Skip to main content

EDITORIAL article

Front. Immunol., 31 January 2025
Sec. Cancer Immunity and Immunotherapy
This article is part of the Research Topic Mechanism Explorations of Enhancing Immunotherapeutic Sensitivity via Mediating Immune Infiltration and Programmed Cell Death in Solid Tumor Microenvironment View all 8 articles

Editorial: Mechanism explorations of enhancing immunotherapeutic sensitivity via mediating immune infiltration and programmed cell death in solid tumor microenvironment

Fang Zheng,,&#x;Fang Zheng1,2,3†Zhiliang Li&#x;Zhiliang Li1†Ruoyu Weng&#x;Ruoyu Weng1†Baochi Ou*Baochi Ou4*Tao Zhang*Tao Zhang5*Ren Zhao,,*Ren Zhao1,2,3*Haoran Feng,,*Haoran Feng1,2,3*
  • 1Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
  • 2Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
  • 3Institute of Medical Robotics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
  • 4Department of Surgery, First Affiliated Hospital, Anhui Medical University, Hefei, China
  • 5Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center (MSK), New York, NY, United States

Immunotherapy has become a research hotspot in the field of cancer therapy in recent years. As a therapy which works by activating and regulating the immune system to recognize and attack tumor cells, it shows significant clinical efficacy especially in some refractory tumors and results in fewer side effects. Despite its potential, immunotherapy still faces several challenges in clinical practice, with one of the widely focused issues being the individual variability of patient response (1). Tumor immune microenvironment (TIME) in solid tumor could be classified into two main types as either “high immunogenicity” or “low immunogenicity”, reflecting the immune infiltration pattern of tumor. The former ones typically exhibit better response to immunotherapy while the latter shows the opposite (2, 3). Therefore, regulating tumor immune infiltration to convert the TIME from “cold” to “hot” is considered a promising approach to enhance the efficacy of immunotherapy (46).

Checkpoint blockage therapy targeting PD-1/PD-L1 is a significant advancement in the field of cancer immunotherapy in recent years. However, multiple post-translational modifications have been found as protective factors of PD-1/PD-L1, contributing to the stability of protein and the prevention of protein degradation (7). Wang et al. reported the therapeutic enhancement mechanism of an active component in ginsenosides named Rg3 in non-small-cell lung cancer (NSCLC), which could upregulate PD-L1 N-glycosylation. It is noted that Rg3 strengthened the anti-tumor ability of T cells and augmented immune cytotoxicity of CD8+T cells. Based on their findings, the combination of Rg3 and immunotherapy is hopeful to enhance the anti-tumor immune response and benefit patients with NSCLC or other solid tumors. Some tumors are inherently resistant to immunotherapies targeting various popular checkpoints, and it is crucial to develop new specific therapeutic targets for these tumors. Tang et al. summarized recent studies about the utilization of single-cell RNA sequencing (scRNA-seq) in uveal melanoma (UM). The analyzed results indicate the presence of intratumoral heterogeneity (ITH), helping tumors cope with challenges. Certain researchers tended to reanalyze existing public data of scRNA-seq to develop advanced predictive models and identify biomarkers for prognosis prediction. In addition, scRNA-seq provided thorough exploration of the complex TME landscape, facilitating potential immunotherapy targets of UM such as LAG-3 for ICIs or specific markers of tumor-associated macrophages (TAMs). Even in the cancers with widespread use of immunotherapy, treatment efficacy varies between patients. Tuersun et al. applied scRNA-seq to analyze the features of certain colorectal cancer (CRC) patients who are more beneficial from neoadjuvant immunotherapy. They trained a novel prognosis model based on a risk score composed of 15 chemokines, performing better in predictive ability than traditional TNM staging. The result of scRNA-seq indicated the difference in tumor stemness between high and low response groups at the same time. CXCL10+ M1 macrophages, which enhance effector T cell migration and convert the TIME from “cold” to “hot,” are reasonable predictors of neoadjuvant therapy. Up to the overall level of TME, adjustment of the proportions of immune cell components also relates to the efficacy of immunotherapy. Ahn et al. focused on mitochondria regulation as enhancement of tumor immunotherapy. The hypoxic glycolysis state in TME refers to mitochondrial metabolic reprogramming, destroying the function of anti-tumor immune cells. However, various immunosuppressive cells develop unique metabolic pathways to adapt the circumstances. Consequently, mitochondrial-targeting drugs, such as metformin and statins, in combination with immunotherapy can regulate the energy metabolism within TME and establish an environment conducive to the activation of anti-tumor immune cells. TLS-TIME is a specific subclass of high immunogenicity TIME, which is correlated to positive prognosis in solid tumor mostly. Yang et al. consolidated potential relationship between gut microbiota and tertiary lymphoid structures (TLSs) in TME. Regarded as significant sites for the activation of adaptive immune responses in the tumor periphery, TLSs are commonly observed in chronic inflammation microenvironment, including the persistent chronic inflammatory status induced by gut microbiota. Regulation on gut microbiome is hopeful to become emerging strategies for modulating the TIME mode by inducing mature TLSs in solid cancer.

