1
|
Sun Y, Liu JQ, Chen WJ, Tang WF, Zhou YL, Liu BJ, Wei Y, Dong JC. Astragaloside III inhibits MAPK-mediated M2 tumor-associated macrophages to suppress the progression of lung Cancer cells via Akt/mTOR signaling pathway. Int Immunopharmacol 2025; 154:114546. [PMID: 40184811 DOI: 10.1016/j.intimp.2025.114546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/25/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025]
Abstract
Tumor-associated macrophages (TAMs) play a key role in facilitating a range of cancerous processes by modulating the tumor microenvironment thus being a target for cancer treatment. Astragaloside III (AS-III), a compound derived from Astragalus triterpenoid saponins, has demonstrated immunomodulatory and anticancer properties, but the underlying mechanism remains unclear. Here, we demonstrated that AS-III suppressed metastasis, angiogenesis and induced apoptosis of lung cancer in vitro and in vivo by inhibiting macrophage M2 polarization and inducing M1 phenotype transformation. This was achieved through the inhibition of the MAPK signaling pathway. Furthermore, the tumor inhibitory effects of AS-III were found to be mediated by the Akt/mTOR pathway. Overall, these results highlight the role of AS-III in modifying the TAMs in TME, offering fresh perspectives on tumor immunotherapy by means of targeting macrophage.
Collapse
Affiliation(s)
- Yan Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Jia-Qi Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Wen-Jing Chen
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Wei-Feng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yao-Long Zhou
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Bao-Jun Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China.
| | - Jing-Cheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
An Y, Ji C, Zhang H, Jiang Q, Maitz MF, Pan J, Luo R, Wang Y. Engineered Cell Membrane Coating Technologies for Biomedical Applications: From Nanoscale to Macroscale. ACS NANO 2025; 19:11517-11546. [PMID: 40126356 DOI: 10.1021/acsnano.4c16280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Cell membrane coating has emerged as a promising strategy for the surface modification of biomaterials with biological membranes, serving as a cloak that can carry more functions. The cloaked biomaterials inherit diverse intrinsic biofunctions derived from different cell sources, including enhanced biocompatibility, immunity evasion, specific targeting capacity, and immune regulation of the regenerative microenvironment. The intrinsic characteristics of biomimicry and biointerfacing have demonstrated the versatility of cell membrane coating technology on a variety of biomaterials, thus, furthering the research into a wide range of biomedical applications and clinical translation. Here, the preparation of cell membrane coatings is emphasized, and different sizes of coated biomaterials from nanoscale to macroscale as well as the engineering strategies to introduce additional biofunctions are summarized. Subsequently, the utilization of biomimetic membrane-cloaked biomaterials in biomedical applications is discussed, including drug delivery, imaging and phototherapy, cancer immunotherapy, anti-infection and detoxification, and implant modification. In conclusion, the latest advancements in clinical and preclinical studies, along with the multiple benefits of cell membrane-coated nanoparticles (NPs) in biomimetic systems, are elucidated.
Collapse
Affiliation(s)
- Yongqi An
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Cheng Ji
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
- College of Materials Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Hao Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Qing Jiang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Manfred F Maitz
- Max Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research Dresden, Dresden 01069, Germany
| | - Junqiang Pan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
- Department of Cardiovascular Medicine, Xi'an Central Hospital, Xi'an 710003, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
- Research Unit. of Minimally Invasive Treatment of Structural Heart-Disease, Chinese Academy of Medical Sciences (2021RU013), Chengdu, 610065, China
| |
Collapse
|
3
|
Mathiesen H, Juul-Madsen K, Tramm T, Vorup-Jensen T, Møller HJ, Etzerodt A, Andersen MN. Prognostic value of CD163 + macrophages in solid tumor malignancies: A scoping review. Immunol Lett 2025; 272:106970. [PMID: 39778658 DOI: 10.1016/j.imlet.2025.106970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/19/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
Tumor-associated macrophages (TAMs) play crucial roles in development and progression of malignant diseases. Notably, CD163+ TAMs likely perform specific pro-tumorigenic functions, suggesting that this subset may serve as both prognostic biomarkers and targets for future anti-cancer therapy. We conducted a scoping review to map the current knowledge on the prognostic role of CD163+ TAMs in the five most lethal cancers worldwide: Lung, colorectal, gastric, liver, and breast cancer. For all cancer types, most studies showed that high tumoral presence of CD163+ cells was associated with poor patient outcome, and this association was more frequently observed when CD163+ cells were measured at the tumor periphery compared to more central parts of the tumor. These results support that CD163+ TAMs represent a biomarker of poor patient outcome across a variety of solid tumors, and highlight the relevance of further investigations of CD163+ TAMs as targets of future immunotherapies.
Collapse
Affiliation(s)
- Henriette Mathiesen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Kristian Juul-Madsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Trine Tramm
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Holger Jon Møller
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Morten Nørgaard Andersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Hematology, Aarhus University Hospital, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
4
|
Whitman MA, Mantri M, Spanos E, Estroff LA, De Vlaminck I, Fischbach C. Bone mineral density affects tumor growth by shaping microenvironmental heterogeneity. Biomaterials 2025; 315:122916. [PMID: 39490060 DOI: 10.1016/j.biomaterials.2024.122916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/09/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Breast cancer bone metastasis is a major cause of mortality in patients with advanced breast cancer. Although decreased mineral density is a known risk factor for bone metastasis, the underlying mechanisms remain poorly understood because studying the isolated effect of bone mineral density on tumor heterogeneity is challenging with conventional approaches. Moreover, mineralized biomaterials are commonly utilized for clinical bone defect repair, but how mineralized biomaterials affect the foreign body response and wound healing is unclear. Here, we investigate how bone mineral affects tumor growth and microenvironmental complexity in vivo by combining single-cell RNA-sequencing with mineral-containing or mineral-free decellularized bone matrices. We discover that the absence of bone mineral significantly influences fibroblast and immune cell heterogeneity, promoting phenotypes that increase tumor growth and alter the response to injury or disease. Importantly, we observe that the stromal response to bone mineral content depends on the murine tumor model used. While lack of bone mineral induces tumor-promoting microenvironments in both immunocompromised and immunocompetent animals, these changes are mediated by altered fibroblast phenotype in immunocompromised mice and macrophage polarization in immunocompetent mice. Collectively, our findings suggest that bone mineral density affects tumor growth by impacting microenvironmental complexity in an organism-dependent manner.
Collapse
Affiliation(s)
- Matthew A Whitman
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Madhav Mantri
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Emmanuel Spanos
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14850, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, 14850, USA
| | - Iwijn De Vlaminck
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA.
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, 14850, USA.
| |
Collapse
|
5
|
Kim D, Allen CA, Chung D, Meng L, Zhang X, Zhang W, Ouyang Y, Li Z, Hong F. A novel TLR4 accessory molecule drives hepatic oncogenesis through tumor-associated macrophages. Cancer Lett 2025; 614:217543. [PMID: 39929433 DOI: 10.1016/j.canlet.2025.217543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/28/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Tumor-associated macrophages (TAMs) play a crucial role in the tumor microenvironment, yet the roles and mechanisms of TAMs in inflammation-associated oncogenesis remain enigmatic. We report that protein canopy homolog 2 (CNPY2) functions as a novel TLR4 regulator, promoting cytokine production in macrophages. CNPY2 binds directly to TLR4. Cnpy2 deficiency reduces cell surface expression of TLR4, nuclear translocation of NFκB and cytokine production in macrophages. Macrophage-specific CNPY2 deficiency significantly decreases cytokine production in macrophages and reduces hepatocarcinogenesis in a diethylnitrosamine (DEN)-induced liver cancer model. RNA-sequencing analysis revealed Cnpy2 knockout decreased the mRNA level and cell surface expression of two VEGF receptors, Flt1 and Kdr, compared to those in WT counterparts, resulting in inhibition of macrophage tumor infiltration. Cnpy2 knockout inhibits NFκB2/p52-mediated transcription of Flt1 and Kdr in macrophages. These findings demonstrate that CNPY2 regulates macrophages in both inflammation and hepatocarcinogenesis and may serve as a therapeutic target for cancer.
Collapse
Affiliation(s)
- Doyeon Kim
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Carter A Allen
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Dongjun Chung
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Lingbin Meng
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Xiaoli Zhang
- Biostatistics Core, College of Nursing, College of Public Health, University of South Florida Health, 12901 Bruce B. Downs Blvd.Tampa, FL, 33612, USA
| | - Wenqing Zhang
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Yuli Ouyang
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Zihai Li
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Feng Hong
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
6
|
Zhao L, Guo J, Xu S, Duan M, Liu B, Zhao H, Wang Y, Liu H, Yang Z, Yuan H, Jiang X, Jiang X. Abnormal changes in metabolites caused by m 6A methylation modification: The leading factors that induce the formation of immunosuppressive tumor microenvironment and their promising potential for clinical application. J Adv Res 2025; 70:159-186. [PMID: 38677545 DOI: 10.1016/j.jare.2024.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/14/2024] [Accepted: 04/14/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND N6-methyladenosine (m6A) RNA methylation modifications have been widely implicated in the metabolic reprogramming of various cell types within the tumor microenvironment (TME) and are essential for meeting the demands of cellular growth and maintaining tissue homeostasis, enabling cells to adapt to the specific conditions of the TME. An increasing number of research studies have focused on the role of m6A modifications in glucose, amino acid and lipid metabolism, revealing their capacity to induce aberrant changes in metabolite levels. These changes may in turn trigger oncogenic signaling pathways, leading to substantial alterations within the TME. Notably, certain metabolites, including lactate, succinate, fumarate, 2-hydroxyglutarate (2-HG), glutamate, glutamine, methionine, S-adenosylmethionine, fatty acids and cholesterol, exhibit pronounced deviations from normal levels. These deviations not only foster tumorigenesis, proliferation and angiogenesis but also give rise to an immunosuppressive TME, thereby facilitating immune evasion by the tumor. AIM OF REVIEW The primary objective of this review is to comprehensively discuss the regulatory role of m6A modifications in the aforementioned metabolites and their potential impact on the development of an immunosuppressive TME through metabolic alterations. KEY SCIENTIFIC CONCEPTS OF REVIEW This review aims to elaborate on the intricate networks governed by the m6A-metabolite-TME axis and underscores its pivotal role in tumor progression. Furthermore, we delve into the potential implications of the m6A-metabolite-TME axis for the development of novel and targeted therapeutic strategies in cancer research.
Collapse
Affiliation(s)
- Liang Zhao
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Department of Colorectal Anal Surgery, Shenyang Coloproctology Hospital, Shenyang 110002, China.
| | - Junchen Guo
- Department of Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Shasha Xu
- Department of Gastroendoscopy, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Meiqi Duan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Baiming Liu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - He Zhao
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Yihan Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Haiyang Liu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Zhi Yang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Hexue Yuan
- Department of Colorectal Anal Surgery, Shenyang Coloproctology Hospital, Shenyang 110002, China.
| | - Xiaodi Jiang
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang 110020, China.
| | - Xiaofeng Jiang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
7
|
Chen Y, Zhang Z, Ji K, Zhang Q, Qian L, Yang C. Role of microplastics in the tumor microenvironment (Review). Oncol Lett 2025; 29:193. [PMID: 40041410 PMCID: PMC11877014 DOI: 10.3892/ol.2025.14939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/07/2025] [Indexed: 03/06/2025] Open
Abstract
Microplastics (MPs) are pervasive in several ecosystems and have the potential to infiltrate multiple aspects of human life through ingestion, inhalation and dermal exposure, thus eliciting substantial concerns regarding their potential implications for human health. Whilst initial research has documented the effects of MPs on disease development across multiple physiological systems, MPs may also facilitate tumor progression by influencing the tumor microenvironment (TME). This evolving focus underscores the growing interest in the role of MPs in tumorigenesis and their interactions within the TME. In the present review, the relationship between MPs and the TME is comprehensively assessed, providing a detailed analysis of their interactions with tumor cells, stromal cells (including macrophages, fibroblasts and endothelial cells), the extracellular matrix and inflammatory processes. Recommendations for future research directions and strategies to address and reduce microplastic pollution are proposed.
