Skip to main content

EDITORIAL article

Front. Oncol., 10 September 2024
Sec. Cancer Metabolism
This article is part of the Research Topic Immunometabolism and Tumor Microenvironment in Hepatocellular Carcinoma View all 6 articles

Editorial: Immunometabolism and tumor microenvironment in hepatocellular carcinoma

Luyun Zhang,Luyun Zhang1,2Lianyuan Tao,Lianyuan Tao3,4Dean TianDean Tian5Dongxiao Li,*Dongxiao Li1,2*
  • 1Department of Gastroenterology, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
  • 2Henan Key Medical Laboratory for Molecular Immunology of Digestive Diseases, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
  • 3Department of Hepatobiliary Pancreatic Surgery, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
  • 4Henan Provincial Key Medical Laboratory for Hepatobiliary and Pancreatic Diseases, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
  • 5Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China

Hepatocellular carcinoma (HCC) remains a highly prevalent and lethal malignancy (1). The historically poor prognosis associated with HCC is undergoing a significant transformation due to the introduction of effective systemic therapies. Despite these advances, the molecular mechanisms underlying immune responses and evasion in HCC remain poorly understood. While the metabolic hallmarks of cancer have been extensively studied, the metabolic states of immune cells are less well characterized. Immunometabolism is an emerging field that investigates the metabolic modulation of immune cells, which is a crucial determinant of their phenotype and function within the tumor microenvironment (TME) (2). Despite remarkable progress in cancer immunotherapy, our understanding of the metabolic interactions between tumor and immune cells within the TME, as well as during immunotherapy, remains limited. Therefore, a comprehensive understanding of immunometabolism in the TME may provide innovative therapeutic perspectives for the treatment of HCC. The papers in this Research Topic illuminate the complex regulatory mechanisms that govern the interactions between tumors and the immune microenvironment. They highlight potential strategies for identifying therapeutic targets for immunotherapy, offering valuable insights into how these interactions can be leveraged to improve treatment outcomes for HCC.

Tumors can hijack existing blood vessels in the surrounding non-malignant tissue through a process known as vessel co-option (VC), which differs from neovascularization and influences disease progression, metastasis, and treatment response (3). In this Research Topic, Dan Yang et al. present a comprehensive analysis of the existing evidence, clinical outcomes, and potential molecular mechanisms associated with VC in liver cancer. Their findings indicate that VC is a significant mechanism contributing to resistance against anti-vascular therapies. Unlike vasogenic tumors, this unique vascular immune niche, characterized by distinct states and distributions of immune cells, fosters the development of an immunosuppressive tumor microenvironment that influences responses to immunotherapy. The authors further investigate the correlation between VC, drug resistance, and the immune microenvironment, offering novel insights for the advancement of liver cancer immunotherapy.

In tumors, hostile microenvironments initiate a persistent state of endoplasmic reticulum stress (ERS) that governs multiple pro-tumor properties in cancer cells while dynamically reprogramming the function of innate and adaptive immune cells (4). Consequently, Zhan et al. conducted an analysis on ERS-associated gene and identified specific gene markers associated with ERS that can effectively stratify patients into high- or low-risk groups for overall survival. The high-risk group is characterized by increased infiltration of immune cells. Furthermore, combining these ERS-associated gene markers with the tumor-node-metastasis (TNM) stage can enhance the accuracy of prognosis prediction. Notably, GP6 gene expression is down-regulated in liver cancer tissues and correlates with poor prognosis; therefore, targeted inhibition of GP6 may hold potential as a therapeutic approach.

As a member of the Forkhead box K transcription factor family, FOXK2 directly regulates the expression of genes involved in nucleotide synthesis, thereby promoting liver cancer growth and conferring chemotherapy resistance in cancer cells (5). In this context, Xing et al. provide a comprehensive overview of the current understanding of the regulatory mechanisms governing FOXK2 and its downstream targets, highlighting the significant impact that dysregulation of FOXK2 can have on cancer initiation and progression. Furthermore, they explore the potential therapeutic implications of targeting FOXK2 for tumor treatment, aiming to offer valuable insights for the diagnosis and management of conditions associated with this protein.

The basement membrane (BM) is a nanoporous matrix layer that physically separates the primary tumor from the surrounding stroma (6). Disruptions in protein expression and turnover are associated with tumor development, while the loss of basement membrane integrity is linked to tumor metastasis (7). Li et al. developed six risk score models based on basement membrane-related genes (BMRGs), revealing that a high BMRGs score correlates significantly with unfavorable prognoses in liver cancer patients, including shorter overall survival, increased extracellular matrix and signal transduction functions, and an immunosuppressive status. Additionally, two key genes, PKM and ITGA3, were found to be closely associated with poor prognosis. Thus, the BMRGs score has potential as a novel clinical predictor to enhance the prognostic evaluation of liver cancer patients.

In TME, cell communication mediated by ligand-receptor interactions orchestrates a wide array of cellular responses, including proliferation, migration, angiogenesis, immune responses, and cell death (8). Research has demonstrated that bidirectional ligand-receptor interactions at the 100 μm-wide boundaries of tumor clusters in HCC play a crucial role in maintaining the tumor’s structural integrity (9). In this context, Dai et al. identified molecular subtypes, prognostic models, and immune microenvironment characteristics based on ligand-receptor interactions involved in malignant HCC cell communication. Additionally, they elucidated the tumorigenic role of HCC cells in recruiting M2-like tumor-associated macrophages via the CCL16-CCR1 axis.

