Extracellular vesicle-based biomarker in head and neck cancer: prospects and challenges

Linlin Wang, Ling Li, Guiquan Zhu

Malignancy Spectrum ›› 2024, Vol. 1 ›› Issue (2) : 75-90.

PDF(596 KB)
PDF(596 KB)
Malignancy Spectrum ›› 2024, Vol. 1 ›› Issue (2) : 75-90. DOI: 10.1002/msp2.24
REVIEW

Extracellular vesicle-based biomarker in head and neck cancer: prospects and challenges

Author information +
History +

Abstract

Head and neck squamous cell carcinoma (HNSCC) represents the sixth most common malignancy worldwide. However, very few established diagnostic biomarkers for HNSCC have been universally applied in clinical practice. Recently, much attention has been paid to extracellular vesicles (EVs) regarding their roles as cancer biomarkers because EVs carry plentiful cargoes, including lipids, proteins, nucleic acids, and metabolites. In HNSCC, several molecules carried by EVs, which are derived from peripheral blood and saliva, have been implicated to be effective in cancer detection, staging, treatment planning, response monitoring, and prognosis prediction. Although several EV molecules have been identified to be significantly correlated with a set of clinicalpathological parameters of HNSCC, several key limitations need to be resolved before the clinical application of EVs as carriers of biomarkers in HNSCC. In this review, we discuss current knowledge in the literature regarding EV-based biomarkers in HNSCC, emphasizing current limitations of their clinical applications.

Keywords

extracellular vesicles / exosomes / liquid biopsy / cancer biomarkers / head and neck cancer

Cite this article

Download citation ▾
Linlin Wang, Ling Li, Guiquan Zhu. Extracellular vesicle-based biomarker in head and neck cancer: prospects and challenges. Malignancy Spectrum, 2024, 1(2): 75‒90 https://doi.org/10.1002/msp2.24

References

[1]
Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharm Ther. 2001;69(3):89-95.
CrossRef Google scholar
[2]
FDA-NIH Biomarker Working Group. BEST (Biomarkers, EndpointS, and other Tools) Resource. Food and Drug Administration. 2016.
[3]
O’Connor JPB, Aboagye EO, Adams JE, et al. Imaging biomarker roadmap for cancer studies. Nat Rev Clin Oncol. 2017;14(3):169-186.
[4]
Vargas AJ, Harris CC. Biomarker development in the precision medicine era: lung cancer as a case study. Nat Rev Cancer. 2016;16(8):525-537.
CrossRef Google scholar
[5]
Henry NL, Hayes DF. Cancer biomarkers. Mol Oncol. 2012;6(2):140-146.
CrossRef Google scholar
[6]
Nonaka T, Wong DTW. Liquid biopsy in head and neck cancer: promises and challenges. J Dent Res. 2018;97(6):701-708.
CrossRef Google scholar
[7]
Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394-424.
CrossRef Google scholar
[8]
Wyss A, Hashibe M, Chuang SC, et al. Cigarette, cigar, and pipe smoking and the risk of head and neck cancers: pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. Am J Epidemiol. 2013;178(5):679-690.
[9]
Taberna M, Mena M, Pavón MA, Alemany L, Gillison ML, Mesía R. Human papillomavirus-related oropharyngeal cancer. Ann Oncol. 2017;28(10):2386-2398.
CrossRef Google scholar
[10]
Kostareli E, Holzinger D, Hess J. New concepts for translational head and neck oncology: lessons from HPV-related oropharyngeal squamous cell carcinomas. Front Oncol. 2012;2:36.
CrossRef Google scholar
[11]
Kreimer AR, Clifford GM, Boyle P, Franceschi S. Human papillomavirus types in head and neck squamous cell carcinomas worldwide: a systematic review. Cancer Epidemiol Biomarkers Prev. 2005;14(2):467-475.
CrossRef Google scholar
[12]
Argiris A, Karamouzis MV, Raben D, Ferris RL. Head and neck cancer. Lancet. 2008;371(9625):1695-1709.
CrossRef Google scholar
[13]
Mehanna H, Jones TM, Gregoire V, Ang KK. Oropharyngeal carcinoma related to human papillomavirus. BMJ. 2010;340: c1439.
CrossRef Google scholar
[14]
Lydiatt WM, Patel SG, O’Sullivan B, et al. Head and neck cancers-major changes in the American Joint Committee on cancer eighth edition cancer staging manual. CA Cancer J Clin. 2017;67(2):122-137.
CrossRef Google scholar
[15]
Colevas AD, Yom SS, Pfister DG, et al. NCCN guidelines insights: head and neck cancers, version 1.2018. J Natl Compr Canc Netw. 2018;16(5):479-490.
CrossRef Google scholar
[16]
Economopoulou P, de Bree R, Kotsantis I, Psyrri A. Diagnostic tumor markers in head and neck squamous cell carcinoma (HNSCC) in the clinical setting. Front Oncol. 2019;9:827.
CrossRef Google scholar
[17]
Ferris RL. Immunology and immunotherapy of head and neck cancer. J Clin Oncol. 2015;33(29):3293-3304.
CrossRef Google scholar
[18]
Seiwert TY, Burtness B, Mehra R, et al. Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): an open-label, multicentre, phase 1b trial. Lancet Oncol. 2016;17(7):956-965.
CrossRef Google scholar
[19]
Larkins E, Blumenthal GM, Yuan W, et al. FDA approval summary: pembrolizumab for the treatment of recurrent or metastatic head and neck squamous cell carcinoma with disease progression on or after platinum-containing chemotherapy. Oncologist. 2017;22(7):873-878.
CrossRef Google scholar
[20]
Burtness B, Harrington KJ, Greil R, et al. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): a randomised, open-label, phase 3 study. Lancet. 2019;394(10212):1915-1928.
[21]
Vermorken JB, Mesia R, Rivera F, et al. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N Engl J Med. 2008;359(11):1116-1127.
CrossRef Google scholar
[22]
Keren S, Shoude Z, Lu Z, Beibei Y. Role of EGFR as a prognostic factor for survival in head and neck cancer: a meta-analysis. Tumor Biol. 2014;35(3):2285-2295.
CrossRef Google scholar
[23]
Bentzen SM, Atasoy BM, Daley FM, et al. Epidermal growth factor receptor expression in pretreatment biopsies from head and neck squamous cell carcinoma as a predictive factor for a benefit from accelerated radiation therapy in a randomized controlled trial. J Clin Oncol. 2005;23(24):5560-5567.
CrossRef Google scholar
[24]
Kang H, Kiess A, Chung CH. Emerging biomarkers in head and neck cancer in the era of genomics. Nat Rev Clin Oncol. 2015;12(1):11-26.
CrossRef Google scholar
[25]
Zhou G, Liu Z, Myers JN. TP53 mutations in head and neck squamous cell carcinoma and their impact on disease progression and treatment response. J Cell Biochem. 2016;117(12):2682-2692.
CrossRef Google scholar
[26]
Poeta ML, Manola J, Goldwasser MA, et al. TP53 mutations and survival in squamous-cell carcinoma of the head and neck. N Engl J Med. 2007;357(25):2552-2561.
CrossRef Google scholar
[27]
Perrone F, Bossi P, Cortelazzi B, et al. TP53 mutations and pathologic complete response to neoadjuvant cisplatin and fluorouracil chemotherapy in resected oral cavity squamous cell carcinoma. J Clin Oncol. 2010;28(5):761-766.
CrossRef Google scholar
[28]
Weinstein JN, Collisson EA, Mills GB, et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nature Genet. 2013;45(10):1113-1120.
CrossRef Google scholar
[29]
Ruivo CF, Adem B, Silva M, Melo SA. The biology of cancer exosomes: insights and new perspectives. Cancer Res. 2017;77(23):6480-6488.
CrossRef Google scholar
[30]
Thery C, Witwer KW, Aikawa E, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7(1):1535750.
CrossRef Google scholar
[31]
Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478):eaau6977.
[32]
van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19:213-228.
CrossRef Google scholar
[33]
Liu Y, Gu Y, Cao X. The exosomes in tumor immunity. Oncoimmunology. 2015;4(9):e1027472.
CrossRef Google scholar
[34]
Wolf P. The nature and significance of platelet products in human plasma. Br J Haematol. 1967;13(3):269-288.
CrossRef Google scholar
[35]
Johnstone RM, Adam M, Hammond JR, Orr L, Turbide C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem. 1987;262(19):9412-9420.
CrossRef Google scholar
[36]
Raposo G, Nijman HW, Stoorvogel W, et al. B lymphocytes secrete antigen-presenting vesicles. J Exp Med. 1996;183(3):1161-1172.
CrossRef Google scholar
[37]
Keerthikumar S, Chisanga D, Ariyaratne D, et al. ExoCarta: a web-based compendium of exosomal cargo. J Mol Biol. 2016;428(4):688-692.
CrossRef Google scholar
[38]
Kim DK, Lee J, Simpson RJ, Lötvall J, Gho YS. EVpedia: a community web resource for prokaryotic and eukaryotic extra-cellular vesicles research. Semin Cell Dev Biol. 2015;40:4-7.
CrossRef Google scholar
[39]
Kalra H, Simpson RJ, Ji H, et al. Vesiclepedia: a compendium for extracellular vesicles with continuous community annotation. PLoS Biol. 2012;10(12):e1001450.
CrossRef Google scholar
[40]
Record M, Carayon K, Poirot M, Silvente-Poirot S. Exosomes as new vesicular lipid transporters involved in cell-cell communication and various pathophysiologies. Biochim Biophys Acta. 2014;1841(1):108-120.
CrossRef Google scholar
[41]
Wubbolts R, Leckie RS, Veenhuizen PTM, et al. Proteomic and biochemical analyses of human B cell-derived exosomes. J Biol Chem. 2003;278(13):10963-10972.
CrossRef Google scholar
[42]
Subra C, Grand D, Laulagnier K, et al. Exosomes account for vesicle-mediated transcellular transport of activatable phospholipases and prostaglandins. J Lipid Res. 2010;51(8):2105-2120.
CrossRef Google scholar
[43]
Théry C, Boussac M, Véron P, et al. Proteomic analysis of dendritic cell-derived exosomes: a secreted subcellular compartment distinct from apoptotic vesicles. J Immunol. 2001;166(12):7309-7318.
CrossRef Google scholar
[44]
Kibria G, Ramos EK, Wan Y, Gius DR, Liu H. Exosomes as a drug delivery system in cancer therapy: potential and challenges. Mol Pharm. 2018;15:3625-3633.
CrossRef Google scholar
[45]
He C, Zheng S, Luo Y, Wang B. Exosome theranostics: biology and translational medicine. Theranostics. 2018;8(1):237-255.
CrossRef Google scholar
[46]
Steinbichler TB, Dudás J, Riechelmann H, Skvortsova II. The role of exosomes in cancer metastasis. Sem Cancer Biol. 2017;44:170-181.
CrossRef Google scholar
[47]
Kalluri R. The biology and function of exosomes in cancer. J Clin Invest. 2016;126(4):1208-1215.
CrossRef Google scholar
[48]
Zhou H, Cheruvanky A, Hu X, et al. Urinary exosomal transcription factors, a new class of biomarkers for renal disease. Kidney Int. 2008;74(5):613-621.
CrossRef Google scholar
[49]
Al-Nedawi K, Meehan B, Micallef J, et al. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell Biol. 2008;10(5):619-624.
CrossRef Google scholar
[50]
Hawari FI, Rouhani FN, Cui X, et al. Release of full-length 55-kDa TNF receptor 1 in exosome-like vesicles: a mechanism for generation of soluble cytokine receptors. Proc Natl Acad Sci USA. 2004;101(5):1297-1302.
CrossRef Google scholar
[51]
Jeppesen DK, Nawrocki A, Jensen SG, et al. Quantitative proteomics of fractionated membrane and lumen exosome proteins from isogenic metastatic and nonmetastatic bladder cancer cells reveal differential expression of EMT factors. Proteomics. 2014;14(6):699-712.
CrossRef Google scholar
[52]
Masciopinto F, Giovani C, Campagnoli S, et al. Association of hepatitis C virus envelope proteins with exosomes. Eur J Immunol. 2004;34(10):2834-2842.
CrossRef Google scholar
[53]
Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biol. 2007;9(6):654-659.
CrossRef Google scholar
[54]
Qu L, Ding J, Chen C, et al. Exosome-transmitted lncARSR promotes sunitinib resistance in renal cancer by acting as a competing endogenous RNA. Cancer Cell. 2016;29(5):653-668.
CrossRef Google scholar
[55]
Li Y, Zheng Q, Bao C, et al. Circular RNA is enriched and stable in exosomes: a promising biomarker for cancer diagnosis. Cell Res. 2015;25(8):981-984.
CrossRef Google scholar
[56]
Li S, Li Y, Chen B, et al. exoRBase: a database of circRNA, lncRNA and mRNA in human blood exosomes. Nucleic Acids Res. 2018;46(D1):D106-D112.
CrossRef Google scholar
[57]
Principe S, Hui ABY, Bruce J, Sinha A, Liu FF, Kislinger T. Tumor-derived exosomes and microvesicles in head and neck cancer: implications for tumor biology and biomarker discovery. Proteomics. 2013;13(10-11):1608-1623.
CrossRef Google scholar
[58]
Wang J, Zhou Y, Lu J, 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.
CrossRef Google scholar
[59]
Mayne GC, Woods CM, Dharmawardana N, et al. Cross validated serum small extracellular vesicle microRNAs for the detection of oropharyngeal squamous cell carcinoma. J Transl Med. 2020;18(1):280.
CrossRef Google scholar
[60]
Qin X, Guo H, 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;20:12.
CrossRef Google scholar
[61]
Li L, Li C, Wang S, 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):1770-1780.
CrossRef Google scholar
[62]
He L, Ping F, Fan Z, et al. Salivary exosomal miR-24-3p serves as a potential detective biomarker for oral squamous cell carcinoma screening. Biomed Pharmacother. 2020;121:109553.
CrossRef Google scholar
[63]
Gai C, Camussi F, Broccoletti R, et al. Salivary extracellular vesicle-associated miRNAs as potential biomarkers in oral squamous cell carcinoma. BMC Cancer. 2018;18:439.
CrossRef Google scholar
[64]
Luo Y, Liu F, Guo J, Gui R. Upregulation of circ_0000199 in circulating exosomes is associated with survival outcome in OSCC. Sci Rep. 2020;10(1):13739.
CrossRef Google scholar
[65]
Langevin S, Kuhnell D, Parry 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):82459-82474.
CrossRef Google scholar
[66]
Chen F, Xu B, Li J, et al. Hypoxic tumour cell-derived exosomal miR-340-5p promotes radioresistance of oesophageal squamous cell carcinoma via KLF10. J Exp Clin Cancer Res. 2021;40(1):38.
CrossRef Google scholar
[67]
Panvongsa W, Siripoon T, Worakitchanon W, et al. Plasma extracellular vesicle microRNA-491-5p as diagnostic and prognostic marker for head and neck squamous cell carcinoma. Cancer Sci. 2021;112(10):4257-4269.
CrossRef Google scholar
[68]
Huang Q, Shen YJ, Hsueh CY, et al. Plasma extracellular vesicles-derived miR-99a-5p: a potential biomarker to predict early head and neck squamous cell carcinoma. Pathol Oncol Res. 2022;28:1610699.
CrossRef Google scholar
[69]
Xi J, Zeng Z, Li X, Zhang X, Xu J. Expression and diagnostic value of tRNA-Derived fragments secreted by extracellular vesicles in hypopharyngeal carcinoma. Onco Targets Ther. 2021;14:4189-4199.
CrossRef Google scholar
[70]
Bigagli E, Locatello LG, Di Stadio A, et al. Extracellular vesicles miR-210 as a potential biomarker for diagnosis and survival prediction of oral squamous cell carcinoma patients. J Oral Pathol Med. 2022;51(4):350-357.
CrossRef Google scholar
[71]
He T, Guo X, Li X, Liao C, Wang X, He K. Plasma-derived exosomal microRNA-130a serves as a noninvasive biomarker for diagnosis and prognosis of oral squamous cell carcinoma. J Oncol. 2021;2021:5547911.
CrossRef Google scholar
[72]
Zhao Q, Zheng X, Guo H, et al. Serum exosomal miR-941 as a promising oncogenic biomarker for laryngeal squamous cell carcinoma. J Cancer. 2020;11(18):5329-5344.
CrossRef Google scholar
[73]
Chen G, Huang AC, Zhang W, et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature. 2018;560(7718):382-386.
CrossRef Google scholar
[74]
Muller L, Simms P, Hong CS, et al. Human tumor-derived exosomes (TEX) regulate Treg functions via cell surface signaling rather than uptake mechanisms. Oncoimmunology. 2017;6(8):e1261243.
CrossRef Google scholar
[75]
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.
CrossRef Google scholar
[76]
Theodoraki MN, Hoffmann TK, Jackson EK, Whiteside TL. Exosomes in HNSCC plasma as surrogate markers of tumour progression and immune competence. Clin Exp Immunol. 2018;194(1):67-78.
CrossRef Google scholar
[77]
Theodoraki MN, Hoffmann TK, Whiteside TL. Separation of plasma-derived exosomes into CD3(+) and CD3(−) fractions allows for association of immune cell and tumour cell markers with disease activity in HNSCC patients. Clin Exp Immunol. 2018;192(3):271-283.
CrossRef Google scholar
[78]
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.
CrossRef Google scholar
[79]
Hofmann L, Ludwig S, Schuler PJ, Hoffmann TK, Brunner C, Theodoraki MN. The potential of CD16 on plasma-derived exosomes as a liquid biomarker in head and neck cancer. Int J Mol Sci. 2020;21(11):3739.
CrossRef Google scholar
[80]
Sanada T, Islam A, Kaminota T, et al. Elevated exosomal lysyl oxidase like 2 is a potential biomarker for head and neck squamous cell carcinoma. Laryngoscope. 2020;130(5): E327-E334.
CrossRef Google scholar
[81]
Tang X, Chang C, Guo J, et al. Tumour-secreted Hsp90α on external surface of exosomes mediates tumour—stromal cell communication via autocrine and paracrine mechanisms. Sci Rep. 2019;9:15108.
CrossRef Google scholar
[82]
Lauwers E, Wang YC, Gallardo R, et al. Hsp90 mediates membrane deformation and exosome release. Mol Cell. 2018;71(5):689-702.
CrossRef Google scholar
[83]
Ono K, Eguchi T, Sogawa C, et al. HSP-enriched properties of extracellular vesicles involve survival of metastatic oral cancer cells. J Cell Biochem. 2018;119(9):7350-7362.
CrossRef Google scholar
[84]
Pickup M, Novitskiy S, Moses HL. The roles of TGFβ in the tumour microenvironment. Nat Rev Cancer. 2013;13(11):788-799.
CrossRef Google scholar
[85]
Rodrigues-Junior DM, Tan SS, Lim SK, et al. Circulating extracellular vesicle-associated TGFβ3 modulates response to cytotoxic therapy in head and neck squamous cell carcinoma. Carcinogenesis. 2019;40(12):1452-1461.
CrossRef Google scholar
[86]
Li C, Zhou Y, Liu J, et al. Potential markers from serumpurified exosomes for detecting oral squamous cell carcinoma metastasis. Cancer Epidemiol Biomarkers Prev. 2019;28(10):1668-1681.
CrossRef Google scholar
[87]
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.
CrossRef Google scholar
[88]
Ludwig N, Yerneni SS, Harasymczuk M, et al. TGFβ carrying exosomes in plasma: potential biomarkers of cancer progression in patients with head and neck squamous cell carcinoma. Br J Cancer. 2023;128(9):1733-1741.
CrossRef Google scholar
[89]
Huang Q, Hsueh CY, Shen YJ, et al. Small extracellular vesicle-packaged TGFβ1 promotes the reprogramming of normal fibroblasts into cancer-associated fibroblasts by regulating fibronectin in head and neck squamous cell carcinoma. Cancer Lett. 2021;517:1-13.
CrossRef Google scholar
[90]
Winck FV, Prado Ribeiro AC, Ramos Domingues R, et al. Insights into immune responses in oral cancer through proteomic analysis of saliva and salivary extracellular vesicles. Sci Rep. 2015;5:16305.
CrossRef Google scholar
[91]
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):1010-1020.
CrossRef Google scholar
[92]
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.
CrossRef Google scholar
[93]
Qu X, Leung TCN, Ngai SM, et al. Proteomic analysis of circulating extracellular vesicles identifies potential biomarkers for lymph node metastasis in oral tongue squamous cell carcinoma. Cells. 2021;10(9):2179.
CrossRef Google scholar
[94]
Nakamichi E, Sakakura H, Mii S, et al. Detection of serum/salivary exosomal Alix in patients with oral squamous cell carcinoma. Oral Dis. 2021;27(3):439-447.
CrossRef Google scholar
[95]
Cecchettini A, Finamore F, Puxeddu I, Ferro F, Baldini C. Salivary extracellular vesicles versus whole saliva: new perspectives for the identification of proteomic biomarkers in Sjögren’s syndrome. Clin Exp Rheumatol. 2019;37 suppl 118(3):240-248.
[96]
Roi A, Roi CI, Negruţiu ML, Riviş M, Sinescu C, Rusu LC. The challenges of OSCC diagnosis: salivary cytokines as potential biomarkers. J Clin Med. 2020;9(9):2866.
CrossRef Google scholar
[97]
Sharma S, Rasool HI, Palanisamy V, et al. Structural-mechanical characterization of nanoparticle exosomes in human saliva, using correlative AFM, FESEM, and force spectroscopy. ACS Nano. 2010;4(4):1921-1926.
CrossRef Google scholar
[98]
Xiao C, Song F, Zheng YL, Lv J, Wang QF, Xu N. Exosomes in head and neck squamous cell carcinoma. Front Oncol. 2019;9:894.
CrossRef Google scholar
[99]
Yap T, Vella L, Seers C, et al. Oral swirl samples—a robust source of microRNA protected by extracellular vesicles. Oral Dis. 2017;23(3):312-317.
CrossRef Google scholar
[100]
Gonzalez-Begne M, Lu B, Han X, et al. Proteomic analysis of human parotid gland exosomes by multidimensional protein identification technology (MudPIT). J Proteome Res. 2009;8(3):1304-1314.
CrossRef Google scholar
[101]
Ogawa Y, Kanai-Azuma M, Akimoto Y, Kawakami H, Yanoshita R. Exosome-like vesicles with dipeptidyl peptidase IV in human saliva. Biol Pharm Bull. 2008;31(6):1059-1062.
CrossRef Google scholar
[102]
Ogawa Y, Miura Y, Harazono A, et al. Proteomic analysis of two types of exosomes in human whole saliva. Biol Pharm Bull. 2011;34(1):13-23.
CrossRef Google scholar
[103]
Zarovni N, Corrado A, Guazzi P, et al. Integrated isolation and quantitative analysis of exosome shuttled proteins and nucleic acids using immunocapture approaches. Methods. 2015;87:46-58.
CrossRef Google scholar
[104]
Ludwig N, Whiteside TL, Reichert TE. Challenges in exosome isolation and analysis in health and disease. Int J Mol Sci. 2019;20(19):4684.
CrossRef Google scholar
[105]
Li P, Kaslan M, Lee SH, Yao J, Gao Z. Progress in exosome isolation techniques. Theranostics. 2017;7(3):789-804.
CrossRef Google scholar
[106]
Royo F, Zuñiga-Garcia P, Sanchez-Mosquera P, et al. Different EV enrichment methods suitable for clinical settings yield different subpopulations of urinary extracellular vesicles from human samples. J Extracell Vesicles. 2016;5:29497.
CrossRef Google scholar
[107]
Tang YT, Huang YY, Zheng L, et al. Comparison of isolation methods of exosomes and exosomal RNA from cell culture medium and serum. Int J Mol Med. 2017;40(3):834-844.
CrossRef Google scholar
[108]
Kalra H, Adda CG, Liem M, et al. Comparative proteomics evaluation of plasma exosome isolation techniques and assessment of the stability of exosomes in normal human blood plasma. Proteomics. 2013;13(22):3354-3364.
CrossRef Google scholar
[109]
Lane RE, Korbie D, Hill MM, Trau M. Extracellular vesicles as circulating cancer biomarkers: opportunities and challenges. Clin Transl Med. 2018;7(1):14.
CrossRef Google scholar
[110]
Gardiner C, Di Vizio D, Sahoo S, et al. Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey. J Extracell Vesicles. 2016;5:32945.
CrossRef Google scholar
[111]
Théry C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. 2006;30:3.22.1-3.22.29.
CrossRef Google scholar
[112]
Nordin JZ, Lee Y, Vader P, et al. Ultrafiltration with size-exclusion liquid chromatography for high yield isolation of extracellular vesicles preserving intact biophysical and functional properties. Nanomed Nanotechnol Biol Med. 2015;11(4):879-883.
CrossRef Google scholar
[113]
Baranyai T, Herczeg K, Onódi Z, et al. Isolation of exosomes from blood plasma: qualitative and quantitative comparison of ultracentrifugation and size exclusion chromatography methods. PLoS One. 2015;10(12):e0145686.
CrossRef Google scholar
[114]
Wang S, Gao X, Liu X, et al. CD133+ cancer stem-like cells promote migration and invasion of salivary adenoid cystic carcinoma by inducing vasculogenic mimicry formation. Oncotarget. 2016;7(20):29051-29062.
CrossRef Google scholar
[115]
Kang D, Oh S, Ahn SM, Lee BH, Moon MH. Proteomic analysis of exosomes from human neural stem cells by flow field-flow fractionation and nanoflow liquid chromatography –tandem mass spectrometry. J Proteome Res. 2008;7(8):3475-3480.
CrossRef Google scholar
[116]
Musante L, Tataruch D, Gu D, et al. A simplified method to recover urinary vesicles for clinical applications, and sample banking. Sci Rep. 2014;4:7532.
CrossRef Google scholar
[117]
Li X, Corbett AL, Taatizadeh E, et al. Challenges and opportunities in exosome research-perspectives from biology, engineering, and cancer therapy. APL Bioeng. 2019;3(1):011503.
CrossRef Google scholar
[118]
Gholizadeh S, Shehata Draz M, Zarghooni M, et al. Micro-fluidic approaches for isolation, detection, and characterization of extracellular vesicles: current status and future directions. Biosens Bioelectron. 2017;91:588-605.
CrossRef Google scholar
[119]
Guo SC, Tao SC, Dawn H. Microfluidics-based on-a-chip systems for isolating and analysing extracellular vesicles. J Extracell Vesicles. 2018;7(1):1508271.
CrossRef Google scholar
[120]
Jablonska J, Pietrowska M, Ludwig S, Lang S, Thakur BK. Challenges in the isolation and proteomic analysis of cancer exosomes-implications for translational research. Proteomes. 2019;7(2):22.
CrossRef Google scholar
[121]
Pitt JM, André F, Amigorena S, et al. Dendritic cell-derived exosomes for cancer therapy. J Clin Invest. 2016;126(4):1224-1232.
CrossRef Google scholar
[122]
Li CC, Eaton SA, Young PE, et al. Glioma microvesicles carry selectively packaged coding and non-coding RNAs which alter gene expression in recipient cells. RNA Biol. 2013;10(8):1333-1344.
CrossRef Google scholar
[123]
Caby MP, Lankar D, Vincendeau-Scherrer C, Raposo G, Bonnerot C. Exosomal-like vesicles are present in human blood plasma. Int Immunol. 2005;17(7):879-887.
CrossRef Google scholar
[124]
Lässer C, Alikhani VS, Ekström K, et al. Human saliva, plasma and breast milk exosomes contain RNA: uptake by macro-phages. J Transl Med. 2011;9:9.
CrossRef Google scholar
[125]
Hornung S, Dutta S, Bitan G. CNS-derived blood exosomes as a promising source of biomarkers: opportunities and challenges. Front Mol Neurosci. 2020;13:38.
CrossRef Google scholar
[126]
Wang W, Luo J, Wang S. Recent progress in isolation and detection of extracellular vesicles for cancer diagnostics. Adv Healthcare Mater. 2018;7:e1800484.
CrossRef Google scholar
[127]
Smith CJ, Osborn AM. Advantages and limitations of quantitative PCR (Q-PCR)-based approaches in microbial ecology. FEMS Microbiol Ecol. 2009;67(1):6-20.
CrossRef Google scholar
[128]
Oksvold MP, Kullmann A, Forfang L, et al. Expression of B-cell surface antigens in subpopulations of exosomes released from B-cell lymphoma cells. Clin Ther. 2014;36(6):847-862.
CrossRef Google scholar
[129]
Occhipinti G, Giulietti M, Principato G, Piva F. The choice of endogenous controls in exosomal microRNA assessments from biofluids. Tumor Biol. 2016;37(9):11657-11665.
CrossRef Google scholar
[130]
Ribeiro IP, de Melo JB, Carreira IM. Head and neck cancer: searching for genomic and epigenetic biomarkers in body fluids—the state of art. Mol Cytogenet. 2019;12:33.
CrossRef Google scholar
[131]
van Ginkel JH, Slieker FJB, de Bree R, van Es RJJ, Van Cann EM, Willems SM. Cell-free nucleic acids in body fluids as biomarkers for the prediction and early detection of recurrent head and neck cancer: a systematic review of the literature. Oral Oncol. 2017;75:8-15.
CrossRef Google scholar
[132]
Kulasinghe A, Hughes BGM, Kenny L, Punyadeera C. An update: circulating tumor cells in head and neck cancer. Expert Rev Mol Diagn. 2019;19(12):1109-1115.
CrossRef Google scholar
[133]
Chai RC, Lim Y, Frazer IH, et al. A pilot study to compare the detection of HPV-16 biomarkers in salivary oral rinses with tumour p16INK4a expression in head and neck squamous cell carcinoma patients. BMC Cancer. 2016;16:178.
CrossRef Google scholar
[134]
Mazurek AM, Rutkowski T, Fiszer-Kierzkowska A, Małusecka E, Składowski K. Assessment of the total cfDNA and HPV16/18 detection in plasma samples of head and neck squamous cell carcinoma patients. Oral Oncol. 2016;54:36-41.
CrossRef Google scholar
[135]
Ahn SM, Chan JY, Zhang Z, et al. Saliva and plasma quantitative polymerase chain reaction-based detection and surveillance of human papillomavirus-related head and neck cancer. JAMA Otolaryngol Head Neck Surg. 2014;140(9):846-854.
CrossRef Google scholar
[136]
Nguyen B, Meehan K, Pereira MR, et al. A comparative study of extracellular vesicle-associated and cell-free DNA and RNA for HPV detection in oropharyngeal squamous cell carcinoma. Sci Rep. 2020;10(1):6083.
CrossRef Google scholar

RIGHTS & PERMISSIONS

2024 2024 The Authors. Malignancy Spectrum published by John Wiley & Sons Australia, Ltd on behalf of Higher Education Press.
AI Summary AI Mindmap
PDF(596 KB)

Accesses

Citations

Detail

Sections
Recommended

/