Pan-cancer analysis of whole genomes (original) (raw)

Data availability

The PCAWG-generated alignments, somatic variant calls, annotations and derived datasets are available for general research use for browsing and download at http://dcc.icgc.org/pcawg/ (Box 1 and Supplementary Table 4). In accordance with the data access policies of the ICGC and TCGA projects, most molecular, clinical and specimen data are in an open tier which does not require access approval. To access potentially identifying information, such as germline alleles and underlying read data, researchers will need to apply to the TCGA Data Access Committee (DAC) via dbGaP (https://dbgap.ncbi.nlm.nih.gov/aa/wga.cgi?page=login) for access to the TCGA portion of the dataset, and to the ICGC Data Access Compliance Office (DACO; http://icgc.org/daco) for the ICGC portion. In addition, to access somatic single nucleotide variants derived from TCGA donors, researchers will also need to obtain dbGaP authorization.

Beyond the core sequence data and variant call-sets, the analyses in this paper used a number of datasets that were derived from the variant calls (Supplementary Table 4). The individual datasets are available at Synapse (https://www.synapse.org/), and are denoted with synXXXXX accession numbers; all these datasets are also mirrored at https://dcc.icgc.org, with full links, filenames, accession numbers and descriptions detailed in Supplementary Table 4. The datasets encompass: clinical data from each patient including demographics, tumour stage and vital status (syn10389158); harmonized tumour histopathology annotations using a standardised hierarchical ontology (syn1038916); inferred purity and ploidy values for each tumour sample (syn8272483); driver mutations for each patient from their cancer genome spanning all classes of variant, and coding versus non-coding drivers (syn11639581); mutational signatures inferred from PCAWG donors (syn11804065), including APOBEC mutagenesis (syn7437313); and transcriptional data from RNA sequencing, including gene expression levels (syn5553985, syn5553991, syn8105922) and gene fusions (syn10003873, syn7221157).

Code availability

Computational pipelines for calling somatic mutations are available to the public at https://dockstore.org/organizations/PCAWG/collections/PCAWG. A range of data-visualization and -exploration tools are also available for the PCAWG data (Box 1).

Change history

A Correction to this paper has been published: https://doi.org/10.1038/s41586-022-05598-w

References

  1. Pleasance, E. D. et al. A comprehensive catalogue of somatic mutations from a human cancer genome. Nature 463, 191–196 (2010).
    Article ADS CAS Google Scholar
  2. Pleasance, E. D. et al. A small-cell lung cancer genome with complex signatures of tobacco exposure. Nature 463, 184–190 (2010).
    Article ADS CAS Google Scholar
  3. Ley, T. J. et al. DNA sequencing of a cytogenetically normal acute myeloid leukaemia genome. Nature 456, 66–72 (2008).
    Article ADS CAS Google Scholar
  4. Rheinbay, E. et al. Analyses of non-coding somatic drivers in 2,693 cancer whole genomes. Nature https://doi.org/10.1038/s41586-020-1965-x (2020).
  5. Alexandrov, L. B. et al. The repertoire of mutational signatures in human cancer. Nature https://doi.org/10.1038/s41586-020-1943-3 (2020).
  6. Li, Y. et al. Patterns of somatic structural variation in human cancer genomes. Nature https://doi.org/10.1038/s41586-019-1913-9 (2020).
  7. Gerstung, M. et al. The evolutionary history of 2,658 cancers. Nature https://doi.org/10.1038/s41586-019-1907-7 (2020).
  8. PCAWG Transcriptome Core Group et al. Genomic basis of RNA alterations in cancer. Nature https://doi.org/10.1038/s41586-020-1970-0 (2020).
  9. Zhang, Y. et al. High-coverage whole-genome analysis of 1,220 cancers reveals hundreds of genes deregulated by rearrangement-mediated _cis_-regulatory alterations. Nat. Commun. https://doi.org/10.1038/s41467-019-13885-w (2020).
  10. Rodriguez-Martin, B. et al. Pan-cancer analysis of whole genomes identifies driver rearrangements promoted by LINE-1 retrotransposition. Nat. Genet. https://doi.org/10.1038/s41588-019-0562-0 (2020).
  11. Zapatka, M. et al. The landscape of viral associations in human cancers. Nat. Genet. https://doi.org/10.1038/s41588-019-0558-9 (2020).
  12. Jiao, W. et al. A deep learning system can accurately classify primary and metastatic cancers based on patterns of passenger mutations. Nat. Commun. https://doi.org/10.1038/s41467-019-13825-8 (2020).
  13. Sieverling, L. et al. Genomic footprints of activated telomere maintenance mechanisms in cancer. Nat. Commun. https://doi.org/10.1038/s41467-019-13824-9 (2020).
  14. Yuan, Y. et al. Comprehensive molecular characterization of mitochondrial genomes in human cancers. Nat. Genet. https://doi.org/10.1038/s41588-019-0557-x (2020).
  15. Akdemir, K. C. et al. Chromatin folding domains disruptions by somatic genomic rearrangements in human cancers. Nat. Genet. https://doi.org/10.1038/s41588-019-0564-y (2020).
  16. Reyna, M. A. et al. Pathway and network analysis of more than 2,500 whole cancer genomes. Nat. Commun. https://doi.org/10.1038/s41467-020-14351-8 (2020).
  17. Bailey, M. H. et al. Retrospective evaluation of whole exome and genome mutation calls in 746 cancer samples. Nat. Commun. (2020).
  18. Cortes-Ciriano, I. et al. Comprehensive analysis of chromothripsis in 2,658 human cancers using whole-genome sequencing. Nat. Genet. https://doi.org/10.1038/s41588-019-0576-7 (2020).
  19. Bray, F., Ren, J.-S., Masuyer, E. & Ferlay, J. Global estimates of cancer prevalence for 27 sites in the adult population in 2008. Int. J. Cancer 132, 1133–1145 (2013).
    Article CAS Google Scholar
  20. Tarver, T. Cancer Facts & Figures 2012. American Cancer Society (ACS). J. Consum. Health Internet 16, 366–367 (2012).
    Article Google Scholar
  21. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).
    Article CAS Google Scholar
  22. International Cancer Genome Consortium. International network of cancer genome projects. Nature 464, 993–998 (2010).
    Article ADS Google Scholar
  23. Bailey, M. H. et al. Comprehensive characterization of cancer driver genes and mutations. Cell 173, 371–385 (2018).
    Article CAS Google Scholar
  24. Sanchez-Vega, F. et al. Oncogenic signaling pathways in The Cancer Genome Atlas. Cell 173, 321–337 (2018).
    Article CAS Google Scholar
  25. Hoadley, K. A. et al. Cell-of-origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancer. Cell 173, 291–304 (2018).
    Article CAS Google Scholar
  26. Stein, L. D., Knoppers, B. M., Campbell, P., Getz, G. & Korbel, J. O. Data analysis: create a cloud commons. Nature 523, 149–151 (2015).
    Article ADS CAS Google Scholar
  27. Phillips, M. et al. Genomics: data sharing needs international code of conduct. Nature https://doi.org/10.1038/d41586-020-00082-9 (2020).
  28. Krochmalski, J. Developing with Docker (Packt Publishing, 2016).
  29. Welch, J. S. et al. The origin and evolution of mutations in acute myeloid leukemia. Cell 150, 264–278 (2012).
    Article CAS Google Scholar
  30. Nik-Zainal, S. et al. Landscape of somatic mutations in 560 breast cancer whole-genome sequences. Nature 534, 47–54 (2016).
    Article ADS CAS Google Scholar
  31. Meier, B. et al. C. elegans whole-genome sequencing reveals mutational signatures related to carcinogens and DNA repair deficiency. Genome Res. 24, 1624–1636 (2014).
    Article CAS Google Scholar
  32. Martincorena, I. et al. Universal patterns of selection in cancer and somatic tissues. Cell 171, 1029–1041 (2017).
    Article CAS Google Scholar
  33. Tamborero, D. et al. Cancer Genome Interpreter annotates the biological and clinical relevance of tumor alterations. Genome Med. 10, 25 (2018).
    Article Google Scholar
  34. Huang, F. W. et al. Highly recurrent TERT promoter mutations in human melanoma. Science 339, 957–959 (2013).
    Article ADS CAS Google Scholar
  35. Rheinbay, E. et al. Recurrent and functional regulatory mutations in breast cancer. Nature 547, 55–60 (2017).
    Article ADS CAS Google Scholar
  36. Fredriksson, N. J., Ny, L., Nilsson, J. A. & Larsson, E. Systematic analysis of noncoding somatic mutations and gene expression alterations across 14 tumor types. Nat. Genet. 46, 1258–1263 (2014).
    Article CAS Google Scholar
  37. Horn, S. et al. TERT promoter mutations in familial and sporadic melanoma. Science 339, 959–961 (2013).
    Article ADS CAS Google Scholar
  38. Ciriello, G. et al. Emerging landscape of oncogenic signatures across human cancers. Nat. Genet. 45, 1127–1133 (2013).
    Article CAS Google Scholar
  39. Rahman, N. Realizing the promise of cancer predisposition genes. Nature 505, 302–308 (2014).
    Article ADS CAS Google Scholar
  40. Pearl, L. H., Schierz, A. C., Ward, S. E., Al-Lazikani, B. & Pearl, F. M. G. Therapeutic opportunities within the DNA damage response. Nat. Rev. Cancer 15, 166–180 (2015).
    Article CAS Google Scholar
  41. Taylor-Weiner, A. et al. DeTiN: overcoming tumor-in-normal contamination. Nat. Methods 15, 531–534 (2018).
    Article CAS Google Scholar
  42. Fujimoto, A. et al. Whole-genome mutational landscape and characterization of noncoding and structural mutations in liver cancer. Nat. Genet. 48, 500–509 (2016).
    Article CAS Google Scholar
  43. Shlush, L. I. Age-related clonal hematopoiesis. Blood 131, 496–504 (2018).
    Article CAS Google Scholar
  44. Northcott, P. A. et al. The whole-genome landscape of medulloblastoma subtypes. Nature 547, 311–317 (2017).
    Article ADS CAS Google Scholar
  45. Scarpa, A. et al. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature 543, 65–71 (2017).
    Article ADS CAS Google Scholar
  46. Davis, C. F. et al. The somatic genomic landscape of chromophobe renal cell carcinoma. Cancer Cell 26, 319–330 (2014).
    Article CAS Google Scholar
  47. Berger, M. F. et al. The genomic complexity of primary human prostate cancer. Nature 470, 214–220 (2011).
    Article ADS CAS Google Scholar
  48. Baca, S. C. et al. Punctuated evolution of prostate cancer genomes. Cell 153, 666–677 (2013).
    Article CAS Google Scholar
  49. Nik-Zainal, S. et al. The life history of 21 breast cancers. Cell 149, 994–1007 (2012).
    Article CAS Google Scholar
  50. Nik-Zainal, S. et al. Mutational processes molding the genomes of 21 breast cancers. Cell 149, 979–993 (2012).
    Article CAS Google Scholar
  51. Roberts, S. A. et al. Clustered mutations in yeast and in human cancers can arise from damaged long single-strand DNA regions. Mol. Cell 46, 424–435 (2012).
    Article CAS Google Scholar
  52. Rausch, T. et al. Genome sequencing of pediatric medulloblastoma links catastrophic DNA rearrangements with TP53 mutations. Cell 148, 59–71 (2012).
    Article CAS Google Scholar
  53. Stephens, P. J. et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 144, 27–40 (2011).
    Article CAS Google Scholar
  54. Korbel, J. O. & Campbell, P. J. Criteria for inference of chromothripsis in cancer genomes. Cell 152, 1226–1236 (2013).
    Article CAS Google Scholar
  55. Zhang, C.-Z. et al. Chromothripsis from DNA damage in micronuclei. Nature 522, 179–184 (2015).
    Article ADS CAS Google Scholar
  56. The Cancer Genome Atlas Research Network. Integrated genomic characterization of papillary thyroid carcinoma. Cell 159, 676–690 (2014).
    Article Google Scholar
  57. Supek, F. & Lehner, B. Clustered mutation signatures reveal that error-prone DNA repair targets mutations to active genes. Cell 170, 534–547 (2017).
    Article CAS Google Scholar
  58. Mardin, B. R. et al. A cell-based model system links chromothripsis with hyperploidy. Mol. Syst. Biol. 11, 828 (2015).
    Article Google Scholar
  59. Weischenfeldt, J. et al. Integrative genomic analyses reveal an androgen-driven somatic alteration landscape in early-onset prostate cancer. Cancer Cell 23, 159–170 (2013).
    Article CAS Google Scholar
  60. Garsed, D. W. et al. The architecture and evolution of cancer neochromosomes. Cancer Cell 26, 653–667 (2014).
    Article CAS Google Scholar
  61. Durinck, S. et al. Temporal dissection of tumorigenesis in primary cancers. Cancer Discov. 1, 137–143 (2011).
    Article CAS Google Scholar
  62. Hayward, N. K. et al. Whole-genome landscapes of major melanoma subtypes. Nature 545, 175–180 (2017).
    Article ADS CAS Google Scholar
  63. The Cancer Genome Atlas Network. Genomic classification of cutaneous melanoma. Cell 161, 1681–1696 (2015).
    Article Google Scholar
  64. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).
    Article CAS Google Scholar
  65. Chan, K. et al. An APOBEC3A hypermutation signature is distinguishable from the signature of background mutagenesis by APOBEC3B in human cancers. Nat. Genet. 47, 1067–1072 (2015).
    Article CAS Google Scholar
  66. Nik-Zainal, S. et al. Association of a germline copy number polymorphism of APOBEC3A and APOBEC3B with burden of putative APOBEC-dependent mutations in breast cancer. Nat. Genet. 46, 487–491 (2014).
    Article CAS Google Scholar
  67. Middlebrooks, C. D. et al. Association of germline variants in the APOBEC3 region with cancer risk and enrichment with APOBEC-signature mutations in tumors. Nat. Genet. 48, 1330–1338 (2016).
    Article CAS Google Scholar
  68. Westra, H.-J. et al. Systematic identification of trans eQTLs as putative drivers of known disease associations. Nat. Genet. 45, 1238–1243 (2013).
    Article CAS Google Scholar
  69. Stranger, B. E. et al. Population genomics of human gene expression. Nat. Genet. 39, 1217–1224 (2007).
    Article CAS Google Scholar
  70. Menghi, F. et al. The tandem duplicator phenotype as a distinct genomic configuration in cancer. Proc. Natl Acad. Sci. USA 113, E2373–E2382 (2016).
    Article CAS Google Scholar
  71. Hendrich, B., Hardeland, U., Ng, H. H., Jiricny, J. & Bird, A. The thymine glycosylase MBD4 can bind to the product of deamination at methylated CpG sites. Nature 401, 301–304 (1999).
    Article ADS CAS Google Scholar
  72. Lee, E. et al. Landscape of somatic retrotransposition in human cancers. Science 337, 967–971 (2012).
    Article ADS CAS Google Scholar
  73. Tubio, J. M. C. et al. Extensive transduction of nonrepetitive DNA mediated by L1 retrotransposition in cancer genomes. Science 345, 1251343–1251343 (2014).
    Article Google Scholar
  74. Helman, E. et al. Somatic retrotransposition in human cancer revealed by whole-genome and exome sequencing. Genome Res. 24, 1053–1063 (2014).
    Article CAS Google Scholar
  75. Shay, J. W. & Wright, W. E. Hayflick, his limit, and cellular ageing. Nat. Rev. Mol. Cell Biol. 1, 72–76 (2000).
    Article CAS Google Scholar
  76. Peifer, M. et al. Telomerase activation by genomic rearrangements in high-risk neuroblastoma. Nature 526, 700–704 (2015).
    Article ADS CAS Google Scholar
  77. Totoki, Y. et al. Trans-ancestry mutational landscape of hepatocellular carcinoma genomes. Nat. Genet. 46, 1267–1273 (2014).
    Article CAS Google Scholar
  78. Paterlini-Bréchot, P. et al. Hepatitis B virus-related insertional mutagenesis occurs frequently in human liver cancers and recurrently targets human telomerase gene. Oncogene 22, 3911–3916 (2003).
    Article Google Scholar
  79. Heaphy, C. M. et al. Prevalence of the alternative lengthening of telomeres telomere maintenance mechanism in human cancer subtypes. Am. J. Pathol. 179, 1608–1615 (2011).
    Article CAS Google Scholar
  80. Barthel, F. P. et al. Systematic analysis of telomere length and somatic alterations in 31 cancer types. Nat. Genet. 49, 349–357 (2017).
    Article CAS Google Scholar
  81. García-Cao, M., Gonzalo, S., Dean, D. & Blasco, M. A. A role for the Rb family of proteins in controlling telomere length. Nat. Genet. 32, 415–419 (2002).
    Article Google Scholar
  82. Tomasetti, C. & Vogelstein, B. Variation in cancer risk among tissues can be explained by the number of stem cell divisions. Science 347, 78–81 (2015).
    Article ADS CAS Google Scholar
  83. Gerstung, M. et al. Precision oncology for acute myeloid leukemia using a knowledge bank approach. Nat. Genet. 49, 332–340 (2017).
    Article CAS Google Scholar
  84. O’Connor, B. D. et al. The Dockstore: enabling modular, community-focused sharing of Docker-based genomics tools and workflows. F1000Res. 6, 52 (2017).
    Article Google Scholar
  85. Zhang, J. et al. The International Cancer Genome Consortium Data Portal. Nat. Biotechnol. 37, 367–369 (2019).
    Article CAS Google Scholar
  86. Miller, C. A., Qiao, Y., DiSera, T., D’Astous, B. & Marth, G. T. bam.iobio: a web-based, real-time, sequence alignment file inspector. Nat. Methods 11, 1189–1189 (2014).
    Article CAS Google Scholar
  87. Goldman, M. et al. The UCSC Xena platform for public and private cancer genomics data visualization and interpretation. Preprint at https://www.biorxiv.org/content/10.1101/326470v6 (2019).
  88. Papatheodorou, I. et al. Expression Atlas: gene and protein expression across multiple studies and organisms. Nucleic Acids Res. 46, D246–D251 (2018).
    Article CAS Google Scholar
  89. NCI SEER. ICD-O-3 Coding Materials (2018).
  90. Li, H. & Durbin, R. Fast and accurate long-read alignment with Burrows–Wheeler transform. Bioinformatics 26, 589–595 (2010).
    Article Google Scholar
  91. 1000 Genomes Project Consortium. A global reference for human genetic variation. Nature 526, 68–74 (2015).
    Article Google Scholar
  92. Raine, K. M. et al. ascatNgs: identifying somatically acquired copy-number alterations from whole-genome sequencing data. Curr. Protoc. Bioinformatics 56, 15.9.1–15.9.17 (2016).
    Article Google Scholar
  93. Jones, D. et al. cgpCaVEManWrapper: simple execution of CaVEMan in order to detect somatic single nucleotide variants in NGS data. Curr. Protoc. Bioinformatics 56, 15.10.1–15.10.18 (2016).
    Article Google Scholar
  94. Raine, K. M. et al. cgpPindel: identifying somatically acquired insertion and deletion events from paired end sequencing. Curr. Protoc. Bioinformatics 52, 15.7.1–15.7.12 (2015).
    Article Google Scholar
  95. 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 Google Scholar
  96. Rausch, T. et al. DELLY: structural variant discovery by integrated paired-end and split-read analysis. Bioinformatics 28, i333–i339 (2012).
    Article CAS Google Scholar
  97. Rimmer, A. et al. Integrating mapping-, assembly- and haplotype-based approaches for calling variants in clinical sequencing applications. Nat. Genet. 46, 912–918 (2014).
    Article CAS Google Scholar
  98. Cibulskis, K. et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 31, 213–219 (2013).
    Article CAS Google Scholar
  99. Carter, S. L. et al. Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol. 30, 413–421 (2012).
    Article CAS Google Scholar
  100. Drier, Y. et al. Somatic rearrangements across cancer reveal classes of samples with distinct patterns of DNA breakage and rearrangement-induced hypermutability. Genome Res. 23, 228–235 (2013).
    Article CAS Google Scholar
  101. Ramos, A. H. et al. Oncotator: cancer variant annotation tool. Hum. Mutat. 36, E2423–E2429 (2015).
    Article Google Scholar
  102. Moncunill, V. et al. Comprehensive characterization of complex structural variations in cancer by directly comparing genome sequence reads. Nat. Biotechnol. 32, 1106–1112 (2014).
    Article CAS Google Scholar
  103. Fan, Y. et al. MuSE: accounting for tumor heterogeneity using a sample-specific error model improves sensitivity and specificity in mutation calling from sequencing data. Genome Biol. 17, 178 (2016).
    Article Google Scholar
  104. Cooke, S. L. et al. Processed pseudogenes acquired somatically during cancer development. Nat. Commun. 5, 3644 (2014).
    Article Google Scholar
  105. Ju, Y. S. et al. Origins and functional consequences of somatic mitochondrial DNA mutations in human cancer. eLife 3, e02935 (2014).
    Article Google Scholar
  106. Sudmant, P. H. et al. An integrated map of structural variation in 2,504 human genomes. Nature 526, 75–81 (2015).
    Article CAS Google Scholar
  107. Garrison, E. & Marth, G. Haplotype-based variant detection from short-read sequencing. Preprint at https://arxiv.org/abs/1207.3907 (2012).
  108. DePristo, M. A. et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 43, 491–498 (2011).
    Article CAS Google Scholar
  109. Yakneen, S., Waszak, S. M., Gertz, M. & Korbel, J. O. & PCAWG Consortium. Butler enables rapid cloud-based analysis of thousands of human genomes. Nat. Biotechnol. https://doi.org/10.1038/s41587-019-0360-3 (2020).
  110. Kim, S. Y., Jacob, L. & Speed, T. P. Combining calls from multiple somatic mutation-callers. BMC Bioinformatics 15, 154 (2014).
    Article Google Scholar
  111. Breiman, L. Stacked regressions. Mach. Learn. 24, 49–64 (1996).
    Article MATH Google Scholar
  112. Campbell, P. J. et al. Identification of somatically acquired rearrangements in cancer using genome-wide massively parallel paired-end sequencing. Nat. Genet. 40, 722–729 (2008).
    Article CAS Google Scholar
  113. Wala, J. A. et al. SvABA: genome-wide detection of structural variants and indels by local assembly. Genome Res. 28, 581–591 (2018).
    Article CAS Google Scholar