Previous clinical studies have shown that the effective rate of single-drug anti-PD-1/PD-L1 treatment is less than 20%, indicating that the majority of patients experience resistance to immunotherapy (10). This underscores the urgent need to identify biomarkers for the early prediction of immunotherapy response. In this context, Liu et al. utilized ultra-high-performance liquid chromatography-mass spectrometry (UHPLC-MS) technology to analyze the relative abundance of glycerophospholipid metabolites. Their work effectively predicts the survival benefit of anti-PD-1/PD-L1 treatment in liver cancer patients, offering a potential avenue for improving patient selection and treatment outcomes.

In summary, the editors of his Research Topic anticipate that the collection of articles will illuminate the complex interactions among the various components within TME and explore the therapeutic potential of elements that regulate the metabolic ecology of the TME. Furthermore, a comprehensive understanding of the dynamic changes in immune metabolism will facilitate the identification of potential therapeutic targets and provide a solid theoretical foundation for the development of novel immunotherapies and metabolic regulation therapies.

Author contributions

DL: Writing – original draft. LZ: Writing – original draft. LT: Writing – review & editing. DT: Writing – review & editing.

Funding

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This work was supported by the National Natural Science Foundation of China (Grant No. 82103617 and 82103618) and the Scientific and Technological Project of Henan Province (Grant No. 222102310001 and 232102311024).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Vogel A, Meyer T, Sapisochin G, Salem R, Saborowski A. Hepatocellular carcinoma. Lancet. (2022) 400:1345–62. doi: 10.1016/S0140-6736(22)01200-4

PubMed Abstract | Crossref Full Text | Google Scholar

2. Leone RD, Powell JD. Metabolism of immune cells in cancer. Nat Rev Cancer. (2020) 20:516–31. doi: 10.1038/s41568-020-0273-y

PubMed Abstract | Crossref Full Text | Google Scholar

3. Kuczynski EA, Vermeulen PB, Pezzella F, Kerbel RS, Reynolds AR. Vessel co-option in cancer. Nat Rev Clin Oncol. (2019) 16:469–93. doi: 10.1038/s41571-019-0181-9

PubMed Abstract | Crossref Full Text | Google Scholar

4. Chen X, Cubillos-Ruiz JR. Endoplasmic reticulum stress signals in the tumour and its microenvironment. Nat Rev Cancer. (2021) 21:71–88. doi: 10.1038/s41568-020-00312-2

PubMed Abstract | Crossref Full Text | Google Scholar

5. Li Y, Chen J, Wang B, Xu Z, Wu C, Ma J, et al. FOXK2 affects cancer cell response to chemotherapy by promoting nucleotide de novo synthesis. Drug Resist Updat. (2023) 67:100926. doi: 10.1016/j.drup.2023.100926

PubMed Abstract | Crossref Full Text | Google Scholar

6. Chang J, Saraswathibhatla A, Song Z, Varma S, Sanchez C, Alyafei NHK, et al. Cell volume expansion and local contractility drive collective invasion of the basement membrane in breast cancer. Nat Mater. (2024) 23:711–22. doi: 10.1038/s41563-023-01716-9

PubMed Abstract | Crossref Full Text | Google Scholar

7. Wisdom KM, Indana D, Chou PE, Desai R, Kim T, Chaudhuri O. Covalent cross-linking of basement membrane-like matrices physically restricts invasive protrusions in breast cancer cells. Matrix Biol. (2020) 85-86:94–111. doi: 10.1016/j.matbio.2019.05.006

PubMed Abstract | Crossref Full Text | Google Scholar

8. Jones DS, Silverman AP, Cochran JR. Developing therapeutic proteins by engineering ligand-receptor interactions. Trends Biotechnol. (2008) 26:498–505. doi: 10.1016/j.tibtech.2008.05.0093

PubMed Abstract | Crossref Full Text | Google Scholar

9. Wu R, Guo W, Qiu X, Wang S, Sui C, Lian Q, et al. Comprehensive analysis of spatial architecture in primary liver cancer. Sci Adv. (2021) 7:eabg3750. doi: 10.1126/sciadv.abg3750

PubMed Abstract | Crossref Full Text | Google Scholar

10. Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med. (2020) 382:1894–905. doi: 10.1056/NEJMoa1915745

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: tumor microenvironment, vessel co-option, endoplasmic reticulum stress (ER stress), FOXK2, basement membrane-related genes (BMRGs), ligand-receptor interactions, UHPLC-MS

Citation: Zhang L, Tao L, Tian D and Li D (2024) Editorial: Immunometabolism and tumor microenvironment in hepatocellular carcinoma. Front. Oncol. 14:1483397. doi: 10.3389/fonc.2024.1483397

Received: 19 August 2024; Accepted: 26 August 2024;
Published: 10 September 2024.

Edited and Reviewed by:

Michael P. Lisanti, University of Salford, United Kingdom

Copyright © 2024 Zhang, Tao, Tian and Li. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Dongxiao Li, lidongxiao@zzu.edu.cn

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.