Skip to main content

EDITORIAL article

Front. Immunol., 05 December 2023
Sec. Cancer Immunity and Immunotherapy
This article is part of the Research Topic The Role of Radiotherapy in Reshaping Tumor Immune Microenvironment View all 5 articles

Editorial: The role of radiotherapy in reshaping tumor immune microenvironment

Xiaoyu Hu&#x;Xiaoyu HuMengyu Zhao&#x;Mengyu ZhaoJinbo Yue*Jinbo Yue*
  • Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China

Immunotherapy, represented by immune checkpoint inhibitors (ICIs), has improved outcomes in several types of cancers. However, many patients still do not respond (1). The anti-tumor response mediated by ICIs depends on the infiltration of cytotoxic T lymphocytes (CTLs) that can recognize and kill tumor cells (2). Tumors can be classified into three primary immune phenotypes based on the spatial distribution of CTLs within the tumor microenvironment (TME): these are the immunoinflammatory phenotype, the immunoexclusionary phenotype, and the immune-desert phenotype (1). Immunoinflammatory tumors, a.k.a “hot tumors”, are characterized by substantial T cell infiltration, amplified IFNγ signaling, and a high tumor mutation burden (TMB). These tumors typically show increased sensitivity to ICIs (3). Conversely, immunoexclusionary and immune-desert tumors, which have less immune activity, are often termed “cold tumors”.

Radiotherapy (RT) can not only directly destroy tumors but also induce a systemic immune response. Radiation induces immunogenic death in tumor cells, activates anti-tumor immune responses, and produces an effect similar to an “in situ vaccine” (4). Moreover, RT has a multifaceted regulatory effect on immune cells and tumor stroma cells. It can also influence the TME by inducing production of various cytokines, ultimately transforming cold tumors into hot and enhancing the efficacy of immunotherapy (5). RT can reshape the immune TME from multiple perspectives, although its specific mechanisms have not been fully elucidated.

In this Research Topic, multiple studies investigate the role of RT in remodeling the immune TME. Gao et al. have identified three autophagy-related genes—SHC1, NAPSA, and AURKA—associated with RT and the prognosis of lung adenocarcinoma. NAPSA and AURKA were also closely associated with TH2 cell infiltration. TH2 cells promote tumor activity by facilitating angiogenesis and inhibiting the cell-mediated immune response that destroys tumor cells. Accumulating evidence suggests that autophagic activity can regulate immune cell infiltration by modulating both the innate and adaptive immune systems. Therefore, it is possible that autophagy-related genes may inhibit antitumor immunity by promoting TH2 cell infiltration following RT. Hence, targeting autophagy-related genes could represent a novel immunomodulatory strategy to enhance the effectiveness of RT in patients with lung adenocarcinoma.

Shi et al. provide a summary of radioimmunotherapy in the treatment of colorectal cancer, discussing its current status and future prospects. Patients with microsatellite instability (MSI) comprise less than 5% of rectal cancer cases, while the majority of colorectal cancers are of the microsatellite stable (MSS) type. These MSS patients derive little-to-no benefit from immunotherapy alone. Due to the synergistic effect between RT and immunotherapy, RT can release tumor antigens, reshape the immune microenvironment, and increase the anti-tumor immune response, thereby producing a synergistic therapeutic effect. Numerous prospective phase II studies suggest that the combination of RT with immunotherapy, used for pMMR/MSS locally advanced rectal cancer, has achieved a higher rate of complete pathological response (pCR) (6).

Chi et al. show that hRT’s regulation of the immune microenvironment depends on the dosage and fractionation pattern of the RT. RT can potentiate antigen presentation, leading to the upregulation of pro-inflammatory cytokine levels and cytotoxic T cell-mediated immunogenic cell death. However, RT can also induce immunosuppressive responses, characterized by the augmented recruitment and infiltration of regulatory T cells, myeloid-derived suppressor cells (MDSCs), and molecules associated with immune checkpoints. Combining RT with immunotherapy may shift this balance, tipping it towards immune activation. This combination could potentially overcome resistance to ICIs and other forms of immunotherapy. Strategies to enhance the efficacy of such combinations warrant further exploration.

In recent years, numerous in vivo studies have revealed the synergistic tumoricidal effects of combining RT with PD-1/-L1 inhibitors. The dynamic nature of the PD-1/-L1 axis, however, has confounded efforts towards a consensus on the optimal timing or dose-fractionation strategy for doing so (6). Understanding the synergistic effects of combining RT with PD-1/PD-L1 blockade requires investigation into the dynamic changes in tumor immune status and circulating biomarkers induced by RT in relation to PD-1/PD-L1 expression. Yoon et al. demonstrate that, following RT, PD-L1 expression levels in tumor cells and intratumoral MDSCs, TAMs, and DCs are upregulated, mainly during the period of tumor regression. With tumor regrowth, PD-L1 levels in tumor cells were found to decrease to levels comparable to those at baseline.

Our understanding of how RT can reshape the immune TME across various levels has continued to grow, informed by a legion of molecular and animal studies as well as clinical trials. As research in this area progresses, it is anticipated that the confluence of RT and immunotherapy will yield increasingly potent therapeutic interactions.

Author contributions

XH: Writing – original draft. MZ: Writing – original draft. JY: Writing – review & editing.

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. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature (2017) 541(7637):321–30. doi: 10.1038/nature21349

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Bruni D, Angell HK, Galon J. The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy. Nat Rev Cancer (2020) 20(11):662–80. doi: 10.1038/s41568-020-0285-7

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Hegde PS, Karanikas V, Evers S. The where, the when, and the how of immune monitoring for cancer immunotherapies in the era of checkpoint inhibition. Clin Cancer Res (2016) 22(8):1865–74. doi: 10.1158/1078-0432.CCR-15-1507

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Goto T. Radiation as an in situ auto-vaccination: current perspectives and challenges. Vaccines (Basel) (2019) 7(3):100. doi: 10.3390/vaccines7030100

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Sharabi AB, Lim M, DeWeese TL, Drake CG. Radiation and checkpoint blockade immunotherapy: radiosensitisation and potential mechanisms of synergy. Lancet Oncol (2015) 16(13):e498–509. doi: 10.1016/S1470-2045(15)00007-8

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Xiao W, Luo H, Yao Y, Wang Y, Liu S, Sun R, et al. Total neoadjuvant treatment and PD-1/PD-L1 checkpoint inhibitor in locally advanced rectal cancer. Front Immunol (2023) 14:1149122. doi: 10.3389/fimmu.2023.1149122

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: radiotherapy, immunotherapy, immune microenvironment, autophagy, colorectal cancer immunotherapy, represented by immune checkpoint inhibitors (ICIs)

Citation: Hu X, Zhao M and Yue J (2023) Editorial: The role of radiotherapy in reshaping tumor immune microenvironment. Front. Immunol. 14:1340844. doi: 10.3389/fimmu.2023.1340844

Received: 19 November 2023; Accepted: 28 November 2023;
Published: 05 December 2023.

Edited and Reviewed by:

Vito Longo, National Cancer Institute Foundation (IRCCS), Italy

Copyright © 2023 Hu, Zhao and Yue. 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: Jinbo Yue, jbyue@sdfmu.edu.cn

These authors have contributed equally to this work and share first authorship

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.