Next Article in Journal
Immunotherapy in Thoracic Malignancies: New Treatment and New Hope
Next Article in Special Issue
Current and Future Biomarkers for Immune Checkpoint Inhibitors in Head and Neck Squamous Cell Carcinoma
Previous Article in Journal
Co-Occurring CSF3R W791* Germline and Somatic T618I Driver Mutations Induce Early CNL and Clonal Progression to Mixed Phenotype Acute Leukemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Micro-RNAs, the Cornerstones of the Future of Radiobiology in Head and Neck Cancers?

by
Camil Ciprian Mireștean
1,2,
Roxana Irina Iancu
3,4,* and
Dragoș Petru Teodor Iancu
5,6
1
Department of Oncology and Radiotherapy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
2
Department of Surgery, Railways Clinical Hospital, 700506 Iasi, Romania
3
Department of Oral Pathology, Faculty of Dentistry, “Gr. T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
4
Clinical Laboratory Department, “St. Spiridon” Emergency Hospital, 700111 Iasi, Romania
5
Department of Radiotherapy, Regional Institute of Oncology, 700483 Iasi, Romania
6
Department of Medical Oncology and Radiotherapy, Faculty of Medicine, “Gr. T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2022, 29(2), 816-833; https://doi.org/10.3390/curroncol29020069
Submission received: 15 December 2021 / Revised: 26 January 2022 / Accepted: 28 January 2022 / Published: 2 February 2022
(This article belongs to the Special Issue Advances in Squamous Cell Carcinoma of the Head and Neck)

Abstract

:
Even though it is only the 6th most common malignancy at the modal level, head and neck cancers are distinguished by a considerable treatment failure rate, especially by locoregional recurrences, the intrinsic tumor radioresistance being one of the causes of this phenomenon. The efforts of radiobiological research of these cancers are oriented towards the identification of biomarkers associated with radioresistance and radiosensitivity in order to modulate the treatment so that the therapeutic benefit is maximum. Micro-RNAs (miRNAs, miRs), small single-stranded non-coding RNA molecules are currently being extensively evaluated as potential biomarkers in numerous diseases, including cancer. The evaluation of the potential of miRNAs to modulate or predict radiosensitivity or radioresistance, to anticipate the risk of recurrence and metastasis, and to differentiate different tumor subtypes is based on multiple mechanisms by which mRNAs control proliferation and apoptosis and interact with cell cycle phases or act as oncogenes with the potential to influence invasion promotion or tumor suppression. A refinement of radiosensitivity based on miRNAs with clinical and radiobiological application in head and neck cancers can lead to a personalization of radiotherapy. Thus, a miRNA signature can anticipate the risk of toxicity associated with chemoradiation, the possibility of obtaining locoregional control after treatment, and the recurrence and distant metastasis risk. The potential of miRNAs as an intrinsic predictor of sensitivity to chemotherapy may also guide the therapeutic decision toward choosing an escalation or de-escalation of concurrent or sequential systemic treatment. The choice of the irradiated dose, the fractional dose, the fractionation scheme, and the refining of the dose-volume constraints depending on the radiosensitivity of each tissue type estimated on a case-by-case basis by miRNAs profile are possible concepts for the future radiotherapy and radiobiology of head and neck cancers.

1. Introduction

Head and neck cancers are the 6th most common malignancy worldwide and squamous cell carcinoma of the head and neck (HNSCC) is currently the most common malignant tumor of the head and neck and accounts for approximately 90% of head and neck cancers. With an annual incidence of over 500,000 new cases worldwide, the severity of HNSCC is given by the unfavorable prognosis, the 5-year survival being about 40% even if maximum treatment is administered [1,2,3]. The pattern of therapeutic failure is mainly locoregional recurrence which occurs in 15–50% of cases, but also distant metastasis can be a cause of disease progression [4]. Resistance to oncological therapies (chemotherapy, radiotherapy, molecular therapy, and immunotherapy) is the main pathophysiological phenomenon that underlies the therapeutic failure of this type of cancer. Radiotherapy is part of the adjuvant or definitive treatment of HNSCC as a single treatment or in combination with therapies with synergistic and radiosensitizing potential, such as chemotherapy or targeted molecular therapy. Although the combination of radiation therapy with chemotherapy demonstrates a potential benefit on tumor control, the toxic effects associated with concurrent treatment are severe and may alter the quality of life (QoL) or may even limit the survival of these patients [5,6]. In this context, the identification of agents that increase tumor radiosensitivity could lead to increased local control rates without increasing the rate of toxicity. Even if radiosensitizing agents are not used, there are significant variations in the tumor response to irradiation, modulated by intrinsic factors that modulate radiosensitivity. Protein-encoding genes can modulate the response of tumor and normal tissues to irradiation, but direct gene manipulation is difficult [2,7]. MiRNAs, a class of small endogenous non-coding RNA, generally composed of 22 nucleotides, can provide new horizons in the control of intrinsic radiosensitivity by post-transcriptional regulation of gene expression. It has been shown that miRNA overexpression or knock-down may both alter radiosensitivity of a tumor or normal tissue [7,8,9]. Thus, evaluation of these miRNAs’ expression can predict DNA damage or lead to cell-cycle checkpoint manipulation. Knowledge of each miRNA role may result in the design of radiobiological models based on miRNAs with direct clinical applications [8].

2. Aim of Study

Without intending to study the depth of the mechanisms of involvement of each miRNA in modulating radiosensitivity, we want to provide a starting point for radiation oncologist clinicians with an interest in translational research in understanding and applying in clinical trials data obtained from fundamental miRNA research in head and neck cancers. By presenting epidemiological and etiopathogenic data, we wish to provide a miRNA-based bridge to understanding the different response to irradiation with a special focus on differentiating HPV+ and HPV− subtypes from oropharyngeal cancers. Last but not least, the current context created by the COVID-19 pandemic with consequences in the rapid implementation of hypofractionation schemes simultaneously with the large-scale implementation of immunotherapy having potentially synergistic with irradiation requires an in-depth understanding and refinement of the head and neck cancers radiobiology, beyond the basics of the linear quadratic model.

3. Head and Neck Cancers: Epidemiology, Etiology, and Pathogenesis

With an incidence of 550,000 cases annually, head and neck cancers represent the 6th neoplasm as the worldwide incidence. The higher death rate (approximately 300,000 per year) is a reason why these cancers require special attention in order to establish measures in order to reduce the incidence and mortality by designing prevention programs but also by refining therapeutic protocols. About 90% of head and neck cancers are squamous cell carcinoma (HNSCC) [6,7,8]. Only a third of these patients are diagnosed in the early stages of the disease (T1-2, NO), the 5-year survival rate being about 40–50% for all HNSCC stages. Smoking is considered the major independent risk factor, both for the development of HNSCC and for development of treatment-related complications such as jaw radionecrosis (in the case of patients receiving radiation therapy). Alcohol is also an independent risk factor for both the development of HNSCC and for the deterioration of quality of life if the patient continues to drink alcohol after completing treatment. A detailed analysis of data from clinical trials published between 2006 and 2009 identifies 55% of 654 oropharyngeal cancer patients as human papilloma virus (HPV) positive. HPV infection has become a growing amplitude risk factor especially in the last decades. Without being able to identify a premalignant clinical lesion, the only effective prevention strategies are the sexual education of young people and the vaccination of 12–13-year-old girls. Oral sex is involved in the transmission of HPV subtypes with a high risk of viral carcinogenesis (subtypes 16 and 18), the prevalence of cases of oral cancer and HPV-positive oropharynx increasing especially in developed countries. Premalignant lesions are also a risk factor for HNSCC, dysplasia being identified by biopsy in 25% of cases of leukoplakia and more commonly in erythroplakia, but the association of HPV infection is not synergistic in these cases. Premalignant conditions such as Fanconi’s anemia, ataxia teleangiectasia Bloom’s syndrome, and Li–Fraumeni syndrome are associated with increased rates of HNSCC and their onset at a young age. The involvement of HPV may be also relevant in HNSCC associated with Fanconi anemia [3,10,11,12,13]. Analyzing data from 1990 to 2017, Auperin highlights a decrease in the incidence of nasopharyngeal and laryngeal cancer worldwide and an increase in the incidence of pharyngeal and lip/oral cavity cancers [14]. An increased incidence of HPV etiology in the oral cavity and oropharynx cancers could justify this change in the ratio of head and neck cancer subtypes. The study mentions the consumption of fruits and vegetables but also the vaccination of boys as strategies to reduce the incidence of HNSCC [14].
HNSCC develops from the mucosal epithelium, the pathophysiological process of carcinogenesis passing successively through the stage of hyperplasia, dysplasia, carcinoma in situ, and finally invasive carcinoma. Being a heterogeneous disease, both by different anatomical locations and by etiological factors (viral or carcinogenic), the cells of origin of HNSCC are usually adult stem cells or progenitors. Even if the pluripotent self-renewal stem cell is represented by only 1–3%, cancers caused by this type of cell with the ability to self-replicate when it is transplanted in other tissues is distinguished by resistance to standard treatment. CD44 (a surface receptor for hyaluronic acid and matrix metalloproteinases), ALDH1 (an intracellular enzyme with a role in detoxification), and CD133 (a membrane-spanning protein) are involved in the process of tumor invasion and metastasis, having a decisive role in prognosis [15,16,17].
The increased frequency of synchronous and metachronous cancers, both in different segments of the head and neck and in the lung or esophagus associated with field cancerization have as substrate molecular abnormalities, the carcinogenesis process in the second primary tumor being amplified by smoking. The presence of ALDH1 in the vicinity of vessels justifies the increased risk of metastasis for cases that have high levels of ALDH1 [18,19].
In the case of HPV-negative cancers, the most incriminated carcinogens are polycyclic aromatic hydrocarbons and especially nitrosamines in the case of tobacco exposure, but for situations where the main carcinogen is betel quid or areca nut, the mechanism is not so much studied. A destabilization of the balance between metabolic activation and detoxification of carcinogen-induced DNA damage underlies carcinogenesis in HPV-negative HNSCC. The metabolism of alcohol to acetic aldehyde increases the number of lesions accumulated, so alcohol is a potentiating factor of carcinogenesis [20]. In the case of HPV+ cancers, carcinogenesis begins in the crypts of the palatal and lingual tonsils. Along with the HPV-16 subtype, which is undoubtedly dominant as a risk factor, in a minority of cases HPV-52, HPV-18, HPV-31, and HPV-33 subtypes could also be identified [21].
In the case of the HPV-16 subtype, the E1–E5 genes encode proteins involved in the replication and transcription of the viral genome and the E6 and E7 genes are associated with carcinogenesis. The E6 gene acts through the synthetic protein product on the tumor suppressor p53, degrading it. E7, via retinoblastoma-associated protein (RB1), acts at the nickel control points of the cell cycle. Alteration of RB1 function also has the effect of upregulating the p16 protein, well known as a surrogate biomarker in the identification of HPV− or HPV+ oropharyngeal cancer discrimination. All these totally different mechanisms in the case of HNSCC HPV− and HPV + highlight that we can talk about two different diseases not only as an etiology, but also as a pathophysiological mechanism of carcinogenesis [22,23].

