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.

Pan-cancer analysis of the extent and consequences of intratumor heterogeneity

Abstract

Intratumor heterogeneity (ITH) drives neoplastic progression and therapeutic resistance. We used the bioinformatics tools 'expanding ploidy and allele frequency on nested subpopulations' (EXPANDS) and PyClone to detect clones that are present at a ≥10% frequency in 1,165 exome sequences from tumors in The Cancer Genome Atlas. 86% of tumors across 12 cancer types had at least two clones. ITH in the morphology of nuclei was associated with genetic ITH (Spearman's correlation coefficient, ρ = 0.24–0.41; P < 0.001). Mutation of a driver gene that typically appears in smaller clones was a survival risk factor (hazard ratio (HR) = 2.15, 95% confidence interval (CI): 1.71–2.69). The risk of mortality also increased when >2 clones coexisted in the same tumor sample (HR = 1.49, 95% CI: 1.20–1.87). In two independent data sets, copy-number alterations affecting either <25% or >75% of a tumor's genome predicted reduced risk (HR = 0.15, 95% CI: 0.08–0.29). Mortality risk also declined when >4 clones coexisted in the sample, suggesting a trade-off between the costs and benefits of genomic instability. ITH and genomic instability thus have the potential to be useful measures that can universally be applied to all cancers.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Tumor metagenomes and subclonal genomes in 12 tumor types from TCGA.
Figure 2: Intratumor genetic heterogeneity in 12 tumor types.
Figure 3: Association of driver-gene mutations with clone size and clone number.
Figure 4: Intratumor nuclear diversity accompanies intratumor genetic diversity.
Figure 5: Clone number and CNV burden appear to be universal prognostic biomarkers.

Similar content being viewed by others

References

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

    CAS  PubMed  Google Scholar 

  2. Bonavia, R., Inda, M.-M., Cavenee, W.K. & Furnari, F.B. Heterogeneity maintenance in glioblastoma: a social network. Cancer Res. 71, 4055–4060 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mroz, E.A., Tward, A.D., Hammon, R.J., Ren, Y. & Rocco, J.W. Intratumor genetic heterogeneity and mortality in head and neck cancer: analysis of data from the Cancer Genome Atlas. PLoS Med. 12, e1001786 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zack, T.I. et al. Pan-cancer patterns of somatic copy number alteration. Nat. Genet. 45, 1134–1140 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kandoth, C. et al. Mutational landscape and significance across 12 major cancer types. Nature 502, 333–339 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ciriello, G. et al. Emerging landscape of oncogenic signatures across human cancers. Nat. Genet. 45, 1127–1133 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Oesper, L., Satas, G. & Raphael, B.J. Quantifying tumor heterogeneity in whole-genome and whole-exome sequencing data. Bioinformatics 30, 3532–3540 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Li, B. & Li, J.Z. A general framework for analyzing tumor subclonality using SNP array and DNA sequencing data. Genome Biol. 15, 473 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. 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).

    Article  CAS  PubMed  Google Scholar 

  19. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Cibulskis, K. et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 31, 213–219 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sathirapongsasuti, J.F. et al. Exome sequencing–based copy-number variation and loss of heterozygosity detection: ExomeCNV. Bioinformatics 27, 2648–2654 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Barber, L.J., Davies, M.N. & Gerlinger, M. Dissecting cancer evolution at the macro-heterogeneity and micro-heterogeneity scale. Curr. Opin. Genet. Dev. 30, 1–6 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Yadav, V.K. & De, S. An assessment of computational methods for estimating purity and clonality using genomic data derived from heterogeneous tumor tissue samples. Brief. Bioinform. 16, 232–241 (2015).

    Article  CAS  PubMed  Google Scholar 

  26. Yoshihara, K. et al. Inferring tumor purity, and stromal and immune cell admixture from expression data. Nat. Commun. 4, 2612 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Tajiri, R. et al. Intratumoral heterogeneous amplification of ERBB2 and subclonal genetic diversity in gastric cancers revealed by multiple ligation-dependent probe amplification and fluorescence in situ hybridization. Hum. Pathol. 45, 725–734.

    Article  CAS  PubMed  Google Scholar 

  28. Sakurada, A., Lara-Guerra, H., Liu, N., Shepherd, F.A. & Tsao, M.-S. Tissue heterogeneity of EGFR mutation in lung adenocarcinoma. J. Thorac. Oncol. 3, 527–529 (2008).

    Article  PubMed  Google Scholar 

  29. Imielinski, M. et al. Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell 150, 1107–1120 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Vitale, M. Intratumor BRAFV600E heterogeneity and kinase inhibitors in the treatment of thyroid cancer: a call for participation. Thyroid 23, 517–519 (2013).

    Article  PubMed  Google Scholar 

  31. Carpenter, A.E. et al. CellProfiler: image analysis software for identifying and quantifying cell phenotypes. Genome Biol. 7, R100 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wang, W., Ozolek, J.A. & Rohde, G.K. Detection and classification of thyroid follicular lesions based on nuclear structure from histopathology images. Cytometry A 77, 485–494 (2010).

    PubMed  PubMed Central  Google Scholar 

  33. Hartwell, K.A. et al. Niche-based screening identifies small-molecule inhibitors of leukemia stem cells. Nat. Chem. Biol. 9, 840–848 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Yamamoto, S. et al. Clinical relevance of Ki67 gene expression analysis using formalin-fixed paraffin-embedded breast cancer specimens. Breast Cancer 20, 262–270 (2013).

    Article  PubMed  Google Scholar 

  35. Cazier, J.-B. et al. Whole-genome sequencing of bladder cancers reveals somatic CDKN1A mutations and clinicopathological associations with mutation burden. Nat. Commun. 5, 3756 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Swanton, C. Cancer evolution constrained by mutation order. N. Engl. J. Med. 372, 661–663 (2015).

    Article  CAS  PubMed  Google Scholar 

  37. Cancer Genome Atlas Research Network. et al. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N. Engl. J. Med. 372, 2481–2498 (2015).

  38. Roylance, R. et al. Relationship of extreme chromosomal instability with long-term survival in a retrospective analysis of primary breast cancer. Cancer Epidemiol. Biomarkers Prev. 20, 2183–2194 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Bochtler, T. et al. Clonal heterogeneity as detected by metaphase karyotyping is an indicator of poor prognosis in acute myeloid leukemia. J. Clin. Oncol. 31, 3898–3905 (2013).

    Article  PubMed  Google Scholar 

  41. Merlo, L.M.F. et al. A comprehensive survey of clonal diversity measures in Barrett’s esophagus as biomarkers of progression to esophageal adenocarcinoma. Cancer Prev. Res. (Phila.) 3, 1388–1397 (2010).

    Article  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  43. Cibulskis, K. et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 31, 213–219 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Roth, A. et al. JointSNVMix: a probabilistic model for accurate detection of somatic mutations in normal-tumor–paired next-generation sequencing data. Bioinformatics 28, 907–913 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Sathirapongsasuti, J.F. et al. Exome sequencing–based copy-number variation and loss of heterozygosity detection: ExomeCNV. Bioinformatics 27, 2648–2654 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. 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).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Goode, A., Gilbert, B., Harkes, J., Jukic, D. & Satyanarayanan, M. OpenSlide: a vendor-neutral software foundation for digital pathology. J. Pathol. Inform. 4, 27 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Kircher, M. et al. A general framework for estimating the relative pathogenicity of human genetic variants. Nat. Genet. 46, 310–315 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Huang, D.W., Sherman, B.T. & Lempicki, R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  CAS  Google Scholar 

  51. Huang, D.W., Sherman, B.T. & Lempicki, R.A. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37, 1–13 (2009).

    Article  CAS  Google Scholar 

  52. Griffith, M. et al. DGIdb: mining the druggable genome. Nat. Methods 10, 1209–1210 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Swanton, C. Cancer evolution constrained by mutation order. N. Engl. J. Med. 372, 661–663 (2015).

    Article  CAS  PubMed  Google Scholar 

  54. Kandoth, C. et al. Mutational landscape and significance across 12 major cancer types. Nature 502, 333–339 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported in part by the US National Institutes of Health (NIH) (grant no. P01 CA91955 (C.C.M.), R01 CA149566 (C.C.M.), R01 CA170595 (C.C.M.), R01 CA185138 (C.C.M.), R01 CA140657 (C.C.M.), P01 HG000205 (H.P.J.), U01CA151920 (H.P.J.), U01CA17629901 (H.P.J.), R01 HG006137 (H.P.J.), R01 CA164746 (C.P.), R01 NS08061904 (C.P.) and R01 HG006137 (L.C.X.)). Additional support to C.C.M. came from the Breast Cancer Research Program Breakthrough Award (award no. BC132057), a Congressionally Directed Medical Research Program (CDMRP). Additional support to H.P.J. came from the Doris Duke Clinical Foundation Clinical Scientist Development Award, a Research Scholar Grant from the American Cancer Society (award no. RSG-13-297-01-TBG) and a Howard Hughes Medical Institute Early Career Grant. N.A. was supported by awards from the Don and Ruth Seiler Fund and the National Cancer Institute (NCI) Cancer Target Discovery and Development (CTDD) Consortium (grant no. U01CA17629901). T.A.G. was supported by the Higher Education Founding Council for England (HEFCE). We are grateful to W. Mewes for advice on the presentation of our results and for insightful discussions about their implications; S.T. Jensen for advice on statistical data analysis; and C.W. Turck and M. Oft for reviewing the manuscript. The results presented here are in part based upon data generated by TCGA Research Network. We thank Hoffmann H. (University of Bonn, Germany) for the availability of the MATLAB function 'violin' that we used to generate the violin plots for the distribution of clone numbers and clone sizes.

Author information

Authors and Affiliations

Authors

Contributions

N.A. developed analytic methods, analyzed data and wrote the manuscript. T.A.G. developed analytic methods, gave technical support and conceptual advice, and wrote the manuscript. M.J. analyzed the histopathology images and provided advice on data visualization and interpretation. L.C.X. provided advice on the choice of statistical methods and the design of the statistical analysis. C.A.A. gave technical support and conceptual advice. C.C.M. developed analytic methods, wrote the manuscript and supervised the project. H.P.J. wrote the manuscript and supervised the project. C.P. supervised the project. All authors edited the manuscript.

Corresponding authors

Correspondence to Hanlee P Ji or Carlo C Maley.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–13, Supplementary Notes 1–4, & Supplementary Tables 1–9 (PDF 12015 kb)

Source data for Table 1 (XLSX 115 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Andor, N., Graham, T., Jansen, M. et al. Pan-cancer analysis of the extent and consequences of intratumor heterogeneity. Nat Med 22, 105–113 (2016). https://doi.org/10.1038/nm.3984

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3984

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