1
|
Kim M, Choi H, Jang DJ, Kim HJ, Sub Y, Gee HY, Choi C. Exploring the clinical transition of engineered exosomes designed for intracellular delivery of therapeutic proteins. Stem Cells Transl Med 2024; 13:637-647. [PMID: 38838263 PMCID: PMC11227971 DOI: 10.1093/stcltm/szae027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/18/2024] [Indexed: 06/07/2024] Open
Abstract
Extracellular vesicles, particularly exosomes, have emerged as promising drug delivery systems owing to their unique advantages, such as biocompatibility, immune tolerability, and target specificity. Various engineering strategies have been implemented to harness these innate qualities, with a focus on enhancing the pharmacokinetic and pharmacodynamic properties of exosomes via payload loading and surface engineering for active targeting. This concise review outlines the challenges in the development of exosomes as drug carriers and offers insights into strategies for their effective clinical translation. We also highlight preclinical studies that have successfully employed anti-inflammatory exosomes and suggest future directions for exosome therapeutics. These advancements underscore the potential for integrating exosome-based therapies into clinical practice, heralding promise for future medical interventions.
Collapse
Affiliation(s)
| | - Hojun Choi
- ILIAS Biologics Inc., Daejeon 34014, Korea
| | - Deok-Jin Jang
- ILIAS Biologics Inc., Daejeon 34014, Korea
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | | | - Yujin Sub
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Heon Yung Gee
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | | |
Collapse
|
2
|
Chen J, Zhang E, Wan Y, Huang T, Wang Y, Jiang H. A quick and innovative pipeline for producing chondrocyte-homing peptide-modified extracellular vesicles by three-dimensional dynamic culture of hADSCs spheroids to modulate the fate of remaining ear chondrocytes in the M1 macrophage-infiltrated microenvironment. J Nanobiotechnology 2024; 22:300. [PMID: 38816719 PMCID: PMC11141023 DOI: 10.1186/s12951-024-02567-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/16/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) derived from human adipose-derived mesenchymal stem cells (hADSCs) have shown great therapeutic potential in plastic and reconstructive surgery. However, the limited production and functional molecule loading of EVs hinder their clinical translation. Traditional two-dimensional culture of hADSCs results in stemness loss and cellular senescence, which is unfavorable for the production and functional molecule loading of EVs. Recent advances in regenerative medicine advocate for the use of three-dimensional culture of hADSCs to produce EVs, as it more accurately simulates their physiological state. Moreover, the successful application of EVs in tissue engineering relies on the targeted delivery of EVs to cells within biomaterial scaffolds. METHODS AND RESULTS The hADSCs spheroids and hADSCs gelatin methacrylate (GelMA) microspheres are utilized to produce three-dimensional cultured EVs, corresponding to hADSCs spheroids-EVs and hADSCs microspheres-EVs respectively. hADSCs spheroids-EVs demonstrate excellent production and functional molecule loading compared with hADSCs microspheres-EVs. The upregulation of eight miRNAs (i.e. hsa-miR-486-5p, hsa-miR-423-5p, hsa-miR-92a-3p, hsa-miR-122-5p, hsa-miR-223-3p, hsa-miR-320a, hsa-miR-126-3p, and hsa-miR-25-3p) and the downregulation of hsa-miR-146b-5p within hADSCs spheroids-EVs show the potential of improving the fate of remaining ear chondrocytes and promoting cartilage formation probably through integrated regulatory mechanisms. Additionally, a quick and innovative pipeline is developed for isolating chondrocyte homing peptide-modified EVs (CHP-EVs) from three-dimensional dynamic cultures of hADSCs spheroids. CHP-EVs are produced by genetically fusing a CHP at the N-terminus of the exosomal surface protein LAMP2B. The CHP + LAMP2B-transfected hADSCs spheroids were cultured with wave motion to promote the secretion of CHP-EVs. A harvesting method is used to enable the time-dependent collection of CHP-EVs. The pipeline is easy to set up and quick to use for the isolation of CHP-EVs. Compared with nontagged EVs, CHP-EVs penetrate the biomaterial scaffolds and specifically deliver the therapeutic miRNAs to the remaining ear chondrocytes. Functionally, CHP-EVs show a major effect on promoting cell proliferation, reducing cell apoptosis and enhancing cartilage formation in remaining ear chondrocytes in the M1 macrophage-infiltrated microenvironment. CONCLUSIONS In summary, an innovative pipeline is developed to obtain CHP-EVs from three-dimensional dynamic culture of hADSCs spheroids. This pipeline can be customized to increase EVs production and functional molecule loading, which meets the requirements for regulating remaining ear chondrocyte fate in the M1 macrophage-infiltrated microenvironment.
Collapse
Affiliation(s)
- Jianguo Chen
- Chinese Academy of Medical Sciences & Peking Union Medical College Plastic Surgery Hospital and Institute, Shijingshan District, Beijing, 100144, China
| | - Enchong Zhang
- Department of Urology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, China.
| | - Yingying Wan
- DongFang Hospital of Beijing University of Chinese Medicine, Fengtai District, Beijing, 100078, China.
| | - Tianyu Huang
- Chinese Academy of Medical Sciences & Peking Union Medical College Plastic Surgery Hospital and Institute, Shijingshan District, Beijing, 100144, China
| | - Yuchen Wang
- Chinese Academy of Medical Sciences & Peking Union Medical College Plastic Surgery Hospital and Institute, Shijingshan District, Beijing, 100144, China
| | - Haiyue Jiang
- Chinese Academy of Medical Sciences & Peking Union Medical College Plastic Surgery Hospital and Institute, Shijingshan District, Beijing, 100144, China.
| |
Collapse
|
3
|
Yue Y, Dai W, Wei Y, Cao S, Liao S, Li A, Liu P, Lin J, Zeng H. Unlocking the potential of exosomes: a breakthrough in the theranosis of degenerative orthopaedic diseases. Front Bioeng Biotechnol 2024; 12:1377142. [PMID: 38699435 PMCID: PMC11064847 DOI: 10.3389/fbioe.2024.1377142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/01/2024] [Indexed: 05/05/2024] Open
Abstract
Degenerative orthopaedic diseases pose a notable worldwide public health issue attributable to the global aging population. Conventional medical approaches, encompassing physical therapy, pharmaceutical interventions, and surgical methods, face obstacles in halting or reversing the degenerative process. In recent times, exosome-based therapy has gained widespread acceptance and popularity as an effective treatment for degenerative orthopaedic diseases. This therapeutic approach holds the potential for "cell-free" tissue regeneration. Exosomes, membranous vesicles resulting from the fusion of intracellular multivesicles with the cell membrane, are released into the extracellular matrix. Addressing challenges such as the rapid elimination of natural exosomes in vivo and the limitation of drug concentration can be effectively achieved through various strategies, including engineering modification, gene overexpression modification, and biomaterial binding. This review provides a concise overview of the source, classification, and preparation methods of exosomes, followed by an in-depth analysis of their functions and potential applications. Furthermore, the review explores various strategies for utilizing exosomes in the treatment of degenerative orthopaedic diseases, encompassing engineering modification, gene overexpression, and biomaterial binding. The primary objective is to provide a fresh viewpoint on the utilization of exosomes in addressing bone degenerative conditions and to support the practical application of exosomes in the theranosis of degenerative orthopaedic diseases.
Collapse
Affiliation(s)
- Yaohang Yue
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Wei Dai
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yihao Wei
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Siyang Cao
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Shuai Liao
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Aikang Li
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Peng Liu
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jianjing Lin
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Hui Zeng
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
4
|
Wang T, Sun L, Ren T, Hou M, Long Y, Jiang JH, He J. Targeted Protein Degradation Mediated by Genetically Engineered Lysosome-Targeting Exosomes. NANO LETTERS 2023; 23:9571-9578. [PMID: 37823825 DOI: 10.1021/acs.nanolett.3c03148] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Protein-degrading chimeras are superior drug modalities compared to traditional protein inhibitors because of their effective therapeutic performance. So far, various targeted protein degradation strategies, including proteolysis-targeting chimeras and lysosome-targeting chimeras, have emerged as essential technologies for tackling diseases caused by abnormal protein expression. Here, we report the development and application of lysosome-targeting exosomes (LYTEXs) for the selective degradation of membrane protein targets. LYTEXs are genetically engineered exosomes expressing multivalent single-chain fragment variables, simultaneously recognizing cell-surface lysosome-targeting and to-be-degraded protein. We show that by targeting the lysosome-directing asialoglycoprotein receptor, bispecific LYTEXs can induce lysosomal degradation of membrane-associated therapeutic targets. This strategy provides a generalizable, easy-to-prepare platform for modulating surface protein expression, with the advantage of therapeutic delivery.
Collapse
Affiliation(s)
- Tao Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, School of Biomedical Sciences, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Liang Sun
- State Key Laboratory of Chemo/Biosensing and Chemometrics, School of Biomedical Sciences, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Tianyu Ren
- State Key Laboratory of Chemo/Biosensing and Chemometrics, School of Biomedical Sciences, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Min Hou
- School of Physics and Chemistry, Hunan First Normal University, Changsha 410205, China
| | - Ying Long
- State Key Laboratory of Chemo/Biosensing and Chemometrics, School of Biomedical Sciences, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Jian-Hui Jiang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, School of Biomedical Sciences, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Jianjun He
- State Key Laboratory of Chemo/Biosensing and Chemometrics, School of Biomedical Sciences, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
5
|
Chen Y, Li Y, Wu Y, Chen S, Jin X, Chen X, Fei B, Xue X, Wu R, Chai K. Yiwei decoction promotes apoptosis of gastric cancer cells through spleen-derived exosomes. Front Pharmacol 2023; 14:1144955. [PMID: 37324462 PMCID: PMC10267389 DOI: 10.3389/fphar.2023.1144955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Yiwei decoction (YWD) is a formula of traditional Chinese medicine (TCM) that is clinically effective for the prevention and treatment of gastric cancer recurrence and metastasis. According to the theory of TCM, YWD tonifies the body and strengthens the body's resistance to gastric cancer recurrence and metastasis potentially via the immune regulation of the spleen. The aims of the present study were to investigate whether YWD-treated spleen-derived exosomes in rats inhibit the proliferation of tumor cells, to elucidate the anticancer effects of YWD, and to provide evidence supporting the use of YWD as a new clinical treatment for gastric cancer. Spleen-derived exosomes were obtained by ultracentrifugation and identified by transmission electron microscopy, nanoparticle tracking analysis, and western blot analysis. The location of the exosomes in tumor cells was then determined by immunofluorescence staining. After tumor cells were treated with different concentrations of exosomes, the effect of exosomes on cell proliferation was determined by cell counting kit 8 (CCK8) and colony formation assays. Tumor cell apoptosis was detected by flow cytometry. Particle analysis and western blot analysis identified the material extracted from spleen tissue supernatant as exosomes. Immunofluorescence staining showed that spleen-derived exosomes were taken up by HGC-27 cells, and the CCK8 assay confirmed that the relative tumor inhibition rate of YWD-treated spleen-derived exosomes in the 30 μg/mL reached 70.78% compared to control exosomes in the 30 μg/mL (p < 0.05). Compared to control exosomes in the 30 μg/mL, the colony formation assay indicated that YWD-treated spleen-derived exosomes in the 30 μg/mL colonies have decreased by 99.03% (p < 0.01). Moreover, flow cytometry analysis showed that treatment with YWD-treated exosomes in the 30 μg/mL increased the apoptosis rate to 43.27%, which was significantly higher than that of the control group in the 30 μg/mL (25.91%) (p < 0.05). In conclusion, spleen-derived exosomes from YWD-treated animals inhibit the proliferation of HGC-27 cells via inducing apoptosis, suggesting that spleen-derived exosomes are involved in mediating the antitumor effect of YWD. These results demonstrated a novel exosome-mediated anticancer effect of YWD as a TCM formula, thereby supporting the use of YWD-treated exosomes as a new approach for the clinical treatment of gastric cancer.
Collapse
Affiliation(s)
- Yingzhi Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Yu Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Yue Wu
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shiyong Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Xiaoming Jin
- Stark Neuroscience Research Institute and Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xuan Chen
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Baoying Fei
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Xiaomin Xue
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Renzhao Wu
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Kequn Chai
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| |
Collapse
|
6
|
Si Q, Wu L, Pang D, Jiang P. Exosomes in brain diseases: Pathogenesis and therapeutic targets. MedComm (Beijing) 2023; 4:e287. [PMID: 37313330 PMCID: PMC10258444 DOI: 10.1002/mco2.287] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 06/15/2023] Open
Abstract
Exosomes are extracellular vesicles with diameters of about 100 nm that are naturally secreted by cells into body fluids. They are derived from endosomes and are wrapped in lipid membranes. Exosomes are involved in intracellular metabolism and intercellular communication. They contain nucleic acids, proteins, lipids, and metabolites from the cell microenvironment and cytoplasm. The contents of exosomes can reflect their cells' origin and allow the observation of tissue changes and cell states under disease conditions. Naturally derived exosomes have specific biomolecules that act as the "fingerprint" of the parent cells, and the contents changed under pathological conditions can be used as biomarkers for disease diagnosis. Exosomes have low immunogenicity, are small in size, and can cross the blood-brain barrier. These characteristics make exosomes unique as engineering carriers. They can incorporate therapeutic drugs and achieve targeted drug delivery. Exosomes as carriers for targeted disease therapy are still in their infancy, but exosome engineering provides a new perspective for cell-free disease therapy. This review discussed exosomes and their relationship with the occurrence and treatment of some neuropsychiatric diseases. In addition, future applications of exosomes in the diagnosis and treatment of neuropsychiatric disorders were evaluated in this review.
Collapse
Affiliation(s)
- Qingying Si
- Department of EndocrinologyTengzhou Central People's HospitalTengzhouChina
| | - Linlin Wu
- Department of OncologyTengzhou Central People's HospitalTengzhouChina
| | - Deshui Pang
- Department of EndocrinologyTengzhou Central People's HospitalTengzhouChina
| | - Pei Jiang
- Translational Pharmaceutical LaboratoryJining First People's HospitalShandong First Medical UniversityJiningChina
- Institute of Translational PharmacyJining Medical Research AcademyJiningChina
| |
Collapse
|
7
|
Johnson V, Vasu S, Kumar US, Kumar M. Surface-Engineered Extracellular Vesicles in Cancer Immunotherapy. Cancers (Basel) 2023; 15:2838. [PMID: 37345176 PMCID: PMC10216164 DOI: 10.3390/cancers15102838] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/14/2023] [Accepted: 05/15/2023] [Indexed: 06/23/2023] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed bodies secreted by all cell types. EVs carry bioactive materials, such as proteins, lipids, metabolites, and nucleic acids, to communicate and elicit functional alterations and phenotypic changes in the counterpart stromal cells. In cancer, cells secrete EVs to shape a tumor-promoting niche. Tumor-secreted EVs mediate communications with immune cells that determine the fate of anti-tumor therapeutic effectiveness. Surface engineering of EVs has emerged as a promising tool for the modulation of tumor microenvironments for cancer immunotherapy. Modification of EVs' surface with various molecules, such as antibodies, peptides, and proteins, can enhance their targeting specificity, immunogenicity, biodistribution, and pharmacokinetics. The diverse approaches sought for engineering EV surfaces can be categorized as physical, chemical, and genetic engineering strategies. The choice of method depends on the specific application and desired outcome. Each has its advantages and disadvantages. This review lends a bird's-eye view of the recent progress in these approaches with respect to their rational implications in the immunomodulation of tumor microenvironments (TME) from pro-tumorigenic to anti-tumorigenic ones. The strategies for modulating TME using targeted EVs, their advantages, current limitations, and future directions are discussed.
Collapse
Affiliation(s)
- Vinith Johnson
- Department of Chemical Engineering, Indian Institute of Technology, Tirupati 517619, India
| | - Sunil Vasu
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Uday S. Kumar
- Department of Chemical Engineering, Indian Institute of Technology, Tirupati 517619, India
| | - Manoj Kumar
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
8
|
Mohammadi AH, Ghazvinian Z, Bagheri F, Harada M, Baghaei K. Modification of Extracellular Vesicle Surfaces: An Approach for Targeted Drug Delivery. BioDrugs 2023; 37:353-374. [PMID: 37093521 DOI: 10.1007/s40259-023-00595-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2023] [Indexed: 04/25/2023]
Abstract
Extracellular vesicles (EVs) are a promising drug delivery vehicle candidate because of their natural origin and intrinsic function of transporting various molecules between different cells. Several advantages of the EV delivery platform include enhanced permeability and retention effect, efficient interaction with recipient cells, the ability to traverse biological barriers, high biocompatibility, high biodegradability, and low immunogenicity. Furthermore, EV membranes share approximately similar structures and contents to the cell membrane, which allows surface modification of EVs, an approach to enable specific targeting. Enhanced drug accumulation in intended sites and reduced adverse effects of chemotherapeutic drugs are the most prominent effects of targeted drug delivery. In order to improve the targeting ability of EVs, chemical modification and genetic engineering are the most adopted methods to date. Diverse chemical methods are employed to decorate EV surfaces with various ligands such as aptamers, carbohydrates, peptides, vitamins, and antibodies. In this review, we introduce the biogenesis, content, and cellular pathway of natural EVs and further discuss the genetic modification of EVs, and its challenges. Furthermore, we provide a comprehensive deliberation on the various chemical modification methods for improved drug delivery, which are directly related to increasing the therapeutic index.
Collapse
Affiliation(s)
- Amir Hossein Mohammadi
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Zeinab Ghazvinian
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Bagheri
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran.
| | - Masako Harada
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA.
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Mondal J, Pillarisetti S, Junnuthula V, Saha M, Hwang SR, Park IK, Lee YK. Hybrid exosomes, exosome-like nanovesicles and engineered exosomes for therapeutic applications. J Control Release 2023; 353:1127-1149. [PMID: 36528193 DOI: 10.1016/j.jconrel.2022.12.027] [Citation(s) in RCA: 88] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/28/2022]
Abstract
Exosomes are endosome-derived nanovesicles involved in cellular communication. They are natural nanocarriers secreted by various cells, making them suitable candidates for diverse drug delivery and therapeutic applications from a material standpoint. They have a phospholipid bilayer decorated with functional molecules and an enclosed parental matrix, which has attracted interest in developing designer/hybrid engineered exosome nanocarriers. The structural versatility of exosomes allows the modification of their original configuration using various methods, including genetic engineering, chemical procedures, physical techniques, and microfluidic technology, to load exosomes with additional cargo for expanded biomedical applications. Exosomes show enormous potential for overcoming the limitations of conventional nanoparticle-based techniques in targeted therapy. This review highlights the exosome sources, characteristics, state of the art in the field of hybrid exosomes, exosome-like nanovesicles and engineered exosomes as potential cargo delivery vehicles for therapeutic applications.
Collapse
Affiliation(s)
- Jagannath Mondal
- Department of Green Bioengineering, Korea National University of Transportation, Chungju 27470, Republic of Korea
| | - Shameer Pillarisetti
- Department of Biomedical Sciences and Biomedical Science Graduate Program (BMSGP), Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 61469, Republic of Korea
| | | | - Monochura Saha
- Media lab, Massachusetts Institute of Technology (MIT), 75 Amherst Street, Cambridge 02139, USA
| | - Seung Rim Hwang
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju 61452, Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Sciences and Biomedical Science Graduate Program (BMSGP), Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 61469, Republic of Korea.
| | - Yong-Kyu Lee
- Department of Green Bioengineering, Korea National University of Transportation, Chungju 27470, Republic of Korea; Department of Chemical & Biological Engineering, Korea National University of Transportation, Chungju 27470, Republic of Korea.
| |
Collapse
|
10
|
Cheng Q, Dai Z, Smbatyan G, Epstein AL, Lenz HJ, Zhang Y. Eliciting anti-cancer immunity by genetically engineered multifunctional exosomes. Mol Ther 2022; 30:3066-3077. [PMID: 35746867 PMCID: PMC9481992 DOI: 10.1016/j.ymthe.2022.06.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/25/2022] [Accepted: 06/17/2022] [Indexed: 12/16/2022] Open
Abstract
Exosomes are cell-derived nanovesicles involved in regulating intercellular communications. In contrast to conventional nanomedicines, exosomes are characterized by unique advantages for therapeutic development. Despite their major successes in drug delivery, the full potential of exosomes for immunotherapy remains untapped. Herein we designed genetically engineered exosomes featured with surfaced-displayed antibody targeting groups and immunomodulatory proteins. Through genetic fusions with exosomal membrane proteins, Expi293F cell-derived exosomes were armed with monoclonal antibodies specific for human T-cell CD3 and epidermal growth factor receptor (EGFR) as well as immune checkpoint modulators, programmed death 1 (PD-1) and OX40 ligand (OX40L). The resulting genetically engineered multifunctional immune-modulating exosomes (GEMINI-Exos) can not only redirect and activate T cells toward killing EGFR-positive triple negative breast cancer (TNBC) cells but also elicit robust anti-cancer immunity, giving rise to highly potent inhibition against established TNBC tumors in mice. GEMINI-Exos represent candidate agents for immunotherapy and may offer a general strategy for generating exosome-based immunotherapeutics with desired functions and properties.
Collapse
Affiliation(s)
- Qinqin Cheng
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Zhefu Dai
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Goar Smbatyan
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Alan L Epstein
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Yong Zhang
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA; Department of Chemistry, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA 90089, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA; Research Center for Liver Diseases, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
11
|
Exosomes as Crucial Players in Pathogenesis of Systemic Lupus Erythematosus. J Immunol Res 2022; 2022:8286498. [PMID: 35910853 PMCID: PMC9328965 DOI: 10.1155/2022/8286498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/29/2022] [Indexed: 11/18/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease that affects multiple systems. Its clinical manifestation varies across patients, from skin mucosa to multiorgan damage to severe central nervous system involvement. The exosome has been shown to play an important role in the pathogenesis of autoimmune diseases, including SLE. We review the recent knowledge of exosomes, including their biology, functions, mechanism, and standardized extraction and purification methods in SLE, to highlight potential therapeutic targets for SLE.
Collapse
|
12
|
Meng F, Xue X, Yin Z, Gao F, Wang X, Geng Z. Research Progress of Exosomes in Bone Diseases: Mechanism, Diagnosis and Therapy. Front Bioeng Biotechnol 2022; 10:866627. [PMID: 35497358 PMCID: PMC9039039 DOI: 10.3389/fbioe.2022.866627] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
With the global escalation of the aging process, the number of patients with bone diseases is increasing year by year. Currently, there are limited effective treatments for bone diseases. Exosome, as a vital medium in cell-cell communication, can mediate tissue metabolism through the paracrine transmission of various cargos (proteins, nucleic acids, lipids, etc.) carried by itself. Recently, an increasing number of researchers have proven that exosomes play essential roles in the formation, metabolism, and pathological changes of bone and cartilage. Because exosomes have the advantages of small size, rich sources, and low immunogenicity, they can be used not only as substitutes for the traditional treatment of bone diseases, but also as biomarkers for the diagnosis of bone diseases. This paper reviews the research progress of several kinds of cells derived-exosomes in bone diseases and provides a theoretical basis for further research and clinical application of exosomes in bone diseases in the future.
Collapse
Affiliation(s)
- Fanying Meng
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Xu Xue
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Fei Gao
- Institute of Translational Medicine, Shanghai University, Shanghai, China,*Correspondence: Fei Gao, ; Xiuhui Wang, ; Zhen Geng,
| | - Xiuhui Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, China,*Correspondence: Fei Gao, ; Xiuhui Wang, ; Zhen Geng,
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, China,*Correspondence: Fei Gao, ; Xiuhui Wang, ; Zhen Geng,
| |
Collapse
|
13
|
Liu K, Cui JJ, Zhan Y, Ouyang QY, Lu QS, Yang DH, Li XP, Yin JY. Reprogramming the tumor microenvironment by genome editing for precision cancer therapy. Mol Cancer 2022; 21:98. [PMID: 35410257 PMCID: PMC8996591 DOI: 10.1186/s12943-022-01561-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/11/2022] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) is essential for immune escape by tumor cells. It plays essential roles in tumor development and metastasis. The clinical outcomes of tumors are often closely related to individual differences in the patient TME. Therefore, reprogramming TME cells and their intercellular communication is an attractive and promising strategy for cancer therapy. TME cells consist of immune and nonimmune cells. These cells need to be manipulated precisely and safely to improve cancer therapy. Furthermore, it is encouraging that this field has rapidly developed in recent years with the advent and development of gene editing technologies. In this review, we briefly introduce gene editing technologies and systematically summarize their applications in the TME for precision cancer therapy, including the reprogramming of TME cells and their intercellular communication. TME cell reprogramming can regulate cell differentiation, proliferation, and function. Moreover, reprogramming the intercellular communication of TME cells can optimize immune infiltration and the specific recognition of tumor cells by immune cells. Thus, gene editing will pave the way for further breakthroughs in precision cancer therapy.
Collapse
|
14
|
Lee J, Lee JH, Chakraborty K, Hwang J, Lee YK. Exosome-based drug delivery systems and their therapeutic applications. RSC Adv 2022; 12:18475-18492. [PMID: 35799926 PMCID: PMC9218984 DOI: 10.1039/d2ra02351b] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/15/2022] [Indexed: 12/13/2022] Open
Abstract
In the past few decades, scientists have actively worked on developing effective drug delivery systems (DDSs) as means to control life-threatening diseases and challenging illnesses.
Collapse
Affiliation(s)
- Jaewook Lee
- 4D Convergence Technology Institute (National Key Technology Institute in University), Korea National University of Transportation, Jeungpyeong, Chung-Buk, 27909, Republic of Korea
| | - Ji-Heon Lee
- 4D Convergence Technology Institute (National Key Technology Institute in University), Korea National University of Transportation, Jeungpyeong, Chung-Buk, 27909, Republic of Korea
| | - Kushal Chakraborty
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju, Chung-Buk 27469, Republic of Korea
| | - Joon Hwang
- 4D Convergence Technology Institute (National Key Technology Institute in University), Korea National University of Transportation, Jeungpyeong, Chung-Buk, 27909, Republic of Korea
- Department of Aeronautical & Mechanical Design Engineering, Korea National University of Transportation, Chungju, Chung-Buk 27469, Republic of Korea
| | - Yong-Kyu Lee
- 4D Convergence Technology Institute (National Key Technology Institute in University), Korea National University of Transportation, Jeungpyeong, Chung-Buk, 27909, Republic of Korea
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju, Chung-Buk 27469, Republic of Korea
| |
Collapse
|