Abstract Heterogeneity in colorectal cancer (CRC) manifests in the diversity and interactions among tumor cells, support stroma, and immune components, critically impacting patient prognosis and therapeutic efficacy. In this study, spatial transcriptomics (ST, 10x Genomics Visium Spatial Gene Expression) was performed on 40 retrospectively collected CRC samples, along with detailed clinicopathological parameters and long-term clinical outcomes, with the goal of unraveling the molecular topography of the tumor and surrounding stromal compartments, and associating their spatial interactions to clinical outcomes. Five primary RNA groups were identified through unsupervised clustering on ST data, correlating with tissue classes detected in H&E-stained images via a pre-trained deep learning model. Markers and pathways specific to these tissues were identified. Furthermore, immune cell infiltration in these regions was deconvoluted using reference single-cell RNA sequencing (scRNA-seq) data. Sub-clustering within the tumor groups revealed five major tumor subtypes, each characterized by a distinct set of differentially expressed marker genes. Enrichment analysis across these subtypes highlighted distinct activation and suppression in pathways including KRAS, p53, MAPK, and Integrin. Tumors exhibited intra- and inter-tumoral heterogeneity. Location-specific tumor subtypes were observed, with those in right-sided CRC displaying increased immune cell infiltration, linked to enriched pathways such as Type II Interferon Signaling and Cytokine Signaling. Conversely, left-sided CRC subtypes had reduced immune cell presence, and were characterized by enhanced pathways related to vascular development, Hypoxia, and p53 Signaling. Inferred clonal lineages from copy number variation (CNV) using ST suggested a spatially structured phylogenetic organization corresponding to transcriptional clusters. Additionally, the ST data unveiled distinct spatial distribution patterns across various cancer-associated fibroblast (CAF) phenotypes, such as matrix, inflammatory, vessel-associated, tumor-promoting, and antigen-presenting CAFs. Transcriptional patterns uncovered through scRNA-seq, including epithelial-mesenchymal transition (EMT) signatures and consensus molecular subtypes (CMS), displayed unique spatial layouts within the tumor microenvironment. To our knowledge, this is the most comprehensive study to date on spatial transcriptomic heterogeneity within CRC patients, revealing distinct tumor, stroma and immune compositions and spatial structures. This spatially detailed map provides novel insights into CRC heterogeneity, critical for advancing personalized treatment. Citation Format: Jie Zhou, Todd B. Sheridan, Sergii Domanskyi, Stephanie L. Cowles, Sunghee Park, Ilham Putra, Olga Anczukow, Jeffrey H. Chuang, Jill C. Rubinstein. Unveiling the spatial landscape of tumor and stroma heterogeneity in colorectal cancer with spatial transcriptomics [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 1150.
Read full abstract