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.

  • Article
  • Published:

A unifying paradigm for transcriptional heterogeneity and squamous features in pancreatic ductal adenocarcinoma

Abstract

Pancreatic cancer expression profiles largely reflect a classical or basal-like phenotype. The extent to which these profiles vary within a patient is unknown. We integrated evolutionary analysis and expression profiling in multiregion-sampled metastatic pancreatic cancers, finding that squamous features are the histologic correlate of an RNA-seq-defined basal-like subtype. In patients with coexisting basal and squamous and classical and glandular morphology, phylogenetic studies revealed that squamous morphology represented a subclonal population in an otherwise classical and glandular tumor. Cancers with squamous features were significantly more likely to have clonal mutations in chromatin modifiers, intercellular heterogeneity for MYC amplification and entosis. These data provide a unifying paradigm for integrating basal-type expression profiles, squamous histology and somatic mutations in chromatin modifier genes in the context of clonal evolution of pancreatic cancer.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Study overview and morphologic heterogeneity for squamous features in PDAC.
Fig. 2: Transcriptional heterogeneity for SF in end-stage PDAC.
Fig. 3: Expressional profiles based on three major classification schemes in end-stage PDAC.
Fig. 4: Genomic landscape of end-stage PDAC with and without SF.
Fig. 5: Integration of transcriptomic and morphologic features with phylogenetic patterns in PDAC PAM55 with clonal KMT2C mutation.
Fig. 6: Integration of transcriptomic and morphologic features with phylogenetic patterns in PDAC PAM02 with clonal ARID1A mutation.
Fig. 7: Integration of transcriptomic and morphologic features with phylogenetic patterns in PDAC PAM46 with MYC amplification.
Fig. 8: Squamous features in pancreatic ductal adenocarcinoma correspond to enhancement of MYC.

Similar content being viewed by others

Data availability

RNA and DNA sequence data for this study have been deposited at the European Genome-phenome Archive under accession number EGAS00001003974. Published gene sets analyzed here are available from previous papers6,10,11. Sequencing data from the MSK IMPACT cohort that were analyzed here18 are publicly available at cBioPortal (https://www.cbioportal.org/). The other human resected pancreatic cancer data were derived from TCGA Research Network: http://cancergenome.nih.gov/. The dataset derived from this resource that supports the findings of this study is available through Firebrowse (http://firebrowse.org/). Source data for Figs. 1, 3, 4, 8 and Extended Data Figs. 24 and 10 have been provided as Source Data Figs. 1, 3, 4 and 8 and Source Data Extended Data Figs. 24 and 10. All other data supporting the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Kamisawa, T., Wood, L. D., Itoi, T. & Takaori, K. Pancreatic cancer. Lancet 388, 73–85 (2016).

    CAS  PubMed  Google Scholar 

  2. Gillen, S., Schuster, T., Meyer Zum Buschenfelde, C., Friess, H. & Kleeff, J. Preoperative/neoadjuvant therapy in pancreatic cancer: a systematic review and meta-analysis of response and resection percentages. PLoS Med. 7, e1000267 (2010).

    PubMed  PubMed Central  Google Scholar 

  3. Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, 2019. CA Cancer J. Clin. 69, 7–34 (2019).

    PubMed  Google Scholar 

  4. 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 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    PubMed Central  Google Scholar 

  8. 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 

  9. Wilentz, R. E. et al. Genetic, immunohistochemical, and clinical features of medullary carcinoma of the pancreas: a newly described and characterized entity. Am. J. Pathol. 156, 1641–1651 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. 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 

  11. 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 

  12. Collisson, E. A., Bailey, P., Chang, D. K. & Biankin, A. V. Molecular subtypes of pancreatic cancer. Nat. Rev. Gastroenterol. Hepatol. 16, 207–220 (2019).

    PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Brody, J. R. et al. Adenosquamous carcinoma of the pancreas harbors KRAS2, DPC4 and TP53 molecular alterations similar to pancreatic ductal adenocarcinoma. Mod. Pathol. 22, 651–659 (2009).

