphology, performance and fitness. Am. Zool. 23, 347–361 (1983).

    Google Scholar 

  66. Basan, M. et al. Overflow metabolism in Escherichia coli results from efficient proteome allocation. Nature 528, 99–104 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Scott, M., Gunderson, C. W., Mateescu, E. M., Zhang, Z. & Hwa, T. Interdependence of cell growth and gene expression: origins and consequences. Science 330, 1099–1102 (2010).

    CAS  PubMed  Google Scholar 

  68. Towbin, B. D. et al. Optimality and sub-optimality in a bacterial growth law. Nat. Commun. 8, 14123 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Reding-Roman, C. et al. The unconstrained evolution of fast and efficient antibiotic-resistant bacterial genomes. Nat. Ecol. Evol. https://doi.org/10.1038/s41559-016-0050 (2017).

    Article  PubMed  Google Scholar 

  70. Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).

    Google Scholar 

  71. Rueffler, C., Hermisson, J. & Wagner, G. P. Evolution of functional specialization and division of labor. Proc. Natl Acad. Sci. USA 109, E326–E335 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Sheftel, H., Shoval, O., Mayo, A. & Alon, U. The geometry of the Pareto front in biological phenotype space. Ecol. Evol. 3, 1471–1483 (2013).

    PubMed  PubMed Central  Google Scholar 

  73. Gillies, R. J., Verduzco, D. & Gatenby, R. A. Evolutionary dynamics of carcinogenesis and why targeted therapy does not work. Nat. Rev. Cancer 12, 487–493 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Halpern, K. B. et al. Single-cell spatial reconstruction reveals global division of labour in the mammalian liver. Nature 542, 352–356 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Gatenby, R. A. & Gillies, R. J. Why do cancers have high aerobic glycolysis? Nat. Rev. Cancer 4, 891–899 (2004).

    CAS  PubMed  Google Scholar 

  76. Sonveaux, P. et al. Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice. J. Clin. Invest. 118, 3930–3942 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Kumar, S. et al. Intra-tumoral metabolic zonation and resultant phenotypic diversification are dictated by blood vessel proximity. Cell Metab. 30, 201–211.e6 (2019).

    CAS  PubMed  Google Scholar 

  78. Dewhirst, M. W. & Secomb, T. W. Transport of drugs from blood vessels to tumour tissue. Nat. Rev. Cancer 17, 738–750 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. McGranahan, N. et al. Clonal status of actionable driver events and the timing of mutational processes in cancer evolution. Sci. Transl. Med. 7, 283ra54 (2015).

    PubMed  PubMed Central  Google Scholar 

  80. Bioucas-Dias, J. A Variable Splitting Augmented Lagrangian Approach to Linear Spectral Unmixing. (IEEE, 2009). https://doi.org/10.1109/WHISPERS.2009.5289072

  81. Kanehisa, M., Sato, Y., Furumichi, M., Morishima, K. & Tanabe, M. New approach for understanding genome variations in KEGG. Nucleic Acids Res. 47, D590–D595 (2019).

    CAS  PubMed  Google Scholar 

  82. Fabregat, A. et al. The reactome pathway knowledgebase. Nucleic Acids Res. 46, D649–D655 (2018).

    CAS  PubMed  Google Scholar 

  83. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. van Dijk, D. et al. Finding Archetypal Spaces for Data Using Neural Networks (Cornell University, 2019).

  85. Laplante, M. & Sabatini, D. M. mTOR signaling in growth control and disease. Cell 149, 274–293 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Azizi, E. et al. Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell 174, 1293–1308 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Crosetto, N., Bienko, M. & van Oudenaarden, A. Spatially resolved transcriptomics and beyond. Nat. Rev. Genet. 16, 57–66 (2015).

    CAS  PubMed  Google Scholar 

  88. Lein, E., Borm, L. E. & Linnarsson, S. The promise of spatial transcriptomics for neuroscience in the era of molecular cell typing. Science 358, 64–69 (2017).

    CAS  PubMed  Google Scholar 

  89. Moor, A. E. et al. Global mRNA polarization regulates translation efficiency in the intestinal epithelium. Science 357, 1299–1303 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Stahl, P. L. et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science 353, 78–82 (2016).

    CAS  PubMed  Google Scholar 

  92. Wang, H. A. et al. Fast chemical imaging at high spatial resolution by laser ablation inductively coupled plasma mass spectrometry. Anal. Chem. 85, 10107–10116 (2013).

    CAS  PubMed  Google Scholar 

  93. Ke, R. et al. In situ sequencing for RNA analysis in preserved tissue and cells. Nat. Methods 10, 857–860 (2013).

    CAS  PubMed  Google Scholar 

  94. Chen, K. H., Boettiger, A. N., Moffitt, J. R., Wang, S. & Zhuang, X. Spatially resolved, highly multiplexed RNA profiling in single cells. Science 348, aaa6090 (2015).

