%0 Journal Article %T PanIN and CAF transitions in pancreatic carcinogenesis revealed with spatial data integration. %A Bell ATF %A Mitchell JT %A Kiemen AL %A Lyman M %A Fujikura K %A Lee JW %A Coyne E %A Shin SM %A Nagaraj S %A Deshpande A %A Wu PH %A Sidiropoulos DN %A Erbe R %A Stern J %A Chan R %A Williams S %A Chell JM %A Ciotti L %A Zimmerman JW %A Wirtz D %A Ho WJ %A Zaidi N %A Thompson E %A Jaffee EM %A Wood LD %A Fertig EJ %A Kagohara LT %J Cell Syst %V 15 %N 8 %D 2024 Aug 21 %M 39116880 %F 11.091 %R 10.1016/j.cels.2024.07.001 %X This study introduces a new imaging, spatial transcriptomics (ST), and single-cell RNA-sequencing integration pipeline to characterize neoplastic cell state transitions during tumorigenesis. We applied a semi-supervised analysis pipeline to examine premalignant pancreatic intraepithelial neoplasias (PanINs) that can develop into pancreatic ductal adenocarcinoma (PDAC). Their strict diagnosis on formalin-fixed and paraffin-embedded (FFPE) samples limited the single-cell characterization of human PanINs within their microenvironment. We leverage whole transcriptome FFPE ST to enable the study of a rare cohort of matched low-grade (LG) and high-grade (HG) PanIN lesions to track progression and map cellular phenotypes relative to single-cell PDAC datasets. We demonstrate that cancer-associated fibroblasts (CAFs), including antigen-presenting CAFs, are located close to PanINs. We further observed a transition from CAF-related inflammatory signaling to cellular proliferation during PanIN progression. We validate these findings with single-cell high-dimensional imaging proteomics and transcriptomics technologies. Altogether, our semi-supervised learning framework for spatial multi-omics has broad applicability across cancer types to decipher the spatiotemporal dynamics of carcinogenesis.