445
Views
26
CrossRef citations to date
0
Altmetric
Review

Extracellular Vesicles – Advanced Nanocarriers in Cancer Therapy: Progress and Achievements

, , , , , & show all
Pages 6485-6502 | Published online: 26 Aug 2020

References

  • Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424. doi:10.3322/caac.2149230207593
  • Maeda H, Khatami M. Analyses of repeated failures in cancer therapy for solid tumors: poor tumor-selective drug delivery, low therapeutic efficacy and unsustainable costs. Clin Transl Med. 2018;7(1):11. doi:10.1186/s40169-018-0185-629541939
  • Magdy T, Burmeister BT, Burridge PW. Validating the pharmacogenomics of chemotherapy-induced cardiotoxicity: what is missing? Pharmacol Ther. 2016;168:113–125. doi:10.1016/j.pharmthera.2016.09.00927609196
  • Marchetti C, De Felice F, Di Pinto A, et al. Dose-dense weekly chemotherapy in advanced ovarian cancer: an updated meta-analysis of randomized controlled trials. Crit Rev Oncol Hematol. 2018;125:30–34. doi:10.1016/j.critrevonc.2018.02.01629650273
  • Tang J, Zhu N, Rao S, Carlson KS. Stem cell damage after chemotherapy- can we do better? Best Pract Res Clin Haematol. 2019;32(1):31–39. doi:10.1016/j.beha.2019.02.00130927973
  • Davoodi P, Lee LY, Xu Q, et al. Drug delivery systems for programmed and on-demand release. Adv Drug Deliv Rev. 2018;132:104–138. doi:10.1016/j.addr.2018.07.00230415656
  • Nikravan G, Haddadi-Asl V, Salami-Kalajahi M. Synthesis of dual temperature - and pH-responsive yolk-shell nanoparticles by conventional etching and new deswelling approaches: DOX release behavior. Colloids Surf B Biointerfaces. 2018;165:1–8. doi:10.1016/j.colsurfb.2018.02.01029448215
  • La-Beck NM, Liu X, Wood LM. Harnessing liposome interactions with the immune system for the next breakthrough in cancer drug delivery. Front Pharmacol. 2019;10:220. doi:10.3389/fphar.2019.0022030914953
  • Das M, Huang L. Liposomal nanostructures for drug delivery in gastrointestinal cancers. J Pharmacol Exp Ther. 2019;370(3):647–656. doi:10.1124/jpet.118.25479730541917
  • Li G, Lei Q, Wang F, et al. Fluorinated polymer mediated transmucosal peptide delivery for intravesical instillation therapy of bladder cancer. Small. 2019;15(25):e1900936. doi:10.1002/smll.20190093631074941
  • Wang H, Agarwal P, Zhao G, et al. Overcoming ovarian cancer drug resistance with a cold responsive nanomaterial. ACS Cent Sci. 2018;4(5):567–581. doi:10.1021/acscentsci.8b0005029806003
  • Kalyane D, Raval N, Maheshwari R, Tambe V, Kalia K, Tekade RK. Employment of enhanced permeability and retention effect (EPR): nanoparticle-based precision tools for targeting of therapeutic and diagnostic agent in cancer. Mater Sci Eng C Mater Biol Appl. 2019;98:1252–1276. doi:10.1016/j.msec.2019.01.06630813007
  • Brun NR, Lenz M, Wehrli B, Fent K. Comparative effects of zinc oxide nanoparticles and dissolved zinc on zebrafish embryos and eleuthero-embryos: importance of zinc ions. Sci Total Environ. 2014;476–477:657–666. doi:10.1016/j.scitotenv.2014.01.053
  • Pullan JE, Confeld MI, Osborn JK, Kim J, Sarkar K, Mallik S. Exosomes as drug carriers for cancer therapy. Mol Pharm. 2019;16(5):1789–1798. doi:10.1021/acs.molpharmaceut.9b0010430951627
  • Batrakova EV, Kim MS. Using exosomes, naturally-equipped nanocarriers, for drug delivery. J Control Release. 2015;219:396–405. doi:10.1016/j.jconrel.2015.07.03026241750
  • Lotvall J, Hill AF, Hochberg F, et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles. J Extracell Vesicles. 2014;3:26913. doi:10.3402/jev.v3.2691325536934