Beyond the above, programmed cell death (PCD) is also closely related to the outcome of immunotherapy. Serving as a planned cell elimination tightly regulated by intricate molecular pathways, it contributes to maintaining physiological homeostasis. PCD could either be activated by immunogenic factors, or adjuvant anti-tumor immunity (8). It is known as PCD could be induced by chemotherapy and radiation, and their combination with immunotherapy leads to enhanced therapeutic effect. Studies have reported that direct administration of gasdermin agonist or other PCD-inducing agents may improve the efficacy of cancer immunotherapy similarly (911).

Exploration of PCD targets and pathways is essential for upgrading current therapeutic approaches and development of novel ones. Huang et al. tracked research hotspots of TGF-β activated kinase 1 (TAK1) by using bibliometric and visualized analysis. TAK1 is involved in the regulation of TNF, IL-1 and TGF-β pathways, which all related to immune response and regulated cell death. Meanwhile, the lack of TAK1 could trigger PANoptosis, a specific inflammatory PCD combining key features of pyroptosis, apoptosis and necroptosis. The involvement of TAK1 in stages of tumorigenesis, progression and treatment renders it critical research value and promising future prospects. Despite theoretical investigation in laboratories, PCD plays a practical role in clinical treatment. Zheng et al. provided a review with the theme of combination of immune checkpoint inhibitors (ICIs) and carbon ion radiotherapy (CIRT) in renal cell carcinoma (RCC). CIRT possesses enhanced physical and biological characters relative to conventional photon radiation, ensuring accurate complete destruction of cancer cells with maximal protection of healthy tissues. With the co-administration of CIRT and ICIs, TIME will be reshaped into a model with enriched immune infiltration. Moreover, CIRT can trigger immunogenic cell death, a type of PCD following with continuous immune response, to recruit immune cells and stimulate the release of pro-inflammatory cytokines.

In conclusion, enhancing sensitivity has emerges as the central topic in the field of immunotherapy therapy recently. The regulation of either immune infiltration or PCD represents a promising strategy. We deeply appreciate the invaluable contributions of all authors, reviewers, and the editorial team throughout the preparation and review of this topic. With the innovation of technology, several novel approaches to enhance immunotherapy have emerged in recent years, such as designing drug delivery systems by nanomaterials and modifying T cells through CRISPR/Cas9 genome editing technology (12, 13). We are optimistic that future progress in this field will provide additional treatment options, enriching the comprehensive treatment plans for cancer patients.

Author contributions

FZ: Writing – original draft. ZL: Writing – original draft. RW: Writing – original draft. BO: Writing – review & editing. TZ: Writing – review & editing. RZ: Writing – review & editing. HF: Writing – review & editing.

Funding

The author(s) declare that financial support was received for the research, authorship, and/or publication of this article. This work was supported by National Natural Science Foundation of China, 82473158 (HF) and 82271766 (RZ).

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. Sun Q, Hong Z, Zhang C, Wang L, Han Z, Ma D. Immune checkpoint therapy for solid tumours: clinical dilemmas and future trends. Signal Transduct Target Ther. (2023) 8:320. doi: 10.1038/s41392-023-01522-4

PubMed Abstract | Crossref Full Text | Google Scholar

2. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. (2018) 24:541–50. doi: 10.1038/s41591-018-0014-x

PubMed Abstract | Crossref Full Text | Google Scholar

3. Ali HR, Chlon L, Pharoah PD, Markowetz F, Caldas C. Patterns of immune infiltration in breast cancer and their clinical implications: A gene-expression-based retrospective study. PloS Med. (2016) 13:e1002194. doi: 10.1371/journal.pmed.1002194

PubMed Abstract | Crossref Full Text | Google Scholar

4. Wang DR, Wu XL, Sun YL. Therapeutic targets and biomarkers of tumor immunotherapy: response versus non-response. Signal Transduct Target Ther. (2022) 7:331. doi: 10.1038/s41392-022-01136-2

PubMed Abstract | Crossref Full Text | Google Scholar

5. Tang T, Huang X, Zhang G, Hong Z, Bai X, Liang T. Advantages of targeting the tumor immune microenvironment over blocking immune checkpoint in cancer immunotherapy. Signal Transduct Target Ther. (2021) 6:72. doi: 10.1038/s41392-020-00449-4

PubMed Abstract | Crossref Full Text | Google Scholar

6. Han C, Zhang A, Liu Z, Moore C, Fu YX. Small molecular drugs reshape tumor microenvironment to synergize with immunotherapy. Oncogene. (2021) 40:885–98. doi: 10.1038/s41388-020-01575-7

PubMed Abstract | Crossref Full Text | Google Scholar

7. Hsu JM, Li CW, Lai YJ, Hung MC. Posttranslational modifications of PD-L1 and their applications in cancer therapy. Cancer Res. (2018) 78:6349–53. doi: 10.1158/0008-5472.Can-18-1892

PubMed Abstract | Crossref Full Text | Google Scholar

8. Liu J, Hong M, Li Y, Chen D, Wu Y, Hu Y. Programmed cell death tunes tumor immunity. Front Immunol. (2022) 13:847345. doi: 10.3389/fimmu.2022.847345

PubMed Abstract | Crossref Full Text | Google Scholar

9. Zhao L, Zhou X, Xie F, Zhang L, Yan H, Huang J, et al. Ferroptosis in cancer and cancer immunotherapy. Cancer Commun (Lond). (2022) 42:88–116. doi: 10.1002/cac2.12250

PubMed Abstract | Crossref Full Text | Google Scholar

10. Hsu SK, Li CY, Lin IL, Syue WJ, Chen YF, Cheng KC, et al. Inflammation-related pyroptosis, a novel programmed cell death pathway, and its crosstalk with immune therapy in cancer treatment. Theranostics. (2021) 11:8813–35. doi: 10.7150/thno.62521

PubMed Abstract | Crossref Full Text | Google Scholar

11. Wang Q, Wang Y, Ding J, Wang C, Zhou X, Gao W, et al. A bioorthogonal system reveals antitumour immune function of pyroptosis. Nature. (2020) 579:421–6. doi: 10.1038/s41586-020-2079-1

PubMed Abstract | Crossref Full Text | Google Scholar

12. Wang SW, Gao C, Zheng YM, Yi L, Lu JC, Huang XY, et al. Current applications and future perspective of CRISPR/Cas9 gene editing in cancer. Mol Cancer. (2022) 21:57. doi: 10.1186/s12943-022-01518-8

PubMed Abstract | Crossref Full Text | Google Scholar

13. Riley RS, June CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discovery. (2019) 18:175–96. doi: 10.1038/s41573-018-0006-z

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: immunotherapy, immune infiltration, programmed cell death, tumor microenvironment, solid cancer

Citation: Zheng F, Li Z, Weng R, Ou B, Zhang T, Zhao R and Feng H (2025) Editorial: Mechanism explorations of enhancing immunotherapeutic sensitivity via mediating immune infiltration and programmed cell death in solid tumor microenvironment. Front. Immunol. 16:1559657. doi: 10.3389/fimmu.2025.1559657

Received: 13 January 2025; Accepted: 20 January 2025;
Published: 31 January 2025.

Edited and Reviewed by:

Peter Brossart, University of Bonn, Germany

Copyright © 2025 Zheng, Li, Weng, Ou, Zhang, Zhao and Feng. 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: Baochi Ou, b3ViYW9jaGlAMTYzLmNvbQ==; Tao Zhang, emhhbmd0NUBtc2tjYy5vcmc=; Ren Zhao, cmp6aGFvcmVuQDEzOS5jb20=; Haoran Feng, d2FuZ3lpX2ZlbmdoYW9yYW5AMTYzLmNvbQ==

These authors have contributed equally to this work

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.