Next Article in Journal
A Cell-Based Systematic Review on the Role of Annexin A1 in Triple-Negative Breast Cancers
Next Article in Special Issue
RNADSN: Transfer-Learning 5-Methyluridine (m5U) Modification on mRNAs from Common Features of tRNA
Previous Article in Journal
BcWRKY33A Enhances Resistance to Botrytis cinerea via Activating BcMYB51-3 in Non-Heading Chinese Cabbage
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Promising Biomarkers in Head and Neck Cancer: The Most Clinically Important miRNAs

by
Arsinoe C. Thomaidou
1,†,
Panagiota Batsaki
2,†,
Maria Adamaki
1,
Maria Goulielmaki
2,
Constantin N. Baxevanis
2,
Vassilis Zoumpourlis
1,* and
Sotirios P. Fortis
2,*
1
Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece
2
Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(15), 8257; https://doi.org/10.3390/ijms23158257
Submission received: 27 June 2022 / Revised: 18 July 2022 / Accepted: 21 July 2022 / Published: 26 July 2022
(This article belongs to the Special Issue RNA Modifications in Disease and Development)

Abstract

:
Head and neck cancers (HNCs) comprise a heterogeneous group of tumors that extend from the oral cavity to the upper gastrointestinal tract. The principal etiologic factors for oral tumors include tobacco smoking and alcohol consumption, while human papillomavirus (HPV) infections have been accused of a high incidence of pharyngeal tumors. Accordingly, HPV detection has been extensively used to categorize carcinomas of the head and neck. The diverse nature of HNC highlights the necessity for novel, sensitive, and precise biomarkers for the prompt diagnosis of the disease, its successful monitoring, and the timely prognosis of patient clinical outcomes. In this context, the identification of certain microRNAs (miRNAs) and/or the detection of alterations in their expression patterns, in a variety of somatic fluids and tissues, could serve as valuable biomarkers for precision oncology. In the present review, we summarize some of the most frequently studied miRNAs (including miR-21, -375, -99, -34a, -200, -31, -125a/b, -196a/b, -9, -181a, -155, -146a, -23a, -16, -29, and let-7), their role as biomarkers, and their implication in HNC pathogenesis. Moreover, we designate the potential of given miRNAs and miRNA signatures as novel diagnostic and prognostic tools for successful patient stratification. Finally, we discuss the currently ongoing clinical trials that aim to identify the diagnostic, prognostic, or therapeutic utility of miRNAs in HNC.

1. Introduction

Head and neck squamous cell carcinoma (HNSCC) is one of the six most common cancers worldwide, counting more than 850,000 new cases and 400,000 deaths, annually [1]. It is considered as one of the most aggressive cancer types, with serious repercussions for patient quality of life, mainly due to advanced local disease and low responsiveness to treatment [2]. HNSCC is an epithelial malignancy that comprises a highly heterogeneous group of tumors located in the oral cavity, larynx, nasopharynx, oropharynx, and hypopharynx [3]. The most common HNSCC risk factors are related to tobacco smoking and alcohol consumption [1]. Human papillomavirus (HPV) infection is also considered a major causative agent, especially in tumors located in the oropharynx [1]. Popular treatment regimens involve surgical removal, radiotherapy (RT), and chemotherapy (CT). Despite recent advances, such as new targeted therapies that include immune checkpoint inhibitors [3], existing therapeutic modalities are often unsuccessful, usually accompanied by a high post-operative recurrence rate and no obvious improvement in five-year survival rates [1,4]. Therefore, HNSCC patients still witness poor prognosis and survival rates remain low, since up to 25% of the affected individuals develop local recurrence or distant metastases [5], as well as lymph node metastases (LNM) [6].
Abnormal expression and accumulation of mutations in oncogenes or tumor suppressor genes are the major steps for head and neck cancer (HNC) development and tumor progression [7,8]. HNC cells have the ability to communicate with external microenvironments and to escape detection by the host immune system. Considering the complexity of HNSCC and the urgent need for improved diagnosis and prognosis of the disease, most researchers have focused on finding novel, clinically relevant biomarkers. Previous studies have shown that certain microRNAs (miRNAs) in HNSCC are differentially expressed, functioning as regulators of tumor suppressors or oncogenes [9,10].
MiRNAs make up a group of non-protein-coding RNAs approximately 18–22 nucleotides long [11]. The biogenesis of miRNAs arises in the nucleus, where they are subsequently exported from into the cytoplasm for further processing [12]. An abundance of human miRNAs has been reported to date, with the majority being able to target and regulate multiple genes. Briefly, miRNAs bind to the 3′-UTR of mRNAs, thereby inducing cleavage and post-transcriptional silencing of these target molecules [13,14]. MiRNAs can control diverse biological functions including embryogenesis, cellular development, and homeostasis, but also appear to play a role in tumorigenesis and cancer development, functioning as regulators of either oncogenes or tumor suppressor genes [15,16]. Aberrantly expressed miRNAs are implicated in several oncogenic processes, including cell proliferation, differentiation, migration, and apoptosis [17,18]. Recently, an increasing number of studies have focused on the differential expression of certain miRNAs in various solid tumors, such as breast [19], lung [20], prostate [21], colon [22], ovarian [23], and HNC [9]. Deregulated miRNA expression levels can be tissue-specific and miRNAs can be characterized as oncogenic (OncomiRs), or tumor suppressor miRNAs, depending on the genes that they regulate and their mode of action. However, it is important to note that each miRNA could exhibit a different expression pattern and function in different cancer types. Furthermore, it has been shown that miRNA signatures can serve as biomarkers for diagnosis and prognosis, and for predicting a patient’s response to treatment [24,25]. There is also enough evidence to suggest that a significant number of miRNAs are abnormally expressed during HNSCC tumorigenesis and progression, either functioning as OncomiRs or as tumor suppressors [26].
In the present review, we summarize some of the most extensively studied miRNAs, the expression levels of which are aberrantly altered and, in this way, involved in the pathogenesis of HNSCC. Furthermore, we highlight the functional characteristics of clinically significant miRNAs that play a crucial role in many cancers, including HNSCC. We also discuss the specific molecular mechanisms of miRNA regulation in an attempt to further unravel their implications in cancer initiation and progression. Finally, we outline how individual miRNAs and miRNA signatures can be used as novel biomarkers for the diagnosis, prognosis, and therapy of HNSCC, as well as their significance in the design and implementation of future therapeutic approaches.

2. The Role of the Most Common miRNAs in HNC

In this section, certain characteristics and biomarker abilities of the most commonly identified miRNAs in HNC and in other malignancies are thoroughly analyzed.

2.1. MiR-21

MiR-21 is currently one of the most extensively studied miRNAs, mainly because it is one of the few miRNAs that are found consistently overexpressed in a number of human cancer types, including ovarian [27], lung [28], gastric [29], breast [30], colorectal [31], B-cell lymphoma [32], and glioblastoma [33]. In a similar manner, miR-21 levels have been consistently found higher in the tumor tissue [34,35,36,37], serum (exosomes) [38,39], plasma [40,41,42,43], saliva [37], and whole blood [44] samples of patients, as compared to those of healthy controls, in the majority of HNC subtypes [40]. miR-21 has clear oncogenic properties in HNC, as it targets a number of known onco-suppressor genes, such as PTEN [45], p53 [46], p63 [47], and PDCD4 [48,49,50], thus playing an important role in several cancer-related processes, including cellular proliferation [51], invasion [51,52], metastasis [53], and apoptosis [48]. miR-21 has been proposed as a potential biomarker of diagnostic, prognostic, and therapeutic value in HNC. miR-21 expression has demonstrated high diagnostic accuracy in distinguishing between oral squamous cell carcinoma (OSCC) tumor tissue and healthy mucosa samples [54], and in evaluating cervical lymph node metastasis in patients with OSCC [37]. A number of different studies have reported that aberrantly increased levels of miR-21 expression correlate with advanced cancer stages, lymph node metastasis, poorer prognosis, and ultimately decreased survival of patients with laryngeal squamous cell carcinoma (LSCC) [34,55] and OSCC [56,57]. In the same context, miR-21 overexpression appears to be an independent prognostic marker of poor survival in patients with squamous cell carcinoma of the tongue (TSCC) [58]. miR-21 has also the potential to be used as a monitoring biomarker, since its expression levels seem to diminish after surgery in HNSCC patients with good prognosis, but remain high in patients with poor prognosis [36,41]. It has been suggested that through the targeting of PDCD4, miR-21 could sensitize chemoresistant TSCC to cisplatin treatment, thereby constituting a potential target for TSCC therapy [59].

2.2. miR-375

miR-375 has been found consistently downregulated in tumor tissue [60,61,62,63], serum [64], plasma [41,42], and saliva [63] samples of patients with HNC. miR-375 appears to play a tumor-suppressing role in HNC, as it has been shown to suppress cancer cell proliferation, migration, and invasion; this is possibly achieved by targeting XPR1 in esophageal squamous cell carcinoma (ESCC) [61], HNF1β in LSCC [65], as well as PDK1 [60] and USP1 [66] in nasopharyngeal carcinoma (NPC). In a study on patients with primary HNSCC, miR-375 expression was successfully utilized in distinguishing HNSCC carcinoma tissues from non-cancerous tumor-adjacent tissues with 87.5% sensitivity and 65% specificity [67]. The low expression of miR-375 was found to significantly correlate with cancer aggressiveness as it is associated with poor overall survival (OS) in ESCC [68] and higher TNM stages in LSCC patients [34]. These findings suggest that miR-375 could potentially serve as a diagnostic and prognostic marker in HNC. The downregulation of plasma miR-375 has been highly associated with disease recurrence after surgery in OSCC patients, suggesting that circulating miR-375 could be utilized as a post-operative surveillance marker for this type of HNSCC [42]. Different anti-cancer drugs (doxorubicin, 5-fluorouracil, trichostatin A, and etoposide) have been shown to reactivate and increase miR-375 expression in tongue cancer cells, possibly indicating that miR-375 may mediate cellular reactions in response to these drugs [69].

2.3. miR-99

The miR-99 family has been shown to function as a tumor suppressor in a number of cancer types, including cervical carcinoma [70], prostate cancer [71], and glioma [72]. In HNSCC, the overexpression of miR-99a and miR-100 suppresses cancer cell proliferation, migration, and invasion [73,74]. Similarly, miR-99*, a passenger strand of miR-99, appears to act as an onco-suppressor in HNC and its expression levels have been found to be significantly downregulated in the tumor tissues of patients with OSCC, compared to adjacent non-cancerous tissues [75,76]. miR-100, miR-99a, and miR-99b, also members of the miR-99 family, have all been found to be downregulated in the tumor tissue samples of HNSCC patients [73,77,78,79,80,81,82,83,84,85], with the exception of one study that reported miR-100 overexpression in the tumor tissue samples of HNSCC patients [86]. miR-99a and miR-100 expression has been found to be lower in the serum and plasma samples of OSCC patients [87,88], with miR-99a also appearing to be downregulated in the plasma samples of HNSCC patients [36], as compared to healthy controls. Notably, miR-99a expression levels have been found to be significantly higher in post-operative OSCC than in pre-operative samples, and have the potential to distinguish OSCC cases from healthy controls, indicating the potential of miR-99 as a diagnostic biomarker [87].
miR-99a, miR-99b, and miR-99* downregulation has been closely associated with LNM [73], advanced clinical stage [73,79,81,87], disease recurrence [75], and worse survival outcomes [89] in patients with HNSCC, while miR-99* overexpression has been shown to correlate with better progression-free survival (PFS) and overall survival (OS) [75]. Interestingly, increased miR-100 expression levels have been reported to significantly correlate with poorer survival outcomes, suggesting that miR-100 might be a prognostic biomarker in OSCC [79]. The aforementioned findings suggest that miR-99 members could be utilized as independent prognostic factors of OS in OSCC patients, and warrant additional follow-up studies to further assess and validate their prognostic value, as well as their potential as therapeutic markers.

2.4. miR-34a

MiR-34a functions as a tumor suppressor and is downregulated in a number of cancer types [90], including colorectal [91], neuroblastoma [92], prostate [93], and thyroid cancer [94]. It interacts with a variety of genes that are known to be involved in oncogenic processes, such as p53 [95,96,97], Bcl-2 [98], cyclin D1 [99], CDK4 [90], and VEGF [90], and has been reported to affect tumor cell proliferation [90,99], apoptosis [95], senescence [100], invasion [98], metastasis [101], and drug resistance [102,103]. In HNSCC, expression levels of miR-34a appear to be significantly lower in tissue samples as compared to normal tissues, and for this reason miR-34a has been suggested as a novel and highly sensitive biomarker for diagnostic use in HNSCC [104]. In a number of other studies, similar expression patterns of miR-34a in HNC have been observed [105,106,107,108], with the exception of a study, comparing site-specific HNSCC samples, reported increased miR-34a expression levels in tumor tissues of oropharyngeal carcinoma [109]. Such findings suggest that miRNA expression profiles could be site-specific, consequently affecting the clinico-pathological features of patients with HNSCC [109]. miR-34a downregulation in NPC has been significantly correlated with bone metastasis and TNM staging, while low levels of miR-34a in NPC and sinonasal squamous cell carcinomas (sinonasal SCC) patients have been correlated with a decreased 5-year survival rate [110,111]. These findings suggest that miR-34a expression could serve as a marker for disease prognosis.

2.5. Let-7

Most members of the let-7 family have been found downregulated in the tumor tissues [112,113,114], serum [115,116], and saliva [117] of HNSCC patients, although several studies have also reported an upregulation of certain let-7 members in the tumor tissues and serum of HNSCC patients [115,116,118]. Let-7 miRNAs have been characterized as tumor suppressors in HNC, targeting a number of important oncogenes, including K-RAS [119] and HMGA2 [120]. A number of let-7 family miRNAs have been linked to the regulation of different molecular pathways that contribute to several oncogenic properties of tumor cells, including invasion [120], metastasis [120], stem-like properties [121], EMT [114], and chemoresistance [114].
Salivary let-7a expression levels have demonstrated high sensitivity and specificity in differentiating between HNSCC patients and healthy controls [117]. In addition, let-7a levels have been reported to correlate with different clinical stages of the disease, since lower let-7a expression was observed in advanced laryngeal cancer compared to early-stage samples [120]. Similarly, low levels of let-7 family miRNAs in HNSCC tumors have been significantly associated with worse survival outcomes and more aggressive forms of cancer, suggesting that let-7 could be a potential prognostic marker [112,122].
With regards to therapeutic applications, let-7 appears to be a particularly prospective target, as several studies have reported that the overexpression or restoration of let-7 levels in tumor cells could suppress different oncogenic characteristics that might enhance treatment outcomes [113,123,124].

2.6. MiR-200

The miR-200 family consists of five members (miR-200a, -200b, -200c, -141, -429), which are grouped into two independent transcriptional clusters (the first one containing miR-200a, miR-200b and miR-429, and the second containing miR-200c and miR-141) [125,126,127]. All miR-200 members have been shown to play an important part in repressing malignant cell transformation and in inhibiting tumor initiation, and have thus been characterized as tumor suppressors [127,128]. More specifically, the miR-200 family has been reported to negatively modulate the progress of EMT in various cancers, including HNSCC [129], by controlling the expression of transcriptional repressors ZEB1 and ZEB2, the so-called EMT master regulators [126,130,131].
In terms of predictive and prognostic applications in the clinical setting, a number of studies have highlighted the miR-200 family as promising potential targets. MiR-200c and miR-141 expression levels appear to be significantly downregulated in HNSCC tumor tissues, as compared to non-cancerous samples [126,132]. MiR-200a is present at significantly lower levels in the saliva samples of OSCC patients, as well as in HNSCC patients before radiotherapy [133], suggesting that miR-200a has the potential to serve as a novel, non-invasive marker of detection and monitoring [133,134]. Furthermore, downregulated miR-200a and miR-200c have demonstrated a significant correlation with disease recurrence in LSCC patients after surgery [130].
Several studies also hint at the therapeutic potential of miR-200 family members. In particular, it has been shown that the induced expression of miR-200 family members successfully inhibits the metastatic ability and malignant cancer stem cell (CSC)-like properties of HNSCC cells [132,135,136]. These findings suggest that the restoration of miR-200c levels could be a promising step towards more successful treatment of advanced malignant HNSCC or HNSCC-derived CSC populations [132].

2.7. MiR-31

The literature on miR-31 is quite controversial, as this particular miRNA appears to be downregulated in certain types of cancer, such as liver [137], prostate [138], and triple negative breast cancer [139], but upregulated in colorectal [140], cervical [141], rectal [142], and certain subtypes of lung cancer [143], suggesting that the mode of miR-31 regulation depends on cancer histology. Overall, in HNC, miR-31 mostly appears to be upregulated [46,47,79,144,145,146], acting as an oncogene and promoting the proliferation and invasion of tumor cells [147]. However, contradicting results have even been reported among different HNC subtypes. In a comparative study, it was shown that miR-31 levels in plasma samples were significantly higher in OSCC patients, as compared to healthy controls [148]. Similarly, miR-31 expression was found to be upregulated in the tissue samples of HNSCC patients [149,150]; it has therefore been proposed that particular miRNAs may promote the development of HNSCC by suppressing the FIH-mediated activation of HIF [149], a gene that promotes angiogenesis. On the other hand, two recent studies reported significantly lower miR-31 expression levels in the peripheral blood samples of patients with NPC, as compared to the healthy controls [151], and a downregulation of salivary miR-31 in oropharynx squamous cell carcinoma (OPSCC) patients, as compared to controls [152], respectively. Furthermore, the downregulated expression of miR-31 has been observed in LSCC tissues, and has been correlated with advanced stages of the disease [153]. These findings suggest that miR-31 expression levels are most likely tumor site-specific and might even vary among different sample types. Particularly, it has been shown that miR-31 is more abundantly expressed in the saliva than in the plasma of OSCC patients, thereby suggesting that the salivary detection of miR-31 could represent a more sensitive marker for the diagnosis of oral malignancy [154].
The utility of miR-31 as a biomarker is further explored in several studies which suggest that, in various HNSCC patient sample types (saliva, plasma, tumor tissues), the expression levels of miR-31 increase and are positively correlated with poor pathological parameters [147,153] and advanced staging [155]. MiR-31 quantification could also serve as a useful marker of post-operative follow-up of OSCC, as salivary levels appear to be remarkably reduced after the excision of the oral carcinoma [154].

2.8. MiR-125a/miR-125b

The miR-125 family, composed of the four homologs, miR-125a-3p, miR-125a-5p, miR-125b-1, and miR-125b-2, has been found to play an important role in a number of cancer types, acting either as promoters or suppressors of tumorigenesis [156]. In HNC, these miRNAs are mainly considered to be tumor suppressors and have been found to be significantly downregulated in the tumor tissue [47,157,158,159,160,161,162], plasma [163], and saliva [133,134] samples of patients; however, the precise functional role and mechanism of action related to cancer progression remain quite unresolved. Although miR-125b-2-3p has been reported to suppress tumor progression, LNM, and distant metastasis in HNSCC [83], in another study, miR-125b levels appear to be significantly higher in the metastatic primary HNSCC tumor samples, as well as in the respective metastatic tumor itself, compared with non-metastatic tumors [164], highlighting the controversial role of this miRNA in HNSCC. It is noteworthy that while these findings suggest a strong potential role for miR-125b as a biomarker of metastases and lower disease-specific survival, it is also indicated that miRΝA expression patterns can shift between initial and later stages as the disease progresses [164]. Last but not least, miR-125b has been proposed as a prognostic marker for OSCC, as its downregulation seems to correlate with tumor stage [165], radioresistance [165], poor prognosis, and worse patient survival [162], while miR-125a-5p has been suggested as an individualized biomarker, as its downregulation has been associated with locoregional recurrence and an overall poor prognosis [166].

2.9. MiR-196a/miR-196b

The miR-196 family, which includes two mature miRNA members, miR-196a and miR-196b, has been found to be aberrantly expressed in a number of cancers [167]. MiR-196a and miR-196b seem to be consistently upregulated in the tumor tissue [57,168,169], plasma [170,171,172], and saliva [168] samples of HNC patients as compared to corresponding healthy controls. Both miRNAs possess oncogenic properties. The overexpression of miR-196a has been shown to significantly increase cell proliferation, migration, and invasion, and to induce EMT, possibly through targeting of ANXA1 [173] and MAMDC2 [174] in HNSCC cells, and of HOXB8 and p27 (CDKN1B) in OSCC cells [175]. MiR-196b has been demonstrated to promote cell proliferation and invasion, and to suppress apoptosis by targeting SOCS2 [176] and PCDH-17 [177] in LSCC cells and ANXA1 in HNSCC cells [178]. The combined detection of plasma miR-196a and miR-196b expression appears highly potent in diagnosing oral cancer patients with high sensitivity and specificity [171]. The upregulation of miR-196a has been shown to strongly correlate with poor prognosis and worse survival outcomes in OSCC patients [57,170,172] and LNM in TSCC patients [179]. It has also been highlighted as a promising marker of HNSCC response to radiotherapy, since miR-196a overexpression appears to increase radioresistance in HNSCC cells [173]. Notably, both miR-196a and miR-196b have been characterized as potentially important therapeutic targets, since the inhibition of these miRNAs has been shown to reduce cell proliferation in oral cancer and LSCC cell lines [175,177,180]. Furthermore, both miR-196a and miR-196b have been suggested as promising diagnostic, prognostic, and therapeutic biomarkers for LC. High miR-196b expression levels have been shown to correlate with worse clinicopathological parameters in patients with LSCC; for this reason, it has been suggested as an independent prognostic factor of OS in LSCC patients [176].

2.10. MiR-9

MiR-9 (miR-9-5p and miR-9-3p) is a miRNA that is very often deregulated in cancer; however, its biological role has proven to be quite complex, as it can behave as an oncomiR in certain cases, and as an onco-suppressor miRNA in other cases [181,182]. MiR-9 appears to be downregulated in gastric [183], colorectal [184], and hepatocellular carcinoma [185], and advanced non-small cell lung cancer [186], but upregulated in breast [187] and cervical cancer [188]. In HNC, the regulation and functional role of miR-9 seem to vary, and might depend on the anatomic site or cellular context. For example, miR-9 levels have also been found overexpressed in the tumor tissue samples of sinonasal SCC [189] and in the saliva samples of HNSCC patients, as compared to corresponding healthy controls [190,191]. However, miR-9 is downregulated in the tumor tissue [192] and plasma samples of NPC patients [193,194], and in the tumor tissue [195,196] and serum samples of OSCC patients [197], which is associated with various clinicopathological parameters and poor OS and disease-free survival (DFS); this suggests that miR-9 downregulation in serum could represent a promising independent prognostic factor for OSCC [197].
Interestingly, a higher expression of miR-9-3p has been shown to correlate with the presence of vascular and perineural invasion, whereas the overexpression of miR-9-5p has been associated with longer survival in sinonasal SCC patients [189]. A recent study has also proposed that miR-9 may represent a valuable predictive biomarker of the response to radio-chemotherapy, since high miR-9 expression appeared to be associated with poor prognosis in HNSCC patients treated with RT+CTX (radiotherapy combined with the anti-EGFR monoclonal antibody cetuximab) [198].

2.11. MiR-181a

The miR-181 family is a group of highly conserved miRNAs that act as regulators in various physiological functions, including proliferation, apoptosis, autophagy, mitochondrial activity, and immune response [199,200,201]. The miR-181 family is differentially expressed in solid tumors and hematological cancers and may act either as cancer cell inhibitors or as cancer promoters, underlining diagnostic and prognostic significance [202]. Four mature forms of the miR-181 family are present in the human genome, including miR-181a, miR-181b, miR-181c, and miR-181d.
miR-181a is normally expressed in the neurons, blood, and lymph nodes. In various cancers, aberrant miR-181a expression causes alterations in the expression patterns of genes, leading to tumor progression and cancer cell migration [202]. In particular, abnormal miR-181a expression has been linked to colorectal [203,204], non-small cell lung [205], pancreatic [206], ovarian [207], and prostate cancer [208], as well as HNSCC [209]. In this context, it has been suggested that laryngeal carcinoma cell invasion and migration is inhibited by miR-181a upregulation, as the latter appears to regulate metastasis-related genes, such as N- and E-cadherin and ATF2 [210].
On the other hand, in OSCC patients, miR-181a plasma levels appear to be aberrantly increased and correlate with advanced LNM and vascular invasion, identifying miR-181a as a valuable non-invasive prognostic biomarker [211]. Interestingly, HNC patients, who are also positive for HPV infection, have decreased levels of miR-181a, as expression is suppressed by HPV subtype 16 [212,213]. The downregulation of miR-181a in plasma could represent a useful biomarker of early disease detection in esophageal cancer patients, and possibly for the follow-up of patients, based on evidence that post-operative plasma miR-181a levels are significantly increased [214]. Similarly, miR-181a expression appears to be increased in the serum of ESCC patients, who respond to radiotherapy [215]. This suggests that serum miR-181a quantification could represent a minimally invasive, predictive marker of response to radiotherapy in ESCC patients. However, miR-181a overexpression in tissue correlates with advanced TNM stages and the detection of LNM; therefore, it can be associated with the development or pathogenesis of ESCC [216].

2.12. MiR-155

miR-155 is known to be implicated in the development of several types of cancer, such as non-small cell lung cancer [217], breast [218], gliοma [219], colorectal [220], and HNSCC [221]. In addition, miR-155 expression levels are increased and correlated with patient survival in hepatocellular carcinoma [222]. Increasing evidence suggests that miR-155 is aberrantly expressed in HNC, indicating a strong correlation with patient survival and the metastatic activity of cancer cells. Specifically, serum and plasma miR-155 levels are upregulated in HNSCC patients [64,223] and circulating miR-155 is characterized as a non-invasive HNSCC biomarker, since patients with higher levels of miR-155 also have a higher risk of relapse [64]. In this context, miR-155 may play a role as an oncogenic miRNA in ESCC and OSCC. Indeed, samples derived from tumor tissues have significantly higher levels of miR-155 [160,224,225,226,227], whereas miR-155 overexpression is highly associated with ESCC tumor aggressiveness [224] and LNM and disease recurrence in OSCC patients, indicating that it could be used as a prognostic biomarker for survival [225,226]. The increased expression of miR-155 in HNSCC tissues is probably associated with immune cell tumor infiltration, since the upregulation of the miR-146a/miR-155 cluster has been highly correlated with the expression of immune cell-related mRNA, including NK cells, B-cells, T regulatory cells (Treg), Th1 cells, CD8+ T-cells, and activated CD4+ T-cells [228]. The increased mRNA expression of these immune cell types has also been significantly associated with prolonged OS after the combined administration of chemo- and radiotherapy [228]. miR-155 is also overexpressed in cultured HNSCC cells, resulting in increased cell proliferation, invasion, and decreased apoptotic activity, via the downregulation of the CDC73 gene, which normally favors the inhibition of cell growth and proliferation [229]. Interestingly, certain studies have also shown a reduction in miR-155 levels in the peripheral blood of HNSCC patients [230]. The downregulation of miR-155 increases the risk of distant metastasis and is correlated with the size and extent of the tumor, suggesting that its expression is also related to the survival of TSCC patients [231]. Such observations also suggest that the type of affected tissue and the tumor site can modify the expression levels of particular miRNAs, including miR-155, implying that distinct tissue- and site-specific miRNA profiles are present in HNC.

2.13. MiR-146a

miR-146a is an abundantly expressed miRNA, involved in physiological processes such as the regulation of the immune system, cell maturation, differentiation, and inflammatory responses [232]. The aberrant expression of miR-146a has been detected in several human malignancies, including B cell lymphomas [32], lung [233,234], breast [235], gastric [236], cervical [237], hepatocellular [238], prostate [239], colorectal [240], glioma cancers [241]. miR-146a has been proposed as a marker of HNSCC in both the tumor site and in circulation; miR-146a overexpression is present in tumor tissues, as compared to neighboring non-cancerous tissue in OSCC and LSCC [7]. By evaluating the levels of miR-146a in oral cancer patients and healthy controls, the plasma miR-146a level was found to be higher in the study group [242]. Interestingly, post-operative miR-146a levels are significantly reduced in oral cancer patients, possibly indicating that miRNAs detected in the circulation might originate from tumor cells [242]. In ESCC, miR-146a is downregulated in tumor tissues and serum and is associated with an increase in TNM stage [243,244]. These observations suggest that miRNA expression highly depends on tissue type, as well as on tumor location.
miR-146a could be characterized as a biomarker of prognosis for distant metastasis, since its expression in the whole blood of HNSSC patients has been negatively correlated with the presence of secondary tumors [230]. The expression profile of miR-146a in HNSCC patients is also correlated with human papillomavirus (HPV) infection, with HPV + HNSCC patients exhibiting significantly higher tumor miR-146a levels, as compared to non-infected patients [245]. Moreover, the evaluation of miRNA levels in the peripheral blood mononuclear cells (PBMCs) of ESCC patients has indicated an upregulation of miR-146a, which appears to be a major component of the miR-146a-IRAK1-NF-κB axis [246].

2.14. MiR-23a

miR-23a belongs to the miR-23a-27a-24-2 cluster and is aberrantly expressed in several malignancies, mainly serving as a biomarker for cancer detection [247,248]. miR-23a is overexpressed in breast [249], gastric [250], hepatocellular carcinoma [251], lung [252], pancreatic and colorectal [253], and ovarian cancers [254], as well as acute myeloid leukemia [255]. The biological role of miR-23a in cancer is highly controversial. MiR-23a has been reported to negatively regulate cancer cell development and metabolic activity [256], and the increased expression of miR-23a stimulates caspase-induced apoptosis [257]. It is also suggested that miR-23a overexpression contributes to enhanced chemosensitivity in hepatocellular carcinoma cell lines [258]. In line with this, miR-23a expression has also been correlated with HNC; specifically, miR-23a negatively regulates glucose metabolism and ATP production in HNSCC cell lines and therefore inhibits cancer cell growth and proliferation [256]. On the contrary, miR-23a has been shown to enhance viability, proliferation, and mobility of renal cancer cell lines [259], while contributing to increased chemoresistance in TSCC [260]. In the tissue samples of ESCC patients, miR-23a is upregulated [261,262] and is associated with advanced TNM staging and the development of LNM, leading to a more aggressive cancer phenotype, thus indicating that the expression patterns of this particular miRNA could affect ESCC progression [261]. The miR-23a is also aberrantly overexpressed in the serum of ESCC patients that exhibit lower response rates to neoadjuvant chemotherapy [263]. Interestingly, the quantification of the serum expression levels of three combined miRNAs, namely of miR-193b, miR-873, and miR-23a, could facilitate the identification of responders to neoadjuvant chemotherapy, thereby representing a non-invasive predictive signature of response to therapy in ESCC patients [263].
ESCC patients with low rates of response to chemotherapy present with significantly higher miR-23a tissue and plasma expression levels, with the latter also being significantly correlated with increased TNM staging and lymph node invasion [264]. This highlights a role for the evaluation of miR-23a levels in the circulation and the tumor samples of ESCC patients for their appropriate stratification based on cancer progression and response to treatment. The inhibition of miR-23a leads to decreased therapeutic resistance in TSCC cells, indicating a possible role for miR-23a as a biomarker for the appropriate stratification of HNSCC patients and the prediction of response to chemotherapy [265].

2.15. MiR-16

miR-16 used to serve as an internal control and a miRNA reference for the normalization of other miRNAs. However, it was suggested that miR-16 is abundantly expressed [266] and highly conserved as one of the first miRNAs to be linked to human cancers [267]. miR-16 is a central regulator of the cell cycle, suppressing cell proliferation and tumorigenicity both in vitro and in vivo and promoting cell apoptosis [268]. It has also been well documented as a prominent tumor suppressor in several types of malignancy, including breast [39], colorectal [269], bladder [270], prostate [271], and non-small lung cancer [272]. In HNSCC, miR-16 can be downregulated or upregulated, depending on the cancer subtype and the tissue in which it is expressed. In OSCC tissues, miR-16 is markedly decreased compared to adjacent non-cancerous tissues [273,274,275]. Salivary miR-16 levels are also downregulated in patients with OSCC [276]. Notably, miR-16 expression is downregulated in patients with metastatic oral cancer and a more advanced TNM stage [162,273], and it is associated with poor overall survival; therefore, it could be used as a robust prognostic biomarker [162].
In NPC patients, plasma miR-16 levels are significantly higher compared to controls, indicating a negative correlation with NPC progression [43]. In ESCC, miR-16 has been characterized as an oncogenic miRNA, significantly upregulated in the serum and tumor samples of patients; the high correlation of aberrant miR-16 expression with LNM and increased tumor size seems to contribute to a decreased OS and DFS in these patients and warrants further investigation into its potential to be used as a prognostic biomarker [277,278,279].

2.16. MiR-29

The miR-29 family includes three miRNAs, namely miR-29a, miR-29b, and miR-29c [280]. Several studies have demonstrated that the miR-29 family is mostly downregulated in cancer. Low miR-29 expression has been observed in several solid tumors, such as glioblastoma [281], hepatic cholangiocarcinoma [282], prostate [283], ovarian [284], endometrial [285], non-small cell lung cancer [286], HNC [287], as well as hematopoietic malignancies [288,289]. On the contrary, the miR-29 family appears to be upregulated in breast cancer [290], suggesting that the mode of miR-29 expression probably depends on the cancer type. In general, however, miR-29 family members are regarded as tumor suppressors and usually function as negative regulators of oncogenes or as enhancers of tumor suppressor genes [280]. Specifically, it has been observed that miR-29 promotes cancer cell apoptosis and inhibits tumor cell proliferation and invasiveness by suppressing EMT activity [280].
miR-29 expression levels are significantly decreased in HNSCC tissues and cell lines, as compared to adjacent non-cancerous samples [291,292]. The induced expression of miR-29 hampers the ability of HNSCC cells to proliferate and migrate [291]. In this context, the miR-29 family could be classified as a miRNA tumor-suppressive signature in HNSCC, achieved via targeting LAMC2 and its ligand ITGA6 [291].
miR-29 is a significantly downregulated LC tumor, compared to adjacent normal tissues [293], as well as in NPC tissue samples and cell lines [294]. It is suggested that miR-29c targets the TIAM1 gene, which is closely related to the metastatic activity of cancer cells [294]. In TSCC, miR-29b functions as a tumor suppressor of miRNA, as it is significantly downregulated in both patient tissues and cell lines [295].
There are also references that correlate miR-29 expression levels with the outcome of radiotherapy in HNC patients. Guo et al. (2019) suggested that miR-29a is significantly downregulated in radioresistant NPC patients and cell lines, while the induced expression of miR-29a results in decreased cell viability and enhanced apoptotic activity following cell irradiation [296]. On the same note, miR-29b has also been found to regulate radiosensitivity in HNSCC. In particular, serum miR-29b levels appear to be significantly decreased in ESCC patients with radioresistance [297]. Overall, the aforementioned studies suggest that the miR-29 family act as tumor-suppressive miRNAs, through the regulation of genes and pathways that are involved in the cell cycle and metastasis of HNSCC cells, and could constitute a promising therapeutic target for HNSCC and related subtypes. Moreover, there is enough evidence to suggest that miR-29 could be used as a prognostic biomarker of improved radiotherapeutic outcomes [296,297]. Currently, there are two clinical trials studying the prognostic and therapeutic significance of the miR-29 family in HNC (discussed in a relevant section of this review).
The most commonly upregulated and downregulated miRNAs that have been reported as potential biomarkers in HNSCC are presented in Table 1, Figure 1, Table 2, and Figure 2, respectively. The identified gene hotspots and/or respective pathways and cellular functions targeted by miRNAs in HNC are presented in Table 3.

3. MiRNA Signatures of Diagnostic, Prognostic, and/or Predictive Value in HNC

Although it has been well established that aberrant miRNA expression could serve as a potential biomarker in cancer patients, it should be noted that miRNA deregulation has also been observed in other pathological conditions. For example, abnormally high levels of miR-21, one of the most extensively studied and upregulated miRNAs in HNC, have also been reported in several non-cancerous pathologies, including cardiorenal syndrome [421], restrictive allograft syndrome (RAS) in lung transplant recipients [422], traumatic brain injury [423], and sepsis-associated cardiac dysfunction [424,425]. This is not entirely surprising since miRNAs are well known for targeting multiple genes and for regulating different cellular functions; it does, however, highlight that miRNA might not constitute cancer-specific biomarkers and that single miRNA expression profiles might not be sufficient for diagnostic and prognostic use. In order to increase the predictive properties of miRNA alterations, recent studies have turned to investigating the expression patterns and ratios of multiple miRNAs combined together as opposed to individual miRNA expression profiles.

3.1. MiRNA Ratios/miRNA Combined Expression

The miR-221 to miR-375 expression ratio has been shown to differentiate between normal tissue and HNC, with 92% sensitivity and 93% specificity, and could therefore be reflective of disease status [62]. In addition, the expression ratio of miR-196a to miR-204 appears to be a very strong predictor of disease recurrence and survival outcome in patients with OSCC, exhibiting 91% specificity and 83% sensitivity in distinguishing aggressive from non-aggressive tumors [57]. In patients with LSCC, the expression ratio of miR-21 and miR-375 has demonstrated 94% sensitivity and specificity in disease detection [55], whereas a high miR-21/miR-375 expression ratio has also been proposed as a prognostic biomarker of a higher tumor stage and worse prognostic outcomes [426]. Similarly, combined differential expression of miR-21 and miR-375 has been demonstrated to accurately predict TSCC both in oral cytology (100% sensitivity and 64% specificity) and in tissue samples (83% sensitivity and 83% specificity), with the former representing a potential non-invasive tool for early diagnosis and possibly for TSCC screening in the future [49]. Additionally, the combined differential expression of miR-6510-3p and miR-34c-5p appears quite promising in distinguishing between healthy tissue and OSCC with 94.6% specificity and 91.9% sensitivity, while the combined expression of miR-449a-5p, miR-6510-3p, and miR-133a-5p seems to differentiate between healthy tissue and LSCC samples with 87.9% specificity and 90.9% sensitivity [7].

3.2. Multiple miRNA Signatures

3.2.1. HNSCC

A 3-miRNA serum signature, consisting of miR-383, miR-615, and miR-877, has shown excellent diagnostic potential in differentiating between patients with HSNCC and healthy controls, with 89.3% sensitivity and 98.9% specificity [277]. Another non-invasive saliva-based panel of three miRNAs, consisting of miR-9, miR-134 and miR-191, has been proposed as a reliable marker for the diagnosis of HNSCC [191], while an 8-miRNA circulating plasma signature, consisting of miR-21-5p, miR-28-3p, miR-142-3p, miR-191-5p, miR-186-5p, miR-197-3p, miR-425-5p, and miR-590-5p, has also demonstrated good performance in distinguishing between HNSCC patients and healthy controls [331].
An 11-miRNA signature (Table 4 and Figure 3a) [24] and a 6-miRNA signature (Table 4 and Figure 3a), and their respective nomogram-based models, have shown great potential in accurately predicting 3- and 5-year survival outcomes in HNSCC patients [427]. Similarly, a 6-miRNA signature (let-7c, miR-125b-2, miR-129-1, miR-337, miR-654, and miR-99a) has been identified as an independent predictor of HNSCC patient survival, as demonstrated in a large The Cancer Genome Atlas dataset (TCGA) [428]. Another signature, composed of four known upregulated miRNAs (miR-21-3p, miR-21-5p, miR-96-5p, and miR-429), when detected in the peritumoral tissue of HNSCC patients, has been associated with shorter local recurrence-free survival and could therefore represent an independent predictive marker of patients at high risk for disease recurrence [429]. On the other hand, a 5-miRNA signature (Table 4 and Figure 3a) has been identified as an independent predictor of disease recurrence (post-radio/chemotherapy) and survival in HPV-negative HNSCC patients [430]. When combined with clinical parameters (TNM stages and extracapsular extension), this same signature appears useful in further categorizing HNSCC patients into distinct risk groups for recurrence, thereby presenting a promising marker for patient stratification in personalized treatment [430]. The combined upregulation of a different 5-miRNA group in the plasma of HNSCC patients, namely miR-142-3p, miR-186-5p, miR-195-5p, miR-374b-5p, and miR-574-3p, has demonstrated significant correlation with poor prognostic outcomes in patients treated with radiochemotherapy [331], while a prognostic model of seven miRNAs (Table 4 and Figure 3a) has shown good specificity and sensitivity in predicting OS in HNSCC patients and in successfully distinguishing between low- and high-risk patient groups, which could potentially improve patient management in the near future [431]. A number of miRNAs have also been associated with patient response to radiotherapy. Two different 5-miRNA signatures (the first consisting of miR-16, miR-29b, miR-150, miR-1254, and let-7e [432] and the second of miR-99a, miR-31, miR-410, miR-424, and miR-495 [433]) have been proposed as markers for predicting radiation responsiveness in HNSCC and could potentially aid in the optimization of radiation strategies.

3.2.2. OSCC

A panel of eight frequently deregulated miRNAs in OSCC, including miR-31-5p, miR-21-5p, miR-125b-5p, miR-99a-5p, miR-100-5p, let-7c-5p, miR-24-3p, and miR-30c, has demonstrated high accuracy in detecting the presence of cancer cells in salivary samples, exhibiting high specificity (100%) in identifying OSCC cases (15/15) [434]. The expression signature of miR-21, miR-181b, and miR-345 has been associated with disease progression in OSCC, as it has been shown to successfully differentiate progressive leukoplakia/OSCC from non-progressive leukoplakias/normal tissue [437]. A combined differential expression of miR-218, miR-125b, and let-7g has in turn been proposed as an important prognostic indicator in patients with OSCC, as it has demonstrated significant association with DFS and disease-specific survival (DSS) [435]. Furthermore, a miRNA-based 5-plex marker panel, consisting of miR-127-3p, miR-4736, miR-655-3p, TNM stage, and histologic grading, has been found useful in assessing the prognostic status in early-stage OSCC patients [436].

3.2.3. OPSCC

A combined expression signature of 6 miRNAs (miR-142-3p, miR-31, miR-146a, miR-26b, miR-24 and miR-193b) appears to have prognostic significance in OPSCC [438]. Similarly, an miRNA-seq profiling analysis has identified a 26-miRNA signature (Table 4 and Figure 3a) that proved highly useful in distinguishing between high- and low-risk HPV-positive patients with OPSCC, further suggesting that the particular signature could potentially improve the HPV-positive OPSCC patient selection for personalized therapies [439]. Three additional groups of miRNAs have been significantly associated with different clinicopathological features in OPSCC patients, including OS (miR-107, miR-151, miR-492), DFS (miR-20b, miR-107, miR-151, miR-182, miR-361), and distant metastasis (miR-151, miR-152, miR-324-5p, miR-361, miR-492) [440].

3.2.4. HSCC/NPC

A 3-miRNA signature, consisting of miR-200a-3p, miR-30b-5p, and miR-4451, has been proposed as a prognostic biomarker for post-operative HSCC patients treated with radiotherapy, since individuals with miR-200a-3p, miR-30b-5p, and miR-4451 upregulation have worse OS and DSS compared to those with lower miRNA expression levels [441].
Several signatures have also been identified as candidate biomarkers for NPC. The miR-29c, miR-30e, and miR-93 expression signature has been suggested as a reliable independent prognostic marker for NPC patients [442], while two different 3-miRNA signatures, the first one consisting of ebv-miR-BART19-3p, miR-135b, and miR-141 [443] and the second entailing miR-142-3p, miR-29c, and miR-30e [334], could be potentially used to predict OS in patients with NPC.

4. Dynamic Correlation between miRNAs and HPV Status in HNSCC

Human papillomavirus (HPV16, HPV18) infection is one of the major risk factors for developing OPSCC, along with smoking and alcohol consumption [366,444]. HPV-positive OPSCC, and its subtype, TSCC, are considered a distinct sub-group of HNSCC, presenting entirely different epidemiological, biological, histopathological, clinical, and molecular characteristics from HPV-negative OPSCC [445,446,447]. Although the true extent of HPV implication in HNSCC is not yet clear [448], HPV presence is a favorable prognostic factor for OPSCC patients, as it is associated with better response to radio-chemotherapy and higher OS [366,446]. Despite their differential disease profiles, both HPV-positive and HPV-negative OPSCC patients usually receive the same treatment (chemotherapy or chemoradiotherapy), meaning that most HPV-positive OPSCC patients are likely overtreated, while patients who belong in the poor prognosis group still present low survival outcomes [446,447]. This highlights the urgent need to identify and establish suitable biomarkers for the early detection of HPV-positive OPSCC cases, as well as for patient selection for personalized treatment options.
MiRNAs could potentially fulfill this need, since the presence of HPV in HNSCC has been shown to correlate with specific miRNA expression profiles that differ from those associated with HPV-negative HNSCC [145,446,449,450]; nonetheless, the studies that correlate the HPV status with the expression levels of particular miRNAs are still quite limited and the results are often controversial. A comparative study between HPV-positive and HPV-negative tonsillar tumors has identified 30 miRNAs exclusively expressed in HPV-positive tonsillar tumors, as well as a core of five miRNAs (miR-141-3p, miR-15b-5p, miR-200a-3p, miR-302c-3p, and miR-9-5p), most commonly found in HPV-positive cancers, including TSCC [451]. MiR-1281 and miR-3194-5p have been found significantly upregulated in HPV-positive primary TSCC compared to HPV-negative tumor tissues [366], while high expression levels of miR-155 have been associated with HPV-positive status in both tonsillar and base of tongue squamous cell carcinoma (BOTSCC) tissues [231]. MiR-99a-3p, miR-411-5p, and miR-4746-5p also appear de-regulated in HPV-positive HNSCC tumor samples, with miR-99a-3p and miR-4746-5p displaying significant upregulation and miR-411-5p equivalent downregulation [452]. Pathway enrichment analysis suggested that these particular miRNAs could be associated with HNSCC progression, as well as patient prognosis [452]. A microarray analysis has identified a total of 17 differentially expressed miRNAs between HPV-positive and HPV-negative TSCC tissues; however, when all tumor sub-sites were included together in one group, only five of those miRNAs (miR-16-3p, miR-29a, miR-29c, miR-150, and miR-363) showed statistically significant differential expression between HPV-positive and HPV-negative HNSCC tumors [245]. Another combination of five miRNAs (miR-16-3p, miR-20b, miR-142-3p, miR-150, and miR-363) accordingly displayed statistical significance between HPV-positive and HPV-negative tumors of exclusively oropharyngeal origin [245]. An additional combination of miRNAs, including miR-363, miR-33, miR-155, miR-181a, miR-181b, miR-29a, miR-218, miR-222, miR-221, and miR-142-5p, has been reported to be differentially expressed between HPV-positive and HPV-negative HNSCC cell lines [449]. Another study has identified a total of 36 differentially expressed miRNAs between HPV-positive and HPV-negative tonsillar squamous cell carcinoma samples, along with a different 14-miRNA core (miR-10b, miR-15a, miR-16, miR-20b, miR-139-3p, miR-139-5p, miR-145, miR-199a-3p, miR-199a-5p, miR-199b-5p, miR-328, miR-379, miR-381, and miR-574-3p) of HPV-positive HNSCC and cervical squamous cell carcinoma (CSCC) [448]. Among these miRNAs, the miR-15a/miR-16/miR-195/miR-497 family, the miR-106-363 cluster, and miR-143/miR-145 seem to have specific target genes associated with HPV pathogenesis, but most of them have never been analyzed in the context of HNSCC, since HPV status is rarely considered; thus, more functional in vitro research is warranted in the future to validate these findings [448].
Of all the miRNAs identified in the aforementioned studies, miR-9 appears to be one of the most frequently associated with HPV-positive head and neck cancer in the literature and the one most likely to be HPV-specific, as it has been reported to be highly expressed in HPV-positive more often than in HPV-negative HNSCC [301,440,453,454]; on the contrary, the majority of miRNAs are found de-regulated regardless of HPV status. Furthermore, it has been demonstrated that HPV is capable of inducing the secretion of miR-9-rich exosomes in HNSCC cells, while exosomal miR-9 derived from HPV-positive HNSCC cells could significantly enhance cell radiosensitivity, possibly through switching macrophages towards the M1 phenotype [455]. Similarly, exosomal miR-9 derived from HPV-positive HNSCC cell lines has been shown to inhibit TGF-β1 signaling-mediated transformation in fibroblasts, which is also associated with a better patient prognosis [453]. While these findings might offer a possible mechanistic explanation as to why HPV-positive HNSCC are generally more sensitive to therapy than their HPV-negative counterparts, they are also indicative of miR-9′s potential as a rather promising therapeutic target for HNSCC patients.
In regards to predictive miRNA biomarkers for HPV-positive HNSCC, a number of studies have specifically focused on identifying miRNA signatures for patient diagnosis, prognosis, and stratification, for the implementation of more efficient treatment approaches. A 5-miRNA salivary panel, consisting of miR-9, miR-134, miR-196b, miR-210, and miR-455, has demonstrated 65% sensitivity and 95% specificity in discriminating between HPV-positive HNSCC patients and healthy controls, while the combination of miR-191, miR-196b, miR-210, and miR-222 has been found capable of distinguishing between HPV-positive and HPV-negative HNSCC patients [190]. A recently developed distinct 6-miRNA signature (Table 5 and Figure 3b) appears to predict the prognosis of individual HPV-positive patients, based on a risk score formula that takes into account the expression and coefficient of miRNAs in the signature [446]. A comparative study has identified an miRNA signature that is differentially expressed and can discriminate between HPV-positive and HPV-negative OPSCCs, while further in silico functional analysis has suggested that a number of genes associated with the aforementioned miRNAs could be potentially targeted for future therapeutic purposes [456]. Furthermore, a set of 14 extracellular vesicle (EV)-derived miRNAs (Table 5 and Figure 3b) has been associated with the presence of HPV in OPSCC cell lines, suggesting that the particular miRNAs could be indicative of HPV status in OPSCC and might potentially present non-invasive blood or salivary future biomarkers for early patient diagnosis and appropriate stratification [457]. A small EV-derived 11-miR ratio signature (Table 5 and Figure 3b), developed through a novel method named stable variable selection (StaVarSel), has demonstrated 90% sensitivity and 79% specificity, when using a high-accuracy model, and 97% specificity and 54% sensitivity, when using a high-specificity model, in detecting HPV-positive OPSCCs. Since the aforementioned panel constitutes a blood-based (serum) marker, it could potentially be utilized for the diagnosis of HPV-positive OPSCCs, as well as for post-therapy surveillance of patients or the earlier detection of possible disease recurrence [458].

5. Clinical Trials Assessing the Utility of miRNAs as Biomarkers for HNC Monitoring

The detection, prediction, and prognosis of HNC is particularly challenging, mainly due to cancer heterogeneity and a significant lack of confirmed molecular biomarkers with high prognostic or diagnostic clinical significance, which could improve patient quality of life and therapeutic approaches. Recent research efforts have focused on the identification of miRNAs that will serve as new biomarkers of prognosis and disease surveillance, and as new therapeutic targets for HNC.
Although miRNAs are the main subject of several in vitro and in vivo studies, only seven clinical trials are currently ongoing with the aim of identifying diagnostic, prognostic, or therapeutic utility of miRNAs in HNC (Table 6). In these trials, miRNA identification and analysis are either the main study subject or among the secondary study goals. Two of these studies (NCT01927354 and NCT02009852) aim to unravel the significance of miR-29b and other members of the miR-29 family in HNSCC prognosis and pathogenesis. In the third miRNA-based clinical trial (NCT04305366), the investigators plan to explore miRNA signatures of several samples (tissue, blood, and saliva) from both HNSCC patients and healthy individuals and to correlate those with diagnosis and disease progression. Finally, in the last miRNA-targeted clinical study (NCT03953443), the primary objective is the delineation of the association between miRNA expression and miRNA promoter methylation, as well as their prognostic and predictive role in primary HPV-negative HNSCC patients. In all of the other ongoing clinical trials (NCT02869399, NCT04453046, and NCT03843515), miRNA identification and evaluation are mainly intended to the adjuvant evaluation of therapeutic efficacy.

6. Conclusions

HNSCC is one of the most frequent malignancies of the upper aerodigestive tract [1]. Despite the new targeted therapies against HNSCC, survival rates have not improved significantly over the last few decades [1]. In this context, it is crucial to develop novel, sensitive, and precise diagnostic and prognostic tools. More than a decade of research has indicated that the aberrant expression of miRNAs deranges the well-regulated RNA complexes in the majority of cancer types. Individual miRNAs or signatures have been considered as suitable biomarkers for clinical applications in cancer prognosis and diagnosis, based on the fact that they exhibit unique and stable expression patterns in cancer tissues [10].
The present review aimed to provide a detailed overview of the current understanding of miRNAs, by summarizing some of the most extensively studied miRNAs that are aberrantly expressed in HNSCC and highlighting their utility as biomarkers. By taking into account that the role of miRNAs is multifunctional, and, as such, that one miRNA can regulate more than one gene targets [15], the scope of this review was to highlight the miRNAs that have so far been associated with evidence of clinical significance. To this purpose, we present the clinical trials that are currently investigating miRNA expression in HNSCC patients and discuss the utility of specific miRNAs as biomarkers for patient stratification in HNSCC. Some of these miRNAs have the potential to serve as reliable biomarkers of diagnosis, prognosis, and therapeutic prediction, in the frame of personalized medicine.
Among the 16 miRNAs that were analyzed in the present review, miR-21 appears to be the most extensively studied and the most consistently upregulated in HNSCC. As it has been associated with poor prognostic outcomes, it could serve as a reliable prognostic biomarker in future clinical applications [55,56]. miR-196a/b and miR-23a, also found upregulated in HNSCC, have been linked to more aggressive cancer types and may therefore confer additional prognostic value [57,261]. Other clinically significant biomarkers seem to include miR-375 and miR-200, as their downregulation is strongly associated with metastasis and worse survival outcomes in HNSCC patients [55,126,132]. miR-29, a consistently downregulated miRNA in HNSCC, is closely correlated with cancer growth and migration [291], and is currently the subject of intense investigation for its potential prognostic significance. Notably, the expression of certain miRNAs (miR-99, -34a, -31, -125a/b, -9, -181a, -155, -146a, -16, and let-7) seems to depend on the HNC subtype and/or the sample type which the miRNAs are extracted from; as such, there is no consistency in the pattern of expression (i.e., exclusively upregulated or downregulated). In such cases, the simultaneous testing of several miRNAs in clinical trials could possibly help to further evaluate and strengthen biomarker significance. MiRNA signatures could provide a more precise approach to the management of HNSCC, leading to the significantly better monitoring of patients.
Additional studies investigating the role of specific miRNAs, individually or as signatures, will undoubtedly shed more light on the utility of miRNAs as biomarkers, leading to improved patient stratification in HNC.

