Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

Open Access 01-12-2021 | Human Papillomavirus | Review

The roles of extracellular vesicles in the development, microenvironment, anticancer drug resistance, and therapy of head and neck squamous cell carcinoma

Authors: Xueying Wang, Junnan Guo, Pingyang Yu, Lunhua Guo, Xionghui Mao, Junrong Wang, Susheng Miao, Ji Sun

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

Abstract

Head and neck squamous cell carcinoma (HNSCC) is one of the main malignant tumours affecting human health, mainly due to delayed diagnosis and high invasiveness. Extracellular vehicles (EVs) are membranous vesicles released by cells into the extracellular matrix that carry important signalling molecules and stably and widely exist in various body fluids, such as plasma, saliva, cerebrospinal fluid, breast milk, urine, semen, lymphatic fluid, synovial fluid, amniotic fluid, and sputum. EVs transport almost all types of bioactive molecules (DNA, mRNAs, microRNAs (miRNAs), proteins, metabolites, and even pharmacological compounds). These “cargoes” can act on recipient cells, reshaping the surrounding microenvironment and altering distant targets, ultimately affecting their biological behaviour. The extensive exploration of EVs has deepened our comprehensive understanding of HNSCC biology. In this review, we not only summarized the effect of HNSCC-derived EVs on the tumour microenvironment but also described the role of microenvironment-derived EVs in HNSCC and discussed how the “mutual dialogue” between the tumour and microenvironment mediates the growth, metastasis, angiogenesis, immune escape, and drug resistance of tumours. Finally, the clinical application of EVS in HNSCC was assessed.
Literature
1.
go back to reference Leemans CR, Snijders PJF, Brakenhoff RH. Publisher Correction: The molecular landscape of head and neck cancer. Nat Rev Cancer. 2018;18(10):662.PubMedCrossRef Leemans CR, Snijders PJF, Brakenhoff RH. Publisher Correction: The molecular landscape of head and neck cancer. Nat Rev Cancer. 2018;18(10):662.PubMedCrossRef
2.
go back to reference Cohen EEW, Bell RB, Bifulco CB, Burtness B, Gillison ML, Harrington KJ, et al. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of squamous cell carcinoma of the head and neck (HNSCC). J Immunother Cancer. 2019;7(1):184.PubMedPubMedCentralCrossRef Cohen EEW, Bell RB, Bifulco CB, Burtness B, Gillison ML, Harrington KJ, et al. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of squamous cell carcinoma of the head and neck (HNSCC). J Immunother Cancer. 2019;7(1):184.PubMedPubMedCentralCrossRef
3.
go back to reference Marur S, Forastiere AA. Head and neck cancer: changing epidemiology, diagnosis, and treatment. Mayo Clin Proc. 2008;83(4):489–501.PubMedCrossRef Marur S, Forastiere AA. Head and neck cancer: changing epidemiology, diagnosis, and treatment. Mayo Clin Proc. 2008;83(4):489–501.PubMedCrossRef
4.
go back to reference Castellsague X, Quintana MJ, Martinez MC, Nieto A, Sanchez MJ, Juan A, et al. The role of type of tobacco and type of alcoholic beverage in oral carcinogenesis. Int J Cancer. 2004;108(5):741–9.PubMedCrossRef Castellsague X, Quintana MJ, Martinez MC, Nieto A, Sanchez MJ, Juan A, et al. The role of type of tobacco and type of alcoholic beverage in oral carcinogenesis. Int J Cancer. 2004;108(5):741–9.PubMedCrossRef
5.
go back to reference Carlander AF, Gronhoj Larsen C, Jensen DH, Garnaes E, Kiss K, Andersen L, et al. Continuing rise in oropharyngeal cancer in a high HPV prevalence area: A Danish population-based study from 2011 to 2014. Eur J Cancer. 2017;70:75–82.PubMedCrossRef Carlander AF, Gronhoj Larsen C, Jensen DH, Garnaes E, Kiss K, Andersen L, et al. Continuing rise in oropharyngeal cancer in a high HPV prevalence area: A Danish population-based study from 2011 to 2014. Eur J Cancer. 2017;70:75–82.PubMedCrossRef
6.
go back to reference D’Souza G, Kreimer AR, Viscidi R, Pawlita M, Fakhry C, Koch WM, et al. Case-control study of human papillomavirus and oropharyngeal cancer. N Engl J Med. 2007;356(19):1944–56.PubMedCrossRef D’Souza G, Kreimer AR, Viscidi R, Pawlita M, Fakhry C, Koch WM, et al. Case-control study of human papillomavirus and oropharyngeal cancer. N Engl J Med. 2007;356(19):1944–56.PubMedCrossRef
7.
go back to reference Mork J, Lie AK, Glattre E, Hallmans G, Jellum E, Koskela P, et al. Human papillomavirus infection as a risk factor for squamous-cell carcinoma of the head and neck. N Engl J Med. 2001;344(15):1125–31.PubMedCrossRef Mork J, Lie AK, Glattre E, Hallmans G, Jellum E, Koskela P, et al. Human papillomavirus infection as a risk factor for squamous-cell carcinoma of the head and neck. N Engl J Med. 2001;344(15):1125–31.PubMedCrossRef
8.
go back to reference Chi AC, Day TA, Neville BW. Oral cavity and oropharyngeal squamous cell carcinoma–an update. CA Cancer J Clin. 2015;65(5):401–21.PubMedCrossRef Chi AC, Day TA, Neville BW. Oral cavity and oropharyngeal squamous cell carcinoma–an update. CA Cancer J Clin. 2015;65(5):401–21.PubMedCrossRef
9.
go back to reference Epstein JB, Thariat J, Bensadoun RJ, Barasch A, Murphy BA, Kolnick L, et al. Oral complications of cancer and cancer therapy: from cancer treatment to survivorship. CA Cancer J Clin. 2012;62(6):400–22.PubMedCrossRef Epstein JB, Thariat J, Bensadoun RJ, Barasch A, Murphy BA, Kolnick L, et al. Oral complications of cancer and cancer therapy: from cancer treatment to survivorship. CA Cancer J Clin. 2012;62(6):400–22.PubMedCrossRef
10.
go back to reference O’Loughlin AJ, Woffindale CA, Wood MJ. Exosomes and the emerging field of exosome-based gene therapy. Curr Gene Ther. 2012;12(4):262–74.PubMedCrossRef O’Loughlin AJ, Woffindale CA, Wood MJ. Exosomes and the emerging field of exosome-based gene therapy. Curr Gene Ther. 2012;12(4):262–74.PubMedCrossRef
11.
go back to reference Ono R, Yoshioka Y, Furukawa Y, Naruse M, Kuwagata M, Ochiya T, et al. Novel hepatotoxicity biomarkers of extracellular vesicle (EV)-associated miRNAs induced by CCl4. Toxicol Rep. 2020;7:685–92.PubMedPubMedCentralCrossRef Ono R, Yoshioka Y, Furukawa Y, Naruse M, Kuwagata M, Ochiya T, et al. Novel hepatotoxicity biomarkers of extracellular vesicle (EV)-associated miRNAs induced by CCl4. Toxicol Rep. 2020;7:685–92.PubMedPubMedCentralCrossRef
13.
go back to reference Willis GR, Kourembanas S, Mitsialis SA. Toward Exosome-Based Therapeutics: Isolation, Heterogeneity, and Fit-for-Purpose Potency. Front Cardiovasc Med. 2017;4:63.PubMedPubMedCentralCrossRef Willis GR, Kourembanas S, Mitsialis SA. Toward Exosome-Based Therapeutics: Isolation, Heterogeneity, and Fit-for-Purpose Potency. Front Cardiovasc Med. 2017;4:63.PubMedPubMedCentralCrossRef
14.
go back to reference D’Acunzo P, Hargash T, Pawlik M, Goulbourne CN, Perez-Gonzalez R, Levy E. Enhanced generation of intraluminal vesicles in neuronal late endosomes in the brain of a Down syndrome mouse model with endosomal dysfunction. Dev Neurobiol. 2019;79(7):656–63.PubMedPubMedCentralCrossRef D’Acunzo P, Hargash T, Pawlik M, Goulbourne CN, Perez-Gonzalez R, Levy E. Enhanced generation of intraluminal vesicles in neuronal late endosomes in the brain of a Down syndrome mouse model with endosomal dysfunction. Dev Neurobiol. 2019;79(7):656–63.PubMedPubMedCentralCrossRef
15.
go back to reference Vietri M, Radulovic M, Stenmark H. The many functions of ESCRTs. Nat Rev Mol Cell Biol. 2020;21(1):25–42.PubMedCrossRef Vietri M, Radulovic M, Stenmark H. The many functions of ESCRTs. Nat Rev Mol Cell Biol. 2020;21(1):25–42.PubMedCrossRef
16.
go back to reference Stuffers S, Sem Wegner C, Stenmark H, Brech A. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic. 2009;10(7):925–37.PubMedCrossRef Stuffers S, Sem Wegner C, Stenmark H, Brech A. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic. 2009;10(7):925–37.PubMedCrossRef
19.
go back to reference Langemeyer L, Frohlich F, Ungermann C. Rab GTPase Function in Endosome and Lysosome Biogenesis. Trends Cell Biol. 2018;28(11):957–70.PubMedCrossRef Langemeyer L, Frohlich F, Ungermann C. Rab GTPase Function in Endosome and Lysosome Biogenesis. Trends Cell Biol. 2018;28(11):957–70.PubMedCrossRef
20.
go back to reference Jongsma ML, Berlin I, Wijdeven RH, Janssen L, Janssen GM, Garstka MA, et al. An ER-Associated Pathway Defines Endosomal Architecture for Controlled Cargo Transport. Cell. 2016;166(1):152–66.PubMedPubMedCentralCrossRef Jongsma ML, Berlin I, Wijdeven RH, Janssen L, Janssen GM, Garstka MA, et al. An ER-Associated Pathway Defines Endosomal Architecture for Controlled Cargo Transport. Cell. 2016;166(1):152–66.PubMedPubMedCentralCrossRef
22.
go back to reference Pollet H, Conrard L, Cloos AS, Tyteca D. Plasma Membrane Lipid Domains as Platforms for Vesicle Biogenesis and Shedding? Biomolecules. 2018;8(3). Pollet H, Conrard L, Cloos AS, Tyteca D. Plasma Membrane Lipid Domains as Platforms for Vesicle Biogenesis and Shedding? Biomolecules. 2018;8(3).
23.
go back to reference Nabhan JF, Hu R, Oh RS, Cohen SN, Lu Q. Formation and release of arrestin domain-containing protein 1-mediated microvesicles (ARMMs) at plasma membrane by recruitment of TSG101 protein. Proc Natl Acad Sci U S A. 2012;109(11):4146–51.PubMedPubMedCentralCrossRef Nabhan JF, Hu R, Oh RS, Cohen SN, Lu Q. Formation and release of arrestin domain-containing protein 1-mediated microvesicles (ARMMs) at plasma membrane by recruitment of TSG101 protein. Proc Natl Acad Sci U S A. 2012;109(11):4146–51.PubMedPubMedCentralCrossRef
24.
go back to reference Stachowiak JC, Schmid EM, Ryan CJ, Ann HS, Sasaki DY, Sherman MB, et al. Membrane bending by protein-protein crowding. Nat Cell Biol. 2012;14(9):944–9.PubMedCrossRef Stachowiak JC, Schmid EM, Ryan CJ, Ann HS, Sasaki DY, Sherman MB, et al. Membrane bending by protein-protein crowding. Nat Cell Biol. 2012;14(9):944–9.PubMedCrossRef
25.
go back to reference Crawford S, Diamond D, Brustolon L, Penarreta R. Effect of increased extracellular ca on microvesicle production and tumor spheroid formation. Cancer Microenviron. 2010;4(1):93–103.PubMedPubMedCentralCrossRef Crawford S, Diamond D, Brustolon L, Penarreta R. Effect of increased extracellular ca on microvesicle production and tumor spheroid formation. Cancer Microenviron. 2010;4(1):93–103.PubMedPubMedCentralCrossRef
26.
go back to reference Dolis D, Moreau C, Zachowski A, Devaux PF. Aminophospholipid translocase and proteins involved in transmembrane phospholipid traffic. Biophys Chem. 1997;68(1–3):221–31.PubMedCrossRef Dolis D, Moreau C, Zachowski A, Devaux PF. Aminophospholipid translocase and proteins involved in transmembrane phospholipid traffic. Biophys Chem. 1997;68(1–3):221–31.PubMedCrossRef
27.
go back to reference Yu SL, Gan XG, Huang JM, Cao Y, Wang YQ, Pan SH, et al. Oxalate impairs aminophospholipid translocase activity in renal epithelial cells via oxidative stress: implications for calcium oxalate urolithiasis. J Urol. 2011;186(3):1114–20.PubMedCrossRef Yu SL, Gan XG, Huang JM, Cao Y, Wang YQ, Pan SH, et al. Oxalate impairs aminophospholipid translocase activity in renal epithelial cells via oxidative stress: implications for calcium oxalate urolithiasis. J Urol. 2011;186(3):1114–20.PubMedCrossRef
28.
go back to reference Usman WM, Pham TC, Kwok YY, Vu LT, Ma V, Peng B, et al. Efficient RNA drug delivery using red blood cell extracellular vesicles. Nat Commun. 2018;9(1):2359.PubMedPubMedCentralCrossRef Usman WM, Pham TC, Kwok YY, Vu LT, Ma V, Peng B, et al. Efficient RNA drug delivery using red blood cell extracellular vesicles. Nat Commun. 2018;9(1):2359.PubMedPubMedCentralCrossRef
29.
go back to reference Cocucci E, Meldolesi J. Ectosomes and exosomes: shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015;25(6):364–72.PubMedCrossRef Cocucci E, Meldolesi J. Ectosomes and exosomes: shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015;25(6):364–72.PubMedCrossRef
30.
