1
|
Wang X, Liu Y, Jiang Y, Li Q. Tumor-derived exosomes as promising tools for cancer diagnosis and therapy. Front Pharmacol 2025; 16:1596217. [PMID: 40444049 PMCID: PMC12119533 DOI: 10.3389/fphar.2025.1596217] [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: 03/19/2025] [Accepted: 05/06/2025] [Indexed: 06/02/2025] Open
Abstract
Mounting evidences indicated that cancer cell-derived exosomes (TDEs) contribute to cancer progression and metastasis by reshaping the tumor microenvironment (TME) and interfering immunity response. TDEs contain unique biomolecular cargo, consisting of protein, nucleic acid, and lipids. In recent years, TDEs have been used as potential disease therapeutics and diagnosis biomarkers and prime candidates as delivery tools for cancer treatment. In the present review, we firstly summarized TDEs biogenesis and characteristic. Also, the role of TDEs in cancer cell metastasis and invasiveness, drug resistance, and immunosuppression was mentioned via cell-cell communication. Additionally, we concluded the current strategies for TDE-based cancer therapy, including TDEs inhibition and clearance, usage as therapeutic drug delivery vector and cancer vaccines. Furthermore, combination therapy with engineered TDEs were summarized, such as radiotherapy, photodynamic therapy, photothermal therapy, and sonodynamic therapy. Consequently, the above opens up novel and interesting opportunities for cancer diagnosis and prognosis based on TDEs, which is prospective to accelerate the clinical translation of TDEs for cancer therapy.
Collapse
Affiliation(s)
- Xirui Wang
- Department of Biomedical Engineering, School of Medical Imaging Xuzhou Medical University, Xuzhou, China
| | - Yanfang Liu
- Department of Central Laboratory, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Yaowen Jiang
- Department of Biomedical Engineering, School of Medical Imaging Xuzhou Medical University, Xuzhou, China
| | - Qinghua Li
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
| |
Collapse
|
2
|
Yang M, Zhou J, Lu L, Deng D, Huang J, Tang Z, Shi X, Lo P, Lovell JF, Zheng Y, Jin H. Tumor cell membrane-based vaccines: A potential boost for cancer immunotherapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230171. [PMID: 39713208 PMCID: PMC11655317 DOI: 10.1002/exp.20230171] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/08/2024] [Indexed: 12/24/2024]
Abstract
Because therapeutic cancer vaccines can, in theory, eliminate tumor cells specifically with relatively low toxicity, they have long been considered for application in repressing cancer progression. Traditional cancer vaccines containing a single or a few discrete tumor epitopes have failed in the clinic, possibly due to challenges in epitope selection, target downregulation, cancer cell heterogeneity, tumor microenvironment immunosuppression, or a lack of vaccine immunogenicity. Whole cancer cell or cancer membrane vaccines, which provide a rich source of antigens, are emerging as viable alternatives. Autologous and allogenic cellular cancer vaccines have been evaluated as clinical treatments. Tumor cell membranes (TCMs) are an intriguing antigen source, as they provide membrane-accessible targets and, at the same time, serve as integrated carriers of vaccine adjuvants and other therapeutic agents. This review provides a summary of the properties and technologies for TCM cancer vaccines. Characteristics, categories, mechanisms, and preparation methods are discussed, as are the demonstrable additional benefits derived from combining TCM vaccines with chemotherapy, sonodynamic therapy, phototherapy, and oncolytic viruses. Further research in chemistry, biomedicine, cancer immunology, and bioinformatics to address current drawbacks could facilitate the clinical adoption of TCM vaccines.
Collapse
Affiliation(s)
- Muyang Yang
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Jie Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Liseng Lu
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Deqiang Deng
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Jing Huang
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Zijian Tang
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Xiujuan Shi
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Pui‐Chi Lo
- Department of Biomedical SciencesCity University of Hong KongKowloonHong KongChina
| | - Jonathan F. Lovell
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNew YorkUSA
| | - Yongfa Zheng
- Department of OncologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Honglin Jin
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| |
Collapse
|
3
|
Lu Y, Zheng J, Lin P, Lin Y, Zheng Y, Mai Z, Chen X, Xia T, Zhao X, Cui L. Tumor Microenvironment-Derived Exosomes: A Double-Edged Sword for Advanced T Cell-Based Immunotherapy. ACS NANO 2024; 18:27230-27260. [PMID: 39319751 DOI: 10.1021/acsnano.4c09190] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer progression and immune evasion, partially mediated by the activity of the TME-derived exosomes. These extracellular vesicles are pivotal in shaping immune responses through the transfer of proteins, lipids, and nucleic acids between cells, facilitating a complex interplay that promotes tumor growth and metastasis. This review delves into the dual roles of exosomes in the TME, highlighting both their immunosuppressive functions and their emerging therapeutic potential. Exosomes can inhibit T cell function and promote tumor immune escape by carrying immune-modulatory molecules, such as PD-L1, yet they also hold promise for cancer therapy as vehicles for delivering tumor antigens and costimulatory signals. Additionally, the review discusses the intricate crosstalk mediated by exosomes among various cell types within the TME, influencing both cancer progression and responses to immunotherapies. Moreover, this highlights current challenges and future directions. Collectively, elucidating the detailed mechanisms by which TME-derived exosomes mediate T cell function offers a promising avenue for revolutionizing cancer treatment. Understanding these interactions allows for the development of targeted therapies that manipulate exosomal pathways to enhance the immune system's response to tumors.
Collapse
Affiliation(s)
- Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Pei Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yucheng Zheng
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Zizhao Mai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Xu Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Tian Xia
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
- School of Dentistry, University of California Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
4
|
Xu C, Jiang C, Li Z, Gao H, Xian J, Guo W, He D, Peng X, Zhou D, Li D. Exosome nanovesicles: biomarkers and new strategies for treatment of human diseases. MedComm (Beijing) 2024; 5:e660. [PMID: 39015555 PMCID: PMC11247338 DOI: 10.1002/mco2.660] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024] Open
Abstract
Exosomes are nanoscale vesicles of cellular origin. One of the main characteristics of exosomes is their ability to carry a wide range of biomolecules from their parental cells, which are important mediators of intercellular communication and play an important role in physiological and pathological processes. Exosomes have the advantages of biocompatibility, low immunogenicity, and wide biodistribution. As researchers' understanding of exosomes has increased, various strategies have been proposed for their use in diagnosing and treating diseases. Here, we provide an overview of the biogenesis and composition of exosomes, describe the relationship between exosomes and disease progression, and focus on the use of exosomes as biomarkers for early screening, disease monitoring, and guiding therapy in refractory diseases such as tumors and neurodegenerative diseases. We also summarize the current applications of exosomes, especially engineered exosomes, for efficient drug delivery, targeted therapies, gene therapies, and immune vaccines. Finally, the current challenges and potential research directions for the clinical application of exosomes are also discussed. In conclusion, exosomes, as an emerging molecule that can be used in the diagnosis and treatment of diseases, combined with multidisciplinary innovative solutions, will play an important role in clinical applications.
Collapse
Affiliation(s)
- Chuan Xu
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| | - Chaoyang Jiang
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| | - Zhihui Li
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| | - Hui Gao
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| | - Jing Xian
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| | - Wenyan Guo
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| | - Dan He
- Department of OncologyThe Second Affiliated Hospital of Chengdu Medical CollegeChina National Nuclear Corporation 416 HospitalChengduSichuanChina
| | - Xingchen Peng
- Department of BiotherapyCancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| | - Daijun Zhou
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| | - Dong Li
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| |
Collapse
|
5
|
Zhao G, Wang Y, Xing S, Jiang Y, Ding J, Cai Y, Ma P, Miao H, Fang Y, Jiang N, Cui D, Yu Y, Tang Q, Wang S, Li N. Exosome-based anticancer vaccines: From Bench to bedside. Cancer Lett 2024; 595:216989. [PMID: 38825162 DOI: 10.1016/j.canlet.2024.216989] [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: 03/14/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 06/04/2024]
Abstract
Exosomes, a subset of extracellular vesicles, are released by all active cells and play a crucial role in intercellular communications. Exosomes could facilitate the transfer of various biologically active molecules, such as DNA, non-coding RNAs, and proteins, from donor to recipient cells, thereby participating in diverse biological and pathological processes. Besides, exosomes possess unique characteristics, including non-toxicity, low-immunogenicity, and stability within biological systems, rendering them highly advantageous for cancer drug development. Meanwhile, accumulating evidence suggests that exosomes originating from tumor cells and immune cells possess distinct composition profiles that play a direct role in anticancer immunotherapy. Of note, exosomes can transport their contents to specific cells, thereby exerting an impact on the phenotype and immune-regulatory functions of targeted cells. Therapeutic cancer vaccines, an emerging therapeutics of immunotherapy, could enhance antitumor immune responses by delivering a large number of tumor antigens, thereby augmenting the immune response against tumor cells. Therefore, the therapeutic rationale of cancer vaccines and exosome-based immunotherapy are almost similar to some extent, but some challenges have hindered their application in the clinical setting. Here, in this review, we first summarized the biogenesis, structure, compositions, and biological functions of exosomes. Then we described the roles of exosomes in cancer biology, particularly in tumor immunity. We also comprehensively reviewed current exosome-based anticancer vaccine development and we divided them into three types. Finally, we give some insights into clinical translation and clinical trial progress of exosome-based anticancer vaccines for future direction.
Collapse
Affiliation(s)
- Guo Zhao
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuning Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shujun Xing
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yale Jiang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiatong Ding
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuanting Cai
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peiwen Ma
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Huilei Miao
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuan Fang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ning Jiang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dandan Cui
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yue Yu
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qiyu Tang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuhang Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Ning Li
- Clinical Trial Center, 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
|
6
|
Takahashi Y, Takakura Y. Extracellular vesicle-based therapeutics: Extracellular vesicles as therapeutic targets and agents. Pharmacol Ther 2023; 242:108352. [PMID: 36702209 DOI: 10.1016/j.pharmthera.2023.108352] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/06/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived membrane vesicles composed of a lipid bilayer. EVs contain biological molecules, such as nucleic acids, lipids, and proteins. As these molecules are transferred to cells that receive EVs, EVs function as intercellular communication tools. EV-mediated intercellular communication is involved in various diseases, such as cancer and neurodegenerative diseases, and biological events, such as immune reactions and inflammation. Therefore, EVs are suggested to be useful as therapeutic targets for various diseases. However, an EV-based drug delivery system (DDS) that utilizes its therapeutic properties has not yet been reported. The biological activities of EVs are derived from their endogenous components; hence, they can be directly applied as drugs. In this review, the basic aspects of EVs, such as their types, methods of isolation, and in vivo behavior, are briefly summarized. Moreover, the potential of using therapeutics targeting EVs has been discussed in cancer and neurodegenerative diseases. Various therapeutics using EVs, including DDSs, are listed and their associated advantages and challenges are discussed.
Collapse
Affiliation(s)
- Yuki Takahashi
- Graduate School of Pharmaceutical Sciences, Kyoto University; 46-29 Yoshida-Shimo-Adachi, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yoshinobu Takakura
- Graduate School of Pharmaceutical Sciences, Kyoto University; 46-29 Yoshida-Shimo-Adachi, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
7
|
Zhu H, Liu Y, Yi X, Zhu C, Fu Y, Huang Z, Zhu K, Zhang W, Hou H, Sun C, Zhong C, Liu W, Li Z, Wang B, Wo J. Novel biomimetic mesoporous silica nanoparticle system possessing targetability and immune synergy facilitates effective solid tumor immuno-chemotherapy. BIOMATERIALS ADVANCES 2023; 144:213229. [PMID: 36502749 DOI: 10.1016/j.bioadv.2022.213229] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 11/12/2022] [Accepted: 11/28/2022] [Indexed: 06/17/2023]
Abstract
New strategies that enhance both the targetability of chemotherapy drugs and the synergistic effects of chemotherapy and immunotherapy are urgently needed for efficacious solid tumor therapy. In this study, a novel biomimetic nanoparticle system possessing the properties of tumor targeting and immune synergy was designed to meet these requirements. Mesoporous silica nanoparticles loaded with the chemotherapeutic drug doxorubicin (DOX) were coated with cell membranes modified by glycosylphosphatidylinositol (GPI)-anchored anti-HER2 single chain variable fragment (scFv) and the GPI-anchored co-stimulatory molecule CD80 (to promote solid tumor-targeted chemotherapy and cooperated immunotherapy, respectively). The impact of the nanotherapeutic system on both tumor-targeted chemotherapy and cellular immune response was investigated through in vitro and in vivo experiments. The results show that the novel biomimetic therapeutic system effectively promoted antitumor efficiency in vitro and in vivo. In addition, this therapeutic system further enhanced antitumor capacity by increasing CD8+ T cell activation and cytokine production and reducing myeloid-derived suppressor cell (MDSC) levels in tumors.
Collapse
Affiliation(s)
- Haoran Zhu
- Department of Orthopedics, The First Affiliated Hospital and The Fifth Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yang Liu
- Department of Orthopedics, The First Affiliated Hospital and The Fifth Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xinfeng Yi
- Department of Neurosurgery, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, China
| | - Chuyun Zhu
- The Biomedical Translational Research Institute, Jinan University Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Yuanyue Fu
- Department of Orthopedics, The First Affiliated Hospital and The Fifth Affiliated Hospital, Jinan University, Guangzhou, China
| | - Zerong Huang
- Department of Orthopedics, The First Affiliated Hospital and The Fifth Affiliated Hospital, Jinan University, Guangzhou, China
| | - Kairui Zhu
- Department of Orthopedics, The First Affiliated Hospital and The Fifth Affiliated Hospital, Jinan University, Guangzhou, China
| | - Wencai Zhang
- Department of Orthopedics, The First Affiliated Hospital and The Fifth Affiliated Hospital, Jinan University, Guangzhou, China
| | - Huige Hou
- Department of Orthopedics, The First Affiliated Hospital and The Fifth Affiliated Hospital, Jinan University, Guangzhou, China
| | - Chenghong Sun
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Cheng Zhong
- The Affiliated Hospital (Jiangmen Traditional Chinese Medicine Hospital), Jinan University, Guangzhou, China
| | - Wei Liu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China.
| | - Zhizhong Li
- Department of Orthopedics, The First Affiliated Hospital and The Fifth Affiliated Hospital, Jinan University, Guangzhou, China.
| | - Baocheng Wang
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, China.
| | - Jin Wo
- Department of Orthopedics, The First Affiliated Hospital and The Fifth Affiliated Hospital, Jinan University, Guangzhou, China; Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China.
| |
Collapse
|
8
|
Zhang X, Cui H, Zhang W, Li Z, Gao J. Engineered tumor cell-derived vaccines against cancer: The art of combating poison with poison. Bioact Mater 2022; 22:491-517. [PMID: 36330160 PMCID: PMC9619151 DOI: 10.1016/j.bioactmat.2022.10.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/07/2022] [Accepted: 10/13/2022] [Indexed: 12/23/2022] Open
Abstract
Tumor vaccination is a promising approach for tumor immunotherapy because it presents high specificity and few side effects. However, tumor vaccines that contain only a single tumor antigen can allow immune system evasion by tumor variants. Tumor antigens are complex and heterogeneous, and identifying a single antigen that is uniformly expressed by tumor cells is challenging. Whole tumor cells can produce comprehensive antigens that trigger extensive tumor-specific immune responses. Therefore, tumor cells are an ideal source of antigens for tumor vaccines. A better understanding of tumor cell-derived vaccines and their characteristics, along with the development of new technologies for antigen delivery, can help improve vaccine design. In this review, we summarize the recent advances in tumor cell-derived vaccines in cancer immunotherapy and highlight the different types of engineered approaches, mechanisms, administration methods, and future perspectives. We discuss tumor cell-derived vaccines, including whole tumor cell components, extracellular vesicles, and cell membrane-encapsulated nanoparticles. Tumor cell-derived vaccines contain multiple tumor antigens and can induce extensive and potent tumor immune responses. However, they should be engineered to overcome limitations such as insufficient immunogenicity and weak targeting. The genetic and chemical engineering of tumor cell-derived vaccines can greatly enhance their targeting, intelligence, and functionality, thereby realizing stronger tumor immunotherapy effects. Further advances in materials science, biomedicine, and oncology can facilitate the clinical translation of tumor cell-derived vaccines.
Collapse
Affiliation(s)
- Xinyi Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Hengqing Cui
- Department of Burns and Plastic Surgery, Shanghai Changzheng Hospital, Shanghai, 200003, China
| | - Wenjun Zhang
- Department of Burns and Plastic Surgery, Shanghai Changzheng Hospital, Shanghai, 200003, China
| | - Zhaoshen Li
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Corresponding author. Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China,Corresponding author. Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200444, China.
| |
Collapse
|
9
|
Xiu H, Nan X, Guo D, Wang J, Li J, Peng Y, Xiong G, Wang S, Wang C, Zhang G, Yang Y, Cai Z. Gp350-anchored extracellular vesicles: promising vehicles for delivering therapeutic drugs of B cell malignancies. Asian J Pharm Sci 2022; 17:462-474. [PMID: 35782327 PMCID: PMC9237600 DOI: 10.1016/j.ajps.2022.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/18/2022] [Accepted: 03/21/2022] [Indexed: 11/27/2022] Open
Abstract
Although chimeric antigen receptor-modified (CAR) T cell therapy has been successfully applied in the treatment of acute B lymphocytic leukemia, its effect on Burkitt lymphoma (BL) and chronic B lymphocytic leukemia (B-CLL) is unsatisfactory. Moreover, fatal side effects greatly impede CAR T cell application. Extracellular vesicles (EVs) are excellent carriers of therapeutic agents. Nevertheless, EVs mainly accumulate in the liver when administered without modification. As an envelope glycoprotein of Epstein-Barr viruses, gp350 can efficiently bind CD21 on B cells. Here, gp350 was directly anchored onto red blood cell EVs (RBC-EVs) via its transmembrane region combined with low-voltage electroporation. The results showed that gp350 could anchor to RBC-EVs with high efficiency and that the resulting gp350-anchored RBC-EVs (RBC-EVs/gp350Etp) exhibited increased targeting to CD21+ BL and B-CLL relative to RBC-EVs. After the loading of doxorubicin or fludarabine, RBC-EVs/gp350Etp had powerful cytotoxicity and therapeutic efficacy on CD21+ BL or B-CLL, respectively. Moreover, RBC-EVs/gp350Etp loaded with a drug did not exhibit any apparent systemic toxicity and specifically induced the apoptosis of tumor B cells but not normal B cells. Therefore, our findings indicate that drug-loaded RBC-EVs/gp350Etp may be adopted in the treatment of CD21+ B cell malignancies.
Collapse
Affiliation(s)
- Huiqing Xiu
- Institute of Immunology, and Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xi Nan
- Institute of Immunology, and Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Danfeng Guo
- Henan Key Laboratory for Digestive Organ Transplantation, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiaoli Wang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University Cancer Centre, Hangzhou, China
| | - Jiahui Li
- Department of Critical Care Medicine of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanmei Peng
- Institute of Immunology, and Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guirun Xiong
- Department of Emergency Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Shibo Wang
- Institute of Immunology, and Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Changjun Wang
- Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China
| | - Gensheng Zhang
- Department of Critical Care Medicine of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunshan Yang
- Department of Medical Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Zhijian Cai
- Institute of Immunology, and Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
10
|
Hosseini R, Sarvnaz H, Arabpour M, Ramshe SM, Asef-Kabiri L, Yousefi H, Akbari ME, Eskandari N. Cancer exosomes and natural killer cells dysfunction: biological roles, clinical significance and implications for immunotherapy. Mol Cancer 2022; 21:15. [PMID: 35031075 PMCID: PMC8759167 DOI: 10.1186/s12943-021-01492-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/26/2021] [Indexed: 12/16/2022] Open
Abstract
Tumor-derived exosomes (TDEs) play pivotal roles in several aspects of cancer biology. It is now evident that TDEs also favor tumor growth by negatively affecting anti-tumor immunity. As important sentinels of immune surveillance system, natural killer (NK) cells can recognize malignant cells very early and counteract the tumor development and metastasis without a need for additional activation. Based on this rationale, adoptive transfer of ex vivo expanded NK cells/NK cell lines, such as NK-92 cells, has attracted great attention and is widely studied as a promising immunotherapy for cancer treatment. However, by exploiting various strategies, including secretion of exosomes, cancer cells are able to subvert NK cell responses. This paper reviews the roles of TDEs in cancer-induced NK cells impairments with mechanistic insights. The clinical significance and potential approaches to nullify the effects of TDEs on NK cells in cancer immunotherapy are also discussed.
Collapse
Affiliation(s)
- Reza Hosseini
- Department of Immunology School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Hamzeh Sarvnaz
- Department of Immunology School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Maedeh Arabpour
- Department of Medical Genetics School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Molaei Ramshe
- Student Research Committee, Department of Medical Genetics, School of Medicine Shahid, Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Asef-Kabiri
- Surgical Oncologist Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, USA
| | - Mohammad Esmaeil Akbari
- Surgical Oncologist Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nahid Eskandari
- Department of Immunology School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
11
|
Amiri A, Pourhanifeh MH, Mirzaei HR, Nahand JS, Moghoofei M, Sahebnasagh R, Mirzaei H, Hamblin MR. Exosomes and Lung Cancer: Roles in Pathophysiology, Diagnosis and Therapeutic Applications. Curr Med Chem 2021; 28:308-328. [PMID: 32013817 DOI: 10.2174/0929867327666200204141952] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/18/2019] [Accepted: 11/29/2019] [Indexed: 11/22/2022]
Abstract
Lung cancer is a malignancy with a high morbidity and mortality rate, and affected patients have low survival and poor prognosis. The therapeutic approaches for the treatment of this cancer, including radiotherapy and chemotherapy, are not particularly effective partly due to late diagnosis. Therefore, the search for new diagnostic and prognostic tools is a critical issue. Novel biomarkers, such as exosomes, could be considered as potential diagnostic tools for malignancies, particularly lung cancer. Exosomes are nanovesicles, which are associated with different physiological and pathological conditions. It has been shown that these particles are released from many cells, such as cancer cells, immune cells and to some degree normal cells. Exosomes could alter the behavior of target cells through intercellular transfer of their cargo (e.g. DNA, mRNA, long non-coding RNAs, microRNAs and proteins). Thus, these vehicles may play pivotal roles in various physiological and pathological conditions. The current insights into lung cancer pathogenesis suggest that exosomes are key players in the pathogenesis of this cancer. Hence, these nanovesicles and their cargos could be used as new diagnostic, prognostic and therapeutic biomarkers in the treatment of lung cancer. Besides the diagnostic roles of exosomes, their use as drug delivery systems and as cancer vaccines is under investigation. The present review summarizes the current information on the diagnostic and pathogenic functions of exosomes in lung cancer.
