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.

  • Letter
  • Published:

Four evolutionary trajectories underlie genetic intratumoral variation in childhood cancer

Subjects

Abstract

A major challenge to personalized oncology is that driver mutations vary among cancer cells inhabiting the same tumor. Whether this reflects principally disparate patterns of Darwinian evolution in different tumor regions has remained unexplored1,2,3,4,5. We mapped the prevalence of genetically distinct clones over 250 regions in 54 childhood cancers. This showed that primary tumors can simultaneously follow up to four evolutionary trajectories over different anatomic areas. The most common pattern consists of subclones with very few mutations confined to a single tumor region. The second most common is a stable coexistence, over vast areas, of clones characterized by changes in chromosome numbers. This is contrasted by a third, less frequent, pattern where a clone with driver mutations or structural chromosome rearrangements emerges through a clonal sweep to dominate an anatomical region. The fourth and rarest pattern is the local emergence of a myriad of clones with TP53 inactivation. Death from disease was limited to tumors exhibiting the two last, most dynamic patterns.

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: Dissection of clonal landscapes over anatomic space.
Fig. 2: Examples of evolutionary trajectories.
Fig. 3: TP53 inactivation causes regional evolutionary explosion and anaplasia.
Fig. 4: Mutational spectra of evolutionary patterns.
Fig. 5: Branching evolution and clinical impact.

Similar content being viewed by others

References

  1. Mengelbier, L. H. et al. Intratumoral genome diversity parallels progression and predicts outcome in pediatric cancer. Nat. Commun. 6, 6125 (2015).

    Article  PubMed  CAS  Google Scholar 

  2. Eleveld, T. F. et al. Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations. Nat. Genet. 47, 864–871 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  4. Schramm, A. et al. Mutational dynamics between primary and relapse neuroblastomas. Nat. Genet. 47, 872–877 (2015).

    Article  PubMed  CAS  Google Scholar 

  5. Padovan-Merhar, O. M. et al. Enrichment of targetable mutations in the relapsed neuroblastoma genome. PLoS Genet. 12, e1006501 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  7. 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).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Kovac, M. et al. Recurrent chromosomal gains and heterogeneous driver mutations characterise papillary renal cancer evolution. Nat. Commun. 6, 6336 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Sottoriva, A. et al. A Big Bang model of human colorectal tumor growth. Nat. Genet. 47, 209–216 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. 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).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Alves, J. M., Prieto, T. & Posada, D. Multiregional tumor trees are not phylogenies. Trends Cancer 3, 546–550 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Molenaar, J. J. et al. Sequencing of neuroblastoma identifies chromothripsis and defects in neuritogenesis genes. Nature 483, 589–593 (2012).

    Article  PubMed  CAS  Google Scholar 

  16. Walz, A. L. et al. Recurrent DGCR8, DROSHA, and SIX homeodomain mutations in favorable histology Wilms tumors. Cancer Cell 27, 286–297 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Wegert, J. et al. Mutations in the SIX1/2 pathway and the DROSHA/DGCR8 miRNA microprocessor complex underlie high-risk blastemal type Wilms tumors. Cancer Cell 27, 298–311 (2015).

    Article  PubMed  CAS  Google Scholar 

  18. Kohsaka, S. et al. A recurrent neomorphic mutation in MYOD1 defines a clinically aggressive subset of embryonal rhabdomyosarcoma associated with PI3K-AKT pathway mutations. Nat. Genet. 46, 595–600 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Staaf, J. et al. Segmentation-based detection of allelic imbalance and loss-of-heterozygosity in cancer cells using whole genome SNP arrays. Genome Biol. 9, R136 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Gisselsson, D. et al. Generation of trisomies in cancer cells by multipolar mitosis and incomplete cytokinesis. Proc. Natl Acad. Sci. USA 107, 20489–20493 (2010).

    Article  PubMed  Google Scholar 

  21. Vujanic, G. M. et al. Revised International Society of Paediatric Oncology (SIOP) working classification of renal tumors of childhood. Med. Pediatr. Oncol. 38, 79–82 (2002).

    Article  PubMed  Google Scholar 

  22. Chagtai, T. et al. Gain of 1q as a prognostic biomarker in Wilms tumors (WTs) treated with preoperative chemotherapy in the International Society of Paediatric Oncology (SIOP) WT 2001 Trial: a SIOP Renal Tumours Biology Consortium Study. J. Clin. Oncol. 34, 3195–3203 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Gratias, E. J. et al. Association of chromosome 1q gain with inferior survival in favorable-histology Wilms tumor: a report from the Children’s Oncology Group. J. Clin. Oncol. 34, 3189–3194 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Ooms, A. H. et al. Significance of TP53 mutation in Wilms tumors with diffuse anaplasia: a report from the Children’s Oncology Group. Clin. Cancer Res. 22, 5582–5591 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Caren, H. et al. High-risk neuroblastoma tumors with 11q-deletion display a poor prognostic, chromosome instability phenotype with later onset. Proc. Natl Acad. Sci. USA 107, 4323–4328 (2010).

    Article  PubMed  Google Scholar 

  26. Caron, H. et al. Allelic loss of chromosome 1p as a predictor of unfavorable outcome in patients with neuroblastoma. N. Engl. J. Med. 334, 225–230 (1996).

    Article  PubMed  CAS  Google Scholar 

  27. Brodeur, G. M., Seeger, R. C., Schwab, M., Varmus, H. E. & Bishop, J. M. Amplification of N-myc in untreated human neuroblastomas correlates with advanced disease stage. Science 224, 1121–1124 (1984).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Baker, A. M. et al. Robust RNA-based in situ mutation detection delineates colorectal cancer subclonal evolution. Nat. Commun. 8, 1998 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Mayrhofer, M., Viklund, B. & Isaksson, A. Rawcopy: Improved copy number analysis with Affymetrix arrays. Sci. Rep. 6, 36158 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Rasmussen, M. et al. Allele-specific copy number analysis of tumor samples with aneuploidy and tumor heterogeneity. Genome Biol. 12, R108 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Koster, J. & Rahmann, S. Snakemake—a scalable bioinformatics workflow engine. Bioinformatics 28, 2520–2522 (2012).

    Article  PubMed  CAS  Google Scholar 

  33. Li, H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. arXiv, https://arxiv.org/abs/1303.3997v2 (2013).

  34. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. DePristo, M. A. et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 43, 491–498 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Narzisi, G. et al. Accurate de novo and transmitted indel detection in exome-capture data using microassembly. Nat. Methods 11, 1033–1036 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Cibulskis, K. et al. ContEst: estimating cross-contamination of human samples in next-generation sequencing data. Bioinformatics 27, 2601–2602 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Okonechnikov, K., Conesa, A. & Garcia-Alcalde, F. Qualimap 2: advanced multi-sample quality control for high-throughput sequencing data. Bioinformatics 32, 292–294 (2016).

    PubMed  CAS  Google Scholar 

  40. Ewels, P., Magnusson, M., Lundin, S. & Kaller, M. MultiQC: summarize analysis results for multiple tools and samples in a single report. Bioinformatics 32, 3047–3048 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Wang, K., Li, M. & Hakonarson, H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Karlsson, A. et al. Mutational and gene fusion analyses of primary large cell and large cell neuroendocrine lung cancer. Oncotarget 6, 22028–22037 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Lindquist, K. E. et al. Clinical framework for next generation sequencing based analysis of treatment predictive mutations and multiplexed gene fusion detection in non-small cell lung cancer. Oncotarget 8, 34796–34810 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Schliep, K. P. phangorn: phylogenetic analysis in R. Bioinformatics 27, 592–593 (2011).

    Article  PubMed  CAS  Google Scholar 

  47. Barde, I., Salmon, P. & Trono, D. Production and titration of lentiviral vectors. Curr. Protoc. Neurosci. 53, 4.21.1–4.21.23 (2010).

    Google Scholar 

  48. Kabadi, A. M., Ousterout, D. G., Hilton, I. B. & Gersbach, C. A. Multiplex CRISPR/Cas9-based genome engineering from a single lentiviral vector. Nucleic Acids Res. 42, e147 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Holm, A., Baldetorp, B., Olde, B., Leeb-Lundberg, L. M. & Nilsson, B. O. The GPER1 agonist G-1 attenuates endothelial cell proliferation by inhibiting DNA synthesis and accumulating cells in the S and G2 phases of the cell cycle. J. Vasc. Res. 48, 327–335 (2011).

    Article  PubMed  CAS  Google Scholar 

  50. Gisselsson, D. Classification of chromosome segregation errors in cancer. Chromosoma 117, 511–519 (2008).

    Article  PubMed  Google Scholar 

  51. Gisselsson, D. et al. When the genome plays dice: circumvention of the spindle assembly checkpoint and near-random chromosome segregation in multipolar cancer cell mitoses. PLoS One 3, e1871 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Mengelbier, L. H. et al. Deletions of 16q in Wilms tumors localize to blastemal-anaplastic cells and are associated with reduced expression of the IRXB renal tubulogenesis gene cluster. Am. J. Pathol. 177, 2609–2621 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Cohn, S. L. et al. The International Neuroblastoma Risk Group (INRG) classification system: an INRG Task Force report. J. Clin. Oncol. 27, 289–297 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Dasgupta, R., Fuchs, J. & Rodeberg, D. Rhabdomyosarcoma. Semin. Pediatr. Surg. 25, 276–283 (2016).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

This study was supported by grants to D.G. from the Swedish Research Foundation (2016-01022), the Swedish Cancer Society (CAN2015/284), the Swedish Childhood Cancer Foundation (PR2016-024, NCP2015-0035), the Crafoord Foundation, the Royal Physiographic Society and the Medical Faculty of Lund University Sweden. We also acknowledge technical support from the Science for Life Laboratory, the Knut and Alice Wallenberg Foundation, the National Genomics Infrastructure founded by the Swedish Research Council and Uppsala Multidisciplinary Center for Advanced Computational Science for assistance with massively parallel sequencing and access to the UPPMAX computational infrastructure. We also thank the Swegene Centre for Integrative Biology at Lund University for assistance.

Author information

Authors and Affiliations

Authors

Contributions

J.K., A.V. and D.G. conceived and designed the project. J.K., A.V., L.H.M., J.S., B.V., T.J. and A.I. coordinated and analyzed whole-genome genotyping and sequencing data, while A.V. performed phylogenetic analysis. S.B., I.Ø., A.B., T.B., B.S., N.P., B.G.L., N.B., D.B. and D.G. performed clinical correlation studies and contributed tumor material. L.C. prepared knockout cell lines. C.J. and A.W. performed technical work.

Corresponding author

Correspondence to David Gisselsson.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–19 and Supplementary Tables 1 and 2

Reporting Summary

Supplementary Data 1

Overview of patients and analytical platforms

Supplementary Data 2

Segmented allelic imbalances

Supplementary Data 3

Mutations detected by whole-exome sequencing

Supplementary Data 4

Variants identified with deep resequencing

Supplementary Data 5

Analysis performed on each sample

Supplementary Data 6

Genome profiles from whole-genome genotyping

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Karlsson, J., Valind, A., Holmquist Mengelbier, L. et al. Four evolutionary trajectories underlie genetic intratumoral variation in childhood cancer. Nat Genet 50, 944–950 (2018). https://doi.org/10.1038/s41588-018-0131-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-018-0131-y

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research