1
|
Zhao Y, Chen X, Zhang X, Liu H. RNA epigenetic modifications as dynamic biomarkers in cancer: from mechanisms to clinical translation. Biomark Res 2025; 13:81. [PMID: 40483535 PMCID: PMC12145623 DOI: 10.1186/s40364-025-00794-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025] Open
Abstract
RNA modifications are crucial for post-transcriptional gene regulation. Research on RNA modifications has become a novel frontier of epitranscriptomics. Up to now, over 170 kinds of modifications have been identified on mRNA and diverse non-coding RNA. Three classes of proteins (writers, erasers, and readers) regulate the addition, removal, and identification of epigenetic marks, thus affecting RNA biological functions. Increasing evidence identifies the dysregulation of RNA modifications in different cancer types and the therapeutic potential of targeting RNA-modifying enzymes. The ability of RNA modifications to improve mRNA stability and translation efficacy and decrease immunogenicity has been exploited for the clinical use of mRNA cancer vaccines. This review aims to shed light on several vital cap, tail, and internal modifications of RNA with a focus on the connection between RNA epigenetic pathways and cancer pathogenesis. We further explore the clinical potential of RNA modifications as dynamic biomarkers for cancer diagnosis, prognosis, and therapeutic response prediction, addressing both technological challenges and translational opportunities. Finally, we analyze the limitations of current studies and discuss the research focus in the future.
Collapse
Affiliation(s)
- Yingchao Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China
| | - Xingli Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China.
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China.
| |
Collapse
|
2
|
Hua Y, Wang C, Li F, Han Y, Zuo D, Lv Y, Sun M, Yuan P, Yuan R, Zhang F, Ma L, Wang Y, Wu H, Zhou G, Lin Q, Wang S, Li N, Lu Y, North China Petroleum Bureau General Hospital. Phase 1, open-label, multicenter, dose escalation safety and tolerability study of oncolytic virus OVV-01 in advanced solid tumors. J Immunother Cancer 2025; 13:e011517. [PMID: 40480657 PMCID: PMC12142144 DOI: 10.1136/jitc-2025-011517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/25/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND OVV-01 is a genetically engineered vesicular stomatitis virus oncolytic virus designed to selectively amplify in tumor cells and express tumor-associated antigen NY-ESO-1. This study was designed to evaluate the safety, tolerability, and efficacy of OVV-01 in patients with advanced solid tumors. METHODS This is a phase 1, first-in-human, open-label, multicenter study of OVV-01 in patients with advanced solid tumors. OVV-01 was intratumorally injected biweekly (every two weeks, Q2W), 3 weeks after the first dose for a total of six doses. Dose escalation follows a 3+3 design at four doses of 6×107 Plaue-Forming Unit (PFU), 6×108 PFU, 6×109 PFU, and 1.2×1011 PFU. The primary endpoints were safety and tolerability. The second endpoints included overall response rate (ORR) and disease control rate (DCR) of OVV-01, by investigators per Response Evaluation Criteria in Solid Tumors V.1.1. RESULTS 18 patients were enrolled into four dose groups, among whom 6 were soft tissue sarcoma (STS). No dose-limiting toxicities and treatment-related severe adverse events were observed. 11 patients were evaluated for efficacy, and the ORR was 27.3%, and the DCR was 63.6%. Among the four evaluable patients with advanced STS, the ORR was 75%. Two patients with STS achieved CR at doses above 6.0×109 PFU. CONCLUSIONS The intratumor injection of OVV-01 was safe and well-tolerated in patients with advanced solid tumors. A significant response was observed in patients with STS. TRIAL REGISTRATION NUMBER NCT04787003.
Collapse
Affiliation(s)
- Yingqi Hua
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chongren Wang
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Li
- GoBroad Medical (Hematology), Beijing Research Center / Beijing GoBroad Boren Hospital, Beijing, China
| | - Yanjie Han
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Dongqing Zuo
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Lv
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengxiong Sun
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Yuan
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruirong Yuan
- Dowlin Biomed, New Hampshire, New Hampshire, USA
| | - Fan Zhang
- Joint Biosciences (SH) Ltd, Shanghai, Shanghai, China
| | - Liang Ma
- Joint Biosciences (SH) Ltd, Shanghai, Shanghai, China
| | - Yan Wang
- Joint Biosciences (SH) Ltd, Shanghai, Shanghai, China
| | - Hui Wu
- Joint Biosciences (SH) Ltd, Shanghai, Shanghai, China
| | - Guoqing Zhou
- Joint Biosciences (SH) Ltd, Shanghai, Shanghai, China
| | - Qiang Lin
- North China Petroleum Bureau General Hospital, Cangzhou, Hebei, China
| | - Shuhang Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Ning Li
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yinying Lu
- The 5th medical Center of PLA general Hospital, Beijing, China
| | | |
Collapse
Collaborators
Haitao Liu,
Collapse
|
3
|
Wu F, Zhan Y, Wang S, Wang X, Hui M, Zhang J, Zhang J, Yang H, Lei Y, Yu S. VSV-CHIKV activates antitumor immunity by inducing pyroptosis in a melanoma model. Discov Oncol 2025; 16:943. [PMID: 40439822 PMCID: PMC12122967 DOI: 10.1007/s12672-025-02788-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 05/22/2025] [Indexed: 06/02/2025] Open
Abstract
Melanoma is the most dangerous skin cancer due to its difficulty in treatment, high recurrence rate and metastatic ability. As a vector for oncolytic viruses (OVs), vesicular stomatitis virus (VSV) has been shown to be effective against malignant melanoma. However, the glycoprotein G protein of VSV has potential neurotoxicity. It has been shown that replacing glycoprotein G with E3-E2-6K-E1 of chikungunya virus (CHIKV) reduces its neurotoxicity and targets gliomas. Therefore, the aim of this study was to investigate the oncolytic effect of recombinant VSV-CHIKV on melanoma and the underlying mechanism. In this study, we found that recombinant VSV-CHIKV triggered GSDMD-mediated melanoma cell pyroptosis. Importantly, the NLRP3/Caspase-1/GSDMD axis was activated after VSV-CHIKV infection in melanoma cell lines and in a xenograft mouse model. Inhibition of GSDMD blocked cell pyroptosis, antitumor immunity and the tumor response in response to VSV-CHIKV treatment, suggesting that VSV-CHIKV act through the GSDMD pathway. VSV-CHIKV-triggered GSDMD-mediated tumor pyroptosis recruited cytotoxic T lymphocytes (CTLs) into the tumor microenvironment, which was accompanied by the release of inflammatory mediators. This remodeled the tumor microenvironment and turned immunologically "cold" tumors into "hot" tumors, thereby sensitized these tumors to checkpoint blockade. Finally, the combination therapy of VSV-CHIKV and an immune checkpoint inhibitor (anti-PD-1) prolonged the survival of mice. In conclusion, the VSV-CHIKV strategy is an attractive biologic therapy against melanoma.
Collapse
Affiliation(s)
- Fan Wu
- College of Life Sciences, Northwest University, Xi'an, Shannxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ying Zhan
- College of Life Sciences, Northwest University, Xi'an, Shannxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Siyu Wang
- School of Stomatology, Guizhou Medical University, Guiyang, 561113, Guizhou, China
| | - Xiaoke Wang
- College of Life Sciences, Northwest University, Xi'an, Shannxi, China
- Department of Microbiology, School of Preclinical Medicine, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Min Hui
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
- College of Medicine, Northwest University, Xi'an, Shannxi, China
| | - Jian Zhang
- Department of Microbiology, School of Preclinical Medicine, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jing Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hongxu Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yingfeng Lei
- Department of Microbiology, School of Preclinical Medicine, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Shibin Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
4
|
Wang C, Zhang Q, Li Q, Wang Y, Chen X. From infection to tumor: genetic evidence of viral antibody immune response' role in urologic cancer development. Discov Oncol 2025; 16:947. [PMID: 40442531 PMCID: PMC12122962 DOI: 10.1007/s12672-025-02768-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 05/21/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Urologic tumors are among the most common malignancies worldwide, and the association between chronic infections and the risk of developing these tumors has garnered significant attention. However, traditional observational studies are prone to confounding factors, making it challenging to establish a clear causal relationship. METHOD This study employs a two-sample bidirectional Mendelian randomization analysis, utilizing genetic data on antibody levels and urologic tumors obtained from GWAS databases. The inverse variance weighted (IVW) method was used to estimate causal relationships, while MR-Egger and MR-PRESSO methods were applied for sensitivity analyses to assess horizontal pleiotropy and heterogeneity. RESULT The results showed that antibody levels associated with various viral infections were significantly correlated with the risk of developing urologic tumors. For example, antibodies related to cytomegalovirus IgG and Epstein-Barr virus (EBV) were found to have complex associations with the risk of prostate cancer, bladder cancer, and testicular cancer. Some antibodies, such as those related to Varicella zoster virus, were associated with a reduced risk of clear cell renal carcinoma. Additionally, sensitivity analyses suggested the potential presence of horizontal pleiotropy in bladder and testicular cancers. CONCLUSION Through Mendelian randomization analysis, we revealed a potential causal relationship between antibody immune responses and urologic tumors. These findings provide new evidence for the role of chronic infections in the pathogenesis of urologic tumors, suggesting that prevention and treatment strategies targeting related pathogens, such as vaccination and antiviral therapies, could offer new avenues for the prevention and management of urologic cancers.
Collapse
Affiliation(s)
- Chen Wang
- Department of Urology, Nanxiang Branch of Ruijin Hospital, Shanghai, China
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qifa Zhang
- Department of Urology, Nanxiang Branch of Ruijin Hospital, Shanghai, China
| | - Qiang Li
- Department of Urology, Nanxiang Branch of Ruijin Hospital, Shanghai, China
| | - Yelong Wang
- Department of Urology, Nanxiang Branch of Ruijin Hospital, Shanghai, China
| | - Xin Chen
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
5
|
Roulstone V, Kyula-Currie J, Wright J, Patin EC, Dean I, Yu L, Barreiro-Alonso A, Melake M, Choudhary J, Elliott R, Lord CJ, Mansfield D, Matthews N, Chauhan R, Jennings V, Chan Wah Hak C, Baldock H, Butera F, Appleton E, Nenclares P, Pederson M, Foo S, Wongariyapak A, Rullan A, Tenev T, Meier P, Vile R, Pandha H, Melcher A, McLaughlin M, Harrington KJ. Palbociclib and dsRNA sensor co-operate to enhance anti-cancer effects through ER stress and modulation of immune evasion. Nat Commun 2025; 16:4855. [PMID: 40413207 PMCID: PMC12103499 DOI: 10.1038/s41467-025-60133-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/15/2025] [Indexed: 05/27/2025] Open
Abstract
Cytoplasmic pattern recognition receptors (PRR) for double-stranded RNA, such as RIG-I/MDA5, are key mediators of anti-viral responses. Here we screen for synergistic drug-virotherapy combinations and find that the reovirus type III Dearing strain (Rt3D)-palbociclib combination augments oncolytic virus-induced stress responses and increases interferon production and signaling. Data from RIG-I agonist and ER stress-inducing agents further confirms the crosstalk between RNA-sensing and ER stress in inducing cancer cell death and interferon production. Combined Rt3D-palbociclib also increases innate immune activation and IFN-induced HLA expression within tumor cells, with accompanying alterations in the epigenetic landscape and endogenous retroviral (ERV) elements. Analysis of the immunopeptidome in treated cells further reveals changes to HLA-captured peptides, including altered expression of peptides from cancer or testis antigens and ERVs. Our findings thus highlight the crosstalk between stress signaling and PRR activation for mediating enhanced anti-cancer efficacy.
