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.

  • Resource
  • Published:

DNA methylation heterogeneity defines a disease spectrum in Ewing sarcoma

Abstract

Developmental tumors in children and young adults carry few genetic alterations, yet they have diverse clinical presentation. Focusing on Ewing sarcoma, we sought to establish the prevalence and characteristics of epigenetic heterogeneity in genetically homogeneous cancers. We performed genome-scale DNA methylation sequencing for a large cohort of Ewing sarcoma tumors and analyzed epigenetic heterogeneity on three levels: between cancers, between tumors, and within tumors. We observed consistent DNA hypomethylation at enhancers regulated by the disease-defining EWS-FLI1 fusion protein, thus establishing epigenomic enhancer reprogramming as a ubiquitous and characteristic feature of Ewing sarcoma. DNA methylation differences between tumors identified a continuous disease spectrum underlying Ewing sarcoma, which reflected the strength of an EWS-FLI1 regulatory signature and a continuum between mesenchymal and stem cell signatures. There was substantial epigenetic heterogeneity within tumors, particularly in patients with metastatic disease. In summary, our study provides a comprehensive assessment of epigenetic heterogeneity in Ewing sarcoma and thereby highlights the importance of considering nongenetic aspects of tumor heterogeneity in the context of cancer biology and personalized medicine.

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: DNA methylation profiling reveals a characteristic epigenomic signature of Ewing sarcoma.
Figure 2: DNA methylation in EwS shows inter-individual heterogeneity without distinct subtypes.
Figure 3: DNA methylation at regulatory elements defines an epigenetic disease spectrum underlying EwS.
Figure 4: DNA methylation patterns identify widespread intra-tumor heterogeneity in EwS.
Figure 5: DNA methylation heterogeneity in EwS is associated with genetic and clinical data.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. de Álava, E., Lessnick, S.L. & Sorensen, P.H.B. Ewing sarcoma. in WHO Classification of Tumours of Soft Tissue and Bone 4th edn. (eds. Fletcher, C.D.M., Bridge, J.A., Hogendoorn, P.C.W. & Mertens, F.) 306–309 (2013).

  2. Delattre, O. et al. Gene fusion with an ETS DNA-binding domain caused by chromosome translocation in human tumours. Nature 359, 162–165 (1992).

    Article  CAS  PubMed  Google Scholar 

  3. Parham, D.M. et al. Neuroectodermal differentiation in Ewing's sarcoma family of tumors does not predict tumor behavior. Hum. Pathol. 30, 911–918 (1999).

    Article  CAS  PubMed  Google Scholar 

  4. Pinto, A., Dickman, P. & Parham, D. Pathobiologic markers of the Ewing sarcoma family of tumors: state of the art and prediction of behaviour. Sarcoma 2011, 856190 (2011).

    Article  PubMed  Google Scholar 

  5. Schmidt, D., Herrmann, C., Jürgens, H. & Harms, D. Malignant peripheral neuroectodermal tumor and its necessary distinction from Ewing's sarcoma. A report from the Kiel Pediatric Tumor Registry. Cancer 68, 2251–2259 (1991).

    Article  CAS  PubMed  Google Scholar 

  6. Lawrence, M.S. et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499, 214–218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Brohl, A.S. et al. The genomic landscape of the Ewing sarcoma family of tumors reveals recurrent STAG2 mutation. PLoS Genet. 10, e1004475 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Crompton, B.D. et al. The genomic landscape of pediatric Ewing sarcoma. Cancer Discov. 4, 1326–1341 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Tirode, F. et al. Genomic landscape of Ewing sarcoma defines an aggressive subtype with co-association of STAG2 and TP53 mutations. Cancer Discov. 4, 1342–1353 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Riggi, N. et al. EWS-FLI1 utilizes divergent chromatin remodeling mechanisms to directly activate or repress enhancer elements in Ewing sarcoma. Cancer Cell 26, 668–681 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Tomazou, E.M. et al. Epigenome mapping reveals distinct modes of gene regulation and widespread enhancer reprogramming by the oncogenic fusion protein EWS-FLI1. Cell Rep. 10, 1082–1095 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Baylin, S.B. & Jones, P.A. A decade of exploring the cancer epigenome - biological and translational implications. Nat. Rev. Cancer 11, 726–734 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Landau, D.A. et al. Locally disordered methylation forms the basis of intratumor methylome variation in chronic lymphocytic leukemia. Cancer Cell 26, 813–825 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Li, S. et al. Distinct evolution and dynamics of epigenetic and genetic heterogeneity in acute myeloid leukemia. Nat. Med. 22, 792–799 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Gu, H. et al. Genome-scale DNA methylation mapping of clinical samples at single-nucleotide resolution. Nat. Methods 7, 133–136 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Meissner, A. et al. Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis. Nucleic Acids Res. 33, 5868–5877 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Bock, C. et al. Quantitative comparison of genome-wide DNA methylation mapping technologies. Nat. Biotechnol. 28, 1106–1114 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Lin, P.P., Wang, Y. & Lozano, G. Mesenchymal stem cells and the origin of Ewing's sarcoma. Sarcoma 2011, 276463 (2011).

