- 1Department of Cardiopulmonary Rehabilitation, Nanjing Zijin Hospital, Nanjing, China
- 2Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
- 3Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
Editorial on the Research Topic
Targeting cancer-associated fibroblasts: disrupting immune evasion and therapy resistance
The tumor microenvironment (TME) is a complex ecosystem that plays a critical role in cancer progression and therapeutic response (1). Within this network, cancer-associated fibroblasts (CAFs) have emerged as master regulators, and they critically shape an immunosuppressive and therapy-resistant niche in TME (2). In the past, CAFs were merely considered as static structural components. But now, CAFs have been recognized as functionally heterogeneous and metabolically active players that promote tumorigenesis through diverse mechanisms such as extracellular matrix (ECM) remodeling, paracrine signaling, and metabolic reprogramming (3). Their profound impact on anti-tumor immunity and chemotherapeutic efficacy makes them as a promising therapeutic target for overcoming treatment failure in multiple cancer types.
This Research Topic, “Targeting Cancer-Associated Fibroblasts: Disrupting Immune Evasion and Therapy Resistance,” was established to compile cutting-edge research and insightful reviews that elucidate the multifaceted roles of CAFs and explore innovative strategies to disrupt the pro-tumorigenic functions of CAFs in different cancer types including esophageal squamous cell carcinoma (ESCC), lung cancer, osteosarcoma, hepatocellular carcinoma (HCC), clear cell renal cell carcinoma (ccRCC), and others—and highlight promising strategies to target these stromal players in cancer therapy.
This Research Topic comprises nine articles, including two original research articles and seven reviews, which systematically elucidate the biology of CAFs across various malignancies. Gao et al. demonstrated that high expression of APE1 in ESCC is associated with increased infiltration of FOXP3+ regulatory T cells (Tregs) and CAFs, correlating with poor survival following adjuvant chemotherapy. This study not only identifies APE1 as a prognostic biomarker but also links it to CAF-mediated immunosuppression, suggesting that stromal-immune crosstalk is a key mechanism of chemoresistance. Similarly, the influence of the stromal environment on tumor behavior is evident in the work of Maeda et al., who showed that an interstitial pneumonia (IP) lung microenvironment promotes metastasis to mediastinal lymph nodes and contralateral lungs in a murine model. This pro-metastatic niche was mitigated by pirfenidone, an anti-fibrotic agent, pointing to the therapeutic potential of modulating the stromal compartment to impede cancer progression. The immunosuppressive functions of CAFs are mediated through diverse mechanisms, including cytokine secretion, metabolic reprogramming, extracellular matrix (ECM) remodeling, and exosome-mediated communication. Luo et al. and Wang et al. elaborate on how CAFs engage in dynamic crosstalk with immune cells—such as T cells, macrophages, and Tregs—to establish an immune-excluded or immune-suppressed TME. These interactions often involve key signaling pathways like TGF-β, IL-6/STAT3, and CXCR4, which not only support tumor survival but also blunt the efficacy of immune checkpoint inhibitors (ICIs). In hepatocellular carcinoma, Shi et al. describe the cellular origins and functional heterogeneity of CAFs, emphasizing their role in immune evasion and chemoresistance. The complexity of CAF subsets—each with distinct marker expression and functional profiles—complicates therapeutic targeting but also offers opportunities for precision medicine. A common theme across these studies is the role of fibroblast activation protein (FAP), a surface protease highly expressed on a subset of CAFs. Lee et al. and Wang et al. discuss FAP as a promising therapeutic target. FAP-positive CAFs contribute to ECM remodeling, angiogenesis, and immune suppression, making them attractive for interventions such as FAP-targeted CAR-T cells, antibody-drug conjugates, or small-molecule inhibitors. However, the heterogeneity of CAF populations and the lack of specific biomarkers remain significant challenges for clinical translation. In addition, nanotechnology offers a promising avenue to overcome these hurdles. Xu et al. review how nano-strategies can deliver CAF-modulating agents—such as cytotoxic drugs, signaling inhibitors, or phenotype-switching molecules—with enhanced specificity and reduced systemic toxicity. Combining CAF-targeted nanotherapies with ICIs has shown synergistic effects in preclinical models, revitalizing T cell infiltration and antitumor immunity.