    CAS  PubMed  Google Scholar 

  15. 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 

  16. Fukushima, N. et al. In WHO Classification of Tumors 4th edn. (ed. F.T. Bosman et al.), 292–296 (2010).

  17. Cheng, D. T. et al. Memorial Sloan Kettering-integrated mutation profiling of actionable cancer targets (MSK IMPACT): a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J. Mol. Diagn. 17, 251–264 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Zehir, A. et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat. Med. 23, 703–713 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Overholtzer, M. et al. A nonapoptotic cell death process, entosis, that occurs by cell-in-cell invasion. Cell 131, 966–979 (2007).

    CAS  PubMed  Google Scholar 

  20. Mackay, H. L. et al. Genomic instability in mutant p53 cancer cells upon entotic engulfment. Nat. Commun. 9, 3070 (2018).

    PubMed  PubMed Central  Google Scholar 

  21. Torphy, R. J. et al. Stromal content is correlated with tissue site, contrast retention, and survival in pancreatic adenocarcinoma. JCO Precis. Oncol. https://doi.org/10.1200/PO.17.00121 (2019).

  22. Liu, C. et al. The UPF1 RNA surveillance gene is commonly mutated in pancreatic adenosquamous carcinoma. Nat. Med. 20, 596–598 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. 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.e518 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Lomberk, G. et al. Distinct epigenetic landscapes underlie the pathobiology of pancreatic cancer subtypes. Nat. Commun. 9, 1978 (2018).

    PubMed  PubMed Central  Google Scholar 

  25. Schleger, C., Verbeke, C., Hildenbrand, R., Zentgraf, H. & Bleyl, U. c-MYC activation in primary and metastatic ductal adenocarcinoma of the pancreas: incidence, mechanisms, and clinical significance. Mod. Pathol. 15, 462–469 (2002).

    CAS  PubMed  Google Scholar 

  26. Wirth, M., Mahboobi, S., Kramer, O. H. & Schneider, G. Concepts to Target MYC in pancreatic cancer. Mol. Cancer Ther. 15, 1792–1798 (2016).

    CAS  PubMed  Google Scholar 

  27. Stine, Z. E., Walton, Z. E., Altman, B. J., Hsieh, A. L. & Dang, C. V. MYC, metabolism, and cancer. Cancer Discov. 5, 1024–1039 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Bielski, C. M. et al. Genome doubling shapes the evolution and prognosis of advanced cancers. Nat. Genet. 50, 1189–1195 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Iacobuzio-Donahue, C. A. et al. Cancer biology as revealed by the research autopsy. Nat. Rev. 19, 686–697 (2019).

    CAS  Google Scholar 

  30. Simone, C. G. et al. Characteristics and outcomes of adenosquamous carcinoma of the pancreas. Gastroint. Cancer Res. 6, 75–79 (2013).

    Google Scholar 

  31. Yamaguchi, K. & Enjoji, M. Adenosquamous carcinoma of the pancreas: a clinicopathologic study. J. Surg. Oncol. 47, 109–116 (1991).

    CAS  PubMed  Google Scholar 

  32. Gonzalez-Vasconcellos, I. et al. The Rb1 tumour suppressor gene modifies telomeric chromatin architecture by regulating TERRA expression. Sci. Rep. 7, 42056 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Versteege, I., Medjkane, S., Rouillard, D. & Delattre, O. A key role of the hSNF5/INI1 tumour suppressor in the control of the G1-S transition of the cell cycle. Oncogene 21, 6403–6412 (2002).

    CAS  PubMed  Google Scholar 

  34. Mu, P. et al. SOX2 promotes lineage plasticity and antiandrogen resistance in TP53- and RB1-deficient prostate cancer. Science 355, 84–88 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Ku, S. Y. et al. Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance. Science 355, 78–83 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Boyd, C. A., Benarroch-Gampel, J., Sheffield, K. M., Cooksley, C. D. & Riall, T. S. 415 patients with adenosquamous carcinoma of the pancreas: a population-based analysis of prognosis and survival. J. Surg. Res. 174, 12–19 (2012).

    PubMed  Google Scholar 

  37. 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 

  38. Dawkins, J. B. et al. Reduced expression of histone methyltransferases KMT2C and KMT2D correlates with improved outcome in pancreatic ductal adenocarcinoma. Cancer Res. 76, 4861–4871 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Sun, Q. et al. Competition between human cells by entosis. Cell Res. 24, 1299–1310 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. de la Cova, C., Abril, M., Bellosta, P., Gallant, P. & Johnston, L. A. Drosophila myc regulates organ size by inducing cell competition. Cell 117, 107–116 (2004).

