Next Article in Journal
Resistance to BRAF Inhibitors: EZH2 and Its Downstream Targets as Potential Therapeutic Options in Melanoma
Next Article in Special Issue
The Therapeutic Potential of a Strategy to Prevent Acute Myeloid Leukemia Stem Cell Reprogramming in Older Patients
Previous Article in Journal
Does the Blood–Brain Barrier Integrity Change in Regard to the Onset of Fetal Growth Restriction?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Roles of Exosomes in the Diagnose, Development and Therapeutic Resistance of Oral Squamous Cell Carcinoma

1
The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
2
Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 1968; https://doi.org/10.3390/ijms24031968
Submission received: 9 December 2022 / Revised: 11 January 2023 / Accepted: 16 January 2023 / Published: 19 January 2023

Abstract

:
Oral cancer is one of the most common cancers worldwide, of which more than half of patients are diagnosed at a locally advanced stage with poor prognosis due to recurrence, metastasis and resistant to treatment. Thus, it is imperative to further explore the potential mechanism of development and drug resistance of oral cancer. Exosomes are small endosome-derived lipid nanoparticles that are released by cells. Since the cargoes of exosomes were inherited from their donor cells, the cargo profiles of exosomes can well recapitulate that of their donor cells. This is the theoretical basis of exosome-based liquid biopsy, providing a tool for early diagnosis of oral cancer. As an important intracellular bioactive cargo delivery vector, exosomes play a critical role in the development of oral cancer by transferring their cargoes to receipt cells. More importantly, recent studies have revealed that exosomes could induce therapy-resistance in oral cancer through multiple ways, including exosome-mediated drug efflux. In this review, we summarize and compare the role of exosomes in the diagnosis, development and therapy-resistant of oral cancer. We also highlight the clinical application of exosomes, and discuss the advantages and challenges of exosomes serving as predictive biomarker, therapy target and therapy vector in oral cancer.

1. Introduction

Oral cancer is one of the top 10 malignant tumors worldwide, leading to over 177,000 deaths annually [1]. More than 90% of oral cancer cases are histologically oral squamous cell carcinoma (OSCC). Accumulating evidence shows that early OSCC patients have a favorable prognosis, with 5-year survival rates up to 80% [2]. Unfortunately, more than half of OSCC patients are diagnosed at a locally advanced stage (Ⅲ/Ⅳ) [3]. Compared with their early counterparts, patients with OSCC in stage Ⅲ/Ⅳ had a significantly poor prognosis and 5-year survival rates less than 30% due to recurrence and metastasis [4]. In addition, despite progress in the available treatments for advanced OSCC [5,6,7,8], the 5-year survival rate has not improved significantly during the past decades. Therefore, it is a major concern to explore the potential mechanism of recurrence and metastasis of OSCC. In recent years, an increasing number of studies have demonstrated that exosomes were closely associated with the progression of various types of cancer including OSCC.
Exosomes are small endosome-derived lipid nanoparticles that are released by both tumor cells and non-tumor cells. Exosomes carry different bioactive molecules and can deliver those cargoes to receipt cells to participate in many biological processes, including the recurrence and metastasis of cancer. Since the cargoes of exosomes are inherited from their donor cells, the cargo profiles of exosomes can well recapitulate that of their donor cells. This is the theoretical basis of exosome-based liquid biopsy. Previous studies [9], including ours [10,11,12], have shown the considerable difference of exosomes between OSCC patients and healthy donors, providing great potential for disease diagnosis. Exosomes are emerging as a new type of cancer biomarkers in recent decades. In addition, as an important intracellular bioactive cargoes delivery vector, exosomes play a critical role in the development of OSCC (e.g., recurrence, metastasis, immune response, angiogenesis and epithelial-mesenchymal transition) by transferring their cargoes to receipt cells [13]. Further, in recent decades, researches [14] have revealed that exosomal contents affect antitumor therapy by regulating sensitivity of the tumors to treatment, usually leading to resistance to therapy and subsequently recurrence and metastasis. In this review, we summarize the role of exosomes in the diagnosis, prognosis, development and treatment of OSCC. In addition, we also highlight the clinical application of exosomes, and discuss the advantages and challenges of exosomes in diagnosis, therapy target and therapy vector in OSCC.

2. Biogenesis and Classification of Exosomes

Extracellular vesicles (EVs) are nano-sized membranous vesicles comprising exosomes, microvesicles and apoptotic bodies. Exosomes, a small extracellular vesicle subtype (30–150 nm in diameter), are formed by activating endosomal pathway. As illustrated in Figure 1, early endosomes are formed through the invagination of the plasma membrane. As endosomes mature, numerous multivesicular bodies (MVBs) can arise by the inward budding of the late endosomal limiting membrane. Finally, the MVBs can fuse with plasma membranes to release contained exosomes or undergo intracellular degradation within lysosomes and autophagosomes. Microvesicles, the larger EVs fraction (100–1000 nm in diameter), are generated directly from the out budding or shedding of the plasma membrane. Apoptotic bodies, the largest EVs subtypes (50–2000 nm in diameter), are formed by apoptotic cell during shrinkage and death [15]. The biogenesis of EVs is represented schematically in Figure 1.
EVs are involved in the interaction between cells, whereas each subpopulation has different biofunctions. Exosomes, the critical mediators of intercellular communication, participate in various biological processes including cell adhesion [16,17], coagulation [18,19], immune response and cell transdifferentiation [20]. Exosomes are also involved in the development of multiple diseases [21,22], especially a variety of tumors [23]. Exosomes play an important role in regulating tumor initiation, growth and therapy. Similarly, microvesicles are involved in various pathophysiological processes. Apoptotic bodies are essential to maintain homeostasis in vivo [24,25]. In addition, different subpopulations of EVs exhibit high heterogeneity in their physical properties. The characteristics of different subsets of EVs are summarized in Table 1.
Regrettably, by virtue of the overlap size and difficult traceability, the taxonomic utility of EVs is limited. In addition, although increasing studies have sought to distinguish EVs subtypes [26,27], there is actually no single reliable marker for discriminating between exosomes and microvesicles. In 2018, the International Society for Extracellular Vesicles (ISEV) suggested that EVs could be subdivided into small extracellular vesicles (<200 nm) and large extracellular vesicles (>200 nm) [28]. Small extracellular vesicles (sEVs) are mainly composed of exosomes. Large extracellular vesicles (lEVs) are mixture of microvesicles and apoptotic bodies.
Table 1. Characteristics of different exosomes, microvesicles and apoptotic bodies.
Table 1. Characteristics of different exosomes, microvesicles and apoptotic bodies.
CharacteristicExosomesMicrovesiclesApoptotic BodiesReferences
MorphologyCup-shapeHeterogeniesHeterogenies[29]
OriginEndosomePlasma membranePlasma membrane[30]
Size30–150 nm100–1000 nm50–2000 nm[30,31,32]
Content1. Nucleic acid (small RNA, DNA)
2. Protein (functional protein)
3. Lipid (glycolipids, free fatty acids)
1. Nucleic acid (RNA, DNA)
2. Protein (functional protein, organelle protein)
3. Lipid (ceramides, sphingomyelins)
1. Nucleic acid (rRNA, DNA)
2. Protein (histone, organelle protein)
3. Lipid
[33,34]
MarkerTetraspanin, TSG101, AlixUnknownApoptosis-related protein[35,36]
FunctionInvolved in various pathophysiological processesInvolved in various pathophysiological processesMaintain the stability of the internal environment[17,18,19,20,21,22,23,24]

3. Exosomes in the Diagnosis of OSCC

So far, tissue biopsy remains the gold standard for the diagnosis of oral cancer. Many OSCC are developed from different types of oral mucosal diseases including leukoplakia, erythema and lichen planus [37,38]. The process of carcinogenesis among different site of the same mucosal diseases is usually inconsistent. Thus, the diagnostic accuracy of biopsy, which is seriously affected by the sample sites of biopsy, is not absolute in these OSCC developed from oral mucosal diseases due to the limited size and different site of sample [39]. In addition, the development of OSCC from mucosal diseases is a prolonged process; it is difficult to decide when does the biopsy should be performed since it is unavoidable associated with invasion, pain and wounds. Thus, early diagnosis of OSCC that developed from mucosal diseases is highly challenging. There has been an increasing and sustained demand for new non-invasive, practical diagnostic strategies. Since exosomes inherit features from their mother cells in both physiological and pathological conditions, they show great potential to serve as diagnostic marker. Furthermore, they are widely present and remarkably stable in human biofluids including plasma and saliva. Exosome-based liquid biopsy has become a promising alternative for diagnosis and prognosis of OSCC (Table 2).

3.1. Exosomes Derived from Saliva

Saliva is presented in the oral cavity and direct contacts with oral cancer, reflecting the relevant state of oral disease and providing the possibility for early diagnosis of OSCC. Ogawa et al. have demonstrated that exosomes are highly abundant in human saliva [56]. In our previous research, we found that the level of salivary microvesicles (SMVs) was significantly increased in OSCC patients. Further analysis found that the elevated SMVs was correlated with prognosis, staging and clinical outcomes [40]. The profile of salivary exosomal miRNA differs considerably between OSCC patients and healthy persons, which may be a promising option for OSCC diagnosis [57,58]. Using qRT-PCR analysis, researchers identified a marked increase of miR-24-3p [41], miR-512-3p [42], miR-412-3p [42] and miR-31 [43] in salivary exosomes from patients with OSCC. Notably, Gai et al. found that miR-302b-3p and miR-517b-3p were highly expressed in exosomes from OSCC patients [42], while undetectable in exosomes from healthy controls. In addition to miRNA, protein cargo in salivary exosomes could also serve as a potential biomarker in the diagnosis and prognosis of OSCC [44]. Fontana et al. found 365 differential protein expression in salivary exosomes isolated from healthy controls and OSCC patients with or without lymph node metastasis (LNM) by performing a proteome quantitative SWATH-MS analysis [45]. Other studies have demonstrated that exosomal protein was associated with immune response, providing new insights into OSCC prognosis [46]. Recently, Zlotogorski-Hurvitz et al. showed the especial IR spectrum of salivary exosomes contributed to the diagnosis of early stage OSCC [47].

3.2. Exosomes Derived from Plasma

Blood is the only fluid in direct contact with all organs and that transports important information throughout the whole bodies [59,60,61]. Many studies have demonstrated the concentration and contents of exosomes in blood vary significantly in pathophysiological conditions. Plasma-derived exosomes have become a promising alternative for the diagnosis of OSCC [57]. We previously reported the significantly increased level of circulating exosomes in OSCC patients compared with healthy donors [10]. Further analysis revealed that the elevated level of exosomes promotes a hypercoagulable state in OSCC via releasing inflammatory factors. Squamous cell carcinoma antigen (SCCA) was a potential marker for diagnosis of OSCC [62]. Yang et al. found SCCA in plasma-derived exosomes was remarkably elevated after saponin treatment [48]. In addition, Li et al., concluded that ApoA1, CXCL7, PF4V1 and F13A1 from serum exosomes could serve as novel diagnosis biomarkers for OSCC with lymph node metastasis [49]. Some scholars found that higher expression of miR-155 [50], miR-21 [50], miR-130a [51], CD63 [52] and CAV1 [52] in serum exosomes was associated with lower survival in oral cancer. They concluded that exosomal miR-155 and miR-21 contributed to tumorigenesis by inhibiting both PTEN and Bcl-6 expression [50]. On the contrary, other scholars proposed that exosomal protein (ALDH7A1, CAD, CANT1, GOT1, MTHFD1, PYGB and ASRS) [53], miRNA (miR-126 [50] and miR-146a [54]) and mtDNA [63] were negatively correlated with survival outcome. As described above, plasma-derived exosome is emerging as a novel diagnostic marker for OSCC.