Collapse
Affiliation(s)
| | | | | | | | - Isaac Dean
- The Institute of Cancer Research, London, UK
| | - Lu Yu
- The Institute of Cancer Research, London, UK
| | | | | | | | - Richard Elliott
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Christopher J Lord
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | | | | | | | | | | | | | | | | | | | | | - Shane Foo
- The Institute of Cancer Research, London, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Mullins-Dansereau V, Myre ML, Bardoul A, Geoffroy K, Rallo Pita MJ, Béland D, Desaulniers KL, Roy DG, Bourgeois-Daigneault MC. Oncolytic VSV-IL-2 has enhanced anticancer vaccination adjuvant abilities. J Immunother Cancer 2025; 13:e010570. [PMID: 40389376 PMCID: PMC12090858 DOI: 10.1136/jitc-2024-010570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 05/07/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Heterologous oncolytic virus prime-boost vaccination is emerging as a promising cancer immunotherapy to establish potent antitumor immunity. With their natural ability to specifically destroy cancer cells, oncolytic viruses also allow for direct oncolysis and our team has previously demonstrated that they can be used as vaccination adjuvants when co-administered with antigenic peptides. As such, adjuvant oncolytic virus vaccines can easily be tailored to target any antigen, which is particularly important in the context of personalized vaccines against patient-specific mutations. Here, we tested if oncolytic viruses engineered to express the immune-stimulating transgene interleukin-2 have improved vaccination adjuvant potential. METHODS For this proof-of-concept study, we generated an oncolytic vesicular stomatitis virus (VSV) variant (MD51) that encodes the T-cell activator cytokine interleukin-2 and measured its vaccination adjuvant potential in a heterologous virus immune boosting approach, 1 week after immune priming compared with the parental virus (also MD51). Tumor-free and B16F10-Ova tumor-bearing mice were vaccinated against the Ova peptide using either virus as adjuvants. The immune response induced by each vaccination regimen was assessed by flow cytometry to characterize antigen-specific CD8 T cells over time. Treatment efficacy was also measured. RESULTS Our data show that VSV-interleukin-2 is superior as a vaccine boosting adjuvant compared with the parental virus as it induces more antigen-specific CD8 T cells with enhanced effector functions that persist over time. VSV-interleukin-2 vaccination also improves tumor control and survival. CONCLUSIONS Overall, we show that engineered oncolytic virus platforms, such as VSV-interleukin-2, have the potential to improve vaccination efficacy and treatment outcomes. Our findings deepen our understanding of the immune mechanisms underlying the use of oncolytic viruses as anticancer vaccination platforms and will allow for the development of a new generation of improved oncolytic virus vaccine adjuvants. TRIAL REGISTRATION NUMBER NCT02285816.
Collapse
Affiliation(s)
- Victor Mullins-Dansereau
- Centre Hospitalier de l'Université de Montréal research center (CRCHUM), Montreal, Quebec, Canada
- Département de Microbiologie, Université de Montréal, Montreal, Quebec, Canada
- Institut du Cancer de Montréal, Montreal, Quebec, Canada
| | - Marie-Lou Myre
- Centre Hospitalier de l'Université de Montréal research center (CRCHUM), Montreal, Quebec, Canada
- Département de Microbiologie, Université de Montréal, Montreal, Quebec, Canada
- Institut du Cancer de Montréal, Montreal, Quebec, Canada
| | - Angelina Bardoul
- Centre Hospitalier de l'Université de Montréal research center (CRCHUM), Montreal, Quebec, Canada
- Département de Microbiologie, Université de Montréal, Montreal, Quebec, Canada
- Institut du Cancer de Montréal, Montreal, Quebec, Canada
| | - Karen Geoffroy
- Centre Hospitalier de l'Université de Montréal research center (CRCHUM), Montreal, Quebec, Canada
- Département de Microbiologie, Université de Montréal, Montreal, Quebec, Canada
- Institut du Cancer de Montréal, Montreal, Quebec, Canada
| | - Marco J Rallo Pita
- Centre Hospitalier de l'Université de Montréal research center (CRCHUM), Montreal, Quebec, Canada
- Département de Microbiologie, Université de Montréal, Montreal, Quebec, Canada
- Institut du Cancer de Montréal, Montreal, Quebec, Canada
| | - Delphine Béland
- Centre Hospitalier de l'Université de Montréal research center (CRCHUM), Montreal, Quebec, Canada
- Département de Microbiologie, Université de Montréal, Montreal, Quebec, Canada
- Institut du Cancer de Montréal, Montreal, Quebec, Canada
| | - Kim Leclerc Desaulniers
- Centre Hospitalier de l'Université de Montréal research center (CRCHUM), Montreal, Quebec, Canada
- Institut du Cancer de Montréal, Montreal, Quebec, Canada
| | - Dominic Guy Roy
- Centre Hospitalier de l'Université de Montréal research center (CRCHUM), Montreal, Quebec, Canada
- Département de Microbiologie, Université de Montréal, Montreal, Quebec, Canada
- Institut du Cancer de Montréal, Montreal, Quebec, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Centre Hospitalier de l'Université de Montréal research center (CRCHUM), Montreal, Quebec, Canada
- Département de Microbiologie, Université de Montréal, Montreal, Quebec, Canada
- Institut du Cancer de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Liu L, Song L, Liu T, Hui K, Hu C, Yang J, Pi X, Yan Y, Liu S, Zhang Y, Chen H, Cao Y, Zhou L, Qiao Y, Yu D, Yin C, Li X, Zhang C, Li D, Wang Z, Liu Z, Jiang X. Recombinant oncolytic virus NDV-anti-VEGFR2 enhances radiotherapy sensitivity in NSCLC by targeting VEGF signaling and impairing DNA repair. Gene Ther 2025:10.1038/s41434-025-00540-x. [PMID: 40382521 DOI: 10.1038/s41434-025-00540-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 04/09/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
Resistance to radiotherapy is a significant challenge in the clinical management of non-small cell lung cancer (NSCLC). This study investigates a novel multimodal therapeutic strategy that combines oncolytic Newcastle disease virus (NDV) with an anti-VEGFR2 single-chain variable fragment (NDV-anti-VEGFR2) to enhance radiosensitivity in NSCLC. We engineered NDV-anti-VEGFR2 and assessed its efficacy in sensitizing Calu-1 cells to radiation. In vitro results demonstrated that NDV-anti-VEGFR2 significantly inhibited tumor cell proliferation when combined with radiotherapy. In vivo experiments revealed that NDV-anti-VEGFR2, combined with radiation, achieved a tumor growth inhibition rate of 86.48%, surpassing the effects of NDV or radiation alone. Mechanistic investigations indicated that NDV-anti-VEGFR2 mitigated hypoxia by downregulating HIF-1α and impaired DNA repair pathways, as evidenced by reduced levels of RAD51 and γ-H2AX. These findings suggest that NDV-anti-VEGFR2 not only normalizes tumor vasculature but also enhances the cytotoxic effects of radiation by compromising DNA repair mechanisms. Collectively, our results support the clinical potential of NDV-anti-VEGFR2 combined with radiotherapy as a promising strategy to overcome radiotherapy resistance in NSCLC. Future studies in immunocompetent models are warranted to elucidate the immune-mediated effects of this innovative therapeutic approach.
Collapse
Affiliation(s)
- Liang Liu
- Lianyungang Clinical College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Liying Song
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Tianyan Liu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Kaiyuan Hui
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Chenxi Hu
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Jiarui Yang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Xuelei Pi
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Yuanyuan Yan
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Shishi Liu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Yating Zhang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Hongna Chen
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Yukai Cao
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Lihua Zhou
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Yun Qiao
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Dan Yu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Chengkai Yin
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Xu Li
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Chenfeng Zhang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Deshan Li
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Zhenzhong Wang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Zhihang Liu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China.
| | - Xiaodong Jiang
- Lianyungang Clinical College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China.
| |
Collapse
|
8
|
Liu DM, Li MH, Zhang J. Letter to the Editor: "Targeted Inhibition of p21 Promotes the Growth of Breast Cancer Cells and Impairs the Tumor-Killing Effect of the Vaccinia Virus". J Breast Cancer 2025; 28:28.e16. [PMID: 40432354 DOI: 10.4048/jbc.2025.0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 04/20/2025] [Indexed: 05/29/2025] Open
Affiliation(s)
- Dong-Mei Liu
- Department of Oncology, Yulin The First Hospital, Second Affiliated Hospital of Yanan University, Yulin, China
| | - Meng-Hui Li
- Department of Cardiology, Yulin The First Hospital, Second Affiliated Hospital of Yanan University, Yulin, China
| | - Jian Zhang
- Department of Cardiology, Yulin The First Hospital, Second Affiliated Hospital of Yanan University, Yulin, China.
| |
Collapse
|
9
|
Gao Y, Zou Y, Wu C, Tao J, Nie Z, Yan J, Wang P, Huang X. Comparative evaluation of immunomodulatory cytokines for oncolytic therapy based on a high-efficient platform for oHSV1 reconstruction. Virol J 2025; 22:133. [PMID: 40325455 PMCID: PMC12054163 DOI: 10.1186/s12985-025-02758-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: 03/20/2024] [Accepted: 04/22/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) presents significant therapeutic challenges due to its immunosuppressive tumor microenvironment (TME). Oncolytic herpes simplex virus type 1 (oHSV1) offers dual mechanisms of tumor lysis and immune activation, yet the optimal cytokine payloads for TNBC remain undefined. METHODS We developed a CRISPR/Cas9-mediated platform for high-efficiency oHSV1 engineering, replacing the ICP47 locus with murine IFN-γ, GM-CSF, or IL-15Rα/IL-15 fusion protein (IL15Fu). Constructs were validated for cytokine secretion, MHC modulation, and cytotoxicity in 4T1 TNBC and a panel of human cancer cell lines. Antitumor efficacy and immune remodeling were evaluated in a syngeneic 4T1 model using RNA sequencing and flow cytometry. RESULTS The CRISPR platform achieved 62.5-71.4% homologous recombination efficiency, enabling rapid virus construction. In vitro, OV-IFNG exhibited upregulated MHC I/II expression and potent cytotoxicity, while OV-GMCSF attenuated oncolysis in subsets of breast cancer cell lines. In the 4T1 model, OV-IL15Fu modestly improved tumor control and extended survival without apparent toxicity, while OV-IFNG induced early mortality associated with systemic toxicity. Transcriptomic profiling revealed divergent immune modulation: OV-IL15Fu enriched T cell/NK cytotoxicity pathways, OV-IFNG amplified cytokine/chemokine signaling, and OV-GMCSF paradoxically enhanced myeloid recruitment while inhibiting MHC-II pathways. Flow cytometry confirmed functional differences in immune activation: OV-IL15Fu expanding cytotoxic lymphocytes (CD8⁺ T/NK cells), OV-IFNG preferentially promote Th1 polarization and innate immune activation, and OV-GMCSF failed to activate T cells despite myeloid infiltration. CONCLUSIONS Our findings underscore the need for rational cytokine selection in oHSV1-based immunotherapy. While IFN-γ increased immunogenic markers, its systemic toxicity and myeloid effects may limit benefit. GM-CSF exacerbated immune suppression in this context, whereas IL15Fu showed favorable immunostimulatory properties without detectable toxicity. These data support IL15Fu as a contextually promising payload for further evaluation in TNBC-targeted oncolytic virotherapy.
Collapse
Affiliation(s)
- Yingzheng Gao
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yufang Zou
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Changjing Wu
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Juan Tao
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zuqing Nie
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jinyuan Yan
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Pengfei Wang
- Kunming Medical University, Kunming, Yunnan, China.
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| | - Xinwei Huang
- Kunming Medical University, Kunming, Yunnan, China.
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
10
|
Sheikh M, Saiyyad A, Aliunui A, Jirvankar PS. The evolving landscape of oncolytic virus immunotherapy: combinatorial strategies and novel engineering approaches. Med Oncol 2025; 42:190. [PMID: 40314865 DOI: 10.1007/s12032-025-02746-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025]
Abstract
Oncolytic viruses (OVs) are a promising class of cancer therapy, exploiting their abilities to selectively infect and kill cancer cells while stimulating antitumor immune responses. The current assessment explores the changing horizons of OV immunotherapy, focusing on recent advances in technology plans to improve OV projects and combined approaches to improve curative efficacy. We discuss how OVs induce direct oncolysis and promote the release of tumor-associated antigens, leading to the activation of both innate and adaptive immunity. Special attention shall be given to programs for arm OVs to express curative genes, modify the tumor microenvironment and overcome immunosuppression. Moreover, we assess the synergies of uniting OVs with other immunotherapeutic techniques, such as immune checkpoint inhibitors and cell therapy, to improve tolerant outcomes. The present assessment provides an understanding of the relevant declaration of the OV analysis, highlighting the main obstacles and the future directions for the development of other capable and targeted cancer immunotherapy.
