Skip to main content
Top
Published in: Molecular Cancer 1/2019

Open Access 01-12-2019 | Review

Microvesicles and chemokines in tumor microenvironment: mediators of intercellular communications in tumor progression

Authors: Xiaojie Bian, Yu-Tian Xiao, Tianqi Wu, Mengfei Yao, Leilei Du, Shancheng Ren, Jianhua Wang

Published in: Molecular Cancer | Issue 1/2019

Login to get access

Abstract

Increasing evidence indicates that the ability of cancer cells to convey biological information to recipient cells within the tumor microenvironment (TME) is crucial for tumor progression. Microvesicles (MVs) are heterogenous vesicles formed by budding of the cellular membrane, which are secreted in larger amounts by cancer cells than normal cells. Recently, several reports have also disclosed that MVs function as important mediators of intercellular communication between cancerous and stromal cells within the TME, orchestrating complex pathophysiological processes. Chemokines are a family of small inflammatory cytokines that are able to induce chemotaxis in responsive cells. MVs which selective incorporate chemokines as their molecular cargos may play important regulatory roles in oncogenic processes including tumor proliferation, apoptosis, angiogenesis, metastasis, chemoresistance and immunomodulation, et al. Therefore, it is important to explore the association of MVs and chemokines in TME, identify the potential prognostic marker of tumor, and develop more effective treatment strategies. Here we review the relevant literature regarding the role of MVs and chemokines in TME.
Literature
2.
go back to reference Ratajczak J, Miekus K, Kucia M, Zhang J, Reca R, Dvorak P, et al. Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: evidence for horizontal transfer of mRNA and protein delivery. Leukemia. 2006;20(5):847–56.PubMedCrossRef Ratajczak J, Miekus K, Kucia M, Zhang J, Reca R, Dvorak P, et al. Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: evidence for horizontal transfer of mRNA and protein delivery. Leukemia. 2006;20(5):847–56.PubMedCrossRef
3.
go back to reference Desrochers LM, Bordeleau F, Reinhart-King CA, Cerione RA, Antonyak MA. Microvesicles provide a mechanism for intercellular communication by embryonic stem cells during embryo implantation. Nat Commun. 2016;7:11958.PubMedPubMedCentralCrossRef Desrochers LM, Bordeleau F, Reinhart-King CA, Cerione RA, Antonyak MA. Microvesicles provide a mechanism for intercellular communication by embryonic stem cells during embryo implantation. Nat Commun. 2016;7:11958.PubMedPubMedCentralCrossRef
4.
go back to reference Clancy JW, Sedgwick A, Rosse C, Muralidharan-Chari V, Raposo G, Method M, et al. Regulated delivery of molecular cargo to invasive tumour-derived microvesicles. Nat Commun. 2015;6:6919.PubMedPubMedCentralCrossRef Clancy JW, Sedgwick A, Rosse C, Muralidharan-Chari V, Raposo G, Method M, et al. Regulated delivery of molecular cargo to invasive tumour-derived microvesicles. Nat Commun. 2015;6:6919.PubMedPubMedCentralCrossRef
5.
go back to reference Stec M, Szatanek R, Baj-Krzyworzeka M, Baran J, Zembala M, Barbasz J, et al. Interactions of tumour-derived micro (nano) vesicles with human gastric cancer cells. J Transl Med. 2015;13:376.PubMedPubMedCentralCrossRef Stec M, Szatanek R, Baj-Krzyworzeka M, Baran J, Zembala M, Barbasz J, et al. Interactions of tumour-derived micro (nano) vesicles with human gastric cancer cells. J Transl Med. 2015;13:376.PubMedPubMedCentralCrossRef
6.
go back to reference D'souza-Schorey C, Clancy JW. Tumor-derived microvesicles: shedding light on novel microenvironment modulators and prospective cancer biomarkers. Genes Dev. 2012;26(12):1287–99.PubMedPubMedCentralCrossRef D'souza-Schorey C, Clancy JW. Tumor-derived microvesicles: shedding light on novel microenvironment modulators and prospective cancer biomarkers. Genes Dev. 2012;26(12):1287–99.PubMedPubMedCentralCrossRef
8.
go back to reference Holm MM, Kaiser J, Schwab ME. Extracellular vesicles: multimodal envoys in neural maintenance and repair. Trends Neurosci. 2018;41(6):360–72.PubMedCrossRef Holm MM, Kaiser J, Schwab ME. Extracellular vesicles: multimodal envoys in neural maintenance and repair. Trends Neurosci. 2018;41(6):360–72.PubMedCrossRef
9.
go back to reference Willms E, Cabanas C, Mager I, Wood MJA, Vader P. Extracellular vesicle heterogeneity: subpopulations, isolation techniques, and diverse functions in Cancer progression. Front Immunol. 2018;9:738.PubMedPubMedCentralCrossRef Willms E, Cabanas C, Mager I, Wood MJA, Vader P. Extracellular vesicle heterogeneity: subpopulations, isolation techniques, and diverse functions in Cancer progression. Front Immunol. 2018;9:738.PubMedPubMedCentralCrossRef
10.
go back to reference Barteneva NS, Fasler-Kan E, Bernimoulin M, Stern JN, Ponomarev ED, Duckett L, et al. Circulating microparticles: square the circle. BMC Cell Biol. 2013;14:23.PubMedPubMedCentralCrossRef Barteneva NS, Fasler-Kan E, Bernimoulin M, Stern JN, Ponomarev ED, Duckett L, et al. Circulating microparticles: square the circle. BMC Cell Biol. 2013;14:23.PubMedPubMedCentralCrossRef
11.
go back to reference Minciacchi VR, Freeman MR, Di Vizio D. Extracellular vesicles in cancer: exosomes, microvesicles and the emerging role of large oncosomes. Semin Cell Dev Biol. 2015;40:41–51.PubMedPubMedCentralCrossRef Minciacchi VR, Freeman MR, Di Vizio D. Extracellular vesicles in cancer: exosomes, microvesicles and the emerging role of large oncosomes. Semin Cell Dev Biol. 2015;40:41–51.PubMedPubMedCentralCrossRef
12.