Collapse
Affiliation(s)
- Yunjie Chen
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zihang Zhang
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Kangming Ji
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Qiuchen Zhang
- Department of Radiology, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Lijun Qian
- Department of Geriatric Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Chuang Yang
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
- Department of Radiology, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| |
Collapse
|
8
|
Liang Y, Zhao Y, Qi Z, Li X, Zhao Y. Ferroptosis: CD8 +T cells' blade to destroy tumor cells or poison for self-destruction. Cell Death Discov 2025; 11:128. [PMID: 40169575 PMCID: PMC11962101 DOI: 10.1038/s41420-025-02415-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/19/2025] [Accepted: 03/19/2025] [Indexed: 04/03/2025] Open
Abstract
Ferroptosis represents an emerging, iron-dependent form of cell death driven by lipid peroxidation. In recent years, it has garnered significant attention in the realm of cancer immunotherapy, particularly in studies involving immune checkpoint inhibitors. This form of cell death not only enhances our comprehension of the tumor microenvironment but is also considered a promising therapeutic strategy to address tumor resistance, investigate immune activation mechanisms, and facilitate the development of cancer vaccines. The combination of immunotherapy with ferroptosis provides innovative targets and fresh perspectives for advancing cancer treatment. Nevertheless, tumor cells appear to possess a wider array of ferroptosis evasion strategies compared to CD8+T cells, which have been conclusively shown to be more vulnerable to ferroptosis. Furthermore, ferroptosis in the TME can create a favorable environment for tumor survival and invasion. Under this premise, both inducing tumor cell ferroptosis and inhibiting T cell ferroptosis will impact antitumor immunity to some extent, and even make the final result run counter to our therapeutic purpose. This paper systematically elucidates the dual-edged sword role of ferroptosis in the antitumor process of T cells, briefly outlining the complexity of ferroptosis within the TME. It explores potential side effects associated with ferroptosis-inducing therapies and critically considers the combined application of ferroptosis-based therapies with ICIs. Furthermore, it highlights the current challenges faced by this combined therapeutic approach and points out future directions for development.
Collapse
Affiliation(s)
- Yuan Liang
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yixin Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhaoyang Qi
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinru Li
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuguang Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
9
|
Liu D, Liu L, Zhao X, Zhang X, Chen X, Che X, Wu G. A comprehensive review on targeting diverse immune cells for anticancer therapy: Beyond immune checkpoint inhibitors. Crit Rev Oncol Hematol 2025; 210:104702. [PMID: 40122356 DOI: 10.1016/j.critrevonc.2025.104702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
Although immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, primary resistance and acquired resistance continue to limit their efficacy for many patients. To address resistance and enhance the anti-tumor activity within the tumor immune microenvironment (TIME), numerous therapeutic strategies targeting both innate and adaptive immune cells have emerged. These include combination therapies with ICIs, chimeric antigen receptor T-cell (CAR-T), chimeric antigen receptor macrophages (CAR-Ms) or chimeric antigen receptor natural killer cell (CAR-NK) therapy, colony stimulating factor 1 receptor (CSF1R) inhibitors, dendritic cell (DC) vaccines, toll-like receptor (TLR) agonists, cytokine therapies, and chemokine inhibition. These approaches underscore the significant potential of the TIME in cancer treatment. This article provides a comprehensive and up-to-date review of the mechanisms of action of various innate and adaptive immune cells within the TIME, as well as the therapeutic strategies targeting each immune cell type, aiming to deepen the understanding of their therapeutic potential.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinming Zhao
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaoman Zhang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaochi Chen
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
10
|
Cannet F, Sequera C, Veloso PM, El Kaoutari A, Methia M, Richelme S, Kaya M, Cherni A, Dupont M, Borg JP, Morel C, Boursier Y, Maina F. Tracing specificity of immune landscape remodeling associated with distinct anticancer treatments. iScience 2025; 28:112071. [PMID: 40124507 PMCID: PMC11930375 DOI: 10.1016/j.isci.2025.112071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 07/18/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Immune cells within the tumor microenvironment impact cancer progression, resistance, response to treatments. Despite remarkable outcomes for some cancer patients, immunotherapies remain unsatisfactory for others. Here, we designed an experimental setting using the Alb-R26 Met "inside-out" mouse model, faithfully recapitulating molecular features of liver cancer patients, to explore the effects of distinct anticancer targeted therapies on the tumor immune landscape. Using two treatments in clinical trials for different cancer types, Decitabine and MEK+BCL-XL blockage, we show their capability to trigger tumor regression in Alb-R26 Met mice and to superimpose distinct profiles of immune cell types and immune-checkpoints, impacting immunotherapy response. A machine learning approach processing tumor imaging and immune profile data identified a putative signature predicting tumor treatment response in mice and patients. Outcomes exemplify how the tumor immune microenvironment is differentially reshaped by distinct anticancer agents and highlight the importance of measuring its modulation during treatment to optimize oncotherapy and immunotherapy combinations.
Collapse
Affiliation(s)
- Floriane Cannet
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| | - Célia Sequera
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| | - Paula Michea Veloso
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Abdessamad El Kaoutari
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Melissa Methia
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Sylvie Richelme
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| | - Muge Kaya
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Afef Cherni
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
| | - Mathieu Dupont
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
| | - Jean-Paul Borg
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Institut Universitaire de France, Paris, France
| | - Christian Morel
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
| | | | - Flavio Maina
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| |
Collapse
|
11
|
Jiang R, Yang L, Liu X, Xu Y, Han L, Chen Y, Gao G, Wang M, Su T, Li H, Fang L, Sun N, Du H, Zheng J, Wang G. Genetically engineered macrophages reverse the immunosuppressive tumor microenvironment and improve immunotherapeutic efficacy in TNBC. Mol Ther 2025:S1525-0016(25)00198-4. [PMID: 40119517 DOI: 10.1016/j.ymthe.2025.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 01/21/2025] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
The main challenges in current immunotherapy for triple-negative breast cancer (TNBC) lie in the immunosuppressive tumor microenvironment (TME). Considering tumor-associated macrophages (TAMs) are the most abundant immune cells in the TME, resetting TAMs is a promising strategy for ameliorating the immunosuppressive TME. Here, we developed genetically engineered macrophages (GEMs) with gene-carrying adenoviruses, to maintain the M1-like phenotype and directly deliver the immune regulators interleukin-12 and CXCL9 into local tumors, thereby reversing the immunosuppressive TME. In tumor-bearing mice, GEMs demonstrated targeted enrichment in tumors and successfully reprogramed TAMs to M1-like macrophages. Moreover, GEMs significantly enhanced the accumulation, proliferation, and activation of CD8+ T cells, mature dendritic cells, and natural killer cells within tumors, while diminishing M2-like macrophages, immunosuppressive myeloid-derived suppressor cells, and regulatory T cells. This treatment efficiently suppressed tumor growth. In addition, combination therapy with GEMs and anti-programmed cell death protein 1 further improved interferon-γ+CD8+ T cell percentages and tumor inhibition efficacy in an orthotopic murine TNBC model. Therefore, this study provides a novel strategy for reversing the immunosuppressive TME and improving immunotherapeutic efficacy through live macrophage-mediated gene delivery.
Collapse
Affiliation(s)
- Ranran Jiang
- Department of Oncology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China; Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Liechi Yang
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Xin Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Department of Urology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Yujun Xu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Lulu Han
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Yuxin Chen
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Ge Gao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Meng Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Tong Su
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Nan Sun
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Hongwei Du
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Junnian Zheng
- Department of Oncology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
12
|
Sholevar CJ, Liu NM, Mukarrama T, Kim J, Lawrence J, Canter RJ. Myeloid Cells in the Immunosuppressive Microenvironment as Immunotargets in Osteosarcoma. Immunotargets Ther 2025; 14:247-258. [PMID: 40125425 PMCID: PMC11930235 DOI: 10.2147/itt.s485672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
Osteosarcoma is an aggressive primary malignant bone tumor associated with high rates of metastasis and poor 5-year survival rates with limited improvements in approximately 40 years. Standard multimodality treatment includes chemotherapy and surgery, and survival rates have remained stagnant. Overall, response rates to immunotherapy like immune checkpoint inhibitors have been disappointing in osteosarcoma despite exciting results in other epithelial tumor types. The poor response of osteosarcoma to current immunotherapies is multifactorial, but a key observation is that the tumor microenvironment in osteosarcoma is profoundly immunosuppressive, and increasing evidence suggests a significant role of suppressive myeloid cells in tumor progression and immune evasion, particularly by myeloid-derived suppressor cells. Targeting suppressive myeloid cells via novel agents are attractive strategies to develop novel immunotherapies for osteosarcoma, and combination strategies will likely be important for durable responses. In this review, we will examine mechanisms of the immunosuppressive microenvironment, highlight pre-clinical and clinical data of combination strategies including colony-stimulating factor 1 (CSF-1) receptor, phosphoinositide 3-kinase (PI3K), CXCR4, and checkpoint inhibition, as well as the role of canine models in elucidating myeloid cells as targets in osteosarcoma immunotherapy.
Collapse
Affiliation(s)
- Cyrus J Sholevar
- Department of Surgery, Division of Surgical Oncology, University of California Davis, Sacramento, CA, USA
| | - Natalie M Liu
- Department of Surgery, Division of Surgical Oncology, University of California Davis, Sacramento, CA, USA
| | - Tasneem Mukarrama
- Biomedical Engineering, University of California Davis, Sacramento, CA, USA
| | - Jinhwan Kim
- Biomedical Engineering, University of California Davis, Sacramento, CA, USA
| | - Jessica Lawrence
- Department of Surgical & Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Robert J Canter
- Department of Surgery, Division of Surgical Oncology, University of California Davis, Sacramento, CA, USA
| |
Collapse
|
13
|
Gao Y, Zhang X, Ding M, Fu Z, Zhong L. Targeting "don't eat me" signal: breast cancer immunotherapy. Breast Cancer Res Treat 2025:10.1007/s10549-025-07659-w. [PMID: 40100495 DOI: 10.1007/s10549-025-07659-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025]
Abstract
PURPOSE Breast cancer ranks as the most prevalent cancer type impacting women globally, both in terms of incidence and mortality rates, making it a major health concern for females. There's an urgent requirement to delve into new cancer treatment methods to improve patient survival rates. METHODS Immunotherapy has gained recognition as a promising area of research in the treatment of breast cancer, with targeted immune checkpoint therapies demonstrating the potential to yield sustained clinical responses and improve overall survival rates. Presently, the predominant immune checkpoints identified on breast cancer cells include CD47, CD24, PD-L1, MHC-I, and STC-1, among others. Nevertheless, the specific roles of these various immune checkpoints in breast carcinogenesis, metastasis, and immune evasion have yet to be comprehensively elucidated. We conducted a comprehensive review of the existing literature pertaining to breast cancer and immune checkpoint inhibitors, providing a summary of findings and an outlook on future research directions. RESULTS This article reviews the advancements in research concerning each immune checkpoint in breast cancer and their contributions to immune evasion, while also synthesizing immunotherapy strategies informed by these mechanisms. Furthermore, it anticipates future research priorities, thereby providing a theoretical foundation to guide immunotherapy as a potential interventional approach for breast cancer treatment. CONCLUSION Knowledge of immune checkpoints will drive the creation of novel cancer therapies, and future breast cancer research will increasingly emphasize personalized treatments tailored to patients' specific tumor characteristics.
Collapse
Affiliation(s)
- Yue Gao
- Department of Breast Surgery, Sixth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyan Zhang
- Department of Breast Surgery, Sixth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mingqiang Ding
- Department of Breast Surgery, Sixth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenkun Fu
- Department of Immunology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China.
| | - Lei Zhong
- Department of Breast Surgery, Sixth Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
14
|
Shi L, Wang H, Sun Y, Xu N, Pei A, Zhang N. Development and validation of a disulfidptosis-related prognostic model for colorectal cancer using multi-omics analysis. Discov Oncol 2025; 16:338. [PMID: 40095116 PMCID: PMC11914417 DOI: 10.1007/s12672-025-02055-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
This study aims to integrate multi-omic and clinical data concerning disulfidptosis-related genes (DRGs) to facilitate molecular typing and prognosis in colorectal cancer (CRC). Public databases provided CRC transcriptome and clinical data, enabling differential expression, genomic analyses, pathway enrichment, survival analysis, and subtyping based on the expression levels of 15 DRGs identified in published studies. Differentially expressed genes (DEGs) between subtypes were identified to create a disulfidptosis prognostic model using LASSO and Cox regression analyses. This model was evaluated by comparing risk scores, survival curves, cellular infiltration, and drug sensitivity between high- and low-risk groups. Analyses revealed differential expression, mutations, and copy number variations (CNV) in DRGs in CRC. Survival analysis demonstrated significant prognostic differences among DRG expression subtypes. GSVA and ssGSEA highlighted DRGs' regulatory roles in CRC. DEGs identified between DRG expression subtypes led to the classification into subtypes A and B. A disulfidptosis prognostic model, including genes VSIG4, SCG2, INHBB, DDC, CXCL13, KLK10, CXCL10, and CCL11A, was developed to stratify patients into high- and low-risk groups. This model displayed strong predictive capability (AUC = 0.700) and calibration. The risk score was also strongly associated with immune cell infiltration, stromal cell score, and stem cell index in the CRC tumor microenvironment. Drug sensitivity analysis indicated that high-risk samples were more responsive to most medications. We established a robust disulfidptosis prognostic model for CRC through comprehensive multi-omics analysis. Our findings provide valuable insights into the role of DRGs in CRC progression and disease management, presenting an important resource for further research.
