1
|
Wan D, Zhang Q, Yang Z, Zhang X, Xie P, Cheng S, Xu L, Liu B, Zhang K, Zhang W. Engineered oncolytic virus OH2-FLT3L enhances antitumor immunity via dendritic cell activation. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200975. [PMID: 40236993 PMCID: PMC11999459 DOI: 10.1016/j.omton.2025.200975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 04/17/2025]
Abstract
The combination of oncolytic viruses (OVs) with other immunotherapies, such as immunostimulatory therapies, is a current research hotspot; however, optimizing their therapeutic potential remains to be fully explored. Here, we designed a novel oncolytic herpes simplex virus 2 expressing Fms-like tyrosine kinase 3 ligand (OH2-FLT3L), which induces an antitumor cytotoxic T cell immune response by activating dendritic cells (DCs). We found that OH2-FLT3L specifically infects tumor cells, induces immunogenic cell death (ICD), and releases a large number of tumor-specific antigens, which bound to danger signals and facilitated antigenic cross-presentation by DCs, significantly enhancing T cell activation and function. Experimental results showed that OH2-FLT3L significantly increased the proportion of activated DCs, enhanced the antitumor immune response, and effectively converted "cold" tumors into "hot" tumors. In addition, when combined with anti-PD-1 antibody, OH2-FLT3L further enhanced therapeutic efficacy. In conclusion, OH2-FLT3L, as a novel oncolytic virus, demonstrates the potential to enhance antitumor immune responses through DC activation.
Collapse
Affiliation(s)
- Duo Wan
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qi Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhenrong Yang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Xiaoli Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Peipei Xie
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shujun Cheng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Libin Xu
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Binlei Liu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan 430068, China
| | - Kaitai Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wen Zhang
- Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
2
|
Franco-Fuquen P, Figueroa-Aguirre J, Martínez DA, Moreno-Cortes EF, Garcia-Robledo JE, Vargas-Cely F, Castro-Martínez DA, Almaini M, Castro JE. Cellular therapies in rheumatic and musculoskeletal diseases. J Transl Autoimmun 2025; 10:100264. [PMID: 39931050 PMCID: PMC11808717 DOI: 10.1016/j.jtauto.2024.100264] [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: 10/29/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 02/13/2025] Open
Abstract
A substantial proportion of patients diagnosed with rheumatologic and musculoskeletal diseases (RMDs) exhibit resistance to conventional therapies or experience recurrent symptoms. These diseases, which include autoimmune disorders such as multiple sclerosis, rheumatoid arthritis, and systemic lupus erythematosus, are marked by the presence of autoreactive B cells that play a critical role in their pathogenesis. The persistence of these autoreactive B cells within lymphatic organs and inflamed tissues impairs the effectiveness of B-cell-depleting monoclonal antibodies like rituximab. A promising therapeutic approach involves using T cells genetically engineered to express chimeric antigen receptors (CARs) that target specific antigens. This strategy has demonstrated efficacy in treating B-cell malignancies by achieving long-term depletion of malignant and normal B cells. Preliminary data from patients with RMDs, particularly those with lupus erythematosus and dermatomyositis, suggest that CAR T-cells targeting CD19 can induce rapid and sustained depletion of circulating B cells, leading to complete clinical and serological responses in cases that were previously unresponsive to conventional therapies. This review will provide an overview of the current state of preclinical and clinical studies on the use of CAR T-cells and other cellular therapies for RMDs. Additionally, it will explore potential future applications of these innovative treatment modalities for managing patients with refractory and recurrent manifestations of these diseases.
Collapse
Affiliation(s)
- Pedro Franco-Fuquen
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Juana Figueroa-Aguirre
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - David A. Martínez
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Eider F. Moreno-Cortes
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Juan E. Garcia-Robledo
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Fabio Vargas-Cely
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | | | - Mustafa Almaini
- Rheumatology, Allergy & Clinical Immunology Division, Mafraq Hospital, United Arab Emirates
| | - Januario E. Castro
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
3
|
Yan C, Wang G. Advances in research on flavonoids in tumor immunotherapy (Review). Mol Med Rep 2025; 31:150. [PMID: 40211703 PMCID: PMC11995692 DOI: 10.3892/mmr.2025.13515] [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: 09/03/2024] [Accepted: 02/25/2025] [Indexed: 04/16/2025] Open
Abstract
Cancer immunotherapy is an approach used in anti‑tumor treatment; however, its efficacy is limited to specific tumor types that are inherently sensitive to immune system modulation. Expanding the scope of indications and enhancing the efficacy of cancer immunotherapy are key goals for continued advancement. Flavonoids modulate the tumor‑immunosuppressive microenvironment. Integrating flavonoids with immunotherapeutic modalities, including cancer vaccines, immune checkpoint inhibitors and adoptive immune‑cell therapy, has potential in terms of augmenting the therapeutic efficacy of immunotherapy. The present review aimed to summarize flavonoids that enhance cancer immunotherapy, focusing on their underlying mechanisms and the application of nanotechnology to overcome inherent limitations such as poor solubility, low bioavailability, rapid metabolism, and instability under physiological conditions, thereby highlighting the potential of flavonoids in advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Chaoguang Yan
- Department of Oncology, Weifang Chinese Medicine Hospital, Weifang, Shandong 261000 P.R. China
| | - Guangchun Wang
- Department of Oncology, Weifang Chinese Medicine Hospital, Weifang, Shandong 261000 P.R. China
| |
Collapse
|
4
|
Tao L, Zhang Y, Zhang J, Tao J, Gong Y, Mao J, Tian Q, Ao P, Zhuo D. Peptides-functionalized gold nanostars enhanced degradation of PD-L1 for improved prostate cancer immunotherapy. J Biomater Appl 2025; 39:1188-1201. [PMID: 39993230 DOI: 10.1177/08853282251319473] [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] [Indexed: 02/26/2025]
Abstract
Blockage of the interaction between programmed death receptor-1 (PD-1) and programmed death ligand-1 (PD-L1) can restore T-cell activity and enhance antitumor immunity. PD-1/PD-L1 pathway inhibitors have promising applications in the treatment of advanced prostate cancer (PCa). We successfully developed a peptides-functionalized gold nanoconstruct (P-AuNS) consisted of PD-L1-binding peptide (PD-L1pep, P) and gold nanostar (AuNS), which could bind to cell-surface PD-L1 specifically and deliver PD-L1 into PCa cells with high efficiency. In PCa cells, P-AuNS can efficiently degrade PD-L1 in a lysosomal-dependent manner. In the co-culture system of Jurkat cells and DU145 cells, P-AuNS restored the proliferative capacity and interferon-gamma (IFN-γ) secretion level of Jurkat cells inhibited by co-cultured DU145 cells, indicating that P-AuNS effectively hampered the interaction between PD-1 and PD-L1. In addition, in PCa-bearing mice, P-AuNS can effectively inhibit tumor growth and down-regulate PD-L1 protein levels, and in vivo experimental results show that P-AuNS has no systemic toxicity. P-AuNS block the interaction between PD-1 and PD-L1 by efficiently degrading PD-L1, thus restoring the antitumor activity of T cells and inhibiting tumor progression of PCa. In all, P-AuNS has great promise as a potential immunotherapy strategy in the treatment of advanced PCa and even other solid tumors.
Collapse
Affiliation(s)
- Liangjun Tao
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Yifei Zhang
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Jingwei Zhang
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Jianping Tao
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Yu Gong
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Jun Mao
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Qixing Tian
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Ping Ao
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Dong Zhuo
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| |
Collapse
|
5
|
Wu H, Liu J, Zhang XH, Jin S, Li P, Liu H, Zhao L, Wang J, Zhao S, Tian HD, Lai JR, Hao Y, Liu GR, Hou K, Yan M, Liu SL, Pang D. The combination of flaxseed lignans and PD-1/ PD-L1 inhibitor inhibits breast cancer growth via modulating gut microbiome and host immunity. Drug Resist Updat 2025; 80:101222. [PMID: 40048957 DOI: 10.1016/j.drup.2025.101222] [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: 12/29/2024] [Revised: 02/22/2025] [Accepted: 02/22/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Patients with breast cancer (BC) who benefit from the PD-1/PD-L1 inhibitor (PDi) is limited, necessitating novel strategies to improve immunotherapy efficacy of BC. Here we aimed to investigate the inhibitory effects of flaxseed lignans (FL) on the biological behaviors of BC and evaluate the roles of FL in enhancing the anticancer effects of PDi. METHODS HPLC was used to detect the content of enterolactone (ENL), the bacterial transformation product of FL. Transcript sequencing was performed and identified CD38 as a downstream target gene of ENL. CD38-overexpressing cells were constructed and cell proliferation, colony formation, wound healing and transwell assays were used to assess the function of ENL/CD38 axis on BC cells in vitro. Multiplexed immunohistochemistry (mIHC) and CyTOF were used to detect the changes of the tumor immune microenvironment (TIM). 16S rDNA sequencing was used to explore the changes of gut microbiota in mice. A series of in vivo experiments were conducted to investigate the anticancer effects and mechanisms of FL and PDi. RESULTS FL was converted to ENL by gut microbiota and FL administration inhibited the progression of BC. ENL inhibited the malignant behaviors of BC by downregulating CD38, a key gene associated with immunosuppression and PD-1/PD-L1 blockade resistance. The mIHC assay revealed that FL administration enhanced CD3+, CD4+ and CD8+ cells and reduced F4/80+ cells in TIM. CyTOF confirmed the regulatory effects of FL and FL in combination with PDi (FLcPDi) on TIM. In addition, 16S rDNA analysis demonstrated that FLcPDi treatment significantly elevated the abundance of Akkermansia and, importantly, Akkermansia administration enhanced the response to PDi in mice treated with antibiotics. CONCLUSIONS The FL/ENL/CD38 axis inhibited BC progression. FL enhanced the anticancer effects of PDi by modulating gut microbiota and host immunity.
Collapse
Affiliation(s)
- Hao Wu
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Jiena Liu
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xing-Hua Zhang
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Shengye Jin
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ping Li
- The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Huidi Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Liuying Zhao
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jianyu Wang
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shilu Zhao
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hong-Da Tian
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Jin-Ru Lai
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Yi Hao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Gui-Rong Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Kaijian Hou
- School of Public Health, Shantou University, Shantou, China; Longhu People's Hospital, Shantou, China.
| | - Meisi Yan
- Department of Pathology, Harbin Medical University, Harbin, China.
| | - Shu-Lin Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China; Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
| | - Da Pang
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
6
|
Zhang W, Yuan S, Zhang Z, Fu S, Liu S, Liu J, Ma Q, Xia Z, Gu P, Gao S, Zhang Z, Zhang X, Liu Y, Zhang N. Regulating tumor cells to awaken T cell antitumor function and enhance melanoma immunotherapy. Biomaterials 2025; 316:123034. [PMID: 39709849 DOI: 10.1016/j.biomaterials.2024.123034] [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: 08/24/2024] [Revised: 11/26/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024]
Abstract
Tumor cells transmit various immunosuppressive signals and induce a dysfunctional state in T cells, which essentially leads to immune escape and tumor progression. However, developing effective strategies to counteract the domestication of T cells by tumor cells remains a challenge. Here, we prepared pH-responsive lipid nanoparticles (NL/PLDs) co-loaded with PCSK9 shRNA, lonidamine (LND), and low-dose doxorubicin (DOX). NL/PLDs can awaken domesticated T cells function by sending pro-activation, pro-recognition, and pro-killing signals by increasing tumor immunogenicity, increasing the expression of major histocompatibility complex I (MHC-I) on tumor cells, and alleviating the suppression effect of tumor-secreted lactic acid (LA) on the T cell effector function, respectively. In melanoma-bearing mice, NL/PLDs effectively relieved tumor immunosuppressive microenvironment (TIME) and enhanced the antitumor immunity mediated by CD8+ T cells. Furthermore, when combined with aPD-1, NL/PLDs demonstrated strong antitumor effects and increased immunotherapeutic efficacy. This regulatory strategy provides new insights for enhancing immunotherapy by regulating tumor immunosuppressive signals and shows significant potential for clinical tumor treatment.
