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:

Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations

This article has been updated

Abstract

The majority of patients with neuroblastoma have tumors that initially respond to chemotherapy, but a large proportion will experience therapy-resistant relapses. The molecular basis of this aggressive phenotype is unknown. Whole-genome sequencing of 23 paired diagnostic and relapse neuroblastomas showed clonal evolution from the diagnostic tumor, with a median of 29 somatic mutations unique to the relapse sample. Eighteen of the 23 relapse tumors (78%) showed mutations predicted to activate the RAS-MAPK pathway. Seven of these events were detected only in the relapse tumor, whereas the others showed clonal enrichment. In neuroblastoma cell lines, we also detected a high frequency of activating mutations in the RAS-MAPK pathway (11/18; 61%), and these lesions predicted sensitivity to MEK inhibition in vitro and in vivo. Our findings provide a rationale for genetic characterization of relapse neuroblastomas and show that RAS-MAPK pathway mutations may function as a biomarker for new therapeutic approaches to refractory disease.

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: Mutational spectra of diagnostic and relapsed neuroblastomas.
Figure 2: RAS-MAPK pathway mutations reside within major relapsed neuroblastoma subclones.
Figure 3: Sensitivity of neuroblastoma cell line models to MEK inhibition therapy.

Similar content being viewed by others

Accession codes

Primary accessions

BioProject

Change history

  • 20 July 2015

    In the version of this article initially published online, affiliation 30 stating that "These authors jointly supervised this work" was incorrectly omitted for author Jan J. Molenaar. The error has been corrected for the print, PDF and HTML versions of this article.

References

  1. Hero, B. et al. Localized infant neuroblastomas often show spontaneous regression: results of the prospective trials NB95-S and NB97. J. Clin. Oncol. 26, 1504–1510 (2008).

    Article  PubMed  Google Scholar 

  2. Park, J.R. et al. Children's Oncology Group's 2013 blueprint for research: neuroblastoma. Pediatr. Blood Cancer 60, 985–993 (2013).

    Article  PubMed  Google Scholar 

  3. Simon, T. et al. Treatment and outcomes of patients with relapsed, high-risk neuroblastoma: results of German trials. Pediatr. Blood Cancer 56, 578–583 (2011).

    Article  PubMed  Google Scholar 

  4. Maris, J.M. Recent advances in neuroblastoma. N. Engl. J. Med. 362, 2202–2211 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. 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 

  6. Bown, N. et al. Gain of chromosome arm 17q and adverse outcome in patients with neuroblastoma. N. Engl. J. Med. 340, 1954–1961 (1999).

    Article  CAS  PubMed  Google Scholar 

  7. Attiyeh, E.F. et al. Chromosome 1p and 11q deletions and outcome in neuroblastoma. N. Engl. J. Med. 353, 2243–2253 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. Seeger, R.C. et al. Association of multiple copies of the N-myc oncogene with rapid progression of neuroblastomas. N. Engl. J. Med. 313, 1111–1116 (1985).

    Article  CAS  PubMed  Google Scholar 

  9. 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  CAS  PubMed  Google Scholar 

  10. Cheung, N.K. et al. Association of age at diagnosis and genetic mutations in patients with neuroblastoma. J. Am. Med. Assoc. 307, 1062–1071 (2012).

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  12. Pugh, T.J. et al. The genetic landscape of high-risk neuroblastoma. Nat. Genet. 45, 279–284 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Sausen, M. et al. Integrated genomic analyses identify ARID1A and ARID1B alterations in the childhood cancer neuroblastoma. Nat. Genet. 45, 12–17 (2013).

    Article  CAS  PubMed  Google Scholar 

  14. Taggart, D., Dubois, S. & Matthay, K.K. Radiolabeled metaiodobenzylguanidine for imaging and therapy of neuroblastoma. Q. J. Nucl. Med. Mol. Imaging 52, 403–418 (2008).

