Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Molecular subtypes of pancreatic cancer

Abstract

Cancers that appear morphologically similar often have dramatically different clinical features, respond variably to therapy and have a range of outcomes. Compelling evidence now demonstrates that differences in the molecular pathology of otherwise indistinguishable cancers substantially impact the clinical characteristics of the disease. Molecular subtypes now guide preclinical and clinical therapeutic development and treatment in many cancer types. The ability to predict optimal therapeutic strategies ahead of treatment improves overall patient outcomes, minimizing treatment-related morbidity and cost. Although clinical decision making based on histopathological criteria underpinned by robust data is well established in many cancer types, subtypes of pancreatic cancer do not currently inform treatment decisions. However, accumulating molecular data are defining subgroups in pancreatic cancer with distinct biology and potential subtype-specific therapeutic vulnerabilities, providing the opportunity to define a de novo clinically applicable molecular taxonomy. This Review summarizes current knowledge concerning the molecular subtyping of pancreatic cancer and explores future strategies for using a molecular taxonomy to guide therapeutic development and ultimately routine therapy with the overall goal of improving outcomes for this disease.

Key points

  • Pancreatic cancer is soon to become the second leading cause of cancer-related death.

  • Histopathological criteria do not adequately inform treatment decisions for pancreatic cancer.

  • A molecular taxonomy could improve outcomes with current treatments and accelerate therapeutic development through better patient selection.

  • Emerging molecular taxonomies define biological differences between subtypes that are associated with prognosis.

  • Genomic and transcriptomic subtypes potentially enrich for therapeutic vulnerabilities and require preclinical and clinical assessment.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Genomic aberrations characteristic of pancreatic cancer.
Fig. 2: The putative actionable genome of pancreatic cancer.
Fig. 3: Transcriptomic subtyping of pancreatic cancer.
Fig. 4: Phylotranscriptomic tree of pancreatic cancer.

Similar content being viewed by others

References

  1. Hudson, T. J. et al. International network of cancer genome projects. Nature 464, 993–998 (2010).

    CAS  PubMed  Google Scholar 

  2. The Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455, 1061–1068 (2008).

    Google Scholar 

  3. Biankin, A. V. & Hudson, T. J. Somatic variation and cancer: therapies lost in the mix. Hum. Genet. 130, 79–91 (2011).

    PubMed  Google Scholar 

  4. Biankin, A. V., Piantadosi, S. & Hollingsworth, S. J. Patient-centric trials for therapeutic development in precision oncology. Nature 526, 361–370 (2015).

    CAS  PubMed  Google Scholar 

  5. Hollingsworth, S. J. & Biankin, A. V. The challenges of precision oncology drug development and implementation. Public Health Genomics 18, 338–348 (2015).

    PubMed  Google Scholar 

  6. Swanton, C. et al. Consensus on precision medicine for metastatic cancers: a report from the MAP conference. Ann. Oncol. 27, 1443–1448 (2016).

    CAS  PubMed  Google Scholar 

  7. American Cancer Society. Key statistics for pancreatic cancer. Cancer.org https://www.cancer.org/cancer/pancreatic-cancer/about/key-statistics.html (2018).

  8. Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, 2017. CA Cancer J. Clin. 67, 7–30 (2017).

    PubMed  Google Scholar 

  9. Rahib, L. et al. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 74, 2913–2921 (2014).

    CAS  PubMed  Google Scholar 

  10. National Cancer Institute SEER. Cancer stat facts: common cancer sites. SEER https://seer.cancer.gov/statfacts/html/common.html (2018).

  11. Ryan, D. P., Hong, T. S. & Bardeesy, N. Pancreatic adenocarcinoma. N. Engl. J. Med. 371, 2140–2141 (2014).

    PubMed  Google Scholar 

  12. The Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature 511, 543–550 (2014).

    PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Petersen, G. M. Familial pancreatic cancer. Semin. Oncol. 43, 548–553 (2016).

    PubMed  PubMed Central  Google Scholar 

  15. Klein, A. P. et al. Prospective risk of pancreatic cancer in familial pancreatic cancer kindreds. Cancer Res. 64, 2634–2638 (2004).