Download references

Acknowledgements

We thank research participants who donated samples and data, the physicians and clinical staff who contributed to sample annotation and collection, and the numerous funding agencies that contributed to the collection and analysis of this dataset.

Author information

Author notes

  1. These authors jointly supervised this work: Peter J. Campbell, Gad Getz, Jan O. Korbel, Joshua M. Stuart, Lincoln D. Stein
  2. A list of members and their affiliations appears in the online version of the paper and lists of working groups appear in the Supplementary Information

Authors and Affiliations

  1. Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
    Lauri A. Aaltonen
  2. Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
    Federico Abascal, David J. Adams, Ludmil B. Alexandrov, Sam Behjati, Shriram G. Bhosle, David T. Bowen, Adam P. Butler, Peter J. Campbell, Peter Clapham, Helen Davies, Kevin J. Dawson, Stefan C. Dentro, Serge Serge, Erik Garrison, Mohammed Ghori, Dominik Glodzik, Jonathan Hinton, David R. Jones, Young Seok Ju, Stian Knappskog, Barbara Kremeyer, Henry Lee-Six, Daniel A. Leongamornlert, Yilong Li, Sancha Martin, Iñigo Martincorena, Ultan McDermott, Andrew Menzies, Thomas J. Mitchell, Sandro Morganella, Jyoti Nangalia, Jonathan Nicholson, Serena Nik-Zainal, Sarah O’Meara, Elli Papaemmanuil, Keiran M. Raine, Manasa Ramakrishna, Kamna Ramakrishnan, Nicola D. Roberts, Rebecca Shepherd, Lucy Stebbings, Michael R. Stratton, Maxime Tarabichi, Jon W. Teague, Ignacio Vázquez-García, David C. Wedge, Lucy Yates, Jorge Zamora & Xueqing Zou
  3. Memorial Sloan Kettering Cancer Center, New York, NY, USA
    Adam Abeshouse, Hikmat Al-Ahmadie, Gunes Gundem, Zachary Heins, Jason Huse, Douglas A. Levine, Eric Minwei Liu & Angelica Ochoa
  4. Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan
    Hiroyuki Aburatani, Genta Nagae, Akihiro Suzuki, Kenji Tatsuno & Shogo Yamamoto
  5. Department of Surgery, University of Chicago, Chicago, IL, USA
    Nishant Agrawal
  6. Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, School of Medicine, Keimyung University Dongsan Medical Center, Daegu, South Korea
    Keun Soo Ahn & Koo Jeong Kang
  7. Department of Oncology, Gil Medical Center, Gachon University, Incheon, South Korea
    Sung-Min Ahn
  8. Hiroshima University, Hiroshima, Japan
    Hiroshi Aikata, Koji Arihiro, Kazuaki Chayama, Yoshiiku Kawakami & Hideki Ohdan
  9. Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Rehan Akbani, Shaolong Cao, Yiwen Chen, Zechen Chong, Yu Fan, Jun Li, Han Liang, Wenyi Wang, Yumeng Wang & Yuan Yuan
  10. University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Kadir C. Akdemir & Ken Chen
  11. King Faisal Specialist Hospital and Research Centre, Al Maather, Riyadh, Saudi Arabia
    Sultan T. Al-Sedairy
  12. Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
    Fatima Al-Shahrour & Elena Piñeiro-Yáñez
  13. Bioinformatics Core Facility, University Medical Center Hamburg, Hamburg, Germany
    Malik Alawi
  14. Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
    Malik Alawi & Adam Grundhoff
  15. Ontario Tumour Bank, Ontario Institute for Cancer Research, Toronto, ON, Canada
    Monique Albert & John Bartlett
  16. Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Kenneth Aldape, Russell R. Broaddus, Bogdan Czerniak, Adel El-Naggar, Savitri Krishnamurthy, Alexander J. Lazar & Xiaoping Su
  17. Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
    Kenneth Aldape
  18. Department of Cellular and Molecular Medicine and Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
    Ludmil B. Alexandrov & Erik N. Bergstrom
  19. UC San Diego Moores Cancer Center, San Diego, CA, USA
    Ludmil B. Alexandrov, Erik N. Bergstrom & Olivier Harismendy
  20. Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
    Adrian Ally, Miruna Balasundaram, Reanne Bowlby, Denise Brooks, Rebecca Carlsen, Eric Chuah, Noreen Dhalla, Robert A. Holt, Steven J. M. Jones, Katayoon Kasaian, Darlene Lee, Haiyan Irene Li, Yussanne Ma, Marco A. Marra, Michael Mayo, Richard A. Moore, Andrew J. Mungall, Karen Mungall, A. Gordon Robertson, Sara Sadeghi, Jacqueline E. Schein, Payal Sipahimalani, Angela Tam, Nina Thiessen & Tina Wong
  21. Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
    Kathryn Alsop, David D. L. Bowtell, Elizabeth L. Christie, Dariush Etemadmoghadam, Sian Fereday, Dale W. Garsed, Linda Mileshkin, Chris Mitchell, Mark Shackleton, Heather Thorne & Nadia Traficante
  22. Centre for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
    Eva G. Alvarez, Alicia L. Bruzos, Bernardo Rodriguez-Martin, Javier Temes, Jose M. C. Tubio & Jorge Zamora
  23. Department of Zoology, Genetics and Physical Anthropology, (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
    Eva G. Alvarez, Alicia L. Bruzos, Bernardo Rodriguez-Martin, Javier Temes, Jose M. C. Tubio & Jorge Zamora
  24. The Biomedical Research Centre (CINBIO), Universidade de Vigo, Vigo, Spain
    Eva G. Alvarez, Alicia L. Bruzos, Bernardo Rodriguez-Martin, Marta Tojo, Jose M. C. Tubio & Jorge Zamora
  25. Royal National Orthopaedic Hospital - Bolsover, London, UK
    Fernanda Amary
  26. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Samirkumar B. Amin, P. Andrew Futreal & Alexander J. Lazar
  27. Quantitative and Computational Biosciences Graduate Program, Baylor College of Medicine, Houston, TX, USA
    Samirkumar B. Amin, Han Liang & Yumeng Wang
  28. The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
    Samirkumar B. Amin, Joshy George & Lucas Lochovsky
  29. Genome Informatics Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
    Brice Aminou, Niall J. Byrne, Aurélien Chateigner, Nodirjon Fayzullaev, Vincent Ferretti, George L. Mihaiescu, Hardeep K. Nahal-Bose, Brian D. O’Connor, B. F. Francis Ouellette, Marc D. Perry, Kevin Thai, Qian Xiang, Christina K. Yung & Junjun Zhang
  30. Institute of Human Genetics, Christian-Albrechts-University, Kiel, Germany
    Ole Ammerpohl, Andrea Haake, Cristina López, Julia Richter & Rabea Wagener
  31. Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
    Ole Ammerpohl, Sietse Aukema, Cristina López, Reiner Siebert & Rabea Wagener
  32. Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, University of Queensland, St. Lucia, Brisbane, QLD, Australia
    Matthew J. Anderson, Timothy J. C. Bruxner, Angelika N. Christ, J. Lynn Fink, Ivon Harliwong, Karin S. Kassahn, David K. Miller, Alan J. Robertson & Darrin F. Taylor
  33. Salford Royal NHS Foundation Trust, Salford, UK
    Yeng Ang, Hsiao-Wei Chen, Ritika Kundra & Francisco Sanchez-Vega
  34. Department of Surgery, Pancreas Institute, University and Hospital Trust of Verona, Verona, Italy
    Davide Antonello, Claudio Bassi, Narong Khuntikeo, Luca Landoni, Giuseppe Malleo, Giovanni Marchegiani, Neil D. Merrett, Marco Miotto, Salvatore Paiella, Antonio Pea, Paolo Pederzoli, Roberto Salvia, Jaswinder S. Samra, Elisabetta Sereni & Samuel Singer
  35. Molecular and Medical Genetics, OHSU Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
    Pavana Anur, Myron Peto & Paul T. Spellman
  36. Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, BC, Canada
    Samuel Aparicio
  37. The McDonnell Genome Institute at Washington University, St. Louis, MO, USA
    Elizabeth L. Appelbaum, Matthew H. Bailey, Matthew G. Cordes, Li Ding, Catrina C. Fronick, Lucinda A. Fulton, Robert S. Fulton, Kuan-lin Huang, Reyka Jayasinghe, Elaine R. Mardis, R. Jay Mashl, Michael D. McLellan, Christopher A. Miller, Heather K. Schmidt, Jiayin Wang, Michael C. Wendl, Richard K. Wilson & Tina Wong
  38. University College London, London, UK
    Elizabeth L. Appelbaum, Jonathan D. Kay, Helena Kilpinen, Laurence B. Lovat, Hayley J. Luxton & Hayley C. Whitaker
  39. Division of Cancer Genomics, National Cancer Center Research Institute, National Cancer Center, Tokyo, Japan
    Yasuhito Arai, Natsuko Hama, Fumie Hosoda, Hiromi Nakamura, Tatsuhiro Shibata, Yasushi Totoki & Shinichi Yachida
  40. DLR Project Management Agency, Bonn, Germany
    Axel Aretz
  41. Tokyo Women’s Medical University, Tokyo, Japan
    Shun-ichi Ariizumi & Masakazu Yamamoto
  42. Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
    Joshua Armenia, Hsiao-Wei Chen, Jianjiong Gao, Ritika Kundra, Francisco Sanchez-Vega, Nikolaus Schultz & Hongxin Zhang
  43. Los Alamos National Laboratory, Los Alamos, NM, USA
    Laurent Arnould
  44. Department of Pathology, University Health Network, Toronto General Hospital, Toronto, ON, Canada
    Sylvia Asa, Michael H. A. Roehrl & Theodorus Van der Kwast
  45. Nottingham University Hospitals NHS Trust, Nottingham, UK
    Sylvia Asa, Simon L. Parsons & Ming Tsao
  46. Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
    Yassen Assenov
  47. Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
    Gurnit Atwal, Philip Awadalla, Jonathan Barenboim, Vinayak Bhandari, Ivan Borozan, Paul C. Boutros, Lewis Jonathan Dursi, Shadrielle M. G. Espiritu, Natalie S. Fox, Michael Fraser, Syed Haider, Vincent Huang, Keren Isaev, Wei Jiao, Christopher M. Lalansingh, Emilie Lalonde, Fabien C. Lamaze, Constance H. Li, Julie Livingstone, Christine P’ng, Marta Paczkowska, Stephenie D. Prokopec, Jüri Reimand, Veronica Y. Sabelnykova, Adriana Salcedo, Yu-Jia Shiah, Solomon I. Shorser, Shimin Shuai, Jared T. Simpson, Lincoln D. Stein, Ren X. Sun, Lina Wadi, Gavin W. Wilson, Adam J. Wright, Takafumi N. Yamaguchi, Fouad Yousif & Denis Yuen
  48. Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
    Gurnit Atwal, Philip Awadalla, Gary D. Bader, Shimin Shuai & Lincoln D. Stein
  49. Vector Institute, Toronto, ON, Canada
    Gurnit Atwal, Quaid D. Morris, Yulia Rubanova & Jeffrey A. Wintersinger
  50. Hematopathology Section, Institute of Pathology, Christian-Albrechts-University, Kiel, Germany
    Sietse Aukema, Wolfram Klapper, Julia Richter & Monika Szczepanowski
  51. Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
    J. Todd Auman & Charles M. Perou
  52. Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
    Miriam R. R. Aure, Anne-Lise Børresen-Dale & Anita Langerød
  53. Pathology, Hospital Clinic, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
    Marta Aymerich
  54. Department of Veterinary Medicine, Transmissible Cancer Group, University of Cambridge, Cambridge, UK
    Adrian Baez-Ortega
  55. Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
    Matthew H. Bailey, Li Ding, Robert S. Fulton, Ramaswamy Govindan & Michael D. McLellan
  56. Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
    Peter J. Bailey, Andrew V. Biankin, David K. Chang, Susanna L. Cooke, Fraser R. Duthie, Janet S. Graham, Nigel B. Jamieson, Elizabeth A. Musgrove & Derek W. Wright
  57. Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
    Saianand Balu, Tom Bodenheimer, D. Neil Hayes, Austin J. Hepperla, Katherine A. Hoadley, Alan P. Hoyle, Stuart R. Jefferys, Shaowu Meng, Lisle E. Mose, Grant Sanders, Yan Shi, Janae V. Simons & Matthew G. Soloway
  58. Broad Institute of MIT and Harvard, Cambridge, MA, USA
    Pratiti Bandopadhayay, Rameen Beroukhim, Angela N. Brooks, Susan Bullman, John Busanovich, Andrew D. Cherniack, Juok Cho, Carrie Cibulskis, Kristian Cibulskis, David Craft, Timothy Defreitas, Andrew J. Dunford, Scott Frazer, Stacey B. Gabriel, Nils Gehlenborg, Gad Getz, Manaswi Gupta, Gavin Ha, Nicholas J. Haradhvala, David I. Heiman, Julian M. Hess, Manolis Kellis, Jaegil Kim, Kiran Kumar, Kirsten Kübler, Eric Lander, Michael S. Lawrence, Ignaty Leshchiner, Pei Lin, Ziao Lin, Dimitri Livitz, Yosef E. Maruvka, Samuel R. Meier, Matthew Meyerson, Michael S. Noble, Chandra Sekhar Pedamallu, Paz Polak, Esther Rheinbay, Daniel Rosebrock, Mara Rosenberg, Gordon Saksena, Richard Sallari, Steven E. Schumacher, Ayellet V. Segre, Ofer Shapira, Juliann Shih, Nasa Sinnott-Armstrong, Oliver Spiro, Chip Stewart, Amaro Taylor-Weiner, Grace Tiao, Douglas Voet, Jeremiah A. Wala, Cheng-Zhong Zhang & Hailei Zhang
  59. Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA, USA
    Pratiti Bandopadhayay
  60. Department of Pediatrics, Harvard Medical School, Boston, MA, USA
    Pratiti Bandopadhayay
  61. Leeds Institute of Medical Research @ St. James’s, University of Leeds, St. James’s University Hospital, Leeds, UK
    Rosamonde E. Banks & Naveen Vasudev
  62. Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
    Stefano Barbi, Vincenzo Corbo & Michele Simbolo
  63. Department of Surgery, Princess Alexandra Hospital, Brisbane, QLD, Australia
    Andrew P. Barbour
  64. Surgical Oncology Group, Diamantina Institute, University of Queensland, Brisbane, QLD, Australia
    Andrew P. Barbour
  65. Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
    Jill Barnholtz-Sloan
  66. Research Health Analytics and Informatics, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
    Jill Barnholtz-Sloan
  67. Gloucester Royal Hospital, Gloucester, UK
    Hugh Barr
  68. European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
    Elisabet Barrera, Wojciech Bazant, Ewan Birney, Rich Boyce, Alvis Brazma, Andy Cafferkey, Claudia Calabrese, Paul Flicek, Nuno A. Fonseca, Anja Füllgrabe, Moritz Gerstung, Santiago Gonzalez, Liliana Greger, Maria Keays, Jan O. Korbel, Alfonso Muñoz, Steven J. Newhouse, David Ocana, Irene Papatheodorou, Robert Petryszak, Roland F. Schwarz, Charles Short, Oliver Stegle & Lara Urban
  69. Diagnostic Development, Ontario Institute for Cancer Research, Toronto, ON, Canada
    John Bartlett & Ilinca Lungu
  70. Barcelona Supercomputing Center (BSC), Barcelona, Spain
    Javier Bartolome, Mattia Bosio, Ana Dueso-Barroso, J. Lynn Fink, Josep L. L. Gelpi, Ana Milovanovic, Montserrat Puiggròs, Javier Bartolomé Rodriguez, Romina Royo, David Torrents, Alfonso Valencia, Miguel Vazquez, David Vicente & Izar Villasante
  71. Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
    Oliver F. Bathe
  72. Departments of Surgery and Oncology, University of Calgary, Calgary, AB, Canada
    Oliver F. Bathe
  73. Department of Pathology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
    Daniel Baumhoer & Bodil Bjerkehagen
  74. PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON, Canada
    Prashant Bavi, Michelle Chan-Seng-Yue, Sean Cleary, Robert E. Denroche, Steven Gallinger, Robert C. Grant, Gun Ho Jang, Sangeetha Kalimuthu, Ilinca Lungu, John D. McPherson, Faiyaz Notta, Michael H. A. Roehrl, Gavin W. Wilson & Julie M. Wilson
  75. Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, MD, USA
    Stephen B. Baylin, Nilanjan Chatterjee, Leslie Cope, Ludmila Danilova & Ralph H. Hruban
  76. University Hospital Southampton NHS Foundation Trust, Southampton, UK
    Stephen B. Baylin & Tim Dudderidge
  77. Royal Stoke University Hospital, Stoke-on-Trent, UK
    Duncan Beardsmore & Christopher Umbricht
  78. Genome Sequence Informatics, Ontario Institute for Cancer Research, Toronto, ON, Canada
    Timothy A. Beck, Bob Gibson, Lawrence E. Heisler, Xuemei Luo & Morgan L. Taschuk
  79. Human Longevity Inc, San Diego, CA, USA
    Timothy A. Beck
  80. Olivia Newton-John Cancer Research Institute, La Trobe University, Heidelberg, VIC, Australia
    Andreas Behren & Jonathan Cebon
  81. Computer Network Information Center, Chinese Academy of Sciences, Beijing, China
    Beifang Niu
  82. Genome Canada, Ottawa, ON, Canada
    Cindy Bell
  83. CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
    Sergi Beltran, Ivo G. Gut, Marta Gut, Simon C. Heath, Tomas Marques-Bonet, Arcadi Navarro, Miranda D. Stobbe, Jean-Rémi Trotta & Justin P. Whalley
  84. Universitat Pompeu Fabra (UPF), Barcelona, Spain
    Sergi Beltran, Mattia Bosio, German M. Demidov, Oliver Drechsel, Ivo G. Gut, Marta Gut, Simon C. Heath, Francesc Muyas, Stephan Ossowski, Aparna Prasad, Raquel Rabionet, Miranda D. Stobbe & Hana Susak
  85. Buck Institute for Research on Aging, Novato, CA, USA
    Christopher Benz & Christina Yau
  86. Duke University Medical Center, Durham, NC, USA
    Andrew Berchuck
  87. Department of Human Genetics, Hannover Medical School, Hannover, Germany
    Anke K. Bergmann
  88. Center for Bioinformatics and Functional Genomics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
    Benjamin P. Berman & Huy Q. Dinh
  89. Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
    Benjamin P. Berman
  90. The Hebrew University Faculty of Medicine, Jerusalem, Israel
    Benjamin P. Berman
  91. Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
    Daniel M. Berney & Yong-Jie Lu
  92. Department of Computer Science, Bioinformatics Group, University of Leipzig, Leipzig, Germany
    Stephan H. Bernhart, Hans Binder, Steve Hoffmann & Peter F. Stadler
  93. Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany
    Stephan H. Bernhart, Hans Binder, Steve Hoffmann, Helene Kretzmer & Peter F. Stadler
  94. Transcriptome Bioinformatics, LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
    Stephan H. Bernhart, Steve Hoffmann, Helene Kretzmer & Peter F. Stadler
  95. Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
    Rameen Beroukhim, Angela N. Brooks, Susan Bullman, Andrew D. Cherniack, Levi Garraway, Matthew Meyerson, Chandra Sekhar Pedamallu, Steven E. Schumacher, Juliann Shih & Jeremiah A. Wala
  96. Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
    Rameen Beroukhim, Aquila Fatima, Andrea L. Richardson, Steven E. Schumacher, Ofer Shapira, Andrew Tutt & Jeremiah A. Wala
  97. Harvard Medical School, Boston, MA, USA
    Rameen Beroukhim, Gad Getz, Kirsten Kübler, Matthew Meyerson, Chandra Sekhar Pedamallu, Paz Polak, Esther Rheinbay & Jeremiah A. Wala
  98. USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
    Mario Berrios, Moiz S. Bootwalla, Andrea Holbrook, Phillip H. Lai, Dennis T. Maglinte, David J. Van Den Berg & Daniel J. Weisenberger
  99. Department of Diagnostics and Public Health, University and Hospital Trust of Verona, Verona, Italy
    Samantha Bersani, Ivana Cataldo, Claudio Luchini & Maria Scardoni
  100. Department of Mathematics, Aarhus University, Aarhus, Denmark
    Johanna Bertl & Asger Hobolth
  101. Department of Molecular Medicine (MOMA), Aarhus University Hospital, Aarhus N, Denmark
    Johanna Bertl, Henrik Hornshøj, Malene Juul, Randi Istrup Juul, Tobias Madsen, Morten Muhlig Nielsen & Jakob Skou Pedersen
  102. Instituto Carlos Slim de la Salud, Mexico City, Mexico
    Miguel Betancourt
  103. Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
    Vinayak Bhandari, Paul C. Boutros, Robert G. Bristow, Keren Isaev, Constance H. Li, Jüri Reimand, Michael H. A. Roehrl & Bradly G. Wouters
  104. Cancer Division, Garvan Institute of Medical Research, Kinghorn Cancer Centre, University of New South Wales (UNSW Sydney), Sydney, NSW, Australia
    Andrew V. Biankin, David K. Chang, Lorraine A. Chantrill, Angela Chou, Anthony J. Gill, Amber L. Johns, James G. Kench, David K. Miller, Adnan M. Nagrial, Marina Pajic, Mark Pinese, Ilse Rooman, Christopher J. Scarlett, Christopher W. Toon & Jianmin Wu
  105. South Western Sydney Clinical School, Faculty of Medicine, University of New South Wales (UNSW Sydney), Liverpool, NSW, Australia
    Andrew V. Biankin
  106. West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
    Andrew V. Biankin & Nigel B. Jamieson
  107. Center for Digital Health, Berlin Institute of Health and Charitè - Universitätsmedizin Berlin, Berlin, Germany
    Matthias Bieg
  108. Heidelberg Center for Personalized Oncology (DKFZ-HIPO), German Cancer Research Center (DKFZ), Heidelberg, Germany
    Matthias Bieg, Ivo Buchhalter, Barbara Hutter & Nagarajan Paramasivam
  109. The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA
    Darell Bigner
  110. Massachusetts General Hospital, Boston, MA, USA
    Michael Birrer, Vikram Deshpande, William C. Faquin, Nicholas J. Haradhvala, Kirsten Kübler, Michael S. Lawrence, David N. Louis, Yosef E. Maruvka, G. Petur Nielsen, Esther Rheinbay, Mara Rosenberg, Dennis C. Sgroi & Chin-Lee Wu
  111. National Institute of Biomedical Genomics, Kalyani, West Bengal, India
    Nidhan K. Biswas, Arindam Maitra & Partha P. Majumder
  112. Institute of Clinical Medicine and Institute of Oral Biology, University of Oslo, Oslo, Norway
    Bodil Bjerkehagen
  113. University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
    Lori Boice, Mei Huang, Sonia Puig & Leigh B. Thorne
  114. ARC-Net Centre for Applied Research on Cancer, University and Hospital Trust of Verona, Verona, Italy
    Giada Bonizzato, Cinzia Cantù, Ivana Cataldo, Vincenzo Corbo, Sonia Grimaldi, Rita T. Lawlor, Andrea Mafficini, Borislav C. Rusev, Aldo Scarpa, Katarzyna O. Sikora, Nicola Sperandio, Alain Viari & Caterina Vicentini
  115. The Institute of Cancer Research, London, UK
    Johann S. De Bono, Niedzica Camacho, Colin S. Cooper, Sandra E. Edwards, Rosalind A. Eeles, Zsofia Kote-Jarai, Daniel A. Leongamornlert, Lucy Matthews & Sue Merson
  116. Centre for Computational Biology, Duke-NUS Medical School, Singapore, Singapore
    Arnoud Boot, Ioana Cutcutache, Mi Ni Huang, John R. McPherson, Steven G. Rozen & Yang Wu
  117. Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
    Arnoud Boot, Ioana Cutcutache, Mi Ni Huang, John R. McPherson, Steven G. Rozen, Patrick Tan, Bin Tean Teh & Yang Wu
  118. Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
    Ake Borg, Markus Ringnér & Johan Staaf
  119. Department of Pediatric Oncology, Hematology and Clinical Immunology, Heinrich-Heine-University, Düsseldorf, Germany
    Arndt Borkhardt & Jessica I. Hoell
  120. Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
    Keith A. Boroevich, Todd A. Johnson, Michael S. Lawrence & Tatsuhiko Tsunoda
  121. RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
    Keith A. Boroevich, Akihiro Fujimoto, Masashi Fujita, Mayuko Furuta, Kazuhiro Maejima, Hidewaki Nakagawa, Kaoru Nakano & Aya Sasaki-Oku
  122. Department of Internal Medicine/Hematology, Friedrich-Ebert-Hospital, Neumünster, Germany
    Christoph Borst & Siegfried Haas
  123. Departments of Dermatology and Pathology, Yale University, New Haven, CT, USA
    Marcus Bosenberg
  124. Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
    Mattia Bosio, German M. Demidov, Oliver Drechsel, Georgia Escaramis, Xavier Estivill, Aliaksei Z. Holik, Francesc Muyas, Stephan Ossowski, Raquel Rabionet & Hana Susak
  125. Radcliffe Department of Medicine, University of Oxford, Oxford, UK
    Jacqueline Boultwood
  126. Canadian Center for Computational Genomics, McGill University, Montreal, QC, Canada
    Guillaume Bourque
  127. Department of Human Genetics, McGill University, Montreal, QC, Canada
    Guillaume Bourque, Mark Lathrop & Yasser Riazalhosseini
  128. Department of Human Genetics, University of California Los Angeles, Los Angeles, CA, USA
    Paul C. Boutros
  129. Department of Pharmacology, University of Toronto, Toronto, ON, Canada
    Paul C. Boutros
  130. Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere University Hospital, Tampere, Finland
    G. Steven Bova & Tapio Visakorpi
  131. Haematology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
    David T. Bowen
  132. Translational Research and Innovation, Centre Léon Bérard, Lyon, France
    Sandrine Boyault
  133. Fox Chase Cancer Center, Philadelphia, PA, USA
    Jeffrey Boyd & Elaine R. Mardis
  134. International Agency for Research on Cancer, World Health Organization, Lyon, France
    Paul Brennan & Ghislaine Scelo
  135. Earlham Institute, Norwich, UK
    Daniel S. Brewer & Colin S. Cooper
  136. Norwich Medical School, University of East Anglia, Norwich, UK
    Daniel S. Brewer & Colin S. Cooper
  137. Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, HB, The Netherlands
    Arie B. Brinkman
  138. CRUK Manchester Institute and Centre, Manchester, UK
    Robert G. Bristow
  139. Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
    Robert G. Bristow
  140. Division of Cancer Sciences, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
    Robert G. Bristow
  141. Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, ON, Canada
    Robert G. Bristow & Fei-Fei Fei Liu
  142. Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
    Jane E. Brock & Sabina Signoretti
  143. Department of Surgery, Division of Thoracic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
    Malcolm Brock
  144. Division of Molecular Pathology, The Netherlands Cancer Institute, Oncode Institute, Amsterdam, CX, The Netherlands
    Annegien Broeks & Jos Jonkers
  145. Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
    Angela N. Brooks, David Haan, Maximillian G. Marin, Thomas J. Matthew, Yulia Newton, Cameron M. Soulette & Joshua M. Stuart
  146. UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, USA
    Angela N. Brooks, Brian Craft, Mary J. Goldman, David Haussler, Joshua M. Stuart & Jingchun Zhu
  147. Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
    Benedikt Brors, Lars Feuerbach, Chen Hong, Charles David Imbusch & Lina Sieverling
  148. German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
    Benedikt Brors, Barbara Hutter, Peter Lichter, Dirk Schadendorf & Holger Sültmann
  149. National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
    Benedikt Brors, Barbara Hutter, Holger Sültmann & Thorsten Zenz
  150. Center for Biological Sequence Analysis, Department of Bio and Health Informatics, Technical University of Denmark, Lyngby, Denmark
    Søren Brunak
  151. Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
    Søren Brunak
  152. Institute for Molecular Bioscience, University of Queensland, St. Lucia, Brisbane, QLD, Australia
    Timothy J. C. Bruxner, Oliver Holmes, Stephen H. Kazakoff, Conrad R. Leonard, Felicity Newell, Katia Nones, Ann-Marie Patch, John V. Pearson, Michael C. Quinn, Nick M. Waddell, Nicola Waddell, Scott Wood & Qinying Xu
  153. Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
    Alex Buchanan & Kyle Ellrott
  154. Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
    Ivo Buchhalter, Calvin Wing Yiu Chan, Roland Eils, Michael C. Heinold, Carl Herrmann, Natalie Jäger, Rolf Kabbe, Jules N. A. Kerssemakers, Kortine Kleinheinz, Nagarajan Paramasivam, Manuel Prinz, Matthias Schlesner & Johannes Werner
  155. Institute of Pharmacy and Molecular Biotechnology and BioQuant, Heidelberg University, Heidelberg, Germany
    Ivo Buchhalter, Roland Eils, Michael C. Heinold, Carl Herrmann, Daniel Hübschmann, Kortine Kleinheinz & Umut H. Toprak
  156. Federal Ministry of Education and Research, Berlin, Germany
    Christiane Buchholz
  157. Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
    Hazel Burke, Ricardo De Paoli-Iseppi, Nicholas K. Hayward, Peter Hersey, Valerie Jakrot, Hojabr Kakavand, Georgina V. Long, Graham J. Mann, Robyn P. M. Saw, Richard A. Scolyer, Ping Shang, Andrew J. Spillane, Jonathan R. Stretch, John F. F. Thompson & James S. Wilmott
  158. Pediatric Hematology and Oncology, University Hospital Muenster, Muenster, Germany
    Birgit Burkhardt
  159. Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
    Kathleen H. Burns & Christopher Umbricht
  160. McKusick-Nathans Institute of Genetic Medicine, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, MD, USA
    Kathleen H. Burns
  161. Foundation Medicine, Inc, Cambridge, MA, USA
    John Busanovich
  162. Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
    Carlos D. Bustamante & Francisco M. De La Vega
  163. Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
    Carlos D. Bustamante, Francisco M. De La Vega, Suyash S. Shringarpure, Nasa Sinnott-Armstrong & Mark H. Wright
  164. Bakar Computational Health Sciences Institute and Department of Pediatrics, University of California, San Francisco, CA, USA
    Atul J. Butte & Jieming Chen
  165. Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
    Anne-Lise Børresen-Dale
  166. National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
    Samantha J. Caesar-Johnson, John A. Demchok, Ina Felau, Roy Tarnuzzer, Zhining Wang, Liming Yang, Jean C. Zenklusen & Jiashan Zhang
  167. Royal Marsden NHS Foundation Trust, London and Sutton, UK
    Declan Cahill, Nening M. Dennis, Tim Dudderidge, Rosalind A. Eeles, Cyril Fisher, Steven Hazell, Vincent Khoo, Pardeep Kumar, Naomi Livni, Erik Mayer, David Nicol, Christopher Ogden, Edward W. Rowe, Sarah Thomas, Alan Thompson & Nicholas van As
  168. Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
    Claudia Calabrese, Serap Erkek, Moritz Gerstung, Santiago Gonzalez, Nina Habermann, Wolfgang Huber, Lara Jerman, Jan O. Korbel, Esa Pitkänen, Benjamin Raeder, Tobias Rausch, Vasilisa A. Rudneva, Oliver Stegle, Stephanie Sungalee, Lara Urban, Sebastian M. Waszak, Joachim Weischenfeldt & Sergei Yakneen
  169. Department of Oncology, University of Cambridge, Cambridge, UK
    Carlos Caldas & Suet-Feung Chin
  170. Li Ka Shing Centre, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
    Carlos Caldas, Suet-Feung Chin, Ruben M. Drews, Paul A. Edwards, Matthew Eldridge, Steve Hawkins, Andy G. Lynch, Geoff Macintyre, Florian Markowetz, Charlie E. Massie, David E. Neal, Simon Tavaré & Ke Yuan
  171. Institut Gustave Roussy, Villejuif, France
    Fabien Calvo
  172. Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
    Peter J. Campbell, Vincent J. Gnanapragasam, William Howat, Thomas J. Mitchell, David E. Neal, Nimish C. Shah & Anne Y. Warren
  173. Department of Haematology, University of Cambridge, Cambridge, UK
    Peter J. Campbell
  174. Anatomia Patológica, Hospital Clinic, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
    Elias Campo
  175. Spanish Ministry of Science and Innovation, Madrid, Spain
    Elias Campo
  176. University of Michigan Comprehensive Cancer Center, Ann Arbor, MI, USA
    Thomas E. Carey
  177. Department for BioMedical Research, University of Bern, Bern, Switzerland
    Joana Carlevaro-Fita
  178. Department of Medical Oncology, Inselspital, University Hospital and University of Bern, Bern, Switzerland
    Joana Carlevaro-Fita, Rory Johnson & Andrés Lanzós
  179. Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
    Joana Carlevaro-Fita & Andrés Lanzós
  180. University of Pavia, Pavia, Italy
    Mario Cazzola & Luca Malcovati
  181. University of Alabama at Birmingham, Birmingham, AL, USA
    Robert Cerfolio
  182. UHN Program in BioSpecimen Sciences, Toronto General Hospital, Toronto, ON, Canada
    Dianne E. Chadwick, Sheng-Ben Liang, Michael H. A. Roehrl & Sagedeh Shahabi
  183. Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
    Dimple Chakravarty
  184. Centre for Law and Genetics, University of Tasmania, Sandy Bay Campus, Hobart, TAS, Australia
    Don Chalmers
  185. Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
    Calvin Wing Yiu Chan, Chen Hong & Lina Sieverling
  186. Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
    Kin Chan
  187. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA
    Vishal S. Chandan
  188. Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
    Stephen J. Chanock, Xing Hua, Lisa Mirabello, Lei Song & Bin Zhu
  189. Illawarra Shoalhaven Local Health District L3 Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, NSW, Australia
    Lorraine A. Chantrill
  190. BioForA, French National Institute for Agriculture, Food, and Environment (INRAE), ONF, Orléans, France
    Aurélien Chateigner
  191. Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
    Nilanjan Chatterjee
  192. University of California San Diego, San Diego, CA, USA
    Zhaohong Chen, Michelle T. Dow, Claudiu Farcas, S. M. Ashiqul Islam, Antonios Koures, Lucila Ohno-Machado, Christos Sotiriou & Ashley Williams
  193. Division of Experimental Pathology, Mayo Clinic, Rochester, MN, USA
    Jeremy Chien
  194. Centre for Cancer Research, The Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
    Yoke-Eng Chiew, Angela Chou, Jillian A. Hung, Catherine J. Kennedy, Graham J. Mann, Gulietta M. Pupo, Sarah-Jane Schramm, Varsha Tembe & Anna deFazio
  195. Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
    Yoke-Eng Chiew, Jillian A. Hung, Catherine J. Kennedy & Anna deFazio
  196. PDXen Biosystems Inc, Seoul, South Korea
    Sunghoon Cho
  197. Korea Advanced Institute of Science and Technology, Daejeon, South Korea
    Jung Kyoon Choi, Young Seok Ju & Christopher J. Yoon
  198. Electronics and Telecommunications Research Institute, Daejeon, South Korea
    Wan Choi, Seung-Hyup Jeon, Hyunghwan Kim & Youngchoon Woo
  199. Institut National du Cancer (INCA), Boulogne-Billancourt, France
    Christine Chomienne & Iris Pauporté
  200. Department of Genetics, Informatics Institute, University of Alabama at Birmingham, Birmingham, AL, USA
    Zechen Chong
  201. Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
    Su Pin Choo
  202. Medical Oncology, University and Hospital Trust of Verona, Verona, Italy
    Sara Cingarlini & Michele Milella
  203. Department of Pediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
    Alexander Claviez
  204. Hepatobiliary/Pancreatic Surgical Oncology Program, University Health Network, Toronto, ON, Canada
    Sean Cleary, Ashton A. Connor & Steven Gallinger
  205. School of Biological Sciences, University of Auckland, Auckland, New Zealand
    Nicole Cloonan
  206. Department of Surgery, University of Melbourne, Parkville, VIC, Australia
    Marek Cmero
  207. The Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC, Australia
    Marek Cmero
  208. Walter and Eliza Hall Institute, Parkville, VIC, Australia
    Marek Cmero
  209. Vancouver Prostate Centre, Vancouver, Canada
    Colin C. Collins, Nilgun Donmez, Faraz Hach, Salem Malikic, S. Cenk Sahinalp, Iman Sarrafi & Raunak Shrestha
  210. Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
    Ashton A. Connor, Steven Gallinger, Robert C. Grant, Treasa A. McPherson & Iris Selander
  211. University of East Anglia, Norwich, UK
    Colin S. Cooper
  212. Norfolk and Norwich University Hospital NHS Trust, Norwich, UK
    Matthew G. Cordes, Catrina C. Fronick & Tom Roques
  213. Victorian Institute of Forensic Medicine, Southbank, VIC, Australia
    Stephen M. Cordner
  214. Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
    Isidro Cortés-Ciriano, Jake June-Koo Lee & Peter J. Park
  215. Department of Chemistry, Centre for Molecular Science Informatics, University of Cambridge, Cambridge, UK
    Isidro Cortés-Ciriano
  216. Ludwig Center at Harvard Medical School, Boston, MA, USA
    Isidro Cortés-Ciriano, Jake June-Koo Lee & Peter J. Park
  217. Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
    Kyle Covington, HarshaVardhan Doddapaneni, Richard A. Gibbs, Jianhong Hu, Joy C. Jayaseelan, Viktoriya Korchina, Lora Lewis, Donna M. Muzny, Linghua Wang, David A. Wheeler & Liu Xi
  218. Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
    Prue A. Cowin, Anne Hamilton, Gisela Mir Arnau & Ravikiran Vedururu
  219. Physics Division, Optimization and Systems Biology Lab, Massachusetts General Hospital, Boston, MA, USA
    David Craft
  220. Department of Medicine, Baylor College of Medicine, Houston, TX, USA
    Chad J. Creighton
  221. University of Cologne, Cologne, Germany
    Yupeng Cun, Martin Peifer & Tsun-Po Yang
  222. International Genomics Consortium, Phoenix, AZ, USA
    Erin Curley & Troy Shelton
  223. Genomics Research Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
    Karolina Czajka, Jenna Eagles, Thomas J. Hudson, Jeremy Johns, Faridah Mbabaali, John D. McPherson, Jessica K. Miller, Danielle Pasternack, Michelle Sam & Lee E. Timms
  224. Barking Havering and Redbridge University Hospitals NHS Trust, Romford, UK
    Bogdan Czerniak, Adel El-Naggar & David Khoo
  225. Children’s Hospital at Westmead, University of Sydney, Sydney, NSW, Australia
    Rebecca A. Dagg
  226. Department of Medicine, Section of Endocrinology, University and Hospital Trust of Verona, Verona, Italy
    Maria Vittoria Davi
  227. Computational Biology Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
    Natalie R. Davidson, Andre Kahles, Kjong-Van Lehmann, Alessandro Pastore, Gunnar Rätsch, Chris Sander, Yasin Senbabaoglu & Nicholas D. Socci
  228. Department of Biology, ETH Zurich, Zürich, Switzerland
    Natalie R. Davidson, Andre Kahles, Kjong-Van Lehmann, Gunnar Rätsch & Stefan G. Stark
  229. Department of Computer Science, ETH Zurich, Zurich, Switzerland
    Natalie R. Davidson, Andre Kahles, Kjong-Van Lehmann & Gunnar Rätsch
  230. SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
    Natalie R. Davidson, Andre Kahles, Kjong-Van Lehmann, Gunnar Rätsch & Stefan G. Stark
  231. Weill Cornell Medical College, New York, NY, USA
    Natalie R. Davidson, Bishoy M. Faltas & Gunnar Rätsch
  232. Academic Department of Medical Genetics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
    Helen Davies & Serena Nik-Zainal
  233. MRC Cancer Unit, University of Cambridge, Cambridge, UK
    Helen Davies, Rebecca C. Fitzgerald, Nicola Grehan, Serena Nik-Zainal & Maria O’Donovan
  234. Departments of Pediatrics and Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
    Ian J. Davis
  235. Seven Bridges Genomics, Charlestown, MA, USA
    Brandi N. Davis-Dusenbery, Sinisa Ivkovic, Milena Kovacevic, Ana Mijalkovic Lazic, Sanja Mijalkovic, Mia Nastic, Petar Radovic & Nebojsa Tijanic
  236. Annai Systems, Inc, Carlsbad, CA, USA
    Francisco M. De La Vega, Tal Shmaya & Dai-Ying Wu
  237. Department of Pathology, General Hospital of Treviso, Department of Medicine, University of Padua, Treviso, Italy
    Angelo P. Dei Tos
  238. Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
    Olivier Delaneau
  239. Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, CH, Switzerland
    Olivier Delaneau
  240. Swiss Institute of Bioinformatics, University of Geneva, Geneva, CH, Switzerland
    Olivier Delaneau
  241. The Francis Crick Institute, London, UK
    Jonas Demeulemeester, Stefan C. Dentro, Matthew W. Fittall, Kerstin Haase, Clemency Jolly, Maxime Tarabichi & Peter Van Loo
  242. University of Leuven, Leuven, Belgium
    Jonas Demeulemeester & Peter Van Loo
  243. Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
    German M. Demidov, Francesc Muyas & Stephan Ossowski
  244. Computational and Systems Biology, Genome Institute of Singapore, Singapore, Singapore
    Deniz Demircioğlu & Jonathan Göke
  245. School of Computing, National University of Singapore, Singapore, Singapore
    Deniz Demircioğlu
  246. Big Data Institute, Li Ka Shing Centre, University of Oxford, Oxford, UK
    Stefan C. Dentro & David C. Wedge
  247. Biomedical Data Science Laboratory, Francis Crick Institute, London, UK
    Nikita Desai
  248. Bioinformatics Group, Department of Computer Science, University College London, London, UK
    Nikita Desai
  249. The Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada
    Amit G. Deshwar
  250. Breast Cancer Translational Research Laboratory JC Heuson, Institut Jules Bordet, Brussels, Belgium
    Christine Desmedt
  251. Department of Oncology, Laboratory for Translational Breast Cancer Research, KU Leuven, Leuven, Belgium
    Christine Desmedt
  252. Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
    Jordi Deu-Pons, Joan Frigola, Abel Gonzalez-Perez, Ferran Muiños, Loris Mularoni, Oriol Pich, Iker Reyes-Salazar, Carlota Rubio-Perez, Radhakrishnan Sabarinathan & David Tamborero
  253. Research Program on Biomedical Informatics, Universitat Pompeu Fabra, Barcelona, Spain
    Jordi Deu-Pons, Abel Gonzalez-Perez, Ferran Muiños, Loris Mularoni, Oriol Pich, Carlota Rubio-Perez, Radhakrishnan Sabarinathan & David Tamborero
  254. Division of Medical Oncology, Princess Margaret Cancer Centre, Toronto, ON, Canada
    Neesha C. Dhani, David Hedley & Malcolm J. Moore
  255. Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
    Priyanka Dhingra, Ekta Khurana, Eric Minwei Liu & Alexander Martinez-Fundichely
  256. Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
    Priyanka Dhingra, Ekta Khurana, Eric Minwei Liu & Alexander Martinez-Fundichely
  257. Department of Pathology, UPMC Shadyside, Pittsburgh, PA, USA
    Rajiv Dhir
  258. Independent Consultant, Wellesley, USA
    Anthony DiBiase
  259. Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
    Klev Diamanti, Jan Komorowski & Husen M. Umer
  260. Department of Medicine and Department of Genetics, Washington University School of Medicine, St. Louis, St. Louis, MO, USA
    Li Ding, Robert S. Fulton, Michael D. McLellan, Michael C. Wendl & Venkata D. Yellapantula
  261. Hefei University of Technology, Anhui, China
    Shuai Ding & Shanlin Yang
  262. Translational Cancer Research Unit, GZA Hospitals St.-Augustinus, Center for Oncological Research, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
    Luc Dirix, Steven Van Laere, Gert G. Van den Eynden & Peter Vermeulen
  263. Simon Fraser University, Burnaby, BC, Canada
    Nilgun Donmez, Ermin Hodzic, Salem Malikic, S. Cenk Sahinalp & Iman Sarrafi
  264. University of Pennsylvania, Philadelphia, PA, USA
    Ronny Drapkin
  265. Faculty of Science and Technology, University of Vic—Central University of Catalonia (UVic-UCC), Vic, Spain
    Ana Dueso-Barroso
  266. The Wellcome Trust, London, UK
    Michael Dunn
  267. The Hospital for Sick Children, Toronto, ON, Canada
    Lewis Jonathan Dursi
  268. Department of Pathology, Queen Elizabeth University Hospital, Glasgow, UK
    Fraser R. Duthie
  269. Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
    Ken Dutton-Regester, Nicholas K. Hayward, Oliver Holmes, Peter A. Johansson, Stephen H. Kazakoff, Conrad R. Leonard, Felicity Newell, Katia Nones, Ann-Marie Patch, John V. Pearson, Antonia L. Pritchard, Michael C. Quinn, Paresh Vyas, Nicola Waddell, Scott Wood & Qinying Xu
  270. Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
    Douglas F. Easton
  271. Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
    Douglas F. Easton
  272. Prostate Cancer Canada, Toronto, ON, Canada
    Stuart Edmonds
  273. University of Cambridge, Cambridge, UK
    Paul A. Edwards, Anthony R. Green, Andy G. Lynch, Florian Markowetz & Thomas J. Mitchell
  274. Department of Laboratory Medicine, Translational Cancer Research, Lund University Cancer Center at Medicon Village, Lund University, Lund, Sweden
    Anna Ehinger
  275. Heidelberg University, Heidelberg, Germany
    Juergen Eils, Roland Eils & Daniel Hübschmann
  276. New BIH Digital Health Center, Berlin Institute of Health (BIH) and Charité - Universitätsmedizin Berlin, Berlin, Germany
    Juergen Eils, Roland Eils & Chris Lawerenz
  277. CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
    Georgia Escaramis
  278. Research Group on Statistics, Econometrics and Health (GRECS), UdG, Barcelona, Spain
    Georgia Escaramis
  279. Quantitative Genomics Laboratories (qGenomics), Barcelona, Spain
    Xavier Estivill
  280. Icelandic Cancer Registry, Icelandic Cancer Society, Reykjavik, Iceland
    Jorunn E. Eyfjord, Holmfridur Hilmarsdottir & Jon G. Jonasson
  281. State Key Laboratory of Cancer Biology, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Shaanxi, China
    Daiming Fan & Yongzhan Nie
  282. Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy
    Matteo Fassan
  283. Rigshospitalet, Copenhagen, Denmark
    Francesco Favero
  284. Center for Cancer Genomics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
    Martin L. Ferguson
  285. Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC, Canada
    Vincent Ferretti
  286. Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
    Matthew A. Field
  287. Department of Neuro-Oncology, Istituto Neurologico Besta, Milano, Italy
    Gaetano Finocchiaro
  288. Bioplatforms Australia, North Ryde, NSW, Australia
    Anna Fitzgerald & Catherine A. Shang
  289. Department of Pathology (Research), University College London Cancer Institute, London, UK
    Adrienne M. Flanagan
  290. Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, ON, Canada
    Neil E. Fleshner
  291. Department of Medical Oncology, Josephine Nefkens Institute and Cancer Genomics Centre, Erasmus Medical Center, Rotterdam, CN, The Netherlands
    John A. Foekens, John W. M. Martens, F. Germán Rodríguez-González, Anieta M. Sieuwerts & Marcel Smid
  292. The University of Queensland Thoracic Research Centre, The Prince Charles Hospital, Brisbane, QLD, Australia
    Kwun M. Fong
  293. CIBIO/InBIO - Research Center in Biodiversity and Genetic Resources, Universidade do Porto, Vairão, Portugal
    Nuno A. Fonseca
  294. HCA Laboratories, London, UK
    Christopher S. Foster
  295. University of Liverpool, Liverpool, UK
    Christopher S. Foster
  296. The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
    Milana Frenkel-Morgenstern
  297. Department of Neurosurgery, University of Florida, Gainesville, FL, USA
    William Friedman
  298. Department of Pathology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
    Masashi Fukayama & Tetsuo Ushiku
  299. University of Milano Bicocca, Monza, Italy
    Carlo Gambacorti-Passerini
  300. BGI-Shenzhen, Shenzhen, China
    Shengjie Gao, Yong Hou, Chang Li, Lin Li, Siliang Li, Xiaobo Li, Xinyue Li, Dongbing Liu, Xingmin Liu, Qiang Pan-Hammarström, Hong Su, Jian Wang, Kui Wu, Heng Xiong, Huanming Yang, Chen Ye, Xiuqing Zhang, Yong Zhou & Shida Zhu
  301. Department of Pathology, Oslo University Hospital Ulleval, Oslo, Norway
    Øystein Garred
  302. Center for Biomedical Informatics, Harvard Medical School, Boston, MA, USA
    Nils Gehlenborg
  303. Department Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain
    Josep L. L. Gelpi
  304. Office of Cancer Genomics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
    Daniela S. Gerhard
  305. Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
    Clarissa Gerhauser, Christoph Plass & Dieter Weichenhan
  306. Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Jeffrey E. Gershenwald
  307. Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Jeffrey E. Gershenwald
  308. Department of Computer Science, Yale University, New Haven, CT, USA
    Mark Gerstein & Fabio C. P. Navarro
  309. Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
    Mark Gerstein, Sushant Kumar, Lucas Lochovsky, Shaoke Lou, Patrick D. McGillivray, Fabio C. P. Navarro, Leonidas Salichos & Jonathan Warrell
  310. Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
    Mark Gerstein, Arif O. Harmanci, Sushant Kumar, Donghoon Lee, Shantao Li, Xiaotong Li, Lucas Lochovsky, Shaoke Lou, William Meyerson, Leonidas Salichos, Jonathan Warrell, Jing Zhang & Yan Zhang
  311. Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
    Gad Getz & Paz Polak
  312. Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
    Gad Getz
  313. Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
    Ronald Ghossein, Dilip D. Giri, Christine A. Iacobuzio-Donahue, Jorge Reis-Filho & Victor Reuter
  314. Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
    Nasra H. Giama, Catherine D. Moser & Lewis R. Roberts
  315. University of Sydney, Sydney, NSW, Australia
    Anthony J. Gill & James G. Kench
  316. University of Oxford, Oxford, UK
    Pelvender Gill, Freddie C. Hamdy, Katalin Karaszi, Adam Lambert, Luke Marsden, Clare Verrill & Paresh Vyas
  317. Department of Surgery, Academic Urology Group, University of Cambridge, Cambridge, UK
    Vincent J. Gnanapragasam
  318. Department of Medicine II, University of Würzburg, Wuerzburg, Germany
    Maria Elisabeth Goebler
  319. Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
    Carmen Gomez
  320. Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), Barcelona, Spain
    Abel Gonzalez-Perez
  321. Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences (NIEHS), Durham, NC, USA
    Dmitry A. Gordenin & Natalie Saini
  322. St. Thomas’s Hospital, London, UK
    James Gossage
  323. Osaka International Cancer Center, Osaka, Japan
    Kunihito Gotoh
  324. Department of Pathology, Skåne University Hospital, Lund University, Lund, Sweden
    Dorthe Grabau
  325. Department of Medical Oncology, Beatson West of Scotland Cancer Centre, Glasgow, UK
    Janet S. Graham
  326. National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
    Eric Green, Carolyn M. Hutter & Heidi J. Sofia
  327. Centre for Cancer Research, Victorian Comprehensive Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
    Sean M. Grimmond
  328. Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
    Robert L. Grossman
  329. German Center for Infection Research (DZIF), Partner Site Hamburg-Borstel-Lübeck-Riems, Hamburg, Germany
    Adam Grundhoff
  330. Bioinformatics Research Centre (BiRC), Aarhus University, Aarhus, Denmark
    Qianyun Guo, Asger Hobolth & Jakob Skou Pedersen
  331. Department of Biotechnology, Ministry of Science and Technology, Government of India, New Delhi, Delhi, India
    Shailja Gupta & K. VijayRaghavan
  332. National Cancer Centre Singapore, Singapore, Singapore
    Jonathan Göke
  333. Brandeis University, Waltham, MA, USA
    James E. Haber
  334. Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
    Faraz Hach
  335. Department of Internal Medicine, Stanford University, Stanford, CA, USA
    Mark P. Hamilton
  336. The University of Texas Health Science Center at Houston, Houston, TX, USA
    Leng Han, Yang Yang & Xuanping Zhang
  337. Imperial College NHS Trust, Imperial College, London, INY, UK
    George B. Hanna
  338. Senckenberg Institute of Pathology, University of Frankfurt Medical School, Frankfurt, Germany
    Martin Hansmann
  339. Department of Medicine, Division of Biomedical Informatics, UC San Diego School of Medicine, San Diego, CA, USA
    Olivier Harismendy
  340. Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center, Houston, TX, USA
    Arif O. Harmanci
  341. Oxford Nanopore Technologies, New York, NY, USA
    Eoghan Harrington & Sissel Juul
  342. Institute of Medical Science, University of Tokyo, Tokyo, Japan
    Takanori Hasegawa, Shuto Hayashi, Seiya Imoto, Mitsuhiro Komura, Satoru Miyano, Naoki Miyoshi, Kazuhiro Ohi, Eigo Shimizu, Yuichi Shiraishi, Hiroko Tanaka & Rui Yamaguchi
  343. Howard Hughes Medical Institute, University of California Santa Cruz, Santa Cruz, CA, USA
    David Haussler
  344. Wakayama Medical University, Wakayama, Japan
    Shinya Hayami, Masaki Ueno & Hiroki Yamaue
  345. Department of Internal Medicine, Division of Medical Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
    D. Neil Hayes
  346. University of Tennessee Health Science Center for Cancer Research, Memphis, TN, USA
    D. Neil Hayes
  347. Department of Histopathology, Salford Royal NHS Foundation Trust, Salford, UK
    Stephen J. Hayes
  348. Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
    Stephen J. Hayes
  349. BIOPIC, ICG and College of Life Sciences, Peking University, Beijing, China
    Yao He & Zemin Zhang
  350. Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
    Yao He & Zemin Zhang
  351. Children’s Hospital of Philadelphia, Philadelphia, PA, USA
    Allison P. Heath
  352. Department of Bioinformatics and Computational Biology and Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Apurva M. Hegde, Yiling Lu & John N. Weinstein
  353. Karolinska Institute, Stockholm, Sweden
    Eva Hellstrom-Lindberg & Jesper Lagergren
  354. The Donnelly Centre, University of Toronto, Toronto, ON, Canada
    Mohamed Helmy & Jeffrey A. Wintersinger
  355. Department of Medical Genetics, College of Medicine, Hallym University, Chuncheon, South Korea
    Seong Gu Heo, Eun Pyo Hong & Ji Wan Park
  356. Department of Experimental and Health Sciences, Institute of Evolutionary Biology (UPF-CSIC), Universitat Pompeu Fabra, Barcelona, Spain
    José María Heredia-Genestar, Tomas Marques-Bonet & Arcadi Navarro
  357. Health Data Science Unit, University Clinics, Heidelberg, Germany
    Carl Herrmann
  358. Massachusetts General Hospital Center for Cancer Research, Charlestown, MA, USA
    Julian M. Hess & Yosef E. Maruvka
  359. Hokkaido University, Sapporo, Japan
    Satoshi Hirano & Toru Nakamura
  360. Department of Pathology and Clinical Laboratory, National Cancer Center Hospital, Tokyo, Japan
    Nobuyoshi Hiraoka
  361. Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
    Katherine A. Hoadley & Tara J. Skelly
  362. Computational Biology, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
    Steve Hoffmann
  363. University of Melbourne Centre for Cancer Research, Melbourne, VIC, Australia
    Oliver Hofmann
  364. University of Nebraska Medical Center, Omaha, NE, USA
    Michael A. Hollingsworth & Sarah P. Thayer
  365. Syntekabio Inc, Daejeon, South Korea
    Jongwhi H. Hong
  366. Department of Pathology, Academic Medical Center, Amsterdam, AZ, The Netherlands
    Gerrit K. Hooijer
  367. China National GeneBank-Shenzhen, Shenzhen, China
    Yong Hou, Chang Li, Siliang Li, Xiaobo Li, Dongbing Liu, Xingmin Liu, Henk G. Stunnenberg, Hong Su, Kui Wu, Heng Xiong, Chen Ye & Shida Zhu
  368. Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
    Volker Hovestadt, Murat Iskar, Peter Lichter, Bernhard Radlwimmer & Marc Zapatka
  369. Division of Life Science and Applied Genomics Center, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
    Taobo Hu, Yogesh Kumar, Eric Z. Ma, Zhenggang Wu & Hong Xue
  370. Icahn School of Medicine at Mount Sinai, New York, NY, USA
    Kuan-lin Huang
  371. Geneplus-Shenzhen, Shenzhen, China
    Yi Huang
  372. School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, China
    Yi Huang, Jiayin Wang, Xiao Xiao & Xuanping Zhang
  373. AbbVie, North Chicago, IL, USA
    Thomas J. Hudson
  374. Institute of Pathology, Charité – University Medicine Berlin, Berlin, Germany
    Michael Hummel & Dido Lenze
  375. Centre for Translational and Applied Genomics, British Columbia Cancer Agency, Vancouver, BC, Canada
    David Huntsman
  376. Edinburgh Royal Infirmary, Edinburgh, UK
    Ted R. Hupp
  377. Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
    Matthew R. Huska, Julia Markowski & Roland F. Schwarz
  378. Department of Pediatric Immunology, Hematology and Oncology, University Hospital, Heidelberg, Germany
    Daniel Hübschmann
  379. German Cancer Research Center (DKFZ), Heidelberg, Germany
    Daniel Hübschmann, Christof von Kalle & Roland F. Schwarz
  380. Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany
    Daniel Hübschmann
  381. Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, USA
    Marcin Imielinski
  382. New York Genome Center, New York, NY, USA
    Marcin Imielinski & Xiaotong Yao
  383. Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
    William B. Isaacs
  384. Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
    Shumpei Ishikawa, Hiroto Katoh & Daisuke Komura
  385. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
    Michael Ittmann
  386. Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
    Michael Ittmann
  387. Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
    Michael Ittmann
  388. Technical University of Denmark, Lyngby, Denmark
    Jose M. G. Izarzugaza
  389. Department of Pathology, College of Medicine, Hanyang University, Seoul, South Korea
    Jocelyne Jacquemier, Hyung-Yong Kim & Gu Kong
  390. Academic Unit of Surgery, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
    Nigel B. Jamieson
  391. Department of Pathology, Asan Medical Center, College of Medicine, Ulsan University, Songpa-gu, Seoul, South Korea
    Se Jin Jang & Hee Jin Lee
  392. Science Writer, Garrett Park, MD, USA
    Karine Jegalian
  393. International Cancer Genome Consortium (ICGC)/ICGC Accelerating Research in Genomic Oncology (ARGO) Secretariat, Ontario Institute for Cancer Research, Toronto, ON, Canada
    Jennifer L. Jennings
  394. University of Ljubljana, Ljubljana, Slovenia
    Lara Jerman
  395. Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
    Yuan Ji
  396. Research Institute, NorthShore University HealthSystem, Evanston, IL, USA
    Yuan Ji
  397. Department for Biomedical Research, University of Bern, Bern, Switzerland
    Rory Johnson, Andrés Lanzós & Mark A. Rubin
  398. Centre of Genomics and Policy, McGill University and Génome Québec Innovation Centre, Montreal, QC, Canada
    Yann Joly, Bartha M. Knoppers, Mark Phillips & Adrian Thorogood
  399. Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
    Corbin D. Jones
  400. Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
    David T. W. Jones, Marcel Kool & Stefan M. Pfister
  401. Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
    David T. W. Jones
  402. Cancer Research UK, London, UK
    Nic Jones & David Scott
  403. Indivumed GmbH, Hamburg, Germany
    Hartmut Juhl
  404. Genome Integration Data Center, Syntekabio, Inc, Daejeon, South Korea
    Jongsun Jung
  405. University Hospital Zurich, Zurich, Switzerland
    Andre Kahles, Kjong-Van Lehmann & Gunnar Rätsch
  406. Clinical Bioinformatics, Swiss Institute of Bioinformatics, Geneva, Switzerland
    Abdullah Kahraman
  407. Institute for Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
    Abdullah Kahraman
  408. Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
    Abdullah Kahraman & Christian von Mering
  409. MRC Human Genetics Unit, MRC IGMM, University of Edinburgh, Edinburgh, UK
    Vera B. Kaiser & Colin A. Semple
  410. Women’s Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
    Beth Karlan
  411. Department of Biology, Bioinformatics Group, Division of Molecular Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
    Rosa Karlić
  412. Department for Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
    Dennis Karsch & Michael Kneba
  413. Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
    Karin S. Kassahn
  414. Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
    Hitoshi Katai
  415. Department of Bioinformatics, Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
    Mamoru Kato, Hirofumi Rokutan & Mihoko Saito-Adachi
  416. A.A. Kharkevich Institute of Information Transmission Problems, Moscow, Russia
    Marat D. Kazanov
  417. Oncology and Immunology, Dmitry Rogachev National Research Center of Pediatric Hematology, Moscow, Russia
    Marat D. Kazanov
  418. Skolkovo Institute of Science and Technology, Moscow, Russia
    Marat D. Kazanov
  419. Department of Surgery, The George Washington University, School of Medicine and Health Science, Washington, DC, USA
    Electron Kebebew
  420. Endocrine Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
    Electron Kebebew
  421. Melanoma Institute Australia, Macquarie University, Sydney, NSW, Australia
    Richard F. Kefford
  422. MIT Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
    Manolis Kellis
  423. Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
    James G. Kench & Richard A. Scolyer
  424. Cholangiocarcinoma Screening and Care Program and Liver Fluke and Cholangiocarcinoma Research Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
    Narong Khuntikeo
  425. Controlled Department and Institution, New York, NY, USA
    Ekta Khurana
  426. Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
    Ekta Khurana & Alexander Martinez-Fundichely
  427. National Cancer Center, Gyeonggi, South Korea
    Hark Kyun Kim
  428. Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul, South Korea
    Hyung-Lae Kim
  429. Health Sciences Department of Biomedical Informatics, University of California San Diego, La Jolla, CA, USA
    Jihoon Kim
  430. Research Core Center, National Cancer Centre Korea, Goyang-si, South Korea
    Jong K. Kim
  431. Department of Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, South Korea
    Youngwook Kim
  432. Samsung Genome Institute, Seoul, South Korea
    Youngwook Kim
  433. Breast Oncology Program, Dana-Farber/Brigham and Women’s Cancer Center, Boston, MA, USA
    Tari A. King
  434. Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
    Tari A. King & Samuel Singer
  435. Division of Breast Surgery, Brigham and Women’s Hospital, Boston, MA, USA
    Tari A. King
  436. Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences (NIEHS), Durham, NC, USA
    Leszek J. Klimczak
  437. Department of Clinical Science, University of Bergen, Bergen, Norway
    Stian Knappskog & Ola Myklebost
  438. Center For Medical Innovation, Seoul National University Hospital, Seoul, South Korea
    Youngil Koh
  439. Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
    Youngil Koh & Sung-Soo Yoon
  440. Institute of Computer Science, Polish Academy of Sciences, Warsawa, Poland
    Jan Komorowski
  441. Functional and Structural Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
    Marcel Kool, Andrey Korshunov, Michael Koscher, Stefan M. Pfister & Qi Wang
  442. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, , National Institutes of Health, Bethesda, MD, USA
    Roelof Koster
  443. Institute for Medical Informatics Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
    Markus Kreuz & Markus Loeffler
  444. Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Savitri Krishnamurthy
  445. Department of Hematology and Oncology, Georg-Augusts-University of Göttingen, Göttingen, Germany
    Dieter Kube & Lorenz H. P. Trümper
  446. Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany
    Ralf Küppers
  447. King’s College London and Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
    Jesper Lagergren
  448. Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA
    Peter W. Laird
  449. The University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, Herston, QLD, Australia
    Sunil R. Lakhani & Peter T. Simpson
  450. Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
    Pablo Landgraf
  451. University of Düsseldorf, Düsseldorf, Germany
    Pablo Landgraf & Guido Reifenberger
  452. Department of Pathology, Institut Jules Bordet, Brussels, Belgium
    Denis Larsimont
  453. Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
    Erik Larsson
  454. Children’s Medical Research Institute, Sydney, NSW, Australia
    Loretta M. S. Lau & Hilda A. Pickett
  455. ILSbio, LLC Biobank, Chestertown, MD, USA
    Xuan Le
  456. Division of Genetics and Genomics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
    Eunjung Alice Lee
  457. Institute for Bioengineering and Biopharmaceutical Research (IBBR), Hanyang University, Seoul, South Korea
    Jeong-Yeon Lee
  458. Department of Statistics, University of California Santa Cruz, Santa Cruz, CA, USA
    Juhee Lee
  459. National Genotyping Center, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
    Ming Ta Michael Lee
  460. Department of Vertebrate Genomics/Otto Warburg Laboratory Gene Regulation and Systems Biology of Cancer, Max Planck Institute for Molecular Genetics, Berlin, Germany
    Hans Lehrach, Hans-Jörg Warnatz & Marie-Laure Yaspo
  461. McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada
    Louis Letourneau
  462. biobyte solutions GmbH, Heidelberg, Germany
    Ivica Letunic
  463. Gynecologic Oncology, NYU Laura and Isaac Perlmutter Cancer Center, New York University, New York, NY, USA
    Douglas A. Levine
  464. Division of Oncology, Stem Cell Biology Section, Washington University School of Medicine, St. Louis, MO, USA
    Tim Ley
  465. Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Han Liang
  466. Harvard University, Cambridge, MA, USA
    Ziao Lin
  467. Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
    W. M. Linehan
  468. University of Oslo, Oslo, Norway
    Ole Christian Lingjærde & Torill Sauer
  469. University of Toronto, Toronto, ON, Canada
    Fei-Fei Fei Liu, Quaid D. Morris, Ruian Shi, Shankar Vembu & Fan Yang
  470. Peking University, Beijing, China
    Fenglin Liu, Fan Zhang, Liangtao Zheng & Xiuqing Zheng
  471. School of Life Sciences, Peking University, Beijing, China
    Fenglin Liu
  472. Leidos Biomedical Research, Inc, McLean, VA, USA
    Jia Liu
  473. Hematology, Hospital Clinic, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
    Armando Lopez-Guillermo
  474. Second Military Medical University, Shanghai, China
    Yong-Jie Lu & Hongwei Zhang
  475. Chinese Cancer Genome Consortium, Shenzhen, China
    Youyong Lu
  476. Department of Medical Oncology, Beijing Hospital, Beijing, China
    Youyong Lu
  477. Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
    Youyong Lu & Rui Xing
  478. School of Medicine/School of Mathematics and Statistics, University of St. Andrews, St, Andrews, Fife, UK
    Andy G. Lynch
  479. Institute for Systems Biology, Seattle, WA, USA
    Lisa Lype, Sheila M. Reynolds & Ilya Shmulevich
  480. Department of Biochemistry and Molecular Biology, Faculty of Medicine, University Institute of Oncology-IUOPA, Oviedo, Spain
    Carlos López-Otín & Xose S. Puente
  481. Institut Bergonié, Bordeaux, France
    Gaetan MacGrogan
  482. Cancer Unit, MRC University of Cambridge, Cambridge, UK
    Shona MacRae
  483. Department of Pathology and Laboratory Medicine, Center for Personalized Medicine, Children’s Hospital Los Angeles, Los Angeles, CA, USA
    Dennis T. Maglinte
  484. John Curtin School of Medical Research, Canberra, ACT, Australia
    Graham J. Mann
  485. MVZ Department of Oncology, PraxisClinic am Johannisplatz, Leipzig, Germany
    Luisa Mantovani-Löffler
  486. Department of Information Technology, Ghent University, Ghent, Belgium
    Kathleen Marchal & Sergio Pulido-Tamayo
  487. Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
    Kathleen Marchal, Sergio Pulido-Tamayo & Lieven P. C. Verbeke
  488. Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, USA
    Elaine R. Mardis
  489. Computational Biology Program, School of Medicine, Oregon Health and Science University, Portland, OR, USA
    Adam A. Margolin & Adam J. Struck
  490. Department of Surgery, Duke University, Durham, NC, USA
    Jeffrey Marks
  491. Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
    Tomas Marques-Bonet, Jose I. Martin-Subero, Arcadi Navarro, David Torrents & Alfonso Valencia
  492. Institut Català de Paleontologia Miquel Crusafont, Universitat Autònoma de Barcelona, Barcelona, Spain
    Tomas Marques-Bonet
  493. University of Glasgow, Glasgow, UK
    Sancha Martin & Ke Yuan
  494. Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
    Jose I. Martin-Subero
  495. Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
    R. Jay Mashl
  496. Department of Surgery and Cancer, Imperial College, London, INY, UK
    Erik Mayer
  497. Applications Department, Oxford Nanopore Technologies, Oxford, UK
    Simon Mayes & Daniel J. Turner
  498. Department of Obstetrics, Gynecology and Reproductive Services, University of California San Francisco, San Francisco, CA, USA
    Karen McCune & Karen Smith-McCune
  499. Department of Biochemistry and Molecular Medicine, University California at Davis, Sacramento, CA, USA
    John D. McPherson
  500. STTARR Innovation Facility, Princess Margaret Cancer Centre, Toronto, ON, Canada
    Alice Meng
  501. Discipline of Surgery, Western Sydney University, Penrith, NSW, Australia
    Neil D. Merrett
  502. Yale School of Medicine, Yale University, New Haven, CT, USA
    William Meyerson
  503. Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
    Piotr A. Mieczkowski, Joel S. Parker, Charles M. Perou, Donghui Tan, Umadevi Veluvolu & Matthew D. Wilkerson
  504. Departments of Neurology and Neurosurgery, Henry Ford Hospital, Detroit, MI, USA
    Tom Mikkelsen
  505. Precision Oncology, OHSU Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
    Gordon B. Mills
  506. Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
    Sarah Minner, Guido Sauter & Ronald Simon
  507. Department of Health Sciences, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
    Shinichi Mizuno
  508. Heidelberg Academy of Sciences and Humanities, Heidelberg, Germany
    Fruzsina Molnár-Gábor
  509. Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
    Carl Morrison, Karin A. Oien, Chawalit Pairojkul, Paul M. Waring & Marc J. van de Vijver
  510. Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
    Carl Morrison
  511. Department of Computer Science, University of Helsinki, Helsinki, Finland
    Ville Mustonen
  512. Institute of Biotechnology, University of Helsinki, Helsinki, Finland
    Ville Mustonen
  513. Organismal and Evolutionary Biology Research Programme, University of Helsinki, Helsinki, Finland
    Ville Mustonen
  514. Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University School of Medicine, St. Louis, MO, USA
    David Mutch
  515. Penrose St. Francis Health Services, Colorado Springs, CO, USA
    Jerome Myers
  516. Institute of Pathology, Ulm University and University Hospital of Ulm, Ulm, Germany
    Peter Möller
  517. National Cancer Center, Tokyo, Japan
    Hitoshi Nakagama
  518. Genome Institute of Singapore, Singapore, Singapore
    Tannistha Nandi & Patrick Tan
  519. 32Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
    Fabio C. P. Navarro
  520. German Cancer Aid, Bonn, Germany
    Gerd Nettekoven & Laura Planko
  521. Programme in Cancer and Stem Cell Biology, Centre for Computational Biology, Duke-NUS Medical School, Singapore, Singapore
    Alvin Wei Tian Ng
  522. The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
    Anthony Ng
  523. Fourth Military Medical University, Shaanxi, China
    Yongzhan Nie
  524. The University of Cambridge School of Clinical Medicine, Cambridge, UK
    Serena Nik-Zainal
  525. St. Jude Children’s Research Hospital, Memphis, TN, USA
    Paul A. Northcott
  526. University Health Network, Princess Margaret Cancer Centre, Toronto, ON, Canada
    Faiyaz Notta & Ming Tsao
  527. Center for Biomolecular Science and Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
    Brian D. O’Connor
  528. Department of Medicine, University of Chicago, Chicago, IL, USA
    Peter O’Donnell
  529. Department of Neurology, Mayo Clinic, Rochester, MN, USA
    Brian Patrick O’Neill
  530. Cambridge Oesophagogastric Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
    J. Robert O’Neill
  531. Department of Computer Science, Carleton College, Northfield, MN, USA
    Layla Oesper
  532. Institute of Cancer Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
    Karin A. Oien
  533. Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
    Akinyemi I. Ojesina
  534. HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
    Akinyemi I. Ojesina
  535. O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
    Akinyemi I. Ojesina
  536. Department of Pathology, Keio University School of Medicine, Tokyo, Japan
    Hidenori Ojima
  537. Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
    Takuji Okusaka
  538. Sage Bionetworks, Seattle, WA, USA
    Larsson Omberg
  539. Lymphoma Genomic Translational Research Laboratory, National Cancer Centre, Singapore, Singapore
    Choon Kiat Ong
  540. Department of Clinical Pathology, Robert-Bosch-Hospital, Stuttgart, Germany
    German Ott
  541. Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
    B. F. Francis Ouellette
  542. Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
    Qiang Pan-Hammarström
  543. Center for Liver Cancer, Research Institute and Hospital, National Cancer Center, Gyeonggi, South Korea
    Joong-Won Park
  544. Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
    Keunchil Park
  545. Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, South Korea
    Keunchil Park
  546. Cheonan Industry-Academic Collaboration Foundation, Sangmyung University, Cheonan, South Korea
    Kiejung Park
  547. NYU Langone Medical Center, New York, NY, USA
    Harvey Pass
  548. Department of Hematology and Medical Oncology, Cleveland Clinic, Cleveland, OH, USA
    Nathan A. Pennell
  549. Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
    Marc D. Perry
  550. Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
    Gloria M. Petersen
  551. Helen F. Graham Cancer Center at Christiana Care Health Systems, Newark, DE, USA
    Nicholas Petrelli
  552. Heidelberg University Hospital, Heidelberg, Germany
    Stefan M. Pfister
  553. CSRA Incorporated, Fairfax, VA, USA
    Todd D. Pihl
  554. Research Department of Pathology, University College London Cancer Institute, London, UK
    Nischalan Pillay
  555. Department of Research Oncology, Guy’s Hospital, King’s Health Partners AHSC, King’s College London School of Medicine, London, UK
    Sarah Pinder
  556. Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
    Andreia V. Pinho
  557. University Hospital of Minjoz, INSERM UMR 1098, Besançon, France
    Xavier Pivot
  558. Spanish National Cancer Research Centre, Madrid, Spain
    Tirso Pons
  559. Center of Digestive Diseases and Liver Transplantation, Fundeni Clinical Institute, Bucharest, Romania
    Irinel Popescu
  560. Cureline, Inc, South San Francisco, CA, USA
    Olga Potapova
  561. St. Luke’s Cancer Centre, Royal Surrey County Hospital NHS Foundation Trust, Guildford, UK
    Shaun R. Preston
  562. Cambridge Breast Unit, Addenbrooke’s Hospital, Cambridge University Hospital NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
    Elena Provenzano
  563. East of Scotland Breast Service, Ninewells Hospital, Aberdeen, UK
    Colin A. Purdie
  564. Department of Genetics, Microbiology and Statistics, University of Barcelona, IRSJD, IBUB, Barcelona, Spain
    Raquel Rabionet
  565. Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
    Janet S. Rader
  566. Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
    Suresh Ramalingam
  567. Department of Computer Science, Princeton University, Princeton, NJ, USA
    Benjamin J. Raphael & Matthew A. Reyna
  568. Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
    W. Kimryn Rathmell
  569. Ohio State University College of Medicine and Arthur G. James Comprehensive Cancer Center, Columbus, OH, USA
    Matthew Ringel
  570. Department of Surgery, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
    Yasushi Rino
  571. Division of Chromatin Networks, German Cancer Research Center (DKFZ) and BioQuant, Heidelberg, Germany
    Karsten Rippe
  572. Research Computing Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
    Jeffrey Roach
  573. School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, WA, USA
    Steven A. Roberts
  574. Finsen Laboratory and Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
    F. Germán Rodríguez-González, Nikos Sidiropoulos & Joachim Weischenfeldt
  575. Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
    Michael H. A. Roehrl & Stefano Serra
  576. Department of Pathology, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
    Michael H. A. Roehrl
  577. University Hospital Giessen, Pediatric Hematology and Oncology, Giessen, Germany
    Marius Rohde
  578. Oncologie Sénologie, ICM Institut Régional du Cancer, Montpellier, France
    Gilles Romieu
  579. Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel, Germany
    Philip C. Rosenstiel & Markus B. Schilhabel
  580. Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
    Andreas Rosenwald
  581. Department of Urology, North Bristol NHS Trust, Bristol, UK
    Edward W. Rowe
  582. SingHealth, Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore, Singapore
    Steven G. Rozen, Patrick Tan & Bin Tean Teh
  583. Department of Computer Science, University of Toronto, Toronto, ON, Canada
    Yulia Rubanova, Jared T. Simpson & Jeffrey A. Wintersinger
  584. Bern Center for Precision Medicine, University Hospital of Bern, University of Bern, Bern, Switzerland
    Mark A. Rubin
  585. Englander Institute for Precision Medicine, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY, USA
    Mark A. Rubin
  586. Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
    Mark A. Rubin
  587. Pathology and Laboratory, Weill Cornell Medical College, New York, NY, USA
    Mark A. Rubin
  588. Vall d’Hebron Institute of Oncology: VHIO, Barcelona, Spain
    Carlota Rubio-Perez
  589. General and Hepatobiliary-Biliary Surgery, Pancreas Institute, University and Hospital Trust of Verona, Verona, Italy
    Andrea Ruzzenente
  590. National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
    Radhakrishnan Sabarinathan
  591. Indiana University, Bloomington, IN, USA
    S. Cenk Sahinalp
  592. Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
    Roberto Salgado
  593. Analytical Biological Services, Inc, Wilmington, DE, USA
    Charles Saller
  594. Sydney Medical School, University of Sydney, Sydney, NSW, Australia
    Jaswinder S. Samra & Richard A. Scolyer
  595. cBio Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
    Chris Sander & Ciyue Shen
  596. Department of Cell Biology, Harvard Medical School, Boston, MA, USA
    Chris Sander & Ciyue Shen
  597. Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
    Rajiv Sarin
  598. School of Environmental and Life Sciences, Faculty of Science, The University of Newcastle, Ourimbah, NSW, Australia
    Christopher J. Scarlett
  599. Department of Dermatology, University Hospital of Essen, Essen, Germany
    Dirk Schadendorf
  600. Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany
    Matthias Schlesner
  601. Department of Urology, Charité Universitätsmedizin Berlin, Berlin, Germany
    Thorsten Schlomm & Joachim Weischenfeldt
  602. Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
    Thorsten Schlomm
  603. Department of General Internal Medicine, University of Kiel, Kiel, Germany
    Stefan Schreiber
  604. German Cancer Consortium (DKTK), Partner site Berlin, Berlin, Germany
    Roland F. Schwarz
  605. Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
    Ralph Scully
  606. University of Pittsburgh, Pittsburgh, PA, USA
    Raja Seethala
  607. Department of Ophthalmology and Ocular Genomics Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
    Ayellet V. Segre
  608. Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA
    Subhajit Sengupta
  609. Van Andel Research Institute, Grand Rapids, MI, USA
    Hui Shen & Wanding Zhou
  610. Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
    Tatsuhiro Shibata, Hirokazu Taniguchi & Tomoko Urushidate
  611. Japan Agency for Medical Research and Development, Tokyo, Japan
    Kiyo Shimizu & Takashi Yugawa
  612. Korea University, Seoul, South Korea
    Seung Jun Shin & Stefan G. Stark
  613. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD, USA
    Craig Shriver
  614. Human Genetics, University of Kiel, Kiel, Germany
    Reiner Siebert
  615. Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
    Sabina Signoretti
  616. Oregon Health and Science University, Portland, OR, USA
    Jaclyn Smith
  617. Center for RNA Interference and Noncoding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Anil K. Sood
  618. Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Anil K. Sood
  619. Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Anil K. Sood
  620. University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
    Sharmila Sothi
  621. Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, GA, The Netherlands
    Paul N. Span
  622. Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL, USA
    Jonathan Spring
  623. Clinic for Hematology and Oncology, St.-Antonius-Hospital, Eschweiler, Germany
    Peter Staib
  624. Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
    Stefan G. Stark
  625. University of Iceland, Reykjavik, Iceland
    Ólafur Andri Stefánsson
  626. Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
    Oliver Stegle
  627. Dundee Cancer Centre, Ninewells Hospital, Dundee, UK
    Alasdair Stenhouse & Alastair M. Thompson
  628. Department for Internal Medicine III, University of Ulm and University Hospital of Ulm, Ulm, Germany
    Stephan Stilgenbauer
  629. Institut Curie, INSERM Unit 830, Paris, France
    Henk G. Stunnenberg & Anne Vincent-Salomon
  630. Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
    Akihiro Suzuki
  631. Department of Laboratory Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, GA, The Netherlands
    Fred Sweep
  632. Division of Cancer Genome Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
    Holger Sültmann
  633. Department of General Surgery, Singapore General Hospital, Singapore, Singapore
    Benita Kiat Tee Tan
  634. Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
    Patrick Tan & Bin Tean Teh
  635. Department of Medical and Clinical Genetics, Genome-Scale Biology Research Program, University of Helsinki, Helsinki, Finland
    Tomas J. Tanskanen
  636. East Anglian Medical Genetics Service, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
    Patrick Tarpey
  637. Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
    Simon Tavaré
  638. Institute of Molecular and Cell Biology, Singapore, Singapore
    Bin Tean Teh
  639. Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Centre Singapore, Singapore, Singapore
    Bin Tean Teh
  640. Universite Lyon, INCa-Synergie, Centre Léon Bérard, Lyon, France
    Gilles Thomas
  641. Department of Urology, Mayo Clinic, Rochester, MN, USA
    R. Houston Thompson
  642. Royal National Orthopaedic Hospital - Stanmore, Stanmore, Middlesex, UK
    Roberto Tirabosco
  643. Department of Biochemistry, Genetics and Immunology, University of Vigo, Vigo, Spain
    Marta Tojo
  644. Giovanni Paolo II / I.R.C.C.S. Cancer Institute, Bari, BA, Italy
    Stefania Tommasi
  645. Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
    Umut H. Toprak
  646. Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy, Rome, Italy
    Giampaolo Tortora
  647. University of Verona, Verona, Italy
    Giampaolo Tortora
  648. Centre National de Génotypage, CEA - Institute de Génomique, Evry, France
    Jörg Tost
  649. CAPHRI Research School, Maastricht University, Maastricht, ER, The Netherlands
    David Townend
  650. Department of Biopathology, Centre Léon Bérard, Lyon, France
    Isabelle Treilleux
  651. Université Claude Bernard Lyon 1, Villeurbanne, France
    Isabelle Treilleux
  652. Core Research for Evolutional Science and Technology (CREST), JST, Tokyo, Japan
    Tatsuhiko Tsunoda
  653. Department of Biological Sciences, Laboratory for Medical Science Mathematics, Graduate School of Science, University of Tokyo, Yokohama, Japan
    Tatsuhiko Tsunoda
  654. Department of Medical Science Mathematics, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
    Tatsuhiko Tsunoda
  655. Cancer Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, UK
    Jose M. C. Tubio
  656. University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
    Olga Tucker
  657. Centre for Cancer Research and Cell Biology, Queen’s University, Belfast, UK
    Richard Turkington
  658. Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Naoto T. Ueno
  659. Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
    Christopher Umbricht
  660. Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
    Husen M. Umer
  661. School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
    Timothy J. Underwood
  662. Department of Gene Technology, Tallinn University of Technology, Tallinn, Estonia
    Liis Uusküla-Reimand
  663. Genetics and Genome Biology Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
    Liis Uusküla-Reimand
  664. Departments of Neurosurgery and Hematology and Medical Oncology, Winship Cancer Institute and School of Medicine, Emory University, Atlanta, GA, USA
    Erwin G. Van Meir
  665. Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
    Miguel Vazquez
  666. Argmix Consulting, North Vancouver, BC, Canada
    Shankar Vembu
  667. Department of Information Technology, Ghent University, Interuniversitair Micro-Electronica Centrum (IMEC), Ghent, Belgium
    Lieven P. C. Verbeke
  668. Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
    Clare Verrill
  669. Institute of Mathematics and Computer Science, University of Latvia, Riga, LV, Latvia
    Juris Viksna
  670. Discipline of Pathology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
    Ricardo E. Vilain
  671. Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge, UK
    Ignacio Vázquez-García
  672. Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
    Ignacio Vázquez-García & Venkata D. Yellapantula
  673. Department of Statistics, Columbia University, New York, NY, USA
    Ignacio Vázquez-García
  674. Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
    Claes Wadelius
  675. School of Electronic and Information Engineering, Xi’an Jiaotong University, Xi’an, China
    Jiayin Wang & Kai Ye
  676. Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
    Anne Y. Warren
  677. Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
    David C. Wedge
  678. Georgia Regents University Cancer Center, Augusta, GA, USA
    Paul Weinberger
  679. Wythenshawe Hospital, Manchester, UK
    Ian Welch
  680. Department of Genetics, Washington University School of Medicine, St.Louis, MO, USA
    Michael C. Wendl
  681. Department of Biological Oceanography, Leibniz Institute of Baltic Sea Research, Rostock, Germany
    Johannes Werner
  682. Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
    Justin P. Whalley
  683. Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
    David A. Wheeler
  684. Thoracic Oncology Laboratory, Mayo Clinic, Rochester, MN, USA
    Dennis Wigle
  685. Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, USA
    Richard K. Wilson
  686. Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Mayo Clinic, Rochester, MN, USA
    Boris Winterhoff
  687. International Institute for Molecular Oncology, Poznań, Poland
    Maciej Wiznerowicz
  688. Poznan University of Medical Sciences, Poznań, Poland
    Maciej Wiznerowicz
  689. Genomics and Proteomics Core Facility High Throughput Sequencing Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
    Stephan Wolf
  690. NCCS-VARI Translational Research Laboratory, National Cancer Centre Singapore, Singapore, Singapore
    Bernice H. Wong
  691. Edison Family Center for Genome Sciences and Systems Biology, Washington University, St. Louis, MO, USA
    Winghing Wong
  692. MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
    Derek W. Wright
  693. Department of Medical Informatics and Clinical Epidemiology, Division of Bioinformatics and Computational Biology, OHSU Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
    Guanming Wu
  694. School of Electronic Information and Communications, Huazhong University of Science and Technology, Wuhan, China
    Tian Xia
  695. Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
    Yanxun Xu
  696. Department of Cancer Genome Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan
    Shinichi Yachida
  697. Institute of Computer Science, Heidelberg University, Heidelberg, Germany
    Sergei Yakneen
  698. School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia
    Jean Y. Yang
  699. Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
    Lixing Yang
  700. Department of Human Genetics, University of Chicago, Chicago, IL, USA
    Lixing Yang
  701. Tri-Institutional PhD Program in Computational Biology and Medicine, Weill Cornell Medicine, New York, NY, USA
    Xiaotong Yao
  702. The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
    Kai Ye
  703. Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
    Jun Yu
  704. Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
    Kaixian Yu & Hongtu Zhu
  705. Duke-NUS Medical School, Singapore, Singapore
    Willie Yu
  706. Department of Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
    Yingyan Yu
  707. School of Computing Science, University of Glasgow, Glasgow, UK
    Ke Yuan
  708. Division of Orthopaedic Surgery, Oslo University Hospital, Oslo, Norway
    Olga Zaikova
  709. Eastern Clinical School, Monash University, Melbourne, VIC, Australia
    Nikolajs Zeps
  710. Epworth HealthCare, Richmond, VIC, Australia
    Nikolajs Zeps
  711. Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
    Cheng-Zhong Zhang
  712. Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, USA
    Yan Zhang
  713. The Ohio State University Comprehensive Cancer Center (OSUCCC – James), Columbus, OH, USA
    Yan Zhang
  714. The University of Texas School of Biomedical Informatics (SBMI) at Houston, Houston, TX, USA
    Zhongming Zhao
  715. Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
    Hongtu Zhu
  716. Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
    Lihua Zou
  717. Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
    Anna deFazio
  718. Department of Pathology, Erasmus Medical Center Rotterdam, Rotterdam, GD, The Netherlands
    Carolien H. M. van Deurzen
  719. Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
    L. van’t Veer
  720. Institute of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
    Christian von Mering

Consortia

The ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium

Contributions

Writing committee leads: Peter J. Campbell, Gad Getz, Jan O. Korbel, Joshua M. Stuart, Jennifer L. Jennings, Lincoln D. Stein. Head of project management: Jennifer L. Jennings. Sample collection: major contributions from Marc D. Perry, Hardeep K. Nahal-Bose; led by B. F. Francis Ouellette. Histopathology harmonization: major contribution from Constance H. Li; further contributions from Esther Rheinbay, G. Petur Nielsen, Dennis C. Sgroi, Chin-Lee Wu, William C. Faquin, Vikram Deshpande, Paul C. Boutros, Alexander J. Lazar, Katherine A. Hoadley; led by Lincoln D. Stein, David N. Louis. Uniform processing, somatic, germline variant calling: major contribution from L. Jonathan Dursi; further contributions from Christina K. Yung, Matthew H. Bailey, Gordon Saksena, Keiran M. Raine, Ivo Buchhalter, Kortine Kleinheinz, Matthias Schlesner, Junjun Zhang, Wenyi Wang, David A. Wheeler; led by Li Ding, Jared T. Simpson. Core alignment, variant calling by cloud computing: major contributions from Christina K. Yung, Brian D. O’Connor, Sergei Yakneen, Junjun Zhang; further contributions from Kyle Ellrott, Kortine Kleinheinz, Naoki Miyoshi, Keiran M. Raine, Adam P. Butler, Romina Royo, Gordon Saksena, Matthias Schlesner, Solomon I. Shorser, Miguel Vazquez. Integration, phasing, validation of germline variant callsets: major contributions from Tobias Rausch, Grace Tiao, Sebastian M. Waszak, Bernardo Rodriguez-Martin, Suyash Shringarpure, Dai-Ying Wu; further contributions from Sergei Yakneen, German M. Demidov, Olivier Delaneau, Shuto Hayashi, Seiya Imoto, Nina Habermann, Ayellet V. Segre, Erik Garrison, Andy Cafferkey, Eva G. Alvarez, José María Heredia-Genestar, Francesc Muyas, Oliver Drechsel, Alicia L. Bruzos, Javier Temes, Jorge Zamora, L. Jonathan Dursi, Adrian Baez-Ortega, Hyung-Lae Kim, Matthew H. Bailey, R. Jay Mashl, Kai Ye, Ivo Buchhalter, Anthony DiBiase, Kuan-lin Huang, Ivica Letunic, Michael D. McLellan, Steven J. Newhouse, Matthias Schlesner, Tal Shmaya, Sushant Kumar, David C. Wedge, Mark H. Wright, Venkata D. Yellapantula, Mark Gerstein, Ekta Khurana, Tomas Marques-Bonet, Arcadi Navarro, Carlos D. Bustamante, Jared T. Simpson, Li Ding, Reiner Siebert, Hidewaki Nakagawa, Douglas F. Easton; led by Stephan Ossowski, Jose M. C. Tubio, Gad Getz, Francisco M. De La Vega, Xavier Estivill, Jan O. Korbel. Validation, benchmarking, merging of somatic variant calls: major contribution from L. Jonathan Dursi; further contributions from David A. Wheeler, Christina K. Yung; led by Li Ding, Jared T. Simpson. Data and code availability: major contribution from Junjun Zhang; further contributions from Christina K. Yung, Sergei Yakneen, Denis Yuen, George L. Mihaiescu, Larsson Omberg; led by Vincent Ferretti. Pan-cancer burden of somatic mutations: major contribution from Junjun Zhang; led by Peter J. Campbell. Panorama of driver mutations in human cancer: led by Radhakrishnan Sabarinathan, Oriol Pich, Abel Gonzalez-Perez. PCAWG tumours with no apparent driver mutations: major contribution from Esther Rheinbay; further contributions from Amaro Taylor-Weiner, Radhakrishnan Sabarinathan; led by Peter J. Campbell, Gad Getz. Patterns, oncogenicity of kataegis, chromoplexy: major contributions from Matthew W. Fittall, Jonas Demeulemeester, Maxime Tarabichi; further contributions from Nicola D. Roberts, Peter J. Campbell, Jan O. Korbel; led by Peter Van Loo. Patterns, oncogenicity of chromothripsis: major contributions from Maxime Tarabichi, Jonas Demeulemeester, Matthew W. Fittall; further contributions from Isidro Cortes-Ciriano, Lara Urban, Peter J. Park, Peter J. Campbell, Jan O. Korbel; led by Peter Van Loo. Timing-clustered mutational processes during tumour evolution: major contributions from Jonas Demeulemeester, Maxime Tarabichi, Matthew W. Fittall; further contributions from Jan O. Korbel, Peter J. Campbell; led by Peter Van Loo. Germline effects on somatic mutation: major contributions from Sebastian M. Waszak, Bin Zhu, Bernardo Rodriguez-Martin, Esa Pitkanen, Tobias Rausch; further contributions from Yilong Li, Natalie Saini, Leszek J. Klimczak, Joachim Weischenfeldt, Nikos Sidiropoulos, Ludmil B. Alexandrov, Francesc Muyas, Raquel Rabionet, Georgia Escaramis, Adrian Baez-Ortega, Mattia Bosio, Aliaksei Z. Holik, Hana Susak, Eva G. Alvarez, Alicia L. Bruzos, Javier Temes, Aparna Prasad, Nina Habermann, Serap Erkek, Lara Urban, Claudia Calabrese, Benjamin Raeder, Eoghan Harrington, Simon Mayes, Daniel Turner, Sissel Juul, Steven A. Roberts, Lei Song, Roelof Koster, Lisa Mirabello, Xing Hua, Tomas J. Tanskanen, Marta Tojo, David C. Wedge, Jorge Zamora, Jieming Chen, Lauri A. Aaltonen, Gunnar Ratsch, Roland F. Schwarz, Atul J. Butte, Alvis Brazma, Peter J. Campbell, Stephen J. Chanock, Nilanjan Chatterjee, Oliver Stegle, Olivier Harismendy; led by G. Steven Bova, Dmitry A. Gordenin, Jose M. C. Tubio, Douglas F. Easton, Xavier Estivill, Jan O. Korbel. Replicative immortality: major contribution from David Haan; further contributions from Lina Sieverling, Lars Feuerbach; led by Lincoln D. Stein, Joshua M. Stuart. Ethical considerations of genomic cloud computing: led by Don Chalmers, Yann Joly, Bartha Knoppers, Fruzsina Molnar-Gabor, Jan O. Korbel, Mark Phillips, Adrian Thorogood, David Townend. Online resources for data access, visualization, exploration and analysis: major contributions from Mary Goldman, Junjun Zhang, Nuno A. Fonseca; further contributions from Qian Xiang, Brian Craft, Elena Pineiro-Yanez, Alfonso Munoz, Robert Petryszak, Anja Fullgrabe, Fatima Al-Shahrour, Maria Keays, David Haussler, John Weinstein, Wolfgang Huber, Alfonso Valencia, Irene Papatheodorou, Jingchun Zhu; led by Brian D. O’Connor, Lincoln D. Stein, Alvis Brazma, Vincent Ferretti, Miguel Vazquez. The 63-sample pilot-analysis validation process: major contribution from L. Jonathan Dursi; further contributions from Christina K. Yung, Matthew H. Bailey, Gordon Saksena, Keiran M. Raine, Ivo Buchhalter, Kortine Kleinheinz, Matthias Schlesner, Yu Fan, David Torrents, Matthias Bieg, Paul C. Boutros, Ken Chen, Zechen Chong, Kristian Cibulskis, Oliver Drechsel, Roland Eils, Robert S. Fulton, Josep Gelpi, Mark Gerstein, Santiago Gonzalez, Gad Getz, Ivo G. Gut, Faraz Hach, Michael Heinold, Taobo Hu, Vincent Huang, Barbara Hutter, Hyung-Lae Kim, Natalie Jager, Jongsun Jung, Sushant Kumar, Yogesh Kumar, Christopher Lalansingh, Ignaty Leshchiner, Ivica Letunic, Dimitri Livitz, Eric Z. Ma, Yosef E. Maruvka, R. Jay Mashl, Michael D. McLellan, Ana Milovanovic, Morten Muhlig Nielsen, Brian D. O’Connor, Stephan Ossowski, Nagarajan Paramasivam, Jakob Skou Pedersen, Marc D. Perry, Montserrat Puiggros, Romina Royo, Esther Rheinbay, S. Cenk Sahinalp, Iman Sarrafi, Chip Stewart, Miranda D. Stobbe, Grace Tiao, Jeremiah A. Wala, Jiayin Wang, Wenyi Wang, Sebastian M. Waszak, Joachim Weischenfeldt, Michael Wendl, Johannes Werner, Zhenggang Wu, Hong Xue, Sergei Yakneen, Takafumi N. Yamaguchi, Kai Ye, Venkata Yellapantula, Junjun Zhang, David A. Wheeler; led by Li Ding, Jared T. Simpson. Processing of validation data: major contributions from Christina K. Yung, Brian D. O’Connor, Sergei Yakneen, Junjun Zhang; further contributions from Kyle Ellrott, Kortine Kleinheinz, Naoki Miyoshi, Keiran M. Raine, Romina Royo, Gordon Saksena, Matthias Schlesner, Solomon I. Shorser, Miguel Vazquez, Joachim Weischenfeldt, Denis Yuen, Adam P. Butler, Brandi N. Davis-Dusenbery, Roland Eils, Vincent Ferretti, Robert L. Grossman, Olivier Harismendy, Youngwook Kim, Hidewaki Nakagawa, Steven J. Newhouse, David Torrents; led by Lincoln D. Stein. Whole-genome sequencing somatic variant calling: major contribution from Junjun Zhang; further contributions from Christina K. Yung, Solomon I. Shorser. Whole-genome alignment: Keiran M. Raine, Junjun Zhang, Brian D. O’Connor. DKFZ pipeline: Kortine Kleinheinz, Tobias Rausch, Jan O. Korbel, Ivo Buchhalter, Michael C. Heinold, Barbara Hutter, Natalie Jager, Nagarajan Paramasivam, Matthias Schlesner. EMBL pipeline: Joachim Weischenfeldt. Sanger pipeline: Keiran M. Raine, Jonathan Hinton, David R. Jones, Andrew Menzies, Lucy Stebbings, Adam P. Butler. Broad pipeline: Gordon Saksena, Dimitri Livitz, Esther Rheinbay, Julian M. Hess, Ignaty Leshchiner, Chip Stewart, Grace Tiao, Jeremiah A. Wala, Amaro Taylor-Weiner, Mara Rosenberg, Andrew J. Dunford, Manaswi Gupta, Marcin Imielinski, Matthew Meyerson, Rameen Beroukhim, Gad Getz. MuSE Pipeline: Yu Fan, Wenyi Wang. Consensus somatic SNV/indel annotation: Andrew Menzies, Matthias Schlesner, Juri Reimand, Priyanka Dhingra, Ekta Khurana. Somatic SNV, indel merging: major contribution from L. Jonathan Dursi; further contributions from Christina K. Yung, Matthew H. Bailey, Gordon Saksena, Keiran M. Raine, Ivo Buchhalter, Kortine Kleinheinz, Matthias Schlesner, Yu Fan, David Torrents, Matthias Bieg, Paul C. Boutros, Ken Chen, Zechen Chong, Kristian Cibulskis, Oliver Drechsel, Roland Eils, Robert S. Fulton, Josep L. Gelpi, Mark Gerstein, Santiago Gonzalez, Gad Getz, Ivo G. Gut, Faraz Hach, Michael Heinold, Taobo Hu, Vincent Huang, Barbara Hutter, Hyung-Lae Kim, Natalie Jager, Jongsun Jung, Sushant Kumar, Yogesh Kumar, Christopher Lalansingh, Ignaty Leshchiner, Ivica Letunic, Dimitri Livitz, Eric Z. Ma, Yosef E. Maruvka, R. Jay Mashl, Michael D. McLellan, Ana Milovanovic, Morten Muhlig Nielsen, Brian D. O’Connor, Stephan Ossowski, Nagarajan Paramasivam, Jakob Skou Pedersen, Marc D. Perry, Montserrat Puiggros, Romina Royo, Esther Rheinbay, S. Cenk Sahinalp, Iman Sarrafi, Chip Stewart, Miranda D. Stobbe, Grace Tiao, Jeremiah A. Wala, Jiayin Wang, Wenyi Wang, Sebastian M. Waszak, Joachim Weischenfeldt, Michael Wendl, Johannes Werner, Zhenggang Wu, Hong Xue, Sergei Yakneen, Takafumi N. Yamaguchi, Kai Ye, Venkata Yellapantula, Junjun Zhang, David A. Wheeler; major contributions from Li Ding, Jared T. Simpson. Somatic SV merging: Joachim Weischenfeldt, Francesco Favero, Yilong Li. Somatic CNA merging: Stefan Dentro, Jeff Wintersinger, Ignaty Leshchiner. Oxidative artefact filtration: Dimitri Livitz, Ignaty Leshchiner, Chip Stewart, Esther Rheinbay, Gordon Saksena, Gad Getz. Strand bias filtration: Matthias Bieg, Ivo Buchhalter, Johannes Werner, Matthias Schlesner. miniBAM generation: Jeremiah A. Wala, Gordon Saksena, Rameen Beroukhim, Gad Getz. Germline variant identification from whole-genome sequencing: major contributions from Tobias Rausch, Grace Tiao, Sebastian M. Waszak, Bernardo Rodriguez-Martin, Suyash Shringarpure, Dai-Ying Wu; further contributions from Sergei Yakneen, German M. Demidov, Olivier Delaneau, Shuto Hayashi, Seiya Imoto, Nina Habermann, Ayellet V. Segre, Erik Garrison, Andy Cafferkey, Eva G. Alvarez, Alicia L. Bruzos, Jorge Zamora, José María Heredia-Genestar, Francesc Muyas, Oliver Drechsel, L. Jonathan Dursi, Adrian Baez-Ortega, Hyung-Lae Kim, Matthew H. Bailey, R. Jay Mashl, Kai Ye, Ivo Buchhalter, Vasilisa Rudneva, Ji Wan Park, Eun Pyo Hong, Seong Gu Heo, Anthony DiBiase, Kuan-lin Huang, Ivica Letunic, Michael D. McLellan, Steven J. Newhouse, Matthias Schlesner, Tal Shmaya, Sushant Kumar, David C. Wedge, Mark H. Wright, Venkata D. Yellapantula, Mark Gerstein, Ekta Khurana, Tomas Marques-Bonet, Arcadi Navarro, Carlos D. Bustamante, Jared T. Simpson, Li Ding, Reiner Siebert, Hidewaki Nakagawa, Douglas F. Easton; led by Stephan Ossowski, Jose M. C. Tubio, Gad Getz, Francisco M. De La Vega, Xavier Estivill, Jan O. Korbel. RNA-sequencing analysis: major contributions from Nuno A. Fonseca, Andre Kahles, Kjong-Van Lehmann, Lara Urban, Cameron M. Soulette, Yuichi Shiraishi, Fenglin Liu, Yao He, Deniz Demircioglu, Natalie R. Davidson, Claudia Calabrese, Junjun Zhang, Marc D. Perry, Qian Xiang; further contributions from Liliana Greger, Siliang Li, Dongbing Liu, Stefan G. Stark, Fan Zhang, Samirkumar B. Amin, Peter Bailey, Aurelien Chateigner, Isidro Cortes-Ciriano, Brian Craft, Serap Erkek, Milana Frenkel-Morgenstern, Mary Goldman, Katherine A. Hoadley, Yong Hou, Matthew R. Huska, Ekta Khurana, Helena Kilpinen, Jan O. Korbel, Fabien C. Lamaze, Chang Li, Xiaobo Li, Xinyue Li, Xingmin Liu, Maximillian G. Marin, Julia Markowski, Tannistha Nandi, Morten Muhlig Nielsen, Akinyemi I. Ojesina, Qiang Pan-Hammarstrom, Peter J. Park, Chandra Sekhar Pedamallu, Jakob Skou Pedersen, Reiner Siebert, Hong Su, Patrick Tan, Bin Tean Teh, Jian Wang, Sebastian M. Waszak, Heng Xiong, Sergei Yakneen, Chen Ye, Christina Yung, Xiuqing Zhang, Liangtao Zheng, Jingchun Zhu, Shida Zhu, Philip Awadalla, Chad J. Creighton, Matthew Meyerson, B. F. Francis Ouellette, Kui Wu, Huanming Yang; led by Jonathan Goke, Roland F. Schwarz, Oliver Stegle, Zemin Zhang, Alvis Brazma, Gunnar Ratsch, Angela N. Brooks. Clustering of tumour genomes based on telomere maintenance-related features: major contribution from David Haan; led by Lincoln D. Stein, Joshua M. Stuart. Clustered mutational processes in PCAWG: major contributions from Jonas Demeulemeester, Maxime Tarabichi, Matthew W. Fittall; led by Peter J. Campbell, Jan O. Korbel, Peter Van Loo. Tumours without detected driver mutations: Esther Rheinbay, Amaro Taylor-Weiner, Radhakrishnan Sabarinathan, Peter J. Campbell, Gad Getz. Panorama of driver mutations in human cancer: major contributions from Radhakrishnan Sabarinathan, Oriol Pich; further contributions from Inigo Martincorena, Carlota Rubio-Perez, Malene Juul, Jeremiah A. Wala, Steven Schumacher, Ofer Shapira, Nikos Sidiropoulos, Sebastian M. Waszak, David Tamborero, Loris Mularoni, Esther Rheinbay, Henrik Hornshoj, Jordi Deu-Pons, Ferran Muinos, Johanna Bertl, Qianyun Guo, Chad J. Creighton, Joachim Weischenfeldt, Jan O. Korbel, Gad Getz, Peter J. Campbell, Jakob Skou Pedersen, Rameen Beroukhim; led by Abel Gonzalez-Perez. Pilot benchmarking, variant consensus development and validation: major contribution from L. Jonathan Dursi; further contributions from Christina K. Yung, Matthew H. Bailey, Gordon Saksena, Keiran M. Raine, Ivo Buchhalter, Kortine Kleinheinz, Matthias Schlesner, Yu Fan, David Torrents, Matthias Bieg, Paul C. Boutros, Ken Chen, Zechen Chong, Kristian Cibulskis, Oliver Drechsel, Roland Eils, Robert S. Fulton, Josep Gelpi, Mark Gerstein, Santiago Gonzalez, Gad Getz, Ivo G. Gut, Faraz Hach, Michael Heinold, Taobo Hu, Vincent Huang, Barbara Hutter, Hyung-Lae Kim, Natalie Jager, Jongsun Jung, Sushant Kumar, Yogesh Kumar, Christopher Lalansingh, Ignaty Leshchiner, Ivica Letunic, Dimitri Livitz, Eric Z. Ma, Yosef E. Maruvka, R. Jay Mashl, Michael D. McLellan, Ana Milovanovic, Morten Muhlig Nielsen, Brian D. O’Connor, Stephan Ossowski, Nagarajan Paramasivam, Jakob Skou Pedersen, Marc D. Perry, Montserrat Puiggros, Romina Royo, Esther Rheinbay, S. Cenk Sahinalp, Iman Sarrafi, Chip Stewart, Miranda D. Stobbe, Grace Tiao, Jeremiah A. Wala, Jiayin Wang, Wenyi Wang, Sebastian M. Waszak, Joachim Weischenfeldt, Michael Wendl, Johannes Werner, Zhenggang Wu, Hong Xue, Sergei Yakneen, Takafumi N. Yamaguchi, Kai Ye, Venkata Yellapantula, Junjun Zhang, David A. Wheeler; led by Li Ding, Jared T. Simpson. Production somatic variant calling on the PCAWG compute cloud: major contributions from Christina K. Yung, Brian D. O’Connor, Sergei Yakneen, Junjun Zhang; further contributions from Kyle Ellrott, Kortine Kleinheinz, Naoki Miyoshi, Keiran M. Raine, Romina Royo, Gordon Saksena, Matthias Schlesner, Solomon I. Shorser, Miguel Vazquez, Joachim Weischenfeldt, Denis Yuen, Adam P. Butler, Brandi N. Davis-Dusenbery, Roland Eils, Vincent Ferretti, Robert L. Grossman, Olivier Harismendy, Youngwook Kim, Hidewaki Nakagawa, Steven J Newhouse, David Torrents; led by Lincoln D. Stein. PCAWG data portals: major contributions from Mary Goldman, Junjun Zhang, Nuno A. Fonseca, Isidro Cortes-Ciriano; further contributions from Qian Xiang, Brian Craft, Elena Pineiro-Yanez, Brian D O’Connor, Wojciech Bazant, Elisabet Barrera, Alfonso Munoz, Robert Petryszak, Anja Fullgrabe, Fatima Al-Shahrour, Maria Keays, David Haussler, John Weinstein, Wolfgang Huber, Alfonso Valencia, Irene Papatheodorou, Jingchun Zhu; led by Vincent Ferretti, Miguel Vazquez.

Corresponding authors

Correspondence toPeter J. Campbell, Gad Getz, Jan O. Korbel, Lincoln D. Stein or Joshua M. Stuart.

Ethics declarations

Competing interests

Gad Getz receives research funds from IBM and Pharmacyclics and is an inventor on patent applications related to MuTect, ABSOLUTE, MutSig, MSMuTect, MSMutSig and POLYSOLVER. Hikmat Al-Ahmadie is consultant for AstraZeneca and Bristol-Myers Squibb. Samuel Aparicio is a founder and shareholder of Contextual Genomics. Pratiti Bandopadhayay receives grant funding from Novartis for an unrelated project. Rameen Beroukhim owns equity in Ampressa Therapeutics. Andrew Biankin receives grant funding from Celgene, AstraZeneca and is a consultant for or on advisory boards of AstraZeneca, Celgene, Elstar Therapeutics, Clovis Oncology and Roche. Ewan Birney is a consultant for Oxford Nanopore, Dovetail and GSK. Marcus Bosenberg is a consultant for Eli Lilly. Atul Butte is a cofounder of and consultant for Personalis, NuMedii, a consultant for Samsung, Geisinger Health, Mango Tree Corporation, Regenstrief Institute and in the recent past a consultant for 10x Genomics and Helix, a shareholder in Personalis, a minor shareholder in Apple, Twitter, Facebook, Google, Microsoft, Sarepta, 10x Genomics, Amazon, Biogen, CVS, Illumina, Snap and Sutro and has received honoraria and travel reimbursement for invited talks from Genentech, Roche, Pfizer, Optum, AbbVie and many academic institutions and health systems. Carlos Caldas has served on the Scientific Advisory Board of Illumina. Lorraine Chantrill acted on an advisory board for AMGEN Australia in the past 2 years. Andrew D. Cherniack receives research funding from Bayer. Helen Davies is an inventor on a number of patent applications that encompass the use of mutational signatures. Francisco De La Vega was employed at Annai Systems during part of the project. Ronny Drapkin serves on the scientific advisory board of Repare Therapeutics and Siamab Therapeutics. Rosalind Eeles has received an honorarium for the GU-ASCO meeting in San Francisco in January 2016 as a speaker, a honorarium and support from Janssen for the RMH FR meeting in November 2017 as a speaker (title: genetics and prostate cancer), a honorarium for an University of Chicago invited talk in May 2018 as speaker and an educational honorarium paid by Bayer & Ipsen to attend GU Connect ‘Treatment sequencing for mCRPC patients within the changing landscape of mHSPC’ at a venue at ESMO, Barcelona, on 28 September 2019. Paul Flicek is a member of the scientific advisory boards of Fabric Genomics and Eagle Genomics. Ronald Ghossein is a consultant for Veracyte. Dominik Glodzik is an inventor on a number of patent applications that encompass the use of mutational signatures. Eoghan Harrington is a full-time employee of Oxford Nanopore Technologies and is a stock holder. Yann Joly is responsible for the Data Access Compliance Office (DACO) of ICGC 2009-2018. Sissel Juul is a full-time employee of Oxford Nanopore Technologies and is a stock holder. Vincent Khoo has received personal fees and non-financial support from Accuray, Astellas, Bayer, Boston Scientific and Janssen. Stian Knappskog is a coprincipal investigator on a clinical trial that receives research funding from AstraZeneca and Pfizer. Ignaty Leshchiner is a consultant for PACT Pharma. Carlos López-Otín has ownership interest (including stock and patents) in DREAMgenics. Matthew Meyerson is a scientific advisory board chair of, and consultant for, OrigiMed, has obtained research funding from Bayer and Ono Pharma and receives patent royalties from LabCorp. Serena Nik-Zainal is an inventor on a number of patent applications that encompass the use of mutational signatures. Nathan Pennell has done consulting work with Merck, Astrazeneca, Eli Lilly and Bristol-Myers Squibb. Xose S. Puente has ownership interest (including stock and patents in DREAMgenics. Benjamin J. Raphael is a consultant for and has ownership interest (including stock and patents) in Medley Genomics. Jorge Reis-Filho is a consultant for Goldman Sachs and REPARE Therapeutics, member of the scientific advisory board of Volition RX and Paige.AI and an ad hoc member of the scientific advisory board of Ventana Medical Systems, Roche Tissue Diagnostics, InVicro, Roche, Genentech and Novartis. Lewis R. Roberts has received grant support from ARIAD Pharmaceuticals, Bayer, BTG International, Exact Sciences, Gilead Sciences, Glycotest, RedHill Biopharma, Target PharmaSolutions and Wako Diagnostics and has provided advisory services to Bayer, Exact Sciences, Gilead Sciences, GRAIL, QED Therapeutics and TAVEC Pharmaceuticals. Richard A. Scolyer has received fees for professional services from Merck Sharp & Dohme, GlaxoSmithKline Australia, Bristol-Myers Squibb, Dermpedia, Novartis Pharmaceuticals Australia, Myriad, NeraCare GmbH and Amgen. Tal Shmaya is employed at Annai Systems. Reiner Siebert has received speaker honoraria from Roche and AstraZeneca. Sabina Signoretti is a consultant for Bristol-Myers Squibb, AstraZeneca, Merck, AACR and NCI and has received funding from Bristol-Myers Squibb, AstraZeneca, Exelixis and royalties from Biogenex. Jared Simpson has received research funding and travel support from Oxford Nanopore Technologies. Anil K. Sood is a consultant for Merck and Kiyatec, has received research funding from M-Trap and is a shareholder in BioPath. Simon Tavaré is on the scientific advisory board of Ipsen and a consultant for Kallyope. John F. Thompson has received honoraria and travel support for attending advisory board meetings of GlaxoSmithKline and Provectus and has received honoraria for participation in advisory boards for MSD Australia and BMS Australia. Daniel Turner is a full-time employee of Oxford Nanopore Technologies and is a stock holder. Naveen Vasudev has received speaker honoraria and/or consultancy fees from Bristol-Myers Squibb, Pfizer, EUSA pharma, MSD and Novartis. Jeremiah A. Wala is a consultant for Nference. Daniel J. Weisenberger is a consultant for Zymo Research. Dai-Ying Wu is employed at Annai Systems. Cheng-Zhong Zhang is a cofounder and equity holder of Pillar Biosciences, a for-profit company that specializes in the development of targeted sequencing assays. The other authors declare no competing interests.

Additional information

Peer review information Nature thanks Arul Chinnaiyan, Ben Lehner, Nicolas Robine and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Flow-chart showing key steps in the analysis of PCAWG genomes.

After alignment to the genome, somatic mutations were identified by three pipelines, with subsequent merging into a consensus variant set used for downstream scientific analyses. Subs, substitutions; DKFZ/EMBL, the German Cancer Research Centre (DKFZ) and Europen Molecular Biology Laboratory (EMBL).

Extended Data Fig. 2 Distribution of accuracy estimates across algorithms and samples from validation data.

a, _F_1 accuracy, precision and sensitivity estimates for somatic SNVs across the core algorithms and different approaches to merging the call-sets. The box plots demarcate the interquartile range and median of estimates across the n = 50 samples in the validation dataset. b, _F_1 accuracy, precision and sensitivity estimates for somatic indels (n = 50 samples). SVM, support vector machine; union, calls made by all variant-calling algorithms; intersect2, calls made by any combination of two variant-calling algorithms; intersect3, calls made by any three variant-calling algorithms.

Extended Data Fig. 3 Distribution of numbers of somatic mutations of different classes across tumour types.

The y axis is on a log scale. The 2,583 donors with the highest quality metrics (white-listed donors) are plotted. SNVs indicate substitutions; indels are taken as insertions or deletions <100 bp in size; retrotranspositions are the combined counts of somatic retrotransposon insertions, transductions and somatic pseudogene insertions.

Extended Data Fig. 4 Patients with no detected driver mutations in PCAWG.

a, Number (red) of patients without detected driver mutations distributed across the different tumour types studied. b, Estimated sensitivity for detecting somatic point mutations genome-wide across tumour types (total sample size: n = 2,583 patients). Each point represents the estimate for a single patient, layered on violin plots that show the estimated density distribution of sensitivity values for that tumour type (the width proportional is to density). c, SETD2 expression levels across different medulloblastoma subtypes. Points represent individual patients, coloured by whether the gene exhibited focal copy number (CN) loss or a truncating point mutation, or was the wild-type gene. The coloured areas are violin plots showing the estimated density distribution of expression values for that medulloblastoma subtype.

Extended Data Fig. 5 Examples of clustered mutational processes.

a, Chromoplexy example in a thyroid adenocarcinoma. Genes at the breakpoints are schematically depicted in their normal genomic context and again in the reconstructed derivative chromosomes below. b, Distinct kataegis signatures in the genome of a pancreatic adenocarcinoma sample. SVs and their classification are shown above the main rainfall plot, as well as the total and minor allele copy number. Tra, translocation; del, deletion; dup, duplication; t2tInv, tail-to-tail inversion; h2hInv, head-to-head inversion. Magnifications of the three foci on chromosomes 1, 8 and 12, respectively, highlight distinct manifestations of kataegis. Left, a novel process similar to signature 17 with T > N mutations at CT or TT dinucleotides. Middle, the prototypical APOBEC3A/B type with C > T (signature 2) and/or C>G/A (signature 13) substitutions at TpC. Right, an alternative cytidine deaminase(s) with a preference for substitutions at C/GpC. Most of the SNVs in each of these foci can be phased to the same allele and no evidence of anti-phasing is observed. c, Example of a chromothripsis event in a melanoma. The black points (top) represent copy-number estimates from individual genomic bins, with SVs shown as coloured arcs (translocation in black, deletion in purple, duplication in brown, tail-to-tail inversion in cyan, head-to-head inversion in green) that mostly demarcate copy-number changes. The mate chromosomes are displayed above translocations. Bottom, the variant allele fractions of somatic mutations distributed along the relevant chromosomal region.

Extended Data Fig. 6 Patterns of intense kataegis.

a, Distribution of the tumour types (colour-coded as in Extended Data Fig. 3) of the samples in the top 5% of kataegis intensity in each of the four identified genome-wide patterns: non-APOBEC, replication stress, rearrangement-associated and the combination of the last two. b, c, Distribution of leading/lagging strand (b) and replication timing bias (c) for rearrangement-(in)dependent APOBEC kataegis, based on n = 2,583 tumours. P values were derived using a two-sided Mann–Whittney _U_-test. d, Example rainfall plots for each of the four identified kataegis patterns.

Extended Data Fig. 7 Association of chromothripsis with covariates and driver events.

a, Odds ratios per cancer type of containing chromothripsis in whole-genome duplicated versus diploid samples (n = 2,583 patients). ***q < 0.001; **q < 0.01; *q < 0.05. Two-sided hypothesis testing was performed using Fisher–Boschloo tests, corrected for multiple-hypothesis testing. b, Same as a for female versus male. c, Proportion of mutations explained by single-base substitution signature 1 and age at diagnosis in prostate cancer samples (n = 210 patients) with or without chromothripsis (q < 0.05). The early-onset prostate cancer project drives the signal and was sequenced at lower depth. For the box-and-whisker plots, the box denotes the interquartile range, with the median marked as a horizontal line. The whiskers extend as far as the range or 1.5× the interquartile range, whichever is less. Two-sided hypothesis testing was performed using Mann–Whitney _U_-tests. d, Counts of co-occurrence of chromothripsis with amplification (blue) and homozygous deletions (red) in driver regions: observed (thick line) versus randomized (shaded area and thin line). The cumulative number of drivers that were hit is plotted as a function of the number of times those drivers were hit. e, For each sample in which chromothripsis coincided with a driver event in those genes, we show the fold change in gene expression compared to the median expression of the gene in non-chromothripsis samples of the same cancer type, coloured by cancer type and shaped by the type of driver event. We show with added transparency the fold changes calculated the same way for samples with driver mutations hitting the same driver genes, but that had no evidence of chromothripsis. Analysis is based on n = 1,222 patients with RNA-sequencing data. f, Enrichment of co-occurrence of chromothripsis with driver events. The x axis shows the association of chromothripsis with a driver in a given cancer type compared with its rate of association with that driver in all other cancer types. The y axis shows the association of chromothripsis with a driver in a given cancer type compared with its rate of association with all other drivers in that type. Exact binomial tests are used and P values are corrected for multiple testing according to the Benjamini–Hochberg method.

Extended Data Fig. 8 Further examples of chromothripsis-induced amplification targeting multiple cancer-associated genes simultaneously in melanoma.

a, Examples of amplifications that occurred early in the development of melanoma. The black points (top) represent copy-number estimates from individual genomic bins, with SVs shown as coloured arcs (translocation in black, deletion in purple, duplication in brown, tail-to-tail inversion in cyan and head-to-head inversion in green) that mostly demarcate copy-number changes. Bottom, the variant allele fractions of SNVs distributed along the relevant chromosomal region. The paucity of somatic mutations at high variant allele fractions in the most-heavily amplified regions indicates that these amplifications began very early in tumour evolution, before the lineage had had opportunity to acquire many SNVs. b, Example of an amplification that occurred late in melanoma development. The large numbers of somatic mutations at high variant allele fractions in the most-heavily amplified regions indicate that these amplifications began late in tumour evolution, after the lineage had already acquired many SNVs.

Extended Data Fig. 9 Timing the amplifications after chromothripsis in molecular time for 10 representative cases.

a, Copy-number plot of chromothriptic regions categorized as ‘liposarc-like’ in five acral melanomas with CCND1 amplification. Segments indicate the copy number of the major allele. Points represent SNV multiplicities, that is, the estimated number of copies carrying each SNV, coloured by base change and shaped by strand. Small vertical arrows link SNVs to their corresponding copy-number segment. Kataegis foci are shown within black boxes and show typical strand specificities (all triangles or all circles), similar multiplicities and base changes of signatures 2 and 13 (red and black, respectively). A coloured bar (top right) represents the molecular timing of the amplification (red bar; high is early, low is late) and is coloured by the fraction of total SNVs assigned to the following timing categories: clonal [early], clonal mutations that occurred before duplications involving the relevant chromosome (including whole-genome duplications); clonal [late], clonal mutations that occurred after such duplications; and clonal [NA], mutations that occurred when no duplication was observed. b, Same as a in two cutaneous melanomas, one shows early amplification, the other late amplification. c, Same as a, b, for three lung squamous cell carcinomas and late amplification of SOX2.

Extended Data Fig. 10 Association between common germline variants and endogenous mutational processes.

Genome-wide association of somatic CpG mutagenesis in individuals of European ancestry (n = 1,201 patients) based on mutational signature analysis (a) and NpCpG motif analysis (b). Two-sided hypothesis testing was performed using PLINK v.1.9. To mitigate multiple-hypothesis testing, the significance threshold was set to genome-wide significance (P < 5 × 10−8). c, d, Locuszoom plot for somatic APOBEC3B-like mutagenesis association results, linkage disequilibrium and recombination rates around the genome-wide significant 22q13.1 locus in individuals with European (c) and East Asian (d) ancestry (n = 1,201 and 318 patients, respectively). Locuszoom plot for somatic APOBEC3B-like mutagenesis association results around the 22q13.1 locus in individuals with European (e) and East Asian (f) ancestry after conditioning on rs12628403. g, h, Association between rs2142833 and expression of APOBEC3 genes in PCAWG tumour samples (adjusted for sex, age at diagnosis, histology and population structure in linear-regression models with two-sided hypothesis testing not corrected for multiple tests). For the box-and-whisker plot, the box denotes the interquartile range, with the median marked as a horizontal line. The whiskers extend as far as the range or 1.5× the interquartile range, whichever is less. Outliers are shown as points.

Extended Data Fig. 11 Association between rare germline PTVs in protein-coding genes and somatic mutational phenotypes.

ad, f, Data are based on two-sided rare-variant association testing across n = 2,583 patients, with a stringent P value threshold of P < 2.5 × 10−6 used to mitigate multiple-hypothesis testing (significant genes marked with coloured circles). Blue/red circles mark genes that decrease/increase somatic mutation rates. The black line represents the identity line that would be followed if the observed _P_ values followed the null expectation, with the shaded area showing the 95% confidence intervals. **a**, _QQ_ plots for the proportion of somatic SV deletions, tandem duplications, inversions and translocation in cancer genomes. **b**, _QQ_ plots for the proportion of somatic SV deletions in cancer genomes stratified by four size groups (1–10 kb, 10–100 kb, 100–1,000 kb and >1,000 kb). c, QQ plots for the proportion of somatic SV tandem duplications in cancer genomes stratified by four size groups (1–10 kb, 10–100 kb, 100–1,000 kb and >1,000 kb). d, QQ plot for the presence or absence of somatic SV templated insertion (cycles) in cancer genomes. e, Number of SV-templated insertion cycles in PCAWG tumours with germline BRCA1 PTVs. Only histological samples with at least one germline BRCA1 PTV carrier are shown (n = 1,095 patients combined). The box denotes the interquartile range, with the median marked as a horizontal line. The whiskers extend as far as the range or 1.5× the interquartile range, whichever is less. Outliers are shown as points. f, QQ plot for somatic CpG mutagenesis in cancer genomes based on NpCpG motif analysis. g, Violin plots show estimated densities of the proportion of somatic CpG mutations in PCAWG donors with germline MBD4 and BRCA2 PTVs. The box denotes the interquartile range, with the median marked as a white point. The whiskers extend as far as the range or 1.5× the interquartile range, whichever is less. Two-sided hypothesis testing, not corrected for multiple testing, was performed using linear regression models. h, Replication of germline MBD4 and BRCA2 PTV associations with somatic CpG mutagenesis in TCGA whole-exome sequencing donors. Violin plots show the estimated density of the proportion of somatic CpG mutations in TCGA exomes with germline MBD4 and BRCA2 PTVs. The box denotes the interquartile range, with the median marked as a white point. The whiskers extend as far as the range or 1.5× the interquartile range, whichever is less. Two-sided hypothesis testing, not corrected for multiple testing, was performed using linear-regression models. i, Correlation between MBD4 expression and somatic CpG mutagenesis in primary solid PCAWG tumours. Hypothesis testing was two-sided and not corrected for multiple testing, using linear-regression models. The box denotes the interquartile range, with the median marked as a horizontal line. The whiskers extend as far as the range or 1.5× the interquartile range, whichever is less. j, Data are mean ± s.e.m. across n = 20 tumour types. The dashed black line shows the fitted line to the data, estimated using linear-regression models. Hypothesis testing was two-sided and not corrected for multiple testing, using Spearman’s rank correlations. k, MBD4 effect sizes (open circles) with 95% confidence intervals (error bars) for individual cancer types were estimated using linear-regression analysis after (if available) accounting for sex, age at diagnosis (young/old) and ICGC project. Hypothesis testing was two-sided and not corrected for multiple testing.

Extended Data Fig. 12 Germline MEI call set.

a, Left, dots show the number of transductions promoted by each hot element in individual samples. Arrows highlight retrotransposition burst. Right, the contribution of each hot locus is represented. The total number of transductions mediated by each source element is shown on the right. b, Source L1 activity rate (that is, measured as the average number of transductions mediated by an element) versus the percentage of samples with retrotransposition activity in which the germline element is active. For visualization purposes, extreme points observed for a source L1 with an activity rate of 49 and for a L1 active in 31% of the samples are shown at ≥20 and ≥10, respectively. c, Contrasting allele frequencies for Strombolian and Plinian source loci (sample sizes shown under each axis label). The box denotes the interquartile range, with the median marked as a white point. The whiskers extend as far as the range or 1.5× the interquartile range, whichever is less. Hypothesis testing was performed using two-sided Mann–Whitney _U_-tests without correction for multiple tests. d, Numbers of active and hot source L1 elements per donor. Data are mean ± s.d. number of elements per donor. e, The novel Plinian source element on 7p12.3 mediates 72 transductions among only 6 cancer samples. This generates a transduction that induces the deletion of the tumour-suppressor gene CDKN2A. f, Violin plots show the estimated number of distinct germline MEI alleles per PCAWG donor. The box denotes the interquartile range, with the median marked as a white point. The whiskers extend as far as the range or 1.5× the interquartile range, whichever is less. Donors are grouped according to their genetic ancestry: AFR, African; AMR, admixed American; EAS, East Asian; EUR, European; SAS, South Asian. Sample sizes are shown under each axis label. g, For each type of MEI (L1, Alu and SVA) identified both in PCAWG and in the 1000 Genomes Project (1KGP), the correlations between allele frequency estimates per ancestry derived from both projects are displayed in a blue (0) to red (1) coloured gradient. n = 2,583 PCAWG patients. Two-sided hypothesis testing was performed using Spearman’s rank correlations without correction for multiple tests. h, Example correlation between MEI allele frequencies derived from PCAWG and the 1000 Genomes Project for individuals with European ancestry (n = 1,201 patients in PCAWG). Two-sided hypothesis testing was performed using Spearman’s rank correlations without correction for multiple tests. i, Evaluation of TraFiC-mem false-discovery rate on a liver hepatocellular carcinoma sample (DO50807) and a cell line (NCI-BL2087) sequenced using single-molecule sequencing with MinION (Oxford Nanopore). For each allele frequency bin (common, >5%; low frequency, 1–5%; rare, <1%), the percentage of events supported by N long reads is represented (N ranges from 0–1 to more than 5). MEIs supported by at least two Nanopore reads were considered to be true positives (blue palette) and were classified as false positives (red) otherwise. The total number of germline MEIs per allele frequency bin is shown on the right. j, Correlation between predicted MEI lengths from Illumina and Nanopore data. Two-sided hypothesis testing was performed using Spearman’s rank correlations without correction for multiple testing.

Extended Data Fig. 13 Different mechanisms of telomere lengthening in cancer.

a, Scatter plot showing the four clusters of tumour-specific telomere patterns identified across PCAWG samples, together with the clusters of matched normal samples, generated by _t_-distributed stochastic neighbour embedding. Circles represent tumour samples and triangles represent matched normal samples. Points are coloured by tissue of origin. Data are based on n = 2,518 tumour samples and their matched normal samples. b, Patterns of comutation of the relevant driver mutations across individual patients. Columns in plot represent individual patients, coloured by type of abnormality observed. c, Distribution of clonality of driver mutations in genes relevant to telomere maintenance across clusters. Clonal [early], clonal mutations that occurred before duplications involving the relevant chromosome (including whole-genome duplications); clonal [late], clonal mutations that occurred after such duplications; and clonal [NA], mutations that occurred when no duplication was observed. d, Relationship between the estimated number of stem cell divisions per year and rate of telomere maintenance abnormalities across tumour types. The analysis uses data on estimated rates of stem cell division per year across n = 19 tissue types previously collated from the literature82. Tumour types are coloured according to the scheme shown in Extended Data Fig. 3. Two-sided hypothesis testing was performed using likelihood ratio tests on Poisson regression models with no correction for multiple tests.

Extended Data Table 1 Overview of the tumour types included in PCAWG project

Full size table

Extended Data Table 2 Ethical considerations of genomic cloud computing

Full size table

Extended Data Table 3 Scientific output using PCAWG data, in bite-size chunks

Full size table

Supplementary information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Cite this article

The ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium. Pan-cancer analysis of whole genomes.Nature 578, 82–93 (2020). https://doi.org/10.1038/s41586-020-1969-6

Download citation