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.

Whole-genome landscape of pancreatic neuroendocrine tumours

A Corrigendum to this article was published on 27 September 2017

Abstract

The diagnosis of pancreatic neuroendocrine tumours (PanNETs) is increasing owing to more sensitive detection methods, and this increase is creating challenges for clinical management. We performed whole-genome sequencing of 102 primary PanNETs and defined the genomic events that characterize their pathogenesis. Here we describe the mutational signatures they harbour, including a deficiency in G:C > T:A base excision repair due to inactivation of MUTYH, which encodes a DNA glycosylase. Clinically sporadic PanNETs contain a larger-than-expected proportion of germline mutations, including previously unreported mutations in the DNA repair genes MUTYH, CHEK2 and BRCA2. Together with mutations in MEN1 and VHL, these mutations occur in 17% of patients. Somatic mutations, including point mutations and gene fusions, were commonly found in genes involved in four main pathways: chromatin remodelling, DNA damage repair, activation of mTOR signalling (including previously undescribed EWSR1 gene fusions), and telomere maintenance. In addition, our gene expression analyses identified a subgroup of tumours associated with hypoxia and HIF signalling.

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: Mutational signatures in pancreatic neuroendocrine tumours.
Figure 2: EWSR1 gene fusions in pancreatic neuroendocrine tumours.
Figure 3: Mutational processes in pancreatic neuroendocrine tumours.
Figure 4: Core pathways in PanNETs.

References

  1. Bosman, F. T., Carneiro, F., Hruban, R. H. & Theise, N. D. WHO Classification of Tumours of the Digestive System 4th edn (International Agency for Research on Cancer, 2010)

  2. Corbo, V. et al. MEN1 in pancreatic endocrine tumors: analysis of gene and protein status in 169 sporadic neoplasms reveals alterations in the vast majority of cases. Endocr. Relat. Cancer 17, 771–783 (2010)

    Article  CAS  PubMed  Google Scholar 

  3. Jiao, Y. et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 331, 1199–1203 (2011)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  4. Missiaglia, E. et al. Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. J. Clin. Oncol. 28, 245–255 (2010)

    Article  CAS  PubMed  Google Scholar 

  5. Neychev, V. et al. Mutation-targeted therapy with sunitinib or everolimus in patients with advanced low-grade or intermediate-grade neuroendocrine tumours of the gastrointestinal tract and pancreas with or without cytoreductive surgery: protocol for a phase II clinical trial. BMJ Open 5, e008248 (2015)

    Article  PubMed  PubMed Central  Google Scholar 

  6. Elsässer, S. J., Allis, C. D. & Lewis, P. W. Cancer. New epigenetic drivers of cancers. Science 331, 1145–1146 (2011)

    Article  ADS  PubMed  Google Scholar 

  7. Heaphy, C. M. et al. Altered telomeres in tumors with ATRX and DAXX mutations. Science 333, 425 (2011)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  8. Marinoni, I. et al. Loss of DAXX and ATRX are associated with chromosome instability and reduced survival of patients with pancreatic neuroendocrine tumors. Gastroenterology 146, 453–460 (2014)

    Article  CAS  PubMed  Google Scholar 

  9. Song, S. et al. qpure: A tool to estimate tumor cellularity from genome-wide single-nucleotide polymorphism profiles. PLoS One 7, e45835 (2012)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  10. Nones, K. et al. Genomic catastrophes frequently arise in esophageal adenocarcinoma and drive tumorigenesis. Nat. Commun . 5, 5224 (2014)

    Article  CAS  ADS  PubMed  Google Scholar 

  11. Waddell, N. et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 518, 495–501 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Popova, T. et al. Genome Alteration Print (GAP): a tool to visualize and mine complex cancer genomic profiles obtained by SNP arrays. Genome Biol . 10, R128 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Mermel, C. H. et al. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol . 12, R41 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Nik-Zainal, S. et al. Mutational processes molding the genomes of 21 breast cancers. Cell 149, 979–993 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Nik-Zainal, S. et al. Landscape of somatic mutations in 560 breast cancer whole-genome sequences. Nature 534, 47–54 (2016)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  17. Patch, A. M. et al. Whole-genome characterization of chemoresistant ovarian cancer. Nature 521, 489–494 (2015)

    Article  CAS  ADS  PubMed  Google Scholar 

  18. Al-Tassan, N. et al. Inherited variants of MYH associated with somatic G:C-->T:A mutations in colorectal tumors. Nat. Genet . 30, 227–232 (2002)

    Article  CAS  PubMed  Google Scholar 

  19. Aretz, S. et al. MUTYH-associated polyposis (MAP): evidence for the origin of the common European mutations p.Tyr179Cys and p.Gly396Asp by founder events. Eur. J. Hum. Genet. 22, 923–929 (2014)

    Article  CAS  PubMed  Google Scholar 

  20. Vogt, S. et al. Expanded extracolonic tumor spectrum in MUTYH-associated polyposis. Gastroenterology 137, 1976–1985 (2009)

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  MathSciNet  PubMed  PubMed Central  Google Scholar 

  22. Rausch, T. et al. Genome sequencing of pediatric medulloblastoma links catastrophic DNA rearrangements with TP53 mutations. Cell 148, 59–71 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Georgitsi, M. et al. Germline CDKN1B/p27Kip1 mutation in multiple endocrine neoplasia. J. Clin. Endocrinol. Metab. 92, 3321–3325 (2007)

    Article  CAS  PubMed  Google Scholar 

  24. Francis, J. M. et al. Somatic mutation of CDKN1B in small intestine neuroendocrine tumors. Nat. Genet . 45, 1483–1486 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lubensky, I. A. et al. Multiple neuroendocrine tumors of the pancreas in von Hippel-Lindau disease patients: histopathological and molecular genetic analysis. Am. J. Pathol . 153, 223–231 (1998)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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 

  27. Gonzalez-Perez, A. et al. IntOGen-mutations identifies cancer drivers across tumor types. Nat. Methods 10, 1081–1082 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med . 366, 883–892 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Li, B. E. et al. Distinct pathways regulated by menin and by MLL1 in hematopoietic stem cells and developing B cells. Blood 122, 2039–2046 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Tang, M. et al. The malignant brain tumor (MBT) domain protein SFMBT1 is an integral histone reader subunit of the LSD1 demethylase complex for chromatin association and epithelial-to-mesenchymal transition. J. Biol. Chem. 288, 27680–27691 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Asai, A. et al. High-resolution 400K oligonucleotide array comparative genomic hybridization analysis of neurofibromatosis type 1-associated cutaneous neurofibromas. Gene 558, 220–226 (2015)

    Article  CAS  PubMed  Google Scholar 

  32. Baba, T. et al. Persephin: A potential key component in human oral cancer progression through the RET receptor tyrosine kinase-mitogen-activated protein kinase signaling pathway. Mol. Carcinog . 54, 608–617 (2015)

    Article  CAS  PubMed  Google Scholar 

  33. Lindahl, M. et al. Human glial cell line-derived neurotrophic factor receptor alpha 4 is the receptor for persephin and is predominantly expressed in normal and malignant thyroid medullary cells. J. Biol. Chem. 276, 9344–9351 (2001)

    Article  CAS  PubMed  Google Scholar 

  34. Alers, S., Löffler, A. S., Wesselborg, S. & Stork, B. Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: cross talk, shortcuts, and feedbacks. Mol. Cell. Biol . 32, 2–11 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sturm, D. et al. New brain tumor entities emerge from molecular classification of CNS-PNETs. Cell 164, 1060–1072 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. 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  ADS  PubMed  Google Scholar 

  37. May, W. A. et al. Ewing sarcoma 11;22 translocation produces a chimeric transcription factor that requires the DNA-binding domain encoded by FLI1 for transformation. Proc. Natl Acad. Sci. USA 90, 5752–5756 (1993)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  38. Sankar, S. & Lessnick, S. L. Promiscuous partnerships in Ewing’s sarcoma. Cancer Genet . 204, 351–365 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Stockman, D. L. et al. Malignant gastrointestinal neuroectodermal tumor: clinicopathologic, immunohistochemical, ultrastructural, and molecular analysis of 16 cases with a reappraisal of clear cell sarcoma-like tumors of the gastrointestinal tract. Am. J. Surg. Pathol. 36, 857–868 (2012)

    Article  PubMed  PubMed Central  Google Scholar 

  40. 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 

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

    Article  CAS  MathSciNet  PubMed  Google Scholar 

  42. 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 

  43. Lovejoy, C. A. et al. Loss of ATRX, genome instability, and an altered DNA damage response are hallmarks of the alternative lengthening of telomeres pathway. PLoS Genet . 8, e1002772 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sadanandam, A. et al. A cross-species analysis in pancreatic neuroendocrine tumors reveals molecular subtypes with distinctive clinical, metastatic, developmental, and metabolic characteristics. Cancer Discov . 5, 1296–1313 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Lin, S. Y. & Elledge, S. J. Multiple tumor suppressor pathways negatively regulate telomerase. Cell 113, 881–889 (2003)

    Article  CAS  PubMed  Google Scholar 

  46. Bar-Peled, L. et al. A Tumor suppressor complex with GAP activity for the Rag GTPases that signal amino acid sufficiency to mTORC1. Science 340, 1100–1106 (2013)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  47. Fang, M. et al. MEN1 is a melanoma tumor suppressor that preserves genomic integrity by stimulating transcription of genes that promote homologous recombination-directed DNA repair. Mol. Cell. Biol . 33, 2635–2647 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Wang, Y. et al. The tumor suppressor protein menin inhibits AKT activation by regulating its cellular localization. Cancer Res . 71, 371–382 (2011)

    Article  CAS  PubMed  Google Scholar 

  49. Matkar, S., Thiel, A. & Hua, X. Menin: a scaffold protein that controls gene expression and cell signaling. Trends Biochem. Sci . 38, 394–402 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Landrum M. J. et al. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res . 44, D862–D868 (2016)

    Article  CAS  PubMed  Google Scholar 

  51. Biankin, A. V. et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491, 399–405 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Lawrence, M. S. et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 505, 495–501 (2014)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  53. Kambara, T. et al. Role of inherited defects of MYH in the development of sporadic colorectal cancer. Genes Chromosom. Cancer 40, 1–9 (2004)

    Article  CAS  PubMed  Google Scholar 

  54. O’Callaghan, N. J. & Fenech, M. A quantitative PCR method for measuring absolute telomere length. Biol. Proced. Online 13, 3 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Bailey, P. et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 531, 47–52 (2016)

    Article  CAS  ADS  PubMed  Google Scholar 

  56. Wilkerson, M. D. & Hayes, D. N. ConsensusClusterPlus: a class discovery tool with confidence assessments and item tracking. Bioinformatics 26, 1572–1573 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Law, C. W., Chen, Y., Shi, W. & Smyth, G. K. voom: Precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol . 15, R29 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Fang, H. & Gough, J. The ‘dnet’ approach promotes emerging research on cancer patient survival. Genome Med . 6, 64 (2014)

    PubMed  PubMed Central  Google Scholar 

  59. Lewis, T. B., Coffin, C. M. & Bernard, P. S. Differentiating Ewing’s sarcoma from other round blue cell tumors using a RT-PCR translocation panel on formalin-fixed paraffin-embedded tissues. Mod. Pathol . 20, 397–404 (2007)

    Article  CAS  PubMed  Google Scholar 

  60. Rossi, S. et al. EWSR1-CREB1 and EWSR1-ATF1 fusion genes in angiomatoid fibrous histiocytoma. Clin. Cancer Res . 13, 7322–7328 (2007)

    Article  CAS  PubMed  Google Scholar 

  61. Cai, Z., Chehab, N. H. & Pavletich, N. P. Structure and activation mechanism of the CHK2 DNA damage checkpoint kinase. Mol. Cell 35, 818–829 (2009)

    Article  CAS  PubMed  Google Scholar 

  62. Krieger, E., Koraimann, G. & Vriend, G. Increasing the precision of comparative models with YASARA NOVA--a self-parameterizing force field. Proteins 47, 393–402 (2002)

    Article  CAS  PubMed  Google Scholar 

  63. Bell, D. W. et al. Genetic and functional analysis of CHEK2 (CHK2) variants in multiethnic cohorts. Int. J. Cancer 121, 2661–2667 (2007)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank E. Missiaglia, S. Beghelli, N. Sperandio, G. Bonizzato, S. Grimaldi, F. Pisani, C. Cantù, G. Zamboni and P. Merlini for assistance at the ARC-Net Research Centre and Verona University; C. Axford, M.-A. Brancato, S. Rowe, M. Thomas, S. Simpson and G. Hammond for central coordination of the Australian Pancreatic Cancer Genome Initiative, data management and quality control; M. Martyn-Smith, L. Braatvedt, H. Tang, V. Papangelis and M. Beilin for biospecimen acquisition; D. Gwynne and D. Stetner for support at the Queensland Centre for Medical Genomics; and The Kinghorn Centre for Clinical Genomics for genome sequencing of validation samples. Funding support was from: Italian Ministry of Research (Cancer Genome Project FIRB RBAP10AHJB); Associazione Italiana Ricerca Cancro (AIRC n. 12182); Fondazione Italiana Malattie Pancreas – Ministero Salute (CUP_J33G13000210001); National Health and Medical Research Council of Australia (NHMRC; 631701, 535903, CDF 1112113, PRF 1025427, SRF 455857, 535903); The Queensland State Government Smart State National and International Research Alliances Program (NIRAP); Institute for Molecular Bioscience/University of Queensland; The Royal Australasian College of Physicians, Sidney Catalyst, NHMRC, Pancare Australia; Australian Government: Department of Innovation, Industry, Science and Research (DIISR); Australian Cancer Research Foundation (ACRF); Cancer Council NSW (SRP06-01, SRP11-01. ICGC); Cancer Institute NSW (10/ECF/2-26; 06/ECF/1-24; 09/CDF/2-40; 07/CDF/1-03; 10/CRF/1-01, 08/RSA/1-15, 07/CDF/1-28, 10/CDF/2-26,10/FRL/2-03, 06/RSA/1-05, 09/RIG/1-02, 10/TPG/1-04, 11/REG/1-10, 11/CDF/3-26); Garvan Institute of Medical Research; Avner Nahmani Pancreatic Cancer Research Foundation; R.T. Hall Trust; Petre Foundation; Philip Hemstritch Foundation; Gastroenterological Society of Australia (GESA Senior Research Fellowship); Royal Australasian College of Surgeons (RACS); Royal Australasian College of Physicians (RACP); Royal College of Pathologists of Australasia (RCPA); QIMR Berghofer Medical Research; The Keith Boden Fellowship (K.N.); NHGRI U54 HG003273; CPRIT grant RP101353-P7; Wellcome Trust Senior Investigator Award (103721/Z/14/Z); CRUK Programme (C29717/A17263 and C29717/A18484); CRUK Glasgow Centre (C596/A18076); CRUK Clinical Training Award (C596/A20921); Pancreatic Cancer UK Future Research Leaders Fund; The Howat Foundation; and the University of Glasgow.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

Biospecimens were collected at affiliated hospitals and processed at each biospecimen core resource centre. Investigator contributions are as follows: A.S., D.K.C., Nicola W., A.V.B., S.M.G. (concept and design); A.S., D.K.C., Nicola W., A.V.B., S.M.G. (project leaders); A.S., D.K.C., K.N., V.Co., Nicola W., A.V.B., S.M.G. (writing team); K.N., A.-M.P., P.B., R.T.L., A.L.J., B.R., S.C., M.C.J.Q, P.J.W., S.H.N., I.D., A.P.D.T, M.V.D., L.L., A.Mal., M.M., M.D.J., J.Hu., L.A.C., V.Ch., A.M.N., M.Pa., M.Pi., C.J.S., A.P., I.R., C.T., V.Ch, A.Maw., E.S.H., E.K.C., A.C., J.A.L., N.B.J., F.D., M.C.G., J.S.S., N.D.M., K.E., N.Q.N., N.Z., M.Fal., M.Fas., G.B., S.P., W.E.F., A. Malp., A. Maw., G.V.B., D.A.W., R.A.G., E.A.M., A.B., C.B., G.T., P.P., A.V.B. (sample collection, processing, quality control & clinical annotation); A.S., D.K.C., J.G.K., A.J.G, A.V.B. (clinico-pathological analyses and interpretation); V.L.J.W., B.A.L. (colon sample collection and clinical annotation); A.S., B.R., I.C., P.C., J.G.K., M.Fas., A.J.G (pathology assessment); V.Co., D.K.M., M.Sc., M.Si., D.A., C.V., T.J.C.B., A.N.C., I.H., S.I., S.McL., C.N., E.N., E.A., S.Be., M.Si. (sequencing); O.H., R.A.D., L.M.S.L., M.L., H.A.P., R.R.R., J.V.P. (telomere analysis); K.N., A.M.P., P.B., R.T.L., A.L.J., A.Maf., S.Ba., K.O.S., S.S., M.C.J.Q., P.J.W., M.J.A., J.L.F., F.N., Nick W., O.H., S.H.K., C.L., S.W., Q.X., J.W., M.Pi., M.C., J.V.P., Nicola W., S.M.G. (bioinformatics); K.K.K, J.Ha. (protein modelling); JLH., K.K.K (functional validation of CHEK2 variants); A.P.D.T. (revision of fusion cases and FISH analysis); A.S., D.K.C., K.N., V.Co., P.B., P.P., N.B.J., F.D., Nicola W., S.M.G., A.V.B. (data interpretation). All authors have read and approved the final manuscript.

Corresponding authors

Correspondence to Aldo Scarpa, Andrew V. Biankin or Sean M. Grimmond.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks S. Chanock and the other anonymous reviewer(s) for their contribution to the peer review of this work.

A list of participants and their affiliations is provided in the Supplementary Information.

Extended data figures and tables

Extended Data Figure 1 Flow chart of the experiments performed on 160 PanNETs.

The chart shows the workflow of analyses conducted on the discovery set of 98 PanNETs and on the validation set of an additional 62 PanNETs and 1 colorectal cancer. CNA, copy-number analysis.

Extended Data Figure 2 Five mutation signatures in pancreatic neuroendocrine tumours.

a, Stability plot indicates there are five mutation signatures (>0.9). b, The profile of the five mutational signatures (A–E) and what function has been assigned to these signatures (MUTYH, APOBEC, BRCA, Age and ‘Signature 5’).

Extended Data Figure 3 Validation of the novel signature in additional MUTYH carriers.

a, Four PanNet samples, three of which harboured a pathogenic MUTYH germline variant, and a colon tumour with a pathogenic MUTYH mutation underwent WGS to validate the association of MUTYH biallelic inactivation with the MUTYH mutation signature. b, Family pedigree of the patient with colon cancer. The 64-year-old male patient with colon cancer was identified as a candidate for MUTYH mutation analysis owing to the presence of two synchronous cancers in the proximal colon, each arising in a contiguous tubulovillous adenoma, as well as approximately 50 adenomatous polyps predominantly in the caecum and ascending colon. The index patient’s brother presented with colorectal cancer at 45 years of age and his sister presented with colorectal cancer at 64 years of age and with breast cancer at 59 years of age. The index patient’s son had polyps removed at 36 years of age. Mutation signature analysis was performed using the 98 discovery PanNET samples and the colon and 4 PanNET validation samples. c, Stability plot showing the solution for the five mutational signatures (>0.75). d, The profile of the five mutational signatures (A–E) and what function has been assigned to these signatures (MUTYH, APOBEC, BRCA, Age and ‘Signature 5’). e, The contribution of each signature (mutations per Mb) and proportion of the signatures in each tumour are shown.

Extended Data Figure 4 Structural rearrangements in pancreatic neuroendocrine tumours.

a, Top, the number and type of somatic structural rearrangements in each tumour. Bottom, tumours with more events tended to have longer telomeres. b, Two methods were used to determine clusters of somatic structural rearrangement breakpoints. Orange squares, chromosomes with a significant cluster of events as determined by a goodness-of-fit test against the expected distribution (P < 0.0001, Kolmogorov–Smirnov test). Blue squares, chromosomes deemed to harbour a high number of breakpoints because they had a chromosomal breakpoint per Mb rate that exceeded the 75th percentile of the chromosomal breakpoint per Mb rate for the cohort by five times the interquartile range. Red squares, chromosomes for which both of these criteria were met. Clusters of events were reviewed and nine tumours were found to harbour regions of chromothripsis. c, Recurrent chromothripsis for chromosome 11 was detected in four tumours. The chromothripsis event caused loss of the MEN1 gene locus in two of these samples.

Extended Data Figure 5 Functional analysis of CHEK2 variants.

a, CHEK2 structure indicating the positions of the germline variants. Mutations are highlighted by rendering as magenta sticks with protein domains coloured as indicated in the adjacent keys. The model includes a superimposed phosphopeptide (red). b, A summary of the CHEK2 variants and their predicted impact on protein structure. To functionally test the CHEK2 variants, a panel of FLAG–CHEK2 constructs encoding P85L, ∆77–82, D177H and E282K was generated. c, d, FLAG western blot of transfected HEK293T whole cell lysates (c) or anti-FLAG immunoprecipitates (d) showed that, compared to the wild type, there was normal expression of P85L but reduced expression of ∆77–82, D177H and E282K. e, Assessment of kinase activity of CHEK2 variants. Immunoprecipitated proteins were incubated either with GST alone (−) or with GST–CDC25C amino acids 200–256 (+) in the presence of γ-P32 ATP. Input and kinase activity were assessed by film radiography (top) and coomassie staining (bottom). Immunoprecipitates of ∆77–82, D177H and E282K had significantly reduced kinase activity in terms of both autophosphorylation and phosphorylation of CDC25C whereas the activity of P85L was normal. f, Quantification of expression levels by western blotting expressed as a fraction of wild type. Data points represent independent experiments. Error bars are mean ± s.e.m. g, h, Quantification of kinase activity. P32 counts for CDC25C (g) and CHEK2 (h) bands were scintillation counted. Corresponding bands from untransfected controls were used for background subtraction. Background-corrected P32 counts per minute were then standardized to wild type for each experiment. Data points represent independent experiments. Error bars are mean ± s.e.m. i, j, Quantification of kinase activity relative to protein expression. Kinase activity (from i and j) was standardized to protein expression level (from f). D177H was not analysed in this manner owing to its very low expression level. Error bars are mean ± s.e.m. Once the low expression level of ∆77–82 is taken into account, it is evident that the expressed protein retains normal kinase activity. On the other hand, E282K is kinase defective even after adjusting for its reduced expression. D177H expression is so low that it is not possible to reliably correct kinase activity for relative expression level, so it is unclear whether D177H is kinase dead as well as unstable. Data are summarized in Supplementary Table 16.

Extended Data Figure 6 Recurrently mutated genes in pancreatic neuroendocrine tumours.

a, The number of SNVs and indels within the genome of each patient (n = 98) is shown in the histogram. The driver plot displays the somatic mutations in key genes or those identified as significantly mutated (Intogen Q < 0.1). SETD2 is also reported, although its Q value was 0.15, as it was recurrently inactivated in six samples and multiple independent deleterious SETD2 mutations were observed in one tumour (a nonsense present at 3%, a missense at 14%, and a frameshift at 11%; only the nonsense is shown but the case is highlighted with a black arrow), suggesting strong selection for SETD2 inactivation in that tumour. b, Somatic mutations in MEN1 are predominantly nonsense mutations or insertions–deletions causing frame shifts and premature protein termination, and occur throughout the protein.

Extended Data Figure 7 Genome characteristics of PanNETs.

Copy number was determined using Illumina SNP arrays in a cohort of 98 PanNETs. a, Copy number events were mainly comprised of whole chromosome arm loss or gain. Cluster analysis of the chromosome arm level copy number state stratified the tumours into four subtypes. Group 1: recurrent pattern of whole chromosomal loss, affecting specific chromosomes (1, 2, 3, 6, 8, 10, 11, 15, 16 and 22); group 2: samples with a limited number of events, many with loss affecting chromosome 11; group 3: polyploid tumours, with gain of all chromosomes; and group 4: aneuploid tumours, containing predominantly whole chromosome gains affecting multiple chromosomes). b, The proportion of bases within the genome affected by copy number change. c, The mutations per Mb (SNPs and small insertion deletions). d, GISTIC analysis showing recurrent gains (red) and losses (blue) of the entire cohort.

Extended Data Figure 8 Telomere length is associated with somatic mutations.

Whole genome sequence data were used to estimate telomere length in PanNETs relative to the matched normal sample. a, Telomere length estimated by whole-genome sequencing correlated with the telomere length calculated from qPCR (R2 = 0.8091). Values are plotted on a log10 scale. be, Boxplots were used to show the association of relative telomere length and DAXX or ATRX and MEN1 mutation status. Mann–Whitney tests were used to determine significant associations (P < 0.05). b, c, Tumours harbouring DAXX or ATRX mutations contain longer telomeres. d, Tumours harbouring MEN1 mutations contain longer telomeres. e, Telomere length is shown in relation to DAXX or ATRX and MEN1 somatic mutations.

Extended Data Figure 9 RNA-seq of PanNET tumours.

Unsupervised clustering, network and gene enrichment analysis for available RNA-seq data identify PanNET subgroups associated with hypoxia and metabolic reprogramming. a, Unsupervised clustering identified three distinct PanNET subgroups (1–3). b, A gene signature defining three expression groups previously described in PanNETs showed enrichment of expression of the intermediate-group genes43 in Group 1 and the metastasis-like PanNET (MLP) genes43 in Group 3. c, Network analysis identified a significant sub-network of genes differentially expressed between Group 3 and other groups (Group 1 and Group 2). Red nodes represent genes upregulated in Group 3 and green nodes represent genes upregulated in other groups. Shaded areas represent network communities. d, Gene enrichment analysis for genes belonging to the sub-network shown in b. e, Heatmap showing the differential expression of genes belong to the identified sub-network. Somatic mutations in some of the recurrently mutated genes are shown (MEN1, DAXX, ATRX and members of the mTOR pathway: DEPDC5, MTOR, PTEN, TSC1 and TSC2).

Extended Data Figure 10 Genomic events associated with outcome.

Kaplan–Meier survival curves. a, b, Tumours harbouring DAXX or ATRX mutations had a poor prognosis in the whole cohort (a) and in the G2 cohort (b). c, Tumours with telomere lengths that were neither short or long had a better prognosis. d, Tumours harbouring mutations in genes that activate the mTOR pathway had a poor prognosis in the G2 cohort (log rank test was used in all instances).

Supplementary information

Supplementary Information

This file contains a list of the participants and their affiliations for the Australian Pancreatic Cancer Genome Initiative. (PDF 134 kb)

Supplementary Data

This file contains Supplementary Tables 1-16. (XLSX 4587 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Scarpa, A., Chang, D., Nones, K. et al. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature 543, 65–71 (2017). https://doi.org/10.1038/nature21063

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature21063

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