In osteosarcoma, Fan et al. highlight CAF-driven resistance mechanisms, including exosome-mediated gene regulation, metabolic reprogramming, and inhibition of ferroptosis. They also discuss emerging CAF-directed therapies, such as TGF-β blockade, FAP depletion, and CAF-reprogramming approaches, which may be combined with conventional chemotherapy or immunotherapy to overcome resistance.
Despite these advances, several challenges still exist. CAF plasticity, functional heterogeneity, and the dual roles of certain CAF subsets—sometimes even exerting tumor-restraining effects—complicate therapeutic targeting. Moreover, the translational gap between preclinical experiments and human trials remains wide (4). As Wang et al. point out, future efforts should integrate single-cell sequencing and spatial multi-omics to decipher the spatiotemporal dynamics of CAF subpopulations and identify context-dependent vulnerabilities, and develop precision treatment strategies based on molecular subtyping to accelerate clinical application.
In conclusion, the collective evidence from these nine articles convincingly establishes CAFs as pivotal regulators of immune evasion and therapy resistance across multiple cancer types. Targeting CAFs—whether through depletion, reprogramming, or disruption of their signaling networks—holds great promise for enhancing the efficacy of existing therapies, particularly immunotherapies. However, achieving success will necessitate a thorough grasp of CAF biology, reliable biomarkers, and innovative combination strategies (5). As we move forward, a multidisciplinary approach that integrates stromal targeting with immune modulation and precision oncology will be essential to disrupt the resilient CAF-TME axis and improve outcomes for cancer patients.
Author contributions
TW: Writing – original draft. ZZ: Writing – original draft. QL: Writing – review & editing.
Acknowledgments
We sincerely thank all the authors who contributed to this Research Topic.
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 Generative AI was used in the creation of this manuscript. The present manuscript was composed independently by the authors; no generative artificial intelligence system was employed in the conception, design, analysis, interpretation, or drafting of its substantive content. The sole intervention consisted of post-composition language refinement using DeepSeek, which was limited to grammatical and language polishing.
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.
References
1. El-Tanani M, Rabbani SA, Babiker R, Rangraze I, Kapre S, Palakurthi SS, et al. Unraveling the tumor microenvironment: Insights into cancer metastasis and therapeutic strategies. Cancer Lett. (2024) 591:216894. doi: 10.1016/j.canlet.2024.216894
2. Chhabra Y and Weeraratna AT. Fibroblasts in cancer: Unity in heterogeneity. Cell. (2023) 186:1580–609. doi: 10.1016/j.cell.2023.03.016
3. Li Y, Liu Q, Jing X, Wang Y, Jia X, Yang X, et al. Cancer-associated fibroblasts: heterogeneity, cancer pathogenesis, and therapeutic targets. MedComm (2020). (2025) 6:e70292. doi: 10.1002/mco2.70292
4. Caligiuri G and Tuveson DA. Activated fibroblasts in cancer: Perspectives and challenges. Cancer Cell. (2023) 41:434–49. doi: 10.1016/j.ccell.2023.02.015
Keywords: cancer-associate fibroblasts, immune evasion, therapy resistance, tumor micro environment (TME), extracellular matrix, targeted therapy
Citation: Wang T, Zhou Z and Lu Q (2025) Editorial: Targeting cancer-associated fibroblasts: disrupting immune evasion and therapy resistance. Front. Immunol. 16:1728782. doi: 10.3389/fimmu.2025.1728782
Received: 20 October 2025; Accepted: 31 October 2025;
Published: 17 November 2025.
Edited and reviewed by:
Peter Brossart, University of Bonn, GermanyCopyright © 2025 Wang, Zhou and Lu. 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: Qiong Lu, Y2hyaXN0eV9sdXFAY3N1LmVkdS5jbg==
†These authors have contributed equally to this work
Ting Wang1†