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:

Spatial mapping of transcriptomic plasticity in metastatic pancreatic cancer

Abstract

Patients with treatment-refractory pancreatic cancer often succumb to systemic metastases1,2,3; however, the transcriptomic heterogeneity that underlies therapeutic recalcitrance remains understudied, particularly in a spatial context. Here we construct high-resolution maps of lineage states, clonal architecture and the tumour microenvironment (TME) using spatially resolved transcriptomics from 55 samples of primary tumour and metastases (liver, lung and peritoneum) collected from rapid autopsies of 13 people. We observe discernible transcriptomic shifts in cancer-cell lineage states as tumours transition from primary sites to organ-specific metastases, with the most pronounced intra-patient distinctions between liver and lung. Phylogenetic trees constructed from inferred copy number variations in primary and metastatic loci in each patient highlight diverse patient-specific evolutionary trajectories and clonal dissemination. We show that multiple tumour lineage states co-exist in each tissue, including concurrent metastatic foci in the same organ. Agnostic to tissue site, lineage states correlate with distinct TME features, such as the spatial proximity of TGFB1-expressing myofibroblastic cancer-associated fibroblasts (myCAFs) to aggressive ‘basal-like’ cancer cells, but not to cells in the ‘classical’ or ‘intermediate’ states. These findings were validated through orthogonal and cross-species analyses using mouse tissues and patient-derived organoids. Notably, basal-like cancer cells aligned with myCAFs correlate with plasma-cell exclusion from the tumour milieu, and neighbouring cell analyses suggest that CXCR4–CXCL12 signalling is the underlying basis for observed immune exclusion. Collectively, our findings underscore the profound transcriptomic heterogeneity and microenvironmental dynamics that characterize treatment-refractory pancreatic cancer.

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: Summary of data cohort and characterization of transcriptomic heterogeneity of PDAC malignant spots from primary tumours to metastatic sites.
Fig. 2: Phylogenetic clone trees reveal diverse evolutionary patterns in PDAC.
Fig. 3: Detection of lineage-transition events from primary to metastatic sites, indicating lineage plasticity of PDAC.
Fig. 4: Comparative analysis of different tumour lineage states and the TME using super-resolution gene expression.
Fig. 5: CosMx data reveal enhanced co-localization between fibroblasts and tumour cells, inhibiting the infiltration of plasma cells.

Similar content being viewed by others

Data availability

The raw and processed spatial transcriptome data and CosMx data generated in this study have been deposited at the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus (GEO) with accession numbers GSE274557 and GSE277782. The combined datasets can be accessed at GEO with accession number GSE277783.

Code availability

All R and Python scripts supporting the findings of this study are available in the GitHub repository at https://github.com/Coolgenome/PDAC.

References

  1. Halbrook, C. J., Lyssiotis, C. A., Pasca di Magliano, M. & Maitra, A. Pancreatic cancer: advances and challenges. Cell 186, 1729–1754 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Hidalgo, M. et al. Addressing the challenges of pancreatic cancer: future directions for improving outcomes. Pancreatology 15, 8–18 (2015).

    Article  PubMed  Google Scholar 

  3. Park, W., Chawla, A. & O’Reilly, E. M. Pancreatic cancer: a review. JAMA 326, 851–862 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Groot, V. P. et al. Implications of the pattern of disease recurrence on survival following pancreatectomy for pancreatic ductal adenocarcinoma. Ann. Surg. Oncol. 25, 2475–2483 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Liu, K. H. et al. Lung metastases in patients with stage IV pancreatic cancer: prevalence, risk factors, and survival impact. J. Clin. Med. 8, 1402 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Sahin, I. H., Elias, H., Chou, J. F., Capanu, M. & O’Reilly, E. M. Pancreatic adenocarcinoma: insights into patterns of recurrence and disease behavior. BMC Cancer 18, 769 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  8. 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).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. 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).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Reiter, J. G. et al. Minimal functional driver gene heterogeneity among untreated metastases. Science 361, 1033–1037 (2018).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  11. Notta, F. et al. A renewed model of pancreatic cancer evolution based on genomic rearrangement patterns. Nature 538, 378–382 (2016).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  12. 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  PubMed  PubMed Central  CAS  Google Scholar 

  13. Kim, M. S. et al. Heterogeneity of pancreatic cancer metastases in a single patient revealed by quantitative proteomics. Mol. Cell. Proteomics 13, 2803–2811 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Erickson, A. et al. Spatially resolved clonal copy number alterations in benign and malignant tissue. Nature 608, 360–367 (2022).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  15. 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).

    Article  PubMed  Google Scholar 

  16. Raghavan, S. et al. Microenvironment drives cell state, plasticity, and drug response in pancreatic cancer. Cell 184, 6119–6137 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Wormann, S. M. et al. APOBEC3A drives deaminase domain-independent chromosomal instability to promote pancreatic cancer metastasis. Nat. Cancer 2, 1338–1356 (2021).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  ADS  PubMed  CAS  Google Scholar 

  20. Gavish, A. et al. Hallmarks of transcriptional intratumour heterogeneity across a thousand tumours. Nature 618, 598–606 (2023).

    Article  ADS  PubMed  CAS  Google Scholar 

  21. Gabitova-Cornell, L. et al. Cholesterol pathway inhibition induces TGF-β signaling to promote basal differentiation in pancreatic cancer. Cancer Cell 38, 567–583 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Zhang, D. et al. Inferring super-resolution tissue architecture by integrating spatial transcriptomics with histology. Nat. Biotechnol. 42, 1372–1377 (2024).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Elyada, E. et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 9, 1102–1123 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Sahai, E. et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer 20, 174–186 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Dilly, J. et al. Mechanisms of resistance to oncogenic KRAS inhibition in pancreatic cancer. Cancer Discov. 14, 2135–2161 (2024).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Zhang, Y. et al. Macropinocytosis in cancer-associated fibroblasts is dependent on CaMKK2/ARHGEF2 signaling and functions to support tumor and stromal cell fitness. Cancer Discov. 11, 1808–1825 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Ma, R. Y., Black, A. & Qian, B. Z. Macrophage diversity in cancer revisited in the era of single-cell omics. Trends Immunol. 43, 546–563 (2022).

    Article  PubMed  CAS  Google Scholar 

  28. Blando, J. et al. Comparison of immune infiltrates in melanoma and pancreatic cancer highlights VISTA as a potential target in pancreatic cancer. Proc. Natl Acad. Sci. USA 116, 1692–1697 (2019).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  29. Carstens, J. L. et al. Spatial computation of intratumoral T cells correlates with survival of patients with pancreatic cancer. Nat. Commun. 8, 15095 (2017).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  30. Falcomata, C. et al. Context-specific determinants of the immunosuppressive tumor microenvironment in pancreatic cancer. Cancer Discov. 13, 278–297 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Biffi, G. et al. IL1-induced JAK/STAT signaling is antagonized by TGFβ to shape CAF heterogeneity in pancreatic ductal adenocarcinoma. Cancer Discov. 9, 282–301 (2019).

    Article  PubMed  Google Scholar 

  32. Feig, C. et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl Acad. Sci. USA 110, 20212–20217 (2013).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  33. Jiang, H. et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat. Med. 22, 851–860 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. He, S. et al. High-plex imaging of RNA and proteins at subcellular resolution in fixed tissue by spatial molecular imaging. Nat. Biotechnol. 40, 1794–1806 (2022).

    Article  PubMed  CAS  Google Scholar 

  35. 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 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. 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).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  Google Scholar 

  39. Lee, J. J. et al. Elucidation of tumor-stromal heterogeneity and the ligand-receptor interactome by single-cell transcriptomics in real-world pancreatic cancer biopsies. Clin. Cancer Res. 27, 5912–5921 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Topham, J. T. et al. Subtype-discordant pancreatic ductal adenocarcinoma tumors show intermediate clinical and molecular characteristics. Clin. Cancer Res. 27, 150–157 (2021).

    Article  PubMed  CAS  Google Scholar 

  41. Williams, H. L. et al. Spatially resolved single-cell assessment of pancreatic cancer expression subtypes reveals co-expressor phenotypes and extensive intratumoral heterogeneity. Cancer Res. 83, 441–455 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Wang, Z. et al. Carcinomas assemble a filamentous CXCL12-keratin-19 coating that suppresses T cell-mediated immune attack. Proc. Natl Acad. Sci. USA 119, e2119463119 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Yan, R., Moresco, P., Gegenhuber, B. & Fearon, D. T. T cell-mediated development of stromal fibroblasts with an immune-enhancing chemokine profile. Cancer Immunol. Res. 11, 1044–1054 (2023).

    Article  CAS  Google Scholar 

  44. Meylan, M. et al. Tertiary lymphoid structures generate and propagate anti-tumor antibody-producing plasma cells in renal cell cancer. Immunity 55, 527–541 (2022).

    Article  PubMed  CAS  Google Scholar 

  45. Law, H. C. et al. The proteomic landscape of pancreatic ductal adenocarcinoma liver metastases identifies molecular subtypes and associations with clinical response. Clin. Cancer Res. 26, 1065–1076 (2020).

    Article  PubMed  CAS  Google Scholar 

  46. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. McInnes, L., Healy, J., Saul, N. & Grossberger, L. UMAP: uniform manifold approximation and projection. J. Open Source Softw. 3, 861 (2018).

    Article  Google Scholar 

  48. Chu, Y. et al. Pan-cancer T cell atlas links a cellular stress response state to immunotherapy resistance. Nat. Med. 29, 1550–1562 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Wang, R. et al. Evolution of immune and stromal cell states and ecotypes during gastric adenocarcinoma progression. Cancer Cell 41, 1407–1426 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Wang, R. et al. Single-cell dissection of intratumoral heterogeneity and lineage diversity in metastatic gastric adenocarcinoma. Nat. Med. 27, 141–151 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Sans, M. et al. Spatial transcriptomics of intraductal papillary mucinous neoplasms of the pancreas identifies NKX6-2 as a driver of gastric differentiation and indolent biological potential. Cancer Discov. 13, 1844–1861 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Yu, G., Smith, D. K., Zhu, H., Guan, Y. & Lam, T. T. Y. ggtree: an R package for visualization and annotation of phylogenetic trees with their covariates and other associated data. Methods Ecol. Evol. 8, 28–36 (2017).

    Article  Google Scholar 

  53. Gao, R. et al. Delineating copy number and clonal substructure in human tumors from single-cell transcriptomes. Nat. Biotechnol. 39, 599–608 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. De Falco, A., Caruso, F., Su, X.-D., Iavarone, A. & Ceccarelli, M. A variational algorithm to detect the clonal copy number substructure of tumors from scRNA-seq data. Nat. Commun. 14, 1074 (2023).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  55. Schliep, K. phangorn: phylogenetic analysis in R. Bioinformatics 27, 592–593 (2011).

    Article  PubMed  CAS  Google Scholar 

  56. Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Hao, D. et al. The single-cell immunogenomic landscape of B and plasma cells in early-stage lung adenocarcinoma. Cancer Discov. 12, 2626–2645 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Liao, Y., Wang, J., Jaehnig, E. J., Shi, Z. & Zhang, B. WebGestalt 2019: gene set analysis toolkit with revamped UIs and APIs. Nucleic Acids Res. 47, W199–W205 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. B 57, 289–300 (1995).

    Article  MathSciNet  Google Scholar 

  60. Ohlund, D. et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med. 214, 579–596 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Cable, D. M. et al. Robust decomposition of cell type mixtures in spatial transcriptomics. Nat. Biotechnol. 40, 517–526 (2022).

    Article  PubMed  CAS  Google Scholar 

  62. Wei, R. et al. Spatial charting of single-cell transcriptomes in tissues. Nat. Biotechnol. 40, 1190–1199 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Vahid, M. R. et al. High-resolution alignment of single-cell and spatial transcriptomes with CytoSPACE. Nat. Biotechnol. 41, 1543–1548 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Pei, G. et al. deCS: a tool for systematic cell type annotations of single-cell RNA sequencing data among human tissues. Genomics Proteomics Bioinformatics 21, 370–384 (2023).

    Article  PubMed  Google Scholar 

  65. Belle, J. I. & DeNardo, D. G. A single-cell window into pancreas cancer fibroblast heterogeneity. Cancer Discov. 9, 1001–1002 (2019).

    Article  PubMed  CAS  Google Scholar 

  66. Boyd, L. N. C., Andini, K. D., Peters, G. J., Kazemier, G. & Giovannetti, E. Heterogeneity and plasticity of cancer-associated fibroblasts in the pancreatic tumor microenvironment. Semin. Cancer Biol. 82, 184–196 (2022).

    Article  PubMed  CAS  Google Scholar 

  67. Hanzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

A.M. was supported by the MD Anderson Pancreatic Cancer Moon Shot Program; the Sheikh Khalifa Bin Zayed Al-Nahyan Foundation; Break Through Cancer; and NIH grants (U54CA274371, U01CA200468 and U24CA274274). L.W. was supported by NIH–NCI grants (R01CA266280, U24CA274274, U01CA294518 and U01CA264583); research funding provided by the James P. Allison Institute; the Institute for Data Science in Oncology; the University of Texas MD Anderson Cancer Center; and Break Through Cancer. L.W. is an Associate Member of the James P. Allison Institute and an Andrew Sabin Family Foundation Fellow at the MD Anderson Cancer Center. G.P. acknowledges support from NIH–NCI grant U01CA294518 and the Program for T Cell-based Therapy at the MD Anderson Cancer Center. A.S. was supported by the the ACCENT (B-487.0012) and BONFOR program (O-112.0070). A.M.L. was supported by a NIH grant (U01CA274295). A.J.A. was funded by the Hale Center for Pancreatic Cancer Research; Break Through Cancer; the Lustgarten Foundation; the Pancreatic Cancer Action Network; NIH–NCI grants (P50CA127003, U01CA274276 and R01CA276268); and the Dana-Farber Cancer Institute Hale Center for Pancreatic Cancer Research. P.M.G. and M.A.H. were supported by the Pancreatic Cancer Detection Consortium (U01CA210240); a NCI Cancer Center Support grant (P30CA36727); and a NCI Research Specialist award (R50CA211462). The cyclic IF staining was performed in the Flow Cytometry and Cellular Imaging Core Facility, which is supported in part by the NIH through MD Anderson’s Cancer Center Support grant (P30CA016672), the NCI’s Research Specialist 1 (R50CA243707-01A1) and a Shared Instrumentation award from the Cancer Prevention Research Institution of Texas (CPRIT). We thank A. S. Multani for the FISH experiments and S. P. So, E. E. Rodriguez, A. T. Reckard, Y. A. Zuberi, A. V. Basi and J. A. Gomez for technical assistance.

Author information

Authors and Affiliations

Authors

Contributions

L.W. and A.M. conceived and jointly supervised the study. V. Branchi, J.L.G., T.C.C., P.M.G. and M.A.H. contributed to sample and patient information collection. V. Branchi and B.C.S. processed tissues and prepared libraries for SRT. A.M., V. Branchi and D.S. contributed to pathology review. L.W. supervised the processing, analysis and interpretation of bioinformatics data. D.Z. and M.L. contributed to tool development. Y.L., K.S.C., Y.C., E.D., V. Bernard, A.S. and G.H. assisted with data analysis. V. Branchi, G.P. and K.I.R. contributed to the processing of sequencing data and integrative analyses. F.T., K.T., B.G., H.T., A.M.L., and A.J.A. provided mouse tissues, human organoid lines, human CAF lines and relevant resources. J.K.B. provided support and resources for cyclic IF. J.M. performed in vitro experiments and data analysis. G.P. and J.M. analysed data and generated figures and tables for the manuscript. G.P., V. Branchi, J.M., K.I.R., C.Y., P.A.G., L.W. and A.M. contributed to data interpretation. J.M., G.P., L.W. and A.M. wrote and revised the manuscript, and all co-authors reviewed the manuscript.