Collapse
Affiliation(s)
- Lei Shi
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China
| | - Huimei Wang
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China
| | - Yongxiao Sun
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China
| | - Na Xu
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China
| | - Aiyue Pei
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China.
| | - Nan Zhang
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China.
| |
Collapse
|
15
|
Hu S, Sun D, Tang L, Kong L, Liu Y, Liu F, Tang D, Lu X, Wang Y. Follicle-stimulating hormone peptide-conjugated liposomes in the treatment of epithelial ovarian cancer through the induction of M2-to-M1 macrophage repolarization. Int J Pharm 2025; 672:125334. [PMID: 39933608 DOI: 10.1016/j.ijpharm.2025.125334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/26/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025]
Abstract
INTRODUCTION The silent killer epithelial ovarian cancer (EOC) is a lethal malignancy with high mortality rate and often diagnosed at an advanced stage. Traditional chemotherapy for EOC remains unsatisfactory as the tumor microenvironment (TME) is complicated and contains multiple factors such as tumor associated macrophages (TAMs). Therefore, a drug delivery system which codelivery chemotherapy drug and immune modulator for EOC treatment is urgently needed. METHODS Follicle-stimulating hormone peptide-conjugated paclitaxel and ginsenoside Rh2 codelivery liposomes (FSH@PTX-Rh2-Lips) were prepared in this study. FSH was decorated on the liposomal surface to enhance cellar uptake, PTX was used to kill cancer cells, and Rh2 was added to induce macrophages repolarization as well as a member material. The targeting, anti-tumor effect and impact on macrophage repolarization of FSH@PTX-Rh2-Lips were evaluated in vitro and in vivo. RESULTS With the ideal physicochemical properties, FSH@PTX-Rh2-Lips displayed increased cellular uptake, strong cytotoxicity to ID8 cells, inhibitory effect of tumor cell metastasis, and ability to induce macrophage repolarization from M2 to M1 in vitro. The tumor-bearing mice model suggested FSH@PTX-Rh2-Lips showed significant effect on antitumor and tumor recurrence, and the mechanism of FSH@PTX-Rh2-Lips in treatment of EOC was related to inhibiting tumor growth and inducing macrophage repolarization. CONCLUSION FSH@PTX-Rh2-Lips with function of affecting TAMs repolarization and altering the TME were successfully prepared and might offer an effective therapeutic strategy against EOC.
Collapse
Affiliation(s)
- Shengxia Hu
- Department of Obstetrics and Gynecology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People's Republic of China
| | - Dan Sun
- Department of Obstetrics and Gynecology, Dalian Women and Children's Medical Center (Group), Dalian, People's Republic of China
| | - Ling Tang
- Department of Obstetrics and Gynecology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People's Republic of China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People's Republic of China
| | - Yang Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People's Republic of China
| | - Fang Liu
- Department of Obstetrics and Gynecology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People's Republic of China
| | - Dongmei Tang
- Department of Obstetrics and Gynecology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People's Republic of China
| | - Xuhong Lu
- Department of Obstetrics and Gynecology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People's Republic of China.
| | - Yuanyuan Wang
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People's Republic of China.
| |
Collapse
|
16
|
Li N, An N, Ma S, Cao J, Zhu F, Qi K, Yan Z, Cheng H, Sang W, Chen W, Li D, Li Z, Xu K, Wang Y. Lymphocyte/monocyte to lactate dehydrogenase ratio prior to lymphodepletion impact the outcomes of patients with diffused large B cell lymphoma undergoing CAR-T cell therapy. Cancer Immunol Immunother 2025; 74:148. [PMID: 40088299 PMCID: PMC11910468 DOI: 10.1007/s00262-025-03987-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/17/2025] [Indexed: 03/17/2025]
Abstract
Factors associated with outcomes of chimeric antigen receptor (CAR)-T cell therapy in patients with relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL) have not been fully elucidated. We explored the impact of the prelymphodepletion (pre-LD) lymphocyte to monocyte ratio (LMR) and its ratio to lactate dehydrogenase (LDH) (LMR/LDH) on the efficacy and prognosis of 60 patients with R/R DLBCL undergoing CAR-T cell therapy. The optimal cutoff values for pre-LD LMR and LMR/LDH were 3.583 and 0.0103, respectively. The overall response rate (ORR)s were higher in patients with high pre-LD LMR or LMR/LDH than those with low pre-LD LMR or LMR/LDH (ORR, 100% vs. 65.79%, P = 0.006 and 96.15% vs. 38.24%, P < 0.0001, respectively). Pre-LD LMR/LDH was an independent factor associated with ORR (P = 0.010, odds ratio = 18.757; 95% confidence interval [CI] 2.046-171.975) by multivariate logistic regression analysis. Patients with high pre-LD LMR/LDH had significantly longer progression-free survival (PFS) (median PFS, 29.73 vs. 2.47 months, P < 0.0001) and overall survival (OS) (median OS, not reached vs. 7.4 months, P = 0.0002) than those with low pre-LD LMR/LDH. Multivariate Cox regression analysis showed that pre-LD LMR/LDH and ORR were independent factors affecting PFS (P = 0.030, hazard ratio [HR] = 2.561; 95% CI 1.093-5.999 and P = 0.024, HR = 2.202; 95% CI 1.22-4.369, respectively); pre-LD LMR/LDH was an independent factor affecting OS (P = 0.029, HR = 3.331; 95% CI 1.131-9.807). In conclusion, the pre-LD LMR/LDH was an independent factor associated with ORR and an independent prognostic factor in patients with R/R DLBCL undergoing CAR-T cell therapy.
Collapse
Affiliation(s)
- Na Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Na An
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Sha Ma
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, China
| | - Jiang Cao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, China
| | - Feng Zhu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, China
| | - Kunming Qi
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, China
| | - Zhiling Yan
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, China
| | - Hai Cheng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, China
| | - Wei Sang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, China
| | - Wei Chen
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, China
| | - Depeng Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, China
| | - Zhenyu Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, China
| | - Ying Wang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, China.
| |
Collapse
|
17
|
Zhang Y, Xie Y, Wang Y, Huang P, Lu Y. The Role of Radiosensitizing Drugs in Osteosarcoma Treatment: Mechanisms and Clinical Perspectives. Drug Des Devel Ther 2025; 19:1927-1942. [PMID: 40110500 PMCID: PMC11920643 DOI: 10.2147/dddt.s512479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/09/2025] [Indexed: 03/22/2025] Open
Abstract
Osteosarcoma is a highly malignant bone tumor that is resistant to radiotherapy and is associated with poor treatment outcomes and prognoses. Understanding the mechanisms of radioresistance and finding strategies to enhance the radiosensitivity is crucial for improving clinical efficacy. The aim of this review was to address the approaches for enhancing the efficacy of radiotherapy in osteosarcoma, thereby improving patient outcomes. Specifically, we have focused on the mechanisms of radiosensitization and the relationship between drugs that enhance radiosensitivity and cancer. These mechanisms involve a delay in DNA damage repair, promotion of apoptosis, inhibition of angiogenesis, and regulation of the tumor microenvironment. In addition, we have summarized the effects of these drugs on the proliferation, migration, invasion and apoptosis of osteosarcoma cell lines. Finally, we have discussed the therapeutic effects and adverse reactions of these drugs in other cancers, providing valuable guidance for clinical treatment strategies tailored to patients with osteosarcoma.
Collapse
Affiliation(s)
- Yilei Zhang
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, People's Republic of China
| | - Yuhuan Xie
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, People's Republic of China
| | - Yiwen Wang
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, People's Republic of China
| | - Panpan Huang
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, People's Republic of China
| | - Yao Lu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, People's Republic of China
| |
Collapse
|
18
|
Greiner D, Xue Q, Waddell TQ, Kurudza E, Chaudhary P, Belote RL, Dotti G, Judson-Torres RL, Reeves MQ, Cheshier SH, Roh-Johnson M. Human CSPG4-targeting CAR-macrophages inhibit melanoma growth. Oncogene 2025:10.1038/s41388-025-03332-0. [PMID: 40082557 DOI: 10.1038/s41388-025-03332-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/12/2025] [Accepted: 02/24/2025] [Indexed: 03/16/2025]
Abstract
Approximately half of melanoma patients relapse or fail to respond to current standards of care, highlighting the need for new treatment options. Engineering T-cells with chimeric antigen receptors (CARs) has revolutionized the treatment of hematological malignancies but has been clinically less effective in solid tumors. We therefore sought to engineer alternative immune cell types to inhibit melanoma progression. Engineering macrophages with CARs has emerged as a promising approach to overcome some of the challenges faced by CAR-T cells; however, whether these engineered macrophages can effectively inhibit melanoma growth is unknown. To determine whether CAR-macrophages (CAR-Ms) specifically target and kill melanoma cells, we engineered CAR-Ms targeting chondroitin sulfate proteoglycan 4 (CSPG4), an antigen expressed in melanoma. CSPG4-targeting CAR-Ms exhibited specific phagocytosis of CSPG4-expressing melanoma cells. We developed 3D approaches to show that CSPG4-targeting CAR-Ms efficiently infiltrated melanoma spheroids. Furthermore, combining CSPG4-targeting CAR-Ms with strategies inhibiting CD47/SIRPα "don't eat me" signaling synergistically enhanced CAR-M-mediated phagocytosis and robustly inhibited melanoma spheroid growth in 3D. Importantly, CSPG4-targeting CAR-Ms inhibited melanoma tumor growth in mouse models. These results suggest engineering macrophages against melanoma antigens is a promising solid tumor immunotherapy approach for treating melanoma.
Collapse
Affiliation(s)
- Daniel Greiner
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Qian Xue
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Trinity Qa Waddell
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Elena Kurudza
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, Salt Lake City, UT, 84112, USA
| | - Piyush Chaudhary
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Rachel L Belote
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
- Department of Molecular Genetics, The Ohio State University College of Arts and Sciences, Columbus, OH, 43210, USA
| | - Gianpietro Dotti
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Robert L Judson-Torres
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
- Department of Dermatology, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Melissa Q Reeves
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Samuel H Cheshier
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
- Division of Pediatric Neurosurgery, Intermountain Primary Children's Hospital, Salt Lake City, UT, 84112, USA
| | - Minna Roh-Johnson
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
19
|
Kaneshige A, Yang Y, Bai L, Wang M, Xu R, Mallik L, Chinnaswamy K, Metwally H, Wang Y, McEachern D, Tošović J, Yang CY, Kirchhoff PD, Meagher JL, Stuckey JA, Wang S. Discovery of AK-1690: A Potent and Highly Selective STAT6 PROTAC Degrader. J Med Chem 2025; 68:5125-5151. [PMID: 39311434 DOI: 10.1021/acs.jmedchem.4c01009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
STAT6 is an attractive therapeutic target for human cancers and other human diseases. Starting from a STAT6 ligand with Ki = 3.5 μM binding affinity, we obtained AK-068 with Ki = 6 nM to STAT6 and at least >85-fold binding selectivity over STAT5. Using AK-068 and cereblon ligands, we discovered AK-1690 as the first, potent and selective PROTAC STAT6 degrader. AK-1690 effectively induces degradation of STAT6 protein in cells with DC50 values of as low as 1 nM while showing minimal effect on other STAT members up to 10 μM. A single dose of AK-1690 effectively depletes STAT6 in mouse tissues. Determination of the first cocrystal structure of STAT6 in complex with AK-1690 provides a structural basis for their interactions. AK-1690 is a powerful tool with which to investigate the roles of STAT6 in human diseases and biological processes and a promising lead compound for further optimization.
