Next Article in Journal
Oligo-Metastatic Cancers: Putative Biomarkers, Emerging Challenges and New Perspectives
Next Article in Special Issue
Circulating Tumor DNA in Head and Neck Squamous Cell Carcinoma
Previous Article in Journal
TRPV6 Calcium Channel Targeting by Antibodies Raised against Extracellular Epitopes Induces Prostate Cancer Cell Apoptosis
Previous Article in Special Issue
Neutrophil to Lymphocyte Ratio in Oropharyngeal Squamous Cell Carcinoma: A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Extracellular Vesicles as Biomarkers in Head and Neck Squamous Cell Carcinoma: From Diagnosis to Disease-Free Survival

Department of Otorhinolaryngology, Head and Neck Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(6), 1826; https://doi.org/10.3390/cancers15061826
Submission received: 21 February 2023 / Revised: 14 March 2023 / Accepted: 15 March 2023 / Published: 17 March 2023
(This article belongs to the Special Issue The Biomarkers and Detection of Head and Neck Cancer)

Abstract

:

Simple Summary

Extracellular vesicles are considered intracellular messengers that exchange proteins, lipids, various types of DNA and RNA, etc. They can be properly collected from many kinds of biofluids, such as blood and saliva. Compared to other substances obtained from liquid biopsy, such as cell-free DNA and circulating tumor cells, extracellular vesicles are a better choice to evaluate the disease since the genetic information is protected by their membranous structure, and thus they are more stable for detection. Extracellular vesicles contain less invasive property compared with other substances, and they are easier to collect from extraction with higher efficiency. Moreover, the roles of extracellular vesicles in multiple physiological and pathological processes are more well-known, entitling them to serve as biomarkers for certain cancers, including head and neck squamous cell carcinoma. This review aims to comprehensively present the current clinical trials at several key stages of tumor diagnosis and treatment, as well as their potential to dynamically monitor head and neck cancers during the whole clinical care process.

Abstract

Head and neck squamous cell carcinomas (HNSCCs) arising from different anatomical sites present with different incidences and characteristics, which requires a personalized treatment strategy. Despite the extensive research that has conducted on this malignancy, HNSCC still has a poor overall survival rate. Many attempts have been made to improve the outcomes, but one of the bottlenecks is thought to be the lack of an effective biomarker with high sensitivity and specificity. Extracellular vesicles (EVs) are secreted by various cells and participate in a great number of intercellular communications. Based on liquid biopsy, EV detection in several biofluids, such as blood, saliva, and urine, has been applied to identify the existence and progression of a variety of cancers. In HNSCC, tumor-derived EVs exhibit many functionalities by transporting diverse cargoes, which highlights their importance in tumor screening, the determination of multidisciplinary therapy, prediction of prognosis, and evaluation of therapeutic effects. This review illustrates the classification and formation of EV subtypes, the cargoes conveyed by these vesicles, and their respective functions in HNSCC cancer biology, and discloses their potential as biomarkers during the whole process of tumor diagnosis, treatment, and follow-up.

1. Introduction to Extracellular Vesicles and Head and Neck Squamous Cell Carcinoma

1.1. Head and Neck Squamous Cell Carcinoma

For the disease condition, more than 90% of head and neck cancers are histologically squamous cell carcinomas (HNSCCs) [1]. HNSCC commonly encompasses the cancers in the mucosal epithelium of the nasal cavity, paranasal sinuses, oral cavity, pharynx, larynx, and salivary glands [2,3]. Cancers arising from different anatomical sites exhibit various incidences: the estimated age-standardized incidence for HNSCC was 4.84 for the lip and oral cavity, 2.59 for laryngeal cancers, and 1.35 for nasopharyngeal cancers [4]. The overall incidence is still growing and is anticipated to rise by 30% by 2030 [1]. HNSCC is also more prevalent in males in certain global regions, such as Hong Kong, India, Central and Eastern Europe, and America [1]. Its prevalence varies across countries and regions with different oncogenic factors: in Southeast Asia and Australia, the consumption of certain carcinogen-containing substances, such as tobacco and alcohol, are the leading risk factors of HNSCC [5]; in the USA and Europe, it is largely attributed to human papillomavirus (HPV) infection [6,7]. HPV-positive HNSCC is associated with carcinomas of the oropharynx. As for the HPV-negative group, they are usually located in the oral cavity and larynx, closely associated with a history of smoking [2]. The reason that the early detection of HNSCC is hard to achieve is partly due to the lack of effective screening strategies, except for opportunistic, careful physical examinations [2].
In some cases of HNSCC, a timely intervention helps to prevent premalignant lesions from developing into HNSCC. Oral premalignant lesions present as leukoplakia (white patches) or erythroplakia (red patches), which are relatively easier to be observed than pathological changes at other sites [2]. However, for most cases, the patients present at a relatively advanced stage, and up to 10% of them undergo metastasis without any premalignant history or lesions [8,9].
The five-year overall survival rate of HNSCC remains dismal: increasing from just 55% during 1992−1996 to 66% in 2002−2006 and staying stagnant since then, especially in older patients [1]. The primary reasons are considered to be the late diagnosis, the high tendency of recurrence or metastasis, and resistance to therapies [10]. Current multidisciplinary treatment for HNSCC is composed of three modalities: surgery, radiotherapy, and anti-cancer drug therapy [10]. The treatment strategies are decided by many factors. For HNSCC in the oral cavity, the first approach is usually surgical resection, potentially followed by adjuvant chemoradiotherapy (CRT); for HNSCC arising from the pharynx or larynx, the primary choice will shift to CRT in lower-volume diseases [9]. Except for early stage HNSCC, the majority of therapeutic strategies involve multiple modalities [9]. Since the advent of cisplatin and the development of cetuximab for the treatment of HNSCC, the diversifying repertoire of anti-cancer drugs has become significantly important in alleviating locally advanced/resistant/metastatic HNSCC (LA- or R/M HNSCC) [11]. For those unresectable, cisplatin-refractory recurrent or metastatic HNSCCs, the immune checkpoint inhibitor pembrolizumab has been approved as a first-line therapy [12,13]. The exploration of the underlying mechanism of the tumor characteristics of HNSCC can shed light on the identification of certain molecular biomarkers, which could aid in the discovery of occult lesions, monitoring of tumor growth, prediction of metastasis, and evaluation of prognosis.

1.2. Extracellular Vesicles

All types of cells possess membrane vesicles, known as extracellular vesicles (EVs), not only in normal homeostatic status but also in pathological conditions as well. EVs used to be considered cellular wastes, but currently they are proving to be a pivotal approach to understanding intercellular communications [14]. Tumor cells, immune cells, epithelial cells, and other complex components of the tumor microenvironment (TME) could be the source of EVs [15].
EVs are divided into several subgroups by their origins, diameter, and surface protein markers, which mainly include exosomes (originating from the endosome, 30−150 nm) [16], microvesicles (MVs) (from the plasma membrane, 100−1000 nm) [17] and apoptotic bodies (from the plasma membrane, 50–5000 nm) (Figure 1) [18]. The classic dichotomy of EVs used to be size-dependent, but a recent expert consensus has placed increasing emphasis on the biogenesis and different molecular contents of EVs, which probably determine their sizes [19]. Exosomes possess relatively small volumes and are thought to participate in endosome biogenesis [20]. Early endosomal membranes sprout inwards to form intraluminal vesicles (ILVs), and ILVs are conveyed by multivesicular bodies (MVBs) during the endosomal maturation. After MVBs move towards the plasma membrane and then fuse with it, MVBs will consequently release exosomes into the extracellular space. The remaining MVBs will be lysed and degraded by lysosomes and hydrolytic enzymes [21,22]. In the late 1980s, exosomes were originally thought to be cell waste, but increasing research has documented the implications of exosomes in intercellular communication and immune regulations, and it has gradually been accepted that exosomes serve in numerous physiological and pathological processes rather than just in certain cell types [23]. MVs are generally larger than exosomes. The formation of MVs is greatly influenced by plasma membrane bending and budding during endocytosis, with its underlying mechanism inconclusive [24]. It used to be called “platelet dust”, since it was found to originate from the serum and participate in blood coagulation [25]. As more was learned about the features, certain MVs from cancer cells, called oncosomes, were proven to trigger or activate related pathways, which likely influence TME and metabolic phenotypes [26]. Besides the cargoes that are conveyed by exosomes and MVs, including DNA, RNA, proteins, and lipids, organelles are specific cargoes only delivered by apoptotic bodies [26].
EVs from different origins consist of various components and present with cell-type-specific proteins. Even from the same cell of origin, the contents of EVs vary with different stimuli and microenvironments, and consequently, they have corresponding fates and functions. Since EVs were discovered, there has been the notion that EVs could potentially contribute to oncogenesis, metastasis, resistance to chemotherapy, and therefore, consequently serve as tumor biomarkers. Rapid detection has become a hot spot in cancer research (Figure 2). However, there are still many barriers to realizing clinical transformation to the detection of biomarkers of EVs, especially in the common widely used isolation methods. For example, with ultracentrifugation, the representative traditional EV isolation method, the main problems are membrane damage and soluble protein contamination with the EV particles [27]. In density gradient ultracentrifugation, a recent common EV purification method, the isolated EV pellets yield a better quality product, but it is time consuming and has low efficiency [28,29]. The precipitation or commercial kit is relatively efficient at isolating EVs, but reports have mentioned that there are miRNA contaminations and a decreased EV subpopulation enrichment [28,29]. For the other EV isolation methods, due to the small sample size for clinical applicability or validation, the detection methods of ultrafiltration, size exclusion chromatography, and immunoaffinity capture have not been put into wide clinical use [28,30]. The leading method to detect EVs or related biomarkers in this field, such as DNA, RNA, and proteins conveyed by EVs, involves size-based and immunoaffinity-based detection. The principle is the combination method that accomplishes the attachment of the “probe”, such as the aptamers of specific EV membrane proteins or EV marker-bound magnetic beads, for detecting EVs or their intravesicular cargoes, to condense them together by the physical or chemical filters [30,31,32,33].
Extracellular vesicles are also called intercellular messengers since they are found to participate in the tumor microenvironment [34]. With EVs, the mutual communication between parent tumor cells and other types of cells is established and then amplified, for instance, paracrine interactions between tumor cells, immune cells (e.g., B-lymphocytes, natural killer cells [35]), epithelial cells, and stromal cells; juxtacrine signaling between tumor cells and tumor-infiltrating T-cells (TILs); and auto-communications between tumor cells and the exosomes released by themselves [36,37]. In the TME, exosomes released by tumor cells re-program some effector cells, such as cancer-associated fibroblasts (CAFs) and mesenchymal stem cells (MSCs), and consequently, they are pro-tumorigenic by promoting tumor growth and metastasis [22,38,39]. As major components of the TME, CAFs and exosomes promote the switch of the metabolic phenotype of cancer cells from mitochondrial oxidative phosphorylation to glycolysis and glutamine-dependent reduction carboxylation [38]. At the level of microRNAs (miRs), the expressions of miR-21, miR-378e, and miR-143 in exosomes derived from CAFs in breast cancer are higher than exosomes from normal fibroblasts, which increases the stemness, epithelial–mesenchymal transition (EMT), invasion and migration of these tumor cells [39]. Additionally, other connective cells, such as MSCs, also play a vital role in the TME. The promotion of tumor growth by MSC-derived EVs, similar to MSCs, was supposed by Zhu et al. [40]. Years later, current studies have been trying to make full use of their paracrine signaling pattern in order to use them as vesicles to deliver therapeutic RNA fragments [41].
Tumor-secreted EVs have their own specific integrin profiles that enable them to move to target organs, and they are vital participants in tumor metastasis [42]. They upregulate pro-inflammatory S100 molecules in the target organs, and their close interactions with metastatic tumor cells promote pre-metastatic niche development. For instance, melanoma-secreted exosomes promote vascular leakiness and inflammation, a hallmark of pre-metastatic niche formation, by upregulating factors such as S100a8, S100a9, and TNF-α [43]. Additionally, EV-RNAs from normal cells can also make the behaviors of malignancies more aggressive. In HCC cells, adipocytes release exosomal circRNA to reduce DNA damage of tumor cells by targeting miR-34a, resulting in metastasis in vivo [44]. Overall, EVs deliver cargoes to stimulate the vasculature and fuse with the parenchymal cells where pre-metastatic niches are colonized to facilitate tumor metastasis.
The regulation of EVs in the immune system, reducing T-cell activity, promotes tumor immune escape via programmed cell death protein-1 (PD-1) and its ligands via a programmed-death-ligand-1 (PD-L1)-related pathway. In oral squamous cell carcinoma (OSCC) patients, mitochondrial Lon, a multiple-function protein that is in charge of protein quality control, metabolism, mitophagy, and stress response, stimulates the cGAS-STING-TBKI pathway and induces interferon (IFN), consequently upregulating PD-L1 [45]. Additionally, EVs secreted by Lon can transport oxidized mitochondrial DNA and PD-L1, which inhibits T-cell activation in the TME and contributes to tumor immune escape [45]. In a similar way, exosomes found in lung cancer containing PD-L1 suppress IFN-γ secretion by Jurkat T-cells and impair immune functions by reducing cytokine production and inducing apoptosis in CD8+ T-cells [46].
The resistance of chemoradiotherapy in certain malignant tumors is enhanced by EVs through multiple mechanisms. In gastric cancers, exosomal miR-155-5p has been proven to induce chemo-resistant phenotype transformations by inhibiting GATA-binding protein 3 (GATA3) and tumor protein p53-inducible nuclear protein 1 (TP53INP1) [47]. As for ovarian cancer, exo-miR-21 mediates the interactions between stromal tissue and tumor cells and potentiates resistance to chemotherapy [48]. Radiotherapy (RT) is the primary treatment for nasopharyngeal carcinoma (NPC) and radio-resistance prevails as a dominant problem in clinical practice. EVs derived from Epstein–Barr-virus (EBV)-encoded latent-membrane-protein-1 (LMP1)-positive NPC cells significantly transmitted LMP1, which could activate the p38 MAPK signaling pathway to induce recipient tumor radio-resistance [49].
With the extensive study of EVs, their potential to work as biomarkers or drug carriers has been gradually explored. There are several merits to EVs as the carriers of therapeutic cargo, since they are naturally compatible with the body, relatively stable in circulation, ideally permeable through various membranes and barriers, and tissue- or cell-type-specific [50]. The targeting ligands are engineered to fuse with EV transmembrane proteins, which make up plasmid fusion constructs and fulfill the specificity of EVs. Therapeutic contents in EVs can be loaded in two modes: endogenous or exogenous. Endogenous loading is based on the overexpression of certain cargo in targeting source cells, and EVs are collected with the drug, whereas exogenous loading collects drug-free EVs [50]. Increasing numbers of trials have been implemented and verified the feasibility of EVs’ therapeutic applications [51,52,53,54]. Since the presence, quantity, and molecular content of EVs are believed to reflect the oncogenesis, progression, and malignant behaviors of tumors, many studies have verified their predictive values in diagnostic or prognostic strategy by in vivo and in vitro cancer biology experiments in different tumors, including cancers of the breast [55], lung [56], gastrointestinal tract [57,58], hepatobiliary system [59,60], prostate [61], head and neck [62,63,64]. The type of sample is determined by the lesion site, which leads to higher cost-efficiency, less invasion, and easier real-time measurement; for OSCC, plasma and saliva are the primary choices [62].
The endeavor to consider EVs as biomarkers in the clinical practice of HNSCC is still in its infancy, but recently, several researchers have supported that EVs with various molecular components have roles in tumorigenesis, progression, metastasis, recurrence, and the alteration of the TME [62,63,64].