  • Thery C, Witwer KW, Aikawa E, 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(1):1535750. doi:10.1080/20013078.2018.153575030637094
  • Harding C, Heuser J, Stahl P. Endocytosis and intracellular processing of transferrin and colloidal gold-transferrin in rat reticulocytes: demonstration of a pathway for receptor shedding. Eur J Cell Biol. 1984;35(2):256–263.6151502
  • 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
  • Minciacchi VR, Freeman MR, Di Vizio D. Extracellular vesicles in cancer: exosomes, microvesicles and the emerging role of large oncosomes. Semin Cell Dev Biol. 2015;40:41–51. doi:10.1016/j.semcdb.2015.02.01025721812
  • Bordeleau F, Chan B, Antonyak MA, Lampi MC, Cerione RA, Reinhart-King CA. Microvesicles released from tumor cells disrupt epithelial cell morphology and contractility. J Biomech. 2016;49(8):1272–1279. doi:10.1016/j.jbiomech.2015.10.00326477404
  • Morello M, Minciacchi VR, de Candia P, et al. Large oncosomes mediate intercellular transfer of functional microRNA. Cell Cycle. 2013;12(22):3526–3536. doi:10.4161/cc.2653924091630
  • Xu X, Lai Y, Hua ZC. Apoptosis and apoptotic body: disease message and therapeutic target potentials. Biosci Rep. 2019;39(1). doi:10.1042/BSR20180992
  • Kowal J, Tkach M, Thery C. Biogenesis and secretion of exosomes. Curr Opin Cell Biol. 2014;29:116–125. doi:10.1016/j.ceb.2014.05.00424959705
  • Choi D, Lee TH, Spinelli C, Chennakrishnaiah S, D’Asti E, Rak J. Extracellular vesicle communication pathways as regulatory targets of oncogenic transformation. Semin Cell Dev Biol. 2017;67:11–22. doi:10.1016/j.semcdb.2017.01.00328077296
  • Kowal J, Arras G, Colombo M, et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci U S A. 2016;113(8):E968–977. doi:10.1073/pnas.152123011326858453
  • Jiang L, Vader P, Schiffelers RM. Extracellular vesicles for nucleic acid delivery: progress and prospects for safe RNA-based gene therapy. Gene Ther. 2017;24(3):157–166. doi:10.1038/gt.2017.828140387
  • Sharma A, Johnson A. Exosome DNA: critical regulator of tumor immunity and a diagnostic biomarker. J Cell Physiol. 2020;235(3):1921–1932. doi:10.1002/jcp.2915331512231
  • Sansone P, Savini C, Kurelac I, et al. Packaging and transfer of mitochondrial DNA via exosomes regulate escape from dormancy in hormonal therapy-resistant breast cancer. Proc Natl Acad Sci U S A. 2017;114(43):E9066–E9075. doi:10.1073/pnas.170486211429073103
  • Jeppesen DK, Fenix AM, Franklin JL, et al. Reassessment of exosome composition. Cell. 2019;177(2):428–445 e418. doi:10.1016/j.cell.2019.02.02930951670
  • Tkach M, Thery C. Communication by extracellular vesicles: where we are and where we need to go. Cell. 2016;164(6):1226–1232. doi:10.1016/j.cell.2016.01.04326967288
  • 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
  • Thery C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. 2006;30:3–22. doi:10.1002/0471143030.cb0322s30
  • Thery C, Ostrowski M, Segura E. Membrane vesicles as conveyors of immune responses. Nat Rev Immunol. 2009;9(8):581–593. doi:10.1038/nri256719498381
  • Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29(4):341–345. doi:10.1038/nbt.180721423189
  • Das CK, Jena BC, Banerjee I, et al. Exosome as a novel shuttle for delivery of therapeutics across biological barriers. Mol Pharm. 2019;16(1):24–40. doi:10.1021/acs.molpharmaceut.8b0090130513203
