98%
921
2 minutes
20
Background And Aims: The prognoses and therapeutic responses of patients with intrahepatic cholangiocarcinoma (iCCA) depend on spatial interactions among tumor microenvironment (TME) components. However, the spatial TME characteristics of iCCA remain poorly understood. The aim of this study was to generate a comprehensive spatial atlas of iCCA using artificial intelligence-assisted spatial multiomics patterns and to identify spatial features associated with prognosis and immunotherapy.
Approach And Results: Spatial multiomics, including imaging mass cytometry (n=155 in-house), spatial proteomics (n=155 in-house), spatial transcriptomics (n=4 in-house), multiplex immunofluorescence (n=20 in-house), single-cell RNA sequencing (scRNA-seq, n=9 in-house and n=34 public), bulk RNA-seq (n=244 public), and bulk proteomics (n=110 in-house and n=214 public), were employed to elucidate the spatial TME of iCCA. More than 1.06 million cells were resolved, and the findings revealed that spatial topology, including cellular deposition patterns, cellular communities, and intercellular communications, profoundly correlates with the prognosis of patients with iCCA. Specifically, CD163 hi M2-like resident-tissue macrophages suppress antitumor immunity by directly interacting with CD8 + T cells, resulting in poorer patient survival. Additionally, 5 spatial subtypes with distinct prognoses were identified, and potential therapeutic options were generated for these subtypes. Furthermore, a spatial TME deep learning system was developed to predict the prognosis of patients with iCCA with high accuracy from a single 1-mm 2 tumor sample.
Conclusions: This study offers preliminary insights into the spatial TME ecosystem of iCCA, providing valuable foundations for precise patient classification and the development of personalized treatment strategies.
Download full-text PDF |
Source |
---|---|
http://dx.doi.org/10.1097/HEP.0000000000001283 | DOI Listing |
Mol Cell Probes
September 2025
Department of Urology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, 453100, China. Electronic address:
Background: Interleukin-1 receptor-like 1 (IL1RL1, also known as ST2) plays a critical role in immune regulation. Pan-cancer analysis has revealed that IL1RL1 is closely associated with cellular immune functions; however, its role in clear cell renal cell carcinoma (ccRCC) and the tumor microenvironment (TME) remains poorly defined.
Methods: We analyzed IL1RL1 expression patterns using data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases.
The tumor microenvironment (TME) of chronic inflammation-associated cancers (CIACs) is shaped by cycles of injury and maladaptive repair, yet the principles organizing fibrotic stroma in these tumors remain unclear. Here, we applied the concept of hot versus cold fibrosis, originally credentialed in non-cancerous fibrosis of heart and kidney, to lung squamous cell carcinoma (LUSC), a prototypical CIAC. Single-cell transcriptomics of matched tumor and adjacent-normal tissue from 16 treatment-naive LUSC patients identified a cold fibrotic architecture in the LUSC TME: cancer-associated fibroblasts (CAFs) expanded and adopted myofibroblast and stress-response states, while macrophages were depleted.
View Article and Find Full Text PDFSmall Methods
September 2025
Despite the availability of numerous approved immunotherapies for various cancers, durable progression-free survival remains relatively uncommon among patients with advanced cancer. As research into immunotherapy intensifies, the heterogeneity and complexity of the tumor microenvironment (TME) have emerged as critical determinants of treatment response and a major obstacle to understanding tumor resistance mechanisms. Recent advances in spatially resolved transcriptomics (SRT) enable transcriptome-wide measurement of gene expression while preserving essential spatial information, which supports the characterization of the features of the TME.
View Article and Find Full Text PDFNeuro Oncol
September 2025
Leeds Institute of Medical Research, University of Leeds, Leeds, UK.
Background: Glioblastoma (GBM), the most aggressive adult brain cancer, comprises a complex tumour microenvironment (TME) with diverse cellular interactions that drive progression and pathobiology. The aim of this study was to understand how these spatial patterns and interactions evolve with treatment.
Methods: To explore these relationships, we employed imaging mass cytometry to measure the expression of 34 protein markers, enabling the identification of GBM-specific cell types and their interactions at single-cell protein level in paired primary (pre-treatment) and recurrent (post-treatment) GBM samples from five patients.
J Exp Clin Cancer Res
September 2025
State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, 400010, China.
The tumor microenvironment (TME) of breast cancer is a complex ecosystem, in which cancer-associated fibroblasts (CAFs), as the most abundant stromal cell type, meticulously construct an ecological niche that supports tumor growth through mechanisms including extracellular matrix (ECM) remodeling, secretion of bioactive factors, and interactions with neighboring cells. High-resolution technologies, including single-cell sequencing and spatial transcriptomics, have revealed the high heterogeneity, functional diversity, and spatial distribution within the CAF population. Significant differences exist in the interactions between distinct CAF subpopulations and immune cells.
View Article and Find Full Text PDF