Corresponding authors

Correspondence to Linghua Wang or Anirban Maitra.

Ethics declarations

Competing interests

A.M. is listed as an inventor on a patent that has been licensed by Johns Hopkins University to Thrive Earlier Detection and serves as a consultant for Tezcat Biosciences. A.J.A. has consulted for Anji Pharmaceuticals, Affini-T Therapeutics, Arrakis Therapeutics, AstraZeneca, Boehringer Ingelheim, Kestrel Therapeutics, Merck, Mirati Therapeutics, Nimbus Therapeutics, Oncorus, Plexium, Quanta Therapeutics, Revolution Medicines, Reactive Biosciences, Riva Therapeutics, Servier Pharmaceuticals, Syros Pharmaceuticals, T-knife Therapeutics, Third Rock Ventures and Ventus Therapeutics; holds equity in Riva Therapeutics and Kestrel Therapeutics; and has research funding from Boehringer Ingelheim, Bristol Myers Squibb, Deerfield, Eli Lilly, Mirati Therapeutics, Novartis, Novo Ventures, Revolution Medicines and Syros Pharmaceuticals. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature thanks Jen Jen Yeh, Nancy Zhang and the other, anonymous, reviewers for their contribution to the peer review of this work.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 UMAP plots of 134,776 SRT spots from 13 patients.

a,b, Each dot represents a single spot coloured by (a) patient and (b) treatment history. c,d, Venn diagrams showing the overlap of significantly upregulated genes in the treated group (c) or untreated group (d) across four different tissue sites.

Extended Data Fig. 2 Workflow for the construction of phylogenetic clone trees based on SRT data.

The workflow comprises four key steps: (1) annotation of tumour spots, (2) inference of CNVs at spot level, (3) construction of phylogenetic tree, and (4) spatial mapping of inferred subclones.

Extended Data Fig. 3 Phylogenetic clone trees reveal diverse evolutionary patterns in PDAC.

a, Pt-3. b, Pt-6. c, Pt-12.

Extended Data Fig. 4 FISH validation of MYC (8q24.2).

a,e, Spatial visualization of subclones from Fig. 2, along with fluorescence images for MYC (red) and control (Ctrl, green) probes targeting the centromere of chromosome 8 in Pt-10 (a) and Pt-1 (e). Dotted boxes and numbers indicate the enlarged areas. b,f, MYC CNV scores from the inferCNV analysis are shown in b (Pt-10) and f (Pt-1). Each dot represents each subclone. Data are presented as mean ± standard deviation (SD). For Pt-10 (b), clone A (n = 2), clone B (n = 3), clone C (n = 5) and clone D (n = 1). For Pt-1 (f), clone A (n = 2), clone B (n = 3) and clone C (n = 5). c,g, Per-cell copy number (CN) ratio for MYC in Pt-10 (c) and Pt-1 (g). d,h, Percentages of cells with MYC amplification (CN ratio over 1) in Pt-10 (d) and Pt-1 (h).

Extended Data Fig. 5 Characterization of lineage-state heterogeneity and plasticity.

a, UMAP plot displaying 67,990 “neoplastic” spots cross lineage states (left), patients (middle) or tissue origin sites (right). b, Bar plots displaying the relative fraction of tumour spots in classical, intermediate, and basal lineage states in each ST sample. The samples were ordered by tissue sites and the fraction of basal lineage. The top pie charts represent the global lineage composition in all Pri, LiM, PerM and LuM sites. c, Co-immunostaining of PanCK (green), S100A2 (red), and GATA6 (blue) in matched liver and lung metastases of three KPCY mice. Dotted boxes indicate the enlarged areas. d, Mean intensity of S100A2 and GATA6 across individual cells in tumour ROIs of liver and lung metastases. Each dot with connected lines represents data from the same mouse (n = 3). Student’s paired t-test. P values are indicated above the plot.

