98%
921
2 minutes
20
Cancer immunoediting reflects the role of the immune system in eliminating tumor cells and shaping tumor immunogenicity, which leaves marks in the genome. In this study, we systematically evaluate four methods for quantifying immunoediting. In colorectal cancer samples from The Cancer Genome Atlas, we found that these methods identified 78.41%, 46.17%, 36.61%, and 4.92% of immunoedited samples, respectively, covering 92.90% of all colorectal cancer samples. Comparison of 10 patient-derived xenografts (PDXs) with their original tumors showed that different methods identified reduced immune selection in PDXs ranging from 44.44% to 60.0%. The proportion of such PDX-tumor pairs increases to 77.78% when considering the union of results from multiple methods, indicating the complementarity of these methods. We find that observed-to-expected ratios highly rely on neoantigen selection criteria and reference datasets. In contrast, HLA-binding mutation ratio, immune dN/dS, and enrichment score of cancer cell fraction were less affected by these factors. Our findings suggest integration of multiple methods may benefit future immunoediting analyses.
Download full-text PDF |
Source |
---|---|
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC12049729 | PMC |
http://dx.doi.org/10.1016/j.crmeth.2025.101006 | DOI Listing |
Cell Res
September 2025
Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
Tumors evolve to avoid immune destruction and establish an immunosuppressive microenvironment. Syngeneic mouse tumor models are critical for understanding tumor immune evasion and testing cancer immunotherapy. Derived from established mouse tumor cell lines that can already evade the immune system, these models cannot simulate early phases of immunoediting during initial tumorigenesis.
View Article and Find Full Text PDFCancer Treat Res
August 2025
Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA.
Artificial intelligence (AI) and machine learning (ML) are revolutionizing cancer immunotherapy by addressing the complex interplay between cancer and the immune system. This chapter explores how AI technologies enhance immunotherapy development across multiple domains: antibody design, response prediction, biomarker identification, and T-cell target discovery. In therapeutic antibody design, AI improves efficiency through predictive modeling of antibody-antigen interactions, structure prediction tools, generative models that create novel antibody sequences, and developability optimization.
View Article and Find Full Text PDFEur J Med Res
August 2025
Department of Thoracic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
Although immune checkpoint inhibitors (ICIs) hold promise for those diagnosed with advanced human lung adenocarcinoma (LUAD), notable heterogeneity in patient responses and the complex tumor microenvironment (TME) limits their clinical utility while providing clinical benefit. To identify new therapeutic targets to pursue in chimeric antigen receptor (CAR) T-cell therapy, we leveraged an integrative multi-omic approach. This study identified three oncogenic drivers, karyopherin α2 (KPNA2), golgi membrane protein 1 (GOLM1) and thymidine kinase 1 (TK1), that are overexpressed in LUAD, spatially enriched in malignant niches and associated with significantly reduced overall survival (log-rank P < 0.
View Article and Find Full Text PDFFront Immunol
August 2025
Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Moscow, Russia.
The anti-tumor role of the immune system has long been associated with interferon-γ-mediated activation of immune cells and their ability to recognize and eliminate transformed cells. Fundamental principles of tumor immunoediting describe a dynamic interplay between the immune system and neoplastic cells, wherein immune pressure can paradoxically shape tumor evolution. Within this context, macrophages, natural killer cells, and T lymphocytes are central effectors of anti-tumor immunity.
View Article and Find Full Text PDFScand J Immunol
August 2025
Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.
A key mechanism of tumour immune escape from CD8 cytotoxic T lymphocytes occurs via downregulation of NLRC5, an IFNγ-induced transcriptional activator of MHC class-I. As NLRC5 deficiency does not abrogate CD8 T cell development, we investigated whether NLRC5-dependent antitumour immune mechanisms are required for immune surveillance. We studied the development of 3-methylcholanthrene (MCA)-induced endogenous fibrosarcoma in Nlrc5 mice with Nlrc5 and Rag1 mice serving as controls.
View Article and Find Full Text PDF