Collapse
Affiliation(s)
- Weihan Zhang
- 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, 250012, China
| | - Shijun Yuan
- 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, 250012, China
| | - Zipeng Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Shunli Fu
- 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, 250012, China
| | - Shujun Liu
- 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, 250012, China
| | - Jinhu Liu
- 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, 250012, China
| | - Qingping Ma
- 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, 250012, China
| | - Zhenxing Xia
- 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, 250012, China
| | - Panpan Gu
- 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, 250012, China
| | - Shuying Gao
- 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, 250012, China
| | - Zhiyue Zhang
- 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, 250012, China
| | - Xinke Zhang
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Yongjun Liu
- 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, 250012, China.
| | - Na Zhang
- 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, 250012, China
| |
Collapse
|
7
|
Qi Y, Ge H, Sun X, Wei Y, Zhai J, Qian H, Mo H, Ma F. Systemic immune characteristics predicting toxicity to immune checkpoint inhibitors in patients with advanced breast cancer. J Autoimmun 2025; 153:103423. [PMID: 40267835 DOI: 10.1016/j.jaut.2025.103423] [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: 01/30/2025] [Revised: 04/12/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are among the most promising treatment options for cancer. However, frequent and sometimes life-threatening immune-related adverse events (irAEs) are associated with ICI treatment. Therefore, it is imperative to establish a model for predicting the risk of irAEs to identify high-risk groups, enable more accurate clinical risk‒benefit analysis for ICI treatment and decrease the incidence of irAEs. However, no ideal model for predicting irAEs has been applied in clinical practice. The aim of this study was to analyze the systemic immune characteristics of patients with irAEs and establish a model for predicting the risk of irAEs. METHODS We conducted a study to monitor irAEs in patients with advanced breast cancer undergoing immunotherapy during and following the treatment course. Peripheral blood mononuclear cells (PBMCs) were collected before and after two cycles of therapy. Mass cytometry time-of-flight (CyTOF) was employed to identify baseline and posttreatment immune cell subpopulations, and the relationships between the proportions of cells in these subpopulations and the occurrence of irAEs were explored. Additionally, we conducted subgroup analyses stratified by the anatomic location and time of onset of irAEs. Furthermore, we developed a logistic regression model to predict the risk of irAEs and validated this model using two independent validation cohorts from the Gene Expression Omnibus (GEO) database (accession numbers GSE189125 and GSE186143). RESULTS By analyzing 106 blood samples and samples from two independent validation cohorts (n = 16 and 60 patients), we found that high proportions of CXCR3+CCR6+CD4+ T cells and CD38+CD86+CXCR3+CCR6+CD8+ T cells and a low proportion of CXCR3lowCD56dim natural killer (NK) cells at baseline were significantly correlated with the incidence of irAEs (P = 0.0029, P < 0.001, and P = 0.0017, respectively). In the subgroup analysis, we observed consistent results in patients with immune-related pneumonitis (ir-pneumonitis) and immune-related thyroiditis (ir-thyroiditis). In the early irAE group, the baseline proportion of CXCR3+CCR6+CD4+ T cells was greater than that in the late irAE group (P = 0.011). An analysis of PBMCs before and after ICI treatment revealed thatthe dynamic changes in the proportions of naïve CD4+ T cells and CXCR3lowCD56dim NK cells were closely related to irAE occurrence. Finally, we ultimately developed a model for predicting the risk of irAEs, which yielded an area under the receiver operating characteristic curve (AUROC) of 0.79 in the training cohort and an AUROC of 0.75 in the single-cell validation cohort (GSE189125). CONCLUSIONS These findings indicate that different populations of immune cells are associated with different irAEs and that characterization of these cells may be used as biomarkers to predict the risk of specific toxicities. This will facilitate the management of irAEs and may lead to a reduction in the incidence of irAEs.
Collapse
Affiliation(s)
- Yalong Qi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Hewei Ge
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xiaoying Sun
- Department of Medical Oncology, Cancer Hospital of HuanXing ChaoYang District, Beijing, China.
| | - Yuhan Wei
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jingtong Zhai
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Haili Qian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Hongnan Mo
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
8
|
Liu N, Wang X, Wang Z, Kan Y, Fang Y, Gao J, Kong X, Wang J. Nanomaterials-driven in situ vaccination: a novel frontier in tumor immunotherapy. J Hematol Oncol 2025; 18:45. [PMID: 40247328 PMCID: PMC12007348 DOI: 10.1186/s13045-025-01692-4] [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: 10/09/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
In situ vaccination (ISV) has emerged as a promising strategy in cancer immunotherapy, offering a targeted approach that uses the tumor microenvironment (TME) to stimulate an immune response directly at the tumor site. This method minimizes systemic exposure while maintaining therapeutic efficacy and enhancing safety. Recent advances in nanotechnology have enabled new approaches to ISV by utilizing nanomaterials with unique properties, including enhanced permeability, retention, and controlled drug release. ISV employing nanomaterials can induce immunogenic cell death and reverse the immunosuppressive and hypoxic TME, thereby converting a "cold" tumor into a "hot" tumor and facilitating a more robust immune response. This review examines the mechanisms through which nanomaterials-based ISV enhances anti-tumor immunity, summarizes clinical applications of these strategies, and evaluates its capacity to serve as a neoadjuvant therapy for eliminating micrometastases in early-stage cancer patients. Challenges associated with the clinical translation of nanomaterials-based ISV, including nanomaterial metabolism, optimization of treatment protocols, and integration with other therapies such as radiotherapy, chemotherapy, and photothermal therapy, are also discussed. Advances in nanotechnology and immunotherapy continue to expand the possible applications of ISV, potentially leading to improved outcomes across a broad range of cancer types.
Collapse
Affiliation(s)
- Naimeng Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiangyu Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhongzhao Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yonemori Kan
- Department of Medical Oncology, National Cancer Center Hospital (NCCH), 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jidong Gao
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518127, China.
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
9
|
Yu J, Fu L, Wu R, Che L, Liu G, Ran Q, Xia Z, Liang X, Zhao G. Immunocytes in the tumor microenvironment: recent updates and interconnections. Front Immunol 2025; 16:1517959. [PMID: 40297580 PMCID: PMC12034658 DOI: 10.3389/fimmu.2025.1517959] [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: 10/27/2024] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
The tumor microenvironment (TME) is a complex, dynamic ecosystem where tumor cells interact with diverse immune and stromal cell types. This review provides an overview of the TME's evolving composition, emphasizing its transition from an early pro-inflammatory, immune-promoting state to a later immunosuppressive milieu characterized by metabolic reprogramming and hypoxia. It highlights the dual roles of key immunocytes-including T lymphocytes, natural killer cells, macrophages, dendritic cells, and myeloid-derived suppressor cells-which can either inhibit or support tumor progression based on their phenotypic polarization and local metabolic conditions. The article further elucidates mechanisms of immune cell plasticity, such as the M1/M2 macrophage switch and the balance between effector T cells and regulatory T cells, underscoring their impact on tumor growth and metastasis. Additionally, emerging therapeutic strategies, including checkpoint inhibitors and chimeric antigen receptor (CAR) T and NK cell therapies, as well as approaches targeting metabolic pathways, are discussed as promising avenues to reinvigorate antitumor immunity. By integrating recent molecular insights and clinical advancements, the review underscores the importance of deciphering the interplay between immunocytes and the TME to develop more effective cancer immunotherapies.
Collapse
Affiliation(s)
- Jiyao Yu
- Department of Ultrasound, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Li Fu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Gastroenterology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Rui Wu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Neurosurgery, Jiangyou People’s Hospital, Mianyang, China
| | - Linyi Che
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qinwen Ran
- General Practice Department, Wufu Town Hospital, Chongqing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China
| | - Xisong Liang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guanjian Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Duminuco A, Novello G, Mauro E, Scalisi E, Del Fabro V, Sambataro D, Palumbo G, Di Raimondo F, Romeo D. Chemotherapy extravasation: diagnosis, prevention and management. J Chemother 2025:1-13. [PMID: 40205769 DOI: 10.1080/1120009x.2025.2488599] [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: 01/03/2025] [Revised: 03/11/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025]
Abstract
Chemotherapy extravasation, the unintended leakage of cytotoxic drugs into surrounding tissues, is a significant complication in oncological treatments, potentially leading to severe tissue damage and long-term consequences. This review explores the factors influencing extravasation risk, including infusion site, patient comorbidities and the physicochemical properties of drugs. Early detection is crucial to prevent irreversible damage. Treatment strategies vary based on the type of drug involved, ranging from topical dimethyl sulfoxide and hyaluronidase to specific antidotes like dexrazoxane for anthracycline extravasations. Preventive measures, including proper catheter placement, drug dilution and patient monitoring, are essential to mitigate risks. Effective management requires a multidisciplinary approach, combining prompt recognition, intervention and ongoing education for healthcare providers to improve patient safety and outcomes in chemotherapy administration. Enhanced training on the early signs of extravasation and advancements in treatment modalities offer critical support in minimizing adverse effects, ensuring timely and appropriate care.
Collapse
Affiliation(s)
- Andrea Duminuco
- Hematology with BMT Unit, A.O.U. Policlinico "G.Rodolico-San Marco", Catania, Italy
| | - Giuseppe Novello
- Department of Medical Oncology, A.O.U. Policlinico "G.Rodolico-San Marco", Catania, Italy
| | - Elisa Mauro
- Hematology with BMT Unit, A.O.U. Policlinico "G.Rodolico-San Marco", Catania, Italy
| | - Elvira Scalisi
- Hematology with BMT Unit, A.O.U. Policlinico "G.Rodolico-San Marco", Catania, Italy
| | | | - Daniela Sambataro
- Faculty of Medicine and Surgery, "Kore" University of Enna, Enna, Italy
| | - Giuseppe Palumbo
- Hematology with BMT Unit, A.O.U. Policlinico "G.Rodolico-San Marco", Catania, Italy
| | | | - Demetria Romeo
- Unità Farmaci Antiblastici, Farmacia I, A.O.U. Policlinico "G.Rodolico-San Marco", Catania, Italy
| |
Collapse
|
11
|
Egberts BK, Ananthakrishna R, Perry R, Woodman RJ, Sutton A, Pasupathy S, Hecker T, Bromley TA, Yang WS, Karapetis C, Selvanayagam JB. Myocardial Injury Following Immune Checkpoint Inhibitors: An Australian Experience. Heart Lung Circ 2025:S1443-9506(24)01917-6. [PMID: 40204532 DOI: 10.1016/j.hlc.2024.11.018] [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: 06/03/2024] [Revised: 10/26/2024] [Accepted: 11/17/2024] [Indexed: 04/11/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have revolutionised cancer treatment, becoming a cornerstone therapy for various malignancies. Despite their efficacy, there are concerns about their potential cardiotoxicity, particularly, subclinical myocardial injury. However, there remains a scarcity of local data on the precise incidence of such cardiac complications. This prospective pilot study aims to assess myocardial injury in patients undergoing ICI therapy without known cardiovascular disease, using biochemical and cardiac imaging markers of myocardial injury. METHOD A total of 41 patients (median age 68 years, 63% male) receiving ICI therapy without previous cardiac disease were prospectively enrolled. Serial assessments were conducted at baseline and 6 weeks, including measurement of cardiac troponin I (cTnI), N-terminal pro-brain natriuretic peptide (NT-proBNP), and two-dimensional transthoracic echocardiography with speckle tracking strain. Cardiac magnetic resonance imaging (CMR) was performed at 6 weeks. A control group (n=10, median age 66 years, 50% male) with no ICI treatment with matched oncological profile was established. The primary outcome was the incidence of cTnI increase at 6 weeks, with secondary outcomes including changes in NT-proBNP, left ventricular (LV) ejection fraction, LV global longitudinal strain (LV-GLS), right ventricular free wall systolic strain, myocardial injury on CMR, and major adverse cardiac events. RESULTS Median baseline cTnI levels measured 4.0 (interquartile range 3.0-7.0) ng/L, with only one patient (2.7%) demonstrating significant cTnI increase after ICI therapy. Follow-up assessments revealed no significant mean differences in cTnI, NT-proBNP, LV ejection fraction, or right ventricular free wall systolic strain. However, a significant change in LV-GLS was noted in the ICI group (-1.2%, p=0.004) compared with controls (-0.7%, p=0.50, p=0.02). A total of 20 patients (54%) underwent CMR, with 11 (55%) exhibiting abnormalities (T1/T2 mapping and late gadolinium enhancement). Of the nine patients (45%) with increased T1 mapping on CMR, all demonstrated a reduction in LV-GLS between baseline and 6 weeks, with a mean reduction of 5% (3%-12%). Within 1 year, major adverse cardiac events occurred in 17 (45.9%) patients. CONCLUSIONS Although the occurrence of significant cTnI increase was low, subclinical myocardial injury can be detected through advanced cardiac imaging after ICI initiation. This underscores the importance of conducting larger studies to further elucidate the prevalence, mechanisms, and clinical implications of myocardial injury in patients receiving ICI treatment.
Collapse
Affiliation(s)
- Briella K Egberts
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; Department of Cardiovascular Medicine, Flinders Medical Centre, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Rajiv Ananthakrishna
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; Department of Cardiovascular Medicine, Flinders Medical Centre, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia; Ballarat Base Hospital, Grampians Health, Ballarat, Vic, Australia; Wimmera Base Hospital, Grampians Health, Horsham, Vic, Australia
| | - Rebecca Perry
- Department of Cardiovascular Medicine, Flinders Medical Centre, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia; Allied Health and Human Performance, University of South Australia, Adelaide, SA, Australia
| | - Richard J Woodman
- Flinders Centre of Epidemiology and Biostatistics, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Alison Sutton
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia; Department of Medical Radiology, Flinders Medical Centre, Adelaide, SA, Australia
| | - Sivabaskari Pasupathy
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Teresa Hecker
- Department of Cardiovascular Medicine, Flinders Medical Centre, Adelaide, SA, Australia
| | - Tonia A Bromley
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; Department of Cardiovascular Medicine, Flinders Medical Centre, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Woo Sze Yang
- Department of Cardiovascular Medicine, Flinders Medical Centre, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Christos Karapetis
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; Department of Medical Oncology, Flinders Medical Centre, Adelaide, SA, Australia
| | - Joseph B Selvanayagam
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; Department of Cardiovascular Medicine, Flinders Medical Centre, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
| |
Collapse
|
12
|
Wu Y, Zhang W, Liang X, Zhang P, Zhang M, Jiang Y, Cui Y, Chen Y, Zhou W, Liang Q, Dai J, Zhang C, Xu J, Li J, Yu T, Zhang Z, Guo R. Habitat radiomics analysis for progression free survival and immune-related adverse reaction prediction in non-small cell lung cancer treated by immunotherapy. J Transl Med 2025; 23:393. [PMID: 40181378 PMCID: PMC11970015 DOI: 10.1186/s12967-024-06057-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/25/2024] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is highly heterogeneous, leading to varied treatment responses and immune-related adverse reactions (irAEs) among patients. Habitat radiomics allows non-invasive quantitative assessment of intratumor heterogeneity (ITH). Therefore, our objective is to employ habitat radiomics techniques to develop a robust approach for predicting the efficacy of Immune checkpoint inhibitors (ICIs) and the likelihood of irAEs in advanced NSCLC patients. METHODS In this retrospective two center study, two independent cohorts of patients with NSCLC were used to develop (n = 248) and validate signatures (n = 95). After applying four kinds of machine learning algorithms to select the key preoperative CT radiomic features, we used clinical, radiomics and habitat radiomic features to develop the clinical signature, radiomics signature and habitat radiomic signature for ICIs prognostics and irAEs prediction. By combining habitat radiomic features with corresponding clinicopathologic information, the nomogram signature was constructed in the training cohort. Next, the internal validation cohort (n = 75) of patients, and the external validation cohort (n = 20) of patients treated with ICIs were included to evaluate the predictive value of the four signatures, and their predictive performance was assessed by the area under operating characteristic curve (AUC). RESULTS Our study introduces a radiomic nomogram model that integrates clinical and habitat radiomic features to identify patients who may benefit from ICIs or experience irAEs. The Radiomics Nomogram model exhibited superior predictive performance in the training, validation, and external validation sets, with AUCs of 0.923, 0.817, and 0.899, respectively. This model outperformed both the Whole-tumor Radiomics Signature model (AUCs of 0.870, 0.736, and 0.626) and the Habitat Signature model (AUCs of 0.900, 0.804, and 0.808). The radiomics model focusing on tumor sub-regional habitat showed better predictive performance than the model derived from the entire tumor. Decision Curve Analysis (DCA) and calibration curves confirmed the nomogram's effectiveness. CONCLUSION By leveraging machine learning to predict the outcomes of ICIs, we can move closer to achieving tailored ICIs for lung cancer. This advancement will assist physicians in selecting and managing subsequent treatment strategies, thereby facilitating clinical decision-making.