Collapse
Affiliation(s)
- Atefeh Amiri
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashahd, Iran
| | | | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Javid Sadri Nahand
- Department of Virology, Faculty of Medicine, University of Medical Sciences, Tehran, Iran
| | - Mohsen Moghoofei
- Department of Microbiology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Roxana Sahebnasagh
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, 40 Blossom Street, Boston, MA, 02114, United States
| |
Collapse
|
12
|
Sun Y, Guo J, Yu L, Guo T, Wang J, Wang X, Chen Y. PD-L1 + exosomes from bone marrow-derived cells of tumor-bearing mice inhibit antitumor immunity. Cell Mol Immunol 2020; 18:2402-2409. [PMID: 32606317 DOI: 10.1038/s41423-020-0487-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 06/03/2020] [Indexed: 12/31/2022] Open
Abstract
Tumors escape immune attack by upregulating the surface expression of PD-L1, which interacts with PD-1 on T cells to activate immune inhibitory signaling. Anti-PD-1 treatments can effectively block this inhibitory signaling and activate antitumor immune responses. However, anti-PD-1 treatment also has a tumor suppressive effect in patients whose tumor cells do not express PD-L1. The underlying mechanisms are poorly defined. Here, we report that exosomes from bone marrow-derived cells (BMDCs) in tumor-bearing mice, but not in healthy mice, carry PD-L1. PD-L1 on these exosomes is biofunctional and can inhibit CD8+ T cell proliferation and activation in vitro and in vivo. The transfer of exogenous exosomes from BMDCs and the inhibition of the production of endogenous exosomes by BMDCs promote and suppress tumor growth, respectively. PD-L1+ exosomes from BMDCs can be found in tumor tissues. In addition, exosomes from BMDCs promote tumor metastasis in a PD-L1-dependent manner. Therefore, our results indicate that exosomes from BMDCs play important roles in tumor immunosuppression via PD-L1.
Collapse
Affiliation(s)
- Yan Sun
- Department of Infectious Disease, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, 310052, Hangzhou, China.,Department of Comprehensive Medical Oncology, Zhejiang Cancer Hospital, 310022, Hangzhou, China.,Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310020, Hangzhou, China
| | - Jufeng Guo
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, China
| | - Lei Yu
- Laboratory of Cancer Biology, The Key Lab of Biotherapy in Zhejiang Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310020, Hangzhou, China
| | - Tianxin Guo
- Department of Respiratory Medicine, Nanjing Medical University, 211166, Nanjing, China
| | - Jiaoli Wang
- Department of Respiratory Medicine, Nanjing Medical University, 211166, Nanjing, China.,Department of Respiratory Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, China
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310020, Hangzhou, China.
| | - Yinghu Chen
- Department of Infectious Disease, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, 310052, Hangzhou, China.
| |
Collapse
|
13
|
Naseri M, Bozorgmehr M, Zöller M, Ranaei Pirmardan E, Madjd Z. Tumor-derived exosomes: the next generation of promising cell-free vaccines in cancer immunotherapy. Oncoimmunology 2020; 9:1779991. [PMID: 32934883 PMCID: PMC7466856 DOI: 10.1080/2162402x.2020.1779991] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Identification of immunogenic tumor antigens that are efficiently processed and delivered by dendritic cells to prime the immune system and to induce an appropriate immune response is a research hotspot in the field of cancer vaccine development. High biosafety is an additional demand. Tumor-derived exosomes (TEXs) are nanosized lipid bilayer encapsulated vesicles that shuttle bioactive information to the tumor microenvironment facilitating tumor progression. However, accumulating evidence points toward the capacity of TEXs to efficiently stimulate immune responses against tumors provided they are appropriately administered. After briefly describing the function of exosomes in cancer biology and their communication with immune cells, we summarize in this review in vitro and preclinical studies eliciting the potency of TEXs in inducing effective anti-tumor responses and recently modified strategies further improving TEX-vaccination efficacy. We interpret the available data as TEXs becoming a lead in cancer vaccination based on tumor antigen-selective high immunogenicity.
Collapse
Affiliation(s)
- Marzieh Naseri
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmood Bozorgmehr
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Margot Zöller
- Section Pancreas Research, University Hospital of Surgery, Heidelberg, Germany
| | - Ehsan Ranaei Pirmardan
- Molecular Biomarkers Nano-imaging Laboratory, Brigham & Women's Hospital, Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Zahra Madjd
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Wan M, Ning B, Spiegel S, Lyon CJ, Hu TY. Tumor-derived exosomes (TDEs): How to avoid the sting in the tail. Med Res Rev 2019; 40:385-412. [PMID: 31318078 PMCID: PMC6917833 DOI: 10.1002/med.21623] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/26/2019] [Accepted: 06/13/2019] [Indexed: 02/05/2023]
Abstract
Exosomes are abundantly secreted extracellular vesicles that accumulate in the circulation and are of great interest for disease diagnosis and evaluation since their contents reflects the phenotype of their cell of origin. Tumor‐derived exosomes (TDEs) are of particular interest for cancer diagnosis and therapy, since most tumor demonstrate highly elevated exosome secretion rates and provide specific information about the genotype of a tumor and its response to treatment. TDEs also contain regulatory factors that can alter the phenotypes of local and distant tissue sites and alter immune cell functions to promote tumor progression. The abundance, information content, regulatory potential, in vivo half‐life, and physical durability of exosomes suggest that TDEs may represent a superior source of diagnostic biomarkers and treatment targets than other materials currently under investigation. This review will summarize current information on mechanisms that may differentially regulate TDE biogenesis, TDE effects on the immune system that promote tumor survival, growth, and metastasis, and new approaches understudy to counteract or utilize TDE properties in cancer therapies.
Collapse
Affiliation(s)
- MeiHua Wan
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Bo Ning
- Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Sarah Spiegel
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona.,Virginia G. Piper Biodesign Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Christopher J Lyon
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona.,Virginia G. Piper Biodesign Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Tony Y Hu
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona.,Virginia G. Piper Biodesign Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, Arizona
| |
Collapse
|
15
|
Sarvizadeh M, Ghasemi F, Tavakoli F, Sadat Khatami S, Razi E, Sharifi H, Biouki NM, Taghizadeh M. Vaccines for colorectal cancer: an update. J Cell Biochem 2018; 120:8815-8828. [PMID: 30536960 DOI: 10.1002/jcb.28179] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 11/12/2018] [Indexed: 12/29/2022]
Abstract
Colorectal cancer (CRC) is known as the third most common and fourth leading cancer associated death worldwide. The occurrence of metastasis has remained as a critical challenge in CRC, so that distant metastasis (mostly to the liver) has been manifested in about 20%-25% of patients. Several screening approaches have introduced for detecting CRC in different stages particularly in early stages. The standard treatments for CRC are surgery, chemotherapy and radiotherapy, in alone or combination. Immunotherapy is a set of novel approaches with the aim of remodeling the immune system battle with metastatic cancer cells, such as immunomodulatory monoclonal antibodies (immune checkpoint inhibitors), adoptive cell transfer (ACT) and cancer vaccine. Cancer vaccines are designed to trigger the intense response of immune system to tumor-specific antigens. In two last decades, introduction of new cancer vaccines and designing several clinical trials with vaccine therapy, have been taken into consideration in colon cancer patients. This review will describe the treatment approaches with the special attention to vaccines applied to treat colorectal cancer.
Collapse
Affiliation(s)
- Mostafa Sarvizadeh
- The Advocate Center for Clinical Research, Ayatollah Yasrebi Hospital, Kashan, Iran
| | - Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Fatemeh Tavakoli
- Department of Biotechnology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Sadat Khatami
- Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Ebrahim Razi
- The Advocate Center for Clinical Research, Ayatollah Yasrebi Hospital, Kashan, Iran
| | - Hossein Sharifi
- The Advocate Center for Clinical Research, Ayatollah Yasrebi Hospital, Kashan, Iran
| | - Nousin Moussavi Biouki
- Department of Surgery, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
16
|
Moradi‐Chaleshtori M, Hashemi SM, Soudi S, Bandehpour M, Mohammadi‐Yeganeh S. Tumor‐derived exosomal microRNAs and proteins as modulators of macrophage function. J Cell Physiol 2018; 234:7970-7982. [DOI: 10.1002/jcp.27552] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 09/14/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Maryam Moradi‐Chaleshtori
- Department of Biotechnology School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology School of Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
- Department of Applied Cell Sciences School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Sara Soudi
- Department of Immunology Faculty of Medical Sciences, Tarbiat Modares University Tehran Iran
| | - Mojgan Bandehpour
- Department of Biotechnology School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Samira Mohammadi‐Yeganeh
- Department of Biotechnology School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences Tehran Iran
| |
Collapse
|
17
|
Yang Y, Hong Y, Cho E, Kim GB, Kim IS. Extracellular vesicles as a platform for membrane-associated therapeutic protein delivery. J Extracell Vesicles 2018; 7:1440131. [PMID: 29535849 PMCID: PMC5844050 DOI: 10.1080/20013078.2018.1440131] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 02/07/2018] [Indexed: 02/08/2023] Open
Abstract
Membrane proteins are of great research interest, particularly because they are rich in targets for therapeutic application. The suitability of various membrane proteins as targets for therapeutic formulations, such as drugs or antibodies, has been studied in preclinical and clinical studies. For therapeutic application, however, a protein must be expressed and purified in as close to its native conformation as possible. This has proven difficult for membrane proteins, as their native conformation requires the association with an appropriate cellular membrane. One solution to this problem is to use extracellular vesicles as a display platform. Exosomes and microvesicles are membranous extracellular vesicles that are released from most cells. Their membranes may provide a favourable microenvironment for membrane proteins to take on their proper conformation, activity, and membrane distribution; moreover, membrane proteins can cluster into microdomains on the surface of extracellular vesicles following their biogenesis. In this review, we survey the state-of-the-art of extracellular vesicle (exosome and small-sized microvesicle)-based therapeutics, evaluate the current biological understanding of these formulations, and forecast the technical advances that will be needed to continue driving the development of membrane protein therapeutics.
Collapse
Affiliation(s)
- Yoosoo Yang
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division for Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Yeonsun Hong
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Eunji Cho
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Gi Beom Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - In-San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| |
Collapse
|
18
|
Guo D, Chen Y, Wang S, Yu L, Shen Y, Zhong H, Yang Y. Exosomes from heat-stressed tumour cells inhibit tumour growth by converting regulatory T cells to Th17 cells via IL-6. Immunology 2018; 154:132-143. [PMID: 29197065 DOI: 10.1111/imm.12874] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 11/17/2017] [Accepted: 11/23/2017] [Indexed: 12/15/2022] Open
Abstract
Exosomes derived from heat-stressed tumour cells (HS-TEXs), which contain abundant heat shock protein (HSP) 70, strongly induce antitumour immune responses. HSP70-induced interleukin (IL)-6 promotes IL-17 expression and causes rejection of established prostate tumours. However, it remains unclear whether HS-TEXs exhibit antitumour effects by converting regulatory T cells (Tregs ) into T helper type 17 (Th17) cells. In this study, we found that compared with TEXs, HS-TEXs were more potent in stimulating secretion of IL-6 from dendritic cells. In vitro, IL-6 blocked tumour cell-derived transforming growth factor beta 1-induced Treg differentiation and promoted Th17 cell differentiation. HS-TEXs exerted strong antitumour effects, converting Tregs into Th17 cells with high efficiency, a process that was entirely dependent upon IL-6. Neutralization of IL-17 completely abolished the antitumour effect of TEXs, but only partially inhibited that of HS-TEXs. In addition, we found higher levels of IL-6 and IL-17 in serum from tumour patients treated with hyperthermia, and an increase in Th17 cells and a decrease in Tregs was detected in peripheral blood mononuclear cells isolated from these patients after hyperthermia. Therefore, our results demonstrate that HS-TEXs possess a powerful capacity to convert immunosuppressive Tregs into Th17 cells via IL-6, which contributes to their potent antitumour effect.
Collapse
Affiliation(s)
- Danfeng Guo
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yinghu Chen
- Zhejiang Key Laboratory for Neonatal Diseases, Division of Infectious Diseases, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shoujie Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Yu
- Laboratory of Cancer Epigenetics, Department of Medical Oncology, Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yingying Shen
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Haijun Zhong
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yunshan Yang
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
19
|
Huang Y, Liu K, Li Q, Yao Y, Wang Y. Exosomes Function in Tumor Immune Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1056:109-122. [PMID: 29754177 DOI: 10.1007/978-3-319-74470-4_7] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Immune cells and mesenchymal stem/stromal cells are the major cellular components in tumor microenvironment that actively migrate to tumor sites by sensing "signals" released from tumor cells. Together with other stromal cells, they form the soil for malignant cell progression. In the crosstalk between tumor cells and its surrounded microenvironment, exosomes exert multiple functions in shaping tumor immune responses. In tumor cells, their exosomes can lead to pro-tumor immune responses, whereas in immune cells, their derived exosomes can operate on tumor cells and regulate their ability to growth, metastasis, even reaction to chemotherapy. Employing exosomes as vehicles for the delivery products to initiate anti-tumor immune responses has striking therapeutic effects on tumor progression. Thus, exosomes are potential therapeutic targets in tumor-related clinical conditions. Here we discuss the role of exosomes in regulating tumor immune microenvironment and future indications for the clinical application of exosomes.
Collapse
Affiliation(s)
- Yin Huang
- Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Keli Liu
- Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Qing Li
- Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yikun Yao
- Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ying Wang
- Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
20
|
Behzadi E, Hosseini HM, Halabian R, Fooladi AAI. Macrophage cell-derived exosomes/staphylococcal enterotoxin B against fibrosarcoma tumor. Microb Pathog 2017; 111:132-138. [DOI: 10.1016/j.micpath.2017.08.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 08/12/2017] [Accepted: 08/16/2017] [Indexed: 01/08/2023]
|
21
|
Xie Y, Wang M, Dong Z, Song H, Li L, Yang M, Li P, Tian J, Zhang K, Xia X, Zhang T, Tang A. In vitro effects of Staphylococcus aureus enterotoxin C3 on T cell activation, proliferation and cytokine production. Mol Med Rep 2017; 16:4744-4750. [PMID: 28849041 PMCID: PMC5647103 DOI: 10.3892/mmr.2017.7199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 06/13/2017] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the effects of Staphylococcus aureus enterotoxin C3 (SEC3), including recombinant (r)SEC3 protein and lentivirus‑mediated SEC3, on the activation, proliferation and cytokine production of human T cells. HeLa cells were infected with SEC3 lentiviral vector (LV‑SEC3) and viability was determined using the Cell Counting Kit‑8 (CCK‑8) assay. Subsequently, infected cells or rSEC3 protein were co‑cultured with human peripheral blood mononuclear cells (PBMCs) for 10 days, after which the culture supernatant and T cells were incubated with untreated HeLa cells, which were subjected to a CCK‑8 assay to determine cytotoxicity. In addition, IL‑6 and IFN‑γ expression was detected by chemiluminescence and enzyme‑linked immunospot analyses, respectively. Subpopulations of activated T cells were sorted by flow cytometry. The results demonstrated that, following infection with LV‑SEC3 or negative control lentiviral vector (LV‑NC), >80% of HeLa cells presented green fluorescent protein‑positive signals. All five groups of co‑cultured T cells exhibited proliferation. Co‑culture of PBMCs with rSEC3 protein or LV‑SEC‑infected cells resulted in elevated IL‑6 and IFN‑γ secretion. In addition, rSEC3‑activated and monocultured T cells were predominantly cluster of differentiation (CD)4+ (62.7 and 59.6%, respectively) whereas phytohemagglutinin‑stimulated T cells were predominantly CD8+ (57.8%). Compared with the LV‑NC group, T cells and culture supernatants from the LV‑SEC3 group significantly attenuated proliferation of HeLa cells. These results suggest that rSEC3 protein, and LV‑SEC3‑infected HeLa cells, are able to potently activate T cells, increasing cytokine production and amplify the antitumor immune response.
Collapse
Affiliation(s)
- Yixin Xie
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Min Wang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Zhihui Dong
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Huan Song
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Lianping Li
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Min Yang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Pengling Li
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Jingjing Tian
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Kan Zhang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Xiaomeng Xia
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Tingting Zhang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Aiguo Tang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
22
|
Di Bonito P, Chiozzini C, Arenaccio C, Anticoli S, Manfredi F, Olivetta E, Ferrantelli F, Falcone E, Ruggieri A, Federico M. Antitumor HPV E7-specific CTL activity elicited by in vivo engineered exosomes produced through DNA inoculation. Int J Nanomedicine 2017; 12:4579-4591. [PMID: 28694699 PMCID: PMC5491702 DOI: 10.2147/ijn.s131309] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We recently proved that exosomes engineered in vitro to deliver high amounts of HPV E7 upon fusion with the Nefmut exosome-anchoring protein elicit an efficient anti-E7 cytotoxic T lymphocyte immune response. However, in view of a potential clinic application of this finding, our exosome-based immunization strategy was faced with possible technical difficulties including industrial manufacturing, cost of production, and storage. To overcome these hurdles, we designed an as yet unproven exosome-based immunization strategy relying on delivery by intramuscular inoculation of a DNA vector expressing Nefmut fused with HPV E7. In this way, we predicted that the expression of the Nefmut/E7 vector in muscle cells would result in a continuous source of endogenous (ie, produced by the inoculated host) engineered exosomes able to induce an E7-specific immune response. To assess this hypothesis, we first demonstrated that the injection of a Nefmut/green fluorescent protein-expressing vector led to the release of fluorescent exosomes, as detected in plasma of inoculated mice. Then, we observed that mice inoculated intramuscularly with a vector expressing Nefmut/E7 developed a CD8+ T-cell immune response against both Nef and E7. Conversely, no CD8+ T-cell responses were detected upon injection of vectors expressing either the wild-type Nef isoform of E7 alone, most likely a consequence of their inefficient exosome incorporation. The production of immunogenic exosomes in the DNA-injected mice was formally demonstrated by the E7-specific CD8+ T-cell immune response we detected in mice inoculated with exosomes isolated from plasma of mice inoculated with the Nefmut/E7 vector. Finally, we provide evidence that the injection of Nefmut/E7 DNA led to the generation of effective antigen-specific cytotoxic T lymphocytes whose activity was likely part of the potent, therapeutic antitumor effect we observed in mice implanted with TC-1 tumor cells. In summary, we established a novel method to generate immunogenic exosomes in vivo by the intramuscular inoculation of DNA vectors expressing the exosome-anchoring protein Nefmut and its derivatives.
Collapse
Affiliation(s)
- Paola Di Bonito
- Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Simona Anticoli
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | - Emiliana Falcone
- Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Rome, Italy
| | - Anna Ruggieri
- Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Rome, Italy
| | | |
Collapse
|
23
|
Manfredi F, di Bonito P, Ridolfi B, Anticoli S, Arenaccio C, Chiozzini C, Baz Morelli A, Federico M. The CD8⁺ T Cell-Mediated Immunity Induced by HPV-E6 Uploaded in Engineered Exosomes Is Improved by ISCOMATRIX TM Adjuvant. Vaccines (Basel) 2016; 4:vaccines4040042. [PMID: 27834857 PMCID: PMC5192362 DOI: 10.3390/vaccines4040042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/23/2016] [Accepted: 11/04/2016] [Indexed: 01/29/2023] Open
Abstract
We recently described the induction of an efficient CD8⁺ T cell-mediated immune response against a tumor-associated antigen (TAA) uploaded in engineered exosomes used as an immunogen delivery tool. This immune response cleared tumor cells inoculated after immunization, and controlled the growth of tumors implanted before immunization. We looked for new protocols aimed at increasing the CD8⁺ T cell specific response to the antigen uploaded in engineered exosomes, assuming that an optimized CD8⁺ T cell immune response would correlate with a more effective depletion of tumor cells in the therapeutic setting. By considering HPV-E6 as a model of TAA, we found that the in vitro co-administration of engineered exosomes and ISCOMATRIXTM adjuvant, i.e., an adjuvant composed of purified ISCOPREPTM saponin, cholesterol, and phospholipids, led to a stronger antigen cross-presentation in both B- lymphoblastoid cell lines ( and monocyte-derived immature dendritic cells compared with that induced by the exosomes alone. Consistently, the co-inoculation in mice of ISCOMATRIXTM adjuvant and engineered exosomes induced a significant increase of TAA-specific CD8⁺ T cells compared to mice immunized with the exosomes alone. This result holds promise for effective usage of exosomes as well as alternative nanovesicles in anti-tumor therapeutic approaches.
Collapse
Affiliation(s)
- Francesco Manfredi
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| | - Paola di Bonito
- Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| | - Barbara Ridolfi
- Department of Therapeutic Research and Medicine Evaluation, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| | - Simona Anticoli
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| | - Claudia Arenaccio
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| | - Chiara Chiozzini
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| | - Adriana Baz Morelli
- CSL, Ltd., Bio21 Institute, 30 Flemington Road, Melbourne, VIC 3010, Australia.
| | - Maurizio Federico
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| |
Collapse
|
24
|
Zhang X, Pei Z, Chen J, Ji C, Xu J, Zhang X, Wang J. Exosomes for Immunoregulation and Therapeutic Intervention in Cancer. J Cancer 2016; 7:1081-1087. [PMID: 27326251 PMCID: PMC4911875 DOI: 10.7150/jca.14866] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 04/26/2016] [Indexed: 02/06/2023] Open
Abstract
Exosomes, as a subset of extracellular vesicles, function as a mode of intercellular communication and molecular transfer, and facilitate the direct extracellular transfer of proteins, lipids, and miRNAs/mRNAs/DNAs between cells. Cancers have adapted exosomes and related microvesicles as a pathway that can suppress the immune system and establish a fertile local and distant environment to support neoplastic growth, invasion, and metastasis; these tumor-derived exosomes affect immunoregulation mechanisms, including immune activation and immune suppression. Immune cell-derived exosomes can modulate the immune response in cancer, which supports the belief that these membranous vesicles are immunotherapeutic reagents. In this review, we discuss the recent advances in the cancer immunotherapy, roles of exosomes in cancer, immunoregulation of tumor-derived exosomes, and immunomodulation by immune cell-derived exosomes. The topics covered here highlight novel insights into the development of efficient exosome-based cancer vaccines for cancer therapeutic intervention.
Collapse
Affiliation(s)
- Xuan Zhang
- 1. Scientific Research Center, Shanghai Public Health Clinical Center, 2901 Caolang Road, Jinshan District, Shanghai 201508, China
| | - Zenglin Pei
- 1. Scientific Research Center, Shanghai Public Health Clinical Center, 2901 Caolang Road, Jinshan District, Shanghai 201508, China
| | - Jinyun Chen
- 2. Departments of Epidemiology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 770030, USA
| | - Chunxia Ji
- 1. Scientific Research Center, Shanghai Public Health Clinical Center, 2901 Caolang Road, Jinshan District, Shanghai 201508, China
| | - Jianqing Xu
- 1. Scientific Research Center, Shanghai Public Health Clinical Center, 2901 Caolang Road, Jinshan District, Shanghai 201508, China
| | - Xiaoyan Zhang
- 1. Scientific Research Center, Shanghai Public Health Clinical Center, 2901 Caolang Road, Jinshan District, Shanghai 201508, China
| | - Jin Wang
- 1. Scientific Research Center, Shanghai Public Health Clinical Center, 2901 Caolang Road, Jinshan District, Shanghai 201508, China
| |
Collapse
|
25
|
Robbins PD, Dorronsoro A, Booker CN. Regulation of chronic inflammatory and immune processes by extracellular vesicles. J Clin Invest 2016; 126:1173-80. [PMID: 27035808 DOI: 10.1172/jci81131] [Citation(s) in RCA: 219] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Almost all cell types release extracellular vesicles (EVs), which are derived either from multivesicular bodies or from the plasma membrane. EVs contain a subset of proteins, lipids, and nucleic acids from the cell from which they are derived. EV factors, particularly small RNAs such as miRNAs, likely play important roles in cell-to-cell communication both locally and systemically. Most of the functions associated with EVs are in the regulation of immune responses to pathogens and cancer, as well as in regulating autoimmunity. This Review will focus on the different modes of immune regulation, both direct and indirect, by EVs. The therapeutic utility of EVs for the regulation of immune responses will also be discussed.
Collapse
|
26
|
Abstract
Engineering exosomes to upload heterologous proteins represents the last frontier in terms of nanoparticle-based technology. A limited number of methods suitable to associate proteins to exosome membrane has been described so far, and very little is known regarding the possibility to upload proteins inside exosomes. We optimized a method of protein incorporation in exosomes by exploiting the unique properties of a nonfunctional mutant of the HIV-1 Nef protein referred to as Nef(mut). It incorporates at high extents in exosomes meanwhile acting as carrier of protein antigens fused at its C-terminus. Manipulating Nef(mut) allows the incorporation into exosomes of high amounts of heterologous proteins which thus remain protected from external neutralization/degradation factors. These features, together with flexibility in terms of incorporation of foreign antigens and ease of production, make Nef(mut)-based exosomes a convenient vehicle for different applications (e.g., protein transduction, immunization) whose performances are comparable with those of alternative, more complex nanoparticle-based delivery systems.
Collapse
|
27
|
The Dichotomy of Tumor Exosomes (TEX) in Cancer Immunity: Is It All in the ConTEXt? Vaccines (Basel) 2015; 3:1019-51. [PMID: 26694473 PMCID: PMC4693230 DOI: 10.3390/vaccines3041019] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/24/2015] [Accepted: 12/05/2015] [Indexed: 02/06/2023] Open
Abstract
Exosomes are virus-sized nanoparticles (30–130 nm) formed intracellularly as intravesicular bodies/intralumenal vesicles within maturing endosomes (“multivesicular bodies”, MVBs). If MVBs fuse with the cell’s plasma membrane, the interior vesicles may be released extracellularly, and are termed “exosomes”. The protein cargo of exosomes consists of cytosolic, membrane, and extracellular proteins, along with membrane-derived lipids, and an extraordinary variety of nucleic acids. As such, exosomes reflect the status and identity of the parent cell, and are considered as tiny cellular surrogates. Because of this closely entwined relationship between exosome content and the source/status of the parental cell, conceivably exosomes could be used as vaccines against various pathologies, as they contain antigens associated with a given disease, e.g., cancer. Tumor-derived exosomes (TEX) have been shown to be potent anticancer vaccines in animal models, driving antigen-specific T and B cell responses, but much recent literature concerning TEX strongly places the vesicles as powerfully immunosuppressive. This dichotomy suggests that the context in which the immune system encounters TEX is critical in determining immune stimulation versus immunosuppression. Here, we review literature on both sides of this immune coin, and suggest that it may be time to revisit the concept of TEX as anticancer vaccines in clinical settings.
Collapse
|
28
|
Mahmoodzadeh Hosseini H, Halabian R, Amin M, Imani Fooladi AA. Texosome-based drug delivery system for cancer therapy: from past to present. Cancer Biol Med 2015; 12:150-62. [PMID: 26487960 PMCID: PMC4607826 DOI: 10.7497/j.issn.2095-3941.2015.0045] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Rising worldwide cancer incidence and resistance to current anti-cancer drugs necessitate the need for new pharmaceutical compounds and drug delivery system. Malfunction of the immune system, particularly in the tumor microenvironment, causes tumor growth and enhances tumor progression. Thus, cancer immunotherapy can be an appropriate approach to provoke the systemic immune system to combat tumor expansion. Texosomes, which are endogenous nanovesicles released by all tumor cells, contribute to cell-cell communication and modify the phenotypic features of recipient cells due to the texosomes' ability to transport biological components. For this reason, texosome-based delivery system can be a valuable strategy for therapeutic purposes. To improve the pharmaceutical behavior of this system and to facilitate its use in medical applications, biotechnology approaches and mimetic techniques have been utilized. In this review, we present the development history of texosome-based delivery systems and discuss the advantages and disadvantages of each system.
Collapse
Affiliation(s)
- Hamideh Mahmoodzadeh Hosseini
- 1 Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran ; 2 Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417653861, Iran
| | - Raheleh Halabian
- 1 Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran ; 2 Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417653861, Iran
| | - Mohsen Amin
- 1 Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran ; 2 Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417653861, Iran
| | - Abbas Ali Imani Fooladi
- 1 Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran ; 2 Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417653861, Iran
| |
Collapse
|
29
|
Staphylococcal enterotoxin B/texosomes as a candidate for breast cancer immunotherapy. Tumour Biol 2015; 37:739-48. [DOI: 10.1007/s13277-015-3877-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 07/30/2015] [Indexed: 10/23/2022] Open
|
30
|
De Toro J, Herschlik L, Waldner C, Mongini C. Emerging roles of exosomes in normal and pathological conditions: new insights for diagnosis and therapeutic applications. Front Immunol 2015; 6:203. [PMID: 25999947 PMCID: PMC4418172 DOI: 10.3389/fimmu.2015.00203] [Citation(s) in RCA: 455] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/14/2015] [Indexed: 12/16/2022] Open
Abstract
From the time when they were first described in the 1970s by the group of Johnstone and Stahl, exosomes are a target of constant research. Exosomes belong to the family of nanovesicles which are of great interest for their many functions and potential for diagnosis and therapy in multiples diseases. Exosomes originate from the intraluminal vesicles of late endosomal compartments named multivesicular bodies and the fusion of these late endosomes with the cell membrane result in the release of the vesicles into the extracellular compartment. Moreover, their generation can be induced by many factors including extracellular stimuli, such as microbial attack and other stress conditions. The primary role attributed to exosomes was the removal of unnecessary proteins from the cells. Now, several studies have demonstrated that exosomes are involved in cell–cell communication, even though their biological function is not completely clear. The participation of exosomes in cancer is the field of microvesicle research that has expanded more over the last years. Evidence proving that exosomes derived from tumor-pulsed dendritic cells, neoplastic cells, and malignant effusions are able to present antigens to T-cells, has led to numerous studies using them as cell-free cancer vaccines. Because exosomes derive from all cell types, they contain proteins, lipids, and micro RNA capable of regulating a variety of target genes. Much research is being conducted, which focuses on the employment of these vesicles as biomarkers in the diagnosis of cancer in addition to innovative biomarkers for diagnosis, prognosis, and management of cardiovascular diseases. Interesting findings indicating the role of exosomes in the pathogenesis of several diseases have encouraged researchers to consider their therapeutic potential not only in oncology but also in the treatment of autoimmune syndromes and neurodegenerative disorders such as Alzheimer’s and Parkinson’s disease, in addition to infectious diseases such as tuberculosis, diphtheria, and toxoplasmosis as well as infections caused by prions or viruses such as HIV. The aim of this review is to disclose the emerging roles of exosomes in normal and pathological conditions and to discuss their potential therapeutic applications.
Collapse
Affiliation(s)
- Julieta De Toro
- Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Leticia Herschlik
- Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Claudia Waldner
- Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Claudia Mongini
- Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires , Buenos Aires , Argentina
| |
Collapse
|
31
|
Liu Y, Gu Y, Cao X. The exosomes in tumor immunity. Oncoimmunology 2015; 4:e1027472. [PMID: 26405598 DOI: 10.1080/2162402x.2015.1027472] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 02/28/2015] [Accepted: 03/03/2015] [Indexed: 02/08/2023] Open
Abstract
Exosomes are a kind of nanometric membrane vesicles and can be released by almost all kinds of cells, including cancer cells. As the important mediators in intercellular communications, exosomes mediate exchange of protein and genetic material derived from parental cells. Emerging evidences show that exosomes secreted by either host cells or cancer cells are involved in tumor initiation, growth, invasion and metastasis. Moreover, communications between immune cells and cancer cells via exosomes play dual roles in modulating tumor immunity. In this review, we focus on exosome-mediated immunosuppression via inhibition of antitumor responses elicited by immune cells (DCs, NK cells, CD4+ and CD8+ T cells, etc.) and induction of immunosuppressive or regulatory cell populations (MDSCs, Tregs and Bregs). Transfer of cytokines, microRNAs (miRNAs) and functional mRNAs by tumor-derived exosomes (TEXs) is crucial in the immune escape. Furthermore, exosomes secreted from several kinds of immune cells (DCs, CD4+ and CD8+ Tregs) also participate in immunosuppression. On the other hand, we summarize the current application of DC-derived and modified tumor-derived exosomes as tumor vaccines. The potential challenges about exosome-based vaccines for clinical application are also discussed.
Collapse
Affiliation(s)
- Yanfang Liu
- National Key Laboratory of Medical Immunology & Institute of Immunology; Second Military Medical University ; Shanghai, China
| | - Yan Gu
- National Key Laboratory of Medical Immunology & Institute of Immunology; Second Military Medical University ; Shanghai, China
| | - Xuetao Cao
- National Key Laboratory of Medical Immunology & Institute of Immunology; Second Military Medical University ; Shanghai, China
| |
Collapse
|
32
|
Di Bonito P, Ridolfi B, Columba-Cabezas S, Giovannelli A, Chiozzini C, Manfredi F, Anticoli S, Arenaccio C, Federico M. HPV-E7 delivered by engineered exosomes elicits a protective CD8⁺ T cell-mediated immune response. Viruses 2015; 7:1079-99. [PMID: 25760140 PMCID: PMC4379561 DOI: 10.3390/v7031079] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 02/20/2015] [Accepted: 02/28/2015] [Indexed: 12/14/2022] Open
Abstract
We developed an innovative strategy to induce a cytotoxic T cell (CTL) immune response against protein antigens of choice. It relies on the production of exosomes, i.e., nanovesicles spontaneously released by all cell types. We engineered the upload of huge amounts of protein antigens upon fusion with an anchoring protein (i.e., HIV-1 Nefmut), which is an inactive protein incorporating in exosomes at high levels also when fused with foreign proteins. We compared the immunogenicity of engineered exosomes uploading human papillomavirus (HPV)-E7 with that of lentiviral virus-like particles (VLPs) incorporating equivalent amounts of the same antigen. These exosomes, whose limiting membrane was decorated with VSV-G, i.e., an envelope protein inducing pH-dependent endosomal fusion, proved to be as immunogenic as the cognate VLPs. It is noteworthy that the immunogenicity of the engineered exosomes remained unaltered in the absence of VSV-G. Most important, we provide evidence that the inoculation in mouse of exosomes uploading HPV-E7 induces production of anti-HPV E7 CTLs, blocks the growth of syngeneic tumor cells inoculated after immunization, and controls the development of tumor cells inoculated before the exosome challenge. These results represent the proof-of-concept about both feasibility and efficacy of the Nefmut-based exosome platform for the induction of CD8+ T cell immunity.
Collapse
Affiliation(s)
- Paola Di Bonito
- Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Barbara Ridolfi
- Department of Therapeutic Research and Medicine Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Sandra Columba-Cabezas
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Andrea Giovannelli
- Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Chiara Chiozzini
- National AIDS Center, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | | | - Simona Anticoli
- National AIDS Center, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Claudia Arenaccio
- National AIDS Center, Istituto Superiore di Sanità, 00161 Rome, Italy.
- Department of Science, University Roma Tre, 00146 Rome, Italy.
| | - Maurizio Federico
- National AIDS Center, Istituto Superiore di Sanità, 00161 Rome, Italy.
| |
Collapse
|
33
|
Mahmoodzadeh Hosseini H, Soleimanirad J, Mehdizadeh Aghdam E, Amin M, Imani Fooladi AA. Texosome-anchored superantigen triggers apoptosis in original ovarian cancer cells. Med Oncol 2014; 32:409. [DOI: 10.1007/s12032-014-0409-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 11/30/2022]
|
34
|
Braicu C, Tomuleasa C, Monroig P, Cucuianu A, Berindan-Neagoe I, Calin GA. Exosomes as divine messengers: are they the Hermes of modern molecular oncology? Cell Death Differ 2014; 22:34-45. [PMID: 25236394 DOI: 10.1038/cdd.2014.130] [Citation(s) in RCA: 231] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 06/24/2014] [Accepted: 07/07/2014] [Indexed: 12/18/2022] Open
Abstract
Exosomes are cell-derived vesicles that convey key elements with the potential to modulate intercellular communication. They are known to be secreted from all types of cells, and are crucial messengers that can regulate cellular processes by 'trafficking' molecules from cells of one tissue to another. The exosomal content has been shown to be broad, composed of different types of cytokines, growth factors, proteins, or nucleic acids. Besides messenger RNA (mRNA) they can also contain noncoding transcripts such as microRNAs (miRNAs), which are small endogenous cellular regulators of protein expression. In diseases such as cancer, exosomes can facilitate tumor progression by altering their vesicular content and supplying the tumor niche with molecules that favor the progression of oncogenic processes such as proliferation, invasion and metastasis, or even drug resistance. The packaging of their molecular content is known to be tissue specific, a fact that makes them interesting tools in clinical diagnostics and ideal candidates for biomarkers. In the current report, we describe the main properties of exosomes and explain their involvement in processes such as cell differentiation and cell death. Furthermore, we emphasize the need of developing patient-targeted treatments by applying the conceptualization of exosomal-derived miRNA-based therapeutics.
Collapse
Affiliation(s)
- C Braicu
- Research Center for Functional Genomics and Translational Medicine, 'Iuliu Hatieganu' University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - C Tomuleasa
- 1] Research Center for Functional Genomics and Translational Medicine, 'Iuliu Hatieganu' University of Medicine and Pharmacy, Cluj-Napoca, Romania [2] Department of Hematology, 'Ion Chiricuta' Oncology Institute, Cluj-Napoca, Romania
| | - P Monroig
- 1] Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA [2] University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - A Cucuianu
- 1] Department of Hematology, 'Ion Chiricuta' Oncology Institute, Cluj-Napoca, Romania [2] Department of Hematology, 'Iuliu Hatieganu' University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - I Berindan-Neagoe
- 1] Research Center for Functional Genomics and Translational Medicine, 'Iuliu Hatieganu' University of Medicine and Pharmacy, Cluj-Napoca, Romania [2] Department of Immunology, 'Iuliu Hatieganu' University of Medicine and Pharmacy, Cluj-Napoca, Romania [3] Department of Functional Genomics and Experimental Pathology, 'Ion Chiricuta' Oncology Institute, Cluj-Napoca, Romania
| | - G A Calin
- 1] Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA [2] Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
35
|
Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat Rev Immunol 2014. [PMID: 24566916 DOI: 10.1038/nri362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Extracellular vesicles, including exosomes, are small membrane vesicles derived from multivesicular bodies or from the plasma membrane. Most, if not all, cell types release extracellular vesicles, which then enter the bodily fluids. These vesicles contain a subset of proteins, lipids and nucleic acids that are derived from the parent cell. It is thought that extracellular vesicles have important roles in intercellular communication, both locally and systemically, as they transfer their contents, including proteins, lipids and RNAs, between cells. Extracellular vesicles are involved in numerous physiological processes, and vesicles from both non-immune and immune cells have important roles in immune regulation. Moreover, extracellular vesicle-based therapeutics are being developed and clinically tested for the treatment of inflammatory diseases, autoimmune disorders and cancer. Given the tremendous therapeutic potential of extracellular vesicles, this Review focuses on their role in modulating immune responses, as well as their potential therapeutic applications.
Collapse
Affiliation(s)
- Paul D Robbins
- Department of Metabolism and Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter, Florida 33458, USA
| | - Adrian E Morelli
- Departments of Surgery and Immunology and T.E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| |
Collapse
|
36
|
Thuma F, Zöller M. Outsmart tumor exosomes to steal the cancer initiating cell its niche. Semin Cancer Biol 2014; 28:39-50. [PMID: 24631836 DOI: 10.1016/j.semcancer.2014.02.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Accepted: 02/22/2014] [Indexed: 12/14/2022]
Abstract
Exosomes are small vesicles that derive from endosomes and are delivered by many cells, including tumor cells that are a particular rich source of exosomes. Exosomes are suggested to be the most potent intercellular communicators. Being recovered in all body fluids, they can communicate with neighboring as well as distant cells. The latter was first described for dendritic cell exosomes that can initiate T cell activation. However, tumor exosomes (TEX) may impede this crosstalk. Besides with hematopoietic cells, TEX communicate with the tumor cell itself, but also with host stroma cells and endothelial cells. This crosstalk received much attention as there is strong evidence that TEX account for angiogenesis and premetastatic niche formation, which may proceed directly via binding and uptake of TEX by cells in the premetastatic organ or indirectly via TEX being taken up by hematopoietic progenitors in the bone marrow (BM), which mature toward lineages with immunosuppressive features or are forced toward premature release from the BM and homing into premetastatic organs. Knowing these deleterious activities of TEX, it becomes demanding to search for modes of therapeutic interference. I here introduce our hypothesis that metastasis formation may be hampered by tailored exosomes that outsmart TEX. The essential prerequisites are an in depth knowledge on TEX binding, uptake, binding-initiated signal transduction and uptake-promoted target cell reprogramming.
Collapse
Affiliation(s)
- Florian Thuma
- Department of Tumor Cell Biology, University Hospital of Surgery and German Cancer Research Center, Heidelberg, Germany
| | - Margot Zöller
- Department of Tumor Cell Biology, University Hospital of Surgery and German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
37
|
Abstract
Extracellular vesicles, including exosomes, are small membrane vesicles derived from multivesicular bodies or from the plasma membrane. Most, if not all, cell types release extracellular vesicles, which then enter the bodily fluids. These vesicles contain a subset of proteins, lipids and nucleic acids that are derived from the parent cell. It is thought that extracellular vesicles have important roles in intercellular communication, both locally and systemically, as they transfer their contents, including proteins, lipids and RNAs, between cells. Extracellular vesicles are involved in numerous physiological processes, and vesicles from both non-immune and immune cells have important roles in immune regulation. Moreover, extracellular vesicle-based therapeutics are being developed and clinically tested for the treatment of inflammatory diseases, autoimmune disorders and cancer. Given the tremendous therapeutic potential of extracellular vesicles, this Review focuses on their role in modulating immune responses, as well as their potential therapeutic applications.
Collapse
Affiliation(s)
- Paul D. Robbins
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, Florida
| | - Adrian E. Morelli
- Departments of Surgery and Immunology and T.E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
38
|
Fleming A, Sampey G, Chung MC, Bailey C, van Hoek ML, Kashanchi F, Hakami RM. The carrying pigeons of the cell: exosomes and their role in infectious diseases caused by human pathogens. Pathog Dis 2014; 71:109-20. [PMID: 24449527 DOI: 10.1111/2049-632x.12135] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 01/07/2014] [Indexed: 12/15/2022] Open
Abstract
Exosomes have recently been classified as the newest family members of 'bioactive vesicles' that function to promote intercellular communication. Long ignored and thought to be only a mechanism by which cellular waste is removed, exosomes have garnered a huge amount of interest in recent years as their critical functions in maintaining homeostasis through intercellular communication and also in different types of diseases have been demonstrated. Many groundbreaking studies of exosome functions have been performed in the cancer field and the infectious disease areas of study, revealing the importance and also the fascinating complexity of exosomal packaging, targeting, and functions. Selective packaging of exosomes in response to the type of infection, exosomal modulation of the immune response and host signaling pathways, exosomal regulation of pathogen spread, and effects of exosomes on the degree of pathogenesis have all been well documented. In this review, we provide a synthesis of the current understanding of the role of exosomes during infections caused by human pathogens and discuss the implications of these findings for a better understanding of pathogenic mechanisms and future therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Adam Fleming
- School of Systems Biology, The National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Wang J, Wang L, Lin Z, Tao L, Chen M. More efficient induction of antitumor T cell immunity by exosomes from CD40L gene-modified lung tumor cells. Mol Med Rep 2013; 9:125-31. [PMID: 24173626 DOI: 10.3892/mmr.2013.1759] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 10/08/2013] [Indexed: 11/06/2022] Open
Abstract
The incidence of lung cancer increases annually. However, the effects of the present methods for the treatment of lung cancer are extremely poor. It has been reported that exosomes from heat‑stressed 3LL Lewis lung tumor cells effectively elicit systemic antitumor immunity. CD40 signaling is critical in the activation of dendritic cells (DCs), which are important in the induction of antitumor immunity. In the present study, exosomes from CD40 ligand gene‑modified 3LL tumor cells (CD40L‑EXO) were identified to be more immunogenic compared with control‑EXO and lac Z-EXO. CD40L‑EXO induced a more mature phenotype of the DCs and promoted them to secrete high levels of interleukin‑12. CD40L‑EXO‑treated DCs induced a greater proliferation of allogeneic T cells in the mixed lymphocyte reaction. Moreover, CD40L‑EXO induced robust tumor antigen‑specific CD4+ T cell proliferation ex vivo. CD40L‑EXO were also extremely effective in the protective and therapeutic antitumor tests in vivo. These results indicate that CD40L‑EXO may be used as an efficient vaccine for lung cancer immunotherapy.
Collapse
Affiliation(s)
- Jiaoli Wang
- Department of Respiratory Medicine, Hangzhou First People's Hospital, Hangzhou, Zhejiang 310006, P.R. China
| | | | | | | | | |
Collapse
|
40
|
Zech D, Rana S, Büchler MW, Zöller M. Tumor-exosomes and leukocyte activation: an ambivalent crosstalk. Cell Commun Signal 2012. [PMID: 23190502 PMCID: PMC3519567 DOI: 10.1186/1478-811x-10-37] [Citation(s) in RCA: 180] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Tumor-exosomes being reported to suppress or promote a cancer-directed immune response, we used exosomes of the rat pancreatic adenocarcinoma BSp73ASML (ASML) to evaluate, whether and which steps in immune response induction can be affected by tumor-exosomes and how the impaired responsiveness can be circumvented. Results ASML-exosomes bind to and are taken up by all leukocyte subpopulations in vivo and in vitro, uptake by CD11b+ leukocytes exceeding that by T and B cells. ASML-exosomes affect leukocyte proliferation via reduced CD44v6 up-regulation and lck, ZAP70 and ERK1,2 phosphorylation, which can be compensated by dendritic cells (DC). ASML-exosomes do not support Treg. Yet, impaired activation of anti-apoptotic signals is accompanied by slightly increased apoptosis susceptibility. IgM secretion is unaffected; NK and CTL activity are strengthened, ASML-exosomes co-operating with DC in CTL activation. ASML-exosomes transiently interfere with leukocyte migration by occupying migration-promoting receptors CD44, CD49d, CD62L and CD54 during binding/internalization. Conclusion ASML-exosomes might well serve as adjuvant in immunotherapy as they support leukocyte effector functions and have only a minor impact on leukocyte activation, which can be overridden by DC. However, exosome-induced modulation of immune cells relies, at least in part, on exosome uptake and message transfer. This implies that depending on the individual tumor's exosome composition, exosomes may distinctly affect the immune system. Nonetheless, whether immunotherapy can profit from using tumor-exosomes as adjuvant can easily be settled beforehand in vitro.
Collapse
Affiliation(s)
- Daniela Zech
- Department of Tumor Cell Biology, University Hospital of Surgery, Im Neuenheimer Feld 365, D-69120, Heidelberg, Germany.
| | | | | | | |
Collapse
|
41
|
Lai CPK, Breakefield XO. Role of exosomes/microvesicles in the nervous system and use in emerging therapies. Front Physiol 2012; 3:228. [PMID: 22754538 PMCID: PMC3384085 DOI: 10.3389/fphys.2012.00228] [Citation(s) in RCA: 244] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 06/06/2012] [Indexed: 12/27/2022] Open
Abstract
Extracellular membrane vesicles (EMVs) are nanometer sized vesicles, including exosomes and microvesicles capable of transferring DNAs, mRNAs, microRNAs, non-coding RNAs, proteins, and lipids among cells without direct cell-to-cell contact, thereby representing a novel form of intercellular communication. Many cells in the nervous system have been shown to release EMVs, implicating their active roles in development, function, and pathologies of this system. While substantial progress has been made in understanding the biogenesis, biophysical properties, and involvement of EMVs in diseases, relatively less information is known about their biological function in the normal nervous system. In addition, since EMVs are endogenous vehicles with low immunogenicity, they have also been actively investigated for the delivery of therapeutic genes/molecules in treatment of cancer and neurological diseases. The present review summarizes current knowledge about EMV functions in the nervous system under both physiological and pathological conditions, as well as emerging EMV-based therapies that could be applied to the nervous system in the foreseeable future.
Collapse
Affiliation(s)
- Charles Pin-Kuang Lai
- Department of Neurology, Neuroscience Center, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School Boston, MA, USA
| | | |
Collapse
|
42
|
Federico M. From virus-like particles to engineered exosomes for a new generation of vaccines. Future Virol 2012. [DOI: 10.2217/fvl.12.29] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Over the last two decades, virus-like particles (VLPs) have been the focus of countless investigations on innovative vaccines. The number of monotypic, multipartite and chimeric VLP-based vaccines proposed have increased even further in the last few years as part of the continuous effort to improve the safety, efficacy and cost–effectiveness of immunogens. As compared with monomer- or subunit-based vaccines, VLPs show several advantages in terms of potency of the elicited immune responses. Chimeric VLPs are quite flexible tools to accommodate foreign peptides, cell proteins and nonself-assembling viral products. However, their use often meets with still unresolved hurdles such as induction of undesired immune responses, neutralization by pre-existing immunity and complex methods of production. Among strategies aimed at developing new nanoparticle-based vaccines, exosomes hold much promise. They are nanovesicles constitutively released by eukaryotic cells that originate from intraluminal vesicles accumulating in multivesicular bodies. Exosomes have immunogenic properties, the strength of which correlates with the amounts of associated antigens. Engineering antigens of interest to target them in exosomes represents the last frontier in terms of nanoparticle-based vaccines.
Collapse
Affiliation(s)
- Maurizio Federico
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
| |
Collapse
|
43
|
Zhang HG, Zhuang X, Sun D, Liu Y, Xiang X, Grizzle WE. Exosomes and immune surveillance of neoplastic lesions: a review. Biotech Histochem 2012; 87:161-8. [PMID: 22216980 DOI: 10.3109/10520291003659042] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The immune system has been reported to suppress the development and progression of neoplastic lesions; however, the exact mechanisms by which neoplastic lesions and the immune system interact are not well understood. Within the last decade, tiny membrane bound particles, approximately 30-100 nm in diameter, have been observed in the blood and other body fluids. These particles, currently called exosomes, are released from many types of tissues including tumors, and they contain and carry many proteins, and mRNAs and microRNA species. We review here how tumors suppress the immune system, especially by the formation of exosomes. Exosomes released from tumors are carried in part by the vascular system to distant cells, which phagocytose them. Depending on the proteins, mRNAs or microRNAs in the exosomes and the cell type, phagocytosis of exosomes may provide a modulating signal to the cell. In the case of exosomes from tumors, uptake of the exosomes by cells of the immune system has been reported to have three main effects: 1) suppression of the number and activity of natural killer cells, 2) suppression of the activity of T cells and 3) suppression of the number and maturation of mature dendritic cells.
Collapse
Affiliation(s)
- H-G Zhang
- James Brown Cancer Center, Department of Microbiology & Immunology, University of Louisville, Louisville, KY 40202, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Zhong H, Yang Y, Ma S, Xiu F, Cai Z, Zhao H, Du L. Induction of a tumour-specific CTL response by exosomes isolated from heat-treated malignant ascites of gastric cancer patients. Int J Hyperthermia 2011; 27:604-11. [PMID: 21846196 DOI: 10.3109/02656736.2011.564598] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Tumour cell-derived exosomes may represent a novel type of cancer vaccine. However, the immunogenicity of exosomes derived from tumour cells has been shown to be poor. Therefore, in this study, exosome immunogenicity following heat treatment of exosomes from malignant ascites obtained from gastric cancer patients was evaluated. MATERIALS AND METHODS Tumour-derived exosomes were isolated from heat-treated and untreated malignant ascites of gastric cancer patients using serial centrifugation and sucrose gradient ultracentrifugation. Next, in vitro experiments were performed to investigate the influence of heat treatment on exosome immunogenicity. RESULTS Exosomes from heat-treated malignant ascites of gastric cancer patients (HS exosomes) were found to contain higher concentrations of heat shock proteins, Hsp70 and Hsp60, than exosomes derived from untreated malignant ascites obtained from gastric cancer patients. Additional in vitro studies suggest that exosomes derived from heat-treated malignant ascites are able to promote dendritic cell (DC) maturation and induce a tumour-specific cytotoxic T lymphocyte (CTL) response. CONCLUSIONS Overall, these results demonstrate that exposure to heat stress can improve the immunogenicity of exosomes obtained from malignant ascites of gastric cancer patients.
Collapse
Affiliation(s)
- Haijun Zhong
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
45
|
The roles of tumor-derived exosomes in cancer pathogenesis. Clin Dev Immunol 2011; 2011:842849. [PMID: 22190973 PMCID: PMC3235485 DOI: 10.1155/2011/842849] [Citation(s) in RCA: 228] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2011] [Accepted: 08/28/2011] [Indexed: 12/15/2022]
Abstract
Exosomes are endosome-derived, 30–100 nm small membrane vesicles released by most cell types including tumor cells. They are enriched in a selective repertoire of proteins and nucleic acids from parental cells and are thought to be actively involved in conferring intercellular signals. Tumor-derived exosomes have been viewed as a source of tumor antigens that can be used to induce antitumor immune responses. However, tumor-derived exosomes also have been found to possess immunosuppressive properties and are able to facilitate tumor growth, metastasis, and the development of drug resistance. These different effects of tumor-derived exosomes contribute to the pathogenesis of cancer. This review will discuss the roles of tumor-derived exosomes in cancer pathogenesis, therapy, and diagnostics.
Collapse
|
46
|
Moon PG, You S, Lee JE, Hwang D, Baek MC. Urinary exosomes and proteomics. MASS SPECTROMETRY REVIEWS 2011; 30:1185-1202. [PMID: 21544848 DOI: 10.1002/mas.20319] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Revised: 07/23/2010] [Accepted: 07/23/2010] [Indexed: 05/30/2023]
Abstract
A number of highly abundant proteins in urine have been identified through proteomics approaches, and some have been considered as disease-biomarker candidates. These molecules might be clinically useful in diagnosis of various diseases. However, none has proven to be specifically indicative of perturbations of cellular processes in cells associated with urogenital diseases. Exosomes could be released into urine which flows through the kidney, ureter, bladder and urethra, with a process of filtration and reabsorption. Urinary exosomes have been recently suggested as alternative materials that offer new opportunities to identify useful biomarkers, because these exosomes secreted from epithelial cells lining the urinary track might reflect the cellular processes associated with the pathogenesis of diseases in their donor cells. Proteomic analysis of such urinary exosomes assists the search of urinary biomarkers reflecting pathogenesis of various diseases and also helps understanding the function of urinary exosomes in urinary systems. Thus, it has been recently suggested that urinary exosomes are one of the most valuable targets for biomarker development and to understand pathophysiology of relevant diseases.
Collapse
Affiliation(s)
- Pyong-Gon Moon
- Department of Molecular Medicine, Cell and Matrix Biology Research Institute, School of Medicine, Kyungpook National University, Daegu 700-422, Republic of Korea
| | | | | | | | | |
Collapse
|
47
|
Yang C, Kim SH, Bianco NR, Robbins PD. Tumor-derived exosomes confer antigen-specific immunosuppression in a murine delayed-type hypersensitivity model. PLoS One 2011; 6:e22517. [PMID: 21829629 PMCID: PMC3149056 DOI: 10.1371/journal.pone.0022517] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 06/25/2011] [Indexed: 12/21/2022] Open
Abstract
Exosomes are endosome-derived small membrane vesicles that are secreted by most cell types including tumor cells. Tumor-derived exosomes usually contain tumor antigens and have been used as a source of tumor antigens to stimulate anti-tumor immune responses. However, many reports also suggest that tumor-derived exosomes can facilitate tumor immune evasion through different mechanisms, most of which are antigen-independent. In the present study we used a mouse model of delayed-type hypersensitivity (DTH) and demonstrated that local administration of tumor-derived exosomes carrying the model antigen chicken ovalbumin (OVA) resulted in the suppression of DTH response in an antigen-specific manner. Analysis of exosome trafficking demonstrated that following local injection, tumor-derived exosomes were internalized by CD11c+ cells and transported to the draining LN. Exosome-mediated DTH suppression is associated with increased mRNA levels of TGF-β1 and IL-4 in the draining LN. The tumor-derived exosomes examined were also found to inhibit DC maturation. Taken together, our results suggest a role for tumor-derived exosomes in inducing tumor antigen-specific immunosuppression, possibly by modulating the function of APCs.
Collapse
Affiliation(s)
- Chenjie Yang
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Seon-Hee Kim
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Nicole R. Bianco
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Paul D. Robbins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
48
|
Kato M, Nakamura Y, Suda T, Ozawa Y, Inui N, Seo N, Nagata T, Koide Y, Kalinski P, Nakamura H, Chida K. Enhanced anti-tumor immunity by superantigen-pulsed dendritic cells. Cancer Immunol Immunother 2011; 60:1029-38. [PMID: 21519830 PMCID: PMC11029592 DOI: 10.1007/s00262-011-1015-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 03/31/2011] [Indexed: 12/25/2022]
Abstract
Staphylococcal enterotoxins A (SEA) and B (SEB) are classical models of superantigens (SAg), which induce potent T-cell-stimulating activity by forming complexes with MHC class II molecules on antigen-presenting cells. This large-scale activation of T-cells is accompanied by increased production of cytokines such as interferon-γ (IFN-γ). Additionally, as we previously reported, IFN-γ-producing CD8(+) T cells act as "helper cells," supporting the ability of dendritic cells to produce interleukin-12 (IL-12)p70. Here, we show that DC pulsed with SAg promote the enhancement of anti-tumor immunity. Murine bone marrow-derived dendritic cells (DC) were pulsed with OVA(257-264) (SIINFEKL), which is an H-2Kb target epitope of EG7 [ovalbumin (OVA)-expressing EL4] cell lines, in the presence of SEA and SEB and were subcutaneously injected into naïve C57BL/6 mice. SAg plus OVA(257-264)-pulsed DC vaccine strongly enhanced peptide-specific CD8(+) T cells exhibiting OVA(257-264)-specific cytotoxic activity and IFN-γ production, leading to the induction of protective immunity against EG7 tumors. Furthermore, cyclophosphamide (CY) added to SAg plus tumor-antigens (OVA(257-264), tumor lysate, or TRP-2) pulsed DC immunization markedly enhanced tumor-specific T-cell expansion and had a significant therapeutic effect against various tumors (EG7, 2LL, and B16). Superantigens are potential candidates for enhancing tumor immunity in DC vaccines.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Antineoplastic Agents, Alkylating/therapeutic use
- CD8-Positive T-Lymphocytes
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/metabolism
- Cyclophosphamide/therapeutic use
- Cytokines/metabolism
- Dendritic Cells/immunology
- Flow Cytometry
- Histocompatibility Antigens Class II/metabolism
- Interferon-gamma/metabolism
- Interleukin-12/metabolism
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/metabolism
- Lymphocyte Activation
- Lymphoma/drug therapy
- Lymphoma/immunology
- Lymphoma/metabolism
- Male
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Mice
- Mice, Inbred C57BL
- Ovalbumin/physiology
- Receptors, G-Protein-Coupled/physiology
- Superantigens/immunology
- Survival Rate
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Helper-Inducer/immunology
- Tumor Cells, Cultured
- Vaccines, Subunit/therapeutic use
Collapse
Affiliation(s)
- Masato Kato
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| | - Yutaro Nakamura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| | - Yuichi Ozawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
- Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naohiro Seo
- Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Toshi Nagata
- Department of Health Science, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yukio Koide
- Department of Infectious Diseases, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Pawel Kalinski
- Department of Surgery, Immunology, Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA USA
| | - Hirotoshi Nakamura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| | - Kingo Chida
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| |
Collapse
|
49
|
Xie Y, Bai O, Zhang H, Yuan J, Zong S, Chibbar R, Slattery K, Qureshi M, Wei Y, Deng Y, Xiang J. Membrane-bound HSP70-engineered myeloma cell-derived exosomes stimulate more efficient CD8(+) CTL- and NK-mediated antitumour immunity than exosomes released from heat-shocked tumour cells expressing cytoplasmic HSP70. J Cell Mol Med 2011; 14:2655-66. [PMID: 19627400 PMCID: PMC4373481 DOI: 10.1111/j.1582-4934.2009.00851.x] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Exosomes (EXO) derived from tumour cells have been used to stimulate antitumour immune responses, but only resulting in prophylatic immunity. Tumour-derived heat shock protein 70 (HSP70) molecules are molecular chaperones with a broad repertoire of tumour antigen peptides capable of stimulating dendritic cell (DC) maturation and T-cell immune responses. To enhance EXO-based antitumour immunity, we generated an engineered myeloma cell line J558HSP expressing endogenous P1A tumour antigen and transgenic form of membrane-bound HSP70 and heat-shocked J558HS expressing cytoplasmic HSP70, and purified EXOHSP and EXOHS from J558HSP and J558HS tumour cell culture supernatants by ultracentrifugation. We found that EXOHSP were able to more efficiently stimulate maturation of DCs with up-regulation of Iab, CD40, CD80 and inflammatory cytokines than EXOHS after overnight incubation of immature bone-marrow-derived DCs (5 × 106 cells) with EXO (100 μg), respectively. We also i.v. immunized BALB/c mice with EXO (30 μg/mouse) and assessed P1A-specific T-cell responses after immunization. We demonstrate that EXOHSP are able to stimulate type 1 CD4+ helper T (Th1) cell responses, and more efficient P1A-specific CD8+ cytotoxic T lymphocyte (CTL) responses and antitumour immunity than EXOHS. In addition, we further elucidate that EXOHSP-stimulated antitumour immunity is mediated by both P1A-specific CD8+ CTL and non-P1A-specific natural killer (NK) responses. Therefore, membrane-bound HSP70-expressing tumour cell-released EXO may represent a more effective EXO-based vaccine in induction of antitumour immunity.
Collapse
Affiliation(s)
- Yufeng Xie
- Research Unit, Division of Health Research, Saskatchewan Cancer Agency, Department of Oncology, University of Saskatchewan, Saskatoon, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Chen T, Guo J, Yang M, Zhu X, Cao X. Chemokine-containing exosomes are released from heat-stressed tumor cells via lipid raft-dependent pathway and act as efficient tumor vaccine. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:2219-28. [PMID: 21242526 DOI: 10.4049/jimmunol.1002991] [Citation(s) in RCA: 177] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Exosomes derived from dendritic cells or tumor cells are a population of nanometer-sized membrane vesicles that can induce specific antitumor immunity. During investigation of the effects of hyperthermia on antitumor immune response, we found that exosomes derived from heat-stressed tumor cells (HS-TEX) could chemoattract and activate dendritic cells (DC) and T cells more potently than that by conventional tumor-derived exosomes. We show that HS-TEX contain chemokines, such as CCL2, CCL3, CCL4, CCL5, and CCL20, and the chemokine-containing HS-TEX are functionally competent in chemoattracting CD11c(+) DC and CD4(+)/CD8(+) T cells both in vitro and in vivo. Moreover, the production of chemokine-containing HS-TEX could be inhibited by ATP inhibitor, calcium chelator, and cholesterol scavenger, indicating that the mobilization of chemokines into exosomes was ATP- and calcium-dependent and via a lipid raft-dependent pathway. We consistently found that the intracellular chemokines could be enriched in lipid rafts after heat stress. Accordingly, intratumoral injection of HS-TEX could induce specific antitumor immune response more efficiently than that by tumor-derived exosomes, thus inhibiting tumor growth and prolonging survival of tumor-bearing mice more significantly. Therefore, our results demonstrate that exosomes derived from HS-TEX represent a kind of efficient tumor vaccine and can chemoattract and activate DC and T cells, inducing more potent antitumor immune response. Release of chemokines through exosomes via lipid raft-dependent pathway may be a new method of chemokine exocytosis.
Collapse
Affiliation(s)
- Taoyong Chen
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | | | | | | | | |
Collapse
|