    PubMed  PubMed Central  Google Scholar 

  95. Eng, C. L. et al. Transcriptome-scale super-resolved imaging in tissues by RNA seqFISH. Nature 568, 235–239 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Nagrath, D. et al. Integrated energy and flux balance based multiobjective framework for large-scale metabolic networks. Ann. Biomed. Eng. 35, 863–885 (2007).

    PubMed  Google Scholar 

  97. Gebhardt, R. & Matz-Soja, M. Liver zonation: novel aspects of its regulation and its impact on homeostasis. World J. Gastroenterol. 20, 8491 (2014).

    PubMed  PubMed Central  Google Scholar 

  98. Junttila, M. R. & De Sauvage, F. J. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature 501, 346–354 (2013).

    CAS  PubMed  Google Scholar 

  99. Maley, C. C. et al. Classifying the evolutionary and ecological features of neoplasms. Nat. Rev. Cancer 17, 605–619 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Nawaz, S., Heindl, A., Koelble, K. & Yuan, Y. Beyond immune density: critical role of spatial heterogeneity in estrogen receptor-negative breast cancer. Mod. Pathol. 28, 766–777 (2015).

    CAS  PubMed  Google Scholar 

  101. Yuan, Y. et al. Quantitative Image Analysis of Cellular Heterogeneity in Breast Tumors Complements Genomic Profiling. Sci. Transl. Med. 4, 157ra143 (2012).

    PubMed  Google Scholar 

  102. 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 

  103. Lloyd, M. C. et al. Vascular measurements correlate with estrogen receptor status. BMC Cancer 14, 279 (2014).

    PubMed  PubMed Central  Google Scholar 

  104. Carmona-Fontaine, C. et al. Emergence of spatial structure in the tumor microenvironment due to the Warburg effect. Proc. Natl Acad. Sci. USA 110, 19402–19407 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Carmona-Fontaine, C. et al. Metabolic origins of spatial organization in the tumor microenvironment. Proc. Natl Acad. Sci. USA 114, 2934–2939 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Tredan, O., Galmarini, C. M., Patel, K. & Tannock, I. F. Drug resistance and the solid tumor microenvironment. J. Natl Cancer Inst. 99, 1441–1454 (2007).

    CAS  PubMed  Google Scholar 

  107. Anderson, A. R. A., Weaver, A. M., Cummings, P. T. & Quaranta, V. Tumor morphology and phenotypic evolution driven by selective pressure from the microenvironment. Cell 127, 905–915 (2006).

    CAS  PubMed  Google Scholar 

  108. Li, X. et al. Temporal and spatial evolution of somatic chromosomal alterations: a case-cohort study of barrett’s esophagus. Cancer Prev. Res. 7, 114–127 (2014).

    Google Scholar 

  109. Lin, J. R. et al. Highly multiplexed immunofluorescence imaging of human tissues and tumors using t-CyCIF and conventional optical microscopes. eLife 7, e31657 (2018).

    PubMed  PubMed Central  Google Scholar 

  110. Rosenbloom, D. I. S., Camara, P. G., Chu, T. & Rabadan, R. Evolutionary scalpels for dissecting tumor ecosystems. Biochim. Biophys. Acta Rev. Cancer 1867, 69–83 (2017).

    CAS  PubMed  Google Scholar 

  111. Azmi, A. S., Bao, B. & Sarkar, F. H. Exosomes in cancer development, metastasis, and drug resistance: a comprehensive review. Cancer Metastasis Rev. 32, 623–642 (2013).

    CAS  PubMed  Google Scholar 

  112. Jones, V. S. et al. Cytokines in cancer drug resistance: cues to new therapeutic strategies. Biochim. Biophys. Acta Rev. Cancer 1865, 255–265 (2016).

    CAS  Google Scholar 

  113. Tabassum, D. P. & Polyak, K. Tumorigenesis: it takes a village. Nat. Rev. Cancer 15, 473–483 (2015).

    CAS  PubMed  Google Scholar 

  114. Chapman, A. et al. Heterogeneous tumor subpopulations cooperate to drive invasion. Cell Rep. 8, 688–695 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank R. Shouval and members of U.A. and J. Joyce’s labs for feedback on the manuscript. This work was supported by the Minerva foundation. U.A. is the incumbent of the Abisch-Frenkel Professorial Chair and acknowledges funding by BSF-NSF-NIH-CRCNS. J.H. acknowledges the support of the Swiss National Science Foundation (#177868), the Swedish Research Council and the Swedish Cancer Society.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding authors

Correspondence to Jean Hausser or Uri Alon.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Cancer thanks A. Anderson, T. Graham and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Related links

Pareto Task Inference software package, Matlab implementation: https://www.weizmann.ac.il/mcb/UriAlon/download/ParTI

An implementation of Pareto Task Inference for the R software: https://github.com/vitkl/ParetoTI

The IMAXT Grand Challenge: https://www.cruk.cam.ac.uk/research-groups/imaxt-laboratory/the-project

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hausser, J., Alon, U. Tumour heterogeneity and the evolutionary trade-offs of cancer. Nat Rev Cancer 20, 247–257 (2020). https://doi.org/10.1038/s41568-020-0241-6

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-020-0241-6

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