Collapse
Affiliation(s)
- Yuemin Wu
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Wei Zhang
- Department of Radiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao Liang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Lung Cancer Institute, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Mengzhe Zhang
- Department of Lung Cancer Surgery, Tianjin Lung Cancer Institute, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuqin Jiang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yanan Cui
- Department of Radiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yi Chen
- Department of Oncology, Pukou Branch of Jiangsu People's Hospital, Nanjing Pukou District Central Hospital, Nanjing, China
| | - Wenxin Zhou
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Qi Liang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jiali Dai
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Chen Zhang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jiali Xu
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jun Li
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Tongfu Yu
- Department of Radiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Zhihong Zhang
- Department of Pathology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Renhua Guo
- Department of Radiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
13
|
Wang S, Huang J, Zeng T, Chen Y, Xu Y, Zhang B. Parps in immune response: Potential targets for cancer immunotherapy. Biochem Pharmacol 2025; 234:116803. [PMID: 39965743 DOI: 10.1016/j.bcp.2025.116803] [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: 10/28/2024] [Revised: 01/24/2025] [Accepted: 02/13/2025] [Indexed: 02/20/2025]
Abstract
Immunotherapy in clinical application faces numerous challenges pertaining to both effectiveness and safety. Poly(ADP-ribose) polymerases (PARPs) exhibit multifunctional characteristics by transferring ADP-ribose units to target proteins or nucleic acids. In recent years, more and more attention has been paid to the biological function of PARPs in immune response. This article reviews the relationship between PARP family members and immune response. PARP1 and PARP2 inhibit anti-tumor immune activity by regulating immune checkpoint expression and the cGAS/STING signaling pathway. PARP7 and PARP11 play an important role in promoting immunosuppressive tumor microenvironment. PARP9 promotes the production of Type I interferon and the infiltration of macrophages. PARP13 is a key tumor suppressor that promotes anti-tumor immune response. PARP14 plays a crucial role in promoting the differentiation of macrophages towards the M2 pro-tumor phenotype. Summarizing the molecular mechanisms of PARP7, PARP9, PARP11, PARP13 and PARP14 in regulating immune response is helpful to deepen our comprehension of the role of PARPs in immune function regulation. This provides a reference and basis for targeted PARP-based cancer treatment strategies and drug development. PARP1, PARP7 inhibitors or other PARP inhibitors in combination with immune checkpoint inhibitors or other immunotherapy strategies may be a more effective cancer therapy.
Collapse
Affiliation(s)
- Shuping Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, PR China.
| | - Jingling Huang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Tingyu Zeng
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, PR China
| | - Yali Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, PR China
| | - Yungen Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Bangzhi Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of New Drug Design and Synthesis, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
14
|
Madera G, Hodge A, Roskelly L, Greenbaum C. What Is the Impact of Novel Systemic Anticancer Therapy on Acute Oncology Education and Service Delivery? Semin Oncol Nurs 2025; 41:151842. [PMID: 39986898 DOI: 10.1016/j.soncn.2025.151842] [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: 12/03/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/24/2025]
Abstract
OBJECTIVES To discuss the importance of educating healthcare professionals about oncological emergencies linked to novel systemic anticancer therapy (SACT) and the impact on acute oncology (AO) services. METHODS This discussion is based on clinical expertise and informed by current literature. RESULTS Novel SACT, such as immune-checkpoint inhibitors, have more complex toxicity profiles and can be challenging to recognize and treat. An increasing prevalence of toxicity is expected as new drugs are developed and the numbers of patients living with and beyond cancer expand; more data are required to capture the full extent of this. There are knowledge gaps within the healthcare workforce, particularly outside oncology-specialist settings. Focused research in this area will provide direction for targeted educational interventions. CONCLUSIONS Insufficient SACT education is a safety issue; severe toxicities can be fatal but initial symptoms can be subtle and may be missed. We argue that emergency care pathways can help to streamline the appropriate management of patients with SACT toxicity, but awareness of AO issues remains "everyone's business." Continuing clinical education is key to maintaining awareness of newly developed SACT. AO service models may vary, but AO competence assessment passports can provide a standardized method of evidencing AO knowledge and skills. IMPLICATIONS FOR NURSING PRACTICE Oncology nurses, as a highly specialized and knowledgeable part of the healthcare workforce, are key in supporting interprofessional education. By using existing cancer nursing frameworks, this can support the implementation of the AO Passports. The learning and development of AO services in the UK can be transferred internationally.
Collapse
Affiliation(s)
- Gina Madera
- Medical Oncology/Networked Services, The Christie NHS Foundation Trust, Manchester, UK.
| | - Ali Hodge
- Immunotherapy and Acute Oncology, Cancer Services Division, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
| | - Lara Roskelly
- Macmillan Acute Oncology Team, Cancer Division, Frimley Health NHS Foundation Trust, UK
| | - Clare Greenbaum
- Workforce and Education, Greater Manchester Cancer Alliance, The Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
15
|
Yang L, Wang X, Wang S, Shen J, Li Y, Wan S, Xiao Z, Wu Z. Targeting lipid metabolism in regulatory T cells for enhancing cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2025; 1880:189259. [PMID: 39798823 DOI: 10.1016/j.bbcan.2025.189259] [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: 06/11/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
As immunosuppressive cells, Regulatory T cells (Tregs) exert their influence on tumor immune escape within the tumor microenvironment (TME) by effectively suppressing the activity of other immune cells, thereby significantly impeding the anti-tumor immune response. In recent years, the metabolic characteristics of Tregs have become a focus of research, especially the important role of lipid metabolism in maintaining the function of Tregs. Consequently, targeted interventions aimed at modulating lipid metabolism in Tregs have been recognized as an innovative and promising approach to enhance the effectiveness of tumor immunotherapy. This review presents a comprehensive overview of the pivotal role of lipid metabolism in regulating the function of Tregs, with a specific focus on targeting Tregs lipid metabolism as an innovative approach to augment anti-tumor immune responses. Furthermore, we discuss potential opportunities and challenges associated with this strategy, aiming to provide novel insights for enhancing the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Liu Yang
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xingyue Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shurong Wang
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yaling Li
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shengli Wan
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Zhigui Wu
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
16
|
Vaghjiani R, Wu R, Tung KH, Ishikawa T, Takabe K. Angiogenesis Is Associated With Aggressive Biology That Counterbalances With Tumor Immunogenicity in Hepatocellular Carcinoma. World J Oncol 2025; 16:173-181. [PMID: 40162113 PMCID: PMC11954604 DOI: 10.14740/wjon2009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 01/17/2025] [Indexed: 04/02/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is an arterialized tumor; thus, anti-angiogenesis targeted therapy is in clinical practice. Herein, we hypothesized that HCC with high angiogenesis is biologically aggressive with worse survival. Methods Angiogenesis score (AS) was derived from the Molecular Signatures Database (MSigDB) Hallmark Angiogenesis Gene Set, and median was used to divide high versus low groups. Transcriptome of HCC patients of The Cancer Genome Atlas (TCGA, n = 386) and GSE76427 (n = 115) cohorts were analyzed. Results High AS correlated with angiogenesis-related gene expressions. Both microvascular and lymphatic endothelial cell infiltrations were higher in high angiogenesis HCC. Surprisingly, no survival difference was seen with varying levels of angiogenesis. High angiogenesis significantly enriched tumor aggravating signaling pathways: glycolysis, Notch, Hedgehog, KRAS, epithelial mesenchymal transition, and transforming growth factor-beta (TGF-β) in Gene Set Enrichment Analysis (GSEA), but also infiltrated less CD8+ T cells and T-helper 1 cells, and higher M1 macrophages and conventional dendritic cells (cDCs) with elevated cytolytic activity score in both cohorts. In agreement, immune response-related gene sets: inflammatory response, tumor necrosis factor-alpha (TNF-α) signaling, allograft rejection, interferon-alpha, and interferon-gamma were all enriched to high angiogenesis HCC. Programmed cell death protein 1 (PD1), programmed death ligand 1 (PD-L1), programmed death ligand 2 (PD-L2), and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) were higher in high angiogenesis HCC in TCGA, but not in GSE76427 cohort. Conclusions Angiogenesis quantified using transcriptome of HCC patients demonstrated that it is associated with aggressive biology but also with tumor immunogenicity and immune response that counterbalance and did not reflect in survival. Given high expression of immune checkpoint molecules, we cannot help but speculate that immunotherapy may be useful for high angiogenesis HCC patients.
Collapse
Affiliation(s)
- Raj Vaghjiani
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- These authors contributed equally to this work
| | - Rongrong Wu
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan
- These authors contributed equally to this work
| | - Kaity H. Tung
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY, USA
| | - Takashi Ishikawa
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan
- Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY, USA
- Department of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Department of Gastroenterological Surgery, Yokohama City University School of Medicine, Yokohama, Japan
- Department of Breast Surgery, Fukushima Medical University, Fukushima, Japan
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
17
|
Georgopoulou S, Droney J, Jaganathan PP, Howell P, Doherty AM, Young K, Cruickshank S. Identification of the PROMs used to assess ICI toxicities and HRQoL in patients receiving immune checkpoint inhibitor treatment in cancer care and their suitability: A systematic review. Cancer Treat Rev 2025; 135:102862. [PMID: 40056766 DOI: 10.1016/j.ctrv.2024.102862] [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: 09/02/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 03/10/2025]
Abstract
BACKGROUND The implementation of patient-reported outcome measures (PROMs) in the clinical identification of immunotherapy toxicities is a complex intervention. There has been very little work evaluating the clinical utility and generalisability of PROMs used after immune checkpoint inhibitor (ICI) treatment to date. We reviewed evidence on the use of PROMs assessing toxicities and health-related quality of life in patients treated with ICIs. METHODS PubMed, EMBASE, MEDLINE, PsycInfo, CINAHL, Web of Knowledge, the Cochrane Library were searched (January 2008 - October 2024). Quantitative studies reporting the use of PROMs to identify, assess and manage toxicities at any timepoint and HRQoL associated with ICI treatment in adult patients with cancer were included. A narrative synthesis describes the key characteristics of the PROMs identified. RESULTS 43 studies were included; 12 on melanoma/skin, 12 on lung and 19 on other cancers. Study designs included 20 randomised controlled trials, 14 cohort studies, six cross-sectional studies and three non-randomised interventional trials. The lack of ICI-specific PROMs was highlighted, particularly as the PROMs used lacked sufficient sensitivity for ICI treatments. CONCLUSIONS There is need for an ICI-specific PROM for effective assessment of toxicities and a tailored PROM for assessment of HRQoL. Some suggested key domains by certain studies for ICI-specific PROMs include: (a) ICI-specific items (e.g. certain USD-I and PRO-CTCAE items) to capture symptoms associated with ICI treatments such as rash, myalgia, (b) role, psychological, emotional and social functioning domains within HRQoL assessments and (c) additional patient-reported toxicities not included in existing PROMs. Findings emphasize the importance of using a disease-specific PROM that is applicable, acceptable and sufficiently sensitive to identify toxicities and HRQoL issues across all stages.
Collapse
Affiliation(s)
| | - Joanne Droney
- The Royal Marsden NHS Foundation Trust, London, United Kingdom; Imperial College London, London, United Kingdom
| | | | - Paul Howell
- The Royal Marsden School, London, United Kingdom
| | | | - Kate Young
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | | |
Collapse
|
18
|
Peng K, Li Y, Yang Q, Yu P, Zeng T, Lin C, Deng Y, Chen J. The therapeutic promise of probiotic Bacteroides fragilis (BF839) in cancer immunotherapy. Front Microbiol 2025; 16:1523754. [PMID: 40231233 PMCID: PMC11995047 DOI: 10.3389/fmicb.2025.1523754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/18/2025] [Indexed: 04/16/2025] Open
Abstract
Background Overwhelming evidence suggests that the gut microbiota modulates tumor response to immune checkpoint inhibitors (ICIs). The probiotic Bacteroides fragilis (BF839) was extensively used in China to improve gut microbiota dysbiosis-related symptoms. We hypothesized that probiotic BF839 could enhance tumor sensitivity to ICIs. Methods In the preclinical studies, mice received BF839 orally, PD-1 intraperitoneal injection, or a combination therapy of the two agents. The antitumor effect of BF839 was investigated by assessing the tumor growth and tumor immune microenvironment. Mice fecal samples were collected for 16S rRNA sequencing. Fresh tumor samples were collected for 16S RNA sequencing. The data of 29 patients with advanced solid tumor who received BF839 adjuvant therapy were retrospectively evaluated. The primary endpoint was overall survival (OS). Results Among patients with advanced solid tumors undergoing ICIs and chemotherapy, patients in BF839 long-term adjuvant treatment group had longer OS (p = 0.0101) than the BF839 short-term adjuvant treatment group. In the preclinical studies, we found that monotherapy with BF839 or anti-PD-1 antibody significantly inhibit tumor growth. Interestingly, BF839 worked synergistically with anti-PD-1 antibody and induced tumor regression, mediated by increased CD8+T cell infiltration. Mechanistically, BF839 induced tumor suppression was regulated by the cGAS-STING pathway. 16S rRNA sequencing results of mice fecal samples showed that BF839 treatment increased gut microbiota diversity. Conclusion Overall, our data suggest that BF839 enhanced tumor sensitivity to ICIs through cGAS-STING signaling. In the future, the application of probiotic BF839 to regulate gut microbiota may be a new strategy to enhance the efficacy of ICIs.
Collapse
Affiliation(s)
- Kunwei Peng
- Department of Medical Oncology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuqing Li
- Department of Medical Oncology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qijun Yang
- Department of Medical Oncology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Peijin Yu
- Department of Medical Oncology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ting Zeng
- Department of Clinical Nutrition, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chuhui Lin
- Department of Clinical Nutrition, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuhong Deng
- Department of Clinical Nutrition, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jingqi Chen
- Department of Medical Oncology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
19
|
Arvanitis P, Tziotis A, Papadimatos S, Farmakiotis D. Pathogenesis, Diagnosis, and Management of Cytokine Release Syndrome in Patients with Cancer: Focus on Infectious Disease Considerations. Curr Oncol 2025; 32:198. [PMID: 40277755 PMCID: PMC12026323 DOI: 10.3390/curroncol32040198] [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: 02/07/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
Background: Cytokine Release Syndrome (CRS) is a hyperinflammatory state triggered by immune therapies like CAR T-cell therapy and bispecific T-cell engagers (BiTEs). Characterized by excessive cytokine release, CRS often mimics infectious and inflammatory conditions, complicating diagnosis and treatment. Immunosuppressive therapies used for CRS further elevate the risk of secondary infections. Methods: A systematic search of PubMed and EMBASE was conducted using terms related to "cytokine release syndrome", "cytokine storm", "infections", and "management". Studies were included if they described infectious complications, diagnostic mimics, or therapeutic approaches related to CRS. Results: Of 19,634 studies, 2572 abstracts were reviewed. Infections occurred in up to 23% of patients post-CAR T therapy and 24% post-BiTE therapy. Pathogens included gram-positive and gram-negative bacteria, herpesviruses (e.g., CMV, HSV), fungi (e.g., Candida, Aspergillus), and parasites (e.g., Toxoplasma gondii). CRS mimics also included non-infectious inflammatory syndromes. Differentiation remains challenging, but cytokine profiling and biomarkers (e.g., ferritin, CRP, sIL-2Rα) may aid in diagnosis. Treatments included tocilizumab, corticosteroids, and empiric antimicrobials. Prophylactic strategies were inconsistently reported. Conclusions: Effective CRS management requires early recognition, differentiation from infectious mimics, and collaboration between oncology and infectious disease (ID) specialists. A multidisciplinary, collaborative, and structured approach, including dedicated ID input and pre-treatment evaluation, is essential for optimizing CRS management and patient outcomes.
Collapse
Affiliation(s)
- Panos Arvanitis
- Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Andreas Tziotis
- Beth Israel Deaconess Medical Center Division of Gastroenterology, Boston, MA 02115, USA; (A.T.); (S.P.)
| | - Spyridon Papadimatos
- Beth Israel Deaconess Medical Center Division of Gastroenterology, Boston, MA 02115, USA; (A.T.); (S.P.)
| | - Dimitrios Farmakiotis
- Beth Israel Deaconess Medical Center Division of Infectious Diseases, Boston, MA 02115, USA
| |
Collapse
|
20
|
Yin N, Yang R, Liu X, Chen X. Efficacy and safety of immunotherapy in real-world patients with advanced non-small cell lung cancer. Cancer Treat Res Commun 2025; 43:100908. [PMID: 40187204 DOI: 10.1016/j.ctarc.2025.100908] [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: 08/05/2024] [Revised: 02/28/2025] [Accepted: 03/20/2025] [Indexed: 04/07/2025]
Abstract
OBJECTIVE The aim of this study was to explore the predictors of immunotherapy efficacy for advanced non-small cell lung cancer (NSCLC) in the real world and to analyze the clinical efficacy and safety of patients receiving immunotherapy for advanced NSCLC. METHODS Clinical pathological data from patients diagnosed with advanced NSCLC treated with immune checkpoint inhibitors (ICIs) were collected. Survival analysis and differential efficacy comparison of progression-free survival (PFS) was performed using the Kaplan-Meier method and Log-rank test. Univariate and multivariate analyses of PFS and objective response rate (ORR) were performed by Cox proportional risk regression models and logistic regression models to explore influence factors associated with the prognosis of immunotherapy. RESULTS (1) Overall, the median PFS (median PFS, mPFS) for 237 patients was 11.3 months (range: 8.5-14.1), the ORR was 55.7 %. Univariate and multivariate analyses of PFS in the overall population found that age ≥65 years, Eastern Cooperative Oncology Group (ECOG) physical status (PS) score of 0-1, clinical stage III, absence of liver metastases, immunotherapy combined with chemotherapy and prognostic nutritional index (PNI) ≥47.8 were independent predictors of longer PFS in immunotherapy-advanced NSCLC. Univariate and multifactorial logistic regression analysis of ORR in 237 patients suggested that ECOG PS score, number of ICI lines and prognostic nutritional index (PNI) were independent influence factors of ORR. (2) In the "stage IV, first-line, ECOG PS 0-1" subgroup, 106(106/237)patients had an mPFS of 10.9 months (range: 9.6-12.2) and an ORR of 59.4 %. Univariate and multivariate analyses of PFS in subgroups found that liver metastases, immunotherapy combined with chemotherapy and PNI were independent influencers of PFS. A univariate analysis of ORR found that only High-PNI was associated with longer PFS. (3) Additional factors affecting the efficacy were explored. A subgroup analysis among 64 (64/237) patients with accessible programmed death-ligand 1 (PD-L1) expression levels showed a trend towards a PFS benefit in patients with PD-L1 tumor cell proportion score (TPS) ≥ 50 % and TPS < 1 % compared to patients with PD-L1 TPS < 1 % (p=0.196); A subgroup analysis among 91 (91/237) patients with traceable genetic test results showed that patients with positive driver genes (KRAS/MET/RET/HER2/EGFR/ALK) had a shorter PFS than patients with negative driver genes (HR=1.712, 95 % CI: 0.994-2.947, p=0.048); Subgroup analyses of efficacy assessment showed significantly prolonged PFS in patients with an initial or best outcome assessment of complete response (CR) or partial remission (PR) compared with stable disease (SD) or progressive disease (PD) (P < 0.001). (4) Immune-related adverse events (irAEs) requiring pharmacological intervention or discontinuation were recorded. 61 (61/237) patients experienced irAEs during treatment. Grade 1∼2 adverse reactions occurred in 27.8 % of patients and grade ≥3 adverse reactions in 3.8 % of patients. There was no statistical difference in the occurrence (P=0.728) and severity (P=0.612) of adverse events between the ICIs. CONCLUSION This study reports the practical experience of ICIs in the treatment of NSCLC in China by analyzing the efficacy and safety of real-world advanced NSCLC treated with ICIs. The results were generally consistent with those of clinical trials, and the factors with the greatest impact on the efficacy of ICIs were ECOG PS, clinical stage, and PNI. Therefore, physicians can predict the future benefit of immunotherapy for NSCLC based on clinical prognostic indicators and make individualized treatment choices.
Collapse
Affiliation(s)
- Na Yin
- Cancer Center, The First Hospital of Jilin University, Jilin University, Changchun City, Jilin Province, PR China.
| | - Ruihan Yang
- Cancer Center, The First Hospital of Jilin University, Jilin University, Changchun City, Jilin Province, PR China
| | - Xiangliang Liu
- Cancer Center, The First Hospital of Jilin University, Jilin University, Changchun City, Jilin Province, PR China
| | - Xiao Chen
- Cancer Center, The First Hospital of Jilin University, Jilin University, Changchun City, Jilin Province, PR China.
| |
Collapse
|
21
|
Da L, Qu Z, Zhang Y, Da J, Gu K. Correlative analysis of immune-related thyroid dysfunction and prognosis in patients with advanced esophageal squamous cell carcinoma. Anticancer Drugs 2025:00001813-990000000-00380. [PMID: 40112209 DOI: 10.1097/cad.0000000000001716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
To explore the clinical characteristics of immune-related thyroid dysfunction (TD) and its correlation with prognosis. By collecting the clinical data of 116 patients with advanced esophageal squamous cell carcinoma (ESCC) who received programmed death receptor-1 (PD-1) inhibitor treatment, we analyzed the clinical characteristics of immune-related TD and its influencing factors and compared the prognostic differences among patients in different groups. Immune-related TD occurred in 45 (38.8%) patients after PD-1 inhibitor treatment, and the median time to its occurrence was 11.3 weeks. The toxicity of immune-related TD was grade 1 or grade 2 and only required symptomatic treatment. Female patients, as well as those with an Eastern Cooperative Oncology Group Performance Status less than equal to 1, no lymph node metastasis, no history of drinking, and high baseline thyroid-stimulating hormone levels, were likely to develop immune-related TD. Compared with the patients in the group without immune-related TD [TD(-)], the median progression-free survival (mPFS) and median overall survival (mOS) of the patients in the immune-related TD [TD(+)] group were significantly prolonged (mPFS: 12.6 vs. 6.5 months, P = 0.001; mOS: 20.2 vs. 11.2 months, P < 0.001). Further subgroup analysis showed that compared with the patients in the group without immune-related overt TD (Overt_TD), the patients in the Overt_TD group had a longer PFS (mPFS: 12.4 vs. 7.3 months, P = 0.015) and OS (mOS: 20.2 vs. 12.2 months, P = 0.001). The 60-, 90-, and 120-day landmark analysis further confirmed that immune-related TD was significantly associated with the improvement of PFS and OS. Multivariate Cox regression analysis indicated that immune-related TD was an independent prognostic factor for PFS (P = 0.015) and OS (P = 0.004). Immune-related TD is a very common immune-related adverse event. It is safe and manageable and has potential prognostic value for patients with advanced ESCC treated with PD-1 inhibitors.
Collapse
Affiliation(s)
- Liangshan Da
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | | | | | | | | |
Collapse
|
22
|
Narote S, Desai SA, Patel VP, Deshmukh R, Raut N, Dapse S. Identification of new immune target and signaling for cancer immunotherapy. Cancer Genet 2025; 294-295:57-75. [PMID: 40154216 DOI: 10.1016/j.cancergen.2025.03.004] [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: 01/01/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
Immunotherapy has become one of the innovative treatments in malignancy as it activates the immune system to find and eliminate malignant cells. The tumor immunology interface has become increasingly intricate, making the identification of new immune targets and signalling pathways on which to base improved therapeutic strategies an ongoing process. This review, we goal to clarify the contacts between cancer and immune system with a focus on immune surveillance as well as immune evasion mechanisms. Comprehensive immunotherapeutic therapies are overviewed with ICI (CTLA-4, PD-1, PD-L1), CAR-T cell therapy, and cancer vaccines whereas, advanced therapies targeting new immune checkpoints are also elucidated including TIM-3, LAG-3, and TIGIT. The JAK/STAT, MAPK and PI3K-AKT-mTOR pathways are reviewed with regards to cancer progression and immunotherapeutic resistance. The dysregulation of these pathways gives hope for the identification of fresh targets for therapy. Genomics, proteomics, immunopeptidomics, single cell mass spectrometry, CRISPR-based functional genomics and bioinformatics are described as essential for immune target identification and for mapping of cancer relevant signaling pathways. This review also considers some emerging issues in the subject area like the tumor heterogeneity, immune-related adverse events (irAEs), and personalized treatment. These barriers are described to facilitate the understanding of ways to overcome them and increase the efficacy of immunotherapies through combination therapies. This means that by developing new knowledge of immunological targets and pathways, immunoprecision medicine for cancer could greatly enhance outcomes.
Collapse
Affiliation(s)
- Sakshi Narote
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Sharav A Desai
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India.
| | - Vipul P Patel
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Rutuja Deshmukh
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Nikita Raut
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Sejal Dapse
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| |
Collapse
|
23
|
Wang R, Wu J, Lin Y, Xiao Y, Yang B, Yao S, Pan T, Fu Z, Li S, Wang C, Zhu Y. An epitope-directed mRNA vaccine inhibits tumor metastasis through the blockade of MICA/B α1/2 shedding. Cell Rep Med 2025; 6:101981. [PMID: 39999840 PMCID: PMC11970329 DOI: 10.1016/j.xcrm.2025.101981] [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: 05/04/2024] [Revised: 11/27/2024] [Accepted: 01/28/2025] [Indexed: 02/27/2025]
Abstract
Antigenic peptide-based mRNA vaccines have been explored for immunotherapeutic use in various types of cancer because of their advantages in activating durable and specific immune responses. However, their role in modulating tumor metastasis is still unclear. Here, we identify a conserved linear epitope-based peptide, Ma3P, located in the proteolytic region of major histocompatibility complex (MHC) class I-related chain A (MICA) α3 and further design mCM10-L, an mRNA vaccine that encodes the carrier protein CRM197 and 10 tandem repeats of Ma3P. We demonstrate that vaccination with mCM10-L induces the production of specific antibodies that block MICA/B α1/2 shedding, activate CD8+ T cells and natural killer (NK) cells, and significantly inhibit MICA/B+ tumor metastasis in mice. Furthermore, mCM10-L stimulation triggers the production of specific antibodies to promote MICA/B-mediated immune killing in an in-vitro-interacting human organoid model and humanized mice. Our results indicate the potential clinical application prospects of the mCM10-L vaccine.
