Open access peer-reviewed chapter

Diagnostic Potential of Salivary Exosomes in Oral Cancer

Written By

Henry Ademola Adeola, Haly Holmes and Dada Oluwaseyi Temilola

Submitted: 07 September 2020 Reviewed: 24 September 2020 Published: 20 November 2020

DOI: 10.5772/intechopen.94192

From the Edited Volume

Oral Cancer - Current Concepts and Future Perspectives

Edited by Gokul Sridharan

Chapter metrics overview

505 Chapter Downloads

View Full Metrics

Abstract

“Omics” based concepts and techniques are gaining momentum in the field of oral medicine, spurred on by rapid advancements within the field of precision diagnostics and therapeutics. Oral cancer, specifically oral squamous cell carcinoma is the most common head and neck cancer, posing both diagnostic and prognostic challenges globally. Saliva offers several advantages as a diagnostic tool and has gained recognition as a biological medium for liquid biopsy. Salivary biomarkers, such as exosomes not only contain the full spectrum of genomic, lipidomic and proteomic material from its cell of origin, but are also more stable and consistently measurable in saliva due to their phospholipid structural protection of their merchandise/contents. Salivary exosomes are mediators in communication and transfer of contents between cancer and normal cells and thus key role players in mediating the tumor environment. Even though exosomes have been widely employed to investigate systemic diseases including head and neck cancers, unraveling the biologic mechanisms, scope of application of salivary tumor-derived exosomes and overcoming restrictions in this emergent field of saliva-exosomics warrants further investigation.

Keywords

  • Saliva
  • exosomes
  • oral cancer
  • exosomics
  • omics-based approaches

1. Introduction

Despite the fact, that there is a relatively wider coverage and application of several emerging molecular and omics-based techniques, in the field of medicine; many of these concepts (and techniques) are only beginning to gain recognition in the field of dentistry [1]. The completion of the human genome project [2, 3, 4], has expanded the trajectory for precision diagnostic and therapeutic potential thereof across many biomedical fields [5, 6]. The clinical lexicon in the post-genomic era is now burgeoning with various personalized application of genomics (and phenomics) to improving the cancer management pipelines [1, 6, 7, 8, 9].

Orthogonal (but complementary) multimodal approaches to conventional histopathology [10, 11], such as liquid biopsies technologies have emerged as useful tools for clinical oncology [11, 12, 13], early tumor diagnosis and biomonitoring [14, 15, 16, 17], as well as therapeutic decision making and delivery [18]. Nano-scaled multivesicular exosomes have emerged as important components of the tumor circulome that has significantly improved the cancer diagnostic field [11, 14]. Furthermore, salivary exosomes have been applied to improve the diagnosis of various cancers [19, 20, 21, 22, 23, 24, 25, 26], including oral cancers [25, 26]. The focus of this chapter is to review the applications and prospects of salivary exosomes in oral cancer detection (Figure 1).

Figure 1.

Overview of origin and theranostic value of exosomes in oral cancer.

Advertisement

2. Salivary diagnostics

Human saliva is a multifunctional biological fluid, which facilitates digestion, swallowing, tasting, tissue lubrication and protection against infectious organisms. It is comprised largely of water (99%) and other biochemical substances such as proteins, nucleic acids, mucins, immunoglobulins, a variety of electrolytes and lipids [27, 28]. Whole saliva production is derived from all the salivary glands including the gingival crevicular fluid [29]. Saliva, which has been used for diagnostic purposes for over 2000 years, plays a key role in the maintenance of general and particularly oral health homeostasis [30]. The health of individuals has been determined by salivary changes such as amount produced, smell, ropiness and gustatory sensation [31, 32].

Components of the salivary glands are responsible for production, modification, transportation and secretion of saliva into the oral cavity (via acinar cells, various ductal system cells, and myoepithelial cells) [33]. The close proximity of a network of highly permeable blood capillaries to the saliva producing acinar cells facilitates the free exchange of blood-derived molecules into the acinar cells [34], which enter the salivary tissues either via transcellular (passive and active transport) or paracellular (extracellular ultrafiltration) routes [35, 36]. This transfer potentially influences the molecular constituency of oral fluids. Diffusion is the most common transport mechanism of molecules from blood into the saliva and this process is driven/influenced by the size and the electric charge of the molecules [37]. Active transport involves the transcellular transportation of blood into saliva via secretory acinar cells of the salivary glands. Ultra-filtration is the major mode by which molecules are transported into saliva via the paracellular route, whereby small sized blood molecules filter into saliva through the spaces between ductal and acinar cells [37]. Salivary acini secrete saliva into collecting ducts, where sodium reabsorption, and bicarbonate and potassium are secretion takes place, thus altering the composition of the saliva [38, 39]. Even though blood molecules transported through ultra-filtration into saliva are usually in low concentration in comparison to their levels in blood, this mode of transport enables blood molecules such as DNA, RNA, proteins, metabolites and exosomes into saliva through the salivary gland. This confers the possibility for oral fluids to harbor molecular information indicative of an individual’s current state of health. This information may be reflected by changes in the concentrations of these molecules or mutation in the genetic constitution of the molecules which are also present in saliva - serving as potential salivary biomarkers for diagnosis, prognosis and monitoring of therapeutic responses [40].

