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:

Pancreatic cancer evolution and heterogeneity: integrating omics and clinical data

Abstract

Pancreatic ductal adenocarcinoma (PDAC), already among the deadliest epithelial malignancies, is rising in both incidence and contribution to overall cancer deaths. Decades of research have improved our understanding of PDAC carcinogenesis, including characterizing germline predisposition, the cell of origin, precursor lesions, the sequence of genetic alterations, including simple and structural alterations, transcriptional changes and subtypes, tumour heterogeneity, metastatic progression and the tumour microenvironment. These fundamental advances inform contemporary translational efforts in primary prevention, screening and early detection, multidisciplinary management and survivorship, as prospective clinical trials begin to adopt molecular-based selection criteria to guide targeted therapies. Genomic and transcriptomic data on PDAC were also included in the international pan-cancer analysis of approximately 2,600 cancers, a milestone in cancer research that allows further insight through comparison with other tumour types. Thus, this is an ideal time to review our current knowledge of PDAC evolution and heterogeneity, gained from the study of preclinical models and patient biospecimens, and to propose a model of PDAC evolution that takes into consideration findings from varied sources, with a particular focus on the genomics of human PDAC.

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: Stepwise model versus punctuated model of pancreatic ductal adenocarcinoma progression.
Fig. 2: Histological stages in pancreatic ductal adenocarcinoma progression.
Fig. 3: Expression-based subtypes of pancreatic ductal adenocarcinoma.
Fig. 4: Evolutionary model of pancreatic ductal adenocarcinoma.

Similar content being viewed by others

References

  1. Connor, A. A. & Gallinger, S. Hereditary pancreatic cancer syndromes. Surg. Oncol. Clin. North Am. 24, 733–764 (2015).

    Google Scholar 

  2. 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 

  3. Hruban, R. H., Goggins, M., Parsons, J. & Kern, S. E. Progression model for pancreatic cancer. Clin. Cancer Res. 6, 2969–2972 (2000). This landmark article describes the stepwise progression model of pancreatic cancer.

    CAS  PubMed  Google Scholar 

  4. Reiter, J. G. & Iacobuzio-Donahue, C. A. Pancreatic cancer: pancreatic carcinogenesis - several small steps or one giant leap? Nat. Rev. Gastroenterol. Hepatol. 14, 7–8 (2016).

    PubMed  Google Scholar 

  5. Notta, F. et al. A renewed model of pancreatic cancer evolution based on genomic rearrangement patterns. Nature 538, 378–382 (2016). This important article describes polyploidization, chromothripsis and the punctuated evolution progression model of pancreatic cancer.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Al-Sukhni, W. et al. Screening for pancreatic cancer in a high-risk cohort: an eight-year experience. J. Gastrointest. Surg. 16, 771–783 (2012).

    PubMed  Google Scholar 

  7. Gall, T. M. H. et al. Circulating tumor cells and cell-free DNA in pancreatic ductal adenocarcinoma. Am. J. Pathol. 189, 71–81 (2019).

    CAS  PubMed  Google Scholar 

  8. Abdallah, R. et al. Plasma circulating tumor DNA in pancreatic adenocarcinoma for screening, diagnosis, prognosis, treatment and follow-up: a systematic review. Cancer Treat. Rev. 87, 102028 (2020).

    CAS  PubMed  Google Scholar 

  9. Hackeng, W. M. et al. Surgical and molecular pathology of pancreatic neoplasms. Diagn. Pathol. 11, 47 (2016).

    PubMed  PubMed Central  Google Scholar 

  10. Grant, R. C. et al. Prevalence of germline mutations in cancer predisposition genes in patients with pancreatic cancer. Gastroenterology 148, 556–564 (2015).

    CAS  PubMed  Google Scholar 

  11. Grant, R. C. et al. Exome-wide association study of pancreatic cancer risk. Gastroenterology 154, 719–722.e3 (2018).

    PubMed  Google Scholar 

  12. Humphris, J. L. & Biankin, A. V. Diagnosis and management of hereditary pancreatic cancer. Recent Results Cancer Res. 205, 61–83 (2016).

    PubMed  Google Scholar 

  13. Klein, A. P. et al. Genome-wide meta-analysis identifies five new susceptibility loci for pancreatic cancer. Nat. Commun. 9, 556 (2018).

    PubMed  PubMed Central  Google Scholar 

  14. Wang, W. et al. PancPRO: risk assessment for individuals with a family history of pancreatic cancer. J. Clin. Oncol. 25, 1417–1422 (2007).

    PubMed  Google Scholar 

  15. Goggins, M. et al. Management of patients with increased risk for familial pancreatic cancer: updated recommendations from the International Cancer of the Pancreas Screening (CAPS) Consortium. Gut 69, 7–17 (2020).

    CAS  PubMed  Google Scholar 

  16. Kozuka, S. et al. Relation of pancreatic duct hyperplasia to carcinoma. Cancer 43, 1418–1428 (1979).

    CAS  PubMed  Google Scholar 

  17. Furukawa, T. et al. Varying grades of epithelial atypia in the pancreatic ducts of humans. Classification based on morphometry and multivariate analysis and correlated with positive reactions of carcinoembryonic antigen. Arch. Pathol. Lab. Med. 118, 227–234 (1994).

    CAS  PubMed  Google Scholar 

  18. 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 

  19. van Heek, N. T. et al. Telomere shortening is nearly universal in pancreatic intraepithelial neoplasia. Am. J. Pathol. 161, 1541–1547 (2002).

    PubMed  PubMed Central  Google Scholar 

  20. Kanda, M. et al. Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology 142, 730–733.e9 (2012).

    CAS  PubMed  Google Scholar 

  21. Murphy, S. J. et al. Genetic alterations associated with progression from pancreatic intraepithelial neoplasia to invasive pancreatic tumor. Gastroenterology 145, 1098–1109.e1 (2013).

    CAS  PubMed  Google Scholar 

  22. Makohon-Moore, A. P. et al. Precancerous neoplastic cells can move through the pancreatic ductal system. Nature 561, 201–205 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. 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 

  24. Jones, S. et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 321, 1801–1806 (2008). This is the first article to describe exome sequencing of pancreatic cancer, reporting frequencies of somatic mutations and altered signalling pathways.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Collisson, E. A. et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat. Med. 17, 500–503 (2011). This article describes expression patterns in pancreatic cancer, and classifies tumours into three prognostic and predictive subtypes.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Moffitt, R. A. et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat. Genet. 47, 1168–1178 (2015). This article describes expression patterns in pancreatic cancer and its associated stroma, and independently classifies tumours into two prognostic and predictive subtypes and stroma into two prognostic subtypes.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Boj, S. F. et al. Organoid models of human and mouse ductal pancreatic cancer. Cell 160, 324–338 (2015).

    CAS  PubMed  Google Scholar 

  28. Huang, L. et al. Ductal pancreatic cancer modeling and drug screening using human pluripotent stem cell- and patient-derived tumor organoids. Nat. Med. 21, 1364–1371 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Lüttges, J. et al. The K-ras mutation pattern in pancreatic ductal adenocarcinoma usually is identical to that in associated normal, hyperplastic, and metaplastic ductal epithelium. Cancer 85, 1703–1710 (1999).

    PubMed  Google Scholar 

  30. Wilentz, R. E. et al. Inactivation of the p16 (INK4A) tumor-suppressor gene in pancreatic duct lesions: loss of intranuclear expression. Cancer Res. 58, 4740–4744 (1998).

    CAS  PubMed  Google Scholar 

  31. Wilentz, R. E. et al. Loss of expression of Dpc4 in pancreatic intraepithelial neoplasia: evidence that DPC4 inactivation occurs late in neoplastic progression. Cancer Res. 60, 2002–2006 (2000).

    CAS  PubMed  Google Scholar 

  32. Hutchings, D. et al. Cancerization of the pancreatic ducts: demonstration of a common and under-recognized process using immunolabeling of paired duct lesions and invasive pancreatic ductal adenocarcinoma for p53 and Smad4 expression. Am. J. Surg. Pathol. 42, 1556–1561 (2018).

    PubMed  PubMed Central  Google Scholar 

  33. Li, C. et al. Identification of pancreatic cancer stem cells. Cancer Res. 67, 1030–1037 (2007).

    CAS  PubMed  Google Scholar 

  34. Hermann, P. C. et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 1, 313–323 (2007).

    CAS  PubMed  Google Scholar 

  35. Huch, M. et al. Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. EMBO J. 32, 2708–2721 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Hingorani, S. R. et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 4, 437–450 (2003).

    CAS  PubMed  Google Scholar 

  37. Hong, S. M. et al. Telomeres are shortened in acinar-to-ductal metaplasia lesions associated with pancreatic intraepithelial neoplasia but not in isolated acinar-to-ductal metaplasias. Mod. Pathol. 24, 256–266 (2011).

    PubMed  Google Scholar 

  38. Matsuda, Y. et al. The prevalence and clinicopathological characteristics of high-grade pancreatic intraepithelial neoplasia: autopsy study evaluating the entire pancreatic parenchyma. Pancreas 46, 658–664 (2017).

    PubMed  Google Scholar 

  39. Goggins, M., Hruban, R. H. & Kern, S. E. BRCA2 is inactivated late in the development of pancreatic intraepithelial neoplasia: evidence and implications. Am. J. Pathol. 156, 1767–1771 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium. Pan-cancer analysis of whole genomes. Nature 578, 82–93 (2020). This article describes many germ line and somatic features of approximately 2,600 genomes amassed from many tumour types, including pancreatic cancer.

    Google Scholar 

  41. 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 

  42. Waddell, N. et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 518, 495–501 (2015). This article describes the patterns of structural variation obtained from whole-genome sequencing of pancreatic cancer, including those predictive of response to platinum-based chemotherapy.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. 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). This article describes the associations between germ line variants, somatic mutational signatures and antitumour immunity in tumours from two cohorts of patients with primary pancreatic cancer.

    PubMed  Google Scholar 

  44. Connor, A. A. et al. Integration of genomic and transcriptional features in pancreatic cancer reveals increased cell cycle progression in metastases. Cancer Cell 35, 267–282.e7 (2019). This article describes the associations between somatic driver gene inactivations, cell cycle progression, hypoxia and expression-based subtypes in primary and metastatic tumours from cohorts of patients with pancreatic cancer, as well as observing Halstedian spread to lymphatic and distant sites, and cases of isolated intraparenchymal spread.

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Whittle, M. C. et al. RUNX3 controls a metastatic switch in pancreatic ductal adenocarcinoma. Cell 161, 1345–1360 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Klein, W. M., Hruban, R. H., Klein-Szanto, A. J. & Wilentz, R. E. Direct correlation between proliferative activity and dysplasia in pancreatic intraepithelial neoplasia (PanIN): additional evidence for a recently proposed model of progression. Mod. Pathol. 15, 441–447 (2002).

    PubMed  Google Scholar 

  48. Wang, J. D. et al. Clinicopathological significance of SMAD4 loss in pancreatic ductal adenocarcinomas: a systematic review and meta-analysis. Oncotarget 8, 16704–16711 (2017).

    PubMed  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. McIntyre, C. A. et al. Alterations in driver genes are predictive of survival in patients with resected pancreatic ductal adenocarcinoma. Cancer 126, 3939–3949 (2020).

    CAS  PubMed  Google Scholar 

  51. The Cancer Genome Atlas Research Network. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell 32, 185–203.e13 (2017).

    PubMed Central  Google Scholar 

  52. Aguirre, A. J. et al. Activated Kras and Ink4a/Arf deficiency cooperate to produce metastatic pancreatic ductal adenocarcinoma. Genes Dev. 17, 3112–3126 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Izeradjene, K. et al. Kras(G12D) and Smad4/Dpc4 haploinsufficiency cooperate to induce mucinous cystic neoplasms and invasive adenocarcinoma of the pancreas. Cancer Cell 11, 229–243 (2007).

    CAS  PubMed  Google Scholar 

  54. Hingorani, S. R. et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 7, 469–483 (2005).

    CAS  PubMed  Google Scholar 

  55. Alexandrov, L. B., Nik-Zainal, S., Wedge, D. C., Campbell, P. J. & Stratton, M. R. Deciphering signatures of mutational processes operative in human cancer. Cell Rep. 3, 246–259 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Zou, X. et al. Validating the concept of mutational signatures with isogenic cell models. Nat. Commun. 9, 1744 (2018).

    PubMed  PubMed Central  Google Scholar 

  57. Alexandrov, L. B. et al. Clock-like mutational processes in human somatic cells. Nat. Genet. 47, 1402–1407 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Alexandrov, L. B., Nik-Zainal, S., Siu, H. C., Leung, S. Y. & Stratton, M. R. A mutational signature in gastric cancer suggests therapeutic strategies. Nat. Commun. 6, 8683 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Grant, R. C. et al. Clinical and genomic characterisation of mismatch repair deficient pancreatic adenocarcinoma. Gut 70, 1894–1903 (2020).

    PubMed  Google Scholar 

  60. Harada, T. et al. Genome-wide DNA copy number analysis in pancreatic cancer using high-density single nucleotide polymorphism arrays. Oncogene 27, 1951–1960 (2008).

    CAS  PubMed  Google Scholar 

  61. Fu, B., Luo, M., Lakkur, S., Lucito, R. & Iacobuzio-Donahue, C. A. Frequent genomic copy number gain and overexpression of GATA-6 in pancreatic carcinoma. Cancer Biol. Ther. 7, 1593–1601 (2008).

    CAS  PubMed  Google Scholar 

  62. Bailey, P. et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 531, 47–52 (2016). This article describes somatic gene expression by RNA sequencing in primary pancreatic cancers and classifies tumours into four prognostic subtypes.

    CAS  PubMed  Google Scholar 

  63. Cortés-Ciriano, I. et al. Comprehensive analysis of chromothripsis in 2,658 human cancers using whole-genome sequencing. Nat. Genet. 52, 331–341 (2020).

    PubMed  PubMed Central  Google Scholar 

  64. McDonald, O. G. et al. Epigenomic reprogramming during pancreatic cancer progression links anabolic glucose metabolism to distant metastasis. Nat. Genet. 49, 367–376 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Feigin, M. E. et al. Recurrent noncoding regulatory mutations in pancreatic ductal adenocarcinoma. Nat. Genet. 49, 825–833 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Rheinbay, E. et al. Analyses of non-coding somatic drivers in 2,658 cancer whole genomes. Nature 578, 102–111 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Rashid, N. U. et al. Purity independent subtyping of tumors (PurIST), a clinically robust, single-sample classifier for tumor subtyping in pancreatic cancer. Clin. Cancer Res. 26, 82–92 (2020).

    PubMed  Google Scholar 

  68. N Kalimuthu, S. et al. Morphological classification of pancreatic ductal adenocarcinoma that predicts molecular subtypes and correlates with clinical outcome. Gut 69, 317–328 (2020).

    PubMed  Google Scholar 

  69. Hayashi, A. et al. A unifying paradigm for transcriptional heterogeneity and squamous features in pancreatic ductal adenocarcinoma. Nat. Cancer 1, 59–74 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Brunton, H. et al. HNF4A and GATA6 loss reveals therapeutically actionable subtypes in pancreatic cancer. Cell Rep. 31, 107625 (2020).

    CAS  PubMed  Google Scholar 

  71. O’Kane, G. M. et al. GATA6 expression distinguishes classical and basal-like subtypes in advanced pancreatic cancer. Clin. Cancer Res. 26, 4901–4910 (2020).

    PubMed  Google Scholar 

  72. Chan-Seng-Yue, M. et al. Transcription phenotypes of pancreatic cancer are driven by genomic events during tumor evolution. Nat. Genet. 52, 231–240 (2020). This article describes bulk and single-cell RNA sequencing of primary and metastatic pancreatic cancers, classifies tumours into five subtypes driven by KRAS allelic imbalance and demonstrates that tumours contain a mixture of cells of each subtype.

    CAS  PubMed  Google Scholar 

  73. Juiz, N. et al. Basal-like and classical cells coexist in pancreatic cancer revealed by single-cell analysis on biopsy-derived pancreatic cancer organoids from the classical subtype. FASEB J. 34, 12214–12228 (2020).

    CAS  PubMed  Google Scholar 

  74. Mueller, S. et al. Evolutionary routes and KRAS dosage define pancreatic cancer phenotypes. Nature 554, 62–68 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. 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 

  76. Yachida, S. et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 467, 1114–1117 (2010). This article describes tumour heterogeneity within primary cancers and its relationship to multifocal metastases by analysis of simple somatic mutations in exome sequencing, and estimates the timing of pancreatic cancer progression from the cell of origin to distant spread.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Campbell, P. J. et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature 467, 1109–1113 (2010). This article describes tumour heterogeneity within primary cancers and its relationship to multifocal metastases by analysis of structural mutations in whole-genome sequencing, with insight into pancreatic carcinogenesis.

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Makohon-Moore, A. P. et al. Limited heterogeneity of known driver gene mutations among the metastases of individual patients with pancreatic cancer. Nat. Genet. 49, 358–366 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Sakamoto, H. et al. The evolutionary origins of recurrent pancreatic cancer. Cancer Discov. 10, 792–805 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. 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 (2018).

    CAS  PubMed  Google Scholar 

  81. Dentro, S. C. et al. Characterizing genetic intra-tumor heterogeneity across 2,658 human cancer genomes. Cell 184, 2239–2254 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Miyabayashi, K. et al. Intraductal transplantation models of human pancreatic ductal adenocarcinoma reveal progressive transition of molecular subtypes. Cancer Discov. 10, 1566–1589 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Brockie, E., Anand, A. & Albores-Saavedra, J. Progression of atypical ductal hyperplasia/carcinoma in situ of the pancreas to invasive adenocarcinoma. Ann. Diagn. Pathol. 2, 286–292 (1998).

    CAS  PubMed  Google Scholar 

  84. Haeno, H. et al. Computational modeling of pancreatic cancer reveals kinetics of metastasis suggesting optimum treatment strategies. Cell 148, 362–375 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Gerstung, M. et al. The evolutionary history of 2,658 cancers. Nature 578, 122–128 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Pushalkar, S. et al. The pancreatic cancer microbiome promotes oncogenesis by induction of innate and adaptive immune suppression. Cancer Discov. 8, 403–416 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Riquelme, E. et al. Tumor microbiome diversity and composition influence pancreatic cancer outcomes. Cell 178, 795–806.e12 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Geller, L. T. et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science 357, 1156–1160 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Knudsen, E. S. et al. Stratification of pancreatic ductal adenocarcinoma: combinatorial genetic, stromal, and immunologic markers. Clin. Cancer Res. 23, 4429–4440 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Balli, D., Rech, A. J., Stanger, B. Z. & Vonderheide, R. H. Immune cytolytic activity stratifies molecular subsets of human pancreatic cancer. Clin. Cancer Res. 23, 3129–3138 (2017).

    CAS  PubMed  Google Scholar 

  91. Bailey, P. et al. Exploiting the neoantigen landscape for immunotherapy of pancreatic ductal adenocarcinoma. Sci. Rep. 6, 35848 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Romero, J. M. et al. A four-chemokine signature is associated with a T-cell-inflamed phenotype in primary and metastatic pancreatic cancer. Clin. Cancer Res. 26, 1997–2010 (2020).

    CAS  PubMed  Google Scholar 

  93. 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 

  94. Özdemir, B. C. et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 25, 719–734 (2014).

    PubMed  PubMed Central  Google Scholar 

  95. Rhim, A. D. et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 25, 735–747 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Van Cutsem, E. et al. Randomized phase III trial of pegvorhyaluronidase alfa with nab-paclitaxel plus gemcitabine for patients with hyaluronan-high metastatic pancreatic adenocarcinoma. J. Clin. Oncol. 38, 3185–3194 (2020).

    PubMed  PubMed Central  Google Scholar 

  97. Hu, Z. I. et al. A randomized phase II trial of nab-paclitaxel and gemcitabine with tarextumab or placebo in patients with untreated metastatic pancreatic cancer. Cancer Med. 8, 5148–5157 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Ligorio, M. et al. Stromal microenvironment shapes the intratumoral architecture of pancreatic cancer. Cell 178, 160–175.e27 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Zapatka, M. et al. The landscape of viral associations in human cancers. Nat. Genet. 52, 320–330 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Rooney, M. S., Shukla, S. A., Wu, C. J., Getz, G. & Hacohen, N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell 160, 48–61 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. 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 

  102. 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 

  103. Sausen, M. et al. Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients. Nat. Commun. 6, 7686 (2015).

    PubMed  Google Scholar 

  104. Shen, S. Y. et al. Sensitive tumour detection and classification using plasma cell-free DNA methylomes. Nature 563, 579–583 (2018).

    CAS  PubMed  Google Scholar 

  105. Moore, M. J. et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J. Clin. Oncol. 25, 1960–1966 (2007).

    CAS  PubMed  Google Scholar 

  106. da Cunha Santos, G. et al. Molecular predictors of outcome in a phase 3 study of gemcitabine and erlotinib therapy in patients with advanced pancreatic cancer: National Cancer Institute of Canada Clinical Trials Group study PA.3. Cancer 116, 5599–5607 (2010).

    PubMed  Google Scholar 

  107. Boeck, S. et al. EGFR pathway biomarkers in erlotinib-treated patients with advanced pancreatic cancer: translational results from the randomised, crossover phase 3 trial AIO-PK0104. Br. J. Cancer 108, 469–476 (2013).

    CAS  PubMed  Google Scholar 

  108. Bryant, K. L. et al. Combination of ERK and autophagy inhibition as a treatment approach for pancreatic cancer. Nat. Med. 25, 628–640 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Canon, J. et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature 575, 217–223 (2019).

    CAS  PubMed  Google Scholar 

  110. Janes, M. R. et al. Targeting KRAS mutant cancers with a covalent G12C-specific inhibitor. Cell 172, 578–589.e17 (2018).

    CAS  PubMed  Google Scholar 

  111. Melisi, D. et al. Galunisertib plus gemcitabine vs. gemcitabine for first-line treatment of patients with unresectable pancreatic cancer. Br. J. Cancer 119, 1208–1214 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Salvador-Barbero, B. et al. CDK4/6 inhibitors impair recovery from cytotoxic chemotherapy in pancreatic adenocarcinoma. Cancer Cell 37, 340–353.e6 (2020).

    CAS  PubMed  Google Scholar 

  113. Laetsch, T. W. et al. Larotrectinib for paediatric solid tumours harbouring NTRK gene fusions: phase 1 results from a multicentre, open-label, phase 1/2 study. Lancet Oncol. 19, 705–714 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Jones, M. R. et al. NRG1 gene fusions are recurrent, clinically actionable gene rearrangements in KRAS wild-type pancreatic ductal adenocarcinoma. Clin. Cancer Res. 25, 4674–4681 (2019).

    CAS  PubMed  Google Scholar 

  115. Heining, C. et al. NRG1 fusions in KRAS wild-type pancreatic cancer. Cancer Discov. 8, 1087–1095 (2018).

    CAS  PubMed  Google Scholar 

  116. Aguirre, A. J. Oncogenic NRG1 fusions: a new hope for targeted therapy in pancreatic cancer. Clin. Cancer Res. 25, 4589–4591 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Golan, T. et al. Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N. Engl. J. Med. 381, 317–327 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Shroff, R. T. et al. Rucaparib monotherapy in patients with pancreatic cancer and a known deleterious BRCA mutation. JCO Precis. Oncol. https://doi.org/10.1200/PO.17.00316 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  119. Golan, T. et al. Increased rate of complete pathologic response after neoadjuvant FOLFIRINOX for BRCA mutation carriers with borderline resectable pancreatic cancer. Ann. Surg. Oncol. 27, 3963–3970 (2020).

    PubMed  Google Scholar 

  120. Pishvaian, M. J. et al. Overall survival in patients with pancreatic cancer receiving matched therapies following molecular profiling: a retrospective analysis of the Know Your Tumor registry trial. Lancet Oncol. 21, 508–518 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. 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 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Pishvaian, M. J. et al. Molecular profiling of patients with pancreatic cancer: initial results from the Know Your Tumor initiative. Clin. Cancer Res. 24, 5018–5027 (2018).

    CAS  PubMed  Google Scholar 

  124. Connor, A. A. & Gallinger, S. Next generation sequencing of pancreatic ductal adenocarcinoma: right or wrong? Expert Rev. Gastroenterol. Hepatol. 11, 683–694 (2017).

    CAS  PubMed  Google Scholar 

  125. Boot, A. et al. In-depth characterization of the cisplatin mutational signature in human cell lines and in esophageal and liver tumors. Genome Res. 28, 654–665 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Pich, O. et al. The mutational footprints of cancer therapies. Nat. Genet. 51, 1732–1740 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Hopkins, J. F. et al. Mutations in mitochondrial DNA from pancreatic ductal adenocarcinomas associate with survival times of patients and accumulate as tumors progress. Gastroenterology 154, 1620–1624.e5 (2018).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge G. O’Kane and F. Notta for proofreading the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Steven Gallinger.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Cancer thanks Andrew Biankin, Helmut Friess 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.

Related links

Know Your Tumor: https://www.pancan.org/for-healthcare-professionals/know-your-tumor/

Precede Consortium: https://precedeconsortium.org/

SEER: https://seer.cancer.gov

Glossary

Pancreatitis

Inflammation of the pancreas, the causes of which include gallstones, alcohol and germline predisposition.

Pancreatic intraepithelial neoplasms

(PanINs). Microscopic lesions, either flat or papillary, arising in the lining of the intrapancreatic ducts and composed of cuboidal or columnar cells with degrees of cytological and architectural atypia.

Intraductal papillary mucinous neoplasms

Microscopic to macroscopic lesions arising in the lining of the intrapancreatic ducts, either main or branch ducts, and composed of mucinous epithelial cells with degrees of cytological and architectural atypia.

Mucinous cystic neoplasms

Macroscopic cystic lesions arising in the pancreas, usually the body and tail, without communication with the pancreatic duct, and composed of mucinous epithelial cells with degrees of cytological and architectural atypia.

Lynch syndrome

The result of heterozygous germline deficiency in mismatch repair genes (MLH1, MSH2, MSH6 and PMS2), which causes an increased risk of certain cancers, especially colorectal, endometrial and pancreatic adenocarcinomas.

Cytological atypia

Abnormal cellular appearance, which could be the shape, colour or size of the entire cell or intracellular contents, often including large, irregularly shaped and hyperchromic nuclei.

Acinar-to-ductal metaplasias

Metaplasias formed from a process that involves pancreatic acinar cells differentiating into duct-like cells.

Mutational signatures

Reproducible patterns of somatic changes in DNA, most commonly identified by considering the 96 possible single-nucleotide substitutions in a trinucleotide context (mutated base and bases immediately 5′ and 3′ to it), that are thought to arise from different mutational processes active during the course of cancer development, including endogenous DNA repair deficiencies and exogenous carcinogens (for example, UV radiation and smoking).

Biallelic inactivation

Two-step process by which both alleles of a gene are lost, often consisting of an inactivating mutation of one copy of the gene and a structural deletion of the other copy, resulting in loss of heterozygosity and loss of that gene’s function, thus commonly observed with tumour suppressor genes during carcinogenesis.

Structural variants

Alteration of a region of DNA, typically 1 kb or greater, by a number of mechanisms, including duplication, deletion, inversion and translocation.

Polyploidization

The acquisition of one or more additional sets of chromosomes by a normally haploid or diploid cell, often consisting of whole-genome duplication in carcinogenesis.

Chromothripsis

A mutational process characterized by up to thousands of structural variations occurring as a single event in localized regions of one chromosome or a few chromosomes.

Allelic imbalance

The unequal expression of two alleles of a gene, which can be caused by several mechanisms, including copy number changes or epigenetic inactivation.

Jaccard similarity index

A statistic used to measure the similarity (or inversely diversity) of two or more sample sets, calculated as the intersection divided by the union of the members of those sets.

Breakage–fusion–bridge cycles

This structural variation arises due to telomere loss at one end of a chromosome, leading to fusion at that identical site on the sister chromatid in prophase, then breakage of the chromosomes at a random site during chromatid separation in anaphase, causing uneven distribution of genetic material to daughter nuclei, and repetition of the cycle with subsequent cell divisions.

Synchronous PDAC

Defined as two or more neoplasms identified simultaneously or up to 6 months apart in the same patient.

Metachronous PDAC

Defined as two or more neoplasms identified more than 6 months apart in the same patient.

Neutral mutations

A DNA mutation that is independent of natural selection, such as a synonymous base substitution.

Synthetic lethality

Cellular or organismal death due to the simultaneous perturbation of two genes, often when inactivation of either gene alone has minimal effect.

Somatic BRCA mutation reversion

Somatic restoration of one or more BRCA1 or BRCA2 alleles, resulting in functional restoration of homologous recombination DNA repair.

Umbrella trials

A clinical trial design involving patient inclusion and exclusion criteria based on tumour biomarkers rather than tissue or site of origin, especially as those biomarkers inform therapeutic vulnerability.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Connor, A.A., Gallinger, S. Pancreatic cancer evolution and heterogeneity: integrating omics and clinical data. Nat Rev Cancer 22, 131–142 (2022). https://doi.org/10.1038/s41568-021-00418-1

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-021-00418-1

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