The Therapeutic Potential and Clinical Significance of Exosomes as Carriers of Drug Delivery System
Abstract
:1. Introduction
2. Formation and Understanding of Exosomes
2.1. Mechanism in the Secretion of Exosomes
2.2. Using Exosome Carriers in Drug Delivery System for Therapeutics
2.3. Advantages of Exosome Carriers for Drug Delivery
3. Tumors
3.1. Delivering Endogenous Biomolecules
3.2. Delivering Chemical Drugs
3.3. Delivering Engineered Drugs
4. Neurological Diseases
5. Autoimmune Diseases
6. Other Systemic Diseases
6.1. Skeletal Diseases
6.2. Cardiovascular Diseases
6.3. Urinary System Diseases
6.4. Cutaneous Disease
6.5. Metabolic Disease
7. The Conclusion and Future Breakthrough Directions of Exosome Carriers
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Boyiadzis, M.; Whiteside, T.L. Information transfer by exosomes: A new frontier in hematologic malignancies. Blood Rev. 2015, 29, 281–290. [Google Scholar] [CrossRef] [PubMed]
- Pan, B.-T.; Johnstone, R.M. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell 1983, 33, 967–978. [Google Scholar] [CrossRef] [PubMed]
- Lässer, C.; Alikhani, V.S.; Ekström, K.; Eldh, M.; Paredes, P.T.; Bossios, A.; Sjöstrand, M.; Gabrielsson, S.; Lötvall, J.; Valadi, H. Human saliva, plasma and breast milk exosomes contain RNA: Uptake by macrophages. J. Transl. Med. 2011, 9, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kowal, J.; Tkach, M.; Théry, C. Biogenesis and secretion of exosomes. Curr. Opin. Cell Biol. 2014, 29, 116–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, M.; Viennois, E.; Xu, C.; Merlin, D. Plant derived edible nanoparticles as a new therapeutic approach against diseases. Tissue Barriers 2016, 4, e1134415. [Google Scholar] [CrossRef] [PubMed]
- Pathan, M.; Fonseka, P.; Chitti, S.V.; Kang, T.; Sanwlani, R.; Van Deun, J.; Hendrix, A.; Mathivanan, S. Vesiclepedia 2019: A compendium of RNA, proteins, lipids and metabolites in extracellular vesicles. Nucleic Acids Res. 2019, 47, D516–D519. [Google Scholar] [CrossRef] [Green Version]
- Liang, Y.; Duan, L.; Lu, J.; Xia, J. Engineering exosomes for targeted drug delivery. Theranostics 2021, 11, 3183–3195. [Google Scholar] [CrossRef]
- Kowal, J.; Arras, G.; Colombo, M.; Jouve, M.; Morath, J.P.; Primdal-Bengtson, B.; Dingli, F.; Loew, D.; Tkach, M.; Théry, C. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc. Natl. Acad. Sci. USA 2016, 113, E968–E977. [Google Scholar] [CrossRef] [Green Version]
- Jeppesen, D.K.; Fenix, A.M.; Franklin, J.L.; Higginbotham, J.N.; Zhang, Q.; Zimmerman, L.J.; Liebler, D.C.; Ping, J.; Liu, Q.; Evans, R.; et al. Reassessment of Exosome Composition. Cell 2019, 177, 428–445.e18. [Google Scholar] [CrossRef] [Green Version]
- Ragusa, M.; Barbagallo, C.; Cirnigliaro, M.; Battaglia, R.; Brex, D.; Caponnetto, A.; Barbagallo, D.; Di Pietro, C.; Purrello, M. Asymmetric RNA Distribution among Cells and Their Secreted Exosomes: Biomedical Meaning and Considerations on Diagnostic Applications. Front. Mol. Biosci. 2017, 4, 66. [Google Scholar] [CrossRef]
- Xu, X.; Xu, L.; Zhang, P.; Ouyang, K.; Xiao, Y.; Xiong, J.; Wang, D.; Liang, Y.; Duan, L. Effects of ATP9A on Extracellular Vesicle Release and Exosomal Lipid Composition. Oxidative Med. Cell. Longev. 2020, 2020, 8865499. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Li, J.; Wang, H.; Zhao, J.; Yan, M.; He, H.; Yu, S. Exosomes: Potential Biomarkers and Functions in Head and Neck Squamous Cell Carcinoma. Front. Mol. Biosci. 2022, 9, 881794. [Google Scholar] [CrossRef] [PubMed]
- Horváth, P.; Müller-Reichert, T. A Structural View on ESCRT-Mediated Abscission. Front. Cell Dev. Biol. 2020, 8, 586880. [Google Scholar] [CrossRef] [PubMed]
- Zhen, Y.; Spangenberg, H.; Munson, M.J.; Brech, A.; Schink, K.O.; Tan, K.-W.; Sørensen, V.; Wenzel, E.M.; Radulovic, M.; Engedal, N.; et al. ESCRT-mediated phagophore sealing during mitophagy. Autophagy 2020, 16, 826–841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jackson, C.E.; Scruggs, B.S.; Schaffer, J.E.; Hanson, P.I. Effects of Inhibiting VPS4 Support a General Role for ESCRTs in Extracellular Vesicle Biogenesis. Biophys. J. 2017, 113, 1342–1352. [Google Scholar] [CrossRef] [Green Version]
- Larios, J.; Mercier, V.; Roux, A.; Gruenberg, J. ALIX- and ESCRT-III–dependent sorting of tetraspanins to exosomes. J. Cell Biol. 2020, 219. [Google Scholar] [CrossRef] [Green Version]
- Meldolesi, J. Exosomes and Ectosomes in Intercellular Communication. Curr. Biol. 2018, 28, R435–R444. [Google Scholar] [CrossRef] [Green Version]
- Higa, L.H.; Jerez, H.E.; de Farias, M.A.; Portugal, R.V.; Romero, E.L.; Morilla, M.J. Ultra-small solid archaeolipid nanoparticles for active targeting to macrophages of the inflamed mucosa. Nanomedicine 2017, 12, 1165–1175. [Google Scholar] [CrossRef]
- Martínez-Carmona, M.; Lozano, D.; Colilla, M.; Vallet-Regí, M. Lectin-conjugated pH-responsive mesoporous silica nanoparticles for targeted bone cancer treatment. Acta Biomater. 2018, 65, 393–404. [Google Scholar] [CrossRef]
- Allen, T.M.; Cullis, P.R. Drug Delivery Systems: Entering the Mainstream. Science 2004, 303, 1818–1822. [Google Scholar] [CrossRef]
- Yang, M.; Wu, S.Y. The Advances and Challenges in Utilizing Exosomes for Delivering Cancer Therapeutics. Front. Pharmacol. 2018, 9, 735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mehryab, F.; Rabbani, S.; Shahhosseini, S.; Shekari, F.; Fatahi, Y.; Baharvand, H.; Haeri, A. Exosomes as a next-generation drug delivery system: An update on drug loading approaches, characterization, and clinical application challenges. Acta Biomater. 2020, 113, 42–62. [Google Scholar] [CrossRef] [PubMed]
- Familtseva, A.; Jeremic, N.; Tyagi, S.C. Exosomes: Cell-created drug delivery systems. Mol. Cell. Biochem. 2019, 459, 1–6. [Google Scholar] [CrossRef] [PubMed]
- Zou, X.; Yuan, M.; Zhang, T.; Wei, H.; Xu, S.; Jiang, N.; Zheng, N.; Wu, Z. Extracellular vesicles expressing a single-chain variable fragment of an HIV-1 specific antibody selectively target Env+ tissues. Theranostics 2019, 9, 5657–5671. [Google Scholar] [CrossRef]
- Mentkowski, K.; Lang, J.K. Exosomes Engineered to Express a Cardiomyocyte Binding Peptide Demonstrate Improved Cardiac Retention in Vivo. Sci. Rep. 2019, 9, 10041. [Google Scholar] [CrossRef] [Green Version]
- Kim, M.S.; Haney, M.J.; Zhao, Y.; Yuan, D.; Deygen, I.; Klyachko, N.L.; Kabanov, A.V.; Batrakova, E.V. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: In vitro and in vivo evaluations. Nanomedicine 2018, 14, 195–204. [Google Scholar] [CrossRef]
- Gao, X.; Ran, N.; Dong, X.; Zuo, B.; Yang, R.; Zhou, Q.; Moulton, H.M.; Seow, Y.; Yin, H. Anchor peptide captures, targets, and loads exosomes of diverse origins for diagnostics and therapy. Sci. Transl. Med. 2018, 10, eaat0195. [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]
- Anand, S.; Samuel, M.; Kumar, S.; Mathivanan, S. Ticket to a bubble ride: Cargo sorting into exosomes and extracellular vesicles. Biochim. Biophys. Acta Proteins Proteom. 2019, 1867, 140203. [Google Scholar] [CrossRef]
- Lai, R.C.; Yeo, R.W.Y.; Tan, K.H.; Lim, S.K. Exosomes for drug delivery—A novel application for the mesenchymal stem cell. Biotechnol. Adv. 2013, 31, 543–551. [Google Scholar] [CrossRef]
- Liu, Q.; Rojas-Canales, D.M.; DiVito, S.J.; Shufesky, W.J.; Stolz, D.B.; Erdos, G.; Sullivan, M.L.; Gibson, G.A.; Watkins, S.C.; Larregina, A.T.; et al. Donor dendritic cell–derived exosomes promote allograft-targeting immune response. J. Clin. Investig. 2016, 126, 2805–2820. [Google Scholar] [CrossRef] [PubMed]
- Qiao, L.; Hu, S.; Huang, K.; Su, T.; Li, Z.; Vandergriff, A.; Cores, J.; Dinh, P.-U.; Allen, T.; Shen, D.; et al. Tumor cell-derived exosomes home to their cells of origin and can be used as Trojan horses to deliver cancer drugs. Theranostics 2020, 10, 3474–3487. [Google Scholar] [CrossRef] [PubMed]
- Xi, X.-M.; Xia, S.-J.; Lu, R. Drug loading techniques for exosome-based drug delivery systems. J. Pharm. Sci. 2021, 76, 61–67. [Google Scholar]
- Peng, Q.; Mu, H. The potential of protein–nanomaterial interaction for advanced drug delivery. J. Control. Release 2016, 225, 121–132. [Google Scholar] [CrossRef] [PubMed]
- Müller, J.; Prozeller, D.; Ghazaryan, A.; Kokkinopoulou, M.; Mailänder, V.; Morsbach, S.; Landfester, K. Beyond the protein corona—Lipids matter for biological response of nanocarriers. Acta Biomater. 2018, 71, 420–431. [Google Scholar] [CrossRef]
- Xiao, W.; Wang, Y.; Zhang, H.; Liu, Y.; Xie, R.; He, X.; Zhou, Y.; Liang, L.; Gao, H. The protein corona hampers the transcytosis of transferrin-modified nanoparticles through blood-brain barrier and attenuates their targeting ability to brain tumor. Biomaterials 2021, 274, 120888. [Google Scholar] [CrossRef]
- Sutaria, D.S.; Badawi, M.; Phelps, M.A.; Schmittgen, T.D. Achieving the Promise of Therapeutic Extracellular Vesicles: The Devil is in Details of Therapeutic Loading. Pharm. Res. 2017, 34, 1053–1066. [Google Scholar] [CrossRef] [Green Version]
- Fernandes, M.; Lopes, I.; Teixeira, J.; Botelho, C.; Gomes, A.C. Exosome-like Nanoparticles: A New Type of Nanocarrier. Curr. Med. Chem. 2020, 27, 3888–3905. [Google Scholar] [CrossRef]
- Zhang, Y.; Chopp, M.; Meng, Y.; Katakowski, M.; Xin, H.; Mahmood, A.; Xiong, Y. Effect of exosomes derived from multipluripotent mesenchymal stromal cells on functional recovery and neurovascular plasticity in rats after traumatic brain injury. J. Neurosurg. 2015, 122, 856–867. [Google Scholar] [CrossRef] [Green Version]
- Li, I.; Nabet, B.Y. Exosomes in the tumor microenvironment as mediators of cancer therapy resistance. Mol. Cancer 2019, 18, 32. [Google Scholar] [CrossRef]
- Tarasov, V.V.; Svistunov, A.A.; Chubarev, V.N.; Dostdar, S.A.; Sokolov, A.V.; Brzecka, A.; Sukocheva, O.; Neganova, M.E.; Klochkov, S.G.; Somasundaram, S.G.; et al. Extracellular vesicles in cancer nanomedicine. Semin. Cancer Biol. 2021, 69, 212–225. [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] [PubMed] [Green Version]
- Aspe, J.R.; Diaz Osterman, C.J.; Jutzy, J.M.; Deshields, S.; Whang, S.; Wall, N.R. Enhancement of Gemcitabine sensitivity in pancreatic adenocarcinoma by novel exosome-mediated delivery of the Survivin-T34A mutant. J. Extracell. Vesicles 2014, 3, 23244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nie, W.; Wu, G.; Zhang, J.; Huang, L.; Ding, J.; Jiang, A.; Zhang, Y.; Liu, Y.; Li, J.; Pu, K.; et al. Responsive Exosome Nano-bioconjugates for Synergistic Cancer Therapy. Angew. Chem. Int. Ed. 2019, 59, 2018–2022. [Google Scholar] [CrossRef] [PubMed]
- Hartman, Z.C.; Wei, J.; Glass, O.K.; Guo, H.; Lei, G.; Yang, X.-Y.; Osada, T.; Hobeika, A.; Delcayre, A.; Le Pecq, J.-B.; et al. Increasing vaccine potency through exosome antigen targeting. Vaccine 2011, 29, 9361–9367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Romagnoli, G.G.; Zelante, B.B.; Toniolo, P.A.; Migliori, I.K.; Barbuto, J.A.M. Dendritic Cell-Derived Exosomes may be a Tool for Cancer Immunotherapy by Converting Tumor Cells into Immunogenic Targets. Front. Immunol. 2015, 5, 692. [Google Scholar]
- Geis-Asteggiante, L.; Belew, A.T.; Clements, V.K.; Edwards, N.J.; Ostrand-Rosenberg, S.; El-Sayed, N.M.; Fenselau, C. Differential Content of Proteins, mRNAs, and miRNAs Suggests that MDSC and Their Exosomes May Mediate Distinct Immune Suppressive Functions. J. Proteome Res. 2018, 17, 486–498. [Google Scholar] [CrossRef]
- Dvorská, D.; Braný, D.; Ňachajová, M.; Halašová, E.; Danková, Z. Breast Cancer and the Other Non-Coding RNAs. Int. J. Mol. Sci. 2021, 22, 3280. [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]
- Ell, B.; Mercatali, L.; Ibrahim, T.; Campbell, N.; Schwarzenbach, H.; Pantel, K.; Amadori, D.; Kang, Y. Tumor-Induced Osteoclast miRNA Changes as Regulators and Biomarkers of Osteolytic Bone Metastasis. Cancer Cell 2013, 24, 542–556. [Google Scholar] [CrossRef] [Green Version]
- O’Brien, K.P.; Khan, S.; Gilligan, K.; Zafar, H.; Lalor, P.; Glynn, C.; O’Flatharta, C.; Ingoldsby, H.; Dockery, P.; De Bhulbh, A.; et al. Employing mesenchymal stem cells to support tumor-targeted delivery of extracellular vesicle (EV)-encapsulated microRNA-379. Oncogene 2018, 37, 2137–2149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ding, Y.; Cao, F.; Sun, H.; Wang, Y.; Liu, S.; Wu, Y.; Cui, Q.; Mei, W.; Li, F. Exosomes derived from human umbilical cord mesenchymal stromal cells deliver exogenous miR-145-5p to inhibit pancreatic ductal adenocarcinoma progression. Cancer Lett. 2019, 442, 351–361. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Kim, S.M.; Yang, Y.; Oh, S.J.; Hong, Y.; Seo, M.; Jang, M. Cancer-derived exosomes as a delivery platform of CRISPR/Cas9 confer cancer cell tropism-dependent targeting. J. Control. Release 2017, 266, 8–16. [Google Scholar] [CrossRef] [PubMed]
- Farooqi, A.A.; Desai, N.N.; Qureshi, M.Z.; Librelotto, D.R.N.; Gasparri, M.L.; Bishayee, A.; Nabavi, S.M.; Curti, V.; Daglia, M. Exosome biogenesis, bioactivities and functions as new delivery systems of natural compounds. Biotechnol. Adv. 2018, 36, 328–334. [Google Scholar] [CrossRef] [PubMed]
- Saari, H.; Lázaro-Ibáñez, E.; Viitala, T.; Vuorimaa-Laukkanen, E.; Siljander, P.; Yliperttula, M. Microvesicle- and exosome-mediated drug delivery enhances the cytotoxicity of Paclitaxel in autologous prostate cancer cells. J. Control. Release 2015, 220, 727–737. [Google Scholar] [CrossRef] [Green Version]
- Garofalo, M.; Saari, H.; Somersalo, P.; Crescenti, D.; Kuryk, L.; Aksela, L.; Capasso, C.; Madetoja, M.; Koskinen, K.; Oksanen, T.; et al. Antitumor effect of oncolytic virus and paclitaxel encapsulated in extracellular vesicles for lung cancer treatment. J. Control. Release 2018, 283, 223–234. [Google Scholar] [CrossRef]
- Tang, K.; Zhang, Y.; Zhang, H.; Xu, P.; Liu, J.; Ma, J.; Lv, M.; Li, D.; Katirai, F.; Shen, G.-X.; et al. Delivery of chemotherapeutic drugs in tumour cell-derived microparticles. Nat. Commun. 2012, 3, 1282. [Google Scholar] [CrossRef] [Green Version]
- Palazzolo, S.; Memeo, L.; Hadla, M.; Duzagac, F.; Steffan, A.; Perin, T.; Canzonieri, V.; Tuccinardi, T.; Caligiuri, I.; Rizzolio, F. Cancer Extracellular Vesicles: Next-Generation Diagnostic and Drug Delivery Nanotools. Cancers 2020, 12, 3165. [Google Scholar] [CrossRef]
- Rayamajhi, S.; Nguyen, T.D.T.; Marasini, R.; Aryal, S. Macrophage-derived exosome-mimetic hybrid vesicles for tumor targeted drug delivery. Acta Biomater. 2019, 94, 482–494. [Google Scholar] [CrossRef]
- Yong, T.; Zhang, X.; Bie, N.; Zhang, H.; Zhang, X.; Li, F.; Hakeem, A.; Hu, J.; Gan, L.; Santos, H.A.; et al. Tumor exosome-based nanoparticles are efficient drug carriers for chemotherapy. Nat. Commun. 2019, 10, 3838. [Google Scholar] [CrossRef] [Green Version]
- Jia, G.; Han, Y.; An, Y.; Ding, Y.; He, C.; Wang, X.; Tang, Q. NRP-1 targeted and cargo-loaded exosomes facilitate simultaneous imaging and therapy of glioma in vitro and in vivo. Biomaterials 2018, 178, 302–316. [Google Scholar] [CrossRef] [PubMed]
- Ghosh, S.; Ghosh, S. Exosome: The “Off-the-Shelf” Cellular Nanocomponent as a Potential Pathogenic Agent, a Disease Biomarker, and Neurotherapeutics. Front. Pharmacol. 2022, 13, 878058. [Google Scholar] [CrossRef] [PubMed]
- Doeppner, T.R.; Herz, J.; Görgens, A.; Schlechter, J.; Ludwig, A.-K.; Radtke, S.; de Miroschedji, K.; Horn, P.A.; Giebel, B.; Hermann, D.M. Extracellular Vesicles Improve Post-Stroke Neuroregeneration and Prevent Postischemic Immunosuppression. Stem Cells Transl. Med. 2015, 4, 1131–1143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kuang, Y.; Zheng, X.; Zhang, L.; Ai, X.; Venkataramani, V.; Kilic, E.; Hermann, D.M.; Majid, A.; Bähr, M.; Doeppner, T.R. Adipose-derived mesenchymal stem cells reduce autophagy in stroke mice by extracellular vesicle transfer of miR-25. J. Extracell. Vesicles 2020, 10, e12024. [Google Scholar] [CrossRef]
- Chen, W.; Wang, H.; Zhu, Z.; Feng, J.; Chen, L. Exosome-Shuttled circSHOC2 from IPASs Regulates Neuronal Autophagy and Ameliorates Ischemic Brain Injury via the miR-7670-3p/SIRT1 Axis. Mol. Ther. Nucleic Acids 2020, 22, 657–672. [Google Scholar] [CrossRef]
- Wu, W.; Liu, J.; Yang, C.; Xu, Z.; Huang, J.; Lin, J. Astrocyte-derived exosome-transported microRNA-34c is neuroprotective against cerebral ischemia/reperfusion injury via TLR7 and the NF-κB/MAPK pathways. Brain Res. Bull. 2020, 163, 84–94. [Google Scholar] [CrossRef]
- Ghosh, S.; Garg, S.; Ghosh, S. Cell-Derived Exosome Therapy: A Novel Approach to Treat Post-traumatic Brain Injury Mediated Neural Injury. ACS Chem. Neurosci. 2020, 11, 2045–2047. [Google Scholar] [CrossRef]
- Moon, G.J.; Sung, J.H.; Kim, D.H.; Kim, E.H.; Cho, Y.H.; Son, J.P.; Cha, J.M.; Bang, O.Y. Application of Mesenchymal Stem Cell-Derived Extracellular Vesicles for Stroke: Biodistribution and MicroRNA Study. Transl. Stroke Res. 2019, 10, 509–521. [Google Scholar] [CrossRef]
- Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [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]
- Wei, H.; Chen, J.; Wang, S.; Fu, F.; Zhu, X.; Wu, C.; Liu, Z.; Zhong, G.; Lin, J. A Nanodrug Consisting of Doxorubicin And Exosome Derived From Mesenchymal Stem Cells For Osteosarcoma Treatment In Vitro. Int. J. Nanomed. 2019, 14, 8603–8610. [Google Scholar] [CrossRef] [PubMed]
- Ferrantelli, F.; Chiozzini, C.; Leone, P.; Manfredi, F.; Federico, M. Engineered Extracellular Vesicles/Exosomes as a New Tool against Neurodegenerative Diseases. Pharmaceutics 2020, 12, 529. [Google Scholar] [CrossRef] [PubMed]
- Qu, M.; Lin, Q.; Huang, L.; Fu, Y.; Wang, L.; He, S.; Fu, Y.; Yang, S.; Zhang, Z.; Zhang, L.; et al. Dopamine-loaded blood exosomes targeted to brain for better treatment of Parkinson’s disease. J. Control. Release 2018, 287, 156–166. [Google Scholar] [CrossRef] [PubMed]
- 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] [Green Version]
- Zhou, X.; Xie, F.; Wang, L.; Zhang, L.; Zhang, S.; Fang, M.; Zhou, F. The function and clinical application of extracellular vesicles in innate immune regulation. Cell. Mol. Immunol. 2020, 17, 323–334. [Google Scholar] [CrossRef]
- Tavasolian, F.; Hosseini, A.Z.; Soudi, S.; Naderi, M. miRNA-146a Improves Immunomodulatory Effects of MSC-derived Exosomes in Rheumatoid Arthritis. Curr. Gene Ther. 2020, 20, 297–312. [Google Scholar] [CrossRef]
- Riazifar, M.; Mohammadi, M.R.; Pone, E.J.; Yeri, A.; Lässer, C.; Segaliny, A.I.; McIntyre, L.L.; Shelke, G.V.; Hutchins, E.; Hamamoto, A.; et al. Stem Cell-Derived Exosomes as Nanotherapeutics for Autoimmune and Neurodegenerative Disorders. ACS Nano 2019, 13, 6670–6688. [Google Scholar] [CrossRef]
- Avnir, Y.; Turjeman, K.; Tulchinsky, D.; Sigal, A.; Kizelsztein, P.; Tzemach, D.; Gabizon, A.; Barenholz, Y. Fabrication Principles and Their Contribution to the Superior In Vivo Therapeutic Efficacy of Nano-Liposomes Remote Loaded with Glucocorticoids. PLoS ONE 2011, 6, e25721. [Google Scholar] [CrossRef] [Green Version]
- Neupane, Y.R.; Mahtab, A.; Siddiqui, L.; Singh, A.; Gautam, N.; Rabbani, S.A.; Goel, H.; Talegaonkar, S. Biocompatible Nanovesicular Drug Delivery Systems with Targeting Potential for Autoimmune Diseases. Curr. Pharm. Des. 2020, 26, 5488–5502. [Google Scholar] [CrossRef]
- Kadry, M.O. Liposomal glutathione as a promising candidate for immunological rheumatoid arthritis therapy. Heliyon 2019, 5, e02162. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Casella, G.; Colombo, F.; Finardi, A.; Descamps, H.; Ill-Raga, G.; Spinelli, A.E.; Podini, P.; Bastoni, M.; Martino, G.; Muzio, L.; et al. Extracellular Vesicles Containing IL-4 Modulate Neuroinflammation in a Mouse Model of Multiple Sclerosis. Mol. Ther. 2018, 26, 2107–2118. [Google Scholar] [CrossRef] [Green Version]
- Kennel, K.A.; Drake, M.T. Adverse effects of bisphosphonates: Implications for osteoporosis management. Mayo. Clin. Proc. 2009, 84, 632–637. [Google Scholar] [CrossRef] [Green Version]
- Yang, X.; Chen, S.; Liu, X.; Yu, M.; Liu, X. Drug Delivery Based on Nanotechnology for Target Bone Disease. Curr. Drug Deliv. 2019, 16, 782–792. [Google Scholar] [CrossRef] [PubMed]
- Zha, Y.; Li, Y.; Lin, T.; Chen, J.; Zhang, S.; Wang, J. Progenitor cell-derived exosomes endowed with VEGF plasmids enhance osteogenic induction and vascular remodeling in large segmental bone defects. Theranostics 2021, 11, 397–409. [Google Scholar] [CrossRef] [PubMed]
- Luo, Z.-W.; Li, F.-X.; Liu, Y.-W.; Rao, S.-S.; Yin, H.; Huang, J.; Chen, C.-Y.; Hu, Y.; Zhang, Y.; Tan, Y.-J.; et al. Aptamer-functionalized exosomes from bone marrow stromal cells target bone to promote bone regeneration. Nanoscale 2019, 11, 20884–20892. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Liu, X.; Li, H.; Chen, C.; Hu, B.; Niu, X.; Li, Q.; Zhao, B.; Xie, Z.; Wang, Y. Exosomes/tricalcium phosphate combination scaffolds can enhance bone regeneration by activating the PI3K/Akt signaling pathway. Stem. Cell Res. Ther. 2016, 7, 136. [Google Scholar] [CrossRef] [Green Version]
- Xu, X.; Liang, Y.; Li, X.; Ouyang, K.; Wang, M.; Cao, T.; Li, W.; Liu, J.; Xiong, J.; Li, B.; et al. Exosome-mediated delivery of kartogenin for chondrogenesis of synovial fluid-derived mesenchymal stem cells and cartilage regeneration. Biomaterials 2021, 269, 120539. [Google Scholar] [CrossRef]
- Liu, Q.; Piao, H.; Wang, Y.; Zheng, D.; Wang, W. Circulating exosomes in cardiovascular disease: Novel carriers of biological information. Biomed. Pharmacother. 2021, 135, 111148. [Google Scholar] [CrossRef]
- Liu, Y.; Wang, M.; Liang, Y.; Wang, C.; Naruse, K.; Takahashi, K. Treatment of Oxidative Stress with Exosomes in Myocardial Ischemia. Int. J. Mol. Sci. 2021, 22, 1729. [Google Scholar] [CrossRef] [PubMed]
- Gollmann-Tepeköylü, C.; Pölzl, L.; Graber, M.; Hirsch, J.; Nägele, F.; Lobenwein, D.; Hess, M.W.; Blumer, M.J.; Kirchmair, E.; Zipperle, J.; et al. miR-19a-3p containing exosomes improve function of ischaemic myocardium upon shock wave therapy. Cardiovasc. Res. 2010, 116, 1226–1236. [Google Scholar] [CrossRef]
- Chen, Q.; Liu, Y.; Ding, X.; Li, Q.; Qiu, F.; Wang, M.; Shen, Z.; Zheng, H.; Fu, G. Bone marrow mesenchymal stem cell-secreted exosomes carrying microRNA-125b protect against myocardial ischemia reperfusion injury via targeting SIRT7. Mol. Cell. Biochem. 2020, 465, 103–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, H.; Wu, J.; Wu, J.; Fan, Q.; Zhou, J.; Wu, J.; Liu, S.; Zang, J.; Ye, J.; Xiao, M.; et al. Exosome-mediated targeted delivery of miR-210 for angiogenic therapy after cerebral ischemia in mice. J. Nanobiotechnol. 2019, 17, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, G.; Zhang, J.; Zhao, Q.; Zhuang, W.; Ding, J.; Zhang, C.; Gao, H.; Pang, D.-W.; Pu, K.; Xie, H.-Y. Molecularly Engineered Macrophage-Derived Exosomes with Inflammation Tropism and Intrinsic Heme Biosynthesis for Atherosclerosis Treatment. Angew. Chem. Int. Ed. 2020, 59, 4068–4074. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Yang, N.; Huang, X.; Zhu, Y.; Gao, S.; Liu, Z.; Cao, F.; Wang, Y. Melatonin Engineered Adipose-Derived Biomimetic Nanovesicles Regulate Mitochondrial Functions and Promote Myocardial Repair in Myocardial Infarction. Front. Cardiovasc. Med. 2022, 9, 789203. [Google Scholar] [CrossRef]
- Lu, M.; Xing, H.; Xun, Z.; Yang, T.; Ding, P.; Cai, C.; Wang, D.; Zhao, X. Exosome-based small RNA delivery: Progress and prospects. Asian J. Pharm. Sci. 2018, 13, 1–11. [Google Scholar] [CrossRef]
- Bruno, S.; Porta, S.; Bussolati, B. Extracellular vesicles in renal tissue damage and regeneration. Eur. J. Pharmacol. 2016, 790, 83–91. [Google Scholar] [CrossRef] [Green Version]
- Kim, S.; A Lee, S.; 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 IκBα ameliorates kidney ischemia-reperfusion injury. Kidney Int. 2021, 100, 570–584. [Google Scholar] [CrossRef]
- Tang, Z.; Tang, C.; Sun, C.; Ying, X.; Shen, R. M1 macrophage-derived exosomes synergistically enhance the anti- bladder cancer effect of gemcitabine. Aging 2022, 14, 7364–7377. [Google Scholar] [CrossRef]
- Wang, W.; Wu, C.; Jin, H. Exosomes in chronic inflammatory skin diseases and skin tumors. Exp. Dermatol. 2019, 28, 213–218. [Google Scholar] [CrossRef] [PubMed]
- Zhao, D.; Yu, Z.; Li, Y.; Wang, Y.; Li, Q.; Han, D. GelMA combined with sustained release of HUVECs derived exosomes for promoting cutaneous wound healing and facilitating skin regeneration. Histochem. J. 2020, 51, 251–263. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.; Bai, X.; Zhao, B.; Li, Y.; Zhang, Y.; Li, Z.; Wang, X.; Luo, L.; Han, F.; Zhang, J.; et al. Cell-free therapy based on adipose tissue stem cell-derived exosomes promotes wound healing via the PI3K/Akt signaling pathway. Exp. Cell Res. 2018, 370, 333–342. [Google Scholar] [CrossRef] [PubMed]
- Yue, T.; Ji, M.; Qu, H.; Guo, M.; Bai, F.; Zhang, Z.; Wang, W.; Gong, X.; Zhang, Z. Comprehensive analyses of long non-coding RNA expression profiles by RNA sequencing and exploration of their potency as biomarkers in psoriatic arthritis patients. BMC Immunol. 2019, 20, 28. [Google Scholar] [CrossRef] [Green Version]
- Shiekh, P.A.; Singh, A.; Kumar, A. Exosome laden oxygen releasing antioxidant and antibacterial cryogel wound dressing OxOBand alleviate diabetic and infectious wound healing. Biomaterials 2020, 249, 120020. [Google Scholar] [CrossRef]
- Wang, Y.; Cao, Z.; Wei, Q.; Ma, K.; Hu, W.; Huang, Q.; Su, J.; Li, H.; Zhang, C.; Fu, X. VH298-loaded extracellular vesicles released from gelatin methacryloyl hydrogel facilitate diabetic wound healing by HIF-1α-mediated enhancement of angiogenesis. Acta Biomater. 2022, 147, 342–355. [Google Scholar] [CrossRef]
- Xia, W.; Li, M.; Jiang, X.; Huang, X.; Gu, S.; Ye, J.; Zhu, L.; Hou, M.; Zan, T. Young fibroblast-derived exosomal microRNA-125b transfers beneficial effects on aged cutaneous wound healing. J. Nanobiotechnol. 2022, 20, 144. [Google Scholar] [CrossRef]
- Warshauer, J.T.; Bluestone, J.A.; Anderson, M.S. New Frontiers in the Treatment of Type 1 Diabetes. Cell Metab. 2020, 31, 46–61. [Google Scholar] [CrossRef]
- Xu, Y.-X.; Pu, S.-D.; Li, X.; Yu, Z.-W.; Zhang, Y.-T.; Tong, X.-W.; Shan, Y.-Y.; Gao, X.-Y. Exosomal ncRNAs: Novel therapeutic target and biomarker for diabetic complications. Pharmacol. Res. 2022, 178, 106135. [Google Scholar] [CrossRef]
- He, C.; Zheng, S.; Luo, Y.; Wang, B. Exosome Theranostics: Biology and Translational Medicine. Theranostics 2018, 8, 237–255. [Google Scholar] [CrossRef]
- Guo, Y.; Wan, Z.; Zhao, P.; Wei, M.; Liu, Y.; Bu, T.; Sun, W.; Li, Z.; Yuan, L. Ultrasound triggered topical delivery of Bmp7 mRNA for white fat browning induction via engineered smart exosomes. J. Nanobiotechnol. 2021, 19, 402. [Google Scholar] [CrossRef] [PubMed]
- Zhou, B.; Xu, K.; Zheng, X.; Chen, T.; Wang, J.; Song, Y.; Shao, Y.; Zheng, S. Application of exosomes as liquid biopsy in clinical diagnosis. Signal Transduct. Target. Ther. 2020, 5, 144. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Zhang, X.; Sai, B.; Wang, F.; Wang, L.; Wang, Y.; Zheng, L.; Li, G.; Tang, J.; Xiang, J. Hypoxic BMSC-derived exosomal miRNAs promote metastasis of lung cancer cells via STAT3-induced EMT. Mol. Cancer 2019, 18, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xunian, Z.; Kalluri, R. Biology and therapeutic potential of mesenchymal stem cell-derived exosomes. Cancer Sci. 2020, 111, 3100–3110. [Google Scholar] [CrossRef]
- Kok, V.C.; Yu, C.-C. Cancer-Derived Exosomes: Their Role in Cancer Biology and Biomarker Development. Int. J. Nanomed. 2020, 15, 8019–8036. [Google Scholar] [CrossRef]
- Zheng, Y.; He, R.; Wang, P.; Shi, Y.; Zhao, L.; Liang, J. Exosomes from LPS-stimulated macrophages induce neuroprotection and functional improvement after ischemic stroke by modulating microglial polarization. Biomater. Sci. 2019, 7, 2037–2049. [Google Scholar] [CrossRef]
- Wahlund, C.J.; Güclüler, G.; Hiltbrunner, S.; Veerman, R.E.; Näslund, T.I.; Gabrielsson, S. Exosomes from antigen-pulsed dendritic cells induce stronger antigen-specific immune responses than microvesicles in vivo. Sci. Rep. 2017, 7, 17095. [Google Scholar] [CrossRef] [Green Version]
- Xie, G.; Yang, H.; Peng, X.; Lin, L.; Wang, J.; Lin, K.; Cui, Z.; Li, J.; Xiao, H.; Liang, Y.; et al. Mast cell exosomes can suppress allergic reactions by binding to IgE. J. Allergy Clin. Immunol. 2018, 141, 788–791. [Google Scholar] [CrossRef] [Green Version]
- Hadley, E.E.; Sheller-Miller, S.; Saade, G.; Salomon, C.; Mesiano, S.; Taylor, R.N.; Taylor, B.D.; Menon, R. Amnion epithelial cell–derived exosomes induce inflammatory changes in uterine cells. Am. J. Obstet. Gynecol. 2018, 219, 478.e1–478.e21. [Google Scholar] [CrossRef]
- Kong, J.; Wang, F.; Zhang, J.; Cui, Y.; Pan, L.; Zhang, W.; Wen, J.; Liu, P. Exosomes of Endothelial Progenitor Cells Inhibit Neointima Formation After Carotid Artery Injury. J. Surg. Res. 2018, 232, 398–407. [Google Scholar] [CrossRef] [PubMed]
- Lin, J.; Li, J.; Huang, B.; Liu, J.; Chen, X.; Chen, X.-M.; Xu, Y.-M.; Huang, L.-F.; Wang, X.-Z. Exosomes: Novel Biomarkers for Clinical Diagnosis. Sci. World J. 2015, 2015, 657086. [Google Scholar] [CrossRef] [PubMed]
- Xiao, J.; Feng, S.; Wang, X.; Long, K.; Luo, Y.; Wang, Y.; Ma, J.; Tang, Q.; Jin, L.; Li, X.; et al. Identification of exosome-like nanoparticle-derived microRNAs from 11 edible fruits and vegetables. Peerj 2018, 6, e5186. [Google Scholar] [CrossRef]
- Yun, B.; Kim, Y.; Park, D.J.; Oh, S. Comparative analysis of dietary exosome-derived microRNAs from human, bovine and caprine colostrum and mature milk. J. Anim. Sci. Technol. 2021, 63, 593–602. [Google Scholar] [CrossRef] [PubMed]
- Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Bedina Zavec, A.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Frolova, L.; Li, I.T.S. Targeting Capabilities of Native and Bioengineered Extracellular Vesicles for Drug Delivery. Bioengineering 2022, 9, 496. [Google Scholar] [CrossRef]
- Abels, E.R.; Breakefield, X.O. Introduction to Extracellular Vesicles: Biogenesis, RNA Cargo Selection, Content, Release, and Uptake. Cell. Mol. Neurobiol. 2016, 36, 301–312. [Google Scholar] [CrossRef] [Green Version]
- Xie, S.; Zhang, Q.; Jiang, L. Current Knowledge on Exosome Biogenesis, Cargo-Sorting Mechanism and Therapeutic Implications. Membranes 2022, 12, 498. [Google Scholar] [CrossRef]
- Tian, Y.; Gong, M.; Hu, Y.; Liu, H.; Zhang, W.; Zhang, M.; Hu, X.; Aubert, D.; Zhu, S.; Wu, L.; et al. Quality and efficiency assessment of six extracellular vesicle isolation methods by nano-flow cytometry. J. Extracell. Vesicles 2019, 9, 1697028. [Google Scholar] [CrossRef]
- Bulut, O.; Gürsel, İ. Mesenchymal stem cell derived extracellular vesicles: Promising immunomodulators against autoimmune, autoinflammatory disorders and SARS-CoV-2 infection. Turk. J. Biol. 2020, 44, 273–282. [Google Scholar] [CrossRef]
Compositions | Reference | ||
---|---|---|---|
Protein | Cell surface protein | MHCI, MHCII, CD63, CD81, CD9, Lamp-2b, lactadherin | [6] |
Cytoplasmic proteins | Ubiquitin, ALIX, ESCRT, HSC70, HSP70, HSP60, HSP90, etc. | [7] | |
Cytoskeletal protein | Actin, tubulin, Keratin 8, 10, 18 and 19, α-Actinin-4 | [8] | |
Enzymes | Lysosomal, AAA ATPase, kinases, fatty acid synthase | [9] | |
Nucleic acid | mRNA, miRNA, ncRNA, siRNA | [10] | |
Lipids | Ceramide, sphingomyelin, phosphatidylserine, phosphatidylcholine, steroid lipids, etc. | [11] |
Exosome Sources | The Main Molecules | Molecules or Pathways Acting in the Process | The Main Mechanism | Reference |
---|---|---|---|---|
Mesenchymal stem cells (MSC) | N/A | N/A | Increased angiogenesis after ischemia | [64] |
Adipose-derived MSCs | miR-25-3p | Autophagy | Reduce neuronal autophagy | [65] |
The astrocyte origin | circSHOC2 | miR-7670-3p/SIRT1 | Inhibition of neuronal cell apoptosis | [66] |
The astrocyte origin | microRNA-34c | TLR7, NF-κB/MAPK | Reduce ischemia/reperfusion (I/R) injury | [67] |
Exosome Sources | Function | Reference |
---|---|---|
Mesenchymal stem cells | Promote tolerant immune responses, suppress inflammatory responses | [115] |
Cancer cell | Involved in tumor microenvironment and cancer development | [116] |
Macrophage | Intercellular communication | [117] |
Dendritic cells | Immune stimulation | [118] |
Mast cell | Participate in immune regulation | [119] |
Other cells (such as human amniotic epithelial cells, endothelial progenitor cells, etc.) | Reflect the pathophysiological state of their cell origin and can be used to diagnose and predict disease | [120,121] |
Body fluid (saliva, ascites, etc.) | Biomarker and therapeutic agent for diseases | [122] |
Heterogeneity (coconut, ginger, pea, bovine or caprine milk, etc.) | Mediate plant–animal intercellular communication | [123,124] |
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. |
© 2022 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
Li, T.; Li, X.; Han, G.; Liang, M.; Yang, Z.; Zhang, C.; Huang, S.; Tai, S.; Yu, S. The Therapeutic Potential and Clinical Significance of Exosomes as Carriers of Drug Delivery System. Pharmaceutics 2023, 15, 21. https://doi.org/10.3390/pharmaceutics15010021
Li T, Li X, Han G, Liang M, Yang Z, Zhang C, Huang S, Tai S, Yu S. The Therapeutic Potential and Clinical Significance of Exosomes as Carriers of Drug Delivery System. Pharmaceutics. 2023; 15(1):21. https://doi.org/10.3390/pharmaceutics15010021
Chicago/Turabian StyleLi, Tianwei, Xiaoqing Li, Guiping Han, Ming Liang, Zongrui Yang, Congyi Zhang, Shizhuan Huang, Sheng Tai, and Shan Yu. 2023. "The Therapeutic Potential and Clinical Significance of Exosomes as Carriers of Drug Delivery System" Pharmaceutics 15, no. 1: 21. https://doi.org/10.3390/pharmaceutics15010021
APA StyleLi, T., Li, X., Han, G., Liang, M., Yang, Z., Zhang, C., Huang, S., Tai, S., & Yu, S. (2023). The Therapeutic Potential and Clinical Significance of Exosomes as Carriers of Drug Delivery System. Pharmaceutics, 15(1), 21. https://doi.org/10.3390/pharmaceutics15010021