    CAS  PubMed  Google Scholar 

  16. Brune, K. A. et al. Importance of age of onset in pancreatic cancer kindreds. J. Natl Cancer Inst. 102, 119–126 (2010).

    PubMed  PubMed Central  Google Scholar 

  17. Jones, S. et al. Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene. Science 324, 217 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Humphris, J. L. et al. Clinical and pathologic features of familial pancreatic cancer. Cancer 120, 3669–3675 (2014).

    PubMed  Google Scholar 

  19. Wolpin, B. M. et al. ABO blood group and the risk of pancreatic cancer. J. Natl Cancer Inst. 101, 424–431 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Hu, Z. I. et al. Evaluating mismatch repair deficiency in pancreatic adenocarcinoma: challenges and recommendations. Clin. Cancer Res. 24, 1326–1336 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Childs, E. J. et al. Association of common susceptibility variants of pancreatic cancer in higher-risk patients: a PACGENE study. Cancer Epidemiol. Biomarkers Prev. 25, 1185–1191 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Grant, R. C. et al. Prevalence of germline mutations in cancer predisposition genes in patients with pancreatic cancer. Gastroenterology 148, 556–564 (2014).

    PubMed  Google Scholar 

  23. Shindo, K. et al. Deleterious germline mutations in patients with apparently sporadic pancreatic adenocarcinoma. J. Clin. Oncol. 35, 3382–3390 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Humphris, J., Chang, D. K. & Biankin, A. V. Inherited susceptibility to pancreatic cancer in the era of next-generation sequencing. Gastroenterology 148, 496–498 (2015).

    PubMed  Google Scholar 

  25. Yurgelun, M. B. et al. Germline cancer susceptibility gene variants, somatic second hits, and survival outcomes in patients with resected pancreatic cancer. Genet. Med. 21, 213–223 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Canto, M. I. et al. International Cancer of the Pancreas Screening (CAPS) Consortium summit on the management of patients with increased risk for familial pancreatic cancer. Gut 62, 339–347 (2013).

    PubMed  Google Scholar 

  27. Langer, P. et al. Five years of prospective screening of high-risk individuals from families with familial pancreatic cancer. Gut 58, 1410–1418 (2009).

    CAS  PubMed  Google Scholar 

  28. McWilliams, R. R. et al. Risk factors for early-onset and very-early-onset pancreatic adenocarcinoma: a pancreatic cancer case-control consortium (PanC4) analysis. Pancreas 45, 311–316 (2016).

    PubMed  PubMed Central  Google Scholar 

  29. Kloppel, G. & Luttges, J. WHO-classification 2000: exocrine pancreatic tumors. Verh. Dtsch. Ges. Pathol. 85, 219–228 (2001).

    CAS  PubMed  Google Scholar 

  30. Kardon, D. E., Thompson, L. D., Przygodzki, R. M. & Heffess, C. S. Adenosquamous carcinoma of the pancreas: a clinicopathologic series of 25 cases. Mod. Pathol. 14, 443–451 (2001).

    CAS  PubMed  Google Scholar 

  31. Basturk, O. et al. Poorly differentiated neuroendocrine carcinomas of the pancreas: a clinicopathologic analysis of 44 cases. Am. J. Surg. Pathol. 38, 437–447 (2014).

    PubMed  PubMed Central  Google Scholar 

  32. Basturk, O. et al. A revised classification system and recommendations from the Baltimore Consensus Meeting for neoplastic precursor lesions in the pancreas. Am. J. Surg. Pathol. 39, 1730–1741 (2015).

    PubMed  PubMed Central  Google Scholar 

  33. Hruban, R. H. et al. An illustrated consensus on the classification of pancreatic intraepithelial neoplasia and intraductal papillary mucinous neoplasms. Am. J. Surg. Pathol. 28, 977–987 (2004).

    PubMed  Google Scholar 

  34. Brugge, W. R., Lauwers, G. Y., Sahani, D., Fernandez-del Castillo, C. & Warshaw, A. L. Cystic neoplasms of the pancreas. N. Engl. J. Med. 351, 1218–1226 (2004).

    CAS  PubMed  Google Scholar 

  35. Balachandran, V. P. et al. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer. Nature 551, 512–516 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Scarpa, A., Real, F. X. & Luchini, C. Genetic unrelatedness of co-occurring pancreatic adenocarcinomas and IPMNs challenges current views of clinical management. Gut 67, 1561–1563 (2018).

