Exosome-Based Drug Delivery: Translation from Bench to Clinic
Abstract
:1. Introduction
2. Structure and Biogenesis of Exosomes
2.1. Exosome Structure
2.2. Exosome Biogenesis
3. Cellular Source of Exosome and Isolation Methods
3.1. Cellular Source of Exosome
3.2. Isolation Methods for Exosome
4. Drug-Loading Approaches to Exosomes
4.1. Pre-Loading/Indirect Method
4.1.1. Co-Incubation
4.1.2. Genetic Modification
4.2. Post-Loading/Direct Method
4.2.1. Passive Loading
4.2.2. Active Loading
Physical Induction
Chemical Induction
5. Exosome Engineering
5.1. Genetic Engineering
5.2. Chemical Engineering
6. Clinical Application of Exosomes-Based Drug Delivery
6.1. Cancer
6.1.1. Small-Molecule Drugs
6.1.2. Therapeutic Nucleic Acids
Nucleic Acid | Cellular Source of Exosome | Transfection Method | Target Disease [Animal Model] | Mechanism | Refs |
---|---|---|---|---|---|
hsa-miR-122 | Human adipose tissue-derived mesenchymal stem cell | Lipofectamine | Hepatocellular carcinma [BALB/c nude mice + HepG2 cells] | Enhancing sensitivity of chemotherapeutic agents (5-fluorouracil and sorafenib) through altering target genes expression | [128] |
miRNA-126 | Human breast cancer cell | ExoFectin | Lung metastasis model [C57BL/6 + A549 cells] | Interaction between surfactant protein C on cancer cell and integrin β4 on exosome Inhibiting proliferation and migration of cancer cells via a PTEN/PI3K/AKT signaling pathway | [129] |
BCR-ABL siRNA | HEK293T cell (engineered using IL3-LAMP-2B plasmid) | Lipofectamine | CML model [NOD/ SCID mice + human LAMA84 cells or K562R cells] | Interaction between IL-3 on exosome and IL3R on CML blast Reverse sensitivity of chemotherapeutic agent (Imatinib) through decreasing BCR-ABL mRNA and protein level | [71] |
CPT1A siRNA | HEK293T cell (engineered using iRGD- LAMP-2B plasmid) | Lipofectamine | Colon cancer mocel [BALB/c nude mice + HCT116-lohp cell] | Interaction between iRGD on exosome and αvβ3 and Neuropilin-1 on colon cancer cell Reverse the sensitivity of chemotherapeutic agent (oxaliplatin) through inhibiting fatty acid oxidation | [130] |
BCL6 siRNA1 | Mouse bone marrow dendritic cell (engineered using iRGD- LAMP-2B plasmid) | Electroporation | Diffuse large B-cell lymphoma (DLBCL) model [BALB/c nude mice + OCI-Ly8 cells] | Interaction between iRGD on exosome and αvβ3 integrin on DLBCL cell Inhibiting DLBCL cell proliferation through inducing G0-G1 cell cycle arrest | [132] |
c-Met siRNA | HEK293T cell | Lipofectamine | Gastric cancer model [BALB/c-nu + SGC7901/DDP] | Enhancing sensitivity of chemotherapeutic agent (cisplatin) through altering target genes expression | [131] |
6.2. Inflammatory Diseases
6.3. Kidney Diseases
6.4. Cardiovascular Diseases
6.5. Neurodegenerative Diseases
7. Challenges Associated with Exosome-Based Drug Delivery and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Zeng, W.; Wen, Z.; Chen, H.; Duan, Y. Exosomes as carriers for drug delivery in cancer therapy. Pharm. Res. 2023, 40, 873–887. [Google Scholar] [CrossRef] [PubMed]
- Elsharkasy, O.M.; Nordin, J.Z.; Hagey, D.W.; de Jong, O.G.; Schiffelers, R.M.; Andaloussi, S.E.; Vader, P. Extracellular vesicles as drug delivery systems: Why and how? Adv. Drug Deliv. Rev. 2020, 159, 332–343. [Google Scholar] [CrossRef] [PubMed]
- Simoes, S.; Filipe, A.; Faneca, H.; Mano, M.; Penacho, N.; Duzgunes, N.; de Lima, M.P. Cationic liposomes for gene delivery. Expert Opin. Drug Deliv. 2005, 2, 237–254. [Google Scholar] [CrossRef] [PubMed]
- Nag, O.K.; Awasthi, V. Surface engineering of liposomes for stealth behavior. Pharmaceutics 2013, 5, 542–569. [Google Scholar] [CrossRef] [Green Version]
- Narayanaswamy, R.; Torchilin, V.P. Targeted delivery of combination therapeutics using monoclonal antibody 2c5-modified immunoliposomes for cancer therapy. Pharm. Res. 2021, 38, 429–450. [Google Scholar] [CrossRef]
- Moholkar, D.N.; Kandimalla, R.; Gupta, R.C.; Aqil, F. Advances in lipid-based carriers for cancer therapeutics: Liposomes, exosomes and hybrid exosomes. Cancer Lett. 2023, 565, 216220. [Google Scholar] [CrossRef]
- Ickenstein, L.M.; Garidel, P. Lipid-based nanoparticle formulations for small molecules and rna drugs. Expert Opin. Drug Deliv. 2019, 16, 1205–1226. [Google Scholar] [CrossRef]
- Daraee, H.; Etemadi, A.; Kouhi, M.; Alimirzalu, S.; Akbarzadeh, A. Application of liposomes in medicine and drug delivery. Artif. Cells Nanomed. Biotechnol. 2016, 44, 381–391. [Google Scholar] [CrossRef] [PubMed]
- Narang, A.S.; Chang, R.K.; Hussain, M.A. Pharmaceutical development and regulatory considerations for nanoparticles and nanoparticulate drug delivery systems. J. Pharm. Sci. 2013, 102, 3867–3882. [Google Scholar] [CrossRef] [PubMed]
- Ha, D.; Yang, N.; Nadithe, V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: Current perspectives and future challenges. Acta Pharm. Sin. B 2016, 6, 287–296. [Google Scholar] [CrossRef] [Green Version]
- Abdelsalam, M.; Ahmed, M.; Osaid, Z.; Hamoudi, R.; Harati, R. Insights into exosome transport through the blood-brain barrier and the potential therapeutical applications in brain diseases. Pharmaceuticals 2023, 16, 571. [Google Scholar] [CrossRef] [PubMed]
- Luan, X.; Sansanaphongpricha, K.; Myers, I.; Chen, H.; Yuan, H.; Sun, D. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacol. Sin. 2017, 38, 754–763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Antimisiaris, S.G.; Mourtas, S.; Marazioti, A. Exosomes and exosome-inspired vesicles for targeted drug delivery. Pharmaceutics 2018, 10, 218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lai, C.P.; Mardini, O.; Ericsson, M.; Prabhakar, S.; Maguire, C.; Chen, J.W.; Tannous, B.A.; Breakefield, X.O. Dynamic biodistribution of extracellular vesicles in vivo using a multimodal imaging reporter. ACS Nano 2014, 8, 483–494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wiklander, O.P.; Nordin, J.Z.; O’Loughlin, A.; Gustafsson, Y.; Corso, G.; Mager, I.; Vader, P.; Lee, Y.; Sork, H.; Seow, Y.; et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J. Extracell. Vesicles 2015, 4, 26316. [Google Scholar] [CrossRef] [Green Version]
- Sidhom, K.; Obi, P.O.; Saleem, A. A review of exosomal isolation methods: Is size exclusion chromatography the best option? Int. J. Mol. Sci. 2020, 21, 6466. [Google Scholar] [CrossRef]
- Jankovicova, J.; Secova, P.; Michalkova, K.; Antalikova, J. Tetraspanins, more than markers of extracellular vesicles in reproduction. Int. J. Mol. Sci. 2020, 21, 7568. [Google Scholar] [CrossRef]
- Kamerkar, S.; LeBleu, V.S.; Sugimoto, H.; Yang, S.; Ruivo, C.F.; Melo, S.A.; Lee, J.J.; Kalluri, R. Exosomes facilitate therapeutic targeting of oncogenic kras in pancreatic cancer. Nature 2017, 546, 498–503. [Google Scholar] [CrossRef] [Green Version]
- Taha, E.A.; Ono, K.; Eguchi, T. Roles of extracellular hsps as biomarkers in immune surveillance and immune evasion. Int. J. Mol. Sci. 2019, 20, 4588. [Google Scholar] [CrossRef] [Green Version]
- Wei, H.; Chen, Q.; Lin, L.; Sha, C.; Li, T.; Liu, Y.; Yin, X.; Xu, Y.; Chen, L.; Gao, W.; et al. Regulation of exosome production and cargo sorting. Int. J. Biol. Sci. 2021, 17, 163–177. [Google Scholar] [CrossRef]
- Minciacchi, V.R.; Freeman, M.R.; Di Vizio, D. Extracellular vesicles in cancer: Exosomes, microvesicles and the emerging role of large oncosomes. Semin. Cell Dev. Biol. 2015, 40, 41–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Record, M. Intercellular communication by exosomes in placenta: A possible role in cell fusion? Placenta 2014, 35, 297–302. [Google Scholar] [CrossRef] [PubMed]
- Henne, W.M.; Buchkovich, N.J.; Emr, S.D. The escrt pathway. Dev. Cell 2011, 21, 77–91. [Google Scholar] [CrossRef] [Green Version]
- Guo, B.B.; Bellingham, S.A.; Hill, A.F. The neutral sphingomyelinase pathway regulates packaging of the prion protein into exosomes. J. Biol. Chem. 2015, 290, 3455–3467. [Google Scholar] [CrossRef] [Green Version]
- Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brugger, B.; Simons, M. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef] [PubMed]
- Stuffers, S.; Sem Wegner, C.; Stenmark, H.; Brech, A. Multivesicular endosome biogenesis in the absence of escrts. Traffic 2009, 10, 925–937. [Google Scholar] [CrossRef] [PubMed]
- Perez-Hernandez, D.; Gutierrez-Vazquez, C.; Jorge, I.; Lopez-Martin, S.; Ursa, A.; Sanchez-Madrid, F.; Vazquez, J.; Yanez-Mo, M. The intracellular interactome of tetraspanin-enriched microdomains reveals their function as sorting machineries toward exosomes. J. Biol. Chem. 2013, 288, 11649–11661. [Google Scholar] [CrossRef] [Green Version]
- Mazurov, D.; Barbashova, L.; Filatov, A. Tetraspanin protein cd9 interacts with metalloprotease cd10 and enhances its release via exosomes. FEBS J. 2013, 280, 1200–1213. [Google Scholar] [CrossRef]
- Zhu, X.; Badawi, M.; Pomeroy, S.; Sutaria, D.S.; Xie, Z.; Baek, A.; Jiang, J.; Elgamal, O.A.; Mo, X.; Perle, K.; et al. Comprehensive toxicity and immunogenicity studies reveal minimal effects in mice following sustained dosing of extracellular vesicles derived from hek293t cells. J. Extracell. Vesicles 2017, 6, 1324730. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Chen, X.; Yi, J.; Liu, Y.; Li, D.; Wang, J.; Hou, D.; Jiang, X.; Zhang, J.; Wang, J.; et al. Identification and characterization of 293t cell-derived exosomes by profiling the protein, mrna and microrna components. PLoS ONE 2016, 11, e0163043. [Google Scholar] [CrossRef] [Green Version]
- Viaud, S.; Thery, C.; Ploix, S.; Tursz, T.; Lapierre, V.; Lantz, O.; Zitvogel, L.; Chaput, N. Dendritic cell-derived exosomes for cancer immunotherapy: What’s next? Cancer Res. 2010, 70, 1281–1285. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Li, J.; Peng, Y.; Du, Y.; Yang, Z.; Qi, X. Dendritic cell derived exosomes loaded neoantigens for personalized cancer immunotherapies. J. Control. Release 2023, 353, 423–433. [Google Scholar] [CrossRef] [PubMed]
- Gatti, S.; Bruno, S.; Deregibus, M.C.; Sordi, A.; Cantaluppi, V.; Tetta, C.; Camussi, G. Microvesicles derived from human adult mesenchymal stem cells protect against ischaemia-reperfusion-induced acute and chronic kidney injury. Nephrol. Dial. Transpl. 2011, 26, 1474–1483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, T.; Yan, Y.; Wang, B.; Qian, H.; Zhang, X.; Shen, L.; Wang, M.; Zhou, Y.; Zhu, W.; Li, W.; et al. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate liver fibrosis. Stem Cells Dev. 2013, 22, 845–854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yamaguchi, T.; Izumi, Y.; Nakamura, Y.; Yamazaki, T.; Shiota, M.; Sano, S.; Tanaka, M.; Osada-Oka, M.; Shimada, K.; Miura, K.; et al. Repeated remote ischemic conditioning attenuates left ventricular remodeling via exosome-mediated intercellular communication on chronic heart failure after myocardial infarction. Int. J. Cardiol. 2015, 178, 239–246. [Google Scholar] [CrossRef] [PubMed]
- Fang, Y.; Zhang, Y.; Zhou, J.; Cao, K. Adipose-derived mesenchymal stem cell exosomes: A novel pathway for tissues repair. Cell Tissue Bank. 2019, 20, 153–161. [Google Scholar] [CrossRef]
- Yeo, R.W.; Lai, R.C.; Zhang, B.; Tan, S.S.; Yin, Y.; Teh, B.J.; Lim, S.K. Mesenchymal stem cell: An efficient mass producer of exosomes for drug delivery. Adv. Drug Deliv. Rev. 2013, 65, 336–341. [Google Scholar]
- Wang, X.; Tian, L.; Lu, J.; Ng, I.O. Exosomes and cancer—Diagnostic and prognostic biomarkers and therapeutic vehicle. Oncogenesis 2022, 11, 54. [Google Scholar] [CrossRef]
- Li, Y.; Chen, Z.K.; Duan, X.; Zhang, H.J.; Xiao, B.L.; Wang, K.M.; Chen, G. Targeted inhibition of tumor-derived exosomes as a novel therapeutic option for cancer. Exp. Mol. Med. 2022, 54, 1379–1389. [Google Scholar] [CrossRef]
- Gao, Y.; Zhang, H.; Zhou, N.; Xu, P.; Wang, J.; Gao, Y.; Jin, X.; Liang, X.; Lv, J.; Zhang, Y.; et al. Methotrexate-loaded tumour-cell-derived microvesicles can relieve biliary obstruction in patients with extrahepatic cholangiocarcinoma. Nat. Biomed. Eng. 2020, 4, 743–753. [Google Scholar] [CrossRef]
- Livshits, M.A.; Khomyakova, E.; Evtushenko, E.G.; Lazarev, V.N.; Kulemin, N.A.; Semina, S.E.; Generozov, E.V.; Govorun, V.M. Isolation of exosomes by differential centrifugation: Theoretical analysis of a commonly used protocol. Sci. Rep. 2015, 5, 17319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, D.; Zhang, W.; Zhang, H.; Zhang, F.; Chen, L.; Ma, L.; Larcher, L.M.; Chen, S.; Liu, N.; Zhao, Q.; et al. Progress, opportunity, and perspective on exosome isolation—Efforts for efficient exosome-based theranostics. Theranostics 2020, 10, 3684–3707. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Wang, L.; Zhu, L.; Xu, Z.; Liu, Y.; Li, Z.; Zhou, J.; Luo, F. Exosomes as drug carriers in anti-cancer therapy. Front. Cell Dev. Biol. 2022, 10, 728616. [Google Scholar] [CrossRef]
- Ding, L.; Yang, X.; Gao, Z.; Effah, C.Y.; Zhang, X.; Wu, Y.; Qu, L. A holistic review of the state-of-the-art microfluidics for exosome separation: An overview of the current status, existing obstacles, and future outlook. Small 2021, 17, e2007174. [Google Scholar] [CrossRef] [PubMed]
- An, M.; Wu, J.; Zhu, J.; Lubman, D.M. Comparison of an optimized ultracentrifugation method versus size-exclusion chromatography for isolation of exosomes from human serum. J. Proteome Res. 2018, 17, 3599–3605. [Google Scholar] [CrossRef]
- Guo, J.; Wu, C.; Lin, X.; Zhou, J.; Zhang, J.; Zheng, W.; Wang, T.; Cui, Y. Establishment of a simplified dichotomic size-exclusion chromatography for isolating extracellular vesicles toward clinical applications. J. Extracell. Vesicles 2021, 10, e12145. [Google Scholar] [CrossRef]
- Joo, H.S.; Suh, J.H.; Lee, H.J.; Bang, E.S.; Lee, J.M. Current knowledge and future perspectives on mesenchymal stem cell-derived exosomes as a new therapeutic agent. Int. J. Mol. Sci. 2020, 21, 727. [Google Scholar] [CrossRef] [Green Version]
- Rider, M.A.; Hurwitz, S.N.; Meckes, D.G., Jr. Extrapeg: A polyethylene glycol-based method for enrichment of extracellular vesicles. Sci. Rep. 2016, 6, 23978. [Google Scholar] [CrossRef]
- Zhang, Y.; Bi, J.; Huang, J.; Tang, Y.; Du, S.; Li, P. Exosome: A review of its classification, isolation techniques, storage, diagnostic and targeted therapy applications. Int. J. Nanomed. 2020, 15, 6917–6934. [Google Scholar] [CrossRef]
- Piffoux, M.; Volatron, J.; Cherukula, K.; Aubertin, K.; Wilhelm, C.; Silva, A.K.A.; Gazeau, F. Engineering and loading therapeutic extracellular vesicles for clinical translation: A data reporting frame for comparability. Adv. Drug Deliv. Rev. 2021, 178, 113972. [Google Scholar] [CrossRef]
- Wang, J.; Chen, D.; Ho, E.A. Challenges in the development and establishment of exosome-based drug delivery systems. J. Control. Release 2021, 329, 894–906. [Google Scholar] [CrossRef] [PubMed]
- Pascucci, L.; Cocce, V.; Bonomi, A.; Ami, D.; Ceccarelli, P.; Ciusani, E.; Vigano, L.; Locatelli, A.; Sisto, F.; Doglia, S.M.; et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: A new approach for drug delivery. J. Control. Release 2014, 192, 262–270. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Wang, Y.; Bai, M.; Wang, J.; Zhu, K.; Liu, R.; Ge, S.; Li, J.; Ning, T.; Deng, T.; et al. Exosomes serve as nanoparticles to suppress tumor growth and angiogenesis in gastric cancer by delivering hepatocyte growth factor sirna. Cancer Sci. 2018, 109, 629–641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, X.; Lu, Y.; Xu, Y.; Hou, S.; Huang, J.; Wang, B.; Zhao, J.; Xia, S.; Fan, S.; Yu, X.; et al. Exosomal transfer of mir-501 confers doxorubicin resistance and tumorigenesis via targeting of blid in gastric cancer. Cancer Lett. 2019, 459, 122–134. [Google Scholar] [CrossRef] [PubMed]
- Kimiz-Gebologlu, I.; Oncel, S.S. Exosomes: Large-scale production, isolation, drug loading efficiency, and biodistribution and uptake. J. Control. Release 2022, 347, 533–543. [Google Scholar] [CrossRef]
- Xi, X.M.; Xia, S.J.; Lu, R. Drug loading techniques for exosome-based drug delivery systems. Pharmazie 2021, 76, 61–67. [Google Scholar]
- Xu, M.; Yang, Q.; Sun, X.; Wang, Y. Recent advancements in the loading and modification of therapeutic exosomes. Front. Bioeng. Biotechnol. 2020, 8, 586130. [Google Scholar] [CrossRef]
- Burgio, S.; Noori, L.; Marino Gammazza, A.; Campanella, C.; Logozzi, M.; Fais, S.; Bucchieri, F.; Cappello, F.; Caruso Bavisotto, C. Extracellular vesicles-based drug delivery systems: A new challenge and the exemplum of malignant pleural mesothelioma. Int. J. Mol. Sci. 2020, 21, 5432. [Google Scholar] [CrossRef]
- Fukuta, T.; Nishikawa, A.; Kogure, K. Low level electricity increases the secretion of extracellular vesicles from cultured cells. Biochem. Biophys. Rep. 2020, 21, 100713. [Google Scholar] [CrossRef]
- Ye, Z.; Zhang, T.; He, W.; Jin, H.; Liu, C.; Yang, Z.; Ren, J. Methotrexate-loaded extracellular vesicles functionalized with therapeutic and targeted peptides for the treatment of glioblastoma multiforme. ACS Appl. Mater. Interfaces 2018, 10, 12341–12350. [Google Scholar] [CrossRef]
- Kanchanapally, R.; Deshmukh, S.K.; Chavva, S.R.; Tyagi, N.; Srivastava, S.K.; Patel, G.K.; Singh, A.P.; Singh, S. Drug-loaded exosomal preparations from different cell types exhibit distinctive loading capability, yield, and antitumor efficacies: A comparative analysis. Int. J. Nanomed. 2019, 14, 531–541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Katakowski, M.; Buller, B.; Zheng, X.; Lu, Y.; Rogers, T.; Osobamiro, O.; Shu, W.; Jiang, F.; Chopp, M. Exosomes from marrow stromal cells expressing mir-146b inhibit glioma growth. Cancer Lett. 2013, 335, 201–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Taganov, K.D.; Boldin, M.P.; Chang, K.J.; Baltimore, D. Nf-kappab-dependent induction of microrna mir-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc. Natl. Acad. Sci. USA 2006, 103, 12481–12486. [Google Scholar] [CrossRef]
- Bhaumik, D.; Scott, G.K.; Schokrpur, S.; Patil, C.K.; Campisi, J.; Benz, C.C. Expression of microrna-146 suppresses nf-kappab activity with reduction of metastatic potential in breast cancer cells. Oncogene 2008, 27, 5643–5647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xia, H.; Qi, Y.; Ng, S.S.; Chen, X.; Li, D.; Chen, S.; Ge, R.; Jiang, S.; Li, G.; Chen, Y.; et al. Microrna-146b inhibits glioma cell migration and invasion by targeting mmps. Brain Res. 2009, 1269, 158–165. [Google Scholar] [CrossRef]
- Wu, P.Y.; Zhang, X.D.; Zhu, J.; Guo, X.Y.; Wang, J.F. Low expression of microrna-146b-5p and microrna-320d predicts poor outcome of large b-cell lymphoma treated with cyclophosphamide, doxorubicin, vincristine, and prednisone. Hum. Pathol. 2014, 45, 1664–1673. [Google Scholar] [CrossRef] [Green Version]
- Hsu, S.D.; Chu, C.H.; Tsou, A.P.; Chen, S.J.; Chen, H.C.; Hsu, P.W.; Wong, Y.H.; Chen, Y.H.; Chen, G.H.; Huang, H.D. Mirnamap 2.0: Genomic maps of micrornas in metazoan genomes. Nucleic Acids Res. 2008, 36, D165–D169. [Google Scholar] [CrossRef] [Green Version]
- Yoon, S.O.; Kim, E.K.; Lee, M.; Jung, W.Y.; Lee, H.; Kang, Y.; Jang, Y.J.; Hong, S.W.; Choi, S.H.; Yang, W.I. Nova1 inhibition by mir-146b-5p in the remnant tissue microenvironment defines occult residual disease after gastric cancer removal. Oncotarget 2016, 7, 2475–2495. [Google Scholar] [CrossRef]
- Shi, L.; Su, Y.; Zheng, Z.; Qi, J.; Wang, W.; Wang, C. Mir-146b-5p promotes colorectal cancer progression by targeting traf6. Exp. Ther. Med. 2022, 23, 231. [Google Scholar] [CrossRef]
- Yim, N.; Ryu, S.W.; Choi, K.; Lee, K.R.; Lee, S.; Choi, H.; Kim, J.; Shaker, M.R.; Sun, W.; Park, J.H.; et al. Exosome engineering for efficient intracellular delivery of soluble proteins using optically reversible protein-protein interaction module. Nat. Commun. 2016, 7, 12277. [Google Scholar] [CrossRef] [Green Version]
- Bellavia, D.; Raimondo, S.; Calabrese, G.; Forte, S.; Cristaldi, M.; Patinella, A.; Memeo, L.; Manno, M.; Raccosta, S.; Diana, P.; et al. Interleukin 3- receptor targeted exosomes inhibit in vitro and in vivo chronic myelogenous leukemia cell growth. Theranostics 2017, 7, 1333–1345. [Google Scholar] [CrossRef]
- Sun, D.; Zhuang, X.; Xiang, X.; Liu, Y.; Zhang, S.; Liu, C.; Barnes, S.; Grizzle, W.; Miller, D.; Zhang, H.G. A novel nanoparticle drug delivery system: The anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol. Ther. 2010, 18, 1606–1614. [Google Scholar] [CrossRef] [PubMed]
- Salarpour, S.; Forootanfar, H.; Pournamdari, M.; Ahmadi-Zeidabadi, M.; Esmaeeli, M.; Pardakhty, A. Paclitaxel incorporated exosomes derived from glioblastoma cells: Comparative study of two loading techniques. Daru 2019, 27, 533–539. [Google Scholar] [CrossRef] [PubMed]
- Hood, J.L. Post isolation modification of exosomes for nanomedicine applications. Nanomedicine 2016, 11, 1745–1756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haney, M.J.; Zhao, Y.; Jin, Y.S.; Li, S.M.; Bago, J.R.; Klyachko, N.L.; Kabanov, A.V.; Batrakova, E.V. Macrophage-derived extracellular vesicles as drug delivery systems for triple negative breast cancer (tnbc) therapy. J. Neuroimmune Pharmacol. 2020, 15, 487–500. [Google Scholar] [CrossRef]
- Zeng, H.; Guo, S.; Ren, X.; Wu, Z.; Liu, S.; Yao, X. Current strategies for exosome cargo loading and targeting delivery. Cells 2023, 12, 1416. [Google Scholar] [CrossRef] [PubMed]
- Fu, S.; Wang, Y.; Xia, X.; Zheng, J.C. Exosome engineering: Current progress in cargo loading and targeted delivery. NanoImpact 2020, 20, 100261. [Google Scholar] [CrossRef]
- Pomatto, M.A.C.; Bussolati, B.; D’Antico, S.; Ghiotto, S.; Tetta, C.; Brizzi, M.F.; Camussi, G. Improved loading of plasma-derived extracellular vesicles to encapsulate antitumor mirnas. Mol. Ther. Methods Clin. Dev. 2019, 13, 133–144. [Google Scholar] [CrossRef] [Green Version]
- Limoni, S.K.; Moghadam, M.F.; Moazzeni, S.M.; Gomari, H.; Salimi, F. Engineered exosomes for targeted transfer of sirna to her2 positive breast cancer cells. Appl. Biochem. Biotechnol. 2019, 187, 352–364. [Google Scholar] [CrossRef]
- Le Saux, S.; Aarrass, H.; Lai-Kee-Him, J.; Bron, P.; Armengaud, J.; Miotello, G.; Bertrand-Michel, J.; Dubois, E.; George, S.; Faklaris, O.; et al. Post-production modifications of murine mesenchymal stem cell (mmsc) derived extracellular vesicles (evs) and impact on their cellular interaction. Biomaterials 2020, 231, 119675. [Google Scholar] [CrossRef]
- Kim, M.S.; Haney, M.J.; Zhao, Y.; Mahajan, V.; Deygen, I.; Klyachko, N.L.; Inskoe, E.; Piroyan, A.; Sokolsky, M.; Okolie, O.; et al. Development of exosome-encapsulated paclitaxel to overcome mdr in cancer cells. Nanomedicine 2016, 12, 655–664. [Google Scholar] [CrossRef] [Green Version]
- Haney, M.J.; Klyachko, N.L.; Zhao, Y.; Gupta, R.; Plotnikova, E.G.; He, Z.; Patel, T.; Piroyan, A.; Sokolsky, M.; Kabanov, A.V.; et al. Exosomes as drug delivery vehicles for parkinson’s disease therapy. J. Control. Release 2015, 207, 18–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, P.; Busatto, S.; Huang, J.; Morad, G.; Moses, M.A. A facile magnetic extrusion method for preparing endosome-derived vesicles for cancer drug delivery. Adv. Funct. Mater. 2021, 31, 2008326. [Google Scholar] [CrossRef] [PubMed]
- Van Deun, J.; Roux, Q.; Deville, S.; Van Acker, T.; Rappu, P.; Miinalainen, I.; Heino, J.; Vanhaecke, F.; De Geest, B.G.; De Wever, O.; et al. Feasibility of mechanical extrusion to coat nanoparticles with extracellular vesicle membranes. Cells 2020, 9, 1797. [Google Scholar]
- Costa, A.P.; Xu, X.; Burgess, D.J. Freeze-anneal-thaw cycling of unilamellar liposomes: Effect on encapsulation efficiency. Pharm. Res. 2014, 31, 97–103. [Google Scholar]
- Cheng, Y.; Zeng, Q.; Han, Q.; Xia, W. Effect of ph, temperature and freezing-thawing on quantity changes and cellular uptake of exosomes. Protein Cell 2019, 10, 295–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lv, L.L.; Cao, Y.; Liu, D.; Xu, M.; Liu, H.; Tang, R.N.; Ma, K.L.; Liu, B.C. Isolation and quantification of micrornas from urinary exosomes/microvesicles for biomarker discovery. Int. J. Biol. Sci. 2013, 9, 1021–1031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sato, Y.T.; Umezaki, K.; Sawada, S.; Mukai, S.A.; Sasaki, Y.; Harada, N.; Shiku, H.; Akiyoshi, K. Engineering hybrid exosomes by membrane fusion with liposomes. Sci. Rep. 2016, 6, 21933. [Google Scholar] [CrossRef] [Green Version]
- Goh, W.J.; Lee, C.K.; Zou, S.; Woon, E.C.; Czarny, B.; Pastorin, G. Doxorubicin-loaded cell-derived nanovesicles: An alternative targeted approach for anti-tumor therapy. Int. J. Nanomed. 2017, 12, 2759–2767. [Google Scholar] [CrossRef] [Green Version]
- Fuhrmann, G.; Serio, A.; Mazo, M.; Nair, R.; Stevens, M.M. Active loading into extracellular vesicles significantly improves the cellular uptake and photodynamic effect of porphyrins. J. Control. Release 2015, 205, 35–44. [Google Scholar] [CrossRef]
- Sarikahya, N.B.; Nalbantsoy, A.; Top, H.; Gokturk, R.S.; Sumbul, H.; Kirmizigul, S. Immunomodulatory, hemolytic and cytotoxic activity potentials of triterpenoid saponins from eight cephalaria species. Phytomedicine 2018, 38, 135–144. [Google Scholar] [CrossRef]
- Shtam, T.A.; Kovalev, R.A.; Varfolomeeva, E.Y.; Makarov, E.M.; Kil, Y.V.; Filatov, M.V. Exosomes are natural carriers of exogenous sirna to human cells in vitro. Cell Commun. Signal. 2013, 11, 88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jafari, D.; Shajari, S.; Jafari, R.; Mardi, N.; Gomari, H.; Ganji, F.; Forouzandeh Moghadam, M.; Samadikuchaksaraei, A. Designer exosomes: A new platform for biotechnology therapeutics. BioDrugs 2020, 34, 567–586. [Google Scholar] [CrossRef] [PubMed]
- Frawley, T.; Piskareva, O. Extracellular vesicle dissemination of epidermal growth factor receptor and ligands and its role in cancer progression. Cancers 2020, 12, 3200. [Google Scholar] [CrossRef]
- Kim, G.; Lee, Y.; Ha, J.; Han, S.; Lee, M. Engineering exosomes for pulmonary delivery of peptides and drugs to inflammatory lung cells by inhalation. J. Control. Release 2021, 330, 684–695. [Google Scholar] [CrossRef]
- Oczypok, E.A.; Perkins, T.N.; Oury, T.D. All the “rage” in lung disease: The receptor for advanced glycation endproducts (rage) is a major mediator of pulmonary inflammatory responses. Paediatr. Respir. Rev. 2017, 23, 40–49. [Google Scholar] [CrossRef]
- Hanford, L.E.; Fattman, C.L.; Schaefer, L.M.; Enghild, J.J. Regulation of receptor for advanced glycation end products during bleomycin-induced lung injury. Am. J. Respir. Cell Mol. Biol. 2003, 29, S77. [Google Scholar]
- Nievergall, E.; Ramshaw, H.S.; Yong, A.S.; Biondo, M.; Busfield, S.J.; Vairo, G.; Lopez, A.F.; Hughes, T.P.; White, D.L.; Hiwase, D.K. Monoclonal antibody targeting of il-3 receptor alpha with csl362 effectively depletes cml progenitor and stem cells. Blood 2014, 123, 1218–1228. [Google Scholar] [CrossRef] [Green Version]
- Testa, U.; Riccioni, R.; Militi, S.; Coccia, E.; Stellacci, E.; Samoggia, P.; Latagliata, R.; Mariani, G.; Rossini, A.; Battistini, A.; et al. Elevated expression of il-3ralpha in acute myelogenous leukemia is associated with enhanced blast proliferation, increased cellularity, and poor prognosis. Blood 2002, 100, 2980–2988. [Google Scholar] [CrossRef] [Green Version]
- Kanki, S.; Jaalouk, D.E.; Lee, S.; Yu, A.Y.; Gannon, J.; Lee, R.T. Identification of targeting peptides for ischemic myocardium by in vivo phage display. J. Mol. Cell. Cardiol. 2011, 50, 841–848. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Chen, Y.; Zhao, Z.; Meng, Q.; Yu, Y.; Sun, J.; Yang, Z.; Chen, Y.; Li, J.; Ma, T.; et al. Engineered exosomes with ischemic myocardium-targeting peptide for targeted therapy in myocardial infarction. J. Am. Heart Assoc. 2018, 7, e008737. [Google Scholar] [CrossRef] [Green Version]
- Liang, Y.; Xu, X.; Li, X.; Xiong, J.; Li, B.; Duan, L.; Wang, D.; Xia, J. Chondrocyte-targeted microrna delivery by engineered exosomes toward a cell-free osteoarthritis therapy. ACS Appl. Mater. Interfaces 2020, 12, 36938–36947. [Google Scholar] [CrossRef]
- Li, Y.; Gao, Y.; Gong, C.; Wang, Z.; Xia, Q.; Gu, F.; Hu, C.; Zhang, L.; Guo, H.; Gao, S. A33 antibody-functionalized exosomes for targeted delivery of doxorubicin against colorectal cancer. Nanomedicine 2018, 14, 1973–1985. [Google Scholar] [CrossRef]
- Longatti, A.; Schindler, C.; Collinson, A.; Jenkinson, L.; Matthews, C.; Fitzpatrick, L.; Blundy, M.; Minter, R.; Vaughan, T.; Shaw, M.; et al. High affinity single-chain variable fragments are specific and versatile targeting motifs for extracellular vesicles. Nanoscale 2018, 10, 14230–14244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, Y.; Mok, H. Citraconylated exosomes for improved internalization into macrophages. Appl. Biol. Chem. 2019, 62, 26. [Google Scholar] [CrossRef]
- Tian, T.; Zhang, H.-X.; He, C.-P.; Fan, S.; Zhu, Y.-L.; Qi, C.; Huang, N.-P.; Xiao, Z.-D.; Lu, Z.-H.; Tannous, B.A.; et al. Surface functionalized exosomes as targeted drug delivery vehicles for cerebral ischemia therapy. Biomaterials 2018, 150, 137–149. [Google Scholar] [CrossRef] [PubMed]
- Cammarata, C.R.; Hughes, M.E.; Ofner, C.M., III. Carbodiimide induced cross-linking, ligand addition, and degradation in gelatin. Mol. Pharm. 2015, 12, 783–793. [Google Scholar] [CrossRef] [Green Version]
- Fang, X.; Tan, W. Aptamers generated from cell-selex for molecular medicine: A chemical biology approach. Acc. Chem. Res. 2010, 43, 48–57. [Google Scholar] [CrossRef] [Green Version]
- Bagheri, E.; Abnous, K.; Farzad, S.A.; Taghdisi, S.M.; Ramezani, M.; Alibolandi, M. Targeted doxorubicin-loaded mesenchymal stem cells-derived exosomes as a versatile platform for fighting against colorectal cancer. Life Sci. 2020, 261, 118369. [Google Scholar] [CrossRef]
- Zou, J.; Shi, M.; Liu, X.; Jin, C.; Xing, X.; Qiu, L.; Tan, W. Aptamer-functionalized exosomes: Elucidating the cellular uptake mechanism and the potential for cancer-targeted chemotherapy. Anal. Chem. 2019, 91, 2425–2430. [Google Scholar] [CrossRef]
- Yvon, A.M.; Wadsworth, P.; Jordan, M.A. Taxol suppresses dynamics of individual microtubules in living human tumor cells. Mol. Biol. Cell 1999, 10, 947–959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barbuti, A.M.; Chen, Z.S. Paclitaxel through the ages of anticancer therapy: Exploring its role in chemoresistance and radiation therapy. Cancers 2015, 7, 2360–2371. [Google Scholar] [CrossRef] [PubMed]
- Kandimalla, R.; Aqil, F.; Alhakeem, S.S.; Jeyabalan, J.; Tyagi, N.; Agrawal, A.; Yan, J.; Spencer, W.; Bondada, S.; Gupta, R.C. Targeted oral delivery of paclitaxel using colostrum-derived exosomes. Cancers 2021, 13, 3700. [Google Scholar] [CrossRef]
- Wang, P.; Wang, H.; Huang, Q.; Peng, C.; Yao, L.; Chen, H.; Qiu, Z.; Wu, Y.; Wang, L.; Chen, W. Exosomes from m1-polarized macrophages enhance paclitaxel antitumor activity by activating macrophages-mediated inflammation. Theranostics 2019, 9, 1714–1727. [Google Scholar] [CrossRef] [PubMed]
- Sheibani, M.; Azizi, Y.; Shayan, M.; Nezamoleslami, S.; Eslami, F.; Farjoo, M.H.; Dehpour, A.R. Doxorubicin-induced cardiotoxicity: An overview on pre-clinical therapeutic approaches. Cardiovasc. Toxicol. 2022, 22, 292–310. [Google Scholar] [CrossRef]
- Hadla, M.; Palazzolo, S.; Corona, G.; Caligiuri, I.; Canzonieri, V.; Toffoli, G.; Rizzolio, F. Exosomes increase the therapeutic index of doxorubicin in breast and ovarian cancer mouse models. Nanomedicine 2016, 11, 2431–2441. [Google Scholar] [CrossRef] [PubMed]
- Gong, C.; Tian, J.; Wang, Z.; Gao, Y.; Wu, X.; Ding, X.; Qiang, L.; Li, G.; Han, Z.; Yuan, Y.; et al. Functional exosome-mediated co-delivery of doxorubicin and hydrophobically modified microrna 159 for triple-negative breast cancer therapy. J. Nanobiotechnol. 2019, 17, 93. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.; Tang, W.; Yang, M.; Yin, Y.; Li, H.; Hu, F.; Tang, L.; Ma, X.; Zhang, Y.; Wang, Y. Inflammatory tumor microenvironment responsive neutrophil exosomes-based drug delivery system for targeted glioma therapy. Biomaterials 2021, 273, 120784. [Google Scholar] [CrossRef]
- Aqil, F.; Munagala, R.; Jeyabalan, J.; Agrawal, A.K.; Gupta, R. Exosomes for the enhanced tissue bioavailability and efficacy of curcumin. AAPS J. 2017, 19, 1691–1702. [Google Scholar] [CrossRef]
- Trotta, T.; Panaro, M.A.; Prifti, E.; Porro, C. Modulation of biological activities in glioblastoma mediated by curcumin. Nutr. Cancer 2019, 71, 1241–1253. [Google Scholar] [CrossRef]
- González-Sarrías, A.; Iglesias-Aguirre, C.E.; Cortés-Martín, A.; Vallejo, F.; Cattivelli, A.; del Pozo-Acebo, L.; Del Saz, A.; López de las Hazas, M.C.; Dávalos, A.; Espín, J.C. Milk-derived exosomes as nanocarriers to deliver curcumin and resveratrol in breast tissue and enhance their anticancer activity. Int. J. Mol. Sci. 2022, 23, 2860. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.G.; Kim, H.; Liu, C.; Yu, S.; Wang, J.; Grizzle, W.E.; Kimberly, R.P.; Barnes, S. Curcumin reverses breast tumor exosomes mediated immune suppression of nk cell tumor cytotoxicity. Biochim. Biophys. Acta 2007, 1773, 1116–1123. [Google Scholar] [CrossRef] [Green Version]
- Wu, H.; Zhou, J.; Zeng, C.; Wu, D.; Mu, Z.; Chen, B.; Xie, Y.; Ye, Y.; Liu, J. Curcumin increases exosomal tcf21 thus suppressing exosome-induced lung cancer. Oncotarget 2016, 7, 87081–87090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Osterman, C.J.; Lynch, J.C.; Leaf, P.; Gonda, A.; Ferguson Bennit, H.R.; Griffiths, D.; Wall, N.R. Curcumin modulates pancreatic adenocarcinoma cell-derived exosomal function. PLoS ONE 2015, 10, e0132845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Naldini, L. Ex vivo gene transfer and correction for cell-based therapies. Nat. Rev. Genet. 2011, 12, 301–315. [Google Scholar] [CrossRef] [PubMed]
- Kim, C.S.; Duncan, B.; Creran, B.; Rotello, V.M. Triggered nanoparticles as therapeutics. Nano Today 2013, 8, 439–447. [Google Scholar] [CrossRef] [Green Version]
- Tsai, W.C.; Hsu, S.D.; Hsu, C.S.; Lai, T.C.; Chen, S.J.; Shen, R.; Huang, Y.; Chen, H.C.; Lee, C.H.; Tsai, T.F.; et al. Microrna-122 plays a critical role in liver homeostasis and hepatocarcinogenesis. J. Clin. Investig. 2012, 122, 2884–2897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lou, G.; Song, X.; Yang, F.; Wu, S.; Wang, J.; Chen, Z.; Liu, Y. Exosomes derived from mir-122-modified adipose tissue-derived mscs increase chemosensitivity of hepatocellular carcinoma. J. Hematol. Oncol. 2015, 8, 122. [Google Scholar] [CrossRef] [Green Version]
- Nie, H.; Xie, X.; Zhang, D.; Zhou, Y.; Li, B.; Li, F.; Li, F.; Cheng, Y.; Mei, H.; Meng, H.; et al. Use of lung-specific exosomes for mirna-126 delivery in non-small cell lung cancer. Nanoscale 2020, 12, 877–887. [Google Scholar] [CrossRef] [PubMed]
- Lin, D.; Zhang, H.; Liu, R.; Deng, T.; Ning, T.; Bai, M.; Yang, Y.; Zhu, K.; Wang, J.; Duan, J.; et al. Irgd-modified exosomes effectively deliver cpt1a sirna to colon cancer cells, reversing oxaliplatin resistance by regulating fatty acid oxidation. Mol. Oncol. 2021, 15, 3430–3446. [Google Scholar] [CrossRef]
- Zhang, Q.; Zhang, H.; Ning, T.; Liu, D.; Deng, T.; Liu, R.; Bai, M.; Zhu, K.; Li, J.; Fan, Q.; et al. Exosome-delivered c-met sirna could reverse chemoresistance to cisplatin in gastric cancer. Int. J. Nanomed. 2020, 15, 2323–2335. [Google Scholar] [CrossRef] [Green Version]
- Liu, Q.; Dai, G.; Wu, Y.; Zhang, M.; Yang, M.; Wang, X.; Song, M.; Li, X.; Xia, R.; Wu, Z. Irgd-modified exosomes-delivered bcl6 sirna inhibit the progression of diffuse large b-cell lymphoma. Front. Oncol. 2022, 12, 822805. [Google Scholar] [CrossRef] [PubMed]
- Wong, W.Y.; Lee, M.M.; Chan, B.D.; Kam, R.K.; Zhang, G.; Lu, A.P.; Tai, W.C. Proteomic profiling of dextran sulfate sodium induced acute ulcerative colitis mice serum exosomes and their immunomodulatory impact on macrophages. Proteomics 2016, 16, 1131–1145. [Google Scholar] [CrossRef] [PubMed]
- Essandoh, K.; Yang, L.; Wang, X.; Huang, W.; Qin, D.; Hao, J.; Wang, Y.; Zingarelli, B.; Peng, T.; Fan, G.C. Blockade of exosome generation with gw4869 dampens the sepsis-induced inflammation and cardiac dysfunction. Biochim. Biophys. Acta 2015, 1852, 2362–2371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Song, J.; Kang, Y.; Chun, C.-H.; Jin, E.-J. Selective loading of exosomal hulc and mir-372 is responsible for chondrocyte death during oa pathogenesis. Anim. Cells Syst. 2017, 21, 397–403. [Google Scholar] [CrossRef] [Green Version]
- Garcia-Contreras, M.; Shah, S.H.; Tamayo, A.; Robbins, P.D.; Golberg, R.B.; Mendez, A.J.; Ricordi, C. Plasma-derived exosome characterization reveals a distinct microrna signature in long duration type 1 diabetes. Sci. Rep. 2017, 7, 5998. [Google Scholar] [CrossRef] [Green Version]
- Tomlinson, P.R.; Zheng, Y.; Fischer, R.; Heidasch, R.; Gardiner, C.; Evetts, S.; Hu, M.; Wade-Martins, R.; Turner, M.R.; Morris, J.; et al. Identification of distinct circulating exosomes in parkinson’s disease. Ann. Clin. Transl. Neurol. 2015, 2, 353–361. [Google Scholar] [CrossRef]
- Zhuang, X.; Xiang, X.; Grizzle, W.; Sun, D.; Zhang, S.; Axtell, R.C.; Ju, S.; Mu, J.; Zhang, L.; Steinman, L.; et al. Treatment of brain inflammatory diseases by delivering exosome encapsulated anti-inflammatory drugs from the nasal region to the brain. Mol. Ther. 2011, 19, 1769–1779. [Google Scholar] [CrossRef]
- Moghadam-Kia, S.; Werth, V.P. Prevention and treatment of systemic glucocorticoid side effects. Int. J. Dermatol. 2010, 49, 239–248. [Google Scholar] [CrossRef] [Green Version]
- Yan, F.; Zhong, Z.; Wang, Y.; Feng, Y.; Mei, Z.; Li, H.; Chen, X.; Cai, L.; Li, C. Exosome-based biomimetic nanoparticles targeted to inflamed joints for enhanced treatment of rheumatoid arthritis. J. Nanobiotechnol. 2020, 18, 115. [Google Scholar] [CrossRef]
- Li, H.; Feng, Y.; Zheng, X.; Jia, M.; Mei, Z.; Wang, Y.; Zhang, Z.; Zhou, M.; Li, C. M2-type exosomes nanoparticles for rheumatoid arthritis therapy via macrophage re-polarization. J. Control. Release 2022, 341, 16–30. [Google Scholar] [CrossRef] [PubMed]
- Oh, K.H.; Kang, M.; Kang, E.; Ryu, H.; Han, S.H.; Yoo, T.H.; Kim, S.W.; Chae, D.W.; Lee, K.B.; Park, S.K.; et al. The know-ckd study: What we have learned about chronic kidney diseases. Kidney Res. Clin. Pract. 2020, 39, 121–135. [Google Scholar] [CrossRef] [PubMed]
- Medina, K.R.P.; Jeong, J.C.; Ryu, J.W.; Kang, E.; Chin, H.J.; Na, K.Y.; Chae, D.W.; Kim, S. Comparison of outcomes of mild and severe community- and hospital-acquired acute kidney injury. Yonsei Med. J. 2022, 63, 902–907. [Google Scholar] [CrossRef] [PubMed]
- Merchant, R.A.; Vathsala, A. Healthy aging and chronic kidney disease. Kidney Res. Clin. Pract. 2022, 41, 644–656. [Google Scholar] [CrossRef]
- Bharati, J.; Jha, V. Global kidney health atlas: A spotlight on the asia-pacific sector. Kidney Res. Clin. Pract. 2022, 41, 22–30. [Google Scholar] [CrossRef]
- Kung, C.W.; Chou, Y.H. Acute kidney disease: An overview of the epidemiology, pathophysiology, and management. Kidney Res. Clin. Pract. 2023. [Google Scholar] [CrossRef]
- Wang, J.; Jiang, L.; Ding, S.; He, S.Y.; Liu, S.B.; Lu, Z.J.; Liu, Y.Z.; Hou, L.W.; Wang, B.S.; Zhang, J.B. Early enteral nutrition and sepsis-associated acute kidney injury: A propensity score matched cohort study based on the mimic-iii database. Yonsei Med. J. 2023, 64, 259–268. [Google Scholar] [CrossRef]
- An, J.N.; Kim, S.G.; Song, Y.R. When and why to start continuous renal replacement therapy in critically ill patients with acute kidney injury. Kidney Res. Clin. Pract. 2021, 40, 566–577. [Google Scholar] [CrossRef]
- Lim, C.C.; Gardner, D.; Ng, R.Z.; Chin, Y.M.; Tan, H.Z.; Mok, I.Y.; Choo, J.C. Synergistic impact of pre-diabetes and immunosuppressants on the risk of diabetes mellitus during treatment of glomerulonephritis and renal vasculitis. Kidney Res. Clin. Pract. 2020, 39, 172–179. [Google Scholar] [CrossRef]
- Han, S.Y.; Jung, C.Y.; Lee, S.H.; Lee, D.W.; Lee, S.; Kim, C.D.; Choi, B.S.; Kim, B.S. A multicenter, randomized, open-label, comparative, phase iv study to evaluate the efficacy and safety of combined treatment with mycophenolate mofetil and corticosteroids in advanced immunoglobulin a nephropathy. Kidney Res. Clin. Pract. 2022, 41, 452–461. [Google Scholar] [CrossRef]
- Kim, H.J.; Han, M.; Song, S.H.; Seong, E.Y. Clinical features and outcomes of elderly patients with antineutrophil cytoplasmic antibody-positive vasculitis: A single-center retrospective study. Kidney Res. Clin. Pract. 2022, 41, 209–218. [Google Scholar] [PubMed]
- Lee, S.A.; Choi, C.; Yoo, T.H. Extracellular vesicles in kidneys and their clinical potential in renal diseases. Kidney Res. Clin. Pract. 2021, 40, 194–207. [Google Scholar] [CrossRef] [PubMed]
- Tang, T.T.; Lv, L.L.; Wang, B.; Cao, J.Y.; Feng, Y.; Li, Z.L.; Wu, M.; Wang, F.M.; Wen, Y.; Zhou, L.T.; et al. Employing macrophage-derived microvesicle for kidney-targeted delivery of dexamethasone: An efficient therapeutic strategy against renal inflammation and fibrosis. Theranostics 2019, 9, 4740–4755. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Lee, S.A.; Yoon, H.; Kim, M.Y.; Yoo, J.K.; Ahn, S.H.; Park, C.H.; Park, J.; Nam, B.Y.; Park, J.T.; et al. Exosome-based delivery of super-repressor ikappabalpha ameliorates kidney ischemia-reperfusion injury. Kidney Int. 2021, 100, 570–584. [Google Scholar] [CrossRef]
- Choi, H.; Kim, Y.; Mirzaaghasi, A.; Heo, J.; Kim, Y.N.; Shin, J.H.; Kim, S.; Kim, N.H.; Cho, E.S.; In Yook, J.; et al. Exosome-based delivery of super-repressor ikappabalpha relieves sepsis-associated organ damage and mortality. Sci. Adv. 2020, 6, eaaz6980. [Google Scholar] [CrossRef] [Green Version]
- Cho, N.J.; Kim, D.Y.; Kwon, S.H.; Ha, T.W.; Kim, H.K.; Lee, M.R.; Chun, S.W.; Park, S.; Lee, E.Y.; Gil, H.W. Urinary exosomal microrna profiling in type 2 diabetes patients taking dipeptidyl peptidase-4 inhibitor compared with sulfonylurea. Kidney Res. Clin. Pract. 2021, 40, 383–391. [Google Scholar] [CrossRef]
- Collino, F.; Bruno, S.; Incarnato, D.; Dettori, D.; Neri, F.; Provero, P.; Pomatto, M.; Oliviero, S.; Tetta, C.; Quesenberry, P.J.; et al. Aki recovery induced by mesenchymal stromal cell-derived extracellular vesicles carrying micrornas. J. Am. Soc. Nephrol. 2015, 26, 2349–2360. [Google Scholar] [CrossRef] [Green Version]
- Tapparo, M.; Bruno, S.; Collino, F.; Togliatto, G.; Deregibus, M.C.; Provero, P.; Wen, S.; Quesenberry, P.J.; Camussi, G. Renal regenerative potential of extracellular vesicles derived from mirna-engineered mesenchymal stromal cells. Int. J. Mol. Sci. 2019, 20, 2381. [Google Scholar] [CrossRef] [Green Version]
- Hu, S.; Hu, H.; Wang, R.; He, H.; Shui, H. Microrna-29b prevents renal fibrosis by attenuating renal tubular epithelial cell-mesenchymal transition through targeting the pi3k/akt pathway. Int. Urol. Nephrol. 2021, 53, 1941–1950. [Google Scholar] [CrossRef]
- Wang, H.; Wang, B.; Zhang, A.; Hassounah, F.; Seow, Y.; Wood, M.; Ma, F.; Klein, J.D.; Price, S.R.; Wang, X.H. Exosome-mediated mir-29 transfer reduces muscle atrophy and kidney fibrosis in mice. Mol. Ther. 2019, 27, 571–583. [Google Scholar] [CrossRef] [Green Version]
- Kura, B.; Kalocayova, B.; Devaux, Y.; Bartekova, M. Potential clinical implications of mir-1 and mir-21 in heart disease and cardioprotection. Int. J. Mol. Sci. 2020, 21, 700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Song, Y.; Zhang, C.; Zhang, J.; Jiao, Z.; Dong, N.; Wang, G.; Wang, Z.; Wang, L. Localized injection of mirna-21-enriched extracellular vesicles effectively restores cardiac function after myocardial infarction. Theranostics 2019, 9, 2346–2360. [Google Scholar] [CrossRef]
- Bazan, H.A.; Hatfield, S.A.; O’Malley, C.B.; Brooks, A.J.; Lightell, D., Jr.; Woods, T.C. Acute loss of mir-221 and mir-222 in the atherosclerotic plaque shoulder accompanies plaque rupture. Stroke 2015, 46, 3285–3287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, Z.; Zhao, Z.; Yang, C.; Song, C. Transfer of microrna-221 from mesenchymal stem cell-derived extracellular vesicles inhibits atherosclerotic plaque formation. Transl. Res. 2020, 226, 83–95. [Google Scholar] [CrossRef] [PubMed]
- Schult, P.; Roth, H.; Adams, R.L.; Mas, C.; Imbert, L.; Orlik, C.; Ruggieri, A.; Pyle, A.M.; Lohmann, V. Microrna-122 amplifies hepatitis c virus translation by shaping the structure of the internal ribosomal entry site. Nat. Commun. 2018, 9, 2613. [Google Scholar] [CrossRef] [Green Version]
- Nazari-Jahantigh, M.; Wei, Y.; Noels, H.; Akhtar, S.; Zhou, Z.; Koenen, R.R.; Heyll, K.; Gremse, F.; Kiessling, F.; Grommes, J.; et al. Microrna-155 promotes atherosclerosis by repressing bcl6 in macrophages. J. Clin. Investig. 2012, 122, 4190–4202. [Google Scholar] [CrossRef] [Green Version]
- Bu, T.; Li, Z.; Hou, Y.; Sun, W.; Zhang, R.; Zhao, L.; Wei, M.; Yang, G.; Yuan, L. Exosome-mediated delivery of inflammation-responsive il-10 mrna for controlled atherosclerosis treatment. Theranostics 2021, 11, 9988–10000. [Google Scholar] [CrossRef]
- Andrade-Guerrero, J.; Santiago-Balmaseda, A.; Jeronimo-Aguilar, P.; Vargas-Rodriguez, I.; Cadena-Suarez, A.R.; Sanchez-Garibay, C.; Pozo-Molina, G.; Mendez-Catala, C.F.; Cardenas-Aguayo, M.D.; Diaz-Cintra, S.; et al. Alzheimer’s disease: An updated overview of its genetics. Int. J. Mol. Sci. 2023, 24, 3754. [Google Scholar] [CrossRef]
- Zong, Y.; Wang, H.; Dong, W.; Quan, X.; Zhu, H.; Xu, Y.; Huang, L.; Ma, C.; Qin, C. Mir-29c regulates bace1 protein expression. Brain Res. 2011, 1395, 108–115. [Google Scholar] [CrossRef]
- Wang, H.; Sui, H.; Zheng, Y.; Jiang, Y.; Shi, Y.; Liang, J.; Zhao, L. Curcumin-primed exosomes potently ameliorate cognitive function in ad mice by inhibiting hyperphosphorylation of the tau protein through the akt/gsk-3beta pathway. Nanoscale 2019, 11, 7481–7496. [Google Scholar] [CrossRef]
- Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.E.; Lang, A.E. Parkinson disease. Nat. Rev. Dis. Primers 2017, 3, 17013. [Google Scholar] [CrossRef]
- Haney, M.J.; Zhao, Y.; Li, S.; Higginbotham, S.M.; Booth, S.L.; Han, H.Y.; Vetro, J.A.; Mosley, R.L.; Kabanov, A.V.; Gendelman, H.E.; et al. Cell-mediated transfer of catalase nanoparticles from macrophages to brain endothelial, glial and neuronal cells. Nanomedicine 2011, 6, 1215–1230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vidal, M.; Sainte-Marie, J.; Philippot, J.R.; Bienvenue, A. Asymmetric distribution of phospholipids in the membrane of vesicles released during in vitro maturation of guinea pig reticulocytes: Evidence precluding a role for “aminophospholipid translocase”. J. Cell. Physiol. 1989, 140, 455–462. [Google Scholar] [CrossRef] [PubMed]
- Lobb, R.J.; Becker, M.; Wen, S.W.; Wong, C.S.; Wiegmans, A.P.; Leimgruber, A.; Moller, A. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J. Extracell. Vesicles 2015, 4, 27031. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Gu, H.; Huang, W.; Peng, J.; Li, Y.; Yang, L.; Qin, D.; Essandoh, K.; Wang, Y.; Peng, T.; et al. Hsp20-mediated activation of exosome biogenesis in cardiomyocytes improves cardiac function and angiogenesis in diabetic mice. Diabetes 2016, 65, 3111–3128. [Google Scholar] [CrossRef] [Green Version]
- Hessvik, N.P.; Overbye, A.; Brech, A.; Torgersen, M.L.; Jakobsen, I.S.; Sandvig, K.; Llorente, A. Pikfyve inhibition increases exosome release and induces secretory autophagy. Cell. Mol. Life Sci. 2016, 73, 4717–4737. [Google Scholar] [CrossRef] [PubMed]
- Messenger, S.W.; Woo, S.S.; Sun, Z.; Martin, T.F.J. A ca(2+)-stimulated exosome release pathway in cancer cells is regulated by munc13-4. J. Cell Biol. 2018, 217, 2877–2890. [Google Scholar] [CrossRef] [Green Version]
- Nakamura, K.; Sawada, K.; Kinose, Y.; Yoshimura, A.; Toda, A.; Nakatsuka, E.; Hashimoto, K.; Mabuchi, S.; Morishige, K.I.; Kurachi, H.; et al. Exosomes promote ovarian cancer cell invasion through transfer of cd44 to peritoneal mesothelial cells. Mol. Cancer Res. 2017, 15, 78–92. [Google Scholar] [CrossRef] [Green Version]
- Usman, W.M.; Pham, T.C.; Kwok, Y.Y.; Vu, L.T.; Ma, V.; Peng, B.; Chan, Y.S.; Wei, L.; Chin, S.M.; Azad, A.; et al. Efficient rna drug delivery using red blood cell extracellular vesicles. Nat. Commun. 2018, 9, 2359. [Google Scholar] [CrossRef] [Green Version]
- Parada, N.; Romero-Trujillo, A.; Georges, N.; Alcayaga-Miranda, F. Camouflage strategies for therapeutic exosomes evasion from phagocytosis. J. Adv. Res. 2021, 31, 61–74. [Google Scholar] [CrossRef]
- Yang, Z.; Shi, J.; Xie, J.; Wang, Y.; Sun, J.; Liu, T.; Zhao, Y.; Zhao, X.; Wang, X.; Ma, Y.; et al. Large-scale generation of functional mrna-encapsulating exosomes via cellular nanoporation. Nat. Biomed. Eng. 2020, 4, 69–83. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Koh, H.B.; Kim, H.J.; Kang, S.-W.; Yoo, T.-H. Exosome-Based Drug Delivery: Translation from Bench to Clinic. Pharmaceutics 2023, 15, 2042. https://doi.org/10.3390/pharmaceutics15082042
Koh HB, Kim HJ, Kang S-W, Yoo T-H. Exosome-Based Drug Delivery: Translation from Bench to Clinic. Pharmaceutics. 2023; 15(8):2042. https://doi.org/10.3390/pharmaceutics15082042
Chicago/Turabian StyleKoh, Hee Byung, Hyo Jeong Kim, Shin-Wook Kang, and Tae-Hyun Yoo. 2023. "Exosome-Based Drug Delivery: Translation from Bench to Clinic" Pharmaceutics 15, no. 8: 2042. https://doi.org/10.3390/pharmaceutics15082042
APA StyleKoh, H. B., Kim, H. J., Kang, S. -W., & Yoo, T. -H. (2023). Exosome-Based Drug Delivery: Translation from Bench to Clinic. Pharmaceutics, 15(8), 2042. https://doi.org/10.3390/pharmaceutics15082042