1
|
Yuan H, Qiu Y, Mei Z, Liu J, Wang L, Zhang K, Liu H, Zhu F. Cancer stem cells and tumor-associated macrophages: Interactions and therapeutic opportunities. Cancer Lett 2025; 624:217737. [PMID: 40274063 DOI: 10.1016/j.canlet.2025.217737] [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: 01/26/2025] [Revised: 03/28/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Cancer stem cells (CSCs) depend on the tumor microenvironment (TME) to sustain their stem-like properties by recruiting monocytes and reprogramming them into tumor-associated macrophages (TAMs), which in turn promote tumor progression. This review explores CSC-TAM interactions, emphasizing how CSCs drive monocyte recruitment and TAM polarization. We discuss how TAMs enhance CSC stemness and niche maintenance through chemokines, cytokines, exosome-mediated miRNA transfer, direct interactions, and extracellular matrix (ECM) remodeling. Furthermore, we examine therapeutic strategies targeting TAMs, including inhibiting TAM differentiation, reprogramming TAM polarization, and leveraging immune checkpoint blockade and CAR-macrophage immunotherapy to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Haitao Yuan
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, PR China
| | - Yun Qiu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, PR China
| | - Zijie Mei
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, PR China
| | - Jiaqing Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, PR China
| | - Lingna Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, PR China
| | - Kaiqing Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, PR China
| | - Huicong Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, PR China
| | - Fangfang Zhu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, PR China.
| |
Collapse
|
2
|
Feng Y, Jiang Y, Yang L, Lu D, Li N, Zhang Q, Yang H, Qin H, Zhang J, Gou X, Jiang F. Interactions and communications in lung tumour microenvironment: chemo/radiotherapy resistance mechanisms and therapeutic targets. J Drug Target 2025; 33:817-836. [PMID: 39815747 DOI: 10.1080/1061186x.2025.2453730] [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: 11/17/2024] [Revised: 12/28/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025]
Abstract
The lung tumour microenvironment (TME) is composed of various cell types, including cancer cells, stromal and immune cells, as well as extracellular matrix (ECM). These cells and surrounding ECM create a stiff, hypoxic, acidic and immunosuppressive microenvironment that can augment the resistance of lung tumours to different forms of cell death and facilitate invasion and metastasis. This environment can induce chemo/radiotherapy resistance by inducing anti-apoptosis mediators such as phosphoinositide 3-kinase (PI3K)/Akt, signal transducer and activator of transcription 3 (STAT3) and nuclear factor kappa B (NF-κB), leading to the exhaustion of antitumor immunity and further resistance to chemo/radiotherapy. In addition, lung tumour cells can resist chemo/radiotherapy by boosting multidrug resistance mechanisms and antioxidant defence systems within cancer cells and other TME components. In this review, we discuss the interactions and communications between these different components of the lung TME and also the effects of hypoxia, immune evasion and ECM remodelling on lung cancer resistance. Finally, we review the current strategies in preclinical and clinical studies, including the inhibition of checkpoint molecules, chemoattractants, cytokines, growth factors and immunosuppressive mediators such as programmed death 1 (PD-1), insulin-like growth factor 2 (IGF-2) for targeting the lung TME to overcome resistance to chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Yuan Feng
- Guangxi University of Chinese Medicine, Nanning, China
| | - Ying Jiang
- Department of Neurology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Lin Yang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Danni Lu
- Guangxi University of Chinese Medicine, Nanning, China
| | - Ning Li
- Guangxi University of Chinese Medicine, Nanning, China
| | - Qun Zhang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Haiyan Yang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Huiyuan Qin
- Guangxi University of Chinese Medicine, Nanning, China
| | - Jiaxin Zhang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Xinyun Gou
- Guangxi University of Chinese Medicine, Nanning, China
| | - Feng Jiang
- Science and Technology Department, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
3
|
Qiu X, Pan T, Kuang T, Shen Y, Zheng Y, Geng H, Ni B, Xia X, Zhu C, Zhang Z, Cao H, Tu L. DEPP1: A prognostic biomarker linked to stroma‑rich and immunosuppressive microenvironment, promoting oxaliplatin resistance in gastric cancer. Oncol Rep 2025; 54:82. [PMID: 40376989 PMCID: PMC12117316 DOI: 10.3892/or.2025.8915] [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: 08/09/2024] [Accepted: 01/02/2025] [Indexed: 05/18/2025] Open
Abstract
Decidual protein induced by progesterone (DEPP1) was identified to exert heterogeneous functions in several cancers, whereas its role in gastric cancer (GC) remains elusive. In the present study, differential expression analysis was conducted using three Gene Expression Omnibus datasets (GSE54129, GSE26942 and GSE3438). Validation of DEPP1 expression was performed using reverse transcription‑quantitative PCR, western blotting and immunofluorescence. Kaplan‑Meier survival and Cox regression analyses were employed to assess the association between DEPP1 expression and the prognosis of patients with GC. Immune infiltration analysis was conducted to explore the correlation between DEPP1 and the tumor microenvironment. The potential of DEPP1 to promote oxaliplatin resistance was assessed using flow cytometry, western blotting, and subcutaneous mouse models. DEPP1 was found to be significantly upregulated in the aforementioned cohorts, which was consistent with the clinical specimens of the present study, and it emerged as an independent risk factor for poor overall survival in patients with GC. A prognostic nomogram was developed to improve prognosis prediction. High DEPP1 expression correlated with increased infiltration of cancer‑associated fibroblasts, endothelial cells, and M2 macrophages, contributing to the development of a stroma‑rich and immunosuppressive microenvironment in GC. Furthermore, high DEPP1 expression was associated with reduced sensitivity to chemotherapy drugs in patients with GC. In vitro and in vivo experiments highlighted DEPP1's crucial role in promoting oxaliplatin resistance in GC. In conclusion, DEPP1 is identified as a promising prognostic biomarker linked to a stroma‑rich and immunosuppressive microenvironment, and it is critical in driving oxaliplatin resistance in GC. These findings may inform personalized therapeutic strategies for patients with GC.
Collapse
Affiliation(s)
- Xudong Qiu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Tao Pan
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Tian Kuang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Yanying Shen
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Yihan Zheng
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Haigang Geng
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Bo Ni
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Xiang Xia
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Chunchao Zhu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Zizhen Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Hui Cao
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Lin Tu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| |
Collapse
|
4
|
Xu L, Li K, Li J, Xu F, Liang S, Kong Y, Chen B. The crosstalk between lung adenocarcinoma cells and M2 macrophages promotes cancer cell development via the SFRS1/miR-708-5p/PD-L1 axis. Life Sci 2025; 371:123599. [PMID: 40185466 DOI: 10.1016/j.lfs.2025.123599] [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: 01/07/2025] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
This study aimed to elucidate the underlying mechanisms regarding microRNA-708-5p (miR-708-5p) in lung adenocarcinoma (LUAD). Here, the co-culture system of LUAD cells and macrophages, as well as a xenograft mouse model, were established. High levels of miR-708-5p were observed in LUAD. Exosomal miR-708-5p facilitated M2-like phenotype polarization, whereas miR-708-5p inhibition blocked the polarization. Exosomal miR-708-5p was identified as a pivotal signaling molecule for macrophages to mediate tumor cell proliferation, invasion, migration and IFN-γ production in T cells. In addition, miR708-5p was observed to induce PD-L1 expression, and PD-L1 silencing inhibited macrophage-induced tumor cell growth behavior and regulated CD8 T cell activity. In xenograft models, miR-708-5p inhibition and PD-L1 silencing attenuated macrophage-induced tumor growth, induced IFN-γ secretion and CD8 expression, and modulated the PTEN/AKT/mTOR pathway. In LUAD patients, there was an upregulation of both miR-708-5p and PD-L1 expression, accompanied by the activation of PTEN/AKT/mTOR. In conclusion, this study demonstrated the induction of M2 macrophage polarization and PD-L1 expression by exosomal miR-708-5p. We observed that exosomal miR-708-5p mediated the PTEN/AKT/mTOR pathway, diminished CD8 T cell activity and accelerated LUAD progression. The inhibition of specific exosomal miRNA secretion and anti-PD-L1 in the LUAD microenvironment may represent a promising avenue for LUAD immunotherapy.
Collapse
Affiliation(s)
- Li Xu
- The Second Department of Thoracic Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Kang Li
- The Second Department of Thoracic Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Jia Li
- The Second Department of Thoracic Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Fang Xu
- The Second Department of Thoracic Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Shuzhi Liang
- The Second Department of Thoracic Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Yi Kong
- The Second Department of Thoracic Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China.
| | - Bolin Chen
- The Second Department of Thoracic Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China.
| |
Collapse
|
5
|
Ye K, Shi G, Xu J, Qiao K, Dai Q, Huo Z, Cao Y, Liu W, Hu Y, Yan L, Zhu Y, Li P, Su R, Xu L, Mi Y. Olaparib reverses prostate cancer resistance to Rapamycin by promoting macrophage polarization towards the M1 phenotype. Mol Cell Biochem 2025; 480:3907-3921. [PMID: 39984794 DOI: 10.1007/s11010-025-05231-0] [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: 11/06/2024] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
Prostate cancer (PCa) is the most common non-cutaneous malignancy and the second leading cause of cancer-related death in men. Despite its prevalence, treatment outcomes are often unsatisfactory, necessitating the search for more effective therapeutic approaches. mTOR inhibitor Rapamycin (RAPA) has shown promise in managing PCa, but the emergence of resistance often undermines its long-term effectiveness. Recent studies suggest that poly ADP-ribose polymerase (PARP) inhibitor Olaparib (OLP) may overcome drug resistance in various tumor types. This study aims to assess the efficacy of OLP in treating RAPA-resistant PCa, with a specific focus on elucidating its underlying molecular mechanisms. This study utilized drug exposure and concentration escalation experiments to establish human RAPA-resistant PCa cell line (PC-3R) based on the human PCa cell line (PC-3). PC-3R cell lines were screened through a cloning assay. The efficacy of OLP in RAPA-resistant PCa, as well as its regulatory impact on tumor-associated macrophages (TAMs), was evaluated through a combination of real-time PCR, ELISA, immunohistochemistry, and fluorescence experiments. This study unveiled that the combination of OLP and RAPA effectively suppressed the proliferation, stemness, invasion, angiogenesis, apoptosis resistance, and anti-oxidative stress capacity of RAPA-resistant PCa. Additionally, it demonstrated the capacity of OLP to regulate macrophage polarization within the tumor microenvironment and reverse drug resistance to RAPA in PCa. The findings of this study lay a theoretical foundation for the potential utilization of OLP in the treatment of RAPA-resistant PCa, offering substantial academic significance and promising application prospects.
Collapse
Affiliation(s)
- Kai Ye
- Clinical School of the Second People'S Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Hepatology, Tianjin Second People'S Hospital, Tianjin, China
- Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin, China
| | - Gang Shi
- Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300021, China
| | - Jian Xu
- Clinical School of the Second People'S Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Hepatology, Tianjin Second People'S Hospital, Tianjin, China
- Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin, China
| | - Kunyan Qiao
- Clinical School of the Second People'S Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Hepatology, Tianjin Second People'S Hospital, Tianjin, China
- Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin, China
| | - Qinghai Dai
- Clinical School of the Second People'S Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Hepatology, Tianjin Second People'S Hospital, Tianjin, China
- Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin, China
| | - Zhixiao Huo
- Clinical School of the Second People'S Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Hepatology, Tianjin Second People'S Hospital, Tianjin, China
- Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin, China
| | - Yu Cao
- Clinical School of the Second People'S Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Hepatology, Tianjin Second People'S Hospital, Tianjin, China
- Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin, China
| | - Wei Liu
- Clinical School of the Second People'S Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Hepatology, Tianjin Second People'S Hospital, Tianjin, China
- Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin, China
| | - Yue Hu
- Clinical School of the Second People'S Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Hepatology, Tianjin Second People'S Hospital, Tianjin, China
- Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin, China
| | - Lihua Yan
- Clinical School of the Second People'S Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Hepatology, Tianjin Second People'S Hospital, Tianjin, China
- Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin, China
| | - Yu Zhu
- Department of Clinical Laboratory, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, The Third Central Hospital of Tianjin, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, 300170, China
| | - Ping Li
- Clinical School of the Second People'S Hospital, Tianjin Medical University, Tianjin, China.
- Tianjin Institute of Hepatology, Tianjin Second People'S Hospital, Tianjin, China.
- Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin, China.
| | - Rui Su
- Clinical School of the Second People'S Hospital, Tianjin Medical University, Tianjin, China.
- Tianjin Institute of Hepatology, Tianjin Second People'S Hospital, Tianjin, China.
- Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin, China.
- School of Precision Instruments and Opto-Electronics Engineering, Tianjin University, Tianjin, China.
| | - Liang Xu
- Clinical School of the Second People'S Hospital, Tianjin Medical University, Tianjin, China.
- Tianjin Institute of Hepatology, Tianjin Second People'S Hospital, Tianjin, China.
- Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin, China.
- Department of Hepatology & Oncology, Tianjin Second People's Hospital, Tianjin, China.
| | - Yuqiang Mi
- Clinical School of the Second People'S Hospital, Tianjin Medical University, Tianjin, China.
- Tianjin Institute of Hepatology, Tianjin Second People'S Hospital, Tianjin, China.
- Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin, China.
| |
Collapse
|
6
|
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] [MESH Headings] [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
|
7
|
Liu Z, Hu J, Han X, Li L, Niu H, Zhang X, Wang N, Shi X, Sang L, Zhang Q, Qian X. SLAMF8 regulates Fc receptor-mediated phagocytosis in mouse macrophage cells through PI3K-Akt signaling. Immunol Lett 2025; 273:106990. [PMID: 39983459 DOI: 10.1016/j.imlet.2025.106990] [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: 08/11/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
Emerging studies have demonstrated that phagocytosis checkpoints, which promote tumor-mediated immune evasion, are potential targets for cancer immunotherapy. In this study, the TCGA colorectal cancer (CRC) dataset and our RNA sequencing dataset suggested that SLAMF8 expression is significantly positively correlated with the expression levels of multiple phagocytosis checkpoint molecules. In vitro, we confirmed that SLAMF8 significantly regulated the phagocytosis of mouse CRC cells. RNA sequencing revealed that the expression of genes that promote Fc receptor (FcR)-mediated phagocytosis, such as FCGR1, FCGR3, FCGR2b, FCGR4, and ITGAM, was significantly upregulated after SLAMF8 knockdown. The Kyoto Encyclopedia of Genes and Genomes (KEGG) results suggested that the significantly enriched signaling pathways after SLAMF8 knockdown or overexpression included the PI3K-Akt signaling pathway. The protein expression levels of p-PI3K and p-Akt were significantly increased after SLAMF8 knockdown. When PI3K inhibitors and Fc blockers were added after SLAMF8 knockdown, mouse macrophage phagocytosis, and FcR expression decreased. Our results suggest that SLAMF8 may impair FcR-mediated phagocytosis through the PI3K-Akt signaling pathway and negatively regulate the antitumor immune response.
Collapse
Affiliation(s)
- Zhihao Liu
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing 210008, PR China; Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Jing Hu
- Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Xingzhi Han
- Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Li Li
- Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Haiqing Niu
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - Xin Zhang
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - Ning Wang
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - Xiao Shi
- Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Liuqi Sang
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing 210008, PR China
| | - Qun Zhang
- Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China.
| | - Xiaoping Qian
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing 210008, PR China; Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China; Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, PR China.
| |
Collapse
|
8
|
Li Q, Sheng M, Chen Y, Yi Q, Yang Z, Chen T. Comprehensive immunogenomic landscape analysis unveils CD33 + myeloid cell-driven immunomodulatory signatures in melanoma development. Pathol Res Pract 2025; 270:155981. [PMID: 40300524 DOI: 10.1016/j.prp.2025.155981] [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: 02/13/2025] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Understanding the causal relationships between immune cell populations and cancer development remains a critical challenge in tumor immunology. METHODS We employed Mendelian Randomization analysis leveraging genome-wide association studies of 612 immune cell traits and 91 cancer types to systematically evaluate causal associations. Single-cell RNA sequencing and computational deconvolution analyses were performed to characterize myeloid cell subpopulations in melanoma samples. FINDINGS Our analysis revealed significant relationships between specific immune cell subsets and cancer risk, particularly highlighting the role of CD33 + myeloid cells in melanoma pathogenesis. Single-cell RNA sequencing identified distinct CD33high myeloid subpopulations characterized by elevated expression of complement cascade components and chemokine signaling pathways. Through computational deconvolution of The Cancer Genome Atlas melanoma cohort, we demonstrated that elevated CD33high monocyte abundance correlates with increased immune dysfunction scores, reduced CD8 + T cell infiltration, and poor survival outcomes. INTERPRETATION Here we delineate the multifaceted mechanisms through which CD33 + myeloid cell populations orchestrate perturbations in the tumor-immune microenvironmental landscape, manifesting in compromised immunosurveillance and enhanced tumor progression. Our findings illuminate novel therapeutic opportunities through targeted modulation of myeloid cell function, while providing a systematic framework for understanding the complex interplay between immune cell populations and oncogenic processes.
Collapse
Affiliation(s)
- Qinke Li
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400010, China; Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Min Sheng
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yiqian Chen
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400010, China; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China
| | - Qiang Yi
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400010, China; Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Zhu Yang
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Tong Chen
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400010, China; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China.
| |
Collapse
|
9
|
Liu X, Jiang R, Xu Y, Xu X, Fang L, Gao G, Han L, Chen Y, Du H, Cai Y, Zhu F, Chen M, Wang K, Li H, Wang G, Quan C. Dual cytokine-engineered macrophages rejuvenate the tumor microenvironment and enhance anti-PD-1 therapy in renal cell carcinoma. Int Immunopharmacol 2025; 156:114725. [PMID: 40294469 DOI: 10.1016/j.intimp.2025.114725] [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: 03/05/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/30/2025]
Abstract
Despite advances in PD-1 blockade therapy, the immunosuppressive tumor microenvironment (TME) limits its efficacy in renal cell carcinoma (RCC). Here, we developed dual-cytokine-engineered macrophages co-delivering IL-12 and CXCL-9 to reprogram TME and enhance anti-PD-1 responsiveness. Single-cell RNA sequencing revealed that RCC harbor abundant M2-like tumor-associated macrophages (TAMs), which correlate with T-cell exhaustion. In vitro, engineered macrophages polarized M2-like TAMs to antitumor M1 phenotypes, secreted CXCL-9 to recruit cytotoxic T cells, and released IL-12 to amplify T/NK cell activation. In vivo, intravenously administered engineered macrophages homed to tumors, reshaped the TME by increasing CD8+ T cells, dendritic cells, and NK cells while reducing immunosuppressive Tregs and MDSCs. This approach synergized with PD-1 blockade, resulting in a 2.5-fold greater tumor growth inhibition compared to anti-PD-1 monotherapy. This dual-cytokine macrophage platform offers a novel strategy to overcome resistance to checkpoint inhibitors in RCC by delivering cytokine and remodeling TME, with implications for clinical translation.
Collapse
Affiliation(s)
- Xin Liu
- Tianjin institute of urology,Tianjin Medical University Second Hospital, Tianjin, China; Department of Urology,The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ranran Jiang
- Department of Oncology,The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yujun Xu
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaodi Xu
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lin Fang
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ge Gao
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lulu Han
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuxin Chen
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongwei Du
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ying Cai
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fei Zhu
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Biotherapy and National Clinical Research Center for Geriatrics, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mingjing Chen
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kaidi Wang
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hailong Li
- Department of Urology,The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Gang Wang
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Changyi Quan
- Tianjin institute of urology,Tianjin Medical University Second Hospital, Tianjin, China.
| |
Collapse
|
10
|
Li D, Rudloff U. Emerging therapeutics targeting tumor-associated macrophages for the treatment of solid organ cancers. Expert Opin Emerg Drugs 2025:1-39. [PMID: 40353504 DOI: 10.1080/14728214.2025.2504376] [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: 12/03/2024] [Revised: 04/29/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Over the last decade, immune checkpoint inhibitors (ICIs) like PD-1/PD-L1 or CTLA-4, which reinvigorate T cells for tumor control have become standard-of-care treatment options. In response to the increasingly recognized mechanisms of resistance to T cell activation in immunologically cold tumors, immuno-oncology drug development has started to shift beyond T cell approaches. These include tumor-associated macrophages (TAMs), a major pro-tumor immune cell population in the tumor microenvironment known to silence immune responses. AREAS COVERED Here we outline anti-TAM therapies in current development, either as monotherapy or in combination with other treatment modalities. We describe emerging drugs targeting TAMs under investigation in phase II and III testing with a focus on their distinguishing mechanism of action which include (1) reprogramming of TAMs toward anti-tumor function and immune surveillance, (2) blockade of recruitment, and (3) reduction and ablation of TAMs. EXPERT OPINION Several new immuno-oncology agents are under investigation to harness anti-tumor functions of TAMs. While robust anti-tumor efficacy of anti-TAM therapies across advanced solid organ cancers remains elusive to-date, TAM reprogramming therapies have yielded benefits in select cancers. The inherent heterogeneity of the diverse TAM population will require enhanced investments into biomarker-driven approaches to fully leverage its therapeutic potential.
Collapse
Affiliation(s)
- Dandan Li
- Developmental Therapeutics Branch (TDB), Biology Group, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Udo Rudloff
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
11
|
Lee C, Lee J, Jeong M, Nam D, Rhee I. Emerging strategies for targeting tumor-associated macrophages in glioblastoma: A focus on chemotaxis blockade. Life Sci 2025; 376:123762. [PMID: 40419107 DOI: 10.1016/j.lfs.2025.123762] [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: 03/04/2025] [Revised: 05/14/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025]
Abstract
Glioblastoma (GBM) remains one of the most aggressive and treatment-resistant brain tumors, with poor prognosis for affected patients. A key player in the GBM tumor microenvironment is the tumor-associated macrophage (TAM), which promotes tumor progression, immune evasion, and therapeutic resistance. The recruitment of TAMs to the tumor site is driven by specific chemotactic signals, including CSF-1/CSF-1R, CXCR4/CXCL12, and HGF/MET pathways. This review explores the current understanding of these chemotaxis mechanisms and their role in GBM progression. It highlights the potential therapeutic benefits of targeting TAM chemotaxis pathways to disrupt TAM infiltration, reduce immunosuppression, and enhance the efficacy of conventional treatments. Additionally, we discuss the preclinical and clinical evidence surrounding key inhibitors, such as PLX3397, AMD3100, and Crizotinib, which have shown promise in reprogramming TAMs and improving treatment outcomes in GBM. While these strategies offer hope for overcoming some of the challenges of GBM therapy, the review also addresses the limitations and obstacles in clinical translation, emphasizing the need for further research and the development of combination therapies to achieve sustained therapeutic benefit.
Collapse
Affiliation(s)
- Chaelin Lee
- Department of Biotechnology and Bioscience, Sejong University, Seoul, Republic of Korea
| | - Jaehyun Lee
- Department of Biotechnology and Bioscience, Sejong University, Seoul, Republic of Korea
| | - Moongyu Jeong
- Department of Biotechnology and Bioscience, Sejong University, Seoul, Republic of Korea
| | - Dayoung Nam
- Department of Biotechnology and Bioscience, Sejong University, Seoul, Republic of Korea
| | - Inmoo Rhee
- Department of Biotechnology and Bioscience, Sejong University, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Chen S, Li Y, Zhou Z, Saiding Q, Zhang Y, An S, Khan MM, Ji X, Qiao R, Tao W, Kong N, Chen W, Xie T. Macrophage hitchhiking nanomedicine for enhanced β-elemene delivery and tumor therapy. SCIENCE ADVANCES 2025; 11:eadw7191. [PMID: 40397726 PMCID: PMC12094207 DOI: 10.1126/sciadv.adw7191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/10/2025] [Indexed: 05/23/2025]
Abstract
Nanoparticle-based drug delivery systems hold promise for tumor therapy; however, they frequently encounter challenges such as low delivery efficiency and suboptimal efficacy. Engineered living cells can redirect drug delivery systems to effectively reach targeted sites. Here, we used living macrophages as vehicles, attaching them with GeS nanosheets (GeSNSs) carrying β-elemene for transport to tumor sites. GeSNSs act as efficient sonosensitizers, enhancing ultrasound-induced reactive oxygen species generation for treating 4T1 breast tumors. Notably, macrophage hitchhiking delivery of β-elemene-loaded GeSNSs not only achieves high accumulation in tumor regions and suppresses tumor growth under ultrasound treatment, but also effectively remodels the immunosuppressive tumor microenvironment by improving M1-like macrophage polarization and enhancing the populations of mature dendritic cells, CD4+, and CD8+ lymphocytes, thereby facilitating enhanced sonodynamic chemoimmunotherapy. These findings underscore the potential of macrophage hitchhiking strategy for drug delivery and suggest broader applicability of engineered living materials-mediated delivery technologies in disease therapy.
Collapse
Affiliation(s)
- Shuying Chen
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Chinese Medicines; and Collaborative Innovation Center of Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Zhuoming Zhou
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yiming Zhang
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Chinese Medicines; and Collaborative Innovation Center of Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Liangzhu Laboratory, Zhejiang University; Hangzhou, Zhejiang 311121, China
| | - Soohwan An
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Muhammad Muzamil Khan
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Ruirui Qiao
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072 Australia
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Na Kong
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Chinese Medicines; and Collaborative Innovation Center of Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Liangzhu Laboratory, Zhejiang University; Hangzhou, Zhejiang 311121, China
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tian Xie
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Chinese Medicines; and Collaborative Innovation Center of Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| |
Collapse
|
13
|
Wang T, Song W, Tang Y, Yi J, Pan H. Breaking the immune desert: Strategies for overcoming the immunological challenges of pancreatic cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189353. [PMID: 40412630 DOI: 10.1016/j.bbcan.2025.189353] [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: 11/08/2024] [Revised: 05/18/2025] [Accepted: 05/18/2025] [Indexed: 05/27/2025]
Abstract
Pancreatic cancer is characterised by its highly aggressive nature and extremely poor prognosis, with a uniquely complex tumour immune microenvironment that manifests as a prototypical "immune desert." This immune-desert phenotype primarily arises from the inherently low immunogenicity of the tumour, the formation of a dense fibrotic stroma, severe deficiency in immune cell infiltration, and profound immunosuppressive effects of the metabolic landscape. Specifically, dysregulated tryptophan metabolism, such as indoleamine 2,3-dioxygenase (IDO)-mediated catabolism, and excessive lactate accumulation contribute to impaired T-cell functionality. Collectively, these factors severely limit the efficacy of current immunotherapy strategies, particularly those based on immune checkpoint inhibitors, which have demonstrated significantly lower clinical response rates in pancreatic cancer than in other malignancies. In response to these therapeutic challenges, this review explores integrated treatment strategies that combine metabolic reprogramming, tumour microenvironment remodelling, and next-generation immune checkpoint blockades, such as LAG-3, TIM-3, and VISTA. These emerging approaches hold substantial promise for clinical application. For example, targeting key metabolic pathways, including glycolysis (Warburg effect) and glutamine metabolism, may help restore T-cell activity by alleviating metabolic stress within the tumour milieu. Additionally, localised administration of immune stimulators such as interleukin-12 (IL-12) and CD40 agonists may enhance immune cell infiltration and promote tumour-specific immune activation. Future research should prioritise large-scale, multicentre clinical trials to validate the therapeutic efficacy of these innovative strategies, aiming to achieve meaningful breakthroughs in pancreatic cancer immunotherapy and significantly improve long-term survival and clinical outcomes in affected patients.
Collapse
Affiliation(s)
- Tianming Wang
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China; Zhengning County Community Health Service Center, Qingyang 745300, Gansu Province, China
| | - Wenjing Song
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Yuan Tang
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Jianfeng Yi
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China.
| | - Haibang Pan
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China.
| |
Collapse
|
14
|
Zhang X, Sun P, Wang Y, Xu L, Li HY, Tong LJ, Tan C, Ding J, Yang CH, Meng LH. JMC14: a novel dual PI3Kδ/CSF1R inhibitor with potent antitumor activity in hematological and solid tumors. Acta Pharmacol Sin 2025:10.1038/s41401-025-01575-x. [PMID: 40389566 DOI: 10.1038/s41401-025-01575-x] [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: 01/25/2025] [Accepted: 04/22/2025] [Indexed: 05/21/2025]
Abstract
PI3Kδ, predominantly expressed in immune cells and markedly dysregulated in B-cell malignancies, emerges as a promising and well-validated therapeutic target in hematologic cancers. Meanwhile, CSF1R regulates the formation and polarization of tumor-associated macrophages (TAMs), facilitating immune suppression and tumor progression in various solid tumors. Although targeting PI3Kδ or CSF1R has shown promise, the clinical application is often constrained by off-target effects, toxicity, and limited efficacy, particularly in solid malignancies. In this study, we identified JMC14, a novel dual inhibitor targeting PI3Kδ and CSF1R with a distinct structure and favorable selectivity among human kinome, yielding IC50 values of 12 nM against PI3Kδ and 143 nM against CSF1R, respectively. JMC14 preferentially inhibited PI3Kδ-mediated signaling at the cellular level and exhibited robust antiproliferative activity across 10 lines of diffuse large B-cell lymphoma (DLBCL) cells, outperforming the approved PI3Kδ inhibitor idelalisib. Notably, its efficacy negatively correlated with the PI3Kα expression among the cell lines tested, suggesting a compensatory pathway mediated by PI3Kα. Daily oral administration of JMC14 (10, 30, or 100 mg/kg, for 21 days) dose-dependently suppressed tumor progression in xenografts derived from TMD8 cells and DLBCL patients, accompanied by good tolerance. Additionally, M-NFS-60 myeloid leukemia cells, which are dependent on the CSF-1-CSF1R axis for survival and proliferation, were effectively inhibited by JMC14 both in vitro and in vivo, further validating its inhibitory activity targeting CSF1R. Furthermore, JMC14 demonstrated potent antitumor activity in murine triple-negative breast cancer (TNBC), which was associated with its activity to reshape the immune microenvironment by reducing M2-like TAMs, enhancing CD8+ T cell infiltration. Collectively, these findings establish JMC14 as a potent dual PI3Kδ/CSF1R inhibitor with remarkable efficacy against both hematologic and solid malignancies with hyperactivation of PI3Kδ and/or CSF1R, highlighting the potential of JMC14 as a useful probe to dissect the interaction of PI3Kδ and CSF1R in tumor progression and immune reprogramming.
Collapse
Affiliation(s)
- Xi Zhang
- Division of Anti-tumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Pu Sun
- Division of Anti-tumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yi Wang
- Division of Anti-tumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lan Xu
- Division of Anti-tumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hui-Yu Li
- Division of Anti-tumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lin-Jiang Tong
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Cun Tan
- Division of Small-molecule Drug Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jian Ding
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Chun-Hao Yang
- Division of Small-molecule Drug Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Ling-Hua Meng
- Division of Anti-tumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
15
|
Chuang CH, Chen CT, Hsu CH, Shao YY. Carry-over effect of immunotherapy in patients with advanced hepatocellular carcinoma. Cancer Immunol Immunother 2025; 74:208. [PMID: 40377687 DOI: 10.1007/s00262-025-04052-w] [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: 12/29/2024] [Accepted: 04/10/2025] [Indexed: 05/18/2025]
Abstract
BACKGROUND Combination immunotherapy is the current standard for treating advanced hepatocellular carcinoma (HCC). The response elicited by upfront immune checkpoint inhibitors (ICIs) might influence the efficacy of salvage therapy, a phenomenon known as the carry-over effect. This effect is thought to stem from immune memory and sustained immune activation, providing extended protection against tumor progression and resulting in a durable response even after discontinuation of ICI. This study aimed to investigate the carry-over effect of first-line ICI therapy in patients with advanced HCC. METHODS Patients who received first-line ICI therapy for advanced HCC from December 2017 to December 2021 were included if they exhibited disease progression and received second-line systemic therapy. We analyzed the associations between clinical benefit (classified as complete, partial response and stable disease) of first-line ICI therapy, post-progression survival (PPS) and second-line progression-free survival (PFS). We used a historical cohort of patients receiving first-line multikinase inhibitor (MKI) for comparison. RESULTS A total of 137 patients were analyzed. We included 60 patients who received first-line ICI therapy, of which clinical benefit was detected in 46 (76.7%). Compared with patients without clinical benefit of first-line ICI therapy, patients with clinical benefit exhibited significantly longer PPS (median: 14.6 vs. 4.9 months, P = 0.024) and second-line PFS (median: 3.6 vs. 1.6 months, P = 0.027). In multivariate analysis, clinical benefit of first-line ICI therapy remained an independent predictor of PPS [hazard ratio (HR): 0.295, P = 0.005] and second-line PFS (HR: 0.484, P = 0.047). Conversely, clinical benefit was not associated with PPS among patients receiving first-line MKI therapy in both univariate and multivariate analysis in historical MKI cohort. CONCLUSIONS Clinical benefit of first-line ICI therapy was associated with PPS and second-line PFS in patients with advanced HCC, suggestive of the carry-over effect of ICI.
Collapse
Affiliation(s)
- Chien-Huai Chuang
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Oncology, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, 10002, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ching-Tso Chen
- Department of Oncology, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, 10002, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Oncology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Chih-Hung Hsu
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Oncology, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, 10002, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Yun Shao
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan.
- Department of Oncology, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, 10002, Taiwan.
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
16
|
Chen J, Cui N, He SH, Xia CY, Li WQ. Single-cell transcriptomics reveals metabolic remodeling and functional specialization in the immune microenvironment of bone tumors. J Transl Med 2025; 23:554. [PMID: 40380260 DOI: 10.1186/s12967-025-06346-0] [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: 12/23/2024] [Accepted: 03/03/2025] [Indexed: 05/19/2025] Open
Abstract
OBJECTIVE To investigate the metabolic remodeling and functional specialization of immune cells within the tumor microenvironment (TME) of bone tumors, including Ewing's sarcoma, osteosarcoma, and giant cell tumor of bone, through high-resolution single-cell RNA sequencing (scRNA-seq) analysis. METHODS Immune cells were isolated from 13 bone tumor samples and profiled via scRNA-seq to delineate cellular compositions, metabolic adaptations, and intercellular communication networks. Differential gene expression analysis, metabolic pathway enrichment, and pseudotime trajectory inference were employed to characterize functional states and differentiation processes of immune cell subsets. RESULTS We identified 12 major immune cell clusters with distinct functional and metabolic characteristics. Naïve T cells exhibited amino acid metabolism-dependent activation potential, whereas NK cells relied on lipid metabolism and the TCA cycle for cytotoxic activity. Macrophage subsets demonstrated functional divergence: C06 macrophages adopted lipid metabolism to facilitate immunosuppression and tissue repair, while C04 macrophages displayed pro-inflammatory characteristics associated with complement activation. Intercellular signaling analysis revealed FN1 as a central regulator of immune coordination, governing cell adhesion, migration, and homeostasis within the TME. CONCLUSION This study provides novel insights into the metabolic and functional plasticity of immune cells in bone tumor TMEs, underscoring the critical role of metabolic remodeling in immune regulation. Our findings highlight potential therapeutic targets for modulating immune cell function and offering new avenues to improve treatment outcomes for patients with bone tumors.
Collapse
Affiliation(s)
- Jun Chen
- Department of Pathology, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Na Cui
- Department of Pathology, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Shao-Hui He
- Department of Orthopaedic Oncology, No.905 Hospital of People's Liberation Army Navy, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Chun-Yan Xia
- Department of Pathology, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Wei-Qing Li
- Department of Pathology, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China.
| |
Collapse
|
17
|
Li YR, Zhu Y, Halladay T, Yang L. In vivo CAR engineering for immunotherapy. Nat Rev Immunol 2025:10.1038/s41577-025-01174-1. [PMID: 40379910 DOI: 10.1038/s41577-025-01174-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2025] [Indexed: 05/19/2025]
Abstract
Chimeric antigen receptor (CAR)-engineered immune cell therapy represents an important advance in cancer treatments. However, the complex ex vivo cell manufacturing process and stringent patient selection criteria curtail its widespread use. In vivo CAR engineering is emerging as a promising off-the-shelf therapy, providing advantages such as streamlined production, elimination of patient-specific manufacturing, reduced costs and simplified logistics. A large set of preclinical findings has inspired further investigation into treatments for hard-to-treat diseases such as solid tumours and has facilitated the development of advanced products to enhance in vivo CAR engineering efficacy, the persistence of the cellular therapeutic and safety. In this Review, we summarize current in vivo CAR engineering strategies, including nanoparticle-based and viral delivery systems as well as bioinstructive implantable scaffolds, and discuss their advantages and disadvantages. Additionally, we provide a systematic comparison between in vivo and conventional ex vivo CAR engineering methods and address the challenges and future prospects of in vivo CAR engineering.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tyler Halladay
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
18
|
Yang Y, Li S, To KKW, Zhu S, Wang F, Fu L. Tumor-associated macrophages remodel the suppressive tumor immune microenvironment and targeted therapy for immunotherapy. J Exp Clin Cancer Res 2025; 44:145. [PMID: 40380196 DOI: 10.1186/s13046-025-03377-9] [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: 01/17/2025] [Accepted: 03/27/2025] [Indexed: 05/19/2025] Open
Abstract
Despite the significant advances in the development of immune checkpoint inhibitors (ICI), primary and acquired ICI resistance remains the primary impediment to effective cancer immunotherapy. Residing in the tumor microenvironment (TME), tumor-associated macrophages (TAMs) play a pivotal role in tumor progression by regulating diverse signaling pathways. Notably, accumulating evidence has confirmed that TAMs interplay with various cellular components within the TME directly or indirectly to maintain the dynamic balance of the M1/M2 ratio and shape an immunosuppressive TME, consequently conferring immune evasion and immunotherapy tolerance. Detailed investigation of the communication network around TAMs could provide potential molecular targets and optimize ICI therapies. In this review, we systematically summarize the latest advances in understanding the origin and functional plasticity of TAMs, with a focus on the key signaling pathways driving macrophage polarization and the diverse stimuli that regulate this dynamic process. Moreover, we elaborate on the intricate interplay between TAMs and other cellular constituents within the TME, that is driving tumor initiation, progression and immune evasion, exploring novel targets for cancer immunotherapy. We further discuss current challenges and future research directions, emphasizing the need to decode TAM-TME interactions and translate preclinical findings into clinical breakthroughs. In conclusion, while TAM-targeted therapies hold significant promise for enhancing immunotherapy outcomes, addressing key challenges-such as TAM heterogeneity, context-dependent plasticity, and therapeutic resistance-remains critical to achieving optimal clinical efficacy.
Collapse
Affiliation(s)
- Yan Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Sijia Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Kenneth K W To
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, 999077, P.R. China
| | - Shuangli Zhu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
19
|
Wu T, Li X, Zheng F, Liu H, Yu Y. Intercellular communication between FAP+ fibroblasts and SPP1+ macrophages in prostate cancer via multi-omics. Front Immunol 2025; 16:1560998. [PMID: 40438108 PMCID: PMC12116517 DOI: 10.3389/fimmu.2025.1560998] [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: 01/15/2025] [Accepted: 04/23/2025] [Indexed: 06/01/2025] Open
Abstract
Background Prostate cancer (PCa) presents substantial heterogeneity and unpredictability in its progression. Despite therapeutic advancements, mortality from advanced PCa remains a significant challenge. Understanding the intercellular communication within the tumor microenvironment (TME) is critical for uncovering mechanisms driving tumorigenesis and identifying novel therapeutic targets. Methods We employed an integrative approach combining bulk RNA sequencing, single-cell RNA sequencing (scRNA-seq), and spatial transcriptomics to investigate interactions between FAP+ fibroblasts and tumor-associated macrophages in PCa. Key findings were validated using immunohistochemical and immunofluorescence staining techniques. Results Analysis of 23,519 scRNA-seq data from 23 prostate samples revealed a pronounced accumulation of FAP+ fibroblasts in tumor tissues. Spatial transcriptomics and bulk RNA sequencing demonstrated strong associations between FAP+ fibroblasts and SPP1+ macrophages. Notably, tumor-specific intercellular signaling pathways, such as CSF1/CSF1R and CXCL/ACKR1, were identified, highlighting their potential role in fostering an immunosuppressive TME. Conclusion Our findings unveil a distinct pattern of crosstalk between FAP+ fibroblasts and SPP1+ macrophages in PCa, shedding light on potential therapeutic targets for advanced PCa.
Collapse
Affiliation(s)
- Tingting Wu
- Department of General Surgery, Shenzhen Qianhai Taikang Hospital, Shenzhen, China
| | - Xinyu Li
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fei Zheng
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hanchao Liu
- Department of Andrology and Urology, Sir Run Shaw Hospital, affiliated with the Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yang Yu
- Department of General Surgery, Chifeng Hospital, Chifeng, Inner Mongolia, China
| |
Collapse
|
20
|
Feng W, Yang K, Zou Y, Xiao Z, Qian R, Qian R. Progress of ursolic acid on the regulation of macrophage: summary and prospect. Front Immunol 2025; 16:1576771. [PMID: 40421013 PMCID: PMC12104263 DOI: 10.3389/fimmu.2025.1576771] [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: 02/14/2025] [Accepted: 04/15/2025] [Indexed: 05/28/2025] Open
Abstract
Ursolic acid (UA), a prevalent pentacyclic triterpenoid found in numerous fruits and herbs, has garnered significant attention for its vital role in anti-inflammatory processes and immune regulation. The study of immune cells has consistently been a focal point, particularly regarding macrophages, which play crucial roles in antigen presentation, immunomodulation, the inflammatory response, and pathogen phagocytosis. This paper reveals the underlying regulatory effects of UA on the function of macrophages and the specific therapeutic effects of UA on a variety of diseases. Owing to the superior effect of UA on macrophages, different types of macrophages in different tissues have been described. Through the multifaceted regulation of macrophage function, UA may provide new ideas for the development of novel anti-inflammatory and immunomodulatory drugs. However, to facilitate its translation into actual medical means, the specific mechanism of UA in macrophages and its clinical application still need to be further studied.
Collapse
Affiliation(s)
- Wenjing Feng
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Kehong Yang
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Ying Zou
- Department of Anatomy, Anatomy Teaching Center of Hunan University of Chinese Medicine, Changsha, China
| | - Zhaohua Xiao
- Xiangya Hospital, Central South University, Changsha, China
| | - Rongkang Qian
- Department of Integrated Traditional Chinese and Western Medicine, Qian Rongkang Clinic, Loudi, China
| | - Ronghua Qian
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
21
|
Zeng J, Wang D, Tong Z, Li Z, Wang G, Du Y, Li J, Miao J, Chen S. Development of a prognostic model for osteosarcoma based on macrophage polarization-related genes using machine learning: implications for personalized therapy. Clin Exp Med 2025; 25:146. [PMID: 40343502 PMCID: PMC12064610 DOI: 10.1007/s10238-024-01530-w] [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: 08/31/2024] [Accepted: 11/25/2024] [Indexed: 05/11/2025]
Abstract
While neoadjuvant chemotherapy combined with surgical resection has improved the prognosis for patients with osteosarcoma, its impact on metastatic and recurrent cases remains limited. Immunotherapy is emerging as a promising alternative. However, the relationship between the phenotype of tumor-associated macrophages and the prognosis of osteosarcoma remains unclear. Differentially expressed gene during macrophage polarization were identified using the Monocle package. Weighted gene co-expression network analysis was conducted to select genes regulating macrophage polarization. The least absolute shrinkage and selection operator algorithm and multivariate Cox regression were used to construct long-term survival predictive strategies. Multiple machine learning algorithms identified target genes for pan-cancer analysis. Lentiviral transfection created stable strains with target gene knockdown, and CCK-8 and transwell migration assays verified the target gene's effects. Western blot and flow cytometry assessed the impact of target genes on macrophage polarization. A total of 141 genes regulating macrophage polarization were identified, from which eight genes were selected to construct prognostic models. Significant differences between high-risk and low-risk groups were observed in immune cell activation, immune-related signaling pathways, and immune function. The prognostic model and target gene were validated to provide more precise immunotherapy options for osteosarcoma and other tumors. BNIP3 knockdown decreased osteosarcoma cell proliferation and migration and promoted macrophage polarization to the M2 phenotype. The constructed prognostic model offers precise immunotherapy regimens and valuable insights into mechanisms underlying current studies. Furthermore, BNIP3 may serve as a potential immunotherapeutic target for osteosarcoma and other tumors.
Collapse
Affiliation(s)
- Jin Zeng
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Dong Wang
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - ZhaoChen Tong
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - ZiXin Li
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - GuoWei Wang
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - YuMeng Du
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Jinsong Li
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Jinglei Miao
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Shijie Chen
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China.
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
22
|
Wang Z, Feng C, Chen G, Cai Q. Astragaloside IV regulates macrophage polarization via the TLR4/NF-κB/STAT3 pathway to inhibit the malignant phenotype of renal clear cell carcinoma. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04219-3. [PMID: 40343452 DOI: 10.1007/s00210-025-04219-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/23/2025] [Indexed: 05/11/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common and aggressive type of kidney cancer. This study aimed to investigate the effect of astragaloside IV (AS-IV) on ccRCC. A variety of cell experimental techniques were used, inducing macrophage polarization, and detecting relevant indicators by flow cytometry, qRT-PCR, immunofluorescence, MTT, wound healing, Transwell, and western blot. A nude mouse xenograft tumor model was constructed for in vivo studies, and siRNA interference technology was used to explore the signaling pathway. The results demonstrated successful macrophage polarization, with AS-IV inhibiting M2 macrophage polarization and promoting the transition from M0 to M1 polarization. Additionally, AS-IV suppressed ccRCC cell proliferation, migration, and invasion, while reversing the malignant effects of M2 macrophages. The study further revealed that AS-IV inhibited M2 polarization through the TLR4/NF-κB/STAT3 signaling pathway. In vivo experiments showed that AS-IV inhibited the growth of ccRCC tumors. This study revealed that AS-IV influences macrophage polarization by regulating the TLR4/NF-κB/STAT3 signaling pathway, thereby inhibiting the malignant phenotype of ccRCC. This finding provides new insights and potential therapeutic strategies for the treatment of ccRCC.
Collapse
Affiliation(s)
- Zhanshi Wang
- Department of Urology, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Xihu District, Hangzhou City, Zhejiang Province, 330012, China
| | - Chao Feng
- Department of Urology, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Xihu District, Hangzhou City, Zhejiang Province, 330012, China
| | - Guodong Chen
- Department of Urology, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Xihu District, Hangzhou City, Zhejiang Province, 330012, China
| | - Qi Cai
- Department of Urology, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Xihu District, Hangzhou City, Zhejiang Province, 330012, China.
| |
Collapse
|
23
|
Hsu CY, Jasim SA, Rasool KH, H M, Kaur J, Jabir MS, Alhajlah S, Kumar A, Jawad SF, Husseen B. Divergent functions of TLRs in gastrointestinal (GI) cancer: Overview of their diagnostic, prognostic and therapeutic value. Semin Oncol 2025; 52:152344. [PMID: 40347779 DOI: 10.1016/j.seminoncol.2025.152344] [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: 02/01/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 05/14/2025]
Abstract
The relationship between the innate immune signal and the start of the adaptive immune response is the central idea of this theory. By controlling the inflammatory and tissue-repair reactions to damage, the Toll-like receptors (TLRs), as a family of PRRs, have attracted increasing attention for its function in protecting the host against infection and preserving tissue homeostasis. Microbial infection, damage, inflammation, and tissue healing have all been linked to the development of malignancies, especially gastrointestinal (GI) cancers. Recently, increased studies on TLR recognition and binding, as well as their ligands, have significantly advanced our knowledge of the various TLR signaling pathways and offered therapy options for GI malignancies. Upon activation by pathogen-associated or damage-associated molecular patterns (DAMPs and PAMPs), TLRs trigger key pathways like NF-κB, MAPK, and IRF. NF-κB activation promotes inflammation, cell survival, and proliferation, often contributing to tumor growth, metastasis, and therapy resistance. MAPK pathways similarly drive uncontrolled cell growth and invasion, while IRF pathways modulate interferon production, yielding both anti-tumor and protumor effects. The resulting chronic inflammatory environment within tumors can foster progression, yet TLR activation can also stimulate beneficial anti-tumor immune responses. However, the functions of TLR expression in GI cancers and their diagnostic and prognostic along with therapeutic value have not yet entirely been elucidated. Understanding how TLR activation contributes to anti-cancer immunity against GI malignancies may hasten immunotherapy developments and increase patient survival.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, Arizona, USA
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-maarif University College, Anbar, Iraq; Biotechnology Department, College of Applied Science, Fallujah University, Fallujah, Iraq
| | - Khetam Habeeb Rasool
- Department of Biology, College of Science, University of Mustansiriyah, Mustansiriyah, Iraq
| | - Malathi H
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Jaswinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Mohali, Punjab, India
| | - Majid S Jabir
- Department of Applied Sciences, University of Technology, Anbar, Iraq
| | - Sharif Alhajlah
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia.
| | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named after the First President of Russia Boris Yeltsin, Ekaterinburg, Russia; Centre for Research Impact & Outcome, Chitkara University, Rajpura, Punjab, India; Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - Sabrean F Jawad
- Department of Pharmacy, Al-Mustaqbal University College, Hillah, Babylon, Iraq
| | - Beneen Husseen
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq; Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
24
|
Liu S, Hu L, Hu J, Qin C, Jiang C, Yu Y. Roles of macrophages and monocytes in resistance to immunotherapy in breast cancers. Postgrad Med J 2025:qgaf065. [PMID: 40327894 DOI: 10.1093/postmj/qgaf065] [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: 01/09/2025] [Revised: 02/20/2025] [Accepted: 03/23/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Immunotherapy is increasingly integral to breast cancer treatment, yet a subset develops resistance, partly mediated by macrophages and monocytes in the tumor immune microenvironment. While macrophages play essential roles in phagocytosis and pathogen clearance, their dual role in breast cancer-acting as both barriers to therapy and potential therapeutic targets-complicates treatment efficacy. STRATEGY Tumor-associated macrophages, polarized by tumor-derived signals, promote cancer progression and metastasis. Monocytes, subdivided into CD14+CD16- and CD14+CD16+ subsets, exhibit distinct functional profiles in cytokine secretion, antigen presentation, and migration. Modulating monocyte subset dynamics and functionality may enhance immunotherapy responsiveness. CONCLUSION A multimodal strategy targeting macrophages, monocytes, and complementary immunotherapies offers promising avenues to overcome resistance. Further research into the heterogeneity and regulatory mechanisms of these cells is critical for developing optimized, safe immunotherapeutic protocols. This review underscores the necessity of combination immunotherapies to improve outcomes in breast cancer.
Collapse
Affiliation(s)
- Siyuan Liu
- Department of Breast Surgery, Zhejiang Cancer Hospital, 1 Banshan East Road, Hangzhou, 310022, Zhejiang, China
- Postgraduate training base Alliance of Wenzhou Medical University, Higher Education Park, Chashan street, Ouhai District, Wenzhou, 325035, Zhejiang, China
| | - Lihong Hu
- Department of Breast Surgery, Zhejiang Cancer Hospital, 1 Banshan East Road, Hangzhou, 310022, Zhejiang, China
- Postgraduate training base Alliance of Wenzhou Medical University, Higher Education Park, Chashan street, Ouhai District, Wenzhou, 325035, Zhejiang, China
| | - Jiejie Hu
- Department of Breast Surgery, Zhejiang Cancer Hospital, 1 Banshan East Road, Hangzhou, 310022, Zhejiang, China
| | - Chengdong Qin
- Department of Breast Surgery, Zhejiang Cancer Hospital, 1 Banshan East Road, Hangzhou, 310022, Zhejiang, China
| | - Chuner Jiang
- Department of Breast Surgery Nurse, Zhejiang Cancer Hospital, 1 Banshan East Road, Hangzhou, 310022, Zhejiang, China
| | - Yang Yu
- Department of Breast Surgery, Zhejiang Cancer Hospital, 1 Banshan East Road, Hangzhou, 310022, Zhejiang, China
| |
Collapse
|
25
|
Xu L, Yang B, Zhang J, Liu S, Zhang Q, Ran L, Li B. Targeting ALPPL2 with a novel CD89 bispecific antibody reprograms macrophages to enhance anti-tumor immunity. Biochem Biophys Res Commun 2025; 762:151761. [PMID: 40209501 DOI: 10.1016/j.bbrc.2025.151761] [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: 04/02/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/12/2025]
Abstract
Immunotherapy holds promise for cancer treatment, but its efficacy in solid tumors is often limited by the immunosuppressive tumor microenvironment (TME). Macrophages, abundant within the TME, can be reprogrammed to elicit anti-tumor immunity. We developed a novel bispecific antibody, ALPPL2-CD89, to specifically target and activate macrophages within the tumor. The ALPPL2-CD89 bispecific antibody demonstrated high binding affinity to both targets and significantly enhanced macrophage-mediated phagocytosis of tumor cells. In vivo studies using human CD89 transgenic mice bearing ALPPL2-expressing tumors showed significant tumor growth inhibition. Analysis of the tumor microenvironment revealed that ALPPL2-CD89 treatment increased CD3+ and CD8+ T cell infiltration, and shifted tumor-associated macrophages toward a pro-inflammatory M1 phenotype. Our findings establish ALPPL2-CD89 as a promising therapeutic candidate that effectively reprograms the myeloid compartment to drive potent anti-tumor immunity against ALPPL2-positive malignancies.
Collapse
Affiliation(s)
- Lijun Xu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Bowen Yang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Junhan Zhang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Shujian Liu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Qi Zhang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Longchao Ran
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Bingyu Li
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China; The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan, China.
| |
Collapse
|
26
|
Jiang H, Pang J, Li T, Akofala A, Zhou X, Yi C, Ning S, Gao X, Qiao Y, Kou J. PD-1 regulates the anti-tumor immune function of macrophages through JAK2-STAT3 signaling pathway in colorectal cancer tumor microenvironment. J Transl Med 2025; 23:502. [PMID: 40317043 PMCID: PMC12048993 DOI: 10.1186/s12967-025-06469-4] [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: 01/24/2025] [Accepted: 04/07/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs), as key immune components of the TME, play a pivotal role in tumor progression by fostering an immunosuppressive environment. Programmed death 1 (PD-1), a critical immune checkpoint molecule predominantly expressed on T cells, mediates immune suppression by binding to programmed death-ligand 1 (PD-L1) on tumor cells within the tumor microenvironment (TME). Emerging evidence reveals that TAMs also express PD-1, however, the expression and functional regulatory mechanisms of PD-1 on TAM remain poorly understood. METHODS In this study, we combined bulk RNA sequencing and single-cell RNA sequencing (scRNA-seq) data to investigate the association between PD-1 expression on macrophages and patient prognosis, while also uncovering the molecular mechanisms by which PD-1 regulates macrophage function. To further investigate the role of PD-1 in macrophage activity, we established a fluorescence-labeled tumor-bearing mouse model using CT26 cells, a murine colorectal cancer cell line, to evaluate the relationship between PD-1 expression on TAMs and their phagocytic activity as well as other functions. Additionally, to mimic the TME in vitro, we cultured bone marrow-derived macrophages (BMDMs) with CT26-conditioned medium (CT26-CM). RESULTS Our results suggest that PD-1 expression on TAMs drives macrophage polarization toward an M2-like phenotype, suppresses their phagocytic activity, inhibits the synthesis of interferon-γ (IFN-γ) signaling molecules, and ultimately promotes tumor progression. Mechanistically, we demonstrated that PD-1 regulates the synthesis of IFN-γ signaling molecules and the polarization of M2-type macrophages in BMDMs through the JAK2-STAT3 signaling pathway. Overall, our study demonstrates that PD-1 expression on TAMs facilitates the formation of an immunosuppressive microenvironment, ultimately accelerating tumor progression. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Han Jiang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, China
- Department of BioinformaticsScience and Technology, Harbin Medical University, Harbin, 150000, China
| | - Jingjing Pang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, China
| | - Tengyue Li
- Department of BioinformaticsScience and Technology, Harbin Medical University, Harbin, 150000, China
| | - Atitso Akofala
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, China
| | - Xiaoxi Zhou
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Changhua Yi
- The Second Hospital of Nanjing, Nanjing, 210003, China
| | - Shangwei Ning
- Department of BioinformaticsScience and Technology, Harbin Medical University, Harbin, 150000, China.
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, China.
- Basic Medical Institute of Heilongjiang Medical Sciences Academy, Harbin, 150086, China.
| | - Yu Qiao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, China.
| | - Jiayuan Kou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, China.
| |
Collapse
|
27
|
Summer M, Riaz S, Ali S, Noor Q, Ashraf R, Khan RRM. Understanding the Dual Role of Macrophages in Tumor Growth and Therapy: A Mechanistic Review. Chem Biodivers 2025; 22:e202402976. [PMID: 39869825 DOI: 10.1002/cbdv.202402976] [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: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/29/2025]
Abstract
Macrophages are heterogeneous cells that are the mediators of tissue homeostasis. These immune cells originated from monocytes and are classified into two basic categories, M1 and M2 macrophages. M1 macrophages exhibit anti-tumorous inflammatory reactions due to the behavior of phagocytosis. M2 macrophages or tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME) and have a basic role in tumor progression by interacting with other immune cells in TME. By the expression of various cytokines, chemokines, and growth factors, TAMs lead to strengthening tumor cell proliferation, angiogenesis, and suppression of the immune system which further support invasion and metastasis. This review discusses recent and updated mechanisms regarding tumor progression by M2 macrophages. Moreover, the current therapeutic approaches targeting TAMs, their advantages, and limitations are also summarized, and further treatment approaches are outlined along with an elaboration of the tumor regression role of macrophages. This comprehensive review article possibly helps to understand the mechanisms underlying the tumor progression and regression role of macrophages in a comparative way from a basic level to the advanced one.
Collapse
Affiliation(s)
- Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Saima Riaz
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Qudsia Noor
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Rimsha Ashraf
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Rana Rashad Mahmood Khan
- Faculty of Chemistry and Life Sciences, Department of Chemistry, Government College University Lahore, Lahore, Pakistan
| |
Collapse
|
28
|
Aquino A, Franzese O. Reciprocal Modulation of Tumour and Immune Cell Motility: Uncovering Dynamic Interplays and Therapeutic Approaches. Cancers (Basel) 2025; 17:1547. [PMID: 40361472 PMCID: PMC12072109 DOI: 10.3390/cancers17091547] [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: 03/31/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Dysregulated cell movement is a hallmark of cancer progression and metastasis, the leading cause of cancer-related mortality. The metastatic cascade involves tumour cell migration, invasion, intravasation, dissemination, and colonisation of distant organs. These processes are influenced by reciprocal interactions between cancer cells and the tumour microenvironment (TME), including immune cells, stromal components, and extracellular matrix proteins. The epithelial-mesenchymal transition (EMT) plays a crucial role in providing cancer cells with invasive and stem-like properties, promoting dissemination and resistance to apoptosis. Conversely, the mesenchymal-epithelial transition (MET) facilitates metastatic colonisation and tumour re-initiation. Immune cells within the TME contribute to either anti-tumour response or immune evasion. These cells secrete cytokines, chemokines, and growth factors that shape the immune landscape and influence responses to immunotherapy. Notably, immune checkpoint blockade (ICB) has transformed cancer treatment, yet its efficacy is often dictated by the immune composition of the tumour site. Elucidating the molecular cross-talk between immune and cancer cells, identifying predictive biomarkers for ICB response, and developing strategies to convert cold tumours into immune-active environments is critical to overcoming resistance to immunotherapy and improving patient survival.
Collapse
Affiliation(s)
| | - Ornella Franzese
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| |
Collapse
|
29
|
Xu Y, Wang B, Huang Y, Liao J, Wu C, Zhou C, Kang Z, Jiang S, Wu B, Zhang D, Xu R, Liu X, Wang F. Targeting Antigen-Presenting Cells to Enhance the Tumor-Spleen Immunity Cycle through Liposome-Neoantigen Vaccine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500021. [PMID: 40125791 PMCID: PMC12097013 DOI: 10.1002/advs.202500021] [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] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 03/03/2025] [Indexed: 03/25/2025]
Abstract
Effective immune responses in both the spleen and the tumor microenvironment are crucial for cancer immunotherapy. However, delivery of neoantigen peptide vaccines to antigen-presenting cells (APCs) at these sites remains challenging. In this study, LNPsD18, a cationic liposomal formulation that targets and enhances APC uptake at both sites without modifying the targeting ligands is developed. By co-delivering tumor-specific neoantigens and a cholesterol-coupled toll-like receptor 9 (TLR9) agonist within LNP-vaxD18, an approximately 60-fold increase in dendritic cell uptake compared to neoantigen-adjuvant mixtures is achieved. Intravenous administration of the liposome-neoantigen peptide vaccine targets both the spleen and the tumor, boosting splenic DC activation, increasing M1-type tumor-associated macrophages, and elevating tumor cytokine levels. This reshapes the tumor microenvironment, enhancing IFN-γ-producing CD8+ T cells and TCF1+CD8+ T cells within tumors. These outcomes significantly inhibit established tumor growth compared to nontargeted lipid-based nanovaccine formulations, resulting in improved survival in orthotopic hepatocellular carcinoma and colorectal cancer models. The findings highlight the importance of targeting APCs in both the spleen and tumors to optimize the therapeutic efficacy of liposome-neoantigen vaccines in cancer treatment.
Collapse
Affiliation(s)
- Yu Xu
- Department of Medical OncologySun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510060P. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhou510060P. R. China
| | - Bing Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Fujian Agriculture and Forestry UniversityFuzhou350002P. R. China
| | - Yue Huang
- Department of Medical OncologySun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510060P. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhou510060P. R. China
| | - JianPing Liao
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510060P. R. China
| | - Chenyi Wu
- Department of Medical OncologySun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510060P. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhou510060P. R. China
| | - Chenxi Zhou
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Zishi Kang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Shiyang Jiang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Bing‐Chen Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Da Zhang
- Department of Medical OncologySun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510060P. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhou510060P. R. China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Ruihua Xu
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhou510060P. R. China
- Department of Medical OncologySun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen UniversityGuangzhou510060P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- CAS Key Laboratory of Design and Assembly of Functional NanostructuresFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhou350002P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Feng Wang
- Department of Medical OncologySun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510060P. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhou510060P. R. China
| |
Collapse
|
30
|
Shi X, Askari Rizvi SF, Yang Y, Liu G. Emerging nanomedicines for macrophage-mediated cancer therapy. Biomaterials 2025; 316:123028. [PMID: 39693782 DOI: 10.1016/j.biomaterials.2024.123028] [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: 10/18/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Tumor-associated macrophages (TAMs) contribute to tumor progression by promoting angiogenesis, remodeling the tumor extracellular matrix, inducing tumor invasion and metastasis, as well as immune evasion. Due to the high plasticity of TAMs, they can polarize into different phenotypes with distinct functions, which are primarily categorized as the pro-inflammatory, anti-tumor M1 type, and the anti-inflammatory, pro-tumor M2 type. Notably, anti-tumor macrophages not only directly phagocytize tumor cells, but also present tumor-specific antigens and activate adaptive immunity. Therefore, targeted regulation of TAMs to unleash their potential anti-tumor capabilities is crucial for improving the efficacy of cancer immunotherapy. Nanomedicine serves as a promising vehicle and can inherently interact with TAMs, hence, emerging as a new paradigm in cancer immunotherapy. Due to their controllable structures and properties, nanomedicines offer a plethora of advantages over conventional drugs, thus enhancing the balance between efficacy and toxicity. In this review, we provide an overview of the hallmarks of TAMs and discuss nanomedicines for targeting TAMs with a focus on inhibiting recruitment, depleting and reprogramming TAMs, enhancing phagocytosis, engineering macrophages, as well as targeting TAMs for tumor imaging. We also discuss the challenges and clinical potentials of nanomedicines for targeting TAMs, aiming to advance the exploitation of nanomedicine for cancer immunotherapy.
Collapse
Affiliation(s)
- Xueying Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Syed Faheem Askari Rizvi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China; Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, 54000, Punjab, Pakistan
| | - Yinxian Yang
- School of Pharmaceutical Sciences, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| |
Collapse
|
31
|
Du Z, Wei W, Lu S, Wang H, Feng C, Li Y, Cui X, Zhe J, Sun K, Liu K, Fan Q, Sun D, Bao W. PCL-PEtOx-based Crystalline-core Micelles for the Targeted Delivery of Paclitaxel and Trabectedin in Ovarian Cancer Therapy. Acta Biomater 2025:S1742-7061(25)00303-4. [PMID: 40306394 DOI: 10.1016/j.actbio.2025.04.050] [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: 01/20/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
Ovarian cancer (OC), which primarily metastasizes through ascites, is both invasive and fatal. Despite its toxicity and drug resistance, the platinum-based chemotherapy Taxol®+Carboplatin has been the first-line standard treatment for decades. Trabectedin (TBD) is a recently developed, highly effective antitumor drug that is also capable of regulating tumor-associated macrophages (TAMs), however, its severe side-effects hinder further clinical application. Here, we developed safe and efficient pH-responsive crystalline-core micelles for the combined treatment of OCs, exploiting parallel delivery of paclitaxel (PTX) and TBD. PCL-PEtOx-COOH was selected as the optimal carrier to encapsulate PTX or TBD, which self-assemble into micelles with internal crystalline cores. The carboxyl group exposed on the surface of the micelles was utilized to react with the amines of Herceptin and hyaluronic acid cross-linked polymer (Herceptin-HA) to form PTX(Target). Similarly, TBD(Target) was formed by reaction with the CD206-targeted peptide mUNO. The low critical micelle concentrations of PTX(Target) and TBD(Target) stabilize the micelles in the bloodstream and normal tissues to prevent drug release. In an acidic microenvironment, the tertiary amide group on PEtOx chain of micelles ionizes, causing disassembly and pH-responsive release. Compared with Taxol®+Carboplatin, the combination therapy displayed dramatically improved safety and efficacy, as evidenced by the elimination of peritoneal tumor spheroids and reduced expression of NOX4, a gene that is overexpressed in most OC tissues. Furthermore, in human tissues, the ROS-response gene NOX4 is linked to the development of M2-type TAMs. Collectively, this study provides a safe and effective non-platinum-based chemotherapy for OC, offering an alternative to traditional Taxol®+Carboplatin. STATEMENT OF SIGNIFICANCE: (1) Significance: This work reports a new approach for ovarian cancer (OC) treatment. We utilized trabectedin (TBD) which a recently developed, highly effective antitumor drug that is also capable of regulating tumor associated macrophages (TAMs) combined with paclitaxel (PTX) to replace platinum-based chemotherapy Taxol®+Carboplatin (TC regimen). Compared to the clinical formulations, Yondelis® and Taxol®, pH-responsive PCL-PEtOx-based crystalline-core micelles were utilized for targeted independent delivery of TBD and PTX to TAMs and tumor cells, which maintained safe and efficient transport, overcoming the challenges posed by TAMs and carboplatin resistance. The system capabilities have also been confirmed in organoid and PDX models. (2) This is the first report demonstrating that this approach simultaneously overcomes the abdominal metastasis and carboplatin resistance of OC.
Collapse
Affiliation(s)
- Zixiu Du
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| | - Wei Wei
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Shuli Lu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Hao Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China
| | - Chenxu Feng
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Yinuo Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China
| | - Xinyi Cui
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jianan Zhe
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910 Hengshan Road, Shanghai, 200030, China
| | - Kuo Sun
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, 1st Minde Road, Nanchang, Jiangxi, 330006, China
| | - Kuai Liu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Qiong Fan
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910 Hengshan Road, Shanghai, 200030, China.
| | - Donglei Sun
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China; Shanghai Key Laboratory for Antibody-Drug Conjugates with Innovative Target, Shanghai, 200240, China.
| | - Wei Bao
- Department of Obstetrics and Gynecology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China.
| |
Collapse
|
32
|
Jiang J, Chen H, Zhao C, Li T, Zhang C, Ma L, Su H, Ma L, Duan Z, Si Q, Chuang TH, Chen C, Luo Y. PRTN3 promotes IL33/Treg-mediated tumor immunosuppression by enhancing the M2 polarization of tumor-associated macrophages in lung adenocarcinoma. Cancer Lett 2025; 616:217584. [PMID: 39993649 DOI: 10.1016/j.canlet.2025.217584] [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: 02/16/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
The immunosuppressive tumor microenvironment (TME) shaped by tumor-associated macrophages (TAMs) is essential for lung adenocarcinoma (LUAD) immune tolerance and tumor progression. Here, we first reported that proteinase 3 (PRTN3) promoted the alternative activation (M2) of TAMs and enhanced IL33/regulatory T cells (Tregs)-mediated tumor immunosuppression in LUAD. Firstly, clinical analysis revealed PRTN3 was highly expressed in TAMs and correlated with the tumor progression and poor prognosis in LUAD patients. Meanwhile, by using the myeloid cells-specific Prtn3-knockout mouse model, we demonstrated Prtn3 deficiency in macrophages remolded the immunosuppressive TME and suppressed tumor growth. The mechanism studies uncovered a novel signaling pathway that PRTN3 up-regulated IL33 expression in TAMs by suppressing AKT-mediated ubiquitinated degradation of FOXO1, which subsequently activated Il33 transcription. Furthermore, lack of PRTN3 or FOXO1 in macrophages greatly restrained IL33-induced Treg differentiation. Importantly, selective knockout of Prtn3 in macrophages significantly enhanced the antitumor effect of anti-PD1 therapy in the mouse model of LUAD. Thus, our work demonstrated that PRTN3 in macrophages, served as a key immunoregulator, contributed to impede the antitumor immune response through reinforcing the TAMs/Tregs crosstalk, which provided valuable insights to improve the immunotherapeutic effect by functional remodeling of TAMs to alleviate immunosuppression in LUAD.
Collapse
Affiliation(s)
- Jiayu Jiang
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou, 213149, China
| | - Huilin Chen
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Chunxing Zhao
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou, 213149, China
| | - Tong Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Chen Zhang
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou, 213149, China; The School of Medicine, Nankai University, Tianjin, 300071, China
| | - Lingyu Ma
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Huifang Su
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou, 213149, China
| | - Lei Ma
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Zhaojun Duan
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou, 213149, China
| | - Qin Si
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou, 213149, China
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Miaoli, Zhunan, Taiwan
| | - Chong Chen
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou, 213149, China.
| | - Yunping Luo
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China; Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou, 213149, China.
| |
Collapse
|
33
|
He Y, Tian T, Li Y, Zeng Y, Wang X, Qian L, Tian T, Jiang M, Li L. From neglect to necessity: the role of innate immunity in cutaneous squamous cell carcinoma therapy. Front Immunol 2025; 16:1570032. [PMID: 40352926 PMCID: PMC12061915 DOI: 10.3389/fimmu.2025.1570032] [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: 02/02/2025] [Accepted: 04/03/2025] [Indexed: 05/14/2025] Open
Abstract
As the second most common non-melanoma skin cancer, cutaneous squamous cell carcinoma (cSCC) has experienced a significant increase in incidence. Although clinical detection is relatively easy, a considerable number of patients are diagnosed at an advanced stage, featuring local tissue infiltration and distant metastasis. Cemiplimab, along with other immune checkpoint inhibitors, enhances T cell activation by blocking the PD-1 pathway, resulting in notable improvements in clinical outcomes. Nonetheless, approximately 50% of the patients with advanced cSCC remain unresponsive to this therapeutic approach. It emphasizes the importance of finding innovative therapeutic targets and strategies to boost the success of immunotherapy across a wider range of patients. Therefore, we focused on frequently neglected functions of innate immune cells. Emerging evidence indicates that innate immune cells exhibit considerable heterogeneity and plasticity, fundamentally contributing to tumor initiation and development. The identification and eradication of cancer cells, along with the modulation of adaptive immune responses, are essential roles of these cells. Consequently, targeting innate immune cells to activate anti-tumor immune responses presents significant potential for enhancing immunotherapeutic strategies in cSCC.
Collapse
Affiliation(s)
- Yong He
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Ting Tian
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Yuancheng Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Yong Zeng
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, The Second Xiangya Hospital of Central South University, Changsha, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Xiaoke Wang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Leqi Qian
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Tian Tian
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Mingjun Jiang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Liming Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| |
Collapse
|
34
|
Zhang G, Yu H, Liu J, Dong G, Cai Z. Myeloid-lineage-specific membrane protein LRRC25 suppresses immunity in solid tumor and is a potential cancer immunotherapy checkpoint target. Cell Rep 2025; 44:115631. [PMID: 40279244 DOI: 10.1016/j.celrep.2025.115631] [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: 09/18/2024] [Revised: 02/20/2025] [Accepted: 04/09/2025] [Indexed: 04/27/2025] Open
Abstract
Leucine-rich repeat containing 25 (LRRC25), a type I membrane protein, is specifically expressed in myeloid cells including neutrophils and macrophages. The anti-inflammatory role of LRRC25 was suggested in a few pathogenic models. However, its role in cancer immunity has not been interrogated. Here, we demonstrate that LRRC25 is robustly expressed in tumor-associated macrophages (TAMs). Lrrc25 deficiency in the tumor microenvironment (TME) suppresses growth of multiple murine tumor models by reprogramming TAMs toward an anti-tumor phenotype and thereby enhancing infiltration and activation of CD8+ T cells. The Nox2-ROS-Nlrp3-Il1β pathway is elevated in Lrrc25-deficient TAMs. Furthermore, a human myeloid cell line or mice with loss of Lrrc25 appear normal, indicating that LRRC25 is a safe immune target. Our results suggest that as an unappreciated immune checkpoint for tumor immunotherapy, the myeloid-specific membrane protein LRRC25 orchestrates the activity of TAMs via the canonical Nlrp3-IL1β inflammatory pathway and influences CD8+ T cell chemotaxis to the TME.
Collapse
Affiliation(s)
- Guorong Zhang
- Tianjin Key Laboratory of Inflammatory Biology, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin Medical University, Tianjin, China; The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Hanzhi Yu
- Tianjin Key Laboratory of Inflammatory Biology, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin Medical University, Tianjin, China; The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Jingjing Liu
- Tianjin Key Laboratory of Inflammatory Biology, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin Medical University, Tianjin, China; The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Ge Dong
- Tianjin Key Laboratory of Inflammatory Biology, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin Medical University, Tianjin, China; The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Zhigang Cai
- Tianjin Key Laboratory of Inflammatory Biology, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin Medical University, Tianjin, China; The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Science, Tianjin Medical University, Tianjin, China; Department of Bioinformatics, School of Basic Medical Science, Tianjin Medical University, Tianjin, China; Department of Hematology, Tianjin Medical University Tianjin General Hospital, Tianjin, China; Department of Rheumatology and Immunology, Tianjin Medical University Tianjin General Hospital, Tianjin, China.
| |
Collapse
|
35
|
Song Z, Wang Q, Xiong H, Xiao J, Zhou Z, Li T, Sun Q, Qiu L, Tan Y, Liu X, Jiang H, Han S, Wang X. Bionic gene delivery system activates tumor autophagy and immunosuppressive niche to sensitize anti-PD-1 treatment against STK11-mutated lung adenocarcinoma. J Nanobiotechnology 2025; 23:312. [PMID: 40275340 PMCID: PMC12020135 DOI: 10.1186/s12951-025-03404-z] [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: 11/21/2024] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
Clinical data have shown that Serine/Threonine Kinase 11 (STK11) mutation may be associated with an immunosuppressive tumor microenvironment (ITEM) and poor prognosis and failure of anti-PD-1 (αPD1) treatment in non-small cell lung cancer (NSCLC). To explore the potential of restoring STK11 protein in immunotherapy, a bionic gene delivery system was prepared by coating the STK11-encoded DNA-cationic polymer complex core with the tumor cell membrane, termed STK11@PPCM. STK11@PPCM could specifically bind with NSCLC cells and achieve precise delivery of STK11-encoded DNA. The released DNA effectively restored the STK11 protein expression, consequently reactivating autophagy and immunogenic cell death (ICD) in cancer cells. The liberated damage-associated molecular patterns (DAMPs) and autophagosome induced dendritic cells (DCs) maturation, which in turn enhanced CD8 + T cell infiltration, M1 macrophage polarization, and proinflammatory factor expression, thereby reversing the ITEM. Moreover, STK11@PPCM was also found to improve the sensitivity of cancer cells to αPD1 by increasing the expression of PD-L1, which was confirmed in STK11-mutated NSCLC cell xenografted mouse models, constructed by CRISPR-Cas9 knockout technology. This work demonstrated for the first time that restoration of functional STK11 can effectively reverse ITME and boost αPD1 efficacy in NSCLC, offering a new therapeutic approach for STK11-mutated lung adenocarcinoma in clinic.
Collapse
Affiliation(s)
- Zhongquan Song
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Qikai Wang
- Health Management Center, Weifang People's Hospital, Shandong Second Medical University, Weifang, Shandong, 261000, China
| | - Hongjie Xiong
- State Key Laboratory of Digital Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Jiang Xiao
- State Key Laboratory of Digital Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Zihan Zhou
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Tianxiang Li
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Qian Sun
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Liping Qiu
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Yue Tan
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China.
| | - Hui Jiang
- State Key Laboratory of Digital Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China.
| | - Shuhua Han
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China.
| | - Xuemei Wang
- State Key Laboratory of Digital Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China.
| |
Collapse
|
36
|
Wei S, Xu G, Zhao S, Zhang C, Feng Y, Yang W, Lu R, Zhou J, Ma Y. EGR2 promotes liver cancer metastasis by enhancing IL-8 expression through transcription regulation of PDK4 in M2 macrophages. Int Immunopharmacol 2025; 153:114484. [PMID: 40139095 DOI: 10.1016/j.intimp.2025.114484] [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: 01/10/2025] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025]
Abstract
Liver tumor is a common digestive system tumor, and its development is closely related to complex cytokines, tumor microenvironment and immunoregulatory mechanisms. Tumor-associated macrophages play a great role in a series of liver cancer development by secreting various cytokines and transmitting multiple signals, but how macrophages regulate the various biological behaviors of liver cancer cells at the microscopic level is a challenge we still need to overcome. In this research, we first identified the Early Growth Response 2 (EGR2) gene, which exhibited significant expression in M2 macrophages in comparison to M0 and M1 cell types, utilizing RNA sequencing. Subsequently, we validated this finding through a battery of methodologies, including WB, qRT-PCR, and immunofluorescence assays. We further employed a co-culture model involving MHCC97L and macrophages to investigate the impact of EGR2 downregulation within M2 cells on the in vivo and in vitro metastatic and invasive capabilities of MHCC97L cells. Subsequently, we directed our attention to investigating the impact of EGR2 on the levels of interleukin-8 (IL-8). Through comprehensive analyses including RNA sequencing, CUT-and-Tag, and ChIP techniques, we demonstrated that EGR2 can bind to the promoter region of the Pyruvate Dehydrogenase Kinase 4 (PDK4) gene. Finally, we introduced a virus overexpressing PDK4 and demonstrated that EGR2 could regulate the transcriptional level of PDK4 to affect the expression of IL-8, and ultimately alter the metastatic ability of hepatocellular carcinoma cells. Our study demonstrates that EGR2 may be a valuable target for future intervention in the disease process of hepatocellular carcinoma and refines the mechanism at the microscopic level of Tumor-associated macrophages.
Collapse
Affiliation(s)
- Song Wei
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Gaoxin Xu
- Department of General Surgery, Affiliated Kunshan Hospital of Jiangsu University,Kunshan,Suzhou,China
| | - Siqi Zhao
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, , Zhejiang, China
| | - Chenwei Zhang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yongheng Feng
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Weijun Yang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Renhe Lu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jin Zhou
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Yong Ma
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
37
|
Li Q, Wang M, Huang X, Wang S, Li C, Li P, Xiang W, Yao L, Deng C, Zhang M, Wang J. 6-Gingerol, an active compound of ginger, attenuates NASH-HCC progression by reprogramming tumor-associated macrophage via the NOX2/Src/MAPK signaling pathway. BMC Complement Med Ther 2025; 25:154. [PMID: 40269843 PMCID: PMC12020160 DOI: 10.1186/s12906-025-04890-2] [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: 10/27/2024] [Accepted: 04/10/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Non-alcoholic steatohepatitis-associated hepatocellular carcinoma (NASH-HCC) accounts for an increasing proportion of HCC cases. Currently, effective pharmacological options for treating both NASH and NASH-HCC remain limited, necessitating the identification of novel therapeutic agents. Our previous studies have demonstrated that ginger can ameliorate nonalcoholic fatty liver disease (NAFLD) and prevent the occurrence of NASH. The therapeutic effects and underlying mechanisms of NASH-HCC, however, remain poorly understood. METHODS Network pharmacology, bioinformatics, single-cell RNA sequencing analysis, and molecular docking were used to identify the main active compounds, targets, and possible mechanisms of ginger in treating NASH-HCC. The anti-tumor efficacy and underlying mechanisms of the selected compound in treating NASH-HCC were validated through in vitro experimentation. RESULTS Network pharmacology, bioinformatics, and molecular docking have revealed that 6-gingerol is the main active compound of ginger in treating NASH-HCC. SRC can be an essential target gene for ginger attenuating NASH-HCC progression, while the mitogen-activated protein kinase (MAPK) signaling pathway and reactive oxygen species (ROS) play equally important roles. Single-cell RNA sequencing of the HCC patients shows that the key targets of ginger in treating NASH-HCC are distributed in tumor-associated macrophage (TAMs). It has been reported that NOX2-derived ROS in macrophages can activate Src and then regulate downstream MAPK signaling cascades. 6-Gingerol can inhibit the proliferation, migration and reduce lipid deposition of liver cancer cells in vitro. More importantly, it induces polarization TAMs to M1 and enhances proinflammatory function, which may be achieved via the NOX2/Src/MAPK signaling pathway. CONCLUSION This study proves that 6-gingerol, the primary active compound in ginger, plays a role in attenuating the progression of NASH-HCC by inhibiting the proliferation and migration of tumor cells, or reprogramming TAMs to the M1 phenotype via the NOX2/Src/MAPK signaling pathway and activating the TAM-mediated immune responses.
Collapse
Affiliation(s)
- Qiurui Li
- College of Traditional Chinese Medicine, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Meng Wang
- College of Traditional Chinese Medicine, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Xuekuan Huang
- College of Traditional Chinese Medicine, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Shang Wang
- College of Traditional Chinese Medicine, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Chunli Li
- School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Pan Li
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Wei Xiang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing University of Chinese Medicine, No. 61 Puguobao Road, Bishan District, Chongqing, 402760, China
| | - Ling Yao
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing University of Chinese Medicine, No. 61 Puguobao Road, Bishan District, Chongqing, 402760, China
| | - Chengdan Deng
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing University of Chinese Medicine, No. 61 Puguobao Road, Bishan District, Chongqing, 402760, China
| | - Mingming Zhang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing University of Chinese Medicine, No. 61 Puguobao Road, Bishan District, Chongqing, 402760, China.
| | - Jianwei Wang
- College of Traditional Chinese Medicine, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing University of Chinese Medicine, No. 61 Puguobao Road, Bishan District, Chongqing, 402760, China.
| |
Collapse
|
38
|
Xie Y, Shi Z, Chen T, Li H, Fan M, Xiang X, Liu F. The Lung Cancer Immune Prognostic Score predicts pathologic complete response and survival in NSCLC patients receiving neoadjuvant immunochemotherapy. Front Immunol 2025; 16:1567565. [PMID: 40308604 PMCID: PMC12040963 DOI: 10.3389/fimmu.2025.1567565] [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: 01/27/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction Neoadjuvant immunochemotherapy (nICT) has significantly improved event-free survival (EFS) and pathologic complete response (pCR) in patients with resectable non-small cell lung cancer (NSCLC). However, the lack of validated biomarkers limits their ability to predict therapeutic efficacy and survival outcomes. This study aimed to develop a novel inflammatory and nutritional index, the Lung Cancer Immune Prognostic Score (LCIPS), to predict pCR and survival prognosis in patients with NSCLC. Methods This retrospective study included 131 patients with clinical stage IB-IIIB NSCLC who underwent neoadjuvant immunochemotherapy between May 2020 and May 2024. Baseline clinical data and hematological parameters were collected. Lasso regression analysis was employed to identify hematological indices associated with pCR, and the LCIPS was constructed based on these factors. Kaplan-Meier survival analysis and log-rank tests were used to assess survival differences. Logistic regression was performed to identify the predictors of pCR, while Cox regression analysis determined independent prognostic factors for disease-free survival (DFS) and overall survival (OS). The predictive performance of the LCIPS was validated using a nomogram. Results Lasso regression identified three core hematological indices: the albumin-to-globulin ratio (A/G), absolute monocyte count (MONO), and absolute lymphocyte count (LYM). The LCIPS formula was as follows: LCIPS=0.900×A/G+0.761×MONO (109/L) -0.065×LYM (109/L). Receiver operating characteristic (ROC) curve analysis showed that the LCIPS had superior predictive efficacy (area under the curve (AUC) = 0.68) compared to other classical markers. Univariate and multivariate logistic regression analyses identified intraoperative lymph node dissection status and A/G and LCIPS as independent predictors of pCR (p < 0.05). Multivariate Cox regression analysis demonstrated that smoking status and LCIPS were independent prognostic factors for DFS and OS. Nomogram validation indicated robust predictive accuracy for LCIPS. Notably, among immune-related adverse events (irAEs), endocrine- and cardiac-related irAEs significantly affected DFS (p < 0.05). Discussion LCIPS is an independent predictor of pCR in patients with NSCLC receiving neoadjuvant immunochemotherapy and is associated with improved DFS and survival outcomes. This novel index offers valuable guidance for personalized treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fang Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
39
|
Liu H, Cai S, Sun Y, Zhao C, Xu X, Gu S, Peng S, Di Q, Zhou H, Gu X, Ai X, Yang C. Macrophage membrane entrapped rapamycin-loaded TPGS/F127 micelles through intratracheal instillation for enhanced drug delivery and therapy to lung cancer with pulmonary fibrosis. Int J Pharm 2025; 674:125429. [PMID: 40049256 DOI: 10.1016/j.ijpharm.2025.125429] [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: 12/31/2024] [Revised: 02/26/2025] [Accepted: 03/02/2025] [Indexed: 03/28/2025]
Abstract
PURPOSE Patients with pulmonary fibrosis are prone to developing lung cancer. Pulmonary fibrosis and lung cancer have many common pathogenic factors and similar pathological features. For patients with IPF combined with lung cancer, there is currently no better treatment method available now. The purpose of this study is to develop a rapamycin pulmonary administration preparation that can treat lung cancer with pulmonary fibrosis, thereby overcoming the limitations of rapamycin treatment. METHODS In this study, rapamycin-loaded mixed micelle nanoparticles (TPGS/F127@RAPA) were first prepared by the film dispersion method. Then biomimetic nanoparticles (MM@TPGS/F127@RAPA) were obtained by coating the surface of TPGS/F127@RAPA with macrophage membranes (MM) using a co-incubation method. RESULTS TPGS/F127@RAPA and MM@TPGS/F127@RAPA showed particle sizes of about 15 nm and 260 nm respectively. Transmission electron microscope results showed that TPGS/F127@RAPA and MM@TPGS/F127@RAPA had homogeneous spherical shape morphologies and that the TPGS/F127@RAPA core was successfully covered with the macrophage membrane. In vitro studies demonstrated that MM@TPGS/F127@RAPA could effectively inhibit the excessive proliferation and migration of A549 cells and activated-Mlg cells. Moreover, MM@TPGS/F127@RAPA could increase the uptake of rapamycin by cells. By inhibiting the TGF-β1/Smad3 and PI3K/AKT/mTOR signaling pathways, TPGS/F127@RAPA and MM@TPGS/F127@RAPA could further reduce collagen deposition, inhibit tumor cell proliferation and improve lung function. Mice suffering from lung cancer with pulmonary fibrosis were treated with MM@TPGS/F127@RAPA through intratracheal instillation. The results showed that compared with TPGS/F127@RAPA, MM@TPGS/F127@RAPA could better reduce the area of pulmonary fibrosis and collagen deposition, inhibit tumor cell proliferation and improve lung function, exhibit longer retention time in lung and better lung distribution and deposition. CONCLUSION Our results revealed that the biomimetic strategy of MM@TPGS/F127@RAPA may be a good choice for the treatment of lung cancer patients with pulmonary fibrosis.
Collapse
Affiliation(s)
- Hongting Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Shihao Cai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Yao Sun
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Conglu Zhao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Xiang Xu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Songtao Gu
- Department of Respiratory & Critical Care Medicine, Tianjin Chest Hospital, No.261, Taierzhuang South Road, Jinnan District, Tianjin 300222, China
| | - Shouchun Peng
- Jinnan Hospital, Tianjin University (Tianjin Jinnan Hospital), Tianjin 300350, China
| | - Qingguo Di
- Department of Respiratory and Critical Care Medicine, Cangzhou Central Hospital, No. 16, Xinhua West Road, Yunhe District, Cangzhou City, Hebei Province 061000, China.
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.
| | - Xiaoting Gu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.
| | - Xiaoyu Ai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| |
Collapse
|
40
|
Wang SL, Chan TA. Navigating established and emerging biomarkers for immune checkpoint inhibitor therapy. Cancer Cell 2025; 43:641-664. [PMID: 40154483 DOI: 10.1016/j.ccell.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/19/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have improved outcomes of patients with many different cancers. These antibodies target molecules such as programmed cell death 1 (PD-1) or cytotoxic T lymphocyte associated protein 4 (CTLA-4) which normally function to limit immune activity. Treatment with ICIs reactivates T cells to destroy tumor cells in a highly specific manner, which in some patients, results in dramatic remissions and durable disease control. Over the last decade, much effort has been directed at characterizing factors that drive efficacy and resistance to ICI therapy. Food and Drug Administration (FDA)-approved biomarkers for ICI therapy have facilitated more judicious treatment of cancer patients and transformed the field of precision oncology. Yet, adaptive immunity against cancers is complex, and newer data have revealed the potential utility of other biomarkers. In this review, we discuss the utility of currently approved biomarkers and highlight how emerging biomarkers can further improve the identification of patients who benefit from ICIs.
Collapse
Affiliation(s)
- Stephen L Wang
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; National Center for Regenerative Medicine, Cleveland, OH, USA.
| |
Collapse
|
41
|
Zhao L, Yan F, Tang D, Li W, An N, Ren C, Wang Y, Xu K, Zhao K. The transition between M1 and M2 macrophage phenotypes is associated with the disease status following CD19 CAR-T therapy for B cell lymphoma/leukemia. Cell Death Dis 2025; 16:275. [PMID: 40216772 PMCID: PMC11992075 DOI: 10.1038/s41419-025-07610-3] [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: 12/18/2024] [Revised: 03/19/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Although anti-CD19 chimeric antigen receptor (CAR-T) cells demonstrate high response rates in relapsed/refractory B-cell lymphomas, a considerable proportion of patients eventually encounter disease progression or relapse. The short-term and long-term outcomes of CAR-T treatment are intricately linked to the tumor microenvironment (TME), wherein macrophages with polarized characteristics can exhibit either anti-tumorigenic or pro-tumorigenic roles. Despite evidence implicating the crucial involvement of macrophages in CAR-T cell-treated lymphoma, their dynamic distribution and immune function related to lymphoma progression remain poorly understood. Immunocompetent mice were utilized to establish syngeneic A20 lymphoma/leukemia models. The distribution and polarization of macrophages were detected using immunohistochemistry (IHC) and flow cytometry techniques. We observed that CD19 CAR-T therapy exhibited significant efficacy in protecting mice against lymphoma, leading to increased infiltration of macrophages into the tumor tissue. Notably, during remission stages, M1-like macrophages (CD11b+F4/80+C206-CD80+) were predominant, whereas in relapsed mice, there was a shift towards M2-like phenotypes (CD11b+F4/80+C206+CD80+). The transition from remissive to relapsed status was accompanied by a reduction in the M1/M2 ratio and a decrease in pro-inflammatory cytokines. Furthermore, quantitative real-time polymerase chain reaction (qRT-PCR) analysis confirmed differential expression levels of CD206 and CD163 between remissive and relapsed mice, while signaling pathways involving PI3K and STAT3 may contribute to the skewing towards M2 polarization. In summary, our findings highlight the dynamic transformation of macrophage polarization during different stages of lymphoma progression and underscore its potential implications for immunotherapeutic interventions.
Collapse
Affiliation(s)
- Li Zhao
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fen Yan
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Donghai Tang
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenwen Li
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Na An
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chunxiao Ren
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ying Wang
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kailin Xu
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Kai Zhao
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
42
|
Wang W, Zhai Y, Yang X, Ye L, Lu G, Shi X, Zhai G. Effective design of therapeutic nanovaccines based on tumor neoantigens. J Control Release 2025; 380:17-35. [PMID: 39892648 DOI: 10.1016/j.jconrel.2025.01.078] [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: 09/05/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Neoantigen vaccines are among the most potent immunotherapies for personalized cancer treatment. Therapeutic vaccines containing tumor-specific neoantigens that elicit specific T cell responses offer the potential for long-term clinical benefits to cancer patients. Unlike immune-checkpoint inhibitors (ICIs), which rely on pre-existing specific T cell responses, personalized neoantigen vaccines not only promote existing specific T cell responses but importantly stimulate the generation of neoantigen-specific T cells, leading to the establishment of a persistent specific memory T cell pool. The review discusses the current state of clinical research on neoantigen nanovaccines, focusing on the application of vectors, adjuvants, and combinational strategies to address a range of challenges and optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Weilin Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84124, United States of America
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guoliang Lu
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Xiaoqun Shi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
43
|
Cui X, Liu S, Song H, Xu J, Sun Y. Single-cell and spatial transcriptomic analyses revealing tumor microenvironment remodeling after neoadjuvant chemoimmunotherapy in non-small cell lung cancer. Mol Cancer 2025; 24:111. [PMID: 40205583 PMCID: PMC11980172 DOI: 10.1186/s12943-025-02287-w] [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: 10/04/2024] [Accepted: 02/28/2025] [Indexed: 04/11/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) represents the most common pathological type of lung cancer, and the combination of neoadjuvant immunotherapy with chemotherapy has emerged as the first-line treatment for NSCLC. Nevertheless, the efficacy of this therapeutic approach remains variable. The present study aims to examine the impact of chemoimmunotherapy in NSCLC patients, with a view to identifying key molecules, critical cell subpopulations, communication patterns and spatial distributions that potentially correlate with therapeutic sensitivity. A total of 16 lung cancer tissue samples were collected from a cohort of 12 NSCLC patients and subjected to single-cell RNA and spatial transcriptome sequencing. Our data demonstrated that the distribution of CD4 + Treg T cells and mCAFs indicated an immunosuppressive tumor microenvironment, while the accumulation of CD4 + Th17 T cells and iCAFs could act as a positive marker for the sensitivity to chemoimmunotherapy. Furthermore, a significant high level of SELENOP-macrophages was observed in tissues from positive responders, and a strong co-localization between SELENOP-macrophages and antigen-presenting cancer associated fibroblasts (CAFs) in the tumor boundaries was identified, indicating the cooperative roles of these two cell types in response to combined therapy. Moreover, SELENOP-macrophages were observed to be accumulated in tertiary lymphoid structures, which further suggested its critical role in recruiting lymphocytes. Furthermore, analysis of cell-cell communication, based on spatial transcriptomics, suggests that the interactions between SELENOP-macrophages, apCAFs, CD4 + and CD8 + T cells were significantly enhanced in responders. In addition, SELENOP-macrophages recruited CD4 + Naïve, Helper and CD8 + Naïve T cells through pathways such as the cholesterol, interleukin, chemokine and HLA when responding to combined therapy. The present study further unveils the dynamic spatial and transcriptional changes in the tumor microenvironment of non-small cell lung cancer in response to combination therapy.
Collapse
Affiliation(s)
- Xiaolu Cui
- Department of Urology, First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China
| | - Siyuan Liu
- Department of Thoracic Surgery, First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China
| | - He Song
- Department of Gastrointestinal Surgery, First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China.
| | - Jingjing Xu
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province , 110004, China.
| | - Yanbin Sun
- Department of Thoracic Surgery, First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China.
| |
Collapse
|
44
|
Ma C, Zhu W, Hu X, Wu D, Zhao X, Du Y, Kong X. Acinar cells modulate the tumor microenvironment through the promotion of M1 macrophage polarization via macrophage endocytosis in pancreatic cancer. Discov Oncol 2025; 16:489. [PMID: 40198509 PMCID: PMC11979042 DOI: 10.1007/s12672-025-02244-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/26/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDA) is a highly aggressive and fatal cancer. M1 macrophages are generally considered to have anti-tumor properties, capable of suppressing tumor growth and metastasis by secreting pro-inflammatory cytokines and enhancing the immune response. AIMS The objective of this research was to pinpoint crucial genes associated with M1 macrophages and search for a new way to activate the M1 phenotype of macrophages in PDA. METHODS The level of immune cell infiltration was assessed using CIBERSORT in TCGA-PAAD cohort and ICGC-PACA cohort. We performed weighted gene co-expression network analysis (WGCNA) to identify the module most correlated with M1 macrophages and we identified hub genes through protein-protein interaction (PPI) analyse. Through survival analysis, correlation analysis and single cell analysis, we obtained the relationship between hub genes and prognosis, and the relationship between key genes and immune cells, as well as its expression in various cells. RESULTS PRSS1 (Cationic trypsinogen) and CTRB1 (Chymosinogen B) were hub genes of the M1 macrophage-associated WGCNA module (211genes) and are closely related to the extension of survival time, which are also verified as cell growth-related genes by DepMap database. Through single-cell sequencing analysis, we determined that the expression levels of PRSS1 and CTRB1 in the acinar cells of tumor tissues were diminished. PRSS1 and CTRB1 are considered to be the signature genes of acinar cells. The proportion of acinar cells was also correlated with the infiltration of CD8T cells and M1 cells. Immunostaining revealed elevated expression levels of PRSS1 and CTRB1 in adjacent normal tissues. Cell line experiments confirmed that macrophages polarize towards M1 by engulfing pancreatic enzyme granules, thereby inhibiting the malignant phenotype of tumor cells. CONCLUSION Our findings highlight the critical role of acinar cells in modulating the immune microenvironment of pancreatic tumors by influencing macrophage polarization. This insight may provide novel opportunities for therapeutic interventions in cancer treatment.
Collapse
Affiliation(s)
- Congjia Ma
- Department of Gastroenterology, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Wenbo Zhu
- Department of Gastroenterology, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Xiulin Hu
- Department of Gastroenterology, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Deli Wu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Soochow University, Soochow, China
| | - Xintong Zhao
- Department of Gastroenterology, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Yiqi Du
- Department of Gastroenterology, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China.
| | - Xiangyu Kong
- Department of Gastroenterology, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China.
| |
Collapse
|
45
|
Peng H, Chen Q, Ye L, Wang W. A Senescence-Associated Gene Signature for Prognostic Prediction and Therapeutic Targeting in Adrenocortical Carcinoma. Biomedicines 2025; 13:894. [PMID: 40299539 PMCID: PMC12025298 DOI: 10.3390/biomedicines13040894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/23/2025] [Accepted: 03/30/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Cellular senescence plays a critical role in tumorigenesis, immune cell infiltration, and treatment response. Adrenocortical carcinoma (ACC) is a malignant tumor that lacks effective therapies. This study aimed to construct and validate a senescence-related gene signature as an independent prognostic predictor for ACC and explore its impact on the tumor microenvironment, immunotherapy, and chemotherapy response. Methods: Data were collected from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) database. Using Kaplan-Meier survival analysis, LASSO penalized Cox regression and multivariable Cox regression, we identified a prognostic model with four senescence-related genes (HJURP, CDK1, FOXM1, and CHEK1). The model's prognostic value was validated through survival analysis, risk score curves, and receiver operating characteristic (ROC) curves. Tumor mutation burden was assessed with maftools, and the tumor microenvironment was analyzed using CIBERSORT and ESTIMATE. Immune and chemotherapeutic responses were assessed through Tumor Immune Dysfunction and Exclusion (TIDE) and OncoPredict. Results: The risk score derived from our model showed a strong association with overall survival (OS) in ACC patients (p < 0.001, HR = 2.478). Higher risk scores were correlated with more advanced tumor stages and a greater frequency of somatic mutations. Differentially expressed genes (DEGs) that were downregulated in the high-risk group were significantly enriched in immune-related pathways. Furthermore, high-risk patients were predicted to have reduced sensitivity to immunotherapy (p = 0.02). Bioinformatics analysis identified potential chemotherapeutic agents, including BI-2536 and MIM1, as more effective treatment options for high-risk patients. Conclusions: Our findings indicate that this prognostic model may serve as a valuable tool for predicting overall survival (OS) and treatment responses in ACC patients, including those receiving chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Hangya Peng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (H.P.); (Q.C.)
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qiujing Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (H.P.); (Q.C.)
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lei Ye
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (H.P.); (Q.C.)
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (H.P.); (Q.C.)
| |
Collapse
|
46
|
Chang X, Huang Y, Qu Y, Guo Y, Fan W, Zhen H. Integrative analysis of mitochondrial-related gene profiling identifies prognostic clusters and drug resistance mechanisms in low-grade glioma. Discov Oncol 2025; 16:465. [PMID: 40186003 PMCID: PMC11971116 DOI: 10.1007/s12672-025-02201-2] [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: 12/16/2024] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
Mitochondrial dysfunction has emerged as a critical factor in the progression and prognosis of low-grade glioma (LGG). In this study, we explored the role of mitochondrial-related genes through consensus clustering analysis using multi-omics data from the TCGA, CGGA, and other independent datasets. Patients were categorized into three clusters (Cluster A, B, and C), with Cluster B consistently associated with poorer prognosis. Mutation landscape analysis revealed distinct genetic alterations and copy number variations among clusters, particularly in Cluster B, which exhibited unique genetic signatures. Immune infiltration analysis showed that Cluster B had higher expression levels of immune checkpoint genes, stronger immune evasion activity, and greater immune cell infiltration, suggesting an immunosuppressive tumor microenvironment. Furthermore, we identified mitochondrial-related prognostic markers and developed a MITscore based on gene expression patterns, which stratified patients into high- and low-risk groups. High MITscore groups displayed stronger stemness characteristics, poorer survival outcomes, and differential responses to chemotherapy and immunotherapy. Cross-validation with drug sensitivity and immunotherapy cohorts indicated that high MITscore patients were more sensitive to certain chemotherapeutic agents and responded better to immunotherapy. Finally, using the SRGA method, we identified novel biomarkers (KDR, LRRK2, SQSTM1) closely associated with mitochondrial function, which may serve as potential targets for therapeutic intervention. These findings highlight the critical role of mitochondrial dysfunction in LGG prognosis, tumor microenvironment regulation, and treatment response, providing new avenues for precision oncology.
Collapse
Affiliation(s)
- Xiaozan Chang
- Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, Zhengzhou, 450003, Henan, China
| | - Yingxuan Huang
- Pediatric Intensive Care Unit, The Affiliated Hospital of Youjiang Medical University for Nationalities; Key Laboratory of Research and Development on Clinical Molecular Diagnosis for High-Incidence Diseases of Baise, Baise, China
| | - Ying Qu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yu Guo
- Nanfang Hospital (ZengCheng Branch), Southern Medical University, No. 28, Innovation Avenue, Ningxi Street, Guangzhou, China.
| | - Wenwen Fan
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Nanli, Panjiayuan, Beijing, 100021, China.
| | - Haining Zhen
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
47
|
Tong W, Qin N, Lu T, Liu L, Liu R, Chen J, Luo N. Integrating bulk and single-cell RNA sequencing reveals SH3D21 promotes hepatocellular carcinoma progression by activating the PI3K/AKT/mTOR pathway. PLoS One 2025; 20:e0302766. [PMID: 40179068 PMCID: PMC11967960 DOI: 10.1371/journal.pone.0302766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 02/16/2025] [Indexed: 04/05/2025] Open
Abstract
As a novel genetic biomarker, the potential role of SH3D21 in hepatocellular carcinoma remains unclear. Here, we decipher the expression and function of SH3D21 in human hepatocellular carcinoma. The expression level and clinical significance of SH3D21 in hepatocellular carcinoma patients, the relationship between SH3D21 and the features of tumor microenvironment (TME) and role of SH3D21 in promoting hepatocellular carcinoma progression were analyzed based on the bulk samples obtained from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases. Single-cell sequencing samples from Gene Expression Omnibus (GEO) database were employed to verify the prediction mechanism. Additionally, different biological effects of SH3D21 on hepatocellular carcinoma cells were investigated by qRT-PCR, CCK-8 assay, colony forming assay and Western blot analysis. Bioinformatics analysis and in vitro experiments revealed that the expression level of SH3D21 was up-regulated in hepatocellular carcinoma and correlated with the poor prognosis in hepatocellular carcinoma patients. SH3D21 effectively promoted the proliferation, invasion, and migration as well as the formation of immunosuppressive microenvironment of hepatocellular carcinoma. In addition, SH3D21 can activate the PI3K/AKT/mTOR signaling pathway. SH3D21 stimulates the progression of hepatocellular carcinoma by activating the PI3K/AKT/mTOR signaling pathway, and SH3D21 can serve as a prognostic biomarker and therapeutic target for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Wangxia Tong
- Department of Hepatology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Na Qin
- The Graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Tao Lu
- Department of hepatobiliary surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Li Liu
- Department of Hepatology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Rong Liu
- Department of Hepatology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Jibing Chen
- Centre for Translational Medical Research in Integrative Chinese and Western Medicine, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Ning Luo
- Department of Neurology, RuiKang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
48
|
Wei QJ, Liang HQ, Liang YW, Huang ZX. TET3 is expressed in prostate cancer tumor-associated macrophages and is associated with anti-androgen resistance. Clin Transl Oncol 2025; 27:1712-1727. [PMID: 39240303 DOI: 10.1007/s12094-024-03708-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: 07/13/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
PURPOSE The aim of this study is to investigate the expression of TET3 in prostate cancer and its effect on the efficacy of anti-androgen therapy (ADT). METHODS The expression of TET3 in 1965 cases of prostate cancer and 493 cases of normal prostate tissues were analyzed. The CIBERSORT algorithm evaluated the abundance of 22 tumor-infiltrating immune cells in 497 prostate cancers. Subsequently, the expression of TET3 in prostate cancer TAMs was analyzed using 21,292 cells from single-cell RNA sequencing (scRNAseq). In addition, the trajectory of the differentiation process was reconstructed based on pseudotime analysis. Sensitivity prediction of prostate cancers to ADT was evaluated based on GDSC2 and CTRP databases. Another dataset GSE111177 was employed for further analysis. RESULTS TET3 was over-expressed in prostate cancer, and the expression of TET3 in metastatic prostate cancer was higher than that in non-metastatic prostate cancer. The scRNAseq analysis of prostate cancer showed that TET3 was mainly expressed in TAM. TET3 expressed in early and active TAMs, with the activation of signaling pathways such as energy metabolism, cell communication, and cytokine production. Prostate cancer in TET3 high expression group was more sensitive to ADT drugs such as Bicalutamide and AZD3514, and was also more sensitive to chemotherapy drugs such as Cyclophosphamide, Paclitaxel, and Vincristine, and MAPK pathway inhibitors of Docetaxel and Dabrafenib. CONCLUSIONS The efficacy of ADT in prostate cancer is related to the expression of TET3 in TAMs, and TET3 may be a potential therapeutic target for coordinating ADT.
Collapse
Affiliation(s)
- Qiu-Ju Wei
- Guangxi Medical University, 22 Shuang-Yong Road, Nanning, 530021, Guangxi, China
| | - Hai-Qi Liang
- Guangxi Medical University, 22 Shuang-Yong Road, Nanning, 530021, Guangxi, China.
| | - Yao-Wen Liang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zu-Xin Huang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
49
|
Hua Q, Li Z, Weng Y, Wu Y, Zheng L. Myeloid cells: key players in tumor microenvironments. Front Med 2025; 19:265-296. [PMID: 40048137 DOI: 10.1007/s11684-025-1124-8] [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: 08/14/2024] [Accepted: 12/16/2024] [Indexed: 05/04/2025]
Abstract
Cancer is the result of evolving crosstalk between neoplastic cell and its immune microenvironment. In recent years, immune therapeutics targeting T lymphocytes, such as immune checkpoint blockade (ICB) and CAR-T, have made significant progress in cancer treatment and validated targeting immune cells as a promising approach to fight human cancers. However, responsiveness to the current immune therapeutic agents is limited to only a small proportion of solid cancer patients. As major components of most solid tumors, myeloid cells played critical roles in regulating the initiation and sustentation of adaptive immunity, thus determining tumor progression as well as therapeutic responses. In this review, we discuss emerging data on the diverse functions of myeloid cells in tumor progression through their direct effects or interactions with other immune cells. We explain how different metabolic reprogramming impacts the characteristics and functions of tumor myeloid cells, and discuss recent progress in revealing different mechanisms-chemotaxis, proliferation, survival, and alternative sources-involved in the infiltration and accumulation of myeloid cells within tumors. Further understanding of the function and regulation of myeloid cells is important for the development of novel strategies for therapeutic exploitation in cancer.
Collapse
Affiliation(s)
- Qiaomin Hua
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zhixiong Li
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yulan Weng
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yan Wu
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Limin Zheng
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
50
|
Li Q, Xu J, Hua R, Xu H, Wu Y, Cheng X. Nano-strategies for Targeting Tumor-Associated Macrophages in Cancer immunotherapy. J Cancer 2025; 16:2261-2274. [PMID: 40302816 PMCID: PMC12036086 DOI: 10.7150/jca.108194] [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: 12/04/2024] [Accepted: 02/12/2025] [Indexed: 05/02/2025] Open
Abstract
Tumor-associated macrophages (TAMs) are one type of the most abundant immune cells within tumor, resulting in immunosuppresive tumor microenvironment and tumor resistance to immunotherapy. Thus, targeting TAMs is a promising therapeutic strategy for boosting cancer immunotherapy. This study provides an overview of current therapeutic strategies targeting TAMs, which focus on blocking the recruitment of TAMs by tumors, regulating the polarization of TAMs, and directly eliminating TAMs using various nanodrugs, especially with a new categorization based on the specific signaling pathways, such as NF-κB, HIF-1α, ROS, STAT, JNK, PI3K, and Notch involved in their regulatory mechanism. The latest developments of nanodrugs modulating these pathways are discussed in determining the polarization of TAMs and their role in the tumor microenvironment. Despite the challenges in clinical translation and the complexity of nanodrug synthesis, the potential of nanodrugs in enhancing the effectiveness of cancer immunotherapy is worthy of expecting.
Collapse
Affiliation(s)
- Qian Li
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, P. R. China
| | - Jingwei Xu
- Department of Thoralic Surgery, Suzhou Municipal Hospital Institution, Suzhou 215000, P. R. China
- Department of Thoralic Surgery, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 215123, P. R. China
| | - Runjia Hua
- Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Hanye Xu
- Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Yongyou Wu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, P. R. China
| | - Xiaju Cheng
- Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|