Collapse
Affiliation(s)
- Mujibullah Sheikh
- Datta Meghe College of Pharmacy DMIHER (Deemed to be University), Wardha, Maharashtra, 442001, India.
| | - Arshiya Saiyyad
- Datta Meghe College of Pharmacy DMIHER (Deemed to be University), Wardha, Maharashtra, 442001, India
| | - Aimé Aliunui
- Datta Meghe College of Pharmacy DMIHER (Deemed to be University), Wardha, Maharashtra, 442001, India
| | - Pranita S Jirvankar
- Datta Meghe College of Pharmacy DMIHER (Deemed to be University), Wardha, Maharashtra, 442001, India
| |
Collapse
|
11
|
Qiu M, Wei R, Zhang Q, Zhao J, Zhang H, Tan J, Qiao W. ISG15 depletion enhances oHSV-1 replication and antitumor efficacy in oral squamous cell carcinoma. Virology 2025; 606:110504. [PMID: 40121989 DOI: 10.1016/j.virol.2025.110504] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Oncolytic viruses (OVs) represent a promising experimental therapy for a range of cancers, including oral squamous cell carcinoma (OSCC). In this study, oHSV refers to an oncolytic virus engineered from HSV-1(Herpes Simplex Virus Type 1). The oHSV-1 is an oncolytic virus derived from HSV-1, where both copies of the ICP34.5 coding sequences have been replaced with the EGFP gene, and the ICP47 gene has been deleted. In previous studies, resistance was observed in certain SCC15 xenograft models treated with oncolytic herpes simplex viruses (oHSV-1). Primary tumor cells were extracted from these resistant models, followed by RNA sequencing with SCC15 cells as controls. Analysis revealed that ISG15 expression was upregulated in the resistant primary cells, as well as in HSV-infected breast cancer cells (GSE137757). In this study, we confirmed that knockdown ISG15 in SCC15 cells enhanced oHSV-1 replication, while ISG15 overexpression suppressed it. Mechanistic studies demonstrated that ISG15 inhibits oHSV-1 replication via ISGylation. To improve the therapeutic efficacy of oHSV-1, an oHSV-1 variant expressing ISG15-targeting short hairpin RNA (shRNA), termed oHSV-1-shISG15, was engineered. oHSV-1-shISG15 exhibited enhanced antitumor efficacy compared to oHSV-1 in vitro and in vivo. These findings suggest that ISG15 depletion augments oHSV-1 replication in OSCC tumor cells through ISGylation inhibition. Meanwhile, this study provides a novel recombinant oncolytic virus to potentiate the efficacy of oncolytic herpes virotherapy.
Collapse
Affiliation(s)
- Manman Qiu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Rongrong Wei
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Qicheng Zhang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Jiawei Zhao
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Hongkai Zhang
- Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Juan Tan
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Wentao Qiao
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| |
Collapse
|
12
|
Schoeps B, Lauer UM, Elbers K. Deciphering permissivity of human tumor ecosystems to oncolytic viruses. Oncogene 2025; 44:1069-1077. [PMID: 40148688 PMCID: PMC11996678 DOI: 10.1038/s41388-025-03357-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/10/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Effective cancer therapy involves initiation of a tumor-specific immune response. Consequently, the interest in oncolytic viruses (OV) capable of triggering immunogenic cell death has sparked in recent years. However, the common use of pre-clinical models that fail to mirror patient tumor ecosystems (TES) hinders clinical translation. Here, we provide a condensed view on the intricate interplay between several aspects of TES and OV action and discuss these considerations in the view of recently developed pre-clinical human model systems. Given the urgent demand for innovative cancer treatments, the purpose of this review is to highlight the so-far overlooked complex impact of the tumor microenvironment (TME) on OV permissivity, with the intent to provide a foundation for future, more effective pre-clinical studies.
Collapse
Affiliation(s)
| | - Ulrich M Lauer
- Department of Medical Oncology and Pneumology, Virotherapy Center Tübingen (VCT), Medical University Hospital, Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Tübingen, Germany
| | | |
Collapse
|
13
|
Admasu TD, Yu JS. Harnessing Immune Rejuvenation: Advances in Overcoming T Cell Senescence and Exhaustion in Cancer Immunotherapy. Aging Cell 2025; 24:e70055. [PMID: 40178455 PMCID: PMC12073907 DOI: 10.1111/acel.70055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/15/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
Immunotherapy has transformed the landscape of cancer treatment, with T cell-based strategies at the forefront of this revolution. However, the durability of these responses is frequently undermined by two intertwined phenomena: T cell exhaustion and senescence. While exhaustion is driven by chronic antigen exposure in the immunosuppressive tumor microenvironment, leading to a reversible state of diminished functionality, senescence reflects a more permanent, age- or stress-induced arrest in cellular proliferation and effector capacity. Together, these processes represent formidable barriers to sustained anti-tumor immunity. In this review, we dissect the molecular underpinnings of T cell exhaustion and senescence, revealing how these dysfunctions synergistically contribute to immune evasion and resistance across a range of solid tumors. We explore cutting-edge therapeutic approaches aimed at rewiring the exhausted and senescent T cell phenotypes. These include advances in immune checkpoint blockade, the engineering of "armored" CAR-T cells, senolytic therapies that selectively eliminate senescent cells, and novel interventions that reinvigorate the immune system's capacity for tumor eradication. By spotlighting emerging strategies that target both exhaustion and senescence, we provide a forward-looking perspective on the potential to harness immune rejuvenation. This comprehensive review outlines the next frontier in cancer immunotherapy: unlocking durable responses by overcoming the immune system's intrinsic aging and exhaustion, ultimately paving the way for transformative therapeutic breakthroughs.
Collapse
Affiliation(s)
| | - John S. Yu
- Department of NeurosurgeryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Kairos PharmaLos AngelesCaliforniaUSA
| |
Collapse
|
14
|
Naffaa MM, Al-Ewaidat OA, Gogia S, Begiashvili V. Neoantigen-based immunotherapy: advancing precision medicine in cancer and glioblastoma treatment through discovery and innovation. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002313. [PMID: 40309350 PMCID: PMC12040680 DOI: 10.37349/etat.2025.1002313] [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: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Neoantigen-based immunotherapy has emerged as a transformative approach in cancer treatment, offering precision medicine strategies that target tumor-specific antigens derived from genetic, transcriptomic, and proteomic alterations unique to cancer cells. These neoantigens serve as highly specific targets for personalized therapies, promising more effective and tailored treatments. The aim of this article is to explore the advances in neoantigen-based therapies, highlighting successful treatments such as vaccines, tumor-infiltrating lymphocyte (TIL) therapy, T-cell receptor-engineered T cells therapy (TCR-T), and chimeric antigen receptor T cells therapy (CAR-T), particularly in cancer types like glioblastoma (GBM). Advances in technologies such as next-generation sequencing, RNA-based platforms, and CRISPR gene editing have accelerated the identification and validation of neoantigens, moving them closer to clinical application. Despite promising results, challenges such as tumor heterogeneity, immune evasion, and resistance mechanisms persist. The integration of AI-driven tools and multi-omic data has refined neoantigen discovery, while combination therapies are being developed to address issues like immune suppression and scalability. Additionally, the article discusses the ongoing development of personalized immunotherapies targeting tumor mutations, emphasizing the need for continued collaboration between computational and experimental approaches. Ultimately, the integration of cutting-edge technologies in neoantigen research holds the potential to revolutionize cancer care, offering hope for more effective and targeted treatments.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ola A Al-Ewaidat
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Sopiko Gogia
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Valiko Begiashvili
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
15
|
Wang X, Zhou Q, Zhang X, Hu H, Liu B, Wang Y. Oncolytic viruses: a promising therapy for malignant pleural effusion and solid tumors. Front Immunol 2025; 16:1570698. [PMID: 40352942 PMCID: PMC12061930 DOI: 10.3389/fimmu.2025.1570698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
Oncolytic viruses (OVs) are natural or recombinant viruses that can directly lyse tumor cells without damaging normal cells. They enhance anti-tumor immunity by releasing antigens and activating inflammatory responses within the tumor microenvironment (TME). This offers a new therapeutic approach for MPE and solid tumors. This review discusses the progress of OVs administered via intrapleural and intratumoral routes, emphasizing their potential in MPE treatment and the challenges posed by the complex intrapleural environment, which affects the direct interaction between OVs, tumor cells, and immune cells. This review also discusses the regulatory barriers, safety concerns and accessibility of oncolytic virus therapy.
Collapse
Affiliation(s)
- Xinya Wang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan, China
| | - Qin Zhou
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan, China
| | - Xuyan Zhang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan, China
| | - Han Hu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan, China
| | - Binlei Liu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan, China
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Yang Wang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan, China
| |
Collapse
|
16
|
Baumrucker CC, Harris N, Hoover S, Czerniecki BJ. Intratumoral Immunotherapy in Breast Cancer. Vaccines (Basel) 2025; 13:429. [PMID: 40333343 PMCID: PMC12031351 DOI: 10.3390/vaccines13040429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 05/09/2025] Open
Abstract
Breast cancer remains the most frequently diagnosed cancer and the second highest cause of cancer death in females. Metastatic recurrence that is resistant to traditional therapies presents a major challenge, necessitating the development of an innovative treatment strategy. Immunotherapy has gained popularity in the treatment of cancer, particularly melanoma, lung cancer, and more recently breast cancer. Major developments in immunotherapy have been made with a better understanding of the tumor microenvironment and how the microenvironment can be manipulated to induce an anti-tumor immune response. Intratumorally delivered immunotherapy can be used to create a local immune response. This review provides a comprehensive overview of intratumoral immunotherapy for breast cancer and its resultant changes in the tumor microenvironment. The discussed immunotherapeutics include oncolytic viruses, nucleic acids, innate immune agonists, bacteria, chimeric antigen receptor T cells, and dendritic cells. The review also evaluates completed clinical trials using these therapies. Lastly, the review offers future perspectives in the development of breast cancer immunotherapy.
Collapse
Affiliation(s)
- Camille C. Baumrucker
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA;
| | - Nicole Harris
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Susan Hoover
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Brian J. Czerniecki
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
17
|
Ghorab BEA, Liu T, Ying M, Wang P, Qin M, Xing J, Wang H, Xu F. Advances in the Drug Development and Quality Evaluation Principles of Oncolytic Herpes Simplex Virus. Viruses 2025; 17:581. [PMID: 40285023 PMCID: PMC12031214 DOI: 10.3390/v17040581] [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: 03/05/2025] [Revised: 04/07/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Oncolytic herpes simplex virus (oHSV) represents a promising therapeutic approach to treating cancers by virtue of its selective replication in and lysis of tumor cells, with stimulation of host antitumor immunity. At present, four OV drugs have been approved for the treatment of cancers worldwide, two of which are oHSV drugs that have received extensive attention, known as T-VEC and Delytact. This review discusses the history, mechanism of action, clinical development, quality control, and evaluation principles of oHSV products, including viral species and genetic modifications that have improved these products' therapeutic potential, limitations, and future directions. Integration of oHSVs with immunotherapeutic agents and conventional therapies has a promising future in the field of treatment of malignant tumors. Although much progress has been achieved, there is still much work to be done regarding the optimization of treatment protocols and the quality control of oncolytic virus drugs. The approval of various oncolytic virus therapies underlines their clinical relevance, safety, and efficacy, thereby paving the way for further research aimed at overcoming the existing limitations and enhancing patient responses.
Collapse
Affiliation(s)
- Basma Eid Abdullah Ghorab
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (B.E.A.G.); (T.L.); (J.X.)
- NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
- Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Guangdong Provincial Key Laboratory of Viral Biotechnology and Application, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tongtan Liu
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (B.E.A.G.); (T.L.); (J.X.)
- NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
- Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Guangdong Provincial Key Laboratory of Viral Biotechnology and Application, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Min Ying
- NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
- Department of Anesthesiology, Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, China
| | - Ping Wang
- Shenzhen Institute for Drug Control, Shenzhen 518057, China; (P.W.); (M.Q.)
| | - Meirong Qin
- Shenzhen Institute for Drug Control, Shenzhen 518057, China; (P.W.); (M.Q.)
| | - Jiayong Xing
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (B.E.A.G.); (T.L.); (J.X.)
- NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
- Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Guangdong Provincial Key Laboratory of Viral Biotechnology and Application, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huadong Wang
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (B.E.A.G.); (T.L.); (J.X.)
- NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
- Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Guangdong Provincial Key Laboratory of Viral Biotechnology and Application, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fuqiang Xu
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (B.E.A.G.); (T.L.); (J.X.)
- NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
- Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Guangdong Provincial Key Laboratory of Viral Biotechnology and Application, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
18
|
Tang S, Yong L, Cui Y, Li H, Bischof E, Cai F. Harnessing Oncolytic Viruses for Targeted Therapy in Triple-Negative Breast Cancer. Int J Med Sci 2025; 22:2186-2207. [PMID: 40303488 PMCID: PMC12035831 DOI: 10.7150/ijms.105683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/19/2025] [Indexed: 05/02/2025] Open
Abstract
Breast cancer is the most prevalent malignant tumor among women, with triple-negative breast cancer (TNBC) being one of the most aggressive forms due to its high invasiveness and metastatic potential. Traditional treatments such as endocrine therapy and anti-HER2-targeted therapy are largely ineffective for TNBC, and while chemotherapy shows some promise, resistance remains a significant hurdle. Recently, there has been increasing interest in biological therapies, especially oncolytic viruses (OVs). OVs promote anti-tumor effects by selectively killing tumor cells and stimulating immune responses, and have achieved notable breakthroughs in breast cancer treatment. OVs have demonstrated effectiveness comparable to surgery, radiotherapy, or chemotherapy in selected cancers, but data are sparse in the context of TNBC. This review provides an overview of recent progress in the application of OVs as a tool for precision TNBC treatment.
Collapse
Affiliation(s)
- Shasha Tang
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Rd, Shanghai 200065, China
| | - Liyun Yong
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Rd, Shanghai 200065, China
| | - Yong Cui
- Department of General Surgery, People's Hospital of Otog Qianqi, Sharita Tara East Street, Aolezhaoqi Town, Otog Qianqi 016200, China
| | - Haibin Li
- Department of General Surgery, People's Hospital of Otog Qianqi, Sharita Tara East Street, Aolezhaoqi Town, Otog Qianqi 016200, China
| | - Evelyne Bischof
- Department of Medical Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Fengfeng Cai
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Rd, Shanghai 200065, China
| |
Collapse
|
19
|
Yan W, Xuan Y, Wang R, Huan Z, Guo Y, Dun H, Xu L, Han R, Sun X, Si L, Lemoine NR, Wang Y, Wang P. Oncolytic Vaccinia Virus Armed with GM-CSF and IL-7 Enhances Antitumor Immunity in Pancreatic Cancer. Biomedicines 2025; 13:882. [PMID: 40299475 PMCID: PMC12024586 DOI: 10.3390/biomedicines13040882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/22/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Objectives: Pancreatic cancer remains a therapeutic challenge due to its immunosuppressive microenvironment and treatment resistance. This study aimed to develop a novel recombinant oncolytic vaccinia virus (VVL-GL7) co-expressing granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-7 (IL-7), designed to enhance anti-tumor immunity and synergize with immune checkpoint inhibitors. Methods: VVL-GL7 was constructed through CRISPR/Cas9-mediated knockout of TK and A49 genes, combined with the simultaneous insertion of dual cytokine-encoding cassettes. Anti-tumor efficacy was evaluated in vitro and in vivo using C57BL/6 mouse and Syrian hamster pancreatic cancer models. Comprehensive immune profiling evaluated CD8+ T-cell and macrophage infiltration dynamics while simultaneously assessing memory T-cell differentiation patterns using flow cytometry. Preclinical combination studies of VVL-GL7 and the PD-1 immune checkpoint inhibitor were systematically evaluated in a syngeneic pancreatic cancer model. Results: VVL-GL7 exhibited potent oncolytic activity, inducing significant tumor regression in both preclinical models. VVL-GL7 therapy significantly augmented CD8+ T-cell and macrophage infiltration within the tumor microenvironment, while concomitantly driving memory T-cell differentiation. The synergistic effects of VVL-GL7 and the PD-1 blockade further improved therapeutic outcomes, resulting in significantly higher tumor remission rates compared to monotherapy and achieving complete tumor regression in pancreatic cancer models. Conclusions: VVL-GL7 reprograms the immunosuppressive tumor microenvironment and synergizes with anti-PD-1 antibodies to overcome resistance in pancreatic cancer.
Collapse
Affiliation(s)
- Wenyi Yan
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.Y.)
| | - Yujing Xuan
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.Y.)
| | - Ruimin Wang
- Department of Pathology, Zhengzhou People’s Hospital, Fifth Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou 450003, China
| | - Ziyan Huan
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.Y.)
| | - Yu Guo
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.Y.)
| | - Huilin Dun
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.Y.)
| | - Lihua Xu
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.Y.)
| | - Ruxia Han
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.Y.)
| | - Xianlei Sun
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.Y.)
| | - Lingling Si
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.Y.)
| | - Nicholas Robert Lemoine
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.Y.)
- Centre for Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Yaohe Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.Y.)
- Centre for Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Pengju Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.Y.)
| |
Collapse
|
20
|
Sun L, Zhao Q, Miao L. Combination therapy with oncolytic viruses for lung cancer treatment. Front Oncol 2025; 15:1524079. [PMID: 40248194 PMCID: PMC12003109 DOI: 10.3389/fonc.2025.1524079] [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: 11/07/2024] [Accepted: 03/18/2025] [Indexed: 04/19/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related death globally. Despite various treatment options, adverse reactions and treatment resistance limit their clinical application and efficacy, therefore, new effective treatment options are still needed. Oncolytic viruses (OVs) are a new anti-cancer option. With a powerful anti-tumor effect, OVs are gradually being applied to the treatment of solid tumor. In clinical practice, we have found that in patients with NSCLC and SCLC, OVs combined with immune checkpoint inhibitors (ICI) treatment make tumor with poor response to immunotherapy become sensitive. Furthermore, studies have shown that OVs combined with chemotherapy, radiation therapy, and other immune approaches (such as anti-pd1 drugs) have synergistic effects. These studies suggest that OVs combined therapy may bring hope for the treatment of lung cancer patients. This article will review the current status and prospect of OVs combination therapy in the field of lung cancer treatment and summarizes the mechanism of action.
Collapse
Affiliation(s)
- Lei Sun
- Department of Pharmacy, Yancheng Branch of Nanjing Drum Tower Hospital, Yancheng, Jiangsu, China
| | - Qi Zhao
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Liyun Miao
- Department of Pharmacy, Yancheng Branch of Nanjing Drum Tower Hospital, Yancheng, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
21
|
Ayele K, Wakimoto H, Nauwynck HJ, Kaufman HL, Rabkin SD, Saha D. Understanding the interplay between oHSV and the host immune system: Implications for therapeutic oncolytic virus development. Mol Ther 2025; 33:1327-1343. [PMID: 39741405 PMCID: PMC11997513 DOI: 10.1016/j.ymthe.2024.12.054] [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: 09/26/2024] [Revised: 12/05/2024] [Accepted: 12/27/2024] [Indexed: 01/03/2025] Open
Abstract
Oncolytic herpes simplex viruses (oHSV) preferentially replicate in cancer cells while inducing antitumor immunity, and thus, they are often referred to as in situ cancer vaccines. OHSV infection of tumors elicits diverse host immune responses comprising both innate and adaptive components. Although the innate and adaptive immune responses primarily target the tumor, they also contribute to antiviral immunity, limiting viral replication/oncolysis. OHSV-encoded proteins use various mechanisms to evade host antiviral pathways and immune recognition, favoring oHSV replication, oncolysis, and spread. In general, oHSV infection and replication within tumors results in a series of sequential events, such as oncolysis and release of tumor and viral antigens, dendritic cell-mediated antigen presentation, T cell priming and activation, T cell trafficking and infiltration to tumors, and T cell recognition of cancer cells, leading to tumor (and viral) clearance. These sequential events align with all steps of the cancer-immunity cycle. However, a comprehensive understanding of the interplay between oHSV and host immune responses is crucial to optimize oHSV-induced antitumor immunity and efficacy. Therefore, this review aims to elucidate oHSV's communication with innate and adaptive immune systems and use such interactions to improve oHSV's potential as a potent immunovirotherapeutic agent against cancer.
Collapse
Affiliation(s)
- Kalkidan Ayele
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, CA 95757, USA
| | - Hiroaki Wakimoto
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Hans J Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Howard L Kaufman
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Samuel D Rabkin
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Dipongkor Saha
- Department of Biology, College of Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA.
| |
Collapse
|
22
|
Sharma R, Sil D, Kumar D, Komal K, Kumar S, Ghosh R, Saini V, Kumar M. Oncolytic virotherapy - a promising approach in cancer treatment. Expert Rev Anticancer Ther 2025; 25:315-318. [PMID: 40022467 DOI: 10.1080/14737140.2025.2474732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/27/2025] [Accepted: 02/27/2025] [Indexed: 03/03/2025]
Affiliation(s)
- Rohit Sharma
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Debayan Sil
- Department of Quality Assurance, ISF College Pharmacy, Moga, India
| | - Dinesh Kumar
- Department of Quality Assurance, ISF College Pharmacy, Moga, India
| | - Kumari Komal
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Rashmi Ghosh
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Vipin Saini
- Department of Pharmaceutics, M M College of Pharmacy, MMDU, Mullana, Ambala, Haryana, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| |
Collapse
|
23
|
Wang X, He J, Sun M, Wang S, Qu J, Shi H, Rao B. High-dose vitamin C as a metabolic treatment of cancer: a new dimension in the era of adjuvant and intensive therapy. Clin Transl Oncol 2025; 27:1366-1382. [PMID: 39259387 DOI: 10.1007/s12094-024-03553-x] [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: 03/22/2024] [Accepted: 06/04/2024] [Indexed: 09/13/2024]
Abstract
The anti-cancer mechanism of High-dose Vitamin C (HDVC) is mainly to participate in the Fenton reaction, hydroxylation reaction, and epigenetic modification, which leads to the energy crisis, metabolic collapse, and severe peroxidation stress that results in the proliferation inhibition or death of cancer cells. However, the mainstream view is that HDVC does not significantly improve cancer treatment outcomes. In clinical work and scientific research, we found that some drugs or therapies can significantly improve the anti-cancer effects of HDVC, such as PD-1 inhibitors that can increase the anti-cancer effects of cancerous HDVC by nearly three times. Here, the adjuvant and intensive therapy and synergistic mechanisms including HDVC combined application of chemoradiotherapies multi-vitamins, targeted drugs, immunotherapies, and oncolytic virus are discussed in detail. Adjuvant and intensive therapy of HDVC can significantly improve the therapeutic effect of HDVC in the metabolic treatment of cancer, but more clinical evidence is needed to support its clinical application.
Collapse
Affiliation(s)
- Xin Wang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Jia He
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Minmin Sun
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Shiwan Wang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Jinxiu Qu
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Hanping Shi
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China.
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China.
- Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
| | - Benqiang Rao
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China.
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China.
| |
Collapse
|
24
|
Sigler GI, Murtha J, Varley PR. Diagnostic Advances and Novel Therapeutics in Peritoneal Metastasis. Surg Oncol Clin N Am 2025; 34:173-194. [PMID: 40015798 DOI: 10.1016/j.soc.2024.12.005] [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: 03/01/2025]
Abstract
Appropriate assessment of disease burden in patients with peritoneal surface malignancy (PSM) is critical for treatment decision-making, and conventional cross-sectional imaging (computed tomography and/or MRI) often underestimates burden of disease. Advances in imaging for PSM include novel functional imaging modalities that target cells unique to the tumor microenvironment. Novel alternative methods of diagnosis and disease monitoring are also potentially applicable to management of PSM. These include forms of "liquid biopsy" targeting circulating tumor DNA. Novel regional therapies include both new therapeutic agents (immune-based and nanoparticle-based), as well as new methods of delivery such as pressurized intraperitoneal aerosolized chemotherapy.
Collapse
Affiliation(s)
- Gregory I Sigler
- Division of Surgical Oncology, Department of General Surgery, Complex General Surgical Oncology, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Mail Code 7375, Madison, WI 53792, USA
| | - Jacqueline Murtha
- Department of General Surgery, General Surgery, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Mail Code 7375, Madison, WI 53792, USA
| | - Patrick R Varley
- Division of Surgical Oncology, Department of General Surgery, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Mail Code 7375, Madison, WI 53792, USA; William S. Middleton Memorial Veterans Affairs Hospital, Madison, WI, USA.
| |
Collapse
|
25
|
Takahashi M, Mukhamejanova D, Jasewicz H, Acharya N, Moon JJ, Hara T. Opportunities to Modulate Tumor Ecosystem Toward Successful Glioblastoma Immunotherapy. Cancer Sci 2025. [PMID: 40123277 DOI: 10.1111/cas.70052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025] Open
Abstract
Over the past decade, the failure of multiple clinical trials has confirmed the need for a systematic and comprehensive understanding of glioblastoma (GBM). Current immunotherapies aiming to harness the immune system to achieve anti-tumor effects remain largely ineffective, highlighting the complexities of the GBM microenvironment. However, our recent understanding of immune niches within the central nervous system provides both opportunities and challenges in translating these insights into successful immunotherapy implementation. We discuss these strategies, including targeting multiple antigens within the heterogeneous GBM microenvironment, identifying new druggable targets to abrogate immunosuppression, and understanding niche-specific immune cell functionality to modulate tumor-immune-stroma interactions.
Collapse
Affiliation(s)
- Mariko Takahashi
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Darina Mukhamejanova
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Department of Neurosurgery, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biology, Nazarbayev University, Astana, Kazakhstan
| | - Himani Jasewicz
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Nandini Acharya
- Pelotonia Institute for Immuno-Oncology, OSUCCC-James, The Ohio State University, Columbus, Ohio, USA
- Department of Neurology, the Neuroscience Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Toshiro Hara
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Department of Neurosurgery, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
26
|
Huberts M, de Graaf JF, Groeneveld D, van Nieuwkoop S, Fouchier RA, van den Hoogen BG. Cell-derived Newcastle disease virus variant with two amino acid substitutions near cleavage site of F shows favorable traits as oncolytic virus. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200915. [PMID: 39802675 PMCID: PMC11719830 DOI: 10.1016/j.omton.2024.200915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/21/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025]
Abstract
Newcastle disease virus (NDV) has shown encouraging effectiveness in in vitro, in vivo, and in early clinical trials as a viro-immunotherapy for pancreatic cancer. Previously, NDV used in clinical trials was produced in embryonated chicken eggs; however, egg-produced viruses are known to be partly neutralized by the human complement system when administered intravenously. Here, an NDV variant (NDV F0) was generated for production in mammalian cells, without passage in eggs. This was achieved by introducing the V-106-M and L-117-S amino acid substitutions upstream of the cleavage site in the F protein, resulting in rNDV F0-M, rNDV F0-S, and NDV F0-M/S. These viruses can be considered non-virulent as determined with in vivo pathogenicity testing and were neutralized less by the human complement system, which is explained by CD46 expression on the viral membrane. The inoculation of 10 pancreatic cancer cell lines demonstrated similar or enhanced replication and cell-killing efficacy of rNDV F0-M/S compared to rNDV F0 and rNDV F0-M. In conclusion, NDV F0 variants with M and S substitutions are non-virulent, effective oncolytic viruses that can be produced in mammalian cells, potentially resulting in a more effective treatment option for pancreatic cancer patients compared to rNDV F0.
Collapse
Affiliation(s)
- Marco Huberts
- Department of Viroscience, Erasmus Medical Centrum, Doctor Molewaterplein 40, 3015 CN Rotterdam, the Netherlands
| | - J. Fréderique de Graaf
- Department of Immunology, Leids Universitair Medisch Centrum, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Daphne Groeneveld
- Department of Viroscience, Erasmus Medical Centrum, Doctor Molewaterplein 40, 3015 CN Rotterdam, the Netherlands
| | - Stefan van Nieuwkoop
- Department of Viroscience, Erasmus Medical Centrum, Doctor Molewaterplein 40, 3015 CN Rotterdam, the Netherlands
| | - Ron A.M. Fouchier
- Department of Viroscience, Erasmus Medical Centrum, Doctor Molewaterplein 40, 3015 CN Rotterdam, the Netherlands
| | - Bernadette G. van den Hoogen
- Department of Viroscience, Erasmus Medical Centrum, Doctor Molewaterplein 40, 3015 CN Rotterdam, the Netherlands
| |
Collapse
|
27
|
Vile R, Kendall B, Liseth O, Sangsuwannukul T, Elliott N, Yerovi MC, Thompson J, Swanson J, Rizk S, Diaz R, Tonne J. Immunodominant antiviral T cell responses outcompete immuno-subdominant antitumor responses to reduce the efficacy of oncolytic viroimmunotherapy. RESEARCH SQUARE 2025:rs.3.rs-6131273. [PMID: 40166032 PMCID: PMC11957203 DOI: 10.21203/rs.3.rs-6131273/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The paradigm in the field of oncolytic virotherapy proposes that tumor cell killing by an oncolytic virus (OV) culminates in the priming of antitumor CD8 T cells. However, this ignores the impact a highly immunodominant antiviral response against the OV has on the antitumor response which has been weakened by mechanisms of central tolerance. Here, we show that inflammatory Vesicular Stomatitis Virus (VSV) failed to prime an adoptively transferred, or pre-existing, population of tumor-reactive T cells. Combination with αPD1 immune checkpoint blockade therapy improved survival only when VSV expressed tumor associated antigens (TAA). These data show that, in this model, the highly inflammatory OV VSV alone actively outcompetes antitumor immunity. However, we also show that viral expression of a mutant near-self TAA can break central tolerance expanding heteroclitic self-reactive and near-self-reactive T cells, thus overcoming viral immunodominance by promoting tumor-specific T cell proliferation in parallel with expanding antiviral T cells.
Collapse
|
28
|
Shah DD, Chorawala MR, Raghani NR, Patel R, Fareed M, Kashid VA, Prajapati BG. Tumor microenvironment: recent advances in understanding and its role in modulating cancer therapies. Med Oncol 2025; 42:117. [PMID: 40102282 DOI: 10.1007/s12032-025-02641-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
Tumor microenvironment (TME) denotes the non-cancerous cells and components presented in the tumor, including molecules produced and released by them. Interactions between cancer cells, immune cells, stromal cells, and the extracellular matrix within the TME create a dynamic ecosystem that can either promote or hinder tumor growth and spread. The TME plays a pivotal role in either promoting or inhibiting tumor growth and dissemination, making it a critical factor to consider in the development of effective cancer therapies. Understanding the intricate interplay within the TME is crucial for devising effective cancer therapies. Combination therapies involving inhibitors of immune checkpoint blockade (ICB), and/or chemotherapy now offer new approaches for cancer therapy. However, it remains uncertain how to best utilize these strategies in the context of the complex tumor microenvironment. Oncogene-driven changes in tumor cell metabolism can impact the TME to limit immune responses and present barriers to cancer therapy. Cellular and acellular components in tumor microenvironment can reprogram tumor initiation, growth, invasion, metastasis, and response to therapies. Components in the TME can reprogram tumor behavior and influence responses to treatments, facilitating immune evasion, nutrient deprivation, and therapeutic resistance. Moreover, the TME can influence angiogenesis, promoting the formation of blood vessels that sustain tumor growth. Notably, the TME facilitates immune evasion, establishes a nutrient-deprived milieu, and induces therapeutic resistance, hindering treatment efficacy. A paradigm shift from a cancer-centric model to a TME-centric one has revolutionized cancer research and treatment. However, effectively targeting specific cells or pathways within the TME remains a challenge, as the complexity of the TME poses hurdles in designing precise and effective therapies. This review highlights challenges in targeting the tumor microenvironment to achieve therapeutic efficacy; explore new approaches and technologies to better decipher the tumor microenvironment; and discuss strategies to intervene in the tumor microenvironment and maximize therapeutic benefits.
Collapse
Affiliation(s)
- Disha D Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India.
| | - Neha R Raghani
- Department of Pharmacology and Pharmacy Practice, Saraswati Institute of Pharmaceutical Sciences, Gandhinagar, Gujarat, 382355, India
| | - Rajanikant Patel
- Department of Product Development, Granules Pharmaceuticals Inc., 3701 Concorde Parkway, Chantilly, VA, 20151, USA
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, 13713, Riyadh, Saudi Arabia
| | - Vivekanand A Kashid
- MABD Institute of Pharmaceutical Education and Research, Babhulgaon, Yeola, Nashik, India
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Kherva, Mehsana, Gujarat, 384012, India.
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand.
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
29
|
Stojchevski R, Sutanto EA, Sutanto R, Hadzi-Petrushev N, Mladenov M, Singh SR, Sinha JK, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Bhaskar R, Avtanski D. Translational Advances in Oncogene and Tumor-Suppressor Gene Research. Cancers (Basel) 2025; 17:1008. [PMID: 40149342 PMCID: PMC11940485 DOI: 10.3390/cancers17061008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cancer, characterized by the uncontrolled proliferation of cells, is one of the leading causes of death globally, with approximately one in five people developing the disease in their lifetime. While many driver genes were identified decades ago, and most cancers can be classified based on morphology and progression, there is still a significant gap in knowledge about genetic aberrations and nuclear DNA damage. The study of two critical groups of genes-tumor suppressors, which inhibit proliferation and promote apoptosis, and oncogenes, which regulate proliferation and survival-can help to understand the genomic causes behind tumorigenesis, leading to more personalized approaches to diagnosis and treatment. Aberration of tumor suppressors, which undergo two-hit and loss-of-function mutations, and oncogenes, activated forms of proto-oncogenes that experience one-hit and gain-of-function mutations, are responsible for the dysregulation of key signaling pathways that regulate cell division, such as p53, Rb, Ras/Raf/ERK/MAPK, PI3K/AKT, and Wnt/β-catenin. Modern breakthroughs in genomics research, like next-generation sequencing, have provided efficient strategies for mapping unique genomic changes that contribute to tumor heterogeneity. Novel therapeutic approaches have enabled personalized medicine, helping address genetic variability in tumor suppressors and oncogenes. This comprehensive review examines the molecular mechanisms behind tumor-suppressor genes and oncogenes, the key signaling pathways they regulate, epigenetic modifications, tumor heterogeneity, and the drug resistance mechanisms that drive carcinogenesis. Moreover, the review explores the clinical application of sequencing techniques, multiomics, diagnostic procedures, pharmacogenomics, and personalized treatment and prevention options, discussing future directions for emerging technologies.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Edward Agus Sutanto
- CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA;
| | - Rinni Sutanto
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY 11545, USA;
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Sajal Raj Singh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Jitendra Kumar Sinha
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, Maharashtra, India;
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, Haryana, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
30
|
He W, Cui K, Farooq MA, Huang N, Zhu S, Jiang D, Zhang X, Chen J, Liu Y, Xu G. TCR-T cell therapy for solid tumors: challenges and emerging solutions. Front Pharmacol 2025; 16:1493346. [PMID: 40129944 PMCID: PMC11931055 DOI: 10.3389/fphar.2025.1493346] [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: 09/09/2024] [Accepted: 02/20/2025] [Indexed: 03/26/2025] Open
Abstract
With the use of T cell receptor T cells (TCR-T cells) and chimeric antigen receptor T cells (CAR-T cells), T-cell immunotherapy for cancer has advanced significantly in recent years. CAR-T cell therapy has demonstrated extraordinary success when used to treat hematologic malignancies. Nevertheless, there are several barriers that prevent this achievement from being applied to solid tumors, such as challenges with tumor targeting and inadequate transit and adaption of genetically modified T-cells, especially in unfavorable tumor microenvironments The deficiencies of CAR-T cell therapy in the treatment of solid tumors are compensated for by TCR-T cells, which have a stronger homing ability to initiate intracellular commands, 90% of the proteins can be used as developmental targets, and they can recognize target antigens more broadly. As a result, TCR-T cells may be more effective in treating solid tumors. In this review, we discussed the structure of TCR-T and have outlined the drawbacks of TCR-T in cancer therapy, and suggested potential remedies. This review is crucial in understanding the current state and future potential of TCR-T cell therapy. We emphasize how important it is to use combinatorial approaches, combining new combinations of various emerging strategies with over-the-counter therapies designed for TCR-T, to increase the anti-tumor efficacy of TCR-T inside the TME and maximize treatment safety, especially when it comes to solid tumor immunotherapies.
Collapse
Affiliation(s)
- Wanjun He
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, Guangdong Medical University, Dongguan, China
| | - Kai Cui
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, Guangdong Medical University, Dongguan, China
| | - Muhammad Asad Farooq
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Na Huang
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, Guangdong Medical University, Dongguan, China
| | - Songshan Zhu
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, Guangdong Medical University, Dongguan, China
| | - Dan Jiang
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, Guangdong Medical University, Dongguan, China
| | - Xiqian Zhang
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, Guangdong Medical University, Dongguan, China
- Yinchuan Guolong Orthopedic Hospital, Yinchuan, China
| | - Jian Chen
- Yinchuan Guolong Orthopedic Hospital, Yinchuan, China
| | - Yinxia Liu
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Guangxian Xu
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, Guangdong Medical University, Dongguan, China
| |
Collapse
|
31
|
He Y, Li W, Zhang X, Cui Z. Oncolytic Virus Targeted Therapy for Glioma via Intravenous Delivery. Adv Healthc Mater 2025; 14:e2404965. [PMID: 39801205 DOI: 10.1002/adhm.202404965] [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] [Indexed: 03/18/2025]
Abstract
Glioma, the deadly primary intracranial tumor, poses challenges in clinical treatment due to its infiltrative growth and resistance to radiation. Oncolytic virus therapy holds potential for the treatment of malignant gliomas, but its application is impeded by the requirement for intracranial injections due to the presence of blood-brain barrier (BBB). In this study, to overcome this limitation, the study develops a nanocapsule encapsulating the recombinant oncolytic virus EV-A71-miR124T, enabling the treatment of glioma through intravenous administration. It is demonstrated that the nanocapsule can cross the BBB and selectively release oncolytic virus at the tumor site, resulting in targeted and specific killing of glioma cells. In mice with implanted intracranial orthotopic gliomas, intravenous administration of the nanocapsule suppresses tumor growth and significantly extends survival time. Consequently, the study establishes an effective treatment method for malignant gliomas using an oncolytic virus nanocapsule through intravenous administration. These findings provide a new strategy for oncolytic virus therapy in glioma treatment and offer perspectives for targeted therapies of other brain tumors and diseases.
Collapse
Affiliation(s)
- Yechenxing He
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Wei Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, P. R. China
| | - Xiaowei Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, P. R. China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
32
|
Chen J, Ma N, Chen B, Huang Y, Li J, Li J, Chen Z, Wang P, Ran B, Yang J, Bai J, Ning S, Ai J, Wei Q, Liu L, Cao D. Synergistic effects of immunotherapy and adjunctive therapies in prostate cancer management. Crit Rev Oncol Hematol 2025; 207:104604. [PMID: 39732304 DOI: 10.1016/j.critrevonc.2024.104604] [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: 11/15/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024] Open
Abstract
In recent years, cancer immunotherapy has received widespread attention due to significant tumor clearance in some malignancies. Various immunotherapy approaches, including vaccines, immune checkpoint inhibitors, oncolytic virotherapy, bispecific T cell engagers, and adoptive T cell transfer, have completed or are undergoing clinical trials for prostate cancer. Despite immune checkpoint blockade's extraordinary effectiveness in treating a variety of cancers, targeted prostate cancer treatment using the immune system is still in its infancy. Multiple factors including the heterogeneity of prostate cancer, the cold tumor microenvironment, and a low level of neoantigens, contribute to the poor immunotherapy response. Significant effort is being devoted to improving immune-based prostate cancer therapy. Recently, several key discoveries demonstrate that prostate cancer immunotherapy agents may be used to promise better prognosis for patients as part of combination strategies with other agents targeting tumor-associated immune mechanism of resistance. Here, this review comprehensively examines the recent advancements in immunotherapy for prostate cancer, exploring its potential synergistic effects when combined with other treatment modalities to enhance clinical efficacy.
Collapse
Affiliation(s)
- Jie Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Ma
- Department of Pediatrics, West China Second University Hospital, Sichuan University, No. 20, 3rd section, South Renmin Road, Chengdu 610041, China
| | - Bo Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yin Huang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinze Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zeyu Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Puze Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Biao Ran
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiahao Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingxing Bai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shu Ning
- Department of Urologic Surgery, University of California Davis, Davis, CA, USA
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liangren Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Dehong Cao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
33
|
Mitrakas AG, Kakouratos C, Lamprou I, Xanthopoulou E, Koukourakis MI. Oncogenic Mutations and the Tumor Microenvironment: Drivers of Non-Small Cell Lung Cancer Progression. Cancers (Basel) 2025; 17:853. [PMID: 40075700 PMCID: PMC11899603 DOI: 10.3390/cancers17050853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Non-small cell lung cancer (NSCLC) is a major cause of cancer-related deaths globally. The study focuses on understanding the interplay between genetic mutations, cancer stem cells (CSCs), and the tumor microenvironment (TME) in driving NSCLC progression, resistance to therapies, and relapse. METHODS A systematic search was conducted in PubMed and Scopus databases to identify significant and valuable studies relevant to NSCLC, focusing on genetic mutations, CSCs, and the TME. Articles were selected based on their relevance, methodological severity, date of publication, and scientific soundness related to NSCLC biology and therapeutic strategies. This review synthesized findings from these sources to highlight key mechanisms and potential therapeutic interventions. RESULTS Mutations in critical genes in KRAS, EGFR, TP53, and other key genes interfere with stem cell regulation, promoting CSC-like behavior, resistance to therapy, and immune evasion. The tumor microenvironment (TME), including immune cells, fibroblasts, and extracellular matrix components, further supports tumor growth and reduction in treatment efficacy. Promising strategies, including CSC targeting, TME modulation, and the development of novel biomarkers, have shown potential in preclinical and clinical studies. CONCLUSIONS The association between genetic alterations, CSCs, the TME, and other cellular pathways-including cell metabolism and immune evasion-plays a crucial role in therapy resistance, highlighting the need for comprehensive treatment strategies. The combination of genomic profiling with TME-targeting therapies could lead to personalized treatment approaches, offering hope for better clinical outcomes and reduced mortality in NSCLC patients.
Collapse
Affiliation(s)
- Achilleas G. Mitrakas
- Department of Radiotherapy/Oncology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (C.K.); (I.L.); (E.X.)
| | | | | | | | - Michael I. Koukourakis
- Department of Radiotherapy/Oncology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (C.K.); (I.L.); (E.X.)
| |
Collapse
|
34
|
Yang Q, Shu Y, Chen Y, Qi Z, Hu S, Zhang Y, Qin Y, Xu X, Hu J, Huang A, Cheng P. Expression of SIRPα-Fc by oncolytic virus enhances antitumor efficacy through tumor microenvironment reprogramming. Front Immunol 2025; 16:1513555. [PMID: 40070841 PMCID: PMC11893986 DOI: 10.3389/fimmu.2025.1513555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
Oncolytic viruses (OVs) selectively replicate within tumors, directly killing cancer cells and promoting a systemic immune response by releasing tumor antigens. These features make OVs a promising approach in tumor immunotherapy, offering targeted treatment with fewer side effects. Despite these advantages, OVs are primarily administered via intratumoral injection, limiting their effectiveness for advanced, systemic cancers. Among OVs, oncolytic adenoviruses (oAdVs) are the most widely studied due to their well-understood gene regulation, safety, and stability. In this study, a modified oAdV vector, pDC316-oAd-SA, was engineered to express the SIRPα-mIgG1Fc gene, designed to remodel tumor-associated macrophages (TAMs) and enhance anti-tumor immunity. This vector, along with a control virus (Ad-ON), was evaluated both in vitro and in vivo. The modified oAd-SA significantly improved macrophage phagocytosis and showed superior tumor regression in murine models. Additionally, while both oAdVs increased T cell infiltration in the tumor microenvironment, oAd-SA specifically enhanced T cell immune function. The study also revealed that oAdVs modulate TAMs differently across tumor types, with oAd-SA therapy particularly increasing TAM phagocytosis and promoting an anti-tumor response.
Collapse
Affiliation(s)
- Qingzhe Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yongheng Shu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yanwei Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongbing Qi
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shichuan Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Qin
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xianglin Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jianchuan Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Anliang Huang
- Department of Pathology, Chengdu Fifth People’s Hospital, Chengdu, China
| | - Ping Cheng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
35
|
Sellers GS, Poirier MA, Mayberry TG, Cowan BC, Wakefield MR, Fang Y. From conventional to cutting edge: an exploration of osteosarcoma treatments. Med Oncol 2025; 42:81. [PMID: 39982613 DOI: 10.1007/s12032-025-02629-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
Osteosarcoma is a highly aggressive cancer in children and young adults that has a remarkably high mortality rate upon metastasis. Current standard treatments have remained largely unchanged for nearly five decades, focusing on a combination of chemotherapy with high-dose methotrexate, doxorubicin, and cisplatin, complemented by aggressive surgical resections. Despite this lack of change, recent advancements in medical research have spurred hope for more effective and less invasive approaches to managing osteosarcoma. In this review, we provide an overview of existing therapeutic modalities, including chemotherapy regimens tailored to tumor stage and patient response, radiation therapies aimed at local tumor control, and advanced surgical techniques such as limb-sparing procedures. Additionally, we explore two promising future treatments that are currently under investigation for osteosarcoma cases: targeted therapies utilizing nanomaterials like graphene oxide and innovative oncolytic viruses. This review highlights potential breakthroughs in treatment options while identifying areas that warrant further investigation in the management of osteosarcoma. Considering the limited advancements in treatment over the past decades, identifying and highlighting novel and effective therapies is vital for improving patient outcomes and survival rates.
Collapse
Affiliation(s)
- Garen S Sellers
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA, 50266, USA
| | - McKade A Poirier
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA, 50266, USA
| | - Trenton G Mayberry
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Braydon C Cowan
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Mark R Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA, 50266, USA.
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA.
| |
Collapse
|
36
|
Zhong L, Gan L, Wang B, Wu T, Yao F, Gong W, Peng H, Deng Z, Xiao G, Liu X, Na J, Xia D, Yu X, Zhang Z, Xiang B, Huo Y, Yan D, Dong Z, Fang F, Ma Y, Jin G, Su D, Liu X, Li Q, Liao H, Tang C, He J, Tang Z, Zhang S, Qiu B, Yang Z, Yang L, Chen Z, Zeng M, Feng R, Jiao J, Liao Y, Wang T, Wu L, Mi Z, Liu Z, Shi S, Zhang K, Shi W, Zhao Y. Hyperacute rejection-engineered oncolytic virus for interventional clinical trial in refractory cancer patients. Cell 2025; 188:1119-1136.e23. [PMID: 39826543 DOI: 10.1016/j.cell.2024.12.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 10/01/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025]
Abstract
Recently, oncolytic virus (OV) therapy has shown great promise in treating malignancies. However, intravenous safety and inherent lack of immunity are two significant limitations in clinical practice. Herein, we successfully developed a recombinant Newcastle disease virus with porcine α1,3GT gene (NDV-GT) triggering hyperacute rejection. We demonstrated its feasibility in preclinical studies. The intravenous NDV-GT showed superior ability to eradicate tumor cells in our innovative CRISPR-mediated primary hepatocellular carcinoma monkeys. Importantly, the interventional clinical trial treating 20 patients with relapsed/refractory metastatic cancer (Chinese Clinical Trial Registry of WHO, ChiCTR2000031980) showed a high rate (90.00%) of disease control and durable responses, without serious adverse events and clinically functional neutralizing antibodies, further suggesting that immunogenicity is minimal under these conditions and demonstrating the feasibility of NDV-GT for immunovirotherapy. Collectively, our results demonstrate the high safety and efficacy of intravenous NDV-GT, thus providing an innovative technology for OV therapy in oncological therapeutics and beyond.
Collapse
Affiliation(s)
- Liping Zhong
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China.
| | - Lu Gan
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Bing Wang
- Department of Spine Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Tao Wu
- The First People's Hospital of Changde City, Changde, Hunan 415000, China
| | - Fei Yao
- Department of Oncology, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi 530023, China
| | - Wenlin Gong
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Hongmei Peng
- The First People's Hospital of Changde City, Changde, Hunan 415000, China
| | - Zhiming Deng
- The First People's Hospital of Changde City, Changde, Hunan 415000, China
| | - Guoyou Xiao
- Department of Nuclear Medicine, The Affiliated Tumor Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Desong Xia
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, The Affiliated Tumor Hospital, Fudan University, Shanghai 200032, China
| | - Zhikun Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Bangde Xiang
- Department of Hepatobiliary Surgery, The Affiliated Tumor Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yu Huo
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Dan Yan
- Department of Oncology, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi 530023, China
| | - Zhixin Dong
- Department of Oncology, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi 530023, China
| | - Fang Fang
- Department of Oncology, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi 530023, China
| | - Yun Ma
- Department of Pathology, The Affiliated Tumor Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Guanqiao Jin
- Department of Radiology, The Affiliated Tumor Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Danke Su
- Department of Radiology, The Affiliated Tumor Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xiuli Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Qiang Li
- Department of Radiology, The Affiliated Tumor Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Hai Liao
- Department of Radiology, The Affiliated Tumor Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Chao Tang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jian He
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zhiping Tang
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Shilai Zhang
- Department of Nuclear Medicine, The Affiliated Tumor Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Bingqing Qiu
- Department of Nuclear Medicine, The Affiliated Tumor Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zhi Yang
- Department of Nuclear Medicine, The Affiliated Tumor Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Lihui Yang
- Fundamental Nursing Teaching and Research Office, Nursing College of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Ziqin Chen
- The First People's Hospital of Changde City, Changde, Hunan 415000, China
| | - Mengsi Zeng
- The First People's Hospital of Changde City, Changde, Hunan 415000, China
| | - Ronghua Feng
- The First People's Hospital of Changde City, Changde, Hunan 415000, China
| | - Jiege Jiao
- Yuandan Biotechnology (Hainan) Co., Ltd., Haikou, Hainan 570100, China
| | - Yuan Liao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Tinghua Wang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Liangliang Wu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zhengcheng Mi
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Ziqun Liu
- Department of Spine Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Si Shi
- Department of Pancreatic Surgery, The Affiliated Tumor Hospital, Fudan University, Shanghai 200032, China
| | - Kun Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China.
| | - Wei Shi
- Department of Oncology, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi 530023, China.
| | - Yongxiang Zhao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
37
|
Luo D, Liu Y, Lu Z, Huang L. Targeted therapy and immunotherapy for gastric cancer: rational strategies, novel advancements, challenges, and future perspectives. Mol Med 2025; 31:52. [PMID: 39923010 PMCID: PMC11806620 DOI: 10.1186/s10020-025-01075-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: 07/16/2024] [Accepted: 01/10/2025] [Indexed: 02/10/2025] Open
Abstract
Gastric cancer (GC) is one of the most common malignant tumors worldwide, and its treatment has been a focus of medical research. Herein we systematically review the current status of and advancements in targeted therapy and immunotherapy for GC, which have emerged as important treatment strategies in recent years with great potential, and summarize the efficacy and safety of such treatments. Targeted therapies against key targets in GC, including epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), and vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR), have shown remarkable therapeutic efficacies by inhibiting tumor progression and/or blood supply. In particular, markable breakthroughs have been made in HER2-targeting drugs for HER2-positive GC patients. To address intrinsic and acquired resistances to HER2-targeting drugs, novel therapeutic agents including bispecific antibodies and antibody-drug conjugates (ADC) targeting HER2 have been developed. Immunotherapy enhances the recognition and elimination of cancer cells by activating body anticancer immune system. Programmed cell death protein 1 (PD-1) and programmed cell death-ligand 1 (PD-L1) antibodies are the most commonly used immunotherapeutic agents and have been used with some success in GC treatment. Innovative immunotherapy modalities, including adoptive immune cell therapy, tumor vaccines, and non-specific immunomodulators therapy, and oncolytic viruses have shown promise in early-stage clinical trials for GC. Clinical trials have supported that targeted therapy and immunotherapy can significantly improve the survival and quality of life of GC patients. However, the effects of such therapies need to be further improved and more personalized, with advancement in researches on tumor immune microenvironment. Further studies remain needed to address the issues of drug resistance and adverse events pertaining to such therapies for GC. The combined application of such therapies and individualized treatment strategies should be further explored with novel drugs developed, to provide more effective treatments for GC patients.
Collapse
Affiliation(s)
- Dong Luo
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
- Center of Structural Heart Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yunmei Liu
- School of Cultural Heritage and Information Management, Shanghai University, Shanghai, 200444, China.
| | - Zhengmao Lu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China.
| | - Lei Huang
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
- National Key Laboratory of Immunity and Inflammation, Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
38
|
Shokoohi M, Sedaghatshoar S, Arian H, Mokarami M, Habibi F, Bamarinejad F. Genetic advancements in breast cancer treatment: a review. Discov Oncol 2025; 16:127. [PMID: 39918655 PMCID: PMC11805739 DOI: 10.1007/s12672-025-01884-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/03/2025] [Indexed: 02/09/2025] Open
Abstract
Breast cancer (BC) remains a leading cause of cancer-related deaths among women globally, highlighting the urgent need for more effective and targeted therapies. Traditional treatments, including surgery, chemotherapy, and radiation, face limitations such as drug resistance, metastasis, and severe side effects. Recent advancements in gene therapy, particularly CRISPR/Cas9 technology and Oncolytic Virotherapy (OVT), are transforming the BC treatment landscape. CRISPR/Cas9 enables precise gene editing to correct mutations in oncogenes like HER2 and MYC, directly addressing tumor growth and immune evasion. Simultaneously, OVT leverages genetically engineered viruses to selectively destroy cancer cells and stimulate robust antitumor immune responses. Despite their potential, gene therapies face challenges, including off-target effects, delivery issues, and ethical concerns. Innovations in delivery systems, combination strategies, and integrating gene therapy with existing treatments offer promising solutions to overcome these barriers. Personalized medicine, guided by genomic profiling, further enhances treatment precision by identifying patient-specific mutations, such as BRCA1 and BRCA2, allowing for more tailored and effective interventions. As research progresses, the constructive interaction between gene therapy, immunotherapy, and traditional approaches is paving the way for groundbreaking advancements in BC care. Continued collaboration between researchers and clinicians is essential to translate these innovations into clinical practice, ultimately improving BC patients' survival rates and quality of life.
Collapse
Affiliation(s)
- Marzieh Shokoohi
- Department of Life Sciences Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, Iran.
- Amino Techno Gene Virtual Private Laboratory, Tehran, Iran.
| | - Sadaf Sedaghatshoar
- Kent School of Social Work and Family Science, University of Louisville, Louisville, KY, USA
| | - Homaira Arian
- Pharmaceutical Biotechnology Department, Pharmacy Faculty, Anadolu University, Eskishehir, Turkey.
| | - Milad Mokarami
- Student Research Committee, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Fatemeh Habibi
- Department of Speech Therapy, School of Rehabilitation, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Bamarinejad
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
39
|
Borella F, Carosso M, Chiparo MP, Ferraioli D, Bertero L, Gallio N, Preti M, Cusato J, Valabrega G, Revelli A, Marozio L, Cosma S. Oncolytic Viruses in Ovarian Cancer: Where Do We Stand? A Narrative Review. Pathogens 2025; 14:140. [PMID: 40005517 PMCID: PMC11858389 DOI: 10.3390/pathogens14020140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Ovarian cancer (OC) remains the most lethal gynecologic malignancy with limited effective treatment options. Oncolytic viruses (OVs) have emerged as a promising therapeutic approach for cancer treatment, capable of selectively infecting and lysing cancer cells while stimulating anti-tumor immune responses. Preclinical studies have demonstrated significant tumor regression and prolonged survival in OC models using various OVs, such as herpes simplex. Early-phase clinical trials have shown a favorable safety profile, though the impact on patient survival has been modest. Current research focuses on combining OVs with other treatments like immune checkpoint inhibitors to enhance their efficacy. We provide a comprehensive overview of the current understanding and future directions for utilizing OVs in the management of OC.
Collapse
Affiliation(s)
- Fulvio Borella
- Gynecology and Obstetrics 1U, Departments of Surgical Sciences, University of Turin, 10126 Turin, Italy; (M.C.); (M.P.C.); (L.M.); (S.C.)
| | - Marco Carosso
- Gynecology and Obstetrics 1U, Departments of Surgical Sciences, University of Turin, 10126 Turin, Italy; (M.C.); (M.P.C.); (L.M.); (S.C.)
| | - Maria Pia Chiparo
- Gynecology and Obstetrics 1U, Departments of Surgical Sciences, University of Turin, 10126 Turin, Italy; (M.C.); (M.P.C.); (L.M.); (S.C.)
| | - Domenico Ferraioli
- Department of Gynecology, Léon Bérard, Comprehensive Cancer Centre, 69008 Lyon, France;
| | - Luca Bertero
- Pathology Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy;
| | - Niccolò Gallio
- Gynecology and Obstetrics 2U, Departments of Surgical Sciences, University of Turin, 10126 Turin, Italy; (N.G.); (A.R.)
| | - Mario Preti
- Gynecology and Obstetrics 1U, Departments of Surgical Sciences, University of Turin, 10126 Turin, Italy; (M.C.); (M.P.C.); (L.M.); (S.C.)
| | - Jessica Cusato
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, University of Turin, 10149 Turin, Italy;
| | - Giorgio Valabrega
- Department of Oncology, University of Turin, Medical Oncology, Ordine Mauriziano Hospital, 10128 Turin, Italy;
| | - Alberto Revelli
- Gynecology and Obstetrics 2U, Departments of Surgical Sciences, University of Turin, 10126 Turin, Italy; (N.G.); (A.R.)
| | - Luca Marozio
- Gynecology and Obstetrics 1U, Departments of Surgical Sciences, University of Turin, 10126 Turin, Italy; (M.C.); (M.P.C.); (L.M.); (S.C.)
| | - Stefano Cosma
- Gynecology and Obstetrics 1U, Departments of Surgical Sciences, University of Turin, 10126 Turin, Italy; (M.C.); (M.P.C.); (L.M.); (S.C.)
| |
Collapse
|
40
|
Zhao X, Li W, Sun Y, Ma J. Oncolytic senecavirus A in tumor immunotherapy: Mechanisms, progress, and future directions. Virology 2025; 603:110338. [PMID: 39667099 DOI: 10.1016/j.virol.2024.110338] [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: 10/15/2024] [Revised: 11/14/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024]
Abstract
Oncolytic virotherapy has emerged as a promising immunotherapy strategy against cancer. As the first picornavirus tested in humans for its oncolytic potential, Senecavirus A (SVA) possesses several advantageous features, including its small size, rapid replication, and ability to penetrate the vascular system of solid tumors, allowing for the specific targeting and lysis of tumor cells. Additionally, SVA does not integrate into the host genome, thus avoiding potential genomic damage, and it lacks oncogenes or other virulence genes. Importantly, no significant pathogenic effects have been observed in humans or companion animals. Due to its simple genetic structure, SVA is amenable to various genetic modifications, allowing it to carry exogenous genes to further enhance tumor therapy. This review summarizes current knowledge of SVA's mechanisms of action and its progress in oncolytic therapy research, while also addressing the challenges and future directions.
Collapse
Affiliation(s)
- Xiaoya Zhao
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Wenjie Li
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Yuan Sun
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| | - Jingyun Ma
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong, 510642, China.
| |
Collapse
|
41
|
Li K, Guo B, Gu J, Ta N, Gu J, Yu H, Sun M, Han T. Emerging advances in drug delivery systems (DDSs) for optimizing cancer complications. Mater Today Bio 2025; 30:101375. [PMID: 39759851 PMCID: PMC11699619 DOI: 10.1016/j.mtbio.2024.101375] [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: 08/02/2024] [Revised: 11/13/2024] [Accepted: 11/29/2024] [Indexed: 01/07/2025] Open
Abstract
The management and treatment of tumor complications pose continuous challenges due to the inherent complexity. However, the advent of drug delivery systems (DDSs) brings promising opportunities to address the tumor complications using innovative technological approaches. This review focuses on common oncological complications, including cancer thrombosis, malignant serous effusion, tumor-associated infections, cancer pain, and treatment-related complications. Emphasis was placed on the application and potential of DDSs in mitigating and treating these tumor complications, and we delved into the underlying mechanisms of common cancer-associated complications, discussed the limitations of conventional treatments, and outlined the current status and potential development of DDSs for various complications in this review. Moreover, we have discussed the existing challenges in DDSs research, underscoring the need for addressing issues related to biocompatibility and targeting of DDSs, optimizing drug delivery routes, and enhancing delivery efficiency and precision. In conclusion, DDSs offer promising avenues for treating cancer complications, offering the potential for the development of more effective and safer drug delivery strategies, thereby improving the quality of life and survival rates of cancer patients.
Collapse
Affiliation(s)
- Kerui Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Bei Guo
- Department of Endocrinology, General Hospital of Northern Theater Command, Shenyang, 110001, China
| | - Junmou Gu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Na Ta
- Department of Neurology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116044, China
| | - Jia Gu
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Hao Yu
- Department of Endocrinology, General Hospital of Northern Theater Command, Shenyang, 110001, China
| | - Mengchi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Tao Han
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| |
Collapse
|
42
|
Diers AR, Guo Q, Li Z, Richardson E, Idris S, Willis C, Tak PP, Withers DR, Barone F. Dynamic Tracking of Tumor Microenvironment Modulation Using Kaede Photoconvertible Transgenic Mice Unveils New Biological Properties of Viral Immunotherapy. CANCER RESEARCH COMMUNICATIONS 2025; 5:327-338. [PMID: 39881590 PMCID: PMC11831061 DOI: 10.1158/2767-9764.crc-24-0434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/06/2024] [Accepted: 01/28/2025] [Indexed: 01/31/2025]
Abstract
SIGNIFICANCE This study utilized a novel photoconvertible mouse tumor model to track immune cell trafficking upon treatment with an investigational viral immunotherapy (CAN-2409), revealing enhanced T-cell responses after viral immunotherapy associated with local proliferation of T cells within tumors that could further enhance antitumor efficacy in combination with immune checkpoint inhibitors. These findings define temporally and spatially distinct interactions of immune cells that could be harnessed by novel therapeutics.
Collapse
Affiliation(s)
| | - Qiuchen Guo
- Candel Therapeutics, Inc., Needham, Massachusetts
| | - Zhi Li
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Erin Richardson
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Suaad Idris
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Claire Willis
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul P. Tak
- Candel Therapeutics, Inc., Needham, Massachusetts
| | - David R. Withers
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | | |
Collapse
|
43
|
Li YR, Lyu Z, Shen X, Fang Y, Yang L. Boosting CAR-T cell therapy through vaccine synergy. Trends Pharmacol Sci 2025; 46:180-199. [PMID: 39755457 DOI: 10.1016/j.tips.2024.12.004] [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: 11/08/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 01/06/2025]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has transformed the treatment landscape for hematological cancers. However, achieving comparable success in solid tumors remains challenging. Factors contributing to these limitations include the scarcity of tumor-specific antigens (TSAs), insufficient CAR-T cell infiltration, and the immunosuppressive tumor microenvironment (TME). Vaccine-based strategies are emerging as potential approaches to address these challenges, enhancing CAR-T cell expansion, persistence, and antitumor efficacy. In this review, we explore diverse vaccine modalities, including mRNA, peptide, viral vector, and dendritic cell (DC)-based vaccines, and their roles in augmenting CAR-T cell responses. Special focus is given to recent clinical advancements combining mRNA-based vaccines with CAR-T therapy for the treatment of genitourinary cancers. In addition, we discuss crucial considerations for optimizing vaccine dosing, scheduling, and delivery to maximize CAR-T synergy, aiming to refine this combination strategy to improve treatment efficacy and safety.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Zibai Lyu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xinyuan Shen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ying Fang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
44
|
Grimes JM, Ghosh S, Manzoor S, Li LX, Moran MM, Clements JC, Alexander SD, Markert JM, Leavenworth JW. Oncolytic reprogramming of tumor microenvironment shapes CD4 T-cell memory via the IL6ra-Bcl6 axis for targeted control of glioblastoma. Nat Commun 2025; 16:1095. [PMID: 39885128 PMCID: PMC11782536 DOI: 10.1038/s41467-024-55455-9] [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: 05/22/2024] [Accepted: 12/13/2024] [Indexed: 02/01/2025] Open
Abstract
Oncolytic viruses (OVs) emerge as a promising cancer immunotherapy. However, the temporal impact on tumor cells and the tumor microenvironment, and the nature of anti-tumor immunity post-therapy remain largely unclear. Here we report that CD4+ T cells are required for durable tumor control in syngeneic murine models of glioblastoma multiforme after treatment with an oncolytic herpes simplex virus (oHSV) engineered to express IL-12. The upregulated MHCII on residual tumor cells facilitates programmed polyfunctional CD4+ T cells for tumor control and for recall responses. Mechanistically, the proper ratio of Bcl-6 to T-bet in CD4+ T cells navigates their enhanced anti-tumor capacity, and a reciprocal IL6ra-Bcl-6 regulatory axis in a memory CD4+ T-cell subset, which requires MHCII signals from reprogrammed tumor cells, tumor-infiltrating and resident myeloid cells, is necessary for the prolonged response. These findings uncover an OV-induced tumor/myeloid-CD4+ T-cell partnership, leading to long-term anti-tumor immune memory, and improved OV therapeutic efficacy.
Collapse
Affiliation(s)
- Jeffrey M Grimes
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
- Graduate Biomedical Sciences Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sadashib Ghosh
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shamza Manzoor
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Li X Li
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Monica M Moran
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
- Graduate Biomedical Sciences Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jennifer C Clements
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sherrie D Alexander
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James M Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
- The O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jianmei W Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA.
- The O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
45
|
Kar S, Mehrotra S, Prajapati VK. From infection to remedy: Harnessing oncolytic viruses in cancer treatment. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 144:213-257. [PMID: 39978967 DOI: 10.1016/bs.apcsb.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Oncolytic virus (OV) mediated immunotherapy is one of the recent techniques used to treat higher grade cancers where conventional therapies like chemotherapy, radiation fail. OVs as a therapeutic tool show high efficacy and fewer side effects than conventional methods as supported by multiple preclinical and clinical studies since they are engineered to target tumours. In this chapter, we discuss the modifications in viruses to make them oncolytic, types of strains commonly administered, mechanisms employed by viruses to specifically target and eradicate malignancy and progress achieved as reported in case studies (preclinical and clinical trials). OVs also face some unique challenges with respect to the malignancy being treated and the varied pathogen exposure of the patients, which is also highlighted here. Since pathogen exposure varies according to population dynamics worldwide, chances of generating a non-specific recall response to an OV cannot be negated. Lastly, the future perspectives and ongoing practises of combination therapies are discussed as they provide a leading edge over monotherapies in terms of tumour clearance, blocking metastasis and enhancing patient survival. Efforts undertaken to overcome current challenges are also highlighted.
Collapse
Affiliation(s)
- Sramona Kar
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
46
|
Novakova A, Morris SA, Vaiarelli L, Frank S. Manufacturing and Financial Evaluation of Peptide-Based Neoantigen Cancer Vaccines for Triple-Negative Breast Cancer in the United Kingdom: Opportunities and Challenges. Vaccines (Basel) 2025; 13:144. [PMID: 40006691 PMCID: PMC11860436 DOI: 10.3390/vaccines13020144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
This review evaluates the financial burden of current treatments for triple-negative breast cancer (TNBC) and projects potential financial scenarios to assess the feasibility of introducing a peptide-based neoantigen cancer vaccine (NCV) targeting the disease, using the UK as a healthcare system model. TNBC, the most aggressive breast cancer subtype, is associated with poor prognosis, worsened by the lack of personalised treatment options. Neoantigen cancer vaccine therapies present a personalised alternative with the potential to enhance T-cell responses independently of genetic factors, unlike approved immunotherapies for TNBC. Through a systematic literature review, the underlying science and manufacturing processes of NCVs are explored, the direct medical costs of existing TNBC treatments are enumerated, and two contrasting pricing scenarios for NCV clinical adoption are evaluated. The findings indicate that limited immunogenicity is the main scientific barrier to NCV clinical advancement, alongside production inefficiencies. Financial analysis shows that the UK spends approximately GBP 230 million annually on TNBC treatments, ranging from GBP 2200 to GBP 54,000 per patient. A best-case pricing model involving government-sponsored NCV therapy appears financially viable, while a worst-case, privately funded model exceeds the National Institute for Health and Care Excellence (NICE) cost thresholds. This study concludes that while NCVs show potential clinical benefits for TNBC, uncertainties about their standalone efficacy make their widespread adoption in the UK unlikely without further clinical research.
Collapse
Affiliation(s)
| | | | - Ludovica Vaiarelli
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK; (A.N.); (S.A.M.)
| | - Stefanie Frank
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK; (A.N.); (S.A.M.)
| |
Collapse
|
47
|
Yang Y, Hu Y, Yang Y, Liu Q, Zheng P, Yang Z, Duan B, He J, Li W, Li D, Zheng X, Wang M, Fu Y, Long Q, Ma Y. Tumor Vaccine Exploiting Membranes with Influenza Virus-Induced Immunogenic Cell Death to Decorate Polylactic Coglycolic Acid Nanoparticles. ACS NANO 2025; 19:3115-3134. [PMID: 39806805 DOI: 10.1021/acsnano.4c00654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Immunogenic cell death (ICD) of tumor cells, which is characterized by releasing immunostimulatory "find me" and "eat me" signals, expressing proinflammatory cytokines and providing personalized and broad-spectrum tumor antigens draws increasing attention in developing a tumor vaccine. In this study, we aimed to investigate whether the influenza virus (IAV) is efficient enough to induce ICD in tumor cells and an extra modification of IAV components such as hemeagglutinin (HA) will be helpful for the ICD-induced cells to elicit robust antitumor effects; in addition, to evaluate whether the membrane-engineering polylactic coglycolic acid nanoparticles (PLGA NPs) simulating ICD immune stimulation mechanisms hold the potential to be a promising vaccine candidate, a mouse melanoma cell line (B16-F10 cell) was infected with IAV rescued by the reverse genetic system, and the prepared cells and membrane-modified PLGA NPs were used separately to immunize the melanoma-bearing mice. IAV-infected tumor cells exhibit dying status, releasing high mobility group box-1 (HMGB1) and adenosine triphosphate (ATP), and exposing calreticulin (CRT), IAV hemeagglutinin (HA), and tumor antigens like tyrosinase-related protein 2 (TRP2). IAV-induced ICD cells enhance biomass-derived carbon (BMDCs) migration, antigen uptake, cross-presentation, and maturation in vitro. Furthermore, immunization with IAV-induced ICD cells effectively suppressed tumor growth in melanoma-bearing mice. The isolated cell membrane inherited the immunological characteristics from the ICD cells and elicited robust antitumor immune responses through decorating PLGA NPs loading with a tumor-specific helper T-cell peptide and supplemented with ATP in a hydrogel system. This study indicated a promising strategy for developing cell-based and personalized tumor vaccines through fully taking advantage of the immune stimulation mechanisms of ICD occurrence in tumor cells, IAV modification, and nanoscale delivery.
Collapse
Affiliation(s)
- Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Yongmao Hu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- Yunnan University, Kunming 650091, China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- Kunming Medical University, Kunming 650500, China
| | - Qingwen Liu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- Kunming Medical University, Kunming 650500, China
| | - Peng Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Zhongqian Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Biao Duan
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- Kunming Medical University, Kunming 650500, China
| | - Jinrong He
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Duo Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- Department of Acute Infectious Diseases Control and Prevention, Yunnan Provincial Center for Disease Control and Prevention, Kunming 650000, China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Mengzhen Wang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Yuting Fu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Qiong Long
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- State Key Laboratory of Respiratory Health and Multimorbidity, Kunming 650031, China
| |
Collapse
|
48
|
Xia Y, Huang C, Zhong M, Zhong H, Ruan R, Xiong J, Yao Y, Zhou J, Deng J. Targeting HGF/c-MET signaling to regulate the tumor microenvironment: Implications for counteracting tumor immune evasion. Cell Commun Signal 2025; 23:46. [PMID: 39856684 PMCID: PMC11762533 DOI: 10.1186/s12964-025-02033-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
The hepatocyte growth factor (HGF) along with its receptor (c-MET) are crucial in preserving standard cellular physiological activities, and imbalances in the c-MET signaling pathway can lead to the development and advancement of tumors. It has been extensively demonstrated that immune checkpoint inhibitors (ICIs) can result in prolonged remission in certain patients. Nevertheless, numerous preclinical studies have shown that MET imbalance hinders the effectiveness of anti-PD-1/PD-L1 treatments through various mechanisms. Consequently, clarifying the link between the c-MET signaling pathway and the tumor microenvironment (TME), as well as uncovering the effects of anti-MET treatment on ICI therapy, is crucial for enhancing the outlook for tumor patients. In this review, we examine the impact of abnormal activation of the HGF/c-MET signaling pathway on the control of the TME and the processes governing PD-L1 expression in cancer cells. The review thoroughly examines both clinical and practical evidence regarding the use of c-MET inhibitors alongside PD-1/PD-L1 inhibitors, emphasizing that focusing on c-MET with immunotherapy enhances the effectiveness of treating MET tumors exhibiting elevated PD-L1 expression.
Collapse
Affiliation(s)
- Yang Xia
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Chunye Huang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Min Zhong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Hongguang Zhong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Ruiwen Ruan
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Yangyang Yao
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| | - Jing Zhou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| |
Collapse
|
49
|
Yang Y, Wang J, Zhong Y, Tian M, Zhang H. Advances in Radionuclide-Labeled Biological Carriers for Tumor Imaging and Treatment. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4316-4336. [PMID: 39792777 DOI: 10.1021/acsami.4c19059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Biological carriers have emerged as significant tools to deliver radionuclides in nuclear medicine, providing a meaningful perspective for tumor imaging and treatment. Various radionuclide-labeled biological carriers have been developed to meet the needs of biomedical applications. This review introduces the principles of radionuclide-mediated imaging and therapy and the selected criteria of them, as well as a comprehensive description of the characteristics and functions of representative biological carriers including bacteria, cells, viruses, and their biological derivatives, emphasizing the labeled strategies of biological carriers combined with radionuclides. Subsequently, we in-depth introduce the application of radionuclide-labeled biological carriers in tumor imaging and treatment, including the imaging of the behaviors of biological carriers in vivo and tumor metastasis and the tumor treatment by radionuclide therapy, plus other strategies and radiation-induced photodynamic therapy. Finally, the challenges and prospects of radionuclide-labeled biological carriers are discussed to improve the shortcomings of this innovative platform and promote clinical transformation in the field of medical imaging.
Collapse
Affiliation(s)
- Yaozhang Yang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
| | - Jing Wang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
| | - Yan Zhong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
- Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang 310014, China
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
50
|
Peng Z, Kalim M, Lu Y. Improving systemic delivery of oncolytic virus by cellular carriers. Cancer Biol Med 2025; 21:j.issn.2095-3941.2024.0390. [PMID: 39831754 PMCID: PMC11745088 DOI: 10.20892/j.issn.2095-3941.2024.0390] [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: 09/13/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025] Open
Abstract
Oncolytic virotherapy (OVT) is a promising option for cancer treatment. OVT involves selective oncolytic virus (OV) replication within cancer cells, which triggers anti-tumor responses and immunostimulation. Despite promising potential, OVT faces critical challenges, including insufficient tumor-specific targeting, which results in limited tumor penetration and variability in therapeutic efficacy. These challenges are particularly pronounced in solid tumors with complex microenvironments and heterogeneous vascularization. A comprehensive research program is currently underway to develop and refine innovative delivery methods to address these issues to enhance OVT precision and efficacy. A principal area of investigation is the utilization of cellular carriers to enhance the delivery and distribution of OVs within tumor microenvironments, thereby optimizing immune system activation and maximizing anti-tumor effects. This review offers a comprehensive overview of the current strategies that are being used to enhance the delivery of OVs via cellular carriers with the goal of improving the clinical impact of OVT in cancer therapy.
Collapse
Affiliation(s)
- Ziyi Peng
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Muhammad Kalim
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Yong Lu
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| |
Collapse
|