Collapse
Affiliation(s)
- Atsunori Kaneshige
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yiqing Yang
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Longchuan Bai
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mi Wang
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Renqi Xu
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Leena Mallik
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | | | - Hoda Metwally
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yu Wang
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Donna McEachern
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jelena Tošović
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Chao-Yie Yang
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Paul D Kirchhoff
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jennifer L Meagher
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jeanne A Stuckey
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Shaomeng Wang
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
20
|
He J, Gao W, Dong R, Lv Y, Zhang Q, Li L, Xie X, Lv Q, Hu L, Wang J. Discovery of Novel Pyrrolo[2,3- b]pyridine-Based CSF-1R Inhibitors with Demonstrated Efficacy against Patient-Derived Colorectal Cancer Organoids. J Med Chem 2025; 68:5655-5674. [PMID: 40013758 DOI: 10.1021/acs.jmedchem.4c02933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Repolarizing M2-like tumor associated macrophages (TAMs) into M1 phenotype by blocking CSF-1/CSF-1R signaling pathway represents a promising strategy to remodel the tumor immune microenvironment. Therefore, the discovery of novel potent CSF-1R inhibitors is of great significance for colorectal cancer immunotherapy. In this work, a series of novel CSF-1R inhibitors were designed and synthesized through rational molecular hybridization strategy and step by step structural optimization based on PLX3397 and BLZ945. Among these derivatives, compound III-1 was strongly bound to CSF-1R and showed potent CSF-1R inhibitory activity. It also effectively inhibited the activation of intracellular CSF-1R pathway and its downstream signaling events. Mechanically, III-1 efficiently repolarized M2-like TAMs into M1-phenotype, and inhibited the proliferation and promoted apoptosis of tumor cells through immunoregulation. More importantly, III-1 showed demonstrated efficacy against patient-derived colorectal cancer organoids and exhibited stronger anticolorectal cancer efficacy in vivo compared to PLX3397, highlighting its potential in the immunotherapy of colorectal cancer.
Collapse
Affiliation(s)
- Jinting He
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Wen Gao
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Rongrong Dong
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Yingshan Lv
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Qiang Zhang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Lin Li
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Xiaolong Xie
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Qi Lv
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Lihong Hu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Junwei Wang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| |
Collapse
|
21
|
Han HJ, Rubio-Alarcon M, Allen T, Lee S, Rahman T. Differential neuropilin isoform expressions highlight plasticity in macrophages in the heterogenous TME through in-silico profiling. Front Immunol 2025; 16:1547330. [PMID: 40134439 PMCID: PMC11933088 DOI: 10.3389/fimmu.2025.1547330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/11/2025] [Indexed: 03/27/2025] Open
Abstract
Introduction The nuanced roles of neuropilin (NRP) isoforms, NRP1 and NRP2, have attracted considerable scientific interest regarding cancer progression. Their differential expressions across various cancer types are specific to NRP isoforms which are shown in a cancer type-dependent manner. It accounts for the different mechanisms involved, driven by a co-expression of gene-sets associated with overexpressed NRP1 or NRP2. Their different expressions on tumour-associated macrophages (TAMs) with disparate markers are associated with the heterogenous tumour microenvironment (TME) through their plasticity and pro-tumorigenic activities. Methods Single-cell RNA sequencing (scRNA-seq) analyses were performed on tumours from clear cell Renal Cell Carcinoma (ccRCC) and skin cutaneous melanoma (SKCM) which exhibit the highest expressions of NRP1 and NRP2, respectively. Datasets were processed using established bioinformatics pipelines, including clustering algorithms, to determine cellular heterogeneity and quantify NRP isoform expression within distinct macrophage populations. Using differential gene expression analysis (DEGA) alongside co-enrichment studies, we explored gene-sets associated with NRP1 or NRP2 overexpression in TAMs. Results Our analysis revealed a marked upregulation of NRP1 in TAMs isolated from ccRCC and elevated NRP2 expression in SKCM-derived TAMs. Both NRP1+ and NRP2+ macrophages showed an M2-like polarisation characterised by immune suppression and extracellular matrix degradation. Coupled with the previously uncharacterised NRP isoform specific- subpopulations within these cancers identified by DEGA, co-enrichment analyses demonstrated that the upregulation of gene-sets associated with NRP1 is associated with angiogenesis and tumour progression through VEGF signalling, while gene-sets with NRP2 showed dual functionality in the TME-dependent manner. Their distinct roles in regulating macrophage plasticity, tumour invasion, and metastasis were highlighted. Discussion These findings underscore distinct isoform-specific mechanisms by which NRP1 and NRP2 contribute to TAM-mediated cancer progression. This study aims to establish a foundation for future research, leading to biological experiments with focused gene-sets derived from our findings. This approach can contribute to the development of immunomodulatory strategies targeting specific NRP isoforms in macrophages, tailored to individual cancer types and abnormal expressions of those gene markers, potentially offering a more effective therapeutic approach compared to broad-spectrum NRP inhibition strategies.
Collapse
Affiliation(s)
- Hyun-Jee Han
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | | | - Thomas Allen
- Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Sunwoo Lee
- Department of Medical Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
22
|
Turlej E, Domaradzka A, Radzka J, Drulis-Fajdasz D, Kulbacka J, Gizak A. Cross-Talk Between Cancer and Its Cellular Environment-A Role in Cancer Progression. Cells 2025; 14:403. [PMID: 40136652 PMCID: PMC11940884 DOI: 10.3390/cells14060403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
The tumor microenvironment is a dynamic and complex three-dimensional network comprising the extracellular matrix and diverse non-cancerous cells, including fibroblasts, adipocytes, endothelial cells and various immune cells (lymphocytes T and B, NK cells, dendritic cells, monocytes/macrophages, myeloid-derived suppressor cells, and innate lymphoid cells). A constantly and rapidly growing number of studies highlight the critical role of these cells in shaping cancer survival, metastatic potential and therapy resistance. This review provides a synthesis of current knowledge on the modulating role of the cellular microenvironment in cancer progression and response to treatment.
Collapse
Affiliation(s)
- Eliza Turlej
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Aleksandra Domaradzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Justyna Radzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Dominika Drulis-Fajdasz
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Julita Kulbacka
- Departament of Molecular and Cellular Biology, Faculty of Pharmacy, Wrocław Medical University, Borowska 211A, 50-556 Wrocław, Poland;
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Agnieszka Gizak
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| |
Collapse
|
23
|
Yuan X, Kang Y, Li R, Niu G, Shi J, Yang Y, Fan Y, Ye J, Han J, Pei Z, Zhang Z, Ji X. Magnetically triggered thermoelectric heterojunctions with an efficient magnetic-thermo-electric energy cascade conversion for synergistic cancer therapy. Nat Commun 2025; 16:2369. [PMID: 40064895 PMCID: PMC11894112 DOI: 10.1038/s41467-025-57672-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Thermoelectric therapy has been emerging as a promising and versatile strategy for targeting malignant tumors treatment. However, the lack of effective time-space controlled triggering of thermoelectric effect in vivo limits the application of thermoelectric therapy. Here a magnetically triggered thermoelectric heterojunction (CuFe2O4/SrTiO3, CFO/STO) for synergistic thermoelectric/chemodynamic/immuno-therapy is developed. The efficient magnetothermal nanoagent (CFO) is synthesized using the hydrothermal method, and thermoelectric nanomaterials (STO) are grown on its surface to create the heterojunction. To enhance oral delivery efficiency, a fusion membrane (M) of Staphylococcus aureus and macrophage cell membranes are coated the CFO/STO heterojunction, enabling effective targeting of orthotopic colorectal cancer. Once the CFO/STO@M reaches the tumor region, in vitro alternating magnetic field (AMF) stimulation activates the catalytic treatment through a magnetic-thermo-electric energy cascade conversion effect. Additionally, the immunogenic death of tumor cells, down-regulating vascular endothelial growth factor and heat shock protein HSP70, increasing expression of endothelial cell adhesion molecule (ICAM-1/VCAM-1), and M1 polarization of macrophages contribute to tumor immunotherapy. Overall, the magnetically triggered thermoelectric heterojunction based on CFO/STO@M shows remarkable antitumor capability in female mice, offering a promising approach to broaden both the scope of application and the effectiveness of catalytic therapy.
Collapse
Affiliation(s)
- Xue Yuan
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Yong Kang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Ruiyan Li
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Gaoli Niu
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Jiacheng Shi
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Yiwen Yang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Yueyue Fan
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Jiamin Ye
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Jingwen Han
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Zhengcun Pei
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Zhuhong Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China.
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China.
- Medical College, Linyi University, Linyi, China.
| |
Collapse
|
24
|
Rodriguez-Baena FJ, Marquez-Galera A, Ballesteros-Martinez P, Castillo A, Diaz E, Moreno-Bueno G, Lopez-Atalaya JP, Sanchez-Laorden B. Microglial reprogramming enhances antitumor immunity and immunotherapy response in melanoma brain metastases. Cancer Cell 2025; 43:413-427.e9. [PMID: 39919736 DOI: 10.1016/j.ccell.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 11/04/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025]
Abstract
Melanoma is one of the tumor types with the highest risk of brain metastasis. However, the biology of melanoma brain metastasis and the role of the brain immune microenvironment in treatment responses are not yet fully understood. Using preclinical models and single-cell transcriptomics, we have identified a mechanism that enhances antitumor immunity in melanoma brain metastasis. We show that activation of the Rela/Nuclear Factor κB (NF-κB) pathway in microglia promotes melanoma brain metastasis. Targeting this pathway elicits microglia reprogramming toward a proinflammatory phenotype, which enhances antitumor immunity and reduces brain metastatic burden. Furthermore, we found that proinflammatory microglial markers in melanoma brain metastasis are associated with improved responses to immune checkpoint inhibitors in patients and targeting Rela/NF-κB pathway in mice improves responses to these therapies in the brain, suggesting a strategy to enhance antitumor immunity and responses to immune checkpoint inhibitors in patients with melanoma brain metastasis.
Collapse
Affiliation(s)
| | | | | | - Alba Castillo
- Instituto de Neurociencias (CSIC-UMH), San Juan de Alicante, Spain
| | - Eva Diaz
- MD Anderson Cancer Center International Foundation, Madrid, Spain
| | - Gema Moreno-Bueno
- MD Anderson Cancer Center International Foundation, Madrid, Spain; Instituto de Investigaciones Biomédicas "Sols-Morreale" CSIC-UAM, Madrid, Spain; CIBERONC Centro de Investigación Biomédica en Red de Cancer, ISCIII, Madrid, Spain; Translational Cancer Research Group, Area 3 Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | | | | |
Collapse
|
25
|
Park SY, Pylaeva E, Bhuria V, Gambardella AR, Schiavoni G, Mougiakakos D, Kim SH, Jablonska J. Harnessing myeloid cells in cancer. Mol Cancer 2025; 24:69. [PMID: 40050933 PMCID: PMC11887392 DOI: 10.1186/s12943-025-02249-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/28/2025] [Indexed: 03/09/2025] Open
Abstract
Cancer-associated myeloid cells due to their plasticity play dual roles in both promoting and inhibiting tumor progression. Myeloid cells with immunosuppressive properties play a critical role in anti-cancer immune regulation. Cells of different origin, such as tumor associated macrophages (TAMs), tumor associated neutrophils (TANs), myeloid derived suppressor cells (also called MDSCs) and eosinophils are often expanded in cancer patients and significantly influence their survival, but also the outcome of anti-cancer therapies. For this reason, the variety of preclinical and clinical studies to modulate the activity of these cells have been conducted, however without successful outcome to date. In this review, pro-tumor activity of myeloid cells, myeloid cell-specific therapeutic targets, in vivo studies on myeloid cell re-polarization and the impact of myeloid cells on immunotherapies/genetic engineering are addressed. This paper also summarizes ongoing clinical trials and the concept of chimeric antigen receptor macrophage (CAR-M) therapies, and suggests future research perspectives, offering new opportunities in the development of novel clinical treatment strategies.
Collapse
Affiliation(s)
- Su-Yeon Park
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ekaterina Pylaeva
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, Essen, 45147, Germany
- German Cancer Consortium (DKTK) Partner Site Düsseldorf/Essen, Essen, Germany
| | - Vikas Bhuria
- Department of Hematology, Oncology, and Cell Therapy, Otto-Von-Guericke University, Magdeburg, Germany
| | | | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Rome, Italy
| | - Dimitrios Mougiakakos
- Department of Hematology, Oncology, and Cell Therapy, Otto-Von-Guericke University, Magdeburg, Germany
| | - Sung-Hoon Kim
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jadwiga Jablonska
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, Essen, 45147, Germany.
- German Cancer Consortium (DKTK) Partner Site Düsseldorf/Essen, Essen, Germany.
| |
Collapse
|
26
|
Soto CA, Lesch ML, Becker JL, Sharipol A, Khan A, Schafer XL, Becker MW, Munger JC, Frisch BJ. Elevated Lactate in the AML Bone Marrow Microenvironment Polarizes Leukemia-Associated Macrophages via GPR81 Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.13.566874. [PMID: 39185193 PMCID: PMC11343108 DOI: 10.1101/2023.11.13.566874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Interactions between acute myeloid leukemia (AML) and the bone marrow microenvironment (BMME) are critical to leukemia progression and chemoresistance. In the solid tumor microenvironment, altered metabolite levels contribute to cancer progression. We performed a metabolomic analysis of AML patient bone marrow serum, revealing increased metabolites compared to age- and sex-matched controls. The most highly elevated metabolite in the AML BMME was lactate. Lactate signaling in solid tumors induces immunosuppressive tumor-associated macrophages and correlates with poor prognosis. This has not yet been studied in the leukemic BMME. Herein, we describe the role of lactate in the polarization of leukemia-associated macrophages (LAMs). Using a murine AML model of blast crisis chronic myelogenous leukemia (bcCML), we characterize the suppressive phenotype of LAMs by surface markers, transcriptomics, and cytokine profiling. Then, mice genetically lacking GPR81, the extracellular lactate receptor, were used to demonstrate GPR81 signaling as a mechanism of both the polarization of LAMs and the direct support of leukemia cells. Furthermore, elevated lactate diminished the function of hematopoietic progenitors and reduced stromal support for normal hematopoiesis. We report microenvironmental lactate as a mechanism of AML-induced immunosuppression and leukemic progression, thus identifying GPR81 signaling as an exciting and novel therapeutic target for treating this devastating disease.
Collapse
Affiliation(s)
- Celia A Soto
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Maggie L Lesch
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine, Rochester, NY, USA
| | - Jennifer L Becker
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Azmeer Sharipol
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester School of Medicine, Rochester, NY, USA
| | - Amal Khan
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine, Rochester, NY, USA
| | - Xenia L Schafer
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, NY, USA
| | - Michael W Becker
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, University of Rochester School of Medicine, Rochester, NY, USA
| | - Joshua C Munger
- Department of Microbiology and Immunology, University of Rochester School of Medicine, Rochester, NY, USA
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, NY, USA
| | - Benjamin J Frisch
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester School of Medicine, Rochester, NY, USA
| |
Collapse
|
27
|
Suliman M, Saleh RO, Chandra M, Rasool KH, Jabir M, Jawad SF, Hasan TF, Singh M, Singh M, Singh A. Macrophage-derived lncRNAs in cancer: regulators of tumor progression and therapeutic targets. Med Oncol 2025; 42:91. [PMID: 40048034 DOI: 10.1007/s12032-025-02643-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Macrophages are key tumor microenvironment (TME) regulators, exhibiting remarkable plasticity that enables them to either suppress or promote cancer progression. Emerging evidence highlights the critical role of macrophage-derived long non-coding RNAs (lncRNAs) in shaping tumor immunity, influencing macrophage polarization, immune evasion, angiogenesis, metastasis, and therapy resistance. This review comprehensively elucidates the functional roles of M1- and M2-associated lncRNAs, detailing their molecular mechanisms and impact on cancer pathogenesis. In summary, elucidating the roles of lncRNAs derived from macrophages in cancer progression offers new avenues for therapeutic strategies, significantly improving patient outcomes in the fight against the disease. Further research into the functional significance of these lncRNAs and the development of targeted therapies is essential to harness their potential fully in clinical applications. We further explore their potential as biomarkers for cancer prognosis and therapeutic targets for modulating macrophage activity to enhance anti-cancer immunity. Targeting macrophage-derived lncRNAs represents a promising avenue for precision oncology, offering novel strategies to reshape the TME and improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Raed Obaid Saleh
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al Maarif, Anbar, Iraq.
| | - Muktesh Chandra
- Marwadi University Research Center, Department of Bioinformatics, Faculty of Engineering and Technology, Marwadi University, Rajkot, Gujarat, 360003, India
| | | | - Majid Jabir
- Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Sabrean F Jawad
- Department of Pharmacy, Al-Mustaqbal University College, 51001, Hillah, Babylon, Iraq
| | - Thikra F Hasan
- College of Health & Medical Technology, Uruk University, Baghdad, Iraq
| | - Mithilesh Singh
- Department of Pharmaceutical Chemistry, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Manmeet Singh
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| |
Collapse
|
28
|
Smith HL, Foxall RB, Duriez PJ, Teal EL, Hoppe AD, Kanczler JM, Gray JC, Beers SA. Comparison of human macrophages derived from peripheral blood and bone marrow. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkae032. [PMID: 40073092 DOI: 10.1093/jimmun/vkae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/21/2024] [Indexed: 03/14/2025]
Abstract
Macrophage differentiation, phenotype, and function have been assessed extensively in vitro by predominantly deriving human macrophages from peripheral blood. It is accepted that there are differences between macrophages isolated from different human tissues; however, the importance of anatomical source for in vitro differentiation and characterization is less clear. Here, phenotype and function were evaluated between human macrophages derived from bone marrow or peripheral blood. Macrophages were differentiated by adherence of heterogenous cell populations or CD14 isolation and polarized with IFNγ and LPS or IL-4 and IL-13 for 48 hours before evaluation of phenotype and phagocytic capacity. The presence of stromal cells in bone marrow heterogenous cultures resulted in a reduction in macrophage purity compared to peripheral blood, which was negated after CD14 isolation. Phenotypically, monocyte-derived macrophages (MDMs) derived from peripheral blood and bone marrow resulted in similar expression of classical and polarized macrophages markers, including CD14, HLA-DR, CD38, and CD40 (increased after IFNγ/LPS), and CD11b and CD206 (elevated after IL-4/IL-13). Functionally, these cells also showed similar levels of Fc-independent and Fc-dependent phagocytosis, although there was a nonsignificant reduction of Fc-dependent phagocytosis in the bone marrow derived macrophages after IFNγ/LPS stimulation. In summary, we have identified that human MDMs differentiated from peripheral blood and bone marrow showed similar characteristics and functionality, suggesting that isolating cells from different anatomical niches does not affect macrophage differentiation after CD14 isolation. Consequently, due to high yield and ready availability peripheral blood derived macrophages are still the most suitable source.
Collapse
Affiliation(s)
- Hannah L Smith
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Bone and Joint Research Group, Human Development and Health, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Russell B Foxall
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Patrick J Duriez
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Emma L Teal
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Adam D Hoppe
- Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, South Dakota, United States
| | - Janos M Kanczler
- Bone and Joint Research Group, Human Development and Health, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Juliet C Gray
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Stephen A Beers
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
29
|
Kim Y, Hur J, Hong SC, Jung J, Park CH, Park JB, Yoon TJ, Kim JB, Yang SH. Modulated electro-hyperthermia therapy combined with Korean mistletoe extract treatment exerts a strong anti-tumor activity by enhancing cellular and humoral immune responses in mice. Anim Cells Syst (Seoul) 2025; 29:163-172. [PMID: 40040867 PMCID: PMC11878165 DOI: 10.1080/19768354.2025.2470455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/04/2025] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
Electro-hyperthermia therapy (EHT) has been known to cause temperature-dependent cell death and enhance the effects of conventional antitumor treatments, such as chemotherapy and radiotherapy. Furthermore, EHT modulates the innate and adaptive immune systems. Mistletoe is one of the most broadly studied complementary and alternative therapeutic agents for cancer treatment due to its ability to stimulate the immune systems. This study aimed to investigate the effects of EHT and mistletoe therapy combination on immune responses. Tumors induced by B16-BL6 melanoma cells were treated twice with modulated EHT (mEHT) (43°C for 10 or 20 min) and with intravenous injection of a Korean mistletoe extract (KME). We examined the level of interferon (IFN)-γ, granzyme, interleukin (IL)-2, IL-10, and tumor-specific antibodies using enzyme-linked immunosorbent assay methods to further study the immunological responses in the combination of mEHT and KME. Additionally, cytotoxic T lymphocyte (CTL) activity is investigated. In this study, we revealed a significant anti-tumor immunological activity elevation in tumor-bearing mice by combined mEHT and KME therapy. Specifically, the combination of mEHT and KME treatment was effective in inhibiting tumor growth in mice. The combination treatment elicited CTL immune response and increased IFN-γ and granzyme secretion. Particularly, the co-treatment appeared to efficiently suppress the immune signal related to tumor-associated macrophage differentiation. Importantly, tumor cell-specific antibodies could be induced in mice after mEHT-treated tumor cell immunization, which represent a promising cancer vaccine strategy. Thus, our results indicate the therapeutic actions of KME as a feasible partner of mEHT, suggesting its potential candidate for cancer immunotherapy. Abbreviations: APC, Antigen-presenting cell; CTL, Cytotoxic T lymphocyte; EHT, Electro-hyperthermia therapy; ELISA, Enzyme-linked immunosorbent assay; HSP, Heat shock protein; KME, Korean mistletoe extract; NK, Natural killer; PBS, Phosphate-buffered saline; QOL, Quality of life; RF, Radio-frequency; TAM, Tumor-associated macrophage.
Collapse
Affiliation(s)
- Yebeen Kim
- Department of Biomedical Engineering, College of Life Science and Biotechnology, Dongguk University, Seoul, Republic of Korea
| | - Jinwoo Hur
- Department of Food and Nutrition, Yuhan University, Buchoen, Republic of Korea
| | - Sung-Chul Hong
- Department of Food Science and Biotechnology, Kunsan National University, Kunsan, Republic of Korea
| | - Jaewoon Jung
- Department of Biomedical Engineering, College of Life Science and Biotechnology, Dongguk University, Seoul, Republic of Korea
| | - Choon-Ho Park
- Graduate School of Clinical Pharmacy and Pharmaceutics, Ajou University, Suwon, South Korea
| | - Joon Beom Park
- Mistletoe Research Center, New Breath Hospital, Seoul, Republic of Korea
| | | | - Jong Bae Kim
- Mistletoe Research Center, New Breath Hospital, Seoul, Republic of Korea
| | - Seung-Hoon Yang
- Department of Biomedical Engineering, College of Life Science and Biotechnology, Dongguk University, Seoul, Republic of Korea
| |
Collapse
|
30
|
Zhang YJN, Xiao Y, Li ZZ, Bu LL. Immunometabolism in head and neck squamous cell carcinoma: Hope and challenge. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167629. [PMID: 39689765 DOI: 10.1016/j.bbadis.2024.167629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/19/2024]
Abstract
Immunotherapy has improved the survival rate of patients with head and neck squamous cell carcinoma (HNSCC), but less than 20 % of them have a durable response to these treatments. Excessive local recurrence and lymph node metastasis ultimately lead to death, making the 5-year survival rate of HNSCC still not optimistic. Cell metabolism has become a key determinant of the viability and function of cancer cells and immune cells. In order to maintain the enormous anabolic demand, tumor cells choose a specialized metabolism different from non-transformed somatic cells, leading to changes in the tumor microenvironment (TME). In recent years, our understanding of immune cell metabolism and cancer cell metabolism has gradually increased, and we have begun to explore the interaction between cancer cell metabolism and immune cell metabolism in a way which is meaningful for treatment. Understanding the different metabolic requirements of different cells that constitute the immune response to HNSCC is beneficial for revealing metabolic heterogeneity and plasticity, thereby enhancing the effect of immunotherapy. In this review, we have concluded that the relevant metabolic processes that affect the function of immune cells in HNSCC TME and proposed our own opinions and prospects on how to use metabolic intervention to enhance anti-tumor immune responses.
Collapse
Affiliation(s)
- Yi-Jia-Ning Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
31
|
Cui X, Song Y, Han J, Yuan Z. The multifaceted role of SMAD4 in immune cell function. Biochem Biophys Rep 2025; 41:101902. [PMID: 39802394 PMCID: PMC11721226 DOI: 10.1016/j.bbrep.2024.101902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/25/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
The Transforming Growth Factor-beta (TGF-β) signaling pathway, with SMAD4 as its central mediator, plays a pivotal role in regulating cellular functions, including growth, differentiation, apoptosis, and immune responses. While extensive research has elucidated SMAD4's role in tumorigenesis, its functions within immune cells remain underexplored. This review synthesizes current knowledge on SMAD4's diverse roles in various immune cells such as T cells, B cells, dendritic cells, and macrophages, highlighting its impact on immune homeostasis and pathogen response. Understanding SMAD4's role in immune cells is crucial, as its dysregulation can lead to autoimmune disorders, chronic inflammation, and immune deficiencies. The review emphasizes the significance of SMAD4 in immune regulation, proposing that deeper investigation could reveal novel therapeutic targets for immune-mediated conditions. Insights into SMAD4's involvement in processes like T cell differentiation, B cell class switch recombination, and macrophage polarization underscore its potential as a therapeutic target for a range of diseases, including autoimmune disorders and cancer.
Collapse
Affiliation(s)
- Xinmu Cui
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| | - Yu Song
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| | - Jianfeng Han
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
- Cellular Biomedicine Group Inc, Shanghai, 201203, China
| | - Zhaoxin Yuan
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| |
Collapse
|
32
|
Jin S, Li T, Liu L, Gao T, Zhang T, Yuan D, Di J, Guo Z, Luo Z, Yuan H, Liu J. V-domain immunoglobulin suppressor of T-cell activation and programmed death receptor 1 dual checkpoint blockade enhances antitumour immunity and survival in glioblastoma. Br J Pharmacol 2025; 182:1306-1323. [PMID: 39626657 DOI: 10.1111/bph.17404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/30/2024] [Accepted: 10/19/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE The current therapy cannot meet the needs of glioblastoma (GBM). V-domain immunoglobulin suppressor of T-cell activation (VISTA) is significantly up-regulated in GBM patients; however, its therapeutic potential in GBM is still unclear. EXPERIMENTAL APPROACH Flow cytometry was used to detect the expression of VISTA and the co-expression pattern of VISTA and programmed death receptor 1 (PD-1) on brain infiltrating lymphocytes of GBM mice. Monoclonal antibody therapy was used to evaluate the therapeutic effect of α-VISTA monotherapy and α-VISTA combined with α-PD-1 on GBM mice. Transcriptome analysis, flow cytometry, and immunofluorescence were used to detect changes of immune microenvironment in mouse brain tumours. Immunofluorescence and TCGA data analysis were used to further validate the combined treatment strategy on patient data. KEY RESULTS Compared with normal mice, the frequency of VISTA expression and co-expression of VISTA and PD-1 on tumour-infiltrating lymphocytes (TILs) in tumour-bearing mice was increased. Anti-VISTA monotherapy significantly up-regulated multiple immune stimulation-related pathways and moderately prolonged mouse survival time. Blocking the immune checkpoint VISTA and PD-1 significantly prolonged the survival time of mice and cured about 80% of the mice; CD8+ T cells played an important role in this process. In addition, we found that the expression of VISTA and PD-1 was significantly up-regulated in GBM patients by immunofluorescence, and patients with high expression of VISTA and PD-1 were associated with poor overall survival. This combination of blocking the immune checkpoint VISTA and PD-1 may achieve clinical transformation in GBM.
Collapse
Affiliation(s)
- Shasha Jin
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Tao Li
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Liu Liu
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ting Gao
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Tingting Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Dingyi Yuan
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jianwen Di
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhanying Guo
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhijie Luo
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Haoliang Yuan
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jun Liu
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
33
|
Wang Y, Liu C, Pang J, Li Z, Zhang J, Dong L. The Extra-Tumoral Vaccine Effects of Apoptotic Bodies in the Advancement of Cancer Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410503. [PMID: 39871756 DOI: 10.1002/smll.202410503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/16/2025] [Indexed: 01/29/2025]
Abstract
The induction of apoptosis in tumor cells is a common target for the development of anti-tumor therapies; however, these therapies still leave patients at increased risk of disease recurrence. For example, apoptotic tumor cells can promote tumor growth and immune evasion via the secretion of metabolites, apoptotic extracellular vesicles, and induction of pro-tumorigenic macrophages. This paradox of apoptosis induction and the pro-tumorigenic effects of tumor cell apoptosis has begged the question of whether apoptosis is a suitable cancer therapy, and led to further explorations into other immunogenic cell death-based approaches. However, these strategies still face multiple challenges, the most critical of which is the tumor microenvironment. Contrary to the promotion of immune tolerance mediated by apoptotic tumor cells, apoptotic bodies with enriched tumor-related antigens have demonstrated great immunogenic potential, as evidenced by their ability to initiate systemic T-cell immune responses. These characteristics indicate that apoptotic body-based therapies could be ideal "in situ" extra-tumoral tumor vaccine candidates for the treatment of cancers, and further address the current issues with apoptosis-based or immunotherapy treatments. Although not yet tested clinically, apoptotic body-based vaccines have the potential to better treatment strategies and patient outcomes in the future.
Collapse
Affiliation(s)
- Yulian Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Chunyan Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Jiayun Pang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Zhenjiang Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Chemistry and Biomedicine Innovative Center, Nanjing University, Nanjing, Jiangsu, 210023, China
| |
Collapse
|
34
|
Yu Z, Fu J, Mantareva V, Blažević I, Wu Y, Wen D, Battulga T, Wang Y, Zhang J. The role of tumor-derived exosomal LncRNA in tumor metastasis. Cancer Gene Ther 2025; 32:273-285. [PMID: 40011710 DOI: 10.1038/s41417-024-00852-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 10/22/2024] [Accepted: 11/05/2024] [Indexed: 02/28/2025]
Abstract
Tumor metastasis regulated by multiple complicated pathways is closely related to variations in the tumor microenvironment. Exosomes can regulate the tumor microenvironment through various mechanisms. Exosomes derived from tumor cells carry a variety of substances, including long non-coding RNAs (lncRNAs), play important roles in intercellular communication and act as critical determinants influencing tumor metastasis. In this review, we elaborate on several pivotal processes through which lncRNAs regulate tumor metastasis, including the regulation of epithelial‒mesenchymal transition, promotion of angiogenesis and lymphangiogenesis, enhancement of the stemness of tumor cells, and evasion of immune clearance. Additionally, we comprehensively summarized a diverse array of potential tumor-derived exosomal lncRNA biomarkers to facilitate accurate diagnosis and prognosis in a clinical setting.
Collapse
Affiliation(s)
- Zhile Yu
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Jiali Fu
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Vanya Mantareva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Bld. 9, 1113, Sofia, Bulgaria
| | - Ivica Blažević
- Department of Organic Chemistry, Faculty of Chemistry and Technology, University of Split, Ruđera Boškovića 35, 21000, Split, Croatia
| | - Yusong Wu
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Dianchang Wen
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Tungalag Battulga
- School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Yuqing Wang
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
- The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 510140, PR China
| | - Jianye Zhang
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China.
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, 511518, PR China.
| |
Collapse
|
35
|
Assi T, Moussa T, Ngo C, Faron M, Verret B, Lévy A, Honoré C, Hénon C, Péchoux CL, Bahleda R, Vibert J, Cesne AL. Therapeutic advances in Tenosynovial giant cell Tumor: Targeting the CSF1/CSF1R axis. Cancer Treat Rev 2025; 134:102904. [PMID: 40020639 DOI: 10.1016/j.ctrv.2025.102904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025]
Abstract
Tenosynovial giant cell tumor is a non-malignant primary locally aggressive articular disease that affects the synovium of joints, tendon sheaths, and bursae. It is characterized by a translocation t (1;2), leading to the overexpression of CSF1 in the tumor microenvironment. CSF1 induces the recruitment of non-malignant cells, mainly macrophages, followed by the differentiation and polarization of these cells into the M2 phenotype. Surgery, particularly total synovectomy, remains the cornerstone of TGCT management. However, recurrence rates vary, reaching 40 to 60% in diffuse disease, often resulting in progressive joint dysfunction, pain, and potential need for joint replacement or limb amputation. Systemic therapy is recommended in recurrent TGCT in patients not amenable to additional surgery. Targeting the CSF1/CSF1R axis has successfully improved tumor responses and enhanced symptomatic function. In this review, we aim to explore contemporary paradigms in inoperable TGCT patients, with a focus on the physiopathology, clinical efficacy, and safety of CSF1 or CSF1R inhibitors.
Collapse
Affiliation(s)
- Tarek Assi
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, France; Radiology Department, Gustave Roussy Cancer Campus, Villejuif, France.
| | - Tania Moussa
- Radiology Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Carine Ngo
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Matthieu Faron
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Benjamin Verret
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Antonin Lévy
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Charles Honoré
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Clémence Hénon
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | | | | | - Julien Vibert
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Axel Le Cesne
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, France; Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
36
|
Guo Y, Jin L, Shen Z, Fan L, Yu X, Kuang Y, Cai L, Zhou J, Chen Z, Yan F, Zhang J, Tong M, Yuan J, Mao Z, Chen G. Biomimetic Membrane Vesicles Reprogram Microglia Polarization and Remodel the Immunosuppressive Microenvironment of Glioblastoma via PERK/HIF-1α/Glycolysis Pathway. Adv Healthc Mater 2025; 14:e2404782. [PMID: 39757442 DOI: 10.1002/adhm.202404782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Indexed: 01/07/2025]
Abstract
The malignant interaction between tumor cells and immune cells is one of the important reasons for the rapid progression and refractoriness of glioblastoma (GBM). As an essential metabolic center of M2 macrophages, the inhibition of protein kinase RNA-like endoplasmic reticulum kinase (PERK) leads to the reduction of M2 macrophages. Nevertheless, the restriction of the blood-brain barrier (BBB) and non-specific cell targeting hinder the application of PERK inhibitors in GBM. Herein, the optimal NP-M-M2pep is developed successfully, which has shown the capacity of BBB penetration and specific targeting of M2 microglia. In addition to inhibiting the polarization of M2 microglia, the administration of iPERK@NP-M-M2pep reprogrammed M2 microglia into M1 ones in vitro via PERK/HIF-1α/glycolysis pathway. Efficient brain accumulation of nanoparticles is achieved after tail vein injection, with effective inhibition of GBM progression after one course of treatment. The glioma-associated microglia and macrophages (GAM) with M2 type are induced to M1 and the immunosuppressive TME is remodeled by upregulating immunostimulatory cells and downregulating immunosuppressive cells. In summary, the biomimetic membrane vesicles (BMVs) specifically delivered iPERK to GAMs offer an inspiring strategy to reprogram microglia polarization, re-educate immunosuppressive TME, and inhibit the progression of GBM.
Collapse
Affiliation(s)
- Yinghan Guo
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhipeng Shen
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Linfeng Fan
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Xian Yu
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Yirui Kuang
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Lingxin Cai
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Jiayin Zhou
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Zihang Chen
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Feng Yan
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Jianmin Zhang
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Minfeng Tong
- Department of Neurosurgery, affiliated Jinhua Hospital, School of Medicine, Zhejiang University, Jinhua, Zhejiang, 321000, China
| | - Jianlie Yuan
- Department of Neurosurgery, affiliated Jinhua Hospital, School of Medicine, Zhejiang University, Jinhua, Zhejiang, 321000, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Gao Chen
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| |
Collapse
|
37
|
Yu X, Qian J, Ding L, Pan C, Liu X, Wu Q, Wang S, Liu J, Shang M, Su R, Guo D, Xie H, Yin S, Zhou L, Zheng S. Galectin-1-Induced Tumor Associated Macrophages Repress Antitumor Immunity in Hepatocellular Carcinoma Through Recruitment of Tregs. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408788. [PMID: 39853961 PMCID: PMC11923918 DOI: 10.1002/advs.202408788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/12/2024] [Indexed: 01/26/2025]
Abstract
Tumor-associated macrophages (TAMs) are commonly considered accomplices in tumorigenesis and tumor development. However, the precise mechanism by which tumor cells prompt TAMs to aid in evading immune surveillance remains to be further investigated. Here, it is elucidated that tumor-secreted galectin-1 (Gal1) conferred immunosuppressive properties to TAMs. Specifically, patient specimens and a public database is first used to analyze the clinical relevance of Gal1 in hepatocellular carcinoma (HCC). Then, it is demonstrated that TAMs functioned as a critical mediator in the Gal1-induced progression of HCC and the establishment of an immunosuppressive tumor microenvironment. Furthermore, RNA-sequencing determined that Gal1 promoted the upregulation of chemokine (C-C motif) ligand 20 (CCL20) in TAMs via activating the PI3K/AKT/NF-κB pathway. Employing an anti-CCL20 neutralizing antibody and Foxp3DTR mice, it is demonstrated that CCR6+Foxp3+ regulatory T cells (Tregs) recruited by Gal1-induced TAMs contributed to reduced infiltration and dysfunctional state of CD8+ T cells, subsequently facilitating tumor progression. Targeting Gal1 dampened the secretion of CCL20 and inhibits the recruitment of Tregs, thereby activating anti-tumor immunity and ameliorating anti-PD-1 resistance. Together, this findings revealed that Gal1-induced TAMs recruited Tregs through the CCL20-CCR6 axis. Inhibition of Gal1 improves the effectiveness of anti-PD1 therapy, shedding important new light on the combination immunotherapy of HCC.