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
31.
go back to reference Oropesa Avila M, Fernandez Vega A, Garrido Maraver J, Villanueva Paz M, De Lavera I, De La Mata M, et al. Emerging roles of apoptotic microtubules during the execution phase of apoptosis. Cytoskeleton (Hoboken). 2015;72(9):435–46.CrossRef Oropesa Avila M, Fernandez Vega A, Garrido Maraver J, Villanueva Paz M, De Lavera I, De La Mata M, et al. Emerging roles of apoptotic microtubules during the execution phase of apoptosis. Cytoskeleton (Hoboken). 2015;72(9):435–46.CrossRef
34.
go back to reference Xu X, Lai Y, Hua ZC. Apoptosis and apoptotic body: disease message and therapeutic target potentials. Biosci Rep. 2019;39(1). Xu X, Lai Y, Hua ZC. Apoptosis and apoptotic body: disease message and therapeutic target potentials. Biosci Rep. 2019;39(1).
36.
go back to reference Xie C, Ji N, Tang Z, Li J, Chen Q. The role of extracellular vesicles from different origin in the microenvironment of head and neck cancers. Mol Cancer. 2019;18(1):83.PubMedPubMedCentralCrossRef Xie C, Ji N, Tang Z, Li J, Chen Q. The role of extracellular vesicles from different origin in the microenvironment of head and neck cancers. Mol Cancer. 2019;18(1):83.PubMedPubMedCentralCrossRef
37.
go back to reference Durcin M, Fleury A, Taillebois E, Hilairet G, Krupova Z, Henry C, et al. Characterisation of adipocyte-derived extracellular vesicle subtypes identifies distinct protein and lipid signatures for large and small extracellular vesicles. J Extracell Vesicles. 2017;6(1):1305677.PubMedPubMedCentralCrossRef Durcin M, Fleury A, Taillebois E, Hilairet G, Krupova Z, Henry C, et al. Characterisation of adipocyte-derived extracellular vesicle subtypes identifies distinct protein and lipid signatures for large and small extracellular vesicles. J Extracell Vesicles. 2017;6(1):1305677.PubMedPubMedCentralCrossRef
38.
go back to reference Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci U S A. 2016;113(8):E968-77.PubMedCrossRef Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci U S A. 2016;113(8):E968-77.PubMedCrossRef
39.
go back to reference Ono K, Eguchi T, Sogawa C, Calderwood SK, Futagawa J, Kasai T, et al. HSP-enriched properties of extracellular vesicles involve survival of metastatic oral cancer cells. J Cell Biochem. 2018;119(9):7350–62.PubMedCrossRef Ono K, Eguchi T, Sogawa C, Calderwood SK, Futagawa J, Kasai T, et al. HSP-enriched properties of extracellular vesicles involve survival of metastatic oral cancer cells. J Cell Biochem. 2018;119(9):7350–62.PubMedCrossRef
40.
go back to reference Overmiller AM, Pierluissi JA, Wermuth PJ, Sauma S, Martinez-Outschoorn U, Tuluc M, et al. Desmoglein 2 modulates extracellular vesicle release from squamous cell carcinoma keratinocytes. FASEB J. 2017;31(8):3412–24.PubMedPubMedCentralCrossRef Overmiller AM, Pierluissi JA, Wermuth PJ, Sauma S, Martinez-Outschoorn U, Tuluc M, et al. Desmoglein 2 modulates extracellular vesicle release from squamous cell carcinoma keratinocytes. FASEB J. 2017;31(8):3412–24.PubMedPubMedCentralCrossRef
41.
go back to reference Shan Y, You B, Shi S, Shi W, Zhang Z, Zhang Q, et al. Hypoxia-Induced Matrix Metalloproteinase-13 Expression in Exosomes from Nasopharyngeal Carcinoma Enhances Metastases. Cell Death Dis. 2018;9(3):382.PubMedPubMedCentralCrossRef Shan Y, You B, Shi S, Shi W, Zhang Z, Zhang Q, et al. Hypoxia-Induced Matrix Metalloproteinase-13 Expression in Exosomes from Nasopharyngeal Carcinoma Enhances Metastases. Cell Death Dis. 2018;9(3):382.PubMedPubMedCentralCrossRef
42.
go back to reference You Y, Shan Y, Chen J, Yue H, You B, Shi S, et al. Matrix metalloproteinase 13-containing exosomes promote nasopharyngeal carcinoma metastasis. Cancer Sci. 2015;106(12):1669–77.PubMedPubMedCentralCrossRef You Y, Shan Y, Chen J, Yue H, You B, Shi S, et al. Matrix metalloproteinase 13-containing exosomes promote nasopharyngeal carcinoma metastasis. Cancer Sci. 2015;106(12):1669–77.PubMedPubMedCentralCrossRef
43.
44.
go back to reference Lousada-Fernandez F, Rapado-Gonzalez O, Lopez-Cedrun JL, Lopez-Lopez R, Muinelo-Romay L, Suarez-Cunqueiro MM. Liquid Biopsy in Oral Cancer. Int J Mol Sci. 2018;19(6). Lousada-Fernandez F, Rapado-Gonzalez O, Lopez-Cedrun JL, Lopez-Lopez R, Muinelo-Romay L, Suarez-Cunqueiro MM. Liquid Biopsy in Oral Cancer. Int J Mol Sci. 2018;19(6).
45.
go back to reference Wang J, Zhou Y, Lu J, Sun Y, Xiao H, Liu M, et al. Combined detection of serum exosomal miR-21 and HOTAIR as diagnostic and prognostic biomarkers for laryngeal squamous cell carcinoma. Med Oncol. 2014;31(9):148.PubMedCrossRef Wang J, Zhou Y, Lu J, Sun Y, Xiao H, Liu M, et al. Combined detection of serum exosomal miR-21 and HOTAIR as diagnostic and prognostic biomarkers for laryngeal squamous cell carcinoma. Med Oncol. 2014;31(9):148.PubMedCrossRef
46.
go back to reference Li L, Li C, Wang S, Wang Z, Jiang J, Wang W, et al. Exosomes Derived from Hypoxic Oral Squamous Cell Carcinoma Cells Deliver miR-21 to Normoxic Cells to Elicit a Prometastatic Phenotype. Cancer Res. 2016;76(7):1770–80.PubMedCrossRef Li L, Li C, Wang S, Wang Z, Jiang J, Wang W, et al. Exosomes Derived from Hypoxic Oral Squamous Cell Carcinoma Cells Deliver miR-21 to Normoxic Cells to Elicit a Prometastatic Phenotype. Cancer Res. 2016;76(7):1770–80.PubMedCrossRef
47.
go back to reference Ludwig S, Sharma P, Theodoraki MN, Pietrowska M, Yerneni SS, Lang S, et al. Molecular and Functional Profiles of Exosomes From HPV(+) and HPV(-) Head and Neck Cancer Cell Lines. Front Oncol. 2018;8:445.PubMedPubMedCentralCrossRef Ludwig S, Sharma P, Theodoraki MN, Pietrowska M, Yerneni SS, Lang S, et al. Molecular and Functional Profiles of Exosomes From HPV(+) and HPV(-) Head and Neck Cancer Cell Lines. Front Oncol. 2018;8:445.PubMedPubMedCentralCrossRef
48.
go back to reference Wang X, Qin X, Yan M, Shi J, Xu Q, Li Z, et al. Loss of exosomal miR-3188 in cancer-associated fibroblasts contributes to HNC progression. J Exp Clin Cancer Res. 2019;38(1):151.PubMedPubMedCentralCrossRef Wang X, Qin X, Yan M, Shi J, Xu Q, Li Z, et al. Loss of exosomal miR-3188 in cancer-associated fibroblasts contributes to HNC progression. J Exp Clin Cancer Res. 2019;38(1):151.PubMedPubMedCentralCrossRef
49.
go back to reference Xiang X, Poliakov A, Liu C, Liu Y, Deng ZB, Wang J, et al. Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer. 2009;124(11):2621–33.PubMedPubMedCentralCrossRef Xiang X, Poliakov A, Liu C, Liu Y, Deng ZB, Wang J, et al. Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer. 2009;124(11):2621–33.PubMedPubMedCentralCrossRef
50.
go back to reference Gu M, Li L, Zhang Z, Chen J, Zhang W, Zhang J, et al. PFKFB3 promotes proliferation, migration and angiogenesis in nasopharyngeal carcinoma. J Cancer. 2017;8(18):3887–96.PubMedPubMedCentralCrossRef Gu M, Li L, Zhang Z, Chen J, Zhang W, Zhang J, et al. PFKFB3 promotes proliferation, migration and angiogenesis in nasopharyngeal carcinoma. J Cancer. 2017;8(18):3887–96.PubMedPubMedCentralCrossRef
51.
go back to reference Huaitong X, Yuanyong F, Yueqin T, Peng Z, Wei S, Kai S. Microvesicles releasing by oral cancer cells enhance endothelial cell angiogenesis via Shh/RhoA signaling pathway. Cancer Biol Ther. 2017;18(10):783–91.PubMedPubMedCentralCrossRef Huaitong X, Yuanyong F, Yueqin T, Peng Z, Wei S, Kai S. Microvesicles releasing by oral cancer cells enhance endothelial cell angiogenesis via Shh/RhoA signaling pathway. Cancer Biol Ther. 2017;18(10):783–91.PubMedPubMedCentralCrossRef
52.
go back to reference Bao L, You B, Shi S, Shan Y, Zhang Q, Yue H, et al. Metastasis-associated miR-23a from nasopharyngeal carcinoma-derived exosomes mediates angiogenesis by repressing a novel target gene TSGA10. Oncogene. 2018;37(21):2873–89.PubMedPubMedCentralCrossRef Bao L, You B, Shi S, Shan Y, Zhang Q, Yue H, et al. Metastasis-associated miR-23a from nasopharyngeal carcinoma-derived exosomes mediates angiogenesis by repressing a novel target gene TSGA10. Oncogene. 2018;37(21):2873–89.PubMedPubMedCentralCrossRef
53.
go back to reference Sento S, Sasabe E, Yamamoto T. Application of a Persistent Heparin Treatment Inhibits the Malignant Potential of Oral Squamous Carcinoma Cells Induced by Tumor Cell-Derived Exosomes. PLoS One. 2016;11(2):e0148454.PubMedPubMedCentralCrossRef Sento S, Sasabe E, Yamamoto T. Application of a Persistent Heparin Treatment Inhibits the Malignant Potential of Oral Squamous Carcinoma Cells Induced by Tumor Cell-Derived Exosomes. PLoS One. 2016;11(2):e0148454.PubMedPubMedCentralCrossRef
54.
go back to reference Kawakubo-Yasukochi T, Morioka M, Hazekawa M, Yasukochi A, Nishinakagawa T, Ono K, et al. miR-200c-3p spreads invasive capacity in human oral squamous cell carcinoma microenvironment. Mol Carcinog. 2018;57(2):295–302.PubMedCrossRef Kawakubo-Yasukochi T, Morioka M, Hazekawa M, Yasukochi A, Nishinakagawa T, Ono K, et al. miR-200c-3p spreads invasive capacity in human oral squamous cell carcinoma microenvironment. Mol Carcinog. 2018;57(2):295–302.PubMedCrossRef
55.
go back to reference Sakha S, Muramatsu T, Ueda K, Inazawa J. Exosomal microRNA miR-1246 induces cell motility and invasion through the regulation of DENND2D in oral squamous cell carcinoma. Sci Rep. 2016;6:38750.PubMedPubMedCentralCrossRef Sakha S, Muramatsu T, Ueda K, Inazawa J. Exosomal microRNA miR-1246 induces cell motility and invasion through the regulation of DENND2D in oral squamous cell carcinoma. Sci Rep. 2016;6:38750.PubMedPubMedCentralCrossRef
56.
go back to reference Guenat D, Hermetet F, Pretet JL, Mougin C. Exosomes and Other Extracellular Vesicles in HPV Transmission and Carcinogenesis. Viruses. 2017;9(8). Guenat D, Hermetet F, Pretet JL, Mougin C. Exosomes and Other Extracellular Vesicles in HPV Transmission and Carcinogenesis. Viruses. 2017;9(8).
57.
go back to reference Harden ME, Munger K. Human papillomavirus 16 E6 and E7 oncoprotein expression alters microRNA expression in extracellular vesicles. Virology. 2017;508:63–9.PubMedCrossRef Harden ME, Munger K. Human papillomavirus 16 E6 and E7 oncoprotein expression alters microRNA expression in extracellular vesicles. Virology. 2017;508:63–9.PubMedCrossRef
58.
go back to reference Honegger A, Schilling D, Bastian S, Sponagel J, Kuryshev V, Sultmann H, et al. Dependence of intracellular and exosomal microRNAs on viral E6/E7 oncogene expression in HPV-positive tumor cells. PLoS Pathog. 2015;11(3):e1004712.PubMedPubMedCentralCrossRef Honegger A, Schilling D, Bastian S, Sponagel J, Kuryshev V, Sultmann H, et al. Dependence of intracellular and exosomal microRNAs on viral E6/E7 oncogene expression in HPV-positive tumor cells. PLoS Pathog. 2015;11(3):e1004712.PubMedPubMedCentralCrossRef
59.
go back to reference Hurwitz SN, Nkosi D, Conlon MM, York SB, Liu X, Tremblay DC, et al. CD63 Regulates Epstein-Barr Virus LMP1 Exosomal Packaging, Enhancement of Vesicle Production, and Noncanonical NF-kappaB Signaling. J Virol. 2017;91(5). Hurwitz SN, Nkosi D, Conlon MM, York SB, Liu X, Tremblay DC, et al. CD63 Regulates Epstein-Barr Virus LMP1 Exosomal Packaging, Enhancement of Vesicle Production, and Noncanonical NF-kappaB Signaling. J Virol. 2017;91(5).
60.
go back to reference Yoshizaki T, Kondo S, Wakisaka N, Murono S, Endo K, Sugimoto H, et al. Pathogenic role of Epstein-Barr virus latent membrane protein-1 in the development of nasopharyngeal carcinoma. Cancer Lett. 2013;337(1):1–7.PubMedCrossRef Yoshizaki T, Kondo S, Wakisaka N, Murono S, Endo K, Sugimoto H, et al. Pathogenic role of Epstein-Barr virus latent membrane protein-1 in the development of nasopharyngeal carcinoma. Cancer Lett. 2013;337(1):1–7.PubMedCrossRef
61.
go back to reference Meckes DG Jr, Shair KH, Marquitz AR, Kung CP, Edwards RH, Raab-Traub N. Human tumor virus utilizes exosomes for intercellular communication. Proc Natl Acad Sci U S A. 2010;107(47):20370–5.PubMedPubMedCentralCrossRef Meckes DG Jr, Shair KH, Marquitz AR, Kung CP, Edwards RH, Raab-Traub N. Human tumor virus utilizes exosomes for intercellular communication. Proc Natl Acad Sci U S A. 2010;107(47):20370–5.PubMedPubMedCentralCrossRef
62.
go back to reference Gourzones C, Gelin A, Bombik I, Klibi J, Verillaud B, Guigay J, et al. Extra-cellular release and blood diffusion of BART viral micro-RNAs produced by EBV-infected nasopharyngeal carcinoma cells. Virol J. 2010;7:271.PubMedPubMedCentralCrossRef Gourzones C, Gelin A, Bombik I, Klibi J, Verillaud B, Guigay J, et al. Extra-cellular release and blood diffusion of BART viral micro-RNAs produced by EBV-infected nasopharyngeal carcinoma cells. Virol J. 2010;7:271.PubMedPubMedCentralCrossRef
63.
go back to reference Bosetti C, Scelo G, Chuang SC, Tonita JM, Tamaro S, Jonasson JG, et al. High constant incidence rates of second primary cancers of the head and neck: a pooled analysis of 13 cancer registries. Int J Cancer. 2011;129(1):173–9.PubMedCrossRef Bosetti C, Scelo G, Chuang SC, Tonita JM, Tamaro S, Jonasson JG, et al. High constant incidence rates of second primary cancers of the head and neck: a pooled analysis of 13 cancer registries. Int J Cancer. 2011;129(1):173–9.PubMedCrossRef
64.
go back to reference Alfieri S, Carenzo A, Platini F, Serafini MS, Perrone F, Galbiati D, et al. Tumor Biomarkers for the Prediction of Distant Metastasis in Head and Neck Squamous Cell Carcinoma. Cancers (Basel). 2020;12(4). Alfieri S, Carenzo A, Platini F, Serafini MS, Perrone F, Galbiati D, et al. Tumor Biomarkers for the Prediction of Distant Metastasis in Head and Neck Squamous Cell Carcinoma. Cancers (Basel). 2020;12(4).
65.
go back to reference Theodoraki MN, Matsumoto A, Beccard I, Hoffmann TK, Whiteside TL. CD44v3 protein-carrying tumor-derived exosomes in HNSCC patients’ plasma as potential noninvasive biomarkers of disease activity. Oncoimmunology. 2020;9(1):1747732.PubMedPubMedCentralCrossRef Theodoraki MN, Matsumoto A, Beccard I, Hoffmann TK, Whiteside TL. CD44v3 protein-carrying tumor-derived exosomes in HNSCC patients’ plasma as potential noninvasive biomarkers of disease activity. Oncoimmunology. 2020;9(1):1747732.PubMedPubMedCentralCrossRef
66.
go back to reference Ohzawa H, Kumagai Y, Yamaguchi H, Miyato H, Sakuma Y, Horie H, et al. Exosomal microRNA in peritoneal fluid as a biomarker of peritoneal metastases from gastric cancer. Ann Gastroenterol Surg. 2020;4(1):84–93.PubMedCrossRef Ohzawa H, Kumagai Y, Yamaguchi H, Miyato H, Sakuma Y, Horie H, et al. Exosomal microRNA in peritoneal fluid as a biomarker of peritoneal metastases from gastric cancer. Ann Gastroenterol Surg. 2020;4(1):84–93.PubMedCrossRef
67.
go back to reference Kobayashi E, Hwang D, Bheda-Malge A, Whitehurst CB, Kabanov AV, Kondo S, et al. Inhibition of UCH-L1 Deubiquitinating Activity with Two Forms of LDN-57444 Has Anti-Invasive Effects in Metastatic Carcinoma Cells. Int J Mol Sci. 2019;20(15). Kobayashi E, Hwang D, Bheda-Malge A, Whitehurst CB, Kabanov AV, Kondo S, et al. Inhibition of UCH-L1 Deubiquitinating Activity with Two Forms of LDN-57444 Has Anti-Invasive Effects in Metastatic Carcinoma Cells. Int J Mol Sci. 2019;20(15).
68.
go back to reference Liu Y, Cao X. Characteristics and Significance of the Pre-metastatic Niche. Cancer Cell. 2016;30(5):668–81.PubMedCrossRef Liu Y, Cao X. Characteristics and Significance of the Pre-metastatic Niche. Cancer Cell. 2016;30(5):668–81.PubMedCrossRef
69.
go back to reference Sceneay J, Smyth MJ, Moller A. The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 2013;32(3–4):449–64.PubMedCrossRef Sceneay J, Smyth MJ, Moller A. The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 2013;32(3–4):449–64.PubMedCrossRef
70.
go back to reference Sanada T, Islam A, Kaminota T, Kirino Y, Tanimoto R, Yoshimitsu H, et al. Elevated exosomal lysyl oxidase like 2 is a potential biomarker for head and neck squamous cell carcinoma. Laryngoscope. 2020;130(5):E327-E34.CrossRef Sanada T, Islam A, Kaminota T, Kirino Y, Tanimoto R, Yoshimitsu H, et al. Elevated exosomal lysyl oxidase like 2 is a potential biomarker for head and neck squamous cell carcinoma. Laryngoscope. 2020;130(5):E327-E34.CrossRef
71.
go back to reference Chen JH, Wu ATH, Bamodu OA, Yadav VK, Chao TY, Tzeng YM, et al. Ovatodiolide Suppresses Oral Cancer Malignancy by Down-Regulating Exosomal Mir-21/STAT3/beta-Catenin Cargo and Preventing Oncogenic Transformation of Normal Gingival Fibroblasts. Cancers (Basel). 2019;12(1). Chen JH, Wu ATH, Bamodu OA, Yadav VK, Chao TY, Tzeng YM, et al. Ovatodiolide Suppresses Oral Cancer Malignancy by Down-Regulating Exosomal Mir-21/STAT3/beta-Catenin Cargo and Preventing Oncogenic Transformation of Normal Gingival Fibroblasts. Cancers (Basel). 2019;12(1).
72.
go back to reference Nieto MA, Huang RY, Jackson RA, Thiery JP. Emt: 2016 Cell. 2016;166(1):21–45.PubMed Nieto MA, Huang RY, Jackson RA, Thiery JP. Emt: 2016 Cell. 2016;166(1):21–45.PubMed
73.
go back to reference Zeng P, Sun S, Li R, Xiao ZX, Chen H. HER2 Upregulates ATF4 to Promote Cell Migration via Activation of ZEB1 and Downregulation of E-Cadherin. Int J Mol Sci. 2019;20(9). Zeng P, Sun S, Li R, Xiao ZX, Chen H. HER2 Upregulates ATF4 to Promote Cell Migration via Activation of ZEB1 and Downregulation of E-Cadherin. Int J Mol Sci. 2019;20(9).
74.
go back to reference Fujiwara T, Eguchi T, Sogawa C, Ono K, Murakami J, Ibaragi S, et al. Anti-EGFR antibody cetuximab is secreted by oral squamous cell carcinoma and alters EGF-driven mesenchymal transition. Biochem Biophys Res Commun. 2018;503(3):1267–72.PubMedCrossRef Fujiwara T, Eguchi T, Sogawa C, Ono K, Murakami J, Ibaragi S, et al. Anti-EGFR antibody cetuximab is secreted by oral squamous cell carcinoma and alters EGF-driven mesenchymal transition. Biochem Biophys Res Commun. 2018;503(3):1267–72.PubMedCrossRef
75.
go back to reference Langley RR, Fidler IJ. Tumor cell-organ microenvironment interactions in the pathogenesis of cancer metastasis. Endocr Rev. 2007;28(3):297–321.PubMedCrossRef Langley RR, Fidler IJ. Tumor cell-organ microenvironment interactions in the pathogenesis of cancer metastasis. Endocr Rev. 2007;28(3):297–321.PubMedCrossRef
76.
go back to reference Huang CH, Yang WH, Chang SY, Tai SK, Tzeng CH, Kao JY, et al. Regulation of membrane-type 4 matrix metalloproteinase by SLUG contributes to hypoxia-mediated metastasis. Neoplasia. 2009;11(12):1371–82.PubMedPubMedCentralCrossRef Huang CH, Yang WH, Chang SY, Tai SK, Tzeng CH, Kao JY, et al. Regulation of membrane-type 4 matrix metalloproteinase by SLUG contributes to hypoxia-mediated metastasis. Neoplasia. 2009;11(12):1371–82.PubMedPubMedCentralCrossRef
77.
go back to reference Chan YK, Zhang H, Liu P, Tsao SW, Lung ML, Mak NK, et al. Proteomic analysis of exosomes from nasopharyngeal carcinoma cell identifies intercellular transfer of angiogenic proteins. Int J Cancer. 2015;137(8):1830–41.PubMedCrossRef Chan YK, Zhang H, Liu P, Tsao SW, Lung ML, Mak NK, et al. Proteomic analysis of exosomes from nasopharyngeal carcinoma cell identifies intercellular transfer of angiogenic proteins. Int J Cancer. 2015;137(8):1830–41.PubMedCrossRef
79.
go back to reference Chen R, Lu X, Li Z, Sun Y, He Z, Li X. Dihydroartemisinin Prevents Progression and Metastasis of Head and Neck Squamous Cell Carcinoma by Inhibiting Polarization of Macrophages in Tumor Microenvironment. Onco Targets Ther. 2020;13:3375–87.PubMedPubMedCentralCrossRef Chen R, Lu X, Li Z, Sun Y, He Z, Li X. Dihydroartemisinin Prevents Progression and Metastasis of Head and Neck Squamous Cell Carcinoma by Inhibiting Polarization of Macrophages in Tumor Microenvironment. Onco Targets Ther. 2020;13:3375–87.PubMedPubMedCentralCrossRef
80.
go back to reference Ludwig N, Yerneni SS, Razzo BM, Whiteside TL. Exosomes from HNSCC Promote Angiogenesis through Reprogramming of Endothelial Cells. Mol Cancer Res. 2018;16(11):1798–808.PubMedCrossRef Ludwig N, Yerneni SS, Razzo BM, Whiteside TL. Exosomes from HNSCC Promote Angiogenesis through Reprogramming of Endothelial Cells. Mol Cancer Res. 2018;16(11):1798–808.PubMedCrossRef
82.
go back to reference Ries J, Vairaktaris E, Agaimy A, Kintopp R, Baran C, Neukam FW, et al. miR-186, miR-3651 and miR-494: potential biomarkers for oral squamous cell carcinoma extracted from whole blood. Oncol Rep. 2014;31(3):1429–36.PubMedCrossRef Ries J, Vairaktaris E, Agaimy A, Kintopp R, Baran C, Neukam FW, et al. miR-186, miR-3651 and miR-494: potential biomarkers for oral squamous cell carcinoma extracted from whole blood. Oncol Rep. 2014;31(3):1429–36.PubMedCrossRef
83.
go back to reference Mao G, Liu Y, Fang X, Liu Y, Fang L, Lin L, et al. Tumor-derived microRNA-494 promotes angiogenesis in non-small cell lung cancer. Angiogenesis. 2015;18(3):373–82.PubMedCrossRef Mao G, Liu Y, Fang X, Liu Y, Fang L, Lin L, et al. Tumor-derived microRNA-494 promotes angiogenesis in non-small cell lung cancer. Angiogenesis. 2015;18(3):373–82.PubMedCrossRef
84.
go back to reference Wang X, Zhang X, Ren XP, Chen J, Liu H, Yang J, et al. MicroRNA-494 targeting both proapoptotic and antiapoptotic proteins protects against ischemia/reperfusion-induced cardiac injury. Circulation. 2010;122(13):1308–18.PubMedPubMedCentralCrossRef Wang X, Zhang X, Ren XP, Chen J, Liu H, Yang J, et al. MicroRNA-494 targeting both proapoptotic and antiapoptotic proteins protects against ischemia/reperfusion-induced cardiac injury. Circulation. 2010;122(13):1308–18.PubMedPubMedCentralCrossRef
85.
go back to reference Liu L, Jiang Y, Zhang H, Greenlee AR, Han Z. Overexpressed miR-494 down-regulates PTEN gene expression in cells transformed by anti-benzo(a)pyrene-trans-7,8-dihydrodiol-9,10-epoxide. Life Sci. 2010;86(5–6):192–8.PubMedCrossRef Liu L, Jiang Y, Zhang H, Greenlee AR, Han Z. Overexpressed miR-494 down-regulates PTEN gene expression in cells transformed by anti-benzo(a)pyrene-trans-7,8-dihydrodiol-9,10-epoxide. Life Sci. 2010;86(5–6):192–8.PubMedCrossRef
86.
go back to reference Ma Y, Hu X, Shang C, Zhong M, Guo Y. Silencing of long non-coding RNA CCAT2 depressed malignancy of oral squamous cell carcinoma via Wnt/beta-catenin pathway. Tumour Biol. 2017;39(7):1010428317717670.PubMedCrossRef Ma Y, Hu X, Shang C, Zhong M, Guo Y. Silencing of long non-coding RNA CCAT2 depressed malignancy of oral squamous cell carcinoma via Wnt/beta-catenin pathway. Tumour Biol. 2017;39(7):1010428317717670.PubMedCrossRef
87.
go back to reference Lang HL, Hu GW, Zhang B, Kuang W, Chen Y, Wu L, et al. Glioma cells enhance angiogenesis and inhibit endothelial cell apoptosis through the release of exosomes that contain long non-coding RNA CCAT2. Oncol Rep. 2017;38(2):785–98.PubMedPubMedCentralCrossRef Lang HL, Hu GW, Zhang B, Kuang W, Chen Y, Wu L, et al. Glioma cells enhance angiogenesis and inhibit endothelial cell apoptosis through the release of exosomes that contain long non-coding RNA CCAT2. Oncol Rep. 2017;38(2):785–98.PubMedPubMedCentralCrossRef
88.
go back to reference Sato S, Vasaikar S, Eskaros A, Kim Y, Lewis JS, Zhang B, et al. EPHB2 carried on small extracellular vesicles induces tumor angiogenesis via activation of ephrin reverse signaling. JCI Insight. 2019;4(23). Sato S, Vasaikar S, Eskaros A, Kim Y, Lewis JS, Zhang B, et al. EPHB2 carried on small extracellular vesicles induces tumor angiogenesis via activation of ephrin reverse signaling. JCI Insight. 2019;4(23).
89.
go back to reference Lu J, Liu QH, Wang F, Tan JJ, Deng YQ, Peng XH, et al. Exosomal miR-9 inhibits angiogenesis by targeting MDK and regulating PDK/AKT pathway in nasopharyngeal carcinoma. J Exp Clin Cancer Res. 2018;37(1):147.PubMedPubMedCentralCrossRef Lu J, Liu QH, Wang F, Tan JJ, Deng YQ, Peng XH, et al. Exosomal miR-9 inhibits angiogenesis by targeting MDK and regulating PDK/AKT pathway in nasopharyngeal carcinoma. J Exp Clin Cancer Res. 2018;37(1):147.PubMedPubMedCentralCrossRef
90.
91.
92.
go back to reference Li YY, Tao YW, Gao S, Li P, Zheng JM, Zhang SE, et al. Cancer-associated fibroblasts contribute to oral cancer cells proliferation and metastasis via exosome-mediated paracrine miR-34a-5p. EBioMedicine. 2018;36:209–20.PubMedPubMedCentralCrossRef Li YY, Tao YW, Gao S, Li P, Zheng JM, Zhang SE, et al. Cancer-associated fibroblasts contribute to oral cancer cells proliferation and metastasis via exosome-mediated paracrine miR-34a-5p. EBioMedicine. 2018;36:209–20.PubMedPubMedCentralCrossRef
93.
go back to reference Wan FZ, Chen KH, Sun YC, Chen XC, Liang RB, Chen L, et al. Exosomes overexpressing miR-34c inhibit malignant behavior and reverse the radioresistance of nasopharyngeal carcinoma. J Transl Med. 2020;18(1):12.PubMedPubMedCentralCrossRef Wan FZ, Chen KH, Sun YC, Chen XC, Liang RB, Chen L, et al. Exosomes overexpressing miR-34c inhibit malignant behavior and reverse the radioresistance of nasopharyngeal carcinoma. J Transl Med. 2020;18(1):12.PubMedPubMedCentralCrossRef
94.
go back to reference Gu J, Wang G, Liu H, Xiong C. SATB2 targeted by methylated miR-34c-5p suppresses proliferation and metastasis attenuating the epithelial-mesenchymal transition in colorectal cancer. Cell Prolif. 2018;51(4):e12455.PubMedPubMedCentralCrossRef Gu J, Wang G, Liu H, Xiong C. SATB2 targeted by methylated miR-34c-5p suppresses proliferation and metastasis attenuating the epithelial-mesenchymal transition in colorectal cancer. Cell Prolif. 2018;51(4):e12455.PubMedPubMedCentralCrossRef
95.
go back to reference Xiao S, Li Y, Pan Q, Ye M, He S, Tian Q, et al. MiR-34c/SOX9 axis regulates the chemoresistance of ovarian cancer cell to cisplatin-based chemotherapy. J Cell Biochem. 2019;120(3):2940–53.PubMedCrossRef Xiao S, Li Y, Pan Q, Ye M, He S, Tian Q, et al. MiR-34c/SOX9 axis regulates the chemoresistance of ovarian cancer cell to cisplatin-based chemotherapy. J Cell Biochem. 2019;120(3):2940–53.PubMedCrossRef
96.
go back to reference Russo V, Paciocco A, Affinito A, Roscigno G, Fiore D, Palma F, et al. Aptamer-miR-34c Conjugate Affects Cell Proliferation of Non-Small-Cell Lung Cancer Cells. Mol Ther Nucleic Acids. 2018;13:334–46.PubMedPubMedCentralCrossRef Russo V, Paciocco A, Affinito A, Roscigno G, Fiore D, Palma F, et al. Aptamer-miR-34c Conjugate Affects Cell Proliferation of Non-Small-Cell Lung Cancer Cells. Mol Ther Nucleic Acids. 2018;13:334–46.PubMedPubMedCentralCrossRef
97.
go back to reference Wu J, Li WZ, Huang ML, Wei HL, Wang T, Fan J, et al. Regulation of cancerous progression and epithelial-mesenchymal transition by miR-34c-3p via modulation of MAP3K2 signaling in triple-negative breast cancer cells. Biochem Biophys Res Commun. 2017;483(1):10–6.PubMedCrossRef Wu J, Li WZ, Huang ML, Wei HL, Wang T, Fan J, et al. Regulation of cancerous progression and epithelial-mesenchymal transition by miR-34c-3p via modulation of MAP3K2 signaling in triple-negative breast cancer cells. Biochem Biophys Res Commun. 2017;483(1):10–6.PubMedCrossRef
98.
go back to reference Wang JX, Yang Y, Li K. Long noncoding RNA DANCR aggravates retinoblastoma through miR-34c and miR-613 by targeting MMP-9. J Cell Physiol. 2018;233(10):6986–95.PubMedCrossRef Wang JX, Yang Y, Li K. Long noncoding RNA DANCR aggravates retinoblastoma through miR-34c and miR-613 by targeting MMP-9. J Cell Physiol. 2018;233(10):6986–95.PubMedCrossRef
99.
go back to reference Peng D, Wang H, Li L, Ma X, Chen Y, Zhou H, et al. miR-34c-5p promotes eradication of acute myeloid leukemia stem cells by inducing senescence through selective RAB27B targeting to inhibit exosome shedding. Leukemia. 2018;32(5):1180–8.PubMedCrossRef Peng D, Wang H, Li L, Ma X, Chen Y, Zhou H, et al. miR-34c-5p promotes eradication of acute myeloid leukemia stem cells by inducing senescence through selective RAB27B targeting to inhibit exosome shedding. Leukemia. 2018;32(5):1180–8.PubMedCrossRef
100.
go back to reference Sun LP, Xu K, Cui J, Yuan DY, Zou B, Li J, et al. Cancerassociated fibroblastderived exosomal miR3825p promotes the migration and invasion of oral squamous cell carcinoma. Oncol Rep. 2019;42(4):1319–28.PubMedPubMedCentral Sun LP, Xu K, Cui J, Yuan DY, Zou B, Li J, et al. Cancerassociated fibroblastderived exosomal miR3825p promotes the migration and invasion of oral squamous cell carcinoma. Oncol Rep. 2019;42(4):1319–28.PubMedPubMedCentral
101.
go back to reference Xie C, Du LY, Guo F, Li X, Cheng B. Exosomes derived from microRNA-101-3p-overexpressing human bone marrow mesenchymal stem cells suppress oral cancer cell proliferation, invasion, and migration. Mol Cell Biochem. 2019;458(1–2):11–26.PubMedCrossRef Xie C, Du LY, Guo F, Li X, Cheng B. Exosomes derived from microRNA-101-3p-overexpressing human bone marrow mesenchymal stem cells suppress oral cancer cell proliferation, invasion, and migration. Mol Cell Biochem. 2019;458(1–2):11–26.PubMedCrossRef
102.
go back to reference Wang L, Yin P, Wang J, Wang Y, Sun Z, Zhou Y, et al. Delivery of mesenchymal stem cells-derived extracellular vesicles with enriched miR-185 inhibits progression of OPMD. Artif Cells Nanomed Biotechnol. 2019;47(1):2481–91.PubMedCrossRef Wang L, Yin P, Wang J, Wang Y, Sun Z, Zhou Y, et al. Delivery of mesenchymal stem cells-derived extracellular vesicles with enriched miR-185 inhibits progression of OPMD. Artif Cells Nanomed Biotechnol. 2019;47(1):2481–91.PubMedCrossRef
103.
go back to reference Principe S, Mejia-Guerrero S, Ignatchenko V, Sinha A, Ignatchenko A, Shi W, et al. Proteomic Analysis of Cancer-Associated Fibroblasts Reveals a Paracrine Role for MFAP5 in Human Oral Tongue Squamous Cell Carcinoma. J Proteome Res. 2018;17(6):2045–59.PubMedCrossRef Principe S, Mejia-Guerrero S, Ignatchenko V, Sinha A, Ignatchenko A, Shi W, et al. Proteomic Analysis of Cancer-Associated Fibroblasts Reveals a Paracrine Role for MFAP5 in Human Oral Tongue Squamous Cell Carcinoma. J Proteome Res. 2018;17(6):2045–59.PubMedCrossRef
105.
go back to reference Fong MY, Zhou W, Liu L, Alontaga AY, Chandra M, Ashby J, et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat Cell Biol. 2015;17(2):183–94.PubMedPubMedCentralCrossRef Fong MY, Zhou W, Liu L, Alontaga AY, Chandra M, Ashby J, et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat Cell Biol. 2015;17(2):183–94.PubMedPubMedCentralCrossRef
107.
109.
110.
111.
go back to reference Ludwig S, Rotter N, Theodoraki MN, Jablonska J, Lammert A, Lang S. [Exosomes as immune regulators in head and neck cancer]. HNO. 2020;68(10):719–25.PubMedCrossRef Ludwig S, Rotter N, Theodoraki MN, Jablonska J, Lammert A, Lang S. [Exosomes as immune regulators in head and neck cancer]. HNO. 2020;68(10):719–25.PubMedCrossRef
112.
go back to reference Bellmunt AM, Lopez-Puerto L, Lorente J, Closa D. Involvement of extracellular vesicles in the macrophage-tumor cell communication in head and neck squamous cell carcinoma. PLoS One. 2019;14(11):e0224710.PubMedPubMedCentralCrossRef Bellmunt AM, Lopez-Puerto L, Lorente J, Closa D. Involvement of extracellular vesicles in the macrophage-tumor cell communication in head and neck squamous cell carcinoma. PLoS One. 2019;14(11):e0224710.PubMedPubMedCentralCrossRef
113.
go back to reference Hsieh CH, Tai SK, Yang MH. Snail-overexpressing Cancer Cells Promote M2-Like Polarization of Tumor-Associated Macrophages by Delivering MiR-21-Abundant Exosomes. Neoplasia. 2018;20(8):775–88.PubMedPubMedCentralCrossRef Hsieh CH, Tai SK, Yang MH. Snail-overexpressing Cancer Cells Promote M2-Like Polarization of Tumor-Associated Macrophages by Delivering MiR-21-Abundant Exosomes. Neoplasia. 2018;20(8):775–88.PubMedPubMedCentralCrossRef
114.
go back to reference Xiao M, Zhang J, Chen W, Chen W. M1-like tumor-associated macrophages activated by exosome-transferred THBS1 promote malignant migration in oral squamous cell carcinoma. J Exp Clin Cancer Res. 2018;37(1):143.PubMedPubMedCentralCrossRef Xiao M, Zhang J, Chen W, Chen W. M1-like tumor-associated macrophages activated by exosome-transferred THBS1 promote malignant migration in oral squamous cell carcinoma. J Exp Clin Cancer Res. 2018;37(1):143.PubMedPubMedCentralCrossRef
115.
go back to reference Tong F, Mao X, Zhang S, Xie H, Yan B, Wang B, et al. HPV + HNSCC-derived exosomal miR-9 induces macrophage M1 polarization and increases tumor radiosensitivity. Cancer Lett. 2020;478:34–44.PubMedCrossRef Tong F, Mao X, Zhang S, Xie H, Yan B, Wang B, et al. HPV + HNSCC-derived exosomal miR-9 induces macrophage M1 polarization and increases tumor radiosensitivity. Cancer Lett. 2020;478:34–44.PubMedCrossRef
116.
go back to reference Zheng Y, Li Y, Tang B, Zhao Q, Wang D, Liu Y, et al. IL-6-induced CD39 expression on tumor-infiltrating NK cells predicts poor prognosis in esophageal squamous cell carcinoma. Cancer Immunol Immunother. 2020;69(11):2371–80.PubMedCrossRef Zheng Y, Li Y, Tang B, Zhao Q, Wang D, Liu Y, et al. IL-6-induced CD39 expression on tumor-infiltrating NK cells predicts poor prognosis in esophageal squamous cell carcinoma. Cancer Immunol Immunother. 2020;69(11):2371–80.PubMedCrossRef
117.
go back to reference Wang Y, Qin X, Zhu X, Chen W, Zhang J, Chen W. Oral cancer-derived exosomal NAP1 enhances cytotoxicity of natural killer cells via the IRF-3 pathway. Oral Oncol. 2018;76:34–41.PubMedCrossRef Wang Y, Qin X, Zhu X, Chen W, Zhang J, Chen W. Oral cancer-derived exosomal NAP1 enhances cytotoxicity of natural killer cells via the IRF-3 pathway. Oral Oncol. 2018;76:34–41.PubMedCrossRef
118.
go back to reference Lundholm M, Schroder M, Nagaeva O, Baranov V, Widmark A, Mincheva-Nilsson L, et al. Prostate tumor-derived exosomes down-regulate NKG2D expression on natural killer cells and CD8 + T cells: mechanism of immune evasion. PLoS One. 2014;9(9):e108925.PubMedPubMedCentralCrossRef Lundholm M, Schroder M, Nagaeva O, Baranov V, Widmark A, Mincheva-Nilsson L, et al. Prostate tumor-derived exosomes down-regulate NKG2D expression on natural killer cells and CD8 + T cells: mechanism of immune evasion. PLoS One. 2014;9(9):e108925.PubMedPubMedCentralCrossRef
119.
go back to reference Hong CS, Muller L, Boyiadzis M, Whiteside TL. Isolation and characterization of CD34 + blast-derived exosomes in acute myeloid leukemia. PLoS One. 2014;9(8):e103310.PubMedPubMedCentralCrossRef Hong CS, Muller L, Boyiadzis M, Whiteside TL. Isolation and characterization of CD34 + blast-derived exosomes in acute myeloid leukemia. PLoS One. 2014;9(8):e103310.PubMedPubMedCentralCrossRef
120.
go back to reference Pei D. Leukolysin/MMP25/MT6-MMP: a novel matrix metalloproteinase specifically expressed in the leukocyte lineage. Cell Res. 1999;9(4):291–303.PubMedCrossRef Pei D. Leukolysin/MMP25/MT6-MMP: a novel matrix metalloproteinase specifically expressed in the leukocyte lineage. Cell Res. 1999;9(4):291–303.PubMedCrossRef
121.
go back to reference Ludwig S, Floros T, Theodoraki MN, Hong CS, Jackson EK, Lang S, et al. Suppression of Lymphocyte Functions by Plasma Exosomes Correlates with Disease Activity in Patients with Head and Neck Cancer. Clin Cancer Res. 2017;23(16):4843–54.PubMedPubMedCentralCrossRef Ludwig S, Floros T, Theodoraki MN, Hong CS, Jackson EK, Lang S, et al. Suppression of Lymphocyte Functions by Plasma Exosomes Correlates with Disease Activity in Patients with Head and Neck Cancer. Clin Cancer Res. 2017;23(16):4843–54.PubMedPubMedCentralCrossRef
122.
go back to reference Tian X, Shen H, Li Z, Wang T, Wang S. Tumor-derived exosomes, myeloid-derived suppressor cells, and tumor microenvironment. J Hematol Oncol. 2019;12(1):84.PubMedPubMedCentralCrossRef Tian X, Shen H, Li Z, Wang T, Wang S. Tumor-derived exosomes, myeloid-derived suppressor cells, and tumor microenvironment. J Hematol Oncol. 2019;12(1):84.PubMedPubMedCentralCrossRef
123.
go back to reference Obermajer N, Muthuswamy R, Odunsi K, Edwards RP, Kalinski P. PGE(2)-induced CXCL12 production and CXCR4 expression controls the accumulation of human MDSCs in ovarian cancer environment. Cancer Res. 2011;71(24):7463–70.PubMedPubMedCentralCrossRef Obermajer N, Muthuswamy R, Odunsi K, Edwards RP, Kalinski P. PGE(2)-induced CXCL12 production and CXCR4 expression controls the accumulation of human MDSCs in ovarian cancer environment. Cancer Res. 2011;71(24):7463–70.PubMedPubMedCentralCrossRef
124.
go back to reference Li L, Cao B, Liang X, Lu S, Luo H, Wang Z, et al. Microenvironmental oxygen pressure orchestrates an anti- and pro-tumoral gammadelta T cell equilibrium via tumor-derived exosomes. Oncogene. 2019;38(15):2830–43.PubMedCrossRef Li L, Cao B, Liang X, Lu S, Luo H, Wang Z, et al. Microenvironmental oxygen pressure orchestrates an anti- and pro-tumoral gammadelta T cell equilibrium via tumor-derived exosomes. Oncogene. 2019;38(15):2830–43.PubMedCrossRef
125.
go back to reference Zhu S, Yang N, Wu J, Wang X, Wang W, Liu YJ, et al. Tumor microenvironment-related dendritic cell deficiency: a target to enhance tumor immunotherapy. Pharmacol Res. 2020;159:104980.PubMedCrossRef Zhu S, Yang N, Wu J, Wang X, Wang W, Liu YJ, et al. Tumor microenvironment-related dendritic cell deficiency: a target to enhance tumor immunotherapy. Pharmacol Res. 2020;159:104980.PubMedCrossRef
126.
go back to reference Zhao Z, Zhang H, Zeng Q, Wang P, Zhang G, Ji J, et al. Exosomes from 5-aminolevulinic acid photodynamic therapy-treated squamous carcinoma cells promote dendritic cell maturation. Photodiagnosis Photodyn Ther. 2020;30:101746.PubMedCrossRef Zhao Z, Zhang H, Zeng Q, Wang P, Zhang G, Ji J, et al. Exosomes from 5-aminolevulinic acid photodynamic therapy-treated squamous carcinoma cells promote dendritic cell maturation. Photodiagnosis Photodyn Ther. 2020;30:101746.PubMedCrossRef
127.
go back to reference Asadirad A, Hashemi SM, Baghaei K, Ghanbarian H, Mortaz E, Zali MR, et al. Phenotypical and functional evaluation of dendritic cells after exosomal delivery of miRNA-155. Life Sci. 2019;219:152–62.PubMedCrossRef Asadirad A, Hashemi SM, Baghaei K, Ghanbarian H, Mortaz E, Zali MR, et al. Phenotypical and functional evaluation of dendritic cells after exosomal delivery of miRNA-155. Life Sci. 2019;219:152–62.PubMedCrossRef
128.
go back to reference Taghikhani A, Hassan ZM, Ebrahimi M, Moazzeni SM. microRNA modified tumor-derived exosomes as novel tools for maturation of dendritic cells. J Cell Physiol. 2019;234(6):9417–27.PubMedCrossRef Taghikhani A, Hassan ZM, Ebrahimi M, Moazzeni SM. microRNA modified tumor-derived exosomes as novel tools for maturation of dendritic cells. J Cell Physiol. 2019;234(6):9417–27.PubMedCrossRef
129.
go back to reference Zahran AM, Youssef MAM, Shafik EA, Zahran ZAM, El-Badawy O, Abo Elgheet AM, et al. Downregulation of B regulatory cells and upregulation of T helper 1 cells in children with Gaucher disease undergoing enzyme replacement therapy. Immunol Res. 2020;68(2):73–80.PubMedCrossRef Zahran AM, Youssef MAM, Shafik EA, Zahran ZAM, El-Badawy O, Abo Elgheet AM, et al. Downregulation of B regulatory cells and upregulation of T helper 1 cells in children with Gaucher disease undergoing enzyme replacement therapy. Immunol Res. 2020;68(2):73–80.PubMedCrossRef
130.
go back to reference Correction. Cell death induced by cytotoxic CD8 + T cells is immunogenic and primes caspase-3-dependent spread immunity against endogenous tumor antigens. J Immunother Cancer. 2020;8(1). Correction. Cell death induced by cytotoxic CD8 + T cells is immunogenic and primes caspase-3-dependent spread immunity against endogenous tumor antigens. J Immunother Cancer. 2020;8(1).
131.
go back to reference Kim JW, Wieckowski E, Taylor DD, Reichert TE, Watkins S, Whiteside TL. Fas ligand-positive membranous vesicles isolated from sera of patients with oral cancer induce apoptosis of activated T lymphocytes. Clin Cancer Res. 2005;11(3):1010–20.PubMedCrossRef Kim JW, Wieckowski E, Taylor DD, Reichert TE, Watkins S, Whiteside TL. Fas ligand-positive membranous vesicles isolated from sera of patients with oral cancer induce apoptosis of activated T lymphocytes. Clin Cancer Res. 2005;11(3):1010–20.PubMedCrossRef
132.
go back to reference Muller L, Mitsuhashi M, Simms P, Gooding WE, Whiteside TL. Tumor-derived exosomes regulate expression of immune function-related genes in human T cell subsets. Sci Rep. 2016;6:20254.PubMedPubMedCentralCrossRef Muller L, Mitsuhashi M, Simms P, Gooding WE, Whiteside TL. Tumor-derived exosomes regulate expression of immune function-related genes in human T cell subsets. Sci Rep. 2016;6:20254.PubMedPubMedCentralCrossRef
133.
go back to reference Theodoraki MN, Yerneni SS, Hoffmann TK, Gooding WE, Whiteside TL. Clinical Significance of PD-L1(+) Exosomes in Plasma of Head and Neck Cancer Patients. Clin Cancer Res. 2018;24(4):896–905.PubMedCrossRef Theodoraki MN, Yerneni SS, Hoffmann TK, Gooding WE, Whiteside TL. Clinical Significance of PD-L1(+) Exosomes in Plasma of Head and Neck Cancer Patients. Clin Cancer Res. 2018;24(4):896–905.PubMedCrossRef
134.
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
135.
go back to reference Wieckowski EU, Visus C, Szajnik M, Szczepanski MJ, Storkus WJ, Whiteside TL. Tumor-derived microvesicles promote regulatory T cell expansion and induce apoptosis in tumor-reactive activated CD8 + T lymphocytes. J Immunol. 2009;183(6):3720–30.PubMedCrossRef Wieckowski EU, Visus C, Szajnik M, Szczepanski MJ, Storkus WJ, Whiteside TL. Tumor-derived microvesicles promote regulatory T cell expansion and induce apoptosis in tumor-reactive activated CD8 + T lymphocytes. J Immunol. 2009;183(6):3720–30.PubMedCrossRef
136.
go back to reference Horton JD, Knochelmann HM, Day TA, Paulos CM, Neskey DM. Immune Evasion by Head and Neck Cancer: Foundations for Combination Therapy. Trends Cancer. 2019;5(4):208–32.PubMedPubMedCentralCrossRef Horton JD, Knochelmann HM, Day TA, Paulos CM, Neskey DM. Immune Evasion by Head and Neck Cancer: Foundations for Combination Therapy. Trends Cancer. 2019;5(4):208–32.PubMedPubMedCentralCrossRef
137.
go back to reference Whiteside TL. Immune modulation of T-cell and NK (natural killer) cell activities by TEXs (tumour-derived exosomes). Biochem Soc Trans. 2013;41(1):245–51.PubMedPubMedCentralCrossRef Whiteside TL. Immune modulation of T-cell and NK (natural killer) cell activities by TEXs (tumour-derived exosomes). Biochem Soc Trans. 2013;41(1):245–51.PubMedPubMedCentralCrossRef
138.
go back to reference Hong CS, Funk S, Muller L, Boyiadzis M, Whiteside TL. Isolation of biologically active and morphologically intact exosomes from plasma of patients with cancer. J Extracell Vesicles. 2016;5:29289.PubMedCrossRef Hong CS, Funk S, Muller L, Boyiadzis M, Whiteside TL. Isolation of biologically active and morphologically intact exosomes from plasma of patients with cancer. J Extracell Vesicles. 2016;5:29289.PubMedCrossRef
139.
140.
go back to reference Ye SB, Zhang H, Cai TT, Liu YN, Ni JJ, He J, et al. Exosomal miR-24-3p impedes T-cell function by targeting FGF11 and serves as a potential prognostic biomarker for nasopharyngeal carcinoma. J Pathol. 2016;240(3):329–40.PubMedCrossRef Ye SB, Zhang H, Cai TT, Liu YN, Ni JJ, He J, et al. Exosomal miR-24-3p impedes T-cell function by targeting FGF11 and serves as a potential prognostic biomarker for nasopharyngeal carcinoma. J Pathol. 2016;240(3):329–40.PubMedCrossRef
141.
go back to reference Mrizak D, Martin N, Barjon C, Jimenez-Pailhes AS, Mustapha R, Niki T, et al. Effect of nasopharyngeal carcinoma-derived exosomes on human regulatory T cells. J Natl Cancer Inst. 2015;107(1):363.PubMedCrossRef Mrizak D, Martin N, Barjon C, Jimenez-Pailhes AS, Mustapha R, Niki T, et al. Effect of nasopharyngeal carcinoma-derived exosomes on human regulatory T cells. J Natl Cancer Inst. 2015;107(1):363.PubMedCrossRef
142.
go back to reference Chaturvedi AK, Engels EA, Pfeiffer RM, Hernandez BY, Xiao W, Kim E, et al. Human papillomavirus and rising oropharyngeal cancer incidence in the United States. J Clin Oncol. 2011;29(32):4294–301.PubMedPubMedCentralCrossRef Chaturvedi AK, Engels EA, Pfeiffer RM, Hernandez BY, Xiao W, Kim E, et al. Human papillomavirus and rising oropharyngeal cancer incidence in the United States. J Clin Oncol. 2011;29(32):4294–301.PubMedPubMedCentralCrossRef
143.
go back to reference Nasman A, Attner P, Hammarstedt L, Du J, Eriksson M, Giraud G, et al. Incidence of human papillomavirus (HPV) positive tonsillar carcinoma in Stockholm, Sweden: an epidemic of viral-induced carcinoma? Int J Cancer. 2009;125(2):362–6.PubMedCrossRef Nasman A, Attner P, Hammarstedt L, Du J, Eriksson M, Giraud G, et al. Incidence of human papillomavirus (HPV) positive tonsillar carcinoma in Stockholm, Sweden: an epidemic of viral-induced carcinoma? Int J Cancer. 2009;125(2):362–6.PubMedCrossRef
144.
go back to reference Nulton TJ, Olex AL, Dozmorov M, Morgan IM, Windle B. Analysis of The Cancer Genome Atlas sequencing data reveals novel properties of the human papillomavirus 16 genome in head and neck squamous cell carcinoma. Oncotarget. 2017;8(11):17684–99.PubMedPubMedCentralCrossRef Nulton TJ, Olex AL, Dozmorov M, Morgan IM, Windle B. Analysis of The Cancer Genome Atlas sequencing data reveals novel properties of the human papillomavirus 16 genome in head and neck squamous cell carcinoma. Oncotarget. 2017;8(11):17684–99.PubMedPubMedCentralCrossRef
145.
go back to reference Shen KY, Liu HY, Yan WL, Wu CC, Lee MH, Leng CH, et al. Liposomal TLR9 Agonist Combined with TLR2 Agonist-Fused Antigen Can Modulate Tumor Microenvironment through Dendritic Cells. Cancers (Basel). 2020;12(4). Shen KY, Liu HY, Yan WL, Wu CC, Lee MH, Leng CH, et al. Liposomal TLR9 Agonist Combined with TLR2 Agonist-Fused Antigen Can Modulate Tumor Microenvironment through Dendritic Cells. Cancers (Basel). 2020;12(4).
146.
go back to reference Welters MJP, Ma W, Santegoets S, Goedemans R, Ehsan I, Jordanova ES, et al. Intratumoral HPV16-Specific T Cells Constitute a Type I-Oriented Tumor Microenvironment to Improve Survival in HPV16-Driven Oropharyngeal Cancer. Clin Cancer Res. 2018;24(3):634–47.PubMedCrossRef Welters MJP, Ma W, Santegoets S, Goedemans R, Ehsan I, Jordanova ES, et al. Intratumoral HPV16-Specific T Cells Constitute a Type I-Oriented Tumor Microenvironment to Improve Survival in HPV16-Driven Oropharyngeal Cancer. Clin Cancer Res. 2018;24(3):634–47.PubMedCrossRef
147.
go back to reference Peacock B, Rigby A, Bradford J, Pink R, Hunter K, Lambert D, et al. Extracellular vesicle microRNA cargo is correlated with HPV status in oropharyngeal carcinoma. J Oral Pathol Med. 2018;47(10):954–63.PubMedCrossRef Peacock B, Rigby A, Bradford J, Pink R, Hunter K, Lambert D, et al. Extracellular vesicle microRNA cargo is correlated with HPV status in oropharyngeal carcinoma. J Oral Pathol Med. 2018;47(10):954–63.PubMedCrossRef
148.
go back to reference Khuu C, Jevnaker AM, Bryne M, Osmundsen H. An investigation into anti-proliferative effects of microRNAs encoded by the miR-106a-363 cluster on human carcinoma cells and keratinocytes using microarray profiling of miRNA transcriptomes. Front Genet. 2014;5:246.PubMedPubMedCentralCrossRef Khuu C, Jevnaker AM, Bryne M, Osmundsen H. An investigation into anti-proliferative effects of microRNAs encoded by the miR-106a-363 cluster on human carcinoma cells and keratinocytes using microarray profiling of miRNA transcriptomes. Front Genet. 2014;5:246.PubMedPubMedCentralCrossRef
149.
150.
go back to reference Gosepath EM, Eckstein N, Hamacher A, Servan K, von Jonquieres G, Lage H, et al. Acquired cisplatin resistance in the head-neck cancer cell line Cal27 is associated with decreased DKK1 expression and can partially be reversed by overexpression of DKK1. Int J Cancer. 2008;123(9):2013–9.PubMedCrossRef Gosepath EM, Eckstein N, Hamacher A, Servan K, von Jonquieres G, Lage H, et al. Acquired cisplatin resistance in the head-neck cancer cell line Cal27 is associated with decreased DKK1 expression and can partially be reversed by overexpression of DKK1. Int J Cancer. 2008;123(9):2013–9.PubMedCrossRef
151.
go back to reference Khoo XH, Paterson IC, Goh BH, Lee WL. Cisplatin-Resistance in Oral Squamous Cell Carcinoma: Regulation by Tumor Cell-Derived Extracellular Vesicles. Cancers (Basel). 2019;11(8). Khoo XH, Paterson IC, Goh BH, Lee WL. Cisplatin-Resistance in Oral Squamous Cell Carcinoma: Regulation by Tumor Cell-Derived Extracellular Vesicles. Cancers (Basel). 2019;11(8).
152.
go back to reference Qin X, Guo H, Wang X, Zhu X, Yan M, Wang X, et al. Exosomal miR-196a derived from cancer-associated fibroblasts confers cisplatin resistance in head and neck cancer through targeting CDKN1B and ING5. Genome Biol. 2019;20(1):12.PubMedPubMedCentralCrossRef Qin X, Guo H, Wang X, Zhu X, Yan M, Wang X, et al. Exosomal miR-196a derived from cancer-associated fibroblasts confers cisplatin resistance in head and neck cancer through targeting CDKN1B and ING5. Genome Biol. 2019;20(1):12.PubMedPubMedCentralCrossRef
153.
go back to reference Liu T, Chen G, Sun D, Lei M, Li Y, Zhou C, et al. Exosomes containing miR-21 transfer the characteristic of cisplatin resistance by targeting PTEN and PDCD4 in oral squamous cell carcinoma. Acta Biochim Biophys Sin (Shanghai). 2017;49(9):808–16.CrossRef Liu T, Chen G, Sun D, Lei M, Li Y, Zhou C, et al. Exosomes containing miR-21 transfer the characteristic of cisplatin resistance by targeting PTEN and PDCD4 in oral squamous cell carcinoma. Acta Biochim Biophys Sin (Shanghai). 2017;49(9):808–16.CrossRef
154.
go back to reference Vermorken JB, Mesia R, Rivera F, Remenar E, Kawecki A, Rottey S, et al. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N Engl J Med. 2008;359(11):1116–27.PubMedCrossRef Vermorken JB, Mesia R, Rivera F, Remenar E, Kawecki A, Rottey S, et al. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N Engl J Med. 2008;359(11):1116–27.PubMedCrossRef
155.
go back to reference Roberts PJ, Der CJ. Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 2007;26(22):3291–310.PubMedCrossRef Roberts PJ, Der CJ. Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 2007;26(22):3291–310.PubMedCrossRef
156.
go back to reference Fujiwara T, Eguchi T, Sogawa C, Ono K, Murakami J, Ibaragi S, et al. Carcinogenic epithelial-mesenchymal transition initiated by oral cancer exosomes is inhibited by anti-EGFR antibody cetuximab. Oral Oncol. 2018;86:251–7.PubMedCrossRef Fujiwara T, Eguchi T, Sogawa C, Ono K, Murakami J, Ibaragi S, et al. Carcinogenic epithelial-mesenchymal transition initiated by oral cancer exosomes is inhibited by anti-EGFR antibody cetuximab. Oral Oncol. 2018;86:251–7.PubMedCrossRef
157.
go back to reference Zheng Y, Song A, Zhou Y, Zhong Y, Zhang W, Wang C, et al. Identification of extracellular vesicles-transported miRNAs in Erlotinib-resistant head and neck squamous cell carcinoma. J Cell Commun Signal. 2020;14(4):389–402.PubMedPubMedCentralCrossRef Zheng Y, Song A, Zhou Y, Zhong Y, Zhang W, Wang C, et al. Identification of extracellular vesicles-transported miRNAs in Erlotinib-resistant head and neck squamous cell carcinoma. J Cell Commun Signal. 2020;14(4):389–402.PubMedPubMedCentralCrossRef
159.
go back to reference Li W, Guo F, Wang P, Hong S, Zhang C. miR-221/222 confers radioresistance in glioblastoma cells through activating Akt independent of PTEN status. Curr Mol Med. 2014;14(1):185–95.PubMedCrossRef Li W, Guo F, Wang P, Hong S, Zhang C. miR-221/222 confers radioresistance in glioblastoma cells through activating Akt independent of PTEN status. Curr Mol Med. 2014;14(1):185–95.PubMedCrossRef
160.
go back to reference Donneys A, Nelson NS, Page EE, Deshpande SS, Felice PA, Tchanque-Fossuo CN, et al. Targeting angiogenesis as a therapeutic means to reinforce osteocyte survival and prevent nonunions in the aftermath of radiotherapy. Head Neck. 2015;37(9):1261–7.PubMedCrossRef Donneys A, Nelson NS, Page EE, Deshpande SS, Felice PA, Tchanque-Fossuo CN, et al. Targeting angiogenesis as a therapeutic means to reinforce osteocyte survival and prevent nonunions in the aftermath of radiotherapy. Head Neck. 2015;37(9):1261–7.PubMedCrossRef
161.
go back to reference Bow H, Hwang LS, Schildhaus N, Xing J, Murray L, Salditch Q, et al. Local delivery of angiogenesis-inhibitor minocycline combined with radiotherapy and oral temozolomide chemotherapy in 9L glioma. J Neurosurg. 2014;120(3):662–9.PubMedCrossRef Bow H, Hwang LS, Schildhaus N, Xing J, Murray L, Salditch Q, et al. Local delivery of angiogenesis-inhibitor minocycline combined with radiotherapy and oral temozolomide chemotherapy in 9L glioma. J Neurosurg. 2014;120(3):662–9.PubMedCrossRef
162.
go back to reference Zheng Y, Liu L, Chen C, Ming P, Huang Q, Li C, et al. The extracellular vesicles secreted by lung cancer cells in radiation therapy promote endothelial cell angiogenesis by transferring miR-23a. PeerJ. 2017;5:e3627.PubMedPubMedCentralCrossRef Zheng Y, Liu L, Chen C, Ming P, Huang Q, Li C, et al. The extracellular vesicles secreted by lung cancer cells in radiation therapy promote endothelial cell angiogenesis by transferring miR-23a. PeerJ. 2017;5:e3627.PubMedPubMedCentralCrossRef
163.
go back to reference Bussink J, van der Kogel AJ, Kaanders JH. Activation of the PI3-K/AKT pathway and implications for radioresistance mechanisms in head and neck cancer. Lancet Oncol. 2008;9(3):288–96.PubMedCrossRef Bussink J, van der Kogel AJ, Kaanders JH. Activation of the PI3-K/AKT pathway and implications for radioresistance mechanisms in head and neck cancer. Lancet Oncol. 2008;9(3):288–96.PubMedCrossRef
164.
go back to reference Pickhard AC, Margraf J, Knopf A, Stark T, Piontek G, Beck C, et al. Inhibition of radiation induced migration of human head and neck squamous cell carcinoma cells by blocking of EGF receptor pathways. BMC Cancer. 2011;11:388.PubMedPubMedCentralCrossRef Pickhard AC, Margraf J, Knopf A, Stark T, Piontek G, Beck C, et al. Inhibition of radiation induced migration of human head and neck squamous cell carcinoma cells by blocking of EGF receptor pathways. BMC Cancer. 2011;11:388.PubMedPubMedCentralCrossRef
165.
go back to reference Mutschelknaus L, Azimzadeh O, Heider T, Winkler K, Vetter M, Kell R, et al. Radiation alters the cargo of exosomes released from squamous head and neck cancer cells to promote migration of recipient cells. Sci Rep. 2017;7(1):12423.PubMedPubMedCentralCrossRef Mutschelknaus L, Azimzadeh O, Heider T, Winkler K, Vetter M, Kell R, et al. Radiation alters the cargo of exosomes released from squamous head and neck cancer cells to promote migration of recipient cells. Sci Rep. 2017;7(1):12423.PubMedPubMedCentralCrossRef
166.
go back to reference Lu SL, Reh D, Li AG, Woods J, Corless CL, Kulesz-Martin M, et al. Overexpression of transforming growth factor beta1 in head and neck epithelia results in inflammation, angiogenesis, and epithelial hyperproliferation. Cancer Res. 2004;64(13):4405–10.PubMedCrossRef Lu SL, Reh D, Li AG, Woods J, Corless CL, Kulesz-Martin M, et al. Overexpression of transforming growth factor beta1 in head and neck epithelia results in inflammation, angiogenesis, and epithelial hyperproliferation. Cancer Res. 2004;64(13):4405–10.PubMedCrossRef
167.
go back to reference Feltl D, Zavadova E, Pala M, Hozak P. The dynamics of plasma transforming growth factor beta 1 (TGF-beta1) level during radiotherapy with or without simultaneous chemotherapy in advanced head and neck cancer. Oral Oncol. 2005;41(2):208–13.PubMedCrossRef Feltl D, Zavadova E, Pala M, Hozak P. The dynamics of plasma transforming growth factor beta 1 (TGF-beta1) level during radiotherapy with or without simultaneous chemotherapy in advanced head and neck cancer. Oral Oncol. 2005;41(2):208–13.PubMedCrossRef
168.
go back to reference Rodrigues-Junior DM, Tan SS, Lim SK, Leong HS, Melendez ME, Ramos CRN, et al. Circulating extracellular vesicle-associated TGFbeta3 modulates response to cytotoxic therapy in head and neck squamous cell carcinoma. Carcinogenesis. 2019;40(12):1452–61.PubMed Rodrigues-Junior DM, Tan SS, Lim SK, Leong HS, Melendez ME, Ramos CRN, et al. Circulating extracellular vesicle-associated TGFbeta3 modulates response to cytotoxic therapy in head and neck squamous cell carcinoma. Carcinogenesis. 2019;40(12):1452–61.PubMed
169.
go back to reference Mutschelknaus L, Peters C, Winkler K, Yentrapalli R, Heider T, Atkinson MJ, et al. Exosomes Derived from Squamous Head and Neck Cancer Promote Cell Survival after Ionizing Radiation. PLoS One. 2016;11(3):e0152213.PubMedPubMedCentralCrossRef Mutschelknaus L, Peters C, Winkler K, Yentrapalli R, Heider T, Atkinson MJ, et al. Exosomes Derived from Squamous Head and Neck Cancer Promote Cell Survival after Ionizing Radiation. PLoS One. 2016;11(3):e0152213.PubMedPubMedCentralCrossRef
170.
go back to reference Mager SELA, Breakefield I, Wood XO. MJ. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 2013;12(5):347–57. Mager SELA, Breakefield I, Wood XO. MJ. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 2013;12(5):347–57.
171.
go back to reference Chapman KB, Prendes MJ, Sternberg H, Kidd JL, Funk WD, Wagner J, et al. COL10A1 expression is elevated in diverse solid tumor types and is associated with tumor vasculature. Future Oncol. 2012;8(8):1031–40.PubMedCrossRef Chapman KB, Prendes MJ, Sternberg H, Kidd JL, Funk WD, Wagner J, et al. COL10A1 expression is elevated in diverse solid tumor types and is associated with tumor vasculature. Future Oncol. 2012;8(8):1031–40.PubMedCrossRef
172.
go back to reference Wei J, Nduom EK, Kong LY, Hashimoto Y, Xu S, Gabrusiewicz K, et al. MiR-138 exerts anti-glioma efficacy by targeting immune checkpoints. Neuro Oncol. 2016;18(5):639–48.PubMedCrossRef Wei J, Nduom EK, Kong LY, Hashimoto Y, Xu S, Gabrusiewicz K, et al. MiR-138 exerts anti-glioma efficacy by targeting immune checkpoints. Neuro Oncol. 2016;18(5):639–48.PubMedCrossRef
173.
go back to reference Li L, Lu S, Liang X, Cao B, Wang S, Jiang J, et al. gammadeltaTDEs: An Efficient Delivery System for miR-138 with Anti-tumoral and Immunostimulatory Roles on Oral Squamous Cell Carcinoma. Mol Ther Nucleic Acids. 2019;14:101–13.PubMedCrossRef Li L, Lu S, Liang X, Cao B, Wang S, Jiang J, et al. gammadeltaTDEs: An Efficient Delivery System for miR-138 with Anti-tumoral and Immunostimulatory Roles on Oral Squamous Cell Carcinoma. Mol Ther Nucleic Acids. 2019;14:101–13.PubMedCrossRef
174.
go back to reference Tomeh MA, Hadianamrei R, Zhao X. A Review of Curcumin and Its Derivatives as Anticancer Agents. Int J Mol Sci. 2019;20(5). Tomeh MA, Hadianamrei R, Zhao X. A Review of Curcumin and Its Derivatives as Anticancer Agents. Int J Mol Sci. 2019;20(5).
175.
go back to reference Sun D, Zhuang X, Xiang X, Liu Y, Zhang S, Liu C, et al. A novel nanoparticle drug delivery system: the anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol Ther. 2010;18(9):1606–14.PubMedPubMedCentralCrossRef Sun D, Zhuang X, Xiang X, Liu Y, Zhang S, Liu C, et al. A novel nanoparticle drug delivery system: the anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol Ther. 2010;18(9):1606–14.PubMedPubMedCentralCrossRef
176.
go back to reference Zhuang X, Xiang X, Grizzle W, Sun D, Zhang S, Axtell RC, et al. Treatment of brain inflammatory diseases by delivering exosome encapsulated anti-inflammatory drugs from the nasal region to the brain. Mol Ther. 2011;19(10):1769–79.PubMedPubMedCentralCrossRef Zhuang X, Xiang X, Grizzle W, Sun D, Zhang S, Axtell RC, et al. Treatment of brain inflammatory diseases by delivering exosome encapsulated anti-inflammatory drugs from the nasal region to the brain. Mol Ther. 2011;19(10):1769–79.PubMedPubMedCentralCrossRef
177.
go back to reference Yang T, Martin P, Fogarty B, Brown A, Schurman K, Phipps R, et al. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio rerio. Pharm Res. 2015;32(6):2003–14.PubMedPubMedCentralCrossRef Yang T, Martin P, Fogarty B, Brown A, Schurman K, Phipps R, et al. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio rerio. Pharm Res. 2015;32(6):2003–14.PubMedPubMedCentralCrossRef
178.
go back to reference Hoornstra D, Vesterlin J, Parnanen P, Al-Samadi A, Zlotogorski-Hurvitz A, Vered M, et al. Fermented Lingonberry Juice Inhibits Oral Tongue Squamous Cell Carcinoma Invasion In Vitro Similarly to Curcumin. Vivo. 2018;32(5):1089–95.CrossRef Hoornstra D, Vesterlin J, Parnanen P, Al-Samadi A, Zlotogorski-Hurvitz A, Vered M, et al. Fermented Lingonberry Juice Inhibits Oral Tongue Squamous Cell Carcinoma Invasion In Vitro Similarly to Curcumin. Vivo. 2018;32(5):1089–95.CrossRef
179.
go back to reference Pascucci L, Cocce V, Bonomi A, Ami D, Ceccarelli P, Ciusani E, et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: a new approach for drug delivery. J Control Release. 2014;192:262–70.PubMedCrossRef Pascucci L, Cocce V, Bonomi A, Ami D, Ceccarelli P, Ciusani E, et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: a new approach for drug delivery. J Control Release. 2014;192:262–70.PubMedCrossRef
180.
go back to reference Rani S, Ryan AE, Griffin MD, Ritter T. Mesenchymal Stem Cell-derived Extracellular Vesicles: Toward Cell-free Therapeutic Applications. Mol Ther. 2015;23(5):812–23.PubMedPubMedCentralCrossRef Rani S, Ryan AE, Griffin MD, Ritter T. Mesenchymal Stem Cell-derived Extracellular Vesicles: Toward Cell-free Therapeutic Applications. Mol Ther. 2015;23(5):812–23.PubMedPubMedCentralCrossRef
181.
go back to reference Sharma A, Khatun Z, Shiras A. Tumor exosomes: cellular postmen of cancer diagnosis and personalized therapy. Nanomedicine (Lond). 2016;11(4):421–37.CrossRef Sharma A, Khatun Z, Shiras A. Tumor exosomes: cellular postmen of cancer diagnosis and personalized therapy. Nanomedicine (Lond). 2016;11(4):421–37.CrossRef
182.
go back to reference Hao S, Bai O, Li F, Yuan J, Laferte S, Xiang J. Mature dendritic cells pulsed with exosomes stimulate efficient cytotoxic T-lymphocyte responses and antitumour immunity. Immunology. 2007;120(1):90–102.PubMedPubMedCentralCrossRef Hao S, Bai O, Li F, Yuan J, Laferte S, Xiang J. Mature dendritic cells pulsed with exosomes stimulate efficient cytotoxic T-lymphocyte responses and antitumour immunity. Immunology. 2007;120(1):90–102.PubMedPubMedCentralCrossRef
183.
go back to reference Kanuma T, Yamamoto T, Kobiyama K, Moriishi E, Masuta Y, Kusakabe T, et al. CD63-Mediated Antigen Delivery into Extracellular Vesicles via DNA Vaccination Results in Robust CD8(+) T Cell Responses. J Immunol. 2017;198(12):4707–15.PubMedCrossRef Kanuma T, Yamamoto T, Kobiyama K, Moriishi E, Masuta Y, Kusakabe T, et al. CD63-Mediated Antigen Delivery into Extracellular Vesicles via DNA Vaccination Results in Robust CD8(+) T Cell Responses. J Immunol. 2017;198(12):4707–15.PubMedCrossRef
184.
go back to reference Becker A, Thakur BK, Weiss JM, Kim HS, Peinado H, Lyden D. Extracellular Vesicles in Cancer: Cell-to-Cell Mediators of Metastasis. Cancer Cell. 2016;30(6):836–48.PubMedPubMedCentralCrossRef Becker A, Thakur BK, Weiss JM, Kim HS, Peinado H, Lyden D. Extracellular Vesicles in Cancer: Cell-to-Cell Mediators of Metastasis. Cancer Cell. 2016;30(6):836–48.PubMedPubMedCentralCrossRef
185.
go back to reference Qadir F, Aziz MA, Sari CP, Ma H, Dai H, Wang X, et al. Transcriptome reprogramming by cancer exosomes: identification of novel molecular targets in matrix and immune modulation. Mol Cancer. 2018;17(1):97.PubMedPubMedCentralCrossRef Qadir F, Aziz MA, Sari CP, Ma H, Dai H, Wang X, et al. Transcriptome reprogramming by cancer exosomes: identification of novel molecular targets in matrix and immune modulation. Mol Cancer. 2018;17(1):97.PubMedPubMedCentralCrossRef
186.
go back to reference Rodriguez Zorrilla S, Perez-Sayans M, Fais S, Logozzi M, Gallas Torreira M, Garcia Garcia A. A Pilot Clinical Study on the Prognostic Relevance of Plasmatic Exosomes Levels in Oral Squamous Cell Carcinoma Patients. Cancers (Basel). 2019;11(3). Rodriguez Zorrilla S, Perez-Sayans M, Fais S, Logozzi M, Gallas Torreira M, Garcia Garcia A. A Pilot Clinical Study on the Prognostic Relevance of Plasmatic Exosomes Levels in Oral Squamous Cell Carcinoma Patients. Cancers (Basel). 2019;11(3).
187.
go back to reference Topkas E, Keith P, Dimeski G, Cooper-White J, Punyadeera C. Evaluation of saliva collection devices for the analysis of proteins. Clin Chim Acta. 2012;413(13–14):1066–70.PubMedCrossRef Topkas E, Keith P, Dimeski G, Cooper-White J, Punyadeera C. Evaluation of saliva collection devices for the analysis of proteins. Clin Chim Acta. 2012;413(13–14):1066–70.PubMedCrossRef
188.
go back to reference Schulz BL, Cooper-White J, Punyadeera CK. Saliva proteome research: current status and future outlook. Crit Rev Biotechnol. 2013;33(3):246–59.PubMedCrossRef Schulz BL, Cooper-White J, Punyadeera CK. Saliva proteome research: current status and future outlook. Crit Rev Biotechnol. 2013;33(3):246–59.PubMedCrossRef
189.
go back to reference Kumeda N, Ogawa Y, Akimoto Y, Kawakami H, Tsujimoto M, Yanoshita R. Characterization of Membrane Integrity and Morphological Stability of Human Salivary Exosomes. Biol Pharm Bull. 2017;40(8):1183–91.PubMedCrossRef Kumeda N, Ogawa Y, Akimoto Y, Kawakami H, Tsujimoto M, Yanoshita R. Characterization of Membrane Integrity and Morphological Stability of Human Salivary Exosomes. Biol Pharm Bull. 2017;40(8):1183–91.PubMedCrossRef
190.
go back to reference Langevin S, Kuhnell D, Parry T, Biesiada J, Huang S, Wise-Draper T, et al. Comprehensive microRNA-sequencing of exosomes derived from head and neck carcinoma cells in vitro reveals common secretion profiles and potential utility as salivary biomarkers. Oncotarget. 2017;8(47):82459–74.PubMedPubMedCentralCrossRef Langevin S, Kuhnell D, Parry T, Biesiada J, Huang S, Wise-Draper T, et al. Comprehensive microRNA-sequencing of exosomes derived from head and neck carcinoma cells in vitro reveals common secretion profiles and potential utility as salivary biomarkers. Oncotarget. 2017;8(47):82459–74.PubMedPubMedCentralCrossRef
191.
go back to reference Zahran F, Ghalwash D, Shaker O, Al-Johani K, Scully C. Salivary microRNAs in oral cancer. Oral Dis. 2015;21(6):739–47.PubMedCrossRef Zahran F, Ghalwash D, Shaker O, Al-Johani K, Scully C. Salivary microRNAs in oral cancer. Oral Dis. 2015;21(6):739–47.PubMedCrossRef
192.
go back to reference Chiabotto G, Gai C, Deregibus MC, Camussi G. Salivary Extracellular Vesicle-Associated exRNA as Cancer Biomarker. Cancers (Basel). 2019;11(7). Chiabotto G, Gai C, Deregibus MC, Camussi G. Salivary Extracellular Vesicle-Associated exRNA as Cancer Biomarker. Cancers (Basel). 2019;11(7).
193.
go back to reference Gai C, Camussi F, Broccoletti R, Gambino A, Cabras M, Molinaro L, et al. Salivary extracellular vesicle-associated miRNAs as potential biomarkers in oral squamous cell carcinoma. BMC Cancer. 2018;18(1):439.PubMedPubMedCentralCrossRef Gai C, Camussi F, Broccoletti R, Gambino A, Cabras M, Molinaro L, et al. Salivary extracellular vesicle-associated miRNAs as potential biomarkers in oral squamous cell carcinoma. BMC Cancer. 2018;18(1):439.PubMedPubMedCentralCrossRef
194.
go back to reference Sharma S, Gillespie BM, Palanisamy V, Gimzewski JK. Quantitative nanostructural and single-molecule force spectroscopy biomolecular analysis of human-saliva-derived exosomes. Langmuir. 2011;27(23):14394–400.PubMedPubMedCentralCrossRef Sharma S, Gillespie BM, Palanisamy V, Gimzewski JK. Quantitative nanostructural and single-molecule force spectroscopy biomolecular analysis of human-saliva-derived exosomes. Langmuir. 2011;27(23):14394–400.PubMedPubMedCentralCrossRef
195.
go back to reference Zlotogorski-Hurvitz A, Dayan D, Chaushu G, Salo T, Vered M. Morphological and molecular features of oral fluid-derived exosomes: oral cancer patients versus healthy individuals. J Cancer Res Clin Oncol. 2016;142(1):101–10.PubMedCrossRef Zlotogorski-Hurvitz A, Dayan D, Chaushu G, Salo T, Vered M. Morphological and molecular features of oral fluid-derived exosomes: oral cancer patients versus healthy individuals. J Cancer Res Clin Oncol. 2016;142(1):101–10.PubMedCrossRef
196.
go back to reference Winck FV, Prado Ribeiro AC, Ramos Domingues R, Ling LY, Riano-Pachon DM, Rivera C, et al. Insights into immune responses in oral cancer through proteomic analysis of saliva and salivary extracellular vesicles. Sci Rep. 2015;5:16305.PubMedPubMedCentralCrossRef Winck FV, Prado Ribeiro AC, Ramos Domingues R, Ling LY, Riano-Pachon DM, Rivera C, et al. Insights into immune responses in oral cancer through proteomic analysis of saliva and salivary extracellular vesicles. Sci Rep. 2015;5:16305.PubMedPubMedCentralCrossRef
197.
go back to reference Katsiougiannis S, Chia D, Kim Y, Singh RP, Wong DT. Saliva exosomes from pancreatic tumor-bearing mice modulate NK cell phenotype and antitumor cytotoxicity. FASEB J. 2017;31(3):998–1010.PubMedCrossRef Katsiougiannis S, Chia D, Kim Y, Singh RP, Wong DT. Saliva exosomes from pancreatic tumor-bearing mice modulate NK cell phenotype and antitumor cytotoxicity. FASEB J. 2017;31(3):998–1010.PubMedCrossRef
198.
go back to reference Reclusa P, Taverna S, Pucci M, Durendez E, Calabuig S, Manca P, et al. Exosomes as diagnostic and predictive biomarkers in lung cancer. J Thorac Dis. 2017;9(Suppl 13):1373-S82. Reclusa P, Taverna S, Pucci M, Durendez E, Calabuig S, Manca P, et al. Exosomes as diagnostic and predictive biomarkers in lung cancer. J Thorac Dis. 2017;9(Suppl 13):1373-S82.
199.
go back to reference Lau CS, Wong DT. Breast cancer exosome-like microvesicles and salivary gland cells interplay alters salivary gland cell-derived exosome-like microvesicles in vitro. PLoS One. 2012;7(3):e33037.PubMedPubMedCentralCrossRef Lau CS, Wong DT. Breast cancer exosome-like microvesicles and salivary gland cells interplay alters salivary gland cell-derived exosome-like microvesicles in vitro. PLoS One. 2012;7(3):e33037.PubMedPubMedCentralCrossRef
Metadata
Title
The roles of extracellular vesicles in the development, microenvironment, anticancer drug resistance, and therapy of head and neck squamous cell carcinoma
Authors
Xueying Wang
Junnan Guo
Pingyang Yu
Lunhua Guo
Xionghui Mao
Junrong Wang
Susheng Miao
Ji Sun
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-021-01840-x

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 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