Many body fluids have been explored as alternative sources for biomarkers in molecular diagnosis of cancers, genetic, immunological and other systemic diseases [41, 42, 43, 44, 45]. However, blood, urine and saliva are the most used media for discovery of biomarkers [46]. Saliva is a rich source of proteins and its DNA, RNA, and protein content is analogous to that of blood with significant commonality in hormones, antibodies and other molecules. Most of the resident salivary protein constituents are synthesized within the salivary glands, with the rest transported from blood or lymph into saliva and used as biomarkers for disease diagnosis and screening purposes [47].

Saliva has the advantages over blood in that it is a readily available specimen which can be collected by non-invasive techniques and is recommended as the diagnostic medium in vulnerable populations such as children. The retrieval of multiple salivary samples from the same individual is possible with minimal discomfort and saliva is safer to work with when compared with blood samples. For example, there are some factors in saliva which help to inhibit HIV infectivity thereby limiting the rate of HIV transmission through the oral cavity [48]. Saliva sample processing is more economical and does not clot making it easier to store and ship with less manipulation. However, the low level of protein detection in saliva is sensitive to the method of saliva collection and specimen contamination. Normal high-abundance salivary constituents such as amylase and proline rich proteins (during stimulated salivary collection methods especially), may dilute the presence of low-level proteins, which may be more important biomarkers.

Salivary biomarkers are miniscule and measured in (picograms), detection of which can only be achieved by techniques which are both sensitive and specific enough to discriminate between them [49]. Technological advances in diagnostic detection methods (next generation sequencing, mass spectrometry, genome wide association studies and other screening techniques) have paralleled the demand for improved diagnostic test accuracy of salivary genomic and proteomic biomarkers, thereby conferring distinct advantages for saliva in the diagnosis and monitoring of diseases such as oral cancer and precancer.

Salivary exosomes, which are nano-sized salivary biomarkers equipped with all the molecular cargo from the parent cells, have become increasingly detectable due to their stability in the circulation and bodily fluids. They have been extensively explored as diagnostic tools for local and systemic diseases [50].

2.1 Salivary exosome physiology

Fusion of nano-sized (30–100 nm in diameter) multivesicular bodies (MVB) [51] derived from the endocytic pathway with plasma membrane was discovered over 30 years ago [52]. Johnstone et al., detected the release of small vesicles into extracellular spaces by reticulocyte MVB’s and observed that their enzymatic activity mirrored that of the cell culture from which they were shed [52]. These bodies (exosomes), originally believed to be involved in waste disposal, due to their resistance to degradation by lysozymes, is now more understood to subserve vital biological functions when released as extracellular vesicles [53]. These functions are influenced/determined by their target cell with which they interact and include cellular communication and homeostasis, immune control (they contain IgA), RNA processing and transport of drugs [53, 54, 55].

Exosomal release (after formation of intraluminal vesicles), can be compared to the reversal of the endocytosis process, permits their evaluation in the extracellular body fluid environments [56]. The exocytotic release of exosomes into the extracellular domain, reveals that they naturally contain key molecular components derived from the parent cell relating to membrane transport, lipid metabolism and extracellular matrix formation [21, 57]. In addition, cytoplasmic nucleic acid contents such as mRNA and microRNA have been found in exosomes [21, 58, 59]. Considering the important contents as highlighted here, exosomes have been identified to play crucial roles in cell-to-cell communication [21]. All exosomes regardless of their origin possess both shared conserved and cell-specific proteins. Emerging knowledge has associated exosomes with the development of physiological and pathological perturbations [60, 61]. For instance, cancer exosomes have been found to be capable of a range of tumor-promoting activities, such as immunomodulation, development of pre-metastatic niches, as well as dysregulation of angiogenesis [62, 63, 64]. Furthermore, cancer exosomes are vital indicators of potentially malignant events in the tumor microenvironment and may exhibit pheno-genomic perturbation biomarkers of cancer [65, 66].

2.2 Diagnostic benefits of exosomes in body fluids

Due to exosomal release into the extracellular compartment, they are abundantly found in most body fluids such as cerebrospinal fluids, blood, breast milk, plasma, urine, serum, saliva, amniotic fluid, semen and ascetic fluid [21, 52, 53, 54, 55]. Exosomes are highly suitable substrates for biomarker signature discovery. Because of the content-protective packaging of their rich cargo (by lipid membranes) from extracellular lytic enzymes, and significantly lower complexity of its contents in comparison to whole tissue analysis [53].

Analyzing exosomal shuttle RNA (esRNA) in maternal blood, has been proposed as a potential surrogate prenatal diagnostic tool, to avoid risky invasive procedures such as chorionic villus sampling and amniocentesis [54]. This could potentially lower the risk of surgical injuries and miscarriages. Even though, cell free fetal DNA (cffDNA) has been previously used for the prenatal diagnostic purposes, the low content of fetal cffDNA has reduced the accuracy of this method [54, 56]. Information about cancer risk and genetic disease predisposition can be potentially gleaned from exosomal esRNA content analysis.

The use of novel liquid biopsy-based cancer diagnostic tools has significantly improved the precision and efficacy of individualized medicine, particularly in resource-limited settings [11]. Exosomes are capable of providing robust molecular tumor information about cells of origin, are retrievable from easily accessible body fluids; and hence are highly useful for early detection and follow-up of cancer [55].

2.3 Salivary exosomes and oral cancer

Exosomes have been successfully isolated from saliva [19, 20, 21, 22, 23, 24, 25]; and the presence of lipids, proteins and nucleic acids in exosomes, makes salivary exosomes attractive substrates for omics analysis (a.k.a Salivaomics) [57, 58, 59, 60]. It has become emergent, that salivary constituents (e.g. mRNAs, proteins, miRNAs, microbes and metabolites such as lipids) may be detected in exosomes and be used as biomarkers for diseases (both local and distal) [54]. Structural, proteomics and transcriptomics analysis of salivary exosomes has been successfully carried out [61, 62, 63, 64, 65]; and salivary exosomes are fast becoming key tools in cancer biomarker theranostics. Exosomes have been revealed as valuable indicators of the micro-environments and perpetrators of cancer intercellular communication [25].

The mean diameter and protein content are used as the basis upon which exosomes isolated from saliva are structurally subdivided into two types (I and II which are ca. 85 nm and 40 nm in diameter, respectively) [20]. Since epithelial barriers between blood vessels and salivary gland structures can be crossed by exosomes [66, 67], they have become important tools for essential transport of key signatures between blood and saliva (which is believed to be an ultrafiltrate of blood) [67, 68, 69]. Important molecular information may be exchanged via transudation, ultrafiltration or selective transport based on their size or presence of transporter molecules [67].

Of all the head and neck cancers, oral squamous cell carcinoma is the most prevalent, often diagnosed in advanced stage and is associated with a low survival and poor prognosis. Early detection of oral cancer is a key goal in epidemiological cancer control and successful management thereof. Using salivary exosomes for identification of oral cancer biomarkers is potentially a highly sensitive, cost-effective and non-invasive point-of-care technique for detecting oral cancer that may be subclinical or missed by routine histological diagnostic approaches [70]. Evaluation of these vesicles shed by cancer cells via multivesicular bodies (MVB’s) into saliva is a viable approach for biomarker detection in oral cancer (Figure 2). Salivary exosomes have played key roles in diagnosis of systemic diseases [51] and have been key player in the molecular characterization of cancer [24]. Due to its reduced complexity as compared to other body fluids, exosomes are reliable tools for early diagnosis of oral cancer and its use as a diagnostic tool may significantly improve cancer survival rates [24].

Figure 2.

Pathways for escape of exosomes into saliva. Oral squamous carcinoma cells (OSCC) may release exosomes into saliva, either by fusion of the multivesicular body (MVB) with the plasma membrane (MF) or by plasma membrane rupture (MR) and direct release through endosomal membrane.

Due to its nano-scaled structure, human salivary exosomes have been identified as potential carriers for non-invasive delivery of cancer biomarkers [22]. For example, electrochemical sensing methods, such as electric field-induced release and measurement (EFIRM) approach, has been used to improve the field of salivary liquid biopsy [23]. Not least, salivary exosomes have been used to enhance the detection of human papilloma virus (HPV) positive oropharyngeal cancers [26].

The critical role of tumor-derived exosome in cancer in largely due to the presence of tumor-specific signatures within its functional cargos, which includes proteins, miRNA and mRNA (Table 1).

BiomarkerTypeSampleMethodsReferences
A2M, HPA, MUC5B, LGALS3BP,
IGHA1, PIP, PKM1/M2, GAPDH
ProteinsalivaMass spectrometry analysis and proteomics data analysisWinck et al. [71]
miRNA-21miRNAOSCC cell linemiRNA sequencingLi et al. [72]
miR-1246miRNAOSCC cell lineMicroRNA microarraySakha et al. [73]
miR-200c-3pmiRNAOSCC cell lineintegrated microarrayKawakubo-Yasukochi et al. [74]
miR-34a-5pmiRNAOSCC cell linemiRNA sequencingLi et al. [75]
PF4V1, CXCL7, F13A1, and ApoA1proteinserumRT-PCRLi et al. [76]
miR-101-3pmiRNAOSCC cell lineMicroarray analysisXie et al. [77]
miR‑382‑5pmiRNAOSCC cell lineRT-PCRSun et al. [78]
miR-24-3pmiRNAsalivaRT-PCRHe et al. [79]
miR-21-5pmiRNAOSCC cell lineRT-PCRChen et al. [80]
miR-155miRNAOSCC cell lineRT-PCR.Kirave et al. [81]

Table 1.

Exosome biomarker for oral cancers.

The significant physiological interaction and overlap of the blood and salivary proteome (ca. 20–30%) makes exosomal protein biomarkers attractive and many cancer-related exosomal proteins have been identified from oral cancer [23, 82, 83]. Potential proteomic biomarkers of oral cancer such as CFB, CD59, A1BG, M2b, CAT, MRP14, PFN, M2BP, ADA, S100, CFL1, IGHG, TF, IL-1B and IL-8S have been identified from whole saliva [84, 85, 86, 87, 88]. However, protein biomarkers such as MUC5B, A2M, LGALS3BP, HPa, GAPDH, IGHA1, PIP and PKM1/M2 have been specifically identified to be exosomal protein biomarkers of oral cancer with a classification accuracy of 90% [80, 89]. Zlotogorski-Hurvitz et al. (2016), identified CD9/−81 downregulation and CD 63 upregulation in exosomes as early diagnostic protein biomarkers of oral cancer [90]. Furthermore, salivary exosomes isolated from HPV-positive oropharyngeal cancer cell lines has been found to underexpress cyclin D1 and p53 and overexpress p16, T-cell inhibitory protein PTPN11 and E6/E7 proteins [91].

Via their interaction with mRNA, micro RNA’s (miRNA) are involved in a number of physiological and disease processes (when there is aberrant expression). MiRNAs are small non-coding RNAs that mediate destabilization and/or translational repression of target messenger RNA (mRNA) molecules thereby reducing final protein output. Exosomes are a rich source of miRNA’s, provides a vehicle for cell to cell transporting to alter cellular functions as well as offer protection in the extracellular environment. Exosomal miRNAs have been investigated as candidate screening tools (miR-24-3p) [92], in chemoresistance (miR-21) [81], regulating tumor progression (miR-34-5p) [93] and miR-342–3p and miR-1246 [93] and miR-382-5p [78].

In oral cancer, the intercellular transfer of molecules (such as miRNA’s) by cancer associated fibroblast influences the tumor microenvironment. miR-21 represents one of the most abundant miRs transported within EV cargos secreted by oral cancer cells and is a well-established oncogenic miR whose major targets include the tumor suppressors. Its exosomal hideout contributes towards chemoresistance due to the camouflage provided by its vehicle during intercellular transfer of the oncogenic miR. It is thus also an important chemotherapeutic precision target in cancers [81]. Li et al. [94], in a study of 108 patients with OSCC, observed that tumor exosomal miR 21 was upregulated in hypoxic cancer cells as well as internalized by normoxic cells. Exosomal miR-34-5p transfer between CAF’s and neighboring OSCC cells played an important role in regulating tumor progression. Sakha et al. [73], demonstrated that effect that intercellular transfer of exosomal oncogenic miRNA’s (miR-342–3p and miR-1246) could be delivered were evaluated for their role in could have on cancer development and progression by influencing cell motility and invasiveness [73]. Exosomal miR-382-5p in cancer-associated fibroblast (CAF) mediated OSSC migration and invasion by evaluation of tissue samples from 47 patients who had OSSC tumor resection. The results showed that CAF’s transfer miR-382-5p associated with migration and invasion [78]. The expression of exosomal miR-24-3p was found to be higher in salivary exosomes from OSCC patients compared to healthy controls. The AUC for miR-24-3p was 0.738 and could significantly distinguish OSCC patients from normal individuals with 64.4% sensitivity and 80% specificity in 49 patients with OSSC.

The functional cargos which include mRNA has been considered as potential biomarker in the diagnosis and monitoring and treatment of cancer. Valadi et al. first described the presence of mRNAs in exosomes in 2007 [95]. Subsequently, studies have shown the transfer of bioactive mRNAs (tumor suppressor genes or oncogenes) from a malignant cell to a normal cell led to a change in the phenotype and malignant transformation of the normal cell [95, 96, 97].

Few studies have identified mRNA in saliva of oral cancer patients. Li et al. identified potential mRNA biomarker which include IL8, SAT, DUSP, IL1B, OAZ1, H3F3A and S100P, in saliva from oral cancer patients [98]. The study showed that the combination of these biomarkers was highly sensitive and specific in differentiating between OSSC and healthy [98]. An in vitro study showed that oral squamous cell carcinoma cell line (PCI-13, UMSCC47) triggered significant increase expression level of IGFBP-3 mRNA and VEGF mRNA in recipient cells [99].

Even though exosomes have been widely employed to investigate head and neck squamous cell carcinomas [100, 101, 102, 103], unraveling the biologic mechanisms and application of salivary tumor -derived exosomes is still an evolving science [67]. This emergent field of saliva-exosomics warrants further investigation.

Advertisement

3. Conclusions

Salivary exosomes provide viable, consistent and stable sources of cancer biomarkers. The scope of its utility as well as understanding the molecular mechanisms which underpins it, requires further investigation. Future studies refining the methodology for extracellular vesicle isolation and cleansing presents the greatest challenge that is needed to overcome the restrictions to exploring the full scope of salivary exosomes in systemic diseases including oral cancer.

Advertisement

Acknowledgments

H.A.A thanks the South African Medical Research Council (SAMRC) for a mid-career scientist and Self-initiated research grant; and the South African National Research Foundation (NRF) for Research Development Grant (RDG) for rated researchers. The authors thank the University Research committee (URC) of the University of Cape Town for funding this book chapter.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Adeola HA, Soyele OO, Adefuye AO, Jimoh SA, Butali A. Omics-based molecular techniques in oral pathology centered cancer: Prospect and challenges in Africa. Cancer Cell International. 2017;17:6
  2. 2. Olson MV. The human genome project. Proceedings of the National Academy of Sciences of the United States of America. 1993;90:4338-434
  3. 3. Collins FS, Morgan M, Patrinos A. The human genome project: Lessons from large-scale biology. Science. 2003;300:286-29
  4. 4. Green ED, Watson JD, Collins FS. Human genome project: Twenty-five years of big biology. Nature. 2015;526:29-3
  5. 5. Hood L, Rowen L. The human genome project: Big science transforms biology and medicine. Genome Medicine. 2013;5:7
  6. 6. Wilson BJ, Nicholls SG. The human genome project, and recent advances in personalized genomics. Risk Manag Healthc Policy. 2015;8:9-2
  7. 7. Cho, J., Ahn, S., Son, D. S., Kim, N. K., et al., Bridging genomics and phenomics of gastric carcinoma. International journal of cancer. Journal international du cancer 2019, 145, 2407-2417
  8. 8. Davatzikos, C., Rathore, S., Bakas, S., Pati, S., et al., Cancer imaging phenomics toolkit: Quantitative imaging analytics for precision diagnostics and predictive modeling of clinical outcome. J Med Imaging (Bellingham) 2018, 5, 011018
  9. 9. Harder N, Athelogou M, Hessel H, Brieu N, et al. Tissue phenomics for prognostic biomarker discovery in low- and intermediate-risk prostate cancer. Scientific Reports. 2018;8:447
  10. 10. Bai Y, Zhao H. Liquid biopsy in tumors: Opportunities and challenges. Annals of translational medicine. 2018;6:S8
  11. 11. Temilola DO, Wium M, Coulidiati TH, Adeola HA, et al. The Prospect and challenges to the flow of liquid biopsy in Africa. Cell. 2019;8
  12. 12. Jacobson, R. A., Munding, E., Hayden, D. M., Levy, M., et al., Evolving clinical utility of liquid biopsy in gastrointestinal cancers. Cancers 2019, 11
  13. 13. Palmirotta, R., Lovero, D., Cafforio, P., Felici, C., et al., Liquid biopsy of cancer: A multimodal diagnostic tool in clinical oncology. Therapeutic advances in medical oncology 2018, 10, 1758835918794630
  14. 14. Wu J, Hu S, Zhang L, Xin J, et al. Tumor circulome in the liquid biopsies for cancer diagnosis and prognosis. Theranostics. 2020;10:4544-455
  15. 15. Chen L, Chen Y, Feng YL, Zhu Y, et al. Tumor circulome in the liquid biopsies for digestive tract cancer diagnosis and prognosis. World Journal of Clinical Cases. 2020;8:2066-208
  16. 16. Bracht JWP. Mayo-de-Las-casas, C., Berenguer, J., Karachaliou, N., Rosell, R., correction to: The present and future of liquid biopsies in non-small cell lung cancer: Combining four biosources for diagnosis, prognosis, prediction, and disease monitoring. Current Oncology Reports. 2020;22:5
  17. 17. De Rubis G, Rajeev Krishnan S, Bebawy M. Liquid biopsies in cancer diagnosis, monitoring, and prognosis. Trends in Pharmacological Sciences. 2019;40:172-18
  18. 18. Saarenheimo J, Eigeliene N, Andersen H, Tiirola M, Jekunen A. The value of liquid biopsies for guiding therapy decisions in non-small cell lung cancer. Frontiers in Oncology. 2019;9:12
  19. 19. Nair S, Tang KD, Kenny L, Punyadeera C. Salivary exosomes as potential biomarkers in cancer. Oral Oncology. 2018;84:31-4
  20. 20. Cheshmi B, Cheshomi H. Salivary exosomes: Properties, medical applications, and isolation methods. Molecular Biology Reports. 202
  21. 21. Zlotogorski-Hurvitz A, Dayan D, Chaushu G, Korvala J, et al. Human saliva-derived exosomes: Comparing methods of isolation. The Journal of Histochemistry and Cytochemistry. 2015;63:181-18
  22. 22. Han Y, Jia L, Zheng Y, Li W. Salivary exosomes: Emerging roles in systemic disease. International Journal of Biological Sciences. 2018;14:633-64
  23. 23. Cheng J, Nonaka T, Wong DTW. Salivary exosomes as nanocarriers for cancer biomarker delivery. Materials. 2019;12
  24. 24. Nonaka T, Wong DTW. Saliva-exosomics in cancer: Molecular characterization of cancer-derived exosomes in Saliva. Enzyme. 2017;42:125-15
  25. 25. Zhan C, Yang X, Yin X, Hou J. Exosomes and other extracellular vesicles in oral and salivary gland cancers. Oral Diseases. 2020;26:865-87
  26. 26. Wang ZY, Li F, Rufo J, Chen CY, et al. Acoustofluidic salivary Exosome isolation a liquid biopsy compatible approach for human papillomavirus-associated oropharyngeal cancer detection. Journal of Molecular Diagnostics. 2020;22:50-5
  27. 27. A review of saliva: normal composition, flow, and function. Humphrey SP, Williamson RT J Prosthet Dent. 2001 Feb; 85(2):162-9.
  28. 28. Structure and biosynthesis of human salivary mucins. Zalewska A, Zwierz K. Zółkowski K, Gindzieński A Acta Biochim Pol. 2000;47(4):1067-107
  29. 29. Saliva and dental health. Clinical implications of saliva: report of a consensus meeting. Edgar WM Br Dent J. 1990 Aug 11-25; 169(3-4):96-8
  30. 30. Tiwari M. Science behind human saliva. Journal of natural science, biology, and medicine. 2011;2(1):53-5
  31. 31. Cove-Smith R, Defective Saliva MA. Proceedings of the Royal Society of Medicine. 1935;29:126-12
  32. 32. Farnaud SJ, Kosti O, Getting SJ, Renshaw D. Saliva: Physiology and diagnostic potential in health and disease. The Scientific World Journal. 2010;10:434-45
  33. 33. Humphrey SP, Williamson RT. A review of saliva: Normal composition, flow, and function. The Journal of prosthetic dentistry. 2001 Feb 1;85(2):162-16
  34. 34. Holsinger F, Bui D. Salivary Gland Disorders. Berlin, Germany: Springer; 200
  35. 35. Pharmacokinetic principles of drug distribution in saliva. Jusko WJ, Milsap RL. Annals of the New York Academy of Sciences. 1993 Sep 20;694:36-4
  36. 36. The application of saliva, sweat and tear fluid for diagnostic purposes. Haeckel R, Hänecke P Ann Biol Clin (Paris). 1993;51(10-11):903-91
  37. 37. Nagarathinam AE, Kumar TD, Kumar AR, Vasanthira K, Lakshmi RS, Jayant VS. Salivary Urea and Creatinine as a Diagnostic Marker of Chronic Kidney Disease – Review. (IOSR-JDMS). 2017;16:95-10
  38. 38. Schneyer LH, Schneyer CA. Secretion of Saliva. Advances in Oral Biology. 1964;1:1-3
  39. 39. Leung SW. A demonstration of the importance of bicarbonate as a salivary buffer. Journal of Dental Research. 1951;30:403-41
  40. 40. Lee JM, Garon E, Wong DT. Salivary diagnostics. Orthodontics & Craniofacial Research. 2009;12:206-21
  41. 41. Hundt S, Haug U, Brenner H. Blood markers for early detection of colorectal cancer: A systematic review. Cancer Epidemiology and Prevention Biomarkers. 2007 Oct 1;16(10):1935-195
  42. 42. De Seny D, Fillet M, Meuwis MA, Geurts P, Lutteri L, Ribbens C, et al. Discovery of new rheumatoid arthritis biomarkers using the surface-enhanced laser desorption/ionization time-of-flight mass spectrometry ProteinChip approach. Arthritis and Rheumatism. 2005 Dec;52(12):3801-381
  43. 43. Hodgetts A, Levin M, Kroll JS, Langford PR. Biomarker discovery in infectious diseases using SELDI. Future Microbiology. 2007;2(1):35-4
  44. 44. Suzuki M, Ross GF, Wiers K, et al. Identification of a urinary proteomic signature for lupus nephritis in children. Pediatric Nephrology. 2007;22(12):2047-205
  45. 45. C. S. Buhimschi, V. Bhandari, B. D. Hamar et al., “Proteomic profiling of the amniotic fluid to detect inflammation, infection, and neonatal sepsis,” PLoS Medicine, vol. 4, no. 1, article e18, 2007
  46. 46. De Bock M, De Seny D, Meuwis MA, Chapelle JP, Louis E. Malaise M. Fillet M. Challenges for biomarker discovery in body fluids using SELDI-TOF-MS. Journal of Biomedicine and Biotechnology: Merville MP; 2009 Dec 6. p. 201
  47. 47. Malamud D. PhDa,b and Isaac R. Rodriguez-Chavez, PhDc, Saliva as a diagnostic fluid. Published in final edited form as: Dent Clin North Am. 2011 January;55(1):159-17
  48. 48. Campo J, Perea MA, Del Romero J, Cano J, Hernando V, Bascones A. Oral transmission of HIV, reality or fiction? An update. Oral Diseases. 2006;12:219-22
  49. 49. Wei F, Yang J, Wong DT. Detection of exosomal biomarker by electric field-induced release and measurement (EFIRM). Biosensors & Bioelectronics. 2013;44:115-12
  50. 50. Wei F, Lin CC, Joon A, Feng Z, Troche G, Lira ME, et al. Noninvasive saliva-based EGFR gene mutation detection in patients with lung cancer. American Journal of Respiratory and Critical Care Medicine. 2014;190:1117-112
  51. 51. Han Y, Jia L, Zheng Y, Li W. Salivary exosomes: Emerging roles in systemic disease. International Journal of Biological Sciences. 2018;14:633-64
  52. 52. Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cellular and molecular life sciences : CMLS. 2018;75:193-20
  53. 53. Baixauli F, Lopez-Otin C, Mittelbrunn M. Exosomes and autophagy: Coordinated mechanisms for the maintenance of cellular fitness. Frontiers in Immunology. 2014;5:40
  54. 54. Lai R, Yeo R, Tan K, Lim S. Exosomes for drug delivery – A novel application for the mesenchymal stem cell. Biotech Adv. 2013;31:543-55
  55. 55. Zheng X, CHEN F, ZHANG J, ZHANG Q , Exosome Analysis LINJ. A promising biomarker system with special attention to saliva. The Journal of Membrane Biology. 2014;247:1129-113
  56. 56. Johnstone RM. Revisiting the road to the discovery of exosomes. Blood Cells, Molecules & Diseases. 2005;34:214-21
  57. 57. Simons M, Raposo G. Exosomes - vesicular carriers for intercellular communication. Current Opinion in Cell Biology. 2009;21:575-58
  58. 58. Valadi H, Ekstrom K, Bossios A, Sjostrand M, et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology. 2007;9:654-U67
  59. 59. Mathivanan S, Fahner CJ, Reid GE, Simpson RJ. ExoCarta 2012: Database of exosomal proteins, RNA and lipids. Nucleic Acids Research. 2012;40:D1241-D124
  60. 60. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles. and friends. The Journal of cell biology. 2013;200:373-38
  61. 61. Record M, Carayon K, Poirot M, Silvente-Poirot S. Exosomes as new vesicular lipid transporters involved in cell-cell communication and various pathophysiologies. Biochimica et Biophysica Acta-Molecular and Cell Biology of Lipids. 2014;1841:108-12
  62. 62. Becker A, Thakur BK, Weiss JM, Kim HS, et al. Extracellular vesicles in cancer: Cell-to-cell mediators of metastasis. Cancer Cell. 2016;30:836-84
  63. 63. Costa-Silva, B., Aiello, N. M., Ocean, A. J., Singh, S., et al., Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol 2015, 17, 816−+
  64. 64. Hoshino, A., Costa-Silva, B., Shen, T. L., Rodrigues, G., et al., Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329−+
  65. 65. Kharaziha P, Ceder S, Li Q , Panaretakis T. Tumor cell-derived exosomes: A message in a bottle. Bba-Rev Cancer. 2012;1826:103-11
  66. 66. Guo Q , Jiang C. Delivery strategies for macromolecular drugs in cancer therapy. Acta Pharmaceutica Sinica B. 2020;10:979-98
  67. 67. Chiappin S, Antonelli G, Gatti R, De Palo EF. Saliva specimen: A new laboratory tool for diagnostic and basic investigation. Clinica Chimica Acta. 2007;383:30-4
  68. 68. Yang, J., Wei, F., Schafer, C., Wong, D. T., Detection of tumor cell-specific mRNA and protein in exosome-like microvesicles from blood and saliva. PLoS One 2014, 9, e110641
  69. 69. Zhou HX, Xu WR, Qian H, Yin Q , et al. Circulating RNA as a novel tumor marker: An in vitro study of the origins and characteristics of extracellular RNA. Cancer Letters. 2008;259:50-6
  70. 70. Aro K, Kaczor-Urbanowicz K, Carreras-Presas CM. Salivaomics in oral cancer. Current Opinion in Otolaryngology & Head and Neck Surgery. 2019;27:91-9
  71. 71. Winck FV, Prado Ribeiro AC, Ramos Domingues R, Ling LY, Riaño-Pachón DM, Rivera C, et al. Insights into immune responses in oral cancer through proteomic analysis of saliva and salivary extracellular vesicles. Scientific Reports. 2015;5:1630
  72. 72. 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 research. 2016 Apr 1;76(7):1770-178
  73. 73. 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. Scientific Reports. 2016 Dec 8;6:3875
  74. 74. Kawakubo-Yasukochi T, Morioka M, Hazekawa M, Yasukochi A, Nishinakagawa T, Ono K. miR-200c-3p spreads invasive capacity in human oral squamous cell carcinoma microenvironment. Molecular Carcinogenesis. 201
  75. 75. 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 Oct 1;36:209-22
  76. 76. Li C, Zhou Y, Liu J, Su X, Qin H, Huang S, et al. Potential markers from serum-purified exosomes for detecting oral squamous cell carcinoma metastasis. Cancer Epidemiology and Prevention Biomarkers. 2019 Oct 1;28(10):1668-168
  77. 77. 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. Molecular and Cellular Biochemistry. 2019 Aug 15;458(1-2):11-2
  78. 78. Sun LP, Xu K, Cui J, Yuan DY, Zou B, Li J, et al. Cancer-associated fibroblast-derived exosomal miR-382-5p promotes the migration and invasion of oral squamous cell carcinoma. Oncology reports. 2019 Oct 1;42(4):1319-132
  79. 79. He L, Ping F, Fan Z, Zhang C, Deng M, Cheng B, Xia J. Salivary exosomal miR-24-3p serves as a potential detective biomarker for oral squamous cell carcinoma screening. Biomedicine & Pharmacotherapy. 2020 Jan 1;121:109553
  80. 80. Chen JH, Wu AT, Bamodu OA, Yadav VK, Chao TY, Tzeng YM, et al. Ovatodiolide suppresses oral cancer malignancy by down-regulating exosomal mir-21/STAT3/β-catenin cargo and preventing oncogenic transformation of normal gingival fibroblasts. Cancers. 2020 Jan;12(1):5
  81. 81. Kirave P, Gondaliya P, Kulkarni B, Rawal R, Garg R, Jain A, et al. Exosome mediated miR-155 delivery confers cisplatin chemoresistance in oral cancer cells via epithelial-mesenchymal transition. Oncotarget. 2020;11(13):1157-1171 https://doi.org/10.18632/oncotarget.27531
  82. 82. Yeh CK, Christodoulides NJ, Floriano PN, Miller CS, et al. Current development of saliva/oral fluid-based diagnostics. Texas Dental Journal. 2010;127:651-66
  83. 83. Yan W, Apweiler R, Balgley BM, Boontheung P, et al. Systematic comparison of the human saliva and plasma proteomes. Proteomics. Clinical Applications. 2009;3:116-13
  84. 84. Ohshiro K, Rosenthal DI, Koomen JM, Streckfus CF, et al. Pre-analytic saliva processing affect proteomic results and biomarker screening of head and neck squamous carcinoma. International Journal of Oncology. 2007;30:743-74
  85. 85. Hu S, Arellano M, Boontheung P, Wang JH, et al. Salivary proteomics for Oral cancer biomarker discovery. Clinical Cancer Research. 2008;14:6246-625
  86. 86. Dowling P, Wormald R, Meleady P, Henry M, et al. Analysis of the saliva proteome from patients with head and neck squamous cell carcinoma reveals differences in abundance levels of proteins associated with tumour progression and metastasis. Journal of Proteomics. 2008;71:168-17
  87. 87. Rai B, Kaur J, Jacobs R, Anand SC. Adenosine deaminase in saliva as a diagnostic marker of squamous cell carcinoma of tongue. Clin Oral Invest. 2011;15:347-34
  88. 88. Elashoff D, Zhou H, Reiss J, Wang JH, et al. Prevalidation of salivary biomarkers for Oral cancer detection. Cancer Epidem Biomar. 2012;21:664-67
  89. 89. Winck FV, Prado Ribeiro AC, Ramos Domingues R, Ling LY, Riaño-Pachón DM, Rivera C, et al. Insights into immune responses in oral cancer through proteomic analysis of saliva and salivary extracellular vesicles. Scientific Reports. 2015;5:1630
  90. 90. 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. Journal of Cancer Research and Clinical Oncology. 2016;142:101-11
  91. 91. Ludwig S, Sharma P, Theodoraki MN, Pietrowska M, et al. Molecular and functional profiles of exosomes from HPV(+) and HPV(−) head and neck cancer cell lines. Frontiers in Oncology. 2018;8
  92. 92. He L, Ping F, Fan ZN, Zhang C, et al. Salivary exosomal miR-24-3p serves as a potential detective biomarker for oral squamous cell carcinoma screening. Biomedicine & Pharmacotherapy. 2020;121
  93. 93. 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. Scientific Reports. 2016;6
  94. 94. Li L, Li C, Wang S, Wang Z, et al. Exosomes derived from hypoxic Oral squamous cell carcinoma cells deliver miR-21 to normoxic cells to elicit a Prometastatic phenotype. Cancer Research. 2016;76:1770-178
  95. 95. Valadi H, Ekstrom K, Bossios A, et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology. 2007;9:654-65
  96. 96. Skog J, Wurdinger T, van Rijn S, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nature Cell Biology. 2008;10:1470-147
  97. 97. Hong BS, Cho JH, Kim H, et al. Colorectal cancer cell-derived microvesicles are enriched in cell cycle-related mRNAs that promote proliferation of endothelial cells. BMC Genomics. 2009;10:55
  98. 98. Li Y, John MA, Zhou X, Kim Y, Sinha U, Jordan RC, et al. Salivary transcriptome diagnostics for oral cancer detection. Clinical Cancer Research. 2004 Dec 15;10(24):8442-845
  99. 99. Ludwig N, Yerneni SS, Razzo BM, Whiteside TL. Exosomes from hnscc promote angiogenesis through reprogramming of endothelial cells. Molecular Cancer Research. 2018;24:24. DOI: 10.1158/1541-7786.MCR-18-035
  100. 100. Xiao C, Song F, Zheng YL, Lv J, et al. Exosomes in head and neck squamous cell carcinoma. Frontiers in Oncology. 2019;9:89
  101. 101. Ludwig N, Gillespie DG, Reichert TE, Jackson EK, Whiteside TL. Purine metabolites in tumor-derived exosomes may facilitate immune escape of head and neck squamous cell carcinoma. Cancers. 2020;12
  102. 102. Jelonek K, Wojakowska A, Marczak L, Muer A, et al. Ionizing radiation affects protein composition of exosomes secreted in vitro from head and neck squamous cell carcinoma. Acta Biochimica Polonica. 2015;62:265-27
  103. 103. Ebnoether E, Muller L. Diagnostic and therapeutic applications of exosomes in cancer with a special focus on head and neck squamous cell carcinoma (HNSCC). International Journal of Molecular Sciences. 2020;21

Written By

Henry Ademola Adeola, Haly Holmes and Dada Oluwaseyi Temilola

Submitted: 07 September 2020 Reviewed: 24 September 2020 Published: 20 November 2020