Epigenetics of oral and oropharyngeal cancers (Review)

Corrigendum in: /10.3892/br.2020.1290

  • Authors:
    • Daniela Russo
    • Francesco Merolla
    • Silvia Varricchio
    • Giovanni Salzano
    • Giovanni Zarrilli
    • Massimo Mascolo
    • Viviana Strazzullo
    • Rosa  Maria Di Crescenzo
    • Angela Celetti
    • Gennaro Ilardi
  • View Affiliations

  • Published online on: July 27, 2018     https://doi.org/10.3892/br.2018.1136
  • Pages: 275-283
  • Copyright : © Russo et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Oral and oropharyngeal cancers represent the two most common malignancies of the head and neck region. The major risk factors for these cancers include alcohol consumption, tobacco use (via smoking or chewing) and high‑risk human papillomavirus infection. The transition from normal epithelium to premalignant tissue and finally carcinoma is in part caused by a summation of genetic and epigenetic modifications. Epigenetic refers to modifications in the way the genome is expressed in cells. The most common examples of epigenetic control of gene expression are DNA methylation, histone modification and regulation by small non‑coding RNAs. The aim of the current paper was to review the recent studies on the main epigenetic changes that have been suggested to serve a role in the carcinogenesis process and progression of oral and oropharyngeal cancers. Furthermore, it is discussed how the epigenetic changes may be used as potential predictive biomarkers and how recent findings in the field may impact the personalized cancer therapy approach for these tumors.

Introduction

Oral and oropharyngeal cancers are the most common malignancies of the head and neck region, with squamous cell carcinoma being the most common histotype (1,2). Reportedly, ~300,000 new cases of oral cancers are diagnosed worldwide annually, of which ~140,000 are oropharyngeal cancers, with a male: female ratio that varies between 2:1 and 4:1 (3). Oral squamous cell carcinoma (OSCC) and oropharyngeal squamous-cell carcinoma (OPSCC) are often listed together and, accounted as a single entity, represent the sixth most common malignancy worldwide (46).

Knowledge of these cancers has improved substantially over recent decades, but the therapeutic outcome has remained mostly unchanged, with a five year survival rate of ~50% (7). The more advanced the stage of cancer at diagnosis, the lower the response rate to therapy; furthermore, patients diagnosed with high stage OSCC or OPSCC have a high risk of developing metastases (both lymph node and distant) and relapses (7).

Oral carcinogenesis is caused by environmental and endogenous factors; categorized in the first group are regular tobacco and alcohol use and persistent high-risk human papillomavirus (HPV) infection. HPV-related OPSCC constitutes a different biological entity compared with HPV-negative cases (8). In particular, HPV-related OPSCC differs significantly in epidemiology from HPV-negative cases and those of the oral cavity: HPV-negative head and neck squamous cell carcinoma (HNSCC) mostly affects individuals of more than 60 years in age, and its incidence is decreasing; conversely, the incidence of HPV-positive HNSCC is increasing, and is often diagnosed in young adults (9,10). HPV status also significantly impacts on prognosis; namely, HPV-positive OPSCC is associated with better prognosis when compared with HPV-negative cases and OSCC (9,11).

Endogenous factors are generally linked to genetic and epigenetic alterations. Genetic alterations refer to changes in the DNA sequence including deletions, amplifications and mutations; these changes are not reversible. Epigenetic alterations are a group of heritable modifications in DNA expression that manifest without direct DNA modification. In the present review, the main epigenetic alterations described recently in the literature on oral and oropharyngeal cancers are discussed.

OSCC and OPSCC carcinogenesis

‘Field cancerization’ (also known as the field effect), is a concept first introduced by Slaughter et al (12) in 1953 for oral cancers. They described field cancerization as being a process in which an area of epithelium has been preconditioned by an, at that time, unknown carcinogenic agent, enabling epidermoid carcinoma of the oral stratified squamous epithelium to originate. Some of the carcinogenic agents involved in neoplastic transformation of oral epithelium have been established, among which tobacco and alcohol are considered as principal risk factors; however, the mechanisms of field cancerization in HPV-related HNSCC remain unknown (9). Tobacco and alcohol seem to cause an anaplastic transformation that involves an area or ‘field’ of mucosa rather than single cells (13). This would explain the tendency of OSCC and OPSCC to recur (10–30% of cases) in different areas relative to the original location, even following surgical removal with tumor free margins (13).

Aside from in HNSCC, the field effect has been also described for cancers of other anatomical regions, including the esophagus (14), colon (15), urinary tract (16), skin (17), breast (18), uterine cervix (19) and airway epithelium (20).

HPV-related carcinogenesis

HPV is an established causative factor in~25% of global cases of HNSCCs. The oropharynx is the anatomical location in which HPV-related cancers are more frequently observed; they amount to ~36% of the total, ~87% of which are related to HPV 16. In the oral cavity, ~23% of cancers are HPV-related, ~69% of which are HPV 16-related. HPV 18 is the second most common papilloma virus type in HNSCC, amounting for ~8% of OSCCs and ~1% of OPSCCs (21). Certain locations appear to have higher HPV-related cancer incidence: For instance, tonsil and base of the tongue cancers have been identified to be HPV-linked in 50–80% of cases (2224).

E6 and E7 are considered the most important HPV proteins involved in carcinogenesis; E6 protein has several functions, with a key and well-established function being to induce the degradation of p53, a widely studied oncosuppressor (2527). E6 binds the ubiquitin-protein ligase, E6AP; the E6/E6AP heterodimer can then bind p53, which is ubiquitinated and degraded in proteasomes (26,27).

E7 binds retinoblastoma protein (pRb). The main function of pRb is to negatively regulate several transcriptional factors, including E2F1-3 (25). When E7 binds pRb, E2F transcriptional factors are no longer inhibited, and have the possibility to activate the transcription of cell cycle-related genes, including cyclin A and cyclin E (25).

Several biomarker expression studies performed in HPV-positive OPSCC have established a clear distinction between HPV-active and HPV-inactive cancers (2834). HPV-active tumors contain and express HPV sequences, are positive for p16 immunohistochemistry as well as for HPV RNA (35) and exhibit gene expression profiles that suggest a ‘HPV signature’, dominated by changes in genes that control the cell cycle, proliferation and mitosis (36). Numerous differences have been described in gene expression profiles between HPV-active and HPV-negative cancers, the latter being dominated by changes in genes that control cell motility, angiogenesis and epithelial-mesenchymal transition, associated with aggressive clinical behavior (36). The gene expression profiles of HPV-inactive tumors lack the characteristic ‘HPV signature’ and are notably more similar to HPV-negative tumors, although some differences can be detected, the clinical significance of which remains to be determined (36,37).

Tomar et al (37), proposed that HPV-inactive cancers may originate as HPV-active lesions that progressively become independent of E6/E7 for proliferation and tumorigenic potential. Methylation mechanisms have been demonstrated as responsible for the silencing of HPV sequences in cells that have acquired mutations that may ‘replace’ E6/E7 function (38). HPV methylation has been investigated in a variety of cancers and cancer cell lines (38); however, the role of HPV methylation and other mechanisms leading to a possible progression from HPV-dependence to HPV-independence in head and neck cancer remains to be determined (37,39).

Tobacco-related carcinogenesis

Tobacco consumption is among the most important risk factors in numerous cancers. Expectedly, it has been widely demonstrated that tobacco consumption is a major risk factor for OSCC and OPSCC (40,41).

Among smoker patients, the risk for the development of oral cancer is greater than three times higher than in non-smokers; for pharyngeal cancers the risk becomes greater than six times higher for smokers (42). The risk of oral and oropharyngeal cancers returns to baseline when smoking is stopped for at least 20 years (43).

Additionally, smokeless tobacco is established as an independent risk factor for oral cancer; an increased risk of oral cancer has been identified with use of numerous tobacco products, including gutka, supari (areca nut) and betel quid (Piper betle), with an odds ratio between 5.1 and 11.4 (44).

Alcohol-related carcinogenesis

Ethanol serves as a risk factor for the development of oral cancer through its local and systemic effects. By its local effects, alcohol may increase the permeability of the mucosa by dissolving the lipid component of the epithelium; it may provide indirect cellular damage through acetaldehyde; it may amplify the toxicity of other carcinogenic agents; and it may cause hyposalivation, leading to a higher exposition to locally acting carcinogens.

As systemic effects, ethanol acts to reduce first pass hepatic metabolism of toxic substances; it may lead to immunosuppression; and it may be associated with malnutrition, which can enhance the risk of developing cancer (45).

Patients who stop drinking exhibit a decreased risk of developing OSCC and OPSCC over time; however, for oropharyngeal cancer, the risk does not return to baseline, remaining higher than in those without a history of alcohol consumption. Otherwise, for oral cancer, the risk in former drinkers after more than 20 years is reportedly lower than in the ‘never drinker’ population (43).

EBV-related carcinogenesis

Epstein-Barr virus (EBV) was identified in 1964 by Epstein et al (46), in a Burkitt lymphoma-derived cell line. It is a herpesvirus (HHV-4), and therefore its genome is composed of DNA (47). EBV is the etiologic agent of mononucleosis, also known as the kissing disease for its transmission path; this is a self-limiting disease, after which the virus may remain latent, with infected individuals potentially becoming lifelong carriers of the virus (48). Worldwide, more than 90% of adults infected with EBV exhibit no sign of the infection (47).

EBV is established as a causative factor in numerous cancers, including Burkitt lymphoma, Hodgkin lymphoma, immunosuppression-related non-Hodgkin lymphoma and nasopharyngeal carcinoma (47,48). EBV-persistent infection has been associated with oral cancers, even though there are few epidemiological data available (49).

Epigenetics

Epigenetics refers to changes in DNA expression not attributable to alterations in DNA sequence. The main events responsible are DNA methylation, histone modification and post-transcriptional gene downregulation by microRNAs (miRNA/miRs). These alterations can persist for a cell lifetime and be inherited by subsequent generations.

The results of epigenetics vary from increased gene expression to complete silencing, depending on the interference of these alterations to activators and suppressors of specific promoters in chromatin. Therefore, epigenetic changes can induce overexpression of oncogenes as much as silencing of tumor suppressor genes (50).

DNA methylation is the most common epigenetic event (51). It is performed by a series of enzymes known as DNA methyltransferases (DNMTs). These enzymes catalyze the covalent addition of a methyl group to the carbon-5 position of cytosine bases that are located 5′ to a guanosine base in a CpG dinucleotide (52).

Hypermethylation has been identified in the promoters of tumor suppressor genes in numerous cancers; this may lead to the reduction of their expression or to their complete silencing (49). Conversely, hypo/demethylation of the promoter of proto-oncogenes may lead to increased expression of these genes (50). An imbalance in tumor suppressor gene methylation status has often been reported following exposure to tobacco-derived carcinogens and other environmental compounds including cadmium, arsenic and nickel (52).

Many genes present an altered DNA methylation profile in OSCC and OPSCC (53). The galanin (GAL) gene maps onto chromosome 11q13.2, and is a neuropeptide widely distributed in the central and peripheral nervous system (54). It has the function of regulating anterior pituitary hormones secretion and acts as neurotransmitter (54). GALR1 and GALR2 are two of the three receptors for galanin; their genes are located on chromosome 18q23 and 17q25.1, respectively. These receptors are members of the G protein-coupled receptor (GPCR) superfamily. Galanin and its receptors regulate growth via the inhibition of extracellular signal-regulated kinase (ERK) 1/2, upregulation of the cyclin-dependent kinase inhibitors p27 and p57, and decreased expression of cyclin D1, which can altogether limit cell proliferation (55). Additionally, GALR2 can induce caspase-3 dependent apoptosis (5557). Hypermethylation of these genes leads to a lack of their expression with a consequent lack of their tumor suppressor activity (57). A previous study identified 25 out of 48 (52%) OSCC cases to have hypermethylation of at least one of the three genes; furthermore in OPSCC, 13 out of 20 cases (65%) were determined to have hypermethylation of at least one of the genes (58).

Calprotectin S100A8/9 is a heterodimer of the two calcium-binding proteins S100A8 and S100A9. S100A8/9 is located both within epithelial cells and in the extracellular space (59). In normal epithelial cells, S100A8/9 works to arrest the cell cycle by activating the G2/M DNA damage checkpoint (59); a function that has been evidenced in the TR146 cell line (an oral cancer-derived cell line) (60). In HNSCC, it has been identified that S100A8/9 level was on average 10-fold lower compared with in normal adjacent tissues (60). Additionally, S100A8/9 in the extracellular space is released from polymorphonuclear, leukocytes and macrophages and works by signaling through the receptor for advanced glycation end products and toll-like receptor (TLR) 4 (61).

Notably, it has been reported that S100A9 promoter hypermethylation correlates with reduced expression of S100A9. S100A8 expression, conversely, does not correlate with its promoter methylation (60).

Saliva-detectable hypermethylation

Saliva is an emerging diagnostic medium for oral and oropharyngeal cancers. It represents an effective diagnostic alternative since it is produced and collected close to the sites of malignancy; this provides the possibility to analyze tumor-specific biomolecules with minimal interferences (61). Additionally, saliva allows analysis of regions including the tonsillar crypts, which are otherwise difficult to sample (62). Ultimately, saliva sampling is easy to perform (63,64).

Several hypermethylated genes have been detected via saliva sampling (Table I). Insulin-like growth factor (IGF)-binding protein 7 (IGFBP-7) is a protein involved in the regulation of free IGF concentration in tissues (65). It is also involved in tumor suppressor activity and in epithelial-mesenchymal transition (EMT) (66,67). In a previous cohort of 47 oral tongue cancer cases, IGFBP-7 promoter was reportedly hypermethylated, and consequently, IGFBP-7 marginally expressed. Furthermore, hypermethylation in these cases was documented to correlate with invasive depth, locoregional recurrence and poor prognosis (67).

Table I.

List of genes found hypermethylated in saliva samples, their reported function and the tumor type where modifications were present.

Table I.

List of genes found hypermethylated in saliva samples, their reported function and the tumor type where modifications were present.

GeneGene productFunctionTumor site (OSCC/OPSCC)(Refs.)
CCNA1Cyclin A1Cell cycle regulationOSCC and OPSCC(62,99)
DAPK1Death-associated protein kinase 1Apoptosis and autophagyOSCC and OPSCC(62,63,99)
DCCDeleted in colorectal carcinomaNetrin 1 receptorOSCC(62,100)
EDNRBEndothelin receptor type BG protein-coupled receptorOSCC(62,100)
ERCC1Excision repair cross-complementation group 1Repair of DNA damage induced by ultraviolet light or cisplatinOSCC and OPSCC(62,101)
HOXA9Homeobox protein Hox-A9Gene expression, morphogenesis and differentiationOSCC(62,101)
KIF1AKinesin family member 1AMembranous organelle transportation along axonal microtubulesOSCC(62,102,103)
MED15Mediator of RNA polymerase II transcription subunit 15Transcriptional coactivator in RNA polymerase II transcriptionOSCC(62,104)
MINT31Spen family transcriptional repressor family transcriptional repressorCalcium channel regulatorOSCC(62,99,105)
NID2Nidogen-2Cell adhesionOSCC(62,102)
PAXIPaxillinCytoskeletal protein involved in actin-membrane attachment at sites of cell adhesion to the extracellular matrixOSCC(62,106)
p16INK2Ap16 (also known as cyclin-dependent kinase inhibitor 2A)Tumor suppressor geneOSCC(62,63,99)
RASSF1αRas association domain family member 1 αGrowth inhibition along the RAS-activated signaling pathwayOSCC(62,63)
TIMP3Metalloproteinase inhibitor 3Inhibitor of the matrix metalloproteinasesOSCC and OPSCC(62,99,105,107)

[i] OSCC, oral squamous cell carcinoma; OPSCC, oropharyngeal squamous cell carcinoma.

Krüppel-like factor 4 (KLF4) is a is a zinc-finger transcription factor encoded by a gene located on chromosome 9q31.2. It is involved in epidermal barrier function (68), cell proliferation, differentiation and apoptosis (69) and stem cell renewal (70). The activity and expression of KLF4 are often associated with mutation in various cancers (7174). Li et al (75), identified that KLF4 was downregulated in OSCC, and its downregulation correlated with cancer differentiation i.e., the poorer the cancer differentiation, the lower the expression of KLF. The downregulation of KLF4 in OSCC has been associated with promoter hypermethylation (75).

Somatostatin (SST) and somatostatin SST receptor type 1 (SSTR1) are encoded by genes located on 3q27.3 and 14q21.1, respectively. SST is a peptide hormone that exists in two forms (14 amino acids and 28 amino acids), both encoded by the same gene. SST has several functions: It inhibits growth hormone (GH) secretion; it works as an immunomodulator to cause an inhibition of tumor necrosis factor-α, interferon-γ, corticotropin releasing hormone and substance P secretion; and it binds µ-opioid receptor (76). Additionally, SST interacts with SSTR1 to cause an antiproliferative effect. In fact, the activity of SSTR1 may lead to the activation of SHP-2 (also known as tyrosine-protein phosphatase non-receptor type 11); SHP-2 dephosphorylates the proto-oncogene tyrosine-protein kinase c-src on Tyr529 (inhibitory site) and makes possible phosphorylation on Tyr418 (stimulatory site). Once activated, c-src may also phosphorylate Raf-1,the function of which is to activate (through phosphorylation) mitogen activated-protein kinase signaling, which can eventually lead to cell cycle inhibition through p21/Cip1. Besides these direct effects, SST and its receptors may inhibit proliferation through indirect effects. In fact, the activation of the SST pathway may cause a reduction of IGF-1 synthesis and inhibition of angiogenesis (77).

SSTR1 is one of the five receptors for SST, and an inhibitory GPCR (78). Misawa et al (79), have demonstrated SST and SSTR1 promoter hypermethylation in OSCC, OPSCC primary tumor samples and in UM-SCC cell lines; these findings suggest that SST and SSTR1 may be considered as important molecules in the tumorigenesis and progression of OSCC and OPSCC.

Other genes observed to be hypermethylated in saliva samples in oral/oropharyngeal cancers are listed in Table I.

miRNA

miRNAs represent a category of non-coding RNA molecules, each made of ~22 nucleotides. These molecules are involved in the control of gene expression by interacting with mRNA (80).

miR-143 is a miRNA capable of interacting with the 3′ untranslated region of hexokinase 2 (HK2) mRNA (81). Elevated concentrations of HK2 have been observed in numerous cancers, including oral and oropharyngeal cancers; in particular, it has been determined in OSCC-derived cell lines that there was a lower concentration of miR-143, compared with in normal keratinocytes and, consequently, higher concentrations of HK2, since miR-143 inhibits HK2 synthesis (81).

Saliva-detectable miRNA. Saliva may be sampled and analyzed for the identification of miRNAs. Several miRNAs have been identified to be overexpressed or underexpressed in patients with oral and/or oropharyngeal cancer. For instance, miR-491-5p is a miRNA that has been implicated in a number of important processes, including migration, invasion and consequently metastasis. The function of miR-491-5p is to target GPCR kinase-interacting protein 1 (GIT1), leading to a downregulation of its expression. A well-established function of GIT1 is to initiate cell motility; this is achieved through interactions with members of the focal adhesion kinase and ERK1/2 pathways.

Huang et al (82), reported that miR-491-5p was downregulated in highly aggressive OSCC cell lines (SAS-I5, OECM-1-I8, SCC25-I6, OC-3-I5- and OC-3-I5-lung-IV2) and in 29 out of 33 OSCC samples compared with in normal tissue. They also demonstrated that cells with low miR-491-5p expression exhibited enhanced capability to migrate and metastasize, and furthermore, that miR-491-5 low expression was correlated with poor prognosis (82).

Examples of miRNAs observed to be over- or underexpressed in OSCC/OPSCC are listed in Table II.

Table II.

List of miRNAs with altered expression, on saliva specimen testing, in oral and/or oropharyngeal cancers.

Table II.

List of miRNAs with altered expression, on saliva specimen testing, in oral and/or oropharyngeal cancers.

NameStatusTumor type(Refs.)
miR-9OverexpressedHPV-negative HNSCC(62)
miR-24OverexpressedOSCC (vs. healthy controls)(62,108)
miR-27bOverexpressedOSCC (vs. healthy controls) OSCC in remission(62,108)
miR-31OverexpressedHPV-related OSCC and OPSCC (vs. healthy controls and leukoplakia)(62,109,110)
miR-134OverexpressedHNSCC(62,111)
miR-191OverexpressedHNSCC(62,111)
miR-125aUnderexpressedOSCC(62,112)
miR-136UnderexpressedOSCC (vs. healthy controls)(62,108)
miR-147UnderexpressedOSCC (vs. healthy controls)(62,108)
miR-148aUnderexpressedOSCC (vs. healthy controls)(62,108)
miR-200aUnderexpressedOSCC (vs. control samples)(62,112,113)
miR-222UnderexpressedOSCC and OPSCC(62,109)
miR-323-5pUnderexpressedOSCC (vs. healthy controls)(62,108)
miR-503UnderexpressedOSCC (vs. healthy controls)(62,108)
miR-622UnderexpressedOSCC (vs. healthy controls)(62,108)
miR-646UnderexpressedOSCC (vs. healthy controls)(62,108)
miR-668UnderexpressedOSCC (vs. healthy controls)(62,108)
miR-877UnderexpressedOSCC (vs. healthy controls)(62,108)
miR-1250UnderexpressedOSCC (vs. healthy controls)(62,108)

[i] miRNA/miR, microRNA; OSCC, oral squamous cell carcinoma; OPSCC, oropharyngeal squamous cell carcinoma; HNSCC, head and neck squamous cell carcinoma; HPV, human papillomavirus.

Therapeutic implications

To date, there are few chemotherapeutic options for oral and oropharyngeal cancers. Platinum-based antineoplastics are the most frequently used; however, their efficacy is not satisfactory (83,84).

The biology of epigenetics can be used for therapeutic purposes; well known among such applications is in the context of viral mimicry. Viral mimicry refers to a characteristic behavior that certain cancer cells acquire when treated with demethylating agents; these treatments can lead to an effect that mimics a viral infection (85). The exact mechanism of action is not fully understood, nor is the reasoning behind why certain cells are more susceptible than others, or why in certain cases the efficacy of demethylating agents decreases over time (85,86).

Viral mimicry is based on a series of immune effects on tumor cells. Demethylating agents may stimulate viral defense and type I interferon signaling; this effect can be achieved through RNA demethylation (85). In fact, demethylated RNA is known to stimulate an interferon response through TLR3 activation; a similar response to DNA demethylation is suggested to occur by a similar effect obtained using demethylating agents which target only DNA (85).

Another immune effect seen with demethylating agents involves double-stranded RNA (dsRNA). In fact, these drugs have been associated with an increase of cytosolic levels of dsRNA; dsRNA molecules are used by epithelial and other cells as a defense mechanism towards viral infection, which in turn may lead to a type I interferon response and apoptosis (85). Viral mimicry has been observed and studied in various cancers, including colon (86) and breast (87) cancers.

Another notable mechanism of action involves endogenous retroviral genes. These genes comprise ~8% of the human genome and are normally silenced through hypermethylation in somatic cells (82). With demethylating drugs these genes are unlocked, and their RNA levels increase in the cell cytoplasm. This may then promote the activation of a viral defense response through RNA sensors (85).

Additionally, demethylating agents may cause toxicity in cancer cells through non-immune mechanisms of action. One of the most plausible and studied mechanisms of action is based on the evidence that cancerous cells share an overall hypermethylation of tumor-suppressor gene promoters; with demethylating agents, these genes are ‘unlocked’ and therefore can provide a tumor-suppressor function (86).

Gene-body methylation may also be important. It affects transcription in a directly proportional way, in that the more a gene-body is methylated, the more it is expressed (86,88). However, this mechanism is poorly understood (86,88).

Another theory is based on the evidence that global demethylation may lead to direct toxicity in cancerous cells, causing their death or interrupting their proliferation; normal cells instead tend to survive against these drugs (86,89). However, the global DNA methylation state cannot predict response to therapy, and thus it is probably not the only mechanism involved (86).

Another mechanism of action describes the cytotoxicity of demethylating agents as the main cause of their efficacy in cancer therapy; specifically, they may trap DNMTs onto DNA, resulting in bulky adducts that can inhibit DNA synthesis and eventually cause cell death (86).

It is likely that all these theories have some involvement, though it is unknown which are more important than others.

5-Azacytidine (5-aza-CR) and 5-aza-2′-deoxycytidine are the main demethylating drugs. These are nucleoside analogs of cytosine and can inhibit DNA methylation by being incorporated in a DNA molecule at the position of cytosine during DNA replication (90). When a DNA methyltransferase interacts with a DNA molecule modified in this way, it ends adhere to the DNA, resulting in a long-lasting demethylation.

miRNA based therapy can be expected to be a critical topic in upcoming research. As discussed, miRNAs have a crucial role in regulating gene expression for both tumor suppressor genes and oncogenes. For therapeutic purposes, the aim is to inhibit the miRNAs that are overexpressed and to replace those that are insufficient.

Use of miRNA mimics is one such strategy to replace underexpressed miRNAs; these molecules must be modified to achieve optimal pharmacokinetics and pharmacodynamics, for example through conjugation with a sugar molecule or utilizing liposomes (91).

Conversely, treatment with miRNA inhibitors is the strategy for lowering the concentration of overexpressed miRNAs. These molecules are capable of binding specific miRNAs, preventing their binding with target mRNA (91).

Conclusions

Among the most considerable problems with treating OSCC and OPSCC is the tendency of these cancers to become chemo-radioresistant. However, this is among the issues that can be bypassed through epigenetic-based therapy; in fact, it is established that certain demethylase inhibitors can induce a chemo-radiotherapy resensitizing effect in various cancer types (9294). HNSCC cell lines (including SQD9, SCC61, Cal27, SC179, SC263, JH011) have been identified to have an increased response to radiation therapy following exposure to demethylating agents in vitro (95,96), suggesting that these drugs may be used efficiently in combination with other drugs and radiation to achieve an improved clinical outcome.

Another issue that may be, at least in part, bypassed with demethylating drugs is radiation-related toxicity; radiation therapy is associated with numerous side effects caused by direct effect of the radiation on healthy tissues (97,98). In oral and oropharyngeal cancers, the most frequent of such effects are mucositis, dysphagia, trismus, dental problems, xerostomia and osteoradionecrosis (97,98). As discussed, demethylating drugs may enhance the radiation sensibility of cancer cells; this would make possible the use of lower doses of radiation to achieve the same therapeutic benefits while potentially minimizing the side effects.

In conclusion, oral and oropharyngeal cancers represent a major health issue, both regarding their epidemiological characteristics and poor clinical outcome. There have been several advances in our understanding of these malignancies, some of which have been used for prevention purposes (for instance by acknowledging the risk factors), and others have had an impact on therapeutic approaches.

Epigenetics influences all phases of cancer development and progression. The understanding of its functioning comprises an important aim of recent research due to the possible therapeutic implications. This is particularly important in oral and oropharyngeal cancer, since the relevant therapeutic approaches and clinical outcomes are still based almost exclusively on classical chemotherapy. This is probably a main reason for the lack of improved survival outcome over time, or at least for the lower extent of improvement than would be expected. In the short term, it is probable that epigenetics will be used as a basis for therapeutic purposes.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

DR, FM, SS and GI designed and wrote the manuscript.SV, GS, GZ, RDC and VS performed the literature search; MM, AC, LC revised the manuscript for intellectual content.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

All authors declare no competing interests.

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Torre LA, Siegel RL, Ward EM and Jemal A: Global cancer incidence and mortality rates and trends - an update. Cancer Epidemiol Biomarkers Prev. 25:16–27. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Warnakulasuriya S: Global epidemiology of oral and oropharyngeal cancer. Oral Oncol. 45:309–316. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Gupta B, Johnson NW and Kumar N: Global epidemiology of head and neck cancers: A continuing challenge. Oncology. 91:13–23. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Mydlarz WK, Hennessey PT and Califano JA: Advances and perspectives in the molecular diagnosis of head and neck cancer. Expert Opin Med Diagn. 4:53–65. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Argiris A, Karamouzis MV, Raben D and Ferris RL: Head and neck cancer. Lancet. 371:1695–1709. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Leemans CR, Braakhuis BJM and Brakenhoff RH: The molecular biology of head and neck cancer. Nat Rev Cancer. 11:9–22. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Chaturvedi AK, Engels EA, Anderson WF and Gillison ML: Incidence trends for human papillomavirus-related and -unrelated oral squamous cell carcinomas in the United States. J Clin Oncol. 26:612–619. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Ang KK, Harris J, Wheeler R, Weber R, Rosenthal DI, Nguyen-Tân PF, Westra WH, Chung CH, Jordan RC, Lu C, et al: Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med. 363:24–35. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Slaughter DP, Southwick HW and Smejkal W: Field cancerization in oral stratified squamous epithelium; clinical implications of multicentric origin. Cancer. 6:963–968. 1953. View Article : Google Scholar : PubMed/NCBI

13 

Tabor MP, Brakenhoff RH, Ruijter-Schippers HJ, Kummer JA, Leemans CR and Braakhuis BJM: Genetically altered fields as origin of locally recurrent head and neck cancer: A retrospective study. Clin Cancer Res. 10:3607–3613. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Prevo LJ, Sanchez CA, Galipeau PC and Reid BJ: p53-mutant clones and field effects in Barrett's esophagus. Cancer Res. 59:4784–4787. 1999.PubMed/NCBI

15 

Hawthorn L, Lan L and Mojica W: Evidence for field effect cancerization in colorectal cancer. Genomics. 103:211–221. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Jones TD, Wang M, Eble JN, MacLennan GT, Lopez-Beltran A, Zhang S, Cocco A and Cheng L: Molecular evidence supporting field effect in urothelial carcinogenesis. Clin Cancer Res. 11:6512–6519. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Torezan LAR and Festa-Neto C: Cutaneous field cancerization: Clinical, histopathological and therapeutic aspects. An Bras Dermatol. 88:775–786. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Rivenbark AG and Coleman WB: Field cancerization in mammary carcinogenesis - Implications for prevention and treatment of breast cancer. Exp Mol Pathol. 93:391–398. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Chu TY, Shen CY, Lee HS and Liu HS: Monoclonality and surface lesion-specific microsatellite alterations in premalignant and malignant neoplasia of uterine cervix: A local field effect of genomic instability and clonal evolution. Genes Chromosomes Cancer. 24:127–134. 1999. View Article : Google Scholar : PubMed/NCBI

20 

Gomperts BN, Spira A, Massion PP, Walser TC, Wistuba II, Minna JD and Dubinett SM: Evolving concepts in lung carcinogenesis. Semin Respir Crit Care Med. 32:32–43. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Kreimer AR, Clifford GM, Boyle P and Franceschi S: Human papillomavirus types in head and neck squamous cell carcinomas worldwide: A systematic review. Cancer Epidemiol Biomarkers Prev. 14:467–475. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Hannisdal K, Schjølberg A, De Angelis PM, Boysen M and Clausen OPF: Human papillomavirus (HPV)-positive tonsillar carcinomas are frequent and have a favourable prognosis in males in Norway. Acta Otolaryngol. 130:293–299. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Attner P, Du J, Näsman A, Hammarstedt L, Ramqvist T, Lindholm J, Marklund L, Dalianis T and Munck-Wikland E: The role of human papillomavirus in the increased incidence of base of tongue cancer. Int J Cancer. 126:2879–2884. 2010.PubMed/NCBI

24 

Ramqvist T, Grün N and Dalianis T: Human papillomavirus and tonsillar and base of tongue cancer. Viruses. 7:1332–1343. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Ghittoni R, Accardi R, Hasan U, Gheit T, Sylla B and Tommasino M: The biological properties of E6 and E7 oncoproteins from human papillomaviruses. Virus Genes. 40:1–13. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Huibregtse JM, Scheffner M and Howley PM: A cellular protein mediates association of p53 with the E6 oncoprotein of human papillomavirus types 16 or 18. EMBO J. 10:4129–4135. 1991. View Article : Google Scholar : PubMed/NCBI

27 

Scheffner M, Huibregtse JM, Vierstra RD and Howley PM: The HPV-16 E6 and E6-AP complex functions as a ubiquitin-protein ligase in the ubiquitination of p53. Cell. 75:495–505. 1993. View Article : Google Scholar : PubMed/NCBI

28 

Mascolo M, Ilardi G, Romano MF, Celetti A, Siano M, Romano S, Luise C, Merolla F, Rocco A, Vecchione ML, et al: Overexpression of chromatin assembly factor-1 p60, poly(ADP-ribose) polymerase 1 and nestin predicts metastasizing behaviour of oral cancer. Histopathology. 61:1089–1105. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Aquino G, Pannone G, Santoro A, Liguori G, Franco R, Serpico R, Florio G, de Rosa A, Mattoni M, Cozza V, et al: pEGFR-Tyr 845 expression as prognostic factors in oral squamous cell carcinoma: A tissue-microarray study with clinic-pathological correlations. Cancer Biol Ther. 13:967–977. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Pannone G, Rodolico V, Santoro A, Lo Muzio L, Franco R, Botti G, Aquino G, Pedicillo MC, Cagiano S, Campisi G, et al: Evaluation of a combined triple method to detect causative HPV in oral and oropharyngeal squamous cell carcinomas: p16 Immunohistochemistry, Consensus PCR HPV-DNA, and In Situ Hybridization. Infect Agent Cancer. 7:42012. View Article : Google Scholar : PubMed/NCBI

31 

Santoro A, Pannone G, Papagerakis S, McGuff HS, Cafarelli B, Lepore S, De Maria S, Rubini C, Mattoni M, Staibano S, et al: Beta-catenin and epithelial tumors: A study based on 374 oropharyngeal cancers. BioMed Res Int. 2014:9482642014. View Article : Google Scholar : PubMed/NCBI

32 

Russo D, Merolla F, Mascolo M, Ilardi G, Romano S, Varricchio S, Napolitano V, Celetti A, Postiglione L, Di Lorenzo PP, et al: FKBP51 immunohistochemical expression: A new prognostic biomarker for OSCC? Int J Mol Sci. 18:4432017. View Article : Google Scholar

33 

Mascolo M, Ilardi G, Merolla F, Russo D, Vecchione ML, de Rosa G and Staibano S: Tissue microarray-based evaluation of Chromatin Assembly Factor-1 (CAF-1)/p60 as tumour prognostic marker. Int J Mol Sci. 13:11044–11062. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Lo Muzio L, Campisi G, Giovannelli L, Ammatuna P, Greco I, Staibano S, Pannone G, De Rosa G, Di Liberto C and D'Angelo M: HPV DNA and survivin expression in epithelial oral carcinogenesis: A relationship? Oral Oncol. 40:1–741. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Weinberger PM, Yu Z, Kountourakis P, Sasaki C, Haffty BG, Kowalski D, Merkley MA, Rimm DL, Camp RL and Psyrri A: Defining molecular phenotypes of human papillomavirus-associated oropharyngeal squamous cell carcinoma: Validation of three-class hypothesis. Otolaryngol Head Neck Surg. 141:382–389. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Jung AC, Briolat J, Millon R, de Reyniès A, Rickman D, Thomas E, Abecassis J, Clavel C and Wasylyk B: Biological and clinical relevance of transcriptionally active human papillomavirus (HPV) infection in oropharynx squamous cell carcinoma. Int J Cancer. 126:1882–1894. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Tomar S, Graves CA, Altomare D, Kowli S, Kassler S, Sutkowski N, Gillespie MB, Creek KE and Pirisi L: Human papillomavirus status and gene expression profiles of oropharyngeal and oral cancers from European American and African American patients. Head Neck. 38 Suppl 1:e694–e704. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Johannsen E and Lambert PF: Epigenetics of human papillomaviruses. Virology. 445:205–212. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Gameiro SF, Kolendowski B, Zhang A, Barrett JW, Nichols AC, Torchia J and Mymryk JS: Human papillomavirus dysregulates the cellular apparatus controlling the methylation status of H3K27 in different human cancers to consistently alter gene expression regardless of tissue of origin. Oncotarget. 8:72564–72576. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Gupta PC, Murti PR, Bhonsle RB, Mehta FS and Pindborg JJ: Effect of cessation of tobacco use on the incidence of oral mucosal lesions in a 10 yr follow-up study of 12,212 users. Oral Dis. 1:54–58. 1995. View Article : Google Scholar : PubMed/NCBI

41 

Saman DM: A review of the epidemiology of oral and pharyngeal carcinoma: Update. Head Neck Oncol. 4:12012. View Article : Google Scholar : PubMed/NCBI

42 

Gandini S, Botteri E, Iodice S, Boniol M, Lowenfels AB, Maisonneuve P and Boyle P: Tobacco smoking and cancer: A meta-analysis. Int J Cancer. 122:155–164. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Marron M, Boffetta P, Zhang Z-F, Zaridze D, Wünsch-Filho V, Winn DM, Wei Q, Talamini R, Szeszenia-Dabrowska N, Sturgis EM, et al: Cessation of alcohol drinking, tobacco smoking and the reversal of head and neck cancer risk. Int J Epidemiol. 39:182–196. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Mahapatra S, Kamath R, Shetty BK and Binu VS: Risk of oral cancer associated with gutka and other tobacco products: A hospital-based case-control study. J Cancer Res Ther. 11:199–203. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Reidy J, McHugh E and Stassen LFA: A review of the relationship between alcohol and oral cancer. Surgeon. 9:278–283. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Epstein MA, Achong BG and Barr YM: Virus particles in cultured lymphoblasts from burkitt's lymphoma. Lancet. 1:702–703. 1964. View Article : Google Scholar : PubMed/NCBI

47 

IARC (International Agency for Research on Cancer), . Epstein-Barr virus. A review o. IARC; Lyon: 2012

48 

Prabhu SR and Wilson DF: Evidence of Epstein-Barr virus association with head and neck cancers: a review. J Can Dent Assoc. 82:g22016.PubMed/NCBI

49 

Higa M, Kinjo T, Kamiyama K, Iwamasa T, Hamada T and Iyama K: Epstein-Barr virus (EBV) subtype in EBV related oral squamous cell carcinoma in Okinawa, a subtropical island in southern Japan, compared with Kitakyushu and Kumamoto in mainland Japan. J Clin Pathol. 55:414–423. 2002. View Article : Google Scholar : PubMed/NCBI

50 

Dawson MA and Kouzarides T: Cancer epigenetics: From mechanism to therapy. Cell. 150:12–27. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Jones PA and Baylin SB: The fundamental role of epigenetic events in cancer. Nat Rev Genet. 3:415–428. 2002. View Article : Google Scholar : PubMed/NCBI

52 

Hema KN, Smitha T, Sheethal HS and Mirnalini SA: Epigenetics in oral squamous cell carcinoma. J Oral Maxillofac Pathol. 21:252–259. 2017. View Article : Google Scholar : PubMed/NCBI

53 

Mascolo M, Siano M, Ilardi G, Russo D, Merolla F, De Rosa G and Staibano S: Epigenetic disregulation in oral cancer. Int J Mol Sci. 13:2331–2353. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Mitsukawa K, Lu X and Bartfai T: Galanin, galanin receptors and drug targets. Cell Mol Life Sci. 65:1796–1805. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Kanazawa T, Misawa K and Carey TE: Galanin receptor subtypes 1 and 2 as therapeutic targets in head and neck squamous cell carcinoma. Expert Opin Ther Targets. 14:289–302. 2010. View Article : Google Scholar : PubMed/NCBI

56 

Kanazawa T, Iwashita T, Kommareddi P, Nair T, Misawa K, Misawa Y, Ueda Y, Tono T and Carey TE: Galanin and galanin receptor type 1 suppress proliferation in squamous carcinoma cells: Activation of the extracellular signal regulated kinase pathway and induction of cyclin-dependent kinase inhibitors. Oncogene. 26:5762–5771. 2007. View Article : Google Scholar : PubMed/NCBI

57 

Kanazawa T, Kommareddi PK, Iwashita T, Kumar B, Misawa K, Misawa Y, Jang I, Nair TS, Iino Y and Carey TE: Galanin receptor subtype 2 suppresses cell proliferation and induces apoptosis in p53 mutant head and neck cancer cells. Clin Cancer Res. 15:2222–2230. 2009. View Article : Google Scholar : PubMed/NCBI

58 

Misawa K, Misawa Y, Kanazawa T, Mochizuki D, Imai A, Endo S, Carey TE and Mineta H: Epigenetic inactivation of galanin and GALR1/2 is associated with early recurrence in head and neck cancer. Clin Exp Metastasis. 33:187–195. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Khammanivong A, Wang C, Sorenson BS, Ross KF and Herzberg MC: S100A8/A9 (calprotectin) negatively regulates G2/M cell cycle progression and growth of squamous cell carcinoma. PLoS One. 8:e693952013. View Article : Google Scholar : PubMed/NCBI

60 

Khammanivong A, Sorenson BS, Ross KF, Dickerson EB, Hasina R, Lingen MW and Herzberg MC: Involvement of calprotectin (S100A8/A9) in molecular pathways associated with HNSCC. Oncotarget. 7:14029–14047. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Hessian PA and Fisher L: The heterodimeric complex of MRP-8 (S100A8) and MRP-14 (S100A9). Antibody recognition, epitope definition and the implications for structure. Eur J Biochem. 268:353–363. 2001. View Article : Google Scholar : PubMed/NCBI

62 

Lim Y, Sun CX, Tran P and Punyadeera C: Salivary epigenetic biomarkers in head and neck squamous cell carcinomas. Biomarkers Med. 10:301–313. 2016. View Article : Google Scholar

63 

Ovchinnikov DA, Cooper MA, Pandit P, Coman WB, Cooper-White JJ, Keith P, Wolvetang EJ, Slowey PD and Punyadeera C: Tumor-suppressor Gene Promoter Hypermethylation in Saliva of Head and Neck Cancer Patients. Transl Oncol. 5:321–326. 2012. View Article : Google Scholar : PubMed/NCBI

64 

Pfaffe T, Cooper-White J, Beyerlein P, Kostner K and Punyadeera C: Diagnostic potential of saliva: Current state and future applications. Clin Chem. 57:675–687. 2011. View Article : Google Scholar : PubMed/NCBI

65 

Collet C and Candy J: How many insulin-like growth factor binding proteins? Mol Cell Endocrinol. 139:1–6. 1998. View Article : Google Scholar : PubMed/NCBI

66 

Chen Y, Cui T, Knösel T, Yang L, Zöller K and Petersen I: IGFBP7 is a p53 target gene inactivated in human lung cancer by DNA hypermethylation. Lung Cancer. 73:38–44. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Chen LH, Liu DW, Chang JL, Chen PR, Hsu LP, Lin HY, Chou YF, Lee CF, Yang MC, Wen YH, et al: Methylation status of insulin-like growth factor-binding protein 7 concurs with the malignance of oral tongue cancer. J Exp Clin Cancer Res. 34:202015. View Article : Google Scholar : PubMed/NCBI

68 

Segre JA, Bauer C and Fuchs E: Klf4 is a transcription factor required for establishing the barrier function of the skin. Nat Genet. 22:356–360. 1999. View Article : Google Scholar : PubMed/NCBI

69 

Feinberg MW, Wara AK, Cao Z, Lebedeva MA, Rosenbauer F, Iwasaki H, Hirai H, Katz JP, Haspel RL, Gray S, et al: The Kruppel-like factor KLF4 is a critical regulator of monocyte differentiation. EMBO J. 26:4138–4148. 2007. View Article : Google Scholar : PubMed/NCBI

70 

Schmidt R and Plath K: The roles of the reprogramming factors Oct4, Sox2 and Klf4 in resetting the somatic cell epigenome during induced pluripotent stem cell generation. Genome Biol. 13:2512012. View Article : Google Scholar : PubMed/NCBI

71 

Wei D, Gong W, Kanai M, Schlunk C, Wang L, Yao JC, Wu TT, Huang S and Xie K: Drastic down-regulation of Krüppel-like factor 4 expression is critical in human gastric cancer development and progression. Cancer Res. 65:2746–2754. 2005. View Article : Google Scholar : PubMed/NCBI

72 

Le Magnen C, Bubendorf L, Ruiz C, Zlobec I, Bachmann A, Heberer M, Spagnoli GC, Wyler S and Mengus C: Klf4 transcription factor is expressed in the cytoplasm of prostate cancer cells. Eur J Cancer. 49:955–963. 2013. View Article : Google Scholar : PubMed/NCBI

73 

Naranjo Gómez JM, Bernal JFV, Arranz PG, Fernández SL and Roman JJG: Alterations in the expression of p53, KLF4, and p21 in neuroendocrine lung tumors. Arch Pathol Lab Med. 138:936–942. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Ohnishi S, Ohnami S, Laub F, Aoki K, Suzuki K, Kanai Y, Haga K, Asaka M, Ramirez F and Yoshida T: Downregulation and growth inhibitory effect of epithelial-type Krüppel-like transcription factor KLF4, but not KLF5, in bladder cancer. Biochem Biophys Res Commun. 308:251–256. 2003. View Article : Google Scholar : PubMed/NCBI

75 

Li W, Liu M, Su Y, Zhou X, Liu Y and Zhang X: The Janus-faced roles of Krüppel-like factor 4 in oral squamous cell carcinoma cells. Oncotarget. 6:44480–44494. 2015. View Article : Google Scholar : PubMed/NCBI

76 

Guillemin R: Hypothalamic hormones a.k.a. hypothalamic releasing factors. J Endocrinol. 184:11–28. 2005. View Article : Google Scholar : PubMed/NCBI

77 

Theodoropoulou M and Stalla GK: Somatostatin receptors: From signaling to clinical practice. Front Neuroendocrinol. 34:228–252. 2013. View Article : Google Scholar : PubMed/NCBI

78 

Lamberts SW, van der Lely AJ, de Herder WW and Hofland LJ: Octreotide. N Engl J Med. 334:246–254. 1996. View Article : Google Scholar : PubMed/NCBI

79 

Misawa K, Misawa Y, Kondo H, Mochizuki D, Imai A, Fukushima H, Uehara T, Kanazawa T and Mineta H: Aberrant methylation inactivates somatostatin and somatostatin receptor type 1 in head and neck squamous cell carcinoma. PLoS One. 10:e01185882015. View Article : Google Scholar : PubMed/NCBI

80 

Esquela-Kerscher A and Slack FJ: Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 6:259–269. 2006. View Article : Google Scholar : PubMed/NCBI

81 

Peschiaroli A, Giacobbe A, Formosa A, Markert EK, Bongiorno-Borbone L, Levine AJ, Candi E, D'Alessandro A, Zolla L, Finazzi Agrò A, et al: miR-143 regulates hexokinase 2 expression in cancer cells. Oncogene. 32:797–802. 2013. View Article : Google Scholar : PubMed/NCBI

82 

Huang WC, Chan SH, Jang TH, Chang JW, Ko YC, Yen TC, Chiang SL, Chiang WF, Shieh TY, Liao CT, et al: miRNA-491-5p and GIT1 serve as modulators and biomarkers for oral squamous cell carcinoma invasion and metastasis. Cancer Res. 74:751–764. 2014. View Article : Google Scholar : PubMed/NCBI

83 

Alterio D, Bacigalupo A, Cantù G, et al: Linee guida - Tumori della testa e del collo. Aiom, Milano. 2016.(In Italian).

84 

Colevas AD, Yom SS, Pfister DG, Spencer S, Adelstein D, Adkins D, Brizel DM, Burtness B, Busse PM, Caudell JJ, et al: Head and neck cancers, version 1.2018. J Natl Compr Canc Netw. 16:479–490. 2018. View Article : Google Scholar : PubMed/NCBI

85 

Chiappinelli KB, Strissel PL, Desrichard A, Li H, Henke C, Akman B, Hein A, Rote NS, Cope LM, Snyder A, et al: Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell. 162:974–986. 2015. View Article : Google Scholar : PubMed/NCBI

86 

Roulois D, Loo Yau H, Singhania R, Wang Y, Danesh A, Shen SY, Han H, Liang G, Jones PA, Pugh TJ, et al: DNA-demethylating agents target colorectal cancer cells by inducing viral mimicry by endogenous transcripts. Cell. 162:961–973. 2015. View Article : Google Scholar : PubMed/NCBI

87 

Nabet BY, Qiu Y, Shabason JE, Wu TJ, Yoon T, Kim BC, Benci JL, DeMichele AM, Tchou J, Marcotrigiano J, et al: Exosome RNA unshielding couples stromal activation to pattern recognition receptor signaling in cancer. Cell. 170:352–366.e13. 2017. View Article : Google Scholar : PubMed/NCBI

88 

Yang X, Han H, De Carvalho DD, Lay FD, Jones PA and Liang G: Gene body methylation can alter gene expression and is a therapeutic target in cancer. Cancer Cell. 26:577–590. 2014. View Article : Google Scholar : PubMed/NCBI

89 

Jones PA, Issa J-PJ and Baylin S: Targeting the cancer epigenome for therapy. Nat Rev Genet. 17:630–641. 2016. View Article : Google Scholar : PubMed/NCBI

90 

Mani S and Herceg Z: DNA demethylating agents and epigenetic therapy of cancer. Adv Genet. 70:327–340. 2010.PubMed/NCBI

91 

Hammond SM: An overview of microRNAs. Adv Drug Deliv Rev. 87:3–14. 2015. View Article : Google Scholar : PubMed/NCBI

92 

Li Y, Geng P, Jiang W, Wang Y, Yao J, Lin X, Liu J, Huang L, Su B and Chen H: Enhancement of radiosensitivity by 5-Aza-CdR through activation of G2/M checkpoint response and apoptosis in osteosarcoma cells. Tumour Biol. 35:4831–4839. 2014. View Article : Google Scholar : PubMed/NCBI

93 

Festuccia C, Gravina GL, D'Alessandro AM, Muzi P, Millimaggi D, Dolo V, Ricevuto E, Vicentini C and Bologna M: Azacitidine improves antitumor effects of docetaxel and cisplatin in aggressive prostate cancer models. Endocr Relat Cancer. 16:401–413. 2009. View Article : Google Scholar : PubMed/NCBI

94 

Qiu H, Yashiro M, Shinto O, Matsuzaki T and Hirakawa K: DNA methyltransferase inhibitor 5-aza-CdR enhances the radiosensitivity of gastric cancer cells. Cancer Sci. 100:181–188. 2009. View Article : Google Scholar : PubMed/NCBI

95 

Brieger J, Mann SA, Pongsapich W, Koutsimpelas D, Fruth K and Mann WJ: Pharmacological genome demethylation increases radiosensitivity of head and neck squamous carcinoma cells. Int J Mol Med. 29:505–509. 2012.PubMed/NCBI

96 

De Schutter H, Kimpe M, Isebaert S and Nuyts S: A systematic assessment of radiation dose enhancement by 5-Aza-2′-deoxycytidine and histone deacetylase inhibitors in head-and-neck squamous cell carcinoma. Int J Radiat Oncol Biol Phys. 73:904–912. 2009. View Article : Google Scholar : PubMed/NCBI

97 

Huang S-H and O'Sullivan B: Oral cancer: Current role of radiotherapy and chemotherapy. Med Oral Patol Oral Cir Bucal. 18:e233–e240. 2013. View Article : Google Scholar : PubMed/NCBI

98 

Basu T, Laskar SG, Gupta T, Budrukkar A, Murthy V and Agarwal JP: Toxicity with radiotherapy for oral cancers and its management: A practical approach. J Cancer Res Ther. 8 Suppl 1:S72–S84. 2012.PubMed/NCBI

99 

Sun W, Zaboli D, Liu Y, Arnaoutakis D, Khan T, Wang H, Koch W, Khan Z and Califano JA: Comparison of promoter hypermethylation pattern in salivary rinses collected with and without an exfoliating brush from patients with HNSCC. PLoS One. 7:e336422012. View Article : Google Scholar : PubMed/NCBI

100 

Schussel J, Zhou XC, Zhang Z, Pattani K, Bermudez F, Jean-Charles G, McCaffrey T, Padhya T, Phelan J, Spivakovsky S, et al: EDNRB and DCC salivary rinse hypermethylation has a similar performance as expert clinical examination in discrimination of oral cancer/dysplasia versus benign lesions. Clin Cancer Res. 19:3268–3275. 2013. View Article : Google Scholar : PubMed/NCBI

101 

Lima LMC, de Souza LR, da Silva TF, Pereira CS, Guimarães ALS, de Paula AMB and de Andrade Carvalho H: DNA repair gene excision repair cross complementing-group 1 (ERCC1) in head and neck squamous cell carcinoma: Analysis of methylation and polymorphism (G19007A), protein expression and association with epidemiological and clinicopathological factors. Histopathology. 60:489–496. 2012. View Article : Google Scholar : PubMed/NCBI

102 

Guerrero-Preston R, Soudry E, Acero J, Orera M, Moreno-López L, Macía-Colón G, Jaffe A, Berdasco M, Ili-Gangas C, Brebi-Mieville P, et al: NID2 and HOXA9 promoter hypermethylation as biomarkers for prevention and early detection in oral cavity squamous cell carcinoma tissues and saliva. Cancer Prev Res (Phila). 4:1061–1072. 2011. View Article : Google Scholar : PubMed/NCBI

103 

Demokan S, Chang X, Chuang A, Mydlarz WK, Kaur J, Huang P, Khan Z, Khan T, Ostrow KL, Brait M, et al: KIF1A and EDNRB are differentially methylated in primary HNSCC and salivary rinses. Int J Cancer. 127:2351–2359. 2010. View Article : Google Scholar : PubMed/NCBI

104 

Ovchinnikov DA, Wan Y, Coman WB, Pandit P, Cooper-White JJ, Herman JG and Punyadeera C: DNA Methylation at the Novel CpG Sites in the Promoter of MED15/PCQAP Gene as a Biomarker for Head and Neck Cancers. Biomark Insight. 9:53–60. 2014. View Article : Google Scholar

105 

Sun W, Zaboli D, Wang H, Liu Y, Arnaoutakis D, Khan T, Khan Z, Koch WM and Califano JA: Detection of TIMP3 promoter hypermethylation in salivary rinse as an independent predictor of local recurrence-free survival in head and neck cancer. Clin Cancer Res. 18:1082–1091. 2012. View Article : Google Scholar : PubMed/NCBI

106 

Huang YK, Peng BY, Wu CY, Su CT, Wang HC and Lai HC: DNA methylation of PAX1 as a biomarker for oral squamous cell carcinoma. Clin Oral Investig. 18:801–808. 2014. View Article : Google Scholar : PubMed/NCBI

107 

Rettori MM, de Carvalho AC, Bomfim Longo AL, de Oliveira CZ, Kowalski LP, Carvalho AL and Vettore AL: Prognostic significance of TIMP3 hypermethylation in post-treatment salivary rinse from head and neck squamous cell carcinoma patients. Carcinogenesis. 34:20–27. 2013. View Article : Google Scholar : PubMed/NCBI

108 

Momen-Heravi F, Trachtenberg AJ, Kuo WP and Cheng YS: Genomewide Study of Salivary MicroRNAs for Detection of Oral Cancer. J Dent Res. 93 Suppl:86S–93S. 2014. View Article : Google Scholar : PubMed/NCBI

109 

Salazar C, Calvopiña D and Punyadeera C: miRNAs in human papilloma virus associated oral and oropharyngeal squamous cell carcinomas. Expert Rev Mol Diagn. 14:1033–1040. 2014. View Article : Google Scholar : PubMed/NCBI

110 

Liu CJ, Lin SC, Yang CC, Cheng HW and Chang KW: Exploiting salivary miR-31 as a clinical biomarker of oral squamous cell carcinoma. Head Neck. 34:219–224. 2012. View Article : Google Scholar : PubMed/NCBI

111 

Salazar C, Nagadia R, Pandit P, Cooper-White J, Banerjee N, Dimitrova N, Coman WB and Punyadeera C: A novel saliva-based microRNA biomarker panel to detect head and neck cancers. Cell Oncol (Dordr). 37:331–338. 2014. View Article : Google Scholar : PubMed/NCBI

112 

Park NJ, Zhou H, Elashoff D, Henson BS, Kastratovic DA, Abemayor E and Wong DT: Salivary microRNA: Discovery, characterization, and clinical utility for oral cancer detection. Clin Cancer Res. 15:5473–5477. 2009. View Article : Google Scholar : PubMed/NCBI

113 

Wiklund ED, Gao S, Hulf T, Sibbritt T, Nair S, Costea DE, Villadsen SB, Bakholdt V, Bramsen JB, Sørensen JA, et al: MicroRNA alterations and associated aberrant DNA methylation patterns across multiple sample types in oral squamous cell carcinoma. PLoS One. 6:e278402011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2018
Volume 9 Issue 4

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Russo D, Merolla F, Varricchio S, Salzano G, Zarrilli G, Mascolo M, Strazzullo V, Di Crescenzo RM, Celetti A, Ilardi G, Ilardi G, et al: Epigenetics of oral and oropharyngeal cancers (Review) Corrigendum in /10.3892/br.2020.1290. Biomed Rep 9: 275-283, 2018
APA
Russo, D., Merolla, F., Varricchio, S., Salzano, G., Zarrilli, G., Mascolo, M. ... Ilardi, G. (2018). Epigenetics of oral and oropharyngeal cancers (Review) Corrigendum in /10.3892/br.2020.1290. Biomedical Reports, 9, 275-283. https://doi.org/10.3892/br.2018.1136
MLA
Russo, D., Merolla, F., Varricchio, S., Salzano, G., Zarrilli, G., Mascolo, M., Strazzullo, V., Di Crescenzo, R. M., Celetti, A., Ilardi, G."Epigenetics of oral and oropharyngeal cancers (Review) Corrigendum in /10.3892/br.2020.1290". Biomedical Reports 9.4 (2018): 275-283.
Chicago
Russo, D., Merolla, F., Varricchio, S., Salzano, G., Zarrilli, G., Mascolo, M., Strazzullo, V., Di Crescenzo, R. M., Celetti, A., Ilardi, G."Epigenetics of oral and oropharyngeal cancers (Review) Corrigendum in /10.3892/br.2020.1290". Biomedical Reports 9, no. 4 (2018): 275-283. https://doi.org/10.3892/br.2018.1136