2. Overview of Biomarkers in HNSCC—cfDNA, Circulating Tumor Cells, EVs

A biomarker can reflect specific biological features and molecular activities of certain diseases, such as in malignancies. Similar to α-fetoprotein in hepatic cell carcinoma [65], prostate-specific antigen in prostate cancer [66], or human chorionic gonadotropin-β in gestational trophoblastic tumors [67], squamous cell carcinoma antigen (SCC-Ag) is a potential biomarker for HNSCC, whereas an elevated level of SCC-Ag also suggests other diseases besides HNSCC, such as skin inflammation, autoimmune diseases and malignancies [68,69,70]. Given the lack of a sensitive and specific biomarker for HNSCC, researchers have continued exploring more potential biomarkers for HNSCC for early diagnosis, to predict therapeutic responses, to facilitate disease surveillance and to ultimately improve patients’ survival.
The biomarkers can be extracted from several kinds of patient samples, including tissue, plasma, saliva, or other biofluids. Tissue samples, which have the highest diagnostic value, are obtained by aspiration biopsy or other invasive procedures, which probably imposes greater mental stress on patients. Therefore, liquid biopsy and appropriate biomarkers are a better choice in light of the potential high efficiency and low invasiveness [71]. Additionally, their emphasis on the dynamic evolution of diseases partly alleviates the spatial and temporal heterogeneity in tumor tissue samples, which provides a general guide for treatment decision making and benefits patient monitoring and follow-ups [71]. The common liquid biopsy analytes include cell-free DNA (cfDNA), circulating tumor cells (CTCs), circulating RNAs, EVs, other proteins, and metabolites.

2.1. Cell-Free DNA

Cell-free DNA (cfDNA) is an extracellular fragment of DNA present in the body fluid, which was first discovered in the circulatory system by Mandel and Metais in 1948 [72]. Subsequently, circulating tumoral DNA (ctDNA), with obviously specific double-strand instability, was found in cfDNA in cancer patients’ DNA by Stroun et al. [73]. Besides tumor-originated, cfDNA, cell-free fetal DNA and cell-free circulating mitochondrial DNA, which have their own characteristics, have also been found. There are many ways for DNA fragments to be released from the inside of cells and finally into circulation or other biofluids, including through necrosis, apoptosis, active DNA release, neutrophil extracellular traps, EV transportation, erythroblast enucleation, and exogenous sources [74]. The first report that demonstrated the predictive values of cfDNA on tumor prognosis was published in the 1970s [75]. It showed that a higher cfDNA level indicates tumor metastasis, and it decreases after the radiation therapy. However, persistent or unchanged cfDNA levels reflect an ineffective treatment and serve as a sign of poor prognosis [75].
During the last few decades, liquid biopsy technology has been developed and its clinical workflow improved. cfDNA can be used for tumor progression, with plasma samples preferred over serum since there is too much non-tumor-derived cfDNA generated during the blood clotting by leukocyte lysis during serum sample preparation [71].
Recently, an increasing number of clinical studies have compared liquid-biopsy-based cfDNA detection and tumor-tissue-based aspiration, showing the distinctive advantages of liquid biopsies over tumor-tissue-based aspiration: less invasiveness, less heterogeneity, great reliability, and more rapid turnaround time [76,77,78,79]. Therefore, cfDNA screening is considered in the surveillance of tumor progression, tracing cancer metastasis, and guiding the selection of treatment strategies.
The clinical samples collected for cfDNA detection in HNSCC patients are mainly blood samples, including plasma and serum, but there are also several trials using saliva, which is more easily acquirable but requires more advanced detection methods [79,80,81]. Comparing HNSCC patients and healthy controls, the overall copy number of cfDNA in HNSCC patient samples is obviously higher than in healthy controls, which emphasizes the screening function and diagnostic value of cfDNA [82,83]. Moreover, a higher total copy number of cfDNA or a greater level of TP53 expression also indicate more aggressive tumor behaviors, such as LNM and advanced stage, in HNSCC patients [82,83]. Intriguingly, in HPV-related HNSCC cases, especially in oropharyngeal squamous cell carcinoma (OPSCC), the quantification of cf-HPV-DNA displays high specificity, positive and negative likelihood ratios, and a diagnostic odds ratio when distinguishing HPV-related OPSCC from normal controls and predicting the post-surgery recurrence [84].
However, so far there are certain limitations to reaching a conclusion about how the detection of cfDNA serves as an efficient tumor-screening tool. Most of the current studies are from North America and Europe, which may have biases by not including other regions, particularly in relation to HPV-negative OPSCC. Another problem is the lack of unified detection method for cfDNA, which is a common obstacle to utilizing a novel technique in clinical practices.

2.2. Circulating Tumor Cells, CTCs

CTCs are tumor cells shedding from the primary cancer site into blood circulation or the lymphatic system, which might extravasate and develop into metastatic sites [85]. Since the concept of CTCs was first proposed, many studies have shown that they are precursors of metastasis in certain cancer types [86,87,88,89]. Therefore, they are also considered to be an ideal tool to monitor tumor progression and therapeutic effects, which are of value in prognosis prediction and offer a choice of treatment strategy to avoid inappropriate surgical intervention or adjuvant drugs.
CTCs can be classified into several subtypes according to multiple factors, including epithelial markers, cell morphology, and apoptotic status. Epithelial-originated CTCs are distinguishable from other phenotypes due to the expression of EpCAM and cytokeratin [90,91,92]. Since mesenchymal CTCs lack cytokeratins and CD45, they are less likely to be isolated from the blood, which also makes them more resistant and more prone to metastasis [93,94]. Apoptotic CTCs result from the process of programmed cell death, the level of which provides a reference for evaluating treatment effects [95,96].
CTCs are few in quantity, up to 10 CTC per milliliter of whole blood compared to a few million white blood cells and a billion red blood cells [97]. Their half-life is less than 3 h, which can also be influenced by external conditions [97]. To achieve a high sensitivity, specificity, and reproducibility, CTC-detecting technologies keep improving, which are broadly classified into biological methods, physical methods, and hybrid methods [98,99,100]. As for the biological mechanism, the isolation of CTCs is based on the antigen–antibody combination in which the common markers include EpCAM as a cancer stem cell marker, and HER2 and PSA as the targets for breast and prostate cancer, respectively [90,91]. The CellSearch system, an immunomagnetic assay with magnetic nanoparticle-based separating technology, is the cleared U.S. Food and Drug Administration (FDA) biological methodology for CTC enumeration [101]. Physical methods generally capture CTCs by various size filtrations, and as the representative, the Parsortix® PC1 system was cleared by the FDA in May 2022 [102]. However, these technologies have their own drawbacks that may have some biases, for instance, the biological extraction of CTCs requires a sufficient expression of the known and selected marker on the cell surface, and physical filtration would probably overlook the other blood components whose diameters and properties are close to those of CTCs [92,100].
Tinhefer et al. and Garrel et al. successfully found the presence of CTCs in HNSCC, which was related to shorter disease-free survival (DFS) [103,104]. In addition, the number of CTCs extracted from peripheral blood could also serve as a predictor of tumor prognosis and a dynamic tracking tool to assess tumor tolerance to personalized therapy [89,105]. CTCs have been explored in patients with relapsing non-operable or metastatic HNSCC [103]. One study reported that the analysis of a patient’s peripheral blood samples collected on day 0 (D0), D7, and D21 after the intervention of chemotherapy and cetuximab dynamically reflected the tumor response to the therapy by CTC kinetics within the first week of treatment, and increasing or stable counts of CTCs suggested poor therapeutic effects and earlier tumor progression [103].
Currently, there are still some limitations and drawbacks for CTCs when acting as biomarkers in HNSCC. HNSCC is an epithelial-originated cancer, but in reality, EpCAM is not always present in every CTC derived from HNSCC cases due to the transition between epithelial and mesenchymal characteristics, which results in the high specificity but low sensitivity of the current detection methods [90,91,100,106]. More innovations in CTC enumeration techniques are needed, such as a new Epithelial ImmunoSPOT (EPISPOT) assay by adding anti-CD45 antibodies to alleviate the influence of leukocytes and enrich the number of CTCs for detection [103].

2.3. Extracellular Vesicles, EVs

Compared to CTCs, the content of EVs in circulation is relatively abundant. Many physicochemical techniques have been developed to isolate and extract EVs from body fluids, including ultracentrifugation, size-based filtration, polymer precipitation, and microfluidics-based isolation [107,108]. Additionally, antigen–antibody-interaction-based immunoaffinity methods have also been adapted to isolate EVs, where magnetic microbeads coated with specific antibodies significantly bind to certain surface markers and extract greater than 10-times-higher quantities of EVs than ultracentrifugation [107,109].
Saliva is an ideal sample type for liquid biopsy due to its easy collection and non-invasiveness, with multiple studies making full use of the components in saliva to screen for malignancy, evaluate progression and prognosis. A study focusing on the values of HPV-16 DNA in distinguishing tumor patients from healthy controls showed a relatively low sensitivity (about 50%) of saliva-based detection and stated that the sensitivity of HPV-16 DNA prediction can be improved by the combination of saliva-based and plasma-based samples (reaching up to 69.5%) [110]. However, as for EVs in the saliva sample, since they were first discovered and successfully isolated, a growing number of studies have demonstrated the advantages of saliva samples as the medium for EV detection; a high number of EVs in saliva makes it simple to collect and present significant differences between patients and the normal population [111,112,113]; the samples are much easier to store and require fewer procedures to complete the identification and quantification of EVs due to fewer protein components compared to plasma [114].
Different subtypes of EVs have shown value in tumor diagnosis. Zlotogorski-Hurvitz et al. successfully established computational-aided models for the Fourier-transform infrared spectra of salivary exosomes from OSCC and indicated that it exhibited both high sensitivity and specificity for diagnosis [115]. Functional signatures of the protein conveyed by small salivary EVs were developed to predict OSCC with the assistance of the sequential window acquisition of all the theoretical mass spectra and a subsequent bioinformatics analysis [116]. The investigations into MVs in HNSCC have been few, and their origins remains controversial. Kim disclosed that the samples collected from clinical patients and cell lines of OSCC contained FasL, a member of the tumor necrosis factor family, which participates in tumor progression and metastasis [117]. FasL-positive MVs can theoretically enhance the immune suppression and apoptosis of activated immune cells, and MVs can maintain a relatively high level in the serum of OSCC patients, where they may act as an indicator of the tumor stage [117,118].
There is no widely recognized biomarker for HNSCC in clinical practice so far, which is probably restricted by the current detection techniques, insufficient data support, and regional disparity due to economic and medical strength, and EVs are expected to lead to a breakthrough in this field.

