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:

The evolution of tumour phylogenetics: principles and practice

Key Points

  • Methods for and applications of phylogenetic tree inference have proliferated in studies of cancer genomics.

  • Tumour phylogeny methods have become important tools for making sense of the complexity of emerging tumour genomic data sets, providing new methods for identifying the order and timing of driver mutations. This has led to new insights and controversies about the nature of the evolutionary processes involved in cancer, and has driven novel approaches to prognostic prediction.

  • Tumour phylogeny methods can be broadly partitioned into several classes of study design, with variations in data source, evolutionary model, and inference algorithm in each class.

  • Productive use of tumour phylogeny methods requires a sophisticated understanding of how to align genomic data sources, evolutionary models, and phylogeny algorithms with research questions about tumour evolution.

  • Key problems of the field remain unresolved, including how to make use of various novel and heterogeneous data sources, the development of more sophisticated models and algorithms appropriate to specific mechanisms of tumour evolution, and the generation of methods and standards for statistically rigorous planning and analysis of tumour phylogeny studies.

Abstract

Rapid advances in high-throughput sequencing and a growing realization of the importance of evolutionary theory to cancer genomics have led to a proliferation of phylogenetic studies of tumour progression. These studies have yielded not only new insights but also a plethora of experimental approaches, sometimes reaching conflicting or poorly supported conclusions. Here, we consider this body of work in light of the key computational principles underpinning phylogenetic inference, with the goal of providing practical guidance on the design and analysis of scientifically rigorous tumour phylogeny studies. We survey the range of methods and tools available to the researcher, their key applications, and the various unsolved problems, closing with a perspective on the prospects and broader implications of this field.

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

Figure 1: Classification of tumour phylogeny methods by study design.
Figure 2: Some challenges in synchronizing data, models and algorithms when applying tumour phylogenetics to a scientific question.

Similar content being viewed by others

References

  1. Hanks, S. et al. Constitutional aneuploidy and cancer predisposition caused by biallelic mutations in (BUB1B). Nat. Genet. 36, 1159–1161 (2004).

    CAS  PubMed  Google Scholar 

  2. Nowell, P. C. The clonal evolution of tumor cell populations. Science 194, 23–28 (1976). This is a seminal paper proposing that solid tumours evolve clonally while accumulating mutations from one mitosis to the next via a process of selection of mutant subpopulations from a common progenitor cell.

    CAS  PubMed  Google Scholar 

  3. Polyak, K. Is breast tumor progression really linear? Clin. Cancer Res. 14, 339–341 (2008).

    PubMed  Google Scholar 

  4. Naxerova, K. & Jain, R. K. Using tumour phylogenetics to identify the roots of metastasis in humans. Nat. Rev. Clin. Oncol. 12, 258–272 (2015).

    CAS  PubMed  Google Scholar 

  5. Foo, J. & Michor, F. Evolution of acquired resistance to anti-cancer therapy. J. Theor. Biol. 355, 10–20 (2014).

    PubMed  Google Scholar 

  6. Enriquez-Naxas, P. M. et al. Exploiting evolutionary principles to prolong tumor control in preclinical models of breast cancer. Sci. Transl Med. 8, 327ra24 (2016).

    Google Scholar 

  7. Merlo, L. M. F., Pepper, J. W., Ried, B. J. & Maley, C. C. Cancer as an evolutionary and ecological process. Nat. Rev. Cancer 6, 924–935 (2006).

    CAS  PubMed  Google Scholar 

  8. Burrell, R. A. & Swanton, C. Re-evaluating clonal dominance in cancer evolution. Trends Cancer 2, 263–276 (2016).

    PubMed  Google Scholar 

  9. Cross, W. C., Graham, T. A. & Wright, N. A. New paradigms in clonal evolution: punctuated equilibrium in cancer. J. Pathol. 240, 126–136 (2016).

    PubMed  Google Scholar 

  10. Podlaha, O., Riester, M., De, S. & Michor, F. Evolution of the cancer genome. Trends Genet. 28, 155–163 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Ding, L., Raphael, B. J., Chen, F. & Wendl, M. C. Advances for studying clonal evolution in cancer. Cancer Lett. 340, 212–219 (2013).

    CAS  PubMed  Google Scholar 

  12. Altrock, P. M., Liu, L. L. & Michor, F. The mathematics of cancer: integrating quantitative models. Nat. Rev. Cancer 15, 730–745 (2015).

    CAS  PubMed  Google Scholar 

  13. Beerenwinkel, N., Schwarz, R. F., Gerstung, M. & Markowetz, F. Cancer evolution: mathematical models and computational inference. Syst. Biol. 64, e1–e25 (2015). This is an in-depth review of applications of mathematical models of evolution to many problems in cancer research, including examples of various techniques drawn from phylogenetics, population genetics, stochastic processes, and game theory and related areas.

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Loeb, L. A. Mutator phenotype may be required for multistage carcinogenesis. Cancer Res. 51, 3075–3079 (1991).

    CAS  PubMed  Google Scholar 

  16. Greenblatt, M. S., Bennett, W. P., Hollstein, M. & Harris, C. C. Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res. 54, 4855–4878 (1994).

    CAS  PubMed  Google Scholar 

  17. Cahill, D. P., Kinzler, K. W., Vogelstein, B. & Lengauer, C. Genetic instability and darwinian selection in tumours. Trends Cell Biol. 9, M57–M60 (1999).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  19. Campbell, P. J. et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature 467, 1109–1113 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Heng, H. H. et al. Chromosome instability (CIN): what it is and why it is crucial to cancer evolution. Cancer Metastasis Rev. 32, 325–340 (2013).

    PubMed  Google Scholar 

  21. de Bruin, E. C. et al. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science 346, 251–256 (2014). This is a particularly instructive study for critically evaluating the application of phylogenetics to bulk tumour samples, in part because it considers multiple phylogenetic methods and recognizes that some samples yield multiple optimal tree topologies.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Gibson, W. J. et al. The genomic landscape and evolution of endometrial carcinoma progression and abdominopelvic metastasis. Nat. Genet. 48, 848–855 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Nik-Zainal, S. et al. Mutational processes molding the genomes of 21 breast cancers. Cell 149, 979–993 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Alexandrov, L. et al. Signatures of mutation processes in human cancers. Nature 500, 415–421 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Alexandrov, L. B. & Stratton, M. R. Mutational signatures: the patterns of somatic mutations hidden in cancer genomes. Curr. Opin. Genet. Devel. 24, 52–60 (2014).

    CAS  Google Scholar 

  28. Hong, M. et al. Tracking the origins and drivers of subclonal metastatic expansion in prostate cancer. Nat. Commun. 6, 6605 (2015).

    CAS  PubMed  Google Scholar 

  29. 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 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Kim, Y. A., Madan, S. & Przytycka, T. M. WeSME: uncovering mutual exclusivity of cancer drivers and beyond. Bioinformatics http://dx.doi.org/10.1093/bioinformatics/btw242 (2016).

  32. Allan, J. M. & Travis, L. B. Mechanisms of therapy-related carcinogenesis. Nat. Rev. Cancer 5, 943–955 (2005).

    CAS  PubMed  Google Scholar 

  33. Johnson, B. E. et al. Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science 343, 189–193 (2014).

    CAS  PubMed  Google Scholar 

  34. Wang, J. et al. Clonal evolution of glioblastoma under therapy. Nat. Genet. 48, 768–776 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Kostadinov, R. L. et al. NSAIDs modulate clonal evolution in Barrett's esophagus. PLoS Genet. 9, e100353 (2013). This is an important investigation for demonstrating the ability of treatment to shape the pre-cancer evolutionary landscape. It provides evidence of a more than order-of-magnitude decrease in mutation rates for patients with Barrett oesophagus who took non-steroidal anti-inflammatory drugs (NSAIDs) versus those who did not.

    Google Scholar 

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

    CAS  PubMed  Google Scholar 

  37. Marusyk, A. & Polyak, K. Tumor heterogeneity: causes and consequences. Biochim. Biophys. Acta 1805, 105–117 (2010).

    CAS  PubMed  Google Scholar 

  38. Park, S. Y., Gönen, M., Kim, H. J., Michor, F. & Polyak, K. Cellular and genetic diversity in the progression of in situ human breast carcinomas to an invasive phenotype. J. Clin. Invest. 120, 636–644 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Anderson, K. et al. Genetic variegation of clonal architecture and propagating cells in leukaemia. Nature 469, 356–361 (2011).

    CAS  PubMed  Google Scholar 

  40. 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 

  41. Heselmeyer-Haddad, K. et al. Single-cell genetic analysis of ductal carcinoma in situ and invasive breast cancer reveals enormous tumor heterogeneity, yet conserved genomic imbalances and gain of MYC during progression. Am. J. Pathol. 181, 1807–1822 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Kim, T. M. et al. Subclonal genomic architectures of primary and metastatic colorectal cancer based on intratumoral genetic heterogeneity. Clin. Cancer Res. 21, 4461–4472 (2015).

    CAS  PubMed  Google Scholar 

  43. Schwarz, R. F. et al. Spatial and temporal heterogeneity in high-grade serous ovarian cancer: a phylogenetic analysis. PLoS Med. 12, e1001789 (2015).

    PubMed  PubMed Central  Google Scholar 

  44. Turajlic, S., McGranahan, N. & Swanton, C. Inferring mutational timing and reconstructing tumour evolutionary histories. Biochim. Biophys. Acta 1855, 264–275 (2015).

    CAS  PubMed  Google Scholar 

  45. Hong, W. S., Shpak, M. & Townsend, J. P. Inferring the origin of metastases from cancer phylogenies. Cancer Res. 75, 4021–4025 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. 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 

  47. Sottoriva, A. et al. A Big Bang model of human colorectal tumor growth. Nat. Genet. 47, 209–216 (2015). This study on the evolution of colorectal cancer illustrates the importance of deep evolutionary theory in interpreting genomic data from tumours. It provides evidence that largely selectively neutral mutations can occur, in contrast to one of the two evolutionary principles of Nowell (reference 2) and others: that cancer evolves by a gradual series of genomic aberrations and that there is strong selection for those aberrations that are more favourable to tumour progression.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. 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 

  49. Shi, H. et al. Acquired resistance and clonal evolution in melanoma during BRAF inhibitor therapy. Cancer Discov. 4, 80–93 (2014).

    CAS  PubMed  Google Scholar 

  50. 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 

  51. Andor, N., Harness, J. V., Müller, S., Mewes, H. W. & Petritsch, C. EXPANDS: expanding ploidy and allele frequency on nested subpopulations. Bioinformatics 30, 50–60 (2014).

    CAS  PubMed  Google Scholar 

  52. Juric, D. et al. Convergent loss of PTEN leads to clinical resistance to a PI(3)Kα inhibitor. Nature 518, 240–244 (2015).

    Article  CAS  PubMed  Google Scholar 

  53. Morrissy, A. S. et al. Divergent clonal selection dominates medulloblastoma at recurrence. Nature 529, 351–357 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Ding, L. et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature 481, 506–510 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Fisher, R., Pusztai, L. & Swanton, C. Cancer heterogeneity: implications for targeted therapeutics. Br. J. Cancer 108, 479–485 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Cooke, S. L. et al. Intra-tumour genetic heterogeneity and poor chemoradiotherapy response in cervical cancer. Br. J. Cancer 104, 361–368 (2011).

    CAS  PubMed  Google Scholar 

  57. Almendro, V. et al. Inference of tumor evolution during chemotherapy by computational modeling and in situ analysis of genetic and phenotypic cellular diversity. Cell Rep. 6, 514–527 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Wangsa, D. et al. Phylogenetic analysis of multiple FISH markers in oral tongue squamous cell carcinoma suggests that a diverse distribution of copy number changes is associated with poor prognosis. Int. J. Cancer 138, 98–109 (2016).

    CAS  PubMed  Google Scholar 

  59. Sottoriva, A. et al. Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc. Natl Acad. Sci. USA 110, 4009–4014 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. McGranahan, N. et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 351, 1463–1469 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Comen, E., Norton, L. & Massagué, J. Clinical implications of cancer self-seeding. Nat. Rev. Clin. Oncol. 8, 369–377 (2011).

    PubMed  Google Scholar 

  63. 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 

  64. Sanborn, J. Z. et al. Phylogenetic analyses of melanoma reveal complex patterns of metastatic dissemination. Proc. Natl Acad. Sci. USA 112, 10995–11000 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Tsao, J. et al. Tracing cell fates in human colorectal tumors from somatic microsatellite mutations: evidence of adenomas with stem cell architecture. Am. J. Pathol. 153, 1189–1200 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Desper, R. et al. Inferring tree models of oncogenesis from comparative genomic hybridization data. J. Comput. Biol. 6, 37–51 (1999). This is the first report to suggest that there might be difficulties with modelling tumour progression as a tree construction problem in phylogenetics.

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Zhao, Z. et al. Early and multiple origins of metastatic lineages within primary tumors. Proc. Natl Acad. Sci. USA 113, 2140–2145 (2016). This study is an important advance over prior bulk sequencing studies for at least three reasons: it compares different phylogenetic methods and draws inferences only when the methods agree on the tree topology; it combines the SNVs in a manner that does not require the use of variant allele frequencies to infer subclones; and it provides clear evidence that some metastases branch early and in parallel, whereas others have a single late origin, reconciling contradictory conclusions reached by earlier studies.

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Pennington, G., Smith, C. A., Shackney, S. & Schwartz, R. Reconstructing tumor phylogenies from heterogeneous single-cell data. J. Bioinform. Comput. Biol. 5, 407–427 (2007).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Navin, N. et al. Tumour evolution inferred by single-cell sequencing. Nature 472, 90–94 (2011). This is the seminal paper in developing and demonstrating the biotechnology to perform scSeq in tumours and apply it to phylogenetic inferences of single tumours.

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Klein, C. A. Selection and adaptation during metastatic cancer progression. Nature 501, 365–372 (2013).

    CAS  PubMed  Google Scholar 

  73. Brocks, D. et al. Intratumor DNA methylation heterogeneity reflects clonal evolution in aggressive prostate cancer. Cell Rep. 8, 798–806 (2014). This bulk tumour phylogeny study is of interest because the authors combine CNV data and DNA methylation data, showing a high correlation of inferred inter-sample evolutionary distances between inferences derived from genetic and from epigenetic data.

