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.

  • Article
  • Published:

Whole-genome sequencing provides new insights into the clonal architecture of Barrett's esophagus and esophageal adenocarcinoma

Abstract

The molecular genetic relationship between esophageal adenocarcinoma (EAC) and its precursor lesion, Barrett's esophagus, is poorly understood. Using whole-genome sequencing on 23 paired Barrett's esophagus and EAC samples, together with one in-depth Barrett's esophagus case study sampled over time and space, we have provided the following new insights: (i) Barrett's esophagus is polyclonal and highly mutated even in the absence of dysplasia; (ii) when cancer develops, copy number increases and heterogeneity persists such that the spectrum of mutations often shows surprisingly little overlap between EAC and adjacent Barrett's esophagus; and (iii) despite differences in specific coding mutations, the mutational context suggests a common causative insult underlying these two conditions. From a clinical perspective, the histopathological assessment of dysplasia appears to be a poor reflection of the molecular disarray within the Barrett's epithelium, and a molecular Cytosponge technique overcomes sampling bias and has the capacity to reflect the entire clonal architecture.

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: Paired Barrett's and EAC samples have varied overlap.
Figure 2: EAC samples display multiple copy number changes in comparison to paired Barrett's esophagus samples.
Figure 3: The mutational context is similar for early and late SNVs.
Figure 4: Summary of the samples sequenced for patient AHM1051.
Figure 5: Six distinct clones are present in the non-dysplastic Barrett's esophagus of patient AHM1051.
Figure 6: Multiple different clones can give rise to dysplasia.

Similar content being viewed by others

References

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

    Article  CAS  Google Scholar 

  2. Esserman, L.J. et al. Addressing overdiagnosis and overtreatment in cancer: a prescription for change. Lancet Oncol. 15, e234–e242 (2014).

    Article  Google Scholar 

  3. Desai, T.K. et al. The incidence of oesophageal adenocarcinoma in non-dysplastic Barrett's oesophagus: a meta-analysis. Gut 61, 970–976 (2012).

    Article  Google Scholar 

  4. Corley, D.A. et al. Impact of endoscopic surveillance on mortality from Barrett's esophagus–associated esophageal adenocarcinomas. Gastroenterology 145, 312–319 (2013).

    Article  Google Scholar 

  5. Shaheen, N.J. & Hur, C. Garlic, silver bullets, and surveillance upper endoscopy for Barrett's esophagus. Gastroenterology 145, 273–276 (2013).

    Article  Google Scholar 

  6. Leedham, S.J. et al. Individual crypt genetic heterogeneity and the origin of metaplastic glandular epithelium in human Barrett's oesophagus. Gut 57, 1041–1048 (2008).

    Article  CAS  Google Scholar 

  7. Maley, C.C. et al. Selectively advantageous mutations and hitchhikers in neoplasms: p16 lesions are selected in Barrett's esophagus. Cancer Res. 64, 3414–3427 (2004).

    Article  CAS  Google Scholar 

  8. Schulmann, K. et al. Inactivation of p16, RUNX3, and HPP1 occurs early in Barrett's-associated neoplastic progression and predicts progression risk. Oncogene 24, 4138–4148 (2005).

    Article  CAS  Google Scholar 

  9. Reid, B.J. et al. Predictors of progression in Barrett's esophagus II: baseline 17p (p53) loss of heterozygosity identifies a patient subset at increased risk for neoplastic progression. Am. J. Gastroenterol. 96, 2839–2848 (2001).

    Article  CAS  Google Scholar 

  10. Kastelein, F. et al. Aberrant p53 protein expression is associated with an increased risk of neoplastic progression in patients with Barrett's oesophagus. Gut 62, 1676–1683 (2013).

    Article  CAS  Google Scholar 

  11. Maley, C.C. Multistage carcinogenesis in Barrett's esophagus. Cancer Lett. 245, 22–32 (2007).

    Article  CAS  Google Scholar 

  12. Fitzgerald, R.C. Dissecting out the genetic origins of Barrett's oesophagus. Gut 57, 1033–1034 (2008).

    Article  Google Scholar 

  13. Weaver, J.M. et al. Ordering of mutations in preinvasive disease stages of esophageal carcinogenesis. Nat. Genet. 46, 837–843 (2014).

    Article  CAS  Google Scholar 

  14. Agrawal, N. et al. Comparative genomic analysis of esophageal adenocarcinoma and squamous cell carcinoma. Cancer Discov. 2, 899–905 (2012).

    Article  CAS  Google Scholar 

  15. Chong, I.Y. et al. The genomic landscape of oesophagogastric junctional adenocarcinoma. J. Pathol. 231, 301–310 (2013).

    Article  CAS  Google Scholar 

  16. Dulak, A.M. et al. Exome and whole-genome sequencing of esophageal adenocarcinoma identifies recurrent driver events and mutational complexity. Nat. Genet. 45, 478–486 (2013).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  18. Chapman, M.A. et al. Initial genome sequencing and analysis of multiple myeloma. Nature 471, 467–472 (2011).

    Article  CAS  Google Scholar 

  19. Ellis, M.J. et al. Whole-genome analysis informs breast cancer response to aromatase inhibition. Nature 486, 353–360 (2012).

    Article  CAS  Google Scholar 

  20. Kan, Z. et al. Whole-genome sequencing identifies recurrent mutations in hepatocellular carcinoma. Genome Res. 23, 1422–1433 (2013).

    Article  CAS  Google Scholar 

  21. Bass, A.J. et al. Genomic sequencing of colorectal adenocarcinomas identifies a recurrent VTI1A-TCF7L2 fusion. Nat. Genet. 43, 964–968 (2011).

    Article  CAS  Google Scholar 

  22. Dulak, A.M. et al. Gastrointestinal adenocarcinomas of the esophagus, stomach, and colon exhibit distinct patterns of genome instability and oncogenesis. Cancer Res. 72, 4383–4393 (2012).

    Article  CAS  Google Scholar 

  23. Schneider, P.M. et al. Mutations of p53 in Barrett's esophagus and Barrett's cancer: a prospective study of ninety-eight cases. J. Thorac. Cardiovasc. Surg. 111, 323–331 discussion 331–333 (1996).

    Article  CAS  Google Scholar 

  24. Dolan, K., Walker, S.J., Gosney, J., Field, J.K. & Sutton, R. TP53 mutations in malignant and premalignant Barrett's esophagus. Dis. Esophagus 16, 83–89 (2003).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  26. Orman, E.S., Li, N. & Shaheen, N.J. Efficacy and durability of radiofrequency ablation for Barrett's Esophagus: systematic review and meta-analysis. Clin. Gastroenterol. Hepatol. 11, 1245–1255 (2013).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  28. Raczy, C. et al. Isaac: ultra-fast whole-genome secondary analysis on Illumina sequencing platforms. Bioinformatics 29, 2041–2043 (2013).

    Article  CAS  Google Scholar 

  29. Saunders, C.T. et al. Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs. Bioinformatics 28, 1811–1817 (2012).

    Article  CAS  Google Scholar 

  30. Gusnanto, A., Wood, H.M., Pawitan, Y., Rabbitts, P. & Berri, S. Correcting for cancer genome size and tumour cell content enables better estimation of copy number alterations from next-generation sequence data. Bioinformatics 28, 40–47 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the Human Research Tissue Bank, which is supported by the National Institute for Health Research (NIHR) Cambridge Biomedical Research Centre. This study was partly funded by a project grant from Cancer Research UK. R.C.F. has programmatic funding from the Medical Research Council and infrastructure support from the Biomedical Research Centre and the Experimental Medicine Centre. We would like to thank all the patients who took part in the study. We thank M. Dunning for his bioinformatics assistance. We thank the Edinburgh Experimental Cancer Medicine Centre.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

R.C.F. conceived the overall study and takes responsibility for the data integrity. C.S.R.-I., J.B. and R.K.C. analyzed the data. C.S.R.-I. extracted the samples for patient AHM1051. A.W. developed the targeted sequencing data visualization tool. C.S.R.-I., J.B., R.K.C., H.N., J.M.J.W., M.R., S.H., D.B. and R.C.F. designed various aspects of the study. H.N. performed the TruSeq Custom Amplicon (TSCA) assay. C.S.R.-I., J.B. and A.G.L. performed the statistical analysis. M.d.P. collected endoscopic samples for patient AHM1051. M.O'D. and S.M. performed the histopathological diagnosis. S.I. developed the copy number pipeline, and R.K.C. and M.H. performed the copy number analysis. Z.K. ran the whole-genome sequencing of patient AHM1051. R.C.F., S.H., D.B. and M.R. supervised the study. C.S.R.-I., J.B., R.K.C. and R.C.F. wrote the manuscript. All authors approved the final version of the manuscript.

Corresponding author

Correspondence to Rebecca C Fitzgerald.

Ethics declarations

Competing interests

R.C.F. developed the Cytosponge technology, which has been licensed by MRC-Technology to Covidien. R.C.F. has no direct pecuniary interest. J.B., A.W., R.K.C., H.N., S.I., M.H., Z.K., M.R., S.H. and D.B. are employees of Illumina.

Integrated supplementary information

Supplementary Figure 1 Multiple samples from five patients (P4, P8, P14, P15 and P17) with Barrett's esophagus and adjacent EAC show that the poor overlap is not a result of sampling bias.

Bar graphs showing the number of SNVs for samples of Barrett's esophagus (BE) and cancer (C) that are either present in all samples (teal), present in all BE samples and absent in all cancer samples (pink), or present in all cancer samples and absent in all BE samples (gray) for each particular patient. Purple bars show the number of remaining variants not accounted for in the other groups (for example, present in a proportion of samples).

Supplementary Figure 2 EAC samples display multiple copy number aberrations compared to paired Barrett's samples.

(a) Copy number plots showing two examples of Barrett's esophagus genomes that are majority copy number 2, compared to the matched EAC samples, which contain multiple gains and losses. (b) Similar plots but for patients where copy number events can be seen in the Barrett's esophagus sample as well as in the EAC sample (but are not significant enough to make a call). (c) Examples of copy number events in the Barrett's esophagus sample that are not seen in the matching EAC sample. The top track shows the depth ratio in a 20-kb window, and the bottom track shows the B-allele ratio.

Supplementary Figure 3 Principal-component analysis of mutational context.

The counts of each different mutational context in each of the three sets of SNVs (common, Barrett's esophagus (BE) unique, EAC unique) were used as input for a principal-component analysis. There is some separation between the common SNVs and the unique SNVs in the second component, but the differences between the groups are not striking (6.9% variance in the second component). (b) Plot showing the weights of the 96 different mutational contexts. Four mutational contexts—C(A>C)C, G(C>G)G and C(A>T)C (all at the top right-hand side of the plot) as well as T(C>G)A (bottom right)—seem to be the main cause for the separation in the second component.

Supplementary Figure 4 Copy number plot for one of the Barrett's esophagus samples with low-grade dysplasia.

Copy number plot for one of the Barrett's esophagus samples with low-grade dysplasia that was sent for whole-genome sequencing. Large deletions on multiple chromosomes, including chromosomes 5, 11, 13, 18 and 21, can be seen.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–4 and Supplementary Tables 1–4. (PDF 1340 kb)

Supplementary Data Set

Nebula: custom-made data browser to investigate and visualize the results from the 1,443 targets that were sequenced for 73 Barrett's esophagus samples from patient AHM1051. (ZIP 7637 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ross-Innes, C., Becq, J., Warren, A. et al. Whole-genome sequencing provides new insights into the clonal architecture of Barrett's esophagus and esophageal adenocarcinoma. Nat Genet 47, 1038–1046 (2015). https://doi.org/10.1038/ng.3357

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3357

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