Collapse
Affiliation(s)
- Xizhi Yu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Zhejiang, 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, 310003, China
| | - Junjie Qian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Zhejiang, 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, 310003, China
| | - Limin Ding
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Zhejiang, 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, 310003, China
| | - Caixu Pan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Zhejiang, 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, 310003, China
| | - Xi Liu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Zhejiang, 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, 310003, China
| | - Qinchuan Wu
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Zhejiang, 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, 310003, China
| | - Shuai Wang
- Department of Hepatobiliary and Pancreatic Surgery, Department of Liver Transplantation, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310000, China
| | - Jianpeng Liu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Zhejiang, 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, 310003, China
| | - Mingge Shang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Zhejiang, 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, 310003, China
| | - Rong Su
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Zhejiang, 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, 310003, China
| | - Danjing Guo
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Zhejiang, 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, 310003, China
| | - Haiyang Xie
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Zhejiang, 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, 310003, China
| | - Shengyong Yin
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Zhejiang, 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, 310003, China
| | - Lin Zhou
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Zhejiang, 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, 310003, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Zhejiang, 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, 310003, China
- Department of Hepatobiliary and Pancreatic Surgery, Department of Liver Transplantation, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310000, China
| |
Collapse
|
38
|
Ruan L, Wang L. Adoptive cell therapy against tumor immune evasion: mechanisms, innovations, and future directions. Front Oncol 2025; 15:1530541. [PMID: 40094019 PMCID: PMC11906336 DOI: 10.3389/fonc.2025.1530541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
Tumors employ a range of strategies to evade detection and eradication by the host's immune system. These include downregulating antigen expression, altering antigen presentation processes, and inhibiting immune checkpoint pathways. etc. Adoptive Cell Therapy (ACT) represents a strategy that boosts anti-tumor immunity. This is achieved by amplifying or genetically engineering immune cells, which are either sourced from the patient or a donor, in a laboratory setting. Subsequently, these cells are reintroduced into the patient to bolster their immune response against cancer. ACT has successfully restored anti-tumor immune responses by amplifying the activity of T cells from patients or donors. This review focuses on the mechanisms underlying tumor escape, including alterations in tumor cell antigens, the immunosuppressive tumor microenvironment (TME), and modulation of immune checkpoint pathways. It further explores how ACT can avddress these factors to enhance therapeutic efficacy. Additionally, the review discusses the application of gene-editing technologies (such as CRISPR) in ACT, highlighting their potential to strengthen the anti-tumor capabilities of T cells. Looking forward, the personalized design of ACT, combined with immune checkpoint inhibitors and targeted therapies, is expected to significantly improve treatment outcomes, positioning this approach as a key strategy in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Liqin Ruan
- Department of Hepatobiliary Surgery, JiuJiang City Key Laboratory of Cell Therapy, JiuJiang No.1 People's Hospital, Jiujiang, Jiangxi, China
| | - Lu Wang
- Department of Oncology, JiuJiang City Key Laboratory of Cell Therapy, JiuJiang No.1 People's Hospital, Jiujiang, Jiangxi, China
| |
Collapse
|
39
|
Zhuang AB, Xi Z, Cheng YX, Zhang CH, Li WG. Current status and future perspectives of immunotherapy for abdominal liposarcoma: From basic research to clinical practice. Shijie Huaren Xiaohua Zazhi 2025; 33:81-88. [DOI: 10.11569/wcjd.v33.i2.81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/06/2024] [Accepted: 12/17/2024] [Indexed: 02/28/2025] Open
Abstract
Liposarcoma is a highly heterogeneous type of soft tissue sarcoma originating from adipose tissue, characterized by complex biological behavior and invasiveness. Traditional treatments have shown limited efficacy in high-grade and metastatic liposarcoma, with unsatisfactory patient outcomes. In recent years, the breakthroughs of immunotherapy in various solid tumors have sparked interest in its potential application to liposarcoma. This review systematically examines the progress in basic research and clinical practice of immunotherapy for liposarcoma, discussing the tumor immune microenvironment, mechanisms of immune evasion, the application of immune checkpoint inhibitors, combination therapy strategies, the challenges faced, as well as the future direction, with an aim to provide a theoretical basis for personalized treatment of liposarcoma, promote the development of novel immunotherapy strategies, and ultimately improve patient prognosis and quality of life.
Collapse
Affiliation(s)
- Ao-Bo Zhuang
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Zhe Xi
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Ying-Xue Cheng
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Chen-He Zhang
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Wen-Gang Li
- Department of Hepatobiliary and Pancreatic Surgery, Xiang'an Hospital of Xiamen University, Xiamen 361102, Fujian Province, China
- Cancer Research Center of Xiamen University, Xiamen 361005, Fujian Province, China
| |
Collapse
|
40
|
Zhang M, Zhou G, Xu Y, Wei B, Liu Q, Zhang G, Chang R. Immunogenic cell death signature predicts survival and reveals the role of VEGFA + Mast cells in lung adenocarcinoma. Sci Rep 2025; 15:7213. [PMID: 40021802 PMCID: PMC11871002 DOI: 10.1038/s41598-025-91401-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/20/2025] [Indexed: 03/03/2025] Open
Abstract
Lung cancer is prevalent worldwide and is a major cause of cancer-related mortality. Despite being the primary model for immunotherapy research, the response rates of lung cancer patients to immunotherapy are unsatisfactory. Furthermore, research on immunogenic cell death (ICD) in lung cancer is limited, which limits the development of strategies that combine ICD-related therapies with immunotherapy. In this study, we compiled and summarized 69 genes associated with ICD and developed an IRS. Across seven independent datasets, the IRS was identified as an independent prognostic factor. IRS was positively associated with multiple tumor proliferation pathways and negatively associated with immune-related pathways. Additionally, IRS negatively correlated with the infiltration of various immune cells, supporting its association with survival outcomes. Based on the correlation between IRS and immune activity, we validated the ability of IRS to predict immunotherapy efficacy across seven immunotherapy datasets and demonstrated that patients who respond to immunotherapy tend to have a lower IRS. Moreover, utilizing single-cell RNA sequencing, we revealed the role of mast cells in the TME with the highest IRS. Through interactions with various receptors on macrophages, endothelial cells, and tumor cells, mast cells promote tumor progression, providing a comprehensive explanation for poor prognosis and lack of response to immunotherapy in patients with high IRS. Our study offers new guidance for combination therapies in lung adenocarcinoma patients and elucidated the mechanism by which mast cells contribute to cancer development within the TME.
Collapse
Affiliation(s)
- Meng Zhang
- The Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Guowei Zhou
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yantao Xu
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Benliang Wei
- Big Data Institute, Central South University, Changsha, Hunan, China
| | - Qian Liu
- Big Data Institute, Central South University, Changsha, Hunan, China
| | - Guanxiong Zhang
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China.
- Furong Laboratory, Changsha, Hunan, China.
| | - Ruimin Chang
- The Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.
| |
Collapse
|
41
|
Das M, Kiruthiga C, Shafreen RB, Nachammai K, Selvaraj C, Langeswaran K. Harnessing the human microbiome and its impact on immuno-oncology and nanotechnology for next-generation cancer therapies. Eur J Pharmacol 2025; 996:177436. [PMID: 40023356 DOI: 10.1016/j.ejphar.2025.177436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
The integration of microbiome research and nanotechnology represents a significant advancement in immuno-oncology, potentially improving the effectiveness of cancer immunotherapies. Recent studies highlight the influential role of the human microbiome in modulating immune responses, presenting new opportunities to enhance immune checkpoint inhibitors (ICIs) and other cancer therapies. Nanotechnology offers precise drug delivery and immune modulation capabilities, minimizing off-target effects while maximizing therapeutic outcomes. This review consolidates current knowledge on the interactions between the microbiome and the immune system, emphasizing the microbiome's impact on ICIs, and explores the incorporation of nanotechnology in cancer treatment strategies. Additionally, it provides a forward-looking perspective on the synergistic potential of microbiome modulation and nanotechnology to overcome existing challenges in immuno-oncology. This integrated approach may enhance the personalization and effectiveness of next-generation cancer treatments, paving the way for transformative patient care.
Collapse
Affiliation(s)
- Mamali Das
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India
| | | | - R Beema Shafreen
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India
| | - Kathiresan Nachammai
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, India
| | - Chandrabose Selvaraj
- CsrDD Lab, Department of Microbiology, Dr. D. Y. Patil Medical College Hospital & Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed to Be University), Pimpri, Pune, 411018, India.
| | - K Langeswaran
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India; Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
42
|
Zhu R, Huang J, Qian F. The role of tumor-associated macrophages in lung cancer. Front Immunol 2025; 16:1556209. [PMID: 40079009 PMCID: PMC11897577 DOI: 10.3389/fimmu.2025.1556209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Lung cancer remains a leading cause of cancer-related deaths worldwide, necessitating innovative treatments. Tumor-associated macrophages (TAMs) are primary immunosuppressive effectors that foster tumor proliferation, angiogenesis, metastasis, and resistance to therapy. They are broadly categorized into proinflammatory M1 and tumor-promoting M2 phenotypes, with elevated M2 infiltration correlating with poor prognosis. Strategies aimed at inhibiting TAM recruitment, depleting TAMs, or reprogramming M2 to M1 are therefore highly promising. Key signaling pathways, such as CSF-1/CSF-1R, IL-4/IL-13-STAT6, TLRs, and CD47-SIRPα, regulate TAM polarization. Additionally, macrophage-based drug delivery systems permit targeted agent transport to hypoxic regions, enhancing therapy. Preclinical studies combining TAM-targeted therapies with chemotherapy or immune checkpoint inhibitors have yielded improved responses and prolonged survival. Several clinical trials have also reported benefits in previously unresponsive patients. Future work should clarify the roles of macrophage-derived exosomes, cytokines, and additional mediators in shaping the immunosuppressive tumor microenvironment. These insights will inform the design of next-generation drug carriers and optimize combination immunotherapies within precision medicine frameworks. Elucidating TAM phenotypes and their regulatory molecules remains central to developing novel strategies that curb tumor progression and ultimately improve outcomes in lung cancer. Importantly, macrophage-based immunomodulation may offer expanded treatment avenues.
Collapse
Affiliation(s)
| | | | - Fenhong Qian
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
43
|
Li P, Huang M, Li M, Li G, Ma Y, Zhao Y, Wang X, Zhang Y, Shi C. Combining molecular characteristics and therapeutic analysis of PDOs predict clinical responses and guide PDAC personalized treatment. J Exp Clin Cancer Res 2025; 44:72. [PMID: 40001264 PMCID: PMC11863571 DOI: 10.1186/s13046-025-03332-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND The emergence of targeted therapies and immunotherapy has broadened treatment options for patients with pancreatic ductal adenocarcinoma (PDAC). Despite this, traditional drug selection, predominantly relies on tumor markers and clinical staging, has underutilized these drugs due to ignoring patient genomic diversity. Patient-derived organoids (PDOs) and corresponding patient-derived organoid xenograft (PDOX) models offer a way to better understand and address this. METHODS In this study, we established PDOs and PDOX models from PDAC clinical samples. These models were analyzed using immunohistochemistry, H&E staining, and genomic profiling. Drug screening with 111 FDA-approved drugs was performed on PDOs, and drug responses in PDOs and PDOX models were compared to assess consistency with clinical treatment outcomes. Gene analysis was conducted to explore the molecular mechanisms underlying variations in drug responses. Additionally, by analyzing the sequencing results from various drug-sensitive groups, the identified differential gene-drug metabolism gene UGT1A10 were modulated in PDOs to evaluate its impact on drug efficacy. A co-culture system of PDOs with immune cells was developed to study the efficacy of immunotherapies. RESULTS PDOs and matched PDOX models retain the morphological, biological, and genomic characteristics of the primary tumor. Exome sequencing and RNA sequencing confirmed both the consistency and heterogeneity among the PDOs. High-throughput drug screening revealed significant variability in drug sensitivity across different organoids, yet PDOs and PDOX derived from the same patient exhibited a high degree of concordance in response to clinical chemotherapy agents. The gene expression analysis of PDOs with significant differences in drug sensitivity revealed UGT1A10 as a crucial regulator. The knockdown of UGT1A10 notably increased drug sensitivity. Furthermore, immune cells demonstrated specific cytotoxicity towards the organoids, underscoring the potential of the co-culture system for application in tumor immunotherapy. CONCLUSION Our results highlight the necessity for personalized treatment strategies that consider genomic diversity beyond tumor markers, thus validating the utility of PDOs and PDOX models in advancing PDAC research and personalized medicine.
Collapse
Affiliation(s)
- Peng Li
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
- Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi, 710032, PR China
| | - Minli Huang
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
- Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Mengyao Li
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
- Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Gen Li
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Yifan Ma
- Gansu University of Chinese Medicine, Lanzhou, 730030, China
| | - Yong Zhao
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi, 710032, PR China
| | - Xiaowu Wang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, PR China.
| | - Yongbin Zhang
- Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China.
- Animal Experiment Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi, 710032, PR China.
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
44
|
Khizar H, Ali K, Wang J. From silent partners to potential therapeutic targets: macrophages in colorectal cancer. Cancer Immunol Immunother 2025; 74:121. [PMID: 39998578 PMCID: PMC11861851 DOI: 10.1007/s00262-025-03965-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/30/2025] [Indexed: 02/27/2025]
Abstract
Cancer cells grow and survive in the tumor microenvironment, which is a complicated process. As a key part of how colorectal cancer (CRC) progresses, tumor-associated macrophages (TAMs) exhibit a double role. Through angiogenesis, this TAM can promote the growth of cancers. Although being able to modify and adjust immune cells is a great advantage, these cells can also exhibit anti-cancer properties including direct killing of cancer cells, presenting antigens, and aiding T cell-mediated responses. The delicate regulatory mechanisms between the immune system and tumors are composed of a complex network of pathways regulated by several factors including hypoxia, metabolic reprogramming, cytokine/chemokine signaling, and cell interactions. Decoding and figuring out these complex systems become significant in building targeted treatment programs. Targeting TAMs in CRC involves disrupting chemokine signaling or adhesion molecules, reprogramming them to an anti-tumor phenotype using TLR agonists, CD40 agonists, or metabolic modulation, and selectively removing TAM subsets that promote tumor growth. Multi-drug resistance, the absence of an accurate biomarker, and drug non-specificity are also major problems. Combining macrophage-targeted therapies with chemotherapy and immunotherapy may revolutionize treatment. Macrophage studies will advance with new technology and multi-omics methodologies to help us understand CRC and build specific and efficient treatments.
Collapse
Affiliation(s)
- Hayat Khizar
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Kamran Ali
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jianwei Wang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, 2nd Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310009, China.
| |
Collapse
|
45
|
Toghraie FS, Bayat M, Hosseini MS, Ramezani A. Tumor-infiltrating myeloid cells; mechanisms, functional significance, and targeting in cancer therapy. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01051-y. [PMID: 39998754 DOI: 10.1007/s13402-025-01051-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor-infiltrating myeloid cells (TIMs), which encompass tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), and tumor-associated dendritic cells (TADCs), are of great importance in tumor microenvironment (TME) and are integral to both pro- and anti-tumor immunity. Nevertheless, the phenotypic heterogeneity and functional plasticity of TIMs have posed challenges in fully understanding their complexity roles within the TME. Emerging evidence suggested that the presence of TIMs is frequently linked to prevention of cancer treatment and improvement of patient outcomes and survival. Given their pivotal function in the TME, TIMs have recently been recognized as critical targets for therapeutic approaches aimed at augmenting immunostimulatory myeloid cell populations while depleting or modifying those that are immunosuppressive. This review will explore the important properties of TIMs related to immunity, angiogenesis, and metastasis. We will also document the latest therapeutic strategies targeting TIMs in preclinical and clinical settings. Our objective is to illustrate the potential of TIMs as immunological targets that may improve the outcomes of existing cancer treatments.
Collapse
Affiliation(s)
- Fatemeh Sadat Toghraie
- Institute of Biotechnology, Faculty of the Environment and Natural Sciences, Brandenburg University of Technology, Cottbus, Germany
| | - Maryam Bayat
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Sadat Hosseini
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Amin Ramezani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
46
|
Cao X, Chen M, Fang T, Deng Y, Wang L, Wang H, Chen Z, Chen G. RSL3-loaded nanoparticles amplify the therapeutic potential of cold atmospheric plasma. J Nanobiotechnology 2025; 23:136. [PMID: 39994619 PMCID: PMC11849213 DOI: 10.1186/s12951-025-03211-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
Cold atmospheric plasma (CAP) has exhibited exciting potential for cancer treatment. Reactive oxygen and nitrogen species (RONS), the primary constituents in CAP, contribute to cancer cell death by elevating oxidative stress in cells. However, several intrinsic cellular antioxidant defense systems exist, such as the glutathione peroxidase 4 (GPX4) enzyme, which dampens the cell-killing efficacy of CAP. RAS-selective lethal 3 (RSL3), also known as a ferroptosis inducer, is a synthetic GPX4 inhibitor. Therefore, we hypothesized that RSL3 can amplify CAP-induced cell death by inhibition of GPX4. In this study, we showed that RSL3 loaded in poly (ethylene glycol)-block-poly(lactide-co-glycolide) (PLGA-PEG) nanoparticles can enhance CAP-induced cell deaths in 4T1 tumor cells. Furthermore, the combination of CAP and RSL3 also promoted cancer immunogenic cell death (ICD), induced dendritic cell (DC) maturation, and macrophage polarization, initiating tumor-specific T-cell mediated immune responses against tumors. For in vivo application, RSL3@NP was co-delivered with CAP via injectable Pluronic hydrogel. In 4T1-bearing mice, hydrogel-mediated delivery of CAP and RSL3-loaded nanoparticles can effectively elicit potent anti-tumor immune responses and inhibit tumor growth.
Collapse
Affiliation(s)
- Xiaona Cao
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- School of Nursing, Tianjin Medical University, Tianjin, China
| | - Mo Chen
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Tianxu Fang
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Yueyang Deng
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Li Wang
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Hanwen Wang
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Zhitong Chen
- Paul C Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Advanced Therapeutic Center, National Innovation Center for Advanced Medical Devices, Shenzhen, China
| | - Guojun Chen
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada.
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
| |
Collapse
|
47
|
Liu J, Cheng P, Xu C, Pu K. Molecular probes for in vivo optical imaging of immune cells. Nat Biomed Eng 2025:10.1038/s41551-024-01275-7. [PMID: 39984703 DOI: 10.1038/s41551-024-01275-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/23/2024] [Indexed: 02/23/2025]
Abstract
Advancing the understanding of the various roles and components of the immune system requires sophisticated methods and technology for the detection of immune cells in their natural states. Recent advancements in the development of molecular probes for optical imaging have paved the way for non-invasive visualization and real-time monitoring of immune responses and functions. Here we discuss recent progress in the development of molecular probes for the selective imaging of specific immune cells. We emphasize the design principles of the probes and their comparative performance when using various optical modalities across disease contexts. We highlight molecular probes for imaging tumour-infiltrating immune cells, and their applications in drug screening and in the prediction of therapeutic outcomes of cancer immunotherapies. We also discuss the use of these probes in visualizing immune cells in atherosclerosis, lung inflammation, allograft rejection and other immune-related conditions, and the translational opportunities and challenges of using optical molecular probes for further understanding of the immune system and disease diagnosis and prognosis.
Collapse
Affiliation(s)
- Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Penghui Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Cheng Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
48
|
Kuhl GC, Tangney M. Bacterial-Mediated In Situ Engineering of Tumour-Associated Macrophages for Cancer Immunotherapy. Cancers (Basel) 2025; 17:723. [PMID: 40075571 PMCID: PMC11899205 DOI: 10.3390/cancers17050723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/29/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Tumour-associated macrophages (TAMs) are critical components of the tumour microenvironment (TME), significantly influencing cancer progression and treatment resistance. This review aims to explore the innovative use of engineered bacteria to reprogram TAMs, enhancing their anti-tumour functions and improving therapeutic outcomes. METHODS We conducted a systematic review following a predefined protocol. Multiple databases were searched to identify relevant studies on TAMs, their phenotypic plasticity, and the use of engineered bacteria for reprogramming. Inclusion and exclusion criteria were applied to select studies, and data were extracted using standardised forms. Data synthesis was performed to summarise the findings, focusing on the mechanisms and therapeutic benefits of using non-pathogenic bacteria to modify TAMs. RESULTS The review summarises the findings that engineered bacteria can selectively target TAMs, promoting a shift from the tumour-promoting M2 phenotype to the tumour-fighting M1 phenotype. This reprogramming enhances pro-inflammatory responses and anti-tumour activity within the TME. Evidence from various studies indicates significant tumour regression and improved immune responses following bacterial therapy. CONCLUSIONS Reprogramming TAMs using engineered bacteria presents a promising strategy for cancer therapy. This approach leverages the natural targeting abilities of bacteria to modify TAMs directly within the tumour, potentially improving patient outcomes and offering new insights into immune-based cancer treatments. Further research is needed to optimise these methods and assess their clinical applicability.
Collapse
Affiliation(s)
- Gabriela Christina Kuhl
- Cancer Research @UCC, College of Medicine and Health, University College Cork, T12 K8AF Cork, Ireland;
| | - Mark Tangney
- Cancer Research @UCC, College of Medicine and Health, University College Cork, T12 K8AF Cork, Ireland;
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland
| |
Collapse
|
49
|
Deng X, Huang Y, Zhang J, Chen Y, Jiang F, Zhang Z, Li T, Hou L, Tan W, Li F. Histone lactylation regulates PRKN-Mediated mitophagy to promote M2 Macrophage polarization in bladder cancer. Int Immunopharmacol 2025; 148:114119. [PMID: 39854875 DOI: 10.1016/j.intimp.2025.114119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Bladder cancer (BCa), particularly muscle-invasive bladder cancer (MIBC), is associated with poor prognosis, partly because of immune evasion driven by M2 tumor-associated macrophages (TAMs). Understanding the regulatory mechanisms of M2 macrophage polarization via PRKN-mediated mitophagy and histone lactylation (H3K18la) is crucial for improving treatment strategies. METHODS A single-cell atlas from 46 human BCa samples was constructed to identify macrophage subpopulations. Bioinformatics analysis and experimental validation, including ChIP-seq and lactylation modulation assays, were used to investigate the role of PRKN in M2 macrophage polarization and its regulation by H3K18la. RESULTS Single-cell analysis revealed distinct macrophage subpopulations, including M1 and M2 types. PRKN was identified as a critical regulator of mitophagy in M2 macrophages, supporting their immunosuppressive function. Bulk RNA-seq and gene intersection analysis revealed a set of mitophagy-related macrophage polarization genes (Mito_Macro_RGs) enriched in mitophagy and immune pathways. Pseudotime analysis revealed that PRKN was upregulated during the M1-to-M2 transition. siRNA-mediated PRKN knockdown impaired M2 polarization, reducing the expression of CD206 and ARG1. ChIP-seq and histone lactylation modulation confirmed that H3K18la enhanced PRKN expression, promoting mitophagy and M2 polarization and thereby facilitating immune suppression and tumor progression. CONCLUSIONS Histone lactylation regulated PRKN-mediated mitophagy, promoting M2 macrophage polarization and contributing to immune evasion in BCa.
Collapse
Affiliation(s)
- Xiaolin Deng
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Yuan Huang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Jinge Zhang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Yuwen Chen
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Feifan Jiang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Zicai Zhang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Tanghua Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Lina Hou
- Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China.
| | - Wanlong Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China.
| | - Fei Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China.
| |
Collapse
|
50
|
Cassali GD, Nakagaki KYR, Salvi M, dos Reys MP, Rocha MAN, de Campos CB, Ferreira E, Rodrigues ACB, dos Reis DC, Damasceno KA, Estrela-Lima A. Canine, Feline, and Murine Mammary Tumors as a Model for Translational Research in Breast Cancer. Vet Sci 2025; 12:189. [PMID: 40005948 PMCID: PMC11860833 DOI: 10.3390/vetsci12020189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/06/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
In veterinary medicine, mammary tumors are the most common neoplasms in female dogs and the third most frequent in cats, representing a significant challenge. Efforts have been directed toward adopting standardized diagnostic criteria to better understand tumor behavior and progression in these species. Meanwhile, the use of animal models has substantially advanced the understanding of comparative mammary carcinogenesis. These models provide critical insights into factors responsible for the disease in humans, with the expectation that such factors can be identified and controlled. In this context, this review presents a work based mainly on articles published by a research group specializing in mammary pathology (Laboratory of Comparative Pathology-Department of General Pathology-ICB/UFMG) and its collaborators, complementing their results with literature findings. The publications were categorized into animal research, experimental research, and human research. These studies addressed topics such as diagnosis, prognostic and predictive factors, tumor microenvironment, inflammation associated with tumors, treatment approaches, and factors influencing tumor growth. The conceptual network analysis underscores the importance of in vivo breast cancer models, both experimental and spontaneous, for understanding tumor progression mechanisms and therapeutic responses, offering valuable contributions to veterinary and human oncology.
Collapse
Affiliation(s)
- Geovanni Dantas Cassali
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Karen Yumi Ribeiro Nakagaki
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Marisa Salvi
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Marina Possa dos Reys
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Marcos André Nino Rocha
- Escola de Medicina Veterinária e Zootecnia, Universidade Federal da Bahia, Salvador 40170-110, Bahia, Brazil
| | | | - Enio Ferreira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | | | - Diego Carlos dos Reis
- Division of Molecular Pathology, The Institute of Cancer Research, London SW7 3RP, UK
| | | | - Alessandra Estrela-Lima
- Escola de Medicina Veterinária e Zootecnia, Universidade Federal da Bahia, Salvador 40170-110, Bahia, Brazil
| |
Collapse
|