frontier-banner
Frontiers
Home>Frontiers>

Molecular Cancer | HBV and Clonorchis sinensis infection reveal tumor microenvironment heterogeneity and immunosuppressive landscape in hepatocellular carcinoma

Molecular Cancer | HBV and Clonorchis sinensis infection reveal tumor microenvironment heterogeneity and immunosuppressive landscape in hepatocellular carcinoma
--

This study integrates single-cell and spatial transcriptomic techniques to systematically analyze the tumor microenvironment of hepatocellular carcinoma patients under different infection backgrounds, providing multi-omics data support and offering new insights into precision treatment and prognosis evaluation for infection-associated liver cancer.

 

Literature Overview

This article, titled "Integrating single cell- and spatial- resolved transcriptomics unravels the inter-tumor heterogeneity and immunosuppressive landscape in HBV- and Clonorchis sinensis-associated hepatocellular carcinoma," published in "Molecular Cancer," reviews and summarizes the impact of HBV and Clonorchis sinensis (C. sinensis) infection on the tumor microenvironment (TME) in hepatocellular carcinoma (HCC). The study integrates scRNA-seq and ST-seq data to evaluate the TME characteristics and intercellular communication patterns of HCC patients under different infection conditions. Furthermore, key signaling pathways and cell types were validated through immunofluorescence and in vitro experiments, providing new perspectives for therapeutic strategies in infection-associated HCC.

Background Knowledge

Liver cancer, specifically hepatocellular carcinoma (HCC), is the third leading cause of cancer-related deaths worldwide. Its high mortality rate is closely related to the complex tumor microenvironment (TME). HBV and Clonorchis sinensis infections are significant risk factors for HCC; however, the combined effect of these two infections on the TME remains unclear. The intricate interactions among immune cells, stromal cells, and cytokines within the TME are crucial for tumor initiation, progression, and therapeutic response. Existing studies have shown that SPP1+ macrophages, cancer-associated fibroblasts (CAFs), and immunosuppressive molecules such as PD-L1+ TANs play critical roles in HCC immune escape. However, the heterogeneity of the TME across different infection backgrounds has not been fully characterized. This study fills this gap, providing a foundation for precision classification and targeted therapies for HCC patients.

 

 

Research Methods and Experiments

The study enrolled 269 HCC patients with different infection backgrounds to analyze their clinical outcomes. scRNA-seq and ST-seq data from Clonorchis sinensis-associated (CP), dual infection (DP), and HBV-associated (HP) HCC patients were integrated for analysis. Through single-cell clustering, gene expression difference analysis, copy number variation (CNV) inference, and GSVA enrichment analysis, the epithelial, stromal, and immune cell heterogeneity among different HCC subtypes were characterized. Key signaling pathways and cell types were further validated using immunofluorescence and in vitro co-culture experiments.

Key Conclusions and Perspectives

  • Clonorchis sinensis (CP) infection is associated with worse clinical prognosis in HCC patients, and dual infection (DP) patients have the poorest survival outcomes
  • HCC cells in the DP group show higher expression of malignant markers, CNV scores, and p53 pathway activation compared to other groups, along with the lowest survival rates
  • The DP group TME exhibits significant enrichment of SPP1+ macrophages, exhausted CD8+ T cells, and COL1A1+ fibroblasts
  • Both CP and HP groups show elevated levels of M2-like macrophages and ENPP2+ liver endothelial cells, indicating distinct immune landscapes under different infection backgrounds
  • Integration of scRNA-seq and ST-seq reveals distinct cellular communication networks among HCC subtypes, such as the interaction between Macro1 (SPP1+) and tumor cells
  • In vitro experiments demonstrate that C. sinensis excretory-secretory products (CsESPs) can induce p53 and EMT pathway activation in HCC cells and promote macrophage M2 polarization


Research Significance and Prospects

This study is the first systematic comparison of HCC subtypes associated with different infection backgrounds, revealing significant heterogeneity in cellular composition, gene expression, pathway activation, and immune profiles. These findings provide theoretical support for HCC subtype classification based on infection features and offer a foundation for developing targeted therapeutic strategies tailored to specific TME profiles. Future studies may further explore the potential applications of these cell subtypes in immunotherapy and evaluate the clinical utility of CsESPs as biomarkers.

 

 

Conclusion

This study systematically reveals the heterogeneity of the tumor microenvironment in HBV- and Clonorchis sinensis-associated HCC through multi-omics integration. It identifies significant differences in immune cell infiltration, signaling pathway activation, and cellular communication patterns among HCC patients with different infection statuses. Notably, dual infection (DP) patients show high enrichment of SPP1+ macrophages and exhausted CD8+ T cells, indicating a highly immunosuppressive TME. The p53 and EMT pathways are specifically activated in dual infection cases, contributing to the poor prognosis observed. These findings enhance our understanding of infection-driven HCC heterogeneity and provide molecular and cellular bases for personalized therapeutic strategies. Future research should focus on the functional mechanisms of these cell subtypes and their potential as therapeutic targets.

 

Reference:
Jiayun Chen, Wenmin Lu, Yanni Lou, Guo-Dong Lu, and Jigang Wang. Integrating single cell- and spatial- resolved transcriptomics unravels the inter-tumor heterogeneity and immunosuppressive landscape in HBV- and Clonorchis sinensis-associated hepatocellular carcinoma. Molecular Cancer.
ImmuneBuilder(AntiBody)
ImmuneBuilder, including ABodyBuilder2, NanoBodyBuilder2, and TCRBuilder2, is specifically designed for predicting the structure of immunoproteins (e.g., antibodies, nanobodies, and T-cell receptors), and employs AlphaFold-Multimer's structural modules with modifications specific to the immunoproteins to improve prediction accuracy.ImmuneBuilder is able to quickly generate immunoprotein structures that resemble experimental data much faster than AlphaFold2 and without the need for large sequence databases or multiple sequence comparisons. The tool's features include high accuracy, fast prediction, and open-source accessibility for structural analysis of large-scale sequence datasets, especially in the study of immunoprotein structures from next-generation sequencing data. immuneBuilder also provides error estimation to help filter out erroneous models, enhancing its value for applications in biotherapeutics and immunology research. Figure 1 shows the architecture of AbBuilder2, and the same architecture is used for NanoBodyBuilder2 and TCRBuilder2.