Pioneer Role of Extracellular Vesicles as Modulators of Cancer Initiation in Progression, Drug Therapy, and Vaccine Prospects
Abstract
:1. Introduction
2. Extracellular Vesicles (EVs)
2.1. History, Functions, and Size of EVs
2.2. Cell Source of EVs
3. Intracellular Transfer of Traits between Microenvironment and Tumor Cells
4. Derived EVs and Epigenetic Biology of EVs
4.1. Methylation of DNA Influenced by EVs
4.2. RNAs and EVs as the Key Players for Cancer Progression
5. Tumor-Derived EVs and Their Approaches in Disease Progression
5.1. Initiation
5.2. Progression
5.3. Metastasis
5.4. Therapy Failure
6. Effects of Tumor-Derived EVs on Different Factors Responsible for Cancer Progressions
6.1. Role of EVs in Aiding Cell to Escape from Apoptosis
6.2. Impact of EVs on Metastasis from Nearby to Far-Flung Tissues and the Breakdown of the Blood–Brain Barrier
6.3. Reprogramming Energy Metabolism
6.4. Angiogenesis
6.5. Evasion of Immune Response
6.5.1. Fibroblasts
6.5.2. T lymphocytes
6.5.3. B Lymphocytes
6.5.4. Macrophages
6.5.5. NK and Dendritic Cells
6.5.6. Transfer of Mutations
7. Derived EVs and Therapy Resistance
7.1. EVs and Multi-Drug Resistance (MDR)
7.2. Drug Efflux Pumps and Their Transfer
7.3. Anti-Apoptotic Pathways and Their Development
7.4. Transfer of Proteins and Lipids
8. EVs as a Diagnostic Tool
Biomarkers
9. EVs as a Tool for Cancer Therapy
9.1. Drug Delivery Vehicle
9.2. EVs as Vaccines
9.3. EVs in Gene Therapy
Future Prospects of EVs in Gene Therapy
10. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- D’Arcy, M.S. Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol. Int. 2019, 43, 582–592. [Google Scholar] [CrossRef] [PubMed]
- Chaffer, C.L.; Weinberg, R.A. A perspective on cancer cell metastasis. Science 2011, 331, 1559–1564. [Google Scholar] [CrossRef] [PubMed]
- Fialkow, P.J. Clonal origin of human tumors. Biochim. Biophys. Acta Rev. Cancer 1976, 458, 283–321. [Google Scholar] [CrossRef]
- Ribatti, D.; Vacca, A. Overview of angiogenesis during tumor growth. In Angiogenesis; Springer: Berlin/Heidelberg, Germany, 2008; pp. 161–168. [Google Scholar]
- Klaus, A.; Birchmeier, W. Wnt signalling and its impact on development and cancer. Nat. Rev. Cancer 2008, 8, 387–398. [Google Scholar] [CrossRef]
- Carnino, J.M.; Hao Kwok, Z.; Jin, Y. Extracellular Vesicles: A Novel Opportunity for Precision Medicine in Respiratory Diseases. Front. Med. 2021, 8, 661679. [Google Scholar] [CrossRef]
- Xavier, C.P.; Caires, H.R.; Barbosa, M.A.; Bergantim, R.; Guimarães, J.E.; Vasconcelos, M.H. The role of extracellular vesicles in the hallmarks of cancer and drug resistance. Cells 2020, 9, 1141. [Google Scholar] [CrossRef]
- Möller, A.; Lobb, R.J. The evolving translational potential of small extracellular vesicles in cancer. Nat. Rev. Cancer 2020, 20, 697–709. [Google Scholar] [CrossRef]
- Bongiovanni, L.; Andriessen, A.; Wauben, M.H.; Hoen, E.N.N.-T.; de Bruin, A. Extracellular Vesicles: Novel Opportunities to Understand and Detect Neoplastic Diseases. Vet. Pathol. 2021, 58, 453–471. [Google Scholar] [CrossRef]
- Yamamoto, T.; Kosaka, N.; Ochiya, T. Latest advances in extracellular vesicles: From bench to bedside. Sci. Technol. Adv. Mater. 2019, 20, 746–757. [Google Scholar] [CrossRef] [Green Version]
- Qian, Z.; Shen, Q.; Yang, X.; Qiu, Y.; Zhang, W. The role of extracellular vesicles: An epigenetic view of the cancer microenvironment. BioMed Res. Int. 2015, 2015. [Google Scholar] [CrossRef] [Green Version]
- Guescini, M.; Genedani, S.; Stocchi, V.; Agnati, L.F. Astrocytes and Glioblastoma cells release exosomes carrying mtDNA. J. Neural Transm. 2010, 117, 1–4. [Google Scholar] [CrossRef] [PubMed]
- O’Loghlen, A. Role for extracellular vesicles in the tumour microenvironment. Philos. Trans. R. Soc. B Biol. Sci. 2018, 373, 20160488. [Google Scholar] [CrossRef] [PubMed]
- Harding, C.; Stahl, P. Transferrin recycling in reticulocytes: pH and iron are important determinants of ligand binding and processing. Biochem. Biophys. Res. Commun. 1983, 113, 650–658. [Google Scholar] [CrossRef]
- Pan, B.-T.; Johnstone, R.M. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell 1983, 33, 967–978. [Google Scholar] [CrossRef]
- Harding, C.V.; Heuser, J.E.; Stahl, P.D. Exosomes: Looking back three decades and into the future. J. Cell Biol. 2013, 200, 367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Trams, E.G.; Lauter, C.J.; Salem, J.N.; Heine, U. Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochim. Biophys. Acta Biomembr. 1981, 645, 63–70. [Google Scholar] [CrossRef]
- Mitchell, P.; Petfalski, E.; Shevchenko, A.; Mann, M.; Tollervey, D. The exosome: A conserved eukaryotic RNA processing complex containing multiple 3′→ 5′ exoribonucleases. Cell 1997, 91, 457–466. [Google Scholar] [CrossRef] [Green Version]
- 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, 256–263. [Google Scholar]
- Pan, B.-T.; Teng, K.; Wu, C.; Adam, M.; Johnstone, R.M. Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J. Cell Biol. 1985, 101, 942–948. [Google Scholar] [CrossRef] [Green Version]
- Raposo, G.; Nijman, H.W.; Stoorvogel, W.; Liejendekker, R.; Harding, C.V.; Melief, C.; Geuze, H.J. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 1996, 183, 1161–1172. [Google Scholar] [CrossRef]
- Zhang, H.-G.; Grizzle, W.E. Exosomes and cancer: A newly described pathway of immune suppression. Clin. Cancer Res. 2011, 17, 959–964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Simons, M.; Raposo, G. Exosomes–vesicular carriers for intercellular communication. Curr. Opin. Cell Biol. 2009, 21, 575–581. [Google Scholar] [CrossRef] [PubMed]
- Théry, C.; Ostrowski, M.; Segura, E. Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 2009, 9, 581–593. [Google Scholar] [CrossRef] [PubMed]
- Chik, F.; Szyf, M.; Rabbani, S.A. Role of epigenetics in cancer initiation and progression. Hum. Cell Transform. 2011, 720, 91–104. [Google Scholar]
- Gould, S.J.; Raposo, G. As we wait: Coping with an imperfect nomenclature for extracellular vesicles. J. Extracell. Vesicles 2013, 2, 20389. [Google Scholar] [CrossRef]
- Minciacchi, V.R.; Freeman, M.R.; Di Vizio, D. Extracellular vesicles in cancer: Exosomes, microvesicles and the emerging role of large oncosomes. Semin. Cell Dev. Biol. 2015, 40, 41–51. [Google Scholar] [CrossRef] [Green Version]
- Whiteside, T.L. The potential of tumor-derived exosomes for noninvasive cancer monitoring: An update. Expert Rev. Mol. Diagn. 2018, 18, 1029–1040. [Google Scholar] [CrossRef]
- Samanta, S.; Rajasingh, S.; Drosos, N.; Zhou, Z.; Dawn, B.; Rajasingh, J. Exosomes: New molecular targets of diseases. Acta Pharmacol. Sin. 2018, 39, 501–513. [Google Scholar] [CrossRef]
- Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Bedina Zavec, A.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [Green Version]
- Ciardiello, C.; Cavallini, L.; Spinelli, C.; Yang, J.; Reis-Sobreiro, M.; De Candia, P.; Minciacchi, V.R.; Di Vizio, D. Focus on extracellular vesicles: New frontiers of cell-to-cell communication in cancer. Int. J. Mol. Sci. 2016, 17, 175. [Google Scholar] [CrossRef] [Green Version]
- Gulinelli, S.; Salaro, E.; Vuerich, M.; Bozzato, D.; Pizzirani, C.; Bolognesi, G.; Idzko, M.; Virgilio, F.D.; Ferrari, D. IL-18 associates to microvesicles shed from human macrophages by a LPS/TLR-4 independent mechanism in response to P2X receptor stimulation. Eur. J. Immunol. 2012, 42, 3334–3345. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.-G.; Liu, C.; Su, K.; Yu, S.; Zhang, L.; Zhang, S.; Wang, J.; Cao, X.; Grizzle, W.; Kimberly, R.P. A membrane form of TNF-α presented by exosomes delays T cell activation-induced cell death. J. Immunol. 2006, 176, 7385–7393. [Google Scholar] [CrossRef] [PubMed]
- Wang, G.-J.; Liu, Y.; Qin, A.; Shah, S.V.; Deng, Z.-b.; Xiang, X.; Cheng, Z.; Liu, C.; Wang, J.; Zhang, L. Thymus exosomes-like particles induce regulatory T cells. J. Immunol. 2008, 181, 5242–5248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miyado, K.; Yoshida, K.; Yamagata, K.; Sakakibara, K.; Okabe, M.; Wang, X.; Miyamoto, K.; Akutsu, H.; Kondo, T.; Takahashi, Y. The fusing ability of sperm is bestowed by CD9-containing vesicles released from eggs in mice. Proc. Natl. Acad. Sci. USA 2008, 105, 12921–12926. [Google Scholar] [CrossRef] [Green Version]
- Bianchi, E.; Doe, B.; Goulding, D.; Wright, G.J. Juno is the egg Izumo receptor and is essential for mammalian fertilization. Nature 2014, 508, 483–487. [Google Scholar] [CrossRef]
- Desrochers, L.M.; Bordeleau, F.; Reinhart-King, C.A.; Cerione, R.A.; Antonyak, M.A. Microvesicles provide a mechanism for intercellular communication by embryonic stem cells during embryo implantation. Nat. Commun. 2016, 7, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Admyre, C.; Bohle, B.; Johansson, S.M.; Focke-Tejkl, M.; Valenta, R.; Scheynius, A.; Gabrielsson, S. B cell–derived exosomes can present allergen peptides and activate allergen-specific T cells to proliferate and produce TH2-like cytokines. J. Allergy Clin. Immunol. 2007, 120, 1418–1424. [Google Scholar] [CrossRef]
- Montecalvo, A.; Larregina, A.T.; Shufesky, W.J.; Beer Stolz, D.; Sullivan, M.L.; Karlsson, J.M.; Baty, C.J.; Gibson, G.A.; Erdos, G.; Wang, Z. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood J. Am. Soc. Hematol. 2012, 119, 756–766. [Google Scholar] [CrossRef] [Green Version]
- Okoye, I.S.; Coomes, S.M.; Pelly, V.S.; Czieso, S.; Papayannopoulos, V.; Tolmachova, T.; Seabra, M.C.; Wilson, M.S. MicroRNA-40. containing T-regulatory-cell-derived exosomes suppress pathogenic T helper 1 cells. Immunity 2014, 41, 89–103. [Google Scholar] [CrossRef] [Green Version]
- Lachenal, G.; Pernet-Gallay, K.; Chivet, M.; Hemming, F.J.; Belly, A.; Bodon, G.; Blot, B.; Haase, G.; Goldberg, Y.; Sadoul, R. Release of exosomes from differentiated neurons and its regulation by synaptic glutamatergic activity. Mol. Cell. Neurosci. 2011, 46, 409–418. [Google Scholar] [CrossRef] [Green Version]
- Morel, L.; Regan, M.; Higashimori, H.; Ng, S.K.; Esau, C.; Vidensky, S.; Rothstein, J.; Yang, Y. Neuronal exosomal miRNA-dependent translational regulation of astroglial glutamate transporter GLT1. J. Biol. Chem. 2013, 288, 7105–7116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fröhlich, D.; Kuo, W.P.; Frühbeis, C.; Sun, J.-J.; Zehendner, C.M.; Luhmann, H.J.; Pinto, S.; Toedling, J.; Trotter, J.; Krämer-Albers, E.-M. Multifaceted effects of oligodendroglial exosomes on neurons: Impact on neuronal firing rate, signal transduction and gene regulation. Philos. Trans. R. Soc. B Biol. Sci. 2014, 369, 20130510. [Google Scholar] [CrossRef] [PubMed]
- Bakhti, M.; Winter, C.; Simons, M. Inhibition of myelin membrane sheath formation by oligodendrocyte-derived exosome-like vesicles. J. Biol. Chem. 2011, 286, 787–796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lopez-Verrilli, M.A.; Picou, F.; Court, F.A. Schwann cell-derived exosomes enhance axonal regeneration in the peripheral nervous system. Glia 2013, 61, 1795–1806. [Google Scholar] [CrossRef]
- Antonucci, F.; Turola, E.; Riganti, L.; Caleo, M.; Gabrielli, M.; Perrotta, C.; Novellino, L.; Clementi, E.; Giussani, P.; Viani, P. Microvesicles released from microglia stimulate synaptic activity via enhanced sphingolipid metabolism. EMBO J. 2012, 31, 1231–1240. [Google Scholar] [CrossRef]
- Biro, E.; Sturk-Maquelin, K.; Vogel, G.; Meuleman, D.; Smit, M.; Hack, C.; Sturk, A.; Nieuwland, R. Human cell-derived microparticles promote thrombus formation in vivo in a tissue factor-dependent manner. J. Thromb. Haemost. 2003, 1, 2561–2568. [Google Scholar] [CrossRef] [Green Version]
- Taraboletti, G.; D’Ascenzo, S.; Borsotti, P.; Giavazzi, R.; Pavan, A.; Dolo, V. Shedding of the matrix metalloproteinases MMP-2, MMP-9, and MT1-MMP as membrane vesicle-associated components by endothelial cells. Am. J. Pathol. 2002, 160, 673–680. [Google Scholar] [CrossRef] [Green Version]
- Brill, A.; Dashevsky, O.; Rivo, J.; Gozal, Y.; Varon, D. Platelet-derived microparticles induce angiogenesis and stimulate post-ischemic revascularization. Cardiovasc. Res. 2005, 67, 30–38. [Google Scholar] [CrossRef]
- Mu, W.; Rana, S.; Zöller, M. Host matrix modulation by tumor exosomes promotes motility and invasiveness. Neoplasia 2013, 15, 875-IN4. [Google Scholar] [CrossRef] [Green Version]
- Ma, Z.; Wang, Y.; Li, H. Applications of extracellular vesicles in tissue regeneration. Biomicrofluidics 2020, 14, 011501. [Google Scholar] [CrossRef]
- Klymenko, Y.; Nephew, K.P. Epigenetic Crosstalk between the Tumor Microenvironment and Ovarian Cancer Cells: A Therapeutic Road Less Traveled. Cancers (Basel) 2018, 10, 295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jahanban-Esfahlan, R.; Seidi, K.; Monhemi, H.; Adli, A.D.F.; Minofar, B.; Zare, P.; Farajzadeh, D.; Farajnia, S.; Behzadi, R.; Abbasi, M.M. RGD delivery of truncated coagulase to tumor vasculature affords local thrombotic activity to induce infarction of tumors in mice. Sci. Rep. 2017, 7, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baghban, R.; Roshangar, L.; Jahanban-Esfahlan, R.; Seidi, K.; Ebrahimi-Kalan, A.; Jaymand, M.; Kolahian, S.; Javaheri, T.; Zare, P. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun. Signal. 2020, 18, 1–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hanahan, D.; Coussens, L.M. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef] [Green Version]
- Frisch, J.; Angenendt, A.; Hoth, M.; Prates Roma, L.; Lis, A. STIM-Orai channels and reactive oxygen species in the tumor microenvironment. Cancers 2019, 11, 457. [Google Scholar] [CrossRef] [Green Version]
- Zhang, D.X.; Vu, L.T.; Ismail, N.N.; Le, M.T.; Grimson, A. Landscape of extracellular vesicles in the tumour microenvironment: Interactions with stromal cells and with non-cell components, and impacts on metabolic reprogramming, horizontal transfer of neoplastic traits, and the emergence of therapeutic resistance. Semin. Cancer Biol. 2021, 74, 24–44. [Google Scholar] [CrossRef]
- Berger, S.L.; Kouzarides, T.; Shiekhattar, R.; Shilatifard, A. An operational definition of epigenetics. Genes Dev. 2009, 23, 781–783. [Google Scholar] [CrossRef] [Green Version]
- Fouse, S.D.; Costello, J.F. Epigenetics of neurological cancers. Future Oncol. 2009, 5, 1615–1629. [Google Scholar] [CrossRef] [Green Version]
- Moran, B.; Silva, R.; Perry, A.S.; Gallagher, W.M. Epigenetics of malignant melanoma. Semin. Cancer Biol. 2018, 51, 80–88. [Google Scholar] [CrossRef]
- Sarkar, D.; Leung, E.Y.; Baguley, B.C.; Finlay, G.J.; Askarian-Amiri, M.E. Epigenetic regulation in human melanoma: Past and future. Epigenetics 2015, 10, 103–121. [Google Scholar] [CrossRef] [Green Version]
- Micevic, G.; Theodosakis, N.; Bosenberg, M. Aberrant DNA methylation in melanoma: Biomarker and therapeutic opportunities. Clin. Epigenetics 2017, 9, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, B.; Hodge, D.R.; Thomas, S.B.; Cherry, J.M.; Munroe, D.J.; Pompeia, C.; Xiao, W.; Farrar, W.L. Epigenetic silencing of the human nucleotide excision repair gene, hHR23B, in interleukin-6-responsive multiple myeloma KAS-6/1 cells. J. Biol. Chem. 2005, 280, 4182–4187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maio, M.; Covre, A.; Fratta, E.; Di Giacomo, A.M.; Taverna, P.; Natali, P.G.; Coral, S.; Sigalotti, L. Molecular pathways: At the crossroads of cancer epigenetics and immunotherapy. Clin. Cancer Res. 2015, 21, 4040–4047. [Google Scholar] [CrossRef] [Green Version]
- Siebenkäs, C.; Chiappinelli, K.B.; Guzzetta, A.A.; Sharma, A.; Jeschke, J.; Vatapalli, R.; Baylin, S.B.; Ahuja, N. Correction: Inhibiting DNA methylation activates cancer testis antigens and expression of the antigen processing and presentation machinery in colon and ovarian cancer cells. PLoS ONE 2020, 15, e0243944. [Google Scholar] [CrossRef] [PubMed]
- Luo, N.; Nixon, M.J.; Gonzalez-Ericsson, P.I.; Sanchez, V.; Opalenik, S.R.; Li, H.; Zahnow, C.A.; Nickels, M.L.; Liu, F.; Tantawy, M.N. DNA methyltransferase inhibition upregulates MHC-I to potentiate cytotoxic T lymphocyte responses in breast cancer. Nat. Commun. 2018, 9, 1–11. [Google Scholar] [CrossRef]
- Ratajczak, J.; Wysoczynski, M.; Hayek, F.; Janowska-Wieczorek, A.; Ratajczak, M. Membrane-derived microvesicles: Important and underappreciated mediators of cell-to-cell communication. Leukemia 2006, 20, 1487–1495. [Google Scholar] [CrossRef]
- Lu, L.; Chen, X.; Tao, H.; Xiong, W.; Jie, S.; Li, H. Regulation of the expression of zinc finger protein genes by microRNAs enriched within acute lymphoblastic leukemia-derived microvesicles. Genet. Mol. Res. 2015, 14, 11884–11895. [Google Scholar] [CrossRef]
- Liu, M.X.; Juan, L.; Ming, X.; Gao, Z.K.; Wang, X.H.; Zhang, Y.; Shang, M.H.; Yin, L.H.; Pu, Y.P.; Ran, L. miR-93-5p transferred by exosomes promotes the proliferation of esophageal cancer cells via intercellular communication by targeting PTEN. Biomed. Environ. Sci. 2018, 31, 171–185. [Google Scholar]
- Cappellesso, R.; Tinazzi, A.; Giurici, T.; Simonato, F.; Guzzardo, V.; Ventura, L.; Crescenzi, M.; Chiarelli, S.; Fassina, A. Programmed cell death 4 and micro RNA 21 inverse expression is maintained in cells and exosomes from ovarian serous carcinoma effusions. Cancer Cytopathol. 2014, 122, 685–693. [Google Scholar] [CrossRef]
- Goyal, B.; Yadav, S.R.M.; Awasthee, N.; Gupta, S.; Kunnumakkara, A.B.; Gupta, S.C. Diagnostic, prognostic, and therapeutic significance of long non-coding RNA MALAT1 in cancer. Biochim. Biophys. Acta Rev. Cancer 2021, 1875, 188502. [Google Scholar] [CrossRef]
- Brockdorff, N. Noncoding RNA and Polycomb recruitment. RNA 2013, 19, 429–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mishra, S.; Verma, S.S.; Rai, V.; Awasthee, N.; Chava, S.; Hui, K.M.; Kumar, A.P.; Challagundla, K.B.; Sethi, G.; Gupta, S.C. Long non-coding RNAs are emerging targets of phytochemicals for cancer and other chronic diseases. Cell. Mol. Life Sci. 2019, 76, 1947–1966. [Google Scholar] [CrossRef] [PubMed]
- Kogure, T.; Yan, I.K.; Lin, W.-L.; Patel, T. Extracellular vesicle–mediated transfer of a novel long noncoding RNA TUC339: A mechanism of intercellular signaling in human hepatocellular cancer. Genes Cancer 2013, 4, 261–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pan, L.; Liang, W.; Fu, M.; Huang, Z.-h.; Li, X.; Zhang, W.; Zhang, P.; Qian, H.; Jiang, P.-C.; Xu, W.-R. Exosomes-mediated transfer of long noncoding RNA ZFAS1 promotes gastric cancer progression. J. Cancer Res. Clin. Oncol. 2017, 143, 991–1004. [Google Scholar] [CrossRef]
- Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef]
- Cao, Y.L.; Zhuang, T.; Xing, B.H.; Li, N.; Li, Q. Exosomal DNMT1 mediates cisplatin resistance in ovarian cancer. Cell Biochem. Funct. 2017, 35, 296–303. [Google Scholar] [CrossRef]
- Yang, S.-J.; Wang, D.-D.; Li, J.; Xu, H.-Z.; Shen, H.-Y.; Chen, X.; Zhou, S.-Y.; Zhong, S.-L.; Zhao, J.-H.; Tang, J.-H. Predictive role of GSTP1-containing exosomes in chemotherapy-resistant breast cancer. Gene 2017, 623, 5–14. [Google Scholar] [CrossRef]
- Sousa, D.; Lima, R.T.; Vasconcelos, M.H. Intercellular transfer of cancer drug resistance traits by extracellular vesicles. Trends Mol. Med. 2015, 21, 595–608. [Google Scholar] [CrossRef]
- Skog, J.; Würdinger, T.; Van Rijn, S.; Meijer, D.H.; Gainche, L.; Curry, W.T.; Carter, B.S.; Krichevsky, A.M.; Breakefield, X.O. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat. Cell Biol. 2008, 10, 1470–1476. [Google Scholar] [CrossRef]
- Taylor, D.D.; Gercel-Taylor, C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 2008, 110, 13–21. [Google Scholar] [CrossRef]
- De Souza, P.S.; Cruz, A.L.; Viola, J.P.; Maia, R.C. Microparticles induce multifactorial resistance through oncogenic pathways independently of cancer cell type. Cancer Sci. 2015, 106, 60–68. [Google Scholar] [CrossRef] [PubMed]
- Wu, H.; Zhou, J.; Mei, S.; Wu, D.; Mu, Z.; Chen, B.; Xie, Y.; Ye, Y.; Liu, J. Circulating exosomal microRNA-96 promotes cell proliferation, migration and drug resistance by targeting LMO7. J. Cell. Mol. Med. 2017, 21, 1228–1236. [Google Scholar] [CrossRef]
- Yu, D.-D.; Wu, Y.; Zhang, X.-H.; Lv, M.-M.; Chen, W.-X.; Chen, X.; Yang, S.-J.; Shen, H.; Zhong, S.-L.; Tang, J.-H. Exosomes from adriamycin-resistant breast cancer cells transmit drug resistance partly by delivering miR-222. Tumor Biol. 2016, 37, 3227–3235. [Google Scholar] [CrossRef] [PubMed]
- Corcoran, C.; Friel, A.M.; Duffy, M.J.; Crown, J.; O’Driscoll, L. Intracellular and extracellular microRNAs in breast cancer. Clin. Chem. 2011, 57, 18–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wei, Y.; Lai, X.; Yu, S.; Chen, S.; Ma, Y.; Zhang, Y.; Li, H.; Zhu, X.; Yao, L.; Zhang, J. Exosomal miR-221/222 enhances tamoxifen resistance in recipient ER-positive breast cancer cells. Breast Cancer Res. Treat. 2014, 147, 423–431. [Google Scholar] [CrossRef] [PubMed]
- Gezer, U.; Özgür, E.; Cetinkaya, M.; Isin, M.; Dalay, N. Long non-coding RNAs with low expression levels in cells are enriched in secreted exosomes. Cell Biol. Int. 2014, 38, 1076–1079. [Google Scholar] [CrossRef] [PubMed]
- Biswas, S.; Guix, M.; Rinehart, C.; Dugger, T.C.; Chytil, A.; Moses, H.L.; Freeman, M.L.; Arteaga, C.L. Inhibition of TGF-β with neutralizing antibodies prevents radiation-induced acceleration of metastatic cancer progression. J. Clin. Investig. 2007, 117, 1305–1313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takahashi, K.; Yan, I.K.; Kogure, T.; Haga, H.; Patel, T. Extracellular vesicle-mediated transfer of long non-coding RNA ROR modulates chemosensitivity in human hepatocellular cancer. FEBS Open Bio 2014, 4, 458–467. [Google Scholar] [CrossRef] [Green Version]
- Takahashi, K.; Yan, I.K.; Wood, J.; Haga, H.; Patel, T. Involvement of extracellular vesicle long noncoding RNA (linc-VLDLR) in tumor cell responses to chemotherapy. Mol. Cancer Res. 2014, 12, 1377–1387. [Google Scholar] [CrossRef] [Green Version]
- Qu, L.; Ding, J.; Chen, C.; Wu, Z.-J.; Liu, B.; Gao, Y.; Chen, W.; Liu, F.; Sun, W.; Li, X.-F. Exosome-transmitted lncARSR promotes sunitinib resistance in renal cancer by acting as a competing endogenous RNA. Cancer Cell 2016, 29, 653–668. [Google Scholar] [CrossRef]
- Wang, J.; Lv, B.; Su, Y.; Wang, X.; Bu, J.; Yao, L. Exosome-mediated transfer of lncRNA HOTTIP promotes cisplatin resistance in gastric cancer cells by regulating HMGA1/miR-218 axis. OncoTargets Ther. 2019, 12, 11325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stoner, S.A.; Duggan, E.; Condello, D.; Guerrero, A.; Turk, J.R.; Narayanan, P.K.; Nolan, J.P. High sensitivity flow cytometry of membrane vesicles. Cytom. Part A 2016, 89, 196–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Lellis, L.; Florio, R.; Di Bella, M.C.; Brocco, D.; Guidotti, F.; Tinari, N.; Grassadonia, A.; Lattanzio, R.; Cama, A.; Veschi, S. Exosomes as pleiotropic players in pancreatic cancer. Biomedicines 2021, 9, 275. [Google Scholar] [CrossRef] [PubMed]
- Siveen, K.S.; Raza, A.; Ahmed, E.I.; Khan, A.Q.; Prabhu, K.S.; Kuttikrishnan, S.; Mateo, J.M.; Zayed, H.; Rasul, K.; Azizi, F. The role of extracellular vesicles as modulators of the tumor microenvironment, metastasis and drug resistance in colorectal cancer. Cancers 2019, 11, 746. [Google Scholar] [CrossRef] [Green Version]
- Rackov, G.; Garcia-Romero, N.; Esteban-Rubio, S.; Carrión-Navarro, J.; Belda-Iniesta, C.; Ayuso-Sacido, A. Vesicle-mediated control of cell function: The role of extracellular matrix and microenvironment. Front. Physiol. 2018, 9, 651. [Google Scholar] [CrossRef] [PubMed]
- Tang, T.-T.; Wang, B.; Wu, M.; Li, Z.-L.; Feng, Y.; Cao, J.-Y.; Yin, D.; Liu, H.; Tang, R.-N.; Crowley, S.D. Extracellular vesicle–encapsulated IL-10 as novel nanotherapeutics against ischemic AKI. Sci. Adv. 2020, 6, eaaz0748. [Google Scholar] [CrossRef] [PubMed]
- Arkhypov, I.; Lasser, S.; Petrova, V.; Weber, R.; Groth, C.; Utikal, J.; Altevogt, P.; Umansky, V. Myeloid cell modulation by tumor-derived extracellular vesicles. Int. J. Mol. Sci. 2020, 21, 6319. [Google Scholar] [CrossRef]
- Pavlyukov, M.S.; Yu, H.; Bastola, S.; Minata, M.; Shender, V.O.; Lee, Y.; Zhang, S.; Wang, J.; Komarova, S.; Wang, J. Apoptotic cell-derived extracellular vesicles promote malignancy of glioblastoma via intercellular transfer of splicing factors. Cancer Cell 2018, 34, 119–135. [Google Scholar] [CrossRef] [Green Version]
- Vella, L.J.; Behren, A.; Coleman, B.; Greening, D.W.; Hill, A.F.; Cebon, J. Intercellular resistance to BRAF inhibition can be mediated by extracellular vesicle–associated PDGFRβ. Neoplasia 2017, 19, 932–940. [Google Scholar] [CrossRef]
- Brzozowski, J.S.; Bond, D.R.; Jankowski, H.; Goldie, B.J.; Burchell, R.; Naudin, C.; Smith, N.D.; Scarlett, C.J.; Larsen, M.R.; Dun, M.D. Extracellular vesicles with altered tetraspanin CD9 and CD151 levels confer increased prostate cell motility and invasion. Sci. Rep. 2018, 8, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Fu, Q.; Zhang, Q.; Lou, Y.; Yang, J.; Nie, G.; Chen, Q.; Chen, Y.; Zhang, J.; Wang, J.; Wei, T. Correction: Primary tumor-derived exosomes facilitate metastasis by regulating adhesion of circulating tumor cells via SMAD3 in liver cancer. Oncogene 2019, 38, 5740–5741. [Google Scholar] [CrossRef] [Green Version]
- Dörsam, B.; Bösl, T.; Reiners, K.S.; Barnert, S.; Schubert, R.; Shatnyeva, O.; Zigrino, P.; Engert, A.; Hansen, H.P.; von Strandmann, E.P. Hodgkin lymphoma-derived extracellular vesicles change the secretome of fibroblasts toward a CAF phenotype. Front. Immunol. 2018, 9, 1358. [Google Scholar] [CrossRef] [PubMed]
- Ning, X.; Zhang, H.; Wang, C.; Song, X. Exosomes released by gastric cancer cells induce transition of pericytes into cancer-associated 104. fibroblasts. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2018, 24, 2350. [Google Scholar]
- Zhou, W.; Fong, M.Y.; Min, Y.; Somlo, G.; Liu, L.; Palomares, M.R.; Yu, Y.; Chow, A.; O’Connor, S.T.F.; Chin, A.R. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell 2014, 25, 501–515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adair, T.H.; Montani, J.-P. Angiogenesis. In Proceedings of the Colloquium Series on Integrated Systems Physiology: From Molecule to Function; Morgan & Claypool Life Sciences: San Rafael, CA, USA, 2010; pp. 1–84. [Google Scholar]
- Nazarenko, I.; Rana, S.; Baumann, A.; McAlear, J.; Hellwig, A.; Trendelenburg, M.; Lochnit, G.; Preissner, K.T.; Zöller, M. Cell surface tetraspanin Tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation. Cancer Res. 2010, 70, 1668–1678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhuang, G.; Wu, X.; Jiang, Z.; Kasman, I.; Yao, J.; Guan, Y.; Oeh, J.; Modrusan, Z.; Bais, C.; Sampath, D. Tumour-secreted miR-9 promotes endothelial cell migration and angiogenesis by activating the JAK-STAT pathway. EMBO J. 2012, 31, 3513–3523. [Google Scholar] [CrossRef]
- Li, B.; Hong, J.; Hong, M.; Wang, Y.; Yu, T.; Zang, S.; Wu, Q. piRNA-823 delivered by multiple myeloma-derived extracellular vesicles promoted tumorigenesis through re-educating endothelial cells in the tumor environment. Oncogene 2019, 38, 5227–5238. [Google Scholar] [CrossRef]
- Masoumi-Dehghi, S.; Babashah, S.; Sadeghizadeh, M. microRNA-141-3p-containing small extracellular vesicles derived from epithelial ovarian cancer cells promote endothelial cell angiogenesis through activating the JAK/STAT3 and NF-κB signaling pathways. J. Cell Commun. Signal. 2020, 14, 233–244. [Google Scholar] [CrossRef]
- Kosaka, N.; Iguchi, H.; Hagiwara, K.; Yoshioka, Y.; Takeshita, F.; Ochiya, T. Neutral sphingomyelinase 2 (nSMase2)-dependent exosomal transfer of angiogenic microRNAs regulate cancer cell metastasis. J. Biol. Chem. 2013, 288, 10849–10859. [Google Scholar] [CrossRef] [Green Version]
- Kosaka, N.; Yoshioka, Y.; Fujita, Y.; Ochiya, T. Versatile roles of extracellular vesicles in cancer. J. Clin. Investig. 2016, 126, 1163–1172. [Google Scholar] [CrossRef] [Green Version]
- Cano, R.L.E.; Lopera, H.D.E. Introduction to T and B lymphocytes. In Autoimmunity: From Bench to Bedside; El Rosario University Press: Bogota, Colombia, 2013. [Google Scholar]
- Giusti, I.; Di Francesco, M.; D’Ascenzo, S.; Palmerini, M.G.; Macchiarelli, G.; Carta, G.; Dolo, V. Ovarian cancer-derived extracellular vesicles affect normal human fibroblast behavior. Cancer Biol. Ther. 2018, 19, 722–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, Y.; Ren, H.; Dai, B.; Li, J.; Shang, L.; Huang, J.; Shi, X. Hepatocellular carcinoma-derived exosomal miRNA-21 contributes to tumor progression by converting hepatocyte stellate cells to cancer-associated fibroblasts. J. Exp. Clin. Cancer Res. 2018, 37, 1–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pang, W.; Su, J.; Wang, Y.; Feng, H.; Dai, X.; Yuan, Y.; Chen, X.; Yao, W. Pancreatic cancer-secreted miR-155 implicates in the conversion from normal fibroblasts to cancer-associated fibroblasts. Cancer Sci. 2015, 106, 1362–1369. [Google Scholar] [CrossRef]
- Lawson, J.; Dickman, C.; Towle, R.; Jabalee, J.; Javer, A.; Garnis, C. Extracellular vesicle secretion of miR-142-3p from lung adenocarcinoma cells induces tumor promoting changes in the stroma through cell-cell communication. Mol. Carcinog. 2019, 58, 376–387. [Google Scholar] [CrossRef] [PubMed]
- Snyder, A.; Makarov, V.; Merghoub, T.; Yuan, J.; Zaretsky, J.M.; Desrichard, A.; Walsh, L.A.; Postow, M.A.; Wong, P.; Ho, T.S. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 2014, 371, 2189–2199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B. Safety, activity, and immune correlates of anti–PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef]
- Pucci, F.; Garris, C.; Lai, C.P.; Newton, A.; Pfirschke, C.; Engblom, C.; Alvarez, D.; Sprachman, M.; Evavold, C.; Magnuson, A. SCS macrophages suppress melanoma by restricting tumor-derived vesicle–B cell interactions. Science 2016, 352, 242–246. [Google Scholar] [CrossRef] [Green Version]
- Kanlikilicer, P.; Bayraktar, R.; Denizli, M.; Rashed, M.H.; Ivan, C.; Aslan, B.; Mitra, R.; Karagoz, K.; Bayraktar, E.; Zhang, X. 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]
- Cooks, T.; Pateras, I.S.; Jenkins, L.M.; Patel, K.M.; Robles, A.I.; Morris, J.; Forshew, T.; Appella, E.; Gorgoulis, V.G.; Harris, C.C. Mutant p53 cancers reprogram macrophages to tumor supporting macrophages via exosomal miR-1246. Nat. Commun. 2018, 9, 1–15. [Google Scholar] [CrossRef] [Green Version]
- Casadei, L.; Calore, F.; Creighton, C.J.; Guescini, M.; Batte, K.; Iwenofu, O.H.; Zewdu, A.; Braggio, D.A.; Bill, K.L.; Fadda, P. Exosome-derived miR-25-3p and miR-92a-3p stimulate liposarcoma progression. Cancer Res. 2017, 77, 3846–3856. [Google Scholar] [CrossRef] [Green Version]
- Chen, X.; Zhou, J.; Li, X.; Wang, X.; Lin, Y.; Wang, X. Exosomes derived from hypoxic epithelial ovarian cancer cells deliver microRNAs to macrophages and elicit a tumor-promoted phenotype. Cancer Lett. 2018, 435, 80–91. [Google Scholar] [CrossRef] [PubMed]
- Van der Vos, K.E.; Abels, E.R.; Zhang, X.; Lai, C.; Carrizosa, E.; Oakley, D.; Prabhakar, S.; Mardini, O.; Crommentuijn, M.H.; Skog, J. Directly visualized glioblastoma-derived extracellular vesicles transfer RNA to microglia/macrophages in the brain. Neuro-Oncology 2015, 18, 58–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Viaud, S.; Terme, M.; Flament, C.; Taieb, J.; Andre, F.; Novault, S.; Escudier, B.; Robert, C.; Caillat-Zucman, S.; Tursz, T. Dendritic cell-derived exosomes promote natural killer cell activation and proliferation: A role for NKG2D ligands and IL-15Rα. PLoS ONE 2009, 4, e4942. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Yuan, X.; Shi, H.; Wu, L.; Qian, H.; Xu, W. Exosomes in cancer: Small particle, big player. J. Hematol. Oncol. 2015, 8, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Czernek, L.; Düchler, M. Functions of cancer-derived extracellular vesicles in immunosuppression. Arch. Immunol. Ther. Exp. 2017, 65, 311–323. [Google Scholar] [CrossRef] [Green Version]
- Szczepanski, M.J.; Szajnik, M.; Welsh, A.; Whiteside, T.L.; Boyiadzis, M. Blast-derived microvesicles in sera from patients with acute myeloid leukemia suppress natural killer cell function via membrane-associated transforming growth factor-β1. Haematologica 2011, 96, 1302. [Google Scholar] [CrossRef]
- Bubeník, J. MHC class I down-regulation: Tumour escape from immune surveillance? Int. J. Oncol. 2004, 25, 487–491. [Google Scholar] [CrossRef]
- Ljunggren, H.-G.; Kärre, K. In search of the ‘missing self’: MHC molecules and NK cell recognition. Immunol. Today 1990, 11, 237–244. [Google Scholar] [CrossRef]
- Clayton, A.; Mitchell, J.P.; Linnane, S.; Mason, M.D.; Tabi, Z. Human tumor-derived exosomes down-modulate NKG2D expression. J. Immunol. 2008, 180, 7249–7258. [Google Scholar] [CrossRef] [Green Version]
- Hedlund, M.; Nagaeva, O.; Kargl, D.; Baranov, V.; Mincheva-Nilsson, L. Thermal-and oxidative stress causes enhanced release of NKG2D ligand-bearing immunosuppressive exosomes in leukemia/lymphoma T and B cells. PLoS ONE 2011, 6, e16899. [Google Scholar] [CrossRef]
- Dörsam, B.; Reiners, K.S.; von Strandmann, E.P. Cancer-derived extracellular vesicles: Friend and foe of tumour immunosurveillance. Philos. Trans. R. Soc. B Biol. Sci. 2018, 373, 20160481. [Google Scholar] [CrossRef]
- Viaud, S.; Théry, C.; Ploix, S.; Tursz, T.; Lapierre, V.; Lantz, O.; Zitvogel, L.; Chaput, N. Dendritic cell-derived exosomes for cancer immunotherapy: What’s next? Cancer Res. 2010, 70, 1281–1285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lamparski, H.G.; Metha-Damani, A.; Yao, J.-Y.; Patel, S.; Hsu, D.-H.; Ruegg, C.; Le Pecq, J.-B. Production and characterization of clinical grade exosomes derived from dendritic cells. J. Immunol. Methods 2002, 270, 211–226. [Google Scholar] [CrossRef]
- Zeng, A.; Yan, W.; Liu, Y.; Wang, Z.; Hu, Q.; Nie, E.; Zhou, X.; Li, R.; Wang, X.; Jiang, T. Tumour exosomes from cells harbouring PTPRZ1–MET fusion contribute to a malignant phenotype and temozolomide chemoresistance in glioblastoma. Oncogene 2017, 36, 5369–5381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Crow, J.; Atay, S.; Banskota, S.; Artale, B.; Schmitt, S.; Godwin, A.K. Exosomes as mediators of platinum resistance in ovarian cancer. Oncotarget 2017, 8, 11917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, X.J.; Ren, Z.J.; Tang, J.H.; Yu, Q. Exosomal MicroRNA MiR-1246 promotes cell proliferation, invasion and drug resistance by targeting CCNG2 in breast cancer. Cell. Physiol. Biochem. 2017, 44, 1741–1748. [Google Scholar] [CrossRef]
- Gregory, P.A.; Bert, A.G.; Paterson, E.L.; Barry, S.C.; Tsykin, A.; Farshid, G.; Vadas, M.A.; Khew-Goodall, Y.; Goodall, G.J. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat. Cell Biol. 2008, 10, 593–601. [Google Scholar] [CrossRef]
- Campos, A.; Salomon, C.; Bustos, R.; Díaz, J.; Martínez, S.; Silva, V.; Reyes, C.; Díaz-Valdivia, N.; Varas-Godoy, M.; Lobos-González, L. Caveolin-1-containing extracellular vesicles transport adhesion proteins and promote malignancy in breast cancer cell lines. Nanomedicine 2018, 13, 2597–2609. [Google Scholar] [CrossRef] [Green Version]
- Fong, M.Y.; Zhou, W.; Liu, L.; Alontaga, A.Y.; Chandra, M.; Ashby, J.; Chow, A.; O’Connor, S.T.F.; Li, S.; Chin, A.R. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat. Cell Biol. 2015, 17, 183–194. [Google Scholar] [CrossRef] [Green Version]
- Kitdumrongthum, S.; Metheetrairut, C.; Charoensawan, V.; Ounjai, P.; Janpipatkul, K.; Panvongsa, W.; Weerachayaphorn, J.; Piyachaturawat, P.; Chairoungdua, A. Dysregulated microRNA expression profiles in cholangiocarcinoma cell-derived exosomes. Life Sci. 2018, 210, 65–75. [Google Scholar] [CrossRef]
- Teng, Y.; Ren, Y.; Hu, X.; Mu, J.; Samykutty, A.; Zhuang, X.; Deng, Z.; Kumar, A.; Zhang, L.; Merchant, M.L. MVP-mediated exosomal sorting of miR-193a promotes colon cancer progression. Nat. Commun. 2017, 8, 1–16. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Xing, T.; Chen, Y.; Xiao, J. Exosome-mediated miR-200b promotes colorectal cancer proliferation upon TGF-β1 exposure. Biomed. Pharmacother. 2018, 106, 1135–1143. [Google Scholar] [CrossRef] [PubMed]
- Zeng, Z.; Li, Y.; Pan, Y.; Lan, X.; Song, F.; Sun, J.; Zhou, K.; Liu, X.; Ren, X.; Wang, F. Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nat. Commun. 2018, 9, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Harada, T.; Yamamoto, H.; Kishida, S.; Kishida, M.; Awada, C.; Takao, T.; Kikuchi, A. Wnt5b-associated exosomes promote cancer cell migration and proliferation. Cancer Sci. 2017, 108, 42–52. [Google Scholar] [CrossRef]
- Higginbotham, J.N.; Beckler, M.D.; Gephart, J.D.; Franklin, J.L.; Bogatcheva, G.; Kremers, G.-J.; Piston, D.W.; Ayers, G.D.; McConnell, R.E.; Tyska, M.J. Amphiregulin exosomes increase cancer cell invasion. Curr. Biol. 2011, 21, 779–786. [Google Scholar] [CrossRef] [Green Version]
- Kalra, H.; Gangoda, L.; Fonseka, P.; Chitti, S.V.; Liem, M.; Keerthikumar, S.; Samuel, M.; Boukouris, S.; Al Saffar, H.; Collins, C. Extracellular vesicles containing oncogenic mutant β-catenin activate Wnt signalling pathway in the recipient cells. J. Extracell. Vesicles 2019, 8, 1690217. [Google Scholar] [CrossRef] [Green Version]
- Muturi, H.T.; Dreesen, J.D.; Nilewski, E.; Jastrow, H.; Giebel, B.; Ergun, S.; Singer, B.B. Tumor and endothelial cell-derived microvesicles carry distinct CEACAMs and influence T-cell behavior. PLoS ONE 2013, 8, e74654. [Google Scholar] [CrossRef] [Green Version]
- Dai, G.; Yao, X.; Zhang, Y.; Gu, J.; Geng, Y.; Xue, F.; Zhang, J. Colorectal cancer cell–derived exosomes containing miR-10b regulate fibroblast cells via the PI3K/Akt pathway. Bull. Cancer 2018, 105, 336–349. [Google Scholar] [CrossRef]
- Guo, K.; Yao, J.; Yu, Q.; Li, Z.; Huang, H.; Cheng, J.; Wang, Z.; Zhu, Y. The expression pattern of long non-coding RNA PVT1 in tumor tissues and in extracellular vesicles of colorectal cancer correlates with cancer progression. Tumor Biol. 2017, 39, 1010428317699122. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Zhou, M.; Bai, L.; Han, R.; Lv, K.; Wang, Z. Extracellular vesicles promote esophageal cancer progression by delivering lncZEB1-AS1 between cells. Eur. Rev. Med. Pharm. Sci. 2018, 22, 2662–2670. [Google Scholar]
- Yang, H.; Fu, H.; Wang, B.; Zhang, X.; Mao, J.; Li, X.; Wang, M.; Sun, Z.; Qian, H.; Xu, W. Exosomal miR-423-5p targets SUFU to promote cancer growth and metastasis and serves as a novel marker for gastric cancer. Mol. Carcinog. 2018, 57, 1223–1236. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Guan, X.; Zhang, Y.; Ge, S.; Zhang, L.; Li, H.; Wang, X.; Liu, R.; Ning, T.; Deng, T. Exosomal miR-27a derived from gastric cancer cells regulates the transformation of fibroblasts into cancer-associated fibroblasts. Cell. Physiol. Biochem. 2018, 49, 869–883. [Google Scholar] [CrossRef]
- Setti, M.; Osti, D.; Richichi, C.; Ortensi, B.; Del Bene, M.; Fornasari, L.; Beznoussenko, G.; Mironov, A.; Rappa, G.; Cuomo, A. Extracellular vesicle-mediated transfer of CLIC1 protein is a novel mechanism for the regulation of glioblastoma growth. Oncotarget 2015, 6, 31413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lang, H.-L.; Hu, G.-W.; Zhang, B.; Kuang, W.; Chen, Y.; Wu, L.; Xu, G.-H. Glioma cells enhance angiogenesis and inhibit endothelial cell apoptosis through the release of exosomes that contain long non-coding RNA CCAT2. Oncol. Rep. 2017, 38, 785–798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lang, H.; Hu, G.; Chen, Y.; Liu, Y.; Tu, W.; Lu, Y.; Wu, L.; Xu, G. Glioma cells promote angiogenesis through the release of exosomes containing long non-coding RNA POU3F3. Eur. Rev. Med. Pharm. Sci. 2017, 21, 959–972. [Google Scholar]
- Sun, X.; Ma, X.; Wang, J.; Zhao, Y.; Wang, Y.; Bihl, J.C.; Chen, Y.; Jiang, C. Glioma stem cells-derived exosomes promote the angiogenic ability of endothelial cells through miR-21/VEGF signal. Oncotarget 2017, 8, 36137. [Google Scholar] [CrossRef] [Green Version]
- Giusti, I.; Delle Monache, S.; Di Francesco, M.; Sanità, P.; D’Ascenzo, S.; Gravina, G.L.; Festuccia, C.; Dolo, V. From glioblastoma to endothelial cells through extracellular vesicles: Messages for angiogenesis. Tumor. Biol. 2016, 37, 12743–12753. [Google Scholar] [CrossRef]
- Cai, Q.; Zhu, A.; Gong, L. Exosomes of glioma cells deliver miR-148a to promote proliferation and metastasis of glioblastoma via targeting CADM1. Bull. Cancer 2018, 105, 643–651. [Google Scholar] [CrossRef]
- Al-Nedawi, K.; Meehan, B.; Micallef, J.; Lhotak, V.; May, L.; Guha, A.; Rak, J. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat. Cell Biol. 2008, 10, 619–624. [Google Scholar] [CrossRef]
- Ricklefs, F.L.; Alayo, Q.; Krenzlin, H.; Mahmoud, A.B.; Speranza, M.C.; Nakashima, H.; Hayes, J.L.; Lee, K.; Balaj, L.; Passaro, C. Immune evasion mediated by PD-L1 on glioblastoma-derived extracellular vesicles. Sci. Adv. 2018, 4, eaar2766. [Google Scholar] [CrossRef] [Green Version]
- Sato, S.; Vasaikar, S.; Eskaros, A.; Kim, Y.; Lewis, J.S.; Zhang, B.; Zijlstra, A.; Weaver, A.M. EPHB2 carried on small extracellular vesicles induces tumor angiogenesis via activation of ephrin reverse signaling. JCI Insight 2019, 4. [Google Scholar] [CrossRef] [PubMed]
- Qu, Z.; Feng, J.; Pan, H.; Jiang, Y.; Duan, Y.; Fa, Z. Exosomes derived from HCC cells with different invasion characteristics mediated EMT through TGF-β/Smad signaling pathway. OncoTargets Ther. 2019, 12, 6897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xue, X.; Wang, X.; Zhao, Y.; Hu, R.; Qin, L. Exosomal miR-93 promotes proliferation and invasion in hepatocellular carcinoma by directly inhibiting TIMP2/TP53INP1/CDKN1A. Biochem. Biophys. Res. Commun. 2018, 502, 515–521. [Google Scholar] [CrossRef] [PubMed]
- Fang, J.H.; Zhang, Z.J.; Shang, L.R.; Luo, Y.W.; Lin, Y.F.; Yuan, Y.; Zhuang, S.M. Hepatoma cell-secreted exosomal microRNA-103 increases vascular permeability and promotes metastasis by targeting junction proteins. Hepatology 2018, 68, 1459–1475. [Google Scholar] [CrossRef] [Green Version]
- Kogure, T.; Lin, W.L.; Yan, I.K.; Braconi, C.; Patel, T. Intercellular nanovesicle-mediated microRNA transfer: A mechanism of environmental modulation of hepatocellular cancer cell growth. Hepatology 2011, 54, 1237–1248. [Google Scholar] [CrossRef] [Green Version]
- Huang, A.; Dong, J.; Li, S.; Wang, C.; Ding, H.; Li, H.; Su, X.; Ge, X.; Sun, L.; Bai, C. Exosomal transfer of vasorin expressed in hepatocellular carcinoma cells promotes migration of human umbilical vein endothelial cells. Int. J. Biol. Sci. 2015, 11, 961. [Google Scholar] [CrossRef]
- Lawson, J.; Dickman, C.; MacLellan, S.; Towle, R.; Jabalee, J.; Lam, S.; Garnis, C. Selective secretion of microRNAs from lung cancer cells via extracellular vesicles promotes CAMK1D-mediated tube formation in endothelial cells. Oncotarget 2017, 8, 83913. [Google Scholar] [CrossRef] [Green Version]
- Zheng, Y.; Liu, L.; Chen, C.; Ming, P.; Huang, Q.; Li, C.; Cao, D.; Xu, X.; Ge, W. The extracellular vesicles secreted by lung cancer cells in radiation therapy promote endothelial cell angiogenesis by transferring miR-23a. PeerJ 2017, 5, e3627. [Google Scholar] [CrossRef] [Green Version]
- Hamid, O.; Robert, C.; Daud, A.; Hodi, F.S.; Hwu, W.-J.; Kefford, R.; Wolchok, J.D.; Hersey, P.; Joseph, R.W.; Weber, J.S. Safety and tumor responses with lambrolizumab (anti–PD-1) in melanoma. N. Engl. J. Med. 2013, 369, 134–144. [Google Scholar] [CrossRef] [Green Version]
- Cesi, G.; Philippidou, D.; Kozar, I.; Kim, Y.J.; Bernardin, F.; Van Niel, G.; Wienecke-Baldacchino, A.; Felten, P.; Letellier, E.; Dengler, S. A new ALK isoform transported by extracellular vesicles confers drug resistance to melanoma cells. Mol. Cancer 2018, 17, 1–14. [Google Scholar] [CrossRef]
- Zhou, X.; Yan, T.; Huang, C.; Xu, Z.; Wang, L.; Jiang, E.; Wang, H.; Chen, Y.; Liu, K.; Shao, Z. Melanoma cell-secreted exosomal miR-155-5p induce proangiogenic switch of cancer-associated fibroblasts via SOCS1/JAK2/STAT3 signaling pathway. J. Exp. Clin. Cancer Res. 2018, 37, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yan, H.; Wu, Q.L.; Sun, C.Y.; Ai, C.-Y.; Deng, J.; Zhang, L.; Chen, L.; Chu, Z.-B.; Tang, B.; Wang, K.; et al. piRNA-823 contributes to tumorigenesis by regulating de novo DNA methylation and angiogenesis in multiple myeloma. Leukemia 2015, 29, 196–206. [Google Scholar] [CrossRef] [PubMed]
- Bao, L.; You, B.; Shi, S.; Shan, Y.; Zhang, Q.; Yue, H.; Zhang, J.; Zhang, W.; Shi, Y.; Liu, Y. Metastasis-associated miR-23a from nasopharyngeal carcinoma-derived exosomes mediates angiogenesis by repressing a novel target gene TSGA10. Oncogene 2018, 37, 2873–2889. [Google Scholar] [CrossRef] [PubMed]
- Ye, S.-B.; Li, Z.-L.; Luo, D.-H.; Huang, B.-J.; Chen, Y.-S.; Zhang, X.-S.; Cui, J.; Zeng, Y.-X.; Li, J. Tumor-derived exosomes promote tumor progression and T-cell dysfunction through the regulation of enriched exosomal microRNAs in human nasopharyngeal carcinoma. Oncotarget 2014, 5, 5439. [Google Scholar] [CrossRef] [Green Version]
- Klibi, J.; Niki, T.; Riedel, A.; Pioche-Durieu, C.; Souquere, S.; Rubinstein, E.; Le Moulec, S.; Guigay, J.; Hirashima, M.; Guemira, F. Blood diffusion and Th1-suppressive effects of galectin-9–containing exosomes released by Epstein-Barr virus–infected nasopharyngeal carcinoma cells. Blood J. Am. Soc. Hematol. 2009, 113, 1957–1966. [Google Scholar] [CrossRef] [Green Version]
- Dickman, C.T.; Lawson, J.; Jabalee, J.; MacLellan, S.A.; LePard, N.E.; Bennewith, K.L.; Garnis, C. Selective extracellular vesicle exclusion of miR-142-3p by oral cancer cells promotes both internal and extracellular malignant phenotypes. Oncotarget 2017, 8, 15252. [Google Scholar] [CrossRef] [Green Version]
- Gong, L.; Bao, Q.; Hu, C.; Wang, J.; Zhou, Q.; Wei, L.; Tong, L.; Zhang, W.; Shen, Y. Exosomal miR-675 from metastatic osteosarcoma promotes cell migration and invasion by targeting CALN1. Biochem. Biophys. Res. Commun. 2018, 500, 170–176. [Google Scholar] [CrossRef]
- Yoshimura, A.; Sawada, K.; Nakamura, K.; Kinose, Y.; Nakatsuka, E.; Kobayashi, M.; Miyamoto, M.; Ishida, K.; Matsumoto, Y.; Kodama, M. Exosomal miR-99a-5p is elevated in sera of ovarian cancer patients and promotes cancer cell invasion by increasing fibronectin and vitronectin expression in neighboring peritoneal mesothelial cells. BMC Cancer 2018, 18, 1–13. [Google Scholar] [CrossRef]
- Czystowska-Kuzmicz, M.; Sosnowska, A.; Nowis, D.; Ramji, K.; Szajnik, M.; Chlebowska-Tuz, J.; Wolinska, E.; Gaj, P.; Grazul, M.; Pilch, Z. Small extracellular vesicles containing arginase-1 suppress T-cell responses and promote tumor growth in ovarian carcinoma. Nat. Commun. 2019, 10, 1–16. [Google Scholar] [CrossRef]
- Chen, D.; Wu, X.; Xia, M.; Wu, F.; Ding, J.; Jiao, Y.; Zhan, Q.; An, F. Upregulated exosomic miR-23b-3p plays regulatory roles in the progression of pancreatic cancer. Oncol. Rep. 2017, 38, 2182–2188. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.C.; Zhao, J.-T.; Gundara, J.; Serpell, J.; Bach, L.A.; Sidhu, S. Papillary thyroid cancer–derived exosomes contain miRNA-146b and miRNA-222. J. Surg. Res. 2015, 196, 39–48. [Google Scholar] [CrossRef] [PubMed]
- Bhagirath, D.; Yang, T.L.; Bucay, N.; Sekhon, K.; Majid, S.; Shahryari, V.; Dahiya, R.; Tanaka, Y.; Saini, S. microRNA-1246 is an exosomal biomarker for aggressive prostate cancer. Cancer Res. 2018, 78, 1833–1844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, L.; Wu, X.; Wang, D.; Luo, C.; Chen, L. Renal carcinoma cell-derived exosomes induce human immortalized line of Jurkat T lymphocyte apoptosis in vitro. Urol. Int. 2013, 91, 363–369. [Google Scholar] [CrossRef]
- Butler, J.T.; Abdelhamed, S.; Kurre, P. Extracellular vesicles in the hematopoietic microenvironment. Haematologica 2018, 103, 382. [Google Scholar] [CrossRef] [PubMed]
- Nehrbas, J.; Butler, J.T.; Chen, D.-W.; Kurre, P. Extracellular vesicles and chemotherapy resistance in the AML microenvironment. Front. Oncol. 2020, 10, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patel, G.K.; Khan, M.A.; Bhardwaj, A.; Srivastava, S.K.; Zubair, H.; Patton, M.C.; Singh, S.; Singh, A.P. Exosomes confer chemoresistance to pancreatic cancer cells by promoting ROS detoxification and miR-155-mediated suppression of key gemcitabine-metabolising enzyme, DCK. Br. J. Cancer 2017, 116, 609–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wojtuszkiewicz, A.; Schuurhuis, G.J.; Kessler, F.L.; Piersma, S.R.; Knol, J.C.; Pham, T.V.; Jansen, G.; Musters, R.J.; van Meerloo, J.; Assaraf, Y.G. Exosomes secreted by apoptosis-resistant acute myeloid leukemia (AML) blasts harbor regulatory network proteins potentially involved in antagonism of apoptosis. Mol. Cell. Proteom. 2016, 15, 1281–1298. [Google Scholar] [CrossRef] [Green Version]
- Bouvy, C.; Wannez, A.; Laloy, J.; Chatelain, C.; Dogné, J.-M. Transfer of multidrug resistance among acute myeloid leukemia cells via extracellular vesicles and their microRNA cargo. Leuk. Res. 2017, 62, 70–76. [Google Scholar] [CrossRef]
- Daver, N.; Schlenk, R.F.; Russell, N.H.; Levis, M.J. Targeting FLT3 mutations in AML: Review of current knowledge and evidence. Leukemia 2019, 33, 299–312. [Google Scholar] [CrossRef] [Green Version]
- Melo, S.A.; Luecke, L.B.; Kahlert, C.; Fernandez, A.F.; Gammon, S.T.; Kaye, J.; LeBleu, V.S.; Mittendorf, E.A.; Weitz, J.; Rahbari, N. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature 2015, 523, 177–182. [Google Scholar] [CrossRef] [Green Version]
- Costa-Silva, B.; Aiello, N.M.; Ocean, A.J.; Singh, S.; Zhang, H.; Thakur, B.K.; Becker, A.; Hoshino, A.; Mark, M.T.; Molina, H. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 2015, 17, 816–826. [Google Scholar] [CrossRef] [PubMed]
- Hu, Y.; Yan, C.; Mu, L.; Huang, K.; Li, X.; Tao, D.; Wu, Y.; Qin, J. Fibroblast-derived exosomes contribute to chemoresistance through priming cancer stem cells in colorectal cancer. PLoS ONE 2015, 10, e0125625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qin, X.; Guo, H.; Wang, X.; Zhu, X.; Yan, M.; Wang, X.; Xu, Q.; Shi, J.; Lu, E.; Chen, W. Exosomal miR-196a derived from cancer-associated fibroblasts confers cisplatin resistance in head and neck cancer through targeting CDKN1B and ING5. Genome Biol. 2019, 20, 1–21. [Google Scholar] [CrossRef] [PubMed]
- Yeung, C.L.A.; Co, N.-N.; Tsuruga, T.; Yeung, T.-L.; Kwan, S.-Y.; Leung, C.S.; Li, Y.; Lu, E.S.; Kwan, K.; Wong, K.-K. Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat. Commun. 2016, 7, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Challagundla, K.B.; Wise, P.M.; Neviani, P.; Chava, H.; Murtadha, M.; Xu, T.; Kennedy, R.; Ivan, C.; Zhang, X.; Vannini, I. Exosome-mediated transfer of microRNAs within the tumor microenvironment and neuroblastoma resistance to chemotherapy. JNCI J. Natl. Cancer Inst. 2015, 107. [Google Scholar] [CrossRef] [Green Version]
- Mc Namee, N.; O’Driscoll, L. Extracellular vesicles and anti-cancer drug resistance. Biochim. Biophys. Acta Rev. Cancer 2018, 1870, 123–136. [Google Scholar] [CrossRef]
- Lopes-Rodrigues, V.; Di Luca, A.; Mleczko, J.; Meleady, P.; Henry, M.; Pesic, M.; Cabrera, D.; van Liempd, S.; Lima, R.T.; O’Connor, R. Identification of the metabolic alterations associated with the multidrug resistant phenotype in cancer and their intercellular transfer mediated by extracellular vesicles. Sci. Rep. 2017, 7, 1–17. [Google Scholar] [CrossRef]
- Wang, X.; Zhang, H.; Chen, X. Drug resistance and combating drug resistance in cancer. Cancer Drug Resist. 2019, 2, 141–160. [Google Scholar] [CrossRef] [Green Version]
- Ye, Q.; Liu, K.; Shen, Q.; Li, Q.; Hao, J.; Han, F.; Jiang, R.-W. Reversal of multidrug resistance in cancer by multi-functional flavonoids. Front. Oncol. 2019, 9, 487. [Google Scholar] [CrossRef] [Green Version]
- Corcoran, C.; Rani, S.; O’Brien, K.; O’Neill, A.; Prencipe, M.; Sheikh, R.; Webb, G.; McDermott, R.; Watson, W.; Crown, J. Docetaxel-resistance in prostate cancer: Evaluating associated phenotypic changes and potential for resistance transfer via exosomes. PLoS ONE 2012, 7, e50999. [Google Scholar] [CrossRef]
- Rahman, M.; Hasan, M.R. Cancer metabolism and drug resistance. Metabolites 2015, 5, 571–600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Castro, I.; Xavier, C.; Vasconcelos, M. Is P-glycoprotein relevant for the release of microvesicles by tumor cells?: PS212. Porto Biomed. J. 2017, 2, 226. [Google Scholar] [CrossRef] [PubMed]
- Fontana, F.; Carollo, E.; Melling, G.E.; Carter, D.R. Extracellular Vesicles: Emerging Modulators of Cancer Drug Resistance. Cancers 2021, 13, 749. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Qiao, D.; Chen, L.; Xu, M.; Chen, S.; Huang, L.; Wang, F.; Chen, Z.; Cai, J.; Fu, L. Chemotherapeutic drugs stimulate the release and recycling of extracellular vesicles to assist cancer cells in developing an urgent chemoresistance. Mol. Cancer 2019, 18, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Levchenko, A.; Mehta, B.M.; Niu, X.; Kang, G.; Villafania, L.; Way, D.; Polycarpe, D.; Sadelain, M.; Larson, S.M. Intercellular transfer of P-glycoprotein mediates acquired multidrug resistance in tumor cells. Proc. Natl. Acad. Sci. USA 2005, 102, 1933–1938. [Google Scholar] [CrossRef] [Green Version]
- Bebawy, M.; Combes, V.; Lee, E.; Jaiswal, R.; Gong, J.; Bonhoure, A.; Grau, G. Membrane microparticles mediate transfer of P-glycoprotein to drug sensitive cancer cells. Leukemia 2009, 23, 1643–1649. [Google Scholar] [CrossRef] [Green Version]
- Chen, W.-X.; Cai, Y.-Q.; Lv, M.-M.; Chen, L.; Zhong, S.-L.; Ma, T.-F.; Zhao, J.-H.; Tang, J.-H. Exosomes from docetaxel-resistant breast cancer cells alter chemosensitivity by delivering microRNAs. Tumor Biol. 2014, 35, 9649–9659. [Google Scholar] [CrossRef]
- Yu, S.; Wei, Y.; Xu, Y.; Zhang, Y.; Li, J.; Zhang, J. Extracellular vesicles in breast cancer drug resistance and their clinical application. Tumor Biol. 2016, 37, 2849–2861. [Google Scholar] [CrossRef]
- Lopes-Rodrigues, V.; Di Luca, A.; Sousa, D.; Seca, H.; Meleady, P.; Henry, M.; Lima, R.T.; O’Connor, R.; Vasconcelos, M.H. Multidrug resistant tumour cells shed more microvesicle-like EVs and less exosomes than their drug-sensitive counterpart cells. Biochim. Biophys. Acta Gen. Subj. 2016, 1860, 618–627. [Google Scholar] [CrossRef]
- Kwok, H.-H.; Ning, Z.; Chong, P.W.-C.; Wan, T.S.-K.; Ng, M.H.-L.; Ho, G.Y.; Ip, M.S.-M.; Lam, D.C.-L. Transfer of extracellular vesicle-associated-RNAs induces drug resistance in ALK-translocated lung adenocarcinoma. Cancers 2019, 11, 104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yan, X.; Yin, J.; Yao, H.; Mao, N.; Yang, Y.; Pan, L. Increased expression of annexin A3 is a mechanism of platinum resistance in ovarian cancer. Cancer Res. 2010, 70, 1616–1624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ciravolo, V.; Huber, V.; Ghedini, G.C.; Venturelli, E.; Bianchi, F.; Campiglio, M.; Morelli, D.; Villa, A.; Mina, P.D.; Menard, S. Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy. J. Cell. Physiol. 2012, 227, 658–667. [Google Scholar] [CrossRef] [PubMed]
- Mikamori, M.; Yamada, D.; Eguchi, H.; Hasegawa, S.; Kishimoto, T.; Tomimaru, Y.; Asaoka, T.; Noda, T.; Wada, H.; Kawamoto, K. MicroRNA-155 controls exosome synthesis and promotes gemcitabine resistance in pancreatic ductal adenocarcinoma. Sci. Rep. 2017, 7, 1–14. [Google Scholar]
- Singh, R.R.; Reindl, K.M. Glutathione S-Transferases in Cancer. Antioxidants (Basel) 2021, 10, 701. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Liu, R.X.; Chan, K.W.; Hu, J.; Zhang, J.; Wei, L.; Tan, H.; Yang, X.; Liu, H. Exosomal transfer of p-STAT3 promotes acquired 5-FU resistance in colorectal cancer cells. J. Exp. Clin. Cancer Res. CR 2019, 38, 320. [Google Scholar] [CrossRef] [PubMed]
- Dong, Y.; Pan, Q.; Jiang, L.; Chen, Z.; Zhang, F.; Liu, Y.; Xing, H.; Shi, M.; Li, J.; Li, X.; et al. Tumor endothelial expression of P-glycoprotein upon microvesicular transfer of TrpC5 derived from adriamycin-resistant breast cancer cells. Biochem. Biophys. Res. Commun. 2014, 446, 85–90. [Google Scholar] [CrossRef] [PubMed]
- Nanayakkara, A.K.; Follit, C.A.; Chen, G.; Williams, N.S.; Vogel, P.D.; Wise, J.G. Targeted inhibitors of P-glycoprotein increase chemotherapeutic-induced mortality of multidrug resistant tumor cells. Sci Rep 2018, 8, 967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hoskin, V.; Ghaffari, A.; Elliott, B.E. Ezrin, more than a metastatic detERMinant? Oncotarget 2019, 10, 6755–6757. [Google Scholar] [CrossRef]
- Yano, K.; Okabe, C.; Fujii, K.; Kato, Y.; Ogihara, T. Regulation of breast cancer resistance protein and P-glycoprotein by ezrin, radixin and moesin in lung, intestinal and renal cancer cell lines. J. Pharm. Pharmacol. 2020, 72, 575–582. [Google Scholar] [CrossRef]
- Pokharel, D.; Padula, M.P.; Lu, J.F.; Jaiswal, R.; Djordjevic, S.P.; Bebawy, M. The Role of CD44 and ERM Proteins in Expression and Functionality of P-glycoprotein in Breast Cancer Cells. Molecules 2016, 21, 290. [Google Scholar] [CrossRef] [Green Version]
- Jiang, Q.H.; Wang, A.X.; Chen, Y. Radixin enhances colon cancer cell invasion by increasing MMP-7 production via Rac1-ERK pathway. Sci. World J. 2014, 2014, 340271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cho, S.H.; Park, Y.S.; Kim, H.J.; Kim, C.H.; Lim, S.W.; Huh, J.W.; Lee, J.H.; Kim, H.R. CD44 enhances the epithelial-mesenchymal transition in association with colon cancer invasion. Int. J. Oncol. 2012, 41, 211–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Szatanek, R.; Baj-Krzyworzeka, M. CD44 and Tumor-Derived Extracellular Vesicles (TEVs). Possible Gateway to Cancer Metastasis. Int. J. Mol. Sci. 2021, 22, 1463. [Google Scholar] [CrossRef] [PubMed]
- Whitlock, B.D.; Leslie, E.M. Efflux transporters in anti-cancer drug resistance: Molecular and functional identification and characterization of multidrug resistance proteins (MRPs/ABCCs). In Drug Efflux Pumps in Cancer Resistance Pathways: From Molecular Recognition and Characterization to Possible Inhibition Strategies in Chemotherapy; Elsevier: Amsterdam, The Netherlands, 2020; pp. 31–65. [Google Scholar]
- Munoz, M.; Henderson, M.; Haber, M.; Norris, M. Role of the MRP1/ABCC1 multidrug transporter protein in cancer. IUBMB Life 2007, 59, 752–757. [Google Scholar] [CrossRef]
- Goler-Baron, V.; Assaraf, Y.G. Structure and function of ABCG2-rich extracellular vesicles mediating multidrug resistance. PLoS ONE 2011, 6, e16007. [Google Scholar] [CrossRef] [Green Version]
- Fukumori, T.; Kanayama, H.O.; Raz, A. The role of galectin-3 in cancer drug resistance. Drug Resist. Updates Rev. Comment. Antimicrob. Anticancer. Chemother. 2007, 10, 101–108. [Google Scholar] [CrossRef] [Green Version]
- Tonissen, K.F.; Poulsen, S.-A. Carbonic anhydrase XII inhibition overcomes P-glycoprotein-mediated drug resistance: A potential new combination therapy in cancer. Cancer Drug Resist. 2021, 4. [Google Scholar] [CrossRef]
- Ning, K.; Wang, T.; Sun, X.; Zhang, P.; Chen, Y.; Jin, J.; Hua, D. UCH-L1-containing exosomes mediate chemotherapeutic resistance transfer in breast cancer. J. Surg. Oncol. 2017, 115, 932–940. [Google Scholar] [CrossRef]
- Gong, J.; Luk, F.; Jaiswal, R.; George, A.M.; Grau, G.E.R.; Bebawy, M. Microparticle drug sequestration provides a parallel pathway in the acquisition of cancer drug resistance. Eur. J. Pharmacol. 2013, 721, 116–125. [Google Scholar] [CrossRef]
- Torreggiani, E.; Roncuzzi, L.; Perut, F.; Zini, N.; Baldini, N. Multimodal transfer of MDR by exosomes in human osteosarcoma. Int. J. Oncol. 2016, 49, 189–196. [Google Scholar] [CrossRef] [Green Version]
- Jaiswal, R.; Luk, F.; Dalla, P.V.; Grau, G.E.R.; Bebawy, M. Breast cancer-derived microparticles display tissue selectivity in the transfer of resistance proteins to cells. PLoS ONE 2013, 8, e61515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hongmei, Z. Extrinsic and Intrinsic Apoptosis Signal Pathway Review; InTechOpen: London, UK, 2012. [Google Scholar]
- Gregory, C.D.; Dransfield, I. Apoptotic tumor cell-derived extracellular vesicles as important regulators of the onco-regenerative niche. Front. Immunol. 2018, 9, 1111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Westhoff, M.-A.; Marschall, N.; Debatin, K.-M. Novel approaches to apoptosis-inducing therapies. Apoptosis Cancer Pathog. Anti-Cancer Ther. 2016, 173–204. [Google Scholar]
- Jiang, N.; Dai, Q.; Su, X.; Fu, J.; Feng, X.; Peng, J. Role of PI3K/AKT pathway in cancer: The framework of malignant behavior. Mol. Biol. Rep. 2020, 47, 4587–4629. [Google Scholar] [CrossRef]
- Dong, H.; Wang, W.; Chen, R.; Zhang, Y.; Zou, K.; Ye, M.; He, X.; Zhang, F.; Han, J. Exosome-mediated transfer of lncRNA-SNHG14 promotes trastuzumab chemoresistance in breast cancer. Int. J. Oncol. 2018, 53, 1013–1026. [Google Scholar] [CrossRef] [Green Version]
- Khoo, X.-H.; Paterson, I.C.; Goh, B.-H.; Lee, W.-L. Cisplatin-resistance in oral squamous cell carcinoma: Regulation by tumor cell-derived extracellular vesicles. Cancers 2019, 11, 1166. [Google Scholar] [CrossRef] [Green Version]
- Brzozowski, J.S.; Jankowski, H.; Bond, D.R.; McCague, S.B.; Munro, B.R.; Predebon, M.J.; Scarlett, C.J.; Skelding, K.A.; Weidenhofer, J. Lipidomic profiling of extracellular vesicles derived from prostate and prostate cancer cell lines. Lipids Health Dis. 2018, 17, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Soekmadji, C.; Nelson, C.C. The emerging role of extracellular vesicle-mediated drug resistance in cancers: Implications in advanced prostate cancer. BioMed Res. Int. 2015, 2015, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brügger, B.; Simons, M. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef]
- Faict, S.; Oudaert, I.; D’Auria, L.; Dehairs, J.; Maes, K.; Vlummens, P.; De Veirman, K.; De Bruyne, E.; Fostier, K.; Vande Broek, I. The transfer of sphingomyelinase contributes to drug resistance in multiple myeloma. Cancers 2019, 11, 1823. [Google Scholar] [CrossRef] [Green Version]
- Srivastava, A.; Amreddy, N.; Pareek, V.; Chinnappan, M.; Ahmed, R.; Mehta, M.; Razaq, M.; Munshi, A.; Ramesh, R. Progress in extracellular vesicle biology and their application in cancer medicine. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2020, 12, e1621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eichelser, C.; Stückrath, I.; Müller, V.; Milde-Langosch, K.; Wikman, H.; Pantel, K.; Schwarzenbach, H. Increased serum levels of circulating exosomal microRNA-373 in receptor-negative breast cancer patients. Oncotarget 2014, 5, 9650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lucien, F.; Lac, V.; Billadeau, D.D.; Borgida, A.; Gallinger, S.; Leong, H.S. Glypican-1 and glycoprotein 2 bearing extracellular vesicles do not discern pancreatic cancer from benign pancreatic diseases. Oncotarget 2019, 10, 1045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vader, P.; Breakefield, X.O.; Wood, M.J. Extracellular vesicles: Emerging targets for cancer therapy. Trends Mol. Med. 2014, 20, 385–393. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Wang, L.; Zhu, Y.; Chen, Z.; Qi, X.; Jin, L.; Jin, J.; Hua, D.; Ma, X. Breast cancer resistance protein (BCRP)-containing circulating microvesicles contribute to chemoresistance in breast cancer. Oncol. Lett. 2015, 10, 3742–3748. [Google Scholar] [CrossRef] [Green Version]
- Corcoran, C.; Rani, S.; O’Driscoll, L. miR-34a is an intracellular and exosomal predictive biomarker for response to docetaxel with clinical relevance to prostate cancer progression. Prostate 2014, 74, 1320–1334. [Google Scholar] [CrossRef] [Green Version]
- Li, L.; Li, C.; Wang, S.; Wang, Z.; Jiang, J.; Wang, W.; Li, X.; Chen, J.; Liu, K.; Li, C. Exosomes derived from hypoxic oral squamous cell carcinoma cells deliver miR-21 to normoxic cells to elicit a prometastatic phenotype. Cancer Res. 2016, 76, 1770–1780. [Google Scholar] [CrossRef] [Green Version]
- Shukuya, T.; Ghai, V.; Amann, J.M.; Okimoto, T.; Shilo, K.; Kim, T.-K.; Wang, K.; Carbone, D.P. Circulating MicroRNAs and Extracellular Vesicle–Containing MicroRNAs as Response Biomarkers of Anti–programmed Cell Death Protein 1 or Programmed Death-Ligand 1 Therapy in NSCLC. J. Thorac. Oncol. 2020, 15, 1773–1781. [Google Scholar] [CrossRef]
- Sidransky, D. Emerging molecular markers of cancer. Nat. Rev. Cancer 2002, 2, 210–219. [Google Scholar] [CrossRef]
- Tutrone, R.; Donovan, M.J.; Torkler, P.; Tadigotla, V.; McLain, T.; Noerholm, M.; Skog, J.; McKiernan, J. Clinical utility of the exosome based ExoDx Prostate (IntelliScore) EPI test in men presenting for initial Biopsy with a PSA 2–10 ng/mL. Prostate Cancer Prostatic Dis. 2020, 23, 607–614. [Google Scholar] [CrossRef]
- Lin, S.-Y.; Chang, C.-H.; Wu, H.-C.; Lin, C.-C.; Chang, K.-P.; Yang, C.-R.; Huang, C.-P.; Hsu, W.-H.; Chang, C.-T.; Chen, C.-J. Proteome profiling of urinary exosomes identifies alpha 1-antitrypsin and H2B1K as diagnostic and prognostic biomarkers for urothelial carcinoma. Sci. Rep. 2016, 6, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Trino, S.; Lamorte, D. Clinical relevance of extracellular vesicles in hematological neoplasms: From liquid biopsy to cell biopsy. Leukemia 2021, 35, 661–678. [Google Scholar] [CrossRef] [PubMed]
- Pang, B.; Zhu, Y.; Ni, J.; Thompson, J.; Malouf, D.; Bucci, J.; Graham, P.; Li, Y. Extracellular vesicles: The next generation of biomarkers for liquid biopsy-based prostate cancer diagnosis. Theranostics 2020, 10, 2309–2326. [Google Scholar] [CrossRef] [PubMed]
- Tavoosidana, G.; Ronquist, G.; Darmanis, S.; Yan, J.; Carlsson, L.; Wu, D.; Conze, T.; Ek, P.; Semjonow, A.; Eltze, E.; et al. Multiple recognition assay reveals prostasomes as promising plasma biomarkers for prostate cancer. Proc. Natl. Acad. Sci. USA 2011, 108, 8809–8814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rabinowits, G.; Gerçel-Taylor, C.; Day, J.M.; Taylor, D.D.; Kloecker, G.H. Exosomal microRNA: A diagnostic marker for lung cancer. Clin. Lung Cancer 2009, 10, 42–46. [Google Scholar] [CrossRef]
- Chang, L.; Ni, J.; Zhu, Y.; Pang, B.; Graham, P.; Zhang, H.; Li, Y. Liquid biopsy in ovarian cancer: Recent advances in circulating extracellular vesicle detection for early diagnosis and monitoring progression. Theranostics 2019, 9, 4130–4140. [Google Scholar] [CrossRef]
- Mathai, R.A.; Vidya, R.V.S.; Reddy, B.S.; Thomas, L.; Udupa, K.; Kolesar, J.; Rao, M. Potential Utility of Liquid Biopsy as a Diagnostic and Prognostic Tool for the Assessment of Solid Tumors: Implications in the Precision Oncology. J. Clin. Med. 2019, 8, 373. [Google Scholar] [CrossRef] [Green Version]
- Hoshino, A.; Kim, H.S.; Bojmar, L.; Gyan, K.E.; Cioffi, M.; Hernandez, J.; Zambirinis, C.P.; Rodrigues, G.; Molina, H.; Heissel, S.; et al. Extracellular Vesicle and Particle Biomarkers Define Multiple Human Cancers. Cell 2020, 182, 1044–1061. [Google Scholar] [CrossRef]
- Wu, S.; Zhu, W.; Thompson, P.; Hannun, Y.A. Evaluating intrinsic and non-intrinsic cancer risk factors. Nat. Commun. 2018, 9, 1–12. [Google Scholar] [CrossRef]
- Kosaka, N.; Iguchi, H.; Ochiya, T. Circulating microRNA in body fluid: A new potential biomarker for cancer diagnosis and prognosis. Cancer Sci. 2010, 101, 2087–2092. [Google Scholar] [CrossRef]
- Cabeza, L.; Perazzoli, G.; Peña, M.; Cepero, A.; Luque, C.; Melguizo, C.; Prados, J. Cancer therapy based on extracellular vesicles as drug delivery vehicles. J. Control Release 2020, 327, 296–315. [Google Scholar] [CrossRef] [PubMed]
- Cao, X.-H.; Liang, M.-X.; Wu, Y.; Yang, K.; Tang, J.-H.; Zhang, W. Extracellular vesicles as drug vectors for precise cancer treatment. Nanomedicine 2021, 16, 1519–1537. [Google Scholar] [CrossRef]
- Agrawal, A.K.; Aqil, F.; Jeyabalan, J.; Spencer, W.A.; Beck, J.; Gachuki, B.W.; Alhakeem, S.S.; Oben, K.; Munagala, R.; Bondada, S. Milk-derived exosomes for oral delivery of paclitaxel. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 1627–1636. [Google Scholar] [CrossRef] [PubMed]
- Van der Meel, R.; Fens, M.H.; Vader, P.; Van Solinge, W.W.; Eniola-Adefeso, O.; Schiffelers, R.M. Extracellular vesicles as drug delivery systems: Lessons from the liposome field. J. Control Release 2014, 195, 72–85. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.; Thompson, D. Stimuli-responsive liposomes for drug delivery. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2017, 9, e1450. [Google Scholar] [CrossRef]
- 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. Oxidative Med. Cell. Longev. 2019, 2019, 1–21. [Google Scholar] [CrossRef]
- Gaurav, I.; Thakur, A.; Iyaswamy, A.; Wang, X.; Chen, X.; Yang, Z. Factors Affecting Extracellular Vesicles Based Drug Delivery Systems. Molecules 2021, 26, 1544. [Google Scholar] [CrossRef]
- Murphy, D.E.; de Jong, O.G.; Brouwer, M.; Wood, M.J.; Lavieu, G.; Schiffelers, R.M.; Vader, P. Extracellular vesicle-based therapeutics: Natural versus engineered targeting and trafficking. Exp. Mol. Med. 2019, 51, 1–12. [Google Scholar] [CrossRef]
- Springer, A.D.; Dowdy, S.F. GalNAc-siRNA conjugates: Leading the way for delivery of RNAi therapeutics. Nucleic Acid Ther. 2018, 28, 109–118. [Google Scholar] [CrossRef]
- 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]
- Tian, H.; Li, W. Dendritic cell-derived exosomes for cancer immunotherapy: Hope and challenges. Ann. Transl. Med. 2017, 5, 221. [Google Scholar] [CrossRef] [PubMed]
- Whiteside, T.L. Exosomes in cancer: Another mechanism of tumor-induced immune suppression. Tumor Immune Microenviron. Cancer Progress. Cancer Ther. 2017, 81–89. [Google Scholar]
- Yang, X.; Chen, J.; Wang, N.; Liu, Z.; Li, Y. Clinical use of dendritic cell-derived exosomes for hepatocellular carcinoma immunotherapy: How far we are? J. Hepatol. 2018, 69, 984–986. [Google Scholar] [CrossRef] [Green Version]
- Luo, R.; Liu, M.; Tan, T.; Yang, Q.; Wang, Y.; Men, L.; Zhao, L.; Zhang, H.; Wang, S.; Xie, T. Emerging Significance and Therapeutic Potential of Extracellular vesicles. Int. J. Biol. Sci. 2021, 17, 2476–2486. [Google Scholar] [CrossRef] [PubMed]
- Campos, J.H.; Soares, R.P.; Ribeiro, K.; Cronemberger Andrade, A.; Batista, W.L.; Torrecilhas, A.C. Extracellular vesicles: Role in inflammatory responses and potential uses in vaccination in cancer and infectious diseases. J. Immunol. Res. 2015, 2015. [Google Scholar] [CrossRef] [Green Version]
- Whiteside, T. Exosomes carrying immunoinhibitory proteins and their role in cancer. Clin. Exp. Immunol. 2017, 189, 259–267. [Google Scholar] [CrossRef] [Green Version]
- Koppers-Lalic, D.; Hogenboom, M.M.; Middeldorp, J.M.; Pegtel, D.M. Virus-modified exosomes for targeted RNA delivery; A new approach in nanomedicine. Adv. Drug Deliv. Rev. 2013, 65, 348–356. [Google Scholar] [CrossRef]
- Wirth, T.; Ylä-Herttuala, S. Gene therapy used in cancer treatment. Biomedicines 2014, 2, 149–162. [Google Scholar] [CrossRef]
- Mizrak, A.; Bolukbasi, M.F.; Ozdener, G.B.; Brenner, G.J.; Madlener, S.; Erkan, E.P.; Ströbel, T.; Breakefield, X.O.; Saydam, O. Genetically engineered microvesicles carrying suicide mRNA/protein inhibit schwannoma tumor growth. Mol. Ther. J. Am. Soc. Gene Ther. 2013, 21, 101–108. [Google Scholar] [CrossRef] [Green Version]
- Erkan, E.P.; Senfter, D.; Madlener, S.; Jungwirth, G.; Ströbel, T.; Saydam, N.; Saydam, O. Extracellular vesicle-mediated suicide mRNA/protein delivery inhibits glioblastoma tumor growth in vivo. Cancer Gene Ther. 2017, 24, 38–44. [Google Scholar] [CrossRef]
- Alexander, M.; Hu, R.; Runtsch, M.C.; Kagele, D.A.; Mosbruger, T.L.; Tolmachova, T.; Seabra, M.C.; Round, J.L.; Ward, D.M.; O’Connell, R.M. Exosome-delivered microRNAs modulate the inflammatory response to endotoxin. Nat. Commun. 2015, 6, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Usman, W.M.; Pham, T.C.; Kwok, Y.Y.; Vu, L.T.; Ma, V.; Peng, B.; Chan, Y.S.; Wei, L.; Chin, S.M.; Azad, A.; et al. Efficient RNA drug delivery using red blood cell extracellular vesicles. Nat. Commun. 2018, 9, 2359. [Google Scholar] [CrossRef]
- Caforio, M.; Sorino, C.; Bertaina, V.; Pitisci, A.; Battafarano, G.; Del Fattore, A.; Fanciulli, M.; Folgiero, V.; Locatelli, F. PB1649 Exosomes-Mediated Delivery of RNA Oligos Directed to Che-1/Aatf Impairs Bcp-All Vitality. HemaSphere 2019, 3, 763. [Google Scholar] [CrossRef]
- Lin, Y.; Wu, J.; Gu, W.; Huang, Y.; Tong, Z.; Huang, L.; Tan, J. Exosome-Liposome Hybrid Nanoparticles Deliver CRISPR/Cas9 System in MSCs. Adv. Sci. 2018, 5, 1700611. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.M.; Yang, Y.; Oh, S.J.; Hong, Y.; Seo, M.; Jang, M. Cancer-derived exosomes as a delivery platform of CRISPR/Cas9 confer cancer cell tropism-dependent targeting. J. Control. Release 2017, 266, 8–16. [Google Scholar] [CrossRef] [PubMed]
- Börger, V.; Weiss, D.J.; Anderson, J.D.; Borràs, F.E.; Bussolati, B.; Carter, D.R.F.; Dominici, M.; Falcón-Pérez, J.M.; Gimona, M.; Hill, A.F.; et al. International Society for Extracellular Vesicles and International Society for Cell and Gene Therapy statement on extracellular vesicles from mesenchymal stromal cells and other cells: Considerations for potential therapeutic agents to suppress coronavirus disease-19. Cytotherapy 2020, 22, 482–485. [Google Scholar] [CrossRef]
- 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]
- Massaro, C.; Sgueglia, G.; Frattolillo, V.; Baglio, S.R.; Altucci, L. Extracellular Vesicle-Based Nucleic Acid Delivery: Current Advances and Future Perspectives in Cancer Therapeutic Strategies. Pharmaceutics 2020, 12, 980. [Google Scholar] [CrossRef]
- Lewis, N.D.; Sia, C.L.; Kirwin, K.; Haupt, S. Exosome Surface Display of IL12 Results in Tumor-Retained Pharmacology with Superior Potency and Limited Systemic Exposure Compared with Recombinant IL12. Mol. Cancer Ther. 2021, 20, 523–534. [Google Scholar] [CrossRef]
- Lewis, N.; Sia, C.L.; Kirwin, K.; Haupt, S.; Mahimkar, G.; Zi, T.; Xu, K.; Dooley, K.; Jang, S.C.; Choi, B. 709 Exosome surface display of IL-12 results in tumor-retained pharmacology with superior potency and limited systemic exposure. BMJ Spec. J. 2020, 8, A751. [Google Scholar] [CrossRef]
- codiakbio.com. Available online: https://ir.codiakbio.com/news-releases/news-release-details/codiak-initiates-patient-dosing-phase-12-clinical-trial (accessed on 19 January 2021).
- codiakbio.com. Available online: https://ir.codiakbio.com/node/6411/pdf (accessed on 19 January 2021).
- biospace.com. Available online: https://www.biospace.com/article/un-stranding-assets-with-evs/ (accessed on 19 January 2021).
- omnispirant.com. Available online: https://www.omnispirant.com/ (accessed on 19 January 2021).
- carminetherapeutics.com. Available online: https://www.carminetherapeutics.com/ (accessed on 19 January 2021).
- Roefs, M.T.; Sluijter, J.P.; Vader, P. Extracellular vesicle-associated proteins in tissue repair. Trends Cell Biol. 2020, 30, 990–1013. [Google Scholar] [CrossRef] [PubMed]
- Meng, W.; He, C.; Hao, Y.; Wang, L.; Li, L.; Zhu, G. Prospects and challenges of extracellular vesicle-based drug delivery system: Considering cell source. Drug Deliv. 2020, 27, 585–598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kennedy, T.L.; Russell, A.J.; Riley, P. Experimental limitations of extracellular vesicle-based therapies for the treatment of myocardial infarction. Trends Cardiovasc. Med. 2021, 31, 405–415. [Google Scholar] [CrossRef] [PubMed]
S. No. | Source of EVs | Functions of EVs/EV Cargoes | References |
---|---|---|---|
1 | CD9-containing vesicles | Sperm and egg fusion | [35] |
2 | Oocyte | Remove sperm receptor to prevent polyspermy | [36] |
3 | Embryonic stem cells | EVs to communicate with environment in blastocyst; trophoblast migration and invasive properties | [37] |
4 | B Lymphocytes | To activate T cells | [38] |
5 | Antigen-presenting cells | RNA cargo influencing immune cell behavior | [39] |
6 | Dendritic cells | miRNA cargo represses mRNA expression target in acceptor dendritic cells | [39] |
7 | Regulatory T cells | miRNA cargo suppresses inflammatory responses of helper T1 cells | [40] |
8 | Neurons and glia cells | Facilitate intercellular communication | [41] |
9 | Neurons | mi-124a cargo uptake by astrocytes upregulates expression of GLT1 | [42] |
10 | Oligodendrocytes | Neurons intake myelin proteins and oxidative stress-protective proteins, causing changes in neuronal firing rates and gene-expression profiles | [43] |
11 | Oligodendrocyte | Inhibiting differentiation and myelin formation | [44] |
12 | Schwann cells | Enhance regeneration capacity after sciatic nerve injury | [45] |
13 | Microglia | Suppress the production of sphingolipid ceramides and sphingosine to regulate neuronal excitability | [46] |
14 | Cells of human blood from wound site | EVs expose a highly procoagulant tissue factor, implying that EVs play a role in hemostasis | [47] |
15 | Endothelial cells | Matrix metalloproteinases cargoes enhance matrix degradation and promotes angiogenesis | [48] |
16 | Platelets | Promotes cell proliferation, the migration of endothelial cells, and vessel formation | [49] |
17 | Lymphocyte-derived EVs | Prohibits VEGF pathway and, thus, suppresses angiogenesis and augments oxidative stress | [50] |
18 | Stem-cell-derived EVs | Bioactive cargoes have regenerative abilities | [51] |
S. No. | Cancer Type | EV Cargo Type | Role/Cancer Hallmarks | References |
---|---|---|---|---|
1 | Acute Leukemia | miR-116 | Cell proliferation and escape from apoptosis | [68] |
2 | Acute Leukemia | miR-118 | Cell proliferation and escape from apoptosis | [68] |
3 | Brain cancer | miR-181c | Cell invasion and metastasis | [105] |
4 | Breast cancer | miR-1246 | Cell proliferation and escape from apoptosis | [139] |
5 | Breast cancer | miR-210 | Sustaining of angiogenesis | [111] |
6 | Breast cancer | miR-200 family | Cell invasion and metastasis (also highly involved in epithelial to mesenchymal transition) | [140] |
7 | Breast cancer | miR- 205 | Cell invasion and metastasis (also highly involved in epithelial to mesenchymal transition) | [140] |
8 | Breast cancer | Caveolin-1 | Cell invasion and metastasis | [141] |
9 | Breast cancer | miR-122 | Reprogramming energy metabolism | [142] |
10 | Breast cancer | GSTP1 | Reprogramming energy metabolism | [78] |
11 | Breast cancer | miR-122 | Reprogramming energy metabolism | [78] |
12 | Cholangiocarcinoma | miR-205 | Cell proliferation and escape from apoptosis | [143] |
13 | Cholangiocarcinoma | miR-205-5p | Cell invasion | [143] |
14 | Colon cancer | DeltaNp73 | Cell proliferation and escape from apoptosis | [143] |
15 | Colon cancer | miR-193a | Cell proliferation and escape from apoptosis | [144] |
16 | Colon cancer | miR-200b | Cell proliferation and escape from apoptosis | [145] |
17 | Colon cancer | miR-25-3p | Sustaining of angiogenesis | [146] |
18 | Colon cancer | Wnt5b | Cell invasion and metastasis | [147] |
19 | Colon cancer | AREG | Cell invasion and metastasis | [148] |
20 | Colon cancer | β-catenin (mutant) | Transfer mutations | [149] |
21 | Colon cancer | miR-1246 | Evade immune response and promote inflammation | [122] |
22 | Colon cancer | Carcinoembryonic antigen related cell adhesion molecule | Evade immune response and promote inflammation | [150] |
23 | Colorectal cancer | miR-10b | Evade immune response and promote inflammation | [151] |
24 | Colorectal cancer | lnRNA PVT1 | Cell proliferation and escape from apoptosis | [152] |
25 | Colorectal cancer | miR200b | Cell proliferation and escape from apoptosis | [152] |
26 | Esophageal cancer | miR-93-5p | Cell proliferation and escape from apoptosis | [69] |
27 | Esophageal cancer | miR-21 | Cell proliferation and escape from apoptosis | [70] |
28 | Esophageal cancer | lncZEB1-AS1 | Cell proliferation and escape from apoptosis | [153] |
29 | Gastric cancer | lncRNAZFAS1 | Cell proliferation and escape from apoptosis | [75] |
30 | Gastric cancer | miR-423-5p | Cell invasion and metastasis | [154] |
31 | Gastric cancer | miR-27a | Evade immune response and promote inflammation | [155] |
32 | Glioblastoma | RBM11 (splicing factor) | Cell proliferation and escape from apoptosis | [99] |
33 | Glioblastoma | CLIC1 | Cell proliferation and escape from apoptosis | [156] |
34 | Glioblastoma | lncRNA CCAT2 | Sustaining of angiogenesis | [157] |
35 | Glioblastoma | lncRNA POU3F3 | Sustaining of angiogenesis | [158] |
36 | Glioblastoma | miR-21 (VEGF- upregulate expression) | Sustaining of angiogenesis | [159] |
37 | Glioblastoma | CXCR4 | Sustaining of angiogenesis | [160] |
38 | Glioblastoma | miR-148a | Cell invasion and metastasis | [161] |
39 | Glioblastoma | PTPRZ1-MET (fusion gene) | Transfer mutations | [137] |
40 | Glioblastoma | EGFRvIII (oncogenic receptor) | Transfer mutations | [162] |
41 | Glioblastoma | miR-210 | Evade immune response and promote inflammation | [125] |
42 | Glioblastoma | PD-L1 | Evade immune response and promote inflammation | [163] |
43 | Head-and-neck squamous cell carcinoma | EPHB2 | Sustaining of angiogenesis | [164] |
44 | Hepatocellular carcinoma | lncRNA TUC339 | Cell proliferation and escape from apoptosis | [164] |
45 | Hepatocellular carcinoma | CXCR4 | Cell proliferation and escape from apoptosis | [164] |
46 | Hepatocellular carcinoma | SMAD3 | Cell proliferation and escape from apoptosis | [165] |
47 | Hepatocellular carcinoma | miR-93 | Cell proliferation and escape from apoptosis | [166] |
48 | Hepatocellular carcinoma | miR-103 | Cell proliferation and escape from apoptosis | [167] |
49 | Hepatocellular carcinoma | miR-584 | Cell proliferation and escape from apoptosis | [168] |
50 | Hepatocellular carcinoma | miR-517c | Cell proliferation and escape from apoptosis | [168] |
51 | Hepatocellular carcinoma | miR-378 | Cell proliferation and escape from apoptosis | [168] |
52 | Hepatocellular carcinoma | miR-520f | Cell proliferation and escape from apoptosis | [168] |
53 | Hepatocellular carcinoma | miR-142-5p | Cell proliferation and escape from apoptosis | [168] |
54 | Hepatocellular carcinoma | miR-451 | Cell proliferation and escape from apoptosis | [168] |
55 | Hepatocellular carcinoma | miR-518d | Cell proliferation and escape from apoptosis | [168] |
56 | Hepatocellular carcinoma | miR-215 | Cell proliferation and escape from apoptosis | [168] |
57 | Hepatocellular carcinoma | miR-376a | Cell proliferation and escape from apoptosis | [168] |
58 | Hepatocellular carcinoma | miR-133b | Cell proliferation and escape from apoptosis | [168] |
59 | Hepatocellular carcinoma | miR-367 | Cell proliferation and escape from apoptosis | [168] |
60 | Hepatocellular carcinoma | Vasorin | Sustaining of angiogenesis | [169] |
61 | Hepatocellular carcinoma | miR-103 | Sustaining of angiogenesis | [167] |
62 | Hepatocellular carcinoma | miR-21 | Evade immune response and promote inflammation | [115] |
63 | Leukemia | miR-20b | Cell proliferation and escape from apoptosis | [68] |
64 | Liposarcoma | miR-25-3p | Evade immune response and promote inflammation | [123] |
65 | Liposarcoma | miR-92a-3p | Evade immune response and promote inflammation | [123] |
66 | Lung cancer | miR-143-3p | Sustaining of angiogenesis | [170] |
67 | Lung cancer | miR-145-5p | Sustaining of angiogenesis | [170] |
68 | Lung cancer | miR-23a | Sustaining of angiogenesis | [171] |
69 | Lung cancer | miR-142-3p | Evade immune response and promote inflammation | [117] |
70 | Melanoma | PGDRF- β | Cell proliferation and escape from apoptosis | [100] |
71 | Melanoma | Programmed cell death protein 1 and cytotoxic T lymphocyte associated antigen-4 | Cell proliferation and escape from apoptosis | [172] |
72 | Melanoma | ALK isoform | Transfer mutations | [173] |
73 | Melanoma | miR-155-5p | Evade immune response and promote inflammation | [174] |
74 | Multiple myeloma | piRNA-823 | Sustaining of angiogenesis | [175] |
75 | Nasopharyngeal carcinoma | miR-23a | Sustaining of angiogenesis | [176] |
76 | Nasopharyngeal carcinoma | miR-24-3p | Evade immune response and promote inflammation | [177] |
77 | Nasopharyngeal carcinoma | Galactin-9 | Evade immune response and promote inflammation | [178] |
78 | Oral cancer | miR-142-3p | Sustaining of angiogenesis | [179] |
79 | Osteosarcoma | miR-675 | Evade immune response and promote inflammation | [180] |
80 | Ovarian cancer | miR-99a-5p | Cell invasion and metastasis | [181] |
81 | Ovarian cancer | SMAD4 | Transfer mutations | [138] |
82 | Ovarian cancer | miR-1246 | Evade immune response and promote inflammation | [121] |
83 | Ovarian cancer | Arginase-1 | Evade immune response and promote inflammation | [182] |
84 | Ovarian carcinoma | miR-141-3p | Sustaining of angiogenesis | [110] |
85 | Ovarian serous carcinoma | miR-21 | Cell proliferation and escape from apoptosis | [70] |
86 | Pancreatic cancer | miR-23b-5p | Cell proliferation and escape from apoptosis | [183] |
87 | Papillary thyroid cancer | miR-146b | Cell proliferation | [184] |
88 | Papillary thyroid cancer | miR-222 | Cell proliferation | [184] |
89 | Prostate cancer | miR1246 | Cell invasion and metastasis | [185] |
90 | Renal cell carcinoma | Fas ligand | Evade immune response and promote inflammation | [186] |
S. No. | Protein/Enzyme from EVs | Role | Cancer | Reference |
---|---|---|---|---|
1 | Glutathione S-transferases (GSTs) | Use glutathione conjugation to detoxify anticancer drugs | Breast cancer Colon cancer | [217,218] |
2 | P-gp (MDR1) | Drug efflux | Leukemia Breast cancer Prostate cancer | [203,219,220] |
3 | TrpC5 | Transcriptional activation of the MDR1 (ABCB1) promoter by NFATc3 | Breast Cancer (MCF7) | [219] |
4 | Ezrin | Modulate P-gp | Lung cancer Breast cancer | [221,222,223] |
5 | Radixin | Modulate P-gp | Colon cancer | [223,224] |
6 | Moesin | Modulate P-gp | Breast cancer | [223] |
7 | CD44 | Modulate P-gp | Colon cancer Gastric cancer Ovarian cancer Pancreatic cancer | [223,225,226] |
8 | Multidrug-resistance-associated proteins (MRP1-9/ABCCs) | Drug efflux | Lung cancer Breast cancer Prostate cancer Childhood neuroblastoma | [227,228] |
9 | ABCG2 | Mediates the efficient pumping and concentration of multiple cytotoxic agents | Breast cancer | [229] |
10 | Galectin-3 | Activation of NF-κB | Neck cancer Thyroid cancer Gastric cancer Colon cancer Uterine cancer Renal cancer | [230] |
11 | Carbonic anhydrase XII (CA XII) | Co-expressed and co-located with P-gp | Renal cancer Breast cancer | [231] |
12 | UCH-L1 | Upregulate P-gp protein expression levels via the MAPK/ERK signaling pathway | Breast cancer | [232] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Jahan, S.; Mukherjee, S.; Ali, S.; Bhardwaj, U.; Choudhary, R.K.; Balakrishnan, S.; Naseem, A.; Mir, S.A.; Banawas, S.; Alaidarous, M.; et al. Pioneer Role of Extracellular Vesicles as Modulators of Cancer Initiation in Progression, Drug Therapy, and Vaccine Prospects. Cells 2022, 11, 490. https://doi.org/10.3390/cells11030490
Jahan S, Mukherjee S, Ali S, Bhardwaj U, Choudhary RK, Balakrishnan S, Naseem A, Mir SA, Banawas S, Alaidarous M, et al. Pioneer Role of Extracellular Vesicles as Modulators of Cancer Initiation in Progression, Drug Therapy, and Vaccine Prospects. Cells. 2022; 11(3):490. https://doi.org/10.3390/cells11030490
Chicago/Turabian StyleJahan, Sadaf, Shouvik Mukherjee, Shaheen Ali, Urvashi Bhardwaj, Ranjay Kumar Choudhary, Santhanaraj Balakrishnan, Asma Naseem, Shabir Ahmad Mir, Saeed Banawas, Mohammed Alaidarous, and et al. 2022. "Pioneer Role of Extracellular Vesicles as Modulators of Cancer Initiation in Progression, Drug Therapy, and Vaccine Prospects" Cells 11, no. 3: 490. https://doi.org/10.3390/cells11030490
APA StyleJahan, S., Mukherjee, S., Ali, S., Bhardwaj, U., Choudhary, R. K., Balakrishnan, S., Naseem, A., Mir, S. A., Banawas, S., Alaidarous, M., Alyenbaawi, H., Iqbal, D., & Siddiqui, A. J. (2022). Pioneer Role of Extracellular Vesicles as Modulators of Cancer Initiation in Progression, Drug Therapy, and Vaccine Prospects. Cells, 11(3), 490. https://doi.org/10.3390/cells11030490