3.3. Exosomes Derived from Other Origins

In addition to saliva and plasma, exosomes derived from drainage fluid also provide guiding significance for the diagnosis and prognosis of OSCC. Drainage fluid enters lymph circulation through lymphatic vessels, playing an important role in immunity [64]. Wang et al., found 313 differential protein expression in drainage fluid-derived exosomes isolated from OSCC patients with or without lymph node metastasis by performing a proteome quantitative analysis [55]. They suggested drainage fluid-derived exosomes may serve as a potential metastasis marker for OSCC.

4. Exosomes in the Development of OSCC

In addition to cancer diagnosis, researchers found that exosomes affected the development of oral cancer by transportation of their contents to target cells [41,65]. However, exosome in body fluids is a hybrid of exosome secreted from multiple cells including tumor cells and non-tumor cells (e.g., immune cells and fibroblasts). Exosomes are significantly diverse in their bioactive cargoes among cell types, leading to different effects on the development of cancer. Thus, it is vital to investigate the impacts of exosomes from different cell sources on OSCC. The functions of exosomes in tumor development are represented schematically in Figure 2.

4.1. Tumor Cell-Derived Exosomes in OSCC

4.1.1. Exosomes-Mediated Malignization

Cancer cells could communicate with each other via tumor cell-derived exosomes (TEXs), affecting proliferation, migration and invasion of cells [66,67] and thereby prompting the malignization. Activation of EGF/EGFR signaling pathway promoted tumor progression and enhanced the malignant potential of OSCC cells through cellular uptake of TEXs [66]. Anti-EGFR agents may be effective for the treatment of patients with OSCC by blocking not only the direct EGF/EGFR signaling pathway but also uptake of TEXs through macropinocytosis. Studies have demonstrated that exosomal miRNA (miR-24-3p [41] and miR-21-5p [68]) remarkedly increased proliferation and viability of OSCC cells. The periodic circadian rhythm adjustment factor (PER1) is an important regulator in cell cycle [69]. Exosomal miR-24-3p could enhance cell proliferation by inhibiting PER1 expression [41]. Chen et al. concluded that exosomal miR-21-5p contributed to the growth rate of OSCC cells by regulating the PI3K/mTOR/STAT3 signaling pathway [68]. In addition, other researchers also found the increased cell invasiveness and migration in the recipient cells after co-incubation with TEXs. Exosomal miR-21 [50,70], miR-155 [50] and miR-200c-3p [71] played a supporting role in migration and invasion of tumor cells, which were significant processes for tumor metastasis. Li et al. found that tumor metastasis in mice was promoted by injection of miR-21-rich TEXs [70].

4.1.2. Exosomes-Mediated Immune Response

TEXs can regulate immune response through direct interaction with immune cells, including Natural killer (NK) cells, T cells and macrophages. The NK cell, a major innate immune cell, exerts a crucial role in early innate immune response of tumor [72,73]. More recently, NK cells have been considered to demonstrate immune memory, playing an important role in adaptive immune responses to tumors [74]. Wang et al., revealed that TEXs increased the cytotoxicity of NK cells toward OSCC cells [75]. They concluded that NAP1, which was found enriched in TEXs, activated NK cells through increasing RF3 expression in the target cell. T cells are the major effector cells of antitumor immunity. Previous study has demonstrated that exosomal PD-L1 promoted tumor growth through inhibiting the activation and function of T cells [76]. Similarly, other researchers found exosomal PD-L1 could reduce T cell infiltration in OSCC [67]. Macrophages, important innate immune cells, can be roughly divided into antitumor M1-like phenotype and pro-tumor M2-like phenotype. Cai et al. found exosomal miR-29a-3p promoted tumor growth by inducing M2 polarization of macrophages in OSCC [77]. Tumor-associated macrophages (TAMs) are the major tumor-infiltration immune cells [78]. Increased TAM infiltration (especially M2-like phenotype) is correlated with frequent metastasis in OSCC patients [79,80]. Increasing evidence have suggested that TEXs promoted tumor growth and metastasis by inducing M2 polarization of TAMs in OSCC. Exosomal CDC37 [81], HSP90 [81] and CMTM6 [82] play an important role in M2 polarization of TAMs. Moreover, exosomal THBS1 induces M1 polarization of TAMs, promoting future malignant phenotypes of OSCC [83].

4.1.3. Exosomes-Mediated Angiogenesis

Angiogenesis is the generation of new blood vessels, which is dependent on the proliferation, migration and invasion of vascular endothelial cells (ECs). Angiogenesis is a crucial process for tumor growth and metastasis [84]. Exosomes released by OSCC cells were able to be internalized by vascular endothelial cells, thereby enhancing angiogenesis. Our previous study also found that EVs in the circulation [11] and tumor tissues [85] of OSCC patients could facilitate angiogenesis by enhancing the proliferation, migration and invasion of ECs. Yan et al. further demonstrated that miR-130b-3p in OSCC cell-derived exosomes promoted the formation of new blood vessels via inhibiting PTEN expression [86].

4.1.4. Exosomes-Mediated Epithelial-to-Mesenchymal Transition

Epithelial-to-Mesenchymal Transition (EMT) is a key cellular process during which epithelial cells gain the mesenchymal phenotype. This process has important biological significance in embryonic development [87]. However, EMT is abnormally activated in diverse tumors, leading to malignant metastasis [88]. Additionally, the role of EMT in drug resistance [89] and immune escape [90] has been emphasized during recent decades. Unfortunately, the mechanism underlying EMT activation in tumors remains unclear. Nevertheless, a growing number of researches found TEXs-induced EMT activation in various tumors, including OSCC [91,92,93]. Fujiwara et al. reported the abnormal activation of EMT in OSCC after co-incubation with EGFR-rich TEXs [94]. Moreover, the activation of EMT was damaged by co-incubating with both EGFR-rich TEXs and EGFR inhibitor, suggesting that exosomal EGFR was a crucial regulator of EMT activation.

4.2. Non-Tumor Cell-Derived Exosomes in OSCC

In addition to TEXs, exosomes secreted by non-tumor cells (e.g., epithelial cells, fibroblasts) were able to regulate tumor development. Cui et al. uncovered exosomal miR-200c from epithelial cells inhibited the proliferation, migration and invasion of tumor cells [95]. The inhibitory effect of exosomal miR-200c could be reversed by the miR-200c inhibitor. However, the result was contradictory to exosomal miR-200c-3p originating from tumor cells, because a previous study [71] found the increased invasiveness in recipient tumor cells. Cancer-associated fibroblasts (CAFs) are one of the main components in tumor stroma [96]. Numerous studies have demonstrated that CAFs are associated with poor prognosis of tumors [97,98]. In OSCC, CAF-derived exosomes could promote tumor growth and metastasis by increasing migration [99], invasion [99] proliferation [100] and angiogenesis of cells [101]. Among them, exosomal miR-382-5p [99], miR-34a-5p [100] and heparan sulfate proteoglycans [101] play a crucial role in this process.

5. Exosomes in the Treatment of OSCC

Currently, surgery, chemotherapy, radiotherapy and immunotherapy are recognized as the main therapeutic regimens for OSCC patients. Despite various attempts, the overall survival rates of OSCC patients have not substantially improved in the last decades [102]. The treatment resistance is a major reason for treatment failure in OSCC. In addition to decreasing response rate, the acquisition of treatment resistance often results in tumor relapse, leading to poor prognosis in OSCC patients. Thus, further exploration of the potential mechanism of drug resistance is urgently needed. Researchers found that exosomes could induce therapy-resistant in proper context [103,104]. In Figure 3, we schematize mechanisms involved in exosome-mediated therapy-resistant.

5.1. Tumor Cell-Derived Exosomes-Mediated Therapy Resistant

TEXs could induce resistant to drug therapy and radiotherapy. Cisplatin (CDDP) and 5-fluorouracil (5-FU) were first-line chemotherapeutic drugs in OSCC treatment. Drug efflux constitutes an important mechanism in drug resistance of tumor cells. Recent studies demonstrated that tumor cells could increase drug efflux and thus decrease the accumulation of anticancer drugs by the secretion of exosomes. A proton pump inhibitor enhances the accumulation of drugs and the susceptibility of tumor cells to CDDP by reducing TEXs secretion [105]. Other authors have reported the supporting function of exosomal miR-21 [68,106], miR-155 [107], ZFAS1 [108] and circ-SCMH1 [109] in CDDP resistance. Kulkarni et al. found decreased expression of circulating exosomal miR-30a in OSCC patients with recurrence, and revealed the inhibitory effect of TEXs-derived miR-30a in CDDP resistance [110]. In addition to CDDP resistance, researchers [66,111] also found increased tolerance of 5-FU in OSCC cells after co-incubation with TEXs. Li et al. proposed exosomal APCDD1L-AS1 conferred 5-FU resistant to 5-FU sensitive cells via miR-1224-5p/nuclear receptor binding SET domain protein 2 (NSD2) axis [111]. In addition, few studies [112] also demonstrated the enhanced radioresistance in radiosensitive OSCC cells after co-incubating with miR-503-3p-rich exosomes.

5.2. Non-Tumor Cell-Derived Exosomes-Mediated Therapy Resistant

Similarly, exosomes derived from non-tumor cells also affected therapy efficacy. miR-200c in exosomes derived from normal tongue epithelial cells impaired drug resistant (Docetaxel) of OSCC cells [95]. On the contrary, Tomita et al. found macrophage-derived exosomes conferred drug resistance (including Cisplatin and 5-FU) to OSCC cells through the activation AKT/GSK-3β signaling pathway in vivo [113]. Qin et al. also found exosomal miR-196 released from CAF remarkably enhances CDDP resistance in OSCC cells (CAL27 cell) [114].

6. Applications and Challenges of Exosomes in OSCC

6.1. Application as Predictive Biomarker

Exosomes inherit features from their mother cells, thus providing a possibility for tumor surveillance. Exosomes are widely present in human biofluids and tissues. Among them, exosomes derived from plasma and saliva have been reported to be reliable markers for OSCC. In contrast to traumatic biopsy, the collection of saliva and blood is procedurally easy and non-invasive, thereby reducing infection rate and other side effects. In addition, collection of exosomes is not obviously impacted by cancer site. This process avoids interference caused by improper sampling, and meanwhile brings new possibilities for detection of deep tumors. In addition, the time point of sample collection can be selected throughout the treatment period, and is not limited to preoperative. Hence, exosome-mediated tumor prediction provides a practical method for real-time monitoring of tumor activity during antitumor therapy. Further, exosome-based detection is not in direct contact with tumor masse, avoiding irritation of tumor tissue, thus reducing the risk of metastasis of cancer caused by invasive biopsy. Moreover, compared to the liquid-based biopsy (e.g., free nucleic acid), the lipid membrane of exosomes protects exosomal contents from degradation, providing a more reliable outcome.
Exosomes are emerging as new type of cancer biomarker; however, some obstacles toward the practical application of exosomes in the clinic exist. Cargo within exosomes differs considerably among healthy donors and OSCC patients with and without good prognosis. However, the differential exosomal contents exhibit high heterogeneity. There is no consensus on the selection of differential content and definition of detectable difference. In addition, non-tumor cell derived exosomes are abundant in biofluids [115,116,117], exacerbating the complexity of molecular source, thereby bringing obstacles for the identification of harbored markers and low abundance markers [118]. In contrast to plasma, saliva is considered as a special fluid associated with oral cancer. To further standardize the application of exosomes, it is vital to summarize the characters and challenges of salivary exosomes.
Saliva mainly concentrates in the oral cavity and is in direct contact with oral cancer, reflecting the relevant state of oral disease, providing the possibility for early oral cancer diagnosis. In contrast to plasma, salivary exosome-secreting cells are mainly tumor cells and oral mucosal cells, increasing concentrations of exosome-based biomarkers, thus enhancing the sensitivity and specificity for tumor detection. Additionally, salivary exosomal proteins are relatively simple [119], further facilitating the identification of exosome-based biomarkers. With regard to multimarker detection, our previous research [12] designed a one-step strategy for diagnosing oral ulcers and oral cancer. By using magnetic capture, EpCAM+ (oral cancer derived) and CD45+ exosomes (oral ulcers derived) are easily separated.
To further assess clinical applications of saliva-derived exosomes, multiple problems need to be considered. First, saliva-derived exosomes directly interact with the extraoral environment and are affected by various environmental factors. The human oral cavity contains complex microbial communities. Keller et al. [120] demonstrated that bacteria interfere with the expression of exosomal protein and secretion of exosomes. The intraoral environment (such as PH, temperature) could also be changed by daily activities such as drinking, eating and speaking. Second, the isolation method of salivary exosomes needs further optimization. At present, ultracentrifugation is the most commonly used method. However, saliva remains sticky after ultracentrifugation, which may impact functional properties of exosomes. Hence, proper pretreatment is needed for the saliva supernatant. Notably, Ohshiro et al. [121] proposed that initial saliva processing affected detection of tumor marker. The amount of saliva can also be affected by antitumor therapy, bringing difficulties for collection of exosomes. For example, irreversible hyposalivation is common in oral cancer patients after radiotherapy.

6.2. Application as Therapy Target

In this review, we summarize the role of exosomes in tumor development and treatment. TEXs enhanced tumor growth by promoting proliferation, migration, invasion and angiogenesis of tumor cells, while inhibiting antitumor immunity. Moreover, TEXs contributed to tumor progression by inducing therapeutic resistant. Hence, TEX-targeted therapies could be considered as promising options in OSCC treatment. Generally speaking, three pathways are included: inhibit biogenesis, tracking and secretion of TEXs, facilitate TEXs clearance and block function of TEXs. These strategies are represented schematically in Figure 4.

6.2.1. Inhibit Biogenesis, Tracking and Secretion of TEXs

The accurate mechanism of biogenesis, tracking and secretion of TEXs still remains unclear. Despite this, three crucial biological processes for exosome biogenesis have been identified: formation of endosome, degradation of MVB and release of MVB. Currently, various inhibitors targeting these processes have been reported. In previous studies, formation of ILV mainly depends on the ESCRT complexes and accessory proteins [122,123]; an ESCRT-independent [124,125] pattern also exists. Datta et al. [126] found the decreased formation of ESCRT-dependent exosomes after stimulation with manumycin A. Ceramide provided an important foundation for ESCRT-independent exosomes biogenesis [127]. GW4869, a noncompetitive inhibitor of nSMAse, effectively inhibited exosomes production by blocking ceramide synthesis. Additionally, the role of sulfisoxazole in lysosomal targeting of MVB inhibited exosomes generation [128]. Furthermore, Catalano et al. [129] proposed the inhibitory activity of cytochalasin D on release of MVB.
However, the majority of the drugs do not specifically target tumor cells, thus impairing the secretion of non-tumor cell-derived exosomes. Hence, a novel strategy to confer tumor-targeted ability is desperately required. By virtue of specific targeting property, nanosized drug delivery systems are considered as emerging options for cancer treatment. Studies have revealed that the accumulation of drugs in tumor cells can be enhanced by tumor-targeted nanoparticles [130]. Inhibition of TEXs biogenesis will be achieved specially when combining exosome inhibitors with the tumor-targeted drug delivery systems. In addition, since its target is enriched in tumor cell [131], manumycin A appeared to exert a stronger inhibitory effect on TEXs biogenesis [126]. Therefore, to develop better tumor-targeted agents, more studies are required to explore the underlying mechanisms of exosome generation, especially the unique mechanism in tumor cells.

6.2.2. Facilitate Clearance of TEXs

Previous research [132] summarized the important application of hemofiltration in TEXs clearance. TEXs are selectively captured and transported to blood circulation, and subsequently removed through the filtration of the microporous membrane. Mesoporous silica nanoparticles (MSNs) exhibit excellent biosafety. Xie et al. [133] functionalized MSNs with EGFR-targeting aptamers (MSN-AP) for specific binding to EGFR-rich exosomes. The bound exosomes are delivered and finally eliminated in the small intestine. However, hemofiltration, a traumatic treatment, inevitably exacerbates infection risk of patients. In addition, the ubiquitously clearance of both TEXs and non-tumor cell-derived exosomes hinder the future application of hemofiltration. With regard to MSN-AP, their applications also have limitations due to the nonhomogeneous expression of EGFR on tumor cells.
Previous studies reported that exosomes were principally metabolized in the liver, spleen and kidney, engulfed by mononuclear phagocyte system [134]. TEXs can enhance their circulation time by inhibiting macrophage-mediated phagocytosis [135]. Shimizu et al. [136] reported that the overexpressed CD47 (“don’t eat me” signal) on TEXs protect them from phagocytosis, extending their lifetime in circulation. In addition, PD-L1, which was recently shown to be another “don’t eat me” signal, was also highly expressed on exosomes released by various cancer cells. Therefore, blocking the “don’t eat me” signal on TEXs will be a promising strategy to enhance macrophage-mediated TEXs clearance.

6.2.3. Block Function of TEXs by Inhibiting Endocytosis

Endocytosis of exosome by receive cells is the key process for exosome to perform biological functions [137,138]. There are multiple pathways that participate in exosome internalization, but the detailed mechanism remains unclear. Exosomes endocytosis can roughly be divided into four forms: clathrin-mediated endocytosis, micropinocytosis, caveolin-dependent endocytosis and lipid raft-mediated endocytosis [139,140,141] (Figure 5). With regard to TEXs internalization, Christianson et al. [142] found TEXs uptake dependent largely on HSPGs (heparan sulfate proteoglycans). Christianson et al. [143] summarized the role of HSPGs in endocytosis. They demonstrated that HSPG endocytosis is not regulated by a specific pathway. Heparan sulfate analogues can attenuate tumor growth by regulating TEXs uptake [144]. In addition, Sasabe et al. [66] proposed that EGF may trigger uptake of TEXs by OSCC cells in a time and dose-dependent manner. They found EGFR inhibitors suppress tumor progression through micropinocytosis-mediated TEXs internalization.
Consequently, blocking endocytosis of TEXs is an excellent prospect for antitumor therapy in the future. However, TEXs internalization is not completely suppressed by blockers. More potential targets of TEXs internalization need future investigation. Additionally, blocker may also affect cell viability and survival. The influence on non-tumor cells should be emphasized at working concentrations. In addition, TEXs can also regulate tumor growth through uptake-independent mechanisms [145]. Thus, blocking function of TEXs by inhibiting endocytosis is still in its infancy.

6.3. Application as Therapy Vector

By virtue of the excellent biocompatibility, specific targeting property and favorable stability in human biofluids, exosomes were considered as promising drug delivery carriers. In our previous study [146], we constructed functional plasma-derived exosomes via efficient antitumor siRNA loading with electroporation. Compared with the control group, functional exosomes treated group had a better potency in OSCC treatment. Moreover, we monitored the distribution and metabolism, and revealed the natural tumor targeting of exosomes isolated from the peripheral blood of parents with OSCC. Other researchers [147,148] also indicated that TEXs could be used as vectors for drug delivery. Kase et al. [149] demonstrated that the selection of exosomes for tumor-targeting was a high priority. They developed a nanoplatform by embedding therapeutic siRNA into exosome isolated from normal fibroblasts. In addition, HEK293T cell-derived exosomes have also been reported as drug delivery vehicle in OSCC treatment [150]. In addition, many studies [151,152] have reported the controlled release of exosomes. Controlled degradation of hydrogels plays an essential role in this phenomenon.
In recent decades, exosomes have made notable progress in therapy vector [153,154,155]. However, there are still some issues which hamper their universal applications in clinical practice. Two major challenges facing their use as therapy vector are high production and efficient drug loading of exosomes. Additionally, therapeutic exosomes are mainly used in animal experiments at present. Dosages of exosomes exhibit high heterogeneity in their animal model, administration method and cellular origin [156]. For future clinical translation, complete guidelines on therapeutic exosome should be developed. In addition, efficiency of exosome-mediated targeted therapy is associated with origin of exosome [157]. TEXs are considered as promising candidates for therapeutic vectors [158]. However, the caution that TEXs contribute to tumor growth via multiple pathways should be taken when using TEXs as therapeutic vectors.

7. Conclusions

Exosomes have wide applicability in oral cancer diagnosis and treatment. Although tissue biopsy remains the gold standard for diagnosis of oral cancer, exosomes derived from saliva or plasma are considered as promising diagnostic biomarkers due to their homogeneity, convenience and safety. However, there is no consensus on the selection of a standard biomarker or a definition of detectable difference. In addition, targeting TEXs therapy is an emerging option in OSCC treatment; however, this novel strategy is still in its infancy. It is an inevitable issue to impair non-tumor cell-derived exosomes. Furthermore, exosomes have made notable progress as a therapy vector; however, commercial acquisition of exosomes is still a major challenge. Despite the excellent prospects, there is still a long way to realize clinical application of exosomes.

Author Contributions

Conceptualization, Z.-L.Y. and J.J.; writing—original draft preparation, S.S.; writing—review and editing, S.S. and Z.-L.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (81801842), and the Young Elite Scientist Sponsorship Program (YESS) by China Association for Science and Technology (CAST) (2018QNRC001).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. All the figures in this review were created by ourselves.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  2. van Lanschot, C.G.F.; Klazen, Y.P.; de Ridder, M.A.J.; Mast, H.; Ten Hove, I.; Hardillo, J.A.; Monserez, D.A.; Sewnaik, A.; Meeuwis, C.A.; Keereweer, S.; et al. Depth of invasion in early stage oral cavity squamous cell carcinoma: The optimal cut-off value for elective neck dissection. Oral Oncol. 2020, 111, 104940. [Google Scholar] [CrossRef] [PubMed]
  3. Pitiphat, W.; Diehl, S.R.; Laskaris, G.; Cartsos, V.; Douglass, C.W.; Zavras, A.I. Factors associated with delay in the diagnosis of oral cancer. J. Dent. Res. 2002, 81, 192–197. [Google Scholar] [CrossRef] [PubMed]
  4. Sciubba, J.J. Oral cancer. The importance of early diagnosis and treatment. Am. J. Clin. Dermatol. 2001, 2, 239–251. [Google Scholar] [CrossRef] [PubMed]
  5. Bhattacharya, D.; Sakhare, K.; Narayan, K.P.; Banerjee, R. The prospects of nanotherapeutic approaches for targeting tumor-associated macrophages in oral cancer. Nanomedicine 2021, 34, 102371. [Google Scholar] [CrossRef]
  6. Wang, S.C.; Wang, Y.Y.; Lin, L.C.; Chang, M.Y.; Yuan, S.F.; Tang, J.Y.; Chang, H.W. Combined Treatment of Sulfonyl Chromen-4-Ones (CHW09) and Ultraviolet-C (UVC) Enhances Proliferation Inhibition, Apoptosis, Oxidative Stress, and DNA Damage against Oral Cancer Cells. Int. J. Mol. Sci. 2020, 21, 6443. [Google Scholar] [CrossRef]
  7. Mosaddad, S.A.; Beigi, K.; Doroodizadeh, T.; Haghnegahdar, M.; Golfeshan, F.; Ranjbar, R.; Tebyanian, H. Therapeutic applications of herbal/synthetic/bio-drug in oral cancer: An update. Eur. J. Pharmacol. 2021, 890, 173657. [Google Scholar] [CrossRef]
  8. Tangthongkum, M.; Kirtsreesakul, V.; Supanimitjaroenporn, P.; Leelasawatsuk, P. Treatment outcome of advance staged oral cavity cancer: Concurrent chemoradiotherapy compared with primary surgery. Eur. Arch. Otorhinolaryngol. 2017, 274, 2567–2572. [Google Scholar] [CrossRef]
  9. Nonaka, T.; Wong, D.T.W. Liquid Biopsy in Head and Neck Cancer: Promises and Challenges. J. Dent. Res. 2018, 97, 701–708. [Google Scholar] [CrossRef]
  10. Ren, J.G.; Man, Q.W.; Zhang, W.; Li, C.; Xiong, X.P.; Zhu, J.Y.; Wang, W.M.; Sun, Z.J.; Jia, J.; Zhang, W.F.; et al. Elevated Level of Circulating Platelet-derived Microparticles in Oral Cancer. J. Dent. Res. 2016, 95, 87–93. [Google Scholar] [CrossRef]
  11. Ren, J.G.; Zhang, W.; Liu, B.; Man, Q.W.; Xiong, X.P.; Li, C.; Zhu, J.Y.; Wang, W.M.; Jia, J.; Sun, Z.J.; et al. Clinical Significance and Roles in Angiogenesis of Circulating Microparticles in Oral Cancer. J. Dent. Res. 2016, 95, 860–867. [Google Scholar] [CrossRef] [PubMed]
  12. Hong, S.-L.; Yu, Z.-L.; Bao, Z.-H.; Zhang, Q.-Y.; Zhang, N.; Tang, M.; Liu, S.-Q.; Jia, J.; Liu, K. One-step detection of oral ulcers and oral cancer derived exosomes on wedge-shaped and high magnetic field gradient mediated chip. Sens. Actuators B Chem. 2022, 357, 131403. [Google Scholar] [CrossRef]
  13. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Vasconcelos, M.H.; Caires, H.R.; Ābols, A.; Xavier, C.P.R.; Linē, A. Extracellular vesicles as a novel source of biomarkers in liquid biopsies for monitoring cancer progression and drug resistance. Drug Resist. Updat. 2019, 47, 100647. [Google Scholar] [CrossRef] [PubMed]
  15. Cotter, T.G.; Al-Rubeai, M. Cell death (apoptosis) in cell culture systems. Trends Biotechnol. 1995, 13, 150–155. [Google Scholar] [CrossRef] [PubMed]
  16. Wang, X.; Shah, F.A.; Vazirisani, F.; Johansson, A.; Palmquist, A.; Omar, O.; Ekström, K.; Thomsen, P. Exosomes influence the behavior of human mesenchymal stem cells on titanium surfaces. Biomaterials 2020, 230, 119571. [Google Scholar] [CrossRef]
  17. Jimenez, L.; Yu, H.; McKenzie, A.J.; Franklin, J.L.; Patton, J.G.; Liu, Q.; Weaver, A.M. Quantitative Proteomic Analysis of Small and Large Extracellular Vesicles (EVs) Reveals Enrichment of Adhesion Proteins in Small EVs. J. Proteome Res. 2019, 18, 947–959. [Google Scholar] [CrossRef]
  18. Berckmans, R.J.; Lacroix, R.; Hau, C.M.; Sturk, A.; Nieuwland, R. Extracellular vesicles and coagulation in blood from healthy humans revisited. J. Extracell. Vesicles 2019, 8, 1688936. [Google Scholar] [CrossRef]
  19. Souza, A.C.; Yuen, P.S.; Star, R.A. Microparticles: Markers and mediators of sepsis-induced microvascular dysfunction, immunosuppression, and AKI. Kidney Int. 2015, 87, 1100–1108. [Google Scholar] [CrossRef] [Green Version]
  20. Huang, R.; Hao, C.; Wang, D.; Zhao, Q.; Li, C.; Wang, C.; Yao, W. SPP1 derived from silica-exposed macrophage exosomes triggers fibroblast transdifferentiation. Toxicol. Appl. Pharmacol. 2021, 422, 115559. [Google Scholar] [CrossRef]
  21. Xu, Y.X.; Pu, S.D.; Li, X.; Yu, Z.W.; Zhang, Y.T.; Tong, X.W.; Shan, Y.Y.; Gao, X.Y. Exosomal ncRNAs: Novel therapeutic target and biomarker for diabetic complications. Pharmacol. Res. 2022, 178, 106135. [Google Scholar] [CrossRef]
  22. Larabi, A.; Barnich, N.; Nguyen, H.T.T. Emerging Role of Exosomes in Diagnosis and Treatment of Infectious and Inflammatory Bowel Diseases. Cells 2020, 9, 1111. [Google Scholar] [CrossRef] [PubMed]
  23. Sasaki, R.; Kanda, T.; Yokosuka, O.; Kato, N.; Matsuoka, S.; Moriyama, M. Exosomes and Hepatocellular Carcinoma: From Bench to Bedside. Int. J. Mol. Sci. 2019, 20, 1406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Li, X.; Liu, Y.; Liu, X.; Du, J.; Bhawal, U.K.; Xu, J.; Guo, L.; Liu, Y. Advances in the Therapeutic Effects of Apoptotic Bodies on Systemic Diseases. Int. J. Mol. Sci. 2022, 23, 8202. [Google Scholar] [CrossRef] [PubMed]
  25. Słomka, A.; Urban, S.K.; Lukacs-Kornek, V.; Żekanowska, E.; Kornek, M. Large Extracellular Vesicles: Have We Found the Holy Grail of Inflammation? Front. Immunol. 2018, 9, 2723. [Google Scholar] [CrossRef] [Green Version]
  26. Tucher, C.; Bode, K.; Schiller, P.; Claßen, L.; Birr, C.; Souto-Carneiro, M.M.; Blank, N.; Lorenz, H.M.; Schiller, M. Extracellular Vesicle Subtypes Released from Activated or Apoptotic T-Lymphocytes Carry a Specific and Stimulus-Dependent Protein Cargo. Front. Immunol. 2018, 9, 534. [Google Scholar] [CrossRef] [Green Version]
  27. Lischnig, A.; Bergqvist, M.; Ochiya, T.; Lässer, C. Quantitative Proteomics Identifies Proteins Enriched in Large and Small Extracellular Vesicles. Mol. Cell Proteom. 2022, 21, 100273. [Google Scholar] [CrossRef]
  28. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; 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. [Google Scholar] [CrossRef] [Green Version]
  29. Durcin, M.; Fleury, A.; Taillebois, E.; Hilairet, G.; Krupova, Z.; Henry, C.; Truchet, S.; Trötzmüller, M.; Köfeler, H.; Mabilleau, G.; et al. Characterisation of adipocyte-derived extracellular vesicle subtypes identifies distinct protein and lipid signatures for large and small extracellular vesicles. J. Extracell. Vesicles 2017, 6, 1305677. [Google Scholar] [CrossRef] [Green Version]
  30. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  31. Wang, Y.; Pang, J.; Wang, Q.; Yan, L.; Wang, L.; Xing, Z.; Wang, C.; Zhang, J.; Dong, L. Delivering Antisense Oligonucleotides across the Blood-Brain Barrier by Tumor Cell-Derived Small Apoptotic Bodies. Adv. Sci. 2021, 8, 2004929. [Google Scholar] [CrossRef] [PubMed]
  32. Kakarla, R.; Hur, J.; Kim, Y.J.; Kim, J.; Chwae, Y.J. Apoptotic cell-derived exosomes: Messages from dying cells. Exp. Mol. Med. 2020, 52, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Crescitelli, R.; Lässer, C.; Szabó, T.G.; Kittel, A.; Eldh, M.; Dianzani, I.; Buzás, E.I.; Lötvall, J. Distinct RNA profiles in subpopulations of extracellular vesicles: Apoptotic bodies, microvesicles and exosomes. J. Extracell. Vesicles 2013, 2, 20677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Haraszti, R.A.; Didiot, M.C.; Sapp, E.; Leszyk, J.; Shaffer, S.A.; Rockwell, H.E.; Gao, F.; Narain, N.R.; DiFiglia, M.; Kiebish, M.A.; et al. High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources. J. Extracell. Vesicles 2016, 5, 32570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Lötvall, J.; Hill, A.F.; Hochberg, F.; Buzás, E.I.; Di Vizio, D.; Gardiner, C.; Gho, Y.S.; Kurochkin, I.V.; Mathivanan, S.; Quesenberry, P.; et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: A position statement from the International Society for Extracellular Vesicles. J. Extracell. Vesicles 2014, 3, 26913. [Google Scholar] [CrossRef]
  36. van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  37. Shi, L.; Yang, Y.; Li, M.; Li, C.; Zhou, Z.; Tang, G.; Wu, L.; Yao, Y.; Shen, X.; Hou, Z.; et al. LncRNA IFITM4P promotes immune escape by up-regulating PD-L1 via dual mechanism in oral carcinogenesis. Mol. Ther. 2022, 30, 1564–1577. [Google Scholar] [CrossRef]
  38. Cai, X.; Zhang, J.; Zhang, H.; Li, T. Overestimated risk of transformation in oral lichen planus. Oral Oncol. 2022, 133, 106025. [Google Scholar] [CrossRef]
  39. Schilling, C.; Stoeckli, S.J.; Haerle, S.K.; Broglie, M.A.; Huber, G.F.; Sorensen, J.A.; Bakholdt, V.; Krogdahl, A.; von Buchwald, C.; Bilde, A.; et al. Sentinel European Node Trial (SENT): 3-year results of sentinel node biopsy in oral cancer. Eur. J. Cancer 2015, 51, 2777–2784. [Google Scholar] [CrossRef]
  40. Zhong, W.Q.; Ren, J.G.; Xiong, X.P.; Man, Q.W.; Zhang, W.; Gao, L.; Li, C.; Liu, B.; Sun, Z.J.; Jia, J.; et al. Increased salivary microvesicles are associated with the prognosis of patients with oral squamous cell carcinoma. J. Cell Mol. Med. 2019, 23, 4054–4062. [Google Scholar] [CrossRef]
  41. 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. Biomed Pharmacother. 2020, 121, 109553. [Google Scholar] [CrossRef] [PubMed]
  42. Gai, C.; Camussi, F.; Broccoletti, R.; Gambino, A.; Cabras, M.; Molinaro, L.; Carossa, S.; Camussi, G.; Arduino, P.G. Salivary extracellular vesicle-associated miRNAs as potential biomarkers in oral squamous cell carcinoma. BMC Cancer 2018, 18, 439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Liu, C.J.; Lin, S.C.; Yang, C.C.; Cheng, H.W.; Chang, K.W. Exploiting salivary miR-31 as a clinical biomarker of oral squamous cell carcinoma. Head Neck 2012, 34, 219–224. [Google Scholar] [CrossRef] [PubMed]
  44. Nakamichi, E.; Sakakura, H.; Mii, S.; Yamamoto, N.; Hibi, H.; Asai, M.; Takahashi, M. Detection of serum/salivary exosomal Alix in patients with oral squamous cell carcinoma. Oral Dis. 2021, 27, 439–447. [Google Scholar] [CrossRef]
  45. Fontana, S.; Mauceri, R.; Novara, M.E.; Alessandro, R.; Campisi, G. Protein Cargo of Salivary Small Extracellular Vesicles as Potential Functional Signature of Oral Squamous Cell Carcinoma. Int. J. Mol. Sci. 2021, 22, 11160. [Google Scholar] [CrossRef]
  46. Winck, F.V.; Prado Ribeiro, A.C.; Ramos Domingues, R.; Ling, L.Y.; Riaño-Pachón, D.M.; Rivera, C.; Brandão, T.B.; Gouvea, A.F.; Santos-Silva, A.R.; Coletta, R.D.; et al. Insights into immune responses in oral cancer through proteomic analysis of saliva and salivary extracellular vesicles. Sci. Rep. 2015, 5, 16305. [Google Scholar] [CrossRef] [Green Version]
  47. Zlotogorski-Hurvitz, A.; Dekel, B.Z.; Malonek, D.; Yahalom, R.; Vered, M. FTIR-based spectrum of salivary exosomes coupled with computational-aided discriminating analysis in the diagnosis of oral cancer. J. Cancer Res. Clin. Oncol. 2019, 145, 685–694. [Google Scholar] [CrossRef]
  48. Yang, M.; Ding, J.; Luo, Q.; Chen, X.; Chen, F. Improving the diagnostic efficacy of squamous cell carcinoma antigen for oral squamous cell carcinoma via saponin disruption of serum extracellular vesicles. Clin. Chim. Acta 2022, 525, 40–45. [Google Scholar] [CrossRef]
  49. Li, C.; Zhou, Y.; Liu, J.; Su, X.; Qin, H.; Huang, S.; Huang, X.; Zhou, N. Potential Markers from Serum-Purified Exosomes for Detecting Oral Squamous Cell Carcinoma Metastasis. Cancer Epidemiol. Biomarkers Prev. 2019, 28, 1668–1681. [Google Scholar] [CrossRef] [Green Version]
  50. Chen, C.M.; Chu, T.H.; Chou, C.C.; Chien, C.Y.; Wang, J.S.; Huang, C.C. Exosome-derived microRNAs in oral squamous cell carcinomas impact disease prognosis. Oral Oncol. 2021, 120, 105402. [Google Scholar] [CrossRef]
  51. He, T.; Guo, X.; Li, X.; Liao, C.; Wang, X.; He, K. Plasma-Derived Exosomal microRNA-130a Serves as a Noninvasive Biomarker for Diagnosis and Prognosis of Oral Squamous Cell Carcinoma. J. Oncol. 2021, 2021, 5547911. [Google Scholar] [CrossRef] [PubMed]
  52. Rodríguez Zorrilla, S.; Pérez-Sayans, M.; Fais, S.; Logozzi, M.; Gallas Torreira, M.; García García, A. A Pilot Clinical Study on the Prognostic Relevance of Plasmatic Exosomes Levels in Oral Squamous Cell Carcinoma Patients. Cancers 2019, 11, 429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Busso-Lopes, A.F.; Carnielli, C.M.; Winck, F.V.; Patroni, F.M.S.; Oliveira, A.K.; Granato, D.C.; Rap, E.C.; Domingues, R.R.; Pauletti, B.A.; Riaño-Pachón, D.M.; et al. A Reductionist Approach Using Primary and Metastatic Cell-Derived Extracellular Vesicles Reveals Hub Proteins Associated with Oral Cancer Prognosis. Mol. Cell Proteom. 2021, 20, 100118. [Google Scholar] [CrossRef]
  54. Flemming, J.P.; Hill, B.L.; Haque, M.W.; Raad, J.; Bonder, C.S.; Harshyne, L.A.; Rodeck, U.; Luginbuhl, A.; Wahl, J.K., 3rd; Tsai, K.Y.; et al. miRNA- and cytokine-associated extracellular vesicles mediate squamous cell carcinomas. J. Extracell. Vesicles 2020, 9, 1790159. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, J.; Man, Q.W.; Fu, Q.Y.; Zhong, N.N.; Wang, H.Q.; Li, S.R.; Gao, X.; Lin, H.; Su, F.C.; Bu, L.L.; et al. Preliminary Extracellular Vesicle Profiling in Drainage Fluid After Neck Dissection in OSCC. J. Dent. Res. 2022. [Google Scholar] [CrossRef] [PubMed]
  56. Ogawa, Y.; Miura, Y.; Harazono, A.; Kanai-Azuma, M.; Akimoto, Y.; Kawakami, H.; Yamaguchi, T.; Toda, T.; Endo, T.; Tsubuki, M.; et al. Proteomic analysis of two types of exosomes in human whole saliva. Biol. Pharm. Bull. 2011, 34, 13–23. [Google Scholar] [CrossRef] [Green Version]
  57. Lousada-Fernandez, F.; Rapado-Gonzalez, O.; Lopez-Cedrun, J.L.; Lopez-Lopez, R.; Muinelo-Romay, L.; Suarez-Cunqueiro, M.M. Liquid Biopsy in Oral Cancer. Int. J. Mol. Sci. 2018, 19, 1704. [Google Scholar] [CrossRef] [Green Version]
  58. Nair, S.; Tang, K.D.; Kenny, L.; Punyadeera, C. Salivary exosomes as potential biomarkers in cancer. Oral Oncol. 2018, 84, 31–40. [Google Scholar] [CrossRef]
  59. Zhong, W.; Edfors, F.; Gummesson, A.; Bergström, G.; Fagerberg, L.; Uhlén, M. Next generation plasma proteome profiling to monitor health and disease. Nat. Commun. 2021, 12, 2493. [Google Scholar] [CrossRef]
  60. Fernández-Pato, A.; Virseda-Berdices, A.; Resino, S.; Ryan, P.; Martínez-González, O.; Pérez-García, F.; Martin-Vicente, M.; Valle-Millares, D.; Brochado-Kith, O.; Blancas, R.; et al. Plasma miRNA profile at COVID-19 onset predicts severity status and mortality. Emerg. Microbes Infect. 2022, 11, 676–688. [Google Scholar] [CrossRef]
  61. Damanti, C.C.; Gaffo, E.; Lovisa, F.; Garbin, A.; Di Battista, P.; Gallingani, I.; Tosato, A.; Pillon, M.; Carraro, E.; Mascarin, M.; et al. MiR-26a-5p as a Reference to Normalize MicroRNA qRT-PCR Levels in Plasma Exosomes of Pediatric Hematological Malignancies. Cells 2021, 10, 101. [Google Scholar] [CrossRef] [PubMed]
  62. Czerninski, R.; Basile, J.R.; Kartin-Gabay, T.; Laviv, A.; Barak, V. Cytokines and tumor markers in potentially malignant disorders and oral squamous cell carcinoma: A pilot study. Oral. Dis. 2014, 20, 477–481. [Google Scholar] [CrossRef] [PubMed]
  63. Balachander, K.; Roy, A.; Priyadharsini, J.V.; Murugan, S.; Paramasivam, A. Mitochondrial DNA in circulating exosomes: A novel biomarker and potential therapeutic target for oral cancer. Oral Oncol. 2022, 128, 105857. [Google Scholar] [CrossRef] [PubMed]
  64. Clement, C.C.; Rotzschke, O.; Santambrogio, L. The lymph as a pool of self-antigens. Trends Immunol. 2011, 32, 6–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Zhao, C.; Zhang, G.; Liu, J.; Zhang, C.; Yao, Y.; Liao, W. Exosomal cargoes in OSCC: Current findings and potential functions. PeerJ 2020, 8, e10062. [Google Scholar] [CrossRef]
  66. Sasabe, E.; Tomomura, A.; Liu, H.; Sento, S.; Kitamura, N.; Yamamoto, T. Epidermal growth factor/epidermal growth factor receptor signaling blockage inhibits tumor cell-derived exosome uptake by oral squamous cell carcinoma through macropinocytosis. Cancer Sci. 2022, 113, 609–621. [Google Scholar] [CrossRef]
  67. Razzo, B.M.; Ludwig, N.; Hong, C.S.; Sharma, P.; Fabian, K.P.; Fecek, R.J.; Storkus, W.J.; Whiteside, T.L. Tumor-derived exosomes promote carcinogenesis of murine oral squamous cell carcinoma. Carcinogenesis 2020, 41, 625–633. [Google Scholar] [CrossRef]
  68. Chen, J.H.; Wu, A.T.H.; Bamodu, O.A.; Yadav, V.K.; Chao, T.Y.; Tzeng, Y.M.; Mukhopadhyay, D.; Hsiao, M.; Lee, J.C. Ovatodiolide Suppresses Oral Cancer Malignancy by Down-Regulating Exosomal Mir-21/STAT3/β-Catenin Cargo and Preventing Oncogenic Transformation of Normal Gingival Fibroblasts. Cancers 2019, 12, 56. [Google Scholar] [CrossRef] [Green Version]
  69. Gery, S.; Komatsu, N.; Baldjyan, L.; Yu, A.; Koo, D.; Koeffler, H.P. The circadian gene per1 plays an important role in cell growth and DNA damage control in human cancer cells. Mol. Cell 2006, 22, 375–382. [Google Scholar] [CrossRef]
  70. Li, L.; Li, C.; Wang, S.; Wang, Z.; Jiang, J.; Wang, W.; Li, X.; Chen, J.; Liu, K.; Li, C.; 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, 1770–1780. [Google Scholar] [CrossRef]
  71. Kawakubo-Yasukochi, T.; Morioka, M.; Hazekawa, M.; Yasukochi, A.; Nishinakagawa, T.; Ono, K.; Kawano, S.; Nakamura, S.; Nakashima, M. miR-200c-3p spreads invasive capacity in human oral squamous cell carcinoma microenvironment. Mol. Carcinog. 2018, 57, 295–302. [Google Scholar] [CrossRef]
  72. Shimasaki, N.; Jain, A.; Campana, D. NK cells for cancer immunotherapy. Nat. Rev. Drug Discov. 2020, 19, 200–218. [Google Scholar] [CrossRef] [PubMed]
  73. Chiossone, L.; Dumas, P.Y.; Vienne, M.; Vivier, E. Natural killer cells and other innate lymphoid cells in cancer. Nat. Rev. Immunol. 2018, 18, 671–688. [Google Scholar] [CrossRef] [PubMed]
  74. Cerwenka, A.; Lanier, L.L. Natural killer cell memory in infection, inflammation and cancer. Nat. Rev. Immunol. 2016, 16, 112–123. [Google Scholar] [CrossRef]
  75. Wang, Y.; Qin, X.; Zhu, X.; Chen, W.; Zhang, J.; Chen, W. Oral cancer-derived exosomal NAP1 enhances cytotoxicity of natural killer cells via the IRF-3 pathway. Oral Oncol. 2018, 76, 34–41. [Google Scholar] [CrossRef] [PubMed]
  76. Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef]
  77. Cai, J.; Qiao, B.; Gao, N.; Lin, N.; He, W. Oral squamous cell carcinoma-derived exosomes promote M2 subtype macrophage polarization mediated by exosome-enclosed miR-29a-3p. Am. J. Physiol. Cell Physiol. 2019, 316, C731–C740. [Google Scholar] [CrossRef] [PubMed]
  78. Chen, Y.; Song, Y.; Du, W.; Gong, L.; Chang, H.; Zou, Z. Tumor-associated macrophages: An accomplice in solid tumor progression. J. Biomed Sci. 2019, 26, 78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Guo, X.Y.; Zhang, J.Y.; Shi, X.Z.; Wang, Q.; Shen, W.L.; Zhu, W.W.; Liu, L.K. Upregulation of CSF-1 is correlated with elevated TAM infiltration and poor prognosis in oral squamous cell carcinoma. Am. J. Transl. Res. 2020, 12, 6235–6249. [Google Scholar]
  80. Haque, A.; Moriyama, M.; Kubota, K.; Ishiguro, N.; Sakamoto, M.; Chinju, A.; Mochizuki, K.; Sakamoto, T.; Kaneko, N.; Munemura, R.; et al. CD206(+) tumor-associated macrophages promote proliferation and invasion in oral squamous cell carcinoma via EGF production. Sci. Rep. 2019, 9, 14611. [Google Scholar] [CrossRef] [Green Version]
  81. Ono, K.; Sogawa, C.; Kawai, H.; Tran, M.T.; Taha, E.A.; Lu, Y.; Oo, M.W.; Okusha, Y.; Okamura, H.; Ibaragi, S.; et al. Triple knockdown of CDC37, HSP90-alpha and HSP90-beta diminishes extracellular vesicles-driven malignancy events and macrophage M2 polarization in oral cancer. J. Extracell. Vesicles 2020, 9, 1769373. [Google Scholar] [CrossRef]
  82. Pang, X.; Wang, S.S.; Zhang, M.; Jiang, J.; Fan, H.Y.; Wu, J.S.; Wang, H.F.; Liang, X.H.; Tang, Y.L. OSCC cell-secreted exosomal CMTM6 induced M2-like macrophages polarization via ERK1/2 signaling pathway. Cancer Immunol. Immunother. 2021, 70, 1015–1029. [Google Scholar] [CrossRef]
  83. Xiao, M.; Zhang, J.; Chen, W.; Chen, W. M1-like tumor-associated macrophages activated by exosome-transferred THBS1 promote malignant migration in oral squamous cell carcinoma. J. Exp. Clin. Cancer Res. 2018, 37, 143. [Google Scholar] [CrossRef] [Green Version]
  84. Kim, K.J.; Li, B.; Winer, J.; Armanini, M.; Gillett, N.; Phillips, H.S.; Ferrara, N. Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature 1993, 362, 841–844. [Google Scholar] [CrossRef]
  85. Yu, Z.L.; Liu, X.C.; Wu, M.; Shi, S.; Fu, Q.Y.; Jia, J.; Chen, G. Untouched isolation enables targeted functional analysis of tumour-cell-derived extracellular vesicles from tumour tissues. J. Extracell. Vesicles 2022, 11, e12214. [Google Scholar] [CrossRef]
  86. Yan, W.; Wang, Y.; Chen, Y.; Guo, Y.; Li, Q.; Wei, X. Exosomal miR-130b-3p Promotes Progression and Tubular Formation Through Targeting PTEN in Oral Squamous Cell Carcinoma. Front. Cell Dev. Biol. 2021, 9, 616306. [Google Scholar] [CrossRef]
  87. von Gise, A.; Pu, W.T. Endocardial and epicardial epithelial to mesenchymal transitions in heart development and disease. Circ. Res. 2012, 110, 1628–1645. [Google Scholar] [CrossRef]
  88. Lu, W.; Kang, Y. Epithelial-Mesenchymal Plasticity in Cancer Progression and Metastasis. Dev. Cell 2019, 49, 361–374. [Google Scholar] [CrossRef]
  89. Wang, J.; Wei, Q.; Wang, X.; Tang, S.; Liu, H.; Zhang, F.; Mohammed, M.K.; Huang, J.; Guo, D.; Lu, M.; et al. Transition to resistance: An unexpected role of the EMT in cancer chemoresistance. Genes Dis. 2016, 3, 3–6. [Google Scholar] [CrossRef] [Green Version]
  90. Jiang, Y.; Zhan, H. Communication between EMT and PD-L1 signaling: New insights into tumor immune evasion. Cancer Lett. 2020, 468, 72–81. [Google Scholar] [CrossRef]
  91. Yang, C.; Dou, R.; Wei, C.; Liu, K.; Shi, D.; Zhang, C.; Liu, Q.; Wang, S.; Xiong, B. Tumor-derived exosomal microRNA-106b-5p activates EMT-cancer cell and M2-subtype TAM interaction to facilitate CRC metastasis. Mol. Ther. 2021, 29, 2088–2107. [Google Scholar] [CrossRef]
  92. Yu, F.; Liang, M.; Huang, Y.; Wu, W.; Zheng, B.; Chen, C. Hypoxic tumor-derived exosomal miR-31-5p promotes lung adenocarcinoma metastasis by negatively regulating SATB2-reversed EMT and activating MEK/ERK signaling. J. Exp. Clin. Cancer Res. 2021, 40, 179. [Google Scholar] [CrossRef]
  93. Hsieh, C.H.; Tai, S.K.; Yang, M.H. Snail-overexpressing Cancer Cells Promote M2-Like Polarization of Tumor-Associated Macrophages by Delivering MiR-21-Abundant Exosomes. Neoplasia 2018, 20, 775–788. [Google Scholar] [CrossRef]
  94. Fujiwara, T.; Eguchi, T.; Sogawa, C.; Ono, K.; Murakami, J.; Ibaragi, S.; Asaumi, J.I.; Calderwood, S.K.; Okamoto, K.; Kozaki, K.I. Carcinogenic epithelial-mesenchymal transition initiated by oral cancer exosomes is inhibited by anti-EGFR antibody cetuximab. Oral Oncol. 2018, 86, 251–257. [Google Scholar] [CrossRef]
  95. Cui, J.; Wang, H.; Zhang, X.; Sun, X.; Zhang, J.; Ma, J. Exosomal miR-200c suppresses chemoresistance of docetaxel in tongue squamous cell carcinoma by suppressing TUBB3 and PPP2R1B. Aging 2020, 12, 6756–6773. [Google Scholar] [CrossRef]
  96. Yu, M.; Tannock, I.F. Targeting tumor architecture to favor drug penetration: A new weapon to combat chemoresistance in pancreatic cancer? Cancer Cell 2012, 21, 327–329. [Google Scholar] [CrossRef] [Green Version]
  97. Herrera, M.; Berral-González, A.; López-Cade, I.; Galindo-Pumariño, C.; Bueno-Fortes, S.; Martín-Merino, M.; Carrato, A.; Ocaña, A.; De La Pinta, C.; López-Alfonso, A.; et al. Cancer-associated fibroblast-derived gene signatures determine prognosis in colon cancer patients. Mol. Cancer 2021, 20, 73. [Google Scholar] [CrossRef]
  98. Li, B.L.; Lu, W.; Qu, J.J.; Ye, L.; Du, G.Q.; Wan, X.P. Loss of exosomal miR-148b from cancer-associated fibroblasts promotes endometrial cancer cell invasion and cancer metastasis. J. Cell Physiol. 2019, 234, 2943–2953. [Google Scholar] [CrossRef]
  99. Sun, L.P.; Xu, K.; Cui, J.; Yuan, D.Y.; Zou, B.; Li, J.; Liu, J.L.; Li, K.Y.; Meng, Z.; Zhang, B. Cancer-associated fibroblast-derived exosomal miR-382-5p promotes the migration and invasion of oral squamous cell carcinoma. Oncol. Rep. 2019, 42, 1319–1328. [Google Scholar] [CrossRef]
  100. Li, Y.Y.; Tao, Y.W.; Gao, S.; Li, P.; Zheng, J.M.; Zhang, S.E.; Liang, J.; Zhang, Y. Cancer-associated fibroblasts contribute to oral cancer cells proliferation and metastasis via exosome-mediated paracrine miR-34a-5p. EBioMedicine 2018, 36, 209–220. [Google Scholar] [CrossRef] [Green Version]
  101. Li, J.; Liu, X.; Zang, S.; Zhou, J.; Zhang, F.; Sun, B.; Qi, D.; Li, X.; Kong, J.; Jin, D.; et al. Small extracellular vesicle-bound vascular endothelial growth factor secreted by carcinoma-associated fibroblasts promotes angiogenesis in a bevacizumab-resistant manner. Cancer Lett. 2020, 492, 71–83. [Google Scholar] [CrossRef]
  102. Sun, L.; Xu, Y.; Zhang, X.; Gao, Y.; Chen, J.; Zhou, A.; Lu, Q.; Wang, Z.; Shao, K.; Wu, H.; et al. Mesenchymal Stem Cells Functionalized Sonodynamic Treatment for Improving Therapeutic Efficacy and Compliance of Orthotopic Oral Cancer. Adv. Mater. 2020, 32, e2005295. [Google Scholar] [CrossRef]
  103. Law, Z.J.; Khoo, X.H.; Lim, P.T.; Goh, B.H.; Ming, L.C.; Lee, W.L.; Goh, H.P. Extracellular Vesicle-Mediated Chemoresistance in Oral Squamous Cell Carcinoma. Front. Mol. Biosci. 2021, 8, 629888. [Google Scholar] [CrossRef]
  104. Yamaguchi, K.; Yamamoto, T.; Chikuda, J.; Shirota, T.; Yamamoto, Y. Impact of Non-Coding RNAs on Chemotherapeutic Resistance in Oral Cancer. Biomolecules 2022, 12, 284. [Google Scholar] [CrossRef]
  105. Khoo, X.H.; Paterson, I.C.; Goh, B.H.; Lee, W.L. Cisplatin-Resistance in Oral Squamous Cell Carcinoma: Regulation by Tumor Cell-Derived Extracellular Vesicles. Cancers 2019, 11, 1166. [Google Scholar] [CrossRef] [Green Version]
  106. Liu, T.; Chen, G.; Sun, D.; Lei, M.; Li, Y.; Zhou, C.; Li, X.; Xue, W.; Wang, H.; Liu, C.; et al. Exosomes containing miR-21 transfer the characteristic of cisplatin resistance by targeting PTEN and PDCD4 in oral squamous cell carcinoma. Acta Biochim. Biophys. Sin. 2017, 49, 808–816. [Google Scholar] [CrossRef] [Green Version]
  107. Kirave, P.; Gondaliya, P.; Kulkarni, B.; Rawal, R.; Garg, R.; Jain, A.; Kalia, K. Exosome mediated miR-155 delivery confers cisplatin chemoresistance in oral cancer cells via epithelial-mesenchymal transition. Oncotarget 2020, 11, 1157–1171. [Google Scholar] [CrossRef] [Green Version]
  108. Wang, X.; Hao, R.; Wang, F.; Wang, F. ZFAS1 Promotes Cisplatin Resistance via Suppressing miR-421 Expression in Oral Squamous Cell Carcinoma. Cancer Manag. Res. 2020, 12, 7251–7262. [Google Scholar] [CrossRef]
  109. Qiu, F.; Qiao, B.; Zhang, N.; Fang, Z.; Feng, L.; Zhang, S.; Qiu, W. Blocking circ-SCMH1 (hsa_circ_0011946) suppresses acquired DDP resistance of oral squamous cell carcinoma (OSCC) cells both in vitro and in vivo by sponging miR-338-3p and regulating LIN28B. Cancer Cell Int. 2021, 21, 412. [Google Scholar] [CrossRef]
  110. Kulkarni, B.; Gondaliya, P.; Kirave, P.; Rawal, R.; Jain, A.; Garg, R.; Kalia, K. Exosome-mediated delivery of miR-30a sensitize cisplatin-resistant variant of oral squamous carcinoma cells via modulating Beclin1 and Bcl2. Oncotarget 2020, 11, 1832–1845. [Google Scholar] [CrossRef]
  111. Li, S.; Shi, Z.; Fu, S.; Li, Q.; Li, B.; Sang, L.; Wu, D. Exosomal-mediated transfer of APCDD1L-AS1 induces 5-fluorouracil resistance in oral squamous cell carcinoma via miR-1224-5p/nuclear receptor binding SET domain protein 2 (NSD2) axis. Bioengineered 2021, 12, 7188–7204. [Google Scholar] [CrossRef]
  112. Yamana, K.; Inoue, J.; Yoshida, R.; Sakata, J.; Nakashima, H.; Arita, H.; Kawaguchi, S.; Gohara, S.; Nagao, Y.; Takeshita, H.; et al. Extracellular vesicles derived from radioresistant oral squamous cell carcinoma cells contribute to the acquisition of radioresistance via the miR-503-3p-BAK axis. J. Extracell. Vesicles 2021, 10, e12169. [Google Scholar] [CrossRef]
  113. Tomita, R.; Sasabe, E.; Tomomura, A.; Yamamoto, T. Macrophage-derived exosomes attenuate the susceptibility of oral squamous cell carcinoma cells to chemotherapeutic drugs through the AKT/GSK-3β pathway. Oncol. Rep. 2020, 44, 1905–1916. [Google Scholar] [CrossRef]
  114. Qin, X.; Guo, H.; Wang, X.; Zhu, X.; Yan, M.; Wang, X.; Xu, Q.; Shi, J.; Lu, E.; Chen, W.; 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. [Google Scholar] [CrossRef]
  115. Sharma, P.; Ludwig, S.; Muller, L.; Hong, C.S.; Kirkwood, J.M.; Ferrone, S.; Whiteside, T.L. Immunoaffinity-based isolation of melanoma cell-derived exosomes from plasma of patients with melanoma. J. Extracell. Vesicles 2018, 7, 1435138. [Google Scholar] [CrossRef]
  116. Theodoraki, M.N.; Hoffmann, T.K.; Jackson, E.K.; Whiteside, T.L. Exosomes in HNSCC plasma as surrogate markers of tumour progression and immune competence. Clin. Exp. Immunol. 2018, 194, 67–78. [Google Scholar] [CrossRef] [Green Version]
  117. Mondal, S.K.; Whiteside, T.L. Immunoaffinity-Based Isolation of Melanoma Cell-Derived and T Cell-Derived Exosomes from Plasma of Melanoma Patients. Methods Mol. Biol. 2021, 2265, 305–321. [Google Scholar]
  118. Li, W.; Li, C.; Zhou, T.; Liu, X.; Liu, X.; Li, X.; Chen, D. Role of exosomal proteins in cancer diagnosis. Mol. Cancer 2017, 16, 145. [Google Scholar] [CrossRef] [Green Version]
  119. Xiao, H.; Wong, D.T. Proteomic analysis of microvesicles in human saliva by gel electrophoresis with liquid chromatography-mass spectrometry. Anal. Chim. Acta 2012, 723, 61–67. [Google Scholar] [CrossRef]
  120. Keller, M.D.; Ching, K.L.; Liang, F.X.; Dhabaria, A.; Tam, K.; Ueberheide, B.M.; Unutmaz, D.; Torres, V.J.; Cadwell, K. Decoy exosomes provide protection against bacterial toxins. Nature 2020, 579, 260–264. [Google Scholar] [CrossRef]
  121. Ohshiro, K.; Rosenthal, D.I.; Koomen, J.M.; Streckfus, C.F.; Chambers, M.; Kobayashi, R.; El-Naggar, A.K. Pre-analytic saliva processing affect proteomic results and biomarker screening of head and neck squamous carcinoma. Int. J. Oncol. 2007, 30, 743–749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Hurley, J.H. ESCRTs are everywhere. Embo J. 2015, 34, 2398–2407. [Google Scholar] [CrossRef] [PubMed]
  123. Hurley, J.H.; Odorizzi, G. Get on the exosome bus with ALIX. Nat. Cell Biol. 2012, 14, 654–655. [Google Scholar] [CrossRef]
  124. Wei, D.; Zhan, W.; Gao, Y.; Huang, L.; Gong, R.; Wang, W.; Zhang, R.; Wu, Y.; Gao, S.; Kang, T. RAB31 marks and controls an ESCRT-independent exosome pathway. Cell Res. 2021, 31, 157–177. [Google Scholar] [CrossRef]
  125. van Niel, G.; Bergam, P.; Di Cicco, A.; Hurbain, I.; Lo Cicero, A.; Dingli, F.; Palmulli, R.; Fort, C.; Potier, M.C.; Schurgers, L.J.; et al. Apolipoprotein E Regulates Amyloid Formation within Endosomes of Pigment Cells. Cell Rep. 2015, 13, 43–51. [Google Scholar] [CrossRef]
  126. Datta, A.; Kim, H.; Lal, M.; McGee, L.; Johnson, A.; Moustafa, A.A.; Jones, J.C.; Mondal, D.; Ferrer, M.; Abdel-Mageed, A.B. Manumycin A suppresses exosome biogenesis and secretion via targeted inhibition of Ras/Raf/ERK1/2 signaling and hnRNP H1 in castration-resistant prostate cancer cells. Cancer Lett. 2017, 408, 73–81. [Google Scholar] [CrossRef]
  127. Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brügger, B.; Simons, M. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef]
  128. Im, E.J.; Lee, C.H.; Moon, P.G.; Rangaswamy, G.G.; Lee, B.; Lee, J.M.; Lee, J.C.; Jee, J.G.; Bae, J.S.; Kwon, T.K.; et al. Sulfisoxazole inhibits the secretion of small extracellular vesicles by targeting the endothelin receptor A. Nat. Commun. 2019, 10, 1387. [Google Scholar] [CrossRef] [Green Version]
  129. Catalano, M.; O’Driscoll, L. Inhibiting extracellular vesicles formation and release: A review of EV inhibitors. J. Extracell. Vesicles 2020, 9, 1703244. [Google Scholar] [CrossRef] [Green Version]
  130. Bala Tannan, N.; Manzari, M.T.; Herviou, L.; Da Silva Ferreira, M.; Hagen, C.; Kiguchi, H.; Manova-Todorova, K.; Seshan, V.; de Stanchina, E.; Heller, D.A.; et al. Tumor-targeted nanoparticles improve the therapeutic index of BCL2 and MCL1 dual inhibition. Blood 2021, 137, 2057–2069. [Google Scholar] [CrossRef]
  131. Sexton, R.E.; Mpilla, G.; Kim, S.; Philip, P.A.; Azmi, A.S. Ras and exosome signaling. Semin. Cancer Biol. 2019, 54, 131–137. [Google Scholar] [CrossRef]
  132. Li, Y.; Chen, Z.K.; Duan, X.; Zhang, H.J.; Xiao, B.L.; Wang, K.M.; Chen, G. Targeted inhibition of tumor-derived exosomes as a novel therapeutic option for cancer. Exp. Mol. Med. 2022, 54, 1379–1389. [Google Scholar] [CrossRef]
  133. Xie, X.; Nie, H.; Zhou, Y.; Lian, S.; Mei, H.; Lu, Y.; Dong, H.; Li, F.; Li, T.; Li, B.; et al. Eliminating blood oncogenic exosomes into the small intestine with aptamer-functionalized nanoparticles. Nat. Commun. 2019, 10, 5476. [Google Scholar] [CrossRef] [Green Version]
  134. Wan, Z.; Zhao, L.; Lu, F.; Gao, X.; Dong, Y.; Zhao, Y.; Wei, M.; Yang, G.; Xing, C.; Liu, L. Mononuclear phagocyte system blockade improves therapeutic exosome delivery to the myocardium. Theranostics 2020, 10, 218–230. [Google Scholar] [CrossRef]
  135. Qiu, X.; Li, Z.; Han, X.; Zhen, L.; Luo, C.; Liu, M.; Yu, K.; Ren, Y. Tumor-derived nanovesicles promote lung distribution of the therapeutic nanovector through repression of Kupffer cell-mediated phagocytosis. Theranostics 2019, 9, 2618–2636. [Google Scholar] [CrossRef]
  136. Shimizu, A.; Sawada, K.; Kobayashi, M.; Yamamoto, M.; Yagi, T.; Kinose, Y.; Kodama, M.; Hashimoto, K.; Kimura, T. Exosomal CD47 Plays an Essential Role in Immune Evasion in Ovarian Cancer. Mol. Cancer Res. 2021, 19, 1583–1595. [Google Scholar] [CrossRef]
  137. Milman, N.; Ginini, L.; Gil, Z. Exosomes and their role in tumorigenesis and anticancer drug resistance. Drug Resist. Updat. 2019, 45, 1–12. [Google Scholar] [CrossRef]
  138. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef]
  139. Costa Verdera, H.; Gitz-Francois, J.J.; Schiffelers, R.M.; Vader, P. Cellular uptake of extracellular vesicles is mediated by clathrin-independent endocytosis and macropinocytosis. J. Control Release 2017, 266, 100–108. [Google Scholar] [CrossRef]
  140. Tian, T.; Zhu, Y.L.; Zhou, Y.Y.; Liang, G.F.; Wang, Y.Y.; Hu, F.H.; Xiao, Z.D. Exosome uptake through clathrin-mediated endocytosis and macropinocytosis and mediating miR-21 delivery. J. Biol. Chem. 2014, 289, 22258–22267. [Google Scholar] [CrossRef] [Green Version]
  141. Nanbo, A.; Kawanishi, E.; Yoshida, R.; Yoshiyama, H. Exosomes derived from Epstein-Barr virus-infected cells are internalized via caveola-dependent endocytosis and promote phenotypic modulation in target cells. J. Virol. 2013, 87, 10334–10347. [Google Scholar] [CrossRef] [Green Version]
  142. Christianson, H.C.; Svensson, K.J.; van Kuppevelt, T.H.; Li, J.P.; Belting, M. Cancer cell exosomes depend on cell-surface heparan sulfate proteoglycans for their internalization and functional activity. Proc. Natl. Acad. Sci. USA 2013, 110, 17380–17385. [Google Scholar] [CrossRef]
  143. Christianson, H.C.; Belting, M. Heparan sulfate proteoglycan as a cell-surface endocytosis receptor. Matrix Biol. 2014, 35, 51–55. [Google Scholar] [CrossRef]
  144. Manandhar, S.; Park, J.; Kothandan, V.K.; Lee, J.; Alam, F.; Jee, J.P.; Hwang, J.; Byun, Y.; Hwang, S.R. Properties of Heparinoids Premixed with Tumor-Derived Extracellular Vesicles. Bioconjug Chem. 2018, 29, 3757–3767. [Google Scholar] [CrossRef]
  145. Muller, L.; Simms, P.; Hong, C.S.; Nishimura, M.I.; Jackson, E.K.; Watkins, S.C.; Whiteside, T.L. Human tumor-derived exosomes (TEX) regulate Treg functions via cell surface signaling rather than uptake mechanisms. Oncoimmunology 2017, 6, e1261243. [Google Scholar] [CrossRef]
  146. Yu, Z.L.; Zhang, W.; Zhao, J.Y.; Zhong, W.Q.; Ren, J.G.; Wu, M.; Zhang, Z.L.; Pang, D.W.; Zhao, Y.F.; Chen, G. Development of a Dual-Modally Traceable Nanoplatform for Cancer Theranostics Using Natural Circulating Cell-Derived Microparticles in Oral Cancer Patients. Adv. Funct. Mater. 2017, 27, 1703482. [Google Scholar] [CrossRef]
  147. Yang, Z.; Liu, D.; Zhou, H.; Tao, B.; Chang, L.; Liu, H.; Luo, H.; Wang, D.; Liu, W. A New Nanomaterial Based on Extracellular Vesicles Containing Chrysin-Induced Cell Apoptosis Through Let-7a in Tongue Squamous Cell Carcinoma. Front. Bioeng. Biotechnol. 2021, 9, 766380. [Google Scholar] [CrossRef]
  148. Sayyed, A.A.; Gondaliya, P.; Mali, M.; Pawar, A.; Bhat, P.; Khairnar, A.; Arya, N.; Kalia, K. MiR-155 Inhibitor-Laden Exosomes Reverse Resistance to Cisplatin in a 3D Tumor Spheroid and Xenograft Model of Oral Cancer. Mol. Pharm. 2021, 18, 3010–3025. [Google Scholar] [CrossRef]
  149. Kase, Y.; Uzawa, K.; Wagai, S.; Yoshimura, S.; Yamamoto, J.I.; Toeda, Y.; Okubo, M.; Eizuka, K.; Ando, T.; Nobuchi, T.; et al. Engineered exosomes delivering specific tumor-suppressive RNAi attenuate oral cancer progression. Sci. Rep. 2021, 11, 5897. [Google Scholar] [CrossRef]
  150. Deng, W.; Meng, Y.; Wang, B.; Wang, C.X.; Hou, C.X.; Zhu, Q.H.; Tang, Y.T.; Ye, J.H. In vitro experimental study on the formation of microRNA-34a loaded exosomes and their inhibitory effect in oral squamous cell carcinoma. Cell Cycle 2022, 21, 1775–1783. [Google Scholar] [CrossRef]
  151. Swanson, W.B.; Zhang, Z.; Xiu, K.; Gong, T.; Eberle, M.; Wang, Z.; Ma, P.X. Scaffolds with controlled release of pro-mineralization exosomes to promote craniofacial bone healing without cell transplantation. Acta Biomater. 2020, 118, 215–232. [Google Scholar] [CrossRef] [PubMed]
  152. Kwak, G.; Cheng, J.; Kim, H.; Song, S.; Lee, S.J.; Yang, Y.; Jeong, J.H.; Lee, J.E.; Messersmith, P.B.; Kim, S.H. Sustained Exosome-Guided Macrophage Polarization Using Hydrolytically Degradable PEG Hydrogels for Cutaneous Wound Healing: Identification of Key Proteins and MiRNAs, and Sustained Release Formulation. Small 2022, 18, e2200060. [Google Scholar] [CrossRef] [PubMed]
  153. Parada, N.; Romero-Trujillo, A.; Georges, N.; Alcayaga-Miranda, F. Camouflage strategies for therapeutic exosomes evasion from phagocytosis. J. Adv. Res. 2021, 31, 61–74. [Google Scholar] [CrossRef] [PubMed]
  154. Jung, K.O.; Jo, H.; Yu, J.H.; Gambhir, S.S.; Pratx, G. Development and MPI tracking of novel hypoxia-targeted theranostic exosomes. Biomaterials 2018, 177, 139–148. [Google Scholar] [CrossRef] [PubMed]
  155. Aimaletdinov, A.M.; Gomzikova, M.O. Tracking of Extracellular Vesicles’ Biodistribution: New Methods and Approaches. Int. J. Mol. Sci. 2022, 23, 11312. [Google Scholar] [CrossRef] [PubMed]
  156. Gupta, D.; Zickler, A.M.; El Andaloussi, S. Dosing extracellular vesicles. Adv. Drug Deliv. Rev. 2021, 178, 113961. [Google Scholar] [CrossRef]
  157. Sancho-Albero, M.; Navascués, N.; Mendoza, G.; Sebastián, V.; Arruebo, M.; Martín-Duque, P.; Santamaría, J. Exosome origin determines cell targeting and the transfer of therapeutic nanoparticles towards target cells. J Nanobiotechnology 2019, 17, 16. [Google Scholar] [CrossRef] [Green Version]
  158. Yong, T.; Zhang, X.; Bie, N.; Zhang, H.; Zhang, X.; Li, F.; Hakeem, A.; Hu, J.; Gan, L.; Santos, H.A.; et al. Tumor exosome-based nanoparticles are efficient drug carriers for chemotherapy. Nat. Commun. 2019, 10, 3838. [Google Scholar] [CrossRef]
Figure 1. Biogenesis and classification of exosomes. Early endosomes are formed through the invagination of the plasma membrane. As endosomes mature, numerous multivesicular bodies (MVBs) can arise by the inward budding of the late endosomal limiting membrane. Finally, the MVBs can fuse with plasma membranes to release contained exosomes. Microvesicles are generated directly from the out budding of the plasma membrane. Apoptotic bodies are formed by apoptotic cell during shrinkage and death.
Figure 1. Biogenesis and classification of exosomes. Early endosomes are formed through the invagination of the plasma membrane. As endosomes mature, numerous multivesicular bodies (MVBs) can arise by the inward budding of the late endosomal limiting membrane. Finally, the MVBs can fuse with plasma membranes to release contained exosomes. Microvesicles are generated directly from the out budding of the plasma membrane. Apoptotic bodies are formed by apoptotic cell during shrinkage and death.
Ijms 24 01968 g001
Figure 2. Exosomes in the development of OSCC. Tumor cell-derived exosomes (TEXs) support tumor development through promoting malignization of tumor cells (a), inhibiting antitumor immune responses (b), promoting angiogenesis (c) and promoting Epithelial-to-mesenchymal transition (EMT) (d). Cancer-associated fibroblasts (CAF)-derived exosomes contribute to tumor growth by promoting malignization of tumor cells (1) and angiogenesis (2). Epithelial cell-derived exosomes impair tumor growth through inhibiting malignization of tumor cells (3).
Figure 2. Exosomes in the development of OSCC. Tumor cell-derived exosomes (TEXs) support tumor development through promoting malignization of tumor cells (a), inhibiting antitumor immune responses (b), promoting angiogenesis (c) and promoting Epithelial-to-mesenchymal transition (EMT) (d). Cancer-associated fibroblasts (CAF)-derived exosomes contribute to tumor growth by promoting malignization of tumor cells (1) and angiogenesis (2). Epithelial cell-derived exosomes impair tumor growth through inhibiting malignization of tumor cells (3).
Ijms 24 01968 g002
Figure 3. Exosomes in the therapeutic resistance of OSCC. Exosomes derived from both tumor cells and non-tumor cells can regulate therapy resistance of OSCC cells through multiple ways.
Figure 3. Exosomes in the therapeutic resistance of OSCC. Exosomes derived from both tumor cells and non-tumor cells can regulate therapy resistance of OSCC cells through multiple ways.
Ijms 24 01968 g003
Figure 4. Challenges and prospects for TEX-targeted therapies. TEX-targeted therapies can be divided in three ways: inhibit biogenesis, tracking and secretion of TEXs (a), facilitate TEXs clearance (b) and block function of TEXs (c). For TEX inhibiting therapy, tumor-targeted drug delivery vectors will be a promising option to promote the tumor-targeted ability of exosomal inhibitor. For TEXs clearance therapy, side effects are inevitable due to invasive procedure. The novel strategy should be developed to further improve macrophage-mediated TEXs clearance. For TEXs blocking therapy, potential targets should be explored to decrease the impact on non-tumor cells.
Figure 4. Challenges and prospects for TEX-targeted therapies. TEX-targeted therapies can be divided in three ways: inhibit biogenesis, tracking and secretion of TEXs (a), facilitate TEXs clearance (b) and block function of TEXs (c). For TEX inhibiting therapy, tumor-targeted drug delivery vectors will be a promising option to promote the tumor-targeted ability of exosomal inhibitor. For TEXs clearance therapy, side effects are inevitable due to invasive procedure. The novel strategy should be developed to further improve macrophage-mediated TEXs clearance. For TEXs blocking therapy, potential targets should be explored to decrease the impact on non-tumor cells.
Ijms 24 01968 g004
Figure 5. Forms of TEXs endocytosis. TEXs endocytosis can roughly be divided into four forms: clathrin-mediated endocytosis (a), caveolin-dependent endocytosis (b), micropinocytosis (c) and lipid raft-mediated endocytosis (d).
Figure 5. Forms of TEXs endocytosis. TEXs endocytosis can roughly be divided into four forms: clathrin-mediated endocytosis (a), caveolin-dependent endocytosis (b), micropinocytosis (c) and lipid raft-mediated endocytosis (d).
Ijms 24 01968 g005
Table 2. Potential applications of exosomes from different samples collected from OSCC patients and healthy donors using different detecting methods.
Table 2. Potential applications of exosomes from different samples collected from OSCC patients and healthy donors using different detecting methods.
SourcesMethodsFindingsClinical ApplicationReferences
SalivaFCMIncreased number in OSCC; higher ratio of apoptotic to non-apoptotic exosomes in lower survivalDiagnosis
Prognosis
[40]
SalivaqRT-PCRHigher expression of miR-24-3p in OSCCDiagnosis[41]
SalivaqRT-PCRmiR-302b-3p and miR-517b-3p only expressed in OSCC; miR-512-3p and miR-412-3p expression level increased in OSCCDiagnosis[42]
SalivaqRT-PCRHigher expression of miR-31 in OSCC Diagnosis[43]
SalivaProteome analysisProteins expression level were correlated with OSCC diagnosis and prognosis Diagnosis
Prognosis
[44,45,46]
SalivaIR spectrumDifferential IR spectrum in OSCC patients compared with normal donorsDiagnosis[47]
PlasmaFCMIncreased number in OSCCDiagnosis[10]
PlasmaChemiluminescence immunoassay analyzerHigher expression of SCCA in OSCC Diagnosis[48]
PlasmaProteome analysisExpression levels of 4 proteins were correlated with metastasis OSCC Diagnosis[49]
PlasmaqRT-PCRHigher expression of miR-155 and miR-21 in OSCC; lower expression of miR-126 in OSCC with lower survivalDiagnosis
Prognosis
[50]
PlasmaqRT-PCRHigher expression of miR-130a in OSCC; higher expression of miR-130a in OSCC with lower survivalDiagnosis
Prognosis
[51]
PlasmaELISADecreased level of CD63+ exosomes or CAV-1+ exosomes in OSCC with higher survivalPrognosis[52]
PlasmaProteome analysis7 proteins expression level decreased in OSCC with lower survival Prognosis[53]
PlasmamiRNA-SeqLower expression of miR-146a was correlated with OSCC malignancy Prognosis[54]
Drainage fluidProteome analysis365 proteins expression level are correlated to lymph node metastasis in OSCCPrognosis[55]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shi, S.; Yu, Z.-L.; Jia, J. The Roles of Exosomes in the Diagnose, Development and Therapeutic Resistance of Oral Squamous Cell Carcinoma. Int. J. Mol. Sci. 2023, 24, 1968. https://doi.org/10.3390/ijms24031968

AMA Style

Shi S, Yu Z-L, Jia J. The Roles of Exosomes in the Diagnose, Development and Therapeutic Resistance of Oral Squamous Cell Carcinoma. International Journal of Molecular Sciences. 2023; 24(3):1968. https://doi.org/10.3390/ijms24031968

Chicago/Turabian Style

Shi, Shan, Zi-Li Yu, and Jun Jia. 2023. "The Roles of Exosomes in the Diagnose, Development and Therapeutic Resistance of Oral Squamous Cell Carcinoma" International Journal of Molecular Sciences 24, no. 3: 1968. https://doi.org/10.3390/ijms24031968

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop