Perspectives in Manipulating EVs for Therapeutic Applications: Focus on Cancer Treatment
Abstract
:1. Introduction
2. Proposed Clinical Applications of EVs in Oncology
2.1. EVs as Cancer Biomarkers
2.1.1. EV-Surface Molecules as Predictors for Patient Responsiveness to Therapy
2.1.2. EV-Expressed Micro RNAs (miRNAs) as Predictors of Drug Resistance and Metastatic Potential
2.2. EVs as Vaccines Inducing Anti-Tumor Immune Responses
2.2.1. DC-Derived EVs
2.2.2. Tumor-Derived EVs
2.2.3. Engineered Nanovesicles
2.3. EVs as a Delivery Tool
EVs for Drug Delivery
3. Perspectives in Manipulating EVs for Therapeutic Applications
3.1. EVs’ Isolation Methods
3.2. Approaches to Loading EVs with Selected Cargo
3.2.1. Passive Loading
3.2.2. Electroporation
3.2.3. Treatment with Saponins
3.2.4. Sonication
3.2.5. Hypotonic Dialysis
3.2.6. Freeze-Thaw Cycles
3.2.7. Extrusion
3.2.8. Novel Approach to Loading of Nucleic Acids
3.3. Directing EVs towards Desired Target
3.3.1. Approaches Based on Receptor-Ligand Interactions
3.3.2. Approaches Based on Antigen-Specific Interactions
3.4. Selecting the Optimal Route of EVs’ Administration
4. Conclusions
Funding
Conflicts of Interest
Abbreviations
CTLA-4 | Cytotoxic T-lymphocyte-associated protein 4 |
DCs | Dendritic cells |
EVs | Extracellular vesicles |
FasL | Fas ligand |
LCs | Light chains |
MHC | Major histocompatibility complex |
miRNA | Micro ribonucleic acid |
PD-1 | Programmed death receptor-1 |
PD-L1 | Programmed death receptor-1 ligand |
siRNA | Small interfering ribonucleic acid |
SIRPα | Signal regulatory protein alpha |
References
- Nazimek, K.; Ptak, W.; Nowak, B.; Ptak, M.; Askenase, P.W.; Bryniarski, K. Macrophages play an essential role in antigen-specific immune suppression mediated by T CD8⁺ cell-derived exosomes. Immunology 2015, 146, 23–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khawar, M.B.; Abbasi, M.H.; Siddique, Z.; Arif, A.; Sheikh, N. An update on novel therapeutic warfronts of extracellular vesicles (EVs) in cancer treatment: Where we are standing right now and where to go in the future. Oxid. Med. Cell Longev. 2019, 2019, 9702562. [Google Scholar] [CrossRef]
- Rahbarghazi, R.; Jabbari, N.; Sani, N.A.; Asghari, R.; Salimi, L.; Kalashani, S.A.; Feghhi, M.; Etemadi, T.; Akbariazar, E.; Mahmoudi, M.; et al. Tumor-derived extracellular vesicles: Reliable tools for cancer diagnosis and clinical applications. Cell. Commun. Signal. 2019, 17, 73. [Google Scholar] [CrossRef] [Green Version]
- Susa, F.; Limongi, T.; Dumontel, B.; Vighetto, V.; Cauda, V. Engineered Extracellular Vesicles as a Reliable Tool in Cancer Nanomedicine. Cancers 2019, 11, 1979. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- He, C.; Zheng, S.; Luo, Y.; Wang, B. Exosome theranostics: Biology and translational medicine. Theranostics 2018, 8, 237–255. [Google Scholar] [CrossRef] [PubMed]
- Junqueira-Neto, S.; Batista, I.A.; Costa, J.L.; Melo, S.A. Liquid biopsy beyond circulating tumor cells and cell-free DNA. Acta Cytol. 2019, 63, 479–488. [Google Scholar] [CrossRef] [PubMed]
- Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef]
- Yang, Y.; Kannisto, E.; Yu, G.; Reid, M.E.; Patnaik, S.K.; Wu, Y. An immuno-biochip selectively captures tumor-derived exosomes and detects exosomal RNAs for cancer diagnosis. ACS Appl. Mater. Interfaces 2018, 10, 43375–43386. [Google Scholar] [CrossRef]
- Kanlikilicer, P.; Bayraktar, R.; Denizli, M.; Rashed, M.H.; Ivan, C.; Aslan, B.; Mitra, R.; Karagoz, K.; Bayraktar, E.; Zhang, X.; et al. Exosomal miRNA confers chemo resistance via targeting Cav1/p-gp/M2-type macrophage axis in ovarian cancer. EBioMedicine 2018, 38, 100–112. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.; Yao, F.; Xiao, Z.; Sun, Y.; Ma, L. MicroRNAs and metastasis: Small RNAs play big roles. Cancer Metastasis Rev. 2018, 37, 5–15. [Google Scholar] [CrossRef]
- Crescioli, S.; Daniels-Wells, T.R.; Dombrowicz, D.; Fiebiger, E.; Gould, H.J.; Irshad, S.; Janda, J.; Josephs, D.H.; Levi-Schaffer, F.; O’ Mahony, L.; et al. AllergoOncology: Opposite outcomes of immune tolerance in allergy and cancer. Allergy 2018, 73, 328–340. [Google Scholar]
- Markov, O.; Oshchepkova, A.; Mironova, N. Immunotherapy based on dendritic cell-targeted/-derived extracellular vesicles - a novel strategy for enhancement of the anti-tumor immune response. Front. Pharmacol. 2019, 10, 1152. [Google Scholar] [CrossRef] [Green Version]
- Butterfield, L.H. Dendritic cells in cancer immunotherapy clinical trials: Are we making progress? Front. Immunol. 2013, 4, 454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santos, P.M.; Butterfield, L.H. Dendritic cell-based cancer vaccines. J. Immunol. 2018, 200, 443–449. [Google Scholar] [CrossRef] [PubMed]
- Pitt, J.M.; André, F.; Amigorena, S.; Soria, J.C.; Eggermont, A.; Kroemer, G.; Zitvogel, L. Dendritic cell-derived exosomes for cancer therapy. J. Clin. Invest. 2016, 126, 1224–1232. [Google Scholar] [CrossRef] [PubMed]
- Le Pecq, J.B. Dexosomes as a therapeutic cancer vaccine: From bench to bedside. Blood Cells Mol. Dis. 2005, 35, 129–135. [Google Scholar] [CrossRef] [PubMed]
- Besse, B.; Charrier, M.; Lapierre, V.; Dansin, E.; Lantz, O.; Planchard, D.; Le Chevalier, T.; Livartoski, A.; Barlesi, F.; Laplanche, A.; et al. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology 2015, 5, e1071008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morse, M.A.; Garst, J.; Osada, T.; Khan, S.; Hobeika, A.; Clay, T.M.; Valente, N.; Shreeniwas, R.; Sutton, M.A.; Delcayre, A.; et al. A phase I study of dexosome immunotherapy in patients with advanced non-small cell lung cancer. J. Transl. Med. 2005, 3, 9. [Google Scholar] [CrossRef] [Green Version]
- Escudier, B.; Dorval, T.; Chaput, N.; André, F.; Caby, M.P.; Novault, S.; Flament, C.; Leboulaire, C.; Borg, C.; Amigorena, S.; et al. Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: Results of the first phase I clinical trial. J. Transl. Med. 2005, 3, 10. [Google Scholar] [CrossRef] [Green Version]
- Chulpanova, D.S.; Kitaeva, K.V.; James, V.; Rizvanov, A.A.; Solovyeva, V.V. Therapeutic prospects of extracellular vesicles in cancer treatment. Front. Immunol. 2018, 9, 1534. [Google Scholar] [CrossRef] [Green Version]
- Mahaweni, N.M.; Kaijen-Lambers, M.E.; Dekkers, J.; Aerts, J.G.; Hegmans, J.P. Tumour-derived exosomes as antigen delivery carriers in dendritic cell-based immunotherapy for malignant mesothelioma. J. Extracell. Vesicles 2013, 2, 22492. [Google Scholar] [CrossRef] [PubMed]
- Taghikhani, A.; Hassan, Z.M.; Ebrahimi, M.; Moazzeni, S.M. microRNA modified tumor-derived exosomes as novel tools for maturation of dendritic cells. J. Cell. Physiol. 2019, 234, 9417–9427. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.; Park, H.; Yu, H.S.; Na, K.; Oh, K.T.; Lee, E.S. Dendritic cell-targeted pH-responsive extracellular vesicles for anticancer vaccination. Pharmaceutics 2019, 11, 54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Manfredi, F.; di Bonito, P.; Ridolfi, B.; Anticoli, S.; Arenaccio, C.; Chiozzini, C.; Baz Morelli, A.; Federico, M. The CD8⁺ T cell-mediated immunity induced by HPV-E6 uploaded in engineered exosomes is improved by ISCOMATRIXTM adjuvant. Vaccines 2016, 4, 42. [Google Scholar] [CrossRef] [Green Version]
- Kroll, A.V.; Jiang, Y.; Zhou, J.; Holay, M.; Fang, R.H.; Zhang, L. Biomimetic nanoparticle vaccines for cancer therapy. Adv. Biosyst. 2019, 3, 1800219. [Google Scholar] [CrossRef] [Green Version]
- Gilligan, K.E.; Dwyer, R.M. Engineering exosomes for cancer therapy. Int. J. Mol. Sci. 2017, 18, 1122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kanchanapally, R.; Deshmukh, S.K.; Chavva, S.R.; Tyagi, N.; Srivastava, S.K.; Patel, G.K.; Singh, A.P.; Singh, S. Drug-loaded exosomal preparations from different cell types exhibit distinctive loading capability, yield, and antitumor efficacies: A comparative analysis. Int. J. Nanomedicine 2019, 14, 531–541. [Google Scholar] [CrossRef] [Green Version]
- Walker, S.; Busatto, S.; Pham, A.; Tian, M.; Suh, A.; Carson, K.; Quintero, A.; Lafrence, M.; Malik, H.; Santana, M.X.; et al. Extracellular vesicle-based drug delivery systems for cancer treatment. Theranostics 2019, 9, 8001–8017. [Google Scholar] [CrossRef]
- Ma, J.; Zhang, Y.; Tang, K.; Zhang, H.; Yin, X.; Li, Y.; Xu, P.; Sun, Y.; Ma, R.; Ji, T.; et al. Reversing drug resistance of soft tumor-repopulating cells by tumor cell-derived chemotherapeutic microparticles. Cell Res. 2016, 26, 713–727. [Google Scholar] [CrossRef] [Green Version]
- de la Torre, P.; Pérez-Lorenzo, M.J.; Alcázar-Garrido, Á.; Flores, A.I. Cell-based nanoparticles delivery systems for targeted cancer therapy: Lessons from anti-angiogenesis treatments. Molecules 2020, 25, 715. [Google Scholar] [CrossRef] [Green Version]
- Sancho-Albero, M.; Encabo-Berzosa, M.D.M.; Beltrán-Visiedo, M.; Fernández-Messina, L.; Sebastián, V.; Sánchez-Madrid, F.; Arruebo, M.; Santamaría, J.; Martín-Duque, P. Efficient encapsulation of theranostic nanoparticles in cell-derived exosomes: Leveraging the exosomal biogenesis pathway to obtain hollow gold nanoparticle-hybrids. Nanoscale 2019, 11, 18825–18836. [Google Scholar] [CrossRef]
- Lara, P.; Palma-Florez, S.; Salas-Huenuleo, E.; Polakovicova, I.; Guerrero, S.; Lobos-Gonzalez, L.; Campos, A.; Muñoz, L.; Jorquera-Cordero, C.; Varas-Godoy, M.; et al. Gold nanoparticle based double-labeling of melanoma extracellular vesicles to determine the specificity of uptake by cells and preferential accumulation in small metastatic lung tumors. J. Nanobiotechnology 2020, 18, 20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rashid, M.H.; Borin, T.F.; Ara, R.; Angara, K.; Cai, J.; Achyut, B.R.; Liu, Y.; Arbab, A.S. Differential in vivo biodistribution of 131I-labeled exosomes from diverse cellular origins and its implication for theranostic application. Nanomedicine 2019, 21, 102072. [Google Scholar] [CrossRef] [PubMed]
- Szatanek, R.; Baran, J.; Siedlar, M.; Baj-Krzyworzeka, M. Isolation of extracellular vesicles: Determining the correct approach (Review). Int. J. Mol. Med. 2015, 36, 11–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Monguió-Tortajada, M.; Gálvez-Montón, C.; Bayes-Genis, A.; Roura, S.; Borràs, F.E. Extracellular vesicle isolation methods: Rising impact of size-exclusion chromatography. Cell. Mol. Life Sci. 2019, 76, 2369–2382. [Google Scholar] [CrossRef] [PubMed]
- Monguió-Tortajada, M.; Morón-Font, M.; Gámez-Valero, A.; Carreras-Planella, L.; Borràs, F.E.; Franquesa, M. Extracellular-vesicle isolation from different biological fluids by size-exclusion chromatography. Curr. Protoc. Stem Cell Biol. 2019, 49, e82. [Google Scholar] [CrossRef] [PubMed]
- Stranska, R.; Gysbrechts, L.; Wouters, J.; Vermeersch, P.; Bloch, K.; Dierickx, D.; Andrei, G.; Snoeck, R. Comparison of membrane affinity-based method with size-exclusion chromatography for isolation of exosome-like vesicles from human plasma. J. Transl. Med. 2018, 16, 1. [Google Scholar] [CrossRef]
- Xu, R.; Greening, D.W.; Rai, A.; Ji, H.; Simpson, R.J. Highly-purified exosomes and shed microvesicles isolated from the human colon cancer cell line LIM1863 by sequential centrifugal ultrafiltration are biochemically and functionally distinct. Methods 2015, 87, 11–25. [Google Scholar] [CrossRef]
- Tauro, B.J.; Greening, D.W.; Mathias, R.A.; Ji, H.; Mathivanan, S.; Scott, A.M.; Simpson, R.J. Comparison of ultracentrifugation, density gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes. Methods 2012, 56, 293–304. [Google Scholar] [CrossRef]
- Bryniarski, K.; Ptak, W.; Jayakumar, A.; Püllmann, K.; Caplan, M.J.; Chairoungdua, A.; Lu, J.; Adams, B.D.; Sikora, E.; Nazimek, K.; et al. Antigen-specific, antibody-coated, exosome-like nanovesicles deliver suppressor T-cell microRNA-150 to effector T cells to inhibit contact sensitivity. J. Allergy Clin. Immunol. 2013, 132, 170–181. [Google Scholar] [CrossRef] [Green Version]
- Wąsik, M.; Nazimek, K.; Nowak, B.; Askenase, P.W.; Bryniarski, K. Delayed-type hypersensitivity underlying casein allergy is suppressed by extracellular vesicles carrying miRNA-150. Nutrients 2019, 11, 907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nazimek, K.; Bustos-Morán, E.; Blas-Rus, N.; Nowak, B.; Ptak, W.; Askenase, P.W.; Sánchez-Madrid, F.; Bryniarski, K. Syngeneic red blood cell-induced extracellular vesicles suppress delayed-type hypersensitivity to self-antigens in mice. Clin. Exp. Allergy 2019, 49, 1487–1499. [Google Scholar] [CrossRef] [PubMed]
- Familtseva, A.; Jeremic, N.; Tyagi, S.C. Exosomes: Cell-created drug delivery systems. Mol. Cell. Biochem. 2019, 459, 1–6. [Google Scholar] [CrossRef]
- Bryniarski, K.; Ptak, W.; Martin, E.; Nazimek, K.; Szczepanik, M.; Sanak, M.; Askenase, P.W. Free extracellular miRNA functionally targets cells by transfecting exosomes from their companion cells. PLoS ONE 2015, 10, e0122991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rufino-Ramos, D.; Albuquerque, P.R.; Carmona, V.; Perfeito, R.; Nobre, R.J.; Pereira de Almeida, L. Extracellular vesicles: Novel promising delivery systems for therapy of brain diseases. J. Control. Release 2017, 262, 247–258. [Google Scholar] [CrossRef] [PubMed]
- Coccè, V.; Farronato, D.; Brini, A.T.; Masia, C.; Giannì, A.B.; Piovani, G.; Sisto, F.; Alessandri, G.; Angiero, F.; Pessina, A. Drug loaded gingival mesenchymal stromal cells (GinPa-MSCs) inhibit in vitro proliferation of oral squamous cell carcinoma. Sci. Rep. 2017, 7, 9376. [Google Scholar] [CrossRef]
- Coccè, V.; Franzè, S.; Brini, A.T.; Giannì, A.B.; Pascucci, L.; Ciusani, E.; Alessandri, G.; Farronato, G.; Cavicchini, L.; Sordi, V.; et al. In vitro anticancer activity of extracellular vesicles (EVs) secreted by gingival mesenchymal stromal cells primed with paclitaxel. Pharmaceutics 2019, 11, 61. [Google Scholar] [CrossRef] [Green Version]
- Pascucci, L.; Coccè, V.; Bonomi, A.; Ami, D.; Ceccarelli, P.; Ciusani, E.; Viganò, L.; Locatelli, A.; Sisto, F.; Doglia, S.M.; et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: A new approach for drug delivery. J. Control. Release 2014, 192, 262–270. [Google Scholar] [CrossRef]
- Tang, K.; Zhang, Y.; Zhang, H.; Xu, P.; Liu, J.; Ma, J.; Lv, M.; Li, D.; Katirai, F.; Shen, G.X.; et al. Delivery of chemotherapeutic drugs in tumour cell-derived microparticles. Nat. Commun. 2012, 3, 1282. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.H.; Forterre, A.V.; Zhao, J.; Frimannsson, D.O.; Delcayre, A.; Antes, T.J.; Efron, B.; Jeffrey, S.S.; Pegram, M.D.; Matin, A.C. Anti-HER2 scFv-directed extracellular vesicle-mediated mRNA-based gene delivery inhibits growth of HER2-positive human breast tumor xenografts by prodrug activation. Mol. Cancer Ther. 2018, 17, 1133–1142. [Google Scholar] [CrossRef] [Green Version]
- Forterre, A.V.; Wang, J.H.; Delcayre, A.; Kim, K.; Green, C.; Pegram, M.D.; Jeffrey, S.S.; Matin, A.C. Extracellular vesicle-mediated in vitro transcribed mRNA delivery for treatment of HER2+ breast cancer xenografts in mice by prodrug CB1954 without general toxicity. Mol. Cancer Ther. 2020, 19, 858–867. [Google Scholar] [CrossRef] [Green Version]
- Luan, X.; Sansanaphongpricha, K.; Myers, I.; Chen, H.; Yuan, H.; Sun, D. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacol. Sin. 2017, 38, 754–763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [PubMed]
- Lamichhane, T.N.; Jay, S.M. Production of extracellular vesicles loaded with therapeutic cargo. Methods Mol. Biol. 2018, 1831, 37–47. [Google Scholar] [PubMed]
- Sun, D.; Zhuang, X.; Xiang, X.; Liu, Y.; Zhang, S.; Liu, C.; Barnes, S.; Grizzle, W.; Miller, D.; Zhang, H.G. A novel nanoparticle drug delivery system: The anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol. Ther. 2010, 18, 1606–1614. [Google Scholar] [CrossRef]
- 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]
- Stremersch, S.; Brans, T.; Braeckmans, K.; De Smedt, S.; Raemdonck, K. Nucleic acid loading and fluorescent labeling of isolated extracellular vesicles requires adequate purification. Int. J. Pharm. 2018, 548, 783–792. [Google Scholar] [CrossRef]
- Pomatto, M.A.C.; Bussolati, B.; D’Antico, S.; Ghiotto, S.; Tetta, C.; Brizzi, M.F.; Camussi, G. Improved loading of plasma-derived extracellular vesicles to encapsulate antitumor miRNAs. Mol. Ther. Methods Clin. Dev. 2019, 13, 133–144. [Google Scholar] [CrossRef] [Green Version]
- Gomari, H.; Forouzandeh Moghadam, M.; Soleimani, M.; Ghavami, M.; Khodashenas, S. Targeted delivery of doxorubicin to HER2 positive tumor models. Int. J. Nanomedicine 2019, 14, 5679–5690. [Google Scholar] [CrossRef] [Green Version]
- Liang, G.; Zhu, Y.; Ali, D.J.; Tian, T.; Xu, H.; Si, K.; Sun, B.; Chen, B.; Xiao, Z. Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J. Nanobiotechnology 2020, 18, 10. [Google Scholar] [CrossRef]
- 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. Pharm. 2015, 12, 3650–3657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Faruqu, F.N.; Xu, L.; Al-Jamal, K.T. Preparation of exosomes for siRNA delivery to cancer cells. J. Vis. Exp. 2018. [Google Scholar] [CrossRef] [PubMed]
- Hood, J.L.; Scott, M.J.; Wickline, S.A. Maximizing exosome colloidal stability following electroporation. Anal. Biochem. 2014, 448, 41–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, Z.; Shi, J.; Xie, J.; Wang, Y.; Sun, J.; Liu, T.; Zhao, Y.; Zhao, X.; Wang, X.; Ma, Y.; et al. Large-scale generation of functional mRNA-encapsulating exosomes via cellular nanoporation. Nat. Biomed. Eng. 2020, 4, 69–83. [Google Scholar] [CrossRef]
- Shandilya, S.; Rani, P.; Onteru, S.K.; Singh, D. Small interfering RNA in milk exosomes is resistant to digestion and crosses the intestinal barrier in vitro. J. Agric. Food Chem. 2017, 65, 9506–9513. [Google Scholar] [CrossRef]
- Oshchepkova, A.; Neumestova, A.; Matveeva, V.; Artemyeva, L.; Morozova, K.; Kiseleva, E.; Zenkova, M.; Vlassov, V. Cytochalasin-B-inducible nanovesicle mimics of natural extracellular vesicles that are capable of nucleic acid transfer. Micromachines 2019, 10, 750. [Google Scholar] [CrossRef] [Green Version]
- Park, O.; Choi, E.S.; Yu, G.; Kim, J.Y.; Kang, Y.Y.; Jung, H.; Mok, H. Efficient delivery of tyrosinase related protein-2 (TRP2) peptides to lymph nodes using serum-derived exosomes. Macromol. Biosci. 2018, 18, e1800301. [Google Scholar] [CrossRef]
- Richter, M.; Fuhrmann, K.; Fuhrmann, G. Evaluation of the storage stability of extracellular vesicles. J. Vis. Exp. 2019. [Google Scholar] [CrossRef] [Green Version]
- Schaar, V.; Paulsson, M.; Mörgelin, M.; Riesbeck, K. Outer membrane vesicles shield Moraxella catarrhalis β-lactamase from neutralization by serum IgG. J. Antimicrob. Chemother. 2013, 68, 593–600. [Google Scholar] [CrossRef] [Green Version]
- Grant, R.; Ansa-Addo, E.; Stratton, D.; Antwi-Baffour, S.; Jorfi, S.; Kholia, S.; Krige, L.; Lange, S.; Inal, J. A filtration-based protocol to isolate human plasma membrane-derived vesicles and exosomes from blood plasma. J. Immunol. Methods 2011, 371, 143–151. [Google Scholar] [CrossRef]
- Kim, M.S.; Haney, M.J.; Zhao, Y.; Mahajan, V.; Deygen, I.; Klyachko, N.L.; Inskoe, E.; Piroyan, A.; Sokolsky, M.; Okolie, O.; et al. Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine 2016, 12, 655–664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, H.; Shen, M.; Zhao, D.; Ru, D.; Duan, Y.; Ding, C.; Li, H. The effect of triptolide-loaded exosomes on the proliferation and apoptosis of human ovarian cancer SKOV3 cells. Biomed. Res. Int. 2019, 2019, 2595801. [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] [PubMed]
- Le Saux, S.; Aarrass, H.; Lai-Kee-Him, J.; Bron, P.; Armengaud, J.; Miotello, G.; Bertrand-Michel, J.; Dubois, E.; George, S.; Faklaris, O.; et al. Post-production modifications of murine mesenchymal stem cell (mMSC) derived extracellular vesicles (EVs) and impact on their cellular interaction. Biomaterials 2020, 231, 119675. [Google Scholar] [CrossRef] [PubMed]
- Balbi, C.; Bolis, S.; Vassalli, G.; Barile, L. Flow cytometric analysis of extracellular vesicles from cell-conditioned media. J. Vis. Exp. 2019. [Google Scholar] [CrossRef] [Green Version]
- Salomon, C.; Yee, S.; Scholz-Romero, K.; Kobayashi, M.; Vaswani, K.; Kvaskoff, D.; Illanes, S.E.; Mitchell, M.D.; Rice, G.E. Extravillous trophoblast cells-derived exosomes promote vascular smooth muscle cell migration. Front. Pharmacol. 2014, 5, 175. [Google Scholar] [CrossRef]
- DeLoach, J.R.; Droleskey, R.E.; Andrews, K. Encapsulation by hypotonic dialysis in human erythrocytes: A diffusion or endocytosis process. Biotechnol. Appl. Biochem. 1991, 13, 72–82. [Google Scholar]
- Bosch, S.; de Beaurepaire, L.; Allard, M.; Mosser, M.; Heichette, C.; Chrétien, D.; Jegou, D.; Bach, J.M. Trehalose prevents aggregation of exosomes and cryodamage. Sci. Rep. 2016, 6, 36162. [Google Scholar] [CrossRef] [Green Version]
- Luo, X.; An, M.; Cuneo, K.C.; Lubman, D.M.; Li, L. High-performance chemical isotope labeling liquid chromatography mass spectrometry for exosome metabolomics. Anal. Chem. 2018, 90, 8314–8319. [Google Scholar] [CrossRef]
- Bæk, R.; Søndergaard, E.K.; Varming, K.; Jørgensen, M.M. The impact of various preanalytical treatments on the phenotype of small extracellular vesicles in blood analyzed by protein microarray. J. Immunol. Methods 2016, 438, 11–20. [Google Scholar] [CrossRef] [Green Version]
- Gigli, I.; Maizon, D.O. microRNAs and the mammary gland: A new understanding of gene expression. Genet. Mol. Biol. 2013, 36, 465–474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jin, Y.; Chen, K.; Wang, Z.; Wang, Y.; Liu, J.; Lin, L.; Shao, Y.; Gao, L.; Yin, H.; Cui, C.; et al. DNA in serum extracellular vesicles is stable under different storage conditions. BMC Cancer 2016, 16, 753. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kubota, S.; Chiba, M.; Watanabe, M.; Sakamoto, M.; Watanabe, N. Secretion of small/microRNAs including miR-638 into extracellular spaces by sphingomyelin phosphodiesterase 3. Oncol. Rep. 2015, 33, 67–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lv, L.L.; Cao, Y.; Liu, D.; Xu, M.; Liu, H.; Tang, R.N.; Ma, K.L.; Liu, B.C. Isolation and quantification of microRNAs from urinary exosomes/microvesicles for biomarker discovery. Int. J. Biol. Sci. 2013, 9, 1021–1031. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Yang, K.; Yuan, W.; Gao, Z. Determination of serum exosomal H19 as a noninvasive biomarker for bladder cancer diagnosis and prognosis. Med. Sci. Monit. 2018, 24, 9307–9316. [Google Scholar] [CrossRef] [PubMed]
- Sato, Y.T.; Umezaki, K.; Sawada, S.; Mukai, S.A.; Sasaki, Y.; Harada, N.; Shiku, H.; Akiyoshi, K. Engineering hybrid exosomes by membrane fusion with liposomes. Sci. Rep. 2016, 6, 21933. [Google Scholar] [CrossRef] [Green Version]
- Lee, H.; Cha, H.; Park, J.H. Derivation of cell-engineered nanovesicles from human induced pluripotent stem cells and their protective effect on the senescence of dermal fibroblasts. Int. J. Mol. Sci. 2020, 21, 343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Park, K.S.; Svennerholm, K.; Shelke, G.V.; Bandeira, E.; Lässer, C.; Jang, S.C.; Chandode, R.; Gribonika, I.; Lötvall, J. Mesenchymal stromal cell-derived nanovesicles ameliorate bacterial outer membrane vesicle-induced sepsis via IL-10. Stem Cell Res. Ther. 2019, 10, 231. [Google Scholar] [CrossRef] [Green Version]
- Thone, M.N.; Kwon, Y.J. Extracellular blebs: Artificially-induced extracellular vesicles for facile production and clinical translation. Methods 2019, S1046-202330220-8. [Google Scholar] [CrossRef]
- Zou, H.; Zhu, J.; Huang, D.S. Cell membrane capsule: A novel natural tool for antitumour drug delivery. Expert Opin. Drug Deliv. 2019, 16, 251–269. [Google Scholar] [CrossRef]
- Jang, S.C.; Kim, O.Y.; Yoon, C.M.; Choi, D.S.; Roh, T.Y.; Park, J.; Nilsson, J.; Lötvall, 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]
- Kalimuthu, S.; Gangadaran, P.; Rajendran, R.L.; Zhu, L.; Oh, J.M.; Lee, H.W.; Gopal, A.; Baek, S.H.; Jeong, S.Y.; Lee, S.W.; et al. A new approach for loading anticancer drugs into mesenchymal stem cell-derived exosome mimetics for cancer therapy. Front. Pharmacol. 2018, 9, 1116. [Google Scholar] [CrossRef]
- Jhan, Y.Y.; Prasca-Chamorro, D.; Palou Zuniga, G.; Moore, D.M.; Arun Kumar, S.; Gaharwar, A.K.; Bishop, C.J. Engineered extracellular vesicles with synthetic lipids via membrane fusion to establish efficient gene delivery. Int. J. Pharm. 2020, 573, 118802. [Google Scholar] [CrossRef]
- Jeyaram, A.; Lamichhane, T.N.; Wang, S.; Zou, L.; Dahal, E.; Kronstadt, S.M.; Levy, D.; Parajuli, B.; Knudsen, D.R.; Chao, W.; et al. Enhanced loading of functional miRNA cargo via pH gradient modification of extracellular vesicles. Mol. Ther. 2020, 28, 975–985. [Google Scholar] [CrossRef]
- Tian, Y.; Li, S.; Song, J.; Ji, T.; Zhu, M.; Anderson, G.J.; Wei, J.; Nie, G. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials 2014, 35, 2383–2390. [Google Scholar] [CrossRef]
- Wang, Y.; Chen, X.; Tian, B.; Liu, J.; Yang, L.; Zeng, L.; Chen, T.; Hong, A.; Wang, X. Nucleolin-targeted extracellular vesicles as a versatile platform for biologics delivery to breast cancer. Theranostics 2017, 7, 1360–1372. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; He, D.; Cheng, L.; Huang, C.; Zhang, Y.; Rao, X.; Kong, Y.; Li, C.; Zhang, Z.; Liu, J.; et al. p300/CBP inhibition enhances the efficacy of programmed death-ligand 1 blockade treatment in prostate cancer. Oncogene 2020. [Google Scholar] [CrossRef] [PubMed]
- Poggio, M.; Hu, T.; Pai, C.C.; Chu, B.; Belair, C.D.; Chang, A.; Montabana, E.; Lang, U.E.; Fu, Q.; Fong, L.; et al. Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell 2019, 177, 414–427.e13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brown, J.A.; Dorfman, D.M.; Ma, F.R.; Sullivan, E.L.; Munoz, O.; Wood, C.R.; Greenfield, E.A.; Freeman, G.J. Blockade of programmed death-1 ligands on dendritic cells enhances T cell activation and cytokine production. J. Immunol. 2003, 170, 1257–1266. [Google Scholar] [CrossRef]
- Versteven, M.; Van den Bergh, J.M.J.; Marcq, E.; Smits, E.L.J.; Van Tendeloo, V.F.I.; Hobo, W.; Lion, E. Dendritic cells and programmed death-1 blockade: A joint venture to combat cancer. Front. Immunol. 2018, 9, 394. [Google Scholar] [CrossRef] [Green Version]
- Yajima, T.; Hoshino, K.; Muranushi, R.; Mogi, A.; Onozato, R.; Yamaki, E.; Kosaka, T.; Tanaka, S.; Shirabe, K.; Yoshikai, Y.; et al. Fas/FasL signaling is critical for the survival of exhausted antigen-specific CD8+ T cells during tumor immune response. Mol. Immunol. 2019, 107, 97–105. [Google Scholar] [CrossRef] [PubMed]
- Zhu, J.; Petit, P.F.; Van den Eynde, B.J. Apoptosis of tumor-infiltrating T lymphocytes: A new immune checkpoint mechanism. Cancer Immunol. Immunother. 2019, 68, 835–847. [Google Scholar] [CrossRef] [PubMed]
- Lakins, M.A.; Ghorani, E.; Munir, H.; Martins, C.P.; Shields, J.D. Cancer-associated fibroblasts induce antigen-specific deletion of CD8+ T Cells to protect tumour cells. Nat. Commun. 2018, 9, 948. [Google Scholar] [CrossRef]
- Dianat-Moghadam, H.; Heidarifard, M.; Mahari, A.; Shahgolzari, M.; Keshavarz, M.; Nouri, M.; Amoozgar, Z. TRAIL in oncology: From recombinant TRAIL to nano- and self-targeted TRAIL-based therapies. Pharmacol. Res. 2020, 155, 104716. [Google Scholar] [CrossRef]
- Peter, M.E.; Hadji, A.; Murmann, A.E.; Brockway, S.; Putzbach, W.; Pattanayak, A.; Ceppi, P. The role of CD95 and CD95 ligand in cancer. Cell Death Differ. 2015, 22, 549–559. [Google Scholar] [CrossRef] [PubMed]
- Raposo, G.; Nijman, H.W.; Stoorvogel, W.; Liejendekker, R.; Harding, C.V.; Melief, C.J.; Geuze, H.J. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 1996, 183, 1161–1172. [Google Scholar] [CrossRef] [PubMed]
- Singh, K.; Ejaz, W.; Dutta, K.; Thayumanavan, S. Antibody delivery for intracellular targets: Emergent therapeutic potential. Bioconjug. Chem. 2019, 30, 1028–1041. [Google Scholar] [CrossRef] [PubMed]
- Shargh, V.H.; Hondermarck, H.; Liang, M. Antibody-targeted biodegradable nanoparticles for cancer therapy. Nanomedicine (Lond.) 2016, 11, 63–79. [Google Scholar] [CrossRef]
- Barile, L.; Vassalli, G. Exosomes: Therapy delivery tools and biomarkers of diseases. Pharmacol. Ther. 2017, 174, 63–78. [Google Scholar] [CrossRef] [Green Version]
- Nazimek, K.; Askenase, P.W.; Bryniarski, K. Antibody light chains dictate the specificity of contact hypersensitivity effector cell suppression mediated by exosomes. Int. J. Mol. Sci. 2018, 19, 2656. [Google Scholar] [CrossRef] [Green Version]
- Nazimek, K.; Bryniarski, K. Approaches to inducing antigen-specific immune tolerance in allergy and autoimmunity: Focus on antigen-presenting cells and extracellular vesicles. Scand. J. Immunol. 2020. [Google Scholar] [CrossRef] [PubMed]
- Nazimek, K.; Bryniarski, K.; Askenase, P.W. Functions of exosomes and microbial extracellular vesicles in allergy and contact and delayed-type hypersensitivity. Int. Arch. Allergy Immunol. 2016, 171, 1–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deng, G. Tumor-infiltrating regulatory T cells: Origins and features. Am. J. Clin. Exp. Immunol. 2018, 7, 81–87. [Google Scholar] [PubMed]
- Li, I.; Nabet, B.Y. Exosomes in the tumor microenvironment as mediators of cancer therapy resistance. Mol. Cancer 2019, 18, 32. [Google Scholar] [CrossRef]
- Patel, S.A.; Minn, A.J. Combination cancer therapy with immune checkpoint blockade: Mechanisms and strategies. Immunity 2018, 48, 417–433. [Google Scholar] [CrossRef] [Green Version]
- Weiskopf, K. Cancer immunotherapy targeting the CD47/SIRPα axis. Eur. J. Cancer 2017, 76, 100–109. [Google Scholar] [CrossRef]
- Topping, L.M.; Thomas, B.L.; Rhys, H.I.; Tremoleda, J.L.; Foster, M.; Seed, M.; Voisin, M.B.; Vinci, C.; Law, H.L.; Perretti, M.; et al. Targeting extracellular vesicles to the arthritic joint using a damaged cartilage-specific antibody. Front. Immunol. 2020, 11, 10. [Google Scholar] [CrossRef]
- Ptak, W.; Nazimek, K.; Askenase, P.W.; Bryniarski, K. From mysterious supernatant entity to miRNA-150 in antigen-specific exosomes: A history of hapten-specific T suppressor factor. Arch. Immunol. Ther. Exp. (Warsz). 2015, 63, 345–356. [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, 26316. [Google Scholar] [CrossRef] [Green Version]
- Brossa, A.; Fonsato, V.; Grange, C.; Tritta, S.; Tapparo, M.; Calvetti, R.; Cedrino, M.; Fallo, S.; Gontero, P.; Camussi, G.; et al. Extracellular vesicles from human liver stem cells inhibit renal cancer stem cell-derived tumor growth in vitro and in vivo. Int. J. Cancer 2020. [Google Scholar] [CrossRef] [Green Version]
- Hao, S.; Ye, Z.; Yang, J.; Bai, O.; Xiang, J. Intradermal vaccination of dendritic cell-derived exosomes is superior to a subcutaneous one in the induction of antitumor immunity. Cancer Biother. Radiopharm. 2006, 21, 146–154. [Google Scholar] [CrossRef] [PubMed]
- Lankford, K.L.; Arroyo, E.J.; Nazimek, K.; Bryniarski, K.; Askenase, P.W.; Kocsis, J.D. Intravenously delivered mesenchymal stem cell-derived exosomes target M2-type macrophages in the injured spinal cord. PLoS ONE 2018, 13, e0190358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kodali, M.; Castro, O.W.; Kim, D.K.; Thomas, A.; Shuai, B.; Attaluri, S.; Upadhya, R.; Gitai, D.; Madhu, L.N.; Prockop, D.J.; et al. Intranasally administered human MSC-derived extracellular vesicles pervasively incorporate into neurons and microglia in both intact and status epilepticus injured forebrain. Int. J. Mol. Sci. 2019, 21, 181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arntz, O.J.; Pieters, B.C.; Oliveira, M.C.; Broeren, M.G.; Bennink, M.B.; de Vries, M.; van Lent, P.L.; Koenders, M.I.; van den Berg, W.B.; van der Kraan, P.M.; et al. Oral administration of bovine milk derived extracellular vesicles attenuates arthritis in two mouse models. Mol. Nutr. Food Res. 2015, 59, 1701–1712. [Google Scholar] [CrossRef]
- Aqil, F.; Munagala, R.; Jeyabalan, J.; Agrawal, A.K.; Gupta, R. Exosomes for the enhanced tissue bioavailability and efficacy of curcumin. AAPS J. 2017, 19, 1691–1702. [Google Scholar] [CrossRef]
- Munagala, R.; Aqil, F.; Jeyabalan, J.; Gupta, R.C. Bovine milk-derived exosomes for drug delivery. Cancer Lett. 2016, 371, 48–61. [Google Scholar] [CrossRef] [Green Version]
- Somiya, M. Where does the cargo go?: Solutions to provide experimental support for the “extracellular vesicle cargo transfer hypothesis”. J. Cell Commun. Signal. 2020. [Google Scholar] [CrossRef]
Method | Principle | Advantages | Disadvantages |
---|---|---|---|
passive loading | joint incubation (sometimes in special media or buffers) | simplicity and very low impact on EVs’ and cargo quality | unpredictable efficacy |
electroporation | transient formation of pores in EV membrane with electrical pulse | high efficacy for RNA loading | very high impact on EVs’ quality and quantity 1 |
treatment with saponins | detergent-induced permeabilization of EVs’ membrane | high efficacy and low impact on EVs’ quality | impact on loaded cargo 2 |
Sonication | sound energy-induced agitation facilitating incorporation of molecules by EVs | simplicity, good efficacy and low impact on EVs’ quality | the need for individual standardization of the protocol |
hypotonic dialysis | subjecting the mixture of EVs and cargo to dialysis in hypotonic conditions | simplicity | possible impact on EVs’ quality |
freeze-thaw cycles | thermal shock-induced transient formation of pores in EV membrane | simplicity and good efficacy | very high impact on EVs’ quality and quantity |
extrusion | serial extrusion of drug-pre-incubated EV-parental cells through the polymeric filter membranes with diminishing pore size | very high quantity of yielded EVs | artificial generation of EVs |
© 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
Nazimek, K.; Bryniarski, K. Perspectives in Manipulating EVs for Therapeutic Applications: Focus on Cancer Treatment. Int. J. Mol. Sci. 2020, 21, 4623. https://doi.org/10.3390/ijms21134623
Nazimek K, Bryniarski K. Perspectives in Manipulating EVs for Therapeutic Applications: Focus on Cancer Treatment. International Journal of Molecular Sciences. 2020; 21(13):4623. https://doi.org/10.3390/ijms21134623
Chicago/Turabian StyleNazimek, Katarzyna, and Krzysztof Bryniarski. 2020. "Perspectives in Manipulating EVs for Therapeutic Applications: Focus on Cancer Treatment" International Journal of Molecular Sciences 21, no. 13: 4623. https://doi.org/10.3390/ijms21134623
APA StyleNazimek, K., & Bryniarski, K. (2020). Perspectives in Manipulating EVs for Therapeutic Applications: Focus on Cancer Treatment. International Journal of Molecular Sciences, 21(13), 4623. https://doi.org/10.3390/ijms21134623