    CAS  PubMed  Google Scholar 

  37. Springer, S. et al. A combination of molecular markers and clinical features improve the classification of pancreatic cysts. Gastroenterology 149, 1501–1510 (2015).

    CAS  PubMed  Google Scholar 

  38. Reid, M. D., Bagci, P. & Adsay, N. V. Histopathologic assessment of pancreatic cancer: does one size fit all? J. Surg. Oncol. 107, 67–77 (2013).

    PubMed  Google Scholar 

  39. Pishvaian, M. J. & Brody, J. R. Therapeutic implications of molecular subtyping for pancreatic cancer. Oncology 31, 159–166 (2017).

    PubMed  Google Scholar 

  40. Biankin, A. V. et al. Expression of S100A2 calcium-binding protein predicts response to pancreatectomy for pancreatic cancer. Gastroenterology 137, 558–568 (2009).

    PubMed  Google Scholar 

  41. FDA News Release. FDA approves first cancer treatment for any solid tumor with a specific genetic feature. FDA.gov https://www.fda.gov/newsevents/newsroom/pressannouncements/ucm560167.htm (2017).

  42. Humphris, J. L. et al. Hypermutation in pancreatic cancer. Gastroenterology 152, 68–74 (2017).

    CAS  PubMed  Google Scholar 

  43. Niu, B. et al. MSIsensor: microsatellite instability detection using paired tumor-normal sequence data. Bioinformatics 30, 1015–1016 (2014).

    CAS  PubMed  Google Scholar 

  44. Garcea, G., Neal, C. P., Pattenden, C. J., Steward, W. P. & Berry, D. P. Molecular prognostic markers in pancreatic cancer: a systematic review. Eur. J. Cancer 41, 2213–2236 (2005).

    CAS  PubMed  Google Scholar 

  45. Iacobuzio-Donahue, C. A. et al. DPC4 gene status of the primary carcinoma correlates with patterns of failure in patients with pancreatic cancer. J. Clin. Oncol. 27, 1806–1813 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Dreyer, S. B. et al. Precision oncology in surgery: patient selection biomarkers for operable pancreatic cancer [abstract 10]. Eur. J. Surg. Oncol. 44, 1838 (2017).

    Google Scholar 

  47. Humphris, J. L. et al. The prognostic and predictive value of serum CA19.9 in pancreatic cancer. Ann. Oncol. 23, 1713–1722 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Aguirre, A. J. et al. Real-time genomic characterization of advanced pancreatic cancer to enable precision medicine. Cancer Discov. 8, 1096–1111 (2018).

    PubMed  PubMed Central  Google Scholar 

  49. Qian, Z. R. et al. Association of alterations in main driver genes with outcomes of patients with resected pancreatic ductal adenocarcinoma. JAMA Oncol. 4, e173420 (2018).

    PubMed  Google Scholar 

  50. Smit, V. T. et al. KRAS codon 12 mutations occur very frequently in pancreatic adenocarcinomas. Nucleic Acids Res. 16, 7773–7782 (1988).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Jones, S. et al. Somatic mutations in the chromatin remodeling gene ARID1A occur in several tumor types. Hum. Mut. 33, 100–103 (2012).

    CAS  PubMed  Google Scholar 

  52. Jones, S. et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 321, 1801–1806 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Chang, D. K., Grimmond, S. M. & Biankin, A. V. Pancreatic cancer genomics. Curr. Opin. Genet. Dev. 24, 74–81 (2014).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  57. Dreyer, S. B., Chang, D. K., Bailey, P. & Biankin, A. V. Pancreatic cancer genomes: implications for clinical management and therapeutic development. Clin. Cancer Res. 23, 1638–1646 (2017).

    PubMed  Google Scholar 

  58. Moffitt, R. A. et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat. Genet. 47, 1168–1178 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Witkiewicz, A. K. et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat. Commun. 6, 6744 (2015).

    CAS  PubMed  Google Scholar 

  60. The Cancer Genome Atlas Research Network. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell 32, 185–203 (2017).

    PubMed Central  Google Scholar 

  61. Cowley, M. J. et al. Understanding pancreatic cancer genomes. J. Hepatobiliary Pancreat. Sci. 20, 549–556 (2013).

    PubMed  Google Scholar 

  62. Kleeff, J. et al. Pancreatic cancer. Nat. Rev. Dis. Primers 2, 16022 (2016).

    PubMed  Google Scholar 

  63. Slamon, D. J. et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N. Engl. J. Med. 344, 783–792 (2001).

    CAS  PubMed  Google Scholar 

  64. Bollag, G. et al. Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma. Nature 467, 596–599 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Planchard, D. et al. Dabrafenib plus trametinib in patients with previously untreated BRAF(V600E)-mutant metastatic non-small-cell lung cancer: an open-label, phase 2 trial. Lancet Oncol. 18, 1307–1316 (2017).

    CAS  PubMed  Google Scholar 

  66. Ledermann, J. et al. Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer. N. Engl. J. Med. 366, 1382–1392 (2012).

    CAS  PubMed  Google Scholar 

  67. Morran, D. C. et al. Targeting mTOR dependency in pancreatic cancer. Gut 63, 1481–1489 (2014).

    CAS  PubMed  Google Scholar 

  68. Weissmueller, S. et al. Mutant p53 drives pancreatic cancer metastasis through cell-autonomous PDGF receptor beta signaling. Cell 157, 382–394 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Miller, B. W. et al. Targeting the LOX/hypoxia axis reverses many of the features that make pancreatic cancer deadly: inhibition of LOX abrogates metastasis and enhances drug efficacy. EMBO Mol. Med. 7, 1063–1076 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Chou, A. et al. Clinical and molecular characterization of HER2 amplified-pancreatic cancer. Genome Med. 5, 78 (2013).

    PubMed  PubMed Central  Google Scholar 

  71. Chmielecki, J. et al. Comprehensive genomic profiling of pancreatic acinar cell carcinomas identifies recurrent RAF fusions and frequent inactivation of DNA repair genes. Cancer Discov. 4, 1398–1405 (2014).

    CAS  PubMed  Google Scholar 

  72. Lowery, M. A. et al. Real-time genomic profiling of pancreatic ductal adenocarcinoma: potential actionability and correlation with clinical phenotype. Clin. Cancer Res. 23, 6094–6100 (2017).

    CAS  PubMed  Google Scholar 

  73. Foster, S. A. et al. Activation mechanism of oncogenic deletion mutations in BRAF, EGFR, and HER2. Cancer Cell 29, 477–493 (2016).

    CAS  PubMed  Google Scholar 

  74. Chang, D. K., Grimmond, S. M., Evans, T. R. J. & Biankin, A. V. Mining the genomes of exceptional responders. Nat. Rev. Cancer 14, 291–292 (2014).

    CAS  PubMed  Google Scholar 

  75. Lord, C. J. & Ashworth, A. The DNA damage response and cancer therapy. Nature 481, 287–294 (2012).

    CAS  PubMed  Google Scholar 

  76. McBride, D. J. et al. Tandem duplication of chromosomal segments is common in ovarian and breast cancer genomes. J. Pathol. 227, 446–455 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Polak, P. et al. A mutational signature reveals alterations underlying deficient homologous recombination repair in breast cancer. Nat. Genet. 49, 1476–1486 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Davies, H. et al. HRDetect is a predictor of BRCA1 and BRCA2 deficiency based on mutational signatures. Nat. Med. 23, 517–525 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Bamford, S. et al. The COSMIC (Catalogue of Somatic Mutations in Cancer) database and website. Br. J. Cancer 91, 355–358 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  81. Catalogue of Somatic Mutations in Cancer. Signatures of mutational processes in human cancer. COSMIC https://cancer.sanger.ac.uk/cosmic/signatures (2017).

  82. Swisher, E. M. et al. Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial. Lancet Oncol. 18, 75–87 (2017).

    CAS  PubMed  Google Scholar 

  83. Alizadeh, A. A. et al. Distinct types of diffuse large B cell lymphoma identified by gene expression profiling. Nature 403, 503–511 (2000).

    CAS  PubMed  Google Scholar 

  84. Perou, C. M. et al. Molecular portraits of human breast tumours. Nature 406, 747–752 (2000).

    CAS  PubMed  Google Scholar 

  85. Sadanandam, A. et al. A colorectal cancer classification system that associates cellular phenotype and responses to therapy. Nat. Med. 19, 619–625 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. De Sousa, E. M. F. et al. Poor-prognosis colon cancer is defined by a molecularly distinct subtype and develops from serrated precursor lesions. Nat. Med. 19, 614–618 (2013).

    Google Scholar 

  87. Marisa, L. et al. Gene expression classification of colon cancer into molecular subtypes: characterization, validation, and prognostic value. PLOS Med. 10, e1001453 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Guinney, J. et al. The consensus molecular subtypes of colorectal cancer. Nat. Med. 21, 1350–1356 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Collisson, E. A. et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat. Med. 17, 500–503 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Decker, K., Goldman, D. C., Grasch, C. L. & Sussel, L. Gata6 is an important regulator of mouse pancreas development. Dev. Biol. 298, 415–429 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Jamieson, N. B., Chang, D. K. & Biankin, A. V. Cancer genetics and implications for clinical management. Surg. Clin. North Am. 95, 919–934 (2015).

    PubMed  Google Scholar 

  92. Vogelstein, B. et al. Cancer genome landscapes. Science 339, 1546–1558 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Kassahn, K. S. et al. Somatic point mutation calling in low cellularity tumors. PLOS ONE 8, e74380 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Song, S. et al. qpure: a tool to estimate tumor cellularity from genome-wide single-nucleotide polymorphism profiles. PLOS ONE 7, e45835 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Nones, K. et al. Genome-wide DNA methylation patterns in pancreatic ductal adenocarcinoma reveal epigenetic deregulation of SLIT-ROBO, ITGA2 and MET signaling. Int. J. Cancer 135, 1110–1118 (2014).

    CAS  PubMed  Google Scholar 

  96. Hoadley, K. A. et al. Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell 158, 929–944 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Biankin, A. V. & Maitra, A. Subtyping pancreatic cancer. Cancer Cell 28, 411–413 (2015).

    CAS  PubMed  Google Scholar 

  98. Puleo, F. et al. Stratification of pancreatic ductal adenocarcinomas based on tumor and microenvironment features. Gastroenterology 155, 1999–2013 (2018).

    PubMed  Google Scholar 

  99. Noll, E. M. et al. CYP3A5 mediates basal and acquired therapy resistance in different subtypes of pancreatic ductal adenocarcinoma. Nat. Med. 22, 278–287 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Knudsen, E. S. et al. Pancreatic cancer cell lines as patient-derived avatars: genetic characterisation and functional utility. Gut 67, 508–520 (2017).

    PubMed  Google Scholar 

  101. Kopp, J. L. et al. Identification of Sox9-dependent acinar-to-ductal reprogramming as the principal mechanism for initiation of pancreatic ductal adenocarcinoma. Cancer Cell 22, 737–750 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Morris, J. P. t., Cano, D. A., Sekine, S., Wang, S. C. & Hebrok, M. Beta-catenin blocks Kras-dependent reprogramming of acini into pancreatic cancer precursor lesions in mice. J. Clin. Invest. 120, 508–520 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Steele, C. W. et al. CXCR2 inhibition profoundly suppresses metastases and augments immunotherapy in pancreatic ductal adenocarcinoma. Cancer Cell 29, 832–845 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Candido, J. B. et al. CSF1R(+) macrophages sustain pancreatic tumor growth through T cell suppression and maintenance of key gene programs that define the squamous subtype. Cell Rep. 23, 1448–1460 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Xu, Z. et al. Role of pancreatic stellate cells in pancreatic cancer metastasis. Am. J. Pathol. 177, 2585–2596 (2010).

    PubMed  PubMed Central  Google Scholar 

  106. Zhao, X. et al. Single-cell RNA-seq reveals a distinct transcriptome signature of aneuploid hematopoietic cells. Blood 130, 2762–2773 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Habib, N. et al. Massively parallel single-nucleus RNA-seq with DroNc-seq. Nat. Methods 14, 955–958 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Sinha, S. et al. PanIN neuroendocrine cells promote tumorigenesis via neuronal cross-talk. Cancer Res. 77, 1868–1879 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Delgiorno, K. E. et al. Identification and manipulation of biliary metaplasia in pancreatic tumors. Gastroenterology 146, 233–244 (2014).

    CAS  PubMed  Google Scholar 

  110. Andricovich, J. et al. Loss of KDM6A activates super-enhancers to induce gender-specific squamous-like pancreatic cancer and confers sensitivity to BET inhibitors. Cancer Cell 33, 512–526 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Sivakumar, S., de Santiago, I., Chlon, L. & Markowetz, F. Master regulators of oncogenic KRAS response in pancreatic cancer: an integrative network biology analysis. PLOS Med. 14, e1002223 (2017).

    PubMed  PubMed Central  Google Scholar 

  112. Kim, S. T. et al. Correlating programmed death ligand 1 (PD-L1) expression, mismatch repair deficiency, and outcomes across tumor types: implications for immunotherapy. Oncotarget 8, 77415–77423 (2017).

    PubMed  PubMed Central  Google Scholar 

  113. Iacobuzio-Donahue, C. A. Genetic evolution of pancreatic cancer: lessons learnt from the pancreatic cancer genome sequencing project. Gut 61, 1085–1094 (2011).

    PubMed  Google Scholar 

  114. Sequist, L. V. et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci. Transl Med. 3, 75ra26 (2011).

    PubMed  PubMed Central  Google Scholar 

  115. Furukawa, T. et al. Whole exome sequencing reveals recurrent mutations in BRCA2 and FAT genes in acinar cell carcinomas of the pancreas. Sci. Rep. 5, 8829 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Jiao, Y. et al. Whole-exome sequencing of pancreatic neoplasms with acinar differentiation. J. Pathol. 232, 428–435 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Hall, J. C. et al. Novel patient-derived xenograft mouse model for pancreatic acinar cell carcinoma demonstrates single agent activity of oxaliplatin. J. Transl Med. 14, 129 (2016).

    PubMed  PubMed Central  Google Scholar 

  118. Botton, T. et al. Recurrent BRAF kinase fusions in melanocytic tumors offer an opportunity for targeted therapy. Pigment Cell Melanoma Res. 26, 845–851 (2013).

    CAS  PubMed  Google Scholar 

  119. Menzies, A. M. et al. Clinical activity of the MEK inhibitor trametinib in metastatic melanoma containing BRAF kinase fusion. Pigment Cell Melanoma Res. 28, 607–610 (2015).

    PubMed  PubMed Central  Google Scholar 

  120. Agaimy, A. et al. Pancreatic undifferentiated rhabdoid carcinoma: KRAS alterations and SMARCB1 expression status define two subtypes. Mod. Pathol. 28, 248–260 (2015).

    CAS  PubMed  Google Scholar 

  121. Connor, A. A. et al. Association of distinct mutational signatures with correlates of increased immune activity in pancreatic ductal adenocarcinoma. JAMA Oncol. 3, 774–783 (2017).

    PubMed  Google Scholar 

  122. Aung, K. L. et al. Genomics-driven precision medicine for advanced pancreatic cancer — early results from the COMPASS trial. Clin. Cancer Res. 24, 1344–1354 (2017).

    PubMed  PubMed Central  Google Scholar 

  123. de Santiago, I. et al. Immuno-phenotypes of pancreatic ductal adenocarcinoma: metaanalysis of transcriptional subtypes. Preprint at https://www.biorxiv.org/content/early/2017/10/05/198903 (2017).

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Andrew V. Biankin.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Related links

Accelerating Research in Genomic Oncology: http://www.icgcargo.org/

Enhanced Pancreatic Cancer Profiling for Individualized Care (EPPIC): https://www.tfri.ca/en/NewsEvents/news/news-releases-detail/2018/03/06/canadian-pancreatic-cancer-research-team-provides-personalized-medicine-new-hope-to-patients

International Cancer Genome Consortium: http://www.icgc.org/

Precision-Panc: http://www.precisionpanc.org/

Precision Promise: http://www.pancan.org/research/precision-promise/

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Collisson, E.A., Bailey, P., Chang, D.K. et al. Molecular subtypes of pancreatic cancer. Nat Rev Gastroenterol Hepatol 16, 207–220 (2019). https://doi.org/10.1038/s41575-019-0109-y

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41575-019-0109-y

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer