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:

Spatially resolved analysis of pancreatic cancer identifies therapy-associated remodeling of the tumor microenvironment

Abstract

In combination with cell-intrinsic properties, interactions in the tumor microenvironment modulate therapeutic response. We leveraged single-cell spatial transcriptomics to dissect the remodeling of multicellular neighborhoods and cell–cell interactions in human pancreatic cancer associated with neoadjuvant chemotherapy and radiotherapy. We developed spatially constrained optimal transport interaction analysis (SCOTIA), an optimal transport model with a cost function that includes both spatial distance and ligand–receptor gene expression. Our results uncovered a marked change in ligand–receptor interactions between cancer-associated fibroblasts and malignant cells in response to treatment, which was supported by orthogonal datasets, including an ex vivo tumoroid coculture system. We identified enrichment in interleukin-6 family signaling that functionally confers resistance to chemotherapy. Overall, this study demonstrates that characterization of the tumor microenvironment using single-cell spatial transcriptomics allows for the identification of molecular interactions that may play a role in the emergence of therapeutic resistance and offers a spatially based analysis framework that can be broadly applied to other contexts.

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: SMI captures pancreatic tumor architecture at subcellular resolution.
Fig. 2: SMI uncovers cell-type diversity in pancreatic cancer.
Fig. 3: SMI reveals glandular heterogeneity and multicellular neighborhoods in pancreatic cancer.
Fig. 4: Deciphering cell–cell interactions using SCOTIA.
Fig. 5: Treatment-associated LR interactions between CAFs and malignant cells.
Fig. 6: Validation analysis of treatment-associated LR interactions between CAFs and malignant cells.
Fig. 7: Substantiation of LR interactions in mouse coculture tumoroids.
Fig. 8: IL-6 family signaling confers chemoresistance in human pancreatic cancer cell lines.

Similar content being viewed by others

Data availability

The raw and processed SMI and coculture snRNA-seq data are available via Mendeley Data at https://doi.org/10.17632/kx6b69n3cb.1 (ref. 81) and Zenodo https://doi.org/10.5281/zenodo.7963531 (ref. 82). The snRNA-seq and GeoMx datasets6 are available from GEO: GSE202051 and GSE199102, respectively. The known ligand–receptor databases are from FANTOM5 (https://fantom.gsc.riken.jp/5), CellChat (http://www.cellchat.org) and CellPhoneDB (https://www.cellphonedb.org). Downstream target gene information was from KEGG (https://www.genome.jp/kegg) and TRRUST (https://www.grnpedia.org/trrust). Source data are provided with this paper.

Code availability

The SCOTIA package and our analysis code have been uploaded to Zenodo at https://doi.org/10.5281/zenodo.12707341 (ref. 83) and GitHub (https://github.com/Caochris/SCOTIA).

References

  1. Siegel, R. L., Giaquinto, A. N. & Jemal, A. Cancer statistics, 2024. CA Cancer J. Clin. 74, 12–49 (2024).

    Article  PubMed  Google Scholar 

  2. Springfeld, C. et al. Neoadjuvant therapy for pancreatic cancer. Nat. Rev. Clin. Oncol. 20, 318–337 (2023).

    Article  PubMed  Google Scholar 

  3. Evan, T., Wang, V. M. Y. & Behrens, A. The roles of intratumour heterogeneity in the biology and treatment of pancreatic ductal adenocarcinoma. Oncogene 41, 4686–4695 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ho, W. J., Jaffee, E. M. & Zheng, L. The tumour microenvironment in pancreatic cancer – clinical challenges and opportunities. Nat. Rev. Clin. Oncol. 17, 527–540 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Hwang, W. L. et al. Single-nucleus and spatial transcriptome profiling of pancreatic cancer identifies multicellular dynamics associated with neoadjuvant treatment. Nat. Genet. 54, 1178–1191 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Grünwald, B. T. et al. Spatially confined sub-tumor microenvironments in pancreatic cancer. Cell 184, 5577–5592.e18 (2021).

    Article  PubMed  Google Scholar 

  8. Falcomatà, C. et al. Selective multi-kinase inhibition sensitizes mesenchymal pancreatic cancer to immune checkpoint blockade by remodeling the tumor microenvironment. Nat. Cancer 3, 318–336 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Tu, M. et al. TNF-α-producing macrophages determine subtype identity and prognosis via AP1 enhancer reprogramming in pancreatic cancer. Nat. Cancer 2, 1185–1203 (2021).

    Article  CAS  PubMed  Google Scholar 

  10. Guo, J. A. et al. Refining the molecular framework for pancreatic cancer with single-cell and spatial technologies. Clin. Cancer Res. 27, 3825–3833 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cui Zhou, D. et al. Spatially restricted drivers and transitional cell populations cooperate with the microenvironment in untreated and chemo-resistant pancreatic cancer. Nat. Genet. 54, 1390–1405 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Bärthel, S., Falcomatà, C., Rad, R., Theis, F. J. & Saur, D. Single-cell profiling to explore pancreatic cancer heterogeneity, plasticity and response to therapy. Nat. Cancer 4, 454–467 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Moncada, R. et al. Integrating microarray-based spatial transcriptomics and single-cell RNA-seq reveals tissue architecture in pancreatic ductal adenocarcinomas. Nat. Biotechnol. 38, 333–342 (2020).

    Article  CAS  PubMed  Google Scholar 

  15. Giladi, A. et al. Dissecting cellular crosstalk by sequencing physically interacting cells. Nat. Biotechnol. 38, 629–637 (2020).

    Article  CAS  PubMed  Google Scholar 

  16. Williams, C. G., Lee, H. J., Asatsuma, T., Vento-Tormo, R. & Haque, A. An introduction to spatial transcriptomics for biomedical research. Genome Med. 14, 68 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Jin, S. et al. Inference and analysis of cell–cell communication using CellChat. Nat. Commun. 12, 1088 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Browaeys, R., Saelens, W. & Saeys, Y. NicheNet: modeling intercellular communication by linking ligands to target genes. Nat. Methods 17, 159–162 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. 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  CAS  PubMed  Google Scholar 

  20. Allen, W. E., Blosser, T. R., Sullivan, Z. A., Dulac, C. & Zhuang, X. Molecular and spatial signatures of mouse brain aging at single-cell resolution. Cell 186, 194–208.e18 (2023).

    Article  CAS  PubMed  Google Scholar 

  21. Petukhov, V. et al. Cell segmentation in imaging-based spatial transcriptomics. Nat. Biotechnol. 40, 345–354 (2022).

    Article  CAS  PubMed  Google Scholar 

  22. Danaher, P. et al. Insitutype: likelihood-based cell typing for single cell spatial transcriptomics. Preprint at bioRxiv https://doi.org/10.1101/2022.10.19.512902 (2022).

  23. 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  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  25. 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  CAS  PubMed  PubMed Central  Google Scholar 

  26. Capucetti, A., Albano, F. & Bonecchi, R. Multiple roles for chemokines in neutrophil biology. Front. Immunol. 11, 1259 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ester, M., Kriegel, H., Sander, J. & Xu, X. A density-based algorithm for discovering clusters in large spatial databases with noise. In Proc. 2nd International Conference on Knowledge Discovery and Data Mining 226–231 (AAAI Press, 1996).

  28. Schubert, E., Sander, J., Ester, M., Kriegel, H. P. & Xu, X. DBSCAN revisited, revisited: why and how you should still use DBSCAN. ACM TODS 42, 1–21 (2017).

    Article  Google Scholar 

  29. Zhu, J., Shang, L. & Zhou, X. SRTsim: spatial pattern preserving simulations for spatially resolved transcriptomics. Genome Biol. 24, 39 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Hosein, A. N., Brekken, R. A. & Maitra, A. Pancreatic cancer stroma: an update on therapeutic targeting strategies. Nat. Rev. Gastroenterol. Hepatol. 17, 487–505 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Chen, Y., Yu, G., Yu, D. & Zhu, M. PKCalpha-induced drug resistance in pancreatic cancer cells is associated with transforming growth factor-beta1. J. Exp. Clin. Cancer Res. 29, 104 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Singh, S., Srivastava, S. K., Bhardwaj, A., Owen, L. B. & Singh, A. P. CXCL12–CXCR4 signalling axis confers gemcitabine resistance to pancreatic cancer cells: a novel target for therapy. Br. J. Cancer 103, 1671–1679 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ren, Y. et al. CXCR3 confers sorafenib resistance of HCC cells through regulating metabolic alteration and AMPK pathway. Am. J. Transl. Res 12, 825 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Li, J. et al. Overexpression of CXCR4 is significantly associated with cisplatin-based chemotherapy resistance and can be a prognostic factor in epithelial ovarian cancer. BMB Rep. 47, 33 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Zhu, S. et al. Expression profile-based screening for critical genes reveals S100A4, ACKR3 and CDH1 in docetaxel-resistant prostate cancer cells. Aging 11, 12754–12772 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Shi, Y. et al. Targeting LIF-mediated paracrine interaction for pancreatic cancer therapy and monitoring. Nature 569, 131–135 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Wu, X. et al. IL-6 secreted by cancer-associated fibroblasts promotes epithelial–mesenchymal transition and metastasis of gastric cancer via JAK2/STAT3 signaling pathway. Oncotarget 8, 20741–20750 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Ebbing, E. A. et al. Stromal-derived interleukin 6 drives epithelial-to-mesenchymal transition and therapy resistance in esophageal adenocarcinoma. Proc. Natl Acad. Sci. USA 116, 2237–2242 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Vicent, S. et al. Cross-species functional analysis of cancer-associated fibroblasts identifies a critical role for CLCF1 and IL-6 in non-small cell lung cancer in vivo. Cancer Res 72, 5744–5756 (2012).

    Article  CAS  PubMed  Google Scholar 

  41. Kim, J. W. et al. Antitumor activity of an engineered decoy receptor targeting CLCF1–CNTFR signaling in lung adenocarcinoma. Nat. Med. 25, 1783–1795 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Jiang, Y. et al. CLCF1 is a novel potential immune-related target with predictive value for prognosis and immunotherapy response in glioma. Front. Immunol. 13, 810832 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Murakami, M., Kamimura, D. & Hirano, T. Pleiotropy and specificity: insights from the interleukin 6 family of cytokines. Immunity 50, 812–831 (2019).

    Article  CAS  PubMed  Google Scholar 

  44. Lelièvre, E. et al. Signaling pathways recruited by the cardiotrophin-like cytokine/cytokine-like factor-1 composite cytokine: specific requirement of the membrane-bound form of ciliary neurotrophic factor receptor alpha component. J. Biol. Chem. 276, 22476–22484 (2001).

    Article  PubMed  Google Scholar 

  45. Jin, W. Role of JAK/STAT3 signaling in the regulation of metastasis, the transition of cancer stem cells, and chemoresistance of cancer by epithelial–mesenchymal transition. Cells 9, 217 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. 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 

  47. Kanehisa, M., Furumichi, M., Tanabe, M., Sato, Y. & Morishima, K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res 45, D353–D361 (2017).

    Article  CAS  PubMed  Google Scholar 

  48. Han, H. et al. TRRUST v2: an expanded reference database of human and mouse transcriptional regulatory interactions. Nucleic Acids Res 46, D380–D386 (2018).

    Article  CAS  PubMed  Google Scholar 

  49. Efremova, M., Vento-Tormo, M., Teichmann, S. A. & Vento-Tormo, R. CellPhoneDB: inferring cell–cell communication from combined expression of multi-subunit ligand–receptor complexes. Nat. Protoc. 15, 1484–1506 (2020).

    Article  CAS  PubMed  Google Scholar 

  50. Dimitrov, D. et al. Comparison of methods and resources for cell–cell communication inference from single-cell RNA-Seq data. Nat. Commun. 13, 3224 (2022).

    Article  PubMed  Google Scholar 

  51. Mao, X. et al. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Mol. Cancer 20, 131 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Joost, P. et al. Heterogenous mismatch-repair status in colorectal cancer. Diagn. Pathol. 9, 126 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Alers, J. C. et al. Cytogenetic heterogeneity and histologic tumor growth patterns in prostatic cancer. Cytometry 21, 84–94 (1995).

    Article  CAS  PubMed  Google Scholar 

  54. Cang, Z. & Nie, Q. Inferring spatial and signaling relationships between cells from single cell transcriptomic data. Nat. Commun. 11, 2084 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Liu, Z., Sun, D. & Wang, C. Evaluation of cell–cell interaction methods by integrating single-cell RNA sequencing data with spatial information. Genome Biol. 23, 218 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Cang, Z. et al. Screening cell–cell communication in spatial transcriptomics via collective optimal transport. Nat. Methods 20, 218–228 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Mantovani, A., Allavena, P., Marchesi, F. & Garlanda, C. Macrophages as tools and targets in cancer therapy. Nat. Rev. Drug Discov. 21, 799–820 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Boraschi-Diaz, I., Wang, J., Mort, J. S. & Komarova, S. V. Collagen type I as a ligand for receptor-mediated signaling. Front. Phys. 5, 12 (2017).

    Article  Google Scholar 

  59. Seino, T. et al. Human pancreatic tumor organoids reveal loss of stem cell niche factor dependence during disease progression. Cell Stem Cell 22, 454–467.e6 (2018).

    Article  CAS  PubMed  Google Scholar 

  60. Fuertes, G. et al. Noncanonical Wnt signaling promotes colon tumor growth, chemoresistance and tumor fibroblast activation. EMBO Rep. 24, e54895 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Martin-Orozco, E., Sanchez-Fernandez, A., Ortiz-Parra, I. & Ayala-San Nicolas, M. WNT signaling in tumors: the way to evade drugs and immunity. Front. Immunol. 10, 2854 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Spada, S., Tocci, A., Di Modugno, F. & Nisticò, P. Fibronectin as a multiregulatory molecule crucial in tumor matrisome: from structural and functional features to clinical practice in oncology. J. Exp. Clin. Cancer Res. 40, 102 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Sun, W. H. et al. CXCR3 isoform A promotes head and neck cancer progression by enhancing stem-like property and chemoresistance. J. Oral. Pathol. Med. 51, 791–800 (2022).

    Article  CAS  PubMed  Google Scholar 

  64. Lu, Y. et al. Spatial transcriptome profiling by MERFISH reveals fetal liver hematopoietic stem cell niche architecture. Cell Discov. 7, 47 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Moffitt, J. R. et al. Molecular, spatial, and functional single-cell profiling of the hypothalamic preoptic region. Science 362, eaau5324 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Marshall, J. L. et al. High-resolution Slide-seqV2 spatial transcriptomics enables discovery of disease-specific cell neighborhoods and pathways. iScience 25, 104097 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

  68. Guo, J. A. et al. Abstract 5775: GLIS3 drives a neural-like malignant state enriched after neoadjuvant treatment in pancreatic cancer. Cancer Res 83, 5775 (2023).

    Article  Google Scholar 

  69. Stringer, C., Wang, T., Michaelos, M. & Pachitariu, M. Cellpose: a generalist algorithm for cellular segmentation. Nat. Methods 18, 100–106 (2021).

    Article  CAS  PubMed  Google Scholar 

  70. Pachitariu, M. & Stringer, C. Cellpose 2.0: how to train your own model. Nat. Methods 19, 1634–1641 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Johnson, W. E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118–127 (2007).

    Article  PubMed  Google Scholar 

  72. Villani, C. Optimal Transport: Old and New Vol. 338 (Springer, 2009).

  73. Ramilowski, J. A. et al. A draft network of ligand–receptor-mediated multicellular signalling in human. Nat. Commun. 6, 7866 (2015).

    Article  CAS  PubMed  Google Scholar 

  74. Thul, P. J. et al. A subcellular map of the human proteome. Science 356, eaal3321 (2017).

    Article  PubMed  Google Scholar 

  75. Chen, Z. et al. Forest fire clustering for single-cell sequencing combines iterative label propagation with parallelized monte carlo simulations. Nat. Commun. 13, 3538 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Gates, A. J. & Ahn, Y.-Y. The impact of random models on clustering similarity. J. Mach. Learn. Res. 18, 3049–3076 (2017).

    Google Scholar 

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

    Article  Google Scholar 

  78. Cokelaer, T. et al. BioServices: a common Python package to access biological Web Services programmatically. Bioinformatics 29, 3241–3242 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Mi, H., Muruganujan, A., Ebert, D., Huang, X. & Thomas, P. D. PANTHER version 14: more genomes, a new PANTHER GO-slim and improvements in enrichment analysis tools. Nucleic Acids Res 47, D419–D426 (2019).

    Article  CAS  PubMed  Google Scholar 

  80. Freed-Pastor, W. A. et al. The CD155/TIGIT axis promotes and maintains immune evasion in neoantigen-expressing pancreatic cancer. Cancer Cell 39, 1342–1360.e14 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Shiau, C. et al. PDAC-SMI and co-culture dataset. Mendeley Data https://doi.org/10.17632/kx6b69n3cb.1 (2024).

  82. Shiau, C., et al. PDAC-SMI transcript locations. Zenodo https://doi.org/10.5281/zenodo.7963531 (2023).

  83. Shiau, C., Cao, J., Hemberg, M. & Hwang, W. L. Codes for PDAC-SMI and co-culture data. Zenodo https://doi.org/10.5281/zenodo.12707341 (2024).

Download references

Acknowledgements

We thank the patients and families who contributed their time and surgical specimens to this study. We thank K. Cormier for assistance with tissue sectioning; T. Oni (Whitehead Institute/Massachusetts Institute of Technology), D. Tuveson (Cold Spring Harbor Laboratory) for providing mouse pancreatic stellate cell lines; P. Danaher, A. Wardhani, M. Rhodes, S. Wiegel, S. He, E. Rueckert, E. Piazza, S. Stephenson, A. Grootsky and E. Miller from NanoString for assistance with project coordination and data analysis; and D. Moschella, T. Balducci, S. McSorley, S. Sullivan, M. Pivovarov, M. Mues, K. Yee, K. Mercer, J. Teixeira and K. Anderson for administrative and technical support. This work was supported in part by National Institutes of Health/National Cancer Institute (NIH/NCI) grant no. K08CA270417 (W.L.H.), Burroughs Wellcome Fund Career Award for Medical Scientists (W.L.H.), Pancreatic Cancer Action Network Career Development Award (W.L.H.), NIH/NCI grant no. 2P50CA127003 (W.L.H.), Krantz Family Center for Cancer Research Quantum Award (W.L.H., D.T.T.), SU2C-Lustgarten Foundation (T.J., T.S.H., D.T.T.), Robert L. Fine Cancer Research Foundation (D.T.T.), the Evergrande Center (J.C., M.H.), National Institute of Arthritis and Musculoskeletal and Skin Diseases grant no. 5UC2AR081023-02 (J-W.C., M.H.), a Bisconti award from Harvard Medical School (M.H.) and the Helmsley Foundation (J-W.C., M.H.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

C.S., J.C., M.H. and W.L.H. developed the study concept. T.K.K., Y.K., N.S. and J.M.B. performed spatial molecular imaging using a panel including custom probes designed by C.S. and W.L.H. C.S., J.C., M.T.G., J-W.C., P.L.W., J.W.R., M.H. and W.L.H. analyzed and interpreted the spatial molecular imaging data. J.C. and M.H. developed the SCOTIA method with input from C.S. and W.L.H. D.G., S.W., J.S., J.A.G., X.Y., P.L.W., N.A.L., J.W.B., R.Z. and W.L.H. designed, performed and analyzed the in vitro experiments, single-nucleus and bulk RNA-sequencing. M.M.-K., N.J.C., J.L.B. and M.L.G. performed histological analyses and guided sectioning and staining. M.Q., T.S.H., J.Y.W., H.R., C.F.-d.C. and M.M-K. provided clinical insights and access to patient specimens. C.S., J.C., D.G. and M.T.G. generated the tables and figures with guidance from M.H. and W.L.H. Funding for the work was provided by W.L.H., D.T.T., T.J., M.H. and R.W. The study was supervised by W.L.H. and M.H. The manuscript was written by J.C., C.S., D.G., P.L.W., M.H. and W.L.H. All authors reviewed the manuscript.

Corresponding authors

Correspondence to Martin Hemberg or William L. Hwang.

Ethics declarations

Competing interests

W.L.H. and C.S. have received conference travel reimbursements from NanoString Technologies related to presentation of some work in this study. M.T.G., J.W.R., T.K.K., Y.K., N.S. and J.M.B. were employees of NanoString Technologies at the time of their contributions to this study. D.T.T. has received an honorarium from NanoString Technologies, which had technology used in this manuscript. D.T.T. has received consulting fees from ROME Therapeutics and Tekla Capital not related to this work. D.T.T. has received honoraria from Moderna, Ikena Oncology, Foundation Medicine, Inc. and Pfizer that are not related to this work. D.T.T. is a founder and has equity in ROME Therapeutics, PanTher Therapeutics and TellBio, Inc., which is not related to this work. D.T.T. receives research support from ACD-Biotechne, PureTech Health LLC, Ribon Therapeutics, AVA LifeScience GmbH and Incyte, which was not used in this work. W.L.H., J.A.G. and T.J. (U.S. Provisional Application No. 63/069,035) and W.L.H., J.A.G., C.S., J.S. and T.J. (U.S. Provisional Application No. 63/346,670) are coinventors on provisional patents related to the pancreatic cancer states used in this study. The interests of W.L.H. and D.T.T. were reviewed and are managed by Mass General Brigham in accordance with their conflict of interest policies. T.J. is a member of the Board of Directors of Amgen and Thermo Fisher Scientific, and a co-Founder of Dragonfly Therapeutics and T2 Biosystems. T.J. serves on the Scientific Advisory Board of Dragonfly Therapeutics, SQZ Biotech and Skyhawk Therapeutics. T.J. is also President of Break Through Cancer. His laboratory currently receives funding from Johnson & Johnson, but these funds did not support the research described in this manuscript. All other authors declare no interests related to this work.

Peer review

Peer review information

Nature Genetics thanks the anonymous reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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 Spatial molecular imaging experimental workflow and probe set.

a, Schematic of the spatial molecular imaging RNA assay workflow. RNA targets in the FFPE tissue slide that are bound to in situ hybridization (ISH) probes are subject to cyclic readout of 16 sets of reporters conjugated to four different fluorophores, which bind to the different reporter-landing domains on the ISH probes. High-resolution images are acquired during each round of reporter hybridization. Fluorophores are then UV cleaved and washed off the reporters before the slide is incubated with the next set of reporters. b, Gene overlap between the seven malignant lineage programs6, the base 960-plex probe set and the 30 custom probes (color legend).

Extended Data Fig. 2 Comparison of cell segmentation and cell type annotation results from different segmentation methods.

a, Overlap of cell boundaries identified by Cellpose (red) and Baysor (blue). b, Barplot showing the adjusted mutual information score across transcripts for each sample. U, untreated; T, treated; b, base panel; a, augmented panel. c, UMAP visualization of the non-malignant cells segmented by Cellpose (left) and Baysor (right). d, Two representative FOVs showing the spatial distributions of all annotated cells (left, Cellpose; right, Baysor). e, Bubble heatmap showing expression levels of select marker genes for annotated cell types (top, Cellpose; bottom, Baysor). f, Comparison of cell type counts (top) and proportions (bottom) between Cellpose (red) and Baysor (blue). g, Heatmap showing the confusion matrix of cell types annotated for cells segmented with Cellpose (x axis) and Baysor (y axis).

Extended Data Fig. 3 Cell type composition across untreated and treated pancreatic cancer samples.

a, UMAP showing subsets of vascular, lymphoid, and myeloid cells. b, Proportions of major cell types (from Fig. 2c-d) across untreated and treated tumors with (left) or without (right) malignant cells included. U, untreated; T, treated; b, base 960-plex panel; a, augmented 990-plex panel. c, Comparison of malignant/non-malignant cell numbers in treated and untreated FOVs (untreated: n = 164; treated: n = 136); two-sided Mann-Whitney U test. d, Proportions of major non-malignant cell types in treated and untreated FOVs (untreated: n = 164; treated: n = 136); Benjamini-Hochberg corrected two-sided Mann-Whitney U tests. e, Comparison of silhouette scores between subsetting malignant cells (n = 27,463) into two (CLS and BSL) or three (CLS, BSL, and NRP) subtypes. Two-sided Mann-Whitney U tests. f, Expression of chemokines with a role in neutrophil recruitment (CXCL1/2/3/5/6/8) from CAFs and malignant cells in treated and untreated FOVs (untreated: n = 164; treated: n = 136); Benjamini-Hochberg corrected two-sided Mann-Whitney U tests. For boxplots in panel c-f, the box limits denote the first and third quartiles, with the median shown in the center and whiskers covering data within 1.5× the interquartile range from the box, with diamonds representing outliers.

Extended Data Fig. 4 Glandular heterogeneity and multicellular neighborhoods in pancreatic cancer.

a, Ten representative side-by-side comparisons between gland assignments manually annotated by a board-certified pathologist (outlined in red) versus those extracted by the DBSCAN algorithm (outlined in different colors). White arrows highlight differences in gland annotations for six highly concordant example FOVs. b, Distribution of the number of cells across malignant glands. c, Observed (black) and expected (gray) distributions of the interspersion of malignant glands, subset to glands in the top quartile of heterogeneity. d, Log2 fold change (y axis) between the observed and expected summed exponential-transformed distances between CAF subtypes and malignant cells across varying decay radii (x axis) for n = 1000 permutations. e, Left: Depiction of the summed exponential functions (z axis height and color bar, decay radius r = 50 μm) that are generated from CD8 T cells for a representative FOV, with spatial locations of malignant subtypes shown as colored dots. Right: Log2 fold change (y axis) between the observed and expected summed exponential-transformed distances between malignant subtypes and CD8 T cells (using a malignant-centric model) for n = 1000 permutations6. f-g, Log2 fold change between the observed and expected summed exponential-transformed distances between malignant subtypes and CD8 T cells (using a malignant-centric model), for varying decay radii (f) and varying quantile thresholds (g), for n = 1000 permutations. Data for (d-g) are presented as mean values ± 95% confidence interval. Color legends for (f) are shared with (g). Significant results for one-sided permutation (p < 0.001) and two-sided K-S (p < 10−16) tests in panels (c-g) are indicated with square and circle symbols, respectively. Statistical test legends for panels (d-g) are shared with panel (c). h-i, Depiction of the summed exponential functions that are generated from CD8 T cells for varying decay radii r (h) and with spatial locations of shuffled malignant subtype annotations, which serves as the null distribution (i).

Extended Data Fig. 5 Performance evaluation of SCOTIA using simulated datasets from SRTsim.

a, The ability of different permutation strategies to identify LR interactions using reference-based (top) and reference-free (bottom) simulated datasets. Strategy A: shuffle gene expression within each cell type; strategy B: shuffle gene expression across all cell types; strategy C: shuffle gene expression across all cell types and permute cell locations. For reference-based simulations, the PDAC SMI dataset was used as the reference. Each scenario contains 3,000 cells across 8 cell types. Gene expression profiles were simulated using negative binomial models with parameters estimated based on the specific cell type in the reference data. To simulate cell type A interacting with cell type B via ligand (L) on A and receptor (R) on B, we randomly assigned the 422 known L-R pairs to specific cell type pairs (A-B), then we increased the expression of the L gene in cell type A and the R gene in adjacent cell type B to a fixed fold change (ranging from 1 to 7). Adjacent cells were defined as the nearest four cells. For reference-free simulations, we used SRTsim to randomly generate cell locations and gene expression profiles with default settings. Each simulation scenario was replicated 5 times. Performance was measured by sensitivity, specificity, F1 score, and precision. Reg is the entropy regularization term, the default is 1.0; regm is the marginal relaxation term, the default is 2.0; Dist_cutff is the distance cutoff for defining ‘adjacent’ cell clusters, the default is 50 µm. b, Performance evaluation of SCOTIA with varied parameters using reference-based simulation datasets from panel a. Fold change of true ligand receptor pairs was set to 2 (top) and 5 (bottom). Error bars in panel a and b indicating standard deviations.

Extended Data Fig. 6 The permutation test strategy used in SCOTIA.

a, Pearson correlation of receptor gene expression between malignant subtypes or ligand gene expression between CAF subtypes (two-sided t test). b, Schematic of the permutation test model used for spatial molecular imaging data. The null distribution was established by randomizing cell locations within a small range (from -20 to 20 µm) while shuffling gene expression in each FOV. c, Neighborhood composition for each cell type from one example FOV with the original (left), permuted (middle) and shuffled negative control (right) data. The negative control was constructed by shuffling cell type labels without any constraints. Neighborhood cells were defined as cells within a radius of 30 µm. d, Boxplots showing the Jaccard index of the top 5% most likely interacting LRs inferred between CAF and malignant cells with varying reg (left) and regm (right) parameters, n = 16. For boxplots, the box limits denote the first and third quartiles, with the median shown in the center and whiskers covering data within 1.5× the interquartile range from the box, with diamonds representing outliers. e, The top five strongest interacting cell type pairs inferred by using cost function Eq. 6 (left) and 7 (right) (Methods). Dot size represents the number of permutation test-significant LR pairs, colored based on the average LR interaction score. Bar plot indicates the average interaction strengths of each cell type pair for the treated and untreated groups. U, untreated; T, treated; b, base 960-plex panel; a, augmented 990-plex panel. f, The top five strongest interacting cell type pairs inferred by using different permutation strategies.

Extended Data Fig. 7 Pathway enrichment and target gene analysis for the SMI data.

a, Ligand–receptor (LR) interactions significantly up- or down- regulated in treated samples between CAF and malignant cells, related to Fig. 5a. The cost functions used were Eq. 6 (left) and 7 (right) (Methods). Benjamini-Hochberg corrected two-sided Mann-Whitney U tests. b, Differentially enriched LR interactions inferred with a mixed effects model test (two-sided, Benjamini-Hochberg adjusted p value, Methods). c, Comparison of ligand and receptor gene expression between SCOTIA-inferred interactions versus non-interacting CAF–malignant cell pairs. Two-sided Mann-Whitney U tests (CLCF1–CNTFR: untreated, n = 158; treated: n = 89. WNT5A–FZD5: untreated, n = 170; treated: n = 98). d, Pathway enrichment analysis with ligand (left) or receptor (right) gene sets from Fig. 5b. Top pathways enriched in untreated (purple) and treated (red) tumors are shown. Benjamini-Hochberg corrected one-sided Fisher’s exact test. e, Boxplots summarizing the difference in target gene expression between significantly up- and down-regulated LR groups for five other abundant cell type pairs. Two-sided Mann-Whitney U test. Malignant–CAF, up: n = 82; down: n = 6. Malignant–macrophage, up: n = 30; down: n = 3. CAF–macrophage, up: n = 20; down: n = 19. Macrophage–CAF, up: n = 56; down: n = 29. Macrophage–malignant, up: n = 10; down: n = 3. For boxplots in panel c and e, the box limits denote the first and third quartiles, with the median shown in the center and whiskers covering data within 1.5× the interquartile range from the box, with diamonds representing outliers.

Extended Data Fig. 8 Pathway enrichment analysis for the co-culture tumoroid data.

a, Pathway enrichment analysis with the ligand gene set (from CAFs, left) or receptor gene set (from malignant cells, right) that were significantly higher (red) or lower (purple) in the treated versus treatment-naïve co-culture tumoroids, related to Fig. 7c. Benjamini-Hochberg corrected one-sided Fisher’s exact test.

Extended Data Fig. 9 Association between IL6 family signaling, inflammatory CAFs and specific malignant subtypes.

a, Log2 fold change (y axis) between the observed and expected summed exponential-transformed distances between malignant subtypes and iCAFs across varying decay radii (x axis) for n = 1000 permutations. Data are presented as mean values ± 95% confidence interval. Significant results of one-sided permutation (p < 0.001) and two-sided K-S (p < 10−16) tests are indicated with square and circle symbols, respectively. b, Proportions of CAF subtypes per FOV (n = 320) in SMI (left) and per sample (n = 43) in single-nucleus RNA-seq6 (right) datasets, stratified by treatment. Box limits denote the first and third quartiles, median shown in the center, and whiskers cover data within 1.5× interquartile range, with diamonds representing outliers. Two-sided two-sample Mann-Whitney U test. c, Number of transcripts of specific IL6 family ligands expressed in CAFs (y axis) in the SMI dataset, stratified by CAF subtype. iCAF: n = 102934. myCAF: n = 149349. Data are presented as mean values ± 95% confidence interval. Two-sided two-sample Mann-Whitney U test.

Extended Data Fig. 10 Performance evaluation of SCOTIA with varied parameters using PDAC SMI and co-culture snRNA-seq datasets.

a, The number of significant LRs enriched in treated samples as a function of varying SCOTIA parameters; adjusted p < 0.05, two-sided Mann-Whitney U. b, The power (y axis) of SCOTIA with varied parameters for identifying treatment-enriched ligand-receptor (LR) interactions. Ground truth was defined with the co-culture snRNA-seq dataset. True LR interactions were defined as those exhibiting enrichment in treated tumoroids. Both ligand and receptor genes were required to display higher expression in treated compared to untreated samples, with at least one gene having a fold change >1.5. Conversely, false LR interactions were defined by enrichment in untreated samples with the same criteria. Performance was measured by sensitivity, specificity, F1 score, and precision. Reg is the entropy regularization term, the default is 1.0; Regm is the marginal relaxation term, the default is 2.0; Dist_cutoff is the distance cutoff for defining ‘adjacent’ cell clusters, the default is 50 µm.

Supplementary information

Supplementary Information

Supplementary Figs. 1–4 and Tables 1–4.

Reporting Summary

Peer Review File

Supplementary Data 1

Full SCOTIA results.

Supplementary Data 2

SMI probe sequences.

Source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 8

Statistical source data.

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

Shiau, C., Cao, J., Gong, D. et al. Spatially resolved analysis of pancreatic cancer identifies therapy-associated remodeling of the tumor microenvironment. Nat Genet (2024). https://doi.org/10.1038/s41588-024-01890-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41588-024-01890-9

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