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:

The genetic landscape of high-risk neuroblastoma

Abstract

Neuroblastoma is a malignancy of the developing sympathetic nervous system that often presents with widespread metastatic disease, resulting in survival rates of less than 50%. To determine the spectrum of somatic mutation in high-risk neuroblastoma, we studied 240 affected individuals (cases) using a combination of whole-exome, genome and transcriptome sequencing as part of the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) initiative. Here we report a low median exonic mutation frequency of 0.60 per Mb (0.48 nonsilent) and notably few recurrently mutated genes in these tumors. Genes with significant somatic mutation frequencies included ALK (9.2% of cases), PTPN11 (2.9%), ATRX (2.5%, and an additional 7.1% had focal deletions), MYCN (1.7%, causing a recurrent p.Pro44Leu alteration) and NRAS (0.83%). Rare, potentially pathogenic germline variants were significantly enriched in ALK, CHEK2, PINK1 and BARD1. The relative paucity of recurrent somatic mutations in neuroblastoma challenges current therapeutic strategies that rely on frequently altered oncogenic drivers.

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: Landscape of genetic variation in neuroblastoma.
Figure 2: Structural variation in neuroblastoma genomes.

Similar content being viewed by others

Accession codes

Accessions

ArrayExpress

Gene Expression Omnibus

References

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Smith, M.A. et al. Outcomes for children and adolescents with cancer: challenges for the twenty-first century. J. Clin. Oncol. 28, 2625–2634 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Matthay, K.K. et al. Treatment of high-risk neuroblastoma with intensive chemotherapy, radiotherapy, autologous bone marrow transplantation, and 13-cis-retinoic acid. Children's Cancer Group. N. Engl. J. Med. 341, 1165–1173 (1999).

    Article  CAS  PubMed  Google Scholar 

  4. Yu, A.L. et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N. Engl. J. Med. 363, 1324–1334 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Oeffinger, K.C. et al. Chronic health conditions in adult survivors of childhood cancer. N. Engl. J. Med. 355, 1572–1582 (2006).

    Article  CAS  PubMed  Google Scholar 

  6. Trochet, D. et al. Germline mutations of the paired-like homeobox 2B (PHOX2B) gene in neuroblastoma. Am. J. Hum. Genet. 74, 761–764 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Mosse, Y.P. et al. Germline PHOX2B mutation in hereditary neuroblastoma. Am. J. Hum. Genet. 75, 727–730 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Mosse, Y.P. et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature 455, 930–935 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Janoueix-Lerosey, I. et al. Somatic and germline activating mutations of the ALK kinase receptor in neuroblastoma. Nature 455, 967–970 (2008).

    Article  CAS  PubMed  Google Scholar 

  10. Diskin, S.J. et al. Common variation at 6q16 within HACE1 and LIN28B influences susceptibility to neuroblastoma. Nat. Genet. 44, 1126–1130 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wang, K. et al. Integrative genomics identifies LMO1 as a neuroblastoma oncogene. Nature 469, 216–220 (2011).

    Article  CAS  PubMed  Google Scholar 

  12. Nguyen, L.B. et al. Phenotype restricted genome-wide association study using a gene-centric approach identifies three low-risk neuroblastoma susceptibility loci. PLoS Genet. 7, e1002026 (2011).

    Article  CAS  PubMed Central  Google Scholar 

  13. Diskin, S.J. et al. Copy number variation at 1q21.1 associated with neuroblastoma. Nature 459, 987–991 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Capasso, M. et al. Common variations in BARD1 influence susceptibility to high-risk neuroblastoma. Nat. Genet. 41, 718–723 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Maris, J.M. et al. Chromosome 6p22 locus associated with clinically aggressive neuroblastoma. N. Engl. J. Med. 358, 2585–2593 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Deyell, R.J. & Attiyeh, E.F. Advances in the understanding of constitutional and somatic genomic alterations in neuroblastoma. Cancer Genet. 204, 113–121 (2011).