Author Contributions

Conceptualization, V.Z. and S.P.F.; writing—original draft preparation, A.C.T. and P.B.; review and editing, M.A., M.G., C.N.B. and S.P.F.; visualization, A.C.T., P.B. and S.P.F.; supervision, C.N.B., V.Z. and S.P.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research has been co-financed by the European Regional Development Fund of the European Union and Greek National Funds through the Operational Program Competitiveness, Entrepreneurship, and Innovation, under the call RESEARCH–CREATE–INNOVATE (project code: Τ2EDK-03266, project acronym and title: “BIOKAKETRA—Identification of genomic and transcriptomic prognostic bio-signatures in head and neck cancer”).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. 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]
  2. Peltanova, B.; Raudenska, M.; Masarik, M. Effect of tumor microenvironment on pathogenesis of the head and neck squamous cell carcinoma: A systematic review. Mol. Cancer 2019, 18, 63. [Google Scholar] [CrossRef]
  3. Chow, L.Q.M. Head and Neck Cancer. N. Engl. J. Med. 2020, 382, 60–72. [Google Scholar] [CrossRef]
  4. Falzone, L.; Salomone, S.; Libra, M. Evolution of Cancer Pharmacological Treatments at the Turn of the Third Millennium. Front. Pharmacol. 2018, 9, 1300. [Google Scholar] [CrossRef] [Green Version]
  5. Takes, R.P.; Rinaldo, A.; Silver, C.E.; Haigentz, M., Jr.; Woolgar, J.A.; Triantafyllou, A.; Mondin, V.; Paccagnella, D.; de Bree, R.; Shaha, A.R.; et al. Distant metastases from head and neck squamous cell carcinoma. Part I. Basic aspects. Oral Oncol. 2012, 48, 775–779. [Google Scholar] [CrossRef]
  6. Kulasinghe, A.; Schmidt, H.; Perry, C.; Whitfield, B.; Kenny, L.; Nelson, C.; Warkiani, M.E.; Punyadeera, C. A Collective Route to Head and Neck Cancer Metastasis. Sci. Rep. 2018, 8, 746. [Google Scholar] [CrossRef] [Green Version]
  7. Piotrowski, I.; Zhu, X.; Saccon, T.D.; Ashiqueali, S.; Schneider, A.; de Carvalho Nunes, A.D.; Noureddine, S.; Sobecka, A.; Barczak, W.; Szewczyk, M.; et al. miRNAs as Biomarkers for Diagnosing and Predicting Survival of Head and Neck Squamous Cell Carcinoma Patients. Cancers 2021, 13, 3980. [Google Scholar] [CrossRef] [PubMed]
  8. Leemans, C.R.; Snijders, P.J.F.; Brakenhoff, R.H. The molecular landscape of head and neck cancer. Nat. Rev. Cancer 2018, 18, 269–282. [Google Scholar] [CrossRef] [PubMed]
  9. Kumarasamy, C.; Madhav, M.R.; Sabarimurugan, S.; Krishnan, S.; Baxi, S.; Gupta, A.; Gothandam, K.M.; Jayaraj, R. Prognostic Value of miRNAs in Head and Neck Cancers: A Comprehensive Systematic and Meta-Analysis. Cells 2019, 8, 772. [Google Scholar] [CrossRef] [Green Version]
  10. Schoof, C.R.; Botelho, E.L.; Izzotti, A.; Vasques Ldos, R. MicroRNAs in cancer treatment and prognosis. Am. J. Cancer Res. 2012, 2, 414–433. [Google Scholar] [PubMed]
  11. Kabzinski, J.; Maczynska, M.; Majsterek, I. MicroRNA as a Novel Biomarker in the Diagnosis of Head and Neck Cancer. Biomolecules 2021, 11, 844. [Google Scholar] [CrossRef]
  12. Kabekkodu, S.P.; Shukla, V.; Varghese, V.K.; D’Souza, J.; Chakrabarty, S.; Satyamoorthy, K. Clustered miRNAs and their role in biological functions and diseases. Biol. Rev. Camb. Philos. Soc. 2018, 93, 1955–1986. [Google Scholar] [CrossRef] [PubMed]
  13. Xie, Y.; Ma, X.; Chen, L.; Li, H.; Gu, L.; Gao, Y.; Zhang, Y.; Li, X.; Fan, Y.; Chen, J.; et al. MicroRNAs with prognostic significance in bladder cancer: A systematic review and meta-analysis. Sci. Rep. 2017, 7, 5619. [Google Scholar] [CrossRef]
  14. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Macfarlane, L.A.; Murphy, P.R. MicroRNA: Biogenesis, Function and Role in Cancer. Curr. Genom. 2010, 11, 537–561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Gross, N.; Kropp, J.; Khatib, H. MicroRNA Signaling in Embryo Development. Biology 2017, 6, 34. [Google Scholar] [CrossRef]
  17. Lai, Y.H.; Liu, H.; Chiang, W.F.; Chen, T.W.; Chu, L.J.; Yu, J.S.; Chen, S.J.; Chen, H.C.; Tan, B.C. MiR-31-5p-ACOX1 Axis Enhances Tumorigenic Fitness in Oral Squamous Cell Carcinoma Via the Promigratory Prostaglandin E2. Theranostics 2018, 8, 486–504. [Google Scholar] [CrossRef]
  18. Zoumpourlis, V.; Skourti, E.; Goulielmaki, M.; Vlahopoulos, S.; Christodoulou, I. The Ideological Frame of the Genetic Basis of Cancer: The Important Role of miRNAs. Crit. Rev. Oncog. 2017, 22, 303–311. [Google Scholar] [CrossRef]
  19. Sabit, H.; Cevik, E.; Tombuloglu, H.; Abdel-Ghany, S.; Tombuloglu, G.; Esteller, M. Triple negative breast cancer in the era of miRNA. Crit. Rev. Oncol. Hematol. 2021, 157, 103196. [Google Scholar] [CrossRef]
  20. Iqbal, M.A.; Arora, S.; Prakasam, G.; Calin, G.A.; Syed, M.A. MicroRNA in lung cancer: Role, mechanisms, pathways and therapeutic relevance. Mol. Asp. Med. 2019, 70, 3–20. [Google Scholar] [CrossRef]
  21. Abramovic, I.; Ulamec, M.; Katusic Bojanac, A.; Bulic-Jakus, F.; Jezek, D.; Sincic, N. miRNA in prostate cancer: Challenges toward translation. Epigenomics 2020, 12, 543–558. [Google Scholar] [CrossRef]
  22. Huang, J.; Yu, S.; Ding, L.; Ma, L.; Chen, H.; Zhou, H.; Zou, Y.; Yu, M.; Lin, J.; Cui, Q. The Dual Role of Circular RNAs as miRNA Sponges in Breast Cancer and Colon Cancer. Biomedicines 2021, 9, 1590. [Google Scholar] [CrossRef]
  23. Deb, B.; Uddin, A.; Chakraborty, S. miRNAs and ovarian cancer: An overview. J. Cell. Physiol. 2018, 233, 3846–3854. [Google Scholar] [CrossRef] [PubMed]
  24. Huang, Y.; Liu, Z.; Zhong, L.; Wen, Y.; Ye, Q.; Cao, D.; Li, P.; Liu, Y. Construction of an 11-microRNA-based signature and a prognostic nomogram to predict the overall survival of head and neck squamous cell carcinoma patients. BMC Genom. 2020, 21, 691. [Google Scholar] [CrossRef] [PubMed]
  25. Zhao, Y.; Xu, L.; Wang, X.; Niu, S.; Chen, H.; Li, C. A novel prognostic mRNA/miRNA signature for esophageal cancer and its immune landscape in cancer progression. Mol. Oncol. 2021, 15, 1088–1109. [Google Scholar] [CrossRef] [PubMed]
  26. Fitriana, M.; Hwang, W.L.; Chan, P.Y.; Hsueh, T.Y.; Liao, T.T. Roles of microRNAs in Regulating Cancer Stemness in Head and Neck Cancers. Cancers 2021, 13, 1742. [Google Scholar] [CrossRef]
  27. Cao, J.; Zhang, Y.; Mu, J.; Yang, D.; Gu, X.; Zhang, J. Exosomal miR-21-5p contributes to ovarian cancer progression by regulating CDK6. Hum. Cell 2021, 34, 1185–1196. [Google Scholar] [CrossRef] [PubMed]
  28. Dai, L.; Chen, F.; Zheng, Y.; Zhang, D.; Qian, B.; Ji, H.; Long, F.; Cretoiu, D. miR-21 regulates growth and EMT in lung cancer cells via PTEN/Akt/GSK3β signaling. Front. Biosci. Landmark 2019, 24, 1426–1439. [Google Scholar] [CrossRef] [Green Version]
  29. Larki, P.; Ahadi, A.; Zare, A.; Tarighi, S.; Zaheri, M.; Souri, M.; Zali, M.R.; Ghaedi, H.; Omrani, M.D. Up-Regulation of miR-21, miR-25, miR-93, and miR-106b in Gastric Cancer. Iran. Biomed. J. 2018, 22, 367–373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Wang, H.; Tan, Z.; Hu, H.; Liu, H.; Wu, T.; Zheng, C.; Wang, X.; Luo, Z.; Wang, J.; Liu, S.; et al. microRNA-21 promotes breast cancer proliferation and metastasis by targeting LZTFL1. BMC Cancer 2019, 19, 738. [Google Scholar] [CrossRef] [Green Version]
  31. Jin, X.H.; Lu, S.; Wang, A.F. Expression and clinical significance of miR-4516 and miR-21-5p in serum of patients with colorectal cancer. BMC Cancer 2020, 20, 241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Drillis, G.; Goulielmaki, M.; Spandidos, D.A.; Aggelaki, S.; Zoumpourlis, V. Non-coding RNAs (miRNAs and lncRNAs) and their roles in lymphogenesis in all types of lymphomas and lymphoid malignancies. Oncol. Lett. 2021, 21, 393. [Google Scholar] [CrossRef]
  33. ParvizHamidi, M.; Haddad, G.; Ostadrahimi, S.; Ostadrahimi, N.; Sadeghi, S.; Fayaz, S.; Fard-Esfahani, P. Circulating miR-26a and miR-21 as biomarkers for glioblastoma multiform. Biotechnol. Appl. Biochem. 2019, 66, 261–265. [Google Scholar] [CrossRef]
  34. Yu, X.; Wu, Y.; Liu, Y.; Deng, H.; Shen, Z.; Xiao, B.; Guo, J. miR-21, miR-106b and miR-375 as novel potential biomarkers for laryngeal squamous cell carcinoma. Curr. Pharm. Biotechnol. 2014, 15, 503–508. [Google Scholar] [CrossRef] [PubMed]
  35. Wei, L.; Mao, M.; Liu, H. Droplet digital PCR and qRT-PCR to detect circulating miR-21 in laryngeal squamous cell carcinoma and pre-malignant laryngeal lesions. Acta Oto-Laryngol. 2016, 136, 923–932. [Google Scholar] [CrossRef]
  36. Hou, B.; Ishinaga, H.; Midorikawa, K.; Shah, S.A.; Nakamura, S.; Hiraku, Y.; Oikawa, S.; Murata, M.; Takeuchi, K. Circulating microRNAs as novel prognosis biomarkers for head and neck squamous cell carcinoma. Cancer Biol. Ther. 2015, 16, 1042–1046. [Google Scholar] [CrossRef] [PubMed]
  37. Jadhav, K.B.; Shah, V.; Chauhan, N.; Shah, N.; Parmar, G. Expression of microRNA-21 in saliva and tumor tissue of patients with oral squamous cell carcinoma: A predictor of cervical lymph node metastasis. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2022, 133, 60–69. [Google Scholar] [CrossRef]
  38. 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]
  39. Zou, X.; Xia, T.; Li, M.; Wang, T.; Liu, P.; Zhou, X.; Huang, Z.; Zhu, W. MicroRNA profiling in serum: Potential signatures for breast cancer diagnosis. Cancer Biomark. 2021, 30, 41–53. [Google Scholar] [CrossRef]
  40. Ishinaga, H.; He, F.; Hou, B.; Shah, S.; Murata, M.; Takeuchi, K. A longitudinal study on circulating miR-21 as a therapeutic effect marker in head and neck squamous cell carcinoma. Carcinogenesis 2019, 40, 1070–1076. [Google Scholar] [CrossRef]
  41. Hsu, C.M.; Lin, P.M.; Wang, Y.M.; Chen, Z.J.; Lin, S.F.; Yang, M.Y. Circulating miRNA is a novel marker for head and neck squamous cell carcinoma. Tumour Biol. 2012, 33, 1933–1942. [Google Scholar] [CrossRef] [PubMed]
  42. Yan, Y.; Wang, X.; Venø, M.T.; Bakholdt, V.; Sørensen, J.A.; Krogdahl, A.; Sun, Z.; Gao, S.; Kjems, J. Circulating miRNAs as biomarkers for oral squamous cell carcinoma recurrence in operated patients. Oncotarget 2017, 8, 8206–8214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Liu, X.; Luo, H.N.; Tian, W.D.; Lu, J.; Li, G.; Wang, L.; Zhang, B.; Liang, B.J.; Peng, X.H.; Lin, S.X.; et al. Diagnostic and prognostic value of plasma microRNA deregulation in nasopharyngeal carcinoma. Cancer Biol. Ther. 2013, 14, 1133–1142. [Google Scholar] [CrossRef]
  44. Ren, W.; Qiang, C.; Gao, L.; Li, S.M.; Zhang, L.M.; Wang, X.L.; Dong, J.W.; Chen, C.; Liu, C.Y.; Zhi, K.Q. Circulating microRNA-21 (MIR-21) and phosphatase and tensin homolog (PTEN) are promising novel biomarkers for detection of oral squamous cell carcinoma. Biomarkers 2014, 19, 590–596. [Google Scholar] [CrossRef] [PubMed]
  45. Darido, C.; Georgy, S.R.; Wilanowski, T.; Dworkin, S.; Auden, A.; Zhao, Q.; Rank, G.; Srivastava, S.; Finlay, M.J.; Papenfuss, A.T.; et al. Targeting of the tumor suppressor GRHL3 by a miR-21-dependent proto-oncogenic network results in PTEN loss and tumorigenesis. Cancer Cell 2011, 20, 635–648. [Google Scholar] [CrossRef] [Green Version]
  46. Yan, Z.Y.; Luo, Z.Q.; Zhang, L.J.; Li, J.; Liu, J.Q. Integrated Analysis and MicroRNA Expression Profiling Identified Seven miRNAs Associated With Progression of Oral Squamous Cell Carcinoma. J. Cell. Physiol. 2017, 232, 2178–2185. [Google Scholar] [CrossRef]
  47. Odar, K.; Boštjančič, E.; Gale, N.; Glavač, D.; Zidar, N. Differential expression of microRNAs miR-21, miR-31, miR-203, miR-125a-5p and miR-125b and proteins PTEN and p63 in verrucous carcinoma of the head and neck. Histopathology 2012, 61, 257–265. [Google Scholar] [CrossRef]
  48. Sun, Z.; Li, S.; Kaufmann, A.M.; Albers, A.E. miR-21 increases the programmed cell death 4 gene-regulated cell proliferation in head and neck squamous carcinoma cell lines. Oncol. Rep. 2014, 32, 2283–2289. [Google Scholar] [CrossRef]
  49. He, Q.; Chen, Z.; Cabay, R.J.; Zhang, L.; Luan, X.; Chen, D.; Yu, T.; Wang, A.; Zhou, X. microRNA-21 and microRNA-375 from oral cytology as biomarkers for oral tongue cancer detection. Oral Oncol. 2016, 57, 15–20. [Google Scholar] [CrossRef] [Green Version]
  50. Ajuyah, P.; Hill, M. MicroRNA (miRNA)-to-miRNA Regulation of Programmed Cell Death 4 (PDCD4). Mol. Cell. Biol. 2019, 39, e00086-19. [Google Scholar] [CrossRef] [Green Version]
  51. Ren, J.; Zhu, D.; Liu, M.; Sun, Y.; Tian, L. Downregulation of miR-21 modulates Ras expression to promote apoptosis and suppress invasion of Laryngeal squamous cell carcinoma. Eur. J. Cancer 2010, 46, 3409–3416. [Google Scholar] [CrossRef] [PubMed]
  52. Yu, E.H.; Tu, H.F.; Wu, C.H.; Yang, C.C.; Chang, K.W. MicroRNA-21 promotes perineural invasion and impacts survival in patients with oral carcinoma. J. Chin. Med. Assoc. JCMA 2017, 80, 383–388. [Google Scholar] [CrossRef]
  53. Wan, Y.; Hoyle, R.G.; Xie, N.; Wang, W.; Cai, H.; Zhang, M.; Ma, Z.; Xiong, G.; Xu, X.; Huang, Z.; et al. A Super-Enhancer Driven by FOSL1 Controls miR-21-5p Expression in Head and Neck Squamous Cell Carcinoma. Front. Oncol. 2021, 11, 656628. [Google Scholar] [CrossRef]
  54. Menderico Junior, G.M.; Theodoro, T.R.; Pasini, F.S.; de Menezes Ishikawa, M.; Santos, N.S.S.; de Mello, E.S.; da Silva Pinhal, M.A.; Moyses, R.A.; Kulcsar, M.A.V. MicroRNA-mediated extracellular matrix remodeling in squamous cell carcinoma of the oral cavity. Head Neck 2021, 43, 2364–2376. [Google Scholar] [CrossRef]
  55. Hu, A.; Huang, J.J.; Xu, W.H.; Jin, X.J.; Li, J.P.; Tang, Y.J.; Huang, X.F.; Cui, H.J.; Sun, G.B. miR-21 and miR-375 microRNAs as candidate diagnostic biomarkers in squamous cell carcinoma of the larynx: Association with patient survival. Am. J. Transl. Res. 2014, 6, 604–613. [Google Scholar] [PubMed]
  56. Hedbäck, N.; Jensen, D.H.; Specht, L.; Fiehn, A.M.; Therkildsen, M.H.; Friis-Hansen, L.; Dabelsteen, E.; von Buchwald, C. MiR-21 expression in the tumor stroma of oral squamous cell carcinoma: An independent biomarker of disease free survival. PLoS ONE 2014, 9, e95193. [Google Scholar] [CrossRef] [PubMed]
  57. Rajan, C.; Roshan, V.G.D.; Khan, I.; Manasa, V.G.; Himal, I.; Kattoor, J.; Thomas, S.; Kondaiah, P.; Kannan, S. MiRNA expression profiling and emergence of new prognostic signature for oral squamous cell carcinoma. Sci. Rep. 2021, 11, 7298. [Google Scholar] [CrossRef] [PubMed]
  58. Li, J.; Huang, H.; Sun, L.; Yang, M.; Pan, C.; Chen, W.; Wu, D.; Lin, Z.; Zeng, C.; Yao, Y.; et al. MiR-21 indicates poor prognosis in tongue squamous cell carcinomas as an apoptosis inhibitor. Clin. Cancer Res. 2009, 15, 3998–4008. [Google Scholar] [CrossRef] [Green Version]
  59. Ren, W.; Wang, X.; Gao, L.; Li, S.; Yan, X.; Zhang, J.; Huang, C.; Zhang, Y.; Zhi, K. MiR-21 modulates chemosensitivity of tongue squamous cell carcinoma cells to cisplatin by targeting PDCD4. Mol. Cell. Biochem. 2014, 390, 253–262. [Google Scholar] [CrossRef] [PubMed]
  60. Jia-yuan, X.; Wei, S.; Fang-fang, L.; Zhi-jian, D.; Long-he, C.; Sen, L. miR-375 Inhibits the Proliferation and Invasion of Nasopharyngeal Carcinoma Cells by Suppressing PDK1. BioMed Res. Int. 2020, 2020, 9704245. [Google Scholar] [CrossRef]
  61. Wu, W.; Zhang, Y.; Li, X.; Wang, X.; Yuan, Y. miR-375 Inhibits the Proliferation, Migration and Invasion of Esophageal Squamous Cell Carcinoma by Targeting XPR1. Curr. Gene Ther. 2021, 21, 290–298. [Google Scholar] [CrossRef]
  62. Avissar, M.; Christensen, B.C.; Kelsey, K.T.; Marsit, C.J. MicroRNA expression ratio is predictive of head and neck squamous cell carcinoma. Curr. Gene Ther. 2009, 15, 2850–2855. [Google Scholar] [CrossRef] [Green Version]
  63. Wiklund, E.D.; Gao, S.; Hulf, T.; Sibbritt, T.; Nair, S.; Costea, D.E.; Villadsen, S.B.; Bakholdt, V.; Bramsen, J.B.; Sørensen, J.A.; et al. MicroRNA alterations and associated aberrant DNA methylation patterns across multiple sample types in oral squamous cell carcinoma. PLoS ONE 2011, 6, e27840. [Google Scholar] [CrossRef]
  64. Avilés-Jurado, F.X.; Muñoz, C.; Meler, C.; Flores, J.C.; Gumà, J.; Benaiges, E.; Mora, J.; Camacho, M.; León, X.; Vilaseca, I.; et al. Circulating microRNAs modulating glycolysis as non-invasive prognostic biomarkers of HNSCC. Eur. Arch. Otorhinolaryngol. 2021, 278, 1585–1594. [Google Scholar] [CrossRef] [PubMed]
  65. Chang, K.; Wei, Z.; Cao, H. miR-375-3p inhibits the progression of laryngeal squamous cell carcinoma by targeting hepatocyte nuclear factor-1β. Oncol. Lett. 2020, 20, 80. [Google Scholar] [CrossRef] [PubMed]
  66. Xu, J.; Li, B.; Song, W.; Cao, L.; Zhu, C.; Lin, S. Tumor suppressor functions of miRNA-375 in nasopharyngeal carcinoma through inhibition of ubiquitin-specific protease 1 expression. Int. J. Biochem. Cell Biol. 2021, 141, 106092. [Google Scholar] [CrossRef]
  67. Hudcova, K.; Raudenska, M.; Gumulec, J.; Binkova, H.; Horakova, Z.; Kostrica, R.; Babula, P.; Adam, V.; Masarik, M. Expression profiles of miR-29c, miR-200b and miR-375 in tumour and tumour-adjacent tissues of head and neck cancers. Tumour Biol. 2016, 37, 12627–12633. [Google Scholar] [CrossRef]
  68. Wang, P.; Xu, L.; Li, L.; Ren, S.; Tang, J.; Zhang, M.; Xu, M. The microRNA-375 as a potentially promising biomarker to predict the prognosis of patients with head and neck or esophageal squamous cell carcinoma: A meta-analysis. Eur. Arch. Oto-Rhino-Laryngol. 2019, 276, 957–968. [Google Scholar] [CrossRef]
  69. Jung, H.M.; Benarroch, Y.; Chan, E.K. Anti-cancer drugs reactivate tumor suppressor miR-375 expression in tongue cancer cells. J. Cell. Biochem. 2015, 116, 836–843. [Google Scholar] [CrossRef]
  70. Xin, J.X.; Yue, Z.; Zhang, S.; Jiang, Z.H.; Wang, P.Y.; Li, Y.J.; Pang, M.; Xie, S.Y. miR-99 inhibits cervical carcinoma cell proliferation by targeting TRIB2. Oncol. Lett. 2013, 6, 1025–1030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Sun, D.; Lee, Y.S.; Malhotra, A.; Kim, H.K.; Matecic, M.; Evans, C.; Jensen, R.V.; Moskaluk, C.A.; Dutta, A. miR-99 family of MicroRNAs suppresses the expression of prostate-specific antigen and prostate cancer cell proliferation. Cancer Res. 2011, 71, 1313–1324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Zhang, M.; Guo, Y.; Wu, J.; Chen, F.; Dai, Z.; Fan, S.; Li, P.; Song, T. Roles of microRNA-99 family in human glioma. OncoTargets Ther. 2016, 9, 3613–3619. [Google Scholar] [CrossRef] [Green Version]
  73. Sun, X.; Yan, H. MicroRNA-99a-5p suppresses cell proliferation, migration, and invasion by targeting isoprenylcysteine carboxylmethyltransferase in oral squamous cell carcinoma. J. Int. Med. Res. 2021, 49, 300060520939031. [Google Scholar] [CrossRef] [PubMed]
  74. Wu, S.H.; Han, L.; Lu, B.C.; Wang, H.Y.; Zheng, C.P. MiR-99a inhibits cell proliferation of nasopharyngeal carcinoma by targeting mTOR and serves as a prognostic factor. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 2053–2061. [Google Scholar] [CrossRef]
  75. Moratin, J.; Hartmann, S.; Brands, R.C.; Horn, D.; Fuchs, A.; Mutzbauer, G.; Seher, A.; Scholz, C.; Müller-Richter, U.D.A.; Linz, C. MicroRNA expression correlates with disease recurrence and overall survival in oral squamous cell carcinoma. J. Craniomaxillofac. Surg. 2019, 47, 523–529. [Google Scholar] [CrossRef] [PubMed]
  76. Moratin, J.; Hartmann, S.; Brands, R.; Brisam, M.; Mutzbauer, G.; Scholz, C.; Seher, A.; Müller-Richter, U.; Kübler, A.C.; Linz, C. Evaluation of miRNA-expression and clinical tumour parameters in oral squamous cell carcinoma (OSCC). J. Craniomaxillofac. Surg. 2016, 44, 876–881. [Google Scholar] [CrossRef]
  77. Chen, Z.; Jin, Y.; Yu, D.; Wang, A.; Mahjabeen, I.; Wang, C.; Liu, X.; Zhou, X. Down-regulation of the microRNA-99 family members in head and neck squamous cell carcinoma. Oral Oncol. 2012, 48, 686–691. [Google Scholar] [CrossRef] [Green Version]
  78. Okada, R.; Koshizuka, K.; Yamada, Y.; Moriya, S.; Kikkawa, N.; Kinoshita, T.; Hanazawa, T.; Seki, N. Regulation of Oncogenic Targets by miR-99a-3p (Passenger Strand of miR-99a-Duplex) in Head and Neck Squamous Cell Carcinoma. Cells 2019, 8, 1535. [Google Scholar] [CrossRef] [Green Version]
  79. Jakob, M.; Mattes, L.M.; Küffer, S.; Unger, K.; Hess, J.; Bertlich, M.; Haubner, F.; Ihler, F.; Canis, M.; Weiss, B.G. MicroRNA expression patterns in oral squamous cell carcinoma: Hsa-mir-99b-3p and hsa-mir-100-5p as novel prognostic markers for oral cancer. Head Neck 2019, 41, 3499–3515. [Google Scholar] [CrossRef] [PubMed]
  80. Manikandan, M.; Deva Magendhra Rao, A.K.; Arunkumar, G.; Rajkumar, K.S.; Rajaraman, R.; Munirajan, A.K. Down Regulation of miR-34a and miR-143 May Indirectly Inhibit p53 in Oral Squamous Cell Carcinoma: A Pilot Study. Asian Pac. J. Cancer Prev. APJCP 2015, 16, 7619–7625. [Google Scholar] [CrossRef] [Green Version]
  81. Chen, Y.T.; Yao, J.N.; Qin, Y.T.; Hu, K.; Wu, F.; Fang, Y.Y. Biological role and clinical value of miR-99a-5p in head and neck squamous cell carcinoma (HNSCC): A bioinformatics-based study. FEBS Open Bio 2018, 8, 1280–1298. [Google Scholar] [CrossRef]
  82. Wei, G.G.; Guo, W.P.; Tang, Z.Y.; Li, S.H.; Wu, H.Y.; Zhang, L.C. Expression level and prospective mechanism of miRNA-99a-3p in head and neck squamous cell carcinoma based on miRNA-chip and miRNA-sequencing data in 1, 167 cases. Pathol. Res. Pract. 2019, 215, 963–976. [Google Scholar] [CrossRef]
  83. Qi, C.-L.; Sheng, J.-F.; Huang, M.-L.; Zou, Y.; Wang, Y.-P.; Wang, F.; Zeng, F.; Hua, Q.-Q.; Chen, S.-M. Integrated analysis of deregulation microRNA expression in head and neck squamous cell carcinoma. Medicine 2021, 100, e24618. [Google Scholar] [CrossRef]
  84. Chen, L.; Liu, S.; Li, K.; Qi, J.; Liu, C.; Zong, L.; Zhang, Y.; Zhao, J.; Zhai, X.; Li, J.; et al. Evaluation of microRNA expression profiling in highly metastatic laryngocarcinoma cells. Acta Oto-Laryngol. 2018, 138, 1105–1111. [Google Scholar] [CrossRef]
  85. Sousa, L.O.; Sobral, L.M.; Matsumoto, C.S.; Saggioro, F.P.; López, R.V.M.; Panepucci, R.A.; Curti, C.; Silva, W.A., Jr.; Greene, L.J.; Leopoldino, A.M. Lymph node or perineural invasion is associated with low miR-15a, miR-34c and miR-199b levels in head and neck squamous cell carcinoma. BBA Clin. 2016, 6, 159–164. [Google Scholar] [CrossRef] [Green Version]
  86. Figueroa-González, G.; Carrillo-Hernández, J.F.; Perez-Rodriguez, I.; De León, D.C.; Campos-Parra, A.D.; Martínez-Gutiérrez, A.D.; Coronel-Hernández, J.; García-Castillo, V.; López-Camarillo, C.; Peralta-Zaragoza, O.; et al. Negative Regulation of Serine Threonine Kinase 11 (STK11) through miR-100 in Head and Neck Cancer. Genes 2020, 11, 1058. [Google Scholar] [CrossRef]
  87. Chen, L.; Hu, J.; Pan, L.; Yin, X.; Wang, Q.; Chen, H. Diagnostic and prognostic value of serum miR-99a expression in oral squamous cell carcinoma. Cancer Biomark. 2018, 23, 333–339. [Google Scholar] [CrossRef]
  88. Le, F.; Ou, Y.; Luo, P.; Zhong, X. LncRNA NCK1-AS1 in plasma distinguishes oral ulcer from early-stage oral squamous cell carcinoma. J. Biol. Res. 2020, 27, 16. [Google Scholar] [CrossRef]
  89. Wang, X.; Yin, Z.; Zhao, Y.; He, M.; Dong, C.; Zhong, M. Identifying potential prognostic biomarkers in head and neck cancer based on the analysis of microRNA expression profiles in TCGA database. Mol. Med. Rep. 2020, 21, 1647–1657. [Google Scholar] [CrossRef]
  90. Kumar, B.; Yadav, A.; Lang, J.; Teknos, T.N.; Kumar, P. Dysregulation of microRNA-34a expression in head and neck squamous cell carcinoma promotes tumor growth and tumor angiogenesis. PLoS ONE 2012, 7, e37601. [Google Scholar] [CrossRef]
  91. Shi, X.; Kaller, M.; Rokavec, M.; Kirchner, T.; Horst, D.; Hermeking, H. Characterization of a p53/miR-34a/CSF1R/STAT3 Feedback Loop in Colorectal Cancer. Cell. Mol. Gastroenterol. Hepatol. 2020, 10, 391–418. [Google Scholar] [CrossRef]
  92. Li, Z.; Chen, H. miR-34a inhibits proliferation, migration and invasion of paediatric neuroblastoma cells via targeting HNF4α. Artif. Cells Nanomed. Biotechnol. 2019, 47, 3072–3078. [Google Scholar] [CrossRef]
  93. Dong, B.; Xu, G.C.; Liu, S.T.; Liu, T.; Geng, B. MiR-34a affects G2 arrest in prostate cancer PC3 cells via Wnt pathway and inhibits cell growth and migration. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 8349–8358. [Google Scholar] [CrossRef]
  94. Liu, H.; Deng, H.; Zhao, Y.; Li, C.; Liang, Y. LncRNA XIST/miR-34a axis modulates the cell proliferation and tumor growth of thyroid cancer through MET-PI3K-AKT signaling. J. Exp. Clin. Cancer Res. 2018, 37, 279. [Google Scholar] [CrossRef]
  95. Chang, T.C.; Wentzel, E.A.; Kent, O.A.; Ramachandran, K.; Mullendore, M.; Lee, K.H.; Feldmann, G.; Yamakuchi, M.; Ferlito, M.; Lowenstein, C.J.; et al. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol. Cell 2007, 26, 745–752. [Google Scholar] [CrossRef] [Green Version]
  96. Raver-Shapira, N.; Marciano, E.; Meiri, E.; Spector, Y.; Rosenfeld, N.; Moskovits, N.; Bentwich, Z.; Oren, M. Transcriptional activation of miR-34a contributes to p53-mediated apoptosis. Mol. Cell 2007, 26, 731–743. [Google Scholar] [CrossRef]
  97. He, L.; He, X.; Lim, L.P.; de Stanchina, E.; Xuan, Z.; Liang, Y.; Xue, W.; Zender, L.; Magnus, J.; Ridzon, D.; et al. A microRNA component of the p53 tumour suppressor network. Nature 2007, 447, 1130–1134. [Google Scholar] [CrossRef] [Green Version]
  98. Zhao, Y.; Wang, X. miR-34a targets BCL-2 to suppress the migration and invasion of sinonasal squamous cell carcinoma. Oncol. Lett. 2018, 16, 6566–6572. [Google Scholar] [CrossRef]
  99. Ye, J.; Li, L.; Feng, P.; Wan, J.; Li, J. Downregulation of miR-34a contributes to the proliferation and migration of laryngeal carcinoma cells by targeting cyclin D1. Oncol. Rep. 2016, 36, 390–398. [Google Scholar] [CrossRef] [Green Version]
  100. Tazawa, H.; Tsuchiya, N.; Izumiya, M.; Nakagama, H. Tumor-suppressive miR-34a induces senescence-like growth arrest through modulation of the E2F pathway in human colon cancer cells. Proc. Natl. Acad. Sci. USA 2007, 104, 15472–15477. [Google Scholar] [CrossRef] [Green Version]
  101. Liu, C.; Kelnar, K.; Liu, B.; Chen, X.; Calhoun-Davis, T.; Li, H.; Patrawala, L.; Yan, H.; Jeter, C.; Honorio, S.; et al. The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat. Med. 2011, 17, 211–215. [Google Scholar] [CrossRef] [Green Version]
  102. Li, Z.H.; Weng, X.; Xiong, Q.Y.; Tu, J.H.; Xiao, A.; Qiu, W.; Gong, Y.; Hu, E.W.; Huang, S.; Cao, Y.L. miR-34a expression in human breast cancer is associated with drug resistance. Oncotarget 2017, 8, 106270–106282. [Google Scholar] [CrossRef] [Green Version]
  103. Deng, X.J.; Zheng, H.L.; Ke, X.Q.; Deng, M.; Ma, Z.Z.; Zhu, Y.; Cui, Y.Y. Hsa-miR-34a-5p reverses multidrug resistance in gastric cancer cells by targeting the 3′-UTR of SIRT1 and inhibiting its expression. Cell. Signal 2021, 84, 110016. [Google Scholar] [CrossRef]
  104. Li, J.; Liu, K.; Zhang, T.; Yang, Z.; Wang, R.; Chen, G.; Kang, M. A comprehensive investigation using meta-analysis and bioinformatics on miR-34a-5p expression and its potential role in head and neck squamous cell carcinoma. Am. J. Transl. Res. 2018, 10, 2246–2263. [Google Scholar] [PubMed]
  105. Li, X.; Zhao, S.; Fu, Y.; Zhang, P.; Zhang, Z.; Cheng, J.; Liu, L.; Jiang, H. miR-34a-5p functions as a tumor suppressor in head and neck squamous cell cancer progression by targeting Flotillin-2. Int. J. Biol. Sci. 2021, 17, 4327–4339. [Google Scholar] [CrossRef]
  106. Wang, Y.; Chen, J.; Chen, X.; Jiang, F.; Sun, Y.; Pan, Y.; Zhang, W.; Zhang, J. MiR-34a suppresses HNSCC growth through modulating cell cycle arrest and senescence. Neoplasma 2017, 64, 543–553. [Google Scholar] [CrossRef] [Green Version]
  107. Wu, X.; Cheng, Y.-S.L.; Matthen, M.; Yoon, A.; Schwartz, G.K.; Bala, S.; Taylor, A.M.; Momen-Heravi, F. Down-regulation of the tumor suppressor miR-34a contributes to head and neck cancer by up-regulating the MET oncogene and modulating tumor immune evasion. J. Exp. Clin. Cancer Res. 2021, 40, 70. [Google Scholar] [CrossRef]
  108. Jiang, C.; Cheng, Z.; Jiang, T.; Xu, Y.; Wang, B. MicroRNA-34a inhibits cell invasion and epithelial-mesenchymal transition via targeting AXL/PI3K/AKT/Snail signaling in nasopharyngeal carcinoma. Genes Genom. 2020, 42, 971–978. [Google Scholar] [CrossRef]
  109. Kalfert, D.; Pesta, M.; Kulda, V.; Topolcan, O.; Ryska, A.; Celakovsky, P.; Laco, J.; Ludvikova, M. MicroRNA profile in site-specific head and neck squamous cell cancer. Anticancer Res. 2015, 35, 2455–2463. [Google Scholar] [PubMed]
  110. Wei, L.; Shi, C.; Zhang, Y. Expression of miR-34a and Ki67 in nasopharyngeal carcinoma and the relationship with clinicopathological features and prognosis. Oncol. Lett. 2020, 19, 1273–1280. [Google Scholar] [CrossRef] [Green Version]
  111. Ogawa, T.; Saiki, Y.; Shiga, K.; Chen, N.; Fukushige, S.; Sunamura, M.; Nagase, H.; Hashimoto, S.; Matsuura, K.; Saijo, S.; et al. miR-34a is downregulated in cis-diamminedichloroplatinum treated sinonasal squamous cell carcinoma patients with poor prognosis. Cancer Sci. 2012, 103, 1737–1743. [Google Scholar] [CrossRef] [Green Version]
  112. Childs, G.; Fazzari, M.; Kung, G.; Kawachi, N.; Brandwein-Gensler, M.; McLemore, M.; Chen, Q.; Burk, R.D.; Smith, R.V.; Prystowsky, M.B.; et al. Low-level expression of microRNAs let-7d and miR-205 are prognostic markers of head and neck squamous cell carcinoma. Am. J. Pathol. 2009, 174, 736–745. [Google Scholar] [CrossRef] [Green Version]
  113. Yu, C.C.; Chen, Y.W.; Chiou, G.Y.; Tsai, L.L.; Huang, P.I.; Chang, C.Y.; Tseng, L.M.; Chiou, S.H.; Yen, S.H.; Chou, M.Y.; et al. MicroRNA let-7a represses chemoresistance and tumourigenicity in head and neck cancer via stem-like properties ablation. Oral Oncol. 2011, 47, 202–210. [Google Scholar] [CrossRef]
  114. Chang, C.J.; Hsu, C.C.; Chang, C.H.; Tsai, L.L.; Chang, Y.C.; Lu, S.W.; Yu, C.H.; Huang, H.S.; Wang, J.J.; Tsai, C.H.; et al. Let-7d functions as novel regulator of epithelial-mesenchymal transition and chemoresistant property in oral cancer. Oncol. Rep. 2011, 26, 1003–1010. [Google Scholar] [CrossRef]
  115. Maclellan, S.A.; Lawson, J.; Baik, J.; Guillaud, M.; Poh, C.F.-Y.; Garnis, C. Differential expression of miRNAs in the serum of patients with high-risk oral lesions. Cancer Med. 2012, 1, 268–274. [Google Scholar] [CrossRef] [PubMed]
  116. Schneider, A.; Victoria, B.; Lopez, Y.N. Tissue and serum microRNA profile of oral squamous cell carcinoma patients. Sci. Rep. 2018, 8, 675. [Google Scholar] [CrossRef] [Green Version]
  117. Fadhil, R.S.; Wei, M.Q.; Nikolarakos, D.; Good, D.; Nair, R.G. Salivary microRNA miR-let-7a-5p and miR-3928 could be used as potential diagnostic bio-markers for head and neck squamous cell carcinoma. PLoS ONE 2020, 15, e0221779. [Google Scholar] [CrossRef] [Green Version]
  118. Chang, S.S.; Jiang, W.W.; Smith, I.; Poeta, L.M.; Begum, S.; Glazer, C.; Shan, S.; Westra, W.; Sidransky, D.; Califano, J.A. MicroRNA alterations in head and neck squamous cell carcinoma. Int. J. Cancer 2008, 123, 2791–2797. [Google Scholar] [CrossRef] [Green Version]
  119. De Ruyck, K.; Duprez, F.; Ferdinande, L.; Mbah, C.; Rios-Velazquez, E.; Hoebers, F.; Praet, M.; Deron, P.; Bonte, K.; Speel, E.J.; et al. A let-7 microRNA polymorphism in the KRAS 3′-UTR is prognostic in oropharyngeal cancer. Cancer Epidemiol. 2014, 38, 591–598. [Google Scholar] [CrossRef]
  120. Song, F.C.; Yang, Y.; Liu, J.X. Expression and significances of MiRNA Let-7 and HMGA2 in laryngeal carcinoma. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 4452–4458. [Google Scholar]
  121. Chien, C.S.; Wang, M.L.; Chu, P.Y.; Chang, Y.L.; Liu, W.H.; Yu, C.C.; Lan, Y.T.; Huang, P.I.; Lee, Y.Y.; Chen, Y.W.; et al. Lin28B/Let-7 Regulates Expression of Oct4 and Sox2 and Reprograms Oral Squamous Cell Carcinoma Cells to a Stem-like State. Cancer Res. 2015, 75, 2553–2565. [Google Scholar] [CrossRef] [Green Version]
  122. Hilly, O.; Pillar, N.; Stern, S.; Strenov, Y.; Bachar, G.; Shomron, N.; Shpitzer, T. Distinctive pattern of let-7 family microRNAs in aggressive carcinoma of the oral tongue in young patients. Oncol. Lett. 2016, 12, 1729–1736. [Google Scholar] [CrossRef] [Green Version]
  123. Lamperska, K.M.; Kolenda, T.; Teresiak, A.; Kowalik, A.; Kruszyna-Mochalska, M.; Jackowiak, W.; Bliźniak, R.; Przybyła, W.; Kapałczyńska, M.; Kozlowski, P. Different levels of let-7d expression modulate response of FaDu cells to irradiation and chemotherapeutics. PLoS ONE 2017, 12, e0180265. [Google Scholar] [CrossRef]
  124. Peng, C.Y.; Wang, T.Y.; Lee, S.S.; Hsieh, P.L.; Liao, Y.W.; Tsai, L.L.; Fang, C.Y.; Yu, C.C. Let-7c restores radiosensitivity and chemosensitivity and impairs stemness in oral cancer cells through inhibiting interleukin-8. J. Oral Pathol. Med. 2018, 47, 590–597. [Google Scholar] [CrossRef]
  125. Feng, X.; Wang, Z.; Fillmore, R.; Xi, Y. MiR-200, a new star miRNA in human cancer. Cancer Lett. 2014, 344, 166–173. [Google Scholar] [CrossRef] [Green Version]
  126. Díaz-Martín, J.; Díaz-López, A.; Moreno-Bueno, G.; Castilla, M.; Rosa-Rosa, J.M.; Cano, A.; Palacios, J. A core microRNA signature associated with inducers of the epithelial-to-mesenchymal transition. J. Pathol. 2014, 232, 319–329. [Google Scholar] [CrossRef]
  127. Skourti, E.; Logotheti, S.; Kontos, C.K.; Pavlopoulou, A.; Dimoragka, P.T.; Trougakos, I.P.; Gorgoulis, V.; Scorilas, A.; Michalopoulos, I.; Zoumpourlis, V. Progression of mouse skin carcinogenesis is associated with the orchestrated deregulation of mir-200 family members, mir-205 and their common targets. Mol. Carcinog. 2016, 55, 1229–1242. [Google Scholar] [CrossRef]
  128. Humphries, B.; Yang, C. The microRNA-200 family: Small molecules with novel roles in cancer development, progression and therapy. Oncotarget 2015, 6, 6472–6498. [Google Scholar] [CrossRef] [Green Version]
  129. Koshizuka, K.; Hanazawa, T.; Arai, T.; Okato, A.; Kikkawa, N.; Seki, N. Involvement of aberrantly expressed microRNAs in the pathogenesis of head and neck squamous cell carcinoma. Cancer Metastasis Rev. 2017, 36, 525–545. [Google Scholar] [CrossRef]
  130. Cappellesso, R.; Marioni, G.; Crescenzi, M.; Giacomelli, L.; Guzzardo, V.; Mussato, A.; Staffieri, A.; Martini, A.; Blandamura, S.; Fassina, A. The prognostic role of the epithelial-mesenchymal transition markers E-cadherin and Slug in laryngeal squamous cell carcinoma. Histopathology 2015, 67, 491–500. [Google Scholar] [CrossRef]
  131. Holt, J.; Walter, V. Integrative Analysis of miRNAs Identifies Clinically Relevant Epithelial and Stromal Subtypes of Head and Neck Squamous Cell Carcinoma. Clin. Cancer Res. 2021, 27, 831–842. [Google Scholar] [CrossRef]
  132. Lo, W.L.; Yu, C.C.; Chiou, G.Y.; Chen, Y.W.; Huang, P.I.; Chien, C.S.; Tseng, L.M.; Chu, P.Y.; Lu, K.H.; Chang, K.W.; et al. MicroRNA-200c attenuates tumour growth and metastasis of presumptive head and neck squamous cell carcinoma stem cells. J. Pathol. 2011, 223, 482–495. [Google Scholar] [CrossRef] [PubMed]
  133. Park, N.J.; Zhou, H.; Elashoff, D.; Henson, B.S.; Kastratovic, D.A.; Abemayor, E.; Wong, D.T. Salivary microRNA: Discovery, characterization, and clinical utility for oral cancer detection. Clin. Cancer Res. 2009, 15, 5473–5477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Greither, T.; Vorwerk, F.; Kappler, M.; Bache, M.; Taubert, H.; Kuhnt, T.; Hey, J.; Eckert, A.W. Salivary miR-93 and miR-200a as post-radiotherapy biomarkers in head and neck squamous cell carcinoma. Oncol. Rep. 2017, 38, 1268–1275. [Google Scholar] [CrossRef] [Green Version]
  135. Kim, E.J.; Kim, J.S.; Lee, S.; Lee, H.; Yoon, J.S.; Hong, J.H.; Chun, S.H.; Sun, S.; Won, H.S.; Hong, S.A.; et al. QKI, a miR-200 target gene, suppresses epithelial-to-mesenchymal transition and tumor growth. Int. J. Cancer 2019, 145, 1585–1595. [Google Scholar] [CrossRef]
  136. Tamagawa, S.; Beder, L.B.; Hotomi, M.; Gunduz, M.; Yata, K.; Grenman, R.; Yamanaka, N. Role of miR-200c/miR-141 in the regulation of epithelial-mesenchymal transition and migration in head and neck squamous cell carcinoma. Int. J. Mol. Med. 2014, 33, 879–886. [Google Scholar] [CrossRef] [Green Version]
  137. Zhang, X.; Xu, L.; Yang, T. miR-31 Modulates Liver Cancer HepG2 Cell Apoptosis and Invasion via ROCK1/F-Actin Pathways. OncoTargets Ther. 2020, 13, 877–888. [Google Scholar] [CrossRef] [Green Version]
  138. Lin, P.-C.; Chiu, Y.-L.; Banerjee, S.; Park, K.; Mosquera, J.M.; Giannopoulou, E.; Alves, P.; Tewari, A.K.; Gerstein, M.B.; Beltran, H.; et al. Epigenetic repression of miR-31 disrupts androgen receptor homeostasis and contributes to prostate cancer progression. Cancer Res. 2013, 73, 1232–1244. [Google Scholar] [CrossRef] [Green Version]
  139. Luo, L.J.; Yang, F.; Ding, J.J.; Yan, D.L.; Wang, D.D.; Yang, S.J.; Ding, L.; Li, J.; Chen, D.; Ma, R.; et al. MiR-31 inhibits migration and invasion by targeting SATB2 in triple negative breast cancer. Gene 2016, 594, 47–58. [Google Scholar] [CrossRef]
  140. Cui, Q. Significance of miR-27a and miR-31 in early diagnosis and prognosis of colorectal cancer. Oncol. Lett. 2019, 18, 3092–3096. [Google Scholar] [CrossRef]
  141. Zheng, W.; Liu, Z.; Zhang, W.; Hu, X. miR-31 functions as an oncogene in cervical cancer. Arch. Gynecol. Obstet. 2015, 292, 1083–1089. [Google Scholar] [CrossRef] [PubMed]
  142. Mu, J.F.; Wang, X.D.; Sun, P.D. Expression of miR-31 in rectal cancer patients and its effect on proliferation ability of rectal cancer cells SW837. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 8675–8681. [Google Scholar] [CrossRef]
  143. Davenport, M.L.; Echols, J.B. miR-31 Displays Subtype Specificity in Lung Cancer. Cancer Res. 2021, 81, 1942–1953. [Google Scholar] [CrossRef] [PubMed]
  144. Kao, S.Y.; Tsai, M.M.; Wu, C.H.; Chen, J.J.; Tseng, S.H.; Lin, S.C.; Chang, K.W. Co-targeting of multiple microRNAs on factor-Inhibiting hypoxia-Inducible factor gene for the pathogenesis of head and neck carcinomas. Head Neck 2016, 38, 522–528. [Google Scholar] [CrossRef]
  145. Lajer, C.B.; Nielsen, F.C.; Friis-Hansen, L.; Norrild, B.; Borup, R.; Garnæs, E.; Rossing, M.; Specht, L.; Therkildsen, M.H.; Nauntofte, B.; et al. Different miRNA signatures of oral and pharyngeal squamous cell carcinomas: A prospective translational study. Br. J. Cancer 2011, 104, 830–840. [Google Scholar] [CrossRef] [PubMed]
  146. Cinpolat, O.; Unal, Z.N.; Ismi, O.; Gorur, A.; Unal, M. Comparison of microRNA profiles between benign and malignant salivary gland tumors in tissue, blood and saliva samples: A prospective, case-control study. Braz. J. Otorhinolaryngol. 2017, 83, 276–284. [Google Scholar] [CrossRef] [Green Version]
  147. Qiang, H.; Zhan, X.; Wang, W.; Cheng, Z.; Ma, S.; Jiang, C. A Study on the Correlations of the miR-31 Expression with the Pathogenesis and Prognosis of Head and Neck Squamous Cell Carcinoma. Cancer Biother. Radiopharm. 2019, 34, 189–195. [Google Scholar] [CrossRef]
  148. Liu, C.J.; Kao, S.Y.; Tu, H.F.; Tsai, M.M.; Chang, K.W.; Lin, S.C. Increase of microRNA miR-31 level in plasma could be a potential marker of oral cancer. Oral Dis. 2010, 16, 360–364. [Google Scholar] [CrossRef]
  149. Liu, C.J.; Tsai, M.M.; Hung, P.S.; Kao, S.Y.; Liu, T.Y.; Wu, K.J.; Chiou, S.H.; Lin, S.C.; Chang, K.W. miR-31 ablates expression of the HIF regulatory factor FIH to activate the HIF pathway in head and neck carcinoma. Cancer Res. 2010, 70, 1635–1644. [Google Scholar] [CrossRef] [Green Version]
  150. Oshima, S.; Asai, S.; Seki, N. Identification of Tumor Suppressive Genes Regulated by miR-31-5p and miR-31-3p in Head and Neck Squamous Cell Carcinoma. Int. J. Mol. Sci. 2021, 22, 6199. [Google Scholar] [CrossRef]
  151. Yi, S.J.; Liu, P.; Chen, B.L.; Ou-Yang, L.; Xiong, W.M.; Su, J.P. Circulating miR-31-5p may be a potential diagnostic biomarker in nasopharyngeal carcinoma. Neoplasma 2019, 66, 825–829. [Google Scholar] [CrossRef]
  152. Mak, M.P.; Pasini, F.S.; Diao, L.; Garcia, F.O.T.; Takahashi, T.K.; Nakazato, D.; Martins, R.E.; Almeida, C.M.; Kulcsar, M.A.V.; Lamounier, V.A.; et al. Valproic acid combined with cisplatin-based chemoradiation in locally advanced head and neck squamous cell carcinoma patients and associated biomarkers. Ecancermedicalscience 2020, 14, 1155. [Google Scholar] [CrossRef] [PubMed]
  153. Yang, S.; Wang, J. Long non-coding RNA LOC554202 promotes laryngeal squamous cell carcinoma progression through regulating miR-31. J. Cell. Biochem. 2018, 119, 6953–6960. [Google Scholar] [CrossRef] [PubMed]
  154. Liu, C.J.; Lin, S.C.; Yang, C.C.; Cheng, H.W.; Chang, K.W. Exploiting salivary miR-31 as a clinical biomarker of oral squamous cell carcinoma. Head Neck 2012, 34, 219–224. [Google Scholar] [CrossRef]
  155. Kyurkchiyan, S.G.; Popov, T.M. A pilot study reveals the potential of miR-31-3p and miR-196a-5p as non-invasive biomarkers in advanced laryngeal cancer. Folia Medica 2021, 63, 355–364. [Google Scholar] [CrossRef]
  156. Yin, H.; Sun, Y.; Wang, X.; Park, J.; Zhang, Y.; Li, M.; Yin, J.; Liu, Q.; Wei, M. Progress on the relationship between miR-125 family and tumorigenesis. Exp. Cell Res. 2015, 339, 252–260. [Google Scholar] [CrossRef] [PubMed]
  157. Henson, B.J.; Bhattacharjee, S.; O’Dee, D.M.; Feingold, E.; Gollin, S.M. Decreased expression of miR-125b and miR-100 in oral cancer cells contributes to malignancy. Genes Chromosomes Cancer 2009, 48, 569–582. [Google Scholar] [CrossRef] [Green Version]
  158. Hui, A.B.; Lenarduzzi, M.; Krushel, T.; Waldron, L.; Pintilie, M.; Shi, W.; Perez-Ordonez, B.; Jurisica, I.; O’Sullivan, B.; Waldron, J.; et al. Comprehensive MicroRNA profiling for head and neck squamous cell carcinomas. Clin. Cancer Res. 2010, 16, 1129–1139. [Google Scholar] [CrossRef] [Green Version]
  159. Cao, P.; Zhou, L.; Zhang, J.; Zheng, F.; Wang, H.; Ma, D.; Tian, J. Comprehensive expression profiling of microRNAs in laryngeal squamous cell carcinoma. Head Neck 2013, 35, 720–728. [Google Scholar] [CrossRef]
  160. Manikandan, M.; Deva Magendhra Rao, A.K.; Rajkumar, K.S.; Rajaraman, R.; Munirajan, A.K. Altered levels of miR-21, miR-125b-2*, miR-138, miR-155, miR-184, and miR-205 in oral squamous cell carcinoma and association with clinicopathological characteristics. J. Oral Pathol. Med. 2015, 44, 792–800. [Google Scholar] [CrossRef]
  161. Mei, L.L.; Wang, W.J.; Qiu, Y.T.; Xie, X.F.; Bai, J.; Shi, Z.Z. miR-125b-5p functions as a tumor suppressor gene partially by regulating HMGA2 in esophageal squamous cell carcinoma. PLoS ONE 2017, 12, e0185636. [Google Scholar] [CrossRef] [PubMed]
  162. Troiano, G.; Mastrangelo, F.; Caponio, V.C.A.; Laino, L.; Cirillo, N.; Lo Muzio, L. Predictive Prognostic Value of Tissue-Based MicroRNA Expression in Oral Squamous Cell Carcinoma: A Systematic Review and Meta-analysis. J. Dent. Res. 2018, 97, 759–766. [Google Scholar] [CrossRef] [PubMed]
  163. Ayaz, L.; Görür, A.; Yaroğlu, H.Y.; Ozcan, C.; Tamer, L. Differential expression of microRNAs in plasma of patients with laryngeal squamous cell carcinoma: Potential early-detection markers for laryngeal squamous cell carcinoma. J. Cancer Res. Clin. Oncol. 2013, 139, 1499–1506. [Google Scholar] [CrossRef]
  164. González-Arriagada, W.; Olivero, P.; Rodriguez, B.; Lozano, C.; Oliveira, C.; Coletta, R. Clinicopathological significance of miR-26, miR-107, miR-125b and miR-203 in head and neck carcinomas. Oral Dis. 2018, 24, 930–939. [Google Scholar] [CrossRef]
  165. Shiiba, M.; Shinozuka, K.; Saito, K.; Fushimi, K.; Kasamatsu, A.; Ogawara, K.; Uzawa, K.; Ito, H.; Takiguchi, Y.; Tanzawa, H. MicroRNA-125b regulates proliferation and radioresistance of oral squamous cell carcinoma. Br. J. Cancer 2013, 108, 1817–1821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Vo, D.T.; Karanam, N.K.; Ding, L.; Saha, D.; Yordy, J.S.; Giri, U.; Heymach, J.V.; Story, M.D. miR-125a-5p Functions as Tumor Suppressor microRNA And Is a Marker of Locoregional Recurrence And Poor prognosis in Head And Neck Cancer. Neoplasia 2019, 21, 849–862. [Google Scholar] [CrossRef] [PubMed]
  167. Lu, Y.-C.; Chang, J.T.; Chan, E.-C.; Chao, Y.-K.; Yeh, T.-S.; Chen, J.-S.; Cheng, A.-J. miR-196, an Emerging Cancer Biomarker for Digestive Tract Cancers. J. Cancer 2016, 7, 650–655. [Google Scholar] [CrossRef] [Green Version]
  168. Álvarez-Teijeiro, S.; Menéndez, S.T.; Villaronga, M.; Rodrigo, J.P.; Manterola, L.; de Villalaín, L.; de Vicente, J.C.; Alonso-Durán, L.; Fernández, M.P.; Lawrie, C.H.; et al. Dysregulation of Mir-196b in Head and Neck Cancers Leads to Pleiotropic Effects in the Tumor Cells and Surrounding Stromal Fibroblasts. Sci. Rep. 2017, 7, 17785. [Google Scholar] [CrossRef]
  169. Wang, Y.; Hu, Y.; Chen, L.; Wu, J.; Wu, K.; Du, J.; Xue, H.; Shen, B. Molecular mechanisms and prognostic markers in head and neck squamous cell carcinoma: A bioinformatic analysis. Int. J. Clin. Exp. Pathol. 2020, 13, 371–381. [Google Scholar]
  170. Liu, C.J.; Tsai, M.M.; Tu, H.F.; Lui, M.T.; Cheng, H.W.; Lin, S.C. miR-196a overexpression and miR-196a2 gene polymorphism are prognostic predictors of oral carcinomas. Ann. Surg. Oncol. 2013, 20 (Suppl. S3), S406–S414. [Google Scholar] [CrossRef]
  171. Lu, Y.C.; Chang, J.T.; Huang, Y.C.; Huang, C.C.; Chen, W.H.; Lee, L.Y.; Huang, B.S.; Chen, Y.J.; Li, H.F.; Cheng, A.J. Combined determination of circulating miR-196a and miR-196b levels produces high sensitivity and specificity for early detection of oral cancer. Clin. Biochem. 2015, 48, 115–121. [Google Scholar] [CrossRef]
  172. Patil, S.; Warnakulasuriya, S. Blood-based circulating microRNAs as potential biomarkers for predicting the prognosis of head and neck cancer-a systematic review. Clin. Oral Investig. 2020, 24, 3833–3841. [Google Scholar] [CrossRef]
  173. Suh, Y.E.; Raulf, N.; Gäken, J.; Lawler, K.; Urbano, T.G.; Bullenkamp, J.; Gobeil, S.; Huot, J.; Odell, E.; Tavassoli, M. MicroRNA-196a promotes an oncogenic effect in head and neck cancer cells by suppressing annexin A1 and enhancing radioresistance. Int. J. Cancer 2015, 137, 1021–1034. [Google Scholar] [CrossRef] [PubMed]
  174. Darda, L.; Hakami, F.; Morgan, R.; Murdoch, C.; Lambert, D.W.; Hunter, K.D. The role of HOXB9 and miR-196a in head and neck squamous cell carcinoma. PLoS ONE 2015, 10, e0122285. [Google Scholar] [CrossRef] [PubMed]
  175. Tsai, S.C.; Huang, S.F.; Chiang, J.H.; Chen, Y.F.; Huang, C.C.; Tsai, M.H.; Tsai, F.J.; Kao, M.C.; Yang, J.S. The differential regulation of microRNAs is associated with oral cancer. Oncol. Rep. 2017, 38, 1613–1620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Zhao, X.; Zhang, W.; Ji, W. miR-196b is a prognostic factor of human laryngeal squamous cell carcinoma and promotes tumor progression by targeting SOCS2. Biochem. Biophys. Res. Commun. 2018, 501, 584–592. [Google Scholar] [CrossRef]
  177. Luo, M.; Sun, G.; Sun, J.W. MiR-196b affects the progression and prognosis of human LSCC through targeting PCDH-17. Auris Nasus Larynx 2019, 46, 583–592. [Google Scholar] [CrossRef] [Green Version]
  178. Álvarez-Teijeiro, S.; Menéndez, S.T.; Villaronga, M.; Pena-Alonso, E.; Rodrigo, J.P.; Morgan, R.O. Annexin A1 down-regulation in head and neck squamous cell carcinoma is mediated via transcriptional control with direct involvement of miR-196a/b. Sci. Rep. 2017, 7, 6790. [Google Scholar] [CrossRef] [Green Version]
  179. Maruyama, T.; Nishihara, K.; Umikawa, M.; Arasaki, A.; Nakasone, T.; Nimura, F.; Matayoshi, A.; Takei, K.; Nakachi, S.; Kariya, K.I.; et al. MicroRNA-196a-5p is a potential prognostic marker of delayed lymph node metastasis in early-stage tongue squamous cell carcinoma. Oncol. Lett. 2018, 15, 2349–2363. [Google Scholar] [CrossRef]
  180. Saito, K.; Inagaki, K.; Kamimoto, T.; Ito, Y.; Sugita, T.; Nakajo, S.; Hirasawa, A.; Iwamaru, A.; Ishikura, T.; Hanaoka, H.; et al. MicroRNA-196a is a putative diagnostic biomarker and therapeutic target for laryngeal cancer. PLoS ONE 2013, 8, e71480. [Google Scholar] [CrossRef] [Green Version]
  181. Coolen, M.; Katz, S.; Bally-Cuif, L. miR-9: A versatile regulator of neurogenesis. Front. Cell. Neurosci. 2013, 7, 220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Wan, Y.; Zhang, X.; Tang, K.D.; Blick, T.; Kenny, L.; Thompson, E.W.; Punyadeera, C. Overexpression of miRNA-9 enhances galectin-3 levels in oral cavity cancers. Mol. Biol. Rep. 2021, 48, 3979–3989. [Google Scholar] [CrossRef] [PubMed]
  183. Tavakolian, S.; Goudarzi, H.; Faghihloo, E. Evaluating the expression level of miR-9-5p and miR-192-5p in gastrointestinal cancer: Introducing novel screening biomarkers for patients. BMC Res. Notes 2020, 13, 226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Milanesi, E.; Dobre, M.; Bucuroiu, A.I.; Herlea, V.; Manuc, T.E. miRNAs-Based Molecular Signature for KRAS Mutated and Wild Type Colorectal Cancer: An Explorative Study. J. Immunol. Res. 2020, 2020, 4927120. [Google Scholar] [CrossRef]
  185. Guan, Q.; Yuan, B.; Zhang, X.; Yan, T.; Li, J.; Xu, W. Long non-coding RNA DUXAP8 promotes tumorigenesis by regulating IGF1R via miR-9-3p in hepatocellular carcinoma. Exp. Ther. Med. 2021, 22, 755. [Google Scholar] [CrossRef]
  186. Dutkowska, A.; Szmyd, B.; Kaszkowiak, M.; Domańska-Senderowska, D.; Pastuszak-Lewandoska, D.; Brzeziańska-Lasota, E.; Kordiak, J.; Antczak, A. Expression of inflammatory interleukins and selected miRNAs in non-small cell lung cancer. Sci. Rep. 2021, 11, 5092. [Google Scholar] [CrossRef]
  187. Li, X.; Tang, X.; Li, K.; Lu, L. Evaluation of Serum MicroRNAs (miR-9-5p, miR-17-5p, and miR-148a-3p) as Potential Biomarkers of Breast Cancer. BioMed Res. Int. 2022, 2022, 9961412. [Google Scholar] [CrossRef]
  188. Farzanehpour, M.; Mozhgani, S.H.; Jalilvand, S.; Faghihloo, E.; Akhavan, S.; Salimi, V.; Azad, T.M. Serum and tissue miRNAs: Potential biomarkers for the diagnosis of cervical cancer. Virol. J. 2019, 16, 116. [Google Scholar] [CrossRef] [Green Version]
  189. Kovaříková, J.; Baranová, I.; Laco, J.; Rozkošová, K.; Vošmíková, H.; Vošmík, M.; Dundr, P.; Němejcová, K.; Michálek, J.; Palička, V.; et al. Deregulation of Selected MicroRNAs in Sinonasal Squamous Cell Carcinoma: Searching for Potential Prognostic Biomarkers. Folia Biol. 2019, 65, 142–151. [Google Scholar]
  190. Wan, Y.; Vagenas, D.; Salazar, C.; Kenny, L.; Perry, C.; Calvopiña, D.; Punyadeera, C. Salivary miRNA panel to detect HPV-positive and HPV-negative head and neck cancer patients. Oncotarget 2017, 8, 99990–100001. [Google Scholar] [CrossRef] [Green Version]
  191. Salazar, C.; Nagadia, R.; Pandit, P.; Cooper-White, J.; Banerjee, N.; Dimitrova, N.; Coman, W.B.; Punyadeera, C. A novel saliva-based microRNA biomarker panel to detect head and neck cancers. Cell. Oncol. 2014, 37, 331–338. [Google Scholar] [CrossRef] [PubMed]
  192. Lu, J.; Luo, H.; Liu, X.; Peng, Y.; Zhang, B.; Wang, L.; Xu, X.; Peng, X.; Li, G.; Tian, W.; et al. miR-9 targets CXCR4 and functions as a potential tumor suppressor in nasopharyngeal carcinoma. Carcinogenesis 2014, 35, 554–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. 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]
  194. Lu, J.; Xu, X.; Liu, X.; Peng, Y.; Zhang, B.; Wang, L.; Luo, H.; Peng, X.; Li, G.; Tian, W.; et al. Predictive value of miR-9 as a potential biomarker for nasopharyngeal carcinoma metastasis. Br. J. Cancer 2014, 110, 392–398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Xiao, C.; Wang, L.; Zhu, L.; Zhang, C.; Zhou, J. Curcumin inhibits oral squamous cell carcinoma SCC-9 cells proliferation by regulating miR-9 expression. Biochem. Biophys. Res. Commun. 2014, 454, 576–580. [Google Scholar] [CrossRef]
  196. Yu, T.; Liu, K.; Wu, Y.; Fan, J.; Chen, J.; Li, C.; Yang, Q.; Wang, Z. MicroRNA-9 inhibits the proliferation of oral squamous cell carcinoma cells by suppressing expression of CXCR4 via the Wnt/β-catenin signaling pathway. Oncogene 2014, 33, 5017–5027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Sun, L.; Liu, L.; Fu, H.; Wang, Q.; Shi, Y. Association of Decreased Expression of Serum miR-9 with Poor Prognosis of Oral Squamous Cell Carcinoma Patients. Med. Sci. Monit. 2016, 22, 289–294. [Google Scholar] [CrossRef]
  198. Citron, F.; Segatto, I.; Musco, L.; Pellarin, I.; Rampioni Vinciguerra, G.L.; Franchin, G.; Fanetti, G.; Miccichè, F.; Giacomarra, V.; Lupato, V.; et al. miR-9 modulates and predicts the response to radiotherapy and EGFR inhibition in HNSCC. EMBO Mol. Med. 2021, 13, e12872. [Google Scholar] [CrossRef]
  199. Ouyang, Y.B.; Lu, Y.; Yue, S.; Giffard, R.G. miR-181 targets multiple Bcl-2 family members and influences apoptosis and mitochondrial function in astrocytes. Mitochondrion 2012, 12, 213–219. [Google Scholar] [CrossRef] [Green Version]
  200. Henao-Mejia, J.; Williams, A.; Goff, L.A.; Staron, M.; Licona-Limon, P.; Kaech, S.M.; Nakayama, M.; Rinn, J.L.; Flavell, R.A. The microRNA miR-181 is a critical cellular metabolic rheostat essential for NKT cell ontogenesis and lymphocyte development and homeostasis. Immunity 2013, 38, 984–997. [Google Scholar] [CrossRef] [Green Version]
  201. Pop-Bica, C.; Pintea, S.; Cojocneanu-Petric, R.; Del Sal, G.; Piazza, S.; Wu, Z.H.; Alencar, A.J.; Lossos, I.S.; Berindan-Neagoe, I.; Calin, G.A. MiR-181 family-specific behavior in different cancers: A meta-analysis view. Cancer Metastasis Rev. 2018, 37, 17–32. [Google Scholar] [CrossRef]
  202. Indrieri, A.; Carrella, S.; Carotenuto, P.; Banfi, S.; Franco, B. The Pervasive Role of the miR-181 Family in Development, Neurodegeneration, and Cancer. Int. J. Mol. Sci. 2020, 21, 2092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Sun, W.; Wang, X.; Li, J.; You, C.; Lu, P.; Feng, H.; Kong, Y.; Zhang, H.; Liu, Y.; Jiao, R.; et al. MicroRNA-181a promotes angiogenesis in colorectal cancer by targeting SRCIN1 to promote the SRC/VEGF signaling pathway. Cell Death Dis. 2018, 9, 438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Li, Z.; Wang, H.; Xu, Z.; Sun, Y.; Han, J. Expression and mechanism of microRNA-181A on incidence and survival in late liver metastases of colorectal cancer. Oncol. Rep. 2016, 35, 1403–1408. [Google Scholar] [CrossRef] [Green Version]
  205. Cao, Y.; Zhao, D.; Li, P.; Wang, L.; Qiao, B.; Qin, X.; Li, L.; Wang, Y. MicroRNA-181a-5p Impedes IL-17-Induced Nonsmall Cell Lung Cancer Proliferation and Migration through Targeting VCAM-1. Cell Physiol. Biochem. 2017, 42, 346–356. [Google Scholar] [CrossRef]
  206. Liu, J.; Xu, D.; Wang, Q.; Zheng, D.; Jiang, X.; Xu, L. LPS induced miR-181a promotes pancreatic cancer cell migration via targeting PTEN and MAP2K4. Dig. Dis. Sci. 2014, 59, 1452–1460. [Google Scholar] [CrossRef] [PubMed]
  207. Parikh, A.; Lee, C.; Joseph, P.; Marchini, S.; Baccarini, A.; Kolev, V.; Romualdi, C.; Fruscio, R.; Shah, H.; Wang, F.; et al. microRNA-181a has a critical role in ovarian cancer progression through the regulation of the epithelial-mesenchymal transition. Nat. Commun. 2014, 5, 2977. [Google Scholar] [CrossRef] [PubMed]
  208. Zhiping, C.; Shijun, T.; Linhui, W.; Yapei, W.; Lianxi, Q.; Qiang, D. MiR-181a promotes epithelial to mesenchymal transition of prostate cancer cells by targeting TGIF2. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 4835–4843. [Google Scholar] [PubMed]
  209. Jamali, Z.; Asl Aminabadi, N.; Attaran, R.; Pournagiazar, F.; Ghertasi Oskouei, S.; Ahmadpour, F. MicroRNAs as prognostic molecular signatures in human head and neck squamous cell carcinoma: A systematic review and meta-analysis. Oral Oncol. 2015, 51, 321–331. [Google Scholar] [CrossRef]
  210. Dai, Y.; Zang, Y.; Li, J.; Liu, Y.; Wan, B. miR-181a and miR-203 inhibit migration and invasion of laryngeal carcinoma cells by interacting with ATF2. Int. J. Clin. Exp. Pathol. 2019, 12, 133–141. [Google Scholar]
  211. Yang, C.C.; Hung, P.S.; Wang, P.W.; Liu, C.J.; Chu, T.H.; Cheng, H.W.; Lin, S.C. miR-181 as a putative biomarker for lymph-node metastasis of oral squamous cell carcinoma. J. Oral Pathol. Med. 2011, 40, 397–404. [Google Scholar] [CrossRef] [PubMed]
  212. Lee, S.H.; Lee, C.R.; Rigas, N.K.; Kim, R.H.; Kang, M.K.; Park, N.H.; Shin, K.H. Human papillomavirus 16 (HPV16) enhances tumor growth and cancer stemness of HPV-negative oral/oropharyngeal squamous cell carcinoma cells via miR-181 regulation. Papillomavirus Res. 2015, 1, 116–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Quabius, E.S.; Merz, I.; Gorogh, T.; Hedderich, J.; Haag, J.; Rocken, C.; Ambrosch, P.; Hoffmann, M. miRNA-expression in tonsillar squamous cell carcinomas in relation to HPV infection and expression of the antileukoproteinase SLPI. Papillomavirus Res. 2017, 4, 26–34. [Google Scholar] [CrossRef]
  214. Lin, Z.; Chen, Y.; Lin, Y.; Lin, H.; Li, H.; Su, X.; Fang, Z.; Wang, J.; Wei, Q.; Teng, J.; et al. Potential miRNA biomarkers for the diagnosis and prognosis of esophageal cancer detected by a novel absolute quantitative RT-qPCR method. Sci. Rep. 2020, 10, 20065. [Google Scholar] [CrossRef]
  215. Zhou, X.; You, M.; Wang, F.; Wang, Z.; Gao, X.; Jing, C.; Liu, J.; Guo, M.; Li, J.; Luo, A.; et al. Multifunctional Graphdiyne-Cerium Oxide Nanozymes Facilitate MicroRNA Delivery and Attenuate Tumor Hypoxia for Highly Efficient Radiotherapy of Esophageal Cancer. Adv. Mater. 2021, 33, e2100556. [Google Scholar] [CrossRef] [PubMed]
  216. Xiang, Z.; Dong, X.; Sun, Q.; Li, X.; Yan, B. Clinical significance of up-regulated miR-181a in prognosis and progression of esophageal cancer. Acta Biochim. Biophys. Sin. 2014, 46, 1007–1010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Wang, F.; Zhou, J.; Zhang, Y.; Wang, Y.; Cheng, L.; Bai, Y.; Ma, H. The Value of MicroRNA-155 as a Prognostic Factor for Survival in Non-Small Cell Lung Cancer: A Meta-Analysis. PLoS ONE 2015, 10, e0136889. [Google Scholar] [CrossRef] [Green Version]
  218. Liu, W.; Zhou, X.; Li, Y.; Jiang, H.; Chen, A. Long Non-Coding RNA NORAD Inhibits Breast Cancer Cell Proliferation and Metastasis by Regulating miR-155-5p/SOCS1 Axis. J. Breast Cancer 2021, 24, 330–343. [Google Scholar] [CrossRef]
  219. Yang, L.; Li, C.; Liang, F.; Fan, Y.; Zhang, S. MiRNA-155 promotes proliferation by targeting caudal-type homeobox 1 (CDX1) in glioma cells. Biomed. Pharmacother. 2017, 95, 1759–1764. [Google Scholar] [CrossRef]
  220. He, B.; Gao, S.Q.; Huang, L.D.; Huang, Y.H.; Zhang, Q.Y.; Zhou, M.T.; Shi, H.Q.; Song, Q.T.; Shan, Y.F. MicroRNA-155 promotes the proliferation and invasion abilities of colon cancer cells by targeting quaking. Mol. Med. Rep. 2015, 11, 2355–2359. [Google Scholar] [CrossRef] [Green Version]
  221. D’Souza, W.; Kumar, A. microRNAs in oral cancer: Moving from bench to bed as next generation medicine. Oral Oncol. 2020, 111, 104916. [Google Scholar] [CrossRef]
  222. Ning, S.; Liu, H.; Gao, B.; Wei, W.; Yang, A.; Li, J.; Zhang, L. miR-155, miR-96 and miR-99a as potential diagnostic and prognostic tools for the clinical management of hepatocellular carcinoma. Oncol. Lett. 2019, 18, 3381–3387. [Google Scholar] [CrossRef] [PubMed]
  223. 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]
  224. Zheng, Y.J.; Liang, T.S.; Wang, J.; Zhao, J.Y.; Zhai, S.N.; Yang, D.K.; Wang, L.D. MicroRNA-155 acts as a diagnostic and prognostic biomarker for oesophageal squamous cell carcinoma. Artif. Cells Nanomed. Biotechnol. 2020, 48, 977–982. [Google Scholar] [CrossRef]
  225. Wu, M.; Duan, Q.; Liu, X.; Zhang, P.; Fu, Y.; Zhang, Z.; Liu, L.; Cheng, J.; Jiang, H. MiR-155-5p promotes oral cancer progression by targeting chromatin remodeling gene ARID2. Biomed. Pharmacother. 2020, 122, 109696. [Google Scholar] [CrossRef] [PubMed]
  226. Baba, O.; Hasegawa, S.; Nagai, H.; Uchida, F.; Yamatoji, M.; Kanno, N.I.; Yamagata, K.; Sakai, S.; Yanagawa, T.; Bukawa, H. MicroRNA-155-5p is associated with oral squamous cell carcinoma metastasis and poor prognosis. J. Oral Pathol. Med. 2016, 45, 248–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Shi, L.J.; Zhang, C.Y.; Zhou, Z.T.; Ma, J.Y.; Liu, Y.; Bao, Z.X.; Jiang, W.W. MicroRNA-155 in oral squamous cell carcinoma: Overexpression, localization, and prognostic potential. Head Neck 2015, 37, 970–976. [Google Scholar] [CrossRef]
  228. Hess, A.K.; Muer, A.; Mairinger, F.D.; Weichert, W.; Stenzinger, A.; Hummel, M.; Budach, V.; Tinhofer, I. MiR-200b and miR-155 as predictive biomarkers for the efficacy of chemoradiation in locally advanced head and neck squamous cell carcinoma. Eur. J. Cancer 2017, 77, 3–12. [Google Scholar] [CrossRef]
  229. Rather, M.I.; Nagashri, M.N.; Swamy, S.S.; Gopinath, K.S.; Kumar, A. Oncogenic microRNA-155 down-regulates tumor suppressor CDC73 and promotes oral squamous cell carcinoma cell proliferation: Implications for cancer therapeutics. J. Biol. Chem. 2013, 288, 608–618. [Google Scholar] [CrossRef] [Green Version]
  230. Lerner, C.; Wemmert, S.; Bochen, F.; Kulas, P.; Linxweiler, M.; Hasenfus, A.; Heinzelmann, J.; Leidinger, P.; Backes, C.; Meese, E.; et al. Characterization of miR-146a and miR-155 in blood, tissue and cell lines of head and neck squamous cell carcinoma patients and their impact on cell proliferation and migration. J. Cancer Res. Clin. Oncol. 2016, 142, 757–766. [Google Scholar] [CrossRef]
  231. Bersani, C.; Mints, M.; Tertipis, N.; Haeggblom, L.; Näsman, A.; Romanitan, M.; Dalianis, T.; Ramqvist, T. MicroRNA-155, -185 and -193b as biomarkers in human papillomavirus positive and negative tonsillar and base of tongue squamous cell carcinoma. Oral Oncol. 2018, 82, 8–16. [Google Scholar] [CrossRef]
  232. Nahand, J.S.; Karimzadeh, M.R.; Nezamnia, M.; Fatemipour, M.; Khatami, A.; Jamshidi, S.; Moghoofei, M.; Taghizadieh, M.; Hajighadimi, S.; Shafiee, A.; et al. The role of miR-146a in viral infection. IUBMB Life 2020, 72, 343–360. [Google Scholar] [CrossRef]
  233. Wani, J.A.; Majid, S.; Khan, A.; Arafah, A.; Ahmad, A.; Jan, B.L.; Shah, N.N.; Kazi, M.; Rehman, M.U. Clinico-Pathological Importance of miR-146a in Lung Cancer. Diagnostics 2021, 11, 274. [Google Scholar] [CrossRef]
  234. Dezfuli, N.K.; Alipoor, S.D.; Dalil Roofchayee, N.; Seyfi, S.; Salimi, B.; Adcock, I.M.; Mortaz, E. Evaluation Expression of miR-146a and miR-155 in Non-Small-Cell Lung Cancer Patients. Front. Oncol. 2021, 11, 715677. [Google Scholar] [CrossRef]
  235. Fortis, S.P.; Vaxevanis, C.K.; Mahaira, L.G.; Sofopoulos, M.; Sotiriadou, N.N.; Dinou, A.; Arnogiannaki, N.; Stavropoulos-Giokas, C.; Thanos, D.; Baxevanis, C.N.; et al. Serum miRNA-based distinct clusters define three groups of breast cancer patients with different clinicopathological and immune characteristics. Cancer Immunol. Immunother. 2019, 68, 57–70. [Google Scholar] [CrossRef]
  236. Adami, B.; Tabatabaeian, H.; Ghaedi, K.; Talebi, A.; Azadeh, M.; Dehdashtian, E. miR-146a is deregulated in gastric cancer. J. Cancer Res. Ther. 2019, 15, 108–114. [Google Scholar] [CrossRef]
  237. Hu, Q.; Song, J.; Ding, B.; Cui, Y.; Liang, J.; Han, S. miR-146a promotes cervical cancer cell viability via targeting IRAK1 and TRAF6. Oncol. Rep. 2018, 39, 3015–3024. [Google Scholar] [CrossRef] [Green Version]
  238. Zu, Y.; Yang, Y.; Zhu, J.; Bo, X.; Hou, S.; Zhang, B.; Qiu, J.; Zheng, J. MiR-146a suppresses hepatocellular carcinoma by downregulating TRAF6. Am. J. Cancer Res. 2016, 6, 2502–2513. [Google Scholar]
  239. Damodaran, M.; Paul, S.F.D.; Venkatesan, V. Genetic Polymorphisms in miR-146a, miR-196a2 and miR-125a Genes and its Association in Prostate Cancer. Pathol. Oncol. Res. 2020, 26, 193–200. [Google Scholar] [CrossRef]
  240. Garo, L.P.; Ajay, A.K.; Fujiwara, M.; Gabriely, G.; Raheja, R.; Kuhn, C.; Kenyon, B.; Skillin, N.; Kadowaki-Saga, R.; Saxena, S.; et al. MicroRNA-146a limits tumorigenic inflammation in colorectal cancer. Nat. Commun. 2021, 12, 2419. [Google Scholar] [CrossRef]
  241. Mei, J.; Bachoo, R.; Zhang, C.L. MicroRNA-146a inhibits glioma development by targeting Notch1. Mol. Cell. Biol. 2011, 31, 3584–3592. [Google Scholar] [CrossRef] [Green Version]
  242. Hung, P.S.; Liu, C.J.; Chou, C.S.; Kao, S.Y.; Yang, C.C.; Chang, K.W.; Chiu, T.H.; Lin, S.C. miR-146a enhances the oncogenicity of oral carcinoma by concomitant targeting of the IRAK1, TRAF6 and NUMB genes. PLoS ONE 2013, 8, e79926. [Google Scholar] [CrossRef] [PubMed]
  243. Wang, C.; Zhang, W.; Zhang, L.; Chen, X.; Liu, F.; Zhang, J.; Guan, S.; Sun, Y.; Chen, P.; Wang, D.; et al. miR-146a-5p mediates epithelial-mesenchymal transition of oesophageal squamous cell carcinoma via targeting Notch2. Br. J. Cancer 2016, 115, 1548–1554. [Google Scholar] [CrossRef]
  244. Wang, C.; Guan, S.; Liu, F.; Chen, X.; Han, L.; Wang, D.; Nesa, E.U.; Wang, X.; Bao, C.; Wang, N.; et al. Prognostic and diagnostic potential of miR-146a in oesophageal squamous cell carcinoma. Br. J. Cancer 2016, 114, 290–297. [Google Scholar] [CrossRef] [Green Version]
  245. Emmett, S.E.; Stark, M.S.; Pandeya, N.; Panizza, B.; Whiteman, D.C.; Antonsson, A. MicroRNA expression is associated with human papillomavirus status and prognosis in mucosal head and neck squamous cell carcinomas. Oral Oncol. 2021, 113, 105136. [Google Scholar] [CrossRef]
  246. Nariman-Saleh-Fam, Z.; Mansoori, Y.; Saadatian, Z.; Tavakkoly-Bazzaz, J.; Daraei, A.; Zununi Vahed, S.; Mahmoodzadeh, H.; Bastami, M. Dysregulated Expression of miR-146a and Its Associated Immune Effectors in Peripheral Blood Mononuclear Cells of Esophageal Carcinoma Patients. Immunol Investig. 2022, 51, 290–300. [Google Scholar] [CrossRef]
  247. Cui, M.; Yao, X.; Lin, Y.; Zhang, D.; Cui, R.; Zhang, X. Interactive functions of microRNAs in the miR-23a-27a-24-2 cluster and the potential for targeted therapy in cancer. J. Cell Physiol. 2020, 235, 6–16. [Google Scholar] [CrossRef]
  248. Roufayel, R.; Kadry, S. Expression of miR-23a by apoptotic regulators in human cancer: A review. Cancer Biol. Ther. 2017, 18, 269–276. [Google Scholar] [CrossRef] [Green Version]
  249. Itani, M.M.; Nassar, F.J.; Tfayli, A.H.; Talhouk, R.S.; Chamandi, G.K.; Itani, A.R.S.; Makoukji, J.; Boustany, R.N.; Hou, L.; Zgheib, N.K.; et al. A Signature of Four Circulating microRNAs as Potential Biomarkers for Diagnosing Early-Stage Breast Cancer. Int. J. Mol. Sci. 2021, 22, 6121. [Google Scholar] [CrossRef]
  250. Hua, K.; Chen, Y.T.; Chen, C.F.; Tang, Y.S.; Huang, T.T.; Lin, Y.C.; Yeh, T.S.; Huang, K.H.; Lee, H.C.; Hsu, M.T.; et al. MicroRNA-23a/27a/24-2 cluster promotes gastric cancer cell proliferation synergistically. Oncol. Lett. 2018, 16, 2319–2325. [Google Scholar] [CrossRef] [Green Version]
  251. Bao, L.; Zhao, J.; Dai, X.; Wang, Y.; Ma, R.; Su, Y.; Cui, H.; Niu, J.; Bai, S.; Xiao, Z.; et al. Correlation between miR-23a and onset of hepatocellular carcinoma. Clin. Res. Hepatol. Gastroenterol. 2014, 38, 318–330. [Google Scholar] [CrossRef] [PubMed]
  252. Fan, X.; Tao, S.; Li, Q.; Deng, B.; Tan, Q.Y.; Jin, H. The miR-23a/27a/24-2 cluster promotes postoperative progression of early-stage non-small cell lung cancer. Mol. Ther. Oncolytics 2022, 24, 205–217. [Google Scholar] [CrossRef]
  253. Piepoli, A.; Tavano, F.; Copetti, M.; Mazza, T.; Palumbo, O.; Panza, A.; di Mola, F.F.; Pazienza, V.; Mazzoccoli, G.; Biscaglia, G.; et al. Mirna expression profiles identify drivers in colorectal and pancreatic cancers. PLoS ONE 2012, 7, e33663. [Google Scholar] [CrossRef] [PubMed]
  254. Su, L.; Liu, M. Correlation analysis on the expression levels of microRNA-23a and microRNA-23b and the incidence and prognosis of ovarian cancer. Oncol. Lett. 2018, 16, 262–266. [Google Scholar] [CrossRef] [Green Version]
  255. Hatzl, S.; Perfler, B.; Wurm, S.; Uhl, B.; Quehenberger, F.; Ebner, S.; Troppmair, J.; Reinisch, A.; Wolfler, A.; Sill, H.; et al. Increased Expression of Micro-RNA-23a Mediates Chemoresistance to Cytarabine in Acute Myeloid Leukemia. Cancers 2020, 12, 496. [Google Scholar] [CrossRef] [Green Version]
  256. Wang, H.; Xue, W.; Ouyang, W.; Jiang, X.; Jiang, X. miR-23a-3p/SIX1 regulates glucose uptake and proliferation through GLUT3 in head and neck squamous cell carcinomas. J. Cancer 2020, 11, 2529–2539. [Google Scholar] [CrossRef]
  257. Chhabra, R.; Adlakha, Y.K.; Hariharan, M.; Scaria, V.; Saini, N. Upregulation of miR-23a-27a-24-2 cluster induces caspase-dependent and -independent apoptosis in human embryonic kidney cells. PLoS ONE 2009, 4, e5848. [Google Scholar] [CrossRef] [Green Version]
  258. Wang, N.; Zhu, M.; Tsao, S.W.; Man, K.; Zhang, Z.; Feng, Y. MiR-23a-mediated inhibition of topoisomerase 1 expression potentiates cell response to etoposide in human hepatocellular carcinoma. Mol. Cancer 2013, 12, 119. [Google Scholar] [CrossRef] [Green Version]
  259. Quan, J.; Pan, X.; Li, Y.; Hu, Y.; Tao, L.; Li, Z.; Zhao, L.; Wang, J.; Li, H.; Lai, Y.; et al. MiR-23a-3p acts as an oncogene and potential prognostic biomarker by targeting PNRC2 in RCC. Biomed. Pharmacother. 2019, 110, 656–666. [Google Scholar] [CrossRef]
  260. Yu, Z.W.; Zhong, L.P.; Ji, T.; Zhang, P.; Chen, W.T.; Zhang, C.P. MicroRNAs contribute to the chemoresistance of cisplatin in tongue squamous cell carcinoma lines. Oral Oncol. 2010, 46, 317–322. [Google Scholar] [CrossRef]
  261. Shi, Z.; Li, G.; Li, Z.; Liu, J.; Tang, Y. TMEM161B-AS1 suppresses proliferation, invasion and glycolysis by targeting miR-23a-3p/HIF1AN signal axis in oesophageal squamous cell carcinoma. J. Cell Mol. Med. 2021, 25, 6535–6549. [Google Scholar] [CrossRef] [PubMed]
  262. Xing, Y.; Zha, W.J.; Li, X.M.; Li, H.; Gao, F.; Ye, T.; Du, W.Q.; Liu, Y.C. Circular RNA circ-Foxo3 inhibits esophageal squamous cell cancer progression via the miR-23a/PTEN axis. J. Cell. Biochem. 2020, 121, 2595–2605. [Google Scholar] [CrossRef] [PubMed]
  263. Niwa, Y.; Yamada, S.; Sonohara, F.; Kurimoto, K.; Hayashi, M.; Tashiro, M.; Iwata, N.; Kanda, M.; Tanaka, C.; Kobayashi, D.; et al. Identification of a serum-based miRNA signature for response of esophageal squamous cell carcinoma to neoadjuvant chemotherapy. J. Transl. Med. 2019, 17, 1. [Google Scholar] [CrossRef] [PubMed]
  264. Komatsu, S.; Ichikawa, D.; Kawaguchi, T.; Takeshita, H.; Miyamae, M.; Ohashi, T.; Okajima, W.; Imamura, T.; Kiuchi, J.; Arita, T.; et al. Plasma microRNA profiles: Identification of miR-23a as a novel biomarker for chemoresistance in esophageal squamous cell carcinoma. Oncotarget 2016, 7, 62034–62048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Peng, F.; Zhang, H.; Du, Y.; Tan, P. miR-23a promotes cisplatin chemoresistance and protects against cisplatin-induced apoptosis in tongue squamous cell carcinoma cells through Twist. Oncol. Rep. 2015, 33, 942–950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Landgraf, P.; Rusu, M.; Sheridan, R.; Sewer, A.; Iovino, N.; Aravin, A.; Pfeffer, S.; Rice, A.; Kamphorst, A.O.; Landthaler, M.; et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 2007, 129, 1401–1414. [Google Scholar] [CrossRef] [Green Version]
  267. Yan, X.; Liang, H.; Deng, T.; Zhu, K.; Zhang, S.; Wang, N.; Jiang, X.; Wang, X.; Liu, R.; Zen, K.; et al. The identification of novel targets of miR-16 and characterization of their biological functions in cancer cells. Mol. Cancer 2013, 12, 92. [Google Scholar] [CrossRef] [Green Version]
  268. Aird, J.; Baird, A.M.; Lim, M.C.J.; McDermott, R.; Finn, S.P.; Gray, S.G. Carcinogenesis in prostate cancer: The role of long non-coding RNAs. Noncoding RNA Res. 2018, 3, 29–38. [Google Scholar] [CrossRef]
  269. Pidikova, P.; Reis, R.; Herichova, I. miRNA Clusters with Down-Regulated Expression in Human Colorectal Cancer and Their Regulation. Int. J. Mol. Sci. 2020, 21, 4633. [Google Scholar] [CrossRef]
  270. Yu, Q.; Liu, P.; Han, G.; Xue, X.; Ma, D. Expression of Concern: CircRNA circPDSS1 promotes bladder cancer by downregulating miR-16. Biosci. Rep. 2021, 41, BSR20191961. [Google Scholar] [CrossRef]
  271. Fredsoe, J.; Rasmussen, A.K.I.; Mouritzen, P.; Bjerre, M.T.; Ostergren, P.; Fode, M.; Borre, M.; Sorensen, K.D. Profiling of Circulating microRNAs in Prostate Cancer Reveals Diagnostic Biomarker Potential. Diagnostics 2020, 10, 188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  272. Ke, Y.; Zhao, W.; Xiong, J.; Cao, R. Downregulation of miR-16 promotes growth and motility by targeting HDGF in non-small cell lung cancer cells. FEBS Lett. 2013, 587, 3153–3157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Hu, S.; Wang, H.; Yan, D.; Lu, W.; Gao, P.; Lou, W.; Kong, X. Loss of miR-16 contributes to tumor progression by activation of tousled-like kinase 1 in oral squamous cell carcinoma. Cell Cycle 2018, 17, 2284–2295. [Google Scholar] [CrossRef] [Green Version]
  274. Manikandan, M.; Deva Magendhra Rao, A.K.; Arunkumar, G.; Manickavasagam, M.; Rajkumar, K.S.; Rajaraman, R.; Munirajan, A.K. Oral squamous cell carcinoma: microRNA expression profiling and integrative analyses for elucidation of tumourigenesis mechanism. Mol. Cancer 2016, 15, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Wang, X.; Li, G.H. MicroRNA-16 functions as a tumor-suppressor gene in oral squamous cell carcinoma by targeting AKT3 and BCL2L2. J. Cell Physiol. 2018, 233, 9447–9457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  276. Koopaie, M.; Manifar, S.; Lahiji, S.S. Assessment of MicroRNA-15a and MicroRNA-16-1 Salivary Level in Oral Squamous Cell Carcinoma Patients. Microrna 2021, 10, 74–79. [Google Scholar] [CrossRef] [PubMed]
  277. Liu, C.; Yu, Z.; Huang, S.; Zhao, Q.; Sun, Z.; Fletcher, C.; Jiang, Y.; Zhang, D. Combined identification of three miRNAs in serum as effective diagnostic biomarkers for HNSCC. EBioMedicine 2019, 50, 135–143. [Google Scholar] [CrossRef] [Green Version]
  278. Hu, Y.; Correa, A.M.; Hoque, A.; Guan, B.; Ye, F.; Huang, J.; Swisher, S.G.; Wu, T.T.; Ajani, J.A.; Xu, X.C. Prognostic significance of differentially expressed miRNAs in esophageal cancer. Int. J. Cancer 2011, 128, 132–143. [Google Scholar] [CrossRef]
  279. Zhu, Y.; Xia, Y.; Niu, H.; Chen, Y. MiR-16 induced the suppression of cell apoptosis while promote proliferation in esophageal squamous cell carcinoma. Cell Physiol. Biochem. 2014, 33, 1340–1348. [Google Scholar] [CrossRef]
  280. Jiang, H.; Zhang, G.; Wu, J.H.; Jiang, C.P. Diverse roles of miR-29 in cancer (review). Oncol. Rep. 2014, 31, 1509–1516. [Google Scholar] [CrossRef] [Green Version]
  281. Shin, J.; Shim, H.G.; Hwang, T.; Kim, H.; Kang, S.H.; Dho, Y.S.; Park, S.H.; Kim, S.J.; Park, C.K. Restoration of miR-29b exerts anti-cancer effects on glioblastoma. Cancer Cell Int. 2017, 17, 104. [Google Scholar] [CrossRef] [PubMed]
  282. Hozaka, Y.; Seki, N.; Tanaka, T.; Asai, S.; Moriya, S.; Idichi, T.; Wada, M.; Tanoue, K.; Kawasaki, Y.; Mataki, Y.; et al. Molecular Pathogenesis and Regulation of the miR-29-3p-Family: Involvement of ITGA6 and ITGB1 in Intra-Hepatic Cholangiocarcinoma. Cancers 2021, 13, 2804. [Google Scholar] [CrossRef] [PubMed]
  283. Nishikawa, R.; Goto, Y.; Kojima, S.; Enokida, H.; Chiyomaru, T.; Kinoshita, T.; Sakamoto, S.; Fuse, M.; Nakagawa, M.; Naya, Y.; et al. Tumor-suppressive microRNA-29s inhibit cancer cell migration and invasion via targeting LAMC1 in prostate cancer. Int. J. Oncol. 2014, 45, 401–410. [Google Scholar] [CrossRef] [Green Version]
  284. Feng, S.; Luo, S.; Ji, C.; Shi, J. miR-29c-3p regulates proliferation and migration in ovarian cancer by targeting KIF4A. World J. Surg. Oncol. 2020, 18, 315. [Google Scholar] [CrossRef]
  285. Li, L.; Shou, H.; Wang, Q.; Liu, S. Investigation of the potential theranostic role of KDM5B/miR-29c signaling axis in paclitaxel resistant endometrial carcinoma. Gene 2019, 694, 76–82. [Google Scholar] [CrossRef] [PubMed]
  286. Chen, B.; Wang, J.; Wang, J.; Wang, H.; Gu, X.; Tang, L.; Feng, X. A regulatory circuitry comprising TP53, miR-29 family, and SETDB1 in non-small cell lung cancer. Biosci. Rep. 2018, 38, 678. [Google Scholar] [CrossRef] [Green Version]
  287. Irani, S. miRNAs Signature in Head and Neck Squamous Cell Carcinoma Metastasis: A Literature Review. J. Dent. 2016, 17, 71–83. [Google Scholar]
  288. Sharma, S.; Pavlasova, G.M.; Seda, V.; Cerna, K.A.; Vojackova, E.; Filip, D.; Ondrisova, L.; Sandova, V.; Kostalova, L.; Zeni, P.F.; et al. miR-29 modulates CD40 signaling in chronic lymphocytic leukemia by targeting TRAF4: An axis affected by BCR inhibitors. Blood 2021, 137, 2481–2494. [Google Scholar] [CrossRef]
  289. Wang, X.; Zhong, H.; Wang, L.; Dong, Y.; Jia, A.; Mo, Q.; Zhang, C. MiR-29 Induces K562 Cell Apoptosis by Down-Regulating FoxM1. Med. Sci. Monit. 2015, 21, 3115–3120. [Google Scholar] [CrossRef] [Green Version]
  290. Piasecka, D.; Braun, M.; Kordek, R.; Sadej, R.; Romanska, H. MicroRNAs in regulation of triple-negative breast cancer progression. J. Cancer Res. Clin. Oncol. 2018, 144, 1401–1411. [Google Scholar] [CrossRef] [Green Version]
  291. Kinoshita, T.; Nohata, N.; Hanazawa, T.; Kikkawa, N.; Yamamoto, N.; Yoshino, H.; Itesako, T.; Enokida, H.; Nakagawa, M.; Okamoto, Y.; et al. Tumour-suppressive microRNA-29s inhibit cancer cell migration and invasion by targeting laminin-integrin signalling in head and neck squamous cell carcinoma. Br. J. Cancer 2013, 109, 2636–2645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  292. Koshizuka, K.; Kikkawa, N.; Hanazawa, T.; Yamada, Y.; Okato, A.; Arai, T.; Katada, K.; Okamoto, Y.; Seki, N. Inhibition of integrin beta1-mediated oncogenic signalling by the antitumor microRNA-29 family in head and neck squamous cell carcinoma. Oncotarget 2018, 9, 3663–3676. [Google Scholar] [CrossRef] [Green Version]
  293. Lu, E.; Su, J.; Zhou, Y.; Zhang, C.; Wang, Y. CCL20/CCR6 promotes cell proliferation and metastasis in laryngeal cancer by activating p38 pathway. Biomed. Pharmacother. 2017, 85, 486–492. [Google Scholar] [CrossRef] [PubMed]
  294. Liu, N.; Tang, L.L.; Sun, Y.; Cui, R.X.; Wang, H.Y.; Huang, B.J.; He, Q.M.; Jiang, W.; Ma, J. MiR-29c suppresses invasion and metastasis by targeting TIAM1 in nasopharyngeal carcinoma. Cancer Lett. 2013, 329, 181–188. [Google Scholar] [CrossRef]
  295. Jia, L.F.; Huang, Y.P.; Zheng, Y.F.; Lyu, M.Y.; Wei, S.B.; Meng, Z.; Gan, Y.H. miR-29b suppresses proliferation, migration, and invasion of tongue squamous cell carcinoma through PTEN-AKT signaling pathway by targeting Sp1. Oral Oncol. 2014, 50, 1062–1071. [Google Scholar] [CrossRef] [PubMed]
  296. Guo, Y.; Zhai, J.; Zhang, J.; Ni, C.; Zhou, H. Improved Radiotherapy Sensitivity of Nasopharyngeal Carcinoma Cells by miR-29-3p Targeting COL1A1 3′-UTR. Med. Sci. Monit. 2019, 25, 3161–3169. [Google Scholar] [CrossRef]
  297. Guo, D.; Jin, J.; Liu, J.; Dong, X.; Li, D.; He, Y. MicroRNA-29b regulates the radiosensitivity of esophageal squamous cell carcinoma by regulating the BTG2-mediated cell cycle. Strahlenther. Onkol. 2021, 197, 829–835. [Google Scholar] [CrossRef] [PubMed]
  298. Lucas Grzelczyk, W.; Szemraj, J.; Kwiatkowska, S.; Jozefowicz-Korczynska, M. Serum expression of selected miRNAs in patients with laryngeal squamous cell carcinoma (LSCC). Diagn. Pathol. 2019, 14, 49. [Google Scholar] [CrossRef]
  299. Al Rawi, N.; Elmabrouk, N.; Abu Kou, R.; Mkadmi, S.; Rizvi, Z.; Hamdoon, Z. The role of differentially expressed salivary microRNA in oral squamous cell carcinoma. A systematic review. Arch. Oral Biol. 2021, 125, 105108. [Google Scholar] [CrossRef]
  300. Cui, Y.; Xue, Y.; Dong, S.; Zhang, P. Plasma microRNA-9 as a diagnostic and prognostic biomarker in patients with esophageal squamous cell carcinoma. J. Int. Med. Res. 2017, 45, 1310–1317. [Google Scholar] [CrossRef]
  301. Bozinovic, K.; Sabol, I.; Dediol, E.; Milutin Gasperov, N.; Manojlovic, S.; Vojtechova, Z.; Tachezy, R.; Grce, M. Genome-wide miRNA profiling reinforces the importance of miR-9 in human papillomavirus associated oral and oropharyngeal head and neck cancer. Sci. Rep. 2019, 9, 2306. [Google Scholar] [CrossRef]
  302. Shen, Y.; Ding, Y.; Ma, Q.; Zhao, L.; Guo, X.; Shao, Y.; Niu, C.; He, Y.; Zhang, F.; Zheng, D.; et al. Identification of Novel Circulating miRNA Biomarkers for the Diagnosis of Esophageal Squamous Cell Carcinoma and Squamous Dysplasia. Cancer Epidemiol. Biomark. Prev. 2019, 28, 1212–1220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  303. Zhou, W.; Chang, A.; Zhao, H.; Ye, H.; Li, D.; Zhuo, X. Identification of a novel microRNA profile including miR-106b, miR-17, miR-20b, miR-18a and miR-93 in the metastasis of nasopharyngeal carcinoma. Cancer Biomark. 2020, 27, 533–539. [Google Scholar] [CrossRef] [PubMed]
  304. Xu, X.L.; Jiang, Y.H.; Feng, J.G.; Su, D.; Chen, P.C.; Mao, W.M. MicroRNA-17, microRNA-18a, and microRNA-19a are prognostic indicators in esophageal squamous cell carcinoma. Ann. Thorac. Surg. 2014, 97, 1037–1045. [Google Scholar] [CrossRef]
  305. Zhou, X.; Wen, W.; Zhu, J.; Huang, Z.; Zhang, L.; Zhang, H.; Qi, L.W.; Shan, X.; Wang, T.; Cheng, W.; et al. A six-microRNA signature in plasma was identified as a potential biomarker in diagnosis of esophageal squamous cell carcinoma. Oncotarget 2017, 8, 34468–34480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  306. Hirajima, S.; Komatsu, S.; Ichikawa, D.; Takeshita, H.; Konishi, H.; Shiozaki, A.; Morimura, R.; Tsujiura, M.; Nagata, H.; Kawaguchi, T.; et al. Clinical impact of circulating miR-18a in plasma of patients with oesophageal squamous cell carcinoma. Br. J. Cancer 2013, 108, 1822–1829. [Google Scholar] [CrossRef]
  307. Christopher, A.F.; Gupta, M.; Bansal, P. Micronome revealed miR-19a/b as key regulator of SOCS3 during cancer related inflammation of oral squamous cell carcinoma. Gene 2016, 594, 30–40. [Google Scholar] [CrossRef]
  308. Li, D.; Liu, K.; Li, Z.; Wang, J.; Wang, X. miR-19a and miR-424 target TGFBR3 to promote epithelial-to-mesenchymal transition and migration of tongue squamous cell carcinoma cells. Cell Adh. Migr. 2018, 12, 236–246. [Google Scholar] [CrossRef] [Green Version]
  309. Bai, Y.; Lin, H.; Fang, Z.; Luo, Q.; Fang, Y.; Su, Y.; Hu, Q.; Duan, H.; Chen, F.; Zhang, Z.Y. Plasma microRNA-19a as a potential biomarker for esophageal squamous cell carcinoma diagnosis and prognosis. Biomark. Med. 2017, 11, 431–441. [Google Scholar] [CrossRef] [Green Version]
  310. Wu, T.Y.; Zhang, T.H.; Qu, L.M.; Feng, J.P.; Tian, L.L.; Zhang, B.H.; Li, D.D.; Sun, Y.N.; Liu, M. MiR-19a is correlated with prognosis and apoptosis of laryngeal squamous cell carcinoma by regulating TIMP-2 expression. Int. J. Clin. Exp. Pathol. 2014, 7, 56–63. [Google Scholar]
  311. Zhang, Y.; Lu, W.; Chen, Y.; Lin, Y.; Yang, X.; Wang, H.; Liu, Z. The miR-19b-3p-MAP2K3-STAT3 feedback loop regulates cell proliferation and invasion in esophageal squamous cell carcinoma. Mol. Oncol. 2021, 15, 1566–1583. [Google Scholar] [CrossRef] [PubMed]
  312. Deng, Y.; Julaiti, A.; Ran, W.; He, Y. Bone marrow mesenchymal stem cells-derived exosomal microRNA-19b-3p targets SOCS1 to facilitate progression of esophageal cancer. Life Sci. 2021, 278, 119491. [Google Scholar] [CrossRef] [PubMed]
  313. Huang, T.; Yin, L.; Wu, J.; Gu, J.J.; Wu, J.Z.; Chen, D.; Yu, H.L.; Ding, K.; Zhang, N.; Du, M.Y.; et al. MicroRNA-19b-3p regulates nasopharyngeal carcinoma radiosensitivity by targeting TNFAIP3/NF-kappaB axis. J. Exp. Clin. Cancer Res. 2016, 35, 188. [Google Scholar] [CrossRef] [PubMed]
  314. Bian, L.H.; Duan, J.L.; Zhou, C.; Shen, G.W.; Wang, X.Y.; Yang, Y.; Zhang, X.L.; Xiao, S.J. MicroRNA19b inhibitors can attenuate the STAT3 signaling pathway in NPC C6661 cells. Mol. Med. Rep. 2020, 22, 51–56. [Google Scholar] [CrossRef]
  315. He, F.C.; Meng, W.W.; Qu, Y.H.; Zhou, M.X.; He, J.; Lv, P.; Ming, L. Expression of circulating microRNA-20a and let-7a in esophageal squamous cell carcinoma. World J. Gastroenterol. 2015, 21, 4660–4665. [Google Scholar] [CrossRef]
  316. Zhang, H.; Zou, X.; Wu, L.; Zhang, S.; Wang, T.; Liu, P.; Zhu, W.; Zhu, J. Identification of a 7-microRNA signature in plasma as promising biomarker for nasopharyngeal carcinoma detection. Cancer Med. 2020, 9, 1230–1241. [Google Scholar] [CrossRef]
  317. Zeng, X.; Xiang, J.; Wu, M.; Xiong, W.; Tang, H.; Deng, M.; Li, X.; Liao, Q.; Su, B.; Luo, Z.; et al. Circulating miR-17, miR-20a, miR-29c, and miR-223 combined as non-invasive biomarkers in nasopharyngeal carcinoma. PLoS ONE 2012, 7, e46367. [Google Scholar] [CrossRef]
  318. Li, B.; Kyung, H.M. Identification of eight meta-signature miRNAs as potential biomarkers for oropharyngeal cancers. Cancer Genet. 2019, 233–234, 75–83. [Google Scholar] [CrossRef]
  319. Nonaka, T.; Wong, D.T.W. Liquid Biopsy in Head and Neck Cancer: Promises and Challenges. J. Dent. Res. 2018, 97, 701–708. [Google Scholar] [CrossRef]
  320. Qiu, F.; Sun, R.; Deng, N.; Guo, T.; Cao, Y.; Yu, Y.; Wang, X.; Zou, B.; Zhang, S.; Jing, T.; et al. miR-29a/b enhances cell migration and invasion in nasopharyngeal carcinoma progression by regulating SPARC and COL3A1 gene expression. PLoS ONE 2015, 10, e0120969. [Google Scholar] [CrossRef] [Green Version]
  321. Wang, L.L.; Li, H.X.; Yang, Y.Y.; Su, Y.L.; Lian, J.S.; Li, T.; Xu, J.; Wang, X.N.; Jin, N.; Liu, X.F. MiR-31 is a potential biomarker for diagnosis of head and neck squamous cell carcinoma. Int. J. Clin. Exp. Pathol. 2018, 11, 4339–4345. [Google Scholar] [PubMed]
  322. Ansari, M.H.; Irani, S.; Edalat, H.; Amin, R.; Mohammadi Roushandeh, A. Deregulation of miR-93 and miR-143 in human esophageal cancer. Tumour Biol. 2016, 37, 3097–3103. [Google Scholar] [CrossRef] [PubMed]
  323. Chen, L.; Fan, X.; Wang, X.; Wu, X. Expression of miR-93-5p in patients with esophageal carcinoma and its relationship with the curative effect and prognosis of radiotherapy. Cell Mol. Biol. 2020, 66, 41–46. [Google Scholar] [CrossRef] [PubMed]
  324. Gao, R.; Wang, Z.; Liu, Q.; Yang, C. MicroRNA-105 plays an independent prognostic role in esophageal cancer and acts as an oncogene. Cancer Biomark. 2020, 27, 173–180. [Google Scholar] [CrossRef]
  325. Huang, G.J.; Yang, B.B. Identification of core miRNA prognostic markers in patients with laryngeal cancer using bioinformatics analysis. Eur. Arch. Oto-Rhino-Laryngol. 2021, 278, 1613–1626. [Google Scholar] [CrossRef]
  326. Yang, F.; Sun, Z.; Wang, D.; Du, T. MiR-106b-5p regulates esophageal squamous cell carcinoma progression by binding to HPGD. BMC Cancer 2022, 22, 308. [Google Scholar] [CrossRef]
  327. Wang, Y.; Zeng, G.; Jiang, Y. The Emerging Roles of miR-125b in Cancers. Cancer Manag. Res. 2020, 12, 1079–1088. [Google Scholar] [CrossRef] [Green Version]
  328. Mazumder, S.; Datta, S.; Ray, J.G.; Chaudhuri, K.; Chatterjee, R. Liquid biopsy: miRNA as a potential biomarker in oral cancer. Cancer Epidemiol. 2019, 58, 137–145. [Google Scholar] [CrossRef]
  329. Yin, X.; Wang, J.; Shan, C.; Jia, Q.; Bian, Y.; Zhang, H. Circular RNA ZNF609 promotes laryngeal squamous cell carcinoma progression by upregulating epidermal growth factor receptor via sponging microRNA-134-5p. Bioengineered 2022, 13, 6929–6941. [Google Scholar] [CrossRef]
  330. Liu, C.J.; Shen, W.G.; Peng, S.Y.; Cheng, H.W.; Kao, S.Y.; Lin, S.C.; Chang, K.W. miR-134 induces oncogenicity and metastasis in head and neck carcinoma through targeting WWOX gene. Int. J. Cancer 2014, 134, 811–821. [Google Scholar] [CrossRef]
  331. Summerer, I.; Unger, K.; Braselmann, H.; Schuettrumpf, L.; Maihoefer, C.; Baumeister, P.; Kirchner, T.; Niyazi, M.; Sage, E.; Specht, H.M.; et al. Circulating microRNAs as prognostic therapy biomarkers in head and neck cancer patients. Br. J. Cancer 2015, 113, 76–82. [Google Scholar] [CrossRef] [PubMed]
  332. Lin, R.J.; Xiao, D.W.; Liao, L.D.; Chen, T.; Xie, Z.F.; Huang, W.Z.; Wang, W.S.; Jiang, T.F.; Wu, B.L.; Li, E.M.; et al. MiR-142-3p as a potential prognostic biomarker for esophageal squamous cell carcinoma. J. Surg. Oncol. 2012, 105, 175–182. [Google Scholar] [CrossRef]
  333. Qi, X.; Li, J.; Zhou, C.; Lv, C.; Tian, M. MiR-142-3p Suppresses SOCS6 Expression and Promotes Cell Proliferation in Nasopharyngeal Carcinoma. Cell Physiol. Biochem. 2015, 36, 1743–1752. [Google Scholar] [CrossRef]
  334. Wang, T.; Wu, J.; Wu, Y.; Cheng, Y.; Deng, Y.; Liao, J.; Liu, H.; Peng, H. A novel microRNA-based signature predicts prognosis among nasopharyngeal cancer patients. Exp. Biol. Med. 2021, 246, 72–83. [Google Scholar] [CrossRef]
  335. Weiss, B.G.; Anczykowski, M.Z.; Ihler, F.; Bertlich, M.; Spiegel, J.L.; Haubner, F.; Canis, M.; Kuffer, S.; Hess, J.; Unger, K.; et al. MicroRNA-182-5p and microRNA-205-5p as potential biomarkers for prognostic stratification of p16-positive oropharyngeal squamous cell carcinoma. Cancer Biomark. 2022, 33, 331–347. [Google Scholar] [CrossRef]
  336. Wong, T.S.; Liu, X.B.; Wong, B.Y.; Ng, R.W.; Yuen, A.P.; Wei, W.I. Mature miR-184 as Potential Oncogenic microRNA of Squamous Cell Carcinoma of Tongue. Clin. Cancer Res. 2008, 14, 2588–2592. [Google Scholar] [CrossRef] [Green Version]
  337. Wong, T.S.; Ho, W.K.; Chan, J.Y.; Ng, R.W.; Wei, W.I. Mature miR-184 and squamous cell carcinoma of the tongue. Sci. World J. 2009, 9, 130–132. [Google Scholar] [CrossRef] [Green Version]
  338. Ghffari, M.; Asadi, M.; Shanaehbandi, D.; Bornehdeli, S.; Sadeghzadeh, M.; Mohammad Reza Khani, H.; Ghasembaglou, S. Aberrant Expression of miR-103, miR-184, miR-378, miR-497 and miR-506 in Tumor Tissue from Patients with Oral Squamous Cell Carcinoma Regulates the Clinical Picture of the Patients. Asian Pac. J. Cancer Prev. APJCP 2020, 21, 1311–1315. [Google Scholar] [CrossRef] [PubMed]
  339. Zahran, F.; Ghalwash, D.; Shaker, O.; Al-Johani, K.; Scully, C. Salivary microRNAs in oral cancer. Oral Dis. 2015, 21, 739–747. [Google Scholar] [CrossRef] [PubMed]
  340. Liu, C.J.; Lin, J.S.; Cheng, H.W.; Hsu, Y.H.; Cheng, C.Y.; Lin, S.C. Plasma miR-187* is a potential biomarker for oral carcinoma. Clin. Oral Investig. 2017, 21, 1131–1138. [Google Scholar] [CrossRef]
  341. Lin, S.C.; Kao, S.Y.; Chang, J.C.; Liu, Y.C.; Yu, E.H.; Tseng, S.H.; Liu, C.J.; Chang, K.W. Up-regulation of miR-187 modulates the advances of oral carcinoma by targeting BARX2 tumor suppressor. Oncotarget 2016, 7, 61355–61365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  342. Sun, G.; Cao, Y.; Wang, P.; Song, H.; Bie, T.; Li, M.; Huai, D. miR-200b-3p in plasma is a potential diagnostic biomarker in oral squamous cell carcinoma. Biomarkers 2018, 23, 137–141. [Google Scholar] [CrossRef] [PubMed]
  343. de Carvalho, A.C.; Scapulatempo-Neto, C.; Maia, D.C.; Evangelista, A.F.; Morini, M.A.; Carvalho, A.L.; Vettore, A.L. Accuracy of microRNAs as markers for the detection of neck lymph node metastases in patients with head and neck squamous cell carcinoma. BMC Med. 2015, 13, 108. [Google Scholar] [CrossRef] [Green Version]
  344. Zhou, Z.; Liu, C.; Liu, K.; Lv, M.; Li, B.; Lan, Z.; Chen, W.; Kang, M. Expression and Possible Molecular Mechanisms of microRNA-205-5p in Patients With Head and Neck Squamous Cell Carcinoma. Technol. Cancer Res. Treat. 2020, 19, 1533033820980110. [Google Scholar] [CrossRef] [PubMed]
  345. Fletcher, A.M.; Heaford, A.C.; Trask, D.K. Detection of metastatic head and neck squamous cell carcinoma using the relative expression of tissue-specific mir-205. Transl. Oncol. 2008, 1, 202–208. [Google Scholar] [CrossRef] [Green Version]
  346. Ibuki, Y.; Nishiyama, Y.; Tsutani, Y.; Emi, M.; Hamai, Y.; Okada, M.; Tahara, H. Circulating microRNA/isomiRs as novel biomarkers of esophageal squamous cell carcinoma. PLoS ONE 2020, 15, e0231116. [Google Scholar] [CrossRef]
  347. Hezova, R.; Kovarikova, A.; Srovnal, J.; Zemanova, M.; Harustiak, T.; Ehrmann, J.; Hajduch, M.; Sachlova, M.; Svoboda, M.; Slaby, O. MiR-205 functions as a tumor suppressor in adenocarcinoma and an oncogene in squamous cell carcinoma of esophagus. Tumour Biol. 2016, 37, 8007–8018. [Google Scholar] [CrossRef]
  348. Li, R.; Lu, C.; Yang, W.; Zhou, Y.; Zhong, J.; Chen, X.; Li, X.; Huang, G.; Peng, X.; Liu, K.; et al. A panel of three serum microRNA can be used as potential diagnostic biomarkers for nasopharyngeal carcinoma. J. Clin. Lab. Anal. 2022, 36, e24194. [Google Scholar] [CrossRef]
  349. Tachibana, H.; Sho, R.; Takeda, Y.; Zhang, X.; Yoshida, Y.; Narimatsu, H.; Otani, K.; Ishikawa, S.; Fukao, A.; Asao, H.; et al. Circulating miR-223 in Oral Cancer: Its Potential as a Novel Diagnostic Biomarker and Therapeutic Target. PLoS ONE 2016, 11, e0159693. [Google Scholar] [CrossRef]
  350. Jiang, L.; Lv, L.; Liu, X.; Jiang, X.; Yin, Q.; Hao, Y.; Xiao, L. MiR-223 promotes oral squamous cell carcinoma proliferation and migration by regulating FBXW7. Cancer Biomark. 2019, 24, 325–334. [Google Scholar] [CrossRef] [Green Version]
  351. Zou, X.; Zhu, D.; Zhang, H.; Zhang, S.; Zhou, X.; He, X.; Zhu, J.; Zhu, W. MicroRNA expression profiling analysis in serum for nasopharyngeal carcinoma diagnosis. Gene 2020, 727, 144243. [Google Scholar] [CrossRef] [PubMed]
  352. Huang, Z.; Zhang, L.; Zhu, D.; Shan, X.; Zhou, X.; Qi, L.W.; Wu, L.; Zhu, J.; Cheng, W.; Zhang, H.; et al. A novel serum microRNA signature to screen esophageal squamous cell carcinoma. Cancer Med. 2017, 6, 109–119. [Google Scholar] [CrossRef] [PubMed]
  353. Chen, C.; Zhao, J.; Liu, J.N.; Sun, C. Mechanism and Role of the Neuropeptide LGI1 Receptor ADAM23 in Regulating Biomarkers of Ferroptosis and Progression of Esophageal Cancer. Dis. Markers 2021, 2021, 9227897. [Google Scholar] [CrossRef]
  354. Tu, H.F.; Chang, K.W.; Cheng, H.W.; Liu, C.J. Upregulation of miR-372 and -373 associates with lymph node metastasis and poor prognosis of oral carcinomas. Laryngoscope 2015, 125, E365–E370. [Google Scholar] [CrossRef]
  355. Chang, Y.A.; Weng, S.L.; Yang, S.F.; Chou, C.H.; Huang, W.C.; Tu, S.J.; Chang, T.H.; Huang, C.N.; Jong, Y.J.; Huang, H.D. A Three-MicroRNA Signature as a Potential Biomarker for the Early Detection of Oral Cancer. Int. J. Mol. Sci. 2018, 19, 758. [Google Scholar] [CrossRef] [Green Version]
  356. Romani, C.; Salviato, E.; Paderno, A.; Zanotti, L.; Ravaggi, A.; Deganello, A.; Berretti, G.; Gualtieri, T.; Marchini, S.; D’Incalci, M.; et al. Genome-wide study of salivary miRNAs identifies miR-423-5p as promising diagnostic and prognostic biomarker in oral squamous cell carcinoma. Theranostics 2021, 11, 2987–2999. [Google Scholar] [CrossRef] [PubMed]
  357. Bolandparva, F.; Hashemi Nasab, M.S.; Mohamadnia, A.; Garajei, A.; Farhadi Nasab, A.; Bahrami, N. Early Diagnosis of Oral Squamous Cell Carcinoma (OSCC) by miR-138 and miR-424-5p Expression as a Cancer Marker. Asian Pac. J. Cancer Prev. APJCP 2021, 22, 2185–2189. [Google Scholar] [CrossRef]
  358. Xu, H.; Yang, Y.; Zhao, H.; Yang, X.; Luo, Y.; Ren, Y.; Liu, W.; Li, N. Serum miR-483-5p: A novel diagnostic and prognostic biomarker for patients with oral squamous cell carcinoma. Tumour Biol. 2016, 37, 447–453. [Google Scholar] [CrossRef]
  359. Xue, L.; Nan, J.; Dong, L.; Zhang, C.; Li, H.; Na, R.; He, H.; Wang, Y. Upregulated miR-483-5p expression as a prognostic biomarker for esophageal squamous cell carcinoma. Cancer Biomark. 2017, 19, 193–197. [Google Scholar] [CrossRef]
  360. Zhou, Y.; Hong, L. Prediction value of miR-483 and miR-214 in prognosis and multidrug resistance of esophageal squamous cell carcinoma. Genet. Test Mol. Biomark. 2013, 17, 470–474. [Google Scholar] [CrossRef] [Green Version]
  361. Wu, C.; Wang, C.; Guan, X.; Liu, Y.; Li, D.; Zhou, X.; Zhang, Y.; Chen, X.; Wang, J.; Zen, K.; et al. Diagnostic and prognostic implications of a serum miRNA panel in oesophageal squamous cell carcinoma. PLoS ONE 2014, 9, e92292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  362. Zheng, X.H.; Cui, C.; Ruan, H.L.; Xue, W.Q.; Zhang, S.D.; Hu, Y.Z.; Zhou, X.X.; Jia, W.H. Plasma microRNA profiling in nasopharyngeal carcinoma patients reveals miR-548q and miR-483-5p as potential biomarkers. Chin. J. Cancer 2014, 33, 330–338. [Google Scholar] [CrossRef]
  363. 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]
  364. Shi, J.; Bao, X.; Liu, Z.; Zhang, Z.; Chen, W.; Xu, Q. Serum miR-626 and miR-5100 are Promising Prognosis Predictors for Oral Squamous Cell Carcinoma. Theranostics 2019, 9, 920–931. [Google Scholar] [CrossRef]
  365. Cui, S.H.; Hu, X.D.; Yan, Y. Wnt/beta-catenin signaling pathway participates in the effect of miR-626 on oral squamous cell carcinoma by targeting RASSF4. J. Oral Pathol. Med. 2021, 50, 1005–1017. [Google Scholar] [CrossRef]
  366. Hufbauer, M.; Maltseva, M.; Meinrath, J.; Lechner, A.; Beutner, D.; Huebbers, C.U.; Akgul, B. HPV16 increases the number of migratory cancer stem cells and modulates their miRNA expression profile in oropharyngeal cancer. Int. J. Cancer 2018, 143, 1426–1439. [Google Scholar] [CrossRef] [Green Version]
  367. Ries, J.; Vairaktaris, E.; Agaimy, A.; Kintopp, R.; Baran, C.; Neukam, F.W.; Nkenke, E. miR-186, miR-3651 and miR-494: Potential biomarkers for oral squamous cell carcinoma extracted from whole blood. Oncol. Rep. 2014, 31, 1429–1436. [Google Scholar] [CrossRef] [Green Version]
  368. Ries, J.; Baran, C.; Wehrhan, F.; Weber, M.; Neukam, F.W.; Krautheim-Zenk, A.; Nkenke, E. Prognostic significance of altered miRNA expression in whole blood of OSCC patients. Oncol. Rep. 2017, 37, 3467–3474. [Google Scholar] [CrossRef] [Green Version]
  369. Luo, C.; Zhang, J.; Zhang, Y.; Zhang, X.; Chen, Y.; Fan, W. Low expression of miR-let-7a promotes cell growth and invasion through the regulation of c-Myc in oral squamous cell carcinoma. Cell Cycle 2020, 19, 1983–1993. [Google Scholar] [CrossRef] [PubMed]
  370. Pang, Y.; Liu, J.; Li, X.; Zhang, Y.; Zhang, B.; Zhang, J.; Du, N.; Xu, C.; Liang, R.; Ren, H.; et al. Nano Let7b sensitization of eliminating esophageal cancer stemlike cells is dependent on blockade of Wnt activation of symmetric division. Int. J. Oncol. 2017, 51, 1077–1088. [Google Scholar] [CrossRef] [Green Version]
  371. Xu, X.; Lu, J.; Wang, F.; Liu, X.; Peng, X.; Yu, B.; Zhao, F.; Li, X. Dynamic Changes in Plasma MicroRNAs Have Potential Predictive Values in Monitoring Recurrence and Metastasis of Nasopharyngeal Carcinoma. BioMed Res. Int. 2018, 2018, 7329195. [Google Scholar] [CrossRef]
  372. Minor, J.; Wang, X.; Zhang, F.; Song, J.; Jimeno, A.; Wang, X.-J.; Lu, X.; Gross, N.; Kulesz-Martin, M.; Wang, D.; et al. Methylation of microRNA-9 is a specific and sensitive biomarker for oral and oropharyngeal squamous cell carcinomas. Oral Oncol. 2012, 48, 73–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  373. Hersi, H.M.; Raulf, N.; Gaken, J.; Folarin, N.; Tavassoli, M. MicroRNA-9 inhibits growth and invasion of head and neck cancer cells and is a predictive biomarker of response to plerixafor, an inhibitor of its target CXCR4. Mol. Oncol. 2018, 12, 2023–2041. [Google Scholar] [CrossRef]
  374. Wang, S.; Pan, Y.; Zhang, R.; Xu, T.; Wu, W.; Zhang, R.; Wang, C.; Huang, H.; Calin, C.A.; Yang, H.; et al. Hsa-miR-24-3p increases nasopharyngeal carcinoma radiosensitivity by targeting both the 3′UTR and 5′UTR of Jab1/CSN5. Oncogene 2016, 35, 6096–6108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  375. Yang, C.; Zheng, S.; Liu, T.; Liu, Q.; Dai, F.; Zhou, J.; Chen, Y.; Sheyhidin, I.; Lu, X. Down-regulated miR-26a promotes proliferation, migration, and invasion via negative regulation of MTDH in esophageal squamous cell carcinoma. FASEB J. 2017, 31, 2114–2122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  376. Jia, L.F.; Wei, S.B.; Gan, Y.H.; Guo, Y.; Gong, K.; Mitchelson, K.; Cheng, J.; Yu, G.Y. Expression, regulation and roles of miR-26a and MEG3 in tongue squamous cell carcinoma. Int. J. Cancer 2014, 135, 2282–2293. [Google Scholar] [CrossRef]
  377. Li, J.; Liang, Y.; Lv, H.; Meng, H.; Xiong, G.; Guan, X.; Chen, X.; Bai, Y.; Wang, K. miR-26a and miR-26b inhibit esophageal squamous cancer cell proliferation through suppression of c-MYC pathway. Gene 2017, 625, 1–9. [Google Scholar] [CrossRef]
  378. Fukumoto, I.; Hanazawa, T.; Kinoshita, T.; Kikkawa, N.; Koshizuka, K.; Goto, Y.; Nishikawa, R.; Chiyomaru, T.; Enokida, H.; Nakagawa, M.; et al. MicroRNA expression signature of oral squamous cell carcinoma: Functional role of microRNA-26a/b in the modulation of novel cancer pathways. Br. J. Cancer 2015, 112, 891–900. [Google Scholar] [CrossRef] [Green Version]
  379. Wei, Z.; Chang, K.; Fan, C.; Zhang, Y. MiR-26a/miR-26b represses tongue squamous cell carcinoma progression by targeting PAK1. Cancer Cell Int. 2020, 20, 82. [Google Scholar] [CrossRef]
  380. Syllaios, A.; Sakellariou, S.; Garmpis, N.; Sarlani, E.; Damaskos, C.; Apostolou, K.; Kykalos, S.; Gazouli, M.; Karavokyros, I.; Schizas, D. The role of miR-101 in esophageal and gastric cancer. Per. Med. 2021, 18, 491–499. [Google Scholar] [CrossRef]
  381. Chiam, K.; Mayne, G.C.; Watson, D.I.; Woodman, R.J.; Bright, T.F.; Michael, M.Z.; Karapetis, C.S.; Irvine, T.; Phillips, W.A.; Hummel, R.; et al. Identification of microRNA Biomarkers of Response to Neoadjuvant Chemoradiotherapy in Esophageal Adenocarcinoma Using Next Generation Sequencing. Ann. Surg. Oncol. 2018, 25, 2731–2738. [Google Scholar] [CrossRef]
  382. Wang, Y.; Jia, R.Z.; Diao, S.; He, J.; Jia, L. miRNA-101 Targets TGF-betaR1 to Retard the Progression of Oral Squamous Cell Carcinoma. Oncol. Res. 2020, 28, 203–212. [Google Scholar] [CrossRef] [PubMed]
  383. Zhou, R.S.; Zhang, E.X.; Sun, Q.F.; Ye, Z.J.; Liu, J.W.; Zhou, D.H.; Tang, Y. Integrated analysis of lncRNA-miRNA-mRNA ceRNA network in squamous cell carcinoma of tongue. BMC Cancer 2019, 19, 779. [Google Scholar] [CrossRef]
  384. Tian, Z.; Li, Z.; Zhu, Y.; Meng, L.; Liu, F.; Sang, M.; Wang, G. Hypermethylation-mediated inactivation of miR-124 predicts poor prognosis and promotes tumor growth at least partially through targeting EZH2/H3K27me3 in ESCC. Clin. Exp. Metastasis 2019, 36, 381–391. [Google Scholar] [CrossRef] [PubMed]
  385. Zhang, Y.H.; Wang, Q.Q.; Li, H.; Ye, T.; Gao, F.; Liu, Y.C. miR-124 radiosensitizes human esophageal cancer cell TE-1 by targeting CDK4. Genet. Mol. Res. 2016, 15, 15027893. [Google Scholar] [CrossRef]
  386. Inoue, H.; Hirasaki, M.; Kogashiwa, Y.; Kuba, K.; Ebihara, Y.; Nakahira, M.; Sakai, A.; Okuda, A.; Sugasawa, M. Predicting the radiosensitivity of HPV-negative oropharyngeal squamous cell carcinoma using miR-130b. Acta Oto-Laryngol. 2021, 141, 640–645. [Google Scholar] [CrossRef]
  387. Duz, M.B.; Karatas, O.F.; Guzel, E.; Turgut, N.F.; Yilmaz, M.; Creighton, C.J.; Ozen, M. Identification of miR-139-5p as a saliva biomarker for tongue squamous cell carcinoma: A pilot study. Cell. Oncol. 2016, 39, 187–193. [Google Scholar] [CrossRef]
  388. Jiang, Q.; Cao, Y.; Qiu, Y.; Li, C.; Liu, L.; Xu, G. Progression of squamous cell carcinoma is regulated by miR-139-5p/CXCR4. Front. Biosci. Landmark Ed. 2020, 25, 1732–1745. [Google Scholar] [CrossRef] [PubMed]
  389. Wang, K.; Jin, J.; Ma, T.; Zhai, H. MiR-139-5p inhibits the tumorigenesis and progression of oral squamous carcinoma cells by targeting HOXA9. J. Cell. Mol. Med. 2017, 21, 3730–3740. [Google Scholar] [CrossRef] [PubMed]
  390. Gao, S.; Zhao, Z.Y.; Zhang, Z.Y.; Zhang, Y.; Wu, R. Prognostic Value of MicroRNAs in Esophageal Carcinoma: A Meta-Analysis. Clin. Transl. Gastroenterol. 2018, 9, 203. [Google Scholar] [CrossRef]
  391. Zheng, S.; Zhang, X.; Wang, X.; Li, J. Downregulation of miR-138 predicts poor prognosis in patients with esophageal squamous cell carcinoma. Cancer Biomark. 2017, 20, 49–54. [Google Scholar] [CrossRef] [PubMed]
  392. Yokobori, T.; Suzuki, S.; Tanaka, N.; Inose, T.; Sohda, M.; Sano, A.; Sakai, M.; Nakajima, M.; Miyazaki, T.; Kato, H.; et al. MiR-150 is associated with poor prognosis in esophageal squamous cell carcinoma via targeting the EMT inducer ZEB1. Cancer Sci. 2013, 104, 48–54. [Google Scholar] [CrossRef]
  393. Zhang, S.; Yue, W.; Xie, Y.; Liu, L.; Li, S.; Dang, W.; Xin, S.; Yang, L.; Zhai, X.; Cao, P.; et al. The fourmicroRNA signature identified by bioinformatics analysis predicts the prognosis of nasopharyngeal carcinoma patients. Oncol. Rep. 2019, 42, 1767–1780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  394. Yue, P.Y.; Ha, W.Y.; Lau, C.C.; Cheung, F.M.; Lee, A.W.; Ng, W.T.; Ngan, R.K.; Yau, C.C.; Kwong, D.L.; Lung, H.L.; et al. MicroRNA profiling study reveals miR-150 in association with metastasis in nasopharyngeal carcinoma. Sci. Rep. 2017, 7, 12012. [Google Scholar] [CrossRef] [Green Version]
  395. Li, G.H.; Ma, Z.H.; Wang, X. Long non-coding RNA CCAT1 is a prognostic biomarker for the progression of oral squamous cell carcinoma via miR-181a-mediated Wnt/beta-catenin signaling pathway. Cell Cycle 2019, 18, 2902–2913. [Google Scholar] [CrossRef] [PubMed]
  396. Wang, H.T.; Tong, X.; Zhang, Z.X.; Sun, Y.Y.; Yan, W.; Xu, Z.M.; Fu, W.N. MYCT1 represses apoptosis of laryngeal cancerous cells through the MAX/miR-181a/NPM1 pathway. FEBS J. 2019, 286, 3892–3908. [Google Scholar] [CrossRef]
  397. Sun, N.; Ye, L.; Chang, T.; Li, X.; Li, X. microRNA-195-Cdc42 axis acts as a prognostic factor of esophageal squamous cell carcinoma. Int. J. Clin. Exp. Pathol. 2014, 7, 6871–6879. [Google Scholar] [PubMed]
  398. Li, C.Y.; Zhang, W.W.; Xiang, J.L.; Wang, X.H.; Li, J.; Wang, J.L. Identification of microRNAs as novel biomarkers for esophageal squamous cell carcinoma: A study based on The Cancer Genome Atlas (TCGA) and bioinformatics. Chin. Med. J. 2019, 132, 2213–2222. [Google Scholar] [CrossRef]
  399. He, R.; Wang, J.; Ye, K.; Du, J.; Chen, J.; Liu, W. Reduced miR-203 predicts metastasis and poor survival in esophageal carcinoma. Aging 2019, 11, 12114–12130. [Google Scholar] [CrossRef]
  400. Lin, J.; Lin, Y.; Fan, L.; Kuang, W.; Zheng, L.; Wu, J.; Shang, P.; Wang, Q.; Tan, J. miR-203 inhibits cell proliferation and promotes cisplatin induced cell death in tongue squamous cancer. Biochem. Biophys. Res. Commun. 2016, 473, 382–387. [Google Scholar] [CrossRef]
  401. Qu, J.Q.; Yi, H.M.; Ye, X.; Zhu, J.F.; Yi, H.; Li, L.N.; Xiao, T.; Yuan, L.; Li, J.Y.; Wang, Y.Y.; et al. MiRNA-203 Reduces Nasopharyngeal Carcinoma Radioresistance by Targeting IL8/AKT Signaling. Mol. Cancer Ther. 2015, 14, 2653–2664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  402. Zhuang, Z.; Yu, P.; Xie, N.; Wu, Y.; Liu, H.; Zhang, M.; Tao, Y.; Wang, W.; Yin, H.; Zou, B.; et al. MicroRNA-204-5p is a tumor suppressor and potential therapeutic target in head and neck squamous cell carcinoma. Theranostics 2020, 10, 1433–1453. [Google Scholar] [CrossRef]
  403. Wang, X.; Li, F.; Zhou, X. miR-204-5p regulates cell proliferation and metastasis through inhibiting CXCR4 expression in OSCC. Biomed. Pharmacother. 2016, 82, 202–207. [Google Scholar] [CrossRef] [PubMed]
  404. Gao, W.; Wu, Y.; He, X.; Zhang, C.; Zhu, M.; Chen, B.; Liu, Q.; Qu, X.; Li, W.; Wen, S.; et al. MicroRNA-204-5p inhibits invasion and metastasis of laryngeal squamous cell carcinoma by suppressing forkhead box C1. J. Cancer 2017, 8, 2356–2368. [Google Scholar] [CrossRef]
  405. Xu, H.; Yao, Y.; Meng, F.; Qian, X.; Jiang, X.; Li, X.; Gao, Z.; Gao, L. Predictive Value of Serum miR-10b, miR-29c, and miR-205 as Promising Biomarkers in Esophageal Squamous Cell Carcinoma Screening. Medicine 2015, 94, e1558. [Google Scholar] [CrossRef]
  406. Harris, T.; Jimenez, L.; Kawachi, N.; Fan, J.-B.; Chen, J.; Belbin, T.; Ramnauth, A.; Loudig, O.; Keller, C.E.; Smith, R.; et al. Low-level expression of miR-375 correlates with poor outcome and metastasis while altering the invasive properties of head and neck squamous cell carcinomas. Am. J. Pathol. 2012, 180, 917–928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  407. Salazar, C.; Calvopina, D.; Punyadeera, C. miRNAs in human papilloma virus associated oral and oropharyngeal squamous cell carcinomas. Expert Rev. Mol. Diagn. 2014, 14, 1033–1040. [Google Scholar] [CrossRef]
  408. Faur, C.I.; Rotaru, H.; Osan, C.; Jurj, A.; Roman, R.C.; Moldovan, M.; Chirila, M.; Hedesiu, M. Salivary exosomal microRNAs as biomarkers for head and neck cancer detection-a literature review. Maxillofac. Plast. Reconstr. Surg. 2021, 43, 19. [Google Scholar] [CrossRef] [PubMed]
  409. Garajei, A.; Parvin, M.; Mohammadi, H.; Allameh, A.; Hamidavi, A.; Sadeghi, M.; Emami, A.; Brand, S. Evaluation of the Expression of miR-486-3p, miR-548-3p, miR-561-5p and miR-509-5p in Tumor Biopsies of Patients with Oral Squamous Cell Carcinoma. Pathogens 2022, 11, 211. [Google Scholar] [CrossRef]
  410. Ren, C.; Chen, H.; Han, C.; Fu, D.; Zhou, L.; Jin, G.; Wang, F.; Wang, D.; Chen, Y.; Ma, L.; et al. miR-486-5p expression pattern in esophageal squamous cell carcinoma, gastric cancer and its prognostic value. Oncotarget 2016, 7, 15840–15853. [Google Scholar] [CrossRef]
  411. Wang, C.; Guan, S.; Chen, X.; Liu, B.; Liu, F.; Han, L.; Un Nesa, E.; Song, Q.; Bao, C.; Wang, X.; et al. Clinical potential of miR-3651 as a novel prognostic biomarker for esophageal squamous cell cancer. Biochem. Biophys. Res. Commun. 2015, 465, 30–34. [Google Scholar] [CrossRef] [PubMed]
  412. Ries, J.; Baran, C.; Wehrhan, F.; Weber, M.; Motel, C.; Kesting, M.; Nkenke, E. The altered expression levels of miR-186, miR-494 and miR-3651 in OSCC tissue vary from those of the whole blood of OSCC patients. Cancer Biomark. 2019, 24, 19–30. [Google Scholar] [CrossRef] [PubMed]
  413. Ge, X.; Gao, J.; Sun, Q.W.; Wang, C.X.; Deng, W.; Mao, G.Y.; Li, H.Q.; Guo, S.S.; Cheng, J.; Wu, Y.N.; et al. MiR-34a inhibits the proliferation, migration, and invasion of oral squamous cell carcinoma by directly targeting SATB2. J. Cell. Physiol. 2020, 235, 4856–4864. [Google Scholar] [CrossRef] [PubMed]
  414. Lei, W.; Liu, Y.E.; Zheng, Y.; Qu, L. MiR-429 inhibits oral squamous cell carcinoma growth by targeting ZEB1. Med. Sci. Monit. 2015, 21, 383–389. [Google Scholar] [CrossRef] [Green Version]
  415. Xu, M.; Zhan, J.; Xie, J.; Zhu, L.; Chen, L.; Luo, X.; Sheng, X.; Liu, T.; Zhang, S.; Lu, Z. MiR-125a-5p inhibits cell proliferation, cell cycle progression, and migration while promoting apoptosis in head and neck cancers by targeting ERBB3. Auris Nasus Larynx 2021, 48, 477–486. [Google Scholar] [CrossRef] [PubMed]
  416. Jin, S.; Liu, M.D.; Wu, H.; Pang, P.; Wang, S.; Li, Z.N.; Sun, C.F.; Liu, F.Y. Overexpression of hsa-miR-125a-5p enhances proliferation, migration and invasion of head and neck squamous cell carcinoma cell lines by upregulating C-C chemokine receptor type 7. Oncol. Lett. 2018, 15, 9703–9710. [Google Scholar] [CrossRef] [Green Version]
  417. Zhu, F.Y.; Gan, C.W.; Wang, M.X.; Sun, B.C.; Li, F.J.; Qiu, Y.H.; Wang, K. MiR-146a-5p inhibits proliferation and promotes apoptosis of oral squamous cell carcinoma cells by regulating NF-kappaB signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 3717–3723. [Google Scholar] [CrossRef]
  418. Chang, H.Y.; Lee, C.H.; Li, Y.S.; Huang, J.T.; Lan, S.H.; Wang, Y.F.; Lai, W.W.; Wang, Y.C.; Lin, Y.J.; Liu, H.S.; et al. MicroRNA-146a suppresses tumor malignancy via targeting vimentin in esophageal squamous cell carcinoma cells with lower fibronectin membrane assembly. J. Biomed. Sci. 2020, 27, 102. [Google Scholar] [CrossRef]
  419. Fukumoto, I.; Kikkawa, N.; Matsushita, R.; Kato, M.; Kurozumi, A.; Nishikawa, R.; Goto, Y.; Koshizuka, K.; Hanazawa, T.; Enokida, H.; et al. Tumor-suppressive microRNAs (miR-26a/b, miR-29a/b/c and miR-218) concertedly suppressed metastasis-promoting LOXL2 in head and neck squamous cell carcinoma. J. Hum. Genet 2016, 61, 109–118. [Google Scholar] [CrossRef]
  420. Chen, L.H.; Hsu, W.L.; Tseng, Y.J.; Liu, D.W.; Weng, C.F. Involvement of DNMT 3B promotes epithelial-mesenchymal transition and gene expression profile of invasive head and neck squamous cell carcinomas cell lines. BMC Cancer 2016, 16, 431. [Google Scholar] [CrossRef] [Green Version]
  421. Huang, C.K.; Bär, C.; Thum, T. miR-21, Mediator, and Potential Therapeutic Target in the Cardiorenal Syndrome. Front. Pharmacol. 2020, 11, 726. [Google Scholar] [CrossRef] [PubMed]
  422. Miyahara, N.; Benazzo, A.; Oberndorfer, F.; Iwasaki, A.; Laszlo, V.; Döme, B.; Hoda, M.A.; Jaksch, P.; Klepetko, W.; Hoetzenecker, K. MiR-21 in Lung Transplant Recipients With Chronic Lung Allograft Dysfunction. Transpl. Int. 2021, 35, 10184. [Google Scholar] [CrossRef] [PubMed]
  423. Wang, L.; Yin, Z.; Wang, F.; Han, Z.; Wang, Y.; Huang, S.; Hu, T.; Guo, M.; Lei, P. Hydrogen exerts neuroprotection by activation of the miR-21/PI3K/AKT/GSK-3β pathway in an in vitro model of traumatic brain injury. J. Cell. Mol. Med. 2020, 24, 4061–4071. [Google Scholar] [CrossRef] [Green Version]
  424. Wang, H.; Bei, Y.; Shen, S.; Huang, P.; Shi, J.; Zhang, J.; Sun, Q.; Chen, Y.; Yang, Y.; Xu, T.; et al. miR-21-3p controls sepsis-associated cardiac dysfunction via regulating SORBS2. J. Mol. Cell. Cardiol. 2016, 94, 43–53. [Google Scholar] [CrossRef] [PubMed]
  425. Zhang, J.; Liu, Y.; Liu, L. Hyperoside prevents sepsis-associated cardiac dysfunction through regulating cardiomyocyte viability and inflammation via inhibiting miR-21. Biomed. Pharmacother. 2021, 138, 111524. [Google Scholar] [CrossRef]
  426. Hu, A.; Huang, J.J.; Xu, W.H.; Jin, X.J.; Li, J.P.; Tang, Y.J.; Huang, X.F.; Cui, H.J.; Sun, G.B.; Li, R.L.; et al. MiR-21/miR-375 ratio is an independent prognostic factor in patients with laryngeal squamous cell carcinoma. Am. J. Cancer Res. 2015, 5, 1775–1785. [Google Scholar]
  427. Zhao, X.; Cui, L. A robust six-miRNA prognostic signature for head and neck squamous cell carcinoma. J. Cell. Physiol. 2020, 235, 8799–8811. [Google Scholar] [CrossRef]
  428. Shi, H.; Chen, J.; Li, Y.; Li, G.; Zhong, R.; Du, D.; Meng, R.; Kong, W.; Lu, M. Identification of a six microRNA signature as a novel potential prognostic biomarker in patients with head and neck squamous cell carcinoma. Oncotarget 2016, 7, 21579–21590. [Google Scholar] [CrossRef]
  429. Ganci, F.; Sacconi, A.; Manciocco, V.; Covello, R.; Benevolo, M.; Rollo, F.; Strano, S.; Valsoni, S. Altered peritumoral microRNA expression predicts head and neck cancer patients with a high risk of recurrence. Mod. Pathol. 2017, 30, 1387–1401. [Google Scholar] [CrossRef] [Green Version]
  430. Hess, J.; Unger, K.; Maihoefer, C. A Five-MicroRNA Signature Predicts Survival and Disease Control of Patients with Head and Neck Cancer Negative for HPV Infection. Clin. Cancer Res. 2019, 25, 1505–1516. [Google Scholar] [CrossRef] [Green Version]
  431. Lu, L.; Wu, Y.; Feng, M.; Xue, X.; Fan, Y. A novel seven-miRNA prognostic model to predict overall survival in head and neck squamous cell carcinoma patients. Mol. Med. Rep. 2019, 20, 4340–4348. [Google Scholar] [CrossRef] [PubMed]
  432. Liu, N.; Boohaker, R.J.; Jiang, C.; Boohaker, J.R.; Xu, B. A radiosensitivity MiRNA signature validated by the TCGA database for head and neck squamous cell carcinomas. Oncotarget 2015, 6, 34649–34657. [Google Scholar] [CrossRef] [Green Version]
  433. Chen, L.; Wen, Y.; Zhang, J.; Sun, W.; Lui, V.W.Y.; Wei, Y.; Chen, F.; Wen, W. Prediction of radiotherapy response with a 5-microRNA signature-based nomogram in head and neck squamous cell carcinoma. Cancer Med. 2018, 7, 726–735. [Google Scholar] [CrossRef] [PubMed]
  434. Yap, T.; Koo, K.; Cheng, L.; Vella, L.J.; Hill, A.F.; Reynolds, E.; Nastri, A.; Cirillo, N.; Seers, C. Predicting the Presence of Oral Squamous Cell Carcinoma Using Commonly Dysregulated MicroRNA in Oral Swirls. Cancer Prev. Res. 2018, 11, 491–502. [Google Scholar] [CrossRef] [Green Version]
  435. Peng, S.C.; Liao, C.T.; Peng, C.H.; Cheng, A.J.; Chen, S.J.; Huang, C.G.; Hsieh, W.P.; Yen, T.C. MicroRNAs MiR-218, MiR-125b, and Let-7g predict prognosis in patients with oral cavity squamous cell carcinoma. PLoS ONE 2014, 9, e102403. [Google Scholar] [CrossRef] [PubMed]
  436. Yoon, A.J.; Wang, S.; Kutler, D.I.; Carvajal, R.D.; Philipone, E.; Wang, T.; Peters, S.M.; LaRoche, D.; Hernandez, B.Y.; McDowell, B.D.; et al. MicroRNA-based risk scoring system to identify early-stage oral squamous cell carcinoma patients at high-risk for cancer-specific mortality. Head Neck 2020, 42, 1699–1712. [Google Scholar] [CrossRef] [PubMed]
  437. Cervigne, N.K.; Reis, P.P.; Machado, J.; Sadikovic, B.; Bradley, G.; Galloni, N.N.; Pintilie, M.; Jurisica, I.; Perez-Ordonez, B.; Gilbert, R.; et al. Identification of a microRNA signature associated with progression of leukoplakia to oral carcinoma. Hum. Mol. Genet. 2009, 18, 4818–4829. [Google Scholar] [CrossRef] [Green Version]
  438. Gao, G.; Gay, H.A.; Chernock, R.D.; Zhang, T.R.; Luo, J.; Thorstad, W.L.; Lewis, J.S., Jr.; Wang, X. A microRNA expression signature for the prognosis of oropharyngeal squamous cell carcinoma. Cancer 2013, 119, 72–80. [Google Scholar] [CrossRef]
  439. Liu, X.; Liu, P.; Chernock, R.D.; Yang, Z.; Lang Kuhs, K.A.; Lewis, J.S.; Luo, J. A MicroRNA Expression Signature as Prognostic Marker for Oropharyngeal Squamous Cell Carcinoma. Cancer 2021, 113, 752–759. [Google Scholar] [CrossRef]
  440. Hui, A.B.; Lin, A.; Xu, W.; Waldron, L.; Perez-Ordonez, B.; Weinreb, I.; Shi, W.; Bruce, J.; Huang, S.H.; O’Sullivan, B.; et al. Potentially prognostic miRNAs in HPV-associated oropharyngeal carcinoma. Clin. Cancer Res. 2013, 19, 2154–2162. [Google Scholar] [CrossRef] [Green Version]
  441. Xu, X.; Lu, Z.; Gross, N.; Li, G. A 3-miRNA signature predicts survival of patients with hypopharyngeal squamous cell carcinoma after post-operative radiotherapy. J. Cell. Mol. Med. 2019, 23, 8280–8291. [Google Scholar] [CrossRef] [Green Version]
  442. Zhang, S.Q.; Liu, J.; Chen, H.B.; Dai, W.J.; Zhou, L.Q.; Xie, C.W.; Li, J.C. A novel three-microRNA signature for predicting survival in patients with nasopharyngeal carcinoma. J. Dent. Sci. 2022, 17, 377–388. [Google Scholar] [CrossRef] [PubMed]
  443. Zhou, J.; Zhang, B.; Zhang, X.; Wang, C.; Xu, Y. Identification of a 3-miRNA Signature Associated With the Prediction of Prognosis in Nasopharyngeal Carcinoma. Front. Oncol. 2021, 11, 823603. [Google Scholar] [CrossRef] [PubMed]
  444. Kreimer, A.R.; Clifford, G.M.; Boyle, P.; Franceschi, S. Human papillomavirus types in head and neck squamous cell carcinomas worldwide: A systematic review. Cancer Epidemiol. Biomark. Prev. 2005, 14, 467–475. [Google Scholar] [CrossRef] [Green Version]
  445. Brakenhoff, R.H.; Wagner, S.; Klussmann, J.P. Molecular Patterns and Biology of HPV-Associated HNSCC. In HPV Infection in Head and Neck Cancer; Golusiński, W., Leemans, C.R., Dietz, A., Eds.; Springer International Publishing: Cham, Switzerland, 2017; pp. 37–56. [Google Scholar]
  446. Luo, X.J.; Zheng, M.; Cao, M.X.; Zhang, W.L.; Huang, M.C.; Dai, L.; Tang, Y.L.; Liang, X.H. Distinguishable Prognostic miRNA Signatures of Head and Neck Squamous Cell Cancer With or Without HPV Infection. Front. Oncol. 2020, 10, 614487. [Google Scholar] [CrossRef] [PubMed]
  447. Näsman, A.; Holzhauser, S.; Kostopoulou, O.N.; Zupancic, M.; Ährlund-Richter, A.; Du, J.; Dalianis, T. Prognostic Markers and Driver Genes and Options for Targeted Therapy in Human-Papillomavirus-Positive Tonsillar and Base-of-Tongue Squamous Cell Carcinoma. Viruses 2021, 13, 910. [Google Scholar] [CrossRef]
  448. Lajer, C.B.; Garnæs, E.; Friis-Hansen, L.; Norrild, B.; Therkildsen, M.H.; Glud, M.; Rossing, M.; Lajer, H.; Svane, D.; Skotte, L.; et al. Erratum: The role of miRNAs in human papilloma virus (HPV)-associated cancers: Bridging between HPV-related head and neck cancer and cervical cancer. Br. J. Cancer 2017, 117, e2. [Google Scholar] [CrossRef] [Green Version]
  449. Wald, A.I.; Hoskins, E.E.; Wells, S.I.; Ferris, R.L.; Khan, S.A. Alteration of microRNA profiles in squamous cell carcinoma of the head and neck cell lines by human papillomavirus. Head Neck 2011, 33, 504–512. [Google Scholar] [CrossRef] [Green Version]
  450. 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]
  451. Vojtechova, Z.; Sabol, I.; Salakova, M.; Smahelova, J.; Zavadil, J.; Turek, L.; Grega, M.; Klozar, J.; Prochazka, B.; Tachezy, R. Comparison of the miRNA profiles in HPV-positive and HPV-negative tonsillar tumors and a model system of human keratinocyte clones. BMC Cancer 2016, 16, 382. [Google Scholar] [CrossRef] [Green Version]
  452. Zhang, Q.; Chen, Y.; Hu, S.Q.; Pu, Y.M.; Zhang, K.; Wang, Y.X. A HPV16-related prognostic indicator for head and neck squamous cell carcinoma. Ann. Transl. Med. 2020, 8, 1492. [Google Scholar] [CrossRef]
  453. Wang, B.; Zhang, S. HPV+ HNSCC-derived exosomal miR-9-5p inhibits TGF-β signaling-mediated fibroblast phenotypic transformation through NOX4. Cancer Sci. 2022, 113, 1475–1487. [Google Scholar] [CrossRef] [PubMed]
  454. Miller, D.L.; Davis, J.W.; Taylor, K.H.; Johnson, J.; Shi, Z.; Williams, R.; Atasoy, U.; Lewis, J.S., Jr.; Stack, M.S. Identification of a human papillomavirus-associated oncogenic miRNA panel in human oropharyngeal squamous cell carcinoma validated by bioinformatics analysis of the Cancer Genome Atlas. Am. J. Pathol. 2015, 185, 679–692. [Google Scholar] [CrossRef] [Green Version]
  455. 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] [PubMed]
  456. Mirghani, H.; Ugolin, N.; Ory, C.; Goislard, M.; Lefevre, M.; Baulande, S.; Hofman, P.; Guily, J.L.; Chevillard, S.; Lacave, R. Comparative analysis of micro-RNAs in human papillomavirus-positive versus -negative oropharyngeal cancers. Head Neck 2016, 38, 1634–1642. [Google Scholar] [CrossRef] [PubMed]
  457. Peacock, B.; Rigby, A.; Bradford, J.; Pink, R.; Hunter, K.; Lambert, D.; Hunt, S. Extracellular vesicle microRNA cargo is correlated with HPV status in oropharyngeal carcinoma. J. Oral Pathol. Med. 2018, 47, 954–963. [Google Scholar] [CrossRef]
  458. 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]
  459. MicroRNA Markers in Head and Neck Cancers. Available online: https://ClinicalTrials.gov/show/NCT04305366 (accessed on 26 June 2022).
  460. Tertiary Prevention of Head and Neck Cancer With a Dietary Intervention. Available online: https://ClinicalTrials.gov/show/NCT02869399 (accessed on 26 June 2022).
  461. Expression & Epigenetic Silencing of MicroRNA for Predicting Therapeutic Response and Prognosis of HPV-negative HNSCC. Available online: https://ClinicalTrials.gov/show/NCT03953443 (accessed on 26 June 2022).
  462. Hemopurifier Plus Pembrolizumab in Head and Neck Cancer. Available online: https://ClinicalTrials.gov/show/NCT04453046 (accessed on 26 June 2022).
  463. Neoadjuvant Nivolumab for Oral Cancer Combined With FDG and Anti-PD-L1 PET/CT Imaging for Response Prediction. Available online: https://ClinicalTrials.gov/show/NCT03843515 (accessed on 26 June 2022).
  464. Study on the Interplay Between Twist1 and Other EMT Regulators Through microRNA-29 Family. Available online: https://ClinicalTrials.gov/show/NCT01927354 (accessed on 26 June 2022).
  465. The Role of microRNA-29b in the Oral Squamous Cell Carcinoma. Available online: https://ClinicalTrials.gov/show/NCT02009852 (accessed on 26 June 2022).
Figure 1. Overview of the most significant miRNAs that are upregulated in HNSCC. The first inner ring represents the subtypes of HNSCC (a); the second, third, and fourth rings represent miRNAs detected in saliva (b); tumor tissues (c); serum, plasma, or peripheral blood (d) of HNSCC patients.
Figure 1. Overview of the most significant miRNAs that are upregulated in HNSCC. The first inner ring represents the subtypes of HNSCC (a); the second, third, and fourth rings represent miRNAs detected in saliva (b); tumor tissues (c); serum, plasma, or peripheral blood (d) of HNSCC patients.
Ijms 23 08257 g001
Figure 2. Overview of the most significant miRNAs that are downregulated in HNSCC. The first inner ring represents the subtypes of HNSCC (a); the second, third, and fourth rings represent miRNAs detected in saliva (b); tumor tissues (c); serum, plasma, or peripheral blood (d) of HNSCC patients.
Figure 2. Overview of the most significant miRNAs that are downregulated in HNSCC. The first inner ring represents the subtypes of HNSCC (a); the second, third, and fourth rings represent miRNAs detected in saliva (b); tumor tissues (c); serum, plasma, or peripheral blood (d) of HNSCC patients.
Ijms 23 08257 g002
Figure 3. (a) Overview of miRNAs signatures in HNSCC subtypes; (b) miRNA signatures detected in HPV+ HNSCC and OPSCC patients. The miRNA signatures are detected and evaluated in the circulation (serum and/or plasma), tumor tissues, and saliva of HNSCC patients and in vitro, in HNSCC cell lines. The term other source refers to data derived from literature and bioinformatic analyses.
Figure 3. (a) Overview of miRNAs signatures in HNSCC subtypes; (b) miRNA signatures detected in HPV+ HNSCC and OPSCC patients. The miRNA signatures are detected and evaluated in the circulation (serum and/or plasma), tumor tissues, and saliva of HNSCC patients and in vitro, in HNSCC cell lines. The term other source refers to data derived from literature and bioinformatic analyses.
Ijms 23 08257 g003
Table 1. Summary of the most common upregulated miRNAs reported as potential biomarkers in HNSCC.
Table 1. Summary of the most common upregulated miRNAs reported as potential biomarkers in HNSCC.
miRNATumor SiteSample TypeBiomarker RoleReference
let-7aLSCCSerumDiagnosis[298]
miR-9OSCCSalivaPrognosis and diagnosis[299]
ESCCPlasmaPrognosis and diagnosis[300]
OPSCCTumor tissueRecurrence and detection of HPV+ patients[301]
miR-16ESCCTumor tissue/serumEarly detection and prognosis[278,302]
miR-18aNPCTumor tissuePrediction metastasis and therapeutic target[303]
ESCCTumor tissue/plasmaPrognosis, detection, and disease monitoring[304,305,306]
miR-19aOSCCSerum/tumor tissueDiagnosis, prognosis, and therapy[115,307]
TSCCTumor tissuePrognosis[308]
ESCCPlasma/tumor tissueEarly detection and prediction of progression-free and overall survival[304,309]
LSCCTumor tissuePrognosis and differential diagnosis[310]
miR-19bESCCPlasma/tumor tissuePrognosis and metastasis prediction[311,312]
NPCTumor tissuePrediction reduced patient survival and therapeutic target[313,314]
miR-20aESCCSerumDiagnosis[315]
NPCSerum/plasmaDetection and prognosis[316,317]
OSCCTumor tissuePrognosis[162]
miR-21OSCCTumor tissue/plasmaPrognosis[36,56,57]
LSCCTumor tissuePrognosis and prediction of lymph node metastasis[34]
TSCCTumor tissuePrediction of chemoresistance[59]
OPSCCTumor tissuePrediction and therapy[318]
miR-23aESCCTumor tissue/plasmaPrognosis and prediction of chemoresistance[261,264]
TSCCTumor tissuePrediction of chemoresistance[265]
miR-24OSCCPlasmaPrognosis[319]
miR-26bOPSCCTumor tissuePrediction and therapy[318]
miR-29bNPCSerumPrognosis[320]
miR-31OSCCTumor tissue/plasma/salivaPrognosis and diagnosis, early detection marker, and disease monitoring[46,148,154]
HNSCCTumor tissue/serumPrognosis and diagnosis[147,321]
LSCCPlasma/tumor tissueEarly detection[155]
miR-34aOPSCCTumor tissueDetection and HPV stratification[109]
miR-93ESCCTumor tissue/serumPrognosis and prediction of metastasis and radiotherapy[322,323]
miR-105ESCC/LSCCTumor tissuePrognosis[324,325]
miR-106ESCCTumor tissueDiagnosis[11,326]
OPSCCTumor tissuePrediction and therapy[318]
miR-125bNPCTumor tissuePrognosis[327]
miR-134OSCCPlasma/serum/saliva/tumor tissueMetastasis detection, prognosis, and diagnosis[299,328]
LSCCTumor tissuePrognosis and targeted therapy[329]
HNSCCTumor tissue/plasmaPrediction of poor survival[330]
miR-142HNSCCPlasma/tumor tissuePrognosis and therapy monitoring, discrimination of HPV-positive patients[245,331]
ESCC/OSCCTumor tissuePrognosis[274,332]
NPCTumor tissue/serumPotential therapy target, prognosis, and prediction of metastasis[333,334]
miR-146aOSCC/LSCCTumor tissuePrognosis[7]
OSCCPlasmaDisease detection[242]
miR-146a/bOPSCCTumor tissuePrediction and therapy[318]
miR-155HNSCCPlasma/serum/tumor tissuePrognosis and therapy[64,229]
ESCC, OSCCTumor tissueDiagnosis and prognosis and therapy target[224,226]
miR-181aOSCCPlasmaPrognosis of lymph node metastasis[211]
miR-182OPSCCTumor tissuePrognosis[335]
miR-184TSCCTumor tissue/plasmaEarly detection[336,337]
OSCCTumor tissue/salivaDiagnosis and prognosis[338,339]
miR-186HNSCCPlasmaPrognosis and therapy monitoring[331]
miR-187OSCCTumor tissue/plasmaDiagnosis and metastasis prediction[340,341]
miR-195HNSCCTumor tissue/plasmaPrognosis and therapy monitoring[331]
OPSCCTumor tissuePrediction and therapy[318]
miR-196aOSCCTumor tissue/plasmaDiagnosis, early detection, and prediction of recurrence and patients’ survival[57,171,175]
HNSCCTumor tissuePrognosis and response to radiotherapy[173]
LSCCTumor tissueDiagnosis and therapy[180]
TSCCTumor tissuePrediction of lymph node metastasis[179]
miR-196bLSCCTumor tissuePrognosis and potential therapeutic target[176,177]
OSCCPlasmaEarly detection[171]
miR-200bOSCCPlasmaDiagnosis[342]
miR-203HNSCCTumor tissueDiagnosis and metastasis detection[164,343]
miR-205HNSCCTumor tissue/plasmaPrognosis, diagnosis, and metastasis detection[343,344,345]
ESCCTumor tissueDiagnosis and therapy[346,347]
NPCPlasma/serumDisease detection[316,348]
OPSCCTumor tissuePrognosis and HPV stratification[335]
miR-223OSCCTumor tissue/serumPrognosis and therapy target[349,350]
NPCSerumPrognosis and detection[351]
ESCCTumor tissue/serum/plasmaPrognosis and diagnosis[305,352]
HNSCCTumor tissue/plasmaTherapy and disease monitoring[36]
miR-372OSCCTumor tissuePrognosis and therapy resistance[162]
ESCCTumor tissuePrognosis[353]
OPSCCTumor tissuePrognosis and metastasis detection [354]
miR-373OPSCCTumor tissueMetastasis detection[354]
miR-374bHNSCCPlasmaPrognosis and disease monitoring[331]
miR-423OSCCPlasma/salivaEarly detection and disease monitoring[355,356]
miR-424OSCCSerumDetection[357]
miR-483OSCCSerumDiagnosis and prognosis[358]
ESCCTumor tissue/serumPrognosis and therapy target[359,360,361]
NPCPlasmaPrognosis[362]
miR-486OPSCCSalivaDetection and early diagnosis[363]
miR-626OSCCSerum/tumor tissuePrognosis[364,365]
miR-1281OPSCCTumor tissueTherapy target and metastasis detection[366]
miR-3194OPSCCTumor tissueHPV patients’ stratification, metastasis detection, and therapy target[366]
miR-3651OSCCWhole bloodPrognosis, metastasis monitoring, and detection of the recurrence[367,368]
miR-5100OSCCSerumPrognosis[364]
miRNA: microRNA; HNSCC: head and neck squamous cell carcinoma; LSCC: laryngeal squamous cell carcinoma; OSCC: oral squamous cell carcinoma; ESCC: esophageal squamous cell carcinoma; OPSCC: oropharyngeal squamous cell carcinoma; TSCC: tongue squamous cell carcinoma; NPC: nasopharyngeal carcinoma; HPV: human papilloma virus.
Table 2. Summary of the most common downregulated miRNAs reported as potential biomarkers in HNSCC.
Table 2. Summary of the most common downregulated miRNAs reported as potential biomarkers in HNSCC.
miRNATumor SiteSampleBiomarker RoleReference
let-7aHNSCC/OSCCSaliva/tumor tissueEarly detection and poor prognosis[117,369]
ESCCSerumDiagnosis[315]
let-7bESCC/OSCCTumor tissueTherapy target[370]
let-7dHNSCC/OSCCTumor tissueProgression[112,114]
miR-9NPCTumor tissue/plasmaRecurrence, metastasis, and disease monitoring[194,371]
OSCCTumor tissue/serumPrognosis[197,372]
HNSCCTumor tissuePrediction and therapy resistance[198,373]
miR-10bOPSCCSalivaDetection[363]
miR-16OSCCTumor tissueTumor suppression and prognosis[273,275]
OSCCSalivaEarly diagnosis[276]
miR-24NPCTumor tissuePrognosis of recurrence, radioresistance[374]
miR-26aESCCTumor tissuePrediction of metastasis[375]
TSCCTumor tissuePrognosis[376]
miR-26bHNSCCPlasma/SerumEarly detection, prognosis, and therapeutic evaluation[41,64]
miR-26a/bESCC/OSCC/TSCCTumor tissuePrevention and therapeutic target[377,378,379]
miR-29TSCCTumor tissueProgression and therapy target[295]
ESCCSerumPrediction and radioresistance[297]
miR-31NPCPeripheral bloodTumor suppression, early diagnosis, and therapeutic target[151]
miR-34aHNSCCTumor tissueTumor suppression, diagnosis, and therapeutic target[104,105]
NPCTumor tissuePrognosis[110]
miR-92bOSCCPlasmaMetastasis detection[42]
miR-93HNSCCSalivaRadiotherapy monitoring[134]
miR-99OSCCTumor tissue/serumClinical outcome, early detection of disease, and prognosis[75,87]
HNSCCPlasmaPrognosis[36]
miR-101HNSCC/ESCCTumor tissue/serumDiagnosis and prediction of metastasis[42,380,381]
OSCCTumor tissuePrognosis and therapeutic target[162,382]
TSCC/LSCCTumor tissueDiagnosis and therapeutic target[383]
miR-124NPC/ESCCTumor tissue/plasmaPrognosis and chemosensitivity[371,384,385]
miR-125aOSCCSalivaDiagnosis and prediction[133,134]
HNSCCTumor tissuePrognosis and therapy target[166]
miR-125bOSCC/HNSCCSerum/tumor tissuePrognosis, early diagnosis, and prediction Development and progression[83,116,162]
LSCCTumor tissue/plasmaEarly detection and progression[159,163]
ESCCTumor tissuePrognosis[161]
miR-130bOPSCCTumor tissueProgression and radioresistance [386]
miR-139OSCC/TSCCSaliva/tumor tissue/serumPrognosis, early detection, disease monitoring, therapeutic targeτ[116,387,388,389]
miR-146aESCCTumor tissue/serumTumor suppression and prognosis[244]
miR-138OSCC/ESCCSerum/tumor tissueEarly detection, metastasis, and prognosis[357,390,391]
miR-150ESCC/OSCCPlasma/tumor tissuePrediction of overall survival and metastasis and early detection[328,355,392]
NPCTumor tissue/serumDiagnosis, prognosis, personalized treatment, and prediction of metastasis[348,393,394]
miR-181aOSCCTumor tissueProgression[395]
LSCCTumor tissueEarly diagnosis and treatment target[210,396]
ESCCPlasma/serumDiagnosis and prognosis[214,215]
miR-195ESCCTumor tissueDiagnosis, patient classification, and prognosis[397,398]
miR-200aHNSCC/OSCCSalivaDetection and radiotherapy monitoring[133,134]
miR-200a/cLCSSTumor tissuePrognosis and disease recurrence[130]
OSCCSaliva/oral rinseDisease detection[63]
miR-203ESCCTumor tissuePrognosis and detection of metastasis[390,399]
OPSCCTumor tissuePrediction and therapy[318]
OSCC/TSCC/NPCTumor tissuePrediction and radiotherapy resistance[400,401]
miR-204HNSCCTumor tissuePrognosis and therapeutic target[402]
OSCC/LSCCTumor tissuePrognosis of overall survival and therapeutic target[175,403,404]
miR-205OSCC/ESCCSaliva/serumDiagnosis, prognosis, and disease monitoring[299,335,405]
miR-222OSCCPlasmaEarly detection[355]
miR-223LSCCSerumDiagnosis[298]
miR-375HNSCCTumor tissuePrognosis and prediction of metastasis[68,406]
LSCC/ESCCTumor tissueOverall survival and prognosis[34,68]
OSCCPlasmaMonitoring recurrence after surgery[42]
OPSCCTumor tissuePrognosis[407]
miR-486OSCCPlasma/saliva/tumor tissueEarly diagnosis and recurrence[42,408,409]
ESCCTumor tissuePrognosis[410]
miR-892bNPCPlasmaMonitor recurrence and metastasis[371]
miR-3651OSCC/ESCCTumor tissueDiagnosis and prognosis[411,412]
miRNA: microRNA; HNSCC: head and neck squamous cell carcinoma; LSCC: laryngeal squamous cell carcinoma; OSCC: oral squamous cell carcinoma; ESCC: esophageal squamous cell carcinoma; OPSCC: oropharyngeal squamous cell carcinoma; TSCC: tongue squamous cell carcinoma; NPC: nasopharyngeal carcinoma.
Table 3. Identified gene hotspots and/or respective pathways and cellular function targeted by miRNAs in HNC.
Table 3. Identified gene hotspots and/or respective pathways and cellular function targeted by miRNAs in HNC.
miRNAFunctionExperimental Set-UpTarget Gene or Signaling PathwayHNC SubtypesReferencesCellular Functions
miR-21OncogenicIn vitroPTENOSCC[45]Proliferation, invasion, metastasis, apoptosis
p53[46]
p63[47]
PDCD4[48,49,50]
miR-375Onco-suppressorIn vitroXPR1ESCC[61]Proliferation, migration, invasion
HNF1βLSCC[65]
PDK1NPC[60]
USP1[66]
miR-99Onco-suppressorIn vitroIGF1R
mTOR
HNSCC[77]Proliferation, migration, invasion
miR-34aOnco-suppressorIn vitroFLOT-2
MEK/ERK1/2
HNSCC[105]Proliferation, migration, invasion, EMT
SATB2OSCC[413]
let-7 familyOnco-suppressorIn vitroNanog, K-RAS, CASPASE3, IL-8HNSCC[113,123,124]Proliferation, metastasis, chemosensitivity
miR-200 familyOnco-suppressorIn vitroZEB1/2, BMI1OSCC[135,414]Proliferation, migration, metastasis, malignant CSC-like properties
miR-31OncogenicIn silicoFIHHNSCC[149]Angiogenesis
miR-125aOnco-suppressor/OncogenicIn vitroERBB2 and ERBB3/CCR7HNSCC[166,415,416]Proliferation, metastasis, invasion
miR-125bOnco-suppressorIn vitroHMGA2ESCC[161]Proliferation, migration, invasion
miR-196aOncogenicIn vitroANXA1, MAMDC2, HOXB8, p27 (CDKN1B)HNSCC/OSCC[173,174]Proliferation, migration, invasion, EMT, radioresistance
miR-196bOncogenicIn vitroSOCS2, PCDH-17, ANXA1LSCC/HNSCC[176,177,178]proliferation, invasion, apoptosis
miR-9Onco-suppressorIn vitroCXCR4, Wnt/β-cateninHNSCC[373]Proliferation, colony formation
miR-181aOnco-suppressorIn vitroMAX/miR-181a/NPM1 pathway, ATF2LSCC[210,396]Proliferation, colony formation, migration, apoptosis
LNC RNA CCAT1OSCC/NPC[395,396]Proliferation, migration, drug resistance
miR-155OncogenicIn vitroCDC73, ARID2OSCC[225,229]Proliferation, invasion, apoptosis
mir-146aOncogenicIn vitroIRAK1-NF-κBESCC
OSCC
[246,417]Proliferation, apoptosis invasion
Onco-suppressorIn vitroSnail
Vimentin
E-cadherin
ESCC[244,418]
miR-23aOnco-suppressorIn vitroSIX1OPSCC[256]Growth, proliferation, chemoresistance, invasion
OncogenicIn vitroHIF1ANOSCC[261]
OncogenicIn vitroPTENESCC[262]
miR-16Onco-suppressorIn vitro/in vivoTLK1, BCL2L2OSCC[273,275]Proliferation, viability, apoptosis
BPDE/RAR-β2ESCC[278]
miR-29Onco-suppressorIn vitro/in vivoLAMC2
ITGA6
HNSCC[291]Proliferation, apoptosis, invasion, EMT
ITGB1HNSCC[292]
LOXL2HNSCC[419]
DNMT3BHNSCC [420]
Sp1/PTEN/p-AKTTSCC[295]
TIAM1NPC[294]
miRNA: microRNA; OSCC: oral squamous cell carcinoma; ESCC: esophageal squamous cell carcinoma; LSCC: laryngeal squamous cell carcinoma; NPC: nasopharyngeal carcinoma; HNSCC: head and neck squamous cell carcinoma; EMT: epithelial–mesenchymal transition; CSC: cancer stem cell.
Table 4. Summary of miRNA signatures and their utility as biomarkers in HNSCC.
Table 4. Summary of miRNA signatures and their utility as biomarkers in HNSCC.
miRNATumor SiteSample TypeBiomarker RoleReference
miR-383, miR-615, miR-877HNSCCSerumDiagnostic[277]
miR-9, miR-134, miR-191HNSCCSalivaDiagnostic[191]
miR-21-5p, miR-28-3p, miR-142-3p, miR-191-5p, miR-186-5p, miR-197-3p, miR-425-5p, miR-590-5pHNSCCPlasmaDiagnostic[331]
miR-204-5p, miR-499a-5p, miR-498-5p, miR-4714-3p, miR-30a-5p, miR-1-5p, miR-548f-3p, miR-518a-3p, miR-155-3p, miR-365a-5p, miR-196b-5pHNSCC(Bioinformatic analysis/TCGA)Prognostic[24]
miR-99a-5p, miR-758-5p, miR-329-3p, miR-137-3p, miR-1229-3p, miR-3187-3pHNSCCTumor tissuePrognostic[427]
let-7c, miR-125b-2, miR-129-1, miR-337, miR-654, miR-99aHNSCCTumor tissue (TCGA)Prognostic[428]
miR-21-3p, miR-21-5p, miR-96-5p, miR-429HNSCCTumor tissuePrognostic[429]
let-7g-3p, miR-6508-5p, miR-210-5p, miR-4306, miR-7161-3pHNSCCTumor tissuePrognostic[430]
miR-142-3p, miR-186-5p, miR-195-5p, miR-374b-5p, miR-574-3pHNSCCPlasmaPrognostic[331]
miR-499a, miR-548k, miR-3619, miR-99a, miR-137, miR-3170, miR-654HNSCCTumor tissue (TCGA)Prognostic[431]
miR-16, miR-29b, miR-150, miR-1254, let-7eHNSCCCell lines (TCGA)Predictive (radiotherapy)[432]
miR-99a, miR-31, miR-410, miR-424, miR-495HNSCCTumor tissue (TCGA)Predictive (radiotherapy)[433]
miR-6510-3p, miR-34c-5pOSCCTumor tissueDiagnostic[7]
miR-31-5p, miR-21-5p, miR-125b-5p, miR-99a-5p, miR-100-5p, let-7c-5p, miR-24-3p, miR-30cOSCCSaliva (Oral Swirls)Diagnostic[434]
miR-196a, miR-196bOSCCPlasmaDiagnostic[171]
miR-218, miR-125b, let-7gOSCCTumor tissuePrognostic[435]
miR-127-3p, miR-4736, miR-655-3pOSCCTumor tissuePrognostic[436]
miR-21, miR-181b, miR-345OSCCTumor tissueSusceptibility/risk[437]
miR-21, miR-375TSCCOral (Brush) Cytology, tumor tissueDiagnostic (screening)[49]
miR-142-3p, miR-31, miR-146a, miR-26b, miR-24, miR-193bOPSCCTumor tissuePrognostic[438]
miR-27a-3p, miR-455-5p, miR-203a-3p, miR-584-5p, miR-24-3p, miR-548k, miR-126-3p, miR-126-5p, miR-365a-5p, miR-98-5p, miR-151b, miR-361-3p, miR-374c-5p, miR-150-5p, miR-374b-5p, miR-107, miR-125b-5p, miR-1287-5p, miR-146a-5p, miR-106a-5p, miR-15b-5p, miR-20b-5p, miR-532-3p, miR-361-5p, miR-363-3p, miR-625-3pOPSCCTumor tissuePrognostic[439]
miR-107, miR-151, miR-492OPSCCTumor tissuePrognostic[440]
miR-20b, miR-107, miR-151, miR-182, miR-361OPSCCTumor tissuePrognostic[440]
miR-151, miR-152, miR-324-5p, miR-361, miR-492OPSCCTumor tissuePrognostic[440]
miR-449a-5p, miR-6510-3p, miR-133a-5pLSCCTumor tissueDiagnostic[7]
miR-31-3p, miR-196a-5pLSCCPlasmaDiagnostic[155]
miR-200a-3p, miR-30b-5p, miR-4451HSCCTumor tissuePrognostic[441]
miR-29c, miR-30e, miR-93NPCTumor tissue (GEO)Prognostic[442]
ebv-miR-BART19-3p, miR-135b, miR-141NPCTumor tissue (GEO)/cell linePrognostic[443]
miR-142-3p, miR-29c, miR-30eNPC(Bioinformatic analysis/GEO)Prognostic[334]
miRNA: microRNA; HNSCC: head and neck squamous cell carcinoma; LSCC: laryngeal squamous cell carcinoma; OSCC: oral squamous cell carcinoma; ESCC: esophageal squamous cell carcinoma; OPSCC: oropharyngeal squamous cell carcinoma; TSCC: tongue squamous cell carcinoma; NPC: nasopharyngeal carcinoma; TCGA: The Cancer Genome Atlas; GEO: gene expression omnibus.
Table 5. Summary of the most common miRNA signatures associated with HPV infection in HNSCC patients.
Table 5. Summary of the most common miRNA signatures associated with HPV infection in HNSCC patients.
miRNATumor SiteSample TypeBiomarker RoleReference
miR-9, miR-134, miR-196b, miR-210, miR-455HNSCCSalivaEarly-stage detection and HPV+ patients’ stratification[190]
miR-191, miR-196b, miR-210, miR-222HNSCCSalivaHPV+ patients’ stratification[190]
miR-378a-3p, miR-16-1-3p, miR-493-3p, miR-380-5p, miR-376c-3p, miR-338-5pHNSCCTumor tissue (TCGA/GDAC)The prognosis and detection of HPV+ patients [446]
miR-324-5p, miR-4764-3p, miR-107, miR-1234, miR-3144-3p, miR-3176, miR-3177-3p, miR-4267, miR-4418, miR-615-3p, miR-668, miR-99b-3p, miR-675-3p, miR-584-5p, miR-212-3p, miR-18b-5p, miR-18a5p, miR-138-1-3p, miR-135b-5p, miR-1246, miR-4764-3p, miR-857, miR-7-2-3pOPSCCTumor tissueThe detection of HPV+ patients [456]
miR-363-3p, miR-551b-3p, miR-20b-5p, miR-20b-3p, miR-143-3p, miR-106a-5p, miR-9-5p (miR-9-1), miR-9-5p (miR-9-2), miR-9-5p (miR-9-3), miR-99a-3p, miR-99a-5p, miR-9-3p (miR-9-1), miR-193a-5p, let-7b-3pOPSCCCell lineEarly detection and patient stratification [457]
miR-206/miR-494-3p, U6 snRNA/miR-150-5p, miR-532-3p/miR-574-3p, miR-125a-5p/miR-193b-3p, miR-1274b/miR-27a-3p, miR-494-3p/miR-150-5p, miR-193a-5p/U6 snRNA, miR-27a-3p/miR-93-5p, ath-miR-159a/miR-152-3p, ath-miR-159a/miR-494-3p, miR-375-3p/miR-483-5pOPSCCSerum (EVs)The detection of HPV+ patients[458]
miRNA: microRNA; HNSCC: head and neck squamous cell carcinoma; OPSCC: oropharyngeal squamous cell carcinoma; TCGA: The Cancer Genome Atlas; GDAC: Genome Data Analysis Centers; HPV: human papilloma virus; EVs: extracellular vesicles; snRNA: small nuclear RNA.
Table 6. Current clinical trials assessing miRNAs’ potential utility as biomarkers.
Table 6. Current clinical trials assessing miRNAs’ potential utility as biomarkers.
IdentifierOfficial TitleClinical PhaseTarget miRNAsSample TypePurposeRef.
NCT04305366MicroRNA Markers in Head and Neck CancersN/AN/DFine needle aspiration biopsy, serum, salivaTo investigate the miRNA signature of samples and to develop biomarkers for surveillance of HNSCC patients.[459]
NCT02869399A Randomized Phase II Study for Tertiary Prevention of Squamocellular Cancer of Head and Neck (SCCHN) With a Dietary InterventionIIN/DSaliva, plasmaTo investigate the role of diet as a risk factor for HNSCC recurrence and secondary tumor development, to identify saliva and plasma miRNAs, and to evaluate their change in inflammatory cytokine profile during the course of dietary intervention.[460]
NCT03953443INST 1008: Expression and Epigenetic Silencing of MicroRNA for Predicting the Therapeutic Response and Prognosis of HPV- negative Head and Neck Squamous Cell Carcinoma (HNSCC)N/AN/DTumor tissue, normal tissueTo assess the association between miR expression and miR promoter methylation and the response to therapy and prognosis in primary HPV-negative HNSCC patients.[461]
NCT04453046Depleting Exosomes to Improve Response to Immune Therapy in Head and Neck Squamous Cell Cancer: An Early Feasibility Phase I Clinical TrialIN/DBloodTo determine whether the use of Hemopurifier before treatment with pembrolizumab is low-risk and well-tolerated by the patients and whether it leads to reduced levels of exosomes in the blood and to identify immunoinhibitory proteins and miRNA profiles for the evaluation of effectiveness of combination treatment in decreasing immune suppression in patients with recurrent/metastatic HNSCC.[462]
NCT03843515Safety and Tolerability of Neoadjuvant Nivolumab for Locally Advanced Resectable Oral Cancer Combined With [18F] BMS-986192/[18F]-FDG PET Imaging and Immunomonitoring for Response PredictionIN/DTumor tissue, plasmaTo further evaluate tumor PD-L1 expression as a predictive biomarker and to investigate the immunophenotype of the patient and tumor, as well as the presence of neoantigens and other potential biomarkers such as plasma vesicle miRNAs.[463]
NCT01927354Observational Study on the Investigation of the Molecular Mechanism and Clinical Significance of the Interplay Between Twist1 and Other EMT Regulators Through microRNA-29 FamilyN/AmiR-29 familyTumor tissueTo delineate the regulatory mechanism of the Twist1-miR-29s-SIN3A axis, to investigate the molecular interplay between Twist1 and Snail through Twist1-miR-29s-SIN3A signal pathway, and to elucidate the molecular basis and pathophysiologic significance of Twist1-Snail interaction under hypoxic environment.[464]
NCT02009852The Role of microRNA-29b in the Oral Squamous Cell CarcinomaN/AmiR-29bTumor tissue, serum, salivaTo identify a prognostic significance for miR-29b in oral cancer.[465]
N/A: not available; N/D: no data; HNSCC: head and neck squamous cell carcinoma; miR: microRNA; HPV: human papilloma virus; PET: positron emission tomography; EMT: epithelial–mesenchymal transition.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Thomaidou, A.C.; Batsaki, P.; Adamaki, M.; Goulielmaki, M.; Baxevanis, C.N.; Zoumpourlis, V.; Fortis, S.P. Promising Biomarkers in Head and Neck Cancer: The Most Clinically Important miRNAs. Int. J. Mol. Sci. 2022, 23, 8257. https://doi.org/10.3390/ijms23158257

AMA Style

Thomaidou AC, Batsaki P, Adamaki M, Goulielmaki M, Baxevanis CN, Zoumpourlis V, Fortis SP. Promising Biomarkers in Head and Neck Cancer: The Most Clinically Important miRNAs. International Journal of Molecular Sciences. 2022; 23(15):8257. https://doi.org/10.3390/ijms23158257

Chicago/Turabian Style

Thomaidou, Arsinoe C., Panagiota Batsaki, Maria Adamaki, Maria Goulielmaki, Constantin N. Baxevanis, Vassilis Zoumpourlis, and Sotirios P. Fortis. 2022. "Promising Biomarkers in Head and Neck Cancer: The Most Clinically Important miRNAs" International Journal of Molecular Sciences 23, no. 15: 8257. https://doi.org/10.3390/ijms23158257

APA Style

Thomaidou, A. C., Batsaki, P., Adamaki, M., Goulielmaki, M., Baxevanis, C. N., Zoumpourlis, V., & Fortis, S. P. (2022). Promising Biomarkers in Head and Neck Cancer: The Most Clinically Important miRNAs. International Journal of Molecular Sciences, 23(15), 8257. https://doi.org/10.3390/ijms23158257

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