2,411
Views
11
CrossRef citations to date
0
Altmetric
Review Article

Exosomes in head and neck cancer. Updating and revisiting

ORCID Icon, , , ORCID Icon, , & ORCID Icon show all
Pages 1641-1651 | Received 04 Jul 2019, Accepted 26 Aug 2019, Published online: 08 Sep 2019

Abstract

Exosomes have gone from being considered simple containers of intracellular waste substances to be considered important carriers of cellular signals. Its broad capacity to promote tumour growth, both in situ and metastatic, has greatly intensified scientific research on them. In the same way and depending on its content, its tumour suppressive properties have opened a window of light and hope in the fight against cancer. In the present review we try to gather in a simple and understandable way the most relevant knowledge to date on the role of exosomes in oral squamous cell carcinoma, helping to understand their process of formation, release and activity on the tumour microenvironment.

Introduction

The most frequent type of head and neck cancer is squamous cell carcinoma (SCC), which accounts for more than 90% of cancers of the head and neckCitation1. One of the primary locations where this disease manifests is in the oral cavity; however, it occurs frequently in the pharynx and larynx as wellCitation2. Oral cancer is the sixth most commonly diagnosed cancer in the world and was responsible for 145, 000 deaths in 2012, 77% of which happened in regions with a poor economic developmentCitation3. Oral squamous cell carcinoma represents 90% of all oral cancer varieties with a very common lymph node involvementCitation4. The five-year survival rate for patients with localised oral squamous cell carcinoma is higher than 80%, however this rate drops dramatically to 40% when the lymph nodes are involved, and to 20% for patients with distant metastasisCitation5.

Recently, numerous studies have implicated the HPV infection as a risk factor for developing oral squamous cell carcinoma, nonetheless the best described risk factors continue to be tobacco and alcoholCitation6. As we have stated, cancer metastasis is related to bad prognosis and it is very frequent in these kinds of tumours, nearly 50% of these patients suffer from metastasisCitation7.

In general, the extracellular vesicle population is comprised of different types of small vesicles which include microvesicles (MVs) and exosomes. Both exosomes and MVs are membrane-bound vesicles, distinguished by their biogenesis and biophysical properties, such as size and surface protein markers, and by their biological roles, including cell-cell communication, the maintenance of normal physiological processes and disease pathologyCitation8. The MISEV2018 position paper from the International Society for Extracellular Vesicles provided clarity on this, as well as serving as a practical guide when carrying out exosome studiesCitation9.

Exosomes are small nanovesicles from 50–150 nm in diameter which are released into the extracellular environment, . These are even released in biological fluids such as saliva, urine and bloodCitation10. Exosomes do not derive from the shedding of plasma membrane fragments, but are secreted by peculiar structures referred to as multivesicular bodies (MVBs). MVBs are the result of a cascade of multifusion phenomena among internal vesicles, including early and late endosomes, lysosomes, and other structures that vary depending on the cellular sourceCitation11.

Figure 1. The diagram reveals the different steps of the formation of exosomes and their release from the cells of an oral squamous cell carcinoma. The intracellular traffic and its release to the extracellular medium is appreciated. In the upper image, the main components of an exosome derived from an OSCC are reflected both at the level of its lipid membrane and inside it.

Figure 1. The diagram reveals the different steps of the formation of exosomes and their release from the cells of an oral squamous cell carcinoma. The intracellular traffic and its release to the extracellular medium is appreciated. In the upper image, the main components of an exosome derived from an OSCC are reflected both at the level of its lipid membrane and inside it.

Exosomes were initially considered as products of a pathway which was used to release undesired material from cellsCitation12 and recent data support the role of exosomes, both in maintaining normal body homeostasis and in the pathophysiology of many diseases. Exosomes perform an important range of extracellular functions which include interactions with the cellular microenvironment through immunologic mediation, morphogen signalling, cell recruitment, and the horizontal transfer of genetic materialCitation13.

Many cell types such as dendritic cells, B cells, T cells, mast cells, epithelial cells, and tumour cells can produce exosomesCitation14. Tumour cells secrete large amounts of exosomes that promote tumour progression through communication between the tumour and the surrounding stromal tissue, the activation of the proliferative and angiogenic pathways, and the initiation of the pre-metastatic siteCitation15–19. For these reasons, exosomes form an important component of the tumour microenvironment and are considered to be one of the main contributors to tumour progression and metastasisCitation13. Exosomes’ external part is mainly comprised of lipids, while the molecules found inside, whose biological activity has been studied recently, include different proteins and RNA fragments.

As we have already stated, the main components of exosomes are lipids. Exosomes are enriched in cholesterol, diglycerides, glycerophospholipids, phospholipids, and sphingolipids or glycosylceramidesCitation20. Besides these lipids, bioactive lipids, such as prostaglandins and leukotrienes, and enzymes activated in lipid metabolism, including phospholipase C, are also found in exosomesCitation21. Exosomes that contain high levels of prostaglandin PGE2 are involved in tumour immune evasion and in the promotion of tumour growthCitation22.

Unlike MVs, exosomes do not initially carry nuclear DNA but they may contain mitochondrial DNACitation23. Valadi et al. was the first author to demonstrate the presence of mRNA and microRNA in exosome preparationsCitation24. MicroRNAs are small, ranging from 22 to 25 nucleotides in length. They are non-coding RNAs with the ability to interfere with RNA post-transcriptionally by binding to the 3′ untranslated region of their target mRNAs to degrade it, or suppress or stimulate translation. Their most common functions are stem cell differentiation, haematopoiesis, differentiation and organogenesis, exocytosis, and tumorigenesisCitation25.

More than 3400 mRNA molecules shuttled by exosomes and 2800 microRNA are reported in ExoCarta entries, however, this is only a small fraction of the large amount of RNA contained in the exosomes.

The tumour suppressor miRNAs (TS-mi RNAs) promote the expression of tumour suppressor genes avoiding the expression of oncogenesCitation26.

Exosome protein composition includes both an ubiquitary and a cell type-specific protein, and covers numerous biological functions. Most of the exosomal proteins which have been identified derive from the endocytic compartment or the plasma membrane of cellular source and only seldomly from other internal compartments such as Golgi, nucleus, endoplasmic reticulum, and mitochondria.

The most relevant protein functions carried out by exosomes in relation with the major biological function are: adhesion molecules, antigen presentation cytoskeletal/structural proteins, enzymes, heat shock proteins/chaperones, lipid raft proteins, membrane trafficking, transport and fusion, MVB formation, signal transduction, transcription/protein synthesis and transmembrane moleculesCitation27.

Exosomes constitutively express tetraspanins (CD63, CD9, CD81), endosomal and lysosomal markers (Rab5, LAMP), and heat shock proteins (HSP 70)Citation28. These nanovesicles are completely different to apoptotic microparticles which bear markers like CD31 or annexin VCitation29.

The tumour milieu is characteristically acidic as a consequence of the fermentative metabolism of glucose that results in massive accumulation of lactic acid within the cytoplasm. Tumour cells get rid of excessive protons through exchangers that are responsible for the extracellular acidification that selects cellular clones that are more apt at surviving in this challenging and culling environment. In vitro studies have recently pointed out that cancer acidity is a major determinant in inducing increased exosome release by human cancer cells, by showing that exosomal release was increased as the pH moved from 7.4 pH to the typical pH of cancer that is 6.5Citation30–32.

A very recent study by Patton et al. demonstrated that small EV and primarly exosomes were the most bioactive in promoting the survival of hypoxic pancreatic cancer cells and hypoxia‐inducible factor‐1α stabilisation was involved in heightened EV release under hypoxia and for their potency to promote hypoxic cell survivalCitation33.

Through an adapted ELISA test, which allows for the detection, characterisation and quantification of exosomes, it has been demonstrated that tumour patients have significantly increased plasmatic levels of exosomes expressing CAV1 compared with the plasma of healthy donorsCitation34 and even CD63Citation35.