  • Monguio-Tortajada M, Galvez-Monton C, Bayes-Genis A, Roura S, Borras FE. Extracellular vesicle isolation methods: rising impact of size-exclusion chromatography. Cell Mol Life Sci. 2019;76(12):2369–2382. doi:10.1007/s00018-019-03071-y30891621
  • Coumans FAW, Brisson AR, Buzas EI, et al. Methodological guidelines to study extracellular vesicles. Circ Res. 2017;120(10):1632–1648. doi:10.1161/CIRCRESAHA.117.30941728495994
  • Xu R, Greening DW, Zhu HJ, Takahashi N, Simpson RJ. Extracellular vesicle isolation and characterization: toward clinical application. J Clin Invest. 2016;126(4):1152–1162. doi:10.1172/JCI8112927035807
  • Tang K, Zhang Y, Zhang H, et al. Delivery of chemotherapeutic drugs in tumour cell-derived microparticles. Nat Commun. 2012;3:1282. doi:10.1038/ncomms228223250412
  • Votteler J, Ogohara C, Yi S, et al. Designed proteins induce the formation of nanocage-containing extracellular vesicles. Nature. 2016;540(7632):292–295. doi:10.1038/nature2060727919066
  • Garcia-Manrique P, Matos M, Gutierrez G, Pazos C, Blanco-Lopez MC. Therapeutic biomaterials based on extracellular vesicles: classification of bio-engineering and mimetic preparation routes. J Extracell Vesicles. 2018;7(1):1422676. doi:10.1080/20013078.2017.142267629372017
  • Li Z, Zhou X, Wei M, et al. In vitro and in vivo RNA inhibition by CD9-HuR functionalized exosomes encapsulated with miRNA or CRISPR/dCas9. Nano Lett. 2018;19(1):19–28. doi:10.1021/acs.nanolett.8b0268930517011
  • Lv P, Liu X, Chen X, et al. Genetically engineered cell membrane nanovesicles for oncolytic adenovirus delivery: a versatile platform for cancer virotherapy. Nano Lett. 2019;19(5):2993–3001. doi:10.1021/acs.nanolett.9b0014530964695
  • Zhang YF, Shi JB, Li C. Small extracellular vesicle loading systems in cancer therapy: current status and the way forward. Cytotherapy. 2019;21(11):1122–1136. doi:10.1016/j.jcyt.2019.10.00231699595
  • Jang SC, Kim OY, Yoon CM, et al. Bioinspired exosome-mimetic nanovesicles for targeted delivery of chemotherapeutics to malignant tumors. ACS Nano. 2013;7(9):7698–7710. doi:10.1021/nn402232g24004438
  • 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
  • Wan Y, Wang L, Zhu C, et al. Aptamer-conjugated extracellular nanovesicles for targeted drug delivery. Cancer Res. 2018;78(3):798–808. doi:10.1158/0008-5472.CAN-17-288029217761
  • Baek G, Choi H, Kim Y, Lee HC, Choi C. Mesenchymal stem cell-derived extracellular vesicles as therapeutics and as a drug delivery platform. Stem Cells Transl Med. 2019;8(9):880–886. doi:10.1002/sctm.18-022631045328
  • Sun D, Zhuang X, Xiang X, et al. A novel nanoparticle drug delivery system: the anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol Ther. 2010;18(9):1606–1614. doi:10.1038/mt.2010.10520571541
  • Saari H, Lazaro-Ibanez 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(Pt B):727–737. doi:10.1016/j.jconrel.2015.09.03126390807
  • Walker S, Busatto S, Pham A, et al. Extracellular vesicle-based drug delivery systems for cancer treatment. Theranostics. 2019;9(26):8001–8017. doi:10.7150/thno.3709731754377
  • Apparailly F, Jorgensen C. siRNA-based therapeutic approaches for rheumatic diseases. Nat Rev Rheumatol. 2013;9(1):56–62. doi:10.1038/nrrheum.2012.17623090506
  • Tatiparti K, Sau S, Kashaw SK, Iyer AK. siRNA delivery strategies: a comprehensive review of recent developments. Nanomaterials. 2017;7(4):77. doi:10.3390/nano7040077
  • Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 2017;16(3):203–222. doi:10.1038/nrd.2016.24628209991
  • Liu T, Zhang X, Du L, et al. Exosome-transmitted miR-128-3p increase chemosensitivity of oxaliplatin-resistant colorectal cancer. Mol Cancer. 2019;18(1):43. doi:10.1186/s12943-019-0981-730890168
  • Baldari S, Di Rocco G, Magenta A, Picozza M, Toietta G. Extracellular vesicles-encapsulated microRNA-125b produced in genetically modified mesenchymal stromal cells inhibits hepatocellular carcinoma cell proliferation. Cells. 2019;8(12):1560. doi:10.3390/cells8121560
  • Zhang K, Dong C, Chen M, et al. Extracellular vesicle-mediated delivery of miR-101 inhibits lung metastasis in osteosarcoma. Theranostics. 2020;10(1):411–425. doi:10.7150/thno.3348231903129
  • Garneau JE, Dupuis ME, Villion M, et al. The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA. Nature. 2010;468(7320):67–71. doi:10.1038/nature0952321048762
  • Lin Y, Wu J, Gu W, et al. Exosome-liposome hybrid nanoparticles deliver CRISPR/Cas9 system in MSCs. Adv Sci. 2018;5(4):1700611. doi:10.1002/advs.201700611
  • Kanada M, Kim BD, Hardy JW, et al. Microvesicle-mediated delivery of minicircle DNA results in effective gene-directed enzyme prodrug cancer therapy. Mol Cancer Ther. 2019;18(12):2331–2342. doi:10.1158/1535-7163.MCT-19-029931451563
  • Batagov AO, Kuznetsov VA, Kurochkin IV. Identification of nucleotide patterns enriched in secreted RNAs as putative cis-acting elements targeting them to exosome nano-vesicles. BMC Genomics. 2011;12(Suppl 3):S18. doi:10.1186/1471-2164-12-S3-S1822369587
  • Bolukbasi MF, Mizrak A, Ozdener GB, et al. miR-1289 and “Zipcode”-like sequence enrich mRNAs in microvesicles. Mol Ther Nucleic Acids. 2012;1:e10. doi:10.1038/mtna.2011.223344721
  • Lee YS, Kim SH, Cho JA, Kim CW. Introduction of the CIITA gene into tumor cells produces exosomes with enhanced anti-tumor effects. Exp Mol Med. 2011;43(5):281–290. doi:10.3858/emm.2011.43.5.02921464590
  • Mizrak A, Bolukbasi MF, Ozdener GB, et al. Genetically engineered microvesicles carrying suicide mRNA/protein inhibit schwannoma tumor growth. Mol Ther. 2013;21(1):101–108. doi:10.1038/mt.2012.16122910294
  • Sterzenbach U, Putz U, Low LH, Silke J, Tan SS, Howitt J. Engineered exosomes as vehicles for biologically active proteins. Mol Ther. 2017;25(6):1269–1278. doi:10.1016/j.ymthe.2017.03.03028412169
  • Barok M, Puhka M, Vereb G, Szollosi J, Isola J, Joensuu H. Cancer-derived exosomes from HER2-positive cancer cells carry trastuzumab-emtansine into cancer cells leading to growth inhibition and caspase activation. BMC Cancer. 2018;18(1):504. doi:10.1186/s12885-018-4418-229720111
  • Noguchi K, Hirano M, Hashimoto T, Yuba E, Takatani-Nakase T, Nakase I. Effects of lyophilization of arginine-rich cell-penetrating peptide-modified extracellular vesicles on intracellular delivery. Anticancer Res. 2019;39(12):6701–6709. doi:10.21873/anticanres.1388531810935
  • Liu B, Gordon WP, Richmond W, Groessl T, Tuntland T. Use of solubilizers in preclinical formulations: effect of Cremophor EL on the pharmacokinetic properties on early discovery compounds. Eur J Pharm Sci. 2016;87:52–57.26499309
  • 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
  • Kim MS, Haney MJ, Zhao Y, et al. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: in vitro and in vivo evaluations. Nanomedicine. 2018;14(1):195–204. doi:10.1016/j.nano.2017.09.01128982587
  • Wei H, Chen J, Wang S, et al. A nanodrug consisting of doxorubicin and exosome derived from mesenchymal stem cells for osteosarcoma treatment in vitro. Int J Nanomedicine. 2019;14:8603–8610. doi:10.2147/IJN.S21898831802872
  • Gong C, Tian J, Wang Z, et al. Functional exosome-mediated co-delivery of doxorubicin and hydrophobically modified microRNA 159 for triple-negative breast cancer therapy. J Nanobiotechnology. 2019;17(1):93. doi:10.1186/s12951-019-0526-731481080
  • Huyan T, Du Y, Huang Q, Huang Q, Li Q. Uptake characterization of tumor cell-derived exosomes by natural killer cells. Iran J Public Health. 2018;47(6):803–813.30087865
  • Ohno S, Takanashi M, Sudo K, et al. Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol Ther. 2013;21(1):185–191. doi:10.1038/mt.2012.18023032975
  • Kooijmans SA, Aleza CG, Roffler SR, van Solinge WW, Vader P, Schiffelers RM. Display of GPI-anchored anti-EGFR nanobodies on extracellular vesicles promotes tumour cell targeting. J Extracell Vesicles. 2016;5:31053. doi:10.3402/jev.v5.3105326979463
  • Gomari H, Forouzandeh Moghadam M, Soleimani M. Targeted cancer therapy using engineered exosome as a natural drug delivery vehicle. Onco Targets Ther. 2018;11:5753–5762. doi:10.2147/OTT.S17311030254468
  • Bellavia D, Raimondo S, Calabrese G, et al. Interleukin 3- receptor targeted exosomes inhibit in vitro and in vivo chronic myelogenous leukemia cell growth. Theranostics. 2017;7(5):1333–1345. doi:10.7150/thno.1709228435469
  • Liang G, Kan S, Zhu Y, Feng S, Feng W, Gao S. Engineered exosome-mediated delivery of functionally active miR-26a and its enhanced suppression effect in HepG2 cells. Int J Nanomedicine. 2018;13:585–599. doi:10.2147/IJN.S15445829430178
  • Yoshida K, Tsuda M, Matsumoto R, et al. Exosomes containing ErbB2/CRK induce vascular growth in premetastatic niches and promote metastasis of bladder cancer. Cancer Sci. 2019;110(7):2119–2132. doi:10.1111/cas.1408031141251
  • Li Y, Liu Y, Xiu F, et al. Characterization of exosomes derived from Toxoplasma gondii and their functions in modulating immune responses. Int J Nanomedicine. 2018;13:467–477. doi:10.2147/IJN.S15111029403276
  • Liu H, Shen M, Zhao D, et al. The effect of triptolide-loaded exosomes on the proliferation and apoptosis of human ovarian cancer SKOV3 cells. Biomed Res Int. 2019;2019:2595801.31240207
  • 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
  • Kanada M, Bachmann MH, Hardy JW, et al. Differential fates of biomolecules delivered to target cells via extracellular vesicles. Proc Natl Acad Sci U S A. 2015;112(12):E1433–1442. doi:10.1073/pnas.141840111225713383
  • Lai CP, Kim EY, Badr CE, et al. Visualization and tracking of tumour extracellular vesicle delivery and RNA translation using multiplexed reporters. Nat Commun. 2015;6:7029. doi:10.1038/ncomms802925967391
  • Yim N, Ryu SW, Choi K, et al. Exosome engineering for efficient intracellular delivery of soluble proteins using optically reversible protein-protein interaction module. Nat Commun. 2016;7:12277. doi:10.1038/ncomms1227727447450
  • Pi F, Binzel DW, Lee TJ, et al. Nanoparticle orientation to control RNA loading and ligand display on extracellular vesicles for cancer regression. Nat Nanotechnol. 2018;13(1):82–89. doi:10.1038/s41565-017-0012-z29230043
  • Smyth T, Petrova K, Payton NM, et al. Surface functionalization of exosomes using click chemistry. Bioconjug Chem. 2014;25(10):1777–1784. doi:10.1021/bc500291r25220352
  • Kooijmans SAA, Fliervoet LAL, van der Meel R, et al. PEGylated and targeted extracellular vesicles display enhanced cell specificity and circulation time. J Control Release. 2016;224:77–85. doi:10.1016/j.jconrel.2016.01.00926773767
  • Ichihara M, Shimizu T, Imoto A, et al. Anti-PEG IgM response against PEGylated liposomes in mice and rats. Pharmaceutics. 2010;3(1):1–11. doi:10.3390/pharmaceutics301000124310423
  • Zhu Q, Ling X, Yang Y, et al. Embryonic stem cells-derived exosomes endowed with targeting properties as chemotherapeutics delivery vehicles for glioblastoma therapy. Adv Sci. 2019;6(6):1801899. doi:10.1002/advs.201801899
  • Li Y, Gao Y, Gong C, et al. A33 antibody-functionalized exosomes for targeted delivery of doxorubicin against colorectal cancer. Nanomedicine. 2018;14(7):1973–1985. doi:10.1016/j.nano.2018.05.02029935333
  • Nakase I, Kobayashi NB, Takatani-Nakase T, Yoshida T. Active macropinocytosis induction by stimulation of epidermal growth factor receptor and oncogenic Ras expression potentiates cellular uptake efficacy of exosomes. Sci Rep. 2015;5:10300. doi:10.1038/srep1030026036864
  • Nakase I, Futaki S. Combined treatment with a pH-sensitive fusogenic peptide and cationic lipids achieves enhanced cytosolic delivery of exosomes. Sci Rep. 2015;5:10112. doi:10.1038/srep1011226011176
  • Nakase I, Noguchi K, Fujii I, Futaki S. Vectorization of biomacromolecules into cells using extracellular vesicles with enhanced internalization induced by macropinocytosis. Sci Rep. 2016;6:34937. doi:10.1038/srep3493727748399
  • Nakase I, Ueno N, Katayama M, et al. Receptor clustering and activation by multivalent interaction through recognition peptides presented on exosomes. Chem Commun. 2017;53(2):317–320. doi:10.1039/C6CC06719K
  • Nakase I, Noguchi K, Aoki A, Takatani-Nakase T, Fujii I, Futaki S. Arginine-rich cell-penetrating peptide-modified extracellular vesicles for active macropinocytosis induction and efficient intracellular delivery. Sci Rep. 2017;7(1):1991. doi:10.1038/s41598-017-02014-628512335
  • Guo M, Wu F, Hu G, et al. Autologous tumor cell-derived microparticle-based targeted chemotherapy in lung cancer patients with malignant pleural effusion. Sci Transl Med. 2019;11(474). doi:10.1126/scitranslmed.aav5519
  • Melnik BC, John SM, Schmitz G. Milk: an exosomal microRNA transmitter promoting thymic regulatory T cell maturation preventing the development of atopy? J Transl Med. 2014;12:43. doi:10.1186/1479-5876-12-4324521175
  • Aqil F, Munagala R, Jeyabalan J, et al. Milk exosomes - Natural nanoparticles for siRNA delivery. Cancer Lett. 2019;449:186–195. doi:10.1016/j.canlet.2019.02.01130771430
  • Li Z, Wang H, Yin H, Bennett C, Zhang HG, Guo P. Arrowtail RNA for ligand display on ginger exosome-like nanovesicles to systemic deliver siRNA for cancer suppression. Sci Rep. 2018;8(1):14644. doi:10.1038/s41598-018-32953-730279553
  • Baldini N, Torreggiani E, Roncuzzi L, Perut F, Zini N, Avnet S. Exosome-like nanovesicles isolated from citrus limon L. exert antioxidative effect. Curr Pharm Biotechnol. 2018;19(11):877–885. doi:10.2174/138920101966618101711575530332948
  • Ju S, Mu J, Dokland T, et al. Grape exosome-like nanoparticles induce intestinal stem cells and protect mice from DSS-induced colitis. Mol Ther. 2013;21(7):1345–1357. doi:10.1038/mt.2013.6423752315
  • Mu J, Zhuang X, Wang Q, et al. Interspecies communication between plant and mouse gut host cells through edible plant derived exosome-like nanoparticles. Mol Nutr Food Res. 2014;58(7):1561–1573. doi:10.1002/mnfr.20130072924842810
  • Zhu L, Gangadaran P, Kalimuthu S, et al. Novel alternatives to extracellular vesicle-based immunotherapy - exosome mimetics derived from natural killer cells. Artif Cells Nanomed Biotechnol. 2018;46(sup3):S166–S179. doi:10.1080/21691401.2018.148982430092165
  • Jo W, Jeong D, Kim J, et al. Microfluidic fabrication of cell-derived nanovesicles as endogenous RNA carriers. Lab Chip. 2014;14(7):1261–1269. doi:10.1039/C3LC50993A24493004
  • Yoon J, Jo W, Jeong D, Kim J, Jeong H, Park J. Generation of nanovesicles with sliced cellular membrane fragments for exogenous material delivery. Biomaterials. 2015;59:12–20. doi:10.1016/j.biomaterials.2015.04.02825941997
  • Goh WJ, Zou S, Ong WY, et al. Bioinspired cell-derived nanovesicles versus exosomes as drug delivery systems: a cost-effective alternative. Sci Rep. 2017;7(1):14322. doi:10.1038/s41598-017-14725-x29085024
  • Goh WJ, Zou S, Czarny B, Pastorin G. nCVTs: a hybrid smart tumour targeting platform. Nanoscale. 2018;10(15):6812–6819. doi:10.1039/C7NR08720A29595203
  • Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Adv Drug Deliv Rev. 2013;65(1):36–48. doi:10.1016/j.addr.2012.09.03723036225
  • Bulbake U, Doppalapudi S, Kommineni N, Khan W. Liposomal formulations in clinical use: an updated review. Pharmaceutics. 2017;9(2):12. doi:10.3390/pharmaceutics9020012
  • Farooq MA, Aquib M, Farooq A, et al. Recent progress in nanotechnology-based novel drug delivery systems in designing of cisplatin for cancer therapy: an overview. Artif Cells Nanomed Biotechnol. 2019;47(1):1674–1692. doi:10.1080/21691401.2019.160453531066300
  • Cheng R, Liu L, Xiang Y, et al. Advanced liposome-loaded scaffolds for therapeutic and tissue engineering applications. Biomaterials. 2020;232:119706. doi:10.1016/j.biomaterials.2019.11970631918220
  • Hoshino A, Costa-Silva B, Shen TL, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527(7578):329–335. doi:10.1038/nature1575626524530
  • 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
  • Piffoux M, Silva AKA, 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(7):6830–6842. doi:10.1021/acsnano.8b0205329975503
  • Yeo RW, Lai RC, Zhang B, et al. Mesenchymal stem cell: an efficient mass producer of exosomes for drug delivery. Adv Drug Deliv Rev. 2013;65(3):336–341. doi:10.1016/j.addr.2012.07.00122780955
  • Cui X, He Z, Liang Z, Chen Z, Wang H, Zhang J. Exosomes from adipose-derived mesenchymal stem cells protect the myocardium against ischemia/reperfusion injury through Wnt/β-catenin signaling pathway. J Cardiovasc Pharmacol. 2017;70(4):225–231. doi:10.1097/FJC.000000000000050728582278
  • Wang B, Yao K, Huuskes BM, et al. Mesenchymal stem cells deliver exogenous microRNA-let7c via exosomes to attenuate renal fibrosis. Mol Ther. 2016;24(7):1290–1301. doi:10.1038/mt.2016.9027203438
  • Bai L, Shao H, Wang H, et al. Effects of mesenchymal stem cell-derived exosomes on experimental autoimmune uveitis. Sci Rep. 2017;7(1):4323. doi:10.1038/s41598-017-04559-y28659587
  • Chen Z, Wang H, Xia Y, Yan F, Lu Y. Therapeutic potential of mesenchymal cell-derived miRNA-150-5p-expressing exosomes in rheumatoid arthritis mediated by the modulation of MMP14 and VEGF. J Immunol. 2018;201(8):2472–2482. doi:10.4049/jimmunol.180030430224512
  • Ding Y, Cao F, Sun H, et al. 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. doi:10.1016/j.canlet.2018.10.03930419348
  • Rialland P, Lankar D, Raposo G, Bonnerot C, Hubert P. BCR-bound antigen is targeted to exosomes in human follicular lymphoma B-cells. Biol Cell. 2006;98(8):491–501. doi:10.1042/BC2006002716677129
  • Blanchard N, Lankar D, Faure F, et al. TCR activation of human T cells induces the production of exosomes bearing the TCR/CD3/ζ complex. J Immunol. 2002;168(7):3235–3241. doi:10.4049/jimmunol.168.7.323511907077
  • Del Cacho E, Gallego M, Lee SH, et al. Induction of protective immunity against Eimeria tenella infection using antigen-loaded dendritic cells (DC) and DC-derived exosomes. Vaccine. 2011;29(21):3818–3825. doi:10.1016/j.vaccine.2011.03.02221439315
  • Lugini L, Cecchetti S, Huber V, et al. Immune surveillance properties of human NK cell-derived exosomes. J Immunol. 2012;189(6):2833–2842. doi:10.4049/jimmunol.110198822904309
  • Jong AY, Wu CH, Li J, et al. Large-scale isolation and cytotoxicity of extracellular vesicles derived from activated human natural killer cells. J Extracell Vesicles. 2017;6(1):1294368. doi:10.1080/20013078.2017.129436828326171
  • Baginska J, Viry E, Paggetti J, et al. The critical role of the tumor microenvironment in shaping natural killer cell-mediated anti-tumor immunity. Front Immunol. 2013;4:490. doi:10.3389/fimmu.2013.0049024400010
  • Ishikawa E, Tsuboi K, Saijo K, et al. Autologous natural killer cell therapy for human recurrent malignant glioma. Anticancer Res. 2004;24(3b):1861–1871.15274367
  • Koehl U, Sorensen J, Esser R, et al. IL-2 activated NK cell immunotherapy of three children after haploidentical stem cell transplantation. Blood Cells Mol Dis. 2004;33(3):261–266. doi:10.1016/j.bcmd.2004.08.01315528141
  • Passweg JR, Tichelli A, Meyer-Monard S, et al. Purified donor NK-lymphocyte infusion to consolidate engraftment after haploidentical stem cell transplantation. Leukemia. 2004;18(11):1835–1838. doi:10.1038/sj.leu.240352415457184
  • Fais S. NK cell-released exosomes: natural nanobullets against tumors. Oncoimmunology. 2013;2(1):e22337. doi:10.4161/onci.2233723482694
  • Li Q, Wang H, Peng H, Huyan T, Cacalano NA. Exosomes: versatile nano mediators of immune regulation. Cancers. 2019;11(10):1557. doi:10.3390/cancers11101557
  • Wang G, Hu W, Chen H, Shou X, Ye T, Xu Y. Cocktail strategy based on NK cell-derived exosomes and their biomimetic nanoparticles for dual tumor therapy. Cancers. 2019;11(10):1560. doi:10.3390/cancers11101560
  • Ortega FG, Roefs MT, de Miguel Perez D, et al. Interfering with endolysosomal trafficking enhances release of bioactive exosomes. Nanomedicine. 2019;20:102014. doi:10.1016/j.nano.2019.10201431152797
  • Ban JJ, Lee M, Im W, Kim M. Low pH increases the yield of exosome isolation. Biochem Biophys Res Commun. 2015;461(1):76–79. doi:10.1016/j.bbrc.2015.03.17225849885
  • Haraszti RA, Miller R, Stoppato M, et al. Exosomes produced from 3D cultures of MSCs by tangential flow filtration show higher yield and improved activity. Mol Ther. 2018;26(12):2838–2847. doi:10.1016/j.ymthe.2018.09.01530341012
  • Caponnetto F, Manini I, Skrap M, et al. Size-dependent cellular uptake of exosomes. Nanomedicine. 2017;13(3):1011–1020. doi:10.1016/j.nano.2016.12.00927993726
  • Shang L, Nienhaus K, Nienhaus GU. Engineered nanoparticles interacting with cells: size matters. J Nanobiotechnology. 2014;12:5. doi:10.1186/1477-3155-12-524491160
  • Zhu X, Badawi M, Pomeroy S, et al. Comprehensive toxicity and immunogenicity studies reveal minimal effects in mice following sustained dosing of extracellular vesicles derived from HEK293T cells. J Extracell Vesicles. 2017;6(1):1324730. doi:10.1080/20013078.2017.132473028717420
  • Barile L, Vassalli G. Exosomes: therapy delivery tools and biomarkers of diseases. Pharmacol Ther. 2017;174:63–78. doi:10.1016/j.pharmthera.2017.02.02028202367