261
Views
55
CrossRef citations to date
0
Altmetric
Expert Opinion

Extracellular Vesicles As Nanomedicine: Hopes And Hurdles In Clinical Translation

ORCID Icon, &
Pages 8847-8859 | Published online: 12 Nov 2019

References

  • Yanez-Mo M, Siljander PR, Andreu Z, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015;4:27066. doi:10.3402/jev.v4.2706625979354
  • van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19(4):213–228. doi:10.1038/nrm.2017.12529339798
  • Agrahari V, Agrahari V, Burnouf PA, Chew CH, Burnouf T. Extracellular microvesicles as new industrial therapeutic frontiers. Trends Biotechnol. 2019;37:707–729. doi:10.1016/j.tibtech.2018.11.01230638682
  • Maas SLN, Breakefield XO, Weaver AM. Extracellular vesicles: unique intercellular delivery vehicles. Trends Cell Biol. 2017;27(3):172–188. doi:10.1016/j.tcb.2016.11.00327979573
  • de Jong OG, Kooijmans SAA, Murphy DE, et al. Drug delivery with extracellular vesicles: from imagination to innovation. Acc Chem Res. 2019;52:1761–1770. doi:10.1021/acs.accounts.9b0010931181910
  • Vader P, Mol EA, Pasterkamp G, Schiffelers RM. Extracellular vesicles for drug delivery. Adv Drug Deliv Rev. 2016;106(Pt A):148–156. doi:10.1016/j.addr.2016.02.00626928656
  • Gudbergsson JM, Jonsson K, Simonsen JB, Johnsen KB. Systematic review of targeted extracellular vesicles for drug delivery - considerations on methodological and biological heterogeneity. J Control Release. 2019;306:108–120. doi:10.1016/j.jconrel.2019.06.00631175896
  • Lener T, Gimona M, Aigner L, et al. Applying extracellular vesicles based therapeutics in clinical trials - an ISEV position paper. J Extracell Vesicles. 2015;4:30087. doi:10.3402/jev.v4.3008726725829
  • Pachler K, Lener T, Streif D, et al. A good manufacturing practice–grade standard protocol for exclusively human mesenchymal stromal cell–derived extracellular vesicles. Cytotherapy. 2017;19(4):458–472. doi:10.1016/j.jcyt.2017.01.00128188071
  • Keshtkar S, Azarpira N, Ghahremani MH. Mesenchymal stem cell-derived extracellular vesicles: novel frontiers in regenerative medicine. Stem Cell Res Ther. 2018;9(1):63. doi:10.1186/s13287-018-0791-729523213
  • Burnouf T, Goubran HA, Chou ML, Devos D, Radosevic M. Platelet microparticles: detection and assessment of their paradoxical functional roles in disease and regenerative medicine. Blood Rev. 2014;28(4):155–166.24826991
  • Tao SC, Guo SC, Zhang CQ. Platelet-derived extracellular vesicles: an emerging therapeutic approach. Int J Biol Sci. 2017;13(7):828–834. doi:10.7150/ijbs.1977628808416
  • Burnouf T, Burnouf PA, Wu YW, Chuang EY, Lu LS, Goubran H. Circulatory-cell-mediated nanotherapeutic approaches in disease targeting. Drug Discov Today. 2018;23(5):934–943. doi:10.1016/j.drudis.2017.08.01228917821
  • Palviainen M, Saari H, Karkkainen O, et al. Metabolic signature of extracellular vesicles depends on the cell culture conditions. J Extracell Vesicles. 2019;8(1):1596669. doi:10.1080/20013078.2018.156080831007875
  • 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(6). doi:10.3390/ijms18061190
  • Pascucci L, Coccè V, Bonomi A, 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. doi:10.1016/j.jconrel.2014.07.04225084218