A recent study has demonstrated that surgical treatment induced a dramatic reduction of the plasmatic levels of exosomes expressing CD63 as early as 1 week after resection. This first result appears to suggest that the tumour mass is responsible for the high levels of circulating exosomes detected in cancer patientsCitation36.

The discovery around 10 years ago that exosome contents can be transferred to another cell via fusion to create phenotypic alterations supports intensive research in this fieldCitation24.

Exosomes in the cancer process

Recent articles have shown that exosomes are present and involved in numerous phases of the cancer process.

It is possible to divide the aforementioned phases in a generic mannerCitation37: tumourigenesis, growth and development, creation of new blood vessels that feed the tumour, evasion of the immune response, development of resistance to chemotherapeutic agents and, finally, metastasis.

In situ tumourigenesis

Exosomes have been defined as promoters of tumour progressionCitation38. Despite the fact that there is abundant in vitro evidence demonstrating the exchange of information between tumour cells by exosomes, in 2015 it was demonstrated, by in vivo techniques using a high resolution image and the Cre-LoxP system, that the exosomes released by malignant tumour cells are taken up by less malignant tumour cells which are located within the same and within distant tumours and that these EVs carry mRNAs involved in migration and metastasisCitation39. Melo et al. have demonstrated how exosomes released by mammary tumour cells can cause cells from adjacent epithelial tissues to transform into tumour cellsCitation40.

The cancer-associated fibroblasts (CAFs) are the most abundant cells in the tumour’s immediate microenvironment. These are capable of releasing exosomes that transfer miRNAs and various proteins which accelerate the growth of these tumoursCitation41. It has also been shown that the tGF-B1 transported by the exosomes is capable of producing a powerful activation of the myofibroblasts, a limiting step in tumour growth and invasionCitation42.

Tumour growth

It has been understood for some time that glioblastomas release exosomes. These vesicles are rich in mRNA, miRNA and angiogenic proteins. They are taken up by normal host cells, such as brain microvascular endothelial cells and glioma cell lines stimulating aggressiveness and tumour growthCitation43.

Osti et al. demonstrated the role of plasma extracellular vesicle concentration levels in glioblastoma clinical diagnosis, and in providing indications about tumour and therapy responseCitation44.

MET oncoproteins which are contained in exosomes can support tumour growth in hepatic carcinomaCitation45. Another study referring to the same type of carcinoma, demonstrated that that the miRNA liberated in exosomes by HCC is an important mechanism for intercellular communication that can modulate TAK1 expression with the subsequent tumour growthCitation46.

Li et al. demonstrated that exosomes carrying miR-1246 can be transferred among different cell lines through direct uptake and can suppress the expression level of its target gene, Cyclin-G2 (CCNG2). By this pathway the tumour volume, migration and chemotherapy resistance of these cells are increasedCitation47.

MiR21 is transferred from cancer-associated adipocytes (CAAs) or fibroblasts (CAFs) to the cancer cells where it suppresses ovarian cancer apoptosis and confers chemoresistance by binding to its direct novel target, APAF1Citation48. In the same way, there are also exosomes with antitumour effect that compete biologically with the pro-tumoural exosomes and which can modulate the tumour growthCitation49.

Angiogenesis

The process of pathological angiogenesis is closely related to the tumour development, providing it with vessels to nourish it and giving the tumour the ability to spread to other tissuesCitation50. Exosome production is enhanced by intratumoural hypoxia, and endothelial cells uptake these cancer cells derived exosomes in order to stimulate the pathological angiogenesisCitation43,Citation51,Citation52.

The aforementioned exosomes not only influence vascular growth, but can also influence their metastatic capacity. These exosomes have the ability to modify vascular fragility, making it easier to penetrate tumour cellsCitation19.

Endothelial cells uptake cancer-secreted miR-105 from breast cancer cells targeting the tight junction protein ZO-1, destroying tight junctions and the integrity of these natural barriers against metastasisCitation53.

On the other hand, it was discovered that mesenchymal stem cell derived exosomes induce a significant and dose-dependent decrease in the expression and secretion of vascular endothelial growth factor (VEGF) through modulating the mTOR/HIF-1α signalling axis in breast cancer-derived cellsCitation54.

Evasion of immune response

Recently, Razzo et al. demonstrated how a single IV injection of tumour-derived exosomes was sufficient to condition mice harbouring premalignant OSCC lesions for accelerated tumour progression in concert with reduced immune cell migration to the tumourCitation55.

Currently the available evidence indicates that there is a dual role in cross communication between tumour cells and the cells of the immune system. On the one hand, it has been noted that there are several genes which stimulate the immune system, these include mesotheline and carcinoembryogenic antigen (CEA) which are released by tumour cells through exosomesCitation56.

On the other hand, many studies have noted that some tumour cells release exosomes which contain proteins and nucleic acids that produce a negative regulation of the immune responseCitation57.

To escape destruction by the immune response, tumours avoid being recognised by cytotoxic cells, directly impair the functioning of APCs (antigen-presenting cells) or cytotoxic cells, or induce suppressor cells which consequently shut down immune reactions. Immune cells are even converted into supporters of tumour growth and survival. Exosomes participate in all these strategies through proteins exposed at their surface, and intra-vesicular cytokines and nucleic acidsCitation58. The most common effects of cancer-derived exosomes activity against the immune system are:

  1. Defective Antigen Presentation: NKG2D ligand-carrying exosomes impair NKG2D-mediated NK-cell cytotoxicity by acting as a decoy, thus contributing to immune evasionCitation59–61.

  2. Suppression of APCs and Cytotoxic T Cells: Suppression of T cell proliferation through the expression of TGF-beta on the surface of exosomesCitation42.

  3. Induction of the apoptosis process in T cells through the receptor-mediated pathwayCitation62–64.

  4. Regulation of T cell mRNA transcription which regulates the expression of key immune function-related genes through miRNAs carried by cancer cell exosomesCitation65. Li et al. recently demonstrated that oxygen pressure in the tumour microenvironment orchestrates an anti- and protumoural γδ T-cell equilibrium by altering TEX content, which subsequently regulates MDSC (myeloid derived suppressor cells) function in a miR-21/PTEN/PD-L1-axis-dependent mannerCitation66.

  5. The extracellular adenosine release by induction of exosomes with CD39 and CD73 on their surface is a potent immune regulatory factor that protects cells and tissues from immune-mediated damage and negatively regulates local immune responsesCitation67. Exosomes induce pro-inflammatory activity of MSCs which in turn get tumour supportive characteristicsCitation68.

Resistance to chemotherapeutic agents

The resistance of tumour cells to chemotherapeutic agents is one of the greatest challenges for its pharmacological treatment. In a heterogeneous tumour environment the way in which less aggressive cells are able to change their phenotype towards greater aggressiveness and resistance has been observed. This is due to the transfer of miRNA, mRNA and proteins released by the exosomes from one cell to another cellCitation69.

Exosomes mediate a horizontal transfer of drug-resistant trait in chronic myeloid leukaemia cell by delivering miR-365Citation70. In this way, it has been demonstrated that exosomes secreted by acute myeloid leukaemia (AML) cells are an essential communicator for the interaction of bone marrow stromal cells and AML which can protect AML cells from chemotherapy drug-induced apoptosisCitation71.

A recent study by Qin et al. found that CAF-derived exosomal miR-196a confers cisplatin resistance in HNC by targeting CDKN1B and ING5. This finding indicates that miR-196a may serve as a predictor of, and potential therapeutic target for cisplatin resistance in HNCCitation72. CAFs exposed to gemcitabine in pancreatic cancer significantly increase the release of exosomes that increase chemoresistance inducing factor, Snail, in recipient epithelial cells and promote proliferation and drug resistanceCitation73. Exosomes can mediate taxol resistance by upregulating Septin-9 in hepatic cell carcinomaCitation74, sunitinib resistance in renal cancerCitation69, and cisplatin resistance in lung cancerCitation75.

Exosomes can also play an important role in drug effluxion by encapsulating and exporting the drugs from inside to outside, avoiding the therapeutic effect of thoseCitation76. Moreover, exosomes regulate the binding of antibodies to cancer cells, therefore playing an important role in downregulating their therapeutic effect.Citation37.

Metastasis

Metastasis is the first cause of death for a large majority of cancer patients despite the advanced techniques in radiotherapy, chemotherapy, immunotherapy and advanced surgical techniquesCitation77.

Recent data have been shown that EVs and specially exosomes may transform the microenvironment of primary tumours thus favouring the selection of cancer cells with a metastatic behaviourCitation78. The release of exosomes and other EVs from resident non-malignant cells may contribute to the metastatic processes as well. However, cancer EVs may induce malignant transformation in resident mesenchymal stem cells, suggesting that the metastatic process is not exclusively due to circulating tumour cellsCitation79.

Metastatic niche refers to the creation of a microenvironment in different organs and tissues that allows for the nesting and development of DTCs (disseminated tumour cellsCitation80–83.

The available scientific evidence indicates that metastatic niches are initiated previously (pre-metastatic niches) through the interaction of factors secreted by tumour cells, the recruitment of tumour progenitor cells, the recruitment of haematopoietic progenitor cells (HPC), myeloid cells, and mesenchymal stem cells (MSC) of the bone marrow, which allows for DTC nesting and subsequent growth, which depends on endothelial precursor cells (EPC) and finally on angiogenic factorsCitation84.

Modulation of vascular permeability and the stimulation of neo-angiogenesis are key steps during pre-metastatic niche formation which favours the initial extravasation and subsequent metastatic growth of tumour cells in secondary organsCitation81.

Oral squamous cell carcinoma hypoxic microenvironment may stimulate tumour cells to release miR-21–rich exosomes that are delivered to normoxic cells to enhance prometastatic behaviourCitation85.

Silencing of the miR-200c-3p targets, CHD9 and WRN, significantly accelerated the invasive potential of SQUU-A cells in squamous tongue carcinoma. miR-200c-3p in exosomes derived from a highly invasive OSCC line can induce a similar phenotype in non-invasive counterpartsCitation86.

A recent paper by Zhou et al. demonstrates that proteomic exosomes cargo PF4V1, CXCL7, F13A1 and ApoA1 from serum is related to the metastasis of OSCCCitation87.

Despite last years and recent discoveries, there still lies a gap in our knowledge of the dynamics of exosomes mediated metastatic processes: from the formation of the pre-metastatic niches, to the metastatic niches to the actual formation of the metastatic lesion.

What we clearly observe is that they play a key role in the preparation of the metastatic niche long before the arrival of any cancer cell in the area, even showing certain exosomes affinity for certain organs for the preparation of the metastatic niche.

The role of exosome in oral squamous cell carcinoma

The effect of exosomes released from oral squamous cell carcinoma (OSCC) into the tumour microenvironment and distant metastasis process is still not completely clear. It has been demonstrated that OSCC cell-derived exosomes are taken up by OSCC cells themselves and significantly promote proliferation, migration, and invasion through, among others, the activation of the PI3K/Akt, MAPK/ERK, and JNK-1/2 pathways in vitroCitation88.

Recent studies have shown an aberrantly expressed pattern of miRNA identified in both tumour and plasma of patients with OSCC, suggesting that this may be a biomarker for OSCC. It has become apparent that aberrations within the noncoding genome drive fundamental cancer phenotypes in addition to the best-known protein coding mutationsCitation89. Circulating exosomes appear to be a reliable method for evaluating circulating tumour-miRNA expressionCitation90. Multiple OSCC cell types exist in a single tumour mass and these secrete exosomes containing a unique set of miRNAs. Through the use of two unique malignant cell clones and by analysing the exosome-derived miRNAs, it was demonstrated that miR-200c-3p is an oncogenic miRNA which is capable of inducing invasive potential in non-invasive cells within an OSCC tumour massCitation86. On the other hand, exosomes derived from mesenchymal stem cells of human bone marrow that overexpress mi-RNA-101-3p suppress the proliferation, invasion and migration of oral cancer cells. Said mi-RNA is under-expressed in the cells of oral squamous cell carcinomasCitation91.

PCR based array methods identified the role of miRNA-26a and miRNA-26b in OSCC that enhances cancer cell migration and invasion through regulation of TMEM184BCitation92. As we stated, miRNAs are detected in the extracellular vesicles in OSCC; for example miRNA-21 was detected in exosomes derived from OSCC under hypoxic conditions. Snail and vimentin expression was significantly enhanced while the E-cadherin levels were decreased both in vitro and in vivo studies. Moreover, circulating exosomal miRNA-21 levels were associated with HIF-1α/HIF-2α expression, T stage, and lymph node metastasis in patients with OSCC. These findings suggest that the hypoxic microenvironment may promote prometastatic behaviours, stimulating tumour cells to generate miRNA-21-rich exosomes that are delivered to normoxic cellsCitation85.

OSCC exosomes are important intercellular communicators, delivering proteins, mRNA and miRNA to selected targets during premetastatic niche. The enrichment of tetraspanins within secreted exosomes hints the role of tetraspanin in the regulation of exosome uptake. Profiling of exosomal CD151 web proteins in the unexplored process of selection and uptake may lead to the early detection of cancer. Exosomal biomarker-based diagnosis is also useful both in laboratory research and clinical medicine, suggesting exciting new directions for future research. Tetraspanins are potential targets for drug development, not only in the area of neoplasias, but also in infectious diseases, given that many tetraspanins are known to facilitate infection processes of various pathogens, for example viral, bacterial and protozoan infectionsCitation93.

Overexpression and increased signalling from the epidermal growth factor receptor (EGFR) often change oral squamous cell carcinoma (OSCC) and thus EGFR is frequently targeted molecularly by the therapeutic antibody cetuximab. Fujiwara et al. assessed the roles of OSCC-derived extracellular vesicles (EVs) including exosomes in the trafficking of cetuximab and in epithelial-mesenchymal transition (EMT) of epithelial cells. OSCC cells abundantly expressed EGFR, which was secreted from cells with OSCC-EVs upon EGF stimulations. The OSCC-EGFR-EVs were then able to enter into and transform epithelial cells leading to increased mesenchymal traits with increased vimentin and spindle-like shapes. EGF priming of OSCC cells further increased this EMT-initiating effect of the OSCC-EVs. The internalisation and pro-EMT effects of the OSCC-EVs were largely blocked by cetuximab. Thus OSCC-derived EVs transform normal epithelial cells into a mesenchymal phenotype and anti-EGFR therapeutic antibody cetuximab inhibits such a carcinogenic effect of the OSCC-EVsCitation94.

Data indicate that miR- 200c-3p in exosomes derived from a highly invasive OSCC line can induce a similar phenotype in non-invasive counterpartsCitation86. On the other hand exosomes derived from cisplatin-resistant OSCC cells were found to enhance the chemoresistance of OSCC cell and reduce the DNA damage signalling in response to cisplatinCitation95.

Chen et al. demonstrated that exosomes released from HIV-infected T cells and those purified from blood of HIV-positive patients stimulate proliferation, migration and invasion of oral/oropharyngeal and lung cancer cells. The HIV transactivation response (TAR) element RNA in HIV-infected T-cell exosomes is responsible for promoting cancer cell proliferation and inducing expression of proto-oncogenes and Toll-like receptor 3 (TLR3)-inducible genesCitation96.

As we stated previously, EVs are detectable in significantly higher quantities in the plasma of patients with OSCC. Oncogenic miRNAs (such as miR-21, miR-27) were detectable in high quantity in the circulating EVs and plasma of patients with OSCC. EVs were taken up by monocytes after co-culture. Mechanistically, uptake of EVs derived from oral cancer cells by monocytes caused activation of the inflammatory pathway, NF-κB activation, and establishment of a pro-inflammatory and protumourigenic microenvironmentCitation89.

Exosomes derived from cancer cells may express surrogate oncogenic markers such as CEP55 membrane protein and carry FOXM1 mRNA cargos. CEP55 protein in saliva or blood could be exploited as a cancer biomarker for non-invasive mode of diagnosis and prognosis of HNSCCCitation97.

Tumour-derived exosomes (TEX) accumulate in the tumour microenvironment (TME) and serve as a communication system between tumour and normal stromal cells. TEX promote angiogenesis and drive HNSCC progressionCitation98.

Mesenchymal stem cells (MSCs) are a major component of the tumour microenvironment (TME) and play a key role in promoting tumour progression. The tumour uses exosomes to co-opt MSCs and re-program their functions. The MSCs re-programmed by TEX become avid producers of their own exosomes that carry and deliver mRNA and miRNA species as well as molecular signals not only back to tumour cells, directly enhancing their growth, but also horizontally to fibroblasts, endothelial cells and immune cells by enhancing their pro-tumour functions. TEX driven cross-talk of MSCs with immune cells blocks their anti-tumour activity and/or converts them into suppressor cells. MSCs re-programmed by TEX mediate pro-angiogenic activity and convert stromal cells into cancer-associated fibroblasts (CAFs)Citation99. In this way Wang et al. recently revealed that CAF-derived exosomes contain lower miR-3188 levels than normal fibroblasts, and the loss of miR-3188 in exosomes contributes to the malignant phenotypes of HNC cells through the derepression of BCL2. Furthermore, these data suggest the potential therapeutic value of exosomal miR-3188 for inhibiting HNC growthCitation100.

It is important to mention long non-coding RNAs that are functionally defined as transcripts bigger than 200 nt in length with no protein-coding potential,Citation101,Citation102, these exert their functions by affecting chromatin remodelling, transcriptional activation or suppression, miRNA sponge and miRNA splicing regulationCitation103. The nc-RNAs below are among those collected by some authors that are up and down-regulated in oral squamous cell carcinoma and his proposed function in literature ().

Table 1. The table is divided into two sections. In the upper section, some of the most important lnc-RNAs collected in the literature appear up-regulated in oral squamous cell carcinomas as well as their main biological effects. In the lower section are those that are down-regulated in the tumours mentioned above.

Recent research has made significant progress, overcoming major barriers for using exosomes as a delivery system. Exosomes are ideal systems for delivering cancer therapeutics, due to their size, surface expression profiles, low immunogenicity, low cytotoxicity, and long-term safety. Their use has opened a new promising avenue for cancer treatmentCitation123. A new study by Rosenberger et al. demonstrated that exosomes secreted by menstrual mesenchymal stem cells have a significant antitumour effect in the intra-tumoural injection of exosomes with a loss of tumour vasculature. The authors of this study claim that menstrual stem cell exosomes are potential anti-angiogenic agents for the treatment of neoplastic conditionsCitation124.

The exosomes whose content is Cav1, CD63, Rab5B and Annexin II are the most commonly described in oral cancer researchCitation125. The caveolin-1 gene is located at D7S522 locus of human chromosome 7q31.1. CAV1 is expressed in most cell typesCitation126 and is present in a variety of cellular and extracellular compartments. CAV1 has a role in both normal and pathological tissue, where it has been shown to be upregulated by the hypoxia-inducible factor (HIF)-αCitation127 that enhances the oncogenic potential of tumour cells by increasing the cell’s proliferative, migratory, and invasive capacitiesCitation128 and even chemotherapy and radiotherapy resistanceCitation129. Some studies on CAV1 in oral squamous cell carcinoma (OSCC) showed an increased immunoexpression of CAV1 in SCC tissue when compared to normal mucosa and precancerous (dysplastic) lesionsCitation130), and it was even found to be significantly over-expressed in OSCC compared to normal oral mucosa (p = 0.002 and p = 0.033, respectively) when using immunohistochemistry to demonstrate that patients with over-expression of Cav-1 protein were associated with poor prognosis (p = 0.030)Citation131. These studies have shown a gradual increased expression of CAV-1 in the different steps of cancerous process in oral cancerCitation132. Authors suggest that accumulation of CAV1-TME in TSCC had a negative prognostic value in vivoCitation133. This was demonstrated in the study by Rodríguez et al. where the negative relationship between plasmatic CAV1 exosomes and the overall survival of OSCC patients was verifiedCitation36. Vered et al. demonstrated how the accumulation of CAV1 protein in tumour microenvironment in patients with tongue squamous cell carcinoma had a negative prognostic value and how CAV1 is involved in fibroblast undergoing trans-differentiation to CAFsCitation134. It is important to note that some authors propose that CAV1 does have a role as a tumour suppressor and that there is a connection between loss of Cav-1 expression and the ability of cells to escape from anchorage growth controlCitation135. The literature suggests that Cav-1 is downregulated in colon cancer, breast cancer, ovarian carcinoma, and soft tissue sarcomas, while increased expression was seen in ductal adenocarcinoma of the pancreas, prostate cancer, squamous cell carcinoma, glioblastoma, non-small cell lung carcinoma (NSCLC) and renal cell carcinomaCitation136.

On the other hand, Western blot analysis confirmed the presence of the known exosome marker CD63Citation85 in OSCC patients. Although CD63 has been originally described as a tumour suppressorCitation137 reassessing its function is worthwhile as it was identified as a receptor of tissue inhibitor of metalloproteinases-1 (TIMP-1)Citation138 which may promote metastasisCitation139.

It is important to note that authors such as Shinya et al. have demonstrated that the uptake of exosomes by OSCC cells and subsequent tumour progression was abrogated in the presence of heparin, and for that reason it may be useful for treatment of these patientsCitation88. In the aforementioned study by Rodríguez et al., a negative survival rate was observed among patients with OSCC in stage IV with plasmatic levels of CD63+ exosomes higher than the mean, when compared to the survival of those patients with levels below the mean. A significant reduction of CD63 exosome levels after tumour resection was also observed, thus proving the existence of a clinical relationship as tumour markers of important prognostic value in OSCC patients and its utility in the clinical follow-up of cancer patientsCitation36.

Exosomes in saliva

Salivaomics integrate the study of saliva and its constituents, functions, and related techniquesCitation140. The first authors to report the presence of exosomes in saliva were Ogawa et al. in the year 2008Citation141.

The collection of exosomes in saliva is simple and non-invasive. It also contains less proteins than blood and therefore its identification and quantification is simplified considerablyCitation142,Citation143. It has been demonstrated that they can be stored at 4° C without the need for them to be supercooled at −80° C, making it much easier to work with them in clinical environmentsCitation144.

In the year 2011, Ogawa et al. described two types of salivary exosomes that differ mainly in terms of their size and protein composition. Type I exosomes are larger and denser to electrons than type II exosomes; however, the latter are more similar to those present in other fluids and body mediaCitation141. Both types of exosomes contain protein markers such as CD63, Alix, Tsg101 and Hsp70, immunoglobulin A and polymeric immunoglobulin receptors, although the specific protein composition is different. Salivary human exosomes participate in the catabolism of biopeptides and play a very important role in the local immune response of the oral cavity. It is believed that the secretion of type I or II exosomes depends, to a large extent, on the type of salivary gland that produces itCitation145. The Next-Generation Sequencing Technology has recently allowed for more exhaustive investigations regarding RNAs that encode long proteins to be carried out. These RNAs form an important part of the salivary exosomes and are able to translationally control tumour protein, likewise they play an important role in cell proliferation and death as well as in the immune responseCitation146. In 2016, Ogawa et al. demonstrated, in vitro, how the content of RNAs and other pseudogenes included in the interior of the salivary exosomes can be transferred horizontally to other cells, modulating the genetic expression of the receptorsCitation147 and increasing their invasion and migration capacityCitation85.

J. Kim et al. checked the amount and type of extracellular vesicles in plasma and saliva in melanoma mice. The authors found that only 38.22% ± 18.55% of the vesicles found in plasma appeared in saliva, unlike the cases in which the tumour pathology is in the oral cavity and the exosome load is much higher due to the direct contact between the saliva and the tumourCitation148.

Several of the exosomal miRNA secreted solely by cancer cells in culture were detected at substantially elevated levels in saliva from HNSCC patients compared to saliva from healthy controls. These findings provide important insight into tumour biology and yield a promising set of candidate HNSCC biomarkers for use with non-invasive saliva samplesCitation149.

Zahran et al. reported a highly significant increase in salivary miRNA-21 and miRNA-184 in saliva of OSCC patients when compared to healthy and disease controls and in fact, the only microRNA they found to discriminate between OSCC and oral potentially malignant disorders was miRNA-184Citation150.

Sharma et al. showed that exosome size, exosome population, and inter-exosome are increased in the saliva of patients with oral cancer. Interestingly, oral cancer exosomes exhibited significantly increased CD63 surface densities and displayed irregular morphologiesCitation151.

Zlotogorski-Hurvitz et al. were able to show morphological as well as molecular differences in salivary exosomes of patients with oral squamous cell carcinoma with respect to healthy patients as a screening measure in patients with a high risk of oral squamous cell carcinomaCitation152.

In tumour cell hypoxic environment, as in the case of oral squamous cell carcinoma, mi-RNA is produced more intensely, this is one of the genetic materials which is most easily identifiable in exosomes and it is of vital importance due to its capacity to regulate the tumour microenvironmentCitation153. miR-21 is one of the most intensively produced RNA genes and it promotes metastatic cell behaviour through its interaction with (PTEN) and (PDCD4), as it transmits tumour-resistant characteristics to chemotherapeutic agents such as cisplatinCitation95. Releasing miR-200c-3p facilitates tumour invasion in tumour-free areasCitation86,Citation154.

Langevin et al. used next-generation sequencing for miRNA, thus differentiating miRNA from HNSCC cells in relation to healthy epithelial cells. They observed that a large amount of mi-RNA was shared in exosomes from healthy and cancerous cells. However, significantly higher concentrations of miR-486-5p, miR-486-3p, and miR-10b-5p were detected with respect to the control groupCitation149.

Some studies by Gai et al. detected by charged based precipitation de presence of miR-412-3p, miR-512-3p, miR-27a-3p, miR-494-3p up-regulated in the saliva of OSCC patients and miR-302b-3p, miR-517b-3p expressed only in exosomes from the saliva OSCC patientsCitation155,Citation156.

As in the blood, CD63 + exosomes were also found in salivaCitation151,Citation152 that have shown prognostic value in patients with OSCC when they are counted in serum prior to and after tumour resection surgeryCitation36. At the same time exosomes PPIA + were detected down-regulated as a poor prognosis factor in the saliva of OSCC patientsCitation157.

A growing body of saliva-exosomics study is highlighting the role of cancer-derived exosomes in saliva. The properties of cancer-derived exosomes in saliva are attracting the attention of scientists since these exosomes could be used as diagnostic biomarkers, potential surrogate markers for other physical conditions, or novel immune regulatory systems through the gastrointestinal tract. However, the utility of salivary exosomes as biomarkers of diseases and conditions requires further investigations due to the current paucity of studies in this emerging area they have a promising role in next year’s investigation and will represent an important future challenge in diagnosis, treatment and prognosis of different cancer type not only OSCC due the capacity of different body cancers exosomes to be secreted by salivary glandsCitation158,Citation159.

Conclusion

In conclusion, we are able to determine that the information available in this field is limited and very recent. Most of it focuses on the quantification and qualification of the exosomes content present in plasma or saliva. However, information about its biological role with respect to squamous cell carcinoma of the mouth is small, although it increases rapidly. As mentioned, there are studies that are able to show their direct relationship with the aggressiveness of the cancer. Also its capacity as a biomarker of the presence of the disease and life prognosis of the patients who suffer it is of great relevance in the diagnosis and treatment of patients with oral squamous cell carcinoma as well as other types of cancers. More research is required in a field that is gaining considerable ground due to its importance in the carcinogenic process.

Disclosure statement

No potential conflict of interest was reported by the authors.

References

  • Marur S, Forastiere AA. Head and neck cancer: changing epidemiology, diagnosis, and treatment. Mayo Clin Proc 2008;83:489–501.
  • Kang H, Kiess A, Chung CH. Emerging biomarkers in head and neck cancer in the era of genomics. Nat Rev Clin Oncol 2015;12:11–26.
  • Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015;136:E359.
  • Rivera C. Essentials of oral cancer. Int J Clin Exp Pathol 2015;8:11884–94.
  • Neville BW, Day TA. Oral cancer and precancerous lesions. CA: Cancer J Clin 2002;52:195–215.
  • Farsi NJ, Rousseau M, Schlecht N, et al. Aetiological heterogeneity of head and neck squamous cell carcinomas: the role of human papillomavirus infections, smoking, and alcohol. Carcinogenesis 2017;38:1188–95.
  • Rivera C. Essentials of oral cancer. Int J Clin Exper Pathol 2015;8:11884.
  • Lee Y, El Andaloussi S, Wood M. Exosomes and microvesicles: extracellular vesicles for genetic information transfer and gene therapy. Hum Mol Genet 2012;21:125.
  • Théry 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;7:1535750.
  • Tkach M, Théry C. Communication by extracellular vesicles: where we are and where we need to go. Cell 2016;164:1226–32.
  • Fais S, Logozzi M, Lugini L, et al. Exosomes: the ideal nanovectors for biodelivery. Biol Chem 2013;394:1–15.
  • Stoorvogel W, Kleijmeer MJ, Geuze HJ, Raposo G. The biogenesis and functions of exosomes. Traffic 2002;3:321–30.
  • Peinado H, Lavotshkin S, Lyden D. The secreted factors responsible for pre-metastatic niche formation: old sayings and new thoughts. Seminar cancer biol 2011;21:139–46.
  • J O'Loughlin A, A Woffindale C, JA Wood M. Exosomes and the emerging field of exosome-based gene therapy. Curr Gene Ther 2012;12:262–74.
  • King HW, Michael MZ, Gleadle JM. Hypoxic enhancement of exosome release by breast cancer cells. BMC Cancer 2012;12:421.
  • Webber J, Steadman R, Mason MD, et al. Cancer exosomes trigger fibroblast to myofibroblast differentiation. Cancer Res 2010;70:9621–30.
  • Liu C, Yu S, Zinn K, et al. Murine mammary carcinoma exosomes promote tumor growth by suppression of NK cell function. J Immunol 2006;176:1375–85.
  • Hood JL, San RS, Wickline SA. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res 2011;71:3792–801.
  • Peinado H, Alečković M, Lavotshkin S, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med 2012;18:883–91.
  • 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;1841:108–20.
  • Subra C, Grand D, Laulagnier K, et al. Exosomes account for vesicle-mediated transcellular transport of activatable phospholipases and prostaglandins. J Lipid Res 2010;51:2105–20.
  • Xiang X, Poliakov A, Liu C, et al. Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer 2009;124:2621–33.
  • Sansone P, Savini C, Kurelac I, et al. Packaging and transfer of mitochondrial DNA via exosomes regulate escape from dormancy in hormonal therapy-resistant breast cancer. Proc Natl Acad Sci USA 2017;114:E906–75.
  • Valadi H, Ekström K, Bossios A, et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 2007;9:654–9.
  • Wang Z, von Au A, Schnolzer M, et al. CD44v6-competent tumor exosomes promote motility, invasion and cancer-initiating cell marker expression in pancreatic and colorectal cancer cells. Oncotarget 2016;7:55409–36.
  • Ling H, Fabbri M, Calin GA. MicroRNAs and other non-coding RNAs as targets for anticancer drug development. Nat Rev Drug Discov 2013;12:847–65.
  • Mathivanan S, Fahner CJ, Reid GE, Simpson RJ. ExoCarta 2012: database of exosomal proteins, RNA and lipids. Nucleic Acids Res 2012;40:1241.
  • Andreu Z, Yáñez-Mó M. Tetraspanins in extracellular vesicle formation and function. Front Immunol 2014;5:442.
  • Simons M, Raposo G. Exosomes–vesicular carriers for intercellular communication. Curr Opin Cell Biol 2009;21:575–81.
  • Gillies RJ, Pilot C, Marunaka Y, Fais S. Targeting acidity in cancer and diabetes. Biochem Biophys Acta Rev Cancer 2019;1871:273–80.
  • Parolini I, Federici C, Raggi C, et al. Microenvironmental pH is a key factor for exosome traffic in tumor cells. J Biol Chem 2009;284:34211–22.
  • Logozzi M, Spugnini E, Mizzoni D, et al. Extracellular acidity and increased exosome release as key phenotypes of malignant tumors. Cancer Metastasis Rev 2019;38:93–101.
  • Patton MC, Zubair H, Khan MA, et al. Hypoxia alters the release and size distribution of extracellular vesicles in pancreatic cancer cells to support their adaptive survival. J Cell Biochem 2019.
  • Properzi F, Logozzi M, Fais S. Exosomes: the future of biomarkers in medicine. Biomarkers 2013;7:769–78.
  • Logozzi M, De Milito A, Lugini L, et al. High levels of exosomes expressing CD63 and caveolin-1 in plasma of melanoma patients. PLoS ONE 2009;4:e5219.
  • Rodríguez Zorrilla S, Pérez-Sayans M, Fais S, et al. A pilot clinical study on the prognostic relevance of plasmatic exosomes levels in oral squamous cell carcinoma patients. Cancers 2019;11:429.
  • Guo W, Gao Y, Li N, et al. Exosomes: new players in cancer (Review). Oncol Rep 2017;38:665–75.
  • Costa-Silva B, Aiello NM, Ocean AJ, et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol 2015;17:816–26.
  • Zomer A, Maynard C, Verweij FJ, et al. In vivo imaging reveals extracellular vesicle-mediated phenocopying of metastatic behavior. Cell 2015;161:1046–57.
  • Melo SA, Sugimoto H, O’Connell JT, et al. Cancer exosomes perform cell-independent MicroRNA biogenesis and promote tumorigenesis. Cancer Cell 2014;26:707–21.
  • Donnarumma E, Fiore D, Nappa M, et al. Cancer-associated fibroblasts release exosomal microRNAs that dictate an aggressive phenotype in breast cancer. Oncotarget 2017;8:19592–608.
  • Webber JP, Spary LK, Sanders AJ, et al. Differentiation of tumour-promoting stromal myofibroblasts by cancer exosomes. Oncogene 2015;34:290–302.
  • Skog J, Würdinger T, van Rijn S, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol 2008;10:1470–6.
  • Osti D, Del Bene M, Rappa G, et al. Clinical significance of extracellular vesicles in plasma from glioblastoma patients. Clin Cancer Res 2019;25:266–76.
  • Matsumoto K, Umitsu M, De Silva DM, et al. Hepatocyte growth factor/MET in cancer progression and biomarker discovery. Cancer Sci 2017;108:296–307.
  • Kogure T, Lin W, Yan IK, et al. Intercellular nanovesicle-mediated microRNA transfer: a mechanism of environmental modulation of hepatocellular cancer cell growth. Hepatology 2011;54:1237–48.
  • Li XJ, Ren ZJ, Tang JH, Yu Q. Exosomal microRNA MiR-1246 promotes cell proliferation, invasion and drug resistance by targeting CCNG2 in breast cancer. Cell Physiol Biochem 2017;44:1741–8.
  • Au Yeung CL, Co N, Tsuruga T, et al. Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat Commun 2016;7:11150.
  • Roccaro AM, Sacco A, Maiso P, et al. BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J Clin Invest 2013;123:1542–55.
  • Fernandes Ribeiro M, Zhu H, Millard RW, Fan G-C. Exosomes function in pro- and anti-angiogenesis. Current Angiogenesis 2013;2:54–9.
  • Park JE, Tan HS, Datta A, et al. Hypoxic tumor cell modulates its microenvironment to enhance angiogenic and metastatic potential by secretion of proteins and exosomes. Mol Cell Proteomics 2010;9:1085–99.
  • Umezu T, Ohyashiki K, Kuroda M, Ohyashiki JH. Leukemia cell to endothelial cell communication via exosomal miRNAs. Oncogene 2013;32:2747–55.
  • Zhou W, Fong MY, Min Y, et al. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell 2014;25:501–15.
  • Pakravan K, Babashah S, Sadeghizadeh M, et al. MicroRNA-100 shuttled by mesenchymal stem cell-derived exosomes suppresses in vitro angiogenesis through modulating the mTOR/HIF-1α/VEGF signaling axis in breast cancer cells. Cell Oncol 2017;40:457–70.
  • Razzo BM, Ludwig N, Hong C, et al. Tumor-derived exosomes promote carcinogenesis of murine oral squamous cell carcinoma. Carcinogenesis 2019; pii: bgz124.
  • Dai S, Wan T, Wang B, et al. More efficient induction of HLA-A*0201-restricted and carcinoembryonic antigen (CEA)-specific CTL response by immunization with exosomes prepared from heat-stressed CEA-positive tumor cells. Clin Cancer Res 2005;11:7554–63.
  • Zitvogel L, Regnault A, Lozier A, et al. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med 1998;4:594–600.
  • Czernek L, Düchler M. Functions of cancer-derived extracellular vesicles in immunosuppression. Arch Immunol Ther Exp 2017;65:311–23.
  • Lundholm M, Schröder M, Nagaeva O, 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:e108925.
  • Hong CS, Muller L, Boyiadzis M, Whiteside TL. Isolation and characterization of CD34+ blast-derived exosomes in acute myeloid leukemia. PLoS ONE 2014;9:e103310.
  • López-Cobo S, Campos-Silva C, Valés-Gómez M. Glycosyl-phosphatidyl-inositol (GPI)-anchors and metalloproteases: their roles in the regulation of exosome composition and NKG2D-mediated immune recognition. Front Cell Dev Biol 2016;4:97.
  • Wieckowski EU, Visus C, Szajnik M, et al. Tumor-derived microvesicles promote regulatory T cell expansion and induce apoptosis in tumor-reactive activated CD8+ T lymphocytes. J Immunol 2009;183:3720–30.
  • Taylor DD, Gerçel-Taylor C, Lyons KS, et al. T-cell apoptosis and suppression of T-cell receptor/CD3-zeta by Fas ligand-containing membrane vesicles shed from ovarian tumors. Clin Cancer Res 2003;9:5113–9.
  • Chen W, Jiang J, Xia W, Huang J. Tumor-related exosomes contribute to tumor-promoting microenvironment: an immunological perspective. J Immunol Res 2017;2017:1.
  • Ding G, Zhou L, Qian Y, et al. Pancreatic cancer-derived exosomes transfer miRNAs to dendritic cells and inhibit RFXAP expression via miR-212-3p. Oncotarget 2015;6:29877–88.
  • Li L, Cao B, Liang X, et al. Microenvironmental oxygen pressure orchestrates an anti- and pro-tumoral γδ T cell equilibrium via tumor-derived exosomes. Oncogene 2018;38:2830–43.
  • Schuler PJ, Saze Z, Hong C, et al. Human CD4+ CD39+ regulatory T cells produce adenosine upon co-expression of surface CD73 or contact with CD73+ exosomes or CD73+ cells. Clin Exp Immunol 2014;177:531–43.
  • Li X, Wang S, Zhu R, et al. Lung tumor exosomes induce a pro-inflammatory phenotype in mesenchymal stem cells via NFκB-TLR signaling pathway. J Hematol Oncol 2016;9:42.
  • 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;29:653–68.
  • Min Q, Wang X, Zhang J, et al. Exosomes derived from imatinib-resistant chronic myeloid leukemia cells mediate a horizontal transfer of drug-resistant trait by delivering miR-365. Exp Cell Res 2017;362:386–93.
  • Chen T, Zhang G, Kong L, et al. Leukemia-derived exosomes induced IL-8 production in bone marrow stromal cells to protect the leukemia cells against chemotherapy. Life Sci 2019;221:187–95.
  • 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.
  • Richards KE, Zeleniak AE, Fishel ML, et al. Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene 2017;36:1770–8.
  • Sun H, Cui R, Zhou J, et al. ERK activation globally downregulates miRNAs through phosphorylating exportin-5. Cancer Cell 2016;30:723–36.
  • Qin X, Yu S, Zhou L, et al. Cisplatin-resistant lung cancer cell–derived exosomes increase cisplatin resistance of recipient cells in exosomal miR-100–5p-dependent manner. Int J Nanomed 2017;12:3721–33.
  • Bach D, Hong J, Park HJ, Lee SK. The role of exosomes and miRNAs in drug-resistance of cancer cells. Int J Cancer 2017;141:220–30.
  • Langley RR, Fidler IJ. Tumor cell-organ microenvironment interactions in the pathogenesis of cancer metastasis. Endocr Rev 2007;28:297–321.
  • Lugini L, Valtieri M, Federici C, et al. Exosomes from human colorectal cancer induce a tumor-like behavior in colonic mesenchymal stromal cells. Oncotarget 2016;7:50086–98.
  • Zhao H, Achreja A, Iessi E, et al. The key role of extracellular vesicles in the metastatic process. Biochim Biophys Acta 2018;1869:64–77.
  • Liu Y, Cao X. Characteristics and significance of the pre-metastatic niche. Cancer Cell 2016;30:668–81.
  • Sceneay J, Smyth MJ, Möller A. The pre-metastatic niche: finding common ground. Cancer Metastasis Rev 2013;32:449–64.
  • Sleeman JP. The lymph node pre-metastatic niche. J Mol Med 2015;93:1173–84.
  • Psaila B, Lyden D. The metastatic niche: adapting the foreign soil. Nat Rev Cancer 2009;9:285–93.
  • Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer 2008;8:618–31.
  • 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;76:1770–80.
  • Kawakubo-Yasukochi T, Morioka M, Hazekawa M, et al. miR-200c-3p spreads invasive capacity in human oral squamous cell carcinoma microenvironment. Mol Carcinog 2017;57:295–302.
  • Zhou N, Li C, Zhou Y, et al. Potential markers from serum-purified exosomes for detecting oral squamous cell carcinoma metastasis. Cancer Epidemiol Biomarkers Prev 2019;pii: cebp.1122.2018.
  • 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:e0148454.
  • Momen-Heravi F, Bala S. miRNA regulation of innate immunity. J Leukoc Biol 2018;103:1205.
  • Giallombardo M, Chacártegui Borrás J, Castiglia M, et al. Exosomal miRNA analysis in non-small cell lung cancer (NSCLC) patients' plasma through qPCR: a feasible liquid biopsy tool. J Vis Exp 2016.
  • Xie C, Du L, Guo F, et al. 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;458:1–16.
  • Fukumoto I, Hanazawa T, Kinoshita T, et al. MicroRNA expression signature of oral squamous cell carcinoma: functional role of microRNA-26a/b in the modulation of novel cancer pathways. Br J Cancer 2015;112:891–900.
  • Malla RR, Pandrangi S, Kumari S, et al. Exosomal tetraspanins as regulators of cancer progression and metastasis and novel diagnostic markers. Asia Pac J Clin Oncol 2018;14:383–91.
  • Fujiwara T, Eguchi T, Sogawa C, et al. Carcinogenic epithelial-mesenchymal transition initiated by oral cancer exosomes is inhibited by anti-EGFR antibody cetuximab. Oral Oncol 2018;86:251–7.
  • Liu T, Chen G, Sun D, 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 2017;49:808–16.
  • Chen L, Feng Z, Yue H, et al. Exosomes derived from HIV-1-infected cells promote growth and progression of cancer via HIV TAR RNA. Nat Commun 2018;9:458–2.
  • Qadir F, Aziz MA, Sari CP, et al. Transcriptome reprogramming by cancer exosomes: identification of novel molecular targets in matrix and immune modulation. Mol Cancer 2018;17:97.
  • Ludwig N, Yerneni SS, Razzo BM, Whiteside TL. Exosomes from HNSCC promote angiogenesis through reprogramming of endothelial cells. Mol Cancer Res 2018;16:1798–808.
  • Whiteside TL. Exosome and mesenchymal stem cell cross-talk in the tumor microenvironment. Semin Immunol 2018;35:69–79.
  • Wang X, Qin X, Yan M, et al. Loss of exosomal miR-3188 in cancer-associated fibroblasts contributes to HNC progression. J Exper Clin Cancer Res 2019;38:151.
  • Iyer MK, Niknafs YS, Malik R, et al. The landscape of long noncoding RNAs in the human transcriptome. Nat Gen 2015;47:199–208.
  • Wilusz JE, Sunwoo H, Spector DL. Long noncoding RNAs: functional surprises from the RNA world. Genes Dev 2009;23:1494–504.
  • Momen-Heravi F, Bala S. Emerging role of non-coding RNA in oral cancer. Cell Signal 2018;42:134–43.
  • Zhou X, Liu S, Cai G, et al. Long non coding RNA MALAT1 promotes tumor growth and metastasis by inducing epithelial-mesenchymal transition in oral squamous cell carcinoma. Sci Rep 2015;5:15972.
  • Feng L, Houck JR, Lohavanichbutr P, Chen C. Transcriptome analysis reveals differentially expressed lncRNAs between oral squamous cell carcinoma and healthy oral mucosa. Oncotarget 2017;8:31521.
  • Arunkumar G, Rao DM, Kuha A, Manikandan M, Arun K, et al. Expression profiling of long non-coding RNA identifies linc-RoR as a prognostic biomarker in oral cancer. Tumour Biol 2017;39. https://doi.org/https://doi.org/10.1177/1010428317698366.
  • Ouyang K, Zou R, Liang J, et al. TUC338 overexpression leads to enhanced proliferation and reduced apoptosis in tongue squamous cell carcinoma cells in vitro. J Oral Maxillofac Surg 2017;75:423–8.
  • Zhang C. Long non-coding RNA FTH1P3 facilitates oral squamous cell carcinoma progression by acting as a molecular sponge of miR-224-5p to modulate fizzled 5 expression. Gene 2017;607:47–55.
  • Yang Y, Wang Y, Lai J, et al. Long non-coding RNA UCA1 contributes to the progression of oral squamous cell carcinoma by regulating the WNT/β-catenin signaling pathway. Cancer Sci 2016;107:1581–9.
  • Yu J, Liu Y, Guo C, et al. Upregulated long non-coding RNA LINC00152 expression is associated with progression and poor prognosis of tongue squamous cell carcinoma. J Cancer 2017;8:523.
  • Li X, Ma C, Zhang L, et al. LncRNAAC132217. 4, a KLF8-regulated long non-coding RNA, facilitates oral squamous cell carcinoma metastasis by upregulating IGF2 expression. Cancer Lett 2017;407:45–56.
  • Shih J, Chiang W, Wu ATH, et al. Long noncoding RNA LncHIFCAR/MIR31HG is a HIF-1α co-activator driving oral cancer progression. Nat Commun 2017;8:15874.
  • Zhang C. Long intergenic non-coding RNA 668 regulates VEGFA signaling through inhibition of miR-297 in oral squamous cell carcinoma. Biochem Biophy Res Commun 2017;489:404–12.
  • Wang L, Ye S, Wang J, et al. HuR stabilizes lnc-Sox5 mRNA to promote tongue carcinogenesis. Biochem Mosc 2017;82:438–45.
  • Yu J, Liu Y, Gong Z, et al. Overexpression long non-coding RNA LINC00673 is associated with poor prognosis and promotes invasion and metastasis in tongue squamous cell carcinoma. Oncotarget 2017;8:16621–32.
  • Li W, Han Y, Zhao Z, et al. Oral mucosal mesenchymal stem cell-derived exosomes: a potential therapeutic target in oral premalignant lesions. Int J Oncol 2019;54:1567–78.
  • Sun L, Xu K, Cui J, et al. Cancer-associated fibroblast-derived exosomal miR-382-5p promotes the migration and invasion of oral squamous cell carcinoma. Oncol Rep 2019;36:209–20.
  • Moon S, Kim DK, Kim J. Apoptosis-related microRNA-145-5p enhances the effects of pheophorbide a-based photodynamic therapy in oral cancer. Oncotarget 2017;8:35184–92.
  • Huang W, Cui X, Chen J, et al. Long non-coding RNA NKILA inhibits migration and invasion of tongue squamous cell carcinoma cells via suppressing epithelial-mesenchymal transition. Oncotarget 2016;7:62520–32.
  • Jia L, Wei S, Gan Y, et al. Expression, regulation and roles of miR‐26a and MEG3 in tongue squamous cell carcinoma. Int J Cancer 2014;135:2282–93.
  • Zhang J, Lin Z, Gao Y, Yao T. Downregulation of long noncoding RNA MEG3 is associated with poor prognosis and promoter hypermethylation in cervical cancer. J Exper Clin Cancer Res 2017;36:5.
  • Liu Z, Wu C, Xie N, Wang P. Long non-coding RNA MEG3 inhibits the proliferation and metastasis of oral squamous cell carcinoma by regulating the WNT/β-catenin signaling pathway. Oncol Lett 2017;14:4053–8.
  • Yang M, Wu SY. The advances and challenges in utilizing exosomes for delivering cancer therapeutics. Front Pharmacol 2018;9:735.
  • Rosenberger L, Ezquer M, Lillo-Vera F, et al. Stem cell exosomes inhibit angiogenesis and tumor growth of oral squamous cell carcinoma. Sci Rep 2019;9:663.
  • Xie C, Ji N, Tang Z, et al. The role of extracellular vesicles from different origin in the microenvironment of head and neck cancers. Mol Cancer 2019;18:83.
  • Thomas CM, Smart EJ. Caveolae structure and function. J Cell Mol Med 2008;12:796–809.
  • Goetz JG, Minguet S, Navarro-Lérida I, et al. Biomechanical remodeling of the microenvironment by stromal caveolin-1 favors tumor invasion and metastasis. Cell 2011;146:148–63.
  • Wang Y, Roche O, Xu C, et al. Hypoxia promotes ligand-independent EGF receptor signaling via hypoxia-inducible factor-mediated upregulation of caveolin-1. Proc Natl Acad Sci USA 2012;109:4892–7.
  • Ketteler J, Klein D. Caveolin-1, cancer and therapy resistance. Int J Cancer 2018;143:2092–104.
  • Hung K, Lin S, Liu C, et al. The biphasic differential expression of the cellular membrane protein, caveolin-1, in oral carcinogenesis. J Oral Pathol Med 2003;32:461–7.
  • Auzair LBM, Vincent-Chong VK, Ghani WMN, et al. Caveolin 1 (Cav-1) and actin-related protein 2/3 complex, subunit 1B (ARPC1B) expressions as prognostic indicators for oral squamous cell carcinoma (OSCC). Eur Arch Oto-Rhino-Laryngol 2016;273:1885–93.
  • Xue J, Chen H, Diao L, et al. Expression of caveolin-1 in tongue squamous cell carcinoma by quantum dots. Eur J Histochem 2010;54:e20.
  • Andrade D, Leite ALD, de Oliveira CE, Dourado MR, et al. Extracellular vesicles from oral squamous carcinoma cells display pro- and antiangiogenic properties. Oral Dis 2018;24:725–31.
  • Vered M, Lehtonen M, Hotakainen L, et al. Caveolin-1 accumulation in the tongue cancer tumor microenvironment is significantly associated with poor prognosis: an in-vivo and in-vitro study. BMC Cancer 2015;15:25.
  • Routray S. Caveolin-1 in oral squamous cell carcinoma microenvironment: an overview. Tumour Biol 2014;35:9487–95.
  • Williams TM, Lisanti MP. Caveolin-1 in oncogenic transformation, cancer, and metastasis. Am J Physiol-Cell Physiol 2005;288:C494–506.
  • Kwon MS, Shin S, Yim S, et al. CD63 as a biomarker for predicting the clinical outcomes in adenocarcinoma of lung. Lung Cancer 2007;57:46–53.
  • Jung K, Liu X, Chirco R, et al. Identification of CD63 as a tissue inhibitor of metalloproteinase-1 interacting cell surface protein. Embo J 2006;25:3934–42.
  • Cruz-Munoz W, Khokha R. The role of tissue inhibitors of metalloproteinases in tumorigenesis and metastasis. Critic Rev Clin Labo Sci 2008;45:291–338.
  • Cheng J, Nonaka T, Wong D. Salivary exosomes as nanocarriers for cancer biomarker delivery. Materials 2019;12:654.
  • Ogawa Y, Kanai-Azuma M, Akimoto Y, et al. Exosome-like vesicles with dipeptidyl peptidase IV in human saliva. Biol Pharm Bull 2008;31:1059–62.
  • Topkas E, Keith P, Dimeski G, et al. Evaluation of saliva collection devices for the analysis of proteins. Clin Chim Acta 2012;413:1066–70.
  • Schulz BL, Cooper-White J, Punyadeera CK. Saliva proteome research: current status and future outlook. Crit Rev Biotechnol 2013;33:246–59.
  • Kumeda N, Ogawa Y, Akimoto Y, et al. Characterization of membrane integrity and morphological stability of human salivary exosomes. Biol Pharm Bull 2017;40:1183–91.
  • Ogawa Y, Taketomi Y, Murakami M, et al. Small RNA transcriptomes of two types of exosomes in human whole saliva determined by next generation sequencing. Biol Pharm Bull 2013;36:66–75.
  • Nair S, Tang KD, Kenny L, Punyadeera C. Salivary exosomes as potential biomarkers in cancer. Oral Oncol 2018;84:31–40.
  • Ogawa Y, Tsujimoto M, Yanoshita R. Next-generation sequencing of protein-coding and long non-protein-coding RNAs in two types of exosomes derived from human whole saliva. Biol Pharm Bull 2016;39:1496–507.
  • Kim J, Shin H, Park J. RNA in salivary extracellular vesicles as a possible tool for systemic disease diagnosis. J Dent Res 2017;96:938–44.
  • 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;8:82459–74.
  • Zahran F, Ghalwash D, Shaker O, et al. Salivary microRNAs in oral cancer. Oral Dis 2015;21:739–47.
  • Sharma S, Gillespie BM, Palanisamy V, Gimzewski JK. Quantitative nanostructural and single-molecule force spectroscopy biomolecular analysis of human-saliva-derived exosomes. Langmuir 2011;27:14394–400.
  • Zlotogorski-Hurvitz A, Dayan D, Chaushu G, et al. Morphological and molecular features of oral fluid-derived exosomes: oral cancer patients versus healthy individuals. J Cancer Res Clin Oncol 2016;142:101–10.
  • Yakob M, Fuentes L, Wang MB, et al. Salivary biomarkers for detection of oral squamous cell carcinoma - current state and recent advances. Curr Oral Health Rep 2014;1:133–41.
  • Han Y, Jia L, Zheng Y, Li W. Salivary exosomes: emerging roles in systemic disease. Int J Biol Sci 2018;14:633–43.
  • Chiabotto G, Gai C, Deregibus MC, Camussi G. Salivary extracellular vesicle-associated exRNA as cancer biomarker. Cancers 2019;11:pii: E891.
  • 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.
  • 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.
  • Nonaka T, Wong D. Saliva-exosomics in cancer: molecular characterization of cancer-derived exosomes in saliva. Enzymes 2017;42:125.
  • Zhan C, Yang X, Yin X, Hou J. Exosomes and other extracellular vesicles in oral and salivary gland cancers. Oral Dis 2019.