Skip to main content
Top
Published in: Cellular Oncology 3/2018

01-06-2018 | Review

The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications

Authors: Vignesh Sundararajan, Fazlul H. Sarkar, Thamil Selvee Ramasamy

Published in: Cellular Oncology | Issue 3/2018

Login to get access

Abstract

Background

Recent advances in cancer biology have highlighted the relevance of exosomes and nanovesicles as carriers of genetic and biological messages between cancer cells and their immediate and/or distant environments. It has been found that these molecular cues may play significant roles in cancer progression and metastasis. Cancer cells secrete exosomes containing diverse molecules that can be transferred to recipient cells and/or vice versa to induce a plethora of biological processes, including angiogenesis, metastasis formation, therapeutic resistance, epithelial-mesenchymal transition and epigenetic/stemness (re)programming. While exosomes interact with cells within the tumour microenvironment to promote tumour growth, these vesicles can also facilitate the process of distant metastasis by mediating the formation of pre-metastatic niches. Next to their tumour promoting effects, exosomes have been found to serve as potential tools for cancer diagnosis and therapy. The ease of isolating exosomes and their content from different body fluids has led to the identification of diagnostic and prognostic biomarker signatures, as well as to predictive biomarker signatures for therapeutic responses. Exosomes can also be used as cargos to deliver therapeutic anti-cancer drugs, and they can be engineered to serve as vaccines for immunotherapy. Additionally, it has been found that inhibition of exosome secretion, and thus the transfer of oncogenic molecules, holds promise for inhibiting tumour growth. Here we provide recent information on the diverse roles of exosomes in various cellular and systemic processes governing cancer progression, and discuss novel strategies to halt this progression using exosome-based targeted therapies and methods to inhibit exosome secretion and the transfer of pro-tumorigenic molecules.

Conclusions

This review highlights the important role of exosomes in cancer progression and its implications for (non-invasive) diagnostics and the development of novel therapeutic strategies, as well as its current and future applications in clinical trials.
Literature
1.
go back to reference S.E.L. Andaloussi, I. Mager, X.O. Breakefield, M.J. Wood, Extracellular vesicles: biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 12, 347–357 (2013)CrossRef S.E.L. Andaloussi, I. Mager, X.O. Breakefield, M.J. Wood, Extracellular vesicles: biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 12, 347–357 (2013)CrossRef
3.
go back to reference D.D. Yu, Y. Wu, H.Y. Shen, M.M. Lv, W.X. Chen, X.H. Zhang, S.L. Zhong, J.H. Tang, J.H. Zhao, Exosomes in development, metastasis and drug resistance of breast cancer. Cancer Sci. 106, 959–964 (2015)PubMedPubMedCentralCrossRef D.D. Yu, Y. Wu, H.Y. Shen, M.M. Lv, W.X. Chen, X.H. Zhang, S.L. Zhong, J.H. Tang, J.H. Zhao, Exosomes in development, metastasis and drug resistance of breast cancer. Cancer Sci. 106, 959–964 (2015)PubMedPubMedCentralCrossRef
4.
go back to reference C. Thery, L. Zitvogel, S. Amigorena, Exosomes: composition, biogenesis and function. Nat. Rev. Immunol. 2, 569–579 (2002)PubMedCrossRef C. Thery, L. Zitvogel, S. Amigorena, Exosomes: composition, biogenesis and function. Nat. Rev. Immunol. 2, 569–579 (2002)PubMedCrossRef
5.
go back to reference L. Muller, M. Mitsuhashi, P. Simms, W.E. Gooding, T.L. Whiteside, Tumor-derived exosomes regulate expression of immune function-related genes in human T cell subsets. Sci. Rep. 6, 20254 (2016)PubMedPubMedCentralCrossRef L. Muller, M. Mitsuhashi, P. Simms, W.E. Gooding, T.L. Whiteside, Tumor-derived exosomes regulate expression of immune function-related genes in human T cell subsets. Sci. Rep. 6, 20254 (2016)PubMedPubMedCentralCrossRef
6.
go back to reference H. Zhao, L. Yang, J. Baddour, A. Achreja, V. Bernard, T. Moss, J.C. Marini, T. Tudawe, E.G. Seviour, F.A. San Lucas, H. Alvarez, S. Gupta, S.N. Maiti, L. Cooper, D. Peehl, P.T. Ram, A. Maitra, D. Nagrath, Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism. elife 5, e10250 (2016)PubMedPubMedCentralCrossRef H. Zhao, L. Yang, J. Baddour, A. Achreja, V. Bernard, T. Moss, J.C. Marini, T. Tudawe, E.G. Seviour, F.A. San Lucas, H. Alvarez, S. Gupta, S.N. Maiti, L. Cooper, D. Peehl, P.T. Ram, A. Maitra, D. Nagrath, Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism. elife 5, e10250 (2016)PubMedPubMedCentralCrossRef
8.
go back to reference S.N. Chatterjee, J. Das, Electron microscopic observations on the excretion of cell-wall material by Vibrio cholerae. J. Gen. Microbiol. 49, 1–11 (1967)PubMedCrossRef S.N. Chatterjee, J. Das, Electron microscopic observations on the excretion of cell-wall material by Vibrio cholerae. J. Gen. Microbiol. 49, 1–11 (1967)PubMedCrossRef
9.
10.
go back to reference X. Yu, S.L. Harris, A.J. Levine, The regulation of exosome secretion: a novel function of the p53 protein. Cancer Res. 66, 4795–4801 (2006)PubMedCrossRef X. Yu, S.L. Harris, A.J. Levine, The regulation of exosome secretion: a novel function of the p53 protein. Cancer Res. 66, 4795–4801 (2006)PubMedCrossRef
11.
go back to reference A. Lespagnol, D. Duflaut, C. Beekman, L. Blanc, G. Fiucci, J.C. Marine, M. Vidal, R. Amson, A. Telerman, Exosome secretion, including the DNA damage-induced p53-dependent secretory pathway, is severely compromised in TSAP6/Steap3-null mice. Cell Death Differ. 15, 1723–1733 (2008)PubMedCrossRef A. Lespagnol, D. Duflaut, C. Beekman, L. Blanc, G. Fiucci, J.C. Marine, M. Vidal, R. Amson, A. Telerman, Exosome secretion, including the DNA damage-induced p53-dependent secretory pathway, is severely compromised in TSAP6/Steap3-null mice. Cell Death Differ. 15, 1723–1733 (2008)PubMedCrossRef
12.
go back to reference C. Thery, M. Ostrowski, E. Segura, Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 9, 581–593 (2009)PubMedCrossRef C. Thery, M. Ostrowski, E. Segura, Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 9, 581–593 (2009)PubMedCrossRef
13.
go back to reference W. Li, Y. Hu, T. Jiang, Y. Han, G. Han, J. Chen, X. Li, Rab27A regulates exosome secretion from lung adenocarcinoma cells A549: involvement of EPI64. APMIS 122, 1080–1087 (2014)PubMed W. Li, Y. Hu, T. Jiang, Y. Han, G. Han, J. Chen, X. Li, Rab27A regulates exosome secretion from lung adenocarcinoma cells A549: involvement of EPI64. APMIS 122, 1080–1087 (2014)PubMed
14.
go back to reference I. Parolini, C. Federici, C. Raggi, L. Lugini, S. Palleschi, A. De Milito, C. Coscia, E. Iessi, M. Logozzi, A. Molinari, M. Colone, M. Tatti, M. Sargiacomo, S. Fais, Microenvironmental pH is a key factor for exosome traffic in tumor cells. J. Biol. Chem. 284, 34211–34222 (2009)PubMedPubMedCentralCrossRef I. Parolini, C. Federici, C. Raggi, L. Lugini, S. Palleschi, A. De Milito, C. Coscia, E. Iessi, M. Logozzi, A. Molinari, M. Colone, M. Tatti, M. Sargiacomo, S. Fais, Microenvironmental pH is a key factor for exosome traffic in tumor cells. J. Biol. Chem. 284, 34211–34222 (2009)PubMedPubMedCentralCrossRef
15.
go back to reference J. Faure, G. Lachenal, M. Court, J. Hirrlinger, C. Chatellard-Causse, B. Blot, J. Grange, G. Schoehn, Y. Goldberg, V. Boyer, F. Kirchhoff, G. Raposo, J. Garin, R. Sadoul, Exosomes are released by cultured cortical neurones. Mol. Cell. Neurosci. 31, 642–648 (2006)PubMedCrossRef J. Faure, G. Lachenal, M. Court, J. Hirrlinger, C. Chatellard-Causse, B. Blot, J. Grange, G. Schoehn, Y. Goldberg, V. Boyer, F. Kirchhoff, G. Raposo, J. Garin, R. Sadoul, Exosomes are released by cultured cortical neurones. Mol. Cell. Neurosci. 31, 642–648 (2006)PubMedCrossRef
16.
go back to reference G. Lachenal, K. Pernet-Gallay, M. Chivet, F.J. Hemming, A. Belly, G. Bodon, B. Blot, G. Haase, Y. Goldberg, R. Sadoul, Release of exosomes from differentiated neurons and its regulation by synaptic glutamatergic activity. Mol. Cell. Neurosci. 46, 409–418 (2011)PubMedCrossRef G. Lachenal, K. Pernet-Gallay, M. Chivet, F.J. Hemming, A. Belly, G. Bodon, B. Blot, G. Haase, Y. Goldberg, R. Sadoul, Release of exosomes from differentiated neurons and its regulation by synaptic glutamatergic activity. Mol. Cell. Neurosci. 46, 409–418 (2011)PubMedCrossRef
17.
go back to reference N. Blanchard, D. Lankar, F. Faure, A. Regnault, C. Dumont, G. Raposo, C. Hivroz, TCR activation of human T cells induces the production of exosomes bearing the TCR/CD3/zeta complex. J. Immunol. 168, 3235–3241 (2002)PubMedCrossRef N. Blanchard, D. Lankar, F. Faure, A. Regnault, C. Dumont, G. Raposo, C. Hivroz, TCR activation of human T cells induces the production of exosomes bearing the TCR/CD3/zeta complex. J. Immunol. 168, 3235–3241 (2002)PubMedCrossRef
18.
go back to reference C.T. Roberts Jr., P. Kurre, Vesicle trafficking and RNA transfer add complexity and connectivity to cell-cell communication. Cancer Res. 73, 3200–3205 (2013)PubMedCrossRef C.T. Roberts Jr., P. Kurre, Vesicle trafficking and RNA transfer add complexity and connectivity to cell-cell communication. Cancer Res. 73, 3200–3205 (2013)PubMedCrossRef
19.
go back to reference E.G. Trams, C.J. Lauter, N. Salem Jr., U. Heine, Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochim. Biophys. Acta 645, 63–70 (1981)PubMedCrossRef E.G. Trams, C.J. Lauter, N. Salem Jr., U. Heine, Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochim. Biophys. Acta 645, 63–70 (1981)PubMedCrossRef
20.
go back to reference B.T. Pan, R.M. Johnstone, Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor. Cell 33, 967–978 (1983)PubMedCrossRef B.T. Pan, R.M. Johnstone, Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor. Cell 33, 967–978 (1983)PubMedCrossRef
21.
go back to reference C. Harding, J. Heuser, P. Stahl, Endocytosis and intracellular processing of transferrin and colloidal gold-transferrin in rat reticulocytes: demonstration of a pathway for receptor shedding. Eur. J. Cell Biol. 35, 256–263 (1984)PubMed C. Harding, J. Heuser, P. Stahl, Endocytosis and intracellular processing of transferrin and colloidal gold-transferrin in rat reticulocytes: demonstration of a pathway for receptor shedding. Eur. J. Cell Biol. 35, 256–263 (1984)PubMed
22.
go back to reference L. Balaj, R. Lessard, L. Dai, Y.J. Cho, S.L. Pomeroy, X.O. Breakefield, J. Skog, Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat. Commun. 2, 180 (2011)PubMedPubMedCentralCrossRef L. Balaj, R. Lessard, L. Dai, Y.J. Cho, S.L. Pomeroy, X.O. Breakefield, J. Skog, Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat. Commun. 2, 180 (2011)PubMedPubMedCentralCrossRef
23.
go back to reference H. Valadi, K. Ekstrom, A. Bossios, M. Sjostrand, J.J. Lee, J.O. Lotvall, Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 9, 654–659 (2007)PubMedCrossRef H. Valadi, K. Ekstrom, A. Bossios, M. Sjostrand, J.J. Lee, J.O. Lotvall, Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 9, 654–659 (2007)PubMedCrossRef
24.
go back to reference C. Admyre, S.M. Johansson, K.R. Qazi, J.J. Filen, R. Lahesmaa, M. Norman, E.P. Neve, A. Scheynius, S. Gabrielsson, Exosomes with immune modulatory features are present in human breast milk. J. Immunol. 179, 1969–1978 (2007)PubMedCrossRef C. Admyre, S.M. Johansson, K.R. Qazi, J.J. Filen, R. Lahesmaa, M. Norman, E.P. Neve, A. Scheynius, S. Gabrielsson, Exosomes with immune modulatory features are present in human breast milk. J. Immunol. 179, 1969–1978 (2007)PubMedCrossRef
25.
go back to reference M.P. Caby, D. Lankar, C. Vincendeau-Scherrer, G. Raposo, C. Bonnerot, Exosomal-like vesicles are present in human blood plasma. Int. Immunol. 17, 879–887 (2005)PubMedCrossRef M.P. Caby, D. Lankar, C. Vincendeau-Scherrer, G. Raposo, C. Bonnerot, Exosomal-like vesicles are present in human blood plasma. Int. Immunol. 17, 879–887 (2005)PubMedCrossRef
26.
go back to reference R. Shi, P.Y. Wang, X.Y. Li, J.X. Chen, Y. Li, X.Z. Zhang, C.G. Zhang, T. Jiang, W.B. Li, W. Ding, S.J. Cheng, Exosomal levels of miRNA-21 from cerebrospinal fluids associated with poor prognosis and tumor recurrence of glioma patients. Oncotarget 6, 26971–26981 (2015)PubMedPubMedCentral R. Shi, P.Y. Wang, X.Y. Li, J.X. Chen, Y. Li, X.Z. Zhang, C.G. Zhang, T. Jiang, W.B. Li, W. Ding, S.J. Cheng, Exosomal levels of miRNA-21 from cerebrospinal fluids associated with poor prognosis and tumor recurrence of glioma patients. Oncotarget 6, 26971–26981 (2015)PubMedPubMedCentral
27.
go back to reference M. Gonzalez-Begne, B. Lu, X. Han, F.K. Hagen, A.R. Hand, J.E. Melvin, J.R. Yates, Proteomic analysis of human parotid gland exosomes by multidimensional protein identification technology (MudPIT). J. Proteome Res. 8, 1304–1314 (2009)PubMedPubMedCentralCrossRef M. Gonzalez-Begne, B. Lu, X. Han, F.K. Hagen, A.R. Hand, J.E. Melvin, J.R. Yates, Proteomic analysis of human parotid gland exosomes by multidimensional protein identification technology (MudPIT). J. Proteome Res. 8, 1304–1314 (2009)PubMedPubMedCentralCrossRef
28.
go back to reference M. Tokuhisa, Y. Ichikawa, N. Kosaka, T. Ochiya, M. Yashiro, K. Hirakawa, T. Kosaka, H. Makino, H. Akiyama, C. Kunisaki, I. Endo, Exosomal miRNAs from peritoneum lavage fluid as potential prognostic biomarkers of peritoneal metastasis in gastric cancer. PLoS One 10, e0130472 (2015)PubMedPubMedCentralCrossRef M. Tokuhisa, Y. Ichikawa, N. Kosaka, T. Ochiya, M. Yashiro, K. Hirakawa, T. Kosaka, H. Makino, H. Akiyama, C. Kunisaki, I. Endo, Exosomal miRNAs from peritoneum lavage fluid as potential prognostic biomarkers of peritoneal metastasis in gastric cancer. PLoS One 10, e0130472 (2015)PubMedPubMedCentralCrossRef
29.
go back to reference T. Pisitkun, R.F. Shen, M.A. Knepper, Identification and proteomic profiling of exosomes in human urine. Proc. Natl. Acad. Sci. U. S. A. 101, 13368–13373 (2004)PubMedPubMedCentralCrossRef T. Pisitkun, R.F. Shen, M.A. Knepper, Identification and proteomic profiling of exosomes in human urine. Proc. Natl. Acad. Sci. U. S. A. 101, 13368–13373 (2004)PubMedPubMedCentralCrossRef
30.
go back to reference D.D. Taylor, C. Gercel-Taylor, MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 110, 13–21 (2008)PubMedCrossRef D.D. Taylor, C. Gercel-Taylor, MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 110, 13–21 (2008)PubMedCrossRef
31.
go back to reference C. Thery, S. Amigorena, G. Raposo and A. Clayton, Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr. Protoc. Cell Biol. Chapter 3, Unit 3 22 (2006) C. Thery, S. Amigorena, G. Raposo and A. Clayton, Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr. Protoc. Cell Biol. Chapter 3, Unit 3 22 (2006)
32.
go back to reference K. Trajkovic, C. Hsu, S. Chiantia, L. Rajendran, D. Wenzel, F. Wieland, P. Schwille, B. Brugger, M. Simons, Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 319, 1244–1247 (2008)PubMedCrossRef K. Trajkovic, C. Hsu, S. Chiantia, L. Rajendran, D. Wenzel, F. Wieland, P. Schwille, B. Brugger, M. Simons, Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 319, 1244–1247 (2008)PubMedCrossRef
34.
go back to reference T. Ravid, J.M. Heidinger, P. Gee, E.M. Khan, T. Goldkorn, c-Cbl-mediated ubiquitinylation is required for epidermal growth factor receptor exit from the early endosomes. J. Biol. Chem. 279, 37153–37162 (2004)PubMedCrossRef T. Ravid, J.M. Heidinger, P. Gee, E.M. Khan, T. Goldkorn, c-Cbl-mediated ubiquitinylation is required for epidermal growth factor receptor exit from the early endosomes. J. Biol. Chem. 279, 37153–37162 (2004)PubMedCrossRef
35.
go back to reference L. Duan, Y. Miura, M. Dimri, B. Majumder, I.L. Dodge, A.L. Reddi, A. Ghosh, N. Fernandes, P. Zhou, K. Mullane-Robinson, N. Rao, S. Donoghue, R.A. Rogers, D. Bowtell, M. Naramura, H. Gu, V. Band, H. Band, Cbl-mediated ubiquitinylation is required for lysosomal sorting of epidermal growth factor receptor but is dispensable for endocytosis. J. Biol. Chem. 278, 28950–28960 (2003)PubMedCrossRef L. Duan, Y. Miura, M. Dimri, B. Majumder, I.L. Dodge, A.L. Reddi, A. Ghosh, N. Fernandes, P. Zhou, K. Mullane-Robinson, N. Rao, S. Donoghue, R.A. Rogers, D. Bowtell, M. Naramura, H. Gu, V. Band, H. Band, Cbl-mediated ubiquitinylation is required for lysosomal sorting of epidermal growth factor receptor but is dispensable for endocytosis. J. Biol. Chem. 278, 28950–28960 (2003)PubMedCrossRef
37.
go back to reference S. Stuffers, C. Sem Wegner, H. Stenmark, A. Brech, Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic 10, 925–937 (2009)PubMedCrossRef S. Stuffers, C. Sem Wegner, H. Stenmark, A. Brech, Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic 10, 925–937 (2009)PubMedCrossRef
38.
go back to reference T. Kajimoto, T. Okada, S. Miya, L. Zhang, S. Nakamura, Ongoing activation of sphingosine 1-phosphate receptors mediates maturation of exosomal multivesicular endosomes. Nat. Commun. 4, 2712 (2013)PubMedCrossRef T. Kajimoto, T. Okada, S. Miya, L. Zhang, S. Nakamura, Ongoing activation of sphingosine 1-phosphate receptors mediates maturation of exosomal multivesicular endosomes. Nat. Commun. 4, 2712 (2013)PubMedCrossRef
39.
go back to reference D. Perez-Hernandez, C. Gutierrez-Vazquez, I. Jorge, S. Lopez-Martin, A. Ursa, F. Sanchez-Madrid, J. Vazquez, M. Yanez-Mo, The intracellular interactome of tetraspanin-enriched microdomains reveals their function as sorting machineries toward exosomes. J. Biol. Chem. 288, 11649–11661 (2013)PubMedPubMedCentralCrossRef D. Perez-Hernandez, C. Gutierrez-Vazquez, I. Jorge, S. Lopez-Martin, A. Ursa, F. Sanchez-Madrid, J. Vazquez, M. Yanez-Mo, The intracellular interactome of tetraspanin-enriched microdomains reveals their function as sorting machineries toward exosomes. J. Biol. Chem. 288, 11649–11661 (2013)PubMedPubMedCentralCrossRef
40.
go back to reference A.V. Vlassov, S. Magdaleno, R. Setterquist, R. Conrad, Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim. Biophys. Acta 1820, 940–948 (2012)PubMedCrossRef A.V. Vlassov, S. Magdaleno, R. Setterquist, R. Conrad, Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim. Biophys. Acta 1820, 940–948 (2012)PubMedCrossRef
41.
go back to reference M. Record, K. Carayon, M. Poirot, S. Silvente-Poirot, Exosomes as new vesicular lipid transporters involved in cell-cell communication and various pathophysiologies. Biochim. Biophys. Acta 1841, 108–120 (2014)PubMedCrossRef M. Record, K. Carayon, M. Poirot, S. Silvente-Poirot, Exosomes as new vesicular lipid transporters involved in cell-cell communication and various pathophysiologies. Biochim. Biophys. Acta 1841, 108–120 (2014)PubMedCrossRef
42.
go back to reference F. Coutant, L. Perrin-Cocon, S. Agaugué, T. Delair, P. André, V. Lotteau, Mature dendritic cell generation promoted by lysophosphatidylcholine. J. Immunol. 169, 1688–1695 (2002)PubMedCrossRef F. Coutant, L. Perrin-Cocon, S. Agaugué, T. Delair, P. André, V. Lotteau, Mature dendritic cell generation promoted by lysophosphatidylcholine. J. Immunol. 169, 1688–1695 (2002)PubMedCrossRef
43.
go back to reference L. Perrin-Cocon, S. Agaugué, F. Coutant, A. Masurel, S. Bezzine, G. Lambeau, P. André, V. Lotteau, Secretory phospholipase A2 induces dendritic cell maturation. Eur. J. Immunol. 34, 2293–2302 (2004)PubMedPubMedCentralCrossRef L. Perrin-Cocon, S. Agaugué, F. Coutant, A. Masurel, S. Bezzine, G. Lambeau, P. André, V. Lotteau, Secretory phospholipase A2 induces dendritic cell maturation. Eur. J. Immunol. 34, 2293–2302 (2004)PubMedPubMedCentralCrossRef
44.
go back to reference Q. Ge, Y. Zhou, J. Lu, Y. Bai, X. Xie, Z. Lu, miRNA in plasma exosome is stable under different storage conditions. Molecules 19, 1568–1575 (2014)PubMedCrossRef Q. Ge, Y. Zhou, J. Lu, Y. Bai, X. Xie, Z. Lu, miRNA in plasma exosome is stable under different storage conditions. Molecules 19, 1568–1575 (2014)PubMedCrossRef
45.
47.
go back to reference T. Tian, Y.L. Zhu, Y.Y. Zhou, G.F. Liang, Y.Y. Wang, F.H. Hu, Z.D. Xiao, Exosome uptake through clathrin-mediated endocytosis and macropinocytosis and mediating miR-21 delivery. J. Biol. Chem. 289, 22258–22267 (2014)PubMedPubMedCentralCrossRef T. Tian, Y.L. Zhu, Y.Y. Zhou, G.F. Liang, Y.Y. Wang, F.H. Hu, Z.D. Xiao, Exosome uptake through clathrin-mediated endocytosis and macropinocytosis and mediating miR-21 delivery. J. Biol. Chem. 289, 22258–22267 (2014)PubMedPubMedCentralCrossRef
48.
go back to reference K.J. Svensson, H.C. Christianson, A. Wittrup, E. Bourseau-Guilmain, E. Lindqvist, L.M. Svensson, M. Morgelin, M. Belting, Exosome uptake depends on ERK1/2-heat shock protein 27 signaling and lipid Raft-mediated endocytosis negatively regulated by caveolin-1. J. Biol. Chem. 288, 17713–17724 (2013)PubMedPubMedCentralCrossRef K.J. Svensson, H.C. Christianson, A. Wittrup, E. Bourseau-Guilmain, E. Lindqvist, L.M. Svensson, M. Morgelin, M. Belting, Exosome uptake depends on ERK1/2-heat shock protein 27 signaling and lipid Raft-mediated endocytosis negatively regulated by caveolin-1. J. Biol. Chem. 288, 17713–17724 (2013)PubMedPubMedCentralCrossRef
49.
go back to reference D. Zech, S. Rana, M.W. Büchler, M. Zöller, Tumor-exosomes and leukocyte activation: an ambivalent crosstalk. Cell Commun. Signaling 10, 37 (2012)CrossRef D. Zech, S. Rana, M.W. Büchler, M. Zöller, Tumor-exosomes and leukocyte activation: an ambivalent crosstalk. Cell Commun. Signaling 10, 37 (2012)CrossRef
50.
go back to reference S. Rana, S. Yue, D. Stadel, M. Zoller, Toward tailored exosomes: the exosomal tetraspanin web contributes to target cell selection. Int. J. Biochem. Cell Biol. 44, 1574–1584 (2012)PubMedCrossRef S. Rana, S. Yue, D. Stadel, M. Zoller, Toward tailored exosomes: the exosomal tetraspanin web contributes to target cell selection. Int. J. Biochem. Cell Biol. 44, 1574–1584 (2012)PubMedCrossRef
51.
go back to reference T.I. Naslund, D. Paquin-Proulx, P.T. Paredes, H. Vallhov, J.K. Sandberg, S. Gabrielsson, Exosomes from breast milk inhibit HIV-1 infection of dendritic cells and subsequent viral transfer to CD4+ T cells. AIDS 28, 171–180 (2014)PubMedCrossRef T.I. Naslund, D. Paquin-Proulx, P.T. Paredes, H. Vallhov, J.K. Sandberg, S. Gabrielsson, Exosomes from breast milk inhibit HIV-1 infection of dendritic cells and subsequent viral transfer to CD4+ T cells. AIDS 28, 171–180 (2014)PubMedCrossRef
52.
go back to reference J.R. Goldenring, A central role for vesicle trafficking in epithelial neoplasia: intracellular highways to carcinogenesis. Nat. Rev. Cancer 13, 813–820 (2013)PubMedPubMedCentralCrossRef J.R. Goldenring, A central role for vesicle trafficking in epithelial neoplasia: intracellular highways to carcinogenesis. Nat. Rev. Cancer 13, 813–820 (2013)PubMedPubMedCentralCrossRef
53.
go back to reference N. Jae, D.G. McEwan, Y. Manavski, R.A. Boon, S. Dimmeler, Rab7a and Rab27b control secretion of endothelial microRNA through extracellular vesicles. FEBS Lett. 589, 3182–3188 (2015)PubMedCrossRef N. Jae, D.G. McEwan, Y. Manavski, R.A. Boon, S. Dimmeler, Rab7a and Rab27b control secretion of endothelial microRNA through extracellular vesicles. FEBS Lett. 589, 3182–3188 (2015)PubMedCrossRef
54.
go back to reference S.N. Hurwitz, M.M. Conlon, M.A. Rider, N.C. Brownstein, D.G. Meckes Jr., Nanoparticle analysis sheds budding insights into genetic drivers of extracellular vesicle biogenesis. J. Extracell. Vesicles 5, 31295 (2016)PubMedCrossRef S.N. Hurwitz, M.M. Conlon, M.A. Rider, N.C. Brownstein, D.G. Meckes Jr., Nanoparticle analysis sheds budding insights into genetic drivers of extracellular vesicle biogenesis. J. Extracell. Vesicles 5, 31295 (2016)PubMedCrossRef
55.
go back to reference C. Hsu, Y. Morohashi, S. Yoshimura, N. Manrique-Hoyos, S. Jung, M.A. Lauterbach, M. Bakhti, M. Gronborg, W. Mobius, J. Rhee, F.A. Barr, M. Simons, Regulation of exosome secretion by Rab35 and its GTPase-activating proteins TBC1D10A-C. J. Cell Biol. 189, 223–232 (2010)PubMedPubMedCentralCrossRef C. Hsu, Y. Morohashi, S. Yoshimura, N. Manrique-Hoyos, S. Jung, M.A. Lauterbach, M. Bakhti, M. Gronborg, W. Mobius, J. Rhee, F.A. Barr, M. Simons, Regulation of exosome secretion by Rab35 and its GTPase-activating proteins TBC1D10A-C. J. Cell Biol. 189, 223–232 (2010)PubMedPubMedCentralCrossRef
56.
go back to reference C.A. Thompson, A. Purushothaman, V.C. Ramani, I. Vlodavsky, R.D. Sanderson, Heparanase regulates secretion, composition, and function of tumor cell-derived exosomes. J. Biol. Chem. 288, 10093–10099 (2013)PubMedPubMedCentralCrossRef C.A. Thompson, A. Purushothaman, V.C. Ramani, I. Vlodavsky, R.D. Sanderson, Heparanase regulates secretion, composition, and function of tumor cell-derived exosomes. J. Biol. Chem. 288, 10093–10099 (2013)PubMedPubMedCentralCrossRef
57.
go back to reference A. Savina, C.M. Fader, M.T. Damiani, M.I. Colombo, Rab11 promotes docking and fusion of multivesicular bodies in a calcium-dependent manner. Traffic 6, 131–143 (2005)PubMedCrossRef A. Savina, C.M. Fader, M.T. Damiani, M.I. Colombo, Rab11 promotes docking and fusion of multivesicular bodies in a calcium-dependent manner. Traffic 6, 131–143 (2005)PubMedCrossRef
58.
go back to reference H.W. King, M.Z. Michael, J.M. Gleadle, Hypoxic enhancement of exosome release by breast cancer cells. BMC Cancer 12, 1–10 (2012)CrossRef H.W. King, M.Z. Michael, J.M. Gleadle, Hypoxic enhancement of exosome release by breast cancer cells. BMC Cancer 12, 1–10 (2012)CrossRef
60.
go back to reference K. Al-Nedawi, B. Meehan, J. Micallef, V. Lhotak, L. May, A. Guha, J. Rak, Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat. Cell Biol. 10, 619–624 (2008)PubMedCrossRef K. Al-Nedawi, B. Meehan, J. Micallef, V. Lhotak, L. May, A. Guha, J. Rak, Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat. Cell Biol. 10, 619–624 (2008)PubMedCrossRef
61.
go back to reference M.M. Valenzuela, H.R. Ferguson Bennit, A. Gonda, C.J. Diaz Osterman, A. Hibma, S. Khan, N.R. Wall, Exosomes secreted from human cancer cell lines contain inhibitors of apoptosis (IAP). Cancer Microenviron. 8, 65–73 (2015)PubMedPubMedCentralCrossRef M.M. Valenzuela, H.R. Ferguson Bennit, A. Gonda, C.J. Diaz Osterman, A. Hibma, S. Khan, N.R. Wall, Exosomes secreted from human cancer cell lines contain inhibitors of apoptosis (IAP). Cancer Microenviron. 8, 65–73 (2015)PubMedPubMedCentralCrossRef
62.
go back to reference E. Donnarumma, D. Fiore, M. Nappa, G. Roscigno, A. Adamo, M. Iaboni, V. Russo, A. Affinito, I. Puoti, C. Quintavalle, A. Rienzo, S. Piscuoglio, R. Thomas, G. Condorelli, Cancer-associated fibroblasts release exosomal microRNAs that dictate an aggressive phenotype in breast cancer. Oncotarget 8, 19592–19608 (2017)PubMedPubMedCentralCrossRef E. Donnarumma, D. Fiore, M. Nappa, G. Roscigno, A. Adamo, M. Iaboni, V. Russo, A. Affinito, I. Puoti, C. Quintavalle, A. Rienzo, S. Piscuoglio, R. Thomas, G. Condorelli, Cancer-associated fibroblasts release exosomal microRNAs that dictate an aggressive phenotype in breast cancer. Oncotarget 8, 19592–19608 (2017)PubMedPubMedCentralCrossRef
63.
go back to reference A. Ramteke, H. Ting, C. Agarwal, S. Mateen, R. Somasagara, A. Hussain, M. Graner, B. Frederick, R. Agarwal, G. Deep, Exosomes secreted under hypoxia enhance invasiveness and stemness of prostate cancer cells by targeting adherens junction molecules. Mol. Carcinog. 54, 554–565 (2015)PubMedCrossRef A. Ramteke, H. Ting, C. Agarwal, S. Mateen, R. Somasagara, A. Hussain, M. Graner, B. Frederick, R. Agarwal, G. Deep, Exosomes secreted under hypoxia enhance invasiveness and stemness of prostate cancer cells by targeting adherens junction molecules. Mol. Carcinog. 54, 554–565 (2015)PubMedCrossRef
64.
go back to reference S. Bao, Q. Wu, S. Sathornsumetee, Y. Hao, Z. Li, A.B. Hjelmeland, Q. Shi, R.E. McLendon, D.D. Bigner, J.N. Rich, Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res. 66, 7843–7848 (2006)PubMedCrossRef S. Bao, Q. Wu, S. Sathornsumetee, Y. Hao, Z. Li, A.B. Hjelmeland, Q. Shi, R.E. McLendon, D.D. Bigner, J.N. Rich, Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res. 66, 7843–7848 (2006)PubMedCrossRef
65.
go back to reference L. Ricci-Vitiani, R. Pallini, M. Biffoni, M. Todaro, G. Invernici, T. Cenci, G. Maira, E.A. Parati, G. Stassi, L.M. Larocca, R. De Maria, Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature 468, 824–828 (2010)PubMedCrossRef L. Ricci-Vitiani, R. Pallini, M. Biffoni, M. Todaro, G. Invernici, T. Cenci, G. Maira, E.A. Parati, G. Stassi, L.M. Larocca, R. De Maria, Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature 468, 824–828 (2010)PubMedCrossRef
66.
67.
go back to reference C. Grange, M. Tapparo, F. Collino, L. Vitillo, C. Damasco, M.C. Deregibus, C. Tetta, B. Bussolati, G. Camussi, Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 71, 5346–5356 (2011)PubMedCrossRef C. Grange, M. Tapparo, F. Collino, L. Vitillo, C. Damasco, M.C. Deregibus, C. Tetta, B. Bussolati, G. Camussi, Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 71, 5346–5356 (2011)PubMedCrossRef
68.
go back to reference A. Conigliaro, V. Costa, A. Lo Dico, L. Saieva, S. Buccheri, F. Dieli, M. Manno, S. Raccosta, C. Mancone, M. Tripodi, G. De Leo, R. Alessandro, CD90+ liver cancer cells modulate endothelial cell phenotype through the release of exosomes containing H19 lncRNA. Mol. Cancer 14, 155 (2015)PubMedPubMedCentralCrossRef A. Conigliaro, V. Costa, A. Lo Dico, L. Saieva, S. Buccheri, F. Dieli, M. Manno, S. Raccosta, C. Mancone, M. Tripodi, G. De Leo, R. Alessandro, CD90+ liver cancer cells modulate endothelial cell phenotype through the release of exosomes containing H19 lncRNA. Mol. Cancer 14, 155 (2015)PubMedPubMedCentralCrossRef
69.
go back to reference E.J. Ekstrom, C. Bergenfelz, V. von Bulow, F. Serifler, E. Carlemalm, G. Jonsson, T. Andersson, K. Leandersson, WNT5A induces release of exosomes containing pro-angiogenic and immunosuppressive factors from malignant melanoma cells. Mol. Cancer 13, 88 (2014)PubMedPubMedCentralCrossRef E.J. Ekstrom, C. Bergenfelz, V. von Bulow, F. Serifler, E. Carlemalm, G. Jonsson, T. Andersson, K. Leandersson, WNT5A induces release of exosomes containing pro-angiogenic and immunosuppressive factors from malignant melanoma cells. Mol. Cancer 13, 88 (2014)PubMedPubMedCentralCrossRef
70.
go back to reference S.K. Gopal, D.W. Greening, E.G. Hanssen, H.J. Zhu, R.J. Simpson, R.A. Mathias, Oncogenic epithelial cell-derived exosomes containing Rac1 and PAK2 induce angiogenesis in recipient endothelial cells. Oncotarget 7, 19709–19722 (2016)PubMedPubMedCentral S.K. Gopal, D.W. Greening, E.G. Hanssen, H.J. Zhu, R.J. Simpson, R.A. Mathias, Oncogenic epithelial cell-derived exosomes containing Rac1 and PAK2 induce angiogenesis in recipient endothelial cells. Oncotarget 7, 19709–19722 (2016)PubMedPubMedCentral
71.
go back to reference Y. Liu, F. Luo, B. Wang, H. Li, Y. Xu, X. Liu, L. Shi, X. Lu, W. Xu, L. Lu, Y. Qin, Q. Xiang, Q. Liu, STAT3-regulated exosomal miR-21 promotes angiogenesis and is involved in neoplastic processes of transformed human bronchial epithelial cells. Cancer Lett. 370, 125–135 (2016)PubMedCrossRef Y. Liu, F. Luo, B. Wang, H. Li, Y. Xu, X. Liu, L. Shi, X. Lu, W. Xu, L. Lu, Y. Qin, Q. Xiang, Q. Liu, STAT3-regulated exosomal miR-21 promotes angiogenesis and is involved in neoplastic processes of transformed human bronchial epithelial cells. Cancer Lett. 370, 125–135 (2016)PubMedCrossRef
72.
go back to reference Y.K. Chan, H. Zhang, P. Liu, S.W. Tsao, M.L. Lung, N.K. Mak, R. Ngok-Shun Wong, P. Ying-Kit Yue, Proteomic analysis of exosomes from nasopharyngeal carcinoma cell identifies intercellular transfer of angiogenic proteins. Int. J. Cancer 137, 1830–1841 (2015)PubMedCrossRef Y.K. Chan, H. Zhang, P. Liu, S.W. Tsao, M.L. Lung, N.K. Mak, R. Ngok-Shun Wong, P. Ying-Kit Yue, Proteomic analysis of exosomes from nasopharyngeal carcinoma cell identifies intercellular transfer of angiogenic proteins. Int. J. Cancer 137, 1830–1841 (2015)PubMedCrossRef
73.
go back to reference K. Pakravan, S. Babashah, M. Sadeghizadeh, S.J. Mowla, M. Mossahebi-Mohammadi, F. Ataei, N. Dana, M. Javan, MicroRNA-100 shuttled by mesenchymal stem cell-derived exosomes suppresses in vitro angiogenesis through modulating the mTOR/HIF-1alpha/VEGF signaling axis in breast cancer cells. Cell. Oncol. 40, 457–470 (2017)CrossRef K. Pakravan, S. Babashah, M. Sadeghizadeh, S.J. Mowla, M. Mossahebi-Mohammadi, F. Ataei, N. Dana, M. Javan, MicroRNA-100 shuttled by mesenchymal stem cell-derived exosomes suppresses in vitro angiogenesis through modulating the mTOR/HIF-1alpha/VEGF signaling axis in breast cancer cells. Cell. Oncol. 40, 457–470 (2017)CrossRef
74.
go back to reference H. Tadokoro, T. Umezu, K. Ohyashiki, T. Hirano, J.H. Ohyashiki, Exosomes derived from hypoxic leukemia cells enhance tube formation in endothelial cells. J. Biol. Chem. 288, 34343–34351 (2013)PubMedPubMedCentralCrossRef H. Tadokoro, T. Umezu, K. Ohyashiki, T. Hirano, J.H. Ohyashiki, Exosomes derived from hypoxic leukemia cells enhance tube formation in endothelial cells. J. Biol. Chem. 288, 34343–34351 (2013)PubMedPubMedCentralCrossRef
75.
go back to reference T. Umezu, H. Tadokoro, K. Azuma, S. Yoshizawa, K. Ohyashiki, J.H. Ohyashiki, Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood 124, 3748–3757 (2014)PubMedPubMedCentralCrossRef T. Umezu, H. Tadokoro, K. Azuma, S. Yoshizawa, K. Ohyashiki, J.H. Ohyashiki, Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood 124, 3748–3757 (2014)PubMedPubMedCentralCrossRef
77.