  • Tian Y, Li S, Song J, et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials. 2014;35(7):2383–2390. doi:10.1016/j.biomaterials.2013.11.08324345736
  • Yang T, Martin P, Fogarty B, et al. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio Rerio. Pharm Res. 2015;32(6):2003–2014. doi:10.1007/s11095-014-1593-y25609010
  • Liao W, Du Y, Zhang C, et al. Exosomes: the next generation of endogenous nanomaterials for advanced drug delivery and therapy. Acta Biomater. 2019;86:1–14. doi:10.1016/j.actbio.2018.12.04530597259
  • Kim MS, Haney MJ, Zhao Y, et al. Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine. 2016;12(3):655–664. doi:10.1016/j.nano.2015.10.01226586551
  • Sato YT, Umezaki K, Sawada S, et al. Engineering hybrid exosomes by membrane fusion with liposomes. Sci Rep. 2016;6:21933. doi:10.1038/srep2193326911358
  • Kim SM, Kim HS. Engineering of extracellular vesicles as drug delivery vehicles. Stem Cell Investig. 2017;4:74. doi:10.21037/sci.2017.08.07
  • Tang TT, Lv LL, Lan HY, Liu BC. Extracellular vesicles: opportunities and challenges for the treatment of renal diseases. Front Physiol. 2019;10:226. doi:10.3389/fphys.2019.0022630941051
  • Paganini C, Palmiero UC, Pocsfalvi G, Touzet N, Bongiovanni A, Arosio P. Scalable production and isolation of extracellular vesicles: available sources and lessons from current industrial bioprocesses. Biotechnol J. 2019;e1800528. doi:10.1002/biot.20180052831140717
  • Konoshenko MY, Lekchnov EA, Vlassov AV, Laktionov PP. Isolation of extracellular vesicles: general methodologies and latest trends. Biomed Res Int. 2018;2018:8545347. doi:10.1155/2018/854534729662902
  • Busatto S, Vilanilam G, Ticer T, et al. Tangential flow filtration for highly efficient concentration of extracellular vesicles from large volumes of fluid. Cells. 2018;7(12):273. doi:10.3390/cells7120273
  • Watson DC, Yung BC, Bergamaschi C, et al. Scalable, cGMP-compatible purification of extracellular vesicles carrying bioactive human heterodimeric IL-15/lactadherin complexes. J Extracell Vesicles. 2018;7(1):1442088. doi:10.1080/20013078.2018.144208829535850
  • Boing AN, van der Pol E, Grootemaat AE, Coumans FA, Sturk A, Nieuwland R. Single-step isolation of extracellular vesicles by size-exclusion chromatography. J Extracell Vesicles. 2014;3. doi:10.3402/jev.v3.24384
  • Ramirez MI, Amorim MG, Gadelha C, et al. Technical challenges of working with extracellular vesicles. Nanoscale. 2018;10(3):881–906. doi:10.1039/c7nr08360b29265147
  • Konoshenko MY, Lekchnov EA, Vlassov AV, Laktionov PP. Isolation of extracellular vesicles: general methodologies and latest trends. Biomed Res Int. 2018;2018:8545347. doi:10.1155/2018/854534729662902
  • Liang K, Liu F, Fan J, et al. Nanoplasmonic quantification of tumor-derived extracellular vesicles in plasma microsamples for diagnosis and treatment monitoring. Nat Biomed Eng. 2017;1. doi:10.1038/s41551-016-0021
  • Sunkara V, Woo HK, Cho YK. Emerging techniques in the isolation and characterization of extracellular vesicles and their roles in cancer diagnostics and prognostics. Analyst. 2016;141(2):371–381. doi:10.1039/c5an01775k26535415
