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:

Mutational dynamics between primary and relapse neuroblastomas

Abstract

Neuroblastoma is a malignancy of the developing sympathetic nervous system that is often lethal when relapse occurs. We here used whole-exome sequencing, mRNA expression profiling, array CGH and DNA methylation analysis to characterize 16 paired samples at diagnosis and relapse from individuals with neuroblastoma. The mutational burden significantly increased in relapsing tumors, accompanied by altered mutational signatures and reduced subclonal heterogeneity. Global allele frequencies at relapse indicated clonal mutation selection during disease progression. Promoter methylation patterns were consistent over disease course and were patient specific. Recurrent alterations at relapse included mutations in the putative CHD5 neuroblastoma tumor suppressor, chromosome 9p losses, DOCK8 mutations, inactivating mutations in PTPN14 and a relapse-specific activity pattern for the PTPN14 target YAP. Recurrent new mutations in HRAS, KRAS and genes mediating cell-cell interaction in 13 of 16 relapse tumors indicate disturbances in signaling pathways mediating mesenchymal transition. Our data shed light on genetic alteration frequency, identity and evolution in neuroblastoma.

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: Overview of genomic events in matched primary and relapse neuroblastomas.
Figure 2: Allele frequency shifts between matched pretreatment primary and relapse neuroblastomas.
Figure 3: Global methylation and expression profiling in matched pretreatment primary and relapse neuroblastomas.

Similar content being viewed by others

Accession codes

Primary accessions

European Nucleotide Archive

Gene Expression Omnibus

Referenced accessions

Gene Expression Omnibus

References

  1. Park, J.R., Eggert, A. & Caron, H. Neuroblastoma: biology, prognosis, and treatment. Hematol. Oncol. Clin. North Am. 24, 65–86 (2010).

    Article  PubMed  Google Scholar 

  2. 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 

  3. 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 

  4. Schulte, J.H. et al. High ALK receptor tyrosine kinase expression supersedes ALK mutation as a determining factor of an unfavorable phenotype in primary neuroblastoma. Clin. Cancer Res. 17, 5082–5092 (2011).

    Article  CAS  PubMed  Google Scholar 

  5. 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 

  6. 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 

  7. Cheung, N.-K.V. 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 

  8. 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 

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

    Article  PubMed  PubMed Central  Google Scholar 

  10. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Cobrinik, D. et al. Recurrent pre-existing and acquired DNA copy number alterations, including focal TERT gains, in neuroblastoma central nervous system metastases. Genes Chromosom. Cancer 52, 1150–1166 (2013).

    Article  CAS  PubMed  Google Scholar 

  13. Vandesompele, J. et al. Genetic heterogeneity of neuroblastoma studied by comparative genomic hybridization. Genes Chromosom. Cancer 23, 141–152 (1998).

    Article  CAS  PubMed  Google Scholar 

  14. Zhang, Q. et al. Combined immunodeficiency associated with DOCK8 mutations. N. Engl. J. Med. 361, 2046–2055 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Belle, L. et al. The tyrosine phosphatase PTPN14 (Pez) inhibits metastasis by altering protein trafficking. Sci. Signal. 8, ra18 (2015).

    Article  CAS  PubMed  Google Scholar 

  16. 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 

  17. Theissen, J. et al. Heterogeneity of the MYCN oncogene in neuroblastoma. Clin. Cancer Res. 15, 2085–2090 (2009).

    Article  CAS  PubMed  Google Scholar 

  18. Shukla, N. et al. Oncogene mutation profiling of pediatric solid tumors reveals significant subsets of embryonal rhabdomyosarcoma and neuroblastoma with mutated genes in growth signaling pathways. Clin. Cancer Res. 18, 748–757 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Koyama, H. et al. Mechanisms of CHD5 inactivation in neuroblastomas. Clin. Cancer Res. 18, 1588–1597 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Fujita, T. et al. CHD5, a tumor suppressor gene deleted from 1p36.31 in neuroblastomas. J. Natl. Cancer Inst. 100, 940–949 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Fischer, A., Vázquez-García, I., Illingworth, C.J. & Mustonen, V. High-definition reconstruction of clonal composition in cancer. Cell Rep. 7, 1740–1752 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Luo, Y. et al. Differences in DNA methylation signatures reveal multiple pathways of progression from adenoma to colorectal cancer. Gastroenterology 147, 418.e8–429.e8 (2014).

    Article  Google Scholar 

  23. Wiestler, B. et al. Assessing CpG island methylator phenotype, 1p/19q codeletion, and MGMT promoter methylation from epigenome-wide data in the biomarker cohort of the NOA-04 trial. Neuro-oncol. 16, 1630–1638 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Miyamoto, Y. & Yamauchi, J. Cellular signaling of Dock family proteins in neural function. Cell. Signal. 22, 175–182 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Griggs, B.L., Ladd, S., Saul, R.A., DuPont, B.R. & Srivastava, A.K. Dedicator of cytokinesis 8 is disrupted in two patients with mental retardation and developmental disabilities. Genomics 91, 195–202 (2008).

    Article  CAS  PubMed  Google Scholar 

  26. Vinci, G. et al. Association of deletion 9p, 46, XY gonadal dysgenesis and autistic spectrum disorder. Mol. Hum. Reprod. 13, 685–689 (2007).

    Article  CAS  PubMed  Google Scholar 

  27. 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 

  28. Shao, D.D. et al. KRAS and YAP1 converge to regulate EMT and tumor survival. Cell 158, 171–184 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kapoor, A. et al. Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer. Cell 158, 185–197 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lin, L. et al. The Hippo effector YAP promotes resistance to RAF- and MEK-targeted cancer therapies. Nat. Genet. 47, 250–256 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Su, X., Zhang, L., Zhang, J., Meric-Bernstam, F. & Weinstein, J.N. PurityEst: estimating purity of human tumor samples using next-generation sequencing data. Bioinformatics 28, 2265–2266 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Peifer, M. et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat. Genet. 44, 1104–1110 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Martin, M. et al. Exome sequencing identifies recurrent somatic mutations in EIF1AX and SF3B1 in uveal melanoma with disomy 3. Nat. Genet. 45, 933–936 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

  35. Theissen, J. et al. Chromosome 17/17q gain and unaltered profiles in high resolution array-CGH are prognostically informative in neuroblastoma. Genes Chromosom. Cancer 53, 639–649 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Oakes, C.C. et al. Evolution of DNA methylation is linked to genetic aberrations in chronic lymphocytic leukemia. Cancer Discov. 4, 348–361 (2014).

    Article  CAS  PubMed  Google Scholar 

  37. Teschendorff, A.E. et al. A β-mixture quantile normalization method for correcting probe design bias in Illumina Infinium 450 k DNA methylation data. Bioinformatics 29, 189–196 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Assenov, Y. et al. Comprehensive analysis of DNA methylation data with RnBeads. Nat. Methods 11, 1138–1140 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kocak, H. et al. Hox-C9 activates the intrinsic pathway of apoptosis and is associated with spontaneous regression in neuroblastoma. Cell Death Dis. 4, e586 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Saitou, N. & Nei, M. The neighbor-joining method: a new method for reconstructing phylogenetic trees. Mol. Biol. Evol. 4, 406–425 (1987).

    CAS  PubMed  Google Scholar 

  41. Boc, A., Diallo, A.B. & Makarenkov, V. T-REX: a web server for inferring, validating and visualizing phylogenetic trees and networks. Nucleic Acids Res. 40, W573–W579 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Schulte, J.H. et al. Microarray analysis reveals differential gene expression patterns and regulation of single target genes contributing to the opposing phenotype of TrkA- and TrkB-expressing neuroblastomas. Oncogene 24, 165–177 (2005).

    Article  CAS  PubMed  Google Scholar 

  43. Althoff, K. et al. miR-542-3p exerts tumor suppressive functions in neuroblastoma by downregulating Survivin. Int. J. Cancer 136, 1308–1320 (2015).

    Article  CAS  PubMed  Google Scholar 

  44. Otto, T. et al. Stabilization of N-Myc is a critical function of Aurora A in human neuroblastoma. Cancer Cell 15, 67–78 (2009).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

A.S., S.R. and S.L. acknowledge the support of the Deutsche Forschungsgemeinschaft (DFG) within Collaborative Research Center SFB876, 'Providing Information by Resource-Constrained Analysis', subproject C1. J.K. and S.R. acknowledge support from the Mercator Foundation for project MERCUR Pe-2013-0012 (UA Ruhr Professorship 'Computational Biology'). M.F. was funded by German Cancer Aid (grant 110122), the Kinderkrebs-Neuroblastomforschung Fördergesellschaft and the Center for Molecular Medicine Cologne (CMMC). A.S., A. Eggert, M.F. and J.H.S. were supported by the German Ministry of Science and Education (BMBF) as part of the e:Med initiative (grant 01ZX1303A,B; 01ZX1307C,D,E; and 01ZX1307). A. Eggert and J.H.S. were supported by the ASSET consortium funded by the European Union (Framework Programme 7, grant 259348). A. Eggert was supported by the German Consortium for Translational Cancer Research (DKTK; joint funding pool and DKTK branch Berlin). A. Eggert and K. Astrahantseff were supported by the ENCCA consortium funded by the European Union (Framework Programme 7, grant 261474). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

A.S., S.R., M.F., A. Eggert and J.H.S. conceived and designed the experiments. K. Althoff, E.M., A.G.H., H.S., K.-O.H., M.G., J.A., P.N. and A.O. performed experiments. A.S., J.K., Y.A., K. Althoff, M.P., D.B., S.L., F.R., K.D.P., F.S., C.E., P.S., C.S., L.H., C.G., C.P., M.F. and J.H.S. analyzed the data. J.K., M.P. and S.R. performed statistical analyses. F.W., S.R., H.N.L., A. Engesser, K. Astrahantseff, Y.K., J.T., B.H. and M.F. contributed reagents, materials and/or analysis tools. A.S., A. Eggert and J.H.S. wrote the manuscript.

Corresponding authors

Correspondence to Alexander Schramm or Johannes H Schulte.

Ethics declarations

Competing interests

M.P. is a cofounder and shareholder of NEO New Oncology; he also received consulting fees from NEO New Oncology. C.G. and L.H. are employees of NEO New Oncology.

Integrated supplementary information

Supplementary Figure 1 Mutation counts in relapse tumors.

Mutation counts in relapse tumors were compared to those detected in primary tumors. The number of mutations in relapse samples was significantly higher when all mutations were considered (P = 1 × 10−42). Note that, although the Venn diagram suggests the presence of 775 SNVs, this is because three genes, APOB, MT-CO2 and MYO9A, are lost at relapse in one patient whereas they are gained in another (Fig. 1b).

Supplementary Figure 2 Heat map representation of gains and losses in the genomes of primary-relapse neuroblastoma pairs.

Blue color indicates losses, and red color depicts gains. DNA content was calculated for 1-Mb intervals compared to normalized control DNA.

Supplementary Figure 3 Number of CNAs affected by genomic aberrations.

The numbers of CNAs affected by genomic aberrations identified by array CGH and detected in the relapse tumors were compared to the numbers detected in primary tumors. The number of CNAs in relapse tumors was significantly higher when all CNAs were considered (P = 1 × 10−17).

Supplementary Figure 4 Fractions of single-nucleotide variants detected by exome sequencing that could be validated by targeted amplicon sequencing.

Phred-scaled mutation call confidences from exome sequencing data were divided into quartiles, with 1 and 4 being the lowest and highest mutation calling confidences, respectively. “All” depicts the overall fraction of variants that could be confirmed by targeted resequencing.

Supplementary Figure 5 Scatterplot of the allele mutational frequencies calculated from amplicon sequencing data for all SNVs detected in primary and relapse neuroblastomas.

Circles close to the x axis represent mutations exclusive to the primary tumor, and circles close to the y axis are relapse specific. Circles on or close to the median line represent mutations with preserved allelic frequencies in primary-relapse tumor pairs.

Supplementary Figure 6 Comparison of the detection limits of mutations using MiSeq amplicon sequencing or exome sequencing.

All mutations that were present in relapse tumors but not detected by our SNV calling algorithm using exome sequencing of primary tumors were considered here. An allele frequency below 0.001 cannot be reliably distinguished from technical sequencing errors for bases with quality Q30 or better, even at extremely high coverage, and no statement can be made concerning the true allele frequency in this range.

Supplementary Figure 7 Comparison of the number of SNVs depending on metastatic patterns.

Surprisingly, loco-regional relapses harbored higher number of SNVs than metastastic relapses.

Supplementary Figure 8 The Hippo-YAP pathway is activated in relapsed neuroblastoma.

The Hippo-YAP pathway is activated in relapsed neuroblastoma. (a) Pathway activity scores for YAP-regulated genes based on mRNA profiling of primary and relapse neuroblastomas. Thin lines connect data obtained from primary and relapse tumors from the same patient. The graph depicts the P values for differences in the pathway activity scores between the set of paired primary and relapse tumors. Patients with relapse tumors that harbored PTPN14 mutations are indicated in red. (b) PTPN14 expression (real-time RT-PCR) in a panel of human neuroblastoma cell lines relative to the cell line with the highest expression. Cell lines derived from transformed embryonic kidney cells (HEK293T), adenocarcinoma cells (A549), colon cancer cells (HCT116) and fibroblasts were included as a reference for other tumor types and normal cells. PTPN14 expression was normalized to GAPDH expression. Error bars, s.d. from three independent measurements. (c) YAP subcellular localization (red) in SK-N-SH cells was monitored by immunocytochemistry with a YAP-specific antibody, and cell nuclei were counterstained with DAPI (blue). YAP was localized predominantly to the nuclei of SK-N-SH cells upon treatment with an siRNA targeting PTPN14, whereas YAP was stained mainly in the cytoplasm when SK-N-SH cells were transfected with unrelated ('scrambled') siRNA. (d) SK-N-SH cells engineered for tetracycline-inducible expression of either wild-type PTPN14 (PTPN14-wt) or a patient-derived variant (PTPN14-T42A) presented with predominant nuclear localization of YAP only when mutant PTPN14 was induced. Detection of YAP was achieved by immunocytochemistry as in c. (e) Immunoblot confirming nuclear localization of YAP upon induction of mutant but not wild-type PTPN14. (f) Clonogenicity of SK-N-SH cells in the absence (–tet) or presence (+ tet) of ectopically expressed PTPN14-wt or mutant PTPN14 (PTPN14-T42A). Expression of mutant but not wild-type PTPN14 significantly enhanced the clonogenic growth of SK-N-SH cells. (g) Schematic of Hippo pathway regulation. PTPN14 controls subcellular YAP localization. If PTPN14 is absent or inactivated, YAP will enter the nucleus and activate transcriptional programs involved in supporting cell survival and clonogenic growth.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8. (PDF 605 kb)

Supplementary Table 1

Patient characteristics. (XLSX 12 kb)

Supplementary Table 2

Quality control values for exome sequencing data. (XLSX 12 kb)

Supplementary Table 3

List of relapse-specific SNVs. (XLSX 54 kb)

Supplementary Table 4

List of all SNVs detected in primary and relapse samples. (XLSX 71 kb)

Supplementary Table 5

List of all SNVs lost at relapse. (XLSX 10 kb)

Supplementary Table 6

Comparison of numbers of mutations at relapse and primary diagnosis. (XLSX 9 kb)

Supplementary Table 7

Subclonal copy numbers. (XLSX 10 kb)

Supplementary Table 8

Top results of GSEA. (XLSX 8 kb)

Supplementary Table 9

Calculation of tumor cell content. (XLSX 10 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schramm, A., Köster, J., Assenov, Y. et al. Mutational dynamics between primary and relapse neuroblastomas. Nat Genet 47, 872–877 (2015). https://doi.org/10.1038/ng.3349

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3349

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