frontier-banner
Frontiers
Home>Frontiers>

Gut | Targeting Treg-CAFs Interaction Enhances Immunotherapy Efficacy in Steatotic Liver Disease-Related Hepatocellular Carcinoma

Gut | Targeting Treg-CAFs Interaction Enhances Immunotherapy Efficacy in Steatotic Liver Disease-Related Hepatocellular Carcinoma
--

This study reveals the interaction mechanism between regulatory T cells (Treg) and cancer-associated fibroblasts (CAFs) in the tumor microenvironment of steatotic liver disease-related hepatocellular carcinoma. Targeting the TNFSF14-TNFRSF14 signaling pathway can effectively enhance antitumor immune responses, providing a theoretical basis for developing novel combination immunotherapy strategies.

 

Literature Overview

The study titled 'Targeting Treg–fibroblast interaction to enhance immunotherapy in steatotic liver disease-related hepatocellular carcinoma', published in Gut, systematically reviews and summarizes the characteristics of the tumor microenvironment in steatotic liver disease-related hepatocellular carcinoma (SLD-HCC) and its impact on immunotherapy response. Utilizing single-cell transcriptomics, cytometry by time-of-flight (CyTOF), and spatial transcriptomics, the research comprehensively analyzed microenvironmental differences between 22 SLD-HCC and 31 non-SLD-HCC cases. Key signaling pathways were further validated through multiplex immunohistochemistry, mouse models, and immunotherapy patient cohorts.

Background Knowledge

Steatotic liver disease (SLD) is a significant predisposing factor for hepatocellular carcinoma (HCC), and its metabolic and immunological microenvironmental adaptations contribute to resistance to immunotherapy. Treg cells and CAFs are recognized as core immunosuppressive cell types, yet their specific interaction mechanisms in SLD-HCC remain unclear. This study focuses on the TNFSF14-TNFRSF14 signal axis to elucidate its role in Treg-CAFs interaction and to explore whether targeting this pathway can enhance the efficacy of immune checkpoint blockade therapy. The study also evaluates molecular features such as Wnt signaling activity and neoantigen load, integrating findings from high-fat diet mouse models and anti-PD-1 treatment responses, to identify new intervention targets for immunotherapy resistance in SLD-HCC.

 

 

Research Methods and Experiments

The study included 22 SLD-HCC and 31 non-SLD-HCC patients, with 2–5 systematic tissue samplings per tumor to evaluate intratumoral heterogeneity. Using single-cell RNA sequencing (scRNA-seq), CyTOF-based immune cell profiling, spatial transcriptomics (Visium and CosMx), and multiplex immunofluorescence (mIF), the research systematically analyzed cellular composition, gene expression, ligand–receptor interaction networks, and their spatial distribution within the tumor microenvironment. Additionally, a high-fat diet-induced HCC mouse model was developed to evaluate antitumor immune responses following treatment with anti-TNFRSF14 antibodies alone or in combination with anti-PD-1 therapy.

Key Conclusions and Perspectives

  • SLD-HCC patients show significant enrichment of Treg and CD4+ memory T cells, while CD8+ T cell numbers are reduced, indicating an immune-excluded microenvironment.
  • scRNA-seq and CyTOF analyses reveal that Treg and CAFs co-localize at the tumor margin in SLD-HCC, where the TNFSF14-TNFRSF14 signaling pathway is significantly activated.
  • Multiplex immunofluorescence further confirms spatial proximity between Treg and CAFs, with significantly reduced average intercellular distance.
  • Targeting TNFRSF14 markedly reduces Treg infiltration, enhances CD8+ T cell and memory CD4+ T cell activity, and improves the efficacy of anti-PD-1 therapy.
  • Mechanistically, the TNFSF14-TNFRSF14 axis drives immunosuppression and influences treatment response by promoting Treg survival and metabolic reprogramming of CAFs.

Research Significance and Prospects

This study provides a new molecular explanation for immunotherapy resistance in steatotic liver disease-related HCC and proposes targeting the TNFSF14-TNFRSF14 axis as a potential therapeutic strategy. Future studies should further evaluate the predictive value of this pathway in clinical settings and explore its synergistic interactions with other immune checkpoint pathways. Additionally, integrating spatial transcriptomic and metabolomic data may facilitate the development of personalized immunomicroenvironment modulation approaches.

 

 

Conclusion

By employing multi-omics and spatial transcriptomic techniques, this study systematically uncovered the spatial interaction network between Treg and cancer-associated fibroblasts in steatotic liver disease-related hepatocellular carcinoma, identifying the TNFSF14-TNFRSF14 signaling pathway as a key driver of immune suppression. Blocking this pathway effectively remodels the immune microenvironment and enhances anti-PD-1 therapeutic response, offering a new target for precision immunotherapy in SLD-HCC patients. These findings deepen our understanding of immune escape mechanisms in SLD-HCC and lay the foundation for developing combination therapies targeting the Treg-CAFs axis.

 

Reference:
Aldo Prawira, Hang Xu, Yu Mei, Jinmiao Chen, and Valerie Chew. Targeting Treg–fibroblast interaction to enhance immunotherapy in steatotic liver disease-related hepatocellular carcinoma. Gut.
PTM Hotspot is a tool designed to identify potential post-translational modification (PTM) risk sites within antibody sequences. By scanning sequences, it accurately locates PTM sites that may affect antibody stability, activity, or immunogenicity, providing critical data support for antibody drug development. Additionally, PTM Hotspot features powerful visualization capabilities, presenting PTM risk site information in an intuitive sequence map. It also enables users to visually assess, via scatter plots, whether sequences fall within therapeutic antibody risk regions. This helps researchers quickly interpret analysis results, optimize antibody sequence design, reduce risks in biopharmaceutical development, and accelerate the R&D process.