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:

Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma

Abstract

Pancreatic ductal adenocarcinoma (PDAC) remains a lethal disease with a 5-year survival rate of 4%. A key hallmark of PDAC is extensive stromal involvement, which makes capturing precise tumor-specific molecular information difficult. Here we have overcome this problem by applying blind source separation to a diverse collection of PDAC gene expression microarray data, including data from primary tumor, metastatic and normal samples. By digitally separating tumor, stromal and normal gene expression, we have identified and validated two tumor subtypes, including a 'basal-like' subtype that has worse outcome and is molecularly similar to basal tumors in bladder and breast cancers. Furthermore, we define 'normal' and 'activated' stromal subtypes, which are independently prognostic. Our results provide new insights into the molecular composition of PDAC, which may be used to tailor therapies or provide decision support in a clinical setting where the choice and timing of therapies are critical.

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

Figure 1: Successful deconvolution of normal tissue with NMF.
Figure 2: The dual action of stroma is described by distinct gene expression patterns, which are not present in PDAC cell lines.
Figure 3: Tumor-specific gene expression suggests two subtypes of PDAC with similarities to other tumor types.
Figure 4: Multivariate survival analysis of tumor and stromal subtypes.
Figure 5: Associations between tumor and stromal subtypes, PDX tumors, KRAS mutations and SMAD4 expression.
Figure 6: Overcoming limited tumor cellularity shows true heterogeneity among matched primary and metastatic sites.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Gene Expression Omnibus

References

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Yachida, S. et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 467, 1114–1117 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Crnogorac-Jurcevic, T. et al. Expression profiling of microdissected pancreatic adenocarcinomas. Oncogene 21, 4587–4594 (2002).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  8. Iacobuzio-Donahue, C.A. et al. Exploration of global gene expression patterns in pancreatic adenocarcinoma using cDNA microarrays. Am. J. Pathol. 162, 1151–1162 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Logsdon, C.D. et al. Molecular profiling of pancreatic adenocarcinoma and chronic pancreatitis identifies multiple genes differentially regulated in pancreatic cancer. Cancer Res. 63, 2649–2657 (2003).

    CAS  PubMed  Google Scholar 

  10. Stuart, R.O. et al. In silico dissection of cell-type-associated patterns of gene expression in prostate cancer. Proc. Natl. Acad. Sci. USA 101, 615–620 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wang, Y. et al. In silico estimates of tissue components in surgical samples based on expression profiling data. Cancer Res. 70, 6448–6455 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yoshihara, K. et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat. Commun. 4, 2612 (2013).

    Article  CAS  PubMed  Google Scholar 

  13. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Biton, A. et al. Independent component analysis uncovers the landscape of the bladder tumor transcriptome and reveals insights into luminal and basal subtypes. Cell Rep. 9, 1235–1245 (2014).

    Article  CAS  PubMed  Google Scholar 

  16. Stratford, J.K. et al. A six-gene signature predicts survival of patients with localized pancreatic ductal adenocarcinoma. PLoS Med. 7, e1000307 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Whitfield, M.L. et al. Identification of genes periodically expressed in the human cell cycle and their expression in tumors. Mol. Biol. Cell 13, 1977–2000 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Froeling, F.E. et al. Retinoic acid—induced pancreatic stellate cell quiescence reduces paracrine Wnt–β-catenin signaling to slow tumor progression. Gastroenterology 141, 1486–1497 (2011).

    Article  CAS  PubMed  Google Scholar 

  19. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lee, J.J. et al. Stromal response to Hedgehog signaling restrains pancreatic cancer progression. Proc. Natl. Acad. Sci. USA 111, E3091–E3100 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Olive, K.P. et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324, 1457–1461 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Erkan, M. et al. The activated stroma index is a novel and independent prognostic marker in pancreatic ductal adenocarcinoma. Clin. Gastroenterol. Hepatol. 6, 1155–1161 (2008).

    Article  PubMed  Google Scholar 

  24. Cohen, S.J. et al. Fibroblast activation protein and its relationship to clinical outcome in pancreatic adenocarcinoma. Pancreas 37, 154–158 (2008).

    Article  CAS  PubMed  Google Scholar 

  25. Hwang, R.F. et al. Cancer-associated stromal fibroblasts promote pancreatic tumor progression. Cancer Res. 68, 918–926 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Vonlaufen, A. et al. Pancreatic stellate cells: partners in crime with pancreatic cancer cells. Cancer Res. 68, 2085–2093 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Herrera, M. et al. Functional heterogeneity of cancer-associated fibroblasts from human colon tumors shows specific prognostic gene expression signature. Clin. Cancer Res. 19, 5914–5926 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. 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).

    Article  CAS  PubMed  Google Scholar 

  29. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature 507, 315–322 (2014).

  30. Damrauer, J.S. et al. Intrinsic subtypes of high-grade bladder cancer reflect the hallmarks of breast cancer biology. Proc. Natl. Acad. Sci. USA 111, 3110–3115 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. McConkey, D.J., Choi, W. & Dinney, C.P. New insights into subtypes of invasive bladder cancer: considerations of the clinician. Eur. Urol. 67, e76–e78 (2015).

    Article  PubMed  Google Scholar 

  32. Parker, J.S. et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J. Clin. Oncol. 27, 1160–1167 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Prat, A. et al. Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res. 12, R68 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Tibshirani, R., Hastie, T., Narasimhan, B. & Chu, G. Diagnosis of multiple cancer types by shrunken centroids of gene expression. Proc. Natl. Acad. Sci. USA 99, 6567–6572 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Isella, C. et al. Stromal contribution to the colorectal cancer transcriptome. Nat. Genet. 47, 312–319 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Rubio-Viqueira, B. et al. An in vivo platform for translational drug development in pancreatic cancer. Clin. Cancer Res. 12, 4652–4661 (2006).

    Article  CAS  PubMed  Google Scholar 

  37. Stolze, B., Reinhart, S., Bulllinger, L., Fröhling, S. & Scholl, C. Comparative analysis of KRAS codon 12, 13, 18, 61, and 117 mutations using human MCF10A isogenic cell lines. Sci. Rep. 5, 8535 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ihle, N.T. et al. Effect of KRAS oncogene substitutions on protein behavior: implications for signaling and clinical outcome. J. Natl. Cancer Inst. 104, 228–239 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Carey, L., Winer, E., Viale, G., Cameron, D. & Gianni, L. Triple-negative breast cancer: disease entity or title of convenience? Nat. Rev. Clin. Oncol. 7, 683–692 (2010).

    Article  PubMed  Google Scholar 

  40. Bardeesy, N. et al. Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes Dev. 20, 3130–3146 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Haeger, S.M. et al. Smad4 loss promotes lung cancer formation but increases sensitivity to DNA topoisomerase inhibitors. Oncogene 10.1038/onc.2015.112 (20 April 2015).

  42. Garrido-Laguna, I. et al. Tumor engraftment in nude mice and enrichment in stroma- related gene pathways predict poor survival and resistance to gemcitabine in patients with pancreatic cancer. Clin. Cancer Res. 17, 5793–5800 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Dal Molin, M. et al. Very long-term survival following resection for pancreatic cancer is not explained by commonly mutated genes: results of whole-exome sequencing analysis. Clin. Cancer Res. 21, 1944–1950 (2015).

    Article  CAS  PubMed  Google Scholar 

  44. Zhang, Y. et al. A Gata6-Wnt pathway required for epithelial stem cell development and airway regeneration. Nat. Genet. 40, 862–870 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zhong, Y. et al. GATA6 activates Wnt signaling in pancreatic cancer by negatively regulating the Wnt antagonist Dickkopf-1. PLoS ONE 6, e22129 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wamunyokoli, F.W. et al. Expression profiling of mucinous tumors of the ovary identifies genes of clinicopathologic importance. Clin. Cancer Res. 12, 690–700 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Ji, H. et al. LKB1 modulates lung cancer differentiation and metastasis. Nature 448, 807–810 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Croft, D. et al. The Reactome pathway knowledgebase. Nucleic Acids Res. 42, D472–D477 (2014).

    Article  CAS  PubMed  Google Scholar 

  49. Ashburner, M. et al. Gene Ontology: tool for the unification of biology. Nat. Genet. 25, 25–29 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Bild, A.H. et al. Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature 439, 353–357 (2006).

    Article  CAS  PubMed  Google Scholar 

  51. Nishimura, D. BioCarta. Biotech. Software Internet Rep. 2, 117–120 (2001).

    Article  Google Scholar 

  52. Shi, L. et al. The MicroArray Quality Control (MAQC)-II study of common practices for the development and validation of microarray-based predictive models. Nat. Biotechnol. 28, 827–838 (2010).

    Article  CAS  PubMed  Google Scholar 

  53. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. 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 

  56. Neel, N.F. et al. Response to MLN8237 in pancreatic cancer is not dependent on RalA phosphorylation. Mol. Cancer Ther. 13, 122–133 (2014).

    Article  CAS  PubMed  Google Scholar 

  57. Bachem, M.G. et al. Pancreatic carcinoma cells induce fibrosis by stimulating proliferation and matrix synthesis of stellate cells. Gastroenterology 128, 907–921 (2005).

    Article  CAS  PubMed  Google Scholar 

  58. Conway, T. et al. Xenome—a tool for classifying reads from xenograft samples. Bioinformatics 28, i172–i178 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc. 7, 562–578 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 490, 61–70 (2012).

Download references

Acknowledgements

We thank the University of North Carolina (UNC) Tissue Procurement, Translational Pathology Laboratory, Animal Studies, Animal Histopathology, and Center for Gastrointestinal Biology and Disease Histopathology core facilities (NIH grant P30-DK034897), the PDX Program and Department of Pharmacology for tremendous technical support. We thank the patients and their families who generously donated their samples to research and in particular to the University of Nebraska Medical Center Rapid Autopsy Pancreatic Program and the Johns Hopkins Gastrointestinal Cancer Rapid Medical Donation Program. This work was partially supported by grant R01-CA140424 from the US National Institutes of Health (NIH) (J.J.Y.), the Kimmel Foundation (J.J.Y.), the American College of Surgeons (J.J.Y.), the University Cancer Research Fund (J.J.Y.) and UNC Lineberger Comprehensive Cancer Center Postdoctoral Training Grant T32-CA009156 from the US NIH (R.A.M.).

Author information

Authors and Affiliations

Authors

Contributions

Designed the study and wrote the manuscript: R.A.M. and J.J.Y. Performed experiments and collected data: R.A.M., R.M., E.L.F., S.G.H.L., L.A.W., S.C.E., A.H.C. and J.K.S. Pathologist: K.E.V. Performed data analysis: R.A.M., R.M., J.J.Y., K.A.H. and N.U.R. Provided samples: J.M.A., H.J.K., D.J.B., M.S.T., C.A.I.-D., M.A.H. and J.J.Y. Performed project oversight: J.J.Y.

Corresponding author

Correspondence to Jen Jen Yeh.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Representative H&E staining of patient tumor samples.

(a) Liver metastases showing regions of tumor and normal tissue. (b) Primary pancreatic tumor sample showing normal pancreatic tissue and tumor cells in the same field. (c) Primary pancreatic tumor with high tumor cellularity. (d) Primary pancreatic tumor with abundant tumor stroma. (e) Percentage of tumor in primary pancreatic tumors in the UNC and ICGC cohorts. Black arrowheads show areas of tumor stroma. Black arrows show areas of tumor. White arrowheads show normal tissue. Scale bars, 200 μm.

Supplementary Figure 2 Deconvolution of a large cohort of PDAC reveals distinct gene expression patterns from multiple tissue types.

Solid color bars above the heat map show the tissue of origin and tumor status of the samples, which were used to order the samples horizontally. Factor weights derived by NMF for selected factors are shown as grayscale bars. Heat maps show the z-normalized gene expression of exemplar genes from each factor. All tumors, cell lines and adjacent normal tissues from our cohort are shown.

Supplementary Figure 3 Correlation of pathology assessments of tumor with factor weights in normal pancreas and primary tumors.

Horizontal axes all show tumor cellularity, while vertical axes show factor weight. Red dashed lines show best linear fits. P values are given for each R2.

Supplementary Figure 4 Primary tumors have higher expression of stroma genes than metastatic samples.

(a) Normalized average gene expression of the 25 exemplar activated stroma genes is higher in primary tumor samples than in metastases (P < 0.001). (b) The average expression of the 25 exemplar normal stroma genes is slightly higher in primary tumors than in metastases (P = 0.031).

Supplementary Figure 5 Immunofluorescence staining of cancer-associated fibroblasts (CAFs).

(a) EpCAM, (b) vimentin and (c) SMAα. Staining of T3M4 pancreatic cancer cells as a (d) positive control for EpCAM, (e) negative control for vimentin and (f) negative control for SMAα. Scale bars, 50 μm.

Supplementary Figure 6 Hierarchical clustering of the Spearman correlation of samples from the UNC, TCGA bladder cancer and Perou data sets shows similarities among basal-like samples.

The color bars above the heat map show subtype, either from the original publication (known tumor subtype) or from our cross-platform classifier (pan-platform classification).

Supplementary Figure 7 Comparison to the subtypes from Collisson et al.

(a) Consensus-clustered heat map of normalized data from UNC and Collisson et al. using the gene sets from Collisson et al. Primary tumors, normal pancreas and cell lines are shown. Samples from Collisson et al. were previously classified as exocrine-like (magenta), classical (cyan) and quasimesenchymal (yellow). (b) Kaplan-Meier plots of UNC samples classified by PAM into the subtypes from Collisson et al. (c) Mouse- and human-specific gene expression of the gene lists from Collisson et al. in PDXs shown in log2 (1 + RPKM). Genes from the classical subtype are expressed by tumor cells, genes from the quasimesenchymal subtype are expressed by a mixture of human and mouse cells, and genes from the exocrine-like subtype are weakly expressed throughout.

Supplementary Figure 8 Collagen I staining of mouse stroma in PDX tumors.

(a) Staining for mouse collagen I of stroma in a representative PDX tumor. (b) Corresponding H&E staining of an adjacent section. Staining for mouse collagen I of (c) mouse skin and (d) human skin. Black arrowheads show areas of tumor stroma. Black arrows show areas of tumor. Scale bars, 200 μm.

Supplementary Figure 9 Tumor gene expression in PDX models.

(a) Mouse- and human-specific gene expression of gene lists from the basal-like and classical subtypes in 37 PDX tumors shown in log2 (1 + RPKM). Both gene sets are robustly expressed by the human (tumor) but not the mouse (stroma) cells in PDX samples. (b) Consensus clustering of these PDX tumors using the gene lists from the basal-like and classical subtypes divides the samples into two groups.

Supplementary Figure 10 SMAD4 staining of representative patient and matched PDX tumors.

Positive SMAD4 staining of a (a) patient adenocarcinoma and (b) corresponding PDX at passage 4. SMAD4 loss in a (c) patient adenocarcinoma and (d) corresponding PDX at passage 2. SMAD4 staining of control tissues: (e) human skin, (f) mouse skin and (g) human normal pancreas. Scale bars, 200 μm.

Supplementary Figure 11 Consensus-clustered heat map of ICGC data for which genetic information was available.

The color bars above the heat map show the subtypes and genetic alterations for key genes in PDAC. The heat maps show the z-normalized gene expression of genes from the basal-like and classical tumor subtypes.

Supplementary Figure 12 Gene signature scores by subtype.

Scores are normalized across the cohort and are calculated as the mean expression across a panel of genes obtained from MSigDB. (a) The basal-like subtype shows downregulation of GATA6. (b) Tumors from the classical subtype are enriched in genes associated with mucinous ovarian cancer. (c) Tumors from the basal-like subtype are enriched in genes related to KRAS activation and STK11 loss.

Supplementary Figure 13 Differences in extracellular mucin in tumors from the classical and basal-like subtypes.

(a) Number of samples with low (<10%) compared to high (10%) extracellular mucin content. (b) Representative H&E staining of samples with a low degree and (c) high degree of extracellular mucin content. Scale bars, 200 μm.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–13, Supplementary Table 3 and Supplementary Note. (PDF 5214 kb)

Supplementary Data Set

Gene expression of samples derived from RNA-seq data and KRAS mutation status. Expression is given as FPKM after processing with Xenome to sort human- and mouse-specific expression. (TXT 18938 kb)

Supplementary Table 1

List of the gene sets enriched in each factor. (XLSX 449 kb)

Supplementary Table 2

List of gene weights for each factor. (XLSX 2551 kb)

Supplementary Table 4

Overlap of the samples with those from Stratford et al. (XLSX 13 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Moffitt, R., Marayati, R., Flate, E. et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet 47, 1168–1178 (2015). https://doi.org/10.1038/ng.3398

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3398

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