Your new experience awaits. Try the new design now and help us make it even better

EDITORIAL article

Front. Immunol., 13 November 2025

Sec. Cancer Immunity and Immunotherapy

Volume 16 - 2025 | https://doi.org/10.3389/fimmu.2025.1737187

This article is part of the Research TopicAdvances in myeloid cell targeting for overcoming tumor immunosuppressionView all 6 articles

Editorial: Advances in myeloid cell targeting for overcoming tumor immunosuppression

  • 1Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
  • 2School of Medicine, South China University of Technology, Guangzhou, China
  • 3School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China

This Research Topic, Advances in myeloid cell targeting for overcoming tumor immunosuppression, comprises one comprehensive review and three original research articles, which collectively explore recent advances in myeloid cell targeting approaches, including nanoparticle-based siRNA and engineered oncolytic viruses. Mechanistically, these new approaches lead to the reprogramming of myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs), thereby enhancing anti-tumor immunity. Moreover, imaging-based biomarkers like imaging-based myeloid response score (iMRS) show promise for patient stratification. Despite progress, challenges remain in overcoming myeloid plasticity, toxicity, and developing reliable biomarkers for effective combination therapies.

Lang et al. developed peptide-based nanoparticles (NPc-Rel) to deliver siRNA targeting the transcription factor c-Rel, a myeloid immune checkpoint, in MDSCs. NPc-Rel effectively silenced c-Rel expression, reduced MDSC numbers and immunosuppressive function, and enhanced CD8+ T cell activity in the tumor microenvironment. In a B16 melanoma mouse model, this strategy significantly inhibited tumor growth, demonstrating that targeting myeloid c-Rel with siRNA nanoparticles is a promising immunotherapy approach to overcome tumor immunosuppression.

Jennings et al. demonstrated that oncolytic rhabdoviruses (ORVs) armed with microRNAs can reprogram TAMs via tumor-derived extracellular vesicles (TDEVs). ORV infection enhanced TDEV production, which was preferentially taken up by myeloid cells. ORVs expressing miR-155 or miR-19a delivered these miRNAs to TAMs, reversing their immunosuppressive function and enhancing T cell activity. While ORV-miR-155 improved antigen presentation and T cell responses, ORV-miR-19a significantly prolonged survival in a murine ovarian cancer model. This approach effectively reprograms the immunosuppressive tumor microenvironment, enhancing the efficacy of oncolytic virotherapy.

Peng et al. developed a noninvasive iMRS using preoperative CT radiomics to predict the myeloid immune contexture in hepatocellular carcinoma (HCC). A multi-phase CT radiomics signature accurately predicted the pathological MRS, which reflects the balance of pro- and anti-tumor myeloid cells. A high iMRS was significantly associated with poor post-surgical overall and recurrence-free survival. Furthermore, in an immunotherapy cohort, a high iMRS predicted a better objective response and improved survival for patients treated with anti-PD-1/PD-L1 therapy, demonstrating its value as a noninvasive biomarker for prognosis and immunotherapy benefit assessment.

The review by Chen et al. summarizes the potential of targeting myeloid cells to overcome immunosuppression in the tumor microenvironment (TME) and enhance cancer immunotherapy. Myeloid populations—such as TAMs, MDSCs, and tumor-associated dendritic cells—promote immune evasion through cytokine secretion, metabolic reprogramming, immune checkpoint expression, and physical exclusion of T cells. The article highlights key therapeutic targets, including CSF1R, PI3Kγ, mTOR, Syk, MerTK/Axl, and immune checkpoints like TREM2, LILRBs, VISTA, and CD40. Preclinical and clinical evidence supports strategies to deplete or reprogram these cells, restoring anti-tumor immunity. Combining myeloid-targeted agents with checkpoint inhibitors, chemotherapy, or radiation shows synergistic promise. However, challenges such as myeloid plasticity, toxicity, and biomarker development remain. Future success hinges on personalized approaches and rational combination therapies.

Future research directions in targeting myeloid cells to overcome tumor immunosuppression will focus on several key areas. A major priority is the development of more selective agents and delivery systems, such as nanoparticles or bispecific antibodies, to precisely modulate specific myeloid subpopulations like TAMs or MDSCs without systemic toxicity. Understanding and targeting the metabolic and epigenetic reprogramming that sustains the immunosuppressive functions of these cells represent another critical avenue. Furthermore, identifying robust predictive biomarkers—through single-cell sequencing or spatial transcriptomics—is essential for patient stratification and for monitoring therapy-induced changes in the myeloid compartment in real-time. Finally, a central challenge is the rational design of combination therapies. This involves integrating myeloid-targeting agents with existing immunotherapies (e.g., checkpoint inhibitors, CAR-T), conventional treatments, or novel modalities like STING agonists to achieve synergistic efficacy, reverse resistance, and create a durable anti-tumor immune response in a wider range of cancers.

Author contributions

DY: Writing – original draft, Writing – review & editing. QY: Writing – original draft, Writing – review & editing. JZ: Writing – original draft, Writing – review & editing.

Acknowledgments

We would like to express our sincere thanks to all the authors who submitted their studies to the present Research. Topic and the reviewers who evaluated the manuscripts. Their efforts help us to communicate current knowledge under this topic to a worldwide audience.

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.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

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.

Keywords: myeloid cells, tumor immunosuppression, immunotherapy resistance, cancer treatment, myeloid-derived suppressor cells (MDSCs)

Citation: Yan D, Yang Q and Zhou J (2025) Editorial: Advances in myeloid cell targeting for overcoming tumor immunosuppression. Front. Immunol. 16:1737187. doi: 10.3389/fimmu.2025.1737187

Received: 01 November 2025; Accepted: 04 November 2025;
Published: 13 November 2025.

Edited and reviewed by:

Peter Brossart, University of Bonn, Germany

Copyright © 2025 Yan, Yang and Zhou. 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: Dehong Yan, ZGgueWFuQHNpYXQuYWMuY24=; Qiong Yang, eWFuZ3FAc2N1dC5lZHUuY24=; Jingying Zhou, emhvdWp5QGN1aGsuZWR1Lmhr

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.