go back to reference S. Shi, Q. Zhang, Y. Xia, B. You, Y. Shan, L. Bao, L. Li, Y. You, Z. Gu, Mesenchymal stem cell-derived exosomes facilitate nasopharyngeal carcinoma progression. Am. J. Cancer Res. 6, 459–472 (2016) S. Shi, Q. Zhang, Y. Xia, B. You, Y. Shan, L. Bao, L. Li, Y. You, Z. Gu, Mesenchymal stem cell-derived exosomes facilitate nasopharyngeal carcinoma progression. Am. J. Cancer Res. 6, 459–472 (2016)
78.
go back to reference C.A. Franzen, R.H. Blackwell, V. Todorovic, K.A. Greco, K.E. Foreman, R.C. Flanigan, P.C. Kuo, G.N. Gupta, Urothelial cells undergo epithelial-to-mesenchymal transition after exposure to muscle invasive bladder cancer exosomes. Oncogene 4, e163 (2015)CrossRef C.A. Franzen, R.H. Blackwell, V. Todorovic, K.A. Greco, K.E. Foreman, R.C. Flanigan, P.C. Kuo, G.N. Gupta, Urothelial cells undergo epithelial-to-mesenchymal transition after exposure to muscle invasive bladder cancer exosomes. Oncogene 4, e163 (2015)CrossRef
79.
go back to reference M. Aga, G.L. Bentz, S. Raffa, M.R. Torrisi, S. Kondo, N. Wakisaka, T. Yoshizaki, J.S. Pagano, J. Shackelford, Exosomal HIF1alpha supports invasive potential of nasopharyngeal carcinoma-associated LMP1-positive exosomes. Oncogene 33, 4613–4622 (2014)PubMedPubMedCentralCrossRef M. Aga, G.L. Bentz, S. Raffa, M.R. Torrisi, S. Kondo, N. Wakisaka, T. Yoshizaki, J.S. Pagano, J. Shackelford, Exosomal HIF1alpha supports invasive potential of nasopharyngeal carcinoma-associated LMP1-positive exosomes. Oncogene 33, 4613–4622 (2014)PubMedPubMedCentralCrossRef
80.
go back to reference W. Qin, Y. Tsukasaki, S. Dasgupta, N. Mukhopadhyay, M. Ikebe, E.R. Sauter, Exosomes in human breast milk promote emt. Clin. Cancer Res. 22, 4517–4524 (2016)PubMedCrossRef W. Qin, Y. Tsukasaki, S. Dasgupta, N. Mukhopadhyay, M. Ikebe, E.R. Sauter, Exosomes in human breast milk promote emt. Clin. Cancer Res. 22, 4517–4524 (2016)PubMedCrossRef
82.
go back to reference D. Xiao, S. Barry, D. Kmetz, M. Egger, J. Pan, S.N. Rai, J. Qu, K.M. McMasters, H. Hao, Melanoma cell-derived exosomes promote epithelial-mesenchymal transition in primary melanocytes through paracrine/autocrine signaling in the tumor microenvironment. Cancer Lett. 376, 318–327 (2016)PubMedPubMedCentralCrossRef D. Xiao, S. Barry, D. Kmetz, M. Egger, J. Pan, S.N. Rai, J. Qu, K.M. McMasters, H. Hao, Melanoma cell-derived exosomes promote epithelial-mesenchymal transition in primary melanocytes through paracrine/autocrine signaling in the tumor microenvironment. Cancer Lett. 376, 318–327 (2016)PubMedPubMedCentralCrossRef
83.
go back to reference I.J. Fidler, The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited. Nat. Rev. Cancer 3, 453–458 (2003)PubMedCrossRef I.J. Fidler, The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited. Nat. Rev. Cancer 3, 453–458 (2003)PubMedCrossRef
84.
go back to reference M. Rodriguez, J. Silva, A. Herrera, M. Herrera, C. Pena, P. Martin, B. Gil-Calderon, M.J. Larriba, M.J. Coronado, B. Soldevilla, V.S. Turrion, M. Provencio, A. Sanchez, F. Bonilla, V. Garcia-Barberan, Exosomes enriched in stemness/metastatic-related mRNAS promote oncogenic potential in breast cancer. Oncotarget 6, 40575–40587 (2015)PubMedPubMedCentralCrossRef M. Rodriguez, J. Silva, A. Herrera, M. Herrera, C. Pena, P. Martin, B. Gil-Calderon, M.J. Larriba, M.J. Coronado, B. Soldevilla, V.S. Turrion, M. Provencio, A. Sanchez, F. Bonilla, V. Garcia-Barberan, Exosomes enriched in stemness/metastatic-related mRNAS promote oncogenic potential in breast cancer. Oncotarget 6, 40575–40587 (2015)PubMedPubMedCentralCrossRef
85.
go back to reference T. Arita, D. Ichikawa, H. Konishi, S. Komatsu, A. Shiozaki, S. Ogino, Y. Fujita, H. Hiramoto, J. Hamada, K. Shoda, T. Kosuga, H. Fujiwara, K. Okamoto, E. Otsuji, Tumor exosome-mediated promotion of adhesion to mesothelial cells in gastric cancer cells. Oncotarget 7, 56855–56863 (2016)PubMedPubMedCentralCrossRef T. Arita, D. Ichikawa, H. Konishi, S. Komatsu, A. Shiozaki, S. Ogino, Y. Fujita, H. Hiramoto, J. Hamada, K. Shoda, T. Kosuga, H. Fujiwara, K. Okamoto, E. Otsuji, Tumor exosome-mediated promotion of adhesion to mesothelial cells in gastric cancer cells. Oncotarget 7, 56855–56863 (2016)PubMedPubMedCentralCrossRef
86.
go back to reference L. Li, C. Li, S. Wang, Z. Wang, J. Jiang, W. Wang, X. Li, J. Chen, K. Liu, C. Li, G. Zhu, Exosomes derived from hypoxic oral squamous cell carcinoma cells deliver mir-21 to normoxic cells to elicit a prometastatic phenotype. Cancer Res. 76, 1770–1780 (2016)PubMedCrossRef L. Li, C. Li, S. Wang, Z. Wang, J. Jiang, W. Wang, X. Li, J. Chen, K. Liu, C. Li, G. Zhu, Exosomes derived from hypoxic oral squamous cell carcinoma cells deliver mir-21 to normoxic cells to elicit a prometastatic phenotype. Cancer Res. 76, 1770–1780 (2016)PubMedCrossRef
87.
go back to reference J. Liao, R. Liu, Y.J. Shi, L.H. Yin, Y.P. Pu, Exosome-shuttling microRNA-21 promotes cell migration and invasion-targeting PDCD4 in esophageal cancer. Int. J. Oncol. 48, 2567–2579 (2016)PubMedCrossRef J. Liao, R. Liu, Y.J. Shi, L.H. Yin, Y.P. Pu, Exosome-shuttling microRNA-21 promotes cell migration and invasion-targeting PDCD4 in esophageal cancer. Int. J. Oncol. 48, 2567–2579 (2016)PubMedCrossRef
88.
go back to reference M. Yang, J. Chen, F. Su, B. Yu, F. Su, L. Lin, Y. Liu, J.-D. Huang, E. Song, Microvesicles secreted by macrophages shuttle invasion-potentiating microRNAs into breast cancer cells. Mol. Cancer 10, 1–13 (2011)CrossRef M. Yang, J. Chen, F. Su, B. Yu, F. Su, L. Lin, Y. Liu, J.-D. Huang, E. Song, Microvesicles secreted by macrophages shuttle invasion-potentiating microRNAs into breast cancer cells. Mol. Cancer 10, 1–13 (2011)CrossRef
89.
go back to reference X.L. Bai, Q. Zhang, L.Y. Ye, F. Liang, X. Sun, Y. Chen, Q.D. Hu, Q.H. Fu, W. Su, Z. Chen, Z.P. Zhuang, T.B. Liang, Myocyte enhancer factor 2C regulation of hepatocellular carcinoma via vascular endothelial growth factor and Wnt/beta-catenin signaling. Oncogene 34, 4089–4097 (2015)PubMedCrossRef X.L. Bai, Q. Zhang, L.Y. Ye, F. Liang, X. Sun, Y. Chen, Q.D. Hu, Q.H. Fu, W. Su, Z. Chen, Z.P. Zhuang, T.B. Liang, Myocyte enhancer factor 2C regulation of hepatocellular carcinoma via vascular endothelial growth factor and Wnt/beta-catenin signaling. Oncogene 34, 4089–4097 (2015)PubMedCrossRef
90.
go back to reference M.Y. Fong, W. Zhou, L. Liu, A.Y. Alontaga, M. Chandra, J. Ashby, A. Chow, S.T. O'Connor, S. Li, A.R. Chin, G. Somlo, M. Palomares, Z. Li, J.R. Tremblay, A. Tsuyada, G. Sun, M.A. Reid, X. Wu, P. Swiderski, X. Ren, Y. Shi, M. Kong, W. Zhong, Y. Chen, S.E. Wang, Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat. Cell Biol. 17, 183–194 (2015)PubMedPubMedCentralCrossRef M.Y. Fong, W. Zhou, L. Liu, A.Y. Alontaga, M. Chandra, J. Ashby, A. Chow, S.T. O'Connor, S. Li, A.R. Chin, G. Somlo, M. Palomares, Z. Li, J.R. Tremblay, A. Tsuyada, G. Sun, M.A. Reid, X. Wu, P. Swiderski, X. Ren, Y. Shi, M. Kong, W. Zhong, Y. Chen, S.E. Wang, Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat. Cell Biol. 17, 183–194 (2015)PubMedPubMedCentralCrossRef
91.
92.
go back to reference W.X. Chen, X.M. Liu, M.M. Lv, L. Chen, J.H. Zhao, S.L. Zhong, M.H. Ji, Q. Hu, Z. Luo, J.Z. Wu, J.H. Tang, Exosomes from drug-resistant breast cancer cells transmit chemoresistance by a horizontal transfer of microRNAs. PLoS One 9, e95240 (2014)PubMedPubMedCentralCrossRef W.X. Chen, X.M. Liu, M.M. Lv, L. Chen, J.H. Zhao, S.L. Zhong, M.H. Ji, Q. Hu, Z. Luo, J.Z. Wu, J.H. Tang, Exosomes from drug-resistant breast cancer cells transmit chemoresistance by a horizontal transfer of microRNAs. PLoS One 9, e95240 (2014)PubMedPubMedCentralCrossRef
93.
go back to reference M.M. Lv, X.Y. Zhu, W.X. Chen, S.L. Zhong, Q. Hu, T.F. Ma, J. Zhang, L. Chen, J.H. Tang, J.H. Zhao, Exosomes mediate drug resistance transfer in MCF-7 breast cancer cells and a probable mechanism is delivery of P-glycoprotein. Tumour Biol. 35, 10773–10779 (2014)PubMedCrossRef M.M. Lv, X.Y. Zhu, W.X. Chen, S.L. Zhong, Q. Hu, T.F. Ma, J. Zhang, L. Chen, J.H. Tang, J.H. Zhao, Exosomes mediate drug resistance transfer in MCF-7 breast cancer cells and a probable mechanism is delivery of P-glycoprotein. Tumour Biol. 35, 10773–10779 (2014)PubMedCrossRef
94.
go back to reference C.L. Au Yeung, N.N. Co, T. Tsuruga, T.L. Yeung, S.Y. Kwan, C.S. Leung, Y. Li, E.S. Lu, K. Kwan, K.K. Wong, R. Schmandt, K.H. Lu, S.C. Mok, Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat. Commun. 7, 11150 (2016)PubMedPubMedCentralCrossRef C.L. Au Yeung, N.N. Co, T. Tsuruga, T.L. Yeung, S.Y. Kwan, C.S. Leung, Y. Li, E.S. Lu, K. Kwan, K.K. Wong, R. Schmandt, K.H. Lu, S.C. Mok, Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat. Commun. 7, 11150 (2016)PubMedPubMedCentralCrossRef
95.
go back to reference Y. Hu, C. Yan, L. Mu, K. Huang, X. Li, D. Tao, Y. Wu, J. Qin, Fibroblast-derived exosomes contribute to chemoresistance through priming cancer stem cells in colorectal cancer. PLoS One 10, e0125625 (2015)PubMedPubMedCentralCrossRef Y. Hu, C. Yan, L. Mu, K. Huang, X. Li, D. Tao, Y. Wu, J. Qin, Fibroblast-derived exosomes contribute to chemoresistance through priming cancer stem cells in colorectal cancer. PLoS One 10, e0125625 (2015)PubMedPubMedCentralCrossRef
96.
go back to reference R. Ji, B. Zhang, X. Zhang, J. Xue, X. Yuan, Y. Yan, M. Wang, W. Zhu, H. Qian, W. Xu, Exosomes derived from human mesenchymal stem cells confer drug resistance in gastric cancer. Cell Cycle 14, 2473–2483 (2015)PubMedPubMedCentralCrossRef R. Ji, B. Zhang, X. Zhang, J. Xue, X. Yuan, Y. Yan, M. Wang, W. Zhu, H. Qian, W. Xu, Exosomes derived from human mesenchymal stem cells confer drug resistance in gastric cancer. Cell Cycle 14, 2473–2483 (2015)PubMedPubMedCentralCrossRef
97.
go back to reference T. Aung, B. Chapuy, D. Vogel, D. Wenzel, M. Oppermann, M. Lahmann, T. Weinhage, K. Menck, T. Hupfeld, R. Koch, L. Trumper, G.G. Wulf, Exosomal evasion of humoral immunotherapy in aggressive B-cell lymphoma modulated by ATP-binding cassette transporter A3. Proc. Natl. Acad. Sci. U. S. A. 108, 15336–15341 (2011)PubMedPubMedCentralCrossRef T. Aung, B. Chapuy, D. Vogel, D. Wenzel, M. Oppermann, M. Lahmann, T. Weinhage, K. Menck, T. Hupfeld, R. Koch, L. Trumper, G.G. Wulf, Exosomal evasion of humoral immunotherapy in aggressive B-cell lymphoma modulated by ATP-binding cassette transporter A3. Proc. Natl. Acad. Sci. U. S. A. 108, 15336–15341 (2011)PubMedPubMedCentralCrossRef
98.
go back to reference V. Ciravolo, V. Huber, G.C. Ghedini, E. Venturelli, F. Bianchi, M. Campiglio, D. Morelli, A. Villa, P. Della Mina, S. Menard, P. Filipazzi, L. Rivoltini, E. Tagliabue, S.M. Pupa, Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy. J. Cell. Physiol. 227, 658–667 (2012)PubMedCrossRef V. Ciravolo, V. Huber, G.C. Ghedini, E. Venturelli, F. Bianchi, M. Campiglio, D. Morelli, A. Villa, P. Della Mina, S. Menard, P. Filipazzi, L. Rivoltini, E. Tagliabue, S.M. Pupa, Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy. J. Cell. Physiol. 227, 658–667 (2012)PubMedCrossRef
99.
go back to reference R. Safaei, B.J. Larson, T.C. Cheng, M.A. Gibson, S. Otani, W. Naerdemann, S.B. Howell, Abnormal lysosomal trafficking and enhanced exosomal export of cisplatin in drug-resistant human ovarian carcinoma cells. Mol. Cancer Ther. 4, 1595–1604 (2005)PubMedCrossRef R. Safaei, B.J. Larson, T.C. Cheng, M.A. Gibson, S. Otani, W. Naerdemann, S.B. Howell, Abnormal lysosomal trafficking and enhanced exosomal export of cisplatin in drug-resistant human ovarian carcinoma cells. Mol. Cancer Ther. 4, 1595–1604 (2005)PubMedCrossRef
100.
go back to reference S. Loewer, M.N. Cabili, M. Guttman, Y.H. Loh, K. Thomas, I.H. Park, M. Garber, M. Curran, T. Onder, S. Agarwal, P.D. Manos, S. Datta, E.S. Lander, T.M. Schlaeger, G.Q. Daley, J.L. Rinn, Large intergenic non-coding RNA-RoR modulates reprogramming of human induced pluripotent stem cells. Nat. Genet. 42, 1113–1117 (2010)PubMedPubMedCentralCrossRef S. Loewer, M.N. Cabili, M. Guttman, Y.H. Loh, K. Thomas, I.H. Park, M. Garber, M. Curran, T. Onder, S. Agarwal, P.D. Manos, S. Datta, E.S. Lander, T.M. Schlaeger, G.Q. Daley, J.L. Rinn, Large intergenic non-coding RNA-RoR modulates reprogramming of human induced pluripotent stem cells. Nat. Genet. 42, 1113–1117 (2010)PubMedPubMedCentralCrossRef
101.
go back to reference Y. Pan, C. Li, J. Chen, K. Zhang, X. Chu, R. Wang, L. Chen, The emerging roles of long noncoding rna ror (lincrna-ror) and its possible mechanisms in human cancers. Cell. Physiol. Biochem. 40, 219–229 (2016)PubMedCrossRef Y. Pan, C. Li, J. Chen, K. Zhang, X. Chu, R. Wang, L. Chen, The emerging roles of long noncoding rna ror (lincrna-ror) and its possible mechanisms in human cancers. Cell. Physiol. Biochem. 40, 219–229 (2016)PubMedCrossRef
102.
go back to reference K. Takahashi, I.K. Yan, T. Kogure, H. Haga, T. Patel, Extracellular vesicle-mediated transfer of long non-coding RNA ROR modulates chemosensitivity in human hepatocellular cancer. FEBS Open Bio. 4, 458–467 (2014)PubMedPubMedCentralCrossRef K. Takahashi, I.K. Yan, T. Kogure, H. Haga, T. Patel, Extracellular vesicle-mediated transfer of long non-coding RNA ROR modulates chemosensitivity in human hepatocellular cancer. FEBS Open Bio. 4, 458–467 (2014)PubMedPubMedCentralCrossRef
103.
go back to reference J. Fan, Y. Xing, X. Wen, R. Jia, H. Ni, J. He, X. Ding, H. Pan, G. Qian, S. Ge, A.R. Hoffman, H. Zhang, X. Fan, Long non-coding RNA ROR decoys gene-specific histone methylation to promote tumorigenesis. Genome Biol. 16, 139 (2015)PubMedPubMedCentralCrossRef J. Fan, Y. Xing, X. Wen, R. Jia, H. Ni, J. He, X. Ding, H. Pan, G. Qian, S. Ge, A.R. Hoffman, H. Zhang, X. Fan, Long non-coding RNA ROR decoys gene-specific histone methylation to promote tumorigenesis. Genome Biol. 16, 139 (2015)PubMedPubMedCentralCrossRef
104.
go back to reference T.H. Cheung, T.A. Rando, Molecular regulation of stem cell quiescence. Nat. Rev. Mol. Cell Biol. 14, 329–340 (2013)PubMedCrossRef T.H. Cheung, T.A. Rando, Molecular regulation of stem cell quiescence. Nat. Rev. Mol. Cell Biol. 14, 329–340 (2013)PubMedCrossRef
105.
go back to reference P.K. Lim, S.A. Bliss, S.A. Patel, M. Taborga, M.A. Dave, L.A. Gregory, S.J. Greco, M. Bryan, P.S. Patel, P. Rameshwar, Gap junction-mediated import of microRNA from bone marrow stromal cells can elicit cell cycle quiescence in breast cancer cells. Cancer Res. 71, 1550–1560 (2011)PubMedCrossRef P.K. Lim, S.A. Bliss, S.A. Patel, M. Taborga, M.A. Dave, L.A. Gregory, S.J. Greco, M. Bryan, P.S. Patel, P. Rameshwar, Gap junction-mediated import of microRNA from bone marrow stromal cells can elicit cell cycle quiescence in breast cancer cells. Cancer Res. 71, 1550–1560 (2011)PubMedCrossRef
106.
go back to reference M. Ono, N. Kosaka, N. Tominaga, Y. Yoshioka, F. Takeshita, R.-u. Takahashi, M. Yoshida, H. Tsuda, K. Tamura, T. Ochiya, Exosomes from bone marrow mesenchymal stem cells contain a microRNA that promotes dormancy in metastatic breast cancer cells. Sci. Signal. 7, ra63 (2014)PubMedCrossRef M. Ono, N. Kosaka, N. Tominaga, Y. Yoshioka, F. Takeshita, R.-u. Takahashi, M. Yoshida, H. Tsuda, K. Tamura, T. Ochiya, Exosomes from bone marrow mesenchymal stem cells contain a microRNA that promotes dormancy in metastatic breast cancer cells. Sci. Signal. 7, ra63 (2014)PubMedCrossRef
107.
go back to reference S.A. Bliss, G. Sinha, O. Sandiford, L. Williams, D.J. Engelberth, K. Guiro, L.L. Isenalumhe, S.J. Greco, S. Ayer, M. Bryan, R. Kumar, N. Ponzio, P. Rameshwar, Mesenchymal stem cell-derived exosomes stimulates cycling quiescence and early breast cancer dormancy in bone marrow. Cancer Res. 76, 5832–5844 (2016)PubMedCrossRef S.A. Bliss, G. Sinha, O. Sandiford, L. Williams, D.J. Engelberth, K. Guiro, L.L. Isenalumhe, S.J. Greco, S. Ayer, M. Bryan, R. Kumar, N. Ponzio, P. Rameshwar, Mesenchymal stem cell-derived exosomes stimulates cycling quiescence and early breast cancer dormancy in bone marrow. Cancer Res. 76, 5832–5844 (2016)PubMedCrossRef
108.
go back to reference M. Collado, M.A. Blasco, M. Serrano, Cellular senescence in cancer and aging. Cell 130, 223–233 (2007)PubMedCrossRef M. Collado, M.A. Blasco, M. Serrano, Cellular senescence in cancer and aging. Cell 130, 223–233 (2007)PubMedCrossRef
110.
go back to reference H.D. Skinner, V.C. Sandulache, T.J. Ow, R.E. Meyn, J.S. Yordy, B.M. Beadle, A.L. Fitzgerald, U. Giri, K.K. Ang, J.N. Myers, TP53 disruptive mutations lead to head and neck cancer treatment failure through inhibition of radiation-induced senescence. Clin. Cancer Res. 18, 290–300 (2012)PubMedCrossRef H.D. Skinner, V.C. Sandulache, T.J. Ow, R.E. Meyn, J.S. Yordy, B.M. Beadle, A.L. Fitzgerald, U. Giri, K.K. Ang, J.N. Myers, TP53 disruptive mutations lead to head and neck cancer treatment failure through inhibition of radiation-induced senescence. Clin. Cancer Res. 18, 290–300 (2012)PubMedCrossRef
111.
go back to reference B. Jonchere, A. Vetillard, B. Toutain, D. Lam, A.C. Bernard, C. Henry, S. De Carne Trecesson, E. Gamelin, P. Juin, C. Guette, O. Coqueret, Irinotecan treatment and senescence failure promote the emergence of more transformed and invasive cells that depend on anti-apoptotic Mcl-1. Oncotarget 6, 409–426 (2015)PubMedCrossRef B. Jonchere, A. Vetillard, B. Toutain, D. Lam, A.C. Bernard, C. Henry, S. De Carne Trecesson, E. Gamelin, P. Juin, C. Guette, O. Coqueret, Irinotecan treatment and senescence failure promote the emergence of more transformed and invasive cells that depend on anti-apoptotic Mcl-1. Oncotarget 6, 409–426 (2015)PubMedCrossRef
112.
go back to reference A.L.C. Ong, T.S. Ramasamy, Role of sirtuin1-p53 regulatory axis in aging, cancer and cellular reprogramming. Ageing Res. Rev. 43, 64–80 (2018)PubMedCrossRef A.L.C. Ong, T.S. Ramasamy, Role of sirtuin1-p53 regulatory axis in aging, cancer and cellular reprogramming. Ageing Res. Rev. 43, 64–80 (2018)PubMedCrossRef
113.
go back to reference X. Yu, T. Riley, A.J. Levine, The regulation of the endosomal compartment by p53 the tumor suppressor gene. FEBS J. 276, 2201–2212 (2009)PubMedCrossRef X. Yu, T. Riley, A.J. Levine, The regulation of the endosomal compartment by p53 the tumor suppressor gene. FEBS J. 276, 2201–2212 (2009)PubMedCrossRef
114.
go back to reference Y. Sun, W. Zheng, Z. Guo, Q. Ju, L. Zhu, J. Gao, L. Zhou, F. Liu, Y. Xu, Q. Zhan, Z. Zhou, W. Sun, X. Zhao, A novel TP53 pathway influences the HGS-mediated exosome formation in colorectal cancer. Sci. Rep. 6, 28083 (2016)PubMedPubMedCentralCrossRef Y. Sun, W. Zheng, Z. Guo, Q. Ju, L. Zhu, J. Gao, L. Zhou, F. Liu, Y. Xu, Q. Zhan, Z. Zhou, W. Sun, X. Zhao, A novel TP53 pathway influences the HGS-mediated exosome formation in colorectal cancer. Sci. Rep. 6, 28083 (2016)PubMedPubMedCentralCrossRef
115.
go back to reference N. Malaquin, A. Martinez, F. Rodier, Keeping the senescence secretome under control: Molecular reins on the senescence-associated secretory phenotype. Exp. Gerontol. 82, 39–49 (2016)PubMedCrossRef N. Malaquin, A. Martinez, F. Rodier, Keeping the senescence secretome under control: Molecular reins on the senescence-associated secretory phenotype. Exp. Gerontol. 82, 39–49 (2016)PubMedCrossRef
116.
go back to reference J.P. Coppe, K. Kauser, J. Campisi, C.M. Beausejour, Secretion of vascular endothelial growth factor by primary human fibroblasts at senescence. J. Biol. Chem. 281, 29568–29574 (2006)PubMedCrossRef J.P. Coppe, K. Kauser, J. Campisi, C.M. Beausejour, Secretion of vascular endothelial growth factor by primary human fibroblasts at senescence. J. Biol. Chem. 281, 29568–29574 (2006)PubMedCrossRef
117.
go back to reference X. Sun, M. Vale, E. Leung, J.R. Kanwar, R. Gupta, G.W. Krissansen, Mouse B7-H3 induces antitumor immunity. Gene Ther. 10, 1728–1734 (2003)PubMedCrossRef X. Sun, M. Vale, E. Leung, J.R. Kanwar, R. Gupta, G.W. Krissansen, Mouse B7-H3 induces antitumor immunity. Gene Ther. 10, 1728–1734 (2003)PubMedCrossRef
118.
go back to reference B.D. Lehmann, M.S. Paine, A.M. Brooks, J.A. McCubrey, R.H. Renegar, R. Wang, D.M. Terrian, Senescence-associated exosome release from human prostate cancer cells. Cancer Res. 68, 7864–7871 (2008)PubMedCrossRef B.D. Lehmann, M.S. Paine, A.M. Brooks, J.A. McCubrey, R.H. Renegar, R. Wang, D.M. Terrian, Senescence-associated exosome release from human prostate cancer cells. Cancer Res. 68, 7864–7871 (2008)PubMedCrossRef
119.
go back to reference K. Weiner-Gorzel, E. Dempsey, M. Milewska, A. McGoldrick, V. Toh, A. Walsh, S. Lindsay, L. Gubbins, A. Cannon, D. Sharpe, J. O'Sullivan, M. Murphy, S.F. Madden, M. Kell, A. McCann, F. Furlong, Overexpression of the microRNA miR-433 promotes resistance to paclitaxel through the induction of cellular senescence in ovarian cancer cells. Cancer Med. 4, 745–758 (2015)PubMedPubMedCentralCrossRef K. Weiner-Gorzel, E. Dempsey, M. Milewska, A. McGoldrick, V. Toh, A. Walsh, S. Lindsay, L. Gubbins, A. Cannon, D. Sharpe, J. O'Sullivan, M. Murphy, S.F. Madden, M. Kell, A. McCann, F. Furlong, Overexpression of the microRNA miR-433 promotes resistance to paclitaxel through the induction of cellular senescence in ovarian cancer cells. Cancer Med. 4, 745–758 (2015)PubMedPubMedCentralCrossRef
120.
go back to reference F. Furlong, P. Fitzpatrick, S. O'Toole, S. Phelan, B. McGrogan, A. Maguire, A. O'Grady, M. Gallagher, M. Prencipe, A. McGoldrick, P. McGettigan, D. Brennan, O. Sheils, C. Martin, E. W. Kay, J. O'Leary, A. McCann, Low MAD2 expression levels associate with reduced progression-free survival in patients with high-grade serous epithelial ovarian cancer. J. Pathol. 226, 746–755 (2012)PubMedPubMedCentralCrossRef F. Furlong, P. Fitzpatrick, S. O'Toole, S. Phelan, B. McGrogan, A. Maguire, A. O'Grady, M. Gallagher, M. Prencipe, A. McGoldrick, P. McGettigan, D. Brennan, O. Sheils, C. Martin, E. W. Kay, J. O'Leary, A. McCann, Low MAD2 expression levels associate with reduced progression-free survival in patients with high-grade serous epithelial ovarian cancer. J. Pathol. 226, 746–755 (2012)PubMedPubMedCentralCrossRef
121.
go back to reference B.W. van Balkom, O.G. de Jong, M. Smits, J. Brummelman, K. den Ouden, P.M. de Bree, M.A. van Eijndhoven, D.M. Pegtel, W. Stoorvogel, T. Wurdinger, M.C. Verhaar, Endothelial cells require miR-214 to secrete exosomes that suppress senescence and induce angiogenesis in human and mouse endothelial cells. Blood 121, 3997–4006 (2013)PubMedCrossRef B.W. van Balkom, O.G. de Jong, M. Smits, J. Brummelman, K. den Ouden, P.M. de Bree, M.A. van Eijndhoven, D.M. Pegtel, W. Stoorvogel, T. Wurdinger, M.C. Verhaar, Endothelial cells require miR-214 to secrete exosomes that suppress senescence and induce angiogenesis in human and mouse endothelial cells. Blood 121, 3997–4006 (2013)PubMedCrossRef
122.
go back to reference S. Baroni, S. Romero-Cordoba, I. Plantamura, M. Dugo, E. D'Ippolito, A. Cataldo, G. Cosentino, V. Angeloni, A. Rossini, M.G. Daidone, M.V. Iorio, Exosome-mediated delivery of miR-9 induces cancer-associated fibroblast-like properties in human breast fibroblasts. Cell Death Dis. 7, e2312 (2016)PubMedPubMedCentralCrossRef S. Baroni, S. Romero-Cordoba, I. Plantamura, M. Dugo, E. D'Ippolito, A. Cataldo, G. Cosentino, V. Angeloni, A. Rossini, M.G. Daidone, M.V. Iorio, Exosome-mediated delivery of miR-9 induces cancer-associated fibroblast-like properties in human breast fibroblasts. Cell Death Dis. 7, e2312 (2016)PubMedPubMedCentralCrossRef
123.
go back to reference A. Gutkin, O. Uziel, E. Beery, J. Nordenberg, M. Pinchasi, H. Goldvaser, S. Henick, M. Goldberg, M. Lahav, Tumor cells derived exosomes contain hTERT mRNA and transform nonmalignant fibroblasts into telomerase positive cells. Oncotarget 7, 59173–59188 (2016)PubMedPubMedCentralCrossRef A. Gutkin, O. Uziel, E. Beery, J. Nordenberg, M. Pinchasi, H. Goldvaser, S. Henick, M. Goldberg, M. Lahav, Tumor cells derived exosomes contain hTERT mRNA and transform nonmalignant fibroblasts into telomerase positive cells. Oncotarget 7, 59173–59188 (2016)PubMedPubMedCentralCrossRef
124.
go back to reference J. Gu, H. Qian, L. Shen, X. Zhang, W. Zhu, L. Huang, Y. Yan, F. Mao, C. Zhao, Y. Shi, W. Xu, Gastric cancer exosomes trigger differentiation of umbilical cord derived mesenchymal stem cells to carcinoma-associated fibroblasts through TGF-beta/Smad pathway. PLoS One 7, e52465 (2012)PubMedPubMedCentralCrossRef J. Gu, H. Qian, L. Shen, X. Zhang, W. Zhu, L. Huang, Y. Yan, F. Mao, C. Zhao, Y. Shi, W. Xu, Gastric cancer exosomes trigger differentiation of umbilical cord derived mesenchymal stem cells to carcinoma-associated fibroblasts through TGF-beta/Smad pathway. PLoS One 7, e52465 (2012)PubMedPubMedCentralCrossRef
125.
go back to reference J. Webber, R. Steadman, M.D. Mason, Z. Tabi, A. Clayton, Cancer exosomes trigger fibroblast to myofibroblast differentiation. Cancer Res. 70, 9621–9630 (2010)PubMedCrossRef J. Webber, R. Steadman, M.D. Mason, Z. Tabi, A. Clayton, Cancer exosomes trigger fibroblast to myofibroblast differentiation. Cancer Res. 70, 9621–9630 (2010)PubMedCrossRef
126.
go back to reference A. Orimo, Y. Tomioka, Y. Shimizu, M. Sato, S. Oigawa, K. Kamata, Y. Nogi, S. Inoue, M. Takahashi, T. Hata, M. Muramatsu, Cancer-associated myofibroblasts possess various factors to promote endometrial tumor progression. Clin. Cancer Res. 7, 3097–3105 (2001)PubMed A. Orimo, Y. Tomioka, Y. Shimizu, M. Sato, S. Oigawa, K. Kamata, Y. Nogi, S. Inoue, M. Takahashi, T. Hata, M. Muramatsu, Cancer-associated myofibroblasts possess various factors to promote endometrial tumor progression. Clin. Cancer Res. 7, 3097–3105 (2001)PubMed
127.
go back to reference L.M. Sobral, A. Bufalino, M.A. Lopes, E. Graner, T. Salo, R.D. Coletta, Myofibroblasts in the stroma of oral cancer promote tumorigenesis via secretion of activin A. Oral Oncol. 47, 840–846 (2011)PubMedCrossRef L.M. Sobral, A. Bufalino, M.A. Lopes, E. Graner, T. Salo, R.D. Coletta, Myofibroblasts in the stroma of oral cancer promote tumorigenesis via secretion of activin A. Oral Oncol. 47, 840–846 (2011)PubMedCrossRef
128.
129.
go back to reference C. Corrado, L. Saieva, S. Raimondo, A. Santoro, G. De Leo, R. Alessandro, Chronic myelogenous leukaemia exosomes modulate bone marrow microenvironment through activation of epidermal growth factor receptor. J. Cell. Mol. Med. 20, 1829–1839 (2016)PubMedPubMedCentralCrossRef C. Corrado, L. Saieva, S. Raimondo, A. Santoro, G. De Leo, R. Alessandro, Chronic myelogenous leukaemia exosomes modulate bone marrow microenvironment through activation of epidermal growth factor receptor. J. Cell. Mol. Med. 20, 1829–1839 (2016)PubMedPubMedCentralCrossRef
130.
go back to reference L. Wu, X. Zhang, B. Zhang, H. Shi, X. Yuan, Y. Sun, Z. Pan, H. Qian, W. Xu, Exosomes derived from gastric cancer cells activate NF-kappaB pathway in macrophages to promote cancer progression. Tumour Biol. 37, 12169–12180 (2016)PubMedCrossRef L. Wu, X. Zhang, B. Zhang, H. Shi, X. Yuan, Y. Sun, Z. Pan, H. Qian, W. Xu, Exosomes derived from gastric cancer cells activate NF-kappaB pathway in macrophages to promote cancer progression. Tumour Biol. 37, 12169–12180 (2016)PubMedCrossRef
131.
132.
go back to reference L.R. Languino, A. Singh, M. Prisco, G.J. Inman, A. Luginbuhl, J.M. Curry, A.P. South, Exosome-mediated transfer from the tumor microenvironment increases TGFbeta signaling in squamous cell carcinoma. Am. J. Transl. Res. 8, 2432–2437 (2016)PubMedPubMedCentral L.R. Languino, A. Singh, M. Prisco, G.J. Inman, A. Luginbuhl, J.M. Curry, A.P. South, Exosome-mediated transfer from the tumor microenvironment increases TGFbeta signaling in squamous cell carcinoma. Am. J. Transl. Res. 8, 2432–2437 (2016)PubMedPubMedCentral
133.
go back to reference M.C. Boelens, T.J. Wu, B.Y. Nabet, B. Xu, Y. Qiu, T. Yoon, D.J. Azzam, C. Twyman-Saint Victor, B.Z. Wiemann, H. Ishwaran, P.J. Ter Brugge, J. Jonkers, J. Slingerland, A.J. Minn, Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways. Cell 159, 499–513 (2014)PubMedPubMedCentralCrossRef M.C. Boelens, T.J. Wu, B.Y. Nabet, B. Xu, Y. Qiu, T. Yoon, D.J. Azzam, C. Twyman-Saint Victor, B.Z. Wiemann, H. Ishwaran, P.J. Ter Brugge, J. Jonkers, J. Slingerland, A.J. Minn, Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways. Cell 159, 499–513 (2014)PubMedPubMedCentralCrossRef
134.
go back to reference D.J. Prockop, Marrow stromal cells as stem cells for nonhematopoietic tissues. Science 276, 71–74 (1997)PubMedCrossRef D.J. Prockop, Marrow stromal cells as stem cells for nonhematopoietic tissues. Science 276, 71–74 (1997)PubMedCrossRef
135.
136.
go back to reference B. Cousin, E. Ravet, S. Poglio, F. De Toni, M. Bertuzzi, H. Lulka, I. Touil, M. Andre, J.L. Grolleau, J.M. Peron, J.P. Chavoin, P. Bourin, L. Penicaud, L. Casteilla, L. Buscail, P. Cordelier, Adult stromal cells derived from human adipose tissue provoke pancreatic cancer cell death both in vitro and in vivo. PLoS One 4, e6278 (2009)PubMedPubMedCentralCrossRef B. Cousin, E. Ravet, S. Poglio, F. De Toni, M. Bertuzzi, H. Lulka, I. Touil, M. Andre, J.L. Grolleau, J.M. Peron, J.P. Chavoin, P. Bourin, L. Penicaud, L. Casteilla, L. Buscail, P. Cordelier, Adult stromal cells derived from human adipose tissue provoke pancreatic cancer cell death both in vitro and in vivo. PLoS One 4, e6278 (2009)PubMedPubMedCentralCrossRef
137.
go back to reference L. Qiao, Z.L. Xu, T.J. Zhao, L.H. Ye, X.D. Zhang, Dkk-1 secreted by mesenchymal stem cells inhibits growth of breast cancer cells via depression of Wnt signalling. Cancer Lett. 269, 67–77 (2008)PubMedCrossRef L. Qiao, Z.L. Xu, T.J. Zhao, L.H. Ye, X.D. Zhang, Dkk-1 secreted by mesenchymal stem cells inhibits growth of breast cancer cells via depression of Wnt signalling. Cancer Lett. 269, 67–77 (2008)PubMedCrossRef
138.
go back to reference Y. Yulyana, I.A. Ho, K.C. Sia, J.P. Newman, X.Y. Toh, B.B. Endaya, J.K. Chan, M. Gnecchi, H. Huynh, A.Y. Chung, K.H. Lim, H.S. Leong, N.G. Iyer, K.M. Hui, P.Y. Lam, Paracrine factors of human fetal MSCs inhibit liver cancer growth through reduced activation of IGF-1R/PI3K/Akt signaling. Mol. Ther. 23, 746–756 (2015)PubMedPubMedCentralCrossRef Y. Yulyana, I.A. Ho, K.C. Sia, J.P. Newman, X.Y. Toh, B.B. Endaya, J.K. Chan, M. Gnecchi, H. Huynh, A.Y. Chung, K.H. Lim, H.S. Leong, N.G. Iyer, K.M. Hui, P.Y. Lam, Paracrine factors of human fetal MSCs inhibit liver cancer growth through reduced activation of IGF-1R/PI3K/Akt signaling. Mol. Ther. 23, 746–756 (2015)PubMedPubMedCentralCrossRef
139.
go back to reference O. Attar-Schneider, V. Zismanov, L. Drucker, M. Gottfried, Secretome of human bone marrow mesenchymal stem cells: an emerging player in lung cancer progression and mechanisms of translation initiation. Tumor Biol. 37, 4755–4765 (2016)CrossRef O. Attar-Schneider, V. Zismanov, L. Drucker, M. Gottfried, Secretome of human bone marrow mesenchymal stem cells: an emerging player in lung cancer progression and mechanisms of translation initiation. Tumor Biol. 37, 4755–4765 (2016)CrossRef
140.