    Article  CAS  PubMed  Google Scholar 

  17. 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 

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

    Article  CAS  Google Scholar 

  19. Sausen, M. et al. Integrated genomic analyses identify ARID1A and ARID1B alterations in the childhood cancer neuroblastoma. Nat. Genet. 45, 12–17 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Bentley, D.R. et al. Accurate whole human genome sequencing using reversible terminator chemistry. Nature 456, 53–59 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. 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 

  22. Pugh, T.J. et al. Medulloblastoma exome sequencing uncovers subtype-specific somatic mutations. Nature 488, 106–110 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wang, L. et al. SF3B1 and other novel cancer genes in chronic lymphocytic leukemia. N. Engl. J. Med. 365, 2497–2506 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Cancer Genome Atlas Research Network. et al. Comprehensive genomic characterization of squamous cell lung cancers. Nature 489, 519–525 (2012); erratum 491, 288 (2012).

  25. Lee, R.S. et al. A remarkably simple genome underlies highly malignant pediatric rhabdoid cancers. J. Clin. Invest. 122, 2983–2988 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Banerji, S. et al. Sequence analysis of mutations and translocations across breast cancer subtypes. Nature 486, 405–409 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Hodis, E. et al. A landscape of driver mutations in melanoma. Cell 150, 251–263 (2012).

    Article  CAS  PubMed  PubMed Central  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. Stransky, N. et al. The mutational landscape of head and neck squamous cell carcinoma. Science 333, 1157–1160 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Nikolaev, S.I. et al. Exome sequencing identifies recurrent somatic MAP2K1 and MAP2K2 mutations in melanoma. Nat. Genet. 44, 133–139 (2012).

    Article  CAS  Google Scholar 

  32. Getz, G. et al. Comment on 'The Consensus Coding Sequences of Human Breast and Colorectal Cancers'. Science 317, 1500 (2007).

    Article  CAS  PubMed  Google Scholar 

  33. Forbes, S.A. et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 39, D945–D950 (2011).

    Article  CAS  PubMed  Google Scholar 

  34. Shi, L. et al. The MicroArray Quality Control (MAQC)-II study of common practices for the development and validation of microarray-based predictive models. Nat. Biotechnol. 28, 827–838 (2010).

    Article  CAS  PubMed  Google Scholar 

  35. 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 

  36. Chen, Y. et al. Oncogenic mutations of ALK kinase in neuroblastoma. Nature 455, 971–974 (2008).

    Article  CAS  PubMed  Google Scholar 

  37. Bentires-Alj, M. et al. Activating mutations of the Noonan syndrome-associated SHP2/PTPN11 gene in human solid tumors and adult acute myelogenous leukemia. Cancer Res. 64, 8816–8820 (2004).

    Article  CAS  PubMed  Google Scholar 

  38. Tartaglia, M. et al. Mutations in PTPN11, encoding the protein tyrosine phosphatase SHP-2, cause Noonan syndrome. Nat. Genet. 29, 465–468 (2001).

    Article  CAS  PubMed  Google Scholar 

  39. Tartaglia, M. et al. Somatic mutations in PTPN11 in juvenile myelomonocytic leukemia, myelodysplastic syndromes and acute myeloid leukemia. Nat. Genet. 34, 148–150 (2003).

    Article  CAS  PubMed  Google Scholar 

  40. Sarkozy, A. et al. A novel PTPN11 gene mutation bridges Noonan syndrome, multiple lentigines/LEOPARD syndrome and Noonan-like/multiple giant cell lesion syndrome. Eur. J. Hum. Genet. 12, 1069–1072 (2004).

    Article  CAS  PubMed  Google Scholar 

  41. De Brouwer, S. et al. Meta-analysis of neuroblastomas reveals a skewed ALK mutation spectrum in tumors with MYCN amplification. Clin. Cancer Res. 16, 4353–4362 (2010).

    Article  CAS  PubMed  Google Scholar 

  42. 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  CAS  PubMed  Google Scholar 

  43. Adzhubei, I.A. et al. A method and server for predicting damaging missense mutations. Nat. Methods 7, 248–249 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ng, P.C. & Henikoff, S. SIFT: predicting amino acid changes that affect protein function. Nucleic Acids Res. 31, 3812–3814 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Schwarz, J.M., Rödelsperger, C., Schuelke, M. & Seelow, D. MutationTaster evaluates disease-causing potential of sequence alterations. Nat. Methods 7, 575–576 (2010).

    Article  CAS  PubMed  Google Scholar 

  46. Tavtigian, S.V. et al. Comprehensive statistical study of 452 BRCA1 missense substitutions with classification of eight recurrent substitutions as neutral. J. Med. Genet. 43, 295–305 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Marchler-Bauer, A. et al. CDD: a Conserved Domain Database for the functional annotation of proteins. Nucleic Acids Res. 39, D225–D229 (2011).

    Article  CAS  PubMed  Google Scholar 

  48. Love, C. et al. The genetic landscape of mutations in Burkitt lymphoma. Nat. Genet. 44, 1321–1325 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Barbosa-Morais, N.L., Carmo-Fonseca, M. & Aparício, S. Systematic genome-wide annotation of spliceosomal proteins reveals differential gene family expansion. Genome Res. 16, 66–77 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chen, M. & Manley, J.L. Mechanisms of alternative splicing regulation: insights from molecular and genomics approaches. Nat. Rev. Mol. Cell Biol. 10, 741–754 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Gabut, M., Chaudhry, S. & Blencowe, B.J. SnapShot: The splicing regulatory machinery. Cell 133, 192 e1 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. 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 

  54. Sijmons, R.H. Encyclopaedia of tumour-associated familial disorders. Part I: from AIMAH to CHIME syndrome. Hered. Cancer Clin. Pract. 6, 22–57 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Wood, R.D., Mitchell, M. & Lindahl, T. Human DNA repair genes, 2005. Mutat. Res. 577, 275–283 (2005).

    Article  CAS  PubMed  Google Scholar 

  56. Tennessen, J.A. et al. Evolution and functional impact of rare coding variation from deep sequencing of human exomes. Science 337, 64–69 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Sodha, N., Mantoni, T.S., Tavtigian, S.V., Eeles, R. & Garrett, M.D. Rare germ line CHEK2 variants identified in breast cancer families encode proteins that show impaired activation. Cancer Res. 66, 8966–8970 (2006).

    Article  CAS  PubMed  Google Scholar 

  58. Lee, S.B. et al. Destabilization of CHK2 by a missense mutation associated with Li-Fraumeni syndrome. Cancer Res. 61, 8062–8067 (2001).

    CAS  PubMed  Google Scholar 

  59. Dong, X. et al. Mutations in CHEK2 associated with prostate cancer risk. Am. J. Hum. Genet. 72, 270–280 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Petitjean, A. et al. Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database. Hum. Mutat. 28, 622–629 (2007).

    Article  CAS  PubMed  Google Scholar 

  61. Birch, J.M. et al. Relative frequency and morphology of cancers in carriers of germline TP53 mutations. Oncogene 20, 4621–4628 (2001).

    Article  CAS  PubMed  Google Scholar 

  62. Choi, J.M. et al. Analysis of PARK genes in a Korean cohort of early-onset Parkinson disease. Neurogenetics 9, 263–269 (2008).

    Article  CAS  PubMed  Google Scholar 

  63. Marongiu, R. et al. PINK1 heterozygous rare variants: prevalence, significance and phenotypic spectrum. Hum. Mutat. 29, 565 (2008).

    Article  PubMed  Google Scholar 

  64. Klein, C. et al. PINK1, Parkin, and DJ-1 mutations in Italian patients with early-onset parkinsonism. Eur. J. Hum. Genet. 13, 1086–1093 (2005).

    Article  CAS  PubMed  Google Scholar 

  65. Fredlund, E., Ringnér, M., Maris, J.M. & Påhlman, S. High Myc pathway activity and low stage of neuronal differentiation associate with poor outcome in neuroblastoma. Proc. Natl. Acad. Sci. USA 105, 14094–14099 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Storlazzi, C.T. et al. Gene amplification as double minutes or homogeneously staining regions in solid tumors: origin and structure. Genome Res. 20, 1198–1206 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Stephens, P.J. et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 144, 27–40 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. 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 

  71. Goya, R. et al. SNVMix: predicting single nucleotide variants from next-generation sequencing of tumors. Bioinformatics 26, 730–736 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Carnevali, P. et al. Computational techniques for human genome resequencing using mated gapped reads. J. Comput. Biol. 19, 279–292 (2012).

    Article  CAS  PubMed  Google Scholar 

  73. Kostic, A.D. et al. PathSeq: software to identify or discover microbes by deep sequencing of human tissue. Nat. Biotechnol. 29, 393–396 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Robertson, G. et al. De novo assembly and analysis of RNA-seq data. Nat. Methods 7, 909–912 (2010).

    Article  CAS  PubMed  Google Scholar 

  75. Lee, W. et al. The mutation spectrum revealed by paired genome sequences from a lung cancer patient. Nature 465, 473–477 (2010).

    Article  CAS  PubMed  Google Scholar 

  76. Morin, R.D. et al. Somatic mutation of EZH2 (Y641) in follicular and diffuse large B-cell lymphomas of germinal center origin. Nat. Genet. 42, 181–185 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Morin, R.D. et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature 476, 298–303 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Pruitt, K.D., Tatusova, T., Klimke, W. & Maglott, D.R. NCBI Reference Sequences: current status, policy and new initiatives. Nucleic Acids Res. 37, D32–D36 (2009).

    Article  CAS  PubMed  Google Scholar 

  79. Morozova, O. et al. System-level analysis of neuroblastoma tumor–initiating cells implicates AURKB as a novel drug target for neuroblastoma. Clin. Cancer Res. 16, 4572–4582 (2010).

    Article  CAS  PubMed  Google Scholar 

  80. Morin, R. et al. Profiling the HeLa S3 transcriptome using randomly primed cDNA and massively parallel short-read sequencing. Biotechniques 45, 81–94 (2008).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Robinson, J.T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Ye, K., Schulz, M.H., Long, Q., Apweiler, R. & Ning, Z. Pindel: a pattern growth approach to detect break points of large deletions and medium sized insertions from paired-end short reads. Bioinformatics 25, 2865–2871 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Shah, S.P. et al. Mutational evolution in a lobular breast tumour profiled at single nucleotide resolution. Nature 461, 809–813 (2009).

    Article  CAS  PubMed  Google Scholar 

  85. Simpson, J.T. et al. ABySS: a parallel assembler for short read sequence data. Genome Res. 19, 1117–1123 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Pruitt, K.D. et al. The consensus coding sequence (CCDS) project: identifying a common protein-coding gene set for the human and mouse genomes. Genome Res. 19, 1316–1323 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Mortazavi, A., Williams, B.A., McCue, K., Schaeffer, L. & Wold, B. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat. Methods 5, 621–628 (2008).

    Article  CAS  PubMed  Google Scholar 

  88. Griffith, M. et al. Alternative expression analysis by RNA sequencing. Nat. Methods 7, 843–847 (2010).

    Article  CAS  PubMed  Google Scholar 

  89. Su, A.I. et al. Large-scale analysis of the human and mouse transcriptomes. Proc. Natl. Acad. Sci. USA 99, 4465–4470 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the Children's Oncology Group for the collection and annotation of samples for this study, and all TARGET co-investigators for scientific support of this project. Funding was provided by US National Institutes of Health grants CA98543 and CA98413 to the Children's Oncology Group, RC1MD004418 to the TARGET consortium, CA124709 (J.M.M.) and CA060104 (R.C.S.) and National Human Genome Research Institute grant U54HG003067 (E.S.L., D.A., S.B.G., G.G. and M.M.), as well as a contract from the National Cancer Institute, US National Institutes of Health (HHSN261200800001E). Additional support included a Canadian Institutes of Health Research Fellowship (T.J.P.), a Roman M. Babicki Fellowship in Medical Research at the University of British Columbia (O.M.), the Canada Research Chair in Genome Science (M.A.M.), the Giulio D'Angio Endowed Chair (J.M.M.), the Alex's Lemonade Stand Foundation (J.M.M.), the Arms Wide Open Foundation (J.M.M.) and the Cookies for Kids Foundation (J.M.M.). We thank E. Nickerson, S. Channer, K. Novik, C. Suragh and R. Roscoe for project management support. We also thank the staff of the Genome Sciences Centre Biospecimen Core, Library Construction, Sequencing and Bioinformatics teams, and the staff of the Broad Institute Biological Samples, Genome Sequencing and Genetic Analysis Platforms for their expertise in genomic processing of samples, and generating the sequencing data used in this analysis.

Author information

Authors and Affiliations

Authors

Contributions

J.M.M., J. Khan, R.C.S., D.S.G. and M.A.S. conceived and led the project. M.A.M. and M.M. conceived of and supervised all aspects of the sequencing work at the British Columbia (BC) Cancer Agency Genome Sciences Centre and Broad Institute, respectively. T.J.P. and O.M. performed the analyses and interpreted the results. E.F.A., S.A., J.S.W., K.A.C., M.D., S.J.D., A.C.W., Y.P.M., L.J., T.B., Y.M., J.M.G.-F. and M.D.H. selected and characterized samples, provided disease-specific expertise in data analysis and edited the manuscript. R.S. and W.B.L. provided statistical support and analyses of clinical covariates. D.A., E.S., C. Sougnez, M.D. and J.M.G.A. provided overall project management and quality control support. S.L.C., K.C., M. Hanna, A.K., J. Kim, M.S.L., L.L., A.M., A.H.R., A.S. and C. Stewart supported analysis of somatic and germline alterations in the exome sequencing data. C.S.P. performed the pathogen discovery analysis. I.B., K.L.M., R.C., S.D.J. and J.Q. performed de novo assembly of Illumina sequencing data. Y.Z. led the library construction effort for the Illumina libraries. A.T. and Y.Z. planned the sequencing verification, and A.A. and B.K. performed the experiments. R.D.C. performed copy number analysis of genome sequencing data. M.K. performed verification of candidate rearrangements. N.T. performed gene- and exon-level quantification analysis of RNA-seq data. A.L. and A.H.K. helped interpret data provided by Complete Genomics. R.A.M. and M. Hirst led the sequencing effort for the Illumina genome and transcriptome libraries. S.B.G. and E.S.L. led the sequencing effort for the exome sequencing libraries. G.G. and S.J.M.J. supervised the bioinformatics group at the Broad Institute and BC Cancer Agency Genome Sciences Centre, respectively. T.J.P., O.M., D.S.G., M.A.M., M.M. and J.M.M. cowrote the manuscript with input from all coauthors.

Corresponding authors

Correspondence to Marco A Marra, Matthew Meyerson or John M Maris.

Ethics declarations

Competing interests

M.M. is a paid consultant for and equity holder in Foundation Medicine, a genomics-based oncology diagnostics company, and is a paid consultant for Novartis.

Supplementary information

Supplementary Text and Figures

Supplementary Note, Supplementary Tables 11–14 and Supplementary Figures 1–10 (PDF 4034 kb)

Supplementary Table 1

Master data table: Clinical and molecular data for all neuroblastoma cases including identifiers from other databases, sequencing technologies used, clinical and biological covariates, and matrix of mutation calls (XLSX 2887 kb)

Supplementary Table 2

Coverage: Fraction of bases in each exon with sufficient coverage for mutation detection (XLSX 184212 kb)

Supplementary Table 3

Full mutation list: All coding somatic mutations called in all cases (XLSX 2377 kb)

Supplementary Table 4

Mutation frequency correlates: Statistical comparison of mutation frequency distributions (Kolmogorov-Smirnov) when comparing cases by clinical and biological variables (XLSX 37 kb)

Supplementary Table 5

Pathogens: Counts of sequencing reads in exome capture libraries corresponding to known viruses (XLS 49 kb)

Supplementary Table 6

MutSig: Significance analysis of somatic mutation frequency in all genes and a focused set of genes listed in the Catalogue of Somatic Mutations in Cancer (XLSX 2052 kb)

Supplementary Table 7

Gene set significance analysis: Full list of pathways, member genes, mutated genes, and significance values as calculated by MutSig with and without significantly mutated genes (XLSX 381 kb)

Supplementary Table 8

Structural rearrangements: All structural variants detected in neuroblastoma genomes or transcriptomes (XLSX 16 kb)

Supplementary Table 9

Significance analysis of germline ClinVar variation: List of all genes tested for enrichment in neuroblastoma of ClinVar variants (XLSX 1622 kb)

Supplementary Table 10

Significance analysis of germline loss-of-function variants in Cancer Census, cancer syndrome, or DNA repair genes (XLSX 632 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Pugh, T., Morozova, O., Attiyeh, E. et al. The genetic landscape of high-risk neuroblastoma. Nat Genet 45, 279–284 (2013). https://doi.org/10.1038/ng.2529

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2529

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