  • Hartjes TA, Mytnyk S, Jenster GW, van Steijn V, van Royen ME. Extracellular vesicle quantification and characterization: common methods and emerging approaches. Bioengineering. 2019;6(1):7. doi:10.3390/bioengineering6020029
  • Rupert DLM, Claudio V, Lässer C, Bally M. Methods for the physical characterization and quantification of extracellular vesicles in biological samples. Biochim Biophys Acta. 2017;1861(1, Part A):3164–3179. doi:10.1016/j.bbagen.2016.07.028
  • Szatanek R, Baj-Krzyworzeka M, Zimoch J, Lekka M, Siedlar M, Baran J. The methods of choice for extracellular vesicles (EVs) characterization. Int J Mol Sci. 2017;18(6):1153. doi:10.3390/ijms18061153
  • Klutz S, Magnus J, Lobedann M, et al. Developing the biofacility of the future based on continuous processing and single-use technology. J Biotechnol. 2015;213:120–130. doi:10.1016/j.jbiotec.2015.06.38826091773
  • Rohde E, Pachler K, Gimona M. Manufacturing and characterization of extracellular vesicles from umbilical cord-derived mesenchymal stromal cells for clinical testing. Cytotherapy. 2019;21:581–592. doi:10.1016/j.jcyt.2018.12.00630979664
  • Cai K, Anderson J, Orchard JD, Afdahl CD, Dickson M, Li Y. Virus removal robustness of ion exchange chromatography. Biologicals. 2019;58:28–34. doi:10.1016/j.biologicals.2019.01.00430661901
  • Burnouf T, Radosevich M. Reducing the risk of infection from plasma products: specific preventative strategies. Blood Rev. 2000;14(2):94–110. doi:10.1054/blre.2000.012911012252
  • Chou ML, Lin LT, Devos D, Burnouf T. Nanofiltration to remove microparticles and decrease the thrombogenicity of plasma: in vitro feasibility assessment. Transfusion. 2015;55(10):2433–2444. doi:10.1111/trf.1316225988671
  • Burnouf T, Strunk D, Koh MB, Schallmoser K. Human platelet lysate: replacing fetal bovine serum as a gold standard for human cell propagation? Biomaterials. 2016;76:371–387. doi:10.1016/j.biomaterials.2015.10.06526561934
  • Henschler R, Gabriel C, Schallmoser K, Burnouf T, Koh MBC. Human platelet lysate current standards and future developments. Transfusion. 2019;59(4):1407–1413. doi:10.1111/trf.1517430741431
  • Schallmoser K, Henschler R, Gabriel C, Koh MBC, Burnouf T. Production and quality requirements of human platelet lysate: a position statement from the working party on cellular therapies of the international society of blood transfusion. Trends Biotechnol. 2019. doi:10.1016/j.tibtech.2019.06.002
  • Aranha H. Virus safety of biopharmaceuticals - absence of evidence is not evidence of absence. Contract Pharma. 2011;13:82–87.
  • Moody M, Alvez W, Varghese J, Khan F. Mouse Minute Virus (MMV) contamination—a case study: detection, root cause determination, and corrective actions. PDA J Pharm Sci Technol. 2011;65(6):580–588. doi:10.5731/pdajpst.2011.0082422294580
  • Kerr A, Nims R. Adventitious viruses detected in biopharmaceutical bulk harvest samples over a 10 year period. PDA J Pharm Sci Technol. 2010;64(5):481–485.21502056
  • Roush JD. Integrated viral clearance strategies — reflecting on the present, projecting to the future. Curr Opin Biotechnol. 2018;53:137–143. doi:10.1016/j.copbio.2018.01.00329367164
  • Carbrello C, Nhiem D, Priest M, Mann K, Greenhalgh P. Supplement: upstream virus safety: protect your bioreactor by media filtration. Genet Eng Biotechnol News. 2017;37(17).
  • Mann K, Royce J, Carbrello C, et al. Protection of bioreactor culture from virus contamination by use of a virus barrier filter. BMC Proc. 2015;9(9):22. doi:10.1186/1753-6561-9-S9-P22
  • Wiklander OP, Nordin JZ, O’Loughlin A, et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J Extracell Vesicles. 2015;4:26316. doi:10.3402/jev.v4.2631625899407
  • Lai CP, Mardini O, Ericsson M, et al. Dynamic biodistribution of extracellular vesicles in vivo using a multimodal imaging reporter. ACS Nano. 2014;8(1):483–494. doi:10.1021/nn404945r24383518
  • Takahashi Y, Nishikawa M, Shinotsuka H, et al. Visualization and in vivo tracking of the exosomes of murine melanoma B16-BL6 cells in mice after intravenous injection. J Biotechnol. 2013;165(2):77–84. doi:10.1016/j.jbiotec.2013.03.01323562828
  • Matsumoto A, Takahashi Y, Nishikawa M, et al. Role of phosphatidylserine-derived negative surface charges in the recognition and uptake of intravenously injected B16BL6-derived exosomes by macrophages. J Pharm Sci. 2017;106(1):168–175. doi:10.1016/j.xphs.2016.07.02227649887
  • Imai T, Takahashi Y, Nishikawa M, et al. Macrophage-dependent clearance of systemically administered B16BL6-derived exosomes from the blood circulation in mice. J Extracell Vesicles. 2015;4:26238. doi:10.3402/jev.v4.2623825669322
  • Matsumoto J, Stewart T, Banks WA, Zhang J. The transport mechanism of extracellular vesicles at the blood-brain barrier. Curr Pharm Des. 2017;23(40):6206–6214. doi:10.2174/138161282366617091316473828914201
  • Pinheiro A, Silva AM, Teixeira JH, et al. Extracellular vesicles: intelligent delivery strategies for therapeutic applications. J Control Release. 2018;289:56–69. doi:10.1016/j.jconrel.2018.09.01930261205
  • Munagala R, Aqil F, Jeyabalan J, Gupta RC. Bovine milk-derived exosomes for drug delivery. Cancer Lett. 2016;371(1):48–61. doi:10.1016/j.canlet.2015.10.02026604130
  • Pieters BC, Arntz OJ, Bennink MB, et al. Commercial cow milk contains physically stable extracellular vesicles expressing immunoregulatory TGF-beta. PLoS One. 2015;10(3):e0121123. doi:10.1371/journal.pone.012112325822997
  • Manca S, Giraud D, Zempleni J. Bioavailability and biodistribution of fluorophore-labeled exosomes from cow’s milk after intravenous and oral administration in C57Bl/6J mice. Faseb J. 2016;30(1_supplement):690.698.
  • Narbute K, Pilipenko V, Pupure J, et al. Intranasal administration of extracellular vesicles derived from human teeth stem cells improves motor symptoms and normalizes tyrosine hydroxylase expression in the substantia nigra and striatum of the 6-hydroxydopamine-treated rats. Stem Cells Transl Med. 2019;8(5):490–499. doi:10.1002/sctm.18-016230706999
  • Zhuang X, Xiang X, Grizzle W, 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(10):1769–1779. doi:10.1038/mt.2011.16421915101
  • Haney MJ, Klyachko NL, Zhao Y, et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J Control Release. 2015;207:18–30. doi:10.1016/j.jconrel.2015.03.03325836593
  • Betzer O, Perets N, Angel A, et al. In vivo neuroimaging of exosomes using gold nanoparticles. ACS Nano. 2017;11(11):10883–10893. doi:10.1021/acsnano.7b0449528960957
  • Ingato D, Lee JU, Sim SJ, Kwon YJ. Good things come in small packages: overcoming challenges to harness extracellular vesicles for therapeutic delivery. J Control Release. 2016;241:174–185. doi:10.1016/j.jconrel.2016.09.01627667180
  • Jeyaram A, Jay SM. Preservation and storage stability of extracellular vesicles for therapeutic applications. Aaps J. 2017;20(1):1. doi:10.1208/s12248-017-0160-y29181730
  • Charoenviriyakul C, Takahashi Y, Nishikawa M, Takakura Y. Preservation of exosomes at room temperature using lyophilization. Int J Pharm. 2018;553(1–2):1–7. doi:10.1016/j.ijpharm.2018.10.03230316791
  • Park SJ, Jeon H, Yoo SM, Lee MS. The effect of storage temperature on the biological activity of extracellular vesicles for the complement system. In Vitro Cell Dev Biol Anim. 2018;54(6):423–429. doi:10.1007/s11626-018-0261-729748909
  • Richter M, Fuhrmann K, Fuhrmann G. Evaluation of the storage stability of extracellular vesicles. J Vis Exp. 2019;(147). doi:10.3791/59584
  • Kusuma GD, Barabadi M, Tan JL, Morton DAV, Frith JE, Lim R. To protect and to preserve: novel preservation strategies for extracellular vesicles. Front Pharmacol. 2018;9(1199). doi:10.3389/fphar.2018.01199
  • Valkonen S, van der Pol E, Boing A, et al. Biological reference materials for extracellular vesicle studies. Eur J Pharm Sci. 2017;98:4–16. doi:10.1016/j.ejps.2016.09.00827622921
  • Witwer KW, Buzas EI, Bemis LT, et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J Extracell Vesicles. 2013;2:20360.
  • Lorincz AM, Timar CI, Marosvari KA, et al. Effect of storage on physical and functional properties of extracellular vesicles derived from neutrophilic granulocytes. J Extracell Vesicles. 2014;3:25465. doi:10.3402/jev.v3.2438425536933
  • Jin Y, Chen K, Wang Z, et al. DNA in serum extracellular vesicles is stable under different storage conditions. BMC Cancer. 2016;16(1):753. doi:10.1186/s12885-016-2783-227662833
  • Bosch S, de Beaurepaire L, Allard M, et al. Trehalose prevents aggregation of exosomes and cryodamage. Sci Rep. 2016;6:36162. doi:10.1038/srep3616227824088
  • Frank J, Richter M, de Rossi C, Lehr CM, Fuhrmann K, Fuhrmann G. Extracellular vesicles protect glucuronidase model enzymes during freeze-drying. Sci Rep. 2018;8(1):12377. doi:10.1038/s41598-018-30786-y30120298
  • Xu R, Rai A, Chen M, Suwakulsiri W, Greening DW, Simpson RJ. Extracellular vesicles in cancer - implications for future improvements in cancer care. Nat Rev Clin Oncol. 2018;15(10):617–638. doi:10.1038/s41571-018-0036-929795272
  • Hinde E, Thammasiraphop K, Duong HT, et al. Pair correlation microscopy reveals the role of nanoparticle shape in intracellular transport and site of drug release. Nat Nanotechnol. 2017;12(1):81. doi:10.1038/nnano.2016.16027618255
  • Erkoc P, Yasa IC, Ceylan H, Yasa O, Alapan Y, Sitti M. Mobile microrobots for active therapeutic delivery. Adv Ther. 2019;2(1):1800064. doi:10.1002/adtp.v2.1
  • Brambilla D, Luciani P, Leroux JC. Breakthrough discoveries in drug delivery technologies: the next 30 years. J Control Release. 2014;190:9–14. doi:10.1016/j.jconrel.2014.03.05624794899
  • Wilhelm S, Tavares AJ, Dai Q, et al. Analysis of nanoparticle delivery to tumours. Nat Rev Mater. 2016;1(5):16014. doi:10.1038/natrevmats.2016.14
  • Kibria G, Ramos EK, Wan Y, Gius DR, Liu H. Exosomes as a drug delivery system in cancer therapy: potential and challenges. Mol Pharm. 2018;15(9):3625–3633. doi:10.1021/acs.molpharmaceut.8b0027729771531
  • Armstrong JPK, Stevens MM. Strategic design of extracellular vesicle drug delivery systems. Adv Drug Deliv Rev. 2018;130:12–16. doi:10.1016/j.addr.2018.06.01729959959
  • Kamerkar S, LeBleu VS, Sugimoto H, et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature. 2017;546(7659):498–503. doi:10.1038/nature2234128607485
  • You Y, Ikezu T. Emerging roles of extracellular vesicles in neurodegenerative disorders. Neurobiol Dis. 2019;130:104512. doi:10.1016/j.nbd.2019.10451231229685
  • Hall J, Prabhakar S, Balaj L, Lai CP, Cerione RA, Breakefield XO. Delivery of therapeutic proteins via extracellular vesicles: review and potential treatments for Parkinson’s disease, Glioma, and Schwannoma. Cell Mol Neurobiol. 2016;36(3):417–427. doi:10.1007/s10571-015-0309-027017608
  • Budnik V, Ruiz-Cañada C, Wendler F. Extracellular vesicles round off communication in the nervous system. Nat Rev Neurosci. 2016;17(3):160–172. doi:10.1038/nrn.2015.2926891626
  • Rufino-Ramos D, Albuquerque PR, Carmona V, Perfeito R, Nobre RJ, Pereira de Almeida L. Extracellular vesicles: novel promising delivery systems for therapy of brain diseases. J Control Release. 2017;262:247–258. doi:10.1016/j.jconrel.2017.07.00128687495
  • 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(4):287–296. doi:10.1016/j.apsb.2016.02.00127471669
  • Qu M, Lin Q, Huang L, et al. Dopamine-loaded blood exosomes targeted to brain for better treatment of Parkinson’s disease. J Control Release. 2018;287:156–166. doi:10.1016/j.jconrel.2018.08.03530165139
  • Tian T, Zhang H-X, He C-P, et al. Surface functionalized exosomes as targeted drug delivery vehicles for cerebral ischemia therapy. Biomaterials. 2018;150:137–149. doi:10.1016/j.biomaterials.2017.10.01229040874
  • Vinaiphat A, Sze SK. Clinical implications of extracellular vesicles in neurodegenerative diseases. Expert Rev Mol Diagn. 2019;1–12.
  • Rayyan M, Zheutlin A, Byrd JB. Clinical research using extracellular vesicles: insights from the International Society for Extracellular Vesicles 2018 annual meeting. J Extracell Vesicles. 2018;7(1):1535744. doi:10.1080/20013078.2018.153574431162489
  • Besse B, Charrier M, Lapierre V, et al. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology. 2016;5(4):e1071008. doi:10.1080/2162402X.2015.107100827141373
  • Shukla AA, Thommes J. Recent advances in large-scale production of monoclonal antibodies and related proteins. Trends Biotechnol. 2010;28(5):253–261. doi:10.1016/j.tibtech.2010.02.00120304511
  • Armstrong JPK, Holme MN, Stevens MM. Re-engineering extracellular vesicles as smart nanoscale therapeutics. ACS Nano. 2017;11(1):69–83. doi:10.1021/acsnano.6b0760728068069
  • Koh E, Lee EJ, Nam GH, et al. Exosome-SIRPalpha, a CD47 blockade increases cancer cell phagocytosis. Biomaterials. 2017;121:121–129. doi:10.1016/j.biomaterials.2017.01.00428086180
  • Riazifar M, Pone EJ, Lotvall J, Zhao W. Stem Cell Extracellular Vesicles: extended Messages of Regeneration. Annu Rev Pharmacol Toxicol. 2017;57:125–154. doi:10.1146/annurev-pharmtox-061616-03014627814025
  • Akyurekli C, Le Y, Richardson RB, Fergusson D, Tay J, Allan DS. A systematic review of preclinical studies on the therapeutic potential of mesenchymal stromal cell-derived microvesicles. Stem Cell Rev. 2015;11(1):150–160. doi:10.1007/s12015-014-9545-9
  • Agrahari V, Burnouf PA, Burnouf T, Agrahari V. Nanoformulation properties, characterization, and behavior in complex biological matrices: Challenges and opportunities for brain-targeted drug delivery applications and enhanced translational potential. Adv Drug Deliv Rev. doi:10.1016/j.addr.2019.02.008 Epub 2019 Feb 22.
  • Agrahari V, Hiremath P. Challenges associated and approaches for successful translation of nanomedicines into commercial products. Nanomedicine (Lond). 2017;12(8):819–823. doi:10.2217/nnm-2017-0039