1
|
Gao C, Luo R, Kwong CHT, Liu J, Tang M, Xie B, Duan T, Wang R. Cancer vaccine from intracellularly gelated tumor cells functionalized with CD47 blockage and damage-associated molecular pattern exposure. Cell Rep Med 2025; 6:102092. [PMID: 40345180 DOI: 10.1016/j.xcrm.2025.102092] [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: 09/30/2024] [Revised: 02/04/2025] [Accepted: 03/31/2025] [Indexed: 05/11/2025]
Abstract
The effectiveness of whole tumor cell vaccines prepared by traditional inactivation methodology is often hindered by insufficient immunogenicity. Here, we report development of a cancer vaccine through the intracellular gelation of tumor cells, combined with CD47 blockade and damage-associated molecular pattern (DAMP) exposure, for effective tumor prevention and treatment. Intracellular hydrogelation preserves the morphology and antigenicity of tumor cells. CD47 blockade and DAMP exposure synergistically enhance the "eat me" signals and inhibit the "don't eat me" signals on tumor cells, significantly improving their immunogenicity. In the context of tumor prevention and treatment of pre-existing tumors, this vaccine polarizes CD4+ T cells toward a TH1 phenotype, reduces regulatory T cells and T cell exhaustion, and elicits a robust tumor-antigen-specific T cell response. When combined with an immune checkpoint inhibitor, this vaccine demonstrates enhanced efficacy in eradicating established tumors. The successful application of this vaccine using ascites and subcutaneous tumor cells supports the feasibility of developing personalized whole tumor cell vaccines for diverse tumor types.
Collapse
Affiliation(s)
- Cheng Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China; School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Ruifeng Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China
| | - Cheryl H T Kwong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China
| | - Jinwei Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China
| | - Mian Tang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China
| | - Beibei Xie
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China.
| | - Tianshun Duan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China.
| |
Collapse
|
2
|
Zhang P, Cui G, Wang T, Zhao X, Wang X, Ye R, Liu T, Zheng Y, Zhong Z. In situ therapeutic vaccines for leukemia by chemo-nanoadjuvant therapy. J Control Release 2025; 383:113851. [PMID: 40389164 DOI: 10.1016/j.jconrel.2025.113851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/25/2025] [Accepted: 05/14/2025] [Indexed: 05/21/2025]
Abstract
Therapeutic vaccines introduce a potentially ultimate cure for cancers including leukemia. The personalized vaccines relying on neoantigens though exhibiting clinical benefits are afflicted by long and delicate manufacture procedure, high cost, and possibly incomplete coverage of heterogeneous tumor cells. Here, we report a facile strategy to generate potent in situ therapeutic vaccines, which effectively eliminate leukemia and induce long-term anti-leukemia immunity, by homoharringtonine-nano-dual-adjuvant (HHT-NDA) therapy. HHT effectively kills leukemia cells and generates abundant tumor antigens via inducing immunogenic cell death. NDAs cooperatively promote the maturation and antigen-presentation of dendritic cells by activating both nucleotide-binding oligomerization domain-containing protein 2 and toll-like receptor 9. The HHT-NDA treatment of murine MLL-AF9 acute myeloid leukemia model leads to 57-71 % complete regression and 100 % protection from rechallenge, in accordance with expansion of memory CD8+ T cells. This standard-of-care chemotherapy in tandem with nano-dual-adjuvant offers a novel strategy to generate in situ therapeutic vaccines for leukemia.
Collapse
Affiliation(s)
- Peng Zhang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
| | - Guanhong Cui
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
| | - Tanzhen Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215000, PR China
| | - Xiaofei Zhao
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
| | - Xinyue Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, PR China
| | - Ruonan Ye
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
| | - Tianhui Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215000, PR China.
| | - Yiran Zheng
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, PR China.
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
3
|
Ou X, Zhou J, Xie HY, Nie W. Fusogenic Lipid Nanovesicles as Multifunctional Immunomodulatory Platforms for Precision Solid Tumor Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2503134. [PMID: 40351077 DOI: 10.1002/smll.202503134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/23/2025] [Indexed: 05/14/2025]
Abstract
Although immunotherapy demonstrates considerable prospect in overcoming solid tumors, its clinical efficacy is limited by several factors, such as poor tumor immunogenicity, inadequate immune activation, and immunosuppressive tumor microenvironment (TME). To overcome these challenges, a versatile and universal immune modulation platform should be developed, and lipid nanovesicles with membrane fusion capabilities (LNV-Fs) have attracted great attention for this purpose. By mimicking natural membrane fusion processes, LNV-Fs enable the precise presentation of immunogenic components on tumor cell membranes, effectively activating anti-tumor immune surveillance. Similarly, LNV-Fs can equip multiple functionalities on autologous and adoptive effector cells for enhanced cell therapies. Additionally, LNV-Fs function as vaccines that elicit robust autologous anti-tumor immunity while promoting long-term immune memory. Furthermore, different LNV-Fs with powerful ability in reprogramming TME have been reported. Given the recent advancements and the absence of comprehensive reviews on this topic, a comprehensive analysis of LNV-F systems, including their structural classifications, membrane fusion mechanisms, and recent applications in cancer immunotherapy is provided. Furthermore, the future prospects of LNV-Fs, with particular emphasis on artificial intelligence-assisted design are explored. This review is intended to engage researchers from diverse interdisciplinary fields and provide valuable insights for advancing precision immunotherapy.
Collapse
Affiliation(s)
- Xu Ou
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Jiaxin Zhou
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Hai-Yan Xie
- Chemical Biology Center Peking University State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Beijing, 100191, P. R. China
| | - Weidong Nie
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| |
Collapse
|
4
|
Huang P, Liu Y, Zhao C, Wang C, Wang L, Luo M, Wang W, Shan W, Liu X, Li B, Wang Z, Deng H, Chen X. Permanent Efferocytosis Prevention by Terminating MerTK Recycle on Tumor-Associated Macrophages for Cancer Immunotherapy. J Am Chem Soc 2025; 147:15901-15914. [PMID: 40294287 DOI: 10.1021/jacs.5c05640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Efferocytosis of apoptotic tumor cells by tumor-associated macrophages mediated through the phosphatidylserine (PtdSer)/MER proto-oncogene tyrosine kinase (MerTK) axis can exacerbate tumor immunosuppression, and conversely, prevention of efferocytosis via blocking PtdSer-MerTK association using prevalent antibodies represents a promising strategy for reversing tumor immunosuppression and boosting antitumor immunity. However, it remains unclear whether the antibody blockade can induce durable efferocytosis prevention and achieve sustained tumor growth inhibition. Here, we have shown that utilizing PtdSer and MerTK antibodies induced only a transient rather than a persistent efferocytosis prevention effect, and little enhancement was observed even after improving antibody enrichment in tumor sites. Further mechanistic studies suggested that degradation of anti-MerTK antibody and recycling of the MerTK receptor to the cell membrane would compromise the therapeutic benefits of antibody blockade. Based on these findings, we developed a CRISPR/Cas9 gene editing system deployed using Cas9 mRNA and MerTK sgRNA to permanently knock out MerTK, which achieved durable efferocytosis prevention, elicited persistent in situ vaccination immune responses via enhancing X-ray irradiation-induced immunogenic cell death, and led to sustained tumor suppression effects together with anti-PtdSer antibody and X-ray irradiation treatment in multiple B16 melanoma tumor models. Our findings provide a reliable gene-editing-mediated strategy for long-term modulating MerTK homeostasis and overcoming MerTK-dependent cancer immune evasion, generating adaptive antitumor immune responses for sustained cancer immunotherapy.
Collapse
Affiliation(s)
- Pei Huang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| | - Yiwen Liu
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of Trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Caiyan Zhao
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of Trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Changrong Wang
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of Trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Lirong Wang
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of Trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Meng Luo
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of Trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Weipeng Wang
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of Trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Wenbo Shan
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of Trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Xiaoqing Liu
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of Trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Bingyu Li
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| | - Zhongliang Wang
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of Trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Hongzhang Deng
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of Trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
- Department of Chemical and Biomolecular Engineering, College of Design and Engineering, National University of Singapore, Singapore 117575, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117575, Singapore
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore 117544, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 138667, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore 138673, Singapore
| |
Collapse
|
5
|
Yang QC, Wang YY, Wang S, Song A, Wang WD, Zhang L, Sun ZJ. Engineered bacterial membrane biomimetic covalent organic framework as nano-immunopotentiator for cancer immunotherapy. Bioact Mater 2025; 47:283-294. [PMID: 39925708 PMCID: PMC11803166 DOI: 10.1016/j.bioactmat.2025.01.018] [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: 09/23/2024] [Revised: 12/27/2024] [Accepted: 01/15/2025] [Indexed: 02/11/2025] Open
Abstract
The cellular uptake and tissue dispersion efficiency of nanomedicines are crucial for realizing their biological functionality. As a cutting-edge category of nanomedicine, covalent organic frameworks (COFs)-based photosensitizers, have been extensively employed in cancer phototherapy in recent years. However, the inherent aggregation tendency of COFs hinders their uptake by tumor cells and dispersion within tumor tissues, thereby limiting their therapeutic efficacy. In this study, we employed Fusobacterium nucleatum (F.n.), a prevalent intratumoral bacterium, to construct a bacterium membrane-wrapped COF, COF-306@FM, which is readily taken up by cancer cells and uniformly dispersed within tumor tissues. Meanwhile, the F.n. membrane can also serve as an immune adjuvant to warm up the "cold" tumor immune microenvironment by enhancing the CD8+ T and B cells infiltration, and inducing the formation of tumor-located tertiary lymphoid structures. Consequently, the response rate of αPD-L1 immunotherapy was drastically promoted to efficiently prevent tumor metastasis and recurrence, causing 84.6 % distant tumor inhibition and complete suppression of tumor metastasis. In summary, this innovative approach not only enhances the therapeutic potential of COFs but also opens up new avenues for integrating microbial and nanotechnological strategies in cancer treatment.
Collapse
Affiliation(s)
- Qi-Chao Yang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Yuan-Yuan Wang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Shuo Wang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - An Song
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Wen-Da Wang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Liang Zhang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Zhi-Jun Sun
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
6
|
Liu N, Wang X, Wang Z, Kan Y, Fang Y, Gao J, Kong X, Wang J. Nanomaterials-driven in situ vaccination: a novel frontier in tumor immunotherapy. J Hematol Oncol 2025; 18:45. [PMID: 40247328 PMCID: PMC12007348 DOI: 10.1186/s13045-025-01692-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
In situ vaccination (ISV) has emerged as a promising strategy in cancer immunotherapy, offering a targeted approach that uses the tumor microenvironment (TME) to stimulate an immune response directly at the tumor site. This method minimizes systemic exposure while maintaining therapeutic efficacy and enhancing safety. Recent advances in nanotechnology have enabled new approaches to ISV by utilizing nanomaterials with unique properties, including enhanced permeability, retention, and controlled drug release. ISV employing nanomaterials can induce immunogenic cell death and reverse the immunosuppressive and hypoxic TME, thereby converting a "cold" tumor into a "hot" tumor and facilitating a more robust immune response. This review examines the mechanisms through which nanomaterials-based ISV enhances anti-tumor immunity, summarizes clinical applications of these strategies, and evaluates its capacity to serve as a neoadjuvant therapy for eliminating micrometastases in early-stage cancer patients. Challenges associated with the clinical translation of nanomaterials-based ISV, including nanomaterial metabolism, optimization of treatment protocols, and integration with other therapies such as radiotherapy, chemotherapy, and photothermal therapy, are also discussed. Advances in nanotechnology and immunotherapy continue to expand the possible applications of ISV, potentially leading to improved outcomes across a broad range of cancer types.
Collapse
Affiliation(s)
- Naimeng Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiangyu Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhongzhao Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yonemori Kan
- Department of Medical Oncology, National Cancer Center Hospital (NCCH), 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jidong Gao
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518127, China.
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
7
|
Liang Y, Zhang J, Wang J, Yang Y, Tan X, Li S, Guo Z, Zhang Z, Liu J, Shi J, Zhang K. Restoring Tumor Cell Immunogenicity Through Ion-Assisted p53 mRNA Domestication for Enhanced In Situ Cancer Vaccination Effect. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500825. [PMID: 39965083 PMCID: PMC11984859 DOI: 10.1002/advs.202500825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 01/28/2025] [Indexed: 02/20/2025]
Abstract
The efficacy of in situ cancer vaccines (ISCVs) is hindered by the poor immunogenicity of tumor cells. Here, PRIZE, a P53-repair nanosystem based on a virus-mimicking nanostructure to deliver p53 mRNA and Zn (II) into tumor cells, domesticating tumor cells by restoring intracellular P53 levels to bolster their immunogenicity, is designed. PRIZE ensures precise delivery to tumor sites, stabilizes p53 mRNA with its biomineralized structure, and extends the half-life of P53. This research highlights that PRIZE can efficiently repair P53 abnormalities in 4T1 (P53-deficient) and MC38 (P53-mutant) cells, subsequently upregulating the expression of major histocompatibility complex (MHC) class I molecules and the surface co-stimulatory molecule CD80 on tumor cells, enhancing antigen presentation and transforming tumor cells into in situ antigen reservoirs. The co-delivered photothermal agent (ICG) can trigger immunogenic cell death under laser irradiation, effectively releasing tumor-associated antigens, and inducing the formation of ISCVs. Importantly, in P53 abnormal tumor mouse models, the induced ISCVs initiate the cancer immune cycle (CIC), demonstrating outstanding tumoricidal immunity and effectively thwarting tumor metastasis and postoperative recurrence, which provides valuable insights for advancing personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Yan Liang
- School of Pharmaceutical SciencesTianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450001P. R. China
| | - Jingge Zhang
- School of Pharmaceutical SciencesTianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450001P. R. China
| | - Jinjin Wang
- School of Pharmaceutical SciencesTianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450001P. R. China
| | - Yuhe Yang
- School of Pharmaceutical SciencesTianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450001P. R. China
| | - Xinyu Tan
- School of Pharmaceutical SciencesTianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450001P. R. China
| | - Shuguang Li
- School of Pharmaceutical SciencesTianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450001P. R. China
| | - Zhenzhen Guo
- School of Pharmaceutical SciencesTianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450001P. R. China
| | - Zhenzhong Zhang
- School of Pharmaceutical SciencesTianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450001P. R. China
| | - Junjie Liu
- School of Pharmaceutical SciencesTianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450001P. R. China
| | - Jinjin Shi
- School of Pharmaceutical SciencesTianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450001P. R. China
| | - Kaixiang Zhang
- School of Pharmaceutical SciencesTianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450001P. R. China
- Beijing Life Science AcademyBeijing102209P. R. China
| |
Collapse
|
8
|
Li M, Liu Y, Liu F, Chen Q, Xu L, Cheng Z, Tan Y, Liu Z. Extracellular Vesicle-Based Antitumor Nanomedicines. Adv Healthc Mater 2025; 14:e2403903. [PMID: 39935134 DOI: 10.1002/adhm.202403903] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/30/2024] [Indexed: 02/13/2025]
Abstract
Extracellular vesicles (EVs) have emerged as promising bioactive carriers for delivering therapeutic agents, including nucleic acids, proteins, and small-molecule drugs, owing to their excellent physicochemical stability and biocompatibility. However, comprehensive reviews on the various types of EV-based nanomedicines for cancer therapy remain scarce. This review explores the potential of EVs as antitumor nanomedicines. Methods for EV extraction, drug loading, and engineering modifications are systematically examined, and the strengths and limitations of these technical approaches are critically assessed. Additionally, key strategies for developing EV-based antitumor therapies are highlighted. Finally, the opportunities and challenges associated with advancing EVs toward clinical translation are discussed. With the integration of multiple disciplines, robust EV-based therapeutic platforms are expected to be manufactured to provide more personalized and effective solutions for oncology patients.
Collapse
Affiliation(s)
- Mingfeng Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Fei Liu
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Qiwen Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Lishang Xu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Zhongyu Cheng
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yifu Tan
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
- Molecular Imaging Research Center of Central South University, Changsha, Hunan, 410008, P. R. China
| |
Collapse
|
9
|
Xiao W, Gao M, Mo B, Huang X, Du Z, Wang S, Chen J, Luo S, Xing H. Endoplasmic Reticulum-Targeted Phototherapy Remodels the Tumor Immunopeptidome to Enhance Immunogenic Cell Death and Adaptive Anti-Tumor Immunity. Pharmaceuticals (Basel) 2025; 18:491. [PMID: 40283929 PMCID: PMC12030737 DOI: 10.3390/ph18040491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Endoplasmic reticulum (ER)-targeted phototherapy has emerged as a promising approach to amplify ER stress, induce immunogenic cell death (ICD), and enhance anti-tumor immunity. However, its impact on the antigenicity of dying tumor cells remains poorly understood. Methods: Laser activation of the ER-targeted photosensitizer ER-Cy-poNO2 was performed to investigate its effects on tumor cell antigenicity. Transcriptomic analysis was carried out to assess gene expression changes. Immunopeptidomics profiling was used to identify high-affinity major histocompatibility complex class I (MHC-I) ligands. In vitro functional studies were conducted to evaluate dendritic cell maturation and T lymphocyte activation, while in vivo experiments were performed by combining the identified peptide with poly IC to evaluate anti-tumor immunity. Results: Laser activation of ER-Cy-poNO2 significantly remodeled the antigenic landscape of 4T-1 tumor cells, enhancing their immunogenicity. Transcriptomic analysis revealed upregulation of antigen processing and presentation pathways. Immunopeptidomics profiling identified multiple high-affinity MHC-I ligands, with IF4G3986-994 (QGPKTIEQI) showing exceptional immunogenicity. In vitro, IF4G3986-994 promoted dendritic cell maturation and enhanced T lymphocytes activation. In vivo, the combination of IF4G3986-994 with poly IC elicited robust anti-tumor immunity, characterized by increased CD8+ T lymphocytes infiltration, reduced regulatory T cells (Tregs) in the tumor microenvironment, elevated systemic Interferon-gamma (IFN-γ) levels, and significant tumor growth inhibition without systemic toxicity. Conclusions: These findings establish a mechanistic link between ER stress-driven ICD, immunopeptidome remodeling, and adaptive immune activation, highlighting the potential of ER-targeted phototherapy as a platform for identifying immunogenic peptides and advancing peptide-based cancer vaccines.
Collapse
Affiliation(s)
- Weidong Xiao
- Department of Pharmacy, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Mingquan Gao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Banghui Mo
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xie Huang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zaizhi Du
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shufeng Wang
- Department of Immunology, College of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jianhong Chen
- Department of Pharmacy, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Shenglin Luo
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Haiyan Xing
- Department of Pharmacy, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| |
Collapse
|
10
|
Li Y, Xu Y, Su W, Xu J, Ye Z, Wang Z, Liu Q, Chen F. Exploring the immuno-nano nexus: A paradigm shift in tumor vaccines. Biomed Pharmacother 2025; 184:117897. [PMID: 39921945 DOI: 10.1016/j.biopha.2025.117897] [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: 10/31/2024] [Revised: 01/17/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025] Open
Abstract
Tumor vaccines have become a crucial strategy in cancer immunotherapy. Challenges of traditional tumor vaccines include inadequate immune activation and low efficacy of antigen delivery. Nanoparticles, with their tunable properties and versatile functionalities, have redefined the landscape of tumor vaccine design. In this review, we outline the multifaceted roles of nanoparticles in tumor vaccines, ranging from their capacity as delivery vehicles to their function as immunomodulatory adjuvants capable of stimulating anti-tumor immunity. We discuss how this innovative approach significantly boosts antigen presentation by leveraging tailored nanoparticles that facilitate efficient uptake by antigen-presenting cells. These nanoparticles have been meticulously designed to overcome biological barriers, ensuring optimal delivery to lymph nodes and effective interaction with the immune system. Overall, this review highlights the transformative power of nanotechnology in redefining the principles of tumor vaccines. The intent is to inform more efficacious and precise cancer immunotherapies. The integration of these advanced nanotechnological strategies should unlock new frontiers in tumor vaccine development, enhancing their potential to elicit robust and durable anti-tumor immunity.
Collapse
Affiliation(s)
- Yuanyuan Li
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yike Xu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Wenwen Su
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Jia Xu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zifei Ye
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zhuoyi Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Qihui Liu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| | - Fangfang Chen
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
11
|
Lin D, Lv W, Qian M, Jiang G, Lin X, Gantulga D, Wang Y. Engineering cell membrane-camouflaged COF-based nanosatellite for enhanced tumor-targeted photothermal chemoimmunotherapy. Biomaterials 2025; 314:122869. [PMID: 39427431 DOI: 10.1016/j.biomaterials.2024.122869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/13/2024] [Accepted: 09/29/2024] [Indexed: 10/22/2024]
Abstract
Dendritic cells (DCs) activation is crucial for regulating the antitumor immune response. However, the tumor's immunosuppressive environment significantly impedes antigen presentation and DCs maturation, thereby limiting the effectiveness of cancer immunotherapy. To address this challenge, we developed tumor cell membrane-coated covalent organic framework (COF) nanoparticles, loaded with mannose-modified gold nanoparticles and doxorubicin (Dox). This created a cell membrane-camouflaged COF-based nanosatellite designed to enhance tumor-targeted chemoimmunotherapy. The nanosatellite exhibits distinct photothermal properties and releases Dox in a pH-sensitive manner, targeting tumor cells to induce immunogenic cell death (ICD) and expose a wealth of antigens. Crucially, the COF structure is selectively degraded to release mannose-modified gold nanoparticles in the acidic environment. These nanoparticles capture antigens from the ICD and efficiently transport them to lymph nodes rich in DCs, facilitated by mannose receptor mediation. As a result, antigens are effectively presented to DCs, activating the immune response, significantly hindering tumor growth and lung metastasis in mice, and extending survival. This study pioneered innovative nano-preparations aimed at enhancing tumor immunotherapy.
Collapse
Affiliation(s)
- Dan Lin
- Center for Advanced Low-dimension Materials, State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China; College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Wenxin Lv
- Center for Advanced Low-dimension Materials, State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Min Qian
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Guangwei Jiang
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Xiaojun Lin
- Center for Advanced Low-dimension Materials, State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Darambazar Gantulga
- Department of Biology, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar, 14210, Mongolia
| | - Yi Wang
- Center for Advanced Low-dimension Materials, State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China.
| |
Collapse
|
12
|
Huang X, Ge W, Li S, Huang R, Wang F. Transferrin-Based Bismuth Nanoparticles for Radiotherapy with Immunomodulation Against Orthotopic Glioma. Adv Healthc Mater 2025; 14:e2404144. [PMID: 39797464 DOI: 10.1002/adhm.202404144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/31/2024] [Indexed: 01/13/2025]
Abstract
Modern radiotherapy frequently employs radiosensitizers for radiation dose deposition and triggers an immunomodulatory effect to enhance tumor destruction. However, developing glioma-targeted sensitizers remains challenging due to the blood-brain barrier (BBB) and multicomponent instability. This study aims to green-synthesize transferrin-bismuth nanoparticles (TBNPs) as biosafe radiosensitizers to enhance X-ray absorption by tumors and stimulate the immune response for glioma therapy. The proposed protein-based strategy provides TBNPs with BBB-crossing ability and prevents off-target toxicity. Cellular experiments following 4 Gy of X-ray irradiation reveal that TBNPs increase DNA damage in glioma cells and trigger immunomodulation, thereby inducing immunogenic cell death. Furthermore, TBNPs effectively inhibit tumor growth through synergistic radiotherapy and immunotherapy in an orthotopic glioma mouse model. The findings highlight TBNPs as promising radiosensitizers for effective and biosafe radiotherapy with immunomodulation.
Collapse
Affiliation(s)
- Xiaoyu Huang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Wei Ge
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Shuxian Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Ruofan Huang
- Department of Oncology, Huashan Hospital, Fudan University, Shanghai, 200240, P. R. China
| | - Fu Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
13
|
Zhang D, Lu B, Ma Q, Xu W, Zhang Q, Xiao Z, Li Y, Chen R, Wang AJ. Identification of a novel immunogenic cell death-related classifier to predict prognosis and optimize precision treatment in hepatocellular carcinoma. Heliyon 2025; 11:e41380. [PMID: 39897773 PMCID: PMC11786863 DOI: 10.1016/j.heliyon.2024.e41380] [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: 03/23/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 02/04/2025] Open
Abstract
Accumulating studies have highlighted the biological significance of immunogenic cell death (ICD) in cancer immunity. However, the influence of ICD on tumor microenvironment (TME) formation and immune response in Hepatocellular carcinoma (HCC) remains largely unexplored. In this study, we systematically analyzed the mRNA profiles of ICD-related genes in 1847 HCC patients and identified three molecular subtypes with significantly different immune features and prognostic stratification. A reliable risk model named ICD score was constructed via machine learning algorithms to assess the immunological status, therapeutic responses, and clinical outcomes of individual HCC patients. High ICD score indicated an immune-excluded TME phenotype, with lower anticancer immunity and shorter survival time. In contrast, low ICD score corresponded to abundant immune cell infiltration, high sensitivity to immunotherapy and a positive prognosis, indicating an "immune-hot" phenotype. Pan-cancer analysis further validated a negative association between ICD score and the immune cell infiltration levels. In conclusion, our findings revealed that the ICD score could serve as a robust prognostic biomarker to predict the benefits of immunotherapy and optimize the clinical decision-making of HCC patients.
Collapse
Affiliation(s)
- Dongjing Zhang
- Department of Gastroenterology and Hepatology, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Bingyun Lu
- Department of Gastroenterology and Hepatology, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Qianqian Ma
- Department of Infectious Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Wen Xu
- Department of Gastroenterology and Hepatology, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Qi Zhang
- Department of Gastroenterology and Hepatology, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Zhiqi Xiao
- Department of Gastroenterology and Hepatology, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Yuanheng Li
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Ren Chen
- Department of Infectious Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - An-jiang Wang
- Department of Gastroenterology and Hepatology, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| |
Collapse
|
14
|
Chen Y, Cen Y, Li XX, Ou XC, Chen XY, Yu BX, Yan MY, Shao ZC, Wang TX, Guo N, Yu R, Li SY. Chimeric Peptide-Engineered Polyprodrug Enhances Cytotoxic T Cell Response by Inducing Immunogenic Cell Death and Upregulating Major Histocompatibility Complex Class I. ACS NANO 2025; 19:837-851. [PMID: 39731541 DOI: 10.1021/acsnano.4c12197] [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: 12/30/2024]
Abstract
Tumor-specific cytotoxic T cell immunity is critically dependent on effective antigen presentation and sustained signal transduction. However, this immune response is frequently compromised by the inherently low immunogenicity of breast cancer and the deficiency in major histocompatibility complex class I (MHC-I) expression. Herein, a chimeric peptide-engineered stoichiometric polyprodrug (PDPP) is fabricated to potentiate the cytotoxic T cell response, characterized by a high drug loading capacity and precise stoichiometric drug ratio, of which the immunogenic cell death (ICD) inducer of protoporphyrin IX (PpIX) and the epigenetic drug of decitabine (DAC) are condensed into a polyprodrug called PpIX-DAC. Furthermore, programmed death ligand 1 (PD-L1) targeting peptide sequence (CVRARTR) is conjugated onto DSPE-PEG2000-Mal for encapsulation of PpIX-DAC, thereby enhancing breast cancer-targeted drug delivery. PDPP exerts its antitumor effects through photodynamic therapy (PDT), ablating breast cancer cells while concurrently inducing the release of damage-associated molecular patterns (DAMPs) to boost tumor immunogenicity. Additionally, PDPP can upregulate MHC-I expression via epigenetic modulation, synergistically augmenting the cytotoxic T cell response together with a PD-L1 blockade. In short, PDPP induces a robust antitumor T cell immunity, causing effective eradication of primary and metastatic breast cancer. This study may inspire the development of stoichiometric nanomedicine for clinical translation.
Collapse
Affiliation(s)
- Ying Chen
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yi Cen
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xin-Xuan Li
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiao-Cheng Ou
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xia-Yun Chen
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Bai-Xue Yu
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Meng-Yi Yan
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhou-Chuan Shao
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Ting-Xin Wang
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Ning Guo
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Rui Yu
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Shi-Ying Li
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
15
|
Ge M, Jiang F, Lin H. Nanocatalytic medicine enabled next-generation therapeutics for bacterial infections. Mater Today Bio 2024; 29:101255. [PMID: 39381264 PMCID: PMC11459013 DOI: 10.1016/j.mtbio.2024.101255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/08/2024] [Accepted: 09/14/2024] [Indexed: 10/10/2024] Open
Abstract
The rapid rise of antibiotic-resistant strains and the persistence of biofilm-associated infections have significantly challenged global public health. Unfortunately, current clinical high-dose antibiotic regimens and combination therapies often fail to completely eradicate these infections, which can lead to adverse side effects and further drug resistance. Amidst this challenge, however, the burgeoning development in nanotechnology and nanomaterials brings hopes. This review provides a comprehensive summary of recent advancements in nanomaterials for treating bacterial infections. Firstly, the research progress of catalytic therapies in the field of antimicrobials is comprehensively discussed. Thereafter, we systematically discuss the strategies of nanomaterials for anti-bacterial infection therapies, including endogenous response catalytic therapy, exogenous stimulation catalytic therapy, and catalytic immunotherapy, in order to elucidate the mechanism of nanocatalytic anti-infections. Based on the current state of the art, we conclude with insights on the remaining challenges and future prospects in this rapidly emerging field.
Collapse
Affiliation(s)
- Min Ge
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Feng Jiang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Han Lin
- Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai, 200050, China
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200331, China
| |
Collapse
|
16
|
An X, Chen Z, Luo Y, Yang P, Yang Z, Ji T, Chi Y, Wang S, Zhang R, Wang Z, Li J. Light-Activated In Situ Vaccine with Enhanced Cytotoxic T Lymphocyte Infiltration and Function for Potent Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403158. [PMID: 38953329 PMCID: PMC11434106 DOI: 10.1002/advs.202403158] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/16/2024] [Indexed: 07/04/2024]
Abstract
In situ cancer vaccination is an attractive strategy that stimulates protective antitumor immunity. Cytotoxic T lymphocytes (CTLs) are major mediators of the adaptive immune defenses, with critical roles in antitumor immune response and establishing immune memory, and are consequently extremely important for in situ vaccines to generate systemic and lasting antitumor efficacy. However, the dense extracellular matrix and hypoxia in solid tumors severely impede the infiltration and function of CTLs, ultimately compromising the efficacy of in situ cancer vaccines. To address this issue, a robust in situ cancer vaccine, Au@MnO2 nanoparticles (AMOPs), based on a gold nanoparticle core coated with a manganese dioxide shell is developed. The AMOPs modulated the unfavorable tumor microenvironment (TME) to restore CTLs infiltration and function and efficiently induced immunogenic cell death. The Mn2+-mediated stimulator of the interferon genes pathway can be activated to further augment the therapeutic efficacy of the AMOPs. Thus, the AMOPs vaccine successfully elicited long-lasting antitumor immunity to considerably inhibit primary, recurrent, and metastatic tumors. This study not only highlights the importance of revitalizing CTLs efficacy against solid tumors but also makes progress toward overcoming TME barriers for sustained antitumor immunity.
Collapse
Affiliation(s)
- Xian An
- Medical School of Chinese PLA & Department of OncologyChinese PLA General HospitalBeijing100193P. R. China
| | - Zhuang Chen
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & Neuro‐imagingMinistry of EducationSchool of Life Science and TechnologyXidian UniversityXi'anShaanxi710126P. R. China
| | - Yi Luo
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & Neuro‐imagingMinistry of EducationSchool of Life Science and TechnologyXidian UniversityXi'anShaanxi710126P. R. China
| | - Peng Yang
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & Neuro‐imagingMinistry of EducationSchool of Life Science and TechnologyXidian UniversityXi'anShaanxi710126P. R. China
| | - Zuo Yang
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & Neuro‐imagingMinistry of EducationSchool of Life Science and TechnologyXidian UniversityXi'anShaanxi710126P. R. China
| | - Tiannan Ji
- Department of EmergencyThe Fifth Medical Center of PLA General HospitalBeijing100193P. R. China
| | - Yajing Chi
- School of MedicineNankai UniversityTianjin300071P. R. China
| | - Shuyuan Wang
- Medical School of Chinese PLA & Department of OncologyChinese PLA General HospitalBeijing100193P. R. China
- School of MedicineNankai UniversityTianjin300071P. R. China
| | - Ruili Zhang
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & Neuro‐imagingMinistry of EducationSchool of Life Science and TechnologyXidian UniversityXi'anShaanxi710126P. R. China
| | - Zhongliang Wang
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & Neuro‐imagingMinistry of EducationSchool of Life Science and TechnologyXidian UniversityXi'anShaanxi710126P. R. China
| | - Jianxiong Li
- Medical School of Chinese PLA & Department of OncologyChinese PLA General HospitalBeijing100193P. R. China
| |
Collapse
|
17
|
Shen N, Li T, Li B, Wang Y, Liu H, Guo C, Chen X, Li J. Dual-functional mediators of high-entropy Prussian blue analogues for lithiophilicity and sulfiphilicity in Li-S batteries. NANOSCALE 2024; 16:7634-7644. [PMID: 38526018 DOI: 10.1039/d4nr00571f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
Lithium-sulfur (Li-S) batteries are considered promising next-generation energy storage systems due to their high energy density (2600 W h kg-1) and cost-effectiveness. However, the shuttle effect of lithium polysulfides in sulfur cathodes and uncontrollable Li dendrite growth in Li metal anodes significantly impede the practical application of Li-S batteries. In this study, we address these challenges by employing a high-entropy Prussian blue analogue Mn0.4Co0.4Ni0.4Cu0.4Zn0.4[Fe(CN)6]2 (HE-PBA) composite containing multiple metal ions as a dual-functional mediator for Li-S batteries. Specifically, the HE-PBA composite provides abundant metal active sites that efficiently chemisorb lithium polysulfides (LiPSs) to facilitate fast redox conversion kinetics of LiPSs. In Li metal anodes, the exceptional lithiophilicity of the HE-PBA ensures a homogeneous Li ion flux, resulting in uniform Li deposition while mitigating the growth of Li dendrites. As a result, our work demonstrates outstanding long-term cycling performance with a decay rate of only 0.05% per cycle over 1000 cycles at 2.0 C. The HE-PBA@Cu/Li anode maintains a stable overpotential even after 600 h at 0.5 mA cm-2 under the total areal capacity of 1.0 mA h cm-2. This study showcases the application potential of the HE-PBA in Li-S batteries and encourages further exploration of prospective high-entropy materials used to engineer next-generation batteries.
Collapse
Affiliation(s)
- Nan Shen
- School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing 210044, Jiangsu, China.
| | - Tianqi Li
- School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing 210044, Jiangsu, China.
| | - Boya Li
- School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing 210044, Jiangsu, China.
| | - Yi Wang
- School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing 210044, Jiangsu, China.
| | - He Liu
- School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing 210044, Jiangsu, China.
| | - Cong Guo
- School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing 210044, Jiangsu, China.
| | - Xiaoyu Chen
- School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing 210044, Jiangsu, China.
- Nanjing Energy Digital Electric Co. Ltd, Nanjing 211106, Jiangsu, China
| | - Jingfa Li
- School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing 210044, Jiangsu, China.
| |
Collapse
|