    CAS  PubMed  Google Scholar 

  74. Hastings, P., Lupski, J. R., Rosenberg, S. M. & Ira, G. Mechanisms of change in gene copy number. Nat. Rev. Genet. 10, 551–564 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Attolini, C. S. O. & Michor, F. Evolutionary theory of cancer. Ann. NY Acad. Sci. 1168, 23–51 (2009).

    CAS  PubMed  Google Scholar 

  76. Frumkin, D. et al. Cell lineage analysis of a mouse tumor. Cancer Res. 68, 5924–5931 (2008).

    CAS  PubMed  Google Scholar 

  77. Salk, J. J., Horwitz, M. S. & Risques, R. A. Passenger mutations as a marker of clonal cell lineages in emerging neoplasia. Semin. Cancer Biol. 20, 294–303 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Shlush, L. I. et al. Cell lineage analysis of acute leukemia relapse uncovers the role of replication-rate heterogeneity and microsatellite instability. Blood 120, 603–612 (2012).

    CAS  PubMed  Google Scholar 

  79. Sottoriva, A., Spiteri, I., Shibata, D., Curtis, C. & Tavaré, S. Single-molecule genomic data delineate patient-specific tumor profiles and cancer stem cell organization. Cancer Res. 73, 41–49 (2013).

    CAS  PubMed  Google Scholar 

  80. Xu, X. et al. Single-cell exome sequencing reveals single-nucleotide mutation characteristics of a kidney tumor. Cell 148, 886–895 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Huelsenbeck, J. P., Ronquist, F., Nielsen, R. & Bollback, J. P. Bayesian inference of phylogeny and its impact on evolutionary biology. Science 294, 2310–2314 (2001).

    CAS  PubMed  Google Scholar 

  82. Felsenstein, J. Inferring Phylogenies (Sinauer Associates, Inc., 2004).

    Google Scholar 

  83. Boutros, P. C. et al. Spatial genomic heterogeneity within localized, multifocal prostate cancer. Nat. Genet. 47, 736–745 (2015).

    CAS  PubMed  Google Scholar 

  84. Chowdhury, S. A. et al. Phylogenetic analysis of multiprobe fluorescence in situ hybridization data from tumor cell populations. Bioinformatics 29, i189–i198 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Schwarz, R. F. et al. Phylogenetic quantification of intra-tumour heterogeneity. PLoS Comput. Biol. 10, e1003535 (2014). This work provides an important example of a robust cross-platform computational tool for tumour-specific phylogenetic inference, MEDICC, which carries out phylogenetic analysis of multiple samples from a tumour by quantifying intra-tumour heterogeneity while taking into account dependencies between genomic changes.

    PubMed  PubMed Central  Google Scholar 

  86. Yuan, K., Sakoparnig, T., Markowetz, F. & Beerenwinkel, N. BitPhylogeny: a probabilistic framework for reconstructing intra-tumor phylogenies. Genome Biol. 16, 36 (2015). This study represents an exciting advance in the development and implementation of tumour phylogeny methods for third-party use, developing a full Bayesian model that can be applied to both bulk sequencing data and single-cell data.

    PubMed  PubMed Central  Google Scholar 

  87. Jahn, K., Kuipers, J. & Beerenwinkel, N. Tree inference for single-cell data. Genome Biol. 17, 96 (2016).

    Google Scholar 

  88. Nicoloau, M., Levine, A. J. & Carlsson, G. Topology based data analysis identifies a subgroup of breast cancers with a unique mutational profile and excellent survival. Proc. Natl Acad. Sci. USA 108, 7265–7270 (2011).

    Google Scholar 

  89. Kim, K. I. & Simon, R. Using single cell sequencing data to model the evolutionary history of a tumor. BMC Bioinformatics 15, 27 (2014).

    PubMed  PubMed Central  Google Scholar 

  90. Misra, N., Szczurek, E. & Vingron, M. Inferring the paths of somatic evolution in cancer. Bioinformatics 30, 2456–2463 (2014).

    CAS  PubMed  Google Scholar 

  91. Lecca, P., Casiraghi, N. & Demichelis, F. Defining order and timing of mutations during cancer progression: the TO-DAG probabilistic graphical model. Front. Genet. 6, 309 (2015).

    PubMed  PubMed Central  Google Scholar 

  92. Urbschat, S. et al. Clonal cytogenetic progression within intratumorally heterogeneous meningiomas predicts tumor recurrence. Int. J. Oncol. 39, 1601–1608 (2011).

    PubMed  Google Scholar 

  93. Fearon, E. & Vogelstein, B. A genetic model for colorectal tumorigenesis. Cell 61, 759–767 (1990).

    CAS  PubMed  Google Scholar 

  94. Tomlinson, I. & Bodmer, W. Selection, the mutation rate and cancer: ensuring that the tail does not wag the dog. Nat. Med. 5, 11–12 (1999).