Collapse
Affiliation(s)
- Rui Wang
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jingni Wu
- Department of International Healthcare Center and General Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yifeng Lin
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yufei Xiao
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Bin Yang
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Sheng Yao
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tianhui Pan
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhixuan Fu
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Shuyu Li
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Caihua Wang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Yongliang Zhu
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
24
|
Zhang Z, Yan Z, Yuan T, Zhao X, Wang M, Liu G, Gan L, Qin W. PD-1 inhibition disrupts collagen homeostasis and aggravates cardiac dysfunction through endothelial-fibroblast crosstalk and EndMT. Front Pharmacol 2025; 16:1549487. [PMID: 40166462 PMCID: PMC11955664 DOI: 10.3389/fphar.2025.1549487] [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: 12/21/2024] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Cardiac immune-related adverse events (irAEs) from PD-1-targeting immune check-point inhibitors (ICIs) are an increasing concern due to their high mortality rate. Collagen plays a crucial role in maintaining cardiac structure, elasticity, and signal transduction; however, the effects and mechanisms of PD-1 inhibitor on cardiac collagen remodeling remain poorly understood. Methods C57BL/6 mice were injected with anti-mouse PD-1 antibody to create a PD-1 inhibitor-treated model. Cardiac function was measured by echocardiography, and collagen distribution was analyzed with Masson's trichrome staining and Sirius Red staining. Single-nucleus RNA sequencing was performed to examine the effects of PD-1 inhibition on gene expression in cardiac fibroblasts (CFs) and endothelial cells (ECs). EC-CF crosstalk was assessed using co-culture experiments and ELISA. ChIP assay was performed to analyze the regulation of TCF12 on TGF-β1 promoter. Western blot, qRT-PCR, and immunofluorescence staining were used to detect the expression of TCF12, TGF-β1, and endothelial-to-mesenchymal transition (EndMT) markers. Reactive oxygen species (ROS) levels were evaluated by DHE staining, MDA content, and SOD activity assays. Results We report a newly discovered cardiotoxic effect of PD-1 inhibitor, which causes aberrant collagen distribution in the heart, marked by a decrease in interstitial collagen and an increase in perivascular collagen deposition. Mechanistically, PD-1 inhibitor does not directly affect CFs but instead impact them through EC-CF crosstalk. PD-1 inhibitor reduces TGF-β1 secretion in ECs by downregulating TCF12, which we identify as a transcriptional promoter of TGF-β1. This subsequently decreases CF activity, leading to reduced interstitial collagen deposition. Additionally, PD-1 inhibitor induces EndMT, increasing perivascular collagen deposition. The endothelial dysfunction induced by PD-1 inhibitor results from ROS accumulation in ECs. Inhibiting ROS with N-acetylcysteine (NAC) preserves normal collagen distribution and cardiac function in PD-1 inhibitor-treated mice by reversing TCF12 downregulation and EndMT in ECs. Conclusion Our results suggest that PD-1 inhibitor causes ROS accumulation in cardiac ECs, leading to imbalanced collagen distribution (decrease in interstitial collagen and increase in perivascular collagen) in the heart by modulating TCF12/TGF-β1-mediated EC-CF crosstalk and EndMT. NAC supplementation could be an effective clinical strategy to mitigate PD-1 inhibitor-induced imbalanced collagen distribution and cardiac dysfunction.
Collapse
Affiliation(s)
- Zejin Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, China
| | - Zhenzhen Yan
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, China
- School of Pharmacy, Shandong University of Traditional Chinese medicine, Jinan, Shandong, China
| | - Tao Yuan
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, China
- School of Pharmacy, Shandong First Medical University, Jinan, Shandong, China
| | - Xiaona Zhao
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, China
- School of Pharmacy, Shandong Second Medical University, Weifang, Shandong, China
| | - Minghui Wang
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, China
- School of Pharmacy, Shandong University of Traditional Chinese medicine, Jinan, Shandong, China
| | - Guoqing Liu
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, China
- School of Pharmacy, Shandong University of Traditional Chinese medicine, Jinan, Shandong, China
| | - Lijun Gan
- Department of Cardiology (Shandong Provincial Key Laboratory for Cardiovascular Disease Diagnosis and Treatment), Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Wei Qin
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, China
- Department of Cardiology (Shandong Provincial Key Laboratory for Cardiovascular Disease Diagnosis and Treatment), Affiliated Hospital of Jining Medical University, Jining, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, Shandong, China
| |
Collapse
|
25
|
Feng S, Li Y, Tan Z, Shen S. Current landscape of metal-organic framework-mediated nucleic acid delivery and therapeutics. Int J Pharm 2025; 672:125295. [PMID: 39914507 DOI: 10.1016/j.ijpharm.2025.125295] [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: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/13/2025]
Abstract
Nucleic acid drugs utilize DNA or RNA molecules to modulate abnormal gene expression or protein translation in cells, enabling precise treatment for specific conditions. In recent years, nucleic acid drugs have demonstrated tremendous potential in vaccine development and treating genetic disorders. Currently, the primary carriers for clinically approved nucleic acid therapies include lipid nanoparticles and viral vectors. Beyond that, metal-organic frameworks (MOFs) are highly ordered, porous nanomaterials formed through the self-assembly of metal ions and organic ligands via coordination bonds. Their porosity structure offers great loading efficiency, stability, tunability, and biocompatibility, making them an attractive option for nucleic acid delivery. Given the research on MOFs as nucleic acid carriers has garnered significant attention in recent years, this review provides an overview of the therapeutic strategies and advancements in MOF-mediated nucleic acid delivery. The unique properties of various MOF carriers are introduced, and different approaches for nucleic acid loading are parallelly compared. Moreover, a systematic classification based on the type of nucleic acid cargo loaded in MOFs and corresponding applications is thoroughly described. This summary outlines the unique mechanisms through MOFs enhance nucleic acid delivery and emphasizes their substantial impact on therapeutic efficacy. In addition, the utilization of MOF-mediated nucleic acid treatment in combination with other therapies against malignant tumors is discussed in particular. Finally, an outlook on the challenges and potential opportunities of this technology in future translational production and clinical implementation is presented and explored.
Collapse
Affiliation(s)
- Shiwei Feng
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yan Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Zheng Tan
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China.
| | - Shiyang Shen
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; Pharmaceutical Preparation Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
26
|
Saberiyan M, Gholami S, Ejlalidiz M, Rezaeian Manshadi M, Noorabadi P, Hamblin MR. The dual role of chaperone-mediated autophagy in the response and resistance to cancer immunotherapy. Crit Rev Oncol Hematol 2025; 210:104700. [PMID: 40086769 DOI: 10.1016/j.critrevonc.2025.104700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025] Open
Abstract
Cancer immunotherapy has become a revolutionary strategy in oncology, utilizing the host immune system to fight malignancies. Notwithstanding major progress, obstacles such as immune evasion by tumors and the development of resistance still remain. This manuscript examines the function of chaperone-mediated autophagy (CMA) in cancer biology, focusing on its effects on tumor immunotherapy response and resistance. CMA is a selective degradation mechanism for cytosolic proteins, which is crucial for sustaining cellular homeostasis and regulating immune responses. By degrading specific proteins, CMA can either facilitate tumor progression in stressful conditions, or promote tumor suppression by removing oncogenic factors. This double-edged sword highlights the complexity of CMA in cancer progression and its possible effect on treatment results. Here we clarify the molecular mechanisms by which CMA can regulate the immune response and its possible role as a therapeutic target for improving the effectiveness of cancer immunotherapy.
Collapse
Affiliation(s)
- Mohammadreza Saberiyan
- Student Research Committee, Hormozgan University of Medical Sciences, Bandar Abbas, Iran; Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Sarah Gholami
- Young Researchers and Ellie Club, Babol Branch. Islamic Azad University, Babol, Iran
| | - Mahsa Ejlalidiz
- Medical Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadsadegh Rezaeian Manshadi
- Clinical Research Development Center, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parisa Noorabadi
- Department of Internal Medicine, School of Medicine, Urmia University of Medical sciences, Urmia, Iran.
| | - Michael R Hamblin
- Laser Research Centre, University of Johannesburg, Doornfontein, South Africa.
| |
Collapse
|
27
|
Abhiraman GC, Householder KD, Rodriguez GE, Glassman CR, Saxton RA, Breuer CB, Wilson SC, Su L, Yen M, Hsu C, Pillarisetty VG, Reticker-Flynn NE, Garcia KC. Redirecting immune signaling with cytokine adaptors. Nat Commun 2025; 16:2432. [PMID: 40069219 PMCID: PMC11897282 DOI: 10.1038/s41467-025-57681-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/28/2025] [Indexed: 03/15/2025] Open
Abstract
Cytokines are signaling molecules that coordinate complex immune processes and are frequently dysregulated in disease. While cytokine blockade has become a common therapeutic modality, cytokine agonism has had limited utility due to the widespread expression of cytokine receptors with pleiotropic effects. To overcome this limitation, we devise an approach to engineer molecular switches, termed cytokine adaptors, that transform one cytokine signal into an alternative signal with a different functional output. Endogenous cytokines act to nucleate the adaptors, converting the cytokine-adaptor complex into a surrogate agonist for a different cytokine pathway. In this way, cytokine adaptors, which have no intrinsic agonist activity, can function as conditional, context-dependent agonists. We develop cytokine adaptors that convert IL-10 or TGF-β into IL-2 receptor agonists to reverse T cell suppression. We also convert the pro-inflammatory cytokines IL-23 or IL-17 into immunosuppressive IL-10 receptor agonists. Thus, we show that cytokine adaptors can convert immunosuppressive cytokines into immunostimulatory cytokines, or vice versa. Unlike other methods of immune conversion that require cell engineering, cytokine adaptors are soluble molecules that leverage endogenous cues from the microenvironment to drive context-specific signaling.
Collapse
Affiliation(s)
- Gita C Abhiraman
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Karsten D Householder
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Grayson E Rodriguez
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Caleb R Glassman
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
| | - Robert A Saxton
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
| | - Cort B Breuer
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Steven C Wilson
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
| | - Leon Su
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
| | - Michelle Yen
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
| | - Cynthia Hsu
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, 98112, USA
| | - Venu G Pillarisetty
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, 98112, USA
| | - Nathan E Reticker-Flynn
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
28
|
Arroyo-Nogales A, Plaza-Palomo G, González-Larre J, Jiménez-Falcao S, Baeza A. Silicasomes in Oncology: From Conventional Chemotherapy to Combined Immunotherapy. Molecules 2025; 30:1257. [PMID: 40142031 PMCID: PMC11945772 DOI: 10.3390/molecules30061257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/21/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
The use of nanoparticles as drug carriers in oncology has evolved from their traditional role as chemotherapy carriers to their application in immunotherapy, exploiting not only their passive accumulation in solid tumors but also their ability to interact with immune cells. Silicasomes are highly versatile nanoplatforms composed of a mesoporous silica core whose external surface is coated with a lipid bilayer that allows the co-delivery of therapeutic agents having different chemical natures (small molecules, proteins, enzymes, or oligonucleotides, among others). Herein, cutting-edge advances carried out in the development and application of silicasomes are presented, providing a general description of the performance of these nanotransporters. Additionally, the specific load of chemotherapeutic drugs is explored, followed by a discussion of the immunotherapeutic application of silicasomes and the combination of different therapeutic strategies, including theragnosis, in a single silicasome platform, highlighting the enormous potential of these nanosystems.
Collapse
Affiliation(s)
| | | | | | | | - Alejandro Baeza
- Materials and Aerospace Production Department, Superior Technic School of Aeronautics and Space Engineering, Politechnic University of Madrid Department Materiales y Producción Aeroespacial, ETSI Aeronáutica y del Espacio, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (A.A.-N.); (G.P.-P.); (J.G.-L.); (S.J.-F.)
| |
Collapse
|
29
|
Xu Y, Sun D, He J, Yao Q. Cancer-associated fibroblasts in ovarian cancer: research progress. Front Oncol 2025; 15:1504762. [PMID: 40104498 PMCID: PMC11913710 DOI: 10.3389/fonc.2025.1504762] [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: 10/01/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Ovarian cancer, known for its high invasiveness and therapeutic resistance, is one of the leading causes of death from gynecological tumors. The tumor microenvironment (TME) plays a crucial role in the development of ovarian cancer, with cancer-associated fibroblasts (CAFs) being a key non-tumor cell component. They significantly affect the prognosis of ovarian cancer by promoting tumor cell proliferation, invasion, metastasis, immune evasion, and drug resistance. The heterogeneity of CAFs provides a new perspective for targeted therapy in ovarian cancer. This review comprehensively analyzes the mechanisms of action, heterogeneity characteristics, and role in the immune microenvironment of CAFs in ovarian cancer, and discusses targeted therapy strategies for CAFs, aiming to provide new theoretical basis and treatment directions for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yuance Xu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Danting Sun
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Junqi He
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qin Yao
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
30
|
Cao X, Wan S, Wu B, Liu Z, Xu L, Ding Y, Huang H. Antitumor Research Based on Drug Delivery Carriers: Reversing the Polarization of Tumor-Associated Macrophages. Mol Pharm 2025; 22:1174-1197. [PMID: 39868820 DOI: 10.1021/acs.molpharmaceut.4c01277] [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] [Indexed: 01/28/2025]
Abstract
The development of malignant tumors is a complex process that involves the tumor microenvironment (TME). An immunosuppressive TME presents significant challenges to current cancer therapies, serving as a key mechanism through which tumor cells evade immune detection and play a crucial role in tumor progression and metastasis. This impedes the optimal effectiveness of immunotherapeutic approaches, including cytokines, immune checkpoint inhibitors, and cancer vaccines. Tumor-associated macrophages (TAMs), a major component of tumor-infiltrating immune cells, exhibit dual functionalities: M1-like TAMs suppress tumorigenesis, while M2-like TAMs promote tumor growth and metastasis. Consequently, the development of various nanocarriers aimed at polarizing M2-like TAMs to M1-like phenotypes through distinct mechanisms has emerged as a promising therapeutic strategy to inhibit tumor immune escape and enhance antitumor responses. This Review covers the origin and types of TAMs, common pathways regulating macrophage polarization, the role of TAMs in tumor progression, and therapeutic strategies targeting TAMs, aiming to provide a comprehensive understanding and guidance for future research and clinical applications.
Collapse
Affiliation(s)
- Xinyu Cao
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Shen Wan
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Bingyu Wu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Zhikuan Liu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Lixing Xu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Yu Ding
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Haiqin Huang
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| |
Collapse
|
31
|
Tang L, Peng S, Zhuang X, He Y, Song Y, Nie H, Zheng C, Pan Z, Lam AK, He M, Shi X, Li B, Xu WW. Tumor Metastasis: Mechanistic Insights and Therapeutic Intervention. MEDCOMM – ONCOLOGY 2025; 4. [DOI: 10.1002/mog2.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/10/2025] [Indexed: 03/04/2025]
Abstract
ABSTRACTMetastasis remains a leading cause of cancer‐related deaths, defined by a complex, multi‐step process in which tumor cells spread and form secondary growths in distant tissues. Despite substantial progress in understanding metastasis, the molecular mechanisms driving this process and the development of effective therapies remain incompletely understood. Elucidating the molecular pathways governing metastasis is essential for the discovery of innovative therapeutic targets. The rapid advancements in sequencing technologies and the expansion of biological databases have significantly deepened our understanding of the molecular drivers of metastasis and associated drug resistance. This review focuses on the molecular drivers of metastasis, particularly the roles of genetic mutations, epigenetic changes, and post‐translational modifications in metastasis progression. We also examine how the tumor microenvironment influences metastatic behavior and explore emerging therapeutic strategies, including targeted therapies and immunotherapies. Finally, we discuss future research directions, stressing the importance of novel treatment approaches and personalized strategies to overcome metastasis and improve patient outcomes. By integrating contemporary insights into the molecular basis of metastasis and therapeutic innovation, this review provides a comprehensive framework to guide future research and clinical advancements in metastatic cancer.
Collapse
Affiliation(s)
- Lin Tang
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Shao‐Cong Peng
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Xiao‐Wan Zhuang
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Yan He
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Yu‐Xiang Song
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Hao Nie
- Department of Radiation Oncology, The Fifth Affiliated Hospital Guangzhou Medical University Guangzhou China
| | - Can‐Can Zheng
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Zhen‐Yu Pan
- Department of Radiation Oncology, The Affiliated Huizhou Hospital Guangzhou Medical University Huizhou China
| | - Alfred King‐Yin Lam
- Cancer Molecular Pathology and Griffith Medical School Griffith University Gold Coast Queensland Australia
| | - Ming‐Liang He
- Department of Biomedical Sciences City University of Hong Kong Hong Kong China
| | - Xing‐Yuan Shi
- Department of Radiation Oncology, The Fifth Affiliated Hospital Guangzhou Medical University Guangzhou China
| | - Bin Li
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Wen Wen Xu
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| |
Collapse
|
32
|
Zhang K, Li S, Li J, Zhou X, Qin Y, Wu L, Ling J. Ultra-pH-sensitive nanoplatform for precise tumor therapy. Biomaterials 2025; 314:122858. [PMID: 39366182 DOI: 10.1016/j.biomaterials.2024.122858] [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: 06/21/2024] [Revised: 09/16/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
The emergence of precision cancer treatment has triggered a paradigm shift in the field of oncology, facilitating the implementation of more effective and personalized therapeutic approaches that enhance patient outcomes. The pH of the tumor microenvironment (TME) plays a pivotal role in both the initiation and progression of cancer, thus emerging as a promising focal point for precision cancer treatment. By specifically targeting the acidic conditions inherent to the tumor microenvironment, innovative therapeutic interventions have been proposed, exhibiting significant potential in augmenting treatment efficacy and ameliorating patient prognosis. The concept of ultra-pH-sensitive (UPS) nanoplatform was proposed several years ago, demonstrating exceptional pH sensitivity and an adjustable pH transition point. Subsequently, diverse UPS nanoplatforms have been actively explored for biomedical applications, enabling the loading of fluorophores, therapeutic drugs, and photosensitizers. This review aims to elucidate the design strategy and response mechanism of the UPS nanoplatform, with a specific emphasis on its applications in surgical therapy, immunotherapy, drug delivery, photodynamic therapy, and photothermal therapy. The potential and challenges of translating in the clinic on UPS nanoplatforms are finally explored. Thanks to its responsive and easily modifiable nature, the integration of multiple functional units within a UPS nanoplatform holds great promise for future advancements in tumor precision theranositcs.
Collapse
Affiliation(s)
- Ke Zhang
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China
| | - Shijie Li
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China
| | - Jiaying Li
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China
| | - Xiaobo Zhou
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China.
| | - Yuling Qin
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China
| | - Li Wu
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China; School of Life Sciences, Nantong University, Nantong, Jiangsu, 226019, China.
| | - Jue Ling
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
33
|
Li J, Wang H, Li Y, Wang C, Feng H, Pang Y, Ren J, Li C, Gao E, Zhang D, Hu D, Zhao P, Ding H, Fan B, Zhang T, Song X, Wei Z, Ning G, Li YQ, Feng S. Novel carbon dots with dual Modulatory effects on the bone marrow and spleen as a potential therapeutic candidate for treating spinal cord injury. Bioact Mater 2025; 45:534-550. [PMID: 39759534 PMCID: PMC11696655 DOI: 10.1016/j.bioactmat.2024.11.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 11/24/2024] [Accepted: 11/24/2024] [Indexed: 01/07/2025] Open
Abstract
Spinal cord injury triggers leukocyte mobilization from the peripheral circulation to the injury site, exacerbating spinal cord damage. Simultaneously, bone marrow hematopoietic stem cells (HSCs) and splenic leukocytes rapidly mobilize to replenish the depleted peripheral blood leukocyte pool. However, current treatments for spinal cord injuries overlook interventions targeting peripheral immune organs and tissues, highlighting the need to develop novel drugs capable of effectively regulating peripheral immunity and treating spinal cord injuries. In this study, we designed, synthesized, and characterized novel Ejiao carbon dots (EJCDs) that inhibit myeloid cell proliferation and peripheral migration by promoting HSC self-renewal, and distinct differentiation into erythroid progenitors in vitro and in vivo. Additionally, EJCDs attenuate the immune response in the spleen, leukocytes' reservoir, following spinal cord injury by diminishing the local infiltration of monocytes and macrophages while promoting motor function recovery. These effects are mediated through the downregulation of CCAAT enhancer binding protein-β expression in the spleen and the upregulation of FZD4 protein expression in Lin- Sca-1+ c-kit+ cells (LSKs) within the bone marrow. Our findings demonstrate that EJCDs effectively reduce myeloid cell infiltration post-spinal cord injury and promote neurological recovery, making them promising therapeutic candidates for treating spinal cord injuries.
Collapse
Affiliation(s)
- Junjin Li
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, 300070, China
| | - Hongda Wang
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, 300070, China
| | - Yuanquan Li
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, 300070, China
| | - Chunzhen Wang
- Institute of Advanced Interdisciplinary Science, School of Physics, Shandong University, Jinan, 250100, China
| | - Haiwen Feng
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, 300070, China
| | - Yilin Pang
- Orthopedic Research Center of Shandong University and Department of Orthopedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Jie Ren
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, 300070, China
| | - Chuanhao Li
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, 300070, China
| | - Erke Gao
- The First Central Clinical School, Tianjin Medical University, Tianjin, 300070, China
| | - Dejing Zhang
- The First Central Clinical School, Tianjin Medical University, Tianjin, 300070, China
| | - Dunxu Hu
- The First Central Clinical School, Tianjin Medical University, Tianjin, 300070, China
| | - Pengtian Zhao
- The First Central Clinical School, Tianjin Medical University, Tianjin, 300070, China
| | - Han Ding
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, 300070, China
| | - Baoyou Fan
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, 300070, China
| | - Tao Zhang
- Department of Orthopaedics, The Second Hospital of Shandong University, No. 247 Beiyuan Street, Tianqiao District, Jinan, 250033, China
| | - Xiaomeng Song
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, 300070, China
| | - Zhijian Wei
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, 300070, China
- Orthopedic Research Center of Shandong University and Department of Orthopedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Guangzhi Ning
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, 300070, China
| | - Yong-Qiang Li
- Institute of Advanced Interdisciplinary Science, School of Physics, Shandong University, Jinan, 250100, China
| | - Shiqing Feng
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, 300070, China
- Orthopedic Research Center of Shandong University and Department of Orthopedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- Department of Orthopaedics, The Second Hospital of Shandong University, No. 247 Beiyuan Street, Tianqiao District, Jinan, 250033, China
| |
Collapse
|
34
|
Xu J, Liu Y. Nanomaterials for liver cancer targeting: research progress and future prospects. Front Immunol 2025; 16:1496498. [PMID: 40092984 PMCID: PMC11906451 DOI: 10.3389/fimmu.2025.1496498] [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: 09/14/2024] [Accepted: 01/07/2025] [Indexed: 03/19/2025] Open
Abstract
The incidence and mortality rates of liver cancer in China remain elevated. Although early-stage liver cancer is amenable to surgical resection, a significant proportion of patients are diagnosed at advanced stages. Currently, in addition to surgical resection for hepatocellular carcinoma, the primary treatment modalities predominantly include chemotherapy. The widespread use of chemotherapy, which non-selectively targets both malignant and healthy cells, often results in substantial immunosuppression. Simultaneously, the accumulation of chemotherapeutic agents can readily induce drug resistance upon reaching the physiological threshold, thereby diminishing the efficacy of these treatments. Besides chemotherapy, there exist targeted therapy, immunotherapy and other therapeutic approaches. Nevertheless, the development of drug resistance remains an inevitable challenge. To address these challenges, we turn to nanomedicine, an emerging and widely utilized discipline that significantly influences medical imaging, antimicrobial strategies, drug delivery systems, and other related areas. Stable and safe nanomaterials serve as effective carriers for delivering anticancer drugs. They enhance the precision of drug targeting, improve bioavailability, and minimize damage to healthy cells. This review focuses on common nanomaterial carriers used in hepatocellular carcinoma (HCC) treatment over the past five years. The following is a summary of the three drugs: Sorafenib, Gefitinib, and lenvatinib. Each drug employs distinct nanomaterial delivery systems, which result in varying levels of bioavailability, drug release rates, and therapeutic efficacy.
Collapse
Affiliation(s)
- Jiahong Xu
- Department of Hepatopancreatobiliary Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Yefu Liu
- Department of Hepatopancreatobiliary Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
35
|
Bandara S, Raveendran S. Current Landscape and Future Directions in Cancer Immunotherapy: Therapies, Trials, and Challenges. Cancers (Basel) 2025; 17:821. [PMID: 40075668 PMCID: PMC11899461 DOI: 10.3390/cancers17050821] [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: 01/10/2025] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a leading global health challenge, placing immense burdens on individuals and healthcare systems. Despite advancements in traditional treatments, significant limitations persist, including treatment resistance, severe side effects, and disease recurrence. Immunotherapy has emerged as a promising alternative, leveraging the immune system to target and eliminate tumour cells. However, challenges such as immunotherapy resistance, patient response variability, and the need for improved biomarkers limit its widespread success. This review provides a comprehensive analysis of the current landscape of cancer immunotherapy, highlighting both FDA-approved therapies and novel approaches in clinical development. It explores immune checkpoint inhibitors, cell and gene therapies, monoclonal antibodies, and nanotechnology-driven strategies, offering insights into their mechanisms, efficacy, and limitations. By integrating emerging research and clinical advancements, this review underscores the need for continued innovation to optimise cancer immunotherapy and overcome existing treatment barriers.
Collapse
Affiliation(s)
- Shehani Bandara
- School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK
- National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
| | - Sreejith Raveendran
- School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK
- National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
| |
Collapse
|
36
|
Yang M, Zhong P, Wei P. Living Bacteria: A New Vehicle for Vaccine Delivery in Cancer Immunotherapy. Int J Mol Sci 2025; 26:2056. [PMID: 40076679 PMCID: PMC11900161 DOI: 10.3390/ijms26052056] [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: 01/26/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer vaccines, aimed at evolving the human immune system to eliminate tumor cells, have long been explored as a method of cancer treatment with significant clinical potential. Traditional delivery systems face significant challenges in directly targeting tumor cells and delivering adequate amounts of antigen due to the hostile tumor microenvironment. Emerging evidence suggests that certain bacteria naturally home in on tumors and modulate antitumor immunity, making bacterial vectors a promising vehicle for precision cancer vaccines. Live bacterial vehicles offer several advantages, including tumor colonization, precise drug delivery, and immune stimulation, making them a compelling option for cancer immunotherapy. In this review, we explore the mechanisms of action behind living bacteria-based vaccines, recent progress in popular bacterial chassis, and strategies for specific payload delivery and biocontainment to ensure safety. These approaches will lay the foundation for developing an affordable, widely applicable cancer vaccine delivery system. This review also discusses the challenges and future opportunities in harnessing bacterial-based vaccines for enhanced therapeutic outcomes in cancer treatment.
Collapse
Affiliation(s)
| | | | - Pengcheng Wei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (M.Y.); (P.Z.)
| |
Collapse
|
37
|
Zhao X, Wang X, Liu S, Cheng P, Chen J, Liu J. Severe thyroiditis induced by sintilimab monotherapy in a patient with non-small cell lung cancer: a case report and literature review. Front Immunol 2025; 16:1548452. [PMID: 40070833 PMCID: PMC11893825 DOI: 10.3389/fimmu.2025.1548452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Thyroid dysfunction is a common immune-related adverse event (irAE) associated with immune checkpoint inhibitors (ICIs) that target PD-1, PD-L1, and CTLA-4. Nevertheless, the incidence of severe cases, defined as grade 3 or higher, remains rare. This report presents a detailed case study of severe thyroiditis in a patient with non-small cell lung cancer (NSCLC) who developed grade 3 thyroiditis following a single cycle of sintilimab monotherapy. The clinical presentation in this patient was remarkable for its early onset, occurring one week after the initiation of sintilimab therapy, and for its severe manifestations. During hospitalization, a prompt and accurate differential diagnosis was performed. Sintilimab treatment was discontinued, and the patient was promptly started on high-dose glucocorticoids, with a tapering schedule implemented as the condition improved or reached Common Terminology Criteria for Adverse Events (CTCAE) grade 1 or lower. The patient subsequently developed overt hypothyroidism, necessitating the initiation of thyroxine replacement therapy. Furthermore, we provide a comprehensive review of the mechanisms and risk factors associated with thyroid dysfunction immune-related adverse events (TD-irAEs). It is imperative for clinicians to meticulously monitor the clinical symptoms exhibited by patients. For those presenting with symptoms, prompt diagnosis and appropriate symptomatic management are essential. Additionally, regular thyroid function testing is recommended for high-risk patients, and we advocate for the assessment of baseline levels of thyroid peroxidase antibodies (TPOAb) and thyroglobulin antibodies (TGAb) prior to initiating ICI treatment.
Collapse
Affiliation(s)
- Xiaolin Zhao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Xiaoyu Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Surui Liu
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Pian Cheng
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Jinjuan Chen
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Jie Liu
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
38
|
Zheng M, Huang W, Wang D, Huang L, Ren Y, Gao Q, Huang Y, Lin W, Chen L. Prognostic assessment of cervical cancer based on biomarkers: the interaction of ERRα and immune microenvironment. Virol J 2025; 22:47. [PMID: 39994715 PMCID: PMC11852515 DOI: 10.1186/s12985-025-02664-3] [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: 12/10/2024] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Cervical cancer poses a substantial global health challenge. Estrogen-related receptor alpha (ERRα) is a central regulator of cellular energy metabolism associated with poor cancer prognosis. However, the effect of ERRα expression on cervical cancer prognosis and immune infiltration has not been explored. This study aims to clarify the expression pattern and role of ERRα in cervical cancer. METHODS We analyzed ERRα expression and its clinical prognosis in cervical cancer using multiple databases, including The Cancer Genome Atlas (TCGA) and Tumor Immune Estimation Resource (TIMER). The results were further validated through immunohistochemistry (IHC) on 221 cervical cancer tissue samples. Furthermore, Kaplan-Meier and Cox regression analyses were used to assess the clinical significance of ERRα in cervical cancer patients. All calculations were performed using the R package. RESULTS ERRα expression was significantly higher in cervical cancer tissues compared to normal tissues. High ERRα expression was associated with poor overall survival (OS), disease-specific survival (DSS), and progression-free survival (PFS). Multivariate Cox regression analysis confirmed ERRα as an independent prognostic factor. Additionally, ERRα expression correlated with various immune cell types and immune checkpoints, indicating its role in the tumor immune microenvironment. CONCLUSIONS ERRα emerges as a promising prognostic biomarker in cervical cancer, influencing immune cell infiltration and potentially guiding personalized therapeutic approaches. Future investigations are warranted to delineate the mechanistic pathways through which ERRα contributes to cervical cancer progression and to assess its viability as a target for innovative immunotherapy strategies.
Collapse
Affiliation(s)
- Meijin Zheng
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Weifeng Huang
- Department of Radiation Oncology, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, 363000, China
| | - Dingjie Wang
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Leyi Huang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350001, Fujian, China
| | - Yuan Ren
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Qiao Gao
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Yuxuan Huang
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Wenyu Lin
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China.
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China.
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China.
| | - Lihua Chen
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China.
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China.
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China.
| |
Collapse
|
39
|
Wang Q, Zhang H, Chen Y, Lv X, Qiao Y, Zhu Q. Impact of baseline glucocorticoids (GCs) on cardiotoxic events and myocardial damage related to immune checkpoint inhibitors: a retrospective clinical research. Expert Opin Drug Saf 2025:1-12. [PMID: 39953683 DOI: 10.1080/14740338.2025.2467814] [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: 06/24/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs)-associated cardiotoxic events (CEs) are of increasing concern. Existing research about glucocorticoids (GCs) on immunotherapy focused on ICIs' efficacy and patients' outcome. The influence of GCs on ICIs-associated CEs and myocardial damage (MD) remains unknown. RESEARCH DESIGN AND METHODS This single-center retrospective study included patients treated with ICIs from 2018 to 2022, with follow-up period ending on 30 June 2023. The incidence, risk factors of ICIs-associated CEs, especially MD were described. Additionally, the impact of baseline GCs was assessed by propensity score matching (PSM) to mitigate intergroup differences and ensure comparability. RESULTS Among 1018 patients, 204 (20.04%) experienced ICIs-associated CEs, including 71 (6.97%) with MD. The mean follow-up time was 40.39 (95% CI 38.47-42.31) weeks. The median time to onset of MD was the shortest at 12.57 weeks (IQR 5.29-25.14). Tumor type, co-medication with platinum and angiogenesis inhibitors may be influential factors of MD. After PSM, the relative risks of CEs (OR 0.4625,95%CI 0.2514-0.7235, p = 0.0020) and MD (OR 0.3254, 95% CI 0.1190-0.8898, p = 0.0378) in GCs1 ≥ 20 mg group were both significantly lower than those in GCs1 < 20 mg. CONCLUSION GCs ≥ 20 mg during the first ICIs treatment cycle is significantly associated with the reduced risks of both ICIs-associated CEs and MD.
Collapse
Affiliation(s)
- Qiaoyun Wang
- Department of Pharmacy, Nanjing Drum Tower Hospital, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Department of Pharmacy, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Haixia Zhang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau, SAR, China
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Nanjing Medical Center for Clinical Pharmacy, Nanjing, China
| | - Yawen Chen
- Department of Pharmacy, Nanjing Drum Tower Hospital, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xin Lv
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yanli Qiao
- Department of Pharmacy, Nanjing Drum Tower Hospital, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qiaoling Zhu
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Nanjing Medical Center for Clinical Pharmacy, Nanjing, China
| |
Collapse
|
40
|
Shi T, Zhang H, Chen Y. The m6A revolution: transforming tumor immunity and enhancing immunotherapy outcomes. Cell Biosci 2025; 15:27. [PMID: 39987091 PMCID: PMC11846233 DOI: 10.1186/s13578-025-01368-z] [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: 12/09/2024] [Accepted: 02/14/2025] [Indexed: 02/24/2025] Open
Abstract
N6-methyladenosine (m6A), the most prevalent RNA modification in eukaryotes, plays a critical role in the development and progression of various diseases, including cancer, through its regulation of RNA degradation, stabilization, splicing, and cap-independent translation. Emerging evidence underscores the significant role of m6A modifications in both pro-tumorigenic and anti-tumorigenic immune responses. In this review, we provide a comprehensive overview of m6A modifications and examine the relationship between m6A regulators and cancer immune responses. Additionally, we summarize recent advances in understanding how m6A modifications influence tumor immune responses by directly modulating immune cells (e.g., dendritic cells, tumor-associated macrophages, and T cells) and indirectly affecting cancer cells via mechanisms such as cytokine and chemokine regulation, modulation of cell surface molecules, and metabolic reprogramming. Furthermore, we explore the potential synergistic effects of targeting m6A regulators in combination with immune checkpoint inhibitor (ICI) therapies. Together, this review consolidates current knowledge on the role of m6A-mediated regulation in tumor immunity, offering insights into how a deeper understanding of these modifications may identify patients who are most likely to benefit from immunotherapies.
Collapse
Affiliation(s)
- Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China.
| | - Huan Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China
| | - Yueqiu Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China.
| |
Collapse
|
41
|
Lv P, Zhang Y, Wu W, Jiang X, Xiang W. Pan-cancer analysis identifies ADAM12 as a prognostic biomarker and indicator of immune infiltration in glioma. Sci Rep 2025; 15:6314. [PMID: 39984619 PMCID: PMC11845722 DOI: 10.1038/s41598-025-90121-0] [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: 11/20/2024] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
ADAM12, part of the adisintegrin and metalloproteases (ADAMs) family, has been widely reported in recent years to be associated with various malignant tumor behaviors, including migration, invasion, and treatment resistance. However, its role at the pan-cancer level remains insufficiently characterized. In this study, pan-cancer data were utilized to elucidate the expression patterns, prognostic significance, and potential roles of ADAM12 within the tumor immune microenvironment. An in-depth analysis of ADAM12 in gliomas was also conducted. Our findings revealed that ADAM12 expression is markedly overexpressed in glioma tissues, enhances glioma cell malignancy, and is associated with a worse prognosis. These results suggest that ADAM12 may serve as a biomarker for predicting glioma malignancy and patient prognosis, as well as a potential therapeutic target in glioma treatment.
Collapse
Affiliation(s)
- Peng Lv
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue NO.1277, Wuhan, 430022, Hubei Province, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yanbin Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue NO.1277, Wuhan, 430022, Hubei Province, China
| | - Wenjie Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue NO.1277, Wuhan, 430022, Hubei Province, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue NO.1277, Wuhan, 430022, Hubei Province, China.
| | - Wei Xiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue NO.1277, Wuhan, 430022, Hubei Province, China.
| |
Collapse
|
42
|
Zheng H, Yu J, Gao L, Wang K, Xu Z, Zeng Z, Zheng K, Tang X, Tian X, Zhao Q, Zhao J, Wan H, Cao Z, Zhang K, Cheng J, Brosius J, Zhang H, Li W, Yan W, Shao Z, Luo F, Deng C. S1PR1-biased activation drives the resolution of endothelial dysfunction-associated inflammatory diseases by maintaining endothelial integrity. Nat Commun 2025; 16:1826. [PMID: 39979282 PMCID: PMC11842847 DOI: 10.1038/s41467-025-57124-x] [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: 05/02/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
G protein-coupled sphingosine-1-phosphate receptor 1 (S1PR1), a drug target for inflammatory bowel disease (IBD), enables immune cells to egress from lymph nodes, but the treatment increases the risk of immunosuppression. The functional signaling pathway triggered by S1PR1 activation in endothelial cells and its therapeutic application remains unclear. Here, we showed that S1PR1 is highly expressed in endothelial cells of IBD patients and positively correlated with endothelial markers. Gi-biased agonist-SAR247799 activated S1PR1 and reversed pathology in male mouse and organoid IBD models by protecting the integrity of the endothelial barrier without affecting immune cell egress. Cryo-electron microscopy structure of S1PR1-Gi signaling complex bound to SAR247799 with a resolution of 3.47 Å revealed the recognition mode for the biased ligand. With the efficacy of SAR247799 in treating other endothelial dysfunction-associated inflammatory diseases, our study offers mechanistic insights into the Gi-biased S1PR1 agonist and represents a strategy for endothelial dysfunction-associated disease treatment.
Collapse
Affiliation(s)
- Huaping Zheng
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jingjing Yu
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Luhua Gao
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Kexin Wang
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Zheng Xu
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Zhen Zeng
- Department of Gastroenterology, Lab of Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Kun Zheng
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoju Tang
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaowen Tian
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Zhao
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Zhao
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Huajing Wan
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongwei Cao
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Kang Zhang
- Center for Biomedicine and Innovations, Faculty of Medicine, Macau University of Science and Technology and University Hospital, Macau, China
| | - Jingqiu Cheng
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jürgen Brosius
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hu Zhang
- Department of Gastroenterology, Lab of Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Li
- Department of Dermatology, Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Yan
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China.
| | - Zhenhua Shao
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China.
| | - Fengming Luo
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| | - Cheng Deng
- Department of Respiratory and Critical Care Medicine, Center for High Altitude Medicine, Institutes for Systems Genetics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
43
|
Verma T, Jawadi A, Ahmed S. Management of multiorgan failure caused by immune checkpoint inhibitor toxicity. BMJ Case Rep 2025; 18:e262209. [PMID: 39961676 PMCID: PMC11848246 DOI: 10.1136/bcr-2024-262209] [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: 07/08/2024] [Accepted: 01/18/2025] [Indexed: 02/26/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) block inhibitory pathways that tumour cells exploit to evade the immune system. They play a key role in the management of aggressive cancers, including non-small cell lung cancer, melanoma and Hodgkin's lymphoma. Despite their efficacy, ICIs can cause severe, potentially fatal, immune-related adverse events (irAEs), including multiorgan failure. We present a case of a woman in her early 70s with metastatic lung adenocarcinoma recently initiated on pembrolizumab who presented with an acute stroke complicated by irAE-related multiorgan failure. Despite aggressive treatment, the patient remained critically ill and passed away the same day after transitioning to end-of-life care. This case emphasises the life-threatening potential of irAEs, the importance of early diagnosis and the challenges of managing these complications. Clinicians should maintain a high index of suspicion for irAEs in ICI-treated patients to optimise timely intervention and outcomes.
Collapse
Affiliation(s)
- Tushita Verma
- Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Altamash Jawadi
- Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Shozab Ahmed
- Pulmonary and Critical Care, University of New Mexico, Albuquerque, New Mexico, USA
| |
Collapse
|
44
|
Li Z, Xi Z, Fan C, Xi X, Zhou Y, Zhao M, Xu L. Nanomaterials evoke pyroptosis boosting cancer immunotherapy. Acta Pharm Sin B 2025; 15:852-875. [PMID: 40177577 PMCID: PMC11959974 DOI: 10.1016/j.apsb.2024.11.011] [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: 07/10/2024] [Revised: 09/12/2024] [Accepted: 10/15/2024] [Indexed: 04/05/2025] Open
Abstract
Cancer immunotherapy is currently a very promising therapeutic strategy for treating tumors. However, its effectiveness is restricted by insufficient antigenicity and an immunosuppressive tumor microenvironment (ITME). Pyroptosis, a unique form of programmed cell death (PCD), causes cells to swell and rupture, releasing pro-inflammatory factors that can enhance immunogenicity and remodel the ITME. Nanomaterials, with their distinct advantages and different techniques, are increasingly popular, and nanomaterial-based delivery systems demonstrate significant potential to potentiate, enable, and augment pyroptosis. This review summarizes and discusses the emerging field of nanomaterials-induced pyroptosis, focusing on the mechanisms of nanomaterials-induced pyroptosis pathways and strategies to activate or enhance specific pyroptosis. Additionally, we provide perspectives on the development of this field, aiming to accelerate its further clinical transition.
Collapse
Affiliation(s)
- Zhenhua Li
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ziyue Xi
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chuanyong Fan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinran Xi
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yao Zhou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ming Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lu Xu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
45
|
Lu Y, Ma N, Cheng K, Liu G, Liang J, Xu C, Li D, Cao C, Gao X, Chen L, Wang X, Wang Y, Zhao X, Jiang K. An OMV-Based Nanovaccine as Antigen Presentation Signal Enhancer for Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413392. [PMID: 39811977 DOI: 10.1002/adma.202413392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Antigen-presenting cells (APCs) process tumor vaccines and present tumor antigens as the first signals to T cells to activate anti-tumor immunity, which process requires the assistance of co-stimulatory second signals on APCs. The immune checkpoint programmed death ligand 1 (PD-L1) not only mediates the immune escape of tumor cells but also acts as a co-inhibitory second signal on APCs. The serious dysfunction of second signals due to the high expression of PD-L1 on APCs in the tumor body results in the inefficiency of tumor vaccines. To overcome this challenge, a previously established Plug-and-Display tumor vaccine platform based on bacterial outer membrane vesicles (OMVs) is developed into an "Antigen Presentation Signal Enhancer" (APSE) by surface-modifying PD-L1 antibodies (αPD-L1). While delivering tumor antigens, APSE can activate the expression of co-stimulatory second signals in APCs due to the high immunogenicity of OMVs. More importantly, the surface-modified αPD-L1 binds to the co-inhibitory signals PD-L1, potentially restoring CD80 function and ensuring efficient co-stimulatory second signals and activation of anti-tumor immunity. The results reveal the importance of PD-L1 blockage in the initiation process of anti-tumor immunity, and the second signal modulation capability of APSE can expand the application potential of cancer vaccines to less immunogenic malignancies.
Collapse
Affiliation(s)
- Yichao Lu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Nana Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chen Xu
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, China
| | - Danrui Li
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
| | - Cheng Cao
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
| | - Xiaoyu Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Liting Chen
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, China
| | - Xinwei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yazhou Wang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Kuirong Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
| |
Collapse
|
46
|
Lin J, Li W, Zhang X, Zhou K, Yang Y, Cheng S, Sun R, Dang C, Diao D. Thromboembolic events associated with immune checkpoint inhibitors in cancer patients: A Bayesian network meta-analysis. Thromb Res 2025; 246:109243. [PMID: 39721224 DOI: 10.1016/j.thromres.2024.109243] [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: 08/20/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs), which offer previously unknown therapeutic advantages, have revolutionized cancer treatment. However, the risk of thromboembolic events (TEEs) associated with ICIs remains unclear. The aim of this network meta-analysis (NMA) was to evaluate the incidence of TEEs in cancer patients receiving different treatment regimens. METHODS We searched for randomized clinical trials (RCTs) between January 2021 and December 2023 without restricting the cancer type. The percentages of TEEs were systematically extracted. An NMA was performed comparing atezolizumab, cemiplimab, durvalumab, ipilimumab, nivolumab, pembrolizumab, conventional therapy (which consists mainly of chemotherapy, targeted therapy, placebo, and their combinations), two ICI drugs, one ICI drug combined with conventional therapy, and two ICI drugs combined with conventional therapy. Additionally, subgroup analysis was conducted based on cancer type. RESULTS Eighty-three RCTs involving 54,736 patients were included. Patients receiving ICIs demonstrated comparable risks of arterial thromboembolism (ATE), deep vein thrombosis (DVT), myocardial infarction (MI), and cerebrovascular accidents (CVAs). Nivolumab (OR 0.39, 95 % CI 0.19 to 0.80) and two ICI drugs (OR 0.52, 95 % CI 0.29 to 0.89) had the lowest risk of venous thromboembolism (VTE) compared to two ICI drugs with conventional therapy. The risk of pulmonary embolism (PE) was greater for ipilimumab (OR 4.09, 95 % CI 1.13 to 15.51) than for nivolumab. For melanoma in the subgroup analysis, nivolumab significantly reduced the risk of VTE (OR 0.07, 95 % CI 0.00 to 0.76) compared to two ICI drugs. Among the single-ICI regimens, durvalumab was associated with the highest incidence of ATE, MI, and CVAs; ipilimumab had the highest incidence of VTE and PE; and pembrolizumab had the highest incidence of DVT. The combination of one ICI drug with conventional therapy was associated with a significantly greater risk of TEEs (except for MI) than the combination of two ICI drugs. CONCLUSIONS Various ICI regimens in cancer patients exhibit clinically significant differences in the risks of TEEs. Nivolumab exhibited a favorable safety profile regarding VTE, while ipilimumab had the highest risk of both VTE and PE. Different ICI regimens require tailored risk management strategies to reduce TEEs.
Collapse
Affiliation(s)
- Jinhe Lin
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Wenxing Li
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xin Zhang
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Kai Zhou
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yanqi Yang
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Shaoli Cheng
- Basic Medical Experiment Teaching Center, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Ruifang Sun
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Chengxue Dang
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Dongmei Diao
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
47
|
Nie F, Guo J, Pan J, Guo Z, Wang C, Yan J, Ma W. Effects of antibiotics on the anti-tumor efficacy of immune checkpoint inhibitor therapy. Clin Transl Oncol 2025; 27:790-799. [PMID: 39046682 DOI: 10.1007/s12094-024-03615-0] [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: 05/05/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024]
Abstract
PURPOSE Immunotherapy using immune checkpoint inhibitors (ICIs) has shown several benefits over traditional therapies. However, the eligible population remains small. Antibiotic (ATB) use might reduce immunotherapy efficacy by disrupting the gut microbiota. However, in China, ATB effect on ICI therapy efficacy remains unelucidated. We aimed to assess the effects of ATBs on the anti-tumor efficacy of ICIs to provide a reference for clinical use. METHODS We included 134 patients with advanced tumors undergoing ICI therapy at Shanghai Jiading District Central Hospital from January 1, 2021, to October 1, 2023. They were divided into Non-ATB and ATB groups based on ATB use within 30 days before and after ICI administration. Moreover, we compared progression-free (PFS) and overall (OS) survival between the groups. RESULTS Median PFS and OS were lower in the ATB than in the Non-ATB group (PFS: 4.0 vs. 5.5 months; OS: 5.4 vs. 6.5 months). Univariate analysis revealed that ATB use significantly affected PFS (hazard ratio [HR] = 2.318, 95% confidence interval [CI] = 1.281-4.194, P = 0.005) and OS (HR = 2.115, 95% CI = 1.161-3.850, P = 0.014). Moreover, multivariate analysis revealed poor PFS (HR = 2.573, 95% CI = 1.373-4.826, P = 0.003) and OS (HR = 2.452, 95% CI = 1.298-4.632, P = 0.006) in patients who received ATBs during ICI therapy. CONCLUSIONS ATB use is negatively correlated with ICI therapy efficacy, leading to reduced PFS and OS in patients undergoing such treatment. Owing to the significant impact of ATBs on the human gut microbiome, regulation of the gut microbiome may emerge as a novel therapeutic target that can enhance the clinical activity of ICIs.
Collapse
Affiliation(s)
- Fangfang Nie
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - JunGang Guo
- Department of Thoracic Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - JiaYi Pan
- Department of Pharmacy, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, No.1 Cheng Bei Road, Jia ding District, Shanghai, 201800, China
| | - ZhaoJiao Guo
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - Chun Wang
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - Jun Yan
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China.
| | - WeiNa Ma
- Department of Pharmacy, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, No.1 Cheng Bei Road, Jia ding District, Shanghai, 201800, China.
| |
Collapse
|
48
|
Guo Z, Liu Y, Chen D, Sun Y, Li D, Meng Y, Zhou Q, Zeng F, Deng G, Chen X. Targeting regulated cell death: Apoptosis, necroptosis, pyroptosis, ferroptosis, and cuproptosis in anticancer immunity. J Transl Int Med 2025; 13:10-32. [PMID: 40115032 PMCID: PMC11921819 DOI: 10.1515/jtim-2025-0004] [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] [Indexed: 03/22/2025] Open
Abstract
In the evolving landscape of cancer treatment, the strategic manipulation of regulated cell death (RCD) pathways has emerged as a crucial component of effective anti-tumor immunity. Evidence suggests that tumor cells undergoing RCD can modify the immunogenicity of the tumor microenvironment (TME), potentially enhancing its ability to suppress cancer progression and metastasis. In this review, we first explore the mechanisms of apoptosis, necroptosis, pyroptosis, ferroptosis, and cuproptosis, along with the crosstalk between these cell death modalities. We then discuss how these processes activate antigen-presenting cells, facilitate the cross-priming of CD8+ T cells, and trigger anti-tumor immune responses, highlighting the complex effects of novel forms of tumor cell death on TME and tumor biology. Furthermore, we summarize potential drugs and nanoparticles that can induce or inhibit these emerging RCD pathways and their therapeutic roles in cancer treatment. Finally, we put forward existing challenges and future prospects for targeting RCD in anti-cancer immunity. Overall, this review enhances our understanding of the molecular mechanisms and biological impacts of RCD-based therapies, providing new perspectives and strategies for cancer treatment.
Collapse
Affiliation(s)
- Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yihuang Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Danyao Chen
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yuming Sun
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Daishi Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yu Meng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Qian Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Furong Zeng
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
49
|
Nie L, Ma J, Yu Y, Tao Y, Song Z, Li J. Exosomes as carriers to stimulate an anti-cancer immune response in immunotherapy and as predictive markers. Biochem Pharmacol 2025; 232:116699. [PMID: 39647605 DOI: 10.1016/j.bcp.2024.116699] [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: 09/29/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
During this era of rapid advancements in cancer immunotherapy, the application of cell-released small vesicles that activate the immune system is of considerable interest. Exosomes are cell-derived nanovesicles that show great promise for the immunological treatment of cancer because of their immunogenicity and molecular transfer capacity. Recent technological advancements have enabled the identification of functional functions that exosome cargoes perform in controlling immune responses. Exosomes are originated specifically from immune cells and tumor cells and they show unique composition patterns directly related to the immunotherapy against cancer. Exosomes can also deliver their cargo to particular cells, which can affect the phenotypic and immune-regulatory functions of those cells. Exosomes can influence the course of cancer and have therapeutic benefits by taking part in several cellular processes; as a result, they have the dual properties of activating and restraining cancer. Exosomes have tremendous potential for cancer immunotherapy; they may develop into the most powerful cancer vaccines and carriers of targeted antigens and drugs. Comprehending the potential applications of exosomes in immune therapy is significant for regulating cancer progression. This review offers an analysis of the function of exosomes in immunotherapy, specifically as carriers that function as diagnostic indicators for immunological activation and trigger an anti-cancer immune response. Moreover, it summarizes the fundamental mechanism and possible therapeutic applications of exosome-based immunotherapy for human cancer.
Collapse
Affiliation(s)
- Lili Nie
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, China
| | - Jingru Ma
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun, China
| | - Yang Yu
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun, China
| | - Ying Tao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhidu Song
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, China
| | - Jian Li
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
50
|
Yang EL, Wang WY, Liu YQ, Yi H, Lei A, Sun ZJ. Tumor-Targeted Catalytic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413210. [PMID: 39676382 DOI: 10.1002/adma.202413210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/30/2024] [Indexed: 12/17/2024]
Abstract
Cancer immunotherapy holds significant promise for improving cancer treatment efficacy; however, the low response rate remains a considerable challenge. To overcome this limitation, advanced catalytic materials offer potential in augmenting catalytic immunotherapy by modulating the immunosuppressive tumor microenvironment (TME) through precise biochemical reactions. Achieving optimal targeting precision and therapeutic efficacy necessitates a thorough understanding of the properties and underlying mechanisms of tumor-targeted catalytic materials. This review provides a comprehensive and systematic overview of recent advancements in tumor-targeted catalytic materials and their critical role in enhancing catalytic immunotherapy. It highlights the types of catalytic reactions, the construction strategies of catalytic materials, and their fundamental mechanisms for tumor targeting, including passive, bioactive, stimuli-responsive, and biomimetic targeting approaches. Furthermore, this review outlines various tumor-specific targeting strategies, encompassing tumor tissue, tumor cell, exogenous stimuli-responsive, TME-responsive, and cellular TME targeting strategies. Finally, the discussion addresses the challenges and future perspectives for transitioning catalytic materials into clinical applications, offering insights that pave the way for next-generation cancer therapies and provide substantial benefits to patients in clinical settings.
Collapse
Affiliation(s)
- En-Li Yang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Wu-Yin Wang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Ying-Qi Liu
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Hong Yi
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Aiwen Lei
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Zhi-Jun Sun
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| |
Collapse
|