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:

Telomerase activation by genomic rearrangements in high-risk neuroblastoma

This article has been updated

Abstract

Neuroblastoma is a malignant paediatric tumour of the sympathetic nervous system1. Roughly half of these tumours regress spontaneously or are cured by limited therapy. By contrast, high-risk neuroblastomas have an unfavourable clinical course despite intensive multimodal treatment, and their molecular basis has remained largely elusive2,3,4. Here we have performed whole-genome sequencing of 56 neuroblastomas (high-risk, n = 39; low-risk, n = 17) and discovered recurrent genomic rearrangements affecting a chromosomal region at 5p15.33 proximal of the telomerase reverse transcriptase gene (TERT). These rearrangements occurred only in high-risk neuroblastomas (12/39, 31%) in a mutually exclusive fashion with MYCN amplifications and ATRX mutations, which are known genetic events in this tumour type1,2,5. In an extended case series (n = 217), TERT rearrangements defined a subgroup of high-risk tumours with particularly poor outcome. Despite a large structural diversity of these rearrangements, they all induced massive transcriptional upregulation of TERT. In the remaining high-risk tumours, TERT expression was also elevated in MYCN-amplified tumours, whereas alternative lengthening of telomeres was present in neuroblastomas without TERT or MYCN alterations, suggesting that telomere lengthening represents a central mechanism defining this subtype. The 5p15.33 rearrangements juxtapose the TERT coding sequence to strong enhancer elements, resulting in massive chromatin remodelling and DNA methylation of the affected region. Supporting a functional role of TERT, neuroblastoma cell lines bearing rearrangements or amplified MYCN exhibited both upregulated TERT expression and enzymatic telomerase activity. In summary, our findings show that remodelling of the genomic context abrogates transcriptional silencing of TERT in high-risk neuroblastoma and places telomerase activation in the centre of transformation in a large fraction of these tumours.

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: Genomic rearrangements are clustered at chromosome 5p15.33 in high-risk neuroblastoma.
Figure 2: Genomic TERT rearrangements are associated with poor patient outcome and high TERT mRNA expression.
Figure 3: Translocation of active enhancers drive TERT expression in TERT-rearranged neuroblastomas.
Figure 4: Telomerase activity is associated with TERT rearrangements and MYCN amplification, while ALT occurs in high-risk neuroblastoma lacking these alterations.

Similar content being viewed by others

Accession codes

Data deposits

All high-throughput data have been deposited at the European Genome-phenome Archive (https://www.ebi.ac.uk/ega/) under accession number EGAS00001001308.

Change history

  • 28 October 2015

    Spelling of author C.G. was corrected.

References

  1. Maris, J. M., Hogarty, M. D., Bagatell, R. & Cohn, S. L. Neuroblastoma. Lancet 369, 2106–2120 (2007)

    Article  CAS  Google Scholar 

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

    Article  CAS  ADS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  5. 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 

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

    Article  ADS  Google Scholar 

  7. Northcott, P. A. et al. Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma. Nature 511, 428–434 (2014)

    Article  CAS  ADS  Google Scholar 

  8. Nagel, I. et al. Deregulation of the telomerase reverse transcriptase (TERT) gene by chromosomal translocations in B-cell malignancies. Blood 116, 1317–1320 (2010)

    Article  CAS  Google Scholar 

  9. Davis, C. F. et al. The somatic genomic landscape of chromophobe renal cell carcinoma. Cancer Cell 26, 319–330 (2014)

    Article  CAS  Google Scholar 

  10. Horn, S. et al. TERT promoter mutations in familial and sporadic melanoma. Science 339, 959–961 (2013)

    Article  CAS  ADS  Google Scholar 

  11. Huang, F. W. et al. Highly recurrent TERT promoter mutations in human melanoma. Science 339, 957–959 (2013)

    Article  CAS  ADS  Google Scholar 

  12. Mac, S. M., D’Cunha, C. A. & Farnham, P. J. Direct recruitment of N-myc to target gene promoters. Mol. Carcinog. 29, 76–86 (2000)

    Article  CAS  Google Scholar 

  13. Zhao, Y., Wang, S., Popova, E. Y., Grigoryev, S. A. & Zhu, J. Rearrangement of upstream sequences of the hTERT gene during cellular immortalization. Genes Chromosom. Cancer 48, 963–974 (2009)

    Article  CAS  Google Scholar 

  14. Kumps, C. et al. Focal DNA copy number changes in neuroblastoma target MYCN regulated genes. PLoS One 8, e52321 (2013)

    Article  CAS  ADS  Google Scholar 

  15. 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  Google Scholar 

  16. Heintzman, N. D. et al. Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome. Nature Genet. 39, 311–318 (2007)

    Article  CAS  Google Scholar 

  17. Ernst, J. et al. Mapping and analysis of chromatin state dynamics in nine human cell types. Nature 473, 43–49 (2011)

    Article  CAS  ADS  Google Scholar 

  18. Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013)

    Article  CAS  Google Scholar 

  19. Castelo-Branco, P. et al. Methylation of the TERT promoter and risk stratification of childhood brain tumours: an integrative genomic and molecular study. Lancet Oncol. 14, 534–542 (2013)

    Article  CAS  Google Scholar 

  20. Farooqi, A. S. et al. Alternative lengthening of telomeres in neuroblastoma cell lines is associated with a lack of MYCN genomic amplification and with p53 pathway aberrations. J. Neurooncol. 119, 17–26 (2014)

    Article  CAS  Google Scholar 

  21. O’Sullivan, R. J. et al. Rapid induction of alternative lengthening of telomeres by depletion of the histone chaperone ASF1. Nature Struct. Mol. Biol. 21, 167–174 (2014)

    Article  Google Scholar 

  22. Cesare, A. J. & Reddel, R. R. Alternative lengthening of telomeres: models, mechanisms and implications. Nature Rev. Genet. 11, 319–330 (2010)

    Article  CAS  Google Scholar 

  23. Martínez, P. & Blasco, M. A. Telomeric and extra-telomeric roles for telomerase and the telomere-binding proteins. Nature Rev. Cancer 11, 161–176 (2011)

    Article  Google Scholar 

  24. Brodeur, G. M. Neuroblastoma: biological insights into a clinical enigma. Nature Rev. Cancer 3, 203–216 (2003)

    Article  CAS  Google Scholar 

  25. Fernandez-Cuesta, L. et al. Frequent mutations in chromatin-remodelling genes in pulmonary carcinoids. Nature Commun. 5, 3518 (2014)

    Article  ADS  Google Scholar 

  26. Zhang, W. et al. Comparison of RNA-seq and microarray-based models for clinical endpoint prediction. Genome Biol. 16, 133 (2015)

    Article  CAS  Google Scholar 

  27. Peng, X. et al. Tissue-specific transcriptome sequencing analysis expands the non-human primate reference transcriptome resource (NHPRTR). Nucleic Acids Res. 43, D737–D742 (2015)

    Article  CAS  Google Scholar 

  28. Blecher-Gonen, R. et al. High-throughput chromatin immunoprecipitation for genome-wide mapping of in vivo protein-DNA interactions and epigenomic states. Nature Protocols 8, 539–554 (2013)

    Article  Google Scholar 

  29. Dahl, J. A. & Collas, P. A rapid micro chromatin immunoprecipitation assay (microChIP). Nature Protocols 3, 1032–1045 (2008)

    Article  CAS  Google Scholar 

  30. Harrel, F. & Lee, K. in Proc. Eleventh Annual SAS User’s Group International 823–838 (SAS Institute, 1986)

    Google Scholar 

Download references

Acknowledgements

We are indebted to the patients and their parents for making available the tumour specimens analysed in this study. We thank the German Neuroblastoma Biobank for providing samples from patients. The Institutional Review Board approved collection and use of all specimens in this study. We also thank our colleagues N. Hemstedt, H. Düren, E. Hess, J. Kreth, and J. Gopalakrishnan; and our collaboration partners I. Amit and F. Paul at the Weizmann Institute of Science for technical assistance. We further acknowledge the Center for Molecular Medicine Cologne light microscope facility for helping us to obtain high-quality data of FISH analyses, and S. Wolf and the Next Generation Sequencing Unit at the German Cancer Research Center (DKFZ) for sequencing. This work was supported by the German Cancer Aid (grant 110122) to M.F., F.W., J.H.S., A.S., and S.A.; the German Ministry of Science and Education (BMBF) as part of the e:Med initiative (grant 01ZX1303A to M.P., M.F., J.H.S., R.B., U.L., and R.K.T., grant 01ZX1406 to M.P., and grant 01ZX1307D to M.F. and F.W.); the BMBF (grant 0316076A to F.W.); the European Union (grant 259348 to F.W.); the Fördergesellschaft Kinderkrebs-Neuroblastom-Forschung e.V. (to M.F.); the German-Israeli Helmholtz Research School in Cancer Biology (to M.G. and F.W.); the Volkswagenstiftung (Lichtenberg Program) (to M.R.S.); and the Center for Molecular Medicine Cologne.

Author information

Authors and Affiliations

Authors

Contributions

Conception and design: M.P., F.H., F.R., D.D., M.G., F.W., R.K.T., and M.F. Administrative support, provision of study materials and patients: M.P., J.H., I.L., T.H., P.N., V.A., U.L., A.Eg., F.B., Z.P., C.Z., L.Sh., R.B., S.P., B.H., A.S., J.H.S., F.W., R.J.O., R.K.T., and M.F. Conduct of the experiments, data analysis, and interpretation: M.P., F.H., F.R., D.D., M.G., R.M., A.K., J.L.R., F.S., J.H., F.I., R.S., S.A., A.En., Y.K., W.V., J.A., J.T.-M., D.T.-M., A.M., S.H., E.M., K.-O.H., C.G., G.B., M.-R.S., L.Sa., S.C.W., C.S., E.B., J.T., R.V., M.S., B.D., M.O., B.H., C.H., R.J.O., F.W., R.K.T., and M.F. Manuscript writing: M.P., F.H., F.R., F.W., R.K.T., and M.F. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Martin Peifer, Frank Westermann, Roman K. Thomas or Matthias Fischer.

Ethics declarations

Competing interests

M.P. is a co-founder and shareholder of NEO New Oncology, and received consulting fees from NEO New Oncology. R.K.T. is a founder and shareholder of NEO New Oncology, and received consulting and lecture fees from NEO New Oncology, Sanofi-Aventis, Merck, Roche, Lilly, Boehringer Ingelheim, AstraZeneca, Atlas Biolabs, Daiichi Sankyo, Johnson & Johnson, Puma and Clovis, as well as research support from Merck, EOS and AstraZeneca.

Extended data figures and tables

Extended Data Figure 1 Validation of rearrangements of the TERT locus by dideoxy-sequencing.

Sequencing chromatograms of the breakpoint regions of 5p15.33 rearrangements along with their genomic coordinates (hg19), and the breakpoint-spanning nucleotide sequences. The sequence mapping to the TERT locus is indicated in yellow, the rearrangement partner is indicated in grey; nucleotides inserted at the breakpoint region are indicated in white.

Extended Data Figure 2 Schematic representation of chromothripsis in four primary neuroblastomas.

a, Circos plots showing chromothripsis of chromosome 5 in the tumours NBL39 and NBL37. b, Circos plots showing chromothripsis of chromosomes 17 and 20 in the tumours NBL21 and NBL08, respectively. Regions showing loss of heterozygosity (LOH) are indicated by black segments.

Extended Data Figure 3 TERT rearrangements are associated with poor event-free survival of patients, and with TERT upregulation independent of the breakpoint distance from the TERT transcriptional start site.

a, Kaplan–Meier estimates for event-free survival of neuroblastoma patient groups defined by TERT rearrangements (TERT), MYCN amplification (MNA), high-risk disease without these alterations (HR), and low-risk or intermediate-risk disease (nonHR). Patients with tumours bearing both a TERT rearrangement and MYCN amplification (n = 5) were excluded. Event-free survival at 5 years: 0.17 ± 0.09 (TERT) versus 0.38 ± 0.09 (MNA) versus 0.43 ± 0.09 (HR) versus 0.83 ± 0.05 (nonHR). b, Multivariable Cox regression analysis of the potential prognostic factors stage, MYCN status, and TERT status for event-free survival in patients aged >18 months (n = 125). c, Validation of TERT expression levels by microarrays in the four neuroblastoma subgroups indicated above. Sample numbers are given at the bottom. **P < 0.01, ***P < 0.001. d, Fold-change of median TERT expression levels in TERT-rearranged and MYCN-amplified tumours compared with low-risk (LR) neuroblastomas as measured by transcriptome sequencing. e, Evidence for monoallelic expression of TERT in five tumours bearing TERT rearrangements. The presence of a heterozygous single nucleotide polymorphism and its allelic fraction measured by whole-genome sequencing (WGS) is shown on the left of each panel; monoallelic expression as established by transcriptome sequencing (RNA-seq) is indicated on the right. The genomic position of the single nucleotide polymorphism is indicated at the top, the number of reads available for the analysis is shown at the bottom. f, TERT expression measured by transcriptome sequencing in relation to the distance of the rearrangement breakpoint from the TERT transcriptional start site (TSS). TERT expression levels and breakpoint distances from the TERT transcriptional start site were not correlated (r = 0, P = 0.97; Spearman’s rank correlation test).

Extended Data Figure 4 TERT rearrangements are maintained in relapsed neuroblastoma.

a, Agarose gel electrophoresis of PCR products representing individual TERT rearrangements in four tumours at initial diagnosis (I), and at relapse (R). The non-template controls are indicated by C. b, TERT expression measured by microarrays in relapsed TERT-rearranged tumours (n = 3) compared with TERT-rearranged tumours biopsied at initial diagnosis (n = 10). c, Sequencing chromatograms of the breakpoint regions for the relapse cases.

Extended Data Figure 5 Regional effects of TERT rearrangements on gene expression patterns.

a, Expression levels of genes around TERT measured by transcriptome sequencing are shown for tumours with TERT rearrangements (TERT, yellow), tumours with MYCN amplifications (MNA, red), and tumours without these aberrations classified as either high-risk (HR, grey) or low-risk (LR, green). Five consecutive genes (TERT, bleyplaby, SLC6A18, SLC6A19, and rarjy) located distal of the breakpoint at chromosome 5p15.33, and three genes (suweeby, blorplaby, and CLPTM1L) located proximal of the breakpoint are shown. Genomic positions of the genes are indicated at the top, and sample sizes are indicated at the bottom. NS, not significant, *P < 0.05, **P < 0.01, ***P < 0.001. b, Averaged expression levels of genes measured by transcriptome sequencing are indicated for an 500 kb region centring around TERT. Colour codes of neuroblastoma subgroups as indicated above. The breakpoint region is indicated in beige.

Extended Data Figure 6 TERT mRNA levels are massively upregulated in TERT-rearranged and MYCN-amplified neuroblastomas.

a, Differential gene expression between TERT-rearranged (n = 10) and low-risk (n = 17) neuroblastomas and b, between MYCN-amplified (MNA, n = 92) and low-risk tumours (n = 238) measured by transcriptome sequencing26. Negative log(P values) are plotted against log2(fold changes) of the genes expressed in the respective subgroups. Genes of interest are indicated at the bottom. log2(fold changes) correspond to the log difference of established gene expression scores. P values were established using Student’s t-tests and applying a Benjamini and Hochberg false discovery rate multiple testing correction. c, Analysis of differential gene expression of a MYCN-amplified neuroblastoma cell line (IMR-5/75) before and after shRNA-mediated knockdown of MYCN using transcriptome sequencing. The number of genes (frequency) up- or downregulated upon MYCN knockdown is plotted against the fold change of regulation. TERT is the most strongly downregulated gene upon shRNA-mediated MYCN knockdown. Only differentially expressed genes (t-test, false discovery rate controlled with P < 0.01) are shown. The insert shows reduced MYCN protein levels after induction of a MYCN-specific shRNA in IMR-5/75 cells as well as β-actin protein levels as control analysed by immunoblotting.

Extended Data Figure 7 Detection of TERT rearrangements in neuroblastoma cell lines.

a, Schematic representation of binding sites of probes used for FISH at the TERT locus. b, FISH analysis of neuroblastoma cell lines with 5p15.33 rearrangements and without these rearrangements; MNA, MYCN-amplified. Arrows indicate the unaltered status of chromosome 5p15.33 (red signals in close proximity to green signals); arrowheads indicate a rearrangement of 5p15.33 (red signals without adjacent green signals).

Extended Data Figure 8 Patterns of H3K27me3 histone modification and DNA methylation at the TERT locus.

a, Survey of the histone mark H3K27me3 in neuroblastomas harbouring TERT rearrangements (cell line GI-ME-N and primary tumour NBL36) and a neuroblastoma cell line lacking these alterations (SK-N-FI). b, DNA methylation patterns of CpG sites at the TERT locus in TERT-rearranged (TERT, n = 6) and MYCN-amplified (MNA, n = 9) primary neuroblastomas, as well as tumours lacking these alterations (others, n = 24) using HumanMethylation450 microrrays. Samples are ordered from top to bottom. CpG sites are indicated relative to their position to the TERT locus. Average methylation levels of each CpG site in the three subgroups are shown at the top. Similar to the highlighted CpG site cg11625005 upstream of the TERT transcriptional start site (Fig. 3d), CpG sites scattered over the TERT gene body were methylated significantly higher in TERT-rearranged and MYCN-amplified cases than in tumours without these alterations (P < 0.001 each).

Extended Data Figure 9 ALT activity in neuroblastoma cell lines lacking TERT rearrangements and MYCN amplifications.

a, Detection of extrachromosomal telomeric repeat DNA by C-circle assays with genomic AluI/MboI digested DNA from the indicated neuroblastoma cell lines. The ALT-positive control cell line, U2OS, is at the left. b, Detection of ALT-associated PML bodies in the indicated cell lines. Telomeric TTAGGG FISH (green) and immunofluorescence for PML (red) were combined and DNA was counterstained with DAPI (blue). c, Telomere restriction-fragment analysis of telomeric DNA from LAN-6, SK-N-FI, and the ALT-positive control U2OS. Telomeric DNA was detected by Southern blot with a [32P]dATP end-labelled (CCCTAA)4 oligonucleotide.

Extended Data Table 1 Prognostic characteristics of the neuroblastoma cohort and prevalence of TERT rearrangements in patient subgroups

Supplementary information

Supplementary Table 1

This table contains WGS samples overview. (XLSX 54 kb)

Supplementary Table 2

This table contains the detected somatic mutations. (XLSX 154 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Peifer, M., Hertwig, F., Roels, F. et al. Telomerase activation by genomic rearrangements in high-risk neuroblastoma. Nature 526, 700–704 (2015). https://doi.org/10.1038/nature14980

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature14980

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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