    CAS  PubMed  Google Scholar 

  95. Nowak, M. A. et al. The role of chromosomal instability in tumor initiation. Proc. Natl Acad. Sci. USA 99, 16226–16231 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Khalique, L. et al. The clonal evolution of metastases from primary serous epithelial ovarian cancers. Int. J. Cancer 124, 1579–1586 (2009).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  98. Brastianos, P. K. et al. Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets. Cancer Discov. 5, 1164–1177 (2015). This is one of the most interesting and instructive bulk tumour phylogeny studies to date because the study design was to investigate the evolution of metastasis across many tumour types, and the sample size (86) is among the largest for the bulk tumour studies carried out thus far.

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Joung, J. G. et al. Nonlinear tumor evolution from dysplastic nodules to hepatocellular carcinoma. Oncotarget http://dx.doi.org/10.18632/oncotarget.10502 (2016).

  100. Paracchini, L. et al. Regional and temporal heterogeneity of epithelial ovarian cancer tumor biopsies: implications for therapeutic strategies. Oncotarget http://dx.doi.org/10.18632/oncotarget.10505 (2016).

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

    PubMed  Google Scholar 

  102. Heselmeyer-Haddad, K. et al. Single-cell genetic analysis reveals insights into clonal development of prostate cancers and indicates loss of PTEN as a marker of poor prognosis. Am. J. Pathol. 184, 2671–2686 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Janocko, L. E. et al. Distinctive patterns of Her-2/neu, c-myc, and cyclin D1 gene amplification by fluorescence in situ hybridization in primary breast cancers. Cytometry 46, 136–149 (2001).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  105. 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 

  106. Naxerova, K. et al. Hypermutable DNA chronicles the evolution of human colon cancer. Proc. Natl Acad. Sci. USA 111, E1889–E1898 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Tsao, J. et al. Colorectal adenoma and cancer divergence: evidence of multilineage progression. Am. J. Pathol. 154, 815–1824 (1999).

    Google Scholar 

  108. von Heydebreck, A., Gunawan, B. & Füzesi, L. Maximum likelihood estimation of oncogenetic tree models. Biostatistics 5, 545–556 (2004).

    PubMed  Google Scholar 

  109. Bilke, S. et al. Inferring a tumor progression model for neuroblastoma from genomic data. J. Clin. Oncol. 23, 7322–7331 (2005).

    CAS  PubMed  Google Scholar 

  110. Beerenwinkel, N. et al. Learning multiple evolutionary pathways from cross-sectional data. J. Comput. Biol. 12, 584–598 (2005).

    CAS  PubMed  Google Scholar 

  111. Beerenwinkel, N. et al. Mtreemix: a software package for learning and using mixture models of mutagenetic trees. Bioinformatics 21, 2106–2107 (2005).

    CAS  PubMed  Google Scholar 

  112. Hjelm, M., Höglund, M. & Lagergren, J. New probabilistic network models and algorithms for oncogenesis. J. Comput. Biol. 13, 853–865 (2006).

    CAS  PubMed  Google Scholar 

  113. Gerstung, M., Baudis, M., Moch, H. & Beerenwinkel, N. Quantifying cancer progression with conjunctive Bayesian networks. Bioinformatics 25, 2809–2815 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Rahnenführer, J. et al. Estimating cancer survival and clinical outcome based on genetic tumor progression scores. Bioinformatics 21, 2438–2446 (2005).

    PubMed  Google Scholar 

  115. Bogojeska, J., Alexa, A., Altmann, A., Lengauer, T. & Rahnenführer, J. Rtreemix: an R package for estimating evolutionary pathways and genetic progression scores. Bioinformatics 24, 2391–2392 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Bogojeska, J., Lengauer, T. & Rahnenführer, J. Stability analysis of mixtures of mutagenetic trees. BMC Bioinformatics 9, 165 (2008).

    PubMed  PubMed Central  Google Scholar 

  117. Attolini, C. S. et al. A mathematical framework to determine the temporal sequence of somatic genetic events in cancer. Proc. Natl Acad. Sci. USA 107, 17604–17609 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Cheng, Y. et al. A mathematical methodology for determining the temporal order of pathway alterations arising during gliomagenesis. PLoS Comput. Biol. 8, e1002337 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Jiao, W., Vembu, S., Deshwar, A. G., Stein, L. & Morris, Q. Inferring clonal evolution of tumors from single nucleotide somatic mutations. BMC Bioinformatics 15, 35 (2014).

    PubMed  PubMed Central  Google Scholar 

  120. Shahrabi Farahani, H. & Lagergren, J. Learning oncogenetic networks by reducing to mixed integer linear programming. PLoS ONE 8, e65773 (2013).

    PubMed  PubMed Central  Google Scholar 

  121. Ramazzotti, D. et al. CAPRI: efficient inference of cancer progression models from cross-sectional data. Bioinformatics 31, 3016–3026 (2015).

    CAS  PubMed  Google Scholar 

  122. Desper, R. et al. Distance-based reconstruction of tree models for oncogenesis. J. Comput. Biol. 7, 789–803 (2000).

    CAS  PubMed  Google Scholar 

  123. Desper, R., Khan, J. & Schäffer, A. A. Tumor classification using phylogenetic methods on expression data. J. Theor. Biol. 228, 477–496 (2004).

    CAS  PubMed  Google Scholar 

  124. Riester, M., Attolini, C., Downey, R. J., Singer, S. & Michor, F. A differentiation-based phylogeny of cancer subtypes. PLoS Comput. Biol. 6, e1000777 (2010).

    PubMed  PubMed Central  Google Scholar 

  125. Liu, J., Bandyopadhyay, N., Ranka, S., Baudis, M. & Kahveci, T. Inferring progression models for CGH data. Bioinformatics 25, 2208–2215 (2009).

    CAS  PubMed  Google Scholar 

  126. Hainke, K., Rahnenführer, J. & Fried, R. Cumulative disease progression models for cross-sectional data: a review and comparison. Biom. J. 54, 617–640 (2012).

    PubMed  Google Scholar 

  127. Diaz-Uriarte, R. Identifying restrictions in the order of accumulation of mutations during tumor progression: effects of passengers, evolutionary models, and sampling. BMC Bioinformatics 16, 41 (2015).

    PubMed  PubMed Central  Google Scholar 

  128. Sprouffske, K., Pepper, J. W. & Maley, C. C. Accurate reconstruction of the temporal order of mutations in neoplastic progression. Cancer Prev. Res. 4, 1135–1144 (2011).

    Google Scholar 

  129. Letouzé, E., Allory, Y., Bollet, M. A., Radvanyi, F. & Guyon, F. Analysis of the copy number profiles of several tumor samples from the same patient reveals the successive steps in tumorigenesis. Genome Biol. 11, R76 (2010).

    PubMed  PubMed Central  Google Scholar 

  130. Hajirasouliha, I., Mahmoody, A. & Raphael, B. J. A combinatorial approach for analyzing intra-tumor heterogeneity from high-throughput sequencing data. Bioinformatics 30, i78–i86 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. El-Kebir, M., Oesper, L., Acheson-Field, H. & Raphael, B. J. Reconstruction of clonal trees and tumor composition from multi-sample sequencing data. Bioinformatics 31, i62–i70 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Popic, V. et al. Fast and scalable inference of multi-sample cancer lineages. Genome Biol. 16, 91 (2015).

    PubMed  PubMed Central  Google Scholar 

  133. Ha, G. et al. TITAN: inference of copy number architectures in clonal cell populations from tumor whole-genome sequence data. Genome Res. 24, 1881–1893 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Greenman, C. D. et al. Estimation of rearrangement phylogeny for cancer genomes. Genome Res. 22, 346–361 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Deshwar, A. G. et al. PhyloWGS: reconstructing subclonal composition and evolution from whole-genome sequencing of tumors. Genome Biol. 16, 35 (2015).

    PubMed  PubMed Central  Google Scholar 

  136. El-Kebir, M., Satas, G., Oesper, L. & Raphael, B. J. Inferring the mutational history of a tumor using multi-state perfect phylogeny mixtures. Cell Syst. 3, 43–53 (2016).

    CAS  PubMed  Google Scholar 

  137. Jiang, Y., Qiu, Y., Minn, A. J. & Zhang, N. R. Assessing intratumor heterogeneity and tracking longitudinal and spatial clonal evolutionary history by next-generation sequencing. Proc. Natl Acad. Sci. USA 113, E5528–E5537 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Qiao, Y. et al. SubcloneSeeker: a computational framework for reconstructing tumor clone structure for cancer variant interpretation and prioritization. Genome Biol. 15, 443 (2014).

    PubMed  PubMed Central  Google Scholar 

  139. Schwartz, R. & Shackney, S. E. Applying unmixing to gene expression data for tumor phylogeny inference. BMC Bioinformatics 11, 42 (2010).

    PubMed  PubMed Central  Google Scholar 

  140. Miller, C. A. et al. SciClone: inferring clonal architecture and tracking the spatial and temporal patterns of tumor evolution. PLoS Comput. Biol. 10, e1003665 (2014).

    PubMed  PubMed Central  Google Scholar 

  141. Roth, A. et al. PyClone: statistical inference of clonal population structure in cancer. Nat. Methods 11, 396–398 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Zare, H. et al. Inferring clonal composition from multiple sections of a breast cancer. PLoS Comput. Biol. 10, e1003703 (2014).

    PubMed  PubMed Central  Google Scholar 

  143. Niknafs, N., Beleva-Guthrie, V., Naiman, D. Q. & Karchin, R. Subclonal hierarchy inference from somatic mutations: automatic reconstruction of cancer evolutionary trees from multi-region next generation sequencing. PLoS Comput. Biol. 11, e1004416 (2015).

    PubMed  PubMed Central  Google Scholar 

  144. Malikic, S., McPherson, A. A., Donmez, N. & Sahinalp, C. S. Clonality inference in multiple tumor samples using phylogeny. Bioinformatics 31, 1349–1356 (2015).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Newburger, D. E. et al. Genome evolution during progression to breast cancer. Genome Res. 23, 1097–1106 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Gertz, E. M. et al. FISHtrees 3.0: tumor phylogenetics using a ploidy probe. PLoS ONE 11, e0158569 (2016).

    PubMed  PubMed Central  Google Scholar 

  150. Spiro, A. & Shapiro, E. Accuracy of answers to cell lineage questions depends on single-cell genomics data quality and quantity. PLoS Comput. Biol. 12, e1004963 (2016).

    Google Scholar 

  151. Chowdhury, S. A. et al. Algorithms to model single gene, single chromosome, and whole genome copy number changes jointly in tumor phylogenetics. PLoS Comput. Biol. 10, e1003740 (2014).

    PubMed  PubMed Central  Google Scholar 

  152. Chowdhury, S. A. et al. Inferring models of multiscale copy number evolution for single-tumor phylogenetics. Bioinformatics 31, i258–i267 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Zhou, J. Lin, Y., Rajan, V., Hoskins, W. & Tang, J. in Proc. 15th Int. Workshop on Algorithms in Bioinformatics. WABI 2015. Lecture Notes in Computer Science Vol. 9289 (eds Pop, M. & Touzet, H.) 108–120 (Springer, 2015).

    Google Scholar 

  154. Ross, E. M. & Markowetz, F. OncoNEM: inferring tumor evolution from single-cell sequencing data. Genome Biol. 17, 69 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Saitou, N. & Nei, M. The neighbor-joining method: a new method for reconstructing phylogenetic trees. Mol. Biol. Evol. 4, 406–425 (1987).

    CAS  PubMed  Google Scholar 

  156. Wang, Y. et al. Clonal evolution in breast cancer revealed by single nucleus genome sequencing. Nature 512, 155–160 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. 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 

  158. Tao, Y. et al. Rapid growth of a hepatocellular carcinoma and the driving mutations revealed by cell-population genetic analysis of whole-genome data. Proc. Natl Acad. Sci. USA 108, 12042–12047 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Lengauer, C., Kinzler, K. W. & Vogelstein, B. Genetic instabilities in human cancers. Nature 396, 643–649 (1998).

    CAS  PubMed  Google Scholar 

  160. Dewhurst, S. M. et al. Tolerance of whole-genome doubling propagates chromosomal instability and accelerates cancer genome evolution. Cancer Discov. 4, 175–185 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Aguilera, A. & Gómez-González, B. Genome instability: a mechanistic view of its causes and consequences. Nat. Rev. Genet. 9, 204–217 (2008).

    CAS  PubMed  Google Scholar 

  162. Youn, A. & Simon, R. Estimating the order of mutations during tumorigenesis from tumor genome sequencing data. Bioinformatics 28, 1555–1561 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Purdom, E. et al. Methods and challenges in timing chromosomal abnormalities within cancer samples. Bioinformatics 29, 3113–3120 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Beaumont, M. A. Approximate Bayesian computation in evolution and ecology. Annu. Rev. Ecol. Evol. Syst. 41, 379–406 (2010).

    Google Scholar 

  165. Tsao, J. et al. Genetic reconstruction of individual colorectal tumor histories. Proc. Natl, Acad. Sci. USA 97, 1236–1241 (2000).

    CAS  Google Scholar 

  166. Ding, L. et al. Genome remodelling in a basal-like breast cancer metastasis and xenograft. Nature 464, 999–1005 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Gerlinger, M. et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat. Genet. 46, 225–233 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Eyles, J. et al. Tumor cells disseminate early, but immunosurveillance limits metastatic outgrowth, in a mouse model of melanoma. J. Clin. Invest. 120, 2030–2039 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Catanzaro, D., Shackney, S., Schäffer, A. A. & Schwartz, R. Classifying the progression of ductal carcinoma from single-cell sampled data via integer linear programming: a case study. IEEE/ACM Trans. Comput. Biol. Bioinform. 13, 643–655 (2016).

    PubMed  Google Scholar 

  170. Garey, M. R. & Johnson, D. S. Computers and Intractability (WH Freeman New York, 2002).

    Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  172. McGlynn, K. A. et al. A phylogenetic analysis identifies heterogeneity among hepatocellular carcinomas. Hepatology 36, 1341–1348 (2002).

    CAS  PubMed  Google Scholar 

  173. Oesper, L., Mahmoody, A. & Raphael, B. J. THetA: inferring intra-tumor heterogeneity from high-throughput DNA sequencing data. Genome Biol. 14, R80 (2013).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Turajlic, S. & Swanton, C. Metastasis as an evolutionary process. Science 352, 169–175 (2016).

    CAS  PubMed  Google Scholar 

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

    Google Scholar 

  177. Subramanian, A., Shackney, S. & Schwartz, R. Inference of tumor phylogenies from genomic assays on heterogeneous samples. J. Biomed. Biotechnol. 2012, 797812 (2012).

    PubMed  PubMed Central  Google Scholar 

  178. Szabo, A. & Boucher, K. Estimating an oncogenetic tree when false negatives and positives are present. Math. Biosci. 176, 219–236 (2002).

    PubMed  Google Scholar 

  179. De Sano, L. et al. TRONCO: an R package for the inference of cancer progression models from heterogeneous genomic data. Bioinformatics 32, 1911–1913 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Caravagna, G. et al. Algorithmic methods to infer the evolutionary trajectories in cancer progression. Proc. Natl Acad. Sci. USA 113, E4025–E4034 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Pennington, G., Smith, C. A., Shackney, S. & Schwartz, R. Expectation-maximization method for reconstructing tumor phylogenies from single-cell data. Comput. Syst. Bioinformatics Conf. 2006, 371–380 (2006).

    Google Scholar 

  182. Potter, N. E. et al. Single cell mutational profiling and clonal phylogeny in cancer. Genome Res. 23, 2115–2125 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Cresswell, G. D. et al. Intra-tumor genetic heterogeneity in Wilms tumor: clonal evolution and clinical implications. EBioMedicine 9, 120–129 (2016).

    PubMed  PubMed Central  Google Scholar 

  185. Yang, Z. et al. Single-cell sequencing reveals variants in ARID1A, GPRC5A and MLL2 driving self-renewal of human bladder cancer stem cells. Eur. Oncol. 71, 8–12 (2017).

    Google Scholar 

  186. Ozawa, T. et al. Most human non-GCIMP glioblastoma subtypes evolve from a common proneural-like recursor glioma. Cancer Cell 26, 288–300 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Eskilsson, E. et al. EGFRvIII mutations can emerge as late and heterogenous events in glioblastoma development and promote angiogenesis through Src activation. Neuro Oncol. 18, 1644–1655 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Notta, F. et al. Evolution of human BCR–ABL1 lymphoblastic leukaemia-initiating cells. Nature 469, 362–367 (2011).

    CAS  PubMed  Google Scholar 

  189. Campbell, P. J. et al. Subclonal phylogenetic structures in cancer revealed by ultra-deep sequencing. Proc. Natl Acad. Sci. USA 105, 13081–13086 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Lamy, P. et al. Paired exome analysis reveals clonal evolution and potential therapeutic targets in urothelial carcinoma. Cancer Res. 76, 5894–5906 (2016).

    CAS  PubMed  Google Scholar 

  191. 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 

Download references

Acknowledgements

This research was supported in part by the Intramural Research Program of the National Library of Medicine (part of the US National Institutes of Health) and by a grant from the Pennsylvania Department of Health (grant number 4100070287). The Pennsylvania Department of Health specifically disclaims responsibility for any analyses, interpretations or conclusions.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Russell Schwartz.

Ethics declarations

Competing interests

R.S. currently receives research funding from UPMC Enterprises, a division of the University of Pittsburgh Medical Center. A.A.S. declares no competing interests.

Supplementary information

Supplementary information S1 (table)

This Table provides more detailed information on all tools mentioned in Table 1 as well as a few other tools of historical significance to the field or of potential value to readers of this review. (XLSX 23 kb)

Supplementary information S2 (table)

This table provides more detailed information on all case studies mentioned in Table 2, some other case studies that are important to the field and are cited in the main text and a handful of other, related studies that were not cited in the main text but are nonetheless notable in the context of this review. (XLSX 27 kb)

PowerPoint slides

Glossary

Selection

An evolutionary process in which one population (or subclone, in the context of cancer) is favoured for growth or survival over another.

Cancer progression

A change of cancer from a less serious to a more serious state, typically in a manner recognizable by pathologists.

Metastasis

A progression in which cancer cells spread to a location in the body that is physically distant from the primary tumour site.

Subclones

Subpopulations of cells in a tumour; the cells in each subclone are almost or completely genetically identical for all measured cancer-related variants.

Hypermutability

An elevated mitotic mutation rate, relative to that in healthy cells; this is often specific to a given mutation type (for example, a single nucleotide variant or a copy number variant).

Intra-tumour heterogeneity

Variation in the genomes of different cells in the same tumour.

Tumour self-seeding

A process by which descendants of cells that escaped the primary tumour re-enter circulation and return to the primary site.

Mathematical model

A formal mathematical abstraction of a physical or biological process, such as a set of evolutionary mechanisms.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schwartz, R., Schäffer, A. The evolution of tumour phylogenetics: principles and practice. Nat Rev Genet 18, 213–229 (2017). https://doi.org/10.1038/nrg.2016.170

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg.2016.170

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