    PubMed  Google Scholar 

  41. Claveria, C., Giovinazzo, G., Sierra, R. & Torres, M. Myc-driven endogenous cell competition in the early mammalian embryo. Nature 500, 39–44 (2013).

    CAS  PubMed  Google Scholar 

  42. Hamann, J. C. et al. Entosis is induced by glucose starvation. Cell Rep. 20, 201–210 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    CAS  PubMed  Google Scholar 

  44. Engstrom, P. G. et al. Systematic evaluation of spliced alignment programs for RNA-seq data. Nat. Meth. 10, 1185–1191 (2013).

    Google Scholar 

  45. Wagner, G. P., Kin, K. & Lynch, V. J. Measurement of mRNA abundance using RNA-seq data: RPKM measure is inconsistent among samples. Theor. Biosci. 131, 281–285 (2012).

    CAS  Google Scholar 

  46. Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Oki, S. et al. ChIP-Atlas: a data-mining suite powered by full integration of public ChIP-seq data. EMBO Rep. 19, e46255 (2018).

    PubMed  PubMed Central  Google Scholar 

  49. Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. 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 

  51. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. DePristo, M. A. et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 43, 491–498 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Mose, L. E., Wilkerson, M. D., Hayes, D. N., Perou, C. M. & Parker, J. S. ABRA: improved coding indel detection via assembly-based realignment. Bioinformatics 30, 2813–2815 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Cibulskis, K. et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 31, 213–219 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Tokheim, C. J., Papadopoulos, N., Kinzler, K. W., Vogelstein, B. & Karchin, R. Evaluating the evaluation of cancer driver genes. Proc. Natl Acad. Sci. USA 113, 14330–14335 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Davoli, T. et al. Cumulative haploinsufficiency and triplosensitivity drive aneuploidy patterns and shape the cancer genome. Cell 155, 948–962 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Lawrence, M. S. et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499, 214–218 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Yachida, S. et al. Clinical significance of the genetic landscape of pancreatic cancer and implications for identification of potential long-term survivors. Clin. Cancer Res. 18, 6339–6347 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).

    PubMed  PubMed Central  Google Scholar 

  61. Reiter, J. G. et al. Reconstructing metastatic seeding patterns of human cancers. Nat. Commun. 8, 14114 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Shen, R. & Seshan, V. E. FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res. 44, e131 (2016).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to G. Askan, A. Yavas and J.V. Egger for assistance in identifying resected adenosquamous samples for use in this study, to S. Yamamoto for analysis tool information and to S. Oki for technical support. We gratefully acknowledge the members of the Molecular Diagnostics Service in the Department of Pathology for MSK IMPACT. This work was supported by National Institutes of Health grant nos. R01 CA179991 and R35 CA220508 to C.I.D., F31 CA180682 and 2T32 CA160001-06 to A.M.M. and CA62924 to R.H.H., the Daiichi-Sankyo Foundation of Life Science Fellowship to A.H., the Mochida Memorial Foundation for Medical and Pharmaceutical Research Fellowship to A.H., Cycle for Survival and the Marie-Josée and Henry R. Kravis Center for Molecular Oncology. MSK IMPACT was funded in part by the Marie-Josée and Henry R. Kravis Center for Molecular Oncology and the National Cancer Institute Cancer Center Core grant no. P30-CA008748.

Author information

Authors and Affiliations

Authors

Contributions

A.H. and C.A.I.-D. designed the study. A.H., J.F., A.P.M.-M., H.S., M.A.A., A.B., R.K., P.B., L.D.W., R.H.H. and C.A.I.-D. collected autopsy samples. A.H. and C.A.I.-D. reviewed the histology of autopsy samples and selected cases. O.B., D.S.K., A.H. and C.A.I.-D. reviewed the pathology of MSK Clinical IMPACT cases. A.H. and C.A.I.-D. reviewed the pathology of surgical cases in TCGA cohort. A.H., R.C., M.O., K.C., M.L., G.J.N. and C.A.I.-D. reviewed the entosis of Immuno-FISH slides. A.H. and J.F. prepared RNA samples. A.P.M.-M., J.Hong, H.S., Z.A.K. and A.H. prepared the DNA samples. A.H., Y.Z. and C.A.I.-D. performed RNA sequencing. Y.H., A.H., L.Z. and J.Huang analyzed RNA sequencing results. A.P.M.-M., J. Ho., Z.A.K., H.S. M.A.A., A.H., and C.A.I.-D. performed DNA sequencing. M.A.A., A.P.M.-M., J.Hong, A.H. and C.A.I.-D. analyzed DNA sequencing results and derived the phylogenies. A.P.M.-M., J.Hong, A.H., J.P.M. and C.A.I.-D. managed the sequencing data. W.W. and E.M.O. collected samples and clinical information for MSK Clinical IMPACT. M.L., K.C. and G.J.N. performed immuno-FISH. J.B. and N.L. performed organoid experiments. A.H., R.C., Y.H., M.O., N.L. and C.A.I.-D. wrote the manuscript. All authors reviewed and edited the final manuscript.

Corresponding author

Correspondence to Christine A. Iacobuzio-Donahue.

Ethics declarations

Competing interests

D.S.K. is a consultant and equity holder to Paige.AI and a consultant to Merck Pharmaceuticals and receives royalties from UpToDate and the American Registry of Pathology.

Additional information

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

Extended Data

Extended Data Fig. 1 Case Selection and Postmortem Diagnosis.

(a) Schematic of case selection for current study. (b) Immunolabelling for glandular and squamous or squamoid components in 13 Representative PDACs. All regions with squamous differentiation (SD) showed positivity for CK5/6 and p63, whereas no labeling was observed in regions with glandular morphology (GL). In two PDACs with the neoplastic cells stained positive for CK5/6 but were negative for p63 and thus classified as having squamoid features. n.d., no immunolabeling performed. IHC was done for 31 case, 167 slides. Scale bar: 100 um. (c) Postmortem case diagnoses. Seven cases corresponded to adenosquamous carcinoma (ASC), two cases showed squamoid features (SF) and four cases showed focal (<30%) squamous differentiation (SD).

Extended Data Fig. 2 Squamous Features of Pancreatic Ductal Adenocarcinoma in the TCGA and MSK Clinical IMPACT Patient Cohorts.

(a) Schematic for histological classification of cases in the TCGA and MSK clinical IMPACT cohorts. (b) Schematic for case selection in the TCGA and MSK clinical IMPACT cohorts. (c) Frequency of case diagnoses in TCGA (n = 145), MSK Clinical IMPACT (n = 617) and our autopsy (n = 123) cohort. (d) Representative digital images of adenosquamous carcinoma (ASC) (out of 3 cases), PDAC with potential squamoid feature or squamous differentiation (PDAC with potential SF/SD) (histologic diagnosis modified based on our re-review) (out of 9 cases), and poorly differentiated ductal adenocarcinoma (out of 57 cases) in TCGA. Alveolar or trabecular pattern was confirmed in ASC or PDAC with potential SF/SD. (e) Kaplan-Meier analysis showed poor prognosis of ASC or PDAC with potential SF/SD (n = 12) compared to conventional PDAC (n = 129) in TCGA cohort (P < 0.0001, Log-rank test). (f) Kaplan-Meier analysis showed poor prognosis of ASC or PDAC with potential SF/SD (n = 70) compared to conventional PDAC (n = 494) in MSK-IMPACT cohort (P = 0.001, Log-rank test).

Source data

Extended Data Fig. 3 Squamous Feature Associated Alteration and Characteristic in Pancreatic Ductal Adenocarcinoma.

(a) Entotic CICs in matched glandular (GL) versus squamoid or squamous morphology (SF/SD) in 10 patients. Statistics are performed using Mann–Whitney U test, two-sided. (62, 62, 73, 38, 50, 26, 33, 21, 8 and 21 blocks/slides were used for entosis evaluation of PAM02, MPAM06, PAM73, PAM55, PAM22, PAM28, PAM53, PAM80, PAM20 and PAM39). (b) mRNA Expression of squamous markers (TP63, KRT5 and KRT6A) in samples with glandular growth pattern (GL) (n = 133), squamoid features (SF) (n = 18) and squamous differentiation (SD) (n = 63). SF have intermediate expression pattern between SD and GL. Each P-value is calculated by Mann–Whitney U test, two-sided. Lines and bars: median with interquartile range. (c) mRNA expression of TP63, KRT5 and KRT6A. ASC (n = 3) and PDAC with potential SF/SD (n = 9) have higher expression of TP63 than conventional PDAC (n = 133) in TCGA (P = 0.007, Mann–Whitney U test, two-sided). (d) Keratin network based on mRNA expression. In GL, KRT19 (normally expressed in ductal epithelia) is a hub in pancreas cancer. In SF, KRT6A and KTR5 (normally expressed in squamous epithelium) have some interaction. In SD, stratified squamous epithelium keratins (KRT4, KRT5, KRT13, KRT14) and heavy weight keratins (KRT1 and KRT10) are expressed in the network. (e)-(g) Tumor purity in PDACs with or without squamous feature. (e) Tumor purity by FACETs in end stage PDAC. Samples with squamous differentiation (SD) (n = 43) have higher tumor purity than samples with squamoid feature (SF) (n = 20) or glandular pattern (GL) (n = 152) (P = 0.012 or P < 0.001, Mann–Whitney U test, two-sided). Lines and bars: median with interquartile range. (f) Intratumoral heterogeneity of tumor purity in end stage PDAC. Samples with SF or SD have higher tumor purity in one tumor (9, 11, 8, 5, 6, 11, 5, 9, 4, 12, 6, 9, 21, 8, 14, 8, 10, 7, 11, 8, 3, 7, 9, 6 and 8 samples were used for PAM46, MPAM06, PAM54, PAM53, PAM32, PAM02, PAM28, PAM22, PAM16, PAM55, PAM20, PAM39, PAM52, PAM48, PAM24, PAM56, PAM51, PAM49, PAM03, PAM29, PAM25, PAM47, PAM50, PAM27, and PAM04) (g) Absolute tumor purity in TCGA cohort. Absolute tumor purity is not different between conventional PDAC (n = 132) and PDAC with potential SF/SD (n = 9) and ASC (n = 3) (P = 0.601, Mann–Whitney U test, two-sided).

Source data

Extended Data Fig. 4 Mutational Characteristics of the MSK clinical IMPACT cohort.

(a) Oncoprint illustrating somatic alterations of chromatin modifier genes, RB1 and MYC amplification in 617 PDAC cases including 26 ASCs and 51 PDACs with potential SF/SD. P-value was tested using two-sided Fisher’s exact test. * indicates P-value if analysis is confined to driver gene mutations only. (b) Entotic CIC are more frequent occur in TP53 mutant PDACs (n = 180) than in TP53 wild type PDACs (n = 42) (P = 0.021, Mann–Whitney U test, two-sided).

Source data

Extended Data Fig. 5 Integration of Transcriptomic and Morphologic Features with Phylogenetic Patterns in Pancreatic Ductal Adenocarcinoma (a-d) PAM54 with clonal KMT2C mutation and (e-h) PAM16 with clonal KDM6A mutation.

(a) Phylogenetic analysis illustrating the clonal relationship of samples analyzed in this patient. The predicted timing of somatic alterations in driver genes and whole genome duplication are also shown. Mutations in chromatin modifier genes are in red font, all others in orange. Clonal driver genes are notable for a KMT2C somatic alteration, whereas mutations in RB1 and SMARCA4 (two independent mutations) are present in a subset of samples. SD in this carcinoma was found in all samples analyzed, although it was admixed with a minor glandular component in some samples. (b) Principal components analysis (214 samples from 27 patients) highly similar expression between samples (all SD, n = 18) in PAM54. (c) Relationship of anatomic location to morphologic and transcriptional profiles. (d) Representative histologic images of tumors (out of total 108 histologic images for PAM54) in the same patient. Scale bar: 100um. (e) Clonal driver genes are notable for a KDM6A somatic alteration. SD in this carcinoma was found in all samples analyzed, although it was admixed with a minor GL component in some samples. (f) Principal components analysis (214 samples from 27 patients) illustrates highly similar expression between samples (all SD, n = 4) in PAM16. (g) Relationship of anatomic location to morphologic and transcriptional profiles. (h) Representative histologic images of metastatic tumors PT3 and PT4 (out of total 15 histologic images for PAM16). Scale bar: 100um.

Extended Data Fig. 6 Integration of Transcriptomic and Morphologic Features with Phylogenetic Patterns in Pancreatic Ductal Adenocarcinoma (a)-(d) PAM39 and (e)-(g) PAM20 with clonal ARID1A mutation.

(a) Phylogenetic analysis illustrating the clonal relationship of samples analyzed in this patient. The predicted timing of somatic alterations in driver genes, whole genome duplication and MYC amplification are shown. Mutations in chromatin modifier genes are in red font, all others in orange. Red outline indicates the one sample with SF based on histology and immunohistochemical analysis (squares) but a classical type expression profile (triangle). Clonal driver genes are notable for an ARID1A somatic alteration. SF is confined to one prostate metastasis sample (PT9). (b) Principal components analysis (214 samples from 27 patients) shows a similar gene expression profile between the samples with GL (n = 7) or SF (n = 1) morphology in PAM39. (c) Relationship of anatomic location to morphologic and transcriptional heterogeneity. (d) Representative histologic and/or immunohistochemical images of the primary (PT1) and metastasis (PT6, PT8, PT9) tumors (out of total 21 histologic images for PAM39). Scale bar: 100um. (e) Clonal driver genes are notable for an ARID1A somatic alteration. MYC amplification (≥ 6 copies) was detected in all samples with SF, and in a phylogenetically distinct sample with GL within the primary tumor. Samples with SF in this carcinoma (PT3-PT6) are clonally related. (f) Relationship of anatomic location to morphologic heterogeneity. The metastasis samples PT3-PT6 showed SF whereas the primary tumor samples showed GL. (g) Representative histologic and/or immunohistochemical images of the primary tumor (PT1) and diaphragm metastasis (PT5) (out of total 8 histologic images for PAM20). Scale bar: 100um.

Extended Data Fig. 7 Integration of Transcriptomic and Morphologic Features with Phylogenetic Patterns in Pancreatic Ductal Adenocarcinoma (a)-(d) PAM28 and (e)-(h) MPAM6 with clonal RB1 Mutation.

(a) Phylogenetic analysis illustrating the clonal relationship of samples analyzed in this patient. The predicted timing of somatic alterations in driver genes and whole genome duplication are shown. The mutation in RB1 is in red font, all others in orange. Purple outline indicates samples that have SD based on histology (squares). Clonal driver genes are notable for an RB1 somatic alteration. Samples with SD are more related to each other than to other samples in this patient. (b) Principal components analysis (214 samples from 27 patients) shows that samples PT1-PT3 with basal-like type expression and SD morphology (n = 5) are distinct from samples PT4 and PT5 that have basal-like type expression but GL morphology (n = 2) in PAM28. (c) Relationship of anatomic location to morphologic and transcriptional heterogeneity. Both GL and SF/SD were seen in the primary tumor, yet liver metastases PT4 and PT5 have GL morphology and a basal-like type expression profile. (d) Representative histologic images and immunohistochemical labeling of primary tumor sample PT1 and liver metastases PT4 and PT5 (out of total 26 histologic images for PAM28). Scale bar: 100um. (e) Clonal driver genes are notable for a deleterious RB1 mutation. Samples with SD (PT5-PT7) are more related to each other than to other samples in the same patient. (f) Principal components analysis (214 samples from 27 patients) indicates distinct gene expression profiles between GL (n = 8) and SD (n = 2) samples in MPAM06. (g) Relationship of anatomic location to morphologic and transcriptional heterogeneity. SD is confined to the liver metastases (PT5-PT7). (h) Representative histologic images of the primary and multiple metastatic tumors in the same patient (out of total 62 histologic images for MPAM6). Scale bar: 100um.

Extended Data Fig. 8 Integration of Transcriptomic and Morphologic Features with Phylogenetic Patterns in Pancreatic Ductal Adenocarcinoma (a)-(d) PAM22 and (e)-(h) PAM53.

(a) Phylogenetic analysis illustrating the clonal relationship of samples analyzed in this patient. Purple outline indicates samples that have SD based on RNAseq (triangles) and histology/immunohistochemistry (squares). The predicted timing of somatic alterations in driver genes, whole genome duplication and MYC amplification are also shown. SD is confined to a single sample within the multiregion sampled primary tumor (PT2). (b) Principal components analysis (214 samples from 27 patients) indicates that SD (n = 2) including PT2 show a different expression profile from all other primary tumor samples that have GL (n = 7) morphology in PAM22. (c) Relationship of anatomic location within the primary tumor to morphologic and/or transcriptional heterogeneity for SF/SD. (d) Representative histologic images of representative tumors in the same patient (out of total 50 histologic images for PAM22). Scale bar: 100um. (e) The one sample with a classical expression profile and GL (PT3) morphology forms the outgroup in the tree. Four samples with basal-like expression and SD correspond to both the primary tumor (PT4 and PT5) and metastasis (PT1 and PT2). (f) Principal components analysis (214 samples from 27 patients) indicates samples PT1 and PT3 have relatively different expression profiles from other SD samples (total 18 samples) in PAM53. (g) Relationship of anatomic location to morphologic and transcriptional heterogeneity. SD was found in one omental metastasis (PT3) which is also showed basal-like expression. (h) Representative histologic and immunohistochemical images of the primary tumor samples PT4 and PT5, liver metastasis PT1 and omental metastasis PT3 (out of total 33 histologic images for PAM22). Scale bar: 100um.

Extended Data Fig. 9 Morphologic Features with Phylogenetic Patterns in Pancreatic Ductal Adenocarcinoma PAM32.

(a) Phylogenetic analysis illustrating the clonal relationship of samples analyzed in this patient. The predicted timing of somatic alterations in driver genes and whole genome duplication are also shown. Purple outline indicates samples that have SD based on histology and immunolabeling (squares). Samples PT3-PT6 with SD are more closely related to each other than to other samples in the same patient. (b) Relationship of anatomic location to morphologic heterogeneity. One liver metastasis (PT7, not sequenced) showed GL morphology. (c) Representative histologic images of the primary and metastatic tumors in this patient (out of total 39 histologic images for PAM32). Scale bar: 100um.

Extended Data Fig. 10 Molecular Characteristics in Squamous Feature and Entosis.

(a)-(d) Impact of MYC-overexpression Using PDAC Organoid Models. (a) Overexpression of MYC and alteration status of chromatin modifier genes in eight PDAC organoids. Center value and bar: mean and SD. Three data points in each organoid means technical triplicates of qPCR data. (b) Representative images of PDAC organoids (out of 8 organoids, 64 images). Images of organoids were acquired 5 days post-sorting (10 days total post-infection). No obvious morphological changes were identified between the MYC-infected vs the mock-infected organoids. Scale bar: 50um. (c) Relative mRNA expression of squamous markers (TP63, KRT5 and KRT6A) after MYC overexpression. Two PDAC organoids with chromatin modifier mutations (HT160c and HT28) shows higher expression of all three markers whereas no effects are seen in the absence of mutations in these genes. Center value and bar: mean and SD. Three data points in each organoid means technical triplicates of qPCR data. **Appropriate KRT5 in HT151 signal was not detected with qPCR due to low expression. (d) Metabolic pathways are enriched in Entotic cases based on five different databases. (e) MYC expression in Entotic CIC. Winner (MYC positive)- Loser (MYC negative) pattern was identified both MYC amplified and non-amplified cases. W(P)-L(P), W(P)-L(N), W(N)-L(P) and W(N)-L(N) are Winner (MYC positive)-Loser (MYC positive), Winner (MYC positive)-Loser (MYC negative), Winner (MYC negative)-Loser (MYC positive) and Winner (MYC negative)-Loser (MYC negative) patterns respectively. (f) Representative image of Winner (positive)- Loser (negative) pattern (out of 9, 6, 1 and 14 images in W(P)-L(P), W(P)-L(N), W(N)-L(P) and W(N)-L(N) patterns).

Source data

Supplementary information

Supplementary Tables 1–10

Reporting Summary

Supplementary Dataset 1

S1 RNA expression of each sample (log2 conversion of DESeq2 normalized data).

Source data

Source Data Fig. 1

Statistical Source Data

Source Data Fig. 3

Statistical Source Data

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 8

Statistical Source Data

Source Data Extended Data Fig. 2

Statistical Source Data

Source Data Extended Data Fig. 3

Statistical Source Data

Source Data Extended Data Fig. 4

Statistical Source Data

Source Data Extended Data Fig. 10

Statistical Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hayashi, A., Fan, J., Chen, R. et al. A unifying paradigm for transcriptional heterogeneity and squamous features in pancreatic ductal adenocarcinoma. Nat Cancer 1, 59–74 (2020). https://doi.org/10.1038/s43018-019-0010-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-019-0010-1

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