    Article  CAS  PubMed  Google Scholar 

  19. Sheffield, N.C. & Bock, C. LOLA: enrichment analysis for genomic region sets and regulatory elements in R and Bioconductor. Bioinformatics 32, 587–589 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Sheffield, N.C. et al. Patterns of regulatory activity across diverse human cell types predict tissue identity, transcription factor binding, and long-range interactions. Genome Res. 23, 777–788 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

  22. Kundaje, A. et al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Liu, T. et al. Cistrome: an integrative platform for transcriptional regulation studies. Genome Biol. 12, R83 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Sánchez-Castillo, M. et al. CODEX: a next-generation sequencing experiment database for the haematopoietic and embryonic stem cell communities. Nucleic Acids Res. 43, D1117–D1123 (2015).

    Article  CAS  PubMed  Google Scholar 

  25. Bilke, S. et al. Oncogenic ETS fusions deregulate E2F3 target genes in Ewing sarcoma and prostate cancer. Genome Res. 23, 1797–1809 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Creyghton, M.P. et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl. Acad. Sci. USA 107, 21931–21936 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Heintzman, N.D. et al. Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature 459, 108–112 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Agirre, X. et al. Whole-epigenome analysis in multiple myeloma reveals DNA hypermethylation of B cell-specific enhancers. Genome Res. 25, 478–487 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Abe, M. et al. CpG island methylator phenotype is a strong determinant of poor prognosis in neuroblastomas. Cancer Res. 65, 828–834 (2005).

    CAS  PubMed  Google Scholar 

  30. Hovestadt, V. et al. Decoding the regulatory landscape of medulloblastoma using DNA methylation sequencing. Nature 510, 537–541 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. Johann, P.D. et al. Atypical teratoid/rhabdoid tumors are comprised of three epigenetic subgroups with distinct enhancer landscapes. Cancer Cell 29, 379–393 (2016).

    Article  CAS  PubMed  Google Scholar 

  32. Kulis, M. et al. Epigenomic analysis detects widespread gene-body DNA hypomethylation in chronic lymphocytic leukemia. Nat. Genet. 44, 1236–1242 (2012).

    Article  CAS  PubMed  Google Scholar 

  33. Mazor, T. et al. DNA methylation and somatic mutations converge on the cell cycle and define similar evolutionary histories in brain tumors. Cancer Cell 28, 307–317 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Aran, D. & Hellman, A. Unmasking risk loci: DNA methylation illuminates the biology of cancer predisposition: analyzing DNA methylation of transcriptional enhancers reveals missed regulatory links between cancer risk loci and genes. BioEssays 36, 184–190 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Bock, C. et al. DNA methylation dynamics during in vivo differentiation of blood and skin stem cells. Mol. Cell 47, 633–647 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Burger, L., Gaidatzis, D., Schübeler, D. & Stadler, M.B. Identification of active regulatory regions from DNA methylation data. Nucleic Acids Res. 41, e155 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hon, G.C. et al. Epigenetic memory at embryonic enhancers identified in DNA methylation maps from adult mouse tissues. Nat. Genet. 45, 1198–1206 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Stadler, M.B. et al. DNA-binding factors shape the mouse methylome at distal regulatory regions. Nature 480, 490–495 (2011).

    Article  CAS  PubMed  Google Scholar 

  39. Mazumdar, C. et al. Leukemia-associated cohesin mutants dominantly enforce stem cell programs and impair human hematopoietic progenitor differentiation. Cell Stem Cell 17, 675–688 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  41. Mazor, T., Pankov, A., Song, J.S. & Costello, J.F. Intratumoral heterogeneity of the epigenome. Cancer Cell 29, 440–451 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Alizadeh, A.A. et al. Toward understanding and exploiting tumor heterogeneity. Nat. Med. 21, 846–853 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bock, C. et al. BLUEPRINT consortium. Quantitative comparison of DNA methylation assays for biomarker development and clinical applications. Nat. Biotechnol. 34, 726–737 (2016).

    Article  CAS  Google Scholar 

  44. Heyn, H. & Esteller, M. DNA methylation profiling in the clinic: applications and challenges. Nat. Rev. Genet. 13, 679–692 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Laird, P.W. The power and the promise of DNA methylation markers. Nat. Rev. Cancer 3, 253–266 (2003).

    Article  CAS  PubMed  Google Scholar 

  46. Chen, X. et al. Targeting oxidative stress in embryonal rhabdomyosarcoma. Cancer Cell 24, 710–724 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ladenstein, R. et al. Primary disseminated multifocal Ewing sarcoma: results of the Euro-EWING 99 trial. J. Clin. Oncol. 28, 3284–3291 (2010).

    Article  CAS  PubMed  Google Scholar 

  48. Ambros, I.M. et al. MIC2 is a specific marker for Ewing's sarcoma and peripheral primitive neuroectodermal tumors. Evidence for a common histogenesis of Ewing's sarcoma and peripheral primitive neuroectodermal tumors from MIC2 expression and specific chromosome aberration. Cancer 67, 1886–1893 (1991).

    Article  CAS  PubMed  Google Scholar 

  49. Amaral, A.T. et al. Characterization of human mesenchymal stem cells from Ewing sarcoma patients. Pathogenetic implications. PLoS One 9, e85814 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Reinisch, A. et al. Epigenetic and in vivo comparison of diverse MSC sources reveals an endochondral signature for human hematopoietic niche formation. Blood 125, 249–260 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Klughammer, J. et al. Differential DNA methylation analysis without a reference genome. Cell Rep. 13, 2621–2633 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Farlik, M. et al. Single-cell DNA methylome sequencing and bioinformatic inference of epigenomic cell-state dynamics. Cell Rep. 10, 1386–1397 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Xi, Y. & Li, W. BSMAP: whole genome bisulfite sequence MAPping program. BMC Bioinformatics 10, 232 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Xi, Y. et al. RRBSMAP: a fast, accurate and user-friendly alignment tool for reduced representation bisulfite sequencing. Bioinformatics 28, 430–432 (2012).

    Article  CAS  PubMed  Google Scholar 

  55. Krueger, F. & Andrews, S.R. Bismark: a flexible aligner and methylation caller for bisulfite-seq applications. Bioinformatics 27, 1571–1572 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Langmead, B. & Salzberg, S.L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  58. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Feng, J., Liu, T., Qin, B., Zhang, Y. & Liu, X.S. Identifying ChIP-seq enrichment using MACS. Nat. Protoc. 7, 1728–1740 (2012).

    Article  CAS  PubMed  Google Scholar 

  60. Speir, M.L. et al. The UCSC Genome Browser database: 2016 update. Nucleic Acids Res. 44, D717–D725 (2016).

    Article  CAS  PubMed  Google Scholar 

  61. Fan, R.-E., Chang, K.-W., Hsieh, C.-J., Wang, X.-R. & Lin, C.-J. LIBLINEAR: A library for large linear classification. J. Mach. Learn. Res. 9, 1871–1874 (2008).

    Google Scholar 

  62. Sing, T., Sander, O., Beerenwinkel, N. & Lengauer, T. ROCR: visualizing classifier performance in R. Bioinformatics 21, 3940–3941 (2005).

    Article  CAS  PubMed  Google Scholar 

  63. Varley, K.E. et al. Dynamic DNA methylation across diverse human cell lines and tissues. Genome Res. 23, 555–567 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Gaujoux, R. & Seoighe, C. A flexible R package for nonnegative matrix factorization. BMC Bioinformatics 11, 367 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Hyvärinen, A. & Oja, E. Independent component analysis: algorithms and applications. Neural Netw. 13, 411–430 (2000).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank all patients who have donated samples for this study. We also thank the team of the Biomedical Sequencing Facility at CeMM for support with next-generation sequencing; the members of the Delattre, Kovar, and Bock labs for discussions; A. Rendeiro and C. Dietz for contributing to the analysis pipelines; K. Clement for sharing his implementation of the PDR score; A. Lankester for providing MSCs; and the following physicians for providing tumor samples: J.M. Guinebretière, L. Brugières, A. de Muret, R. Tichit, N. Sirvent, F. Millot, F. Guilhot, J.P. Vannier, C. Michot, E. Plouvier, A. Gomez-Brouchet, J. Rivel, B. Petit, F. Dijoud, F. Larousserie, A. Kurt, A. Foulet, A.S. Desfachelles, H. Sartelet, I. Quintin Roue, J. Otten, J. Chasles, C. Bouvier, C. Soler, M. Peuchmaur, and X. Rialland. This study was funded by a grant from the Austrian National Bank's Jubiläumsfonds to E.M.T. (OeNB project number: 15714) and by a peer-reviewed institutional grant to E.M.T., which was based on a charitable donation of the Kapsch group (http://www.kapsch.net/kapschgroup) to St. Anna Kinderkrebsforschung. The French samples were collected in the context of the Plateforme Hospitalière de Génétique Moléculaire des Cancers of the Institut Curie and Centre Hospitalier de Versailles, with support by grants from INSERM within the framework of the International Cancer Genome Consortium program and from the Ligue Nationale Contre Le Cancer (Equipe labellisée), and the Société Française des Cancers de l'Enfant. The following associations supported this work: Courir pour Mathieu, Dans les pas du Géant, Olivier Chape, Les Bagouzamanon, Enfants et Santé, and les Amis de Claire. The study was performed in the context of the following European Union consortia: Euro Ewing (grant agreement no. 602856), BLUEPRINT (grant agreement no. 282510), PROVABES (grant agreement no. 01KT1310), ASSET (grant agreement no. 259348), and TECHNOBEAT (grant agreement no. 668724). N.C.S. was supported by a long-term fellowship of the Human Frontier Science Program (LT000211/2014). J.K. was supported by a DOC Fellowship of the Austrian Academy of Sciences. D. Surdez was supported by the Institut Curie-SIRIC (Site de Recherche Intégrée en Cancérologie) program. E.d.A. was supported by Ministry of Economy and Competitiveness of Spain-FEDER grants (CIBERONC, RD12/0036/0017, PI14/01466), María García-Estrada, CRIS contra el Cáncer Foundations, and Pablo Ugarte Association. C.B. was supported by a New Frontiers Group award of the Austrian Academy of Sciences and by a European Research Council (ERC) Starting Grant (European Union's Horizon 2020 research and innovation program; grant 679146). E.M.T. was supported by fellowships of the Austrian Science Fund (FWF, Lise Meitner Fellowship M1448-B13; and Elise Richter Fellowship V506-B28).

Author information

Authors and Affiliations

Authors

Contributions

N.C.S., O.D., H.K., C.B., and E.M.T. designed the study. N.C.S. performed the data analysis with contributions from J.K., A.S., and M.S. G.P., D. Surdez, D.G., E.L., P.F., J.C., R.B., I.M.A., C.H., E.S., A.T.A., E.d.A., K.S., D. Strunk, B.R., B.L.-A., B.H., A.L., G.d.P., P.T., V.L., J.M., R.L., W.H., R.W., U.D., P.F.A., and O.D. provided materials such as tumor samples, clinical data, cell lines, and MSC samples. P.D., J.H., D.W., and E.M.T. performed the experiments. N.C.S., C.B., and E.M.T. wrote the manuscript with contributions from all authors.

Corresponding authors

Correspondence to Christoph Bock or Eleni M Tomazou.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–12 (PDF 13593 kb)

Supplementary Table 1

Sample annotations and clinical data for the analyzed EwS tumors, EwS cell lines, and MSC samples. (XLSX 27 kb)

Supplementary Table 2

Data processing statistics for the RRBS experiments on EwS tumors, EwS cell lines, and MSC samples performed in this study (sheet 1); for publicly available RRBS profiles of various cancer samples (sheet 2); and for a diverse collection of cell types (sheet 3). (XLSX 101 kb)

Supplementary Table 3

Full results of the LOLA analyses (Figure 1d,e), showing enriched region sets among EwS hypomethylated CpGs (sheet 1) and among EwS hypermethylated CpGs (sheet 2), based on the LOLA Core database. (XLSX 802 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sheffield, N., Pierron, G., Klughammer, J. et al. DNA methylation heterogeneity defines a disease spectrum in Ewing sarcoma. Nat Med 23, 386–395 (2017). https://doi.org/10.1038/nm.4273

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4273

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