    CAS  PubMed  Google Scholar 

  15. Schleiermacher, G. et al. Emergence of new ALK mutations at relapse of neuroblastoma. J. Clin. Oncol. 32, 2727–2734 (2014).

    Article  CAS  PubMed  Google Scholar 

  16. Carter, S.L. et al. Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol. 30, 413–421 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Boca, S.M., Kinzler, K.W., Velculescu, V.E., Vogelstein, B. & Parmigiani, G. Patient-oriented gene set analysis for cancer mutation data. Genome Biol. 11, R112 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Futreal, P.A. et al. A census of human cancer genes. Nat. Rev. Cancer 4, 177–183 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Bresler, S.C. et al. ALK mutations confer differential oncogenic activation and sensitivity to ALK inhibition therapy in neuroblastoma. Cancer Cell 26, 682–694 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hölzel, M. et al. NF1 is a tumor suppressor in neuroblastoma that determines retinoic acid response and disease outcome. Cell 142, 218–229 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Tartaglia, M. et al. Diversity and functional consequences of germline and somatic PTPN11 mutations in human disease. Am. J. Hum. Genet. 78, 279–290 (2006).

    Article  CAS  PubMed  Google Scholar 

  22. Carr-Wilkinson, J. et al. High frequency of p53/MDM2/p14ARF pathway abnormalities in relapsed neuroblastoma. Clin. Cancer Res. 16, 1108–1118 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Garnett, M.J. et al. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature 483, 570–575 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Schleiermacher, G. et al. Accumulation of segmental alterations determines progression in neuroblastoma. J. Clin. Oncol. 28, 3122–3130 (2010).

    Article  PubMed  Google Scholar 

  25. 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 

  26. Long, G.V. Increased MAPK reactivation in early resistance to dabrafenib/trametinib combination therapy of BRAF-mutant metastatic melanoma. Nat. Commun. 5, 5694 (2014).

    Article  CAS  PubMed  Google Scholar 

  27. George, R.E. et al. Activating mutations in ALK provide a therapeutic target in neuroblastoma. Nature 455, 975–978 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Mossé, Y.P. et al. Safety and activity of crizotinib for paediatric patients with refractory solid tumours or anaplastic large-cell lymphoma: a Children's Oncology Group phase 1 consortium study. Lancet Oncol. 14, 472–480 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Berry, T. et al. The ALKF1174L mutation potentiates the oncogenic activity of MYCN in neuroblastoma. Cancer Cell 22, 117–130 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Moore, N.F. et al. Molecular rationale for the use of PI3K/AKT/mTOR pathway inhibitors in combination with crizotinib in ALK-mutated neuroblastoma. Oncotarget 5, 8737–8749 (2014).

  31. Drmanac, R. et al. Human genome sequencing using unchained base reads on self-assembling DNA nanoarrays. Science 327, 78–81 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Favero, F. et al. Sequenza: allele-specific copy number and mutation profiles from tumor sequencing data. Ann. Oncol. 26, 64–70 (2015).

    Article  CAS  PubMed  Google Scholar 

  33. Krzywinski, M. et al. Circos: an information aesthetic for comparative genomics. Genome Res. 19, 1639–1645 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. 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  CAS  PubMed  PubMed Central  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  CAS  PubMed  PubMed Central  Google Scholar 

  37. 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  Google Scholar 

  38. 1000 Genomes Project Consortium. An integrated map of genetic variation from 1,092 human genomes. Nature 491, 56–65 (2012).

  39. Rausch, T. et al. DELLY: structural variant discovery by integrated paired-end and split-read analysis. Bioinformatics 28, i333–i339 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Carter, S.L. et al. Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol. 30, 413–421 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. 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 

  42. Lamers, F. et al. Targeted BIRC5 silencing using YM155 causes cell death in neuroblastoma cells with low ABCB1 expression. Eur. J. Cancer 48, 763–771 (2012).

    Article  CAS  PubMed  Google Scholar 

  43. Gnirke, A. et al. Solution hybrid selection with ultra-long oligonucleotides for massively parallel targeted sequencing. Nat. Biotechnol. 27, 182–189 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors would like to thank N. Ross for sample preparation and quality control. This study was supported in part by US National Institutes of Health grants RC1MD004418 to the TARGET consortium and CA98543 and CA98413 to the Children's Oncology Group and was supported in part by a grant throughout the University of Pennsylvania Genome Frontiers Institute. In addition, this project was funded in part with federal funds from the National Cancer Institute, US National Institutes of Health, under contract HHSN261200800001E. The content of this publication does not necessarily reflect the views or policies of the US Department of Health and Human Services nor does mention of trade names, commercial products or organizations imply endorsement by the US government. The binimetinib xenograft studies were supported through a research collaborative agreement between the Children's Hospital of Philadelphia and Novartis Pharmaceuticals. In France, this study was supported by the Annenberg Foundation and the Nelia and Amadeo Barletta Foundation. Funding was also obtained from SiRIC/INCa (grant INCa-DGOS-4654) and from the Comité d'Evaluation et Suivi des Projets de Recherche de Transfert (CEST) of Institut Curie. This study was also funded by the Associations Enfants et Santé, Association Hubert Gouin Enfance et Cancer, Les Bagouz à Manon and Les Amis de Claire. Deep sequencing experiments were conducted on the ICGex next-generation sequencing platform at the Institut Curie funded by the EQUIPEX Investissements d'Avenir program (ANR-10-EQPX-03) and ANR10-INBS-09-08 from the Agence Nationale de le Recherche and by the Canceropôle Ile-de-France. In the Netherlands, this study was supported by grants from the Villa Joep Foundation, the Kinderen Kankervrij Foundation and the Netherlands Cancer Foundation.

Author information

Authors and Affiliations

Authors

Contributions

J.M.M., J.J.M. and G.S. co-coordinated the project. T.F.E., D.A.O., V.B., J. Koster, R.V., G.S., J.J.M. and J.M.M. prepared the manuscript. D.A.O., J. Koster, L.C.D., V.B., N.B.B., P.L.-N., S.J.D., T.J.P., E.F.A., J.S.W., S.Z., S.A., T.B.K.W., D.A.Z. and A.N. generated microarray and/or next-generation sequencing data and/or performed computational data analysis. T.F.E., L.S.H., J.R. and J.M.M. conceived drug testing and gene manipulation experiments and analyzed the data. P.v.S., M.E.E., E.L., V.C., A.B. and M.C. performed sample preparation. L.S.H., J.R., L.S., A.H., M.E.M.D. and T.F.E. contributed to in vitro drug testing and manipulation experiments. A.B., M.C., P.L.-N., D.A.O., T.J.P., E.v.W. and C.E.v.d.S. performed subclonal detection of mutational and structural variants. J.M.G.-F., M.D.H., E.M.W., J.H.S., G.A.T., H.N.C., J.M. and V.C. contributed patient tissue and/or other biological reagents. M.A.S., J.M.G.A., D.S.G., J. Khan and J.M.M. organized the US neuroblastoma TARGET consortium sequencing effort. J.J.M., R.V., J. Koster. and H.N.C. organized the Dutch consortium sequencing effort. G.S., J.M., I.J.-L. and O.D. organized the French consortium sequencing effort.

Corresponding authors

Correspondence to Gudrun Schleiermacher, Jan J Molenaar or John M Maris.

Ethics declarations

Competing interests

J.M.M. has commercial research grants from Novartis Pharmaceuticals and GlaxoSmithKline. H.N.C. is employed by Roche.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–17 and Supplementary Note. (PDF 10668 kb)

Supplementary Tables 1–6

Supplementary Tables 1–6. (XLSX 393 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Eleveld, T., Oldridge, D., Bernard, V. et al. Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations. Nat Genet 47, 864–871 (2015). https://doi.org/10.1038/ng.3333

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3333

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing