
This study systematically reveals the key mechanisms of immune metabolic reprogramming and Treg interaction with cancer-associated fibroblasts (CAFs) leading to immunosuppression in steatotic liver disease-related hepatocellular carcinoma (SLD-HCC), by integrating single-cell transcriptomics, spatial transcriptomics, and mass cytometry (CyTOF). Blocking TNFRSF14 enhances anti-PD-1 therapy efficacy, offering novel therapeutic strategies for patients with SLD-HCC.
Literature Overview
This article, titled 'Targeting Treg–fibroblast interaction to enhance immunotherapy in steatotic liver disease-related hepatocellular carcinoma', published in the journal 'Gut', reviews and summarizes the characteristics of the immune microenvironment in steatotic liver disease-related hepatocellular carcinoma (SLD-HCC) and its mechanisms of resistance to immunotherapy. The article uses multi-omics approaches to reveal how Treg and CAF interactions contribute to tumor immune exclusion and immunosuppression, and proposes the TNFSF14–TNFRSF14 axis as a potential therapeutic target, providing theoretical support for developing novel combination immunotherapy strategies.
Background Knowledge
Steatotic liver disease-related hepatocellular carcinoma (SLD-HCC) is a subtype of liver cancer with rapidly increasing incidence in recent years. Its characteristic immune-excluded microenvironment leads to poor efficacy of immune checkpoint inhibitors. This study uses scRNA-seq, CyTOF, and spatial transcriptomics to reveal the Treg–CAFs interaction mechanism in SLD-HCC, and validates the therapeutic effect of targeting TNFRSF14 in mouse models. The research provides critical insights into understanding immunotherapy resistance and developing novel interventions.
Research Methods and Experiments
The research team employed single-cell RNA sequencing (scRNA-seq), mass cytometry (CyTOF), spatial transcriptomics (Visium and CosMx technologies), and multiplex immunofluorescence techniques to systematically analyze the tumor microenvironment in 22 cases of SLD-HCC and 31 non-SLD-HCC samples. Additionally, the therapeutic impact of TNFRSF14 blockade on the tumor microenvironment and anti-PD-1 efficacy was evaluated in a high-fat diet-induced mouse model of HCC.
Key Conclusions and Perspectives
Research Significance and Prospects
This study provides the first systematic analysis of the immunosuppressive microenvironment in SLD-HCC and its connection to metabolic adaptation, offering preclinical evidence for therapeutic strategies targeting Treg–CAF interactions. Future work can explore the clinical application of combining inhibitors of this signaling axis with immune checkpoint blockade, paving the way for precision immunotherapy in SLD-HCC patients.
Conclusion
This study reveals a unique immune metabolic adaptation and Treg–CAF interaction-mediated immunosuppressive mechanism in steatotic liver disease-related hepatocellular carcinoma. Targeting the TNFSF14–TNFRSF14 signaling pathway effectively enhances antitumor immunity and overcomes immunotherapy resistance. These findings provide a theoretical basis for developing SLD-HCC-specific combination immunotherapy regimens and advancing precision cancer immunotherapy.