3. EV Cargoes and Their Instructive Effects in HNSCC

In tumor-derived EVs (TDEs), the cargoes conveyed by EVs could be DNA molecules, different kinds of RNAs, proteins, and even artificially synthesized pharmacological compounds, which have different functions in numerous vital pathways in HNSCC biology.

3.1. DNA

Double-stranded DNA (dsDNA) delivered by EVs in circulation possesses the same features and oncogenic gene mutations as the primary tumor DNA [119]. KRAS and TP53 expression have both been found in circulating EVs and pancreatic cancerous cells, which confirm their potential to be tumor biomarkers [119,120]. However, the genetic information about the whole genome sequence carried by EV-DNA seems to be limited when compared to formalin-fixed paraffin-embedded (FFPE) tumor samples, since the efficiency of DNA extraction is be significantly impacted by the various methods of EV isolation and DNA extraction [121]. Therefore, so far, it is not appropriate to select EV-associated DNA alone as the marker for tumor progression [121]. However, novel research in this field is focused on the detection of viral DNA. As a pivotal transmitter in intercellular communication, EVs play a significant role in tumor cells and virally infected cells in their microenvironment [122,123]. Viruses can also make use of EVs to escape from immune attack and disseminate infections, which has also been confirmed by several reports that EVs containing EBV-originated DNA and miRNA could be detected in the peripheral blood samples of NPC patients [122,123]. More than 90% of NPCs in endemic regions are associated with EBV, a γ-Herpes virus with long, linear, double-stranded DNA genomes [124]. EBV infection is commonly asymptomatic, but when activated, EBV may present as inflammation, such as mononucleosis [125,126], and induce the development of NPC and other malignancies, including Burkitt’s lymphoma and NK/T-cell lymphoma [127,128]. In EBV-infected NPC patients, the virus persists as a type II latency [129], and the expression of certain genes, including EBNA1, LMP1, LMP2A, EBER 1 & 2, and BARTS, can be detected [130]. The expression of LMP1, LMP2A, BARTS, and EBV DNA have been found in and delivered by EVs [49,130,131], which is a potential indicator of viral infection and tumor progression.

3.2. RNA

Ribonucleic acids (RNAs) are classified by many methodologies, such as by function, including messenger RNA (mRNA), transfer RNA (tRNA), and ribosomal RNA (rRNA) [132]; or by length, including small RNA (shorter than 200 nt; e.g., some rRNA, tRNA, microRNA (miRNA), and small interfering RNA (siRNA)) [133] and long RNAs (greater than 200 nt, e.g., long non-coding RNA (lncRNA) and mRNA) [134]. There are also some special RNAs with unique structures, such as double-stranded RNA and circular RNA (circRNA) [135].
Ma et al. investigated the mediation of cell-to-cell communication by exosomal shuttle RNA; they also verified the regulation of tumor growth by miRNAs (exo-miR-17/20) by interacting with target downstream proteins, such as IL-8, CXCL1, CK8 and α-ENO [136]. Despite the lack of a coding function, miRNAs can induce mRNA degradation and greatly influence protein expression at the post-transcription level [137]. It has been suggested that an active sorting mechanism of miRNAs into EVs exists that confers differing EV-miRNA regulations of tumor behaviors [137]. MiRNA can be extracted from many kinds of biofluids, such as saliva. Gallo et al. demonstrated that the majority of miRNA was enveloped by exosomes, and could be detected in a just small amount of saliva; additionally, it more stably exists in the saliva with the protection by a bi-layer membranous structure, which facilitates its development as a biomarker for HNSCC [138,139]. Distinguishable levels of oncogenic miRNA are anticipated to facilitate the early detection of HNSCC, which has been illustrated by numerous studies, as shown in Table 1. There were also some studies that analyzed particular miRNAs rich in HPV(+) and HPV(-) HNSCCs, respectively. Ludwig et al. reported that miR-205-5p was exclusively expressed in HPV(+) oropharyngeal squamous cell carcinoma (OPSCC) cell lines (UD-SCC-2, UM-SCC47, UPCI: SCC90)-derived exosomes, and miR-1972 was extraordinarily presented in HPV(−) cell lines (PCI-13 and PCI-30) [140]. When it comes to EBV, it was confirmed that some viral miRNAs were selectively packaged into virus-derived exosomes so that the viral infection could be promoted and transferred [141]. The diverse effects of miRNA in each stage of HNSCC diagnosis and treatment have been overviewed, and a literature review based on the biological source of the samples of the clinical patient cohort is shown in Table 1.
LncRNA is a class of RNA longer than 200 nucleotides but without protein-coding capacity [134]. EVs could envelop and transport lncRNA from tumor cells to target cells, consequently influencing the expression of miRNA and changing the tumor microenvironment (TME) [142]. One of the most representative lncRNAs was HOTTIP, and it caused various changes according to its origin: the M1 macrophage-derived exosomal lncRNA HOTTIP proved to strongly suppressed tumor progression via the upregulation of the TLR5/NF-κB pathway [143]; however, when it was secreted by cancer stem cells in OSCC, it then promoted M2 macrophage polarization, inhibited CD4+ T-cell proliferation and IFN-γ production, and facilitated tumor progression and immunosuppression via the LAMC2-mediated PI3K/AKT pathway [144].
CircRNA possesses a covalently bonded structure with a closed loop and without 5′ caps or 3′ poly(A) tails. Due to its relatively stable structure, wide distribution, and tissue specificity, it is superior when selected to act as molecular diagnostic marker and drug treatment target [145]. There are, however, few reports on circRNA detection in HNSCC screening. In OSCC-derived EVs, circRNA proved to be stably present in abundance [146], to be oncogenic through the upregulation of certain circRNAs in HNSCC, such as circCORO1C via the let-7c-5p/PBX3 axis [147], circRNA MTCL1 by inhibiting C1QBP ubiquitin degradation and beta-catenin activation [148], circPARD3 in autophagy through the PRKCI-Akt-mTOR pathway [149], and circPVT1 via the p53/YAP/TEAD transcription-competent complex [150].
Table 1. The performance of EV-miRNA in the diagnosis, progression, prognosis prediction and treatment evaluation of HNSCC.
Table 1. The performance of EV-miRNA in the diagnosis, progression, prognosis prediction and treatment evaluation of HNSCC.
First AuthorYearCancer TypeRNASample TypeGroupingEVs Isolation
Method
RNA Detection
Method
Clinical UseRef.
Wang2014LSCC *miR-21SerumLSCC (n = 52);
benign laryngeal disease (n = 49)
PrecipitationqRT-PCRDiagnosis, prognosis[151]
Li2016OSCC *miR-21SerumOSCC (n = 108);
healthy control (n = 108)
ExoQuick kit (SBI)/precipitationqRT-PCRProgression (metastasis)[152]
Ye2016NPC *miR-24-3pPlasmaNPC (n = 65);
healthy control (n = 20)
Ultra-centrifugation (UC)qRT-PCR array,
qRT-PCR
Prognosis[153]
Langevin2017HNSCC *miR-486-5p,
miR-486-3p,
miR-10b-5p, miR-31
SalivaHNSCC (n = 11);
healthy control (n = 9)
UCSequencing, ddPCRDiagnosis[154]
Ramayanti2019NPCBART7-3p,
BART9-3p,
BART13-3p
SerumNPC (n = 39);
healthy control (n = 33)
SEC Sepharose CL-2B columnLightCycler stem-loop RT-PCRDiagnosis[155]
Momen-Heravi2018OSCCmiR-21,
miR-27,
miR-412-3p, miR-512-3p
SalivaOSCC (n = 24);
healthy control (n = 10)
ExoQuick kit (SBI)/precipitationqRT-PCR and TaqMan miRNA assayDiagnosis[156]
Bao2018NPCmiR-23aTissue, serumNPC (n = 150)UCqRT-PCRProgression (metastasis), prognosis[157]
Gai2018OSCCmiR-512-3p, miR-412-3p, miR-302b-3p, miR-517b-3p, miR-21, HOTAIRSalivaOSCC (n = 21);
healthy control (n = 11)
PrecipitationqRT-PCR array,
qRT-PCR
Diagnosis[112]
Lu2018NPCmiR-9PlasmaNPC (n = 110);
healthy control (n = 60)
UCqRT-PCRDiagnosis, prognosis[158]
Xie2019OSCCmiR-101-3pTissueOSCC (n = 36, tumor tissue vs. adjacent normal tissue)UCqRT-PCRProgression (stage)[159]
Qin2019HNSCCmiR-196aPlasmaHNSCC (n = 74);
healthy control (n = 30)
ExoQuick kit (SBI), precipitationqRT-PCRDiagnosis, prognosis, treatment response (cisplatin)[160]
Sun2019OSCCmiR-382-5pCocultured tissue47 OSCC patients
(CAF-rich vs. CAF-poor)
PrecipitationqRT-PCRProgression[161]
Mayne2020OPSCC *11-miR (ratio) panelSerumOPSCC (n = 40);
GORD (n = 20);
healthy control (n = 20)
PrecipitationMicroarrayDiagnosis[162]
Wang2020OSCCmiR-210-3pTissueOSCC (n = 80);
healthy control (n = 7)
UCqRT-PCRProgression (stage)[163]
He2020OSCCmiR-24-3pSalivaHNSCC (n = 49);
healthy control (n = 14)
ExoQuick kit (SBI), precipitationMicroarray, qRT-PCRDiagnosis[164]
Shuang2022LSCCmiR-27aPlasmaLSCC (n = 62);
healthy control uk
ExoQuick kit (SBI), precipitationqRT-PCR array,
qRT-PCR
Diagnosis, prognosis[165]
Yin2021NPCmiR-433-3pTissueLSCC (n = 13, tumor tissue vs. adjacent normal tissue)UCqRT-PCRProgression (stage), prognosis[166]
Tong2020HNSCCmiR-9TissueHNSCC (n = 303)UCqRT-PCRPrognosis, treatment response (RT *)[167]
Jiang2021NPCmiR-134-5p, miR-205-5p, miR-409-3pPlasmaNPC (n = 16);
healthy control (n = 5)
PrecipitationNanoparticle tracking analysis (NTA) and qRT-PCRDiagnosis[168]
Chen2021OSCCmiR-155, miR-21,
miR-126
SerumOSCC (n = 35);
healthy control (n = 11)
Exosome Isolation KitMicroarray,
qRT-PCR
Progression (stage), prognosis[169]
Liu2021NPCmiR-181aTissueNPC (n = 134)UCqRT-PCRProgression (metastasis, staging)[170]
Bigagli2021OSCCmiR-210PlasmaOSCC (n = 30);
healthy control (n = 14)
PrecipitationqRT-PCRDiagnosis, prognosis[138]
Panvongsa2021HNSCCmiR-491-5pPlasmaHNSCC (n = 73);
healthy control (n = 20)
ExoQuick kit (SBI), precipitationmiRNA microarray analysis, qRT-PCRDiagnosis, prognosis[171]
Yu2022LSCCmiR-15b-5pCocultured tissueLSCC (n = 16, tumor tissue vs. adjacent normal tissue)ExoQuick kit (SBI), precipitationqRT-PCRProgression (LNM *)[172]
Jiang2022NPCmiR-197.3pTissueNPC (n = 40,
with good vs. poor RT effects)
ExoQuick kit (SBI), precipitationqRT-PCRTreatment (RT *)[173]
Lou2022OSCCmiR-626Serum, tissueOSCC (n = 65, tumor and serum; n = 10, tumor tissue vs. adjacent normal tissue)UCqRT-PCRPrognosis[174]
* LSCC, laryngeal squamous cell carcinoma; OSCC, oral squamous cell carcinoma; NPC, nasopharyngeal carcinoma; HNSCC, head and neck squamous cell carcinoma; OPSCC, oropharyngeal squamous cell carcinoma; RT, radiotherapy; LNM, lymph node metastasis.

3.3. Protein

There is a broad array of proteins present in biofluids. The limitation of proteins acting as biomarkers for certain diseases includes its rapid degradation by proteases in circulation and its relatively poor specificity. However, with the assistance of EVs, EVs could be identified by membrane proteins, which helps to tag enveloped proteins and protects them from lysis. As mentioned, one of the obstacles to elevating the survival rate of HNSCC patients is the early detection of cancer; therefore, many efforts have been made in this field with respect to exosomal proteins [175].
Liu et al. proposed that the detection of exosomal cyclophilin A (CYPA), a protein from immunophilin, helped distinguish NPC patients from healthy controls, which could greatly improve diagnostic precision when combined with the EBV antibody test [141]. You et al. explained that HS1-associated protein X-1 (HAX-1) was overexpressed in many human malignancies, such as HNSCC and especially NPC [176], and they also announced that exosome-packaged HAX-1 had a better performance when serving as a biomarker for LNM, clinical stage, and the prognosis of NPC [177].
The therapeutic effects of these immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 pathway led to survival benefits for platinum-pretreated recurrent and metastatic HNSCC patients [178]. The levels of PD-L1 expressed on the exosomes in circulation was proved to be correlated with patients’ UICC stage and LNM status [179]. Patients with higher expressions of PD-L1 exhibited longer DFS, and postoperative patients faced a higher risk of relapse or recurrence if they had a low PD-L1 expression status. PD-L1 was identified as the earliest indicator of treatment failure and prognosis prediction [179].
Likewise, Kannan et al. noticed that, in OPSCC, the exosomal proteins SIRPA and HPV-16-E7 were elevated in the serum of HPV-16-associated OPSCC [180]. Li et al. disclosed that the protein expression levels of MMP-1 significantly differed with the stratification of malignant potential; it only increased from MSC-derived exosomes collected from normal oral mucosa, oral leukoplakia, and OSCC [54]. These studies suggested that virus-derived and exosome-carried components and proteins could act as biomarkers for HNSCC [54,180].

4. EV as Prospective Biomarkers in HNSCC

Cancer-derived EVs have promising potential to be used as tumor biomarkers, not only for screening and diagnosis but also for monitoring cancer progression and the prediction of patients’ responses to therapy, which has been widely demonstrated in some cancers, such as non-small cell lung cancer [181,182,183]. However, EV detections and applications have not been widely adopted for clinical use since we are still pursuing a greater platform that could ensure highly sensitive and specific methods, but these require minimal sample volume for testing. Additionally, the platform should be capable of detecting different cargoes delivered by EVs (i.e., DNA, RNA, proteins), since they might also be specific biomarkers for certain diseases, and if a platform for detecting EVs could realize such a high-throughput analysis for development, then the clinical application for EVs as biomarkers would be applied in future [30].
The profiles of EV components between malignant tumor cells and normal cells vary significantly, and differences also exist between HNSCCs with and without specific virus infections; these ensure the significance of EVs as competitive candidate biomarkers for HNSCC diagnosis and prognosis.
The merits of EV detection in liquid biopsies in HNSCC include easy sampling, less invasiveness and high acceptance by patients. Compared to CTCs or cfDNA, the concentration of EVs is relatively higher, and EVs can be found in more kinds of biofluids, such as urine and sweat, not just in blood. Despite abundant studies supporting and discovering a great number of EV-conveyed components to be screening tools, one of the limitations that restrict wide clinical application is the interference caused by other endogenous substances and current EV-detection technologies; it is still challenging to identify and extract the tumor-derived components. EVs are likely to exhibit varying sensitivity and specificity due to different sample types and targeting substances, and there is still a lack of standardization of techniques for detection and isolation; these are all problems to solve before they are widely used. In addition, it is also a major obstacle to reaching a well-established quantified standard for tumor screening because the number of circulating EVs differs from person to person, and the abundance of EVs could be affected by confounding effects from nonmalignant cells, such as platelets and megakaryocytes, under certain stimuli. Unlike exosomes, the knowledge about microvesicles still remains occult, especially in head and neck cancer, which probably requires greater attention and effort. However, it is believed that the rapidly developing technologies will continuously and gradually improve the use of EVs as biomarkers in liquid biopsies.
Since it is difficult to set a defined range of values, the individual variance in EV numbers could be partly eliminated if we simply evaluate them case by case instead of comparing them to the mean or a certain range. For instance, the changes in these EV-containing cargoes could be used as indicators for distinguishing different stages of the disease, such as following tumor progression and observing effective treatment. When the goal is not to precisely discriminate patients from the normal population but to monitor the therapeutic effects of interventions, disease diagnosis and treatment, the advantages of liquid biopsy such as easy sampling and minimal invasiveness are easily demonstrable. However, as shown, the clinical trials to investigate EV assessments in disease monitoring are lacking, which requires further study (Table 1).

5. Conclusions

EVs, intricate structures that contain diverse components, such as DNA, RNA, and proteins, participate in many pivotal pathways of HNSCC. The tumor microenvironment and metabolic phenotype are greatly altered after receiving the message molecules delivered by EVs, which are released by connective tissue cells (such as cancer-associated fibroblasts and mesenchymal stem cells), as well as immune cells. The assistance of EVs facilitates the formation of pre-metastatic niches, activation of the vasculature, and fusion with parenchymal cells, which greatly promotes tumor metastasis. The cargoes transported by vesicles also influence the resistance to chemoradiotherapy, and it sheds light on the possibility that EVs can serve as therapeutic target or drug carriers. When compared to tissue biopsy, the advantages of EV detection are easy sampling and minimal invasiveness, which make it more acceptable by patients; when compared to cfDNA, CTC, or other detections based on liquid biopsy, EVs can be extracted from more diverse biofluids, and their membranous structure enables cargoes conveyed by EVs to be preserved more stably and detected even in small amounts. However, the clinical application and formal approval for EV assessment in the screening and diagnosis of HNSCC is restricted by the stagnant development of detection technologies and individual differences in terms of the abundance and expression of the tumor-specific components of EVs. There is a good prospect for their application in the prediction of the response to interventions and relapse or prognosis, since it focuses on the dynamic monitoring of the treatment process and the changes in patient conditions.

Author Contributions

Conceptualization, J.Y.-K.C. and B.C.; writing—original draft preparation, B.C.; writing—review and editing, J.Y.-K.C., L.L.L. and X.Q.; final approval—J.Y.-K.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

EV, extracellular vesicles; HNSCC, head and neck squamous cell carcinoma; DNA, deoxyribonucleic acid; RNA, ribonucleic acid; TNF, tumor necrosis factors; MAPK, mitogen-activated protein kinase; LNM, lymph node metastasis; EpCAM, epithelial cell adhesion molecule; PSA, prostate specific antigen; FFPE, formalin fixed paraffin-embedded.

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] [PubMed] [Green Version]
  2. Johnson, D.E.; Burtness, B.; Leemans, C.R.; Lui, V.W.Y.; Bauman, J.E.; Grandis, J.R. Head and neck squamous cell carcinoma. Nat. Rev. Dis. Primers 2020, 6, 92. [Google Scholar] [CrossRef] [PubMed]
  3. Castellsagué, X.; Quintana, M.J.; Martínez, M.C.; Nieto, A.; Sánchez, M.J.; Juan, A.; Monner, A.; Carrera, M.; Agudo, A.; Quer, M.; et al. The role of type of tobacco and type of alcoholic beverage in oral carcinogenesis. Int. J. Cancer 2004, 108, 741–749. [Google Scholar] [CrossRef] [PubMed]
  4. Aupérin, A. Epidemiology of head and neck cancers: An update. Curr. Opin. Oncol. 2020, 32, 178–186. [Google Scholar] [CrossRef] [PubMed]
  5. Jiang, H.; Livingston, M.; Room, R.; Gan, Y.; English, D.; Chenhall, R. Can public health policies on alcohol and tobacco reduce a cancer epidemic? Australia’s experience. BMC Med. 2019, 17, 213. [Google Scholar] [CrossRef]
  6. Marur, S.; D’Souza, G.; Westra, W.H.; Forastiere, A.A. HPV-associated head and neck cancer: A virus-related cancer epidemic. Lancet Oncol. 2010, 11, 781–789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Rassy, E.; Nicolai, P.; Pavlidis, N. Comprehensive management of HPV-related squamous cell carcinoma of the head and neck of unknown primary. Head Neck 2019, 41, 3700–3711. [Google Scholar] [CrossRef]
  8. Cohen, E.E.W.; Bell, R.B.; Bifulco, C.B.; Burtness, B.; Gillison, M.L.; Harrington, K.J.; Le, Q.T.; Lee, N.Y.; Leidner, R.; Lewis, R.L.; et al. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of squamous cell carcinoma of the head and neck (HNSCC). J. Immunother. Cancer 2019, 7, 184. [Google Scholar] [CrossRef] [Green Version]
  9. Marur, S.; Forastiere, A.A. Head and neck cancer: Changing epidemiology, diagnosis, and treatment. Mayo Clin. Proc. 2008, 83, 489–501. [Google Scholar] [CrossRef] [PubMed]
  10. Bhat, G.R.; Hyole, R.G.; Li, J. Head and neck cancer: Current challenges and future perspectives. Adv. Cancer Res. 2021, 152, 67–102. [Google Scholar] [CrossRef]
  11. Bonner, J.A.; Harari, P.M.; Giralt, J.; Azarnia, N.; Shin, D.M.; Cohen, R.B.; Jones, C.U.; Sur, R.; Raben, D.; Jassem, J.; et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N. Engl. J. Med. 2006, 354, 567–578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Burtness, B.; Harrington, K.J.; Greil, R.; Soulières, D.; Tahara, M.; de Castro, G., Jr.; Psyrri, A.; Basté, N.; Neupane, P.; Bratland, Å.; et al. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): A randomised, open-label, phase 3 study. Lancet 2019, 394, 1915–1928. [Google Scholar] [CrossRef]
  13. Seiwert, T.Y.; Burtness, B.; Mehra, R.; Weiss, J.; Berger, R.; Eder, J.P.; Heath, K.; McClanahan, T.; Lunceford, J.; Gause, C.; et al. Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): An open-label, multicentre, phase 1b trial. Lancet Oncol. 2016, 17, 956–965. [Google Scholar] [CrossRef] [PubMed]
  14. Lee, D.D.; Schwarz, M.A. Cell-Cell Communication Breakdown and Endothelial Dysfunction. Crit. Care Clin. 2020, 36, 189–200. [Google Scholar] [CrossRef] [PubMed]
  15. O’Loughlin, A.J.; Woffindale, C.A.; Wood, M.J. Exosomes and the emerging field of exosome-based gene therapy. Curr. Gene Ther. 2012, 12, 262–274. [Google Scholar] [CrossRef]
  16. Pegtel, D.M.; Gould, S.J. Exosomes. Annu. Rev. Biochem. 2019, 88, 487–514. [Google Scholar] [CrossRef]
  17. Becker, A.; Thakur, B.K.; Weiss, J.M.; Kim, H.S.; Peinado, H.; Lyden, D. Extracellular Vesicles in Cancer: Cell-to-Cell Mediators of Metastasis. Cancer Cell 2016, 30, 836–848. [Google Scholar] [CrossRef] [Green Version]
  18. Müller Bark, J.; Kulasinghe, A.; Amenábar, J.M.; Punyadeera, C. Exosomes in cancer. Adv. Clin. Chem. 2021, 101, 1–40. [Google Scholar] [CrossRef]
  19. Vago, R.; Salonia, A. Re: Circulating Extracellular Vesicles in Human Disease. Eur. Urol. 2019, 75, 342–343. [Google Scholar] [CrossRef]
  20. Willis, G.R.; Kourembanas, S.; Mitsialis, S.A. Toward Exosome-Based Therapeutics: Isolation, Heterogeneity, and Fit-for-Purpose Potency. Front. Cardiovasc. Med. 2017, 4, 63. [Google Scholar] [CrossRef] [Green Version]
  21. D’Acunzo, P.; Hargash, T.; Pawlik, M.; Goulbourne, C.N.; Pérez-González, R.; Levy, E. Enhanced generation of intraluminal vesicles in neuronal late endosomes in the brain of a Down syndrome mouse model with endosomal dysfunction. Dev. Neurobiol. 2019, 79, 656–663. [Google Scholar] [CrossRef] [PubMed]
  22. Elkin, S.R.; Lakoduk, A.M.; Schmid, S.L. Endocytic pathways and endosomal trafficking: A primer. Wien Med. Wochenschr. 2016, 166, 196–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Trams, E.G.; Lauter, C.J.; Salem, N., Jr.; Heine, U. Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochim. Biophys. Acta 1981, 645, 63–70. [Google Scholar] [CrossRef]
  24. Pollet, H.; Conrard, L.; Cloos, A.S.; Tyteca, D. Plasma Membrane Lipid Domains as Platforms for Vesicle Biogenesis and Shedding? Biomolecules 2018, 8, 94. [Google Scholar] [CrossRef] [Green Version]
  25. Wolf, P. The nature and significance of platelet products in human plasma. Br. J. Haematol. 1967, 13, 269–288. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, X.; Guo, J.; Yu, P.; Guo, L.; Mao, X.; Wang, J.; Miao, S.; Sun, J. The roles of extracellular vesicles in the development, microenvironment, anticancer drug resistance, and therapy of head and neck squamous cell carcinoma. J. Exp. Clin. Cancer Res. 2021, 40, 35. [Google Scholar] [CrossRef]
  27. Baranyai, T.; Herczeg, K.; Onódi, Z.; Voszka, I.; Módos, K.; Marton, N.; Nagy, G.; Mäger, I.; Wood, M.J.; El Andaloussi, S.; et al. Isolation of Exosomes from Blood Plasma: Qualitative and Quantitative Comparison of Ultracentrifugation and Size Exclusion Chromatography Methods. PLoS ONE 2015, 10, e0145686. [Google Scholar] [CrossRef] [Green Version]
  28. Lobb, R.J.; Becker, M.; Wen, S.W.; Wong, C.S.; Wiegmans, A.P.; Leimgruber, A.; Möller, A. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J. Extracell. Vesicles 2015, 4, 27031. [Google Scholar] [CrossRef]
  29. Van Deun, J.; Mestdagh, P.; Sormunen, R.; Cocquyt, V.; Vermaelen, K.; Vandesompele, J.; Bracke, M.; De Wever, O.; Hendrix, A. The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling. J. Extracell. Vesicles 2014, 3, 24858. [Google Scholar] [CrossRef] [Green Version]
  30. Visan, K.S.; Wu, L.Y.; Voss, S.; Wuethrich, A.; Möller, A. Status quo of Extracellular Vesicle isolation and detection methods for clinical utility. Semin. Cancer Biol. 2023, 88, 157–171. [Google Scholar] [CrossRef]
  31. Woo, H.K.; Sunkara, V.; Park, J.; Kim, T.H.; Han, J.R.; Kim, C.J.; Choi, H.I.; Kim, Y.K.; Cho, Y.K. Exodisc for Rapid, Size-Selective, and Efficient Isolation and Analysis of Nanoscale Extracellular Vesicles from Biological Samples. ACS Nano 2017, 11, 1360–1370. [Google Scholar] [CrossRef]
  32. Xu, H.; Liao, C.; Zuo, P.; Liu, Z.; Ye, B.C. Magnetic-Based Microfluidic Device for On-Chip Isolation and Detection of Tumor-Derived Exosomes. Anal. Chem. 2018, 90, 13451–13458. [Google Scholar] [CrossRef] [PubMed]
  33. Jin, D.; Yang, F.; Zhang, Y.; Liu, L.; Zhou, Y.; Wang, F.; Zhang, G.J. ExoAPP: Exosome-Oriented, Aptamer Nanoprobe-Enabled Surface Proteins Profiling and Detection. Anal. Chem. 2018, 90, 14402–14411. [Google Scholar] [CrossRef] [PubMed]
  34. Whiteside, T.L. Exosome and mesenchymal stem cell cross-talk in the tumor microenvironment. Semin. Immunol. 2018, 35, 69–79. [Google Scholar] [CrossRef] [PubMed]
  35. Szczepanski, M.J.; Szajnik, M.; Welsh, A.; Whiteside, T.L.; Boyiadzis, M. Blast-derived microvesicles in sera from patients with acute myeloid leukemia suppress natural killer cell function via membrane-associated transforming growth factor-beta1. Haematologica 2011, 96, 1302–1309. [Google Scholar] [CrossRef] [Green Version]
  36. Whiteside, T.L. Exosomes carrying immunoinhibitory proteins and their role in cancer. Clin. Exp. Immunol. 2017, 189, 259–267. [Google Scholar] [CrossRef] [Green Version]
  37. Boyiadzis, M.; Whiteside, T.L. Information transfer by exosomes: A new frontier in hematologic malignancies. Blood Rev. 2015, 29, 281–290. [Google Scholar] [CrossRef]
  38. Zhao, H.; Yang, L.; Baddour, J.; Achreja, A.; Bernard, V.; Moss, T.; Marini, J.C.; Tudawe, T.; Seviour, E.G.; San Lucas, F.A.; et al. Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism. Elife 2016, 5, e10250. [Google Scholar] [CrossRef]
  39. Yang, X.; Li, Y.; Zou, L.; Zhu, Z. Role of Exosomes in Crosstalk between Cancer-Associated Fibroblasts and Cancer Cells. Front. Oncol. 2019, 9, 356. [Google Scholar] [CrossRef]
  40. Zhu, T.; Hu, Z.; Wang, Z.; Ding, H.; Li, R.; Wang, J.; Wang, G. microRNA-301b-3p from mesenchymal stem cells-derived extracellular vesicles inhibits TXNIP to promote multidrug resistance of gastric cancer cells. Cell Biol. Toxicol. 2022, 1–15. [Google Scholar] [CrossRef]
  41. Weng, Z.; Zhang, B.; Wu, C.; Yu, F.; Han, B.; Li, B.; Li, L. Therapeutic roles of mesenchymal stem cell-derived extracellular vesicles in cancer. J. Hematol. Oncol. 2021, 14, 136. [Google Scholar] [CrossRef]
  42. Hoshino, A.; Costa-Silva, B.; Shen, T.L.; Rodrigues, G.; Hashimoto, A.; Tesic Mark, M.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef] [Green Version]
  43. Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.; et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef] [Green Version]
  44. Zhang, H.; Deng, T.; Ge, S.; Liu, Y.; Bai, M.; Zhu, K.; Fan, Q.; Li, J.; Ning, T.; Tian, F.; et al. Exosome circRNA secreted from adipocytes promotes the growth of hepatocellular carcinoma by targeting deubiquitination-related USP7. Oncogene 2019, 38, 2844–2859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Cheng, A.N.; Cheng, L.C.; Kuo, C.L.; Lo, Y.K.; Chou, H.Y.; Chen, C.H.; Wang, Y.H.; Chuang, T.H.; Cheng, S.J.; Lee, A.Y. Mitochondrial Lon-induced mtDNA leakage contributes to PD-L1-mediated immunoescape via STING-IFN signaling and extracellular vesicles. J. Immunother. Cancer 2020, 8, e001372. [Google Scholar] [CrossRef] [PubMed]
  46. Kim, D.H.; Kim, H.; Choi, Y.J.; Kim, S.Y.; Lee, J.E.; Sung, K.J.; Sung, Y.H.; Pack, C.G.; Jung, M.K.; Han, B.; et al. Exosomal PD-L1 promotes tumor growth through immune escape in non-small cell lung cancer. Exp. Mol. Med. 2019, 51, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Huang, T.; Song, C.; Zheng, L.; Xia, L.; Li, Y.; Zhou, Y. The roles of extracellular vesicles in gastric cancer development, microenvironment, anti-cancer drug resistance, and therapy. Mol. Cancer 2019, 18, 62. [Google Scholar] [CrossRef] [Green Version]
  48. Au Yeung, C.L.; Co, N.N.; Tsuruga, T.; Yeung, T.L.; Kwan, S.Y.; Leung, C.S.; Li, Y.; Lu, E.S.; Kwan, K.; Wong, K.K.; et al. Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat. Commun. 2016, 7, 11150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Zhang, Z.; Yu, X.; Zhou, Z.; Li, B.; Peng, J.; Wu, X.; Luo, X.; Yang, L. LMP1-positive extracellular vesicles promote radioresistance in nasopharyngeal carcinoma cells through P38 MAPK signaling. Cancer Med. 2019, 8, 6082–6094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. EL Andaloussi, S.; Mäger, I.; Breakefield, X.O.; Wood, M.J. Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 2013, 12, 347–357. [Google Scholar] [CrossRef]
  51. Besse, B.; Charrier, M.; Lapierre, V.; Dansin, E.; Lantz, O.; Planchard, D.; Le Chevalier, T.; Livartoski, A.; Barlesi, F.; Laplanche, A.; et al. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology 2016, 5, e1071008. [Google Scholar] [CrossRef] [Green Version]
  52. Dai, S.; Wei, D.; Wu, Z.; Zhou, X.; Wei, X.; Huang, H.; Li, G. Phase I clinical trial of autologous ascites-derived exosomes combined with GM-CSF for colorectal cancer. Mol. Ther. 2008, 16, 782–790. [Google Scholar] [CrossRef]
  53. Escudier, B.; Dorval, T.; Chaput, N.; André, F.; Caby, M.P.; Novault, S.; Flament, C.; Leboulaire, C.; Borg, C.; Amigorena, S.; et al. Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: Results of thefirst phase I clinical trial. J. Transl. Med. 2005, 3, 10. [Google Scholar] [CrossRef] [Green Version]
  54. Li, S.; Han, Y.; Lu, M.; Liu, Z.; Jin, J.; Guo, Q.; Wang, Y.; Liu, H. Mesenchymal stem cell-exosome-mediated matrix metalloproteinase 1 participates in oral leukoplakia and carcinogenesis by inducing angiogenesis. J. Oral Pathol. Med. 2022, 51, 638–648. [Google Scholar] [CrossRef] [PubMed]
  55. Lakshmi, S.; Hughes, T.A.; Priya, S. Exosomes and exosomal RNAs in breast cancer: A status update. Eur. J. Cancer 2021, 144, 252–268. [Google Scholar] [CrossRef] [PubMed]
  56. Kubo, H. Extracellular Vesicles in Lung Disease. Chest 2018, 153, 210–216. [Google Scholar] [CrossRef]
  57. Wu, H.; Fu, M.; Liu, J.; Chong, W.; Fang, Z.; Du, F.; Liu, Y.; Shang, L.; Li, L. The role and application of small extracellular vesicles in gastric cancer. Mol. Cancer 2021, 20, 71. [Google Scholar] [CrossRef]
  58. Zheng, X.; Xu, K.; Zhou, B.; Chen, T.; Huang, Y.; Li, Q.; Wen, F.; Ge, W.; Wang, J.; Yu, S.; et al. A circulating extracellular vesicles-based novel screening tool for colorectal cancer revealed by shotgun and data-independent acquisition mass spectrometry. J. Extracell. Vesicles 2020, 9, 1750202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Arbelaiz, A.; Azkargorta, M.; Krawczyk, M.; Santos-Laso, A.; Lapitz, A.; Perugorria, M.J.; Erice, O.; Gonzalez, E.; Jimenez-Agüero, R.; Lacasta, A.; et al. Serum extracellular vesicles contain protein biomarkers for primary sclerosing cholangitis and cholangiocarcinoma. Hepatology 2017, 66, 1125–1143. [Google Scholar] [CrossRef] [Green Version]
  60. Thietart, S.; Rautou, P.E. Extracellular vesicles as biomarkers in liver diseases: A clinician’s point of view. J. Hepatol. 2020, 73, 1507–1525. [Google Scholar] [CrossRef]
  61. Logozzi, M.; Angelini, D.F.; Iessi, E.; Mizzoni, D.; Di Raimo, R.; Federici, C.; Lugini, L.; Borsellino, G.; Gentilucci, A.; Pierella, F.; et al. Increased PSA expression on prostate cancer exosomes in in vitro condition and in cancer patients. Cancer Lett. 2017, 403, 318–329. [Google Scholar] [CrossRef]
  62. Leung, L.L.; Riaz, M.K.; Qu, X.; Chan, J.; Meehan, K. Profiling of extracellular vesicles in oral cancer, from transcriptomics to proteomics. Semin. Cancer Biol. 2021, 74, 3–23. [Google Scholar] [CrossRef] [PubMed]
  63. Qu, X.; Leung, T.C.N.; Ngai, S.M.; Tsai, S.N.; Thakur, A.; Li, W.K.; Lee, Y.; Leung, L.; Ng, T.H.; Yam, J.; et al. Proteomic Analysis of Circulating Extracellular Vesicles Identifies Potential Biomarkers for Lymph Node Metastasis in Oral Tongue Squamous Cell Carcinoma. Cells 2021, 10, 2179. [Google Scholar] [CrossRef] [PubMed]
  64. Qu, X.; Li, J.W.; Chan, J.; Meehan, K. Extracellular Vesicles in Head and Neck Cancer: A Potential New Trend in Diagnosis, Prognosis, and Treatment. Int. J. Mol. Sci. 2020, 21, 8260. [Google Scholar] [CrossRef]
  65. Galle, P.R.; Foerster, F.; Kudo, M.; Chan, S.L.; Llovet, J.M.; Qin, S.; Schelman, W.R.; Chintharlapalli, S.; Abada, P.B.; Sherman, M.; et al. Biology and significance of alpha-fetoprotein in hepatocellular carcinoma. Liver Int. 2019, 39, 2214–2229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Saini, S. PSA and beyond: Alternative prostate cancer biomarkers. Cell Oncol. 2016, 39, 97–106. [Google Scholar] [CrossRef] [Green Version]
  67. Vaitukaitis, J.L. Human chorionic gonadotropin—A hormone secreted for many reasons. N. Engl. J. Med. 1979, 301, 324–326. [Google Scholar] [CrossRef]
  68. Izuhara, K.; Yamaguchi, Y.; Ohta, S.; Nunomura, S.; Nanri, Y.; Azuma, Y.; Nomura, N.; Noguchi, Y.; Aihara, M. Squamous Cell Carcinoma Antigen 2 (SCCA2, SERPINB4): An Emerging Biomarker for Skin Inflammatory Diseases. Int. J. Mol. Sci. 2018, 19, 1102. [Google Scholar] [CrossRef] [Green Version]
  69. Kommu, S.; Hadway, P.; Watkin, N. Squamous cell carcinoma antigen as a biomarker for penile cancer. BJU Int. 2005, 95, 478–479. [Google Scholar] [CrossRef]
  70. Sun, Z.; Shi, X.; Wang, Y.; Zhao, Y. Serum Squamous Cell Carcinoma Antigen in Psoriasis: A Potential Quantitative Biomarker for Disease Severity. Dermatology 2018, 234, 120–126. [Google Scholar] [CrossRef]
  71. Ignatiadis, M.; Sledge, G.W.; Jeffrey, S.S. Liquid biopsy enters the clinic—Implementation issues and future challenges. Nat. Rev. Clin. Oncol. 2021, 18, 297–312. [Google Scholar] [CrossRef]
  72. Mandel, P.; Metais, P. Nuclear Acids In Human Blood Plasma. C. R. Seances Soc. Biol. Fil. 1948, 142, 241–243. [Google Scholar]
  73. Stroun, M.; Anker, P.; Maurice, P.; Lyautey, J.; Lederrey, C.; Beljanski, M. Neoplastic characteristics of the DNA found in the plasma of cancer patients. Oncology 1989, 46, 318–322. [Google Scholar] [CrossRef] [PubMed]
  74. De Miranda, F.S.; Barauna, V.G.; Dos Santos, L.; Costa, G.; Vassallo, P.F.; Campos, L.C.G. Properties and Application of Cell-Free DNA as a Clinical Biomarker. Int. J. Mol. Sci. 2021, 22, 9110. [Google Scholar] [CrossRef] [PubMed]
  75. Leon, S.A.; Shapiro, B.; Sklaroff, D.M.; Yaros, M.J. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 1977, 37, 646–650. [Google Scholar]
  76. Corcoran, R.B. Liquid biopsy versus tumor biopsy for clinical-trial recruitment. Nat. Med. 2020, 26, 1815–1816. [Google Scholar] [CrossRef]
  77. Leighl, N.B.; Page, R.D.; Raymond, V.M.; Daniel, D.B.; Divers, S.G.; Reckamp, K.L.; Villalona-Calero, M.A.; Dix, D.; Odegaard, J.I.; Lanman, R.B.; et al. Clinical Utility of Comprehensive Cell-free DNA Analysis to Identify Genomic Biomarkers in Patients with Newly Diagnosed Metastatic Non-small Cell Lung Cancer. Clin. Cancer Res. 2019, 25, 4691–4700. [Google Scholar] [CrossRef] [Green Version]
  78. Nakamura, Y.; Taniguchi, H.; Ikeda, M.; Bando, H.; Kato, K.; Morizane, C.; Esaki, T.; Komatsu, Y.; Kawamoto, Y.; Takahashi, N.; et al. Clinical utility of circulating tumor DNA sequencing in advanced gastrointestinal cancer: SCRUM-Japan GI-SCREEN and GOZILA studies. Nat. Med. 2020, 26, 1859–1864. [Google Scholar] [CrossRef]
  79. Yang, X.; Song, H.; Ji, T.; Du, G.; Liu, W. The implications of gene mutations in salivary DNA for noninvasive diagnosis of head and neck cancer with a focus on oral cancer. Oral. Oncol. 2022, 130, 105924. [Google Scholar] [CrossRef] [PubMed]
  80. Arantes, L.; De Carvalho, A.C.; Melendez, M.E.; Lopes Carvalho, A. Serum, plasma and saliva biomarkers for head and neck cancer. Expert. Rev. Mol. Diagn. 2018, 18, 85–112. [Google Scholar] [CrossRef] [PubMed]
  81. Langevin, S.M.; Stone, R.A.; Bunker, C.H.; Grandis, J.R.; Sobol, R.W.; Taioli, E. MicroRNA-137 promoter methylation in oral rinses from patients with squamous cell carcinoma of the head and neck is associated with gender and body mass index. Carcinogenesis 2010, 31, 864–870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Schirmer, M.A.; Beck, J.; Leu, M.; Oellerich, M.; Rave-Fränk, M.; Walson, P.D.; Schütz, E.; Canis, M. Cell-Free Plasma DNA for Disease Stratification and Prognosis in Head and Neck Cancer. Clin. Chem. 2018, 64, 959–970. [Google Scholar] [CrossRef]
  83. Wilson, H.L.; D’Agostino, R.B., Jr.; Meegalla, N.; Petro, R.; Commander, S.; Topaloglu, U.; Zhang, W.; Porosnicu, M. The Prognostic and Therapeutic Value of the Mutational Profile of Blood and Tumor Tissue in Head and Neck Squamous Cell Carcinoma. Oncologist 2021, 26, e279–e289. [Google Scholar] [CrossRef]
  84. Wuerdemann, N.; Jain, R.; Adams, A.; Speel, E.M.; Wagner, S.; Joosse, S.A.; Klussmann, J.P. Cell-Free HPV-DNA as a Biomarker for Oropharyngeal Squamous Cell Carcinoma-A Step Towards Personalized Medicine? Cancers 2020, 12, 2997. [Google Scholar] [CrossRef] [PubMed]
  85. Gkountela, S.; Castro-Giner, F.; Szczerba, B.M.; Vetter, M.; Landin, J.; Scherrer, R.; Krol, I.; Scheidmann, M.C.; Beisel, C.; Stirnimann, C.U.; et al. Circulating Tumor Cell Clustering Shapes DNA Methylation to Enable Metastasis Seeding. Cell 2019, 176, 98–112.e14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Aceto, N.; Toner, M.; Maheswaran, S.; Haber, D.A. En Route to Metastasis: Circulating Tumor Cell Clusters and Epithelial-to-Mesenchymal Transition. Trends Cancer 2015, 1, 44–52. [Google Scholar] [CrossRef]
  87. Alix-Panabières, C.; Pantel, K. Challenges in circulating tumour cell research. Nat. Rev. Cancer 2014, 14, 623–631. [Google Scholar] [CrossRef]
  88. Hristozova, T.; Konschak, R.; Stromberger, C.; Fusi, A.; Liu, Z.; Weichert, W.; Stenzinger, A.; Budach, V.; Keilholz, U.; Tinhofer, I. The presence of circulating tumor cells (CTCs) correlates with lymph node metastasis in nonresectable squamous cell carcinoma of the head and neck region (SCCHN). Ann. Oncol. 2011, 22, 1878–1885. [Google Scholar] [CrossRef] [PubMed]
  89. Strati, A.; Koutsodontis, G.; Papaxoinis, G.; Angelidis, I.; Zavridou, M.; Economopoulou, P.; Kotsantis, I.; Avgeris, M.; Mazel, M.; Perisanidis, C.; et al. Prognostic significance of PD-L1 expression on circulating tumor cells in patients with head and neck squamous cell carcinoma. Ann. Oncol. 2017, 28, 1923–1933. [Google Scholar] [CrossRef]
  90. De Wit, S.; van Dalum, G.; Lenferink, A.T.; Tibbe, A.G.; Hiltermann, T.J.; Groen, H.J.; van Rijn, C.J.; Terstappen, L.W. The detection of EpCAM(+) and EpCAM(−) circulating tumor cells. Sci. Rep. 2015, 5, 12270. [Google Scholar] [CrossRef] [Green Version]
  91. Mikolajczyk, S.D.; Millar, L.S.; Tsinberg, P.; Coutts, S.M.; Zomorrodi, M.; Pham, T.; Bischoff, F.Z.; Pircher, T.J. Detection of EpCAM-Negative and Cytokeratin-Negative Circulating Tumor Cells in Peripheral Blood. J. Oncol. 2011, 2011, 252361. [Google Scholar] [CrossRef] [PubMed]
  92. Pauken, C.M.; Kenney, S.R.; Brayer, K.J.; Guo, Y.; Brown-Glaberman, U.A.; Marchetti, D. Heterogeneity of Circulating Tumor Cell Neoplastic Subpopulations Outlined by Single-Cell Transcriptomics. Cancers 2021, 13, 4885. [Google Scholar] [CrossRef]
  93. Mego, M.; Cierna, Z.; Janega, P.; Karaba, M.; Minarik, G.; Benca, J.; Sedlácková, T.; Sieberova, G.; Gronesova, P.; Manasova, D.; et al. Relationship between circulating tumor cells and epithelial to mesenchymal transition in early breast cancer. BMC Cancer 2015, 15, 533. [Google Scholar] [CrossRef] [Green Version]
  94. Smolkova, B.; Cierna, Z.; Kalavska, K.; Miklikova, S.; Plava, J.; Minarik, G.; Sedlackova, T.; Cholujova, D.; Gronesova, P.; Cihova, M.; et al. Increased Stromal Infiltrating Lymphocytes Are Associated with the Risk of Disease Progression in Mesenchymal Circulating Tumor Cell-Positive Primary Breast Cancer Patients. Int. J. Mol. Sci. 2020, 21, 9460. [Google Scholar] [CrossRef]
  95. Deutsch, T.M.; Riethdorf, S.; Nees, J.; Hartkopf, A.D.; Schönfisch, B.; Domschke, C.; Sprick, M.R.; Schütz, F.; Brucker, S.Y.; Stefanovic, S.; et al. Impact of apoptotic circulating tumor cells (aCTC) in metastatic breast cancer. Breast Cancer Res. Treat. 2016, 160, 277–290. [Google Scholar] [CrossRef]
  96. Kallergi, G.; Konstantinidis, G.; Markomanolaki, H.; Papadaki, M.A.; Mavroudis, D.; Stournaras, C.; Georgoulias, V.; Agelaki, S. Apoptotic circulating tumor cells in early and metastatic breast cancer patients. Mol. Cancer Ther. 2013, 12, 1886–1895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Strilic, B.; Offermanns, S. Intravascular Survival and Extravasation of Tumor Cells. Cancer Cell 2017, 32, 282–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Chen, K.; Dopico, P.; Varillas, J.; Zhang, J.; George, T.J.; Fan, Z.H. Integration of Lateral Filter Arrays with Immunoaffinity for Circulating-Tumor-Cell Isolation. Angew. Chem. Int. Ed. Engl. 2019, 58, 7606–7610. [Google Scholar] [CrossRef]
  99. Cho, H.; Kim, J.; Song, H.; Sohn, K.Y.; Jeon, M.; Han, K.H. Microfluidic technologies for circulating tumor cell isolation. Analyst 2018, 143, 2936–2970. [Google Scholar] [CrossRef]
  100. Gabriel, M.T.; Calleja, L.R.; Chalopin, A.; Ory, B.; Heymann, D. Circulating Tumor Cells: A Review of Non-EpCAM-Based Approaches for Cell Enrichment and Isolation. Clin. Chem. 2016, 62, 571–581. [Google Scholar] [CrossRef] [Green Version]
  101. Lin, M.; Chen, J.F.; Lu, Y.T.; Zhang, Y.; Song, J.; Hou, S.; Ke, Z.; Tseng, H.R. Nanostructure embedded microchips for detection, isolation, and characterization of circulating tumor cells. Acc. Chem. Res. 2014, 47, 2941–2950. [Google Scholar] [CrossRef] [Green Version]
  102. Cohen, E.N.; Jayachandran, G.; Moore, R.G.; Cristofanilli, M.; Lang, J.E.; Khoury, J.D.; Press, M.F.; Kim, K.K.; Khazan, N.; Zhang, Q.; et al. A Multi-Center Clinical Study to Harvest and Characterize Circulating Tumor Cells from Patients with Metastatic Breast Cancer Using the Parsortix(®) PC1 System. Cancers 2022, 14, 5238. [Google Scholar] [CrossRef] [PubMed]
  103. Garrel, R.; Mazel, M.; Perriard, F.; Vinches, M.; Cayrefourcq, L.; Guigay, J.; Digue, L.; Aubry, K.; Alfonsi, M.; Delord, J.P.; et al. Circulating Tumor Cells as a Prognostic Factor in Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma: The CIRCUTEC Prospective Study. Clin. Chem. 2019, 65, 1267–1275. [Google Scholar] [CrossRef] [PubMed]
  104. Tinhofer, I.; Konschak, R.; Stromberger, C.; Raguse, J.D.; Dreyer, J.H.; Jöhrens, K.; Keilholz, U.; Budach, V. Detection of circulating tumor cells for prediction of recurrence after adjuvant chemoradiation in locally advanced squamous cell carcinoma of the head and neck. Ann. Oncol. 2014, 25, 2042–2047. [Google Scholar] [CrossRef]
  105. Wang, H.M.; Wu, M.H.; Chang, P.H.; Lin, H.C.; Liao, C.D.; Wu, S.M.; Hung, T.M.; Lin, C.Y.; Chang, T.C.; Tzu-Tsen, Y.; et al. The change in circulating tumor cells before and during concurrent chemoradiotherapy is associated with survival in patients with locally advanced head and neck cancer. Head Neck 2019, 41, 2676–2687. [Google Scholar] [CrossRef] [PubMed]
  106. Yang, W.Y.; Feng, L.F.; Meng, X.; Chen, R.; Xu, W.H.; Hou, J.; Xu, T.; Zhang, L. Liquid biopsy in head and neck squamous cell carcinoma: Circulating tumor cells, circulating tumor DNA, and exosomes. Expert Rev. Mol. Diagn. 2020, 20, 1213–1227. [Google Scholar] [CrossRef]
  107. Gurunathan, S.; Kang, M.H.; Jeyaraj, M.; Qasim, M.; Kim, J.H. Review of the Isolation, Characterization, Biological Function, and Multifarious Therapeutic Approaches of Exosomes. Cells 2019, 8, 307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Li, P.; Kaslan, M.; Lee, S.H.; Yao, J.; Gao, Z. Progress in Exosome Isolation Techniques. Theranostics 2017, 7, 789–804. [Google Scholar] [CrossRef]
  109. Zarovni, N.; Corrado, A.; Guazzi, P.; Zocco, D.; Lari, E.; Radano, G.; Muhhina, J.; Fondelli, C.; Gavrilova, J.; Chiesi, A. Integrated isolation and quantitative analysis of exosome shuttled proteins and nucleic acids using immunocapture approaches. Methods 2015, 87, 46–58. [Google Scholar] [CrossRef]
  110. Ahn, S.M.; Chan, J.Y.; Zhang, Z.; Wang, H.; Khan, Z.; Bishop, J.A.; Westra, W.; Koch, W.M.; Califano, J.A. Saliva and plasma quantitative polymerase chain reaction-based detection and surveillance of human papillomavirus-related head and neck cancer. JAMA Otolaryngol. Head Neck Surg. 2014, 140, 846–854. [Google Scholar] [CrossRef] [Green Version]
  111. Chiabotto, G.; Gai, C.; Deregibus, M.C.; Camussi, G. Salivary Extracellular Vesicle-Associated exRNA as Cancer Biomarker. Cancers 2019, 11, 891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. 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] [Green Version]
  113. Nonaka, T.; Wong, D.T.W. Saliva-Exosomics in Cancer: Molecular Characterization of Cancer-Derived Exosomes in Saliva. Enzymes 2017, 42, 125–151. [Google Scholar] [CrossRef]
  114. Topkas, E.; Keith, P.; Dimeski, G.; Cooper-White, J.; Punyadeera, C. Evaluation of saliva collection devices for the analysis of proteins. Clin. Chim. Acta 2012, 413, 1066–1070. [Google Scholar] [CrossRef]
  115. 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]
  116. 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]
  117. Kim, J.W.; Wieckowski, E.; Taylor, D.D.; Reichert, T.E.; Watkins, S.; Whiteside, T.L. Fas ligand-positive membranous vesicles isolated from sera of patients with oral cancer induce apoptosis of activated T lymphocytes. Clin. Cancer Res. 2005, 11, 1010–1020. [Google Scholar] [CrossRef] [PubMed]
  118. Bergmann, C.; Strauss, L.; Wieckowski, E.; Czystowska, M.; Albers, A.; Wang, Y.; Zeidler, R.; Lang, S.; Whiteside, T.L. Tumor-derived microvesicles in sera of patients with head and neck cancer and their role in tumor progression. Head Neck 2009, 31, 371–380. [Google Scholar] [CrossRef] [Green Version]
  119. Bigler, L.R.; Streckfus, C.F.; Dubinsky, W.P. Salivary biomarkers for the detection of malignant tumors that are remote from the oral cavity. Clin. Lab. Med. 2009, 29, 71–85. [Google Scholar] [CrossRef] [PubMed]
  120. Alvarez, M.L.; Khosroheidari, M.; Kanchi Ravi, R.; DiStefano, J.K. Comparison of protein, microRNA, and mRNA yields using different methods of urinary exosome isolation for the discovery of kidney disease biomarkers. Kidney Int. 2012, 82, 1024–1032. [Google Scholar] [CrossRef] [Green Version]
  121. Nguyen, B.; Wong, N.C.; Semple, T.; Clark, M.; Wong, S.Q.; Leslie, C.; Mirzai, B.; Millward, M.; Meehan, K.; Lim, A.M. Low-coverage whole-genome sequencing of extracellular vesicle-associated DNA in patients with metastatic cancer. Sci. Rep. 2021, 11, 4016. [Google Scholar] [CrossRef]
  122. Bauer, M.; Jasinski-Bergner, S.; Mandelboim, O.; Wickenhauser, C.; Seliger, B. Epstein-Barr Virus-Associated Malignancies and Immune Escape: The Role of the Tumor Microenvironment and Tumor Cell Evasion Strategies. Cancers 2021, 13, 5189. [Google Scholar] [CrossRef] [PubMed]
  123. Sun, L.; Meckes, D.G., Jr. Methodological Approaches to Study Extracellular Vesicle miRNAs in Epstein-Barr Virus-Associated Cancers. Int. J. Mol. Sci. 2018, 19, 2810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Tsao, S.W.; Tsang, C.M.; Lo, K.W. Epstein-Barr virus infection and nasopharyngeal carcinoma. Philos. Trans. R. Soc. B Biol. Sci. 2017, 372, 20160270. [Google Scholar] [CrossRef] [Green Version]
  125. Dunmire, S.K.; Hogquist, K.A.; Balfour, H.H. Infectious Mononucleosis. Curr. Top. Microbiol. Immunol. 2015, 390, 211–240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Dunmire, S.K.; Verghese, P.S.; Balfour, H.H., Jr. Primary Epstein-Barr virus infection. J. Clin. Virol. 2018, 102, 84–92. [Google Scholar] [CrossRef]
  127. Grande, B.M.; Gerhard, D.S.; Jiang, A.; Griner, N.B.; Abramson, J.S.; Alexander, T.B.; Allen, H.; Ayers, L.W.; Bethony, J.M.; Bhatia, K.; et al. Genome-wide discovery of somatic coding and noncoding mutations in pediatric endemic and sporadic Burkitt lymphoma. Blood 2019, 133, 1313–1324. [Google Scholar] [CrossRef]
  128. Xiong, J.; Cui, B.W.; Wang, N.; Dai, Y.T.; Zhang, H.; Wang, C.F.; Zhong, H.J.; Cheng, S.; Ou-Yang, B.S.; Hu, Y.; et al. Genomic and Transcriptomic Characterization of Natural Killer T Cell Lymphoma. Cancer Cell 2020, 37, 403–419.e6. [Google Scholar] [CrossRef]
  129. Png, Y.T.; Yang, A.Z.Y.; Lee, M.Y.; Chua, M.J.M.; Lim, C.M. The Role of NK Cells in EBV Infection and EBV-Associated NPC. Viruses 2021, 13, 300. [Google Scholar] [CrossRef]
  130. Cheerathodi, M.; Nkosi, D.; Cone, A.S.; York, S.B.; Meckes, D.G., Jr. Epstein-Barr Virus LMP1 Modulates the CD63 Interactome. Viruses 2021, 13, 675. [Google Scholar] [CrossRef]
  131. Teow, S.Y.; Liew, K.; Khoo, A.S.; Peh, S.C. Pathogenic Role of Exosomes in Epstein-Barr Virus (EBV)-Associated Cancers. Int. J. Biol. Sci. 2017, 13, 1276–1286. [Google Scholar] [CrossRef] [Green Version]
  132. Higgs, P.G.; Lehman, N. The RNA World: Molecular cooperation at the origins of life. Nat. Rev. Genet. 2015, 16, 7–17. [Google Scholar] [CrossRef]
  133. Carthew, R.W.; Sontheimer, E.J. Origins and Mechanisms of miRNAs and siRNAs. Cell 2009, 136, 642–655. [Google Scholar] [CrossRef] [Green Version]
  134. Saw, P.E.; Xu, X.; Chen, J.; Song, E.W. Non-coding RNAs: The new central dogma of cancer biology. Sci. China Life Sci. 2021, 64, 22–50. [Google Scholar] [CrossRef] [PubMed]
  135. Zhou, W.Y.; Cai, Z.R.; Liu, J.; Wang, D.S.; Ju, H.Q.; Xu, R.H. Circular RNA: Metabolism, functions and interactions with proteins. Mol. Cancer 2020, 19, 172. [Google Scholar] [CrossRef] [PubMed]
  136. Ma, H.; Liang, C.; Wang, G.; Jia, S.; Zhao, Q.; Xiang, Z.; Li, Y.; Cho, W.C.; Pestell, R.G.; Liang, L.; et al. MicroRNA-mediated cancer metastasis regulation via heterotypic signals in the microenvironment. Curr. Pharm. Biotechnol. 2014, 15, 455–458. [Google Scholar] [CrossRef]
  137. 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]
  138. Bigagli, E.; Locatello, L.G.; Di Stadio, A.; Maggiore, G.; Valdarnini, F.; Bambi, F.; Gallo, O.; Luceri, C. Extracellular vesicles miR-210 as a potential biomarker for diagnosis and survival prediction of oral squamous cell carcinoma patients. J. Oral. Pathol. Med. 2022, 51, 350–357. [Google Scholar] [CrossRef]
  139. Zhan, C.; Yang, X.; Yin, X.; Hou, J. Exosomes and other extracellular vesicles in oral and salivary gland cancers. Oral. Dis. 2020, 26, 865–875. [Google Scholar] [CrossRef]
  140. Ludwig, S.; Sharma, P.; Wise, P.; Sposto, R.; Hollingshead, D.; Lamb, J.; Lang, S.; Fabbri, M.; Whiteside, T.L. mRNA and miRNA Profiles of Exosomes from Cultured Tumor Cells Reveal Biomarkers Specific for HPV16-Positive and HPV16-Negative Head and Neck Cancer. Int. J. Mol. Sci. 2020, 21, 8570. [Google Scholar] [CrossRef] [PubMed]
  141. Meckes, D.G., Jr.; Shair, K.H.; Marquitz, A.R.; Kung, C.P.; Edwards, R.H.; Raab-Traub, N. Human tumor virus utilizes exosomes for intercellular communication. Proc. Natl. Acad. Sci. USA 2010, 107, 20370–20375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Zhang, W.L.; Liu, Y.; Jiang, J.; Tang, Y.J.; Tang, Y.L.; Liang, X.H. Extracellular vesicle long non-coding RNA-mediated crosstalk in the tumor microenvironment: Tiny molecules, huge roles. Cancer Sci. 2020, 111, 2726–2735. [Google Scholar] [CrossRef] [PubMed]
  143. Jiang, H.; Zhou, L.; Shen, N.; Ning, X.; Wu, D.; Jiang, K.; Huang, X. M1 macrophage-derived exosomes and their key molecule lncRNA HOTTIP suppress head and neck squamous cell carcinoma progression by upregulating the TLR5/NF-κB pathway. Cell Death Dis. 2022, 13, 183. [Google Scholar] [CrossRef] [PubMed]
  144. Wu, L.; Ye, S.; Yao, Y.; Zhang, C.; Liu, W. Oral Cancer Stem Cell-Derived Small Extracellular Vesicles Promote M2 Macrophage Polarization and Suppress CD4(+) T-Cell Activity by Transferring UCA1 and Targeting LAMC2. Stem. Cells Int. 2022, 2022, 5817684. [Google Scholar] [CrossRef]
  145. Ye, D.; Gong, M.; Deng, Y.; Fang, S.; Cao, Y.; Xiang, Y.; Shen, Z. Roles and clinical application of exosomal circRNAs in the diagnosis and treatment of malignant tumors. J. Transl. Med. 2022, 20, 161. [Google Scholar] [CrossRef] [PubMed]
  146. Li, Y.; Zheng, Q.; Bao, C.; Li, S.; Guo, W.; Zhao, J.; Chen, D.; Gu, J.; He, X.; Huang, S. Circular RNA is enriched and stable in exosomes: A promising biomarker for cancer diagnosis. Cell Res. 2015, 25, 981–984. [Google Scholar] [CrossRef] [Green Version]
  147. Wu, Y.; Zhang, Y.; Zheng, X.; Dai, F.; Lu, Y.; Dai, L.; Niu, M.; Guo, H.; Li, W.; Xue, X.; et al. Circular RNA circCORO1C promotes laryngeal squamous cell carcinoma progression by modulating the let-7c-5p/PBX3 axis. Mol. Cancer 2020, 19, 99. [Google Scholar] [CrossRef]
  148. Wang, Z.; Sun, A.; Yan, A.; Yao, J.; Huang, H.; Gao, Z.; Han, T.; Gu, J.; Li, N.; Wu, H.; et al. Circular RNA MTCL1 promotes advanced laryngeal squamous cell carcinoma progression by inhibiting C1QBP ubiquitin degradation and mediating beta-catenin activation. Mol. Cancer 2022, 21, 92. [Google Scholar] [CrossRef]
  149. Gao, W.; Guo, H.; Niu, M.; Zheng, X.; Zhang, Y.; Xue, X.; Bo, Y.; Guan, X.; Li, Z.; Guo, Y.; et al. circPARD3 drives malignant progression and chemoresistance of laryngeal squamous cell carcinoma by inhibiting autophagy through the PRKCI-Akt-mTOR pathway. Mol. Cancer 2020, 19, 166. [Google Scholar] [CrossRef]
  150. Verduci, L.; Ferraiuolo, M.; Sacconi, A.; Ganci, F.; Vitale, J.; Colombo, T.; Paci, P.; Strano, S.; Macino, G.; Rajewsky, N.; et al. The oncogenic role of circPVT1 in head and neck squamous cell carcinoma is mediated through the mutant p53/YAP/TEAD transcription-competent complex. Genome Biol. 2017, 18, 237. [Google Scholar] [CrossRef] [Green Version]
  151. Wang, J.; Zhou, Y.; Lu, J.; Sun, Y.; Xiao, H.; Liu, M.; Tian, L. Combined detection of serum exosomal miR-21 and HOTAIR as diagnostic and prognostic biomarkers for laryngeal squamous cell carcinoma. Med. Oncol. 2014, 31, 148. [Google Scholar] [CrossRef]
  152. 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] [Green Version]
  153. Ye, S.B.; Zhang, H.; Cai, T.T.; Liu, Y.N.; Ni, J.J.; He, J.; Peng, J.Y.; Chen, Q.Y.; Mo, H.Y.; Jun, C.; et al. Exosomal miR-24-3p impedes T-cell function by targeting FGF11 and serves as a potential prognostic biomarker for nasopharyngeal carcinoma. J. Pathol. 2016, 240, 329–340. [Google Scholar] [CrossRef] [PubMed]
  154. Langevin, S.; Kuhnell, D.; Parry, T.; Biesiada, J.; Huang, S.; Wise-Draper, T.; Casper, K.; Zhang, X.; Medvedovic, M.; Kasper, S. 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, 82459–82474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Ramayanti, O.; Verkuijlen, S.; Novianti, P.; Scheepbouwer, C.; Misovic, B.; Koppers-Lalic, D.; van Weering, J.; Beckers, L.; Adham, M.; Martorelli, D.; et al. Vesicle-bound EBV-BART13-3p miRNA in circulation distinguishes nasopharyngeal from other head and neck cancer and asymptomatic EBV-infections. Int. J. Cancer 2019, 144, 2555–2566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Momen-Heravi, F.; Bala, S. Extracellular vesicles in oral squamous carcinoma carry oncogenic miRNA profile and reprogram monocytes via NF-κB pathway. Oncotarget 2018, 9, 34838–34854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Bao, L.; You, B.; Shi, S.; Shan, Y.; Zhang, Q.; Yue, H.; Zhang, J.; Zhang, W.; Shi, Y.; Liu, Y.; et al. Metastasis-associated miR-23a from nasopharyngeal carcinoma-derived exosomes mediates angiogenesis by repressing a novel target gene TSGA10. Oncogene 2018, 37, 2873–2889. [Google Scholar] [CrossRef] [PubMed]
  158. Lu, J.; Liu, Q.H.; Wang, F.; Tan, J.J.; Deng, Y.Q.; Peng, X.H.; Liu, X.; Zhang, B.; Xu, X.; Li, X.P. Exosomal miR-9 inhibits angiogenesis by targeting MDK and regulating PDK/AKT pathway in nasopharyngeal carcinoma. J. Exp. Clin. Cancer Res. 2018, 37, 147. [Google Scholar] [CrossRef] [Green Version]
  159. Xie, C.; Du, L.Y.; Guo, F.; Li, X.; Cheng, B. Exosomes derived from microRNA-101-3p-overexpressing human bone marrow mesenchymal stem cells suppress oral cancer cell proliferation, invasion, and migration. Mol. Cell Biochem. 2019, 458, 11–26. [Google Scholar] [CrossRef]
  160. 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]
  161. 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] [PubMed]
  162. Mayne, G.C.; Woods, C.M.; Dharmawardana, N.; Wang, T.; Krishnan, S.; Hodge, J.C.; Foreman, A.; Boase, S.; Carney, A.S.; Sigston, E.A.W.; et al. Cross validated serum small extracellular vesicle microRNAs for the detection of oropharyngeal squamous cell carcinoma. J. Transl. Med. 2020, 18, 280. [Google Scholar] [CrossRef] [PubMed]
  163. Wang, H.; Wang, L.; Zhou, X.; Luo, X.; Liu, K.; Jiang, E.; Chen, Y.; Shao, Z.; Shang, Z. OSCC Exosomes Regulate miR-210-3p Targeting EFNA3 to Promote Oral Cancer Angiogenesis through the PI3K/AKT Pathway. Biomed. Res. Int. 2020, 2020, 2125656. [Google Scholar] [CrossRef] [PubMed]
  164. 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]
  165. Shuang, Y.; Yao, X.; Liu, J.; Niu, J.; Guo, W.; Li, C. Serum-derived extracellular vesicles mediate Smad4 expression through shuttling microRNA-27a in the progression of laryngeal squamous cell carcinoma. Hum. Cell 2022, 35, 1084–1099. [Google Scholar] [CrossRef]
  166. Yin, H.; Qiu, X.; Shan, Y.; You, B.; Xie, L.; Zhang, P.; Zhao, J.; You, Y. HIF-1α downregulation of miR-433-3p in adipocyte-derived exosomes contributes to NPC progression via targeting SCD1. Cancer Sci. 2021, 112, 1457–1470. [Google Scholar] [CrossRef]
  167. Tong, F.; Mao, X.; Zhang, S.; Xie, H.; Yan, B.; Wang, B.; Sun, J.; Wei, L. HPV + HNSCC-derived exosomal miR-9 induces macrophage M1 polarization and increases tumor radiosensitivity. Cancer Lett. 2020, 478, 34–44. [Google Scholar] [CrossRef]
  168. Jiang, L.; Zhang, Y.; Li, B.; Kang, M.; Yang, Z.; Lin, C.; Hu, K.; Wei, Z.; Xu, M.; Mi, J.; et al. miRNAs derived from circulating small extracellular vesicles as diagnostic biomarkers for nasopharyngeal carcinoma. Cancer Sci. 2021, 112, 2393–2404. [Google Scholar] [CrossRef]
  169. 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] [PubMed]
  170. Liu, J.; Zhu, M.; Tang, Q. Human umbilical cord mesenchymal stem cells-derived exosomal microRNA-181a retards nasopharyngeal carcinoma development by mediating KDM5C. J. Cancer Res. Clin. Oncol. 2021, 147, 2867–2877. [Google Scholar] [CrossRef]
  171. Panvongsa, W.; Siripoon, T.; Worakitchanon, W.; Arsa, L.; Trachu, N.; Jinawath, N.; Ngamphaiboon, N.; Chairoungdua, A. Plasma extracellular vesicle microRNA-491-5p as diagnostic and prognostic marker for head and neck squamous cell carcinoma. Cancer Sci. 2021, 112, 4257–4269. [Google Scholar] [CrossRef] [PubMed]
  172. Yu, F.; Lin, Y.; Tan, G.; Ai, M.; Gong, H.; Liu, W.; Huang, J.; Zou, Z. Tumor-derived exosomal microRNA-15b-5p augments laryngeal cancer by targeting TXNIP. Cell Cycle 2022, 21, 730–740. [Google Scholar] [CrossRef] [PubMed]
  173. Jiang, J.; Tang, Q.; Gong, J.; Jiang, W.; Chen, Y.; Zhou, Q.; Aldeen, A.; Wang, S.; Li, C.; Lv, W.; et al. Radiosensitizer EXO-miR-197-3p Inhibits Nasopharyngeal Carcinoma Progression and Radioresistance by Regulating the AKT/mTOR Axis and HSPA5-mediated Autophagy. Int. J. Biol. Sci. 2022, 18, 1878–1895. [Google Scholar] [CrossRef] [PubMed]
  174. Lou, C.; Shi, J.; Xu, Q. Exosomal miR-626 promotes the malignant behavior of oral cancer cells by targeting NFIB. Mol. Biol. Rep. 2022, 49, 4829–4840. [Google Scholar] [CrossRef] [PubMed]
  175. Sahu, S.; Routray, S. Assessing the analytical efficacy of TEX in diagnosing oral cancer using a systematic review approach. J. Oral. Pathol. Med. 2021, 50, 123–128. [Google Scholar] [CrossRef]
  176. Yu, Z.; Ni, F.; Chen, Y.; Zhang, J.; Cai, J.; Shi, W. miR-125b suppresses cell proliferation and metastasis by targeting HAX-1 in esophageal squamous cell carcinoma. Pathol. Res. Pract. 2020, 216, 152792. [Google Scholar] [CrossRef]
  177. You, B.; Cao, X.; Shao, X.; Ni, H.; Shi, S.; Shan, Y.; Gu, Z.; You, Y. Clinical and biological significance of HAX-1 overexpression in nasopharyngeal carcinoma. Oncotarget 2016, 7, 12505–12524. [Google Scholar] [CrossRef] [Green Version]
  178. Ferris, R.L.; Blumenschein, G., Jr.; Fayette, J.; Guigay, J.; Colevas, A.D.; Licitra, L.; Harrington, K.; Kasper, S.; Vokes, E.E.; Even, C.; et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2016, 375, 1856–1867. [Google Scholar] [CrossRef] [Green Version]
  179. Theodoraki, M.N.; Laban, S.; Jackson, E.K.; Lotfi, R.; Schuler, P.J.; Brunner, C.; Hoffmann, T.K.; Whiteside, T.L.; Hofmann, L. Changes in circulating exosome molecular profiles following surgery/(chemo)radiotherapy: Early detection of response in head and neck cancer patients. Br. J. Cancer 2021, 125, 1677–1686. [Google Scholar] [CrossRef]
  180. Kannan, A.; Hertweck, K.L.; Philley, J.V.; Wells, R.B.; Dasgupta, S. Genetic Mutation and Exosome Signature of Human Papilloma Virus Associated Oropharyngeal Cancer. Sci. Rep. 2017, 7, 46102. [Google Scholar] [CrossRef] [Green Version]
  181. Visan, K.S.; Lobb, R.J.; Wen, S.W.; Bedo, J.; Lima, L.G.; Krumeich, S.; Palma, C.; Ferguson, K.; Green, B.; Niland, C.; et al. Blood-Derived Extracellular Vesicle-Associated miR-3182 Detects Non-Small Cell Lung Cancer Patients. Cancers 2022, 14, 257. [Google Scholar] [CrossRef] [PubMed]
  182. Nguyen, L.T.H.; Zhang, J.; Rima, X.Y.; Wang, X.; Kwak, K.J.; Okimoto, T.; Amann, J.; Yoon, M.J.; Shukuya, T.; Chiang, C.L.; et al. An immunogold single extracellular vesicular RNA and protein ((Au) SERP) biochip to predict responses to immunotherapy in non-small cell lung cancer patients. J. Extracell. Vesicles 2022, 11, e12258. [Google Scholar] [CrossRef] [PubMed]
  183. Peng, X.X.; Yu, R.; Wu, X.; Wu, S.Y.; Pi, C.; Chen, Z.H.; Zhang, X.C.; Gao, C.Y.; Shao, Y.W.; Liu, L.; et al. Correlation of plasma exosomal microRNAs with the efficacy of immunotherapy in EGFR/ALK wild-type advanced non-small cell lung cancer. J. Immunother. Cancer 2020, 8, e000376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Biogenesis of EV subtypes and their transporting mode. Early endosomes sprout inwards from the cell membrane to form intraluminal vesicles, engulf intracellular components (such as RNAs, proteins, and other metabolites), transform into multivesicular bodies and then release exosomes into extracellular space. As for microvesicles, they bud from plasma membrane, and accepted by recipient cells via (a) direct fusion, (b) phagocytosis, (c) receptor-mediated or not endocytosis. EVs could be identified and extracted by liquid biopsy as cell-free DNA and circulating tumor cells.
Figure 1. Biogenesis of EV subtypes and their transporting mode. Early endosomes sprout inwards from the cell membrane to form intraluminal vesicles, engulf intracellular components (such as RNAs, proteins, and other metabolites), transform into multivesicular bodies and then release exosomes into extracellular space. As for microvesicles, they bud from plasma membrane, and accepted by recipient cells via (a) direct fusion, (b) phagocytosis, (c) receptor-mediated or not endocytosis. EVs could be identified and extracted by liquid biopsy as cell-free DNA and circulating tumor cells.
Cancers 15 01826 g001
Figure 2. EV cargoes and the clinical application of EV-miRNA during HNSCC diagnosis and treatment processes. The exact influences caused by EVs depend on their cargoes. Oncogenic EVs activate the vasculature, promote epithelial–mesenchymal transition, facilitate formation of pre-metastatic niche, protect tumor cells from immune supervision and realize many other functions. Based on current technologies, EVs could be detected and extracted from saliva, blood, urine and tissue samples. Much evidence supports the great significance of EV-miRNAs in the diagnosis, progression, prognosis prediction and treatment evaluation of HNSCC. The miRNAs shown in bold (e.g., BART7-3p) are EBV-derived.
Figure 2. EV cargoes and the clinical application of EV-miRNA during HNSCC diagnosis and treatment processes. The exact influences caused by EVs depend on their cargoes. Oncogenic EVs activate the vasculature, promote epithelial–mesenchymal transition, facilitate formation of pre-metastatic niche, protect tumor cells from immune supervision and realize many other functions. Based on current technologies, EVs could be detected and extracted from saliva, blood, urine and tissue samples. Much evidence supports the great significance of EV-miRNAs in the diagnosis, progression, prognosis prediction and treatment evaluation of HNSCC. The miRNAs shown in bold (e.g., BART7-3p) are EBV-derived.
Cancers 15 01826 g002
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

Chen, B.; Leung, L.L.; Qu, X.; Chan, J.Y.-K. Extracellular Vesicles as Biomarkers in Head and Neck Squamous Cell Carcinoma: From Diagnosis to Disease-Free Survival. Cancers 2023, 15, 1826. https://doi.org/10.3390/cancers15061826

AMA Style

Chen B, Leung LL, Qu X, Chan JY-K. Extracellular Vesicles as Biomarkers in Head and Neck Squamous Cell Carcinoma: From Diagnosis to Disease-Free Survival. Cancers. 2023; 15(6):1826. https://doi.org/10.3390/cancers15061826

Chicago/Turabian Style

Chen, Bojie, Leanne Lee Leung, Xinyu Qu, and Jason Ying-Kuen Chan. 2023. "Extracellular Vesicles as Biomarkers in Head and Neck Squamous Cell Carcinoma: From Diagnosis to Disease-Free Survival" Cancers 15, no. 6: 1826. https://doi.org/10.3390/cancers15061826

APA Style

Chen, B., Leung, L. L., Qu, X., & Chan, J. Y. -K. (2023). Extracellular Vesicles as Biomarkers in Head and Neck Squamous Cell Carcinoma: From Diagnosis to Disease-Free Survival. Cancers, 15(6), 1826. https://doi.org/10.3390/cancers15061826

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