go back to reference J.K. Lee, S.R. Park, B.K. Jung, Y.K. Jeon, Y.S. Lee, M.K. Kim, Y.G. Kim, J.Y. Jang, C.W. Kim, Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells. PLoS One 8, e84256 (2013)PubMedPubMedCentralCrossRef J.K. Lee, S.R. Park, B.K. Jung, Y.K. Jeon, Y.S. Lee, M.K. Kim, Y.G. Kim, J.Y. Jang, C.W. Kim, Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells. PLoS One 8, e84256 (2013)PubMedPubMedCentralCrossRef
141.
go back to reference A.Y. Khakoo, S. Pati, S.A. Anderson, W. Reid, M.F. Elshal, I.I. Rovira, A.T. Nguyen, D. Malide, C.A. Combs, G. Hall, J. Zhang, M. Raffeld, T.B. Rogers, W. Stetler-Stevenson, J.A. Frank, M. Reitz, T. Finkel, Human mesenchymal stem cells exert potent antitumorigenic effects in a model of Kaposi's sarcoma. J. Exp. Med. 203, 1235–1247 (2006)PubMedPubMedCentralCrossRef A.Y. Khakoo, S. Pati, S.A. Anderson, W. Reid, M.F. Elshal, I.I. Rovira, A.T. Nguyen, D. Malide, C.A. Combs, G. Hall, J. Zhang, M. Raffeld, T.B. Rogers, W. Stetler-Stevenson, J.A. Frank, M. Reitz, T. Finkel, Human mesenchymal stem cells exert potent antitumorigenic effects in a model of Kaposi's sarcoma. J. Exp. Med. 203, 1235–1247 (2006)PubMedPubMedCentralCrossRef
142.
go back to reference J.F. Ji, B.P. He, S.T. Dheen, S.S.W. Tay, Interactions of chemokines and chemokine receptors mediate the migration of mesenchymal stem cells to the impaired site in the brain after hypoglossal nerve injury. Stem Cells 22, 415–427 (2004)PubMedCrossRef J.F. Ji, B.P. He, S.T. Dheen, S.S.W. Tay, Interactions of chemokines and chemokine receptors mediate the migration of mesenchymal stem cells to the impaired site in the brain after hypoglossal nerve injury. Stem Cells 22, 415–427 (2004)PubMedCrossRef
143.
go back to reference B.M. Beckermann, G. Kallifatidis, A. Groth, D. Frommhold, A. Apel, J. Mattern, A.V. Salnikov, G. Moldenhauer, W. Wagner, A. Diehlmann, R. Saffrich, M. Schubert, A.D. Ho, N. Giese, M.W. Buchler, H. Friess, P. Buchler, I. Herr, VEGF expression by mesenchymal stem cells contributes to angiogenesis in pancreatic carcinoma. Br. J. Cancer 99, 622–631 (2008)PubMedPubMedCentralCrossRef B.M. Beckermann, G. Kallifatidis, A. Groth, D. Frommhold, A. Apel, J. Mattern, A.V. Salnikov, G. Moldenhauer, W. Wagner, A. Diehlmann, R. Saffrich, M. Schubert, A.D. Ho, N. Giese, M.W. Buchler, H. Friess, P. Buchler, I. Herr, VEGF expression by mesenchymal stem cells contributes to angiogenesis in pancreatic carcinoma. Br. J. Cancer 99, 622–631 (2008)PubMedPubMedCentralCrossRef
144.
go back to reference A. Schmidt, D. Ladage, T. Schinköthe, U. Klausmann, C. Ulrichs, F.J. Klinz, K. Brixius, S. Arnhold, B. Desai, U. Mehlhorn, R.H.G. Schwinger, P. Staib, K. Addicks, W. Bloch, Basic fibroblast growth factor controls migration in human mesenchymal stem cells. Stem Cells 24, 1750–1758 (2006)PubMedCrossRef A. Schmidt, D. Ladage, T. Schinköthe, U. Klausmann, C. Ulrichs, F.J. Klinz, K. Brixius, S. Arnhold, B. Desai, U. Mehlhorn, R.H.G. Schwinger, P. Staib, K. Addicks, W. Bloch, Basic fibroblast growth factor controls migration in human mesenchymal stem cells. Stem Cells 24, 1750–1758 (2006)PubMedCrossRef
145.
go back to reference C. Ke, J. Chen, Y. Guo, Z.W. Chen, J. Cai, Migration mechanism of mesenchymal stem cells studied by QD/NSOM. Biochim. Biophys. Acta Biomembr. 1848, 859–868 (2015)CrossRef C. Ke, J. Chen, Y. Guo, Z.W. Chen, J. Cai, Migration mechanism of mesenchymal stem cells studied by QD/NSOM. Biochim. Biophys. Acta Biomembr. 1848, 859–868 (2015)CrossRef
146.
go back to reference G. Ren, X. Zhao, Y. Wang, X. Zhang, X. Chen, C. Xu, Z.R. Yuan, A.I. Roberts, L. Zhang, B. Zheng, T. Wen, Y. Han, A.B. Rabson, J.A. Tischfield, C. Shao, Y. Shi, CCR2-dependent recruitment of macrophages by tumor-educated mesenchymal stromal cells promotes tumor development and is mimicked by TNFalpha. Cell Stem Cell 11, 812–824 (2012)PubMedPubMedCentralCrossRef G. Ren, X. Zhao, Y. Wang, X. Zhang, X. Chen, C. Xu, Z.R. Yuan, A.I. Roberts, L. Zhang, B. Zheng, T. Wen, Y. Han, A.B. Rabson, J.A. Tischfield, C. Shao, Y. Shi, CCR2-dependent recruitment of macrophages by tumor-educated mesenchymal stromal cells promotes tumor development and is mimicked by TNFalpha. Cell Stem Cell 11, 812–824 (2012)PubMedPubMedCentralCrossRef
147.
go back to reference B.G. Cuiffo, A. Campagne, G.W. Bell, A. Lembo, F. Orso, E.C. Lien, M.K. Bhasin, M. Raimo, S.E. Hanson, A. Marusyk, D. El-Ashry, P. Hematti, K. Polyak, F. Mechta-Grigoriou, O. Mariani, S. Volinia, A. Vincent-Salomon, D. Taverna, A.E. Karnoub, MSC-regulated microRNAs converge on the transcription factor FOXP2 and promote breast cancer metastasis. Cell Stem Cell 15, 762–774 (2014)PubMedCrossRef B.G. Cuiffo, A. Campagne, G.W. Bell, A. Lembo, F. Orso, E.C. Lien, M.K. Bhasin, M. Raimo, S.E. Hanson, A. Marusyk, D. El-Ashry, P. Hematti, K. Polyak, F. Mechta-Grigoriou, O. Mariani, S. Volinia, A. Vincent-Salomon, D. Taverna, A.E. Karnoub, MSC-regulated microRNAs converge on the transcription factor FOXP2 and promote breast cancer metastasis. Cell Stem Cell 15, 762–774 (2014)PubMedCrossRef
148.
go back to reference A. De Boeck, P. Pauwels, K. Hensen, J.L. Rummens, W. Westbroek, A. Hendrix, D. Maynard, H. Denys, K. Lambein, G. Braems, C. Gespach, M. Bracke, O. De Wever, Bone marrow-derived mesenchymal stem cells promote colorectal cancer progression through paracrine neuregulin 1/HER3 signalling. Gut 62, 550–560 (2013)PubMedCrossRef A. De Boeck, P. Pauwels, K. Hensen, J.L. Rummens, W. Westbroek, A. Hendrix, D. Maynard, H. Denys, K. Lambein, G. Braems, C. Gespach, M. Bracke, O. De Wever, Bone marrow-derived mesenchymal stem cells promote colorectal cancer progression through paracrine neuregulin 1/HER3 signalling. Gut 62, 550–560 (2013)PubMedCrossRef
149.
go back to reference Y. Huang, P. Yu, W. Li, G. Ren, A.I. Roberts, W. Cao, X. Zhang, J. Su, X. Chen, Q. Chen, P. Shou, C. Xu, L. Du, L. Lin, N. Xie, L. Zhang, Y. Wang, Y. Shi, p53 regulates mesenchymal stem cell-mediated tumor suppression in a tumor microenvironment through immune modulation. Oncogene 33, 3830–3838 (2014)PubMedCrossRef Y. Huang, P. Yu, W. Li, G. Ren, A.I. Roberts, W. Cao, X. Zhang, J. Su, X. Chen, Q. Chen, P. Shou, C. Xu, L. Du, L. Lin, N. Xie, L. Zhang, Y. Wang, Y. Shi, p53 regulates mesenchymal stem cell-mediated tumor suppression in a tumor microenvironment through immune modulation. Oncogene 33, 3830–3838 (2014)PubMedCrossRef
150.
go back to reference K. McLean, Y. Gong, Y. Choi, N. Deng, K. Yang, S. Bai, L. Cabrera, E. Keller, L. McCauley, K.R. Cho, R.J. Buckanovich, Human ovarian carcinoma-associated mesenchymal stem cells regulate cancer stem cells and tumorigenesis via altered BMP production. J. Clin. Invest. 121, 3206–3219 (2011)PubMedPubMedCentralCrossRef K. McLean, Y. Gong, Y. Choi, N. Deng, K. Yang, S. Bai, L. Cabrera, E. Keller, L. McCauley, K.R. Cho, R.J. Buckanovich, Human ovarian carcinoma-associated mesenchymal stem cells regulate cancer stem cells and tumorigenesis via altered BMP production. J. Clin. Invest. 121, 3206–3219 (2011)PubMedPubMedCentralCrossRef
151.
go back to reference F. Vianello, F. Villanova, V. Tisato, S. Lymperi, K.-K. Ho, A.R. Gomes, D. Marin, D. Bonnet, J. Apperley, E.W.F. Lam, F. Dazzi, Bone marrow mesenchymal stromal cells non-selectively protect chronic myeloid leukemia cells from imatinib-induced apoptosis via the CXCR4/CXCL12 axis. Haematologica 95, 1081–1089 (2010)PubMedPubMedCentralCrossRef F. Vianello, F. Villanova, V. Tisato, S. Lymperi, K.-K. Ho, A.R. Gomes, D. Marin, D. Bonnet, J. Apperley, E.W.F. Lam, F. Dazzi, Bone marrow mesenchymal stromal cells non-selectively protect chronic myeloid leukemia cells from imatinib-induced apoptosis via the CXCR4/CXCL12 axis. Haematologica 95, 1081–1089 (2010)PubMedPubMedCentralCrossRef
152.
go back to reference L.Y. Lin, L.M. Du, K. Cao, Y. Huang, P.F. Yu, L.Y. Zhang, F.Y. Li, Y. Wang, Y.F. Shi, Tumour cell-derived exosomes endow mesenchymal stromal cells with tumour-promotion capabilities. Oncogene 35, 6038–6042 (2016)PubMedPubMedCentralCrossRef L.Y. Lin, L.M. Du, K. Cao, Y. Huang, P.F. Yu, L.Y. Zhang, F.Y. Li, Y. Wang, Y.F. Shi, Tumour cell-derived exosomes endow mesenchymal stromal cells with tumour-promotion capabilities. Oncogene 35, 6038–6042 (2016)PubMedPubMedCentralCrossRef
153.
go back to reference X. Song, Y. Ding, G. Liu, X. Yang, R. Zhao, Y. Zhang, X. Zhao, G.J. Anderson, G. Nie, Cancer Cell-derived exosomes induce mitogen-activated protein kinase-dependent monocyte survival by transport of functional receptor tyrosine kinases. J. Biol. Chem. 291, 8453–8464 (2016)PubMedPubMedCentralCrossRef X. Song, Y. Ding, G. Liu, X. Yang, R. Zhao, Y. Zhang, X. Zhao, G.J. Anderson, G. Nie, Cancer Cell-derived exosomes induce mitogen-activated protein kinase-dependent monocyte survival by transport of functional receptor tyrosine kinases. J. Biol. Chem. 291, 8453–8464 (2016)PubMedPubMedCentralCrossRef
154.
go back to reference J. Choi, J. Gyamfi, H. Jang and J.S. Koo, The role of tumor-associated macrophage in breast cancer biology. Histol. Histopathol. 33, 133–145 (2018) J. Choi, J. Gyamfi, H. Jang and J.S. Koo, The role of tumor-associated macrophage in breast cancer biology. Histol. Histopathol. 33, 133–145 (2018)
155.
go back to reference F. Leonard, L.T. Curtis, M.J. Ware, T. Nosrat, X. Liu, K. Yokoi, H.B. Frieboes, B. Godin, Macrophage polarization contributes to the anti-tumoral efficacy of mesoporous nanovectors loaded with albumin-bound paclitaxel. Front. Immunol. 8, 693 (2017)PubMedPubMedCentralCrossRef F. Leonard, L.T. Curtis, M.J. Ware, T. Nosrat, X. Liu, K. Yokoi, H.B. Frieboes, B. Godin, Macrophage polarization contributes to the anti-tumoral efficacy of mesoporous nanovectors loaded with albumin-bound paclitaxel. Front. Immunol. 8, 693 (2017)PubMedPubMedCentralCrossRef
156.
go back to reference Z. Chen, X. Feng, C.J. Herting, V. Alvarez Garcia, K. Nie, W.W. Pong, R. Rasmussen, B. Dwivedi, S. Seby, S.A. Wolf, D.H. Gutmann, D. Hambardzumyan, Cellular and molecular identity of tumor-associated macrophages in glioblastoma. Cancer Res. 77, 2266–2278 (2017)PubMedPubMedCentralCrossRef Z. Chen, X. Feng, C.J. Herting, V. Alvarez Garcia, K. Nie, W.W. Pong, R. Rasmussen, B. Dwivedi, S. Seby, S.A. Wolf, D.H. Gutmann, D. Hambardzumyan, Cellular and molecular identity of tumor-associated macrophages in glioblastoma. Cancer Res. 77, 2266–2278 (2017)PubMedPubMedCentralCrossRef
157.
go back to reference M. Yin, X. Li, S. Tan, H.J. Zhou, W. Ji, S. Bellone, X. Xu, H. Zhang, A.D. Santin, G. Lou, W. Min, Tumor-associated macrophages drive spheroid formation during early transcoelomic metastasis of ovarian cancer. J. Clin. Invest. 126, 4157–4173 (2016)PubMedPubMedCentralCrossRef M. Yin, X. Li, S. Tan, H.J. Zhou, W. Ji, S. Bellone, X. Xu, H. Zhang, A.D. Santin, G. Lou, W. Min, Tumor-associated macrophages drive spheroid formation during early transcoelomic metastasis of ovarian cancer. J. Clin. Invest. 126, 4157–4173 (2016)PubMedPubMedCentralCrossRef
158.
go back to reference H. Shinohara, Y. Kuranaga, M. Kumazaki, N. Sugito, Y. Yoshikawa, T. Takai, K. Taniguchi, Y. Ito, Y. Akao, Regulated polarization of tumor-associated macrophages by mir-145 via colorectal cancer–derived extracellular vesicles. J. Immunol. 199, 1505–1515 (2017)PubMedCrossRef H. Shinohara, Y. Kuranaga, M. Kumazaki, N. Sugito, Y. Yoshikawa, T. Takai, K. Taniguchi, Y. Ito, Y. Akao, Regulated polarization of tumor-associated macrophages by mir-145 via colorectal cancer–derived extracellular vesicles. J. Immunol. 199, 1505–1515 (2017)PubMedCrossRef
159.
go back to reference J. Wang, K. De Veirman, S. Faict, M.A. Frassanito, D. Ribatti, A. Vacca, E. Menu, Multiple myeloma exosomes establish a favourable bone marrow microenvironment with enhanced angiogenesis and immunosuppression. J. Pathol. 239, 162–173 (2016)PubMedCrossRef J. Wang, K. De Veirman, S. Faict, M.A. Frassanito, D. Ribatti, A. Vacca, E. Menu, Multiple myeloma exosomes establish a favourable bone marrow microenvironment with enhanced angiogenesis and immunosuppression. J. Pathol. 239, 162–173 (2016)PubMedCrossRef
160.
go back to reference U. Putz, J. Howitt, A. Doan, C.-P. Goh, L.-H. Low, J. Silke, S.-S. Tan, The tumor suppressor pten is exported in exosomes and has phosphatase activity in recipient cells. Sci. Signal. 5, ra70 (2012)PubMedCrossRef U. Putz, J. Howitt, A. Doan, C.-P. Goh, L.-H. Low, J. Silke, S.-S. Tan, The tumor suppressor pten is exported in exosomes and has phosphatase activity in recipient cells. Sci. Signal. 5, ra70 (2012)PubMedCrossRef
161.
go back to reference A.M.M.T. Reza, Y.-J. Choi, H. Yasuda, J.-H. Kim, Human adipose mesenchymal stem cell-derived exosomal-miRNAs are critical factors for inducing anti-proliferation signalling to A2780 and SKOV-3 ovarian cancer cells. Sci. Rep. 6, 38498 (2016)PubMedPubMedCentralCrossRef A.M.M.T. Reza, Y.-J. Choi, H. Yasuda, J.-H. Kim, Human adipose mesenchymal stem cell-derived exosomal-miRNAs are critical factors for inducing anti-proliferation signalling to A2780 and SKOV-3 ovarian cancer cells. Sci. Rep. 6, 38498 (2016)PubMedPubMedCentralCrossRef
162.
go back to reference S.F. Ko, H.K. Yip, Y.Y. Zhen, C.C. Lee, C.C. Lee, C.C. Huang, S.H. Ng, J.W. Lin, Adipose-derived mesenchymal stem cell exosomes suppress hepatocellular carcinoma growth in a rat model: apparent diffusion coefficient, natural killer t-cell responses, and histopathological features. Stem Cells Int. 2015, 853506 (2015)PubMedPubMedCentralCrossRef S.F. Ko, H.K. Yip, Y.Y. Zhen, C.C. Lee, C.C. Lee, C.C. Huang, S.H. Ng, J.W. Lin, Adipose-derived mesenchymal stem cell exosomes suppress hepatocellular carcinoma growth in a rat model: apparent diffusion coefficient, natural killer t-cell responses, and histopathological features. Stem Cells Int. 2015, 853506 (2015)PubMedPubMedCentralCrossRef
163.
go back to reference F. Alcayaga-Miranda, P.L. Gonzalez, A. Lopez-Verrilli, M. Varas-Godoy, C. Aguila-Diaz, L. Contreras, M. Khoury, Prostate tumor-induced angiogenesis is blocked by exosomes derived from menstrual stem cells through the inhibition of reactive oxygen species. Oncotarget 7, 44462–44477 (2016)PubMedPubMedCentralCrossRef F. Alcayaga-Miranda, P.L. Gonzalez, A. Lopez-Verrilli, M. Varas-Godoy, C. Aguila-Diaz, L. Contreras, M. Khoury, Prostate tumor-induced angiogenesis is blocked by exosomes derived from menstrual stem cells through the inhibition of reactive oxygen species. Oncotarget 7, 44462–44477 (2016)PubMedPubMedCentralCrossRef
164.
go back to reference H.D. Lee, B.H. Koo, Y.H. Kim, O.H. Jeon, D.S. Kim, Exosome release of ADAM15 and the functional implications of human macrophage-derived ADAM15 exosomes. FASEB J. 26, 3084–3095 (2012)PubMedCrossRef H.D. Lee, B.H. Koo, Y.H. Kim, O.H. Jeon, D.S. Kim, Exosome release of ADAM15 and the functional implications of human macrophage-derived ADAM15 exosomes. FASEB J. 26, 3084–3095 (2012)PubMedCrossRef
165.
go back to reference B. Costa-Silva, N.M. Aiello, A.J. Ocean, S. Singh, H. Zhang, B.K. Thakur, A. Becker, A. Hoshino, M.T. Mark, H. Molina, J. Xiang, T. Zhang, T.M. Theilen, G. Garcia-Santos, C. Williams, Y. Ararso, Y. Huang, G. Rodrigues, T.L. Shen, K.J. Labori, I.M. Lothe, E.H. Kure, J. Hernandez, A. Doussot, S.H. Ebbesen, P.M. Grandgenett, M.A. Hollingsworth, M. Jain, K. Mallya, S.K. Batra, W.R. Jarnagin, R.E. Schwartz, I. Matei, H. Peinado, B.Z. Stanger, J. Bromberg, D. Lyden, Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 17, 816–826 (2015)PubMedPubMedCentralCrossRef B. Costa-Silva, N.M. Aiello, A.J. Ocean, S. Singh, H. Zhang, B.K. Thakur, A. Becker, A. Hoshino, M.T. Mark, H. Molina, J. Xiang, T. Zhang, T.M. Theilen, G. Garcia-Santos, C. Williams, Y. Ararso, Y. Huang, G. Rodrigues, T.L. Shen, K.J. Labori, I.M. Lothe, E.H. Kure, J. Hernandez, A. Doussot, S.H. Ebbesen, P.M. Grandgenett, M.A. Hollingsworth, M. Jain, K. Mallya, S.K. Batra, W.R. Jarnagin, R.E. Schwartz, I. Matei, H. Peinado, B.Z. Stanger, J. Bromberg, D. Lyden, Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 17, 816–826 (2015)PubMedPubMedCentralCrossRef
166.
go back to reference J. Sceneay, M.J. Smyth, A. Moller, The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 32, 449–464 (2013)PubMedCrossRef J. Sceneay, M.J. Smyth, A. Moller, The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 32, 449–464 (2013)PubMedCrossRef
167.
go back to reference H. Peinado, M. Aleckovic, S. Lavotshkin, I. Matei, B. Costa-Silva, G. Moreno-Bueno, M. Hergueta-Redondo, C. Williams, G. Garcia-Santos, C. Ghajar, A. Nitadori-Hoshino, C. Hoffman, K. Badal, B.A. Garcia, M.K. Callahan, J. Yuan, V.R. Martins, J. Skog, R.N. Kaplan, M.S. Brady, J.D. Wolchok, P.B. Chapman, Y. Kang, J. Bromberg, D. Lyden, Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 18, 883–891 (2012)PubMedPubMedCentralCrossRef H. Peinado, M. Aleckovic, S. Lavotshkin, I. Matei, B. Costa-Silva, G. Moreno-Bueno, M. Hergueta-Redondo, C. Williams, G. Garcia-Santos, C. Ghajar, A. Nitadori-Hoshino, C. Hoffman, K. Badal, B.A. Garcia, M.K. Callahan, J. Yuan, V.R. Martins, J. Skog, R.N. Kaplan, M.S. Brady, J.D. Wolchok, P.B. Chapman, Y. Kang, J. Bromberg, D. Lyden, Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 18, 883–891 (2012)PubMedPubMedCentralCrossRef
168.
go back to reference T. Jung, D. Castellana, P. Klingbeil, I. Cuesta Hernandez, M. Vitacolonna, D.J. Orlicky, S.R. Roffler, P. Brodt, M. Zoller, CD44v6 dependence of premetastatic niche preparation by exosomes. Neoplasia 11, 1093–1105 (2009)PubMedPubMedCentralCrossRef T. Jung, D. Castellana, P. Klingbeil, I. Cuesta Hernandez, M. Vitacolonna, D.J. Orlicky, S.R. Roffler, P. Brodt, M. Zoller, CD44v6 dependence of premetastatic niche preparation by exosomes. Neoplasia 11, 1093–1105 (2009)PubMedPubMedCentralCrossRef
169.
go back to reference J.L. Hood, R.S. San and S.A. Wickline, Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 71, 3792–3801 (2011) J.L. Hood, R.S. San and S.A. Wickline, Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 71, 3792–3801 (2011)
170.
go back to reference C.A. Sanchez, E.I. Andahur, R. Valenzuela, E.A. Castellon, J.A. Fulla, C.G. Ramos, J.C. Trivino, Exosomes from bulk and stem cells from human prostate cancer have a differential microRNA content that contributes cooperatively over local and pre-metastatic niche. Oncotarget 7, 3993–4008 (2016)PubMedCrossRef C.A. Sanchez, E.I. Andahur, R. Valenzuela, E.A. Castellon, J.A. Fulla, C.G. Ramos, J.C. Trivino, Exosomes from bulk and stem cells from human prostate cancer have a differential microRNA content that contributes cooperatively over local and pre-metastatic niche. Oncotarget 7, 3993–4008 (2016)PubMedCrossRef
171.
go back to reference A. Hoshino, B. Costa-Silva, T.L. Shen, G. Rodrigues, A. Hashimoto, M. Tesic Mark, H. Molina, S. Kohsaka, A. Di Giannatale, S. Ceder, S. Singh, C. Williams, N. Soplop, K. Uryu, L. Pharmer, T. King, L. Bojmar, A.E. Davies, Y. Ararso, T. Zhang, H. Zhang, J. Hernandez, J.M. Weiss, V.D. Dumont-Cole, K. Kramer, L.H. Wexler, A. Narendran, G.K. Schwartz, J.H. Healey, P. Sandstrom, K.J. Labori, E.H. Kure, P.M. Grandgenett, M.A. Hollingsworth, M. de Sousa, S. Kaur, M. Jain, K. Mallya, S.K. Batra, W.R. Jarnagin, M.S. Brady, O. Fodstad, V. Muller, K. Pantel, A.J. Minn, M.J. Bissell, B.A. Garcia, Y. Kang, V.K. Rajasekhar, C.M. Ghajar, I. Matei, H. Peinado, J. Bromberg, D. Lyden, Tumour exosome integrins determine organotropic metastasis. Nature 527, 329–335 (2015)PubMedPubMedCentralCrossRef A. Hoshino, B. Costa-Silva, T.L. Shen, G. Rodrigues, A. Hashimoto, M. Tesic Mark, H. Molina, S. Kohsaka, A. Di Giannatale, S. Ceder, S. Singh, C. Williams, N. Soplop, K. Uryu, L. Pharmer, T. King, L. Bojmar, A.E. Davies, Y. Ararso, T. Zhang, H. Zhang, J. Hernandez, J.M. Weiss, V.D. Dumont-Cole, K. Kramer, L.H. Wexler, A. Narendran, G.K. Schwartz, J.H. Healey, P. Sandstrom, K.J. Labori, E.H. Kure, P.M. Grandgenett, M.A. Hollingsworth, M. de Sousa, S. Kaur, M. Jain, K. Mallya, S.K. Batra, W.R. Jarnagin, M.S. Brady, O. Fodstad, V. Muller, K. Pantel, A.J. Minn, M.J. Bissell, B.A. Garcia, Y. Kang, V.K. Rajasekhar, C.M. Ghajar, I. Matei, H. Peinado, J. Bromberg, D. Lyden, Tumour exosome integrins determine organotropic metastasis. Nature 527, 329–335 (2015)PubMedPubMedCentralCrossRef
172.
go back to reference S. Keller, J. Ridinger, A.K. Rupp, J.W. Janssen, P. Altevogt, Body fluid derived exosomes as a novel template for clinical diagnostics. J. Transl. Med. 9, 86 (2011)PubMedPubMedCentralCrossRef S. Keller, J. Ridinger, A.K. Rupp, J.W. Janssen, P. Altevogt, Body fluid derived exosomes as a novel template for clinical diagnostics. J. Transl. Med. 9, 86 (2011)PubMedPubMedCentralCrossRef
173.
go back to reference C.L. Chen, Y.F. Lai, P. Tang, K.Y. Chien, J.S. Yu, C.H. Tsai, H.W. Chen, C.C. Wu, T. Chung, C.W. Hsu, C.D. Chen, Y.S. Chang, P.L. Chang, Y.T. Chen, Comparative and targeted proteomic analyses of urinary microparticles from bladder cancer and hernia patients. J. Proteome Res. 11, 5611–5629 (2012)PubMedCrossRef C.L. Chen, Y.F. Lai, P. Tang, K.Y. Chien, J.S. Yu, C.H. Tsai, H.W. Chen, C.C. Wu, T. Chung, C.W. Hsu, C.D. Chen, Y.S. Chang, P.L. Chang, Y.T. Chen, Comparative and targeted proteomic analyses of urinary microparticles from bladder cancer and hernia patients. J. Proteome Res. 11, 5611–5629 (2012)PubMedCrossRef
174.
go back to reference A. Kannan, R.B. Wells, S. Sivakumar, S. Komatsu, K.P. Singh, B. Samten, J.V. Philley, E.R. Sauter, M. Ikebe, S. Idell, S. Gupta, S. Dasgupta, Mitochondrial reprogramming regulates breast cancer progression. Clin. Cancer Res. 22, 3348–3360 (2016)PubMedCrossRef A. Kannan, R.B. Wells, S. Sivakumar, S. Komatsu, K.P. Singh, B. Samten, J.V. Philley, E.R. Sauter, M. Ikebe, S. Idell, S. Gupta, S. Dasgupta, Mitochondrial reprogramming regulates breast cancer progression. Clin. Cancer Res. 22, 3348–3360 (2016)PubMedCrossRef
175.
go back to reference M.J. Donovan, M. Noerholm, S. Bentink, S. Belzer, J. Skog, V. O'Neill, J.S. Cochran, G.A. Brown, A molecular signature of PCA3 and ERG exosomal RNA from non-DRE urine is predictive of initial prostate biopsy result. Prostate Cancer Prostatic Dis. 18, 370–375 (2015)PubMedCrossRef M.J. Donovan, M. Noerholm, S. Bentink, S. Belzer, J. Skog, V. O'Neill, J.S. Cochran, G.A. Brown, A molecular signature of PCA3 and ERG exosomal RNA from non-DRE urine is predictive of initial prostate biopsy result. Prostate Cancer Prostatic Dis. 18, 370–375 (2015)PubMedCrossRef
176.
go back to reference M. He, H. Qin, T.C. Poon, S.C. Sze, X. Ding, N.N. Co, S.M. Ngai, T.F. Chan, N. Wong, Hepatocellular carcinoma-derived exosomes promote motility of immortalized hepatocyte through transfer of oncogenic proteins and RNAs. Carcinogenesis 36, 1008–1018 (2015)PubMedCrossRef M. He, H. Qin, T.C. Poon, S.C. Sze, X. Ding, N.N. Co, S.M. Ngai, T.F. Chan, N. Wong, Hepatocellular carcinoma-derived exosomes promote motility of immortalized hepatocyte through transfer of oncogenic proteins and RNAs. Carcinogenesis 36, 1008–1018 (2015)PubMedCrossRef
177.
go back to reference G.K. Joshi, S. Deitz-McElyea, T. Liyanage, K. Lawrence, S. Mali, R. Sardar, M. Korc, Label-free nanoplasmonic-based short noncoding rna sensing at attomolar concentrations allows for quantitative and highly specific assay of microrna-10b in biological fluids and circulating exosomes. ACS Nano 9, 11075–11089 (2015)PubMedPubMedCentralCrossRef G.K. Joshi, S. Deitz-McElyea, T. Liyanage, K. Lawrence, S. Mali, R. Sardar, M. Korc, Label-free nanoplasmonic-based short noncoding rna sensing at attomolar concentrations allows for quantitative and highly specific assay of microrna-10b in biological fluids and circulating exosomes. ACS Nano 9, 11075–11089 (2015)PubMedPubMedCentralCrossRef
178.
go back to reference L. Manterola, E. Guruceaga, J. Gallego Perez-Larraya, M. Gonzalez-Huarriz, P. Jauregui, S. Tejada, R. Diez-Valle, V. Segura, N. Sampron, C. Barrena, I. Ruiz, A. Agirre, A. Ayuso, J. Rodriguez, A. Gonzalez, E. Xipell, A. Matheu, A. Lopez de Munain, T. Tunon, I. Zazpe, J. Garcia-Foncillas, S. Paris, J.Y. Delattre, M.M. Alonso, A small noncoding RNA signature found in exosomes of GBM patient serum as a diagnostic tool. Neuro-Oncology 16, 520–527 (2014)PubMedPubMedCentralCrossRef L. Manterola, E. Guruceaga, J. Gallego Perez-Larraya, M. Gonzalez-Huarriz, P. Jauregui, S. Tejada, R. Diez-Valle, V. Segura, N. Sampron, C. Barrena, I. Ruiz, A. Agirre, A. Ayuso, J. Rodriguez, A. Gonzalez, E. Xipell, A. Matheu, A. Lopez de Munain, T. Tunon, I. Zazpe, J. Garcia-Foncillas, S. Paris, J.Y. Delattre, M.M. Alonso, A small noncoding RNA signature found in exosomes of GBM patient serum as a diagnostic tool. Neuro-Oncology 16, 520–527 (2014)PubMedPubMedCentralCrossRef
179.
go back to reference J. Skog, T. Wurdinger, S. van Rijn, D.H. Meijer, L. Gainche, W.T. Curry, B.S. Carter, A.M. Krichevsky, X.O. Breakefield, Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat. Cell Biol. 10, 1470–1476 (2008)PubMedPubMedCentralCrossRef J. Skog, T. Wurdinger, S. van Rijn, D.H. Meijer, L. Gainche, W.T. Curry, B.S. Carter, A.M. Krichevsky, X.O. Breakefield, Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat. Cell Biol. 10, 1470–1476 (2008)PubMedPubMedCentralCrossRef
180.
go back to reference P. Kharaziha, D. Chioureas, D. Rutishauser, G. Baltatzis, L. Lennartsson, P. Fonseca, A. Azimi, K. Hultenby, R. Zubarev, A. Ullen, J. Yachnin, S. Nilsson, T. Panaretakis, Molecular profiling of prostate cancer derived exosomes may reveal a predictive signature for response to docetaxel. Oncotarget 6, 21740–21754 (2015)PubMedPubMedCentral P. Kharaziha, D. Chioureas, D. Rutishauser, G. Baltatzis, L. Lennartsson, P. Fonseca, A. Azimi, K. Hultenby, R. Zubarev, A. Ullen, J. Yachnin, S. Nilsson, T. Panaretakis, Molecular profiling of prostate cancer derived exosomes may reveal a predictive signature for response to docetaxel. Oncotarget 6, 21740–21754 (2015)PubMedPubMedCentral
181.
go back to reference K. Kawakami, Y. Fujita, T. Kato, K. Mizutani, K. Kameyama, H. Tsumoto, Y. Miura, T. Deguchi, M. Ito, Integrin beta4 and vinculin contained in exosomes are potential markers for progression of prostate cancer associated with taxane-resistance. Int. J. Oncol. 47, 384–390 (2015)PubMedCrossRef K. Kawakami, Y. Fujita, T. Kato, K. Mizutani, K. Kameyama, H. Tsumoto, Y. Miura, T. Deguchi, M. Ito, Integrin beta4 and vinculin contained in exosomes are potential markers for progression of prostate cancer associated with taxane-resistance. Int. J. Oncol. 47, 384–390 (2015)PubMedCrossRef
182.
go back to reference Y.Y. Yeh, H.G. Ozer, A.M. Lehman, K. Maddocks, L. Yu, A.J. Johnson, J.C. Byrd, Characterization of CLL exosomes reveals a distinct microRNA signature and enhanced secretion by activation of BCR signaling. Blood 125, 3297–3305 (2015)PubMedPubMedCentralCrossRef Y.Y. Yeh, H.G. Ozer, A.M. Lehman, K. Maddocks, L. Yu, A.J. Johnson, J.C. Byrd, Characterization of CLL exosomes reveals a distinct microRNA signature and enhanced secretion by activation of BCR signaling. Blood 125, 3297–3305 (2015)PubMedPubMedCentralCrossRef
183.
go back to reference J. Silva, V. Garcia, M. Rodriguez, M. Compte, E. Cisneros, P. Veguillas, J.M. Garcia, G. Dominguez, Y. Campos-Martin, J. Cuevas, C. Pena, M. Herrera, R. Diaz, N. Mohammed, F. Bonilla, Analysis of exosome release and its prognostic value in human colorectal cancer. Genes Chromosom. Cancer 51, 409–418 (2012)PubMedCrossRef J. Silva, V. Garcia, M. Rodriguez, M. Compte, E. Cisneros, P. Veguillas, J.M. Garcia, G. Dominguez, Y. Campos-Martin, J. Cuevas, C. Pena, M. Herrera, R. Diaz, N. Mohammed, F. Bonilla, Analysis of exosome release and its prognostic value in human colorectal cancer. Genes Chromosom. Cancer 51, 409–418 (2012)PubMedCrossRef
184.
go back to reference N. Kosaka, H. Iguchi, Y. Yoshioka, F. Takeshita, Y. Matsuki, T. Ochiya, Secretory mechanisms and intercellular transfer of microRNAs in living cells. J. Biol. Chem. 285, 17442–17452 (2010)PubMedPubMedCentralCrossRef N. Kosaka, H. Iguchi, Y. Yoshioka, F. Takeshita, Y. Matsuki, T. Ochiya, Secretory mechanisms and intercellular transfer of microRNAs in living cells. J. Biol. Chem. 285, 17442–17452 (2010)PubMedPubMedCentralCrossRef
185.
go back to reference M. Fabbri, A. Paone, F. Calore, R. Galli, E. Gaudio, R. Santhanam, F. Lovat, P. Fadda, C. Mao, G.J. Nuovo, N. Zanesi, M. Crawford, G.H. Ozer, D. Wernicke, H. Alder, M.A. Caligiuri, P. Nana-Sinkam, D. Perrotti, C.M. Croce, MicroRNAs bind to toll-like receptors to induce prometastatic inflammatory response. Proc. Natl. Acad. Sci. U. S. A. 109, E2110–E2116 (2012)PubMedPubMedCentralCrossRef M. Fabbri, A. Paone, F. Calore, R. Galli, E. Gaudio, R. Santhanam, F. Lovat, P. Fadda, C. Mao, G.J. Nuovo, N. Zanesi, M. Crawford, G.H. Ozer, D. Wernicke, H. Alder, M.A. Caligiuri, P. Nana-Sinkam, D. Perrotti, C.M. Croce, MicroRNAs bind to toll-like receptors to induce prometastatic inflammatory response. Proc. Natl. Acad. Sci. U. S. A. 109, E2110–E2116 (2012)PubMedPubMedCentralCrossRef
186.
go back to reference F. Chalmin, S. Ladoire, G. Mignot, J. Vincent, M. Bruchard, J.P. Remy-Martin, W. Boireau, A. Rouleau, B. Simon, D. Lanneau, A. De Thonel, G. Multhoff, A. Hamman, F. Martin, B. Chauffert, E. Solary, L. Zitvogel, C. Garrido, B. Ryffel, C. Borg, L. Apetoh, C. Rebe, F. Ghiringhelli, Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J. Clin. Invest. 120, 457–471 (2010)PubMedPubMedCentral F. Chalmin, S. Ladoire, G. Mignot, J. Vincent, M. Bruchard, J.P. Remy-Martin, W. Boireau, A. Rouleau, B. Simon, D. Lanneau, A. De Thonel, G. Multhoff, A. Hamman, F. Martin, B. Chauffert, E. Solary, L. Zitvogel, C. Garrido, B. Ryffel, C. Borg, L. Apetoh, C. Rebe, F. Ghiringhelli, Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J. Clin. Invest. 120, 457–471 (2010)PubMedPubMedCentral
187.
go back to reference A. Bobrie, S. Krumeich, F. Reyal, C. Recchi, L.F. Moita, M.C. Seabra, M. Ostrowski, C. Théry, Rab27a supports exosome-dependent and -independent mechanisms that modify the tumor microenvironment and can promote tumor progression. Cancer Res. 72, 4920–4930 (2012)PubMedCrossRef A. Bobrie, S. Krumeich, F. Reyal, C. Recchi, L.F. Moita, M.C. Seabra, M. Ostrowski, C. Théry, Rab27a supports exosome-dependent and -independent mechanisms that modify the tumor microenvironment and can promote tumor progression. Cancer Res. 72, 4920–4930 (2012)PubMedCrossRef
188.
go back to reference M. Ruiz-Martinez, A. Navarro, R.M. Marrades, N. Vinolas, S. Santasusagna, C. Munoz, J. Ramirez, L. Molins, M. Monzo, YKT6 expression, exosome release, and survival in non-small cell lung cancer. Oncotarget 7, 51515–51524 (2016)PubMedPubMedCentral M. Ruiz-Martinez, A. Navarro, R.M. Marrades, N. Vinolas, S. Santasusagna, C. Munoz, J. Ramirez, L. Molins, M. Monzo, YKT6 expression, exosome release, and survival in non-small cell lung cancer. Oncotarget 7, 51515–51524 (2016)PubMedPubMedCentral
189.
go back to reference A. Bosque, L. Dietz, A. Gallego-Lleyda, M. Sanclemente, M. Iturralde, J. Naval, M.A. Alava, L. Martinez-Lostao, H.J. Thierse, A. Anel, Comparative proteomics of exosomes secreted by tumoral Jurkat T cells and normal human T cell blasts unravels a potential tumorigenic role for valosin-containing protein. Oncotarget 7, 29287–29305 (2016)PubMedPubMedCentralCrossRef A. Bosque, L. Dietz, A. Gallego-Lleyda, M. Sanclemente, M. Iturralde, J. Naval, M.A. Alava, L. Martinez-Lostao, H.J. Thierse, A. Anel, Comparative proteomics of exosomes secreted by tumoral Jurkat T cells and normal human T cell blasts unravels a potential tumorigenic role for valosin-containing protein. Oncotarget 7, 29287–29305 (2016)PubMedPubMedCentralCrossRef
190.
go back to reference C. Federici, F. Petrucci, S. Caimi, A. Cesolini, M. Logozzi, M. Borghi, S. D'Ilio, L. Lugini, N. Violante, T. Azzarito, C. Majorani, D. Brambilla, S. Fais, Exosome release and low pH belong to a framework of resistance of human melanoma cells to cisplatin. PLoS One 9, e88193 (2014)PubMedPubMedCentralCrossRef C. Federici, F. Petrucci, S. Caimi, A. Cesolini, M. Logozzi, M. Borghi, S. D'Ilio, L. Lugini, N. Violante, T. Azzarito, C. Majorani, D. Brambilla, S. Fais, Exosome release and low pH belong to a framework of resistance of human melanoma cells to cisplatin. PLoS One 9, e88193 (2014)PubMedPubMedCentralCrossRef
191.
go back to reference X.Q. Li, J.T. Liu, L.L. Fan, Y. Liu, L. Cheng, F. Wang, H.Q. Yu, J. Gao, W. Wei, H. Wang, G.P. Sun, Exosomes derived from gefitinib-treated EGFR-mutant lung cancer cells alter cisplatin sensitivity via up-regulating autophagy. Oncotarget 7, 24585–24595 (2016)PubMedPubMedCentral X.Q. Li, J.T. Liu, L.L. Fan, Y. Liu, L. Cheng, F. Wang, H.Q. Yu, J. Gao, W. Wei, H. Wang, G.P. Sun, Exosomes derived from gefitinib-treated EGFR-mutant lung cancer cells alter cisplatin sensitivity via up-regulating autophagy. Oncotarget 7, 24585–24595 (2016)PubMedPubMedCentral
192.
go back to reference H.G. Zhang, H. Kim, C. Liu, S. Yu, J. Wang, W.E. Grizzle, R.P. Kimberly, S. Barnes, Curcumin reverses breast tumor exosomes mediated immune suppression of NK cell tumor cytotoxicity. Biochim. Biophys. Acta 1773, 1116–1123 (2007)PubMedPubMedCentralCrossRef H.G. Zhang, H. Kim, C. Liu, S. Yu, J. Wang, W.E. Grizzle, R.P. Kimberly, S. Barnes, Curcumin reverses breast tumor exosomes mediated immune suppression of NK cell tumor cytotoxicity. Biochim. Biophys. Acta 1773, 1116–1123 (2007)PubMedPubMedCentralCrossRef
193.
go back to reference T.S. Ramasamy, A.Z. Ayob, H.H. Myint, S. Thiagarajah, F. Amini, Targeting colorectal cancer stem cells using curcumin and curcumin analogues: insights into the mechanism of the therapeutic efficacy. Cancer Cell Int. 15, 96 (2015)PubMedPubMedCentralCrossRef T.S. Ramasamy, A.Z. Ayob, H.H. Myint, S. Thiagarajah, F. Amini, Targeting colorectal cancer stem cells using curcumin and curcumin analogues: insights into the mechanism of the therapeutic efficacy. Cancer Cell Int. 15, 96 (2015)PubMedPubMedCentralCrossRef
194.
go back to reference S. Amigorena, Cancer immunotherapy using dendritic cell-derived exosomes. Medicina (B Aires) 60 Suppl 2, 51–54 (2000) S. Amigorena, Cancer immunotherapy using dendritic cell-derived exosomes. Medicina (B Aires) 60 Suppl 2, 51–54 (2000)
195.
go back to reference G.G. Romagnoli, B.B. Zelante, P.A. Toniolo, I.K. Migliori, J.A.M. Barbuto, Dendritic cell-derived exosomes may be a tool for cancer immunotherapy by converting tumor cells into immunogenic targets. Front. Immunol. 5, 692 (2014)PubMed G.G. Romagnoli, B.B. Zelante, P.A. Toniolo, I.K. Migliori, J.A.M. Barbuto, Dendritic cell-derived exosomes may be a tool for cancer immunotherapy by converting tumor cells into immunogenic targets. Front. Immunol. 5, 692 (2014)PubMed
196.
go back to reference Q. Rao, B. Zuo, Z. Lu, X. Gao, A. You, C. Wu, Z. Du, H. Yin, Tumor-derived exosomes elicit tumor suppression in murine hepatocellular carcinoma models and human in vitro. Hepatology 64, 456–472 (2016)PubMedCrossRef Q. Rao, B. Zuo, Z. Lu, X. Gao, A. You, C. Wu, Z. Du, H. Yin, Tumor-derived exosomes elicit tumor suppression in murine hepatocellular carcinoma models and human in vitro. Hepatology 64, 456–472 (2016)PubMedCrossRef
197.
go back to reference J. Wang, L. Wang, Z. Lin, L. Tao, M. Chen, More efficient induction of antitumor T cell immunity by exosomes from CD40L gene-modified lung tumor cells. Mol. Med. Rep. 9, 125–131 (2014)PubMedCrossRef J. Wang, L. Wang, Z. Lin, L. Tao, M. Chen, More efficient induction of antitumor T cell immunity by exosomes from CD40L gene-modified lung tumor cells. Mol. Med. Rep. 9, 125–131 (2014)PubMedCrossRef
198.
go back to reference M. Damo, D.S. Wilson, E. Simeoni, J.A. Hubbell, TLR-3 stimulation improves anti-tumor immunity elicited by dendritic cell exosome-based vaccines in a murine model of melanoma. Sci. Rep. 5, 17622 (2015)PubMedPubMedCentralCrossRef M. Damo, D.S. Wilson, E. Simeoni, J.A. Hubbell, TLR-3 stimulation improves anti-tumor immunity elicited by dendritic cell exosome-based vaccines in a murine model of melanoma. Sci. Rep. 5, 17622 (2015)PubMedPubMedCentralCrossRef
199.
go back to reference Y. Xie, O. Bai, H. Zhang, J. Yuan, S. Zong, R. Chibbar, K. Slattery, M. Qureshi, Y. Wei, Y. Deng, J. Xiang, Membrane-bound HSP70-engineered myeloma cell-derived exosomes stimulate more efficient CD8(+) CTL- and NK-mediated antitumour immunity than exosomes released from heat-shocked tumour cells expressing cytoplasmic HSP70. J. Cell. Mol. Med. 14, 2655–2666 (2010)PubMedCrossRef Y. Xie, O. Bai, H. Zhang, J. Yuan, S. Zong, R. Chibbar, K. Slattery, M. Qureshi, Y. Wei, Y. Deng, J. Xiang, Membrane-bound HSP70-engineered myeloma cell-derived exosomes stimulate more efficient CD8(+) CTL- and NK-mediated antitumour immunity than exosomes released from heat-shocked tumour cells expressing cytoplasmic HSP70. J. Cell. Mol. Med. 14, 2655–2666 (2010)PubMedCrossRef
200.
go back to reference L.H. Lv, Y.L. Wan, Y. Lin, W. Zhang, M. Yang, G.L. Li, H.M. Lin, C.Z. Shang, Y.J. Chen, J. Min, Anticancer drugs cause release of exosomes with heat shock proteins from human hepatocellular carcinoma cells that elicit effective natural killer cell antitumor responses in vitro. J. Biol. Chem. 287, 15874–15885 (2012)PubMedPubMedCentralCrossRef L.H. Lv, Y.L. Wan, Y. Lin, W. Zhang, M. Yang, G.L. Li, H.M. Lin, C.Z. Shang, Y.J. Chen, J. Min, Anticancer drugs cause release of exosomes with heat shock proteins from human hepatocellular carcinoma cells that elicit effective natural killer cell antitumor responses in vitro. J. Biol. Chem. 287, 15874–15885 (2012)PubMedPubMedCentralCrossRef
201.
go back to reference G. Fuhrmann, A. Serio, M. Mazo, R. Nair, M.M. Stevens, Active loading into extracellular vesicles significantly improves the cellular uptake and photodynamic effect of porphyrins. J. Control. Release 205, 35–44 (2015)PubMedCrossRef G. Fuhrmann, A. Serio, M. Mazo, R. Nair, M.M. Stevens, Active loading into extracellular vesicles significantly improves the cellular uptake and photodynamic effect of porphyrins. J. Control. Release 205, 35–44 (2015)PubMedCrossRef
202.
go back to reference F. Aqil, H. Kausar, A.K. Agrawal, J. Jeyabalan, A.H. Kyakulaga, R. Munagala, R. Gupta, Exosomal formulation enhances therapeutic response of celastrol against lung cancer. Exp. Mol. Pathol. 101, 12–21 (2016)PubMedCrossRef F. Aqil, H. Kausar, A.K. Agrawal, J. Jeyabalan, A.H. Kyakulaga, R. Munagala, R. Gupta, Exosomal formulation enhances therapeutic response of celastrol against lung cancer. Exp. Mol. Pathol. 101, 12–21 (2016)PubMedCrossRef
203.
go back to reference M.S. Kim, M.J. Haney, Y. Zhao, V. Mahajan, I. Deygen, N.L. Klyachko, E. Inskoe, A. Piroyan, M. Sokolsky, O. Okolie, S.D. Hingtgen, A.V. Kabanov, E.V. Batrakova, Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine 12, 655–664 (2016)PubMedCrossRef M.S. Kim, M.J. Haney, Y. Zhao, V. Mahajan, I. Deygen, N.L. Klyachko, E. Inskoe, A. Piroyan, M. Sokolsky, O. Okolie, S.D. Hingtgen, A.V. Kabanov, E.V. Batrakova, Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine 12, 655–664 (2016)PubMedCrossRef
204.
go back to reference R. Munagala, F. Aqil, J. Jeyabalan, R.C. Gupta, Bovine milk-derived exosomes for drug delivery. Cancer Lett. 371, 48–61 (2016)PubMedCrossRef R. Munagala, F. Aqil, J. Jeyabalan, R.C. Gupta, Bovine milk-derived exosomes for drug delivery. Cancer Lett. 371, 48–61 (2016)PubMedCrossRef
205.
go back to reference S.C. Jang, O.Y. Kim, C.M. Yoon, D.S. Choi, T.Y. Roh, J. Park, J. Nilsson, J. Lotvall, Y.K. Kim, Y.S. Gho, Bioinspired exosome-mimetic nanovesicles for targeted delivery of chemotherapeutics to malignant tumors. ACS Nano 7, 7698–7710 (2013)PubMedCrossRef S.C. Jang, O.Y. Kim, C.M. Yoon, D.S. Choi, T.Y. Roh, J. Park, J. Nilsson, J. Lotvall, Y.K. Kim, Y.S. Gho, Bioinspired exosome-mimetic nanovesicles for targeted delivery of chemotherapeutics to malignant tumors. ACS Nano 7, 7698–7710 (2013)PubMedCrossRef
206.
go back to reference K. O'Brien, M.C. Lowry, C. Corcoran, V.G. Martinez, M. Daly, S. Rani, W.M. Gallagher, M.W. Radomski, R.A. MacLeod, L. O'Driscoll, miR-134 in extracellular vesicles reduces triple-negative breast cancer aggression and increases drug sensitivity. Oncotarget 6, 32774–32789 (2015)PubMedPubMedCentral K. O'Brien, M.C. Lowry, C. Corcoran, V.G. Martinez, M. Daly, S. Rani, W.M. Gallagher, M.W. Radomski, R.A. MacLeod, L. O'Driscoll, miR-134 in extracellular vesicles reduces triple-negative breast cancer aggression and increases drug sensitivity. Oncotarget 6, 32774–32789 (2015)PubMedPubMedCentral
207.
go back to reference S. Ohno, M. Takanashi, K. Sudo, S. Ueda, A. Ishikawa, N. Matsuyama, K. Fujita, T. Mizutani, T. Ohgi, T. Ochiya, N. Gotoh, M. Kuroda, Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol. Ther. 21, 185–191 (2013)PubMedCrossRef S. Ohno, M. Takanashi, K. Sudo, S. Ueda, A. Ishikawa, N. Matsuyama, K. Fujita, T. Mizutani, T. Ohgi, T. Ochiya, N. Gotoh, M. Kuroda, Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol. Ther. 21, 185–191 (2013)PubMedCrossRef
208.
go back to reference K.A. Greco, C.A. Franzen, K.E. Foreman, R.C. Flanigan, P.C. Kuo, G.N. Gupta, PLK-1 silencing in bladder cancer by sirna delivered with exosomes. Urology 91, e241–e247 (2016)CrossRef K.A. Greco, C.A. Franzen, K.E. Foreman, R.C. Flanigan, P.C. Kuo, G.N. Gupta, PLK-1 silencing in bladder cancer by sirna delivered with exosomes. Urology 91, e241–e247 (2016)CrossRef
209.
go back to reference J.L. Munoz, S.A. Bliss, S.J. Greco, S.H. Ramkissoon, K.L. Ligon, P. Rameshwar, Delivery of functional anti-mir-9 by mesenchymal stem cell-derived exosomes to glioblastoma multiforme cells conferred chemosensitivity. Mol. Ther. Nucleic Acids 2, e126 (2013)PubMedPubMedCentralCrossRef J.L. Munoz, S.A. Bliss, S.J. Greco, S.H. Ramkissoon, K.L. Ligon, P. Rameshwar, Delivery of functional anti-mir-9 by mesenchymal stem cell-derived exosomes to glioblastoma multiforme cells conferred chemosensitivity. Mol. Ther. Nucleic Acids 2, e126 (2013)PubMedPubMedCentralCrossRef
210.
go back to reference H. Qi, C. Liu, L. Long, Y. Ren, S. Zhang, X. Chang, X. Qian, H. Jia, J. Zhao, J. Sun, X. Hou, X. Yuan, C. Kang, Blood exosomes endowed with magnetic and targeting properties for cancer therapy. ACS Nano 10, 3323–3333 (2016)PubMedCrossRef H. Qi, C. Liu, L. Long, Y. Ren, S. Zhang, X. Chang, X. Qian, H. Jia, J. Zhao, J. Sun, X. Hou, X. Yuan, C. Kang, Blood exosomes endowed with magnetic and targeting properties for cancer therapy. ACS Nano 10, 3323–3333 (2016)PubMedCrossRef
211.
go back to reference Y. Tian, S. Li, J. Song, T. Ji, M. Zhu, G.J. Anderson, J. Wei, G. Nie, A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials 35, 2383–2390 (2014)PubMedCrossRef Y. Tian, S. Li, J. Song, T. Ji, M. Zhu, G.J. Anderson, J. Wei, G. Nie, A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials 35, 2383–2390 (2014)PubMedCrossRef
212.
go back to reference T. Yang, P. Martin, B. Fogarty, A. Brown, K. Schurman, R. Phipps, V.P. Yin, P. Lockman, S. Bai, Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio rerio. Pharm. Res. 32, 2003–2014 (2015)PubMedPubMedCentralCrossRef T. Yang, P. Martin, B. Fogarty, A. Brown, K. Schurman, R. Phipps, V.P. Yin, P. Lockman, S. Bai, Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio rerio. Pharm. Res. 32, 2003–2014 (2015)PubMedPubMedCentralCrossRef
213.
go back to reference C.S. Hong, L. Muller, M. Boyiadzis, T.L. Whiteside, Isolation and characterization of CD34+ blast-derived exosomes in acute myeloid leukemia. PLoS One 9, e103310 (2014)PubMedPubMedCentralCrossRef C.S. Hong, L. Muller, M. Boyiadzis, T.L. Whiteside, Isolation and characterization of CD34+ blast-derived exosomes in acute myeloid leukemia. PLoS One 9, e103310 (2014)PubMedPubMedCentralCrossRef
214.
go back to reference C.J. Beckham, J. Olsen, P.N. Yin, C.H. Wu, H.J. Ting, F.K. Hagen, E. Scosyrev, E.M. Messing, Y.F. Lee, Bladder cancer exosomes contain EDIL-3/Del1 and facilitate cancer progression. J. Urol. 192, 583–592 (2014)PubMedCrossRef C.J. Beckham, J. Olsen, P.N. Yin, C.H. Wu, H.J. Ting, F.K. Hagen, E. Scosyrev, E.M. Messing, Y.F. Lee, Bladder cancer exosomes contain EDIL-3/Del1 and facilitate cancer progression. J. Urol. 192, 583–592 (2014)PubMedCrossRef
215.
go back to reference C. Berrondo, J. Flax, V. Kucherov, A. Siebert, T. Osinski, A. Rosenberg, C. Fucile, S. Richheimer, C.J. Beckham, Expression of the long non-coding rna hotair correlates with disease progression in bladder cancer and is contained in bladder cancer patient urinary exosomes. PLoS One 11, e0147236 (2016)PubMedPubMedCentralCrossRef C. Berrondo, J. Flax, V. Kucherov, A. Siebert, T. Osinski, A. Rosenberg, C. Fucile, S. Richheimer, C.J. Beckham, Expression of the long non-coding rna hotair correlates with disease progression in bladder cancer and is contained in bladder cancer patient urinary exosomes. PLoS One 11, e0147236 (2016)PubMedPubMedCentralCrossRef
216.
go back to reference C. Eichelser, I. Stuckrath, V. Muller, K. Milde-Langosch, H. Wikman, K. Pantel, H. Schwarzenbach, Increased serum levels of circulating exosomal microRNA-373 in receptor-negative breast cancer patients. Oncotarget 5, 9650–9663 (2014)PubMedPubMedCentralCrossRef C. Eichelser, I. Stuckrath, V. Muller, K. Milde-Langosch, H. Wikman, K. Pantel, H. Schwarzenbach, Increased serum levels of circulating exosomal microRNA-373 in receptor-negative breast cancer patients. Oncotarget 5, 9650–9663 (2014)PubMedPubMedCentralCrossRef
217.
go back to reference S. Khan, H.F. Bennit, D. Turay, M. Perez, S. Mirshahidi, Y. Yuan, N.R. Wall, Early diagnostic value of survivin and its alternative splice variants in breast cancer. BMC Cancer 14, 176 (2014)PubMedPubMedCentralCrossRef S. Khan, H.F. Bennit, D. Turay, M. Perez, S. Mirshahidi, Y. Yuan, N.R. Wall, Early diagnostic value of survivin and its alternative splice variants in breast cancer. BMC Cancer 14, 176 (2014)PubMedPubMedCentralCrossRef
218.
go back to reference I. Vardaki, S. Ceder, D. Rutishauser, G. Baltatzis, T. Foukakis, T. Panaretakis, Periostin is identified as a putative metastatic marker in breast cancer-derived exosomes. Oncotarget 7, 74966–74978 (2016)PubMedPubMedCentralCrossRef I. Vardaki, S. Ceder, D. Rutishauser, G. Baltatzis, T. Foukakis, T. Panaretakis, Periostin is identified as a putative metastatic marker in breast cancer-derived exosomes. Oncotarget 7, 74966–74978 (2016)PubMedPubMedCentralCrossRef
219.
go back to reference J. Liu, H. Sun, X. Wang, Q. Yu, S. Li, X. Yu, W. Gong, Increased exosomal microRNA-21 and microRNA-146a levels in the cervicovaginal lavage specimens of patients with cervical cancer. Int. J. Mol. Sci. 15, 758–773 (2014)PubMedPubMedCentralCrossRef J. Liu, H. Sun, X. Wang, Q. Yu, S. Li, X. Yu, W. Gong, Increased exosomal microRNA-21 and microRNA-146a levels in the cervicovaginal lavage specimens of patients with cervical cancer. Int. J. Mol. Sci. 15, 758–773 (2014)PubMedPubMedCentralCrossRef
220.
go back to reference T. Matsumura, K. Sugimachi, H. Iinuma, Y. Takahashi, J. Kurashige, G. Sawada, M. Ueda, R. Uchi, H. Ueo, Y. Takano, Y. Shinden, H. Eguchi, H. Yamamoto, Y. Doki, M. Mori, T. Ochiya, K. Mimori, Exosomal microRNA in serum is a novel biomarker of recurrence in human colorectal cancer. Br. J. Cancer 113, 275–281 (2015)PubMedPubMedCentralCrossRef T. Matsumura, K. Sugimachi, H. Iinuma, Y. Takahashi, J. Kurashige, G. Sawada, M. Ueda, R. Uchi, H. Ueo, Y. Takano, Y. Shinden, H. Eguchi, H. Yamamoto, Y. Doki, M. Mori, T. Ochiya, K. Mimori, Exosomal microRNA in serum is a novel biomarker of recurrence in human colorectal cancer. Br. J. Cancer 113, 275–281 (2015)PubMedPubMedCentralCrossRef
221.
go back to reference H. Ogata-Kawata, M. Izumiya, D. Kurioka, Y. Honma, Y. Yamada, K. Furuta, T. Gunji, H. Ohta, H. Okamoto, H. Sonoda, M. Watanabe, H. Nakagama, J. Yokota, T. Kohno, N. Tsuchiya, Circulating exosomal microRNAs as biomarkers of colon cancer. PLoS One 9, e92921 (2014)PubMedPubMedCentralCrossRef H. Ogata-Kawata, M. Izumiya, D. Kurioka, Y. Honma, Y. Yamada, K. Furuta, T. Gunji, H. Ohta, H. Okamoto, H. Sonoda, M. Watanabe, H. Nakagama, J. Yokota, T. Kohno, N. Tsuchiya, Circulating exosomal microRNAs as biomarkers of colon cancer. PLoS One 9, e92921 (2014)PubMedPubMedCentralCrossRef
222.
go back to reference Q. Li, Y. Shao, X. Zhang, T. Zheng, M. Miao, L. Qin, B. Wang, G. Ye, B. Xiao, J. Guo, Plasma long noncoding RNA protected by exosomes as a potential stable biomarker for gastric cancer. Tumour Biol. 36, 2007–2012 (2015)PubMedCrossRef Q. Li, Y. Shao, X. Zhang, T. Zheng, M. Miao, L. Qin, B. Wang, G. Ye, B. Xiao, J. Guo, Plasma long noncoding RNA protected by exosomes as a potential stable biomarker for gastric cancer. Tumour Biol. 36, 2007–2012 (2015)PubMedCrossRef
223.
go back to reference H. Wang, L. Hou, A. Li, Y. Duan, H. Gao, X. Song, Expression of serum exosomal microRNA-21 in human hepatocellular carcinoma. Biomed. Res. Int. 2014, 864894 (2014)PubMedPubMedCentral H. Wang, L. Hou, A. Li, Y. Duan, H. Gao, X. Song, Expression of serum exosomal microRNA-21 in human hepatocellular carcinoma. Biomed. Res. Int. 2014, 864894 (2014)PubMedPubMedCentral
224.
go back to reference K. Sugimachi, T. Matsumura, H. Hirata, R. Uchi, M. Ueda, H. Ueo, Y. Shinden, T. Iguchi, H. Eguchi, K. Shirabe, T. Ochiya, Y. Maehara, K. Mimori, Identification of a bona fide microRNA biomarker in serum exosomes that predicts hepatocellular carcinoma recurrence after liver transplantation. Br. J. Cancer 112, 532–538 (2015)PubMedPubMedCentralCrossRef K. Sugimachi, T. Matsumura, H. Hirata, R. Uchi, M. Ueda, H. Ueo, Y. Shinden, T. Iguchi, H. Eguchi, K. Shirabe, T. Ochiya, Y. Maehara, K. Mimori, Identification of a bona fide microRNA biomarker in serum exosomes that predicts hepatocellular carcinoma recurrence after liver transplantation. Br. J. Cancer 112, 532–538 (2015)PubMedPubMedCentralCrossRef
225.
go back to reference J. Wang, Y. Zhou, J. Lu, Y. Sun, H. Xiao, M. Liu, L. Tian, Combined detection of serum exosomal miR-21 and HOTAIR as diagnostic and prognostic biomarkers for laryngeal squamous cell carcinoma. Med. Oncol. 31, 148 (2014)PubMedCrossRef J. Wang, Y. Zhou, J. Lu, Y. Sun, H. Xiao, M. Liu, L. Tian, Combined detection of serum exosomal miR-21 and HOTAIR as diagnostic and prognostic biomarkers for laryngeal squamous cell carcinoma. Med. Oncol. 31, 148 (2014)PubMedCrossRef
226.
go back to reference M. Guan, X. Chen, Y. Ma, L. Tang, L. Guan, X. Ren, B. Yu, W. Zhang, B. Su, MDA-9 and GRP78 as potential diagnostic biomarkers for early detection of melanoma metastasis. Tumour Biol. 36, 2973–2982 (2015)PubMedCrossRef M. Guan, X. Chen, Y. Ma, L. Tang, L. Guan, X. Ren, B. Yu, W. Zhang, B. Su, MDA-9 and GRP78 as potential diagnostic biomarkers for early detection of melanoma metastasis. Tumour Biol. 36, 2973–2982 (2015)PubMedCrossRef
227.
go back to reference E. Alegre, M.F. Sanmamed, C. Rodriguez, O. Carranza, S. Martin-Algarra, A. Gonzalez, Study of circulating microRNA-125b levels in serum exosomes in advanced melanoma. Arch. Pathol. Lab. Med. 138, 828–832 (2014)PubMedCrossRef E. Alegre, M.F. Sanmamed, C. Rodriguez, O. Carranza, S. Martin-Algarra, A. Gonzalez, Study of circulating microRNA-125b levels in serum exosomes in advanced melanoma. Arch. Pathol. Lab. Med. 138, 828–832 (2014)PubMedCrossRef
228.
go back to reference J. Klibi, T. Niki, A. Riedel, C. Pioche-Durieu, S. Souquere, E. Rubinstein, S. Le Moulec, J. Guigay, M. Hirashima, F. Guemira, D. Adhikary, J. Mautner, P. Busson, Blood diffusion and Th1-suppressive effects of galectin-9-containing exosomes released by Epstein-Barr virus-infected nasopharyngeal carcinoma cells. Blood 113, 1957–1966 (2009)PubMedCrossRef J. Klibi, T. Niki, A. Riedel, C. Pioche-Durieu, S. Souquere, E. Rubinstein, S. Le Moulec, J. Guigay, M. Hirashima, F. Guemira, D. Adhikary, J. Mautner, P. Busson, Blood diffusion and Th1-suppressive effects of galectin-9-containing exosomes released by Epstein-Barr virus-infected nasopharyngeal carcinoma cells. Blood 113, 1957–1966 (2009)PubMedCrossRef
229.
go back to reference Y. Li, Y. Zhang, F. Qiu, Z. Qiu, Proteomic identification of exosomal LRG1: a potential urinary biomarker for detecting NSCLC. Electrophoresis 32, 1976–1983 (2011)PubMedCrossRef Y. Li, Y. Zhang, F. Qiu, Z. Qiu, Proteomic identification of exosomal LRG1: a potential urinary biomarker for detecting NSCLC. Electrophoresis 32, 1976–1983 (2011)PubMedCrossRef
230.
go back to reference Y. Tanaka, H. Kamohara, K. Kinoshita, J. Kurashige, T. Ishimoto, M. Iwatsuki, M. Watanabe, H. Baba, Clinical impact of serum exosomal microRNA-21 as a clinical biomarker in human esophageal squamous cell carcinoma. Cancer 119, 1159–1167 (2013)PubMedCrossRef Y. Tanaka, H. Kamohara, K. Kinoshita, J. Kurashige, T. Ishimoto, M. Iwatsuki, M. Watanabe, H. Baba, Clinical impact of serum exosomal microRNA-21 as a clinical biomarker in human esophageal squamous cell carcinoma. Cancer 119, 1159–1167 (2013)PubMedCrossRef
231.
go back to reference D.D. Taylor and C. Gercel-Taylor, MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 110, 13–21 (2008) D.D. Taylor and C. Gercel-Taylor, MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 110, 13–21 (2008)
232.
go back to reference X. Ying, Q. Wu, X. Wu, Q. Zhu, X. Wang, L. Jiang, X. Chen and X. Wang, Epithelial ovarian cancer-secreted exosomal miR-222-3p induces polarization of tumor-associated macrophages. Oncotarget 7, 43076–43087 (2016) X. Ying, Q. Wu, X. Wu, Q. Zhu, X. Wang, L. Jiang, X. Chen and X. Wang, Epithelial ovarian cancer-secreted exosomal miR-222-3p induces polarization of tumor-associated macrophages. Oncotarget 7, 43076–43087 (2016)
233.
go back to reference C. Kahlert, S.A. Melo, A. Protopopov, J. Tang, S. Seth, M. Koch, J. Zhang, J. Weitz, L. Chin, A. Futreal, R. Kalluri, Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J. Biol. Chem. 289, 3869–3875 (2014)PubMedPubMedCentralCrossRef C. Kahlert, S.A. Melo, A. Protopopov, J. Tang, S. Seth, M. Koch, J. Zhang, J. Weitz, L. Chin, A. Futreal, R. Kalluri, Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J. Biol. Chem. 289, 3869–3875 (2014)PubMedPubMedCentralCrossRef
234.
go back to reference S.A. Melo, L.B. Luecke, C. Kahlert, A.F. Fernandez, S.T. Gammon, J. Kaye, V.S. LeBleu, E.A. Mittendorf, J. Weitz, N. Rahbari, C. Reissfelder, C. Pilarsky, M.F. Fraga, D. Piwnica-Worms, R. Kalluri, Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature 523, 177–182 (2015)PubMedPubMedCentralCrossRef S.A. Melo, L.B. Luecke, C. Kahlert, A.F. Fernandez, S.T. Gammon, J. Kaye, V.S. LeBleu, E.A. Mittendorf, J. Weitz, N. Rahbari, C. Reissfelder, C. Pilarsky, M.F. Fraga, D. Piwnica-Worms, R. Kalluri, Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature 523, 177–182 (2015)PubMedPubMedCentralCrossRef
235.
go back to reference P.J. Mitchell, J. Welton, J. Staffurth, J. Court, M.D. Mason, Z. Tabi, A. Clayton, Can urinary exosomes act as treatment response markers in prostate cancer? J. Transl. Med. 7, 4 (2009)PubMedPubMedCentralCrossRef P.J. Mitchell, J. Welton, J. Staffurth, J. Court, M.D. Mason, Z. Tabi, A. Clayton, Can urinary exosomes act as treatment response markers in prostate cancer? J. Transl. Med. 7, 4 (2009)PubMedPubMedCentralCrossRef
236.
go back to reference T. Kato, K. Mizutani, K. Kameyama, K. Kawakami, Y. Fujita, K. Nakane, Y. Kanimoto, H. Ehara, H. Ito, M. Seishima, T. Deguchi, M. Ito, Serum exosomal P-glycoprotein is a potential marker to diagnose docetaxel resistance and select a taxoid for patients with prostate cancer. Urol. Oncol. 33, e315–e320 (2015)CrossRef T. Kato, K. Mizutani, K. Kameyama, K. Kawakami, Y. Fujita, K. Nakane, Y. Kanimoto, H. Ehara, H. Ito, M. Seishima, T. Deguchi, M. Ito, Serum exosomal P-glycoprotein is a potential marker to diagnose docetaxel resistance and select a taxoid for patients with prostate cancer. Urol. Oncol. 33, e315–e320 (2015)CrossRef
237.
go back to reference X. Huang, T. Yuan, M. Liang, M. Du, S. Xia, R. Dittmar, D. Wang, W. See, B.A. Costello, F. Quevedo, W. Tan, D. Nandy, G.H. Bevan, S. Longenbach, Z. Sun, Y. Lu, T. Wang, S.N. Thibodeau, L. Boardman, M. Kohli, L. Wang, Exosomal miR-1290 and miR-375 as prognostic markers in castration-resistant prostate cancer. Eur. Urol. 67, 33–41 (2015)PubMedCrossRef X. Huang, T. Yuan, M. Liang, M. Du, S. Xia, R. Dittmar, D. Wang, W. See, B.A. Costello, F. Quevedo, W. Tan, D. Nandy, G.H. Bevan, S. Longenbach, Z. Sun, Y. Lu, T. Wang, S.N. Thibodeau, L. Boardman, M. Kohli, L. Wang, Exosomal miR-1290 and miR-375 as prognostic markers in castration-resistant prostate cancer. Eur. Urol. 67, 33–41 (2015)PubMedCrossRef
239.
go back to reference James Graham Brown Cancer Center. preliminary clinical trial investigating the ability of plant exosomes to abrogate oral mucositis induced by combined chemotherapy and radiation in head and neck cancer patients. http://clinicaltrials.gov/show/NCT01668849. Accessed 16 September 2016 James Graham Brown Cancer Center. preliminary clinical trial investigating the ability of plant exosomes to abrogate oral mucositis induced by combined chemotherapy and radiation in head and neck cancer patients. http://​clinicaltrials.​gov/​show/​NCT01668849. Accessed 16 September 2016
240.
go back to reference B. Besse, M. Charrier, V. Lapierre, E. Dansin, O. Lantz, D. Planchard, T. Le Chevalier, A. Livartoski, F. Barlesi, A. Laplanche, S. Ploix, N. Vimond, I. Peguillet, C. Thery, L. Lacroix, I. Zoernig, K. Dhodapkar, M. Dhodapkar, S. Viaud, J.C. Soria, K.S. Reiners, E. Pogge von Strandmann, F. Vely, S. Rusakiewicz, A. Eggermont, J.M. Pitt, L. Zitvogel, N. Chaput, Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology 5, e1071008 (2016)PubMedCrossRef B. Besse, M. Charrier, V. Lapierre, E. Dansin, O. Lantz, D. Planchard, T. Le Chevalier, A. Livartoski, F. Barlesi, A. Laplanche, S. Ploix, N. Vimond, I. Peguillet, C. Thery, L. Lacroix, I. Zoernig, K. Dhodapkar, M. Dhodapkar, S. Viaud, J.C. Soria, K.S. Reiners, E. Pogge von Strandmann, F. Vely, S. Rusakiewicz, A. Eggermont, J.M. Pitt, L. Zitvogel, N. Chaput, Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology 5, e1071008 (2016)PubMedCrossRef
241.
go back to reference S. Viaud, M. Terme, C. Flament, J. Taieb, F. Andre, S. Novault, B. Escudier, C. Robert, S. Caillat-Zucman, T. Tursz, L. Zitvogel, N. Chaput, Dendritic cell-derived exosomes promote natural killer cell activation and proliferation: a role for NKG2D ligands and IL-15Ralpha. PLoS One 4, e4942 (2009)PubMedPubMedCentralCrossRef S. Viaud, M. Terme, C. Flament, J. Taieb, F. Andre, S. Novault, B. Escudier, C. Robert, S. Caillat-Zucman, T. Tursz, L. Zitvogel, N. Chaput, Dendritic cell-derived exosomes promote natural killer cell activation and proliferation: a role for NKG2D ligands and IL-15Ralpha. PLoS One 4, e4942 (2009)PubMedPubMedCentralCrossRef
242.
go back to reference B. Escudier, T. Dorval, N. Chaput, F. Andre, M.P. Caby, S. Novault, C. Flament, C. Leboulaire, C. Borg, S. Amigorena, C. Boccaccio, C. Bonnerot, O. Dhellin, M. Movassagh, S. Piperno, C. Robert, V. Serra, N. Valente, J.B. Le Pecq, A. Spatz, O. Lantz, T. Tursz, E. Angevin, L. Zitvogel, Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: results of thefirst phase I clinical trial. J. Transl. Med. 3, 10 (2005)PubMedPubMedCentralCrossRef B. Escudier, T. Dorval, N. Chaput, F. Andre, M.P. Caby, S. Novault, C. Flament, C. Leboulaire, C. Borg, S. Amigorena, C. Boccaccio, C. Bonnerot, O. Dhellin, M. Movassagh, S. Piperno, C. Robert, V. Serra, N. Valente, J.B. Le Pecq, A. Spatz, O. Lantz, T. Tursz, E. Angevin, L. Zitvogel, Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: results of thefirst phase I clinical trial. J. Transl. Med. 3, 10 (2005)PubMedPubMedCentralCrossRef
245.
go back to reference New Mexico Cancer Care Alliance. An observational, single-institution pilot/feasibility study of exosome testing as a screening modality for human papillomavirus-positive oropharyngeal squamous cell carcinoma. http://clinicaltrials.gov/show/NCT02147418. Accessed 16 September 2016 New Mexico Cancer Care Alliance. An observational, single-institution pilot/feasibility study of exosome testing as a screening modality for human papillomavirus-positive oropharyngeal squamous cell carcinoma. http://​clinicaltrials.​gov/​show/​NCT02147418. Accessed 16 September 2016
247.
go back to reference National Taiwan University Hospital. Anaplastic thyroid cancer and follicular thyroid cancer-derived exosomal analysis via treatment of lovastatin and vildagliptin and pilot prognostic study via urine exosomal biological markers in thyroid cancer patients. http://clinicaltrials.gov/show/NCT02862470. Accessed 16 September 2016 National Taiwan University Hospital. Anaplastic thyroid cancer and follicular thyroid cancer-derived exosomal analysis via treatment of lovastatin and vildagliptin and pilot prognostic study via urine exosomal biological markers in thyroid cancer patients. http://​clinicaltrials.​gov/​show/​NCT02862470. Accessed 16 September 2016
248.
go back to reference Thomas Jefferson University. Phase 1 study in humans evaluating the safety of rectus sheath implantation of diffusion chambers encapsulating autologous malignant glioma cells treated with insulin-like growth factor receptor-1 antisense oligodeoxynucleotide in 12 patients with recurrent malignant glioma. http://www.clinicaltrials.gov/show/NCT01550523. Accessed 16 September 2016 Thomas Jefferson University. Phase 1 study in humans evaluating the safety of rectus sheath implantation of diffusion chambers encapsulating autologous malignant glioma cells treated with insulin-like growth factor receptor-1 antisense oligodeoxynucleotide in 12 patients with recurrent malignant glioma. http://​www.​clinicaltrials.​gov/​show/​NCT01550523. Accessed 16 September 2016
249.
go back to reference Thomas Jefferson University. Phase I study in humans evaluating the safety of rectus sheath implantation of diffusion chambers encapsulating autologous malignant glioma cells treated with insulin-like growth factor receptor-1 antisense oligodeoxynucleotide (igf-1r/as odn) in 32 patients with newly diagnosed malignant glioma. http://clinicaltrials.gov/show/NCT02507583. Accessed 16 September 2016 Thomas Jefferson University. Phase I study in humans evaluating the safety of rectus sheath implantation of diffusion chambers encapsulating autologous malignant glioma cells treated with insulin-like growth factor receptor-1 antisense oligodeoxynucleotide (igf-1r/as odn) in 32 patients with newly diagnosed malignant glioma. http://​clinicaltrials.​gov/​show/​NCT02507583. Accessed 16 September 2016
250.
go back to reference S. Dai, D. Wei, Z. Wu, X. Zhou, X. Wei, H. Huang, G. Li, I. Phase, clinical trial of autologous ascites-derived exosomes combined with GM-CSF for colorectal cancer. Mol. Ther. 16, 782–790 (2008)PubMedCrossRef S. Dai, D. Wei, Z. Wu, X. Zhou, X. Wei, H. Huang, G. Li, I. Phase, clinical trial of autologous ascites-derived exosomes combined with GM-CSF for colorectal cancer. Mol. Ther. 16, 782–790 (2008)PubMedCrossRef
Metadata
Title
The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications
Authors
Vignesh Sundararajan
Fazlul H. Sarkar
Thamil Selvee Ramasamy
Publication date
01-06-2018
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 3/2018
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-018-0378-4

Other articles of this Issue 3/2018

Cellular Oncology 3/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine