Engineered Extracellular Vesicles: Tailored-Made Nanomaterials for Medical Applications
Abstract
:1. Introduction
2. Re-Engineering of the Parental Cell
2.1. Genetic-Manipulation of Parental Cells
2.2. Exogenous Stimulation
2.3. Fusogenic Liposomes
2.4. Environmental Stimulation
2.4.1. External Stimulation
2.4.2. 3D Culture Platforms
2.4.3. Bioreactors
3. Modification of Isolated EVs
3.1. Passive Loading of EVs
EVs Incubation
3.2. Active Loading of EVs
3.2.1. Electroporation
3.2.2. Sonication
3.2.3. Extrusion
3.2.4. Freeze–Thawing
3.2.5. Saponin-Assisted Loading
3.3. Surface Modifications of EVs
3.3.1. Hydrophobic Insertion
3.3.2. Synthetic Lipid Nanoparticle Fusion
3.3.3. Covalent Modifications
3.3.4. Non-Covalent Modifications
4. Fabrication of Artificial EVs
4.1. Cell-Derived Nanovesicles (CDNs)
4.2. EV-Inspired Liposomes (EVLs)
5. EV-Functionalised Biomaterials
5.1. Considerations for Biomaterials Design
5.2. Challenges and Future Perspective
6. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Hourd, P.; Chandra, A.; Medcalf, N.; Williams, D.J. Regulatory challenges for the manufacture and scale-out of autologous cell therapies. In StemBook; Harvard Stem Cell Institute: Cambridge, UK, 2008. [Google Scholar] [CrossRef]
- Yanez-Mo, M.; Siljander, P.R.M.; Andreu, Z.; Zavec, A.B.; Borras, 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Williams, C.; Rodriguez-Barrueco, R.; Silva, J.M.; Zhang, W.J.; Hearn, S.; Elemento, O.; Paknejad, N.; Manova-Todorova, K.; Welte, K.; Bromberg, J.; et al. Double-stranded DNA in exosomes: A novel biomarker in cancer detection. Cell Res. 2014, 24, 766–769. [Google Scholar] [CrossRef] [Green Version]
- Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jurj, A.; Zanoaga, O.; Braicu, C.; Lazar, V.; Tomuleasa, C.; Irimie, A.; Berindan-Neagoe, I. A Comprehensive Picture of Extracellular Vesicles and Their Contents. Molecular Transfer to Cancer Cells. Cancers 2020, 12, 298. [Google Scholar] [CrossRef] [Green Version]
- Robbins, P.D.; Morelli, A.E. Regulation of immune responses by extracellular vesicles. Nat. Rev. Immunol. 2014, 14, 195–208. [Google Scholar] [CrossRef] [Green Version]
- Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [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] [Green Version]
- Azoidis, I.; Cox, S.C.; Davies, O.G. The role of extracellular vesicles in biomineralisation: Current perspective and application in regenerative medicine. J. Tissue Eng. 2018, 9. [Google Scholar] [CrossRef]
- Thery, 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. [Google Scholar] [CrossRef] [Green Version]
- Hass, R.; Kasper, C.; Bohm, S.; Jacobs, R. Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC. Cell Commun. Signal. 2011, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef] [PubMed]
- Anderson, H.C.; Mulhall, D.; Garimella, R. Role of extracellular membrane vesicles in the pathogenesis of various diseases, including cancer, renal diseases, atherosclerosis, and arthritis. Lab. Investig. 2010, 90, 1549–1557. [Google Scholar] [CrossRef] [PubMed]
- Gnecchi, M.; He, H.M.; Liang, O.D.; Melo, L.G.; Morello, F.; Mu, H.; Noiseux, N.; Zhang, L.N.; Pratt, R.E.; Ingwall, J.S.; et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat. Med. 2005, 11, 367–368. [Google Scholar] [CrossRef]
- Rani, S.; Ryan, A.E.; Griffin, M.D.; Ritter, T. Mesenchymal Stem Cell-derived Extracellular Vesicles: Toward Cell-free Therapeutic Applications. Mol. Ther. 2015, 23, 812–823. [Google Scholar] [CrossRef] [Green Version]
- EL Andaloussi, S.; Maeger, I.; Breakefield, X.O.; Wood, M.J.A. Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 2013, 12, 348–358. [Google Scholar] [CrossRef]
- Cha, J.M.; Shin, E.K.; Sung, J.H.; Moon, G.J.; Kim, E.H.; Cho, Y.H.; Park, H.D.; Bae, H.; Kim, J.; Bang, O.Y. Efficient scalable production of therapeutic microvesicles derived from human mesenchymal stem cells. Sci. Rep. 2018, 8. [Google Scholar] [CrossRef]
- van den Boorn, J.G.; Schlee, M.; Coch, C.; Hartmann, G. SiRNA delivery with exosome nanoparticles. Nat. Biotechnol. 2011, 29, 325–326. [Google Scholar] [CrossRef]
- Kikuchi, S.; Yoshioka, Y.; Prieto-Vila, M.; Ochiya, T. Involvement of Extracellular Vesicles in Vascular-Related Functions in Cancer Progression and Metastasis. Int. J. Mol. Sci. 2019, 20, 2584. [Google Scholar] [CrossRef] [Green Version]
- Clayton, A.; Harris, C.L.; Court, J.; Mason, M.D.; Morgan, B.P. Antigen-presenting cell exosomes are protected from complement-mediated lysis by expression of CD55 and CD59. Eur. J. Immunol. 2003, 33, 522–531. [Google Scholar] [CrossRef]
- Verweij, F.J.; Revenu, C.; Arras, G.; Dingli, F.; Loew, D.; Pegtel, D.M.; Follain, G.; Allio, G.; Goetz, J.G.; Zimmermann, P.; et al. Live Tracking of Inter-organ Communication by Endogenous Exosomes In Vivo. Dev. Cell 2019, 48, 573–589. [Google Scholar] [CrossRef] [Green Version]
- Xu, J.; Camfield, R.; Gorski, S.M. The interplay between exosomes and autophagy-partners in crime. J. Cell Sci. 2018, 131. [Google Scholar] [CrossRef] [Green Version]
- Quah, B.J.C.; O’Neill, H.C. The immunogenicity of dendritic cell-derived exosomes. Blood Cell Mol. Dis. 2005, 35, 94–110. [Google Scholar] [CrossRef]
- Di Rocco, G.; Baldari, S.; Toietta, G. Towards Therapeutic Delivery of Extracellular Vesicles: Strategies for In Vivo Tracking and Biodistribution Analysis. Stem Cells Int. 2016, 2016. [Google Scholar] [CrossRef] [Green Version]
- Wiklander, O.P.; Nordin, J.Z.; O’Loughlin, A.; Gustafsson, Y.; Corso, G.; Mäger, 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. [Google Scholar] [CrossRef] [Green Version]
- Gimona, M.; Pachler, K.; Laner-Plamberger, S.; Schallmoser, K.; Rohde, E. Manufacturing of Human Extracellular Vesicle-Based Therapeutics for Clinical Use. Int. J. Mol. Sci. 2017, 18, 1190. [Google Scholar] [CrossRef]
- Kim, H.; Kim, D.; Nam, H.; Moon, S.; Kwon, Y.J.; Lee, J.B. Engineered extracellular vesicles and their mimetics for clinical translation. Methods 2020, 177, 80–94. [Google Scholar] [CrossRef]
- Vader, P.; Mol, E.A.; Pasterkamp, G.; Schiffelers, R.M. Extracellular vesicles for drug delivery. Adv. Drug Deliver Rev. 2016, 106, 148–156. [Google Scholar] [CrossRef]
- Armstrong, J.P.K.; Holme, M.N.; Stevens, M.M. Re-Engineering Extracellular Vesicles as Smart Nanoscale Therapeutics. ACS Nano 2017, 11, 69–83. [Google Scholar] [CrossRef] [Green Version]
- Tao, S.C.; Guo, S.C.; Li, M.; Ke, Q.F.; Guo, Y.P.; Zhang, C.Q. Chitosan Wound Dressings Incorporating Exosomes Derived from MicroRNA-126-Overexpressing Synovium Mesenchymal Stem Cells Provide Sustained Release of Exosomes and Heal Full-Thickness Skin Defects in a Diabetic Rat Model. Stem Cell Transl. Med. 2017, 6, 736–747. [Google Scholar] [CrossRef]
- Kang, K.; Ma, R.L.; Cai, W.F.; Huang, W.; Paul, C.; Liang, J.L.; Wang, Y.H.; Zhao, T.J.; Kim, H.W.; Xu, M.F.; et al. Exosomes Secreted from CXCR4 Overexpressing Mesenchymal Stem Cells Promote Cardioprotection via Akt Signaling Pathway following Myocardial Infarction. Stem Cells Int. 2015, 2015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, C.Y.; Su, C.Q. Design strategies and application progress of therapeutic exosomes. Theranostics 2019, 9, 1015–1028. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Zhou, X.; Wei, M.; Gao, X.; Zhao, L.; Shi, R.; Sun, W.; Duan, Y.; Yang, G.; Yuan, L. In Vitro and in Vivo RNA Inhibition by CD9-HuR Functionalized Exosomes Encapsulated with miRNA or CRISPR/dCas9. Nano Lett. 2019, 19, 19–28. [Google Scholar] [CrossRef] [PubMed]
- Santangelo, L.; Giurato, G.; Cicchini, C.; Montaldo, C.; Mancone, C.; Tarallo, R.; Battistelli, C.; Alonzi, T.; Weisz, A.; Tripodi, M. The RNA-Binding Protein SYNCRIP Is a Component of the Hepatocyte Exosomal Machinery Controlling MicroRNA Sorting. Cell Rep. 2016, 17, 799–808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Villarroya-Beltri, C.; Gutierrez-Vazquez, C.; Sanchez-Cabo, F.; Perez-Hernandez, D.; Vazquez, J.; Martin-Cofreces, N.; Martinez-Herrera, D.J.; Pascual-Montano, A.; Mittelbrunn, M.; Sanchez-Madrid, F. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nat. Commun. 2013, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sutaria, D.S.; Jiang, J.; Elgamal, O.A.; Pomeroy, S.M.; Badawi, M.; Zhu, X.; Pavlovicz, R.; Azevedo-Pouly, A.C.P.; Chalmers, J.; Li, C.; et al. Low active loading of cargo into engineered extracellular vesicles results in inefficient miRNA mimic delivery. J. Extracell Vesicles 2017, 6, 1333882. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Ostrowski, M.; Carmo, N.B.; Krumeich, S.; Fanget, I.; Raposo, G.; Savina, A.; Moita, C.F.; Schauer, K.; Hume, A.N.; Freitas, R.P.; et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat. Cell Biol. 2010, 12, 19–30. [Google Scholar] [CrossRef] [Green Version]
- Hsu, C.; Morohashi, Y.; Yoshimura, S.; Manrique-Hoyos, N.; Jung, S.; Lauterbach, M.A.; Bakhti, M.; Gronborg, M.; Mobius, W.; Rhee, J.; et al. Regulation of exosome secretion by Rab35 and its GTPase-activating proteins TBC1D10A-C. J. Cell Biol. 2010, 189, 223–232. [Google Scholar] [CrossRef]
- Laulagnier, K.; Grand, D.; Dujardin, A.; Hamdi, S.; Vincent-Schneider, H.; Lankar, D.; Salles, J.P.; Bonnerot, C.; Perret, B.; Record, M. PLD2 is enriched on exosomes and its activity is correlated to the release of exosomes. FEBS Lett. 2004, 572, 11–14. [Google Scholar] [CrossRef] [Green Version]
- Hu, Q.; Su, H.; Li, J.; Lyon, C.; Tang, W.; Wan, M.; Hu, T.Y. Clinical applications of exosome membrane proteins. Precis Clin. Med. 2020, 3, 54–66. [Google Scholar] [CrossRef] [PubMed]
- Kooijmans, S.A.A.; Schiffelers, R.M.; Zarovni, N.; Vago, R. Modulation of tissue tropism and biological activity of exosomes and other extracellular vesicles: New nanotools for cancer treatment. Pharmacol. Res. 2016, 111, 487–500. [Google Scholar] [CrossRef] [PubMed]
- Hanna, E.; Remuzat, C.; Auquier, P.; Toumi, M. Advanced therapy medicinal products: Current and future perspectives. J. Mark. Access Health Policy 2016, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Y.; Lu, X.; He, J.; Zhao, W. Influence of erythropoietin on microvesicles derived from mesenchymal stem cells protecting renal function of chronic kidney disease. Stem Cell Res. Ther. 2015, 6, 100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lopatina, T.; Bruno, S.; Tetta, C.; Kalinina, N.; Porta, M.; Camussi, G. Platelet-derived growth factor regulates the secretion of extracellular vesicles by adipose mesenchymal stem cells and enhances their angiogenic potential. Cell Commun. Signal. 2014, 12. [Google Scholar] [CrossRef] [Green Version]
- Qu, Y.; Dubyak, G.R. P2X7 receptors regulate multiple types of membrane trafficking responses and non-classical secretion pathways. Purinerg. Signal. 2009, 5, 163–173. [Google Scholar] [CrossRef] [Green Version]
- Carreira, S.C.; Armstrong, J.P.K.; Seddon, A.M.; Perriman, A.W.; Hartley-Davies, R.; Schwarzacher, W. Ultra-fast stem cell labelling using cationised magnetoferritin. Nanoscale 2016, 8, 7474–7483. [Google Scholar] [CrossRef] [Green Version]
- Sarkar, D.; Vemula, P.K.; Zhao, W.; Gupta, A.; Karnik, R.; Karp, J.M. Engineered mesenchymal stem cells with self-assembled vesicles for systemic cell targeting. Biomaterials 2010, 31, 5266–5274. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.; Lee, H.; Goh, U.; Kim, J.; Jeong, M.; Lee, J.; Park, J.H. Cellular Engineering with Membrane Fusogenic Liposomes to Produce Functionalized Extracellular Vesicles. ACS Appl. Mater. Interfaces 2016, 8, 6790–6795. [Google Scholar] [CrossRef]
- Lee, J.; Kim, J.; Jeong, M.; Lee, H.; Goh, U.; Kim, H.; Kim, B.; Park, J.H. Liposome-based engineering of cells to package hydrophobic compounds in membrane vesicles for tumor penetration. Nano Lett. 2015, 15, 2938–2944. [Google Scholar] [CrossRef]
- Kolasinac, R.; Kleusch, C.; Braun, T.; Merkel, R.; Csiszar, A. Deciphering the Functional Composition of Fusogenic Liposomes. Int. J. Mol. Sci. 2018, 19, 346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kolasinac, R.; Jaksch, S.; Dreissen, G.; Braeutigam, A.; Merkel, R.; Csiszar, A. Influence of Environmental Conditions on the Fusion of Cationic Liposomes with Living Mammalian Cells. Nanomaterials 2019, 9, 1025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barenholz, Y. Liposome application: Problems and prospects. Curr. Opin. Colloid Interface Sci. 2001, 6, 66–77. [Google Scholar] [CrossRef]
- Burnley-Hall, N.; Willis, G.; Davis, J.; Rees, D.A.; James, P.E. Nitrite-derived nitric oxide reduces hypoxia-inducible factor 1alpha-mediated extracellular vesicle production by endothelial cells. Nitric Oxide 2017, 63, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Sun, L.; Wang, H.X.; Zhu, X.J.; Wu, P.H.; Chen, W.Q.; Zou, P.; Li, Q.B.; Chen, Z.C. Serum deprivation elevates the levels of microvesicles with different size distributions and selectively enriched proteins in human myeloma cells in vitro. Acta Pharmacol. Sin. 2014, 35, 381–393. [Google Scholar] [CrossRef]
- Li, J.; Lee, Y.; Johansson, H.J.; Mager, I.; Vader, P.; Nordin, J.Z.; Wiklander, O.P.; Lehtio, J.; Wood, M.J.; Andaloussi, S.E. Serum-free culture alters the quantity and protein composition of neuroblastoma-derived extracellular vesicles. J. Extracell Vesicles 2015, 4, 26883. [Google Scholar] [CrossRef] [PubMed]
- Eichholz, K.F.; Woods, I.; Riffault, M.; Johnson, G.P.; Corrigan, M.; Lowry, M.C.; Shen, N.; Labour, M.-N.; Wynne, K.; O’Driscoll, L.; et al. Human bone marrow stem/stromal cell osteogenesis is regulated via mechanically activated osteocyte-derived extracellular vesicles. Stem Cells Transl. Med. 2020, 1–17. [Google Scholar] [CrossRef]
- Davies, O.G.; Cox, S.C.; Williams, R.L.; Tsaroucha, D.; Dorrepaal, R.M.; Lewis, M.P.; Grover, L.M. Annexin-enriched osteoblast-derived vesicles act as an extracellular site of mineral nucleation within developing stem cell cultures. Sci. Rep. 2017, 7. [Google Scholar] [CrossRef]
- Ader, M.; Tanaka, E.M. Modeling human development in 3D culture. Curr. Opin. Cell Biol. 2014, 31, 23–28. [Google Scholar] [CrossRef]
- Zhang, Y.; Chopp, M.; Zhang, Z.G.; Katakowski, M.; Xin, H.; Qu, C.; Ali, M.; Mahmood, A.; Xiong, Y. Systemic administration of cell-free exosomes generated by human bone marrow derived mesenchymal stem cells cultured under 2D and 3D conditions improves functional recovery in rats after traumatic brain injury. Neurochem. Int. 2017, 111, 69–81. [Google Scholar] [CrossRef]
- Patel, D.B.; Santoro, M.; Born, L.J.; Fisher, J.P.; Jay, S.M. Towards rationally designed biomanufacturing of therapeutic extracellular vesicles: Impact of the bioproduction microenvironment. Biotechnol. Adv. 2018, 36, 2051–2059. [Google Scholar] [CrossRef] [PubMed]
- Tabata, Y. Biomaterial technology for tissue engineering applications. J. R Soc. Interface 2009, 6, S311–S324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patel, D.B.; Luthers, C.R.; Lerman, M.J.; Fisher, J.P.; Jay, S.M. Enhanced extracellular vesicle production and ethanol-mediated vascularization bioactivity via a 3D-printed scaffold-perfusion bioreactor system. Acta Biomater. 2019, 95, 236–244. [Google Scholar] [CrossRef] [PubMed]
- Pigeau, G.M.; Csaszar, E.; Dulgar-Tulloch, A. Commercial Scale Manufacturing of Allogeneic Cell Therapy. Front. Med. 2018, 5. [Google Scholar] [CrossRef] [PubMed]
- Stephenson, M.; Grayson, W. Recent advances in bioreactors for cell-based therapies. F1000Research 2018, 7, 517. [Google Scholar] [CrossRef] [PubMed]
- Palviainen, M.; Saari, H.; Karkkainen, O.; Pekkinen, J.; Auriola, S.; Yliperttula, M.; Puhka, M.; Hanhineva, K.; Siljander, P.R. Metabolic signature of extracellular vesicles depends on the cell culture conditions. J. Extracell Vesicles 2019, 8, 1596669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haraszti, R.A.; Miller, R.; Stoppato, M.; Sere, Y.Y.; Coles, A.; Didiot, M.C.; Wollacott, R.; Sapp, E.; Dubuke, M.L.; Li, X.N.; et al. Exosomes Produced from 3D Cultures of MSCs by Tangential Flow Filtration Show Higher Yield and Improved Activity. Mol. Ther. 2018, 26, 2838–2847. [Google Scholar] [CrossRef] [Green Version]
- Watson, D.C.; Bayik, D.; Srivatsan, A.; Bergamaschi, C.; Valentin, A.; Niu, G.; Bear, J.; Monninger, M.; Sun, M.; Morales-Kastresana, A.; et al. Efficient production and enhanced tumor delivery of engineered extracellular vesicles. Biomaterials 2016, 105, 195–205. [Google Scholar] [CrossRef] [Green Version]
- Akuma, P.; Okagu, O.D.; Udenigwe, C.C. Naturally Occurring Exosome Vesicles as Potential Delivery Vehicle for Bioactive Compounds. Front. Sustain. Food Syst. 2019, 3. [Google Scholar] [CrossRef]
- Sun, D.M.; Zhuang, X.Y.; Xiang, X.Y.; Liu, Y.L.; Zhang, S.Y.; Liu, C.R.; 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]
- Didiot, M.C.; Hall, L.M.; Coles, A.H.; Haraszti, R.A.; Godinho, B.M.; Chase, K.; Sapp, E.; Ly, S.; Alterman, J.F.; Hassler, M.R.; et al. Exosome-mediated Delivery of Hydrophobically Modified siRNA for Huntingtin mRNA Silencing. Mol. Ther. 2016, 24, 1836–1847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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.-Meth. Clin. Dev. 2019, 13, 133–144. [Google Scholar] [CrossRef] [Green Version]
- Wahlgren, J.; Karlson, T.D.; Brisslert, M.; Sani, F.V.; Telemo, E.; Sunnerhagen, P.; Valadi, H. Plasma exosomes can deliver exogenous short interfering RNA to monocytes and lymphocytes. Nucleic Acids Res. 2012, 40. [Google Scholar] [CrossRef] [Green Version]
- Kooijmans, S.A.A.; Stremersch, S.; Braeckmans, K.; de Smedt, S.C.; Hendrix, A.; Wood, M.J.A.; Schiffelers, R.M.; Raemdonck, K.; Vader, P. Electroporation-induced siRNA precipitation obscures the efficiency of siRNA loading into extracellular vesicles. J. Control. Release 2013, 172, 229–238. [Google Scholar] [CrossRef] [PubMed]
- Lamichhane, T.N.; Jeyaram, A.; Patel, D.B.; Parajuli, B.; Livingston, N.K.; Arumugasaamy, N.; Schardt, J.S.; Jay, S.M. Oncogene Knockdown via Active Loading of Small RNAs into Extracellular Vesicles by Sonication. Cell Mol. Bioeng. 2016, 9, 315–324. [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. Nanomed-Nanotechnol. Biol. Med. 2016, 12, 655–664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lamichhane, T.N.; Raiker, R.S.; Jay, S.M. Exogenous DNA Loading into Extracellular Vesicles via Electroporation is Size-Dependent and Enables Limited Gene Delivery. Mol. Pharmaceut. 2015, 12, 3650–3657. [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] [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]
- Cheng, Y.R.; Zeng, Q.Y.; Han, Q.; Xia, W.L. Effect of pH, temperature and freezing-thawing on quantity changes and cellular uptake of exosomes. Protein Cell 2019, 10, 295–299. [Google Scholar] [CrossRef] [Green Version]
- Luan, X.; Sansanaphongpricha, K.; Myers, I.; Chen, H.W.; Yuan, H.B.; Sun, D.X. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacol. Sin. 2017, 38, 754–763. [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] [PubMed] [Green Version]
- Podolak, I.; Galanty, A.; Sobolewska, D. Saponins as cytotoxic agents: A review. Wiley Interdisciplinary Rev. Nanomed. Nanobiotechnol. 2010, 9, 425–474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Loughlin, A.J.; Mager, I.; de Jong, O.G.; Varela, M.A.; Schiffelers, R.M.; El Andaloussi, S.; Wood, M.J.A.; Vader, P. Functional Delivery of Lipid-Conjugated siRNA by Extracellular Vesicles. Mol. Ther. 2017, 25, 1580–1587. [Google Scholar] [CrossRef] [Green Version]
- Batrakova, E.V.; Kim, M.S. Development and regulation of exosome-based therapy products. Wires Nanomed. Nanobi 2016, 8, 744–757. [Google Scholar] [CrossRef]
- Huang, L.; Gu, N.; Zhang, X.E.; Wang, D.B. Light-Inducible Exosome-Based Vehicle for Endogenous RNA Loading and Delivery to Leukemia Cells. Adv. Funct Mater. 2019, 29. [Google Scholar] [CrossRef]
- Kim, M.S.; Haney, M.J.; Zhao, Y.L.; Yuan, D.F.; 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. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 195–204. [Google Scholar] [CrossRef]
- Piffoux, M.; Silva, A.K.A.; Wilhelm, C.; Gazeau, F.; Tareste, D. Modification of Extracellular Vesicles by Fusion with Liposomes for the Design of Personalized Biogenic Drug Delivery Systems. ACS Nano 2018, 12, 6830–6842. [Google Scholar] [CrossRef]
- Kooijmans, S.A.A.; Fliervoet, L.A.L.; van der Meel, R.; Fens, M.H.A.M.; Heijnen, H.F.G.; Henegouwen, P.M.P.V.E.; Vader, P.; Schiffelers, R.M. PEGylated and targeted extracellular vesicles display enhanced cell specificity and circulation time. J. Control. Release 2016, 224, 77–85. [Google Scholar] [CrossRef]
- Smyth, T.; Petrova, K.; Payton, N.M.; Persaud, I.; Redzic, J.S.; Gruner, M.W.; Smith-Jones, P.; Anchordoquy, T.J. Surface Functionalization of Exosomes Using Click Chemistry. Bioconjugate Chem. 2014, 25, 1777–1784. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Wan, Y.; Wang, L.X.; Zhu, C.D.; Zheng, Q.; Wang, G.X.; Tong, J.L.; Fang, Y.; Xia, Y.Q.; Cheng, G.; He, X.; et al. Aptamer-Conjugated Extracellular Nanovesicles for Targeted Drug Delivery. Cancer Res. 2018, 78, 798–808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Biscans, A.; Haraszti, R.A.; Echeverria, D.; Miller, R.; Didiot, M.C.; Nikan, M.; Roux, L.; Aronin, N.; Khvorova, A. Hydrophobicity of Lipid-Conjugated siRNAs Predicts Productive Loading to Small Extracellular Vesicles. Mol. Ther. 2018, 26, 1520–1528. [Google Scholar] [CrossRef] [Green Version]
- Qi, H.Z.; Liu, C.Y.; Long, L.X.; Ren, Y.; Zhang, S.S.; Chang, X.D.; Qian, X.M.; Jia, H.H.; Zhao, J.; Sun, J.J.; et al. Blood Exosomes Endowed with Magnetic and Targeting Properties for Cancer Therapy. ACS Nano 2016, 10, 3323–3333. [Google Scholar] [CrossRef] [PubMed]
- Maguire, C.A.; Balaj, L.; Sivaraman, S.; Crommentuijn, M.H.W.; Ericsson, M.; Mincheva-Nilsson, L.; Baranov, V.; Gianni, D.; Tannous, B.A.; Sena-Esteves, M.; et al. Microvesicle-associated AAV Vector as a Novel Gene Delivery System. Mol. Ther. 2012, 20, 960–971. [Google Scholar] [CrossRef] [Green Version]
- Nakase, I.; Futaki, S. Combined treatment with a pH-sensitive fusogenic peptide and cationic lipids achieves enhanced cytosolic delivery of exosomes. Sci. Rep. 2015, 5. [Google Scholar] [CrossRef] [PubMed]
- Nel, A.E.; Madler, L.; Velegol, D.; Xia, T.; Hoek, E.M.V.; Somasundaran, P.; Klaessig, F.; Castranova, V.; Thompson, M. Understanding biophysicochemical interactions at the nano-bio interface. Nat. Mater. 2009, 8, 543–557. [Google Scholar] [CrossRef]
- Leroueil, P.R.; Berry, S.A.; Duthie, K.; Han, G.; Rotello, V.M.; McNerny, D.Q.; Baker, J.R.; Orr, B.G.; Holl, M.M.B. Wide varieties of cationic nanoparticles induce defects in supported lipid bilayers. Nano Lett. 2008, 8, 420–424. [Google Scholar] [CrossRef]
- Goh, W.J.; Zou, S.; Ong, W.Y.; Torta, F.; Alexandra, A.F.; Schiffelers, R.M.; Storm, G.; Wang, J.W.; Czarny, B.; Pastorin, G. Bioinspired Cell-Derived Nanovesicles versus Exosomes as Drug Delivery Systems: A Cost-Effective Alternative. Sci. Rep. 2017, 7. [Google Scholar] [CrossRef]
- Lunavat, T.R.; Jang, S.C.; Nilsson, L.; Park, H.T.; Repiska, G.; Lasser, C.; Nilsson, J.A.; Gho, Y.S.; Lotvall, J. RNAi delivery by exosome-mimetic nanovesicles-Implications for targeting c-Myc in cancer. Biomaterials 2016, 102, 231–238. [Google Scholar] [CrossRef]
- Jang, S.C.; Kim, O.Y.; Yoon, C.M.; Choi, D.S.; Roh, T.Y.; Park, J.; Nilsson, J.; Lotvall, J.; Kim, Y.K.; Gho, Y.S. Bioinspired Exosome-Mimetic Nanovesicles for Targeted Delivery of Chemotherapeutics to Malignant Tumors. ACS Nano 2013, 7, 7698–7710. [Google Scholar] [CrossRef] [PubMed]
- Ilahibaks, N.F.; Lei, Z.Y.; Mol, E.A.; Deshantri, A.K.; Jiang, L.L.; Schiffelers, R.M.; Vader, P.; Sluijter, J.P.G. Biofabrication of Cell-Derived Nanovesicles: A Potential Alternative to Extracellular Vesicles for Regenerative Medicine. Cells 2019, 8, 1509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Han, C.; Jeong, D.; Kim, B.; Jo, W.; Kang, H.; Cho, S.; Kim, K.H.; Park, J. Mesenchymal Stem Cell Engineered Nanovesicles for Accelerated Skin Wound Closure. ACS Biomater. Sci. Eng. 2019, 5, 1534–1543. [Google Scholar] [CrossRef]
- Garcia-Manrique, P.; Matos, M.; Gutierrez, G.; Pazos, C.; Blanco-Lopez, M.C. Therapeutic biomaterials based on extracellular vesicles: Classification of bio-engineering and mimetic preparation routes. J. Extracell. Vesicles 2018, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, J.Y.; Ji, A.L.; Wang, Z.X.; Qiang, G.H.; Qu, Z.; Wu, J.H.; Jiang, C.P. Exosome-Mimetic Nanovesicles from Hepatocytes promote hepatocyte proliferation in vitro and liver regeneration in vivo. Sci. Rep. 2018, 8. [Google Scholar] [CrossRef] [Green Version]
- Kooijmans, S.A.; Vader, P.; van Dommelen, S.M.; van Solinge, W.W.; Schiffelers, R.M. Exosome mimetics: A novel class of drug delivery systems. Int. J. Nanomed. 2012, 7, 1525–1541. [Google Scholar] [CrossRef] [Green Version]
- Wagner, A.; Vorauer-Uhl, K. Liposome technology for industrial purposes. J. Drug Deliv. 2011, 2011, 591325. [Google Scholar] [CrossRef] [Green Version]
- Lu, M.; Zhao, X.; Xing, H.; Xun, Z.; Zhu, S.; Lang, L.; Yang, T.; Cai, C.; Wang, D.; Ding, P. Comparison of exosome-mimicking liposomes with conventional liposomes for intracellular delivery of siRNA. Int. J. Pharm 2018, 550, 100–113. [Google Scholar] [CrossRef]
- De la Pena, H.; Madrigal, J.A.; Rusakiewicz, S.; Bencsik, M.; Cave, G.W.V.; Selman, A.; Rees, R.C.; Travers, P.J.; Dodi, I.A. Artificial exosomes as tools for basic and clinical immunology. J. Immunol. Methods 2009, 344, 121–132. [Google Scholar] [CrossRef]
- Martinez-Lorenzo, M.J.; Anel, A.; Saez-Gutierrez, B.; Royo-Canas, M.; Bosque, A.; Alava, M.A.; Pineiro, A.; Lasierra, P.; Asin-Ungria, J.; Larrad, L. Rheumatoid synovial fluid T cells are sensitive to APO2L/TRAIL. Clin. Immunol. 2007, 122, 28–40. [Google Scholar] [CrossRef]
- Martinez-Lostao, L.; Garcia-Alvarez, F.; Basanez, G.; Alegre-Aguaron, E.; Desportes, P.; Larrad, L.; Naval, J.; Martinez-Lorenzo, M.J.; Anel, A. Liposome-Bound APO2L/TRAIL Is an Effective Treatment in a Rabbit Model of Rheumatoid Arthritis. Arthritis Rheum. 2010, 62, 2272–2282. [Google Scholar] [CrossRef] [PubMed]
- De Miguel, D.; Basanez, G.; Sanchez, D.; Malo, P.G.; Marzo, I.; Larrad, L.; Naval, J.; Pardo, J.; Anel, A.; Martinez-Lostao, L. Liposomes Decorated with Apo2L/TRAIL Overcome Chemoresistance of Human Hematologic Tumor Cells. Mol. Pharmaceut. 2013, 10, 893–904. [Google Scholar] [CrossRef] [PubMed]
- Imai, T.; Takahashi, Y.; Nishikawa, M.; Kato, K.; Morishita, M.; Yamashita, T.; Matsumoto, A.; Charoenviriyakul, C.; Takakura, Y. Macrophage-dependent clearance of systemically administered B16BL6-derived exosomes from the blood circulation in mice. J. Extracell Vesicles 2015, 4, 26238. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Chu, W.C.; Lai, R.C.; Lim, S.K.; Hui, J.H.P.; Toh, W.S. Exosomes derived from human embryonic mesenchymal stem cells promote osteochondral regeneration. Osteoarthr. Cartil. 2016, 24, 2135–2140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Han, C.; Zhou, J.; Liang, C.; Liu, B.; Pan, X.; Zhang, Y.; Wang, Y.; Yan, B.; Xie, W.; Liu, F.; et al. Human umbilical cord mesenchymal stem cell derived exosomes encapsulated in functional peptide hydrogels promote cardiac repair. Biomater. Sci. 2019, 7, 2920–2933. [Google Scholar] [CrossRef]
- Mardpour, S.; Ghanian, M.H.; Sadeghi-Abandansari, H.; Mardpour, S.; Nazari, A.; Shekari, F.; Baharvand, H. Hydrogel-Mediated Sustained Systemic Delivery of Mesenchymal Stem Cell-Derived Extracellular Vesicles Improves Hepatic Regeneration in Chronic Liver Failure. ACS Appl. Mater. Interfaces 2019, 11, 37421–37433. [Google Scholar] [CrossRef]
- De Witte, T.M.; Fratila-Apachitei, L.E.; Zadpoor, A.A.; Peppas, N.A. Bone tissue engineering via growth factor delivery: From scaffolds to complex matrices. Regen. Biomater. 2018, 5, 197–211. [Google Scholar] [CrossRef] [Green Version]
- Sabir, M.I.; Xu, X.X.; Li, L. A review on biodegradable polymeric materials for bone tissue engineering applications. J. Mater. Sci. 2009, 44, 5713–5724. [Google Scholar] [CrossRef]
- Bose, S.; Roy, M.; Bandyopadhyay, A. Recent advances in bone tissue engineering scaffolds. Trends Biotechnol. 2012, 30, 546–554. [Google Scholar] [CrossRef]
- Brennan, M.Á.; Layrolle, P.; Mooney, D.J. Biomaterials Functionalized with MSC Secreted Extracellular Vesicles and Soluble Factors for Tissue Regeneration. Adv. Funct. Mater. 2020, 1909125. [Google Scholar] [CrossRef]
- Huang, C.C.; Narayanan, R.; Alapati, S.; Ravindran, S. Exosomes as biomimetic tools for stem cell differentiation: Applications in dental pulp tissue regeneration. Biomaterials 2016, 111, 103–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deregibus, M.C.; Figliolini, F.; D’antico, S.; Manzini, P.M.; Pasquino, C.; De Lena, M.; Tetta, C.; Brizzi, M.F.; Camussi, G. Charge-based precipitation of extracellular vesicles. Int. J. Mol. Med. 2016, 38, 1359–1366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- He, X.Z.; Ma, J.Y.; Jabbari, E. Effect of Grafting RGD and BMP-2 Protein-Derived Peptides to a Hydrogel Substrate on Osteogenic Differentiation of Marrow Stromal Cells. Langmuir 2008, 24, 12508–12516. [Google Scholar] [CrossRef] [PubMed]
- Di Luca, A.; Klein-Gunnewiek, M.; Vancso, J.G.; van Blitterswijk, C.A.; Benetti, E.M.; Moroni, L. Covalent Binding of Bone Morphogenetic Protein-2 and Transforming Growth Factor-3 to 3D Plotted Scaffolds for Osteochondral Tissue Regeneration. Biotechnol. J. 2017, 12. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.W.; Wang, L.L.; Zaman, S.; Gordon, J.; Arisi, M.F.; Venkataraman, C.M.; Chung, J.J.; Hung, G.; Gaffey, A.C.; Spruce, L.A.; et al. Sustained release of endothelial progenitor cell-derived extracellular vesicles from shear-thinning hydrogels improves angiogenesis and promotes function after myocardial infarction. Cardiovasc. Res. 2018, 114, 1029–1040. [Google Scholar] [CrossRef] [Green Version]
- Liu, B.H.; Lee, B.W.; Nakanishi, K.; Villasante, A.; Williamson, R.; Metz, J.; Kim, J.; Kanai, M.; Bi, L.; Brown, K.; et al. Cardiac recovery via extended cell-free delivery of extracellular vesicles secreted by cardiomyocytes derived from induced pluripotent stem cells. Nat. Biomed. Eng. 2018, 2, 293–303. [Google Scholar] [CrossRef]
- Xu, N.; Wang, L.L.; Guan, J.J.; Tang, C.; He, N.; Zhang, W.; Fu, S.P. Wound healing effects of a Curcuma zedoaria polysaccharide with platelet-rich plasma exosomes assembled on chitosan/silk hydrogel sponge in a diabetic rat model. Int. J. Biol. Macromol. 2018, 117, 102–107. [Google Scholar] [CrossRef]
- Fang, S.; Xu, C.; Zhang, Y.T.; Xue, C.Y.; Yang, C.; Bi, H.D.; Qian, X.J.; Wu, M.J.; Ji, K.H.; Zhao, Y.P.; et al. Umbilical Cord-Derived Mesenchymal Stem Cell-Derived Exosomal MicroRNAs Suppress Myofibroblast Differentiation by Inhibiting the Transforming Growth Factor-beta/SMAD2 Pathway During Wound Healing. Stem Cell Transl. Med. 2016, 5, 1425–1439. [Google Scholar] [CrossRef]
- Zhang, K.Y.; Zhao, X.N.; Chen, X.N.; Wei, Y.Z.; Du, W.; Wang, Y.B.; Liu, L.A.; Zhao, W.A.; Han, Z.B.; Kong, D.L.; et al. Enhanced Therapeutic Effects of Mesenchymal Stem Cell-Derived Exosomes with an Injectable Hydrogel for Hindlimb Ischemia Treatment. ACS Appl. Mater. Inter. 2018, 10, 30081–30091. [Google Scholar] [CrossRef]
- Qin, Y.H.; Wang, L.; Gao, Z.L.; Chen, G.Y.; Zhang, C.Q. Bone marrow stromal/stem cell-derived extracellular vesicles regulate osteoblast activity and differentiation in vitro and promote bone regeneration in vivo. Sci. Rep. 2016, 6. [Google Scholar] [CrossRef]
- Chen, S.; Tang, Y.M.; Liu, Y.S.; Zhang, P.; Lv, L.W.; Zhang, X.; Jia, L.F.; Zhou, Y.S. Exosomes derived from miR-375-overexpressing human adipose mesenchymal stem cells promote bone regeneration. Cell Proliferat. 2019, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gangadaran, P.; Rajendran, R.L.; Lee, H.W.; Kalimuthu, S.; Hong, C.M.; Jeong, S.Y.; Lee, S.W.; Lee, J.; Ahn, B.C. Extracellular vesicles from mesenchymal stem cells activates VEGF receptors and accelerates recovery of hindlimb ischemia. J. Control. Release 2017, 264, 112–126. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.J.; Zhang, K.Y.; Zhao, X.N.; Kong, D.L.; Zhao, Q.; Liu, N.; Ma, F.X. Enhanced Therapeutic Effects of MSC-derived Exosomes with an Injectable Hydrogel for Hindlimb Ischemia Treatment. Circ. Res. 2018, 123. [Google Scholar] [CrossRef]
- Zhang, Y.T.; Hao, Z.C.; Wang, P.F.; Xia, Y.; Wu, J.H.; Xia, D.M.; Fang, S.; Xu, S.G. Exosomes from human umbilical cord mesenchymal stem cells enhance fracture healing through HIF-1 alpha-mediated promotion of angiogenesis in a rat model of stabilized fracture. Cell Proliferat. 2019, 52. [Google Scholar] [CrossRef] [Green Version]
- Liu, X.L.; Yang, Y.L.; Li, Y.; Niu, X.; Zhao, B.Z.; Wang, Y.; Bao, C.Y.; Xie, Z.P.; Lin, Q.N.; Zhu, L.Y. Integration of stem cell-derived exosomes with in situ hydrogel glue as a promising tissue patch for articular cartilage regeneration. Nanoscale 2017, 9, 4430–4438. [Google Scholar] [CrossRef]
- Nikravesh, N.; Davies, O.G.; Azoidis, I.; Moakes, R.J.A.; Marani, L.; Turner, M.; Kearney, C.J.; Eisenstein, N.M.; Grover, L.M.; Cox, S.C. Physical Structuring of Injectable Polymeric Systems to Controllably Deliver Nanosized Extracellular Vesicles. Adv. Healthc. Mater. 2019, 8. [Google Scholar] [CrossRef] [Green Version]
- Fuhrmann, G.; Herrmann, I.K.; Stevens, M.M. Cell-derived vesicles for drug therapy and diagnostics: Opportunities and challenges. Nano Today 2015, 10, 397–409. [Google Scholar] [CrossRef] [Green Version]
- Adjei, I.M.; Blanka, S. Modulation of the tumor microenvironment for cancer treatment: A biomaterials approach. J. Funct. Biomater. 2015, 6, 81–103. [Google Scholar] [CrossRef] [Green Version]
- Thippabhotla, S.; Zhong, C.C.; He, M. 3D cell culture stimulates the secretion of in vivo like extracellular vesicles. Sci. Rep. 2019, 9. [Google Scholar] [CrossRef] [Green Version]
- Yan, I.K.; Shukla, N.; Borrelli, D.A.; Patel, T. Use of a Hollow Fiber Bioreactor to Collect Extracellular Vesicles from Cells in Culture. Methods Mol. Biol. 2018, 1740, 35–41. [Google Scholar] [CrossRef]
- Colao, I.L.; Corteling, R.; Bracewell, D.; Wall, I. Manufacturing Exosomes: A Promising Therapeutic Platform. Trends Mol. Med. 2018, 24, 242–256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pachler, K.; Lener, T.; Streif, D.; Dunai, Z.A.; Desgeorges, A.; Feichtner, M.; Oller, M.; Schallmoser, K.; Rohde, E.; Gimona, M. A Good Manufacturing Practice-grade standard protocol for exclusively human mesenchymal stromal cell-derived extracellular vesicles. Cytotherapy 2017, 19, 458–472. [Google Scholar] [CrossRef] [Green Version]
- Mendt, M.; Kamerkar, S.; Sugimoto, H.; McAndrews, K.M.; Wu, C.C.; Gagea, M.; Yang, S.J.; Blanko, E.V.R.; Peng, Q.; Ma, X.Y.; et al. Generation and testing of clinical-grade exosomes for pancreatic cancer. JCI Insight 2018, 3, e99263. [Google Scholar] [CrossRef]
Engineering Method | Advantages | Disadvantages | Ref. |
---|---|---|---|
Genetic-manipulation | Allows the enrichment of a certain therapeutic molecule of interest | Cumbersome and time-consuming | [31,32] |
High-associated costs | |||
Issue regarding transduction and EV loading efficiency | |||
Exogenous stimulation | Avoids the issues associated with genetic reprogramming | Long compound exposure/concentration may affect cell viability | [45,46,47] |
EV loading efficiency | |||
Fusogenic liposomes | Delivery of both hydrophobic and hydrophilic cargo into the cell | Issues with loading large cargo | [50,51] |
Difficult to control proportion packaged into EVs | |||
External stimulation | Enhanced EVs yield/therapeutic potency | May alter composition of EVs produced | [55,56,57] |
3D culture platforms | Enhanced EVs yield/therapeutic potency | Possible difficulty in extracting EVs | [18,61,64] |
Bioreactors | Scalability/reproducibility of EV manufacture | Issues with the use of primary cells | [67,68,69] |
May alter composition of EVs |
Technique | Advantages | Disadvantages | Ref. |
---|---|---|---|
Co-incubation | Simple method Membrane not compromised Inexpensive | Low loading efficacy | [71] |
Require hydrophobic cargo | |||
Electroporation | Allows loading of large molecules (siRNA, miRNA) | Disrupts EV integrity | [74] |
RNA aggregation | |||
Sonication | Higher loading efficiency | Possible membrane deformation | [78] |
Low-efficiency loading of hydrophobic cargo | |||
Extrusion | High loading efficiency | Possible membrane deformation | [79] |
Simple method | |||
Freeze/thaw | Medium loading efficiency | Aggregation of EVs | [83] |
Reduced loading efficacy | |||
Saponin-assisted loading | High loading efficiency | Possible membrane deformation | [80] |
Possible in vivo toxicity |
Technique | Advantages | Disadvantages | Ref. |
---|---|---|---|
Hydrophobic insertion | Highly effective for the addition of lipophilic molecules | Dependent upon hydrophobic/amphiphilic properties | [87,88] |
Simple co-incubation | |||
Synthetic lipid nanoparticle fusion | Facilitate the transfer of membranes proteins | Risk of cargo leakage during fusion | [89,90] |
Allow more complex membrane modifications | More complex approach with the development of synthetic nanoparticles | ||
Covalent modifications | Rapid and simple | Possible alteration of the active site of surface proteins | [91,92] |
Good stability due to the strong covalent bond | |||
Easily scalable | |||
Non-covalent modifications | High loading efficiency | Weaker bond strength compared to covalent link (less stable) | [95,96] |
Simple method | Potential membrane disruption when using cationic molecules |
Clinical Application | EV Source | Biomaterial | Study Observations | Ref. |
---|---|---|---|---|
Cardiovascular | Endothelial progenitor cells | Adamantane and β-cyclodextrin-modified hyaluronic acid hydrogel | Enhanced myocardium regeneration | [126] |
Cardiomyocyte-derived iPSCs | Collagen type I Gelfoam sponge | Increased cardiac recovery in a rat myocardial infarction model | [127] | |
Skin | Blood plasma | Chitosan/silk fibroin sponge | Enhanced wound healing in a diabetic rat model | [128] |
Umbilical cord-derived MSCs | HydroMatrix hydrogel | Reduced myofibroblast accumulation/scar formation | [129] | |
Angiogenic | Human placental MSCs | Chitosan hydrogel | Improved EV retention and capillary formation in hindlimb ischemic model | [130] |
Bone | BMSCs | HyStem-HP hydrogel | Increased bone healing of critical-sized calvaria defects | [131] |
MSCs | Thiol-modified hyaluronan | Accelerated regeneration of rat calvaria defect | [132] | |
Muscular | MSCs | Matrigel | Enhanced angiogenesis and muscle recovery in an ischemia model | [133] |
MSCs | Silk fibroin hydrogel | Prevented ischemia-induced vascular dysfunction | [116] |
© 2020 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Man, K.; Brunet, M.Y.; Jones, M.-C.; Cox, S.C. Engineered Extracellular Vesicles: Tailored-Made Nanomaterials for Medical Applications. Nanomaterials 2020, 10, 1838. https://doi.org/10.3390/nano10091838
Man K, Brunet MY, Jones M-C, Cox SC. Engineered Extracellular Vesicles: Tailored-Made Nanomaterials for Medical Applications. Nanomaterials. 2020; 10(9):1838. https://doi.org/10.3390/nano10091838
Chicago/Turabian StyleMan, Kenny, Mathieu Y. Brunet, Marie-Christine Jones, and Sophie C. Cox. 2020. "Engineered Extracellular Vesicles: Tailored-Made Nanomaterials for Medical Applications" Nanomaterials 10, no. 9: 1838. https://doi.org/10.3390/nano10091838
APA StyleMan, K., Brunet, M. Y., Jones, M. -C., & Cox, S. C. (2020). Engineered Extracellular Vesicles: Tailored-Made Nanomaterials for Medical Applications. Nanomaterials, 10(9), 1838. https://doi.org/10.3390/nano10091838