Extended Data Fig. 6 Subtyping of mesenchymal and squamous components in the basal-lineage-enriched tumour spots.

a, Pearson correlation analysis among classical, basal lineage, mesenchymal, squamous, and 8 core basal gene signature scores across all tumour spots. b, Bar plots comparing the relative fraction of tumour spots with mesenchymal and squamous lineages in each ST sample. c, Overview of 41 MPs among classical, intermediate, mesenchymal, and squamous lineage tumour enriched spots.

Extended Data Fig. 7 Comparison of tumour lineage compositions and tumour locations.

a, Schematic diagram illustrating the redefinition of tumour regions based on their distance from non-tumour areas, categorizing them into tumour edge, intermediate, and core regions. b,c, Total spot number (left) and relative composition (right) of tumour spots from the three main different lineages (b), with further subtyping of the basal lineage into mesenchymal and squamous lineages (c), among tumour-edge, tumour-intermediate, and tumour-core regions. d, Overview of 41 MPs based on tumour regions.

Extended Data Fig. 8 Spatial proximity between basal-like tumour cells and myCAFs.

a, Co-immunostaining of PanCK (green), S100A2 (red), and DAPI (blue) in matched liver and lung metastases of three different KPCY mice. White lines denote the tumour bed, and yellow lines denote the juxtalesional areas. b, Mean intensity of α-SMA across individual cells in juxtalesional ROIs between liver (LiM) and lung (LuM) metastases. For mouse 1, n = 86,763 cells (LiM) and n = 4,125 cells (LuM). For mouse 2, n = 21,310 cells (LiM) and n = 5,101 cells (LuM). For mouse 3, n = 194,729 cells (LiM) and n = 11,861 cells (LuM). Each dot with connected lines represents data from the same mouse. Student’s paired t-test. ****P < 0.0001. c, A representative image of E-cadherin (white), α-SMA (green), S100A2 (red), and GATA6 (blue) in a liver metastasis containing both classical-like and basal-like tumour cells. White arrows with numbers denote the enlarged areas. d, Phase contrast, H&E staining, and co-immunostaining for S100A2 (red), α-SMA (green), and DAPI (blue) in classical-like and basal-like PDOs cultured with human CAFs. Full PDO IDs are as follows: 185; PANFR0185_T2, 332; PANFR0332_T1, 172; PANFR0172_T4, and 440; PANFR0440_T1. e, Quantification of the percentage of organoids displaying direct attachments of α-SMA+ CAFs in each PDO line. Each dot represents an individual CAF line. Data are presented as mean ± SD (n = 3, independent experiments). Student’s unpaired t-test. *P < 0.05 (P = 0.014 in 185 vs. 172, P = 0.01 in 185 vs. 440), **P < 0.01 (P = 0.006 in 332 vs. 172, P = 0.005 in 332 vs. 440).

Extended Data Fig. 9 Absence of T cell and B cell infiltration in PDAC SRT samples.

From top to bottom, the images show tumour lineage distribution, iStar-derived T cell signature, iStar-derived B cell signature, and expression levels of CD3D and MS4A1.

Supplementary information

Supplementary Figures

This file contains Supplementary Figures 1–12.

Reporting Summary

Supplementary Tables

This file contains Supplementary Tables 1–10.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pei, G., Min, J., Rajapakshe, K.I. et al. Spatial mapping of transcriptomic plasticity in metastatic pancreatic cancer. Nature 642, 212–221 (2025). https://doi.org/10.1038/s41586-025-08927-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41586-025-08927-x

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