go back to reference Ciardiello C, Cavallini L, Spinelli C, Yang J, Reis-Sobreiro M, De Candia P, et al. Focus on extracellular vesicles: new Frontiers of cell-to-cell communication in Cancer. Int J Mol Sci. 2016;17(2):175.PubMedPubMedCentralCrossRef Ciardiello C, Cavallini L, Spinelli C, Yang J, Reis-Sobreiro M, De Candia P, et al. Focus on extracellular vesicles: new Frontiers of cell-to-cell communication in Cancer. Int J Mol Sci. 2016;17(2):175.PubMedPubMedCentralCrossRef
13.
go back to reference Karin N. Chemokines and cancer: new immune checkpoints for cancer therapy. Curr Opin Immunol. 2018;51:140–5.PubMedCrossRef Karin N. Chemokines and cancer: new immune checkpoints for cancer therapy. Curr Opin Immunol. 2018;51:140–5.PubMedCrossRef
14.
go back to reference Weitzenfeld P, Ben-Baruch A. The chemokine system, and its CCR5 and CXCR4 receptors, as potential targets for personalized therapy in cancer. Cancer Lett. 2014;352(1):36–53.PubMedCrossRef Weitzenfeld P, Ben-Baruch A. The chemokine system, and its CCR5 and CXCR4 receptors, as potential targets for personalized therapy in cancer. Cancer Lett. 2014;352(1):36–53.PubMedCrossRef
15.
go back to reference Wendler F, Favicchio R, Simon T, Alifrangis C, Stebbing J, Giamas G. Extracellular vesicles swarm the cancer microenvironment: from tumor-stroma communication to drug intervention. Oncogene. 2017;36(7):877–84.PubMedCrossRef Wendler F, Favicchio R, Simon T, Alifrangis C, Stebbing J, Giamas G. Extracellular vesicles swarm the cancer microenvironment: from tumor-stroma communication to drug intervention. Oncogene. 2017;36(7):877–84.PubMedCrossRef
16.
go back to reference Ratajczak J, Wysoczynski M, Hayek F, Janowska-Wieczorek A, Ratajczak MZ. Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication. Leukemia. 2006;20(9):1487–95.PubMedCrossRef Ratajczak J, Wysoczynski M, Hayek F, Janowska-Wieczorek A, Ratajczak MZ. Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication. Leukemia. 2006;20(9):1487–95.PubMedCrossRef
17.
go back to reference Surman M, Stepien E, Hoja-Lukowicz D, Przybylo M. Deciphering the role of ectosomes in cancer development and progression: focus on the proteome. Clin Exp Metastasis. 2017;34(3–4):273–89.PubMedPubMedCentralCrossRef Surman M, Stepien E, Hoja-Lukowicz D, Przybylo M. Deciphering the role of ectosomes in cancer development and progression: focus on the proteome. Clin Exp Metastasis. 2017;34(3–4):273–89.PubMedPubMedCentralCrossRef
18.
go back to reference Morel O, Jesel L, Freyssinet JM, Toti F. Cellular mechanisms underlying the formation of circulating microparticles. Arterioscler Thromb Vasc Biol. 2011;31(1):15–26.PubMedCrossRef Morel O, Jesel L, Freyssinet JM, Toti F. Cellular mechanisms underlying the formation of circulating microparticles. Arterioscler Thromb Vasc Biol. 2011;31(1):15–26.PubMedCrossRef
19.
go back to reference Pap E, Pallinger E, Pasztoi M, Falus A. Highlights of a new type of intercellular communication: microvesicle-based information transfer. Inflamm Res. 2009;58(1):1–8.PubMedCrossRef Pap E, Pallinger E, Pasztoi M, Falus A. Highlights of a new type of intercellular communication: microvesicle-based information transfer. Inflamm Res. 2009;58(1):1–8.PubMedCrossRef
21.
go back to reference Turiak L, Misjak P, Szabo TG, Aradi B, Paloczi K, Ozohanics O, et al. Proteomic characterization of thymocyte-derived microvesicles and apoptotic bodies in BALB/c mice. J Proteome. 2011;74(10):2025–33.CrossRef Turiak L, Misjak P, Szabo TG, Aradi B, Paloczi K, Ozohanics O, et al. Proteomic characterization of thymocyte-derived microvesicles and apoptotic bodies in BALB/c mice. J Proteome. 2011;74(10):2025–33.CrossRef
22.
go back to reference Lane JD, Allan VJ, Woodman PG. Active relocation of chromatin and endoplasmic reticulum into blebs in late apoptotic cells. J Cell Sci. 2005;118(Pt 17):4059–71.PubMedCrossRef Lane JD, Allan VJ, Woodman PG. Active relocation of chromatin and endoplasmic reticulum into blebs in late apoptotic cells. J Cell Sci. 2005;118(Pt 17):4059–71.PubMedCrossRef
23.
go back to reference Di Vizio D, Kim J, Hager MH, Morello M, Yang W, Lafargue CJ, et al. Oncosome formation in prostate cancer: association with a region of frequent chromosomal deletion in metastatic disease. Cancer Res. 2009;69(13):5601–9.PubMedPubMedCentralCrossRef Di Vizio D, Kim J, Hager MH, Morello M, Yang W, Lafargue CJ, et al. Oncosome formation in prostate cancer: association with a region of frequent chromosomal deletion in metastatic disease. Cancer Res. 2009;69(13):5601–9.PubMedPubMedCentralCrossRef
24.
go back to reference Di Vizio D, Morello M, Dudley AC, Schow PW, Adam RM, Morley S, et al. Large oncosomes in human prostate cancer tissues and in the circulation of mice with metastatic disease. Am J Pathol. 2012;181(5):1573–84.PubMedPubMedCentralCrossRef Di Vizio D, Morello M, Dudley AC, Schow PW, Adam RM, Morley S, et al. Large oncosomes in human prostate cancer tissues and in the circulation of mice with metastatic disease. Am J Pathol. 2012;181(5):1573–84.PubMedPubMedCentralCrossRef
25.
go back to reference Tokunaga R, Zhang W, Naseem M, Puccini A, Berger MD, Soni S, et al. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation - a target for novel cancer therapy. Cancer Treat Rev. 2018;63:40–7.PubMedCrossRef Tokunaga R, Zhang W, Naseem M, Puccini A, Berger MD, Soni S, et al. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation - a target for novel cancer therapy. Cancer Treat Rev. 2018;63:40–7.PubMedCrossRef
26.
go back to reference Namas RA, Mi Q, Namas R, Almahmoud K, Zaaqoq AM, Abdul-Malak O, et al. Insights into the role of chemokines, damage-associated molecular patterns, and lymphocyte-derived mediators from computational models of trauma-induced inflammation. Antioxid Redox Signal. 2015;23(17):1370–87.PubMedPubMedCentralCrossRef Namas RA, Mi Q, Namas R, Almahmoud K, Zaaqoq AM, Abdul-Malak O, et al. Insights into the role of chemokines, damage-associated molecular patterns, and lymphocyte-derived mediators from computational models of trauma-induced inflammation. Antioxid Redox Signal. 2015;23(17):1370–87.PubMedPubMedCentralCrossRef
28.
go back to reference Sun X, Cheng G, Hao M, Zheng J, Zhou X, Zhang J, et al. CXCL12 / CXCR4 / CXCR7 chemokine axis and cancer progression. Cancer Metastasis Rev. 2010;29(4):709–22.PubMedPubMedCentralCrossRef Sun X, Cheng G, Hao M, Zheng J, Zhou X, Zhang J, et al. CXCL12 / CXCR4 / CXCR7 chemokine axis and cancer progression. Cancer Metastasis Rev. 2010;29(4):709–22.PubMedPubMedCentralCrossRef
29.
go back to reference Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol. 2017;17(9):559–72.PubMedPubMedCentralCrossRef Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol. 2017;17(9):559–72.PubMedPubMedCentralCrossRef
31.
go back to reference Chen K, Bao Z, Tang P, Gong W, Yoshimura T, Wang JM. Chemokines in homeostasis and diseases. Cell Mol Immunol. 2018;15(4):324–34.PubMedCrossRef Chen K, Bao Z, Tang P, Gong W, Yoshimura T, Wang JM. Chemokines in homeostasis and diseases. Cell Mol Immunol. 2018;15(4):324–34.PubMedCrossRef
33.
go back to reference Webber JP, Spary LK, Sanders AJ, Chowdhury R, Jiang WG, Steadman R, et al. Differentiation of tumour-promoting stromal myofibroblasts by cancer exosomes. Oncogene. 2015;34(3):290–302.PubMedCrossRef Webber JP, Spary LK, Sanders AJ, Chowdhury R, Jiang WG, Steadman R, et al. Differentiation of tumour-promoting stromal myofibroblasts by cancer exosomes. Oncogene. 2015;34(3):290–302.PubMedCrossRef
34.
go back to reference Emon B, Bauer J, Jain Y, Jung B, Saif T. Biophysics of tumor microenvironment and Cancer metastasis - a mini review. Comput Struct Biotechnol J. 2018;16:279–87.PubMedPubMedCentralCrossRef Emon B, Bauer J, Jain Y, Jung B, Saif T. Biophysics of tumor microenvironment and Cancer metastasis - a mini review. Comput Struct Biotechnol J. 2018;16:279–87.PubMedPubMedCentralCrossRef
35.
go back to reference Whiteside TL. Tumour-derived exosomes or microvesicles: another mechanism of tumour escape from the host immune system? Br J Cancer. 2005;92(2):209–11.PubMedPubMedCentralCrossRef Whiteside TL. Tumour-derived exosomes or microvesicles: another mechanism of tumour escape from the host immune system? Br J Cancer. 2005;92(2):209–11.PubMedPubMedCentralCrossRef
36.
go back to reference Baj-Krzyworzeka M, Szatanek R, Weglarczyk K, Baran J, Zembala M. Tumour-derived microvesicles modulate biological activity of human monocytes. Immunol Lett. 2007;113(2):76–82.PubMedCrossRef Baj-Krzyworzeka M, Szatanek R, Weglarczyk K, Baran J, Zembala M. Tumour-derived microvesicles modulate biological activity of human monocytes. Immunol Lett. 2007;113(2):76–82.PubMedCrossRef
37.
go back to reference Jardim LL, Chaves DG, Silveira-Cassette ACO, Simoes ESAC, Santana MP, Cerqueira MH, et al. Immune status of patients with haemophilia a before exposure to factor VIII: first results from the HEMFIL study. Br J Haematol. 2017;178(6):971–8.PubMedCrossRef Jardim LL, Chaves DG, Silveira-Cassette ACO, Simoes ESAC, Santana MP, Cerqueira MH, et al. Immune status of patients with haemophilia a before exposure to factor VIII: first results from the HEMFIL study. Br J Haematol. 2017;178(6):971–8.PubMedCrossRef
38.
go back to reference Akuthota P, Carmo LA, Bonjour K, Murphy RO, Silva TP, Gamalier JP, et al. Extracellular microvesicle production by human eosinophils activated by "inflammatory" stimuli. Front Cell Dev Biol. 2016;4:117.PubMedPubMedCentralCrossRef Akuthota P, Carmo LA, Bonjour K, Murphy RO, Silva TP, Gamalier JP, et al. Extracellular microvesicle production by human eosinophils activated by "inflammatory" stimuli. Front Cell Dev Biol. 2016;4:117.PubMedPubMedCentralCrossRef
39.
go back to reference Baj-Krzyworzeka M, Szatanek R, Weglarczyk K, Baran J, Urbanowicz B, Branski P, et al. Tumour-derived microvesicles carry several surface determinants and mRNA of tumour cells and transfer some of these determinants to monocytes. Cancer Immunol Immunother. 2006;55(7):808–18.PubMedCrossRef Baj-Krzyworzeka M, Szatanek R, Weglarczyk K, Baran J, Urbanowicz B, Branski P, et al. Tumour-derived microvesicles carry several surface determinants and mRNA of tumour cells and transfer some of these determinants to monocytes. Cancer Immunol Immunother. 2006;55(7):808–18.PubMedCrossRef
40.
go back to reference Baj-Krzyworzeka M, Weglarczyk K, Mytar B, Szatanek R, Baran J, Zembala M. Tumour-derived microvesicles contain interleukin-8 and modulate production of chemokines by human monocytes. Anticancer Res. 2011;31(4):1329–35.PubMed Baj-Krzyworzeka M, Weglarczyk K, Mytar B, Szatanek R, Baran J, Zembala M. Tumour-derived microvesicles contain interleukin-8 and modulate production of chemokines by human monocytes. Anticancer Res. 2011;31(4):1329–35.PubMed
41.
go back to reference Richards DM, Hettinger J, Feuerer M. Monocytes and macrophages in cancer: development and functions. Cancer Microenviron. 2013;6(2):179–91.PubMedCrossRef Richards DM, Hettinger J, Feuerer M. Monocytes and macrophages in cancer: development and functions. Cancer Microenviron. 2013;6(2):179–91.PubMedCrossRef
43.
go back to reference Lin LY, Du LM, Cao K, Huang Y, Yu PF, Zhang LY, et al. Tumour cell-derived exosomes endow mesenchymal stromal cells with tumour-promotion capabilities. Oncogene. 2016;35(46):6038–42.PubMedPubMedCentralCrossRef Lin LY, Du LM, Cao K, Huang Y, Yu PF, Zhang LY, et al. Tumour cell-derived exosomes endow mesenchymal stromal cells with tumour-promotion capabilities. Oncogene. 2016;35(46):6038–42.PubMedPubMedCentralCrossRef
44.
go back to reference Allinen M, Beroukhim R, Cai L, Brennan C, Lahti-Domenici J, Huang H, et al. Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell. 2004;6(1):17–32.PubMedCrossRef Allinen M, Beroukhim R, Cai L, Brennan C, Lahti-Domenici J, Huang H, et al. Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell. 2004;6(1):17–32.PubMedCrossRef
45.
go back to reference Wang Y, Weng X, Wang L, Hao M, Li Y, Hou L, et al. HIC1 deletion promotes breast cancer progression by activating tumor cell/fibroblast crosstalk. J Clin Invest. 2018;128(12):5235–50.PubMedPubMedCentralCrossRef Wang Y, Weng X, Wang L, Hao M, Li Y, Hou L, et al. HIC1 deletion promotes breast cancer progression by activating tumor cell/fibroblast crosstalk. J Clin Invest. 2018;128(12):5235–50.PubMedPubMedCentralCrossRef
46.
go back to reference Lee NH, Nikfarjam M, He H. Functions of the CXC ligand family in the pancreatic tumor microenvironment. Pancreatology. 2018;18(7):705–16.PubMedCrossRef Lee NH, Nikfarjam M, He H. Functions of the CXC ligand family in the pancreatic tumor microenvironment. Pancreatology. 2018;18(7):705–16.PubMedCrossRef
47.
go back to reference Morello M, Minciacchi VR, De Candia P, Yang J, Posadas E, Kim H, et al. Large oncosomes mediate intercellular transfer of functional microRNA. Cell Cycle. 2013;12(22):3526–36.PubMedPubMedCentralCrossRef Morello M, Minciacchi VR, De Candia P, Yang J, Posadas E, Kim H, et al. Large oncosomes mediate intercellular transfer of functional microRNA. Cell Cycle. 2013;12(22):3526–36.PubMedPubMedCentralCrossRef
48.
go back to reference Forst B, Hansen MT, Klingelhofer J, Moller HD, Nielsen GH, Grum-Schwensen B, et al. Metastasis-inducing S100A4 and RANTES cooperate in promoting tumor progression in mice. PLoS One. 2010;5(4):e10374.PubMedPubMedCentralCrossRef Forst B, Hansen MT, Klingelhofer J, Moller HD, Nielsen GH, Grum-Schwensen B, et al. Metastasis-inducing S100A4 and RANTES cooperate in promoting tumor progression in mice. PLoS One. 2010;5(4):e10374.PubMedPubMedCentralCrossRef
49.
go back to reference Rana S, Yue S, Stadel D, Zoller M. Toward tailored exosomes: the exosomal tetraspanin web contributes to target cell selection. Int J Biochem Cell Biol. 2012;44(9):1574–84.PubMedCrossRef Rana S, Yue S, Stadel D, Zoller M. Toward tailored exosomes: the exosomal tetraspanin web contributes to target cell selection. Int J Biochem Cell Biol. 2012;44(9):1574–84.PubMedCrossRef
50.
go back to reference Dong W, Zhang H, Yin X, Liu Y, Chen D, Liang X, et al. Oral delivery of tumor microparticle vaccines activates NOD2 signaling pathway in ileac epithelium rendering potent antitumor T cell immunity. Oncoimmunology. 2017;6(3):e1282589.PubMedPubMedCentralCrossRef Dong W, Zhang H, Yin X, Liu Y, Chen D, Liang X, et al. Oral delivery of tumor microparticle vaccines activates NOD2 signaling pathway in ileac epithelium rendering potent antitumor T cell immunity. Oncoimmunology. 2017;6(3):e1282589.PubMedPubMedCentralCrossRef
51.
go back to reference Ruben JM, Van Den Ancker W, Bontkes HJ, Westers TM, Hooijberg E, Ossenkoppele GJ, et al. Apoptotic blebs from leukemic cells as a preferred source of tumor-associated antigen for dendritic cell-based vaccines. Cancer Immunol Immunother. 2014;63(4):335–45.PubMedCrossRef Ruben JM, Van Den Ancker W, Bontkes HJ, Westers TM, Hooijberg E, Ossenkoppele GJ, et al. Apoptotic blebs from leukemic cells as a preferred source of tumor-associated antigen for dendritic cell-based vaccines. Cancer Immunol Immunother. 2014;63(4):335–45.PubMedCrossRef
52.
go back to reference Chen T, Guo J, Yang M, Zhu X, Cao X. Chemokine-containing exosomes are released from heat-stressed tumor cells via lipid raft-dependent pathway and act as efficient tumor vaccine. J Immunol. 2011;186(4):2219–28.PubMedCrossRef Chen T, Guo J, Yang M, Zhu X, Cao X. Chemokine-containing exosomes are released from heat-stressed tumor cells via lipid raft-dependent pathway and act as efficient tumor vaccine. J Immunol. 2011;186(4):2219–28.PubMedCrossRef
53.
go back to reference Baran J, Baj-Krzyworzeka M, Weglarczyk K, Szatanek R, Zembala M, Barbasz J, et al. Circulating tumour-derived microvesicles in plasma of gastric cancer patients. Cancer Immunol Immunother. 2010;59(6):841–50.PubMedCrossRef Baran J, Baj-Krzyworzeka M, Weglarczyk K, Szatanek R, Zembala M, Barbasz J, et al. Circulating tumour-derived microvesicles in plasma of gastric cancer patients. Cancer Immunol Immunother. 2010;59(6):841–50.PubMedCrossRef
54.
go back to reference Berenguer J, Lagerweij T, Zhao XW, Dusoswa S, Van Der Stoop P, Westerman B, et al. Glycosylated extracellular vesicles released by glioblastoma cells are decorated by CCL18 allowing for cellular uptake via chemokine receptor CCR8. J Extracell Vesicles. 2018;7(1):1446660.PubMedPubMedCentralCrossRef Berenguer J, Lagerweij T, Zhao XW, Dusoswa S, Van Der Stoop P, Westerman B, et al. Glycosylated extracellular vesicles released by glioblastoma cells are decorated by CCL18 allowing for cellular uptake via chemokine receptor CCR8. J Extracell Vesicles. 2018;7(1):1446660.PubMedPubMedCentralCrossRef
55.
go back to reference Al-Nedawi K, Meehan B, Micallef J, Lhotak V, May L, Guha A, et al. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell Biol. 2008;10(5):619–24.PubMedCrossRef Al-Nedawi K, Meehan B, Micallef J, Lhotak V, May L, Guha A, et al. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell Biol. 2008;10(5):619–24.PubMedCrossRef
56.
go back to reference Skog J, Wurdinger T, Van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10(12):1470–6.PubMedPubMedCentralCrossRef Skog J, Wurdinger T, Van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10(12):1470–6.PubMedPubMedCentralCrossRef
57.
go back to reference Antonyak MA, Li B, Boroughs LK, Johnson JL, Druso JE, Bryant KL, et al. Cancer cell-derived microvesicles induce transformation by transferring tissue transglutaminase and fibronectin to recipient cells. Proc Natl Acad Sci U S A. 2011;108(12):4852–7.PubMedPubMedCentralCrossRef Antonyak MA, Li B, Boroughs LK, Johnson JL, Druso JE, Bryant KL, et al. Cancer cell-derived microvesicles induce transformation by transferring tissue transglutaminase and fibronectin to recipient cells. Proc Natl Acad Sci U S A. 2011;108(12):4852–7.PubMedPubMedCentralCrossRef
58.
go back to reference Al-Nedawi K, Meehan B, Kerbel RS, Allison AC, Rak J. Endothelial expression of autocrine VEGF upon the uptake of tumor-derived microvesicles containing oncogenic EGFR. Proc Natl Acad Sci U S A. 2009;106(10):3794–9.PubMedPubMedCentralCrossRef Al-Nedawi K, Meehan B, Kerbel RS, Allison AC, Rak J. Endothelial expression of autocrine VEGF upon the uptake of tumor-derived microvesicles containing oncogenic EGFR. Proc Natl Acad Sci U S A. 2009;106(10):3794–9.PubMedPubMedCentralCrossRef
59.
go back to reference Ko SF, Yip HK, Zhen YY, Lee CC, Li JH, Lee CC, et al. Cancer patient-derived circulating microparticles enhance lung metastasis in a rat model: dual-source CT, cellular, and molecular studies. Mol Imaging Biol. 2016;18(4):490–9.PubMedCrossRef Ko SF, Yip HK, Zhen YY, Lee CC, Li JH, Lee CC, et al. Cancer patient-derived circulating microparticles enhance lung metastasis in a rat model: dual-source CT, cellular, and molecular studies. Mol Imaging Biol. 2016;18(4):490–9.PubMedCrossRef
60.
go back to reference Liu Y, Zhu XJ, Zeng C, Wu PH, Wang HX, Chen ZC, et al. Microvesicles secreted from human multiple myeloma cells promote angiogenesis. Acta Pharmacol Sin. 2014;35(2):230–8.PubMedCrossRef Liu Y, Zhu XJ, Zeng C, Wu PH, Wang HX, Chen ZC, et al. Microvesicles secreted from human multiple myeloma cells promote angiogenesis. Acta Pharmacol Sin. 2014;35(2):230–8.PubMedCrossRef
61.
go back to reference Giusti I, Delle Monache S, Di Francesco M, Sanita P, D'ascenzo S, Gravina GL, et al. From glioblastoma to endothelial cells through extracellular vesicles: messages for angiogenesis. Tumour Biol. 2016;37(9):12743–53.PubMedCrossRef Giusti I, Delle Monache S, Di Francesco M, Sanita P, D'ascenzo S, Gravina GL, et al. From glioblastoma to endothelial cells through extracellular vesicles: messages for angiogenesis. Tumour Biol. 2016;37(9):12743–53.PubMedCrossRef
62.
go back to reference Hong BS, Cho JH, Kim H, Choi EJ, Rho S, Kim J, et al. Colorectal cancer cell-derived microvesicles are enriched in cell cycle-related mRNAs that promote proliferation of endothelial cells. BMC Genomics. 2009;10:556.PubMedPubMedCentralCrossRef Hong BS, Cho JH, Kim H, Choi EJ, Rho S, Kim J, et al. Colorectal cancer cell-derived microvesicles are enriched in cell cycle-related mRNAs that promote proliferation of endothelial cells. BMC Genomics. 2009;10:556.PubMedPubMedCentralCrossRef
63.
go back to reference Grange C, Tapparo M, Collino F, Vitillo L, Damasco C, Deregibus MC, et al. Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 2011;71(15):5346–56.PubMedCrossRef Grange C, Tapparo M, Collino F, Vitillo L, Damasco C, Deregibus MC, et al. Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 2011;71(15):5346–56.PubMedCrossRef
64.
go back to reference Zhang H, Bai M, Deng T, Liu R, Wang X, Qu Y, et al. Cell-derived microvesicles mediate the delivery of miR-29a/c to suppress angiogenesis in gastric carcinoma. Cancer Lett. 2016;375(2):331–9.PubMedCrossRef Zhang H, Bai M, Deng T, Liu R, Wang X, Qu Y, et al. Cell-derived microvesicles mediate the delivery of miR-29a/c to suppress angiogenesis in gastric carcinoma. Cancer Lett. 2016;375(2):331–9.PubMedCrossRef
65.
go back to reference Yamada N, Tsujimura N, Kumazaki M, Shinohara H, Taniguchi K, Nakagawa Y, et al. Colorectal cancer cell-derived microvesicles containing microRNA-1246 promote angiogenesis by activating Smad 1/5/8 signaling elicited by PML down-regulation in endothelial cells. Biochim Biophys Acta. 2014;1839(11):1256–72.PubMedCrossRef Yamada N, Tsujimura N, Kumazaki M, Shinohara H, Taniguchi K, Nakagawa Y, et al. Colorectal cancer cell-derived microvesicles containing microRNA-1246 promote angiogenesis by activating Smad 1/5/8 signaling elicited by PML down-regulation in endothelial cells. Biochim Biophys Acta. 2014;1839(11):1256–72.PubMedCrossRef
66.
go back to reference Schlienger S, Campbell S, Claing A. ARF1 regulates the Rho/MLC pathway to control EGF-dependent breast cancer cell invasion. Mol Biol Cell. 2014;25(1):17–29.PubMedPubMedCentralCrossRef Schlienger S, Campbell S, Claing A. ARF1 regulates the Rho/MLC pathway to control EGF-dependent breast cancer cell invasion. Mol Biol Cell. 2014;25(1):17–29.PubMedPubMedCentralCrossRef
67.
go back to reference Li B, Antonyak MA, Zhang J, Cerione RA. RhoA triggers a specific signaling pathway that generates transforming microvesicles in cancer cells. Oncogene. 2012;31(45):4740–9.PubMedPubMedCentralCrossRef Li B, Antonyak MA, Zhang J, Cerione RA. RhoA triggers a specific signaling pathway that generates transforming microvesicles in cancer cells. Oncogene. 2012;31(45):4740–9.PubMedPubMedCentralCrossRef
68.
go back to reference El-Sayed IY, Daher A, Destouches D, Firlej V, Kostallari E, Maille P, et al. Extracellular vesicles released by mesenchymal-like prostate carcinoma cells modulate EMT state of recipient epithelial-like carcinoma cells through regulation of AR signaling. Cancer Lett. 2017;410:100–11.PubMedCrossRef El-Sayed IY, Daher A, Destouches D, Firlej V, Kostallari E, Maille P, et al. Extracellular vesicles released by mesenchymal-like prostate carcinoma cells modulate EMT state of recipient epithelial-like carcinoma cells through regulation of AR signaling. Cancer Lett. 2017;410:100–11.PubMedCrossRef
69.
go back to reference Zheng J, Wang J, Sun X, Hao M, Ding T, Xiong D, et al. HIC1 modulates prostate cancer progression by epigenetic modification. Clin Cancer Res. 2013;19(6):1400–10.PubMedCrossRef Zheng J, Wang J, Sun X, Hao M, Ding T, Xiong D, et al. HIC1 modulates prostate cancer progression by epigenetic modification. Clin Cancer Res. 2013;19(6):1400–10.PubMedCrossRef
70.
go back to reference Wang J, Shiozawa Y, Wang J, Wang Y, Jung Y, Pienta KJ, et al. The role of CXCR7/RDC1 as a chemokine receptor for CXCL12/SDF-1 in prostate cancer. J Biol Chem. 2008;283(7):4283–94.PubMedCrossRef Wang J, Shiozawa Y, Wang J, Wang Y, Jung Y, Pienta KJ, et al. The role of CXCR7/RDC1 as a chemokine receptor for CXCL12/SDF-1 in prostate cancer. J Biol Chem. 2008;283(7):4283–94.PubMedCrossRef
71.
go back to reference Kalinkovich A, Tavor S, Avigdor A, Kahn J, Brill A, Petit I, et al. Functional CXCR4-expressing microparticles and SDF-1 correlate with circulating acute myelogenous leukemia cells. Cancer Res. 2006;66(22):11013–20.PubMedCrossRef Kalinkovich A, Tavor S, Avigdor A, Kahn J, Brill A, Petit I, et al. Functional CXCR4-expressing microparticles and SDF-1 correlate with circulating acute myelogenous leukemia cells. Cancer Res. 2006;66(22):11013–20.PubMedCrossRef
72.
go back to reference Jaszczynska-Nowinka K, Rucinski M, Ziolkowska A, Markowska A, Malendowicz LK. Expression of SDF-1 and CXCR4 transcript variants and CXCR7 in epithelial ovarian cancer. Oncol Lett. 2014;7(5):1618–24.PubMedPubMedCentralCrossRef Jaszczynska-Nowinka K, Rucinski M, Ziolkowska A, Markowska A, Malendowicz LK. Expression of SDF-1 and CXCR4 transcript variants and CXCR7 in epithelial ovarian cancer. Oncol Lett. 2014;7(5):1618–24.PubMedPubMedCentralCrossRef
73.
go back to reference Lima LG, Leal AC, Vargas G, Porto-Carreiro I, Monteiro RQ. Intercellular transfer of tissue factor via the uptake of tumor-derived microvesicles. Thromb Res. 2013;132(4):450–6.PubMedCrossRef Lima LG, Leal AC, Vargas G, Porto-Carreiro I, Monteiro RQ. Intercellular transfer of tissue factor via the uptake of tumor-derived microvesicles. Thromb Res. 2013;132(4):450–6.PubMedCrossRef
74.
go back to reference Pang A, Cui Y, Chen Y, Cheng N, Delaney MK, Gu M, et al. Shear-induced integrin signaling in platelet phosphatidylserine exposure, microvesicle release, and coagulation. Blood. 2018;132(5):533–43.PubMedCrossRef Pang A, Cui Y, Chen Y, Cheng N, Delaney MK, Gu M, et al. Shear-induced integrin signaling in platelet phosphatidylserine exposure, microvesicle release, and coagulation. Blood. 2018;132(5):533–43.PubMedCrossRef
75.
go back to reference Crompot E, Van Damme M, Pieters K, Vermeersch M, Perez-Morga D, Mineur P, et al. Extracellular vesicles of bone marrow stromal cells rescue chronic lymphocytic leukemia B cells from apoptosis, enhance their migration and induce gene expression modifications. Haematologica. 2017;102(9):1594–604.PubMedPubMedCentralCrossRef Crompot E, Van Damme M, Pieters K, Vermeersch M, Perez-Morga D, Mineur P, et al. Extracellular vesicles of bone marrow stromal cells rescue chronic lymphocytic leukemia B cells from apoptosis, enhance their migration and induce gene expression modifications. Haematologica. 2017;102(9):1594–604.PubMedPubMedCentralCrossRef
76.
go back to reference Liebhardt S, Ditsch N, Nieuwland R, Rank A, Jeschke U, Von Koch F, et al. CEA-, Her2/neu-, BCRP- and Hsp27-positive microparticles in breast cancer patients. Anticancer Res. 2010;30(5):1707–12.PubMed Liebhardt S, Ditsch N, Nieuwland R, Rank A, Jeschke U, Von Koch F, et al. CEA-, Her2/neu-, BCRP- and Hsp27-positive microparticles in breast cancer patients. Anticancer Res. 2010;30(5):1707–12.PubMed
77.
go back to reference Lu JF, Luk F, Gong J, Jaiswal R, Grau GE, Bebawy M. Microparticles mediate MRP1 intercellular transfer and the re-templating of intrinsic resistance pathways. Pharmacol Res. 2013;76:77–83.PubMedCrossRef Lu JF, Luk F, Gong J, Jaiswal R, Grau GE, Bebawy M. Microparticles mediate MRP1 intercellular transfer and the re-templating of intrinsic resistance pathways. Pharmacol Res. 2013;76:77–83.PubMedCrossRef
78.
go back to reference Wei Y, Lai X, Yu S, Chen S, Ma Y, Zhang Y, et al. Exosomal miR-221/222 enhances tamoxifen resistance in recipient ER-positive breast cancer cells. Breast Cancer Res Treat. 2014;147(2):423–31.PubMedCrossRef Wei Y, Lai X, Yu S, Chen S, Ma Y, Zhang Y, et al. Exosomal miR-221/222 enhances tamoxifen resistance in recipient ER-positive breast cancer cells. Breast Cancer Res Treat. 2014;147(2):423–31.PubMedCrossRef
79.
go back to reference Jaiswal R, Luk F, Dalla PV, Grau GE, Bebawy M. Breast cancer-derived microparticles display tissue selectivity in the transfer of resistance proteins to cells. PLoS One. 2013;8(4):e61515.PubMedPubMedCentralCrossRef Jaiswal R, Luk F, Dalla PV, Grau GE, Bebawy M. Breast cancer-derived microparticles display tissue selectivity in the transfer of resistance proteins to cells. PLoS One. 2013;8(4):e61515.PubMedPubMedCentralCrossRef
80.
go back to reference De Souza PS, Cruz AL, Viola JP, Maia RC. Microparticles induce multifactorial resistance through oncogenic pathways independently of cancer cell type. Cancer Sci. 2015;106(1):60–8.PubMedCrossRef De Souza PS, Cruz AL, Viola JP, Maia RC. Microparticles induce multifactorial resistance through oncogenic pathways independently of cancer cell type. Cancer Sci. 2015;106(1):60–8.PubMedCrossRef
81.
go back to reference Shedden K, Xie XT, Chandaroy P, Chang YT, Rosania GR. Expulsion of small molecules in vesicles shed by cancer cells: association with gene expression and chemosensitivity profiles. Cancer Res. 2003;63(15):4331–7.PubMed Shedden K, Xie XT, Chandaroy P, Chang YT, Rosania GR. Expulsion of small molecules in vesicles shed by cancer cells: association with gene expression and chemosensitivity profiles. Cancer Res. 2003;63(15):4331–7.PubMed
82.
go back to reference Jorfi S, Ansa-Addo EA, Kholia S, Stratton D, Valley S, Lange S, et al. Inhibition of microvesiculation sensitizes prostate cancer cells to chemotherapy and reduces docetaxel dose required to limit tumor growth in vivo. Sci Rep. 2015;5:13006.PubMedPubMedCentralCrossRef Jorfi S, Ansa-Addo EA, Kholia S, Stratton D, Valley S, Lange S, et al. Inhibition of microvesiculation sensitizes prostate cancer cells to chemotherapy and reduces docetaxel dose required to limit tumor growth in vivo. Sci Rep. 2015;5:13006.PubMedPubMedCentralCrossRef
83.
go back to reference Kholia S, Jorfi S, Thompson PR, Causey CP, Nicholas AP, Inal JM, et al. A novel role for peptidylarginine deiminases in microvesicle release reveals therapeutic potential of PAD inhibition in sensitizing prostate cancer cells to chemotherapy. J Extracell Vesicles. 2015;4:26192.PubMedCrossRef Kholia S, Jorfi S, Thompson PR, Causey CP, Nicholas AP, Inal JM, et al. A novel role for peptidylarginine deiminases in microvesicle release reveals therapeutic potential of PAD inhibition in sensitizing prostate cancer cells to chemotherapy. J Extracell Vesicles. 2015;4:26192.PubMedCrossRef
84.
go back to reference Ma J, Zhang Y, Tang K, Zhang H, Yin X, Li Y, et al. Reversing drug resistance of soft tumor-repopulating cells by tumor cell-derived chemotherapeutic microparticles. Cell Res. 2016;26(6):713–27.PubMedPubMedCentralCrossRef Ma J, Zhang Y, Tang K, Zhang H, Yin X, Li Y, et al. Reversing drug resistance of soft tumor-repopulating cells by tumor cell-derived chemotherapeutic microparticles. Cell Res. 2016;26(6):713–27.PubMedPubMedCentralCrossRef
85.
go back to reference Zeelenberg IS, Ostrowski M, Krumeich S, Bobrie A, Jancic C, Boissonnas A, et al. Targeting tumor antigens to secreted membrane vesicles in vivo induces efficient antitumor immune responses. Cancer Res. 2008;68(4):1228–35.PubMedCrossRef Zeelenberg IS, Ostrowski M, Krumeich S, Bobrie A, Jancic C, Boissonnas A, et al. Targeting tumor antigens to secreted membrane vesicles in vivo induces efficient antitumor immune responses. Cancer Res. 2008;68(4):1228–35.PubMedCrossRef
86.
go back to reference Napoletano C, Rughetti A, Landi R, Pinto D, Bellati F, Rahimi H, et al. Immunogenicity of Allo-vesicle carrying ERBB2 tumor antigen for dendritic cell-based anti-tumor immunotherapy. Int J Immunopathol Pharmacol. 2009;22(3):647–58.PubMedCrossRef Napoletano C, Rughetti A, Landi R, Pinto D, Bellati F, Rahimi H, et al. Immunogenicity of Allo-vesicle carrying ERBB2 tumor antigen for dendritic cell-based anti-tumor immunotherapy. Int J Immunopathol Pharmacol. 2009;22(3):647–58.PubMedCrossRef
87.
go back to reference Moore C, Kosgodage U, Lange S, Inal JM. The emerging role of exosome and microvesicle- (EMV-) based cancer therapeutics and immunotherapy. Int J Cancer. 2017;141(3):428–36.PubMedCrossRef Moore C, Kosgodage U, Lange S, Inal JM. The emerging role of exosome and microvesicle- (EMV-) based cancer therapeutics and immunotherapy. Int J Cancer. 2017;141(3):428–36.PubMedCrossRef
88.
go back to reference Maus RLG, Jakub JW, Nevala WK, Christensen TA, Noble-Orcutt K, Sachs Z, et al. Human melanoma-derived extracellular vesicles regulate dendritic cell maturation. Front Immunol. 2017;8:358.PubMedPubMedCentralCrossRef Maus RLG, Jakub JW, Nevala WK, Christensen TA, Noble-Orcutt K, Sachs Z, et al. Human melanoma-derived extracellular vesicles regulate dendritic cell maturation. Front Immunol. 2017;8:358.PubMedPubMedCentralCrossRef
89.
go back to reference Valenti R, Huber V, Filipazzi P, Pilla L, Sovena G, Villa A, et al. Human tumor-released microvesicles promote the differentiation of myeloid cells with transforming growth factor-beta-mediated suppressive activity on T lymphocytes. Cancer Res. 2006;66(18):9290–8.PubMedCrossRef Valenti R, Huber V, Filipazzi P, Pilla L, Sovena G, Villa A, et al. Human tumor-released microvesicles promote the differentiation of myeloid cells with transforming growth factor-beta-mediated suppressive activity on T lymphocytes. Cancer Res. 2006;66(18):9290–8.PubMedCrossRef
90.
go back to reference Rughetti A, Rahimi H, Belleudi F, Napoletano C, Battisti F, Zizzari IG, et al. Microvesicle cargo of tumor-associated MUC1 to dendritic cells allows cross-presentation and specific carbohydrate processing. Cancer Immunol Res. 2014;2(2):177–86.PubMedCrossRef Rughetti A, Rahimi H, Belleudi F, Napoletano C, Battisti F, Zizzari IG, et al. Microvesicle cargo of tumor-associated MUC1 to dendritic cells allows cross-presentation and specific carbohydrate processing. Cancer Immunol Res. 2014;2(2):177–86.PubMedCrossRef
91.
go back to reference Battisti F, Napoletano C, Rahimi Koshkaki H, Belleudi F, Zizzari IG, Ruscito I, et al. Tumor-derived microvesicles modulate antigen cross-processing via reactive oxygen species-mediated Alkalinization of Phagosomal compartment in dendritic cells. Front Immunol. 2017;8:1179.PubMedPubMedCentralCrossRef Battisti F, Napoletano C, Rahimi Koshkaki H, Belleudi F, Zizzari IG, Ruscito I, et al. Tumor-derived microvesicles modulate antigen cross-processing via reactive oxygen species-mediated Alkalinization of Phagosomal compartment in dendritic cells. Front Immunol. 2017;8:1179.PubMedPubMedCentralCrossRef
92.
go back to reference Ruffner MA, Kim SH, Bianco NR, Francisco LM, Sharpe AH, Robbins PD. B7-1/2, but not PD-L1/2 molecules, are required on IL-10-treated tolerogenic DC and DC-derived exosomes for in vivo function. Eur J Immunol. 2009;39(11):3084–90.PubMedPubMedCentralCrossRef Ruffner MA, Kim SH, Bianco NR, Francisco LM, Sharpe AH, Robbins PD. B7-1/2, but not PD-L1/2 molecules, are required on IL-10-treated tolerogenic DC and DC-derived exosomes for in vivo function. Eur J Immunol. 2009;39(11):3084–90.PubMedPubMedCentralCrossRef
93.
go back to reference Ricklefs FL, Alayo Q, Krenzlin H, Mahmoud AB, Speranza MC, Nakashima H, et al. Immune evasion mediated by PD-L1 on glioblastoma-derived extracellular vesicles. Sci Adv. 2018;4;(3):eaar2766. Ricklefs FL, Alayo Q, Krenzlin H, Mahmoud AB, Speranza MC, Nakashima H, et al. Immune evasion mediated by PD-L1 on glioblastoma-derived extracellular vesicles. Sci Adv. 2018;4;(3):eaar2766.
94.
go back to reference Martinez VG, O'neill S, Salimu J, Breslin S, Clayton A, Crown J, et al. Resistance to HER2-targeted anti-cancer drugs is associated with immune evasion in cancer cells and their derived extracellular vesicles. Oncoimmunology. 2017;6(12):e1362530.PubMedPubMedCentralCrossRef Martinez VG, O'neill S, Salimu J, Breslin S, Clayton A, Crown J, et al. Resistance to HER2-targeted anti-cancer drugs is associated with immune evasion in cancer cells and their derived extracellular vesicles. Oncoimmunology. 2017;6(12):e1362530.PubMedPubMedCentralCrossRef
95.
go back to reference Iorgulescu JB, Ivan ME, Safaee M, Parsa AT. The limited capacity of malignant glioma-derived exosomes to suppress peripheral immune effectors. J Neuroimmunol. 2016;290:103–8.PubMedCrossRef Iorgulescu JB, Ivan ME, Safaee M, Parsa AT. The limited capacity of malignant glioma-derived exosomes to suppress peripheral immune effectors. J Neuroimmunol. 2016;290:103–8.PubMedCrossRef
96.
go back to reference Zhang H, Tang K, Zhang Y, Ma R, Ma J, Li Y, et al. Cell-free tumor microparticle vaccines stimulate dendritic cells via cGAS/STING signaling. Cancer Immunol Res. 2015;3(2):196–205.PubMedCrossRef Zhang H, Tang K, Zhang Y, Ma R, Ma J, Li Y, et al. Cell-free tumor microparticle vaccines stimulate dendritic cells via cGAS/STING signaling. Cancer Immunol Res. 2015;3(2):196–205.PubMedCrossRef
Metadata
Title
Microvesicles and chemokines in tumor microenvironment: mediators of intercellular communications in tumor progression
Authors
Xiaojie Bian
Yu-Tian Xiao
Tianqi Wu
Mengfei Yao
Leilei Du
Shancheng Ren
Jianhua Wang
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2019
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-019-0973-7

Other articles of this Issue 1/2019

Molecular Cancer 1/2019 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