4. MicroRNA Involvement in Head and Neck Cancer Development

Human papillomaviruses (HPV) are involved in the etiopathogenesis of at least three type of cancers (cervical, anal, and HNSCC). Although the involvement of HPV infection in the case of cervical cancer is incriminated in almost all cases, there is only a part of head and neck cancers associated with this viral infection. It is obvious that the HPV infection causes the hijacking of the host cell pathways, but the identification of the targets within this cell as well as their contribution to the malignancy process is also of major importance. The Hippo pathway, involved in epithelial homeostasis, is thought to be involved in the carcinogenesis and progression of HPV-induced cancer. Morgan and collaborators propose the activation of the Hippo pathway as a therapeutic target in cancers associated with HPV infection. Serine/threonine-protein kinase 4 (STK4), the master Hippo kinase is identified as low in HPV-associated cancers and HPV-associated proteins E6 and E7 upregulate the miR-18, thus promoting tumor genesis by inhibiting STK4. The suppressive effect of miR-18 on STK4 and indirectly tumor promotion is identified not only in HPV-induced cancers but also in prostate cancer [24,25].
The development of the tumor phenotype is based on alterations in tumor suppressor oncogenes in tumor cells and stromal cells. These phenomena have the effect of transforming the normal epithelium into carcinoma in situ and subsequently into invasive squamous cell carcinoma. Loss of p53 control by viral mutations has the final effect of carcinogenesis by inhibiting apoptosis. In HPV-positive cancers, inhibition of p53 is mediated by the E6 protein characteristic of the HPV16 subtype. Tumor proliferation is also supported by p16 inactivation and overexpression of cyclin D in HNSCC. Epidermal growth factor (EGFR) is involved in the control of multiple pathways that mediate both proliferation and invasion and migration. EGFR also modulates tumor survival and angiogenesis, and EGF-induced STAT3 signaling initiates the transcription of genes that modulate cell growth, survival, and angiogenesis (Cyclyn D1, Bcl-XL, and VEGF, respectively) [25,26,27,28,29,30,31,32]. Progression and metastasis involve the interaction of the tumor with the tumor microenvironment and the remodeling of it and the stromal cells, a cytokine-mediated interaction. Angiogenesis, basement membrane modification, and tumor proliferation are also processes in which cytokines are involved. Infiltrating immune and endothelial cells but also cancer fibroblasts have been associated with the process of distant metastasis. Cancer-associated fibroblasts already shown to be associated with tumor progression have been correlated by Shi et al. with the occurrence of lung metastasis. Evaluated in a preclinical model, TGFβ-enriched fibroblasts and TGFβ activation were correlated with the risk of micrometastases [32,33,34].
The role of miRNA in the carcinogenesis process is both to regulate genes that suppress tumors, their blocking being associated with carcinogenesis, but also to block oncogenes. Deletion of miRNA genes leads to increased oncogenic production, favoring tumor progression [30,32]. Moreover, miR-15a and miR-16-1 are the first miRNAs identified as involved in carcinogenesis, being detected in more than 50% of cases of chronic leukemia. The oncogenic effect of these two miRNAs is mediated by the modulation of the BCL-2 antiapoptotic gene [28,29]. MiR-21 has been shown to be frequently identified in HNSCC and has the role of reducing the expression of PTEN, a modulator of the phosphoinositide 3-kinasePI3K pathway, the most commonly mutated pathway in HNSCC. Another miRNA involved in the carcinogenesis of head and neck cancers is miR-31. It promotes tumor progression and angioneogenesis by activating the hypoxia-inducible factor (HIF) pathway. MiR-375, identified in over 90% of HNSCC, is considered a tumor suppressor [30,31,32]. miR-34a is involved in tumor suppression, its reduced expression being identified in pancreatic, breast, and lung cancer [35,36,37].
MiR-34a demonstrated a significant role in mediating apoptosis, senescence, and related to p53-mediated cell cycle arrest, directly targeting cyclin E2, BCL-2cyclin-dependent kinases (CDK4 and CDK6). MiR-34 family loss in tumors was also associated with tumorigenesis [35,36]. Recent studies demonstrate the potential of miR-34a to modulate tumor growth by denervating the p53-mediated tumor microenvironment [37]. P53 modulates cell survival through indirect control of hypoxia and angiogenesis. Knockdown of endogenous miR-107 may act to amplify inducible factor-1beta hypoxia (HIF-1beta) and overexpression of miR-107 reduces angiosis. MiR-192 and miR-215 are effectors and regulators of p53 and may suppress tumor genesis by cell cycle arrest [38]. No miRNA has been shown to directly modulate capillary tumor extravasation to favor metastasis, but miR-520/373, miR-204, and miR-200 modulate tumor angiosis via TGFβ by tumor-associated fibroblasts (TAFs) [39]. MiR-30a-5p interacts with the MET and EGFR pathways thus being an indirect suppressor of tumor growth [40]. Mammalian target of rapamycin (mTOR) of serine-threonine kinase protein with PI3K/Akt pathway signaling role also has a role in tumor proliferation independent of EGFR and p53. The mTOR pathway can also modulate epithelial-mesenchymal transition (EMT), thus being a promoter of tumor migration. MiR-7, miR-99a, miR-100, and miR-101 are just some of the miRNAs involved in modulating this pathway in cancer [41]. MiRNA-7 inhibits tumor growth and metastasis by targeting the phosphatidylinositol 3-kinase/Akt pathway in hepatocellular carcinoma [42]. Silencing of miR-17-5p can block HNSCC tumor cells in the G2/M phase, thus demonstrating the potential of this miRNA to promote tumor growth and progression [43].

5. miRNAs and Cancer-Implications in Clinical Practice—Focus on Radiobiology:

The interest in the value of miRNA in the radiobiology of the future is justified by the vast number of reports that mention the involvement of these small non-coding RNAs both in the development and normal function of organs (brain development and functioning) and in the pathogenesis of diseases such as neuropsychiatric disorders, schizophrenia or bipolar disorder, atherosclerosis, cardiac hypertrophy, and systemic lupus erythematosus. Their huge potential for regulatory molecules opens new horizons in the medicine of the future and in the treatment and diagnosis of many diseases [33,34,44]. The modulation potential of radiosensitivity is given by the involvement of miRNAs in the differentiation and proliferation and cell death. By specifically altering these cellular functions of both cancer and other diseases, miRNA demonstrates its potential to be used as a circulating noninvasive biomarker. Even if the many prognostic and predictive risk factors for the cancer evolution are currently used in daily clinical practice (such as tumor staging according to TNM classification, histological type, degree of cell differentiation, genetic mutations, and expressions of some proteins with biomarker value), a refining of these biomarkers is necessary, especially regarding both the tumor and the normal tissue response to irradiation. Thus, it will be easier to anticipate as accurately as possible the risk of toxicity but also the possibility of tumor control for each individual case [33,44,45]. As early as 25 years ago, the different response to irradiation was considered a consequence of the variation in intrinsic radiosensitivity. Pekkola-Heino and collaborators evaluated the average value of radiosensitivity at 1.9 Gy with variations between 1.0 Gy and 2.8 Gy, finding sensitive differences even between cell populations [46]. Cancers are also among the diseases in which miRNAs are involved both in down-regulation and up-regulation of different genes involved in tumor radiosensitivity by activating mechanisms of radioresistance or radiosensitization. MiRNAs’ impact can be augmented via a carcinogenesis effect exercised by down-regulation of the tumor suppressor gene or by modulating cell proliferation and apoptosis. The large amount of evidence that makes the connection between down-regulation, up-regulation, knockdown, overexpression, or other dysregulation of miRNAs in various cancers justifies the hypothesis that these small single-stranded non-coding RNA molecules will play an essential role in the oncology of the near future. MiRNAs are modulators of radiosensitivity through effects on phenomena such as cell damage repair, apoptosis, and free radical generation [47,48,49].
Mi-R-139-5p is associated with the accumulation of DNA damage caused by irradiation via the methionine adenosyltransferase 2A (MAT2A) gene. Another pathway for modulating DNA damage involved in radiosensitivity is the mutant ataxia-telangiectasia (ATM) and ataxia-telangiectasia (ATR) genes that modulate DNA damage repair via cyclin-dependent kinase (CDK) [41,42]. MiR-16 and miiR-15/ab are involved in the modulation of this pathway. Blocking the formation of CDK/Cyclin complexes limits the transition of the cell from phase G1 to phase S and from phase G2 to phase M. MiR-15 family and miR-449 are involved in G2 and G2/M phase arrest. ATM/P53/P21 is another pathway with cell cycle implications and miR-200c, miR-375, and miR-106b controlled this pathway [50,51,52]. Also worth mentioning is miR-208a, a radiation-induced mi-RNA that can produce radioresistance by activating the AKT/mTOR pathway [50,51,52,53,54]. A direct or inverse correlation between miRNA and specific proteins, determinants of tumor radiosensitivity in breast cancer, has been identified. Among these proteins, we mention proteins that are therapeutic targets in several types of cancers: vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR), human epidermal growth factor receptor-2 (Her-2/neu) whose status is involved in the molecular classification of breast cancer, and p53, a well-known predictor both of radiosensitivity and of sensitivity to platinum-based chemotherapy. The mechanism of direct and inverse correlation is given by the role of these proteins in regulating the expression of some miRNAs, but miRNAs also have a role in regulating proteins at the post-transcriptional level [55,56]. MiR-18a has the potential to be a radiosensitivity biomarker and an actor that modulates radioresistance, having a suppressive effect on ATM genes, with direct influence on the ability to repair double-strand DNA breaks after irradiation [33,34]. Without considering that we cover the entire vast field of radiosensitivity modulation, the purpose of the example was to highlight the role of miRNAs in the radiosensitivity of one of the most heterogeneous cancers, HNSCC. MiR-218 identified in tumor tissue is associated with unfavorable evolution and cancer progression. In fact, it has been shown that up-regulation of miR-218 is also involved in potentiating irradiation-induced apoptosis and due to this mechanism, miR-218 becomes a potential biomarker of radiosensitivity in cervical cancer. EMT and angiogenesis are regulated in colorectal cancer by miR-218 [57]. MiR-145 expression is associated with radiosensitivity in high-risk human papilloma virus (HPV) associated with cervical cancer, the mechanism being one of synergistic interaction with long non-coding RNA mucosa-associated lymphoid tissue via the lymphoma translocation protein 1 (MALAT1) [58,59,60]. MiR-139 can modulate radiosensitivity by accumulating DNA damage, the mechanism being mediated by the MAT2A gene [29]. The accumulation of radiation-induced DNA damage has as its substrate the inhibition of two mechanisms: non-homologous end joining (NHEJ) and homologous recombination (HR) pathways, methods by which under normal conditions DNA damage is repaired. If the total repair of the lesions has occurred, the cell may die by entering in the apoptosis, suffering a mitotic catastrophe, or it may continue the normal cell cycle [61]. MiR-208a-modulated radiosensitivity has been demonstrated in both preclinical cell and animal models and miR-208a has been associated with radioresistance and proliferation in lung cancer. In patients with non-small cell lung carcinoma (NSCLC), overexpression of hsa-miR-96-5p and hsa-miR-874-3p associated with irradiation may potentiate the tumoricidal effect, the results being similar to the situation where systemic agents targeting HR and NHEJ pathways are added to radiotherapy [50,62]. PTEN down-regulation has as a consequence the activation of the PI3K/AKT pathway, associated with radioresistance and tumor proliferation. MiR-10b reduces the antiproliferative effect of irradiation by activating caspase 3/7 and inhibiting Bcl-2 expression [63,64,65,66]. Thus, activating p-AKT, miR-10b expression reduces the sensitivity of glioblastoma to irradiation promoting proliferation and tumoral invasion [64,65].

6. miRNA in Head and Neck Cancers—From the Biomarker of the Future to the Orchestrator of Radiosensitivity

The huge potential of miRNAs to become valuable and accurate biomarkers for diagnostic and prognosis in the future is supported by several basic characteristics: they are rapidly synthesized in a certain clinical situation, have a high specificity, and remain in the environment from which they are identified a long period of time. It is generally considered that there are three types of diseases in which miRNAs have and will have a key role as a biomarker (cardiovascular diseases, infectious diseases, and neoplasms) [57,58,59,60]. Nearly a decade after the launch of the first miRNA panel that opened the horizons for the widespread use of miRNAs in medical practice, there are still problems standardizing and establishing relationships between a miRNA or set of miRNAs and a particular disease. In clinical practice, the first use of miRNA as a biomarker is attributed to Lawrie and collaborators who, in 2008, identified higher levels of tumor-specific miRNAs in patients with large B-cell lymphoma [66,67,68,69,70,71,72].
MiR-138 was associated with the control of transcriptional activity of E-cadherin and elevated levels were negatively correlated with the risk of metastasis. Being commonly reported in HNSCC miR-138, associated with EMT this miRNA is a potential prognostic biomarker for HNSCC [64,65,66,67]. Different results are reported for miR-205-5p; identified in peritumoral tissue, this miRNA is associated with early detection of minimal residual disease involved in tumor development. MiR-205-5p is also considered a tumor suppressor and a limiter of tumor migration and invasion in squamous cell carcinoma of the oral cavity. An association with let-7d of this miRNA may constitute a combined biomarker of survival and prognosis in HNSCC [73,74,75,76,77]. Reduced miR-29c-5p expression is associated not only with the malignant phenotype in HNSCC but also has prognostic and therapeutic implications. Up-regulation of miR-29c-5p is correlated with the arrest of tumor cells in the G2/M phase, being associated with reduced tumor proliferation both in vivo and in vitro. This miRNA proves to be not only a biomarker but also a potential therapeutic target [78].
Intensity Modulated Radiation Therapy (IMRT) in a dose of at least 70Gy in 35 daily fraction, 5 fractions per week over 7 weeks, and concurrent chemotherapy with cisplatin is currently the therapeutic standard for the treatment of locally advanced non-metastatic head and neck cancers. Cisplatin, a platinum-based alkylating agent, has the potential for radiosensitization, being, so far, the backbone of the concurrent association between radiation therapy and systemic therapy. Even if given weekly at a dose of 40mg/m2 or 100 mg/m2 every 3 weeks, new evidence indicates that a cumulative dose of at least 200 mg/m2 is associated with therapeutic benefit. In addition, the stratification of patients with squamous cell carcinomas of the head and neck (HNSCCs) according to human papilloma virus (HPV) status in HPV+ and HPV− tumors showed differences in response to chemoradiation with a higher response rate in non-smokers, HPV+ [79,80,81,82,83,84]. Although the role of induction chemotherapy is still controversial, the triple combination of TPF (docetaxel, carboplatin, and fluorouracil) followed by platinum-based chemoradiation (CCRT) may be considered with benefits in locoregional control for certain categories of patients. Contrary to expectations, trials that proposed de-escalating treatment for oropharyngeal HNSCC HPV + cases were negative. However, some controversial topics exist regarding the concept of de-escalation, requiring a refinement of results. Analyzing the data from a small Italian phase II trial, the significantly lower (50% vs. 83%) OS at 2 years for patients who received cetuximab and not cisplatin suggests that it can be estimated that it is not the reduction of irradiation dose that would be the cause of treatment failure but de-escalation of chemotherapy by substituting cisplatin with cetuximab [85,86,87]. If currently these biomarkers of radiosensitivity have no implications in the clinical decision, the de-escalation of chemo-radiotherapy treatment in certain groups of patients being only the subject of clinical trials, miRNAs open new horizons in customizing treatments based on different radio-chemo-sensitivity [88,89,90].
The COVID-19 pandemic has brought back the need for an irradiation regimen to limit patient and staff exposure during prolonged treatment. Thus, hypofractionated accelerated chemoradiation was evaluated on a batch of 564 patients in the PET NECK study trial. Even if in this pandemic context, a moderately hypofractionated treatment regimen (fractions less than or equal to 2.4 Gy per fraction) is feasible for chemo-irradiation, if hypofractionated regimens are used such as 55Gy in 20 daily fractions, a careful prediction of the benefit and risk in terms of tumor control and the risk of toxicity is necessary. In the context of an accelerated implementation of hypofractionation schemes, the development of new radiosensitivity biomarkers is essential [91,92,93].
Immunotherapy with immune checkpoint inhibitors (ICIs) has recently become the therapeutic standard in recurrent and metastatic HNSCC. The interest for the addition of radiotherapy in order to potentiate the effect of radiotherapy is increased considering the relatively low response rate. The mechanisms involved are multiple, with both theories on the immunostimulatory and immunosuppressive effect of tumor irradiation and tumor microenvironment existing. The implications of other factors including the microbiome in the synergistic effect of radiotherapy and immunotherapy are still little known. In the context of this new possible indication of radiotherapy, the identification of biomarkers both for the response to treatment with ICI or for radiotherapy–immunotherapy combination is one of maximum interest [94,95].
Analyzing data obtained from 515 HNSCC samples and 44 normal tissues, Luo et al. highlights deregulated miRNAs for both positive and negative HPV HNSCC cases [96]. In the patient lot including 97 HPV + patients, 282 miRNAs were identified and after statistical analysis, a 7 miRNA signature was considered proper for its prognostic value. In the lot including 418 HPV− HNSCC patients, among 289 miRNAs, 6 were included in the prognostic signature. Importantly, the signature of miRNAs for the two types of HNSCC is completely different. In the HPV+ group, miRNAs associated with the unfavorable prognosis are negatively correlated with CD8+ T lymphocytes. MiRNAs associated with a better OS have been associated with NK cells and T regulatory cells (Tregs). In the HPV− tumors group, miR-605-5p was associated with CD8 + T cells, activated CD4 T cells (considered tumor suppressors), and M1 macrophages. MiR-135-3p was associated with a better survival and it was negatively correlated with M2 macrophages. The authors also propose a risk score based on miRNA, mentioning the involvement of miRNAs correlated with CD8 + T lymphocytes and NK cells, Tregs, and T follicular helper cells (TFH) in the tumor microenvironment in pre-existing antitumor immune response. Cases of HPV− with a high risk of miRNAs could benefit more from target therapies and HPV+ patients with a low risk of miRNAs could benefit more from immunotherapy. The authors mention metabolic disorder as a possible cause of therapeutic failure in HPV− patients with an increased miRNA risk score [66,67,96].
Resistance to cisplatin is modulated by cancer-associated fibroblasts (CAF) which in turn regulate cell survival and proliferation in head and neck cancers. The mechanism of restoration of miR-196a sensitivity to cisplatin is related to depletion of miR-196a in CAF, and the authors demonstrate the exosomal capacity of miR-196a to be a biomarker of cisplatin sensitivity [97].
Factors involved in HNSCC radioresistance or radiosensitivity are numerous and are generally common to all cancers including traditional variable like cells lesions repair capacity, hypoxia, cell cycle position, and cell growth fraction, but also new “actors” such as hepatocyte growth factor receptor (HGFR) or programmed cell death protein 1 and programmed cell death protein 1 (PD-1) and its ligand, programmed death-ligand 1 (PD-L1) [98,99,100,101]. HNSCC is a malignancy of a special interest for the improvement of therapeutic ratio due to the importance of radiotherapy in both adjuvant treatment and definitive treatment in association with chemotherapy or in particular cases with target therapy. The proximity of radiosensitive organs which are exposed to potentially severe or even life-threatening toxicities by irradiation with tumoricidal doses is a serious argument both for improving the technique and conformity of delivered radiation dose as accurately as possible to target volumes in prescribed isodose but also for improving prediction by radiobiological modeling based on new evidence of radiosensitivity [79,80,81,82,83]. The RAS, EGFR, and PI3K/AKT pathways are involved in this process of modulating tumor radiosensitivity. EGFR is currently a clinically validated therapeutic target by using anti-EGFR monoclonal antibodies (cetuximab) in both metastatic and locally advanced settings but also has prognostic value, increased EGFR expression being associated with radioresistance and therapeutic failure. The TP53 suppressor gene, a regulator of genome stability and consequently a manipulator of DNA degradation, is mutant in 40–70% of HNSCC and thus inactivates the product or protein with effect on the cell cycle. Tumor hypoxia induces neovascularization and, by modulating the response to DNA damage, indirectly influences intrinsic radiosensitivity [91,92]. The mutation in the TP53 gene leads to the inactivation of its protein (p53) product and the change in p53 leads to an impaired ability to stop the cell cycle and inhibit apoptosis. Hypoxia-induced neovascularization and epithelial-mesenchymal transition (EMT), a phenomenon that modulates invasion and resistance to apoptosis, are also involved in tumor radioresistance in HNSCC. Cancer stem cell (SCC) populations, by their ability to renew and proliferate indefinitely and the potential for differentiation, are also associated with radioresistance [102,103,104]. A signature consisting of 4 and 12 miRNAs associated with TP53 were identified in HNSCC as being correlated with non-recurrent survival and cancer-specific survival, respectively. MiR-96-5p is overexpressed in tumors that show a p53 mutant, being involved in chemoresistance and radioresistance by activating the PI3K-AKT pathway [105,106,107].
The role of miRNAs in HNSCC radioresistance phenomena has been demonstrated for a group of miR-16, miR-29b, miR-1254, and miR-150 up-regulated miRNAs and the down-regulated let-7e miRNA for the situation where radiosensitivity is not mediated by ATM gene [79,81,89]. MiR-196a and miR-1323 are considered to have oncogenic potential and down-regulation of miR-205 and up-regulation of miR-96 have been associated with radioresistance but through different signaling pathways. Of note is the different effect that the same miRNA has in two different anatomical localizations of cancer for the same histopathological tumoral type. The low level of miR-203 predicts an unfavorable prognosis and early recurrence after radiotherapy in laryngeal cancer and the same miRNA is associated with radiosensitivity in nasopharyngeal cancer cell populations [66,73,89,90]. Given the need for a validated biomarker to be accessible and evaluable in dynamics, the conceptual use of miRNAs as biomarkers obtained from body fluids is a goal of translational research in personalized medicine and particularly in oncology. One of the most studied miRNAs that can be obtained from serum, plasma, or saliva is miR-21. In head and neck cancers, the high expression of this miRNA is associated with an unfavorable prognosis, the risk of distant metastasis. As demonstrated by a study that included 50 HNSCC patients, elevated postoperative miR-21 levels were associated with unfavorable 1-year survival and this observation offers the opportunity for miRNAs to be used as biomarkers in head and neck cancer surgery [89,107]. Down-regulated serum miR-9 has been associated with recurrence and metastasis in nasopharyngeal cancer and miR-31 is involved in the regulation of the hypoxia pathway, a well-known radioresistance factor. A low level of miR-31 in the blood was also observed in nasopharyngeal cancer compared to the level of this miRNA in normal nasopharyngeal tissues. HPV status with an essential role in radiosensitivity of head and neck cancers can be differentiated based on miRNA (miR-9, miR-122, miR-124, miR-134) in p16 positive and negative, as was demonstrated by Salazar-Ruales et al. and Wan et al. [97,105,106,108,109,110]. In oral cancer, miR-802 was identified as down-regulated in approximately 60% of cases using this level of miRNA in normal tissue as a reference. For a relatively “orphan” tumor subtype in terms of systemic treatments with curative potential (cystic adenoid carcinoma of the head and neck), it has been shown that high levels of miR-374c are associated with reduced recurrence rate and miR-21 inhibitors in association with sinvastatin have shown antiproliferative potential in lung metastases of cystic adenoid carcinoma of the salivary glands [89,97,111,112].
Without proposing to include the vast number of research papers that analyze the involvement of miRNAs in the radiosensitivity of head and neck cancers, we present in a table the main studies in the field, also mentioning the mechanism of action, binding site, and the radiosensitizing or the radio-resistance augmentation effect for each mentioned miRNA. (Table 1).

7. Conclusions

MiRNAs are currently intensively evaluated as potential biomarkers in numerous diseases and cancers. Determining the ability of miRNAs to modulate or predict radiosensitivity and resistance, to anticipate the risk of recurrence and metastasis, and to differentiate different tumor subtypes is based on multiple mechanisms by which mRNAs control proliferation and apoptosis, interact with cell cycle phases, and mechanisms by which miRNAs act as oncogenes with the potential to influence invasion promotion or tumor suppression. A refining of biomarkers of radiosensitivity miRNAs with clinical application in head and neck cancers can lead to a personalization of radiotherapy by anticipating the risk of toxicity, locoregional tumor control, recurrence, and metastasis. The potential to be an intrinsic predictor of sensitivity to chemotherapy may also guide the decision to choose an escalation or de-escalation of concurrent or sequential systemic treatment. Choosing the total irradiation dose, the dose per fraction, the fractionation scheme, and refining the dose-volume constraints according to the radiosensitivity of each tissue type estimated on a case-by-case basis by miRNAs profile are possible concepts and may be the basis for radiotherapy customization and radiobiology of head and neck cancers in the near future. miRNAs will be able to answer questions about the groups that will benefit from de-escalation of treatment and miRNA signatures could stratify patients who will benefit from hypofractionated radiotherapy and we can anticipate that they will answer the controversies regarding synergistic irradiation with immunotherapy in HNSCC.

Author Contributions

Conceptualization, methodology, validation, writing—original draft preparation, writing—review and editing, supervision, C.C.M., R.I.I. and D.P.T.I. These authors contributed equally to this work. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Epidemiology and Risk Factors for Head and Neck Cancer, Global Cancer Observatory. International Agency for Research on Cancer. World Health Organization. Available online: https://gco.iarc.fr/ (accessed on 6 June 2021).
  2. Brockstein, B.; Haraf, D.J.; Rademaker, A.W.; Kies, M.S.; Stenson, K.M.; Rosen, F.; Mittal, B.B.; Pelzer, H.; Fung, B.B.; Witt, M.E.; et al. Patterns of failure, prognostic factors and survival in locoregionally advanced head and neck cancer treated with concomitant chemoradiotherapy: A 9-year, 337-patient, multi-institutional experience. Ann Oncol. 2004, 15, 1179–1186. [Google Scholar] [CrossRef]
  3. Vigneswaran, N.; Williams, M.D. Epidemiologic trends in head and neck cancer and aids in diagnosis. Oral Maxillofac. Surg. Clin. N. Am. 2014, 26, 123–141. [Google Scholar] [CrossRef]
  4. Chang, J.H.; Wu, C.C.; Yuan, K.S.; Wu, A.T.H.; Wu, S.Y. Locoregionally recurrent head and neck squamous cell carcinoma: Incidence, survival, prognostic factors, and treatment outcomes. Oncotarget 2017, 8, 55600–55612. [Google Scholar] [CrossRef] [Green Version]
  5. Langendijk, J.A.; Doornaert, P.; Verdonck-de Leeuw, I.M.; Leemans, C.R.; Aaronson, N.K.; Slotman, B.J. Impact of late treatment-related toxicity on quality of life among patients with head and neck cancer treated with radiotherapy. J. Clin. Oncol. 2008, 26, 3770–3776. [Google Scholar] [CrossRef]
  6. Iqbal, M.S.; Chaw, C.; Kovarik, J.; Aslam, S.; Jackson, A.; Kelly, J.; Dobrowsky, W.; Kelly, C. Primary Concurrent Chemoradiation in Head and Neck Cancers with Weekly Cisplatin Chemotherapy: Analysis of Compliance, Toxicity and Survival. Int. Arch. Otorhinolaryngol. 2017, 21, 171–177. [Google Scholar] [CrossRef] [Green Version]
  7. Pardo-Reoyo, S.; Roig-Lopez, J.L.; Yang, E.S. Potential biomarkers for radiosensitivity in head and neck cancers. Ann. Transl. Med. 2016, 4, 524. [Google Scholar] [CrossRef] [Green Version]
  8. Bourhis, J.; Le Maître, A.; Baujat, B.; Audry, H.; Pignon, J.P. Meta-Analysis of Chemotherapy in Head, Neck Cancer Collaborative Group, Meta-Analysis of Radiotherapy in Carcinoma of Head, Neck Collaborative Group, Meta-Analysis of Chemotherapy in Nasopharynx Carcinoma Collaborative Group. Curr. Opin. Oncol. 2007, 19, 188. [Google Scholar] [CrossRef]
  9. Zhao, L.; Bode, A.M.; Cao, Y.; Dong, Z. Regulatory mechanisms and clinical perspectives of miRNA in tumor radiosensitivity. Carcinogenesis 2012, 33, 2220–2227. [Google Scholar] [CrossRef]
  10. Jemal, A.; Bray, F.; Center, M.M.; Ferlay, J.; Ward, E.; Forman, D. Global cancer statistics. CA Cancer J. Clin. 2011, 61, 69–90. [Google Scholar] [CrossRef] [Green Version]
  11. Jemal, A.; Siegel, R.; Ward, E.; Murray, T.; Xu, J.; Thun, M.J. Cancer statistics, 2007. CA Cancer J. Clin. 2007, 57, 43–66. [Google Scholar] [CrossRef]
  12. WHO. Head and Neck Cancer. Union for International Cancer Control. 2014 Review of Cancer Medicines on the WHO List of Essential Medicines. Available online: https://www.who.int/selection_medicines/committees/expert/20/applications/HeadNeck.pdf. (accessed on 20 December 2021).
  13. Shaw, R.; Beasley, N. Aetiology and risk factors for head and neck cancer: United Kingdom National Multidisciplinary Guidelines. J. Laryngol. Otol. 2016, 130 (Suppl. 2), S9–S12. [Google Scholar] [CrossRef] [PubMed]
  14. Aupérin, A. Epidemiology of head and neck cancers: An update. Curr. Opin. Oncol. 2020, 32, 178–186. [Google Scholar] [CrossRef] [PubMed]
  15. Yu, S.S.; Cirillo, N. The molecular markers of cancer stem cells in head and neck tumors. J. Cell Physiol. 2020, 235, 65–73. [Google Scholar] [CrossRef] [PubMed]
  16. Leinung, M.; Ernst, B.; Döring, C.; Wagenblast, J.; Tahtali, A.; Diensthuber, M.; Stöver, T.; Geissler, C. Expression of ALDH1A1 and CD44 in primary head and neck squamous cell carcinoma and their value for carcinogenesis, tumor progression and cancer stem cell identification. Oncol. Lett. 2015, 10, 2289–2294. [Google Scholar] [CrossRef] [PubMed]
  17. Zhang, Q.; Shi, S.; Yen, Y.; Brown, J.; Ta, J.Q.; Le, A.D. A subpopulation of CD133(+) cancer stem-like cells characterized in human oral squamous cell carcinoma confer resistance to chemotherapy. Cancer Lett. 2010, 289, 151–160. [Google Scholar] [CrossRef] [PubMed]
  18. Krishnamurthy, S.; Nör, J.E. Head and neck cancer stem cells. J. Dent. Res. 2012, 91, 334–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Khariwala, S.S.; Ma, B.; Ruszczak, C.; Carmella, S.G.; Lindgren, B.; Hatsukami, D.K.; Hecht, S.S.; Stepanov, I. High Level of Tobacco Carcinogen-Derived DNA Damage in Oral Cells Is an Independent Predictor of Oral/Head and Neck Cancer Risk in Smokers. Cancer Prev. Res. 2017, 10, 507–513. [Google Scholar] [CrossRef] [Green Version]
  20. Brooks, P.J.; Theruvathu, J.A. DNA adducts from acetaldehyde: Implications for alcohol-related carcinogenesis. Alcohol 2005, 35, 187–193. [Google Scholar] [CrossRef]
  21. Michaud, D.S.; Langevin, S.M.; Eliot, M.; Nelson, H.H.; Pawlita, M.; McClean, M.D.; Kelsey, K.T. High-risk HPV types and head and neck cancer. Int. J. Cancer 2014, 135, 1653–1661. [Google Scholar] [CrossRef]
  22. Scheffner, M.; Huibregtse, J.M.; Vierstra, R.D.; Howley, P.M. The HPV-16 E6 and E6-AP complex functions as a ubiquitin-protein ligase in the ubiquitination of p53. Cell 1993, 75, 495–505. [Google Scholar] [CrossRef]
  23. Johnson, D.E.; Burtness, B.; Leemans, C.R.; Lui, V.W.Y.; Bauman, J.E.; Grandis, J.R. Head and neck squamous cell carcinoma. Nat. Rev. Dis. Primers 2020, 6, 92. [Google Scholar] [CrossRef] [PubMed]
  24. Hsu, T.I.; Hsu, C.H.; Lee, K.H.; Lin, J.T.; Chen, C.S.; Chang, K.C.; Su, C.Y.; Hsiao, M.; Lu, P.J. MicroRNA-18a is elevated in prostate cancer and promotes tumorigenesis through suppressing STK4 in vitro and in vivo. Oncogenesis 2014, 3, e99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Morgan, E.L.; Patterson, M.R.; Ryder, E.L.; Lee, S.Y.; Wasson, C.W.; Harper, K.L.; Li, Y.; Griffin, S.; Blair, G.E.; Whitehouse, A.; et al. MicroRNA-18a targeting of the STK4/MST1 tumour suppressor is necessary for transformation in HPV positive cervical cancer. PLoS Pathog. 2020, 16, e1008624. [Google Scholar] [CrossRef] [PubMed]
  26. Klein, J.D.; Grandis, J.R. The molecular pathogenesis of head and neck cancer. Cancer Biol. Ther. 2010, 9, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Shi, X.; Luo, J.; Weigel, K.J.; Hall, S.C.; Du, D.; Wu, F.; Rudolph, M.C.; Zhou, H.; Young, C.D.; Wang, X.-J. Cancer-Associated Fibroblasts Facilitate Squamous Cell Carcinoma Lung Metastasis in Mice by Providing TGFβ-Mediated Cancer Stem Cell Niche. Front. Cell Dev. Biol. 2021, 9, 668164. [Google Scholar] [CrossRef]
  28. Acunzo, M.; Croce, C.M. Downregulation of miR-15a and miR-16-1 at 13q14 in Chronic Lymphocytic Leukemia. Clin. Chem. 2016, 62, 655–656. [Google Scholar] [CrossRef] [Green Version]
  29. Jérôme, T.; Laurie, P.; Louis, B.; Pierre, C. Enjoy the Silence: The Story of let-7 MicroRNA and Cancer. Curr. Genomics. 2007, 8, 229–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Hui, A.B.Y.; Lenarduzzi, M.; Krushel, T.; Waldron, L.; Pintilie, M.; Shi, W.; Perez-Ordonez, B.; Jurisica, I.; O’sullivan, B.; Waldrom, J.; et al. Comprehensive microRNA profiling for head and neck squamous cell carcinomas. Clin. Cancer Res. 2009, 16, 1129–1139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Spadaccino, F.; Gigante, M.; Netti, G.; Rocchetti, M.; Franzin, R.; Gesualdo, L.; Castellano, G.; Stallone, G.; Ranieri, E. The Ambivalent Role of miRNAs in Carcinogenesis: Involvement in Renal Cell Carcinoma and Their Clinical Applications. Pharmaceuticals 2021, 14, 322. [Google Scholar] [CrossRef]
  32. Cai, Y.; Dodhia, S.; Su, G.H. Dysregulations in the PI3K pathway and targeted therapies for head and neck squamous cell carcinoma. Oncotarget 2017, 8, 22203–22217. [Google Scholar] [CrossRef] [Green Version]
  33. Alural, B.; Genc, S.; Haggarty, S.J. Diagnostic and therapeutic potential of microRNAs in neuropsychiatric disorders: Past, present, and future. Prog. Neuropsychopharmacol Biol. Psychiatry 2017, 73, 87–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Kishore, A.; Borucka, J.; Petrkova, J.; Petrek, M. Novel insights into miRNA in lung and heart inflammatory diseases. Mediat. Inflamm. 2014, 2014, 259131. [Google Scholar] [CrossRef] [PubMed]
  35. 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] [PubMed] [Green Version]
  36. Bommer, G.T.; Gerin, I.; Feng, Y.; Kaczorowski, A.J.; Kuick, R.; Love, R.E.; Zhai, Y.; Giordano, T.J.; Qin, Z.S.; Moore, B.B.; et al. p53-mediated activation of miRNA34 candidate tumor-suppressor genes. Curr. Biol. 2007, 17, 1298–1307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Amit, M.; Takahashi, H.; Dragomir, M.P.; Lindemann, A.; Gleber-Netto, F.O.; Pickering, C.R.; Anfossi, S.; Osman, A.A.; Cai, Y.; Wang, R.; et al. Loss of p53 drives neuron reprogramming in head and neck cancer. Nature 2020, 578, 449–454. [Google Scholar] [CrossRef]
  38. Yamakuchi, M.; Lotterman, C.D.; Bao, C.; Hruban, R.H.; Karim, B.; Mendell, J.T.; Huso, D.; Lowenstein, C.J. P53-induced microRNA-107 inhibits HIF-1 and tumor angiogenesis. Proc. Natl. Acad. Sci. USA 2010, 107, 6334–6339. [Google Scholar] [CrossRef] [Green Version]
  39. Braun, C.J.; Zhang, X.; Savelyeva, I.; Wolff, S.; Moll, U.M.; Schepeler, T.; Ørntoft, T.F.; Andersen, C.L.; Dobbelstein, M. p53-Responsive micrornas 192 and 215 are capable of inducing cell cycle arrest. Cancer Res. 2008, 68, 10094–10104. [Google Scholar] [CrossRef] [Green Version]
  40. Kanchan, R.K.; Siddiqui, J.A.; Mahapatra, S.; Batra, S.K.; Nasser, M.W. microRNAs Orchestrate Pathophysiology of Breast Cancer Brain Metastasis: Advances in Therapy. Mol. Cancer 2020, 19, 29. [Google Scholar] [CrossRef] [Green Version]
  41. Saleh, A.D.; Cheng, H.; Martin, S.E.; Si, H.; Ormanoglu, P.; Carlson, S.; Clavijo, P.E.; Yang, X.; Das, R.; Cornelius, S.; et al. Integrated Genomic and Functional microRNA Analysis Identifies miR-30-5p as a Tumor Suppressor and Potential Therapeutic Nanomedicine in Head and Neck Cancer. Clin. Cancer Res. 2019, 25, 2860–2873. [Google Scholar] [CrossRef] [Green Version]
  42. Dong, P.; Konno, Y.; Watari, H.; Hosaka, M.; Noguchi, M.; Sakuragi, N. The impact of microRNA-mediated PI3K/AKT signaling on epithelial-mesenchymal transition and cancer stemness in endometrial cancer. J. Transl. Med. 2014, 12, 231. [Google Scholar] [CrossRef]
  43. Fang, Y.; Xue, J.L.; Shen, Q.; Chen, J.; Tian, L. MicroRNA-7 inhibits tumor growth and metastasis by targeting the phosphoinositide 3-kinase/Akt pathway in hepatocellular carcinoma. Hepatology 2012, 55, 1852–1862, Erratum in Hepatology 2014, 59, 1216. [Google Scholar] [CrossRef] [PubMed]
  44. Ardekani, A.M.; Naeini, M.M. The Role of MicroRNAs in Human Diseases. Avicenna J. Med. Biotechnol. 2010, 2, 161–179. [Google Scholar] [PubMed]
  45. Aravin, A.; Tuschl, T. Identification and characterization of small RNAs involved in RNA silencing. FEBS Lett. 2005, 579, 5830–5840. [Google Scholar] [CrossRef] [Green Version]
  46. Pekkola-Heino, K.; Jaakkola, M.; Kulmala, J.; Grénman, R. Comparison of cellular radiosensitivity between different localizations of head and neck squamous-cell carcinoma. J. Cancer Res. Clin. Oncol. 1995, 121, 452–456. [Google Scholar] [CrossRef] [PubMed]
  47. Lhakhang, T.W.; Chaudhry, M.A. Interactome of Radiation-Induced microRNA-Predicted Target Genes. Int. J. Genom. 2012, 2021, 569731. [Google Scholar] [CrossRef] [Green Version]
  48. Petrović, N.; Stanojković, T.P.; Nikitović, M. MicroRNAs in prostate cancer following radiotherapy: Towards predicting response to radiation treatment. Curr. Med. Chem. 2021. [Google Scholar] [CrossRef]
  49. Metheetrairut, C.; Slack, F.J. MicroRNAs in the ionizing radiation response and in radiotherapy. Curr. Opin. Genet. Dev. 2013, 23, 12–19. [Google Scholar] [CrossRef] [Green Version]
  50. Chen, Y.; Cui, J.; Gong, Y.; Wei, S.; Wei, Y.; Yi, L. MicroRNA: A novel implication for damage and protection against ionizing radiation. Env. Sci. Pollut. Res. Int. 2021, 28, 15584–15596. [Google Scholar] [CrossRef]
  51. Pajic, M.; Froio, D.; Daly, S.; Doculara, L.; Millar, E.; Graham, P.H.; Drury, A.; Steinmann, A.; de Bock, C.E.; Boulghourjian, A.; et al. miR-139-5p Modulates Radiotherapy Resistance in Breast Cancer by Repressing Multiple Gene Networks of DNA Repair and ROS Defense. Cancer Res. 2018, 78, 501–515. [Google Scholar] [CrossRef] [Green Version]
  52. Yoo, H.Y.; Kumagai, A.; Shevchenko, A.; Shevchenko, A.; Dunphy, W.G. Ataxia-telangiectasia mutated (ATM)-dependent activation of ATR occurs through phosphorylation of TopBP1 by ATM. J. Biol. Chem. 2007, 282, 17501–17506. [Google Scholar] [CrossRef] [Green Version]
  53. Zheng, L.; Zhang, Y.; Liu, Y.; Zhou, M.; Lu, Y.; Yuan, L.; Zhang, C.; Hong, M.; Wang, S.; Li, X. MiR-106b induces cell radioresistance via the PTEN/PI3K/AKT pathways and p21 in colorectal cancer. J. Transl. Med. 2015, 13, 252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Tang, Y.; Wallace, M.; Sanchez-Gurmaches, J.; Hsiao, W.Y.; Li, H.; Lee, P.L.; Vernia, S.; Metallo, C.M.; Guertin, D.A. Adipose tissue mTORC2 regulates ChREBP-driven de novo lipogenesis and hepatic glucose metabolism. Nat. Commun. 2016, 7, 11365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Dittmann, K.; Mayer, C.; Czemmel, S.; Huber, S.M.; Rodemann, H.P. New roles for nuclear EGFR in regulating the stability and translation of mRNAs associated with VEGF signaling. PLoS ONE 2017, 12, e0189087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Shi, L.; Zhang, S.; Wu, H.; Zhang, L.; Dai, X.; Hu, J.; Xue, J.; Liu, T.; Liang, Y.; Wu, G. MiR-200c increases the radiosensitivity of non-small-cell lung cancer cell line A549 by targeting VEGF-VEGFR2 pathway. PLoS ONE 2013, 8, e78344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Lun, W.; Wu, X.; Deng, Q.; Zhi, F. MiR-218 regulates epithelial-mesenchymal transition and angiogenesis in colorectal cancer via targeting CTGF. Cancer Cell Int. 2018, 18, 83. [Google Scholar] [CrossRef] [PubMed]
  58. Liu, S.; Pan, X.; Yang, Q.; Wen, L.; Jiang, Y.; Zhao, Y.; Li, G. MicroRNA-18a enhances the radiosensitivity of cervical cancer cells by promoting radiation-induced apoptosis. Oncol. Rep. 2015, 33, 2853–2862. [Google Scholar] [CrossRef] [Green Version]
  59. Yuan, W.; Xiaoyun, H.; Haifeng, Q.; Jing, L.; Weixu, H.; Ruofan, D.; Jinjin, Y.; Zongji, S. MicroRNA-218 enhances the radiosensitivity of human cervical cancer via promoting radiation induced apoptosis. Int. J. Med. Sci. 2014, 11, 691–696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Lu, H.; He, Y.; Lin, L.; Qi, Z.; Ma, L.; Li, L.; Su, Y. Long non-coding RNA MALAT1 modulates radiosensitivity of HR-HPV+ cervical cancer via sponging miR-145. Tumour Biol. 2016, 37, 1683–1691. [Google Scholar] [CrossRef] [PubMed]
  61. Maier, P.; Hartmann, L.; Wenz, F.; Herskind, C. Cellular Pathways in Response to Ionizing Radiation and Their Targetability for Tumor Radiosensitization. Int. J. Mol. Sci. 2016, 17, 102. [Google Scholar] [CrossRef] [Green Version]
  62. Piotto, C.; Biscontin, A.; Millino, C.; Mognato, M. Functional validation of miRNAs targeting genes of DNA double-strand break repair to radiosensitize non-small lung cancer cells. Biochim. Biophys. Acta Gene Regul. Mech. 2018, 1861, 1102–1118. [Google Scholar] [CrossRef]
  63. Zhang, Y.; Zheng, L.; Ding, Y.; Li, Q.; Wang, R.; Liu, T.; Sun, Q.; Yang, H.; Peng, S.; Wang, W.; et al. MiR-20a Induces Cell Radioresistance by Activating the PTEN/PI3K/Akt Signaling Pathway in Hepatocellular Carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2015, 92, 1132–1140. [Google Scholar] [CrossRef] [PubMed]
  64. Li, H.F.; Kim, J.S.; Waldman, T. Radiation-induced Akt activation modulates radioresistance in human glioblastoma cells. Radiat. Oncol. 2009, 4, 43. [Google Scholar] [CrossRef] [Green Version]
  65. Zhen, L.; Li, J.; Zhang, M.; Yang, K. MiR-10b decreases sensitivity of glioblastoma cells to radiation by targeting AKT. J. Biol. Res. 2016, 23, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. 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] [PubMed]
  67. Bartels, C.L.; Tsongalis, G.J. MicroRNAs: Novel biomarkers for human cancer. Clin. Chem. 2009, 55, 623–631. [Google Scholar] [CrossRef] [Green Version]
  68. Colpaert, R.M.W.; Calore, M. MicroRNAs in Cardiac Diseases. Cells. 2019, 8, 737. [Google Scholar] [CrossRef] [Green Version]
  69. Drury, R.E.; O’Connor, D.; Pollard, A.J. The Clinical Application of MicroRNAs in Infectious Disease. Front. Immunol. 2017, 8, 1182. [Google Scholar] [CrossRef] [PubMed]
  70. Mitchell, P.S.; Parkin, R.K.; Kroh, E.M.; Fritz, B.R.; Wyman, S.K.; Pogosova-Agadjanyan, E.L.; Peterson, A.; Noteboom, J.; O’Briant, K.C.; Allen, A.; et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc. Natl. Acad. Sci. USA 2008, 105, 10513–10518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Meiri, E.; Mueller, W.C.; Rosenwald, S.; Zepeniuk, M.; Klinke, E.; Edmonston, T.B.; Werner, M.; Lass, U.; Barshack, I.; Feinmesser, M.; et al. A second-generation microRNA-based assay for diagnosing tumor tissue origin. Oncologist 2012, 17, 801–812. [Google Scholar] [CrossRef] [Green Version]
  72. Lawrie, C.H.; Gal, S.; Dunlop, H.M.; Pushkaran, B.; Liggins, A.P.; Pulford, K.; Banham, A.H.; Pezzella, F.; Boultwood, J.; Wainscoat, J.S.; et al. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. Br. J. Haematol. 2008, 141, 672–675. [Google Scholar] [CrossRef] [PubMed]
  73. Vahabi, M.; Blandino, G.; Di Agostino, S. MicroRNAs in head and neck squamous cell carcinoma: A possible challenge as biomarkers, determinants for the choice of therapy and targets for personalized molecular therapies. Transl. Cancer Res. 2021, 10, 3090–3110. [Google Scholar] [CrossRef]
  74. Liu, X.; Wang, C.; Chen, Z.; Jin, Y.; Wang, Y.; Kolokythas, A.; Dai, Y.; Zhou, X. MicroRNA-138 suppresses epithelial-mesenchymal transition in squamous cell carcinoma cell lines. Biochem. J. 2011, 440, 23–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Sha, H.-H.; Wang, D.-D.; Chen, D.; Liu, S.-W.; Wang, Z.; Yan, D.-L.; Dong, S.-C.; Feng, J.-F. MiR-138: A promising therapeutic target for cancer. Tumor Biol. 2017. [Google Scholar] [CrossRef] [Green Version]
  76. Bao, L.; Miao, W.; Yu, Y. Reduced serum miR-138 is associated with poor prognosis of head and neck squamous cell carcinoma. Int. J. Clin. Exp. Pathol. 2017, 10, 10276–10281. [Google Scholar] [PubMed]
  77. Childs, G.; Fazzari, M.; Kung, G.; Fazzari, M.; Kung, G.; Kawachi, N.; Brandwein-Gensler, M.; McLemore, M.; Chen, Q.; Burk, R.D.; 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]
  78. Li, J.; Chen, W.; Luo, L.; Liao, L.; Deng, X.; Wang, Y. The microRNA miR-29c-5p inhibits cell proliferation and migration by targeting TMEM98 in head and neck carcinoma. Aging 2020, 13, 769–781. [Google Scholar] [CrossRef] [PubMed]
  79. Van Gestel, D.; Van Den Weyngaert, D.; Schrijvers, D.; Weyler, J.; Vermorken, J.B. Intensity-modulated radiotherapy in patients with head and neck cancer: A European single-centre experience. Br. J. Radiol. 2011, 84, 367–374. [Google Scholar] [CrossRef] [Green Version]
  80. Rühle, A.; Grosu, A.L.; Nicolay, N.H. De-Escalation Strategies of (Chemo)Radiation for Head-and-Neck Squamous Cell Cancers-HPV and Beyond. Cancers 2021, 13, 2204. [Google Scholar] [CrossRef]
  81. Tao, Y.; Auperin, A.; Blanchard, P.; Alfonsi, M.; Sun, X.S.; Rives, M.; Pointreau, Y.; Castelli, J.; Graff, P.; Wong Hee Kam, S.; et al. Concurrent cisplatin and dose escalation with intensity-modulated radiotherapy (IMRT) versus conventional radiotherapy for locally advanced head and neck squamous cell carcinomas (HNSCC): GORTEC 2004-01 randomized phase III trial. Radiother Oncol. 2020, 150, 18–25. [Google Scholar] [CrossRef]
  82. Wittekindt, C.; Klussmann, J.P. Tumor Staging and HPV-Related Oropharyngeal Cancer. Recent Results Cancer Res. 2017, 206, 123–133. [Google Scholar]
  83. Strojan, P.; Vermorken, J.B.; Beitler, J.J.; Saba, N.F.; Haigentz, M., Jr.; Bossi, P.; Worden, F.P.; Langendijk, J.A.; Eisbruch, A.; Mendenhall, W.M.; et al. Cumulative cisplatin dose in concurrent chemoradiotherapy for head and neck cancer: A systematic review. Head Neck 2016, 38 (Suppl 1), E2151–E21588. [Google Scholar] [CrossRef] [Green Version]
  84. Al-Mamgani, A.; de Ridder, M.; Navran, A.; Klop, W.M.; de Boer, J.P.; Tesselaar, M.E. The impact of cumulative dose of cisplatin on outcome of patients with head and neck squamous cell carcinoma. Eur. Arch Otorhinolaryngol. 2017, 274, 3757–3765. [Google Scholar] [CrossRef] [PubMed]
  85. Anderson, C.M.; Kimple, R.J.; Lin, A.; Karam, S.D.; Margalit, D.N.; Chua, M.L.K. De-Escalation Strategies in HPV-Associated Oropharynx Cancer-Are we Putting the Cart Before the Horse? Int. J. Radiat. Oncol. Biol. Phys. 2019, 104, 705–709. [Google Scholar] [CrossRef] [PubMed]
  86. Magrini, S.M.; Buglione, M.; Corvò, R.; Pirtoli, L.; Paiar, F.; Ponticelli, P.; Petrucci, A.; Bacigalupo, A.; Crociani, M.; Lastrucci, L.; et al. Cetuximab and Radiotherapy Versus Cisplatin and Radiotherapy for Locally Advanced Head and Neck Cancer: A Randomized Phase II Trial. J. Clin. Oncol. 2016, 34, 427–435. [Google Scholar] [CrossRef] [PubMed]
  87. Haddad, R.; Posner, M.; Hitt, R.; Cohen, E.; Schulten, J.; Lefebvre, J.-L.; Vermorken, J. Induction chemotherapy in locally advanced squamous cell carcinoma of the head and neck: Role, controversy, and future directions. Ann. Oncol. 2018, 29, 1130–1140. [Google Scholar] [CrossRef] [PubMed]
  88. Korpela, E.; Vesprini, D.; Liu, S.K. MicroRNA in radiotherapy: miRage or miRador? Br. J. Cancer 2015, 112, 777–782. [Google Scholar] [CrossRef] [Green Version]
  89. Ahmad, P.; Sana, J.; Slavik, M.; Slampa, P.; Smilek, P.; Slaby, O. MicroRNAs Involvement in Radioresistance of Head and Neck Cancer. Dis. Markers 2017, 2017, 8245345. [Google Scholar] [CrossRef] [Green Version]
  90. Nowicka, Z.; Stawiski, K.; Tomasik, B.; Fendler, W. Extracellular miRNAs as Biomarkers of Head and Neck Cancer Progression and Metastasis. Int. J. Mol. Sci. 2019, 20, 4799. [Google Scholar] [CrossRef] [Green Version]
  91. Mehanna, H.; McConkey, C.C.; Rahman, J.K.; Wong, W.L.; Smith, A.F.; Nutting, C.; Hartley, A.G.; Hall, P.; Hulme, C.; Patel, D.K.; et al. PET-NECK: A multicentre randomised Phase III non-inferiority trial comparing a positron emission tomography-computerised tomography-guided watch-and-wait policy with planned neck dissection in the management of locally advanced (N2/N3) nodal metastases in patients with squamous cell head and neck cancer. Health Technol. Assess. 2017, 21, 1–122. [Google Scholar] [PubMed] [Green Version]
  92. Huang, S.H.; O’Sullivan, B.; Su, J.; Ringash, J.; Bratman, S.V.; Kim, J.; Hosni, A.; Bayley, A.; Cho, J.; Giuliani, M.; et al. Hypofractionated radiotherapy alone with 2.4 Gy per fraction for head and neck cancer during the COVID-19 pandemic: The Princess Margaret experience and proposal. Cancer 2020, 126, 3426–3437. [Google Scholar] [CrossRef] [PubMed]
  93. Gupta, T.; Ghosh-Laskar, S.; Agarwal, J.P. Resource-sparing curative-intent hypofractionated-accelerated radiotherapy in head and neck cancer: More relevant than ever before in the COVID era. Oral Oncol. 2020, 111, 105045. [Google Scholar] [CrossRef] [PubMed]
  94. Biau, J.; Bourhis, J. Combining immunotherapy and radiotherapy in head and neck squamous cell cancers: Which perspectives? Curr. Opin. Oncol. 2020, 32, 196–202. [Google Scholar] [CrossRef] [PubMed]
  95. Plavc, G.; Jesenko, T.; Oražem, M.; Strojan, P. Challenges in Combining Immunotherapy with Radiotherapy in Recurrent/Metastatic Head and Neck Cancer. Cancers 2020, 12, 3197. [Google Scholar] [CrossRef]
  96. Luo, X.J.; Zheng, M.; Cao, M.X.; Zhang, W.L.; Huang, M.C.; Dai, L.; Tang, Y.L.; Liang, X.L. Distinguishable Prognostic miRNA Signatures of Head and Neck Squamous Cell Cancer With or Without HPV Infection. Front. Oncol. 2021, 10, 614487. [Google Scholar] [CrossRef] [PubMed]
  97. Salazar-Ruales, C.; Arguello, J.V.; López-Cortés, A.; Cabrera-Andrade, A.; García-Cárdenas, J.M.; Guevara-Ramírez, P.; Peralta, P.; Leone, P.E.; Paz-Y-Miño, C. Salivary MicroRNAs for Early Detection of Head and Neck Squamous Cell Carcinoma: A Case-Control Study in the High Altitude Mestizo Ecuadorian Population. BioMed Res Int. 2018, 2018, 9792730. [Google Scholar] [CrossRef] [Green Version]
  98. Menegakis, A.; Klompmaker, R.; Vennin, C.; Arbusà, A.; Damen, M.; van den Broek, B.; Zips, D.; van Rheenen, J.; Krenning, L.; Medema, R.H. Resistance of Hypoxic Cells to Ionizing Radiation Is Mediated in Part via Hypoxia-Induced Quiescence. Cells 2021, 10, 610. [Google Scholar] [CrossRef] [PubMed]
  99. Zölzer, F.; Streffer, C. Increased radiosensitivity with chronic hypoxia in four human tumor cell lines. Int. J. Radiat. Oncol. Biol. Phys. 2002, 54, 910–920. [Google Scholar] [CrossRef]
  100. Fiedler, M.; Weber, F.; Hautmann, M.G.; Haubner, F.; Reichert, T.E.; Klingelhöffer, C.; Schreml, S.; Meier, J.K.; Hartmann, A.; Ettl, T. Biological predictors of radiosensitivity in head and neck squamous cell carcinoma. Clin. Oral. Investig. 2018, 22, 189–200. [Google Scholar] [CrossRef]
  101. Chang, L.; Graham, P.H.; Hao, J.; Ni, J.; Bucci, J.; Cozzi, P.J.; Kearsley, J.H.; Li, Y. PI3K/Akt/mTOR pathway inhibitors enhance radiosensitivity in radioresistant prostate cancer cells through inducing apoptosis, reducing autophagy, suppressing NHEJ and HR repair pathways. Cell Death Dis. 2014, 5, e1437. [Google Scholar] [CrossRef]
  102. Horn, D.; Hess, J.; Freier, K.; Hoffmann, J.; Freudlsperger, C. Targeting EGFR-PI3K-AKT-mTOR signaling enhances radiosensitivity in head and neck squamous cell carcinoma. Expert Opin. Ther. Targets 2015, 19, 795–805. [Google Scholar] [CrossRef]
  103. Mohammadi, C.; Gholamzadeh Khoei, S.; Fayazi, N.; Mohammadi, Y.; Najafi, R. miRNA as promising theragnostic biomarkers for predicting radioresistance in cancer: A systematic review and meta-analysis. Crit. Rev. Oncol. Hematol. 2021, 157, 103183. [Google Scholar] [CrossRef] [PubMed]
  104. Steinbichler, T.B.; Alshaimaa, A.; Maria, M.V.; Daniel, D.; Herbert, R.; Jozsef, D.; Ira-Ida, S. Epithelial-mesenchymal crosstalk induces radioresistance in HNSCC cells. Oncotarget 2017, 9, 3641–3652, Correction in Oncotarget 2019, 10, 5889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Vahabi, M.; Pulito, C.; Sacconi, A.; Donzelli, S.; D’Andrea, M.; Manciocco, V.; Pellini, R.; Paci, P.; Sanguineti, G.; Strigari, L.; et al. miR-96-5p targets PTEN expression affecting radio-chemosensitivity of HNSCC cells. J. Exp. Clin. Cancer Res. 2019, 38, 141. [Google Scholar] [CrossRef] [Green Version]
  106. John, K.; Wu, J.; Lee, B.W.; Farah, C.S. MicroRNAs in Head and Neck Cancer. Int. J. Dent. 2013. [Google Scholar] [CrossRef]
  107. Luo, X.; Stock, C.; Burwinkel, B.; Brenner, H. Identification and evaluation of plasma microRNAs for early detection of colorectal cancer. PLoS ONE 2013, 8, e62880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. 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]
  109. Arantes, L.M.; Laus, A.C.; Melendez, M.E.; de Carvalho, A.C.; Sorroche, B.P.; De Marchi, P.R.M.; Evangelista, A.F.; Scapulatempo-Neto, C.; de Souza Viana, L.; Carvalho, A.L. MiR-21 as prognostic biomarker in head and neck squamous cell carcinoma patients undergoing an organ preservation protocol. Oncotarget 2017, 8, 9911–9921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. 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]
  111. Andreasen, S.; Tan, Q.; Agander, T.K.; Hansen, T.V.O.; Steiner, P.; Bjørndal, K.; Høgdall, E.; Larsen, S.R.; Erentaite, D.; Olsen, C.H.; et al. MicroRNA dysregulation in adenoid cystic carcinoma of the salivary gland in relation to prognosis and gene fusion status: A cohort study. Virchows Arch. 2018, 473, 329–340. [Google Scholar] [CrossRef]
  112. Wang, C.; Li, T.; Yan, F.; Cai, W.; Zheng, J.; Jiang, X.; Sun, J. Effect of simvastatin and microRNA-21 inhibitor on metastasis and progression of human salivary adenoid cystic carcinoma. Biomed Pharmacother. 2018, 105, 1054–1061. [Google Scholar] [CrossRef]
  113. Maia, D.; de Carvalho, A.C.; Horst, M.A.; Carvalho, A.L.; Scapulatempo-Neto, C.; Vettore, A.L. Expression of miR-296-5p as predictive marker for radiotherapy resistance in early-stage laryngeal carcinoma. J. Transl. Med. 2015, 13, 262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Liu, C.; Li, G.; Yang, N.; Su, Z.; Zhang, S.; Deng, T.; Ren, S.; Lu, S.; Tian, Y.; Liu, Y.; et al. miR-324-3p suppresses migration and invasion by targeting WNT2B in nasopharyngeal carcinoma. Cancer Cell Int. 2017, 17, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. de Jong, M.C.; Ten Hoeve, J.J.; Grénman, R.; Wessels, L.F.; Kerkhoven, R.; Te Riele, H.; van den Brekel, M.W.; Verheij, M.; Begg, A.C. Pretreatment microRNA Expression Impacting on Epithelial-to-Mesenchymal Transition Predicts Intrinsic Radiosensitivity in Head and Neck Cancer Cell Lines and Patients. Clin. Cancer Res. 2015, 21, 5630–5638. [Google Scholar] [CrossRef] [Green Version]
  116. Li, G.; Qiu, Y.; Su, Z.; Ren, S.; Liu, C.; Tian, Y.; Liu, Y. Genome-wide analyses of radioresistance-associated miRNA expression profile in nasopharyngeal carcinoma using next generation deep sequencing. PLoS ONE 2014, 9, e110099. [Google Scholar] [CrossRef]
  117. Long, D.; Xu, L.; Deng, Z.; Guo, D.; Zhang, Y.; Liu, Z.; Zhang, C. HPV16 E6 enhances the radiosensitivity in HPV-positive human head and neck squamous cell carcinoma by regulating the miR-27a-3p/SMG1 axis. Infect. Agents Cancer 2021, 16, 56. [Google Scholar] [CrossRef]
  118. Zhang, C.; Chen, H.; Deng, Z.; Long, D.; Xu, L.; Liu, Z. DGCR8/miR-106 Axis Enhances Radiosensitivity of Head and Neck Squamous Cell Carcinomas by Downregulating RUNX3. Front. Med. 2020, 7, 582097. [Google Scholar] [CrossRef] [PubMed]
  119. Qu, C.; Liang, Z.; Huang, J.; Zhao, R.; Su, C.; Wang, S.; Wang, X.; Zhang, R.; Lee, M.H.; Yang, H. MiR-205 determines the radioresistance of human nasopharyngeal carcinoma by directly targeting PTEN. Cell Cycle 2012, 11, 785–796. [Google Scholar] [CrossRef] [Green Version]
  120. Zhang, B.; Li, Y.; Hou, D.; Shi, Q.; Yang, S.; Li, Q. MicroRNA-375 Inhibits Growth and Enhances Radiosensitivity in Oral Squamous Cell Carcinoma by Targeting Insulin Like Growth Factor 1 Receptor. Cell. Physiol. Biochem. 2017, 42, 2105–2117. [Google Scholar] [CrossRef] [Green Version]
  121. Gee, H.E.; Camps, C.; Buffa, F.M.; Patiar, S.; Winter, S.C.; Betts, G.; Homer, J.; Corbridge, R.; Cox, G.; West, C.M.; et al. hsa-mir-210 is a marker of tumor hypoxia and a prognostic factor in head and neck cancer. Cancer 2010, 116, 2148–2158. [Google Scholar] [CrossRef]
  122. 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]
  123. 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]
  124. Feng, X.; Lv, W.; Wang, S.; He, Q. miR 495 enhances the efficacy of radiotherapy by targeting GRP78 to regulate EMT in nasopharyngeal carcinoma cells. Oncol. Rep. 2018, 40, 1223–1232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Huang, Y.; Tan, D.; Xiao, J.; Li, Q.; Zhang, X.; Luo, Z. miR-150 contributes to the radioresistance in nasopharyngeal carcinoma cells by targeting glycogen synthase kinase-3β. J. Cancer Res. Ther. 2018, 14, 111–118. [Google Scholar] [CrossRef]
  126. Qu, J.Q.; Yi, H.M.; Ye, X.; Li, L.N.; Zhu, J.F.; Xiao, T.; Yuan, L.; Li, J.Y.; Wang, Y.Y.; Feng, J.; et al. MiR-23a sensitizes nasopharyngeal carcinoma to irradiation by targeting IL-8/Stat3 pathway. Oncotarget 2015, 6, 28341–28356. [Google Scholar] [CrossRef] [Green Version]
  127. 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] [Green Version]
  128. Lin, M.Y.; Chang, Y.C.; Wang, S.Y.; Yang, M.H.; Chang, C.H.; Hsiao, M.; Kitsis, R.N.; Lee, Y.J. OncomiR miR-182-5p Enhances Radiosensitivity by Inhibiting the Radiation-Induced Antioxidant Effect through SESN2 in Head and Neck Cancer. Antioxidants 2021, 10, 1808. [Google Scholar] [CrossRef]
  129. Ma, H.; Lian, R.; Wu, Z.; Li, X.; Yu, W.; Shang, Y.; Guo, X. MiR-503 enhances the radiosensitivity of laryngeal carcinoma cells via the inhibition of WEE1. Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Medicine. 2017, 39, 1010428317706224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Du, T.; Jiang, J.; Chen, Y.; Zhang, N.; Chen, G.; Wang, X.; Long, X.; Feng, X. MiR-138-1-3p alters the stemness and radiosensitivity of tumor cells by targeting CRIPTO and the JAK2/STAT3 pathway in nasopharyngeal carcinoma. Ann. Transl. Med. 2021, 9, 485. [Google Scholar] [CrossRef]
  131. Xie, F.; Xiao, W.; Tian, Y.; Lan, Y.; Zhang, C.; Bai, L. MicroRNA-195-3p inhibits cyclin dependent kinase 1 to induce radiosensitivity in nasopharyngeal carcinoma. Bioengineered 2021, 12, 7325–7334. [Google Scholar] [CrossRef]
  132. 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-κB axis. J. Exp. Clin. Cancer Res. 2016, 35, 188. [Google Scholar] [CrossRef]
  133. Wu, Q.; Han, T.; Sheng, X.; Zhang, N.; Wang, P. Downregulation of EB virus miR-BART4 inhibits proliferation and aggressiveness while promoting radiosensitivity of nasopharyngeal carcinoma. Biomed. Pharmacother. 2018, 108, 741–751. [Google Scholar] [CrossRef] [PubMed]
  134. 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] [PubMed]
  135. Lee, K.H.; Lin, F.C.; Hsu, T.I.; Lin, J.T.; Guo, J.H.; Tsai, C.H.; Lee, Y.C.; Lee, Y.C.; Chen, C.L.; Hsiao, M.; et al. MicroRNA-296-5p (miR-296-5p) functions as a tumor suppressor in prostate cancer by directly targeting Pin1. Biochim. Biophys. Acta 2014, 1843, 2055–2066. [Google Scholar] [CrossRef] [Green Version]
  136. Dharap, A.; Pokrzywa, C.; Murali, S.; Pandi, G.; Vemuganti, R. MicroRNA miR-324-3p induces promoter-mediated expression of RelA gene. PLoS ONE 2013, 8, e79467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Wang, C.; Wang, X.; Liang, H.; Wang, T.; Yan, X.; Cao, M.; Wang, N.; Zhang, S.; Zen, K.; Zhang, C.; et al. miR-203 inhibits cell proliferation and migration of lung cancer cells by targeting PKCα. PLoS ONE 2013, 8, e73985. [Google Scholar] [CrossRef]
  138. Wang, L.; Yang, G.; Zhu, X.; Wang, Z.; Wang, H.; Bai, Y.; Sun, P.; Peng, L.; Wei, W.; Chen, G.; et al. miR-93-3p inhibition suppresses clear cell renal cell carcinoma proliferation, metastasis and invasion. Oncotarget 2017, 8, 82824–82834. [Google Scholar] [CrossRef] [Green Version]
  139. Du, E.; Cao, Y.; Feng, C.; Lu, J.; Yang, H.; Zhang, Y. The Possible Involvement of miR-371a-5p Regulating XIAP in the Pathogenesis of Recurrent Pregnancy Loss. Reprod. Sci. 2019, 26, 1468–1475. [Google Scholar] [CrossRef]
  140. Bartoszewska, S.; Cabaj, A.; Dąbrowski, M.; Collawn, J.F.; Bartoszewski, R. miR-34c-5p modulates X-box-binding protein 1 (XBP1) expression during the adaptive phase of the unfolded protein response. FASEB J. 2019, 33, 11541–11554. [Google Scholar] [CrossRef]
  141. Xie, H.; Liu, M.; Jin, Y.; Lin, H.; Zhang, Y.; Zheng, S. miR-1323 suppresses bone mesenchymal stromal cell osteogenesis and fracture healing via inhibiting BMP4/SMAD4 signaling. J. Orthop. Surg. Res. 2020, 15, 237. [Google Scholar] [CrossRef]
  142. Li, Y.; Han, W.; Ni, T.T.; Lu, L.; Huang, M.; Zhang, Y.; Cao, H.; Zhang, H.Q.; Luo, W.; Li, H. Knockdown of microRNA-1323 restores sensitivity to radiation by suppression of PRKDC activity in radiation-resistant lung cancer cells. Oncol. Rep. 2015, 33, 2821–2828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Lu, B.; Feng, Z.; Fan, B.; Shi, Y. Blocking miR-27a-3p sensitises Taxol resistant osteosarcoma cells through targeting Fbxw7. Bull. Cancer 2021, 108, 596–604. [Google Scholar] [CrossRef]
  144. Sharma, A.; Kumar, M.; Aich, J.; Hariharan, M.; Brahmachari, S.K.; Agrawal, A.; Ghosh, B. Posttranscriptional regulation of interleukin-10 expression by hsa-miR-106a. Proc. Natl. Acad. Sci. USA 2009, 106, 5761–5766, Erratum in Proc. Natl. Acad. Sci. USA 2020, 117, 12498–12499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Hong, L.; Peng, S.; Li, Y.; Fang, Y.; Wang, Q.; Klausen, C.; Yin, C.; Wang, S.; Leung, P.C.K.; Yang, X. miR-106a Increases Granulosa Cell Viability and Is Downregulated in Women With Diminished Ovarian Reserve. J. Clin. Endocrinol. Metab. 2018, 103, 2157–2166. [Google Scholar] [CrossRef] [PubMed]
  146. Liu, S.L.; Sui, Y.F.; Lin, M.Z. MiR-375 is epigenetically downregulated due to promoter methylation and modulates multi-drug resistance in breast cancer cells via targeting YBX1. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 3223–3229. [Google Scholar] [PubMed]
  147. Zheng, L.; Qi, T.; Yang, D.; Qi, M.; Li, D.; Xiang, X.; Huang, K.; Tong, Q. microRNA-9 suppresses the proliferation, invasion and metastasis of gastric cancer cells through targeting cyclin D1 and Ets1. PLoS ONE 2013, 8, e55719. [Google Scholar] [CrossRef]
  148. Zhang, Z.; Sun, H.; Dai, H.; Walsh, R.M.; Imakura, M.; Schelter, J.; Burchard, J.; Dai, X.; Chang, A.N.; Diaz, R.L.; et al. MicroRNA miR-210 modulates cellular response to hypoxia through the MYC antagonist MNT. Cell Cycle 2009, 8, 2756–2768. [Google Scholar] [CrossRef] [Green Version]
  149. Wang, K.; Li, J.; Guo, H.; Xu, X.; Xiong, G.; Guan, X.; Liu, B.; Li, J.; Chen, X.; Yang, K.; et al. MiR-196a binding-site SNP regulates RAP1A expression contributing to esophageal squamous cell carcinoma risk and metastasis. Carcinogenesis 2012, 33, 2147–2154. [Google Scholar] [CrossRef] [PubMed]
  150. Mishra, P.J.; Humeniuk, R.; Mishra, P.J.; Longo-Sorbello, G.S.; Banerjee, D.; Bertino, J.R. A miR-24 microRNA binding-site polymorphism in dihydrofolate reductase gene leads to methotrexate resistance. Proc. Natl. Acad. Sci. USA 2007, 104, 13513–13518. [Google Scholar] [CrossRef] [Green Version]
  151. Lee, S.; Yoon, D.S.; Paik, S.; Lee, K.M.; Jang, Y.; Lee, J.W. microRNA-495 inhibits chondrogenic differentiation in human mesenchymal stem cells by targeting Sox9. Stem Cells Dev. 2014, 23, 1798–1808. [Google Scholar] [CrossRef] [Green Version]
  152. Liu, X.; Fu, B.; Chen, D.; Hong, Q.; Cui, J.; Li, J.; Bai, X.; Chen, X. miR-184 and miR-150 promote renal glomerular mesangial cell aging by targeting Rab1a and Rab31. Exp. Cell Res. 2015, 336, 192–203. [Google Scholar] [CrossRef]
  153. Wu, H.; Zhu, S.; Mo, Y.Y. Suppression of cell growth and invasion by miR-205 in breast cancer. Cell Res. 2009, 19, 439–448. [Google Scholar] [CrossRef]
  154. Gindin, Y.; Jiang, Y.; Francis, P.; Walker, R.L.; Abaan, O.D.; Zhu, Y.J.; Meltzer, P.S. miR-23a impairs bone differentiation in osteosarcoma via down-regulation of GJA1. Front. Genet. 2015, 6, 233. [Google Scholar] [CrossRef] [Green Version]
  155. Li, J.; Aung, L.H.; Long, B.; Qin, D.; An, S.; Li, P. miR-23a binds to p53 and enhances its association with miR-128 promoter. Sci. Rep. 2015, 5, 16422. [Google Scholar] [CrossRef] [Green Version]
  156. Zeng, Q.; Zeng, J. Inhibition of miR-494-3p alleviates oxidative stress-induced cell senescence and inflammation in the primary epithelial cells of COPD patients. Int. Immunopharmacol. 2021, 92, 107044. [Google Scholar] [CrossRef] [PubMed]
  157. Gyvyte, U.; Lukosevicius, R.; Inciuraite, R.; Streleckiene, G.; Gudoityte, G.; Bekampyte, J.; Valentini, S.; Salteniene, V.; Ruzgys, P.; Satkauskas, S.; et al. The Role of miR-375-3p and miR-200b-3p in Gastrointestinal Stromal Tumors. Int. J. Mol. Sci. 2020, 21, 5151. [Google Scholar] [CrossRef]
  158. Ding, L.; Xu, Y.; Zhang, W.; Deng, Y.; Si, M.; Du, Y.; Yao, H.; Liu, X.; Ke, Y.; Si, J.; et al. MiR-375 frequently downregulated in gastric cancer inhibits cell proliferation by targeting JAK2. Cell Res. 2010, 20, 784–793. [Google Scholar] [CrossRef]
  159. Wu, X.; Wang, W.; Wu, G.; Peng, C.; Liu, J. miR-182-5p Serves as an Oncogene in Lung Adenocarcinoma through Binding to STARD13. Comput. Math. Methods Med. 2021, 2021, 7074343. [Google Scholar] [CrossRef]
  160. Cui, Y.H.; Xiao, L.; Rao, J.N.; Zou, T.; Liu, L.; Chen, Y.; Turner, D.J.; Gorospe, M.; Wang, J.Y. miR-503 represses CUG-binding protein 1 translation by recruiting CUGBP1 mRNA to processing bodies. Mol. Biol. Cell 2012, 23, 151–162. [Google Scholar] [CrossRef]
  161. Xu, Y.Y.; Wu, H.J.; Ma, H.D.; Xu, L.P.; Huo, Y.; Yin, L.R. MicroRNA-503 suppresses proliferation and cell-cycle progression of endometrioid endometrial cancer by negatively regulating cyclin D1. FEBS J. 2013, 280, 3768–3779. [Google Scholar] [CrossRef] [PubMed]
  162. Ikari, J.; Nelson, A.J.; Obaid, J.; Giron-Martinez, A.; Ikari, K.; Makino, F.; Iwasawa, S.; Gunji, Y.; Farid, M.; Wang, X.; et al. Reduced microRNA-503 expression augments lung fibroblast VEGF production in chronic obstructive pulmonary disease. PLoS ONE 2017, 12, e0184039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Liu, L.; Yan, L.N.; Sui, Z. MicroRNA-150 affects endoplasmic reticulum stress via MALAT1-miR-150 axis-mediated NF-κB pathway in LPS-challenged HUVECs and septic mice. Life Sci. 2021, 265, 118744. [Google Scholar] [CrossRef] [PubMed]
  164. 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]
  165. Xu, L.; Chen, Z.; Xue, F.; Chen, W.; Ma, R.; Cheng, S.; Cui, P. MicroRNA-24 inhibits growth, induces apoptosis, and reverses radioresistance in laryngeal squamous cell carcinoma by targeting X-linked inhibitor of apoptosis protein. Cancer Cell Int. 2015, 15, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Weng, J.H.; Yu, C.C.; Lee, Y.C.; Lin, C.W.; Chang, W.W.; Kuo, Y.L. miR-494-3p Induces Cellular Senescence and Enhances Radiosensitivity in Human Oral Squamous Carcinoma Cells. Int. J. Mol. Sci. 2016, 17, 1092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. MiRNAs that modulate the radiosensitivity of head and neck cancers [113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163].
Table 1. MiRNAs that modulate the radiosensitivity of head and neck cancers [113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163].
mi-RNATissue of OriginCancer TypeMechanism of ActionBinding SiteRadiosensitivityReferences
miR-296-5ptissuelarynxnot specified
PIN1
PIN1(Lee et al. 2014) [135]radioresistanceMaia et al. 2015 [113]
miR-324-3pcells and tissuenasopharynxtargeting WNT2BRelA promoter
(Dharap et al. 2013) [136]
radiosensitivityLiu et al. 2017 [114]
miR-203CellsHNSCC
nasopharyngeal
EMT modulation
targeting IL8/AKT
PKCα
(Wang et al. 2013) [137]
radiosesitivityde Jong et al. 2015 [115]
Qu et al. 2015 [119]
miR-324-3p, miR-93-3p miR-4501tissuenasopharynxdown-regulation/CDH1, PTENP1 and HSP90AA1PEDF (Wang et al. 2017) [138]radioresistanceLi et al. 2013 [116]
miR-371a-5p, miR-34c-5p and miR-1323tissuenasopharynxup-regulation/ICAM1, WNT2B, MYC, HLA-F, and TGF-β1 pathwaysXiap (Du et al. 2016) [139]
XBP1(Bartoszewska et al. 2019) [140]
BMP4 and SMAD4; PRKDC (Xie et al. 2020) [141]
radioresistanceLi et al. 2013 [116]
miR-27a-3pcellsHPV+ HNSCCup-regulation/DGCR8/miR-27a-3p/SMG1 axisFBXW7 (Lu et al. 2021) [143]radiosensitivityLong et al. 2021 [117]
miR-106acellsHPV+ HNSCCup-regulation/DGCR8/miR-106a/RUNX3 axisL10; ASK1 (Sharma et al. 2020) [144]
(Hong et al. 2018) [145]
radiosensitivityZhang et al. 2020 [118]
miR-375tissueoropharyngealtargeting IGF-1R/cycle arrest in G0/G1 phase, increases apoptosisYBX1 (Liu et al. 2016) [146]radiosensitivityZhang et al. 2017 [120]
miR-9cells
tissue, cells, saliva
HPV + HNSCCinducing M1 macrophage polarization via down-regulation of PPARδ
by targeting KLF5, positively regulates the expression of Sp1
cyclin D1 and Ets1 (Zheng et al. 2013) [147]radiosensitivity
radioreistance
Tong et al. 2020 [164]
Citron et al. 2021 [134]
miR-210cellsHNSCCmodulation of hypoxiaMNT (Zhang et al. 2009) [148]radioresistanceGee et al. 2010 [121]
miR-196atissue and cellsHNSCCsuppressing annexin A1
/induce EMT
SNP (Wang et al. 2012) [149]radioresistanceSuh et al. 2015 [122]
miR-24tissue and cellsLarynx SCCtargeting X-linked inhibitor of apoptosis proteindihydrofolate reductase gene (Mihsra et al. 2007) [150]radiosensitivityXu et al. 2015 [165]
miR-495tissue and cellsnasphparynxtargeting GRP78 to regulate EMTSox9 (Lee et al. 2014) [151]radiosensitivityFeng et al. 2018 [124]
miR-150cellsnasopharynxtargeting glycogen synthase kinase-3βRab1a and Rab31 (Liu et al. 2015) [152]radiresistanceHuang et al. 2018 [125]
miR-205cellsnasopharynxdirectly targeting PTENErbB3; VEGF-A (Wu et al. 2009) [153]radioresistanceQu et al. 2012 [119]
miR-23acellsnasopharynxtargeting IL-8/Stat3 pathwayGJA1; p53 (Gindin et al. 2015) [154]radiosensitivityQu et al. 2015 [126]
miR-24tissue and cellsnasopharynxinhibits Jab1/CSN5 translationdihydrofolate reductase gene (Mihsra et al. 2007) [150]radiosensitivityWang et al.2016 [127]
miR-494-3pcellsoral cavity SCCdownregulation of Bmi1 pathwaySIRT3 (Zeng et al. 2021) [156]radiosensitivityWeng et al. 2016 [166]
miR-375tissueoral cavity SCCtargeting insulin-like growth factor 1 receptorKIT; JAK2 (Gyvyte et al. 2020) [157]
(Ding et al. 2010) [158]
radiosensitivityZhang et al. 2017 [120]
miR-182-5pcellsHNSCCradiation-induced antioxidant effect through SESN2STARD13 (Wu et al. 2021) [159]radiosensitivityLin et al. 2021 [128]
miR-503cellsHNSCCinhibition of WEE1CUGBP1; CCND1; VEGF; E2 F3 (Cui et al. 2021) (Xu et al. 2013) (Ikari et. al 2017) [160,161,162]radiosensitivityMa et al. 2017 [129]
miR-150cellsnasopharyngealtargeting glycogen synthase kinase-3βMALAT1 (Liu et al. 2021) [163]radioresistanceHuang et al. 2018 [125]
miR-138-1-3pcellsnasopharynxEMT and the JAK2/STAT3 signaling pathwayCRIPTO (Du et al. 2021) [130]radiosensitivityDu et al. 2021 [130]
miR-195-3ptissue and cellsnasopharynxinhibits cyclin dependent kinase 1CDK1 (Xie at al. 2021) [131]radiosensitivityXie at al. 2021 [131]
miR-19b-3pcellsnasopharynxactivating the TNFAIP3/NF-κB axisTNFAIP3 (Huang et al.) 2016 [132]radioresistanceHuang et al. 2016 [132]
miR-BART4tissue and cellsnasopharynxtargeting PTENPTEN (Wu et al. 2018) [133]radioresistanceWu et al. 2018 [133]
miR-96-5pcellsHNSCCPI3K-Akt signaling pathway PTEN (Vahabi et al. 2019) [105]radioresistanceVahabi et al. 2019 [105]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mireștean, C.C.; Iancu, R.I.; Iancu, D.P.T. Micro-RNAs, the Cornerstones of the Future of Radiobiology in Head and Neck Cancers? Curr. Oncol. 2022, 29, 816-833. https://doi.org/10.3390/curroncol29020069

AMA Style

Mireștean CC, Iancu RI, Iancu DPT. Micro-RNAs, the Cornerstones of the Future of Radiobiology in Head and Neck Cancers? Current Oncology. 2022; 29(2):816-833. https://doi.org/10.3390/curroncol29020069

Chicago/Turabian Style

Mireștean, Camil Ciprian, Roxana Irina Iancu, and Dragoș Petru Teodor Iancu. 2022. "Micro-RNAs, the Cornerstones of the Future of Radiobiology in Head and Neck Cancers?" Current Oncology 29, no. 2: 816-833. https://doi.org/10.3390/curroncol29020069

Article Metrics

Back to TopTop