Review Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Methodol. Mar 26, 2016; 6(1): 77-86
Published online Mar 26, 2016. doi: 10.5662/wjm.v6.i1.77
Updated overview of current biomarkers in head and neck carcinoma
Kiran Dahiya, Department of Biochemistry, Pt. B.D.Sharma PGIMS, Rohtak 124001, Haryana, India
Rakesh Dhankhar, Department of Radiotherapy, Pt. B.D.Sharma PGIMS, Rohtak 124001, Haryana, India
Author contributions: Dahiya K and Dhankhar R contributed to this article.
Conflict-of-interest statement: Authors declare no conflict of interests for this article.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Dr. Kiran Dahiya, Department of Biochemistry, Pt. B.D.Sharma PGIMS, 778/28, Bharat Colony, Rohtak 124001, Haryana, India. kirandahiya_2002@yahoo.com
Telephone: +91-98-96111985
Received: September 9, 2015
Peer-review started: September 9, 2015
First decision: November 7, 2015
Revised: January 20, 2016
Accepted: March 7, 2016
Article in press: March 9, 2016
Published online: March 26, 2016

Abstract

Squamous cell cancer is the most common type of malignancy arising from the epithelial cells of the head and neck region. Head and neck squamous cell carcinoma (HNSCC) is one of the predominant causes of cancer related casualties worldwide. Overall prognosis in this disease has improved to some extent with the advancements in therapeutic modalities but detection of primary tumor at its initial stage and prevention of relapse are the major targets to be achieved for further improvement in terms of survival rate of patients. Latest achievements in basic research regarding molecular characterization of the disease has helped in better perception of the molecular mechanisms involved in HNSCC progression and also in recognizing and targeting various molecular biomarkers associated with HNSCC. In the present article, we review the information regarding latest and potential biomarkers for the early detection of HNSCC. A detailed molecular characterization, ultimately, is likely to improve the development of new therapeutic strategies, potentially relevant to diagnosis and prognosis of head and neck cancers. The need for more accurate and timely disease prediction has generated enormous research interests in this field.

Key Words: Head and neck squamous cell carcinoma, Early detection, Prognosis, Biomarkers, Molecular level

Core tip: Early detection of head and neck squamous cell carcinoma is vital in improving the overall survival and prognosis. It can be achieved by use of latest biomarkers. With advancement in knowledge of molecular characteristics of this disease, various biomarkers acting at molecular level have been identified. This review compiles information regarding the potential players in this field.



INTRODUCTION

The term head and neck carcinoma encompasses all malignancies arising in the nasal and oral cavities, pharynx, larynx and the paranasal sinuses. Majority of these (approximately 95%) epithelial cancers are squamous cell carcinomas[1]. Smoking and alcoholism are two well known predisposing factors[2]. Head and neck squamous cell carcinoma (HNSCC) is reported to be the sixth common cause of cancer mortality throughout the world[2].

There is no significant improvement in the mortality rates even with continuous research and trials in the field of diagnostics and therapeutics[3]. As compared to other cancers like breast, cervix and colorectal, the five-year survival rate of HNSCC after diagnosis is significantly lower[4,5]. The reason for this could be failure in early diagnosis and insufficient effectiveness of therapeutic modalities[6,7]. The predominant cause of mortality in HNSCC is regional and/or distant metastatic spreading of tumor cells from primary site[8]. Therefore, the vital area in the treatment of head and neck cancers is ability to diagnose it at an early stage.

EARLY DIAGNOSIS OF HNSCC

Till date only one third cases of HNSCC are being diagnosed at an early stage and rest land up with an advanced disease in the United States[9,10]. The major reason put forward for this trend include a lack of appropriate screening biomarkers[11]. The treatment of neoplasia is most effective in its early stage when the tumor size at primary site is lowest with least lymphatic and hematogenous spread. Therefore, early diagnosis and intervention is of utmost significance in the treatment of HNSCC. Here comes the role of biomarkers. Biomarkers may be analyzed in the tissue itself, plasma or other body fluids like saliva in case of HNSCC. The drawback of biomarkers may include lack of specificity and sensitivity but these may prove to be essential tools in timely diagnosis of the disease[12]. A variety of biomarkers have been reported in literature with a promising potential but these are still in the need of clinical validation. In this article, we present a review of different biomarkers which may be utilized in early diagnosis and timely decision-making for intervention in patients of HNSCC.

ALTERATION IN EXPRESSION OF CHEMOKINE RECEPTORS

Recently, the importance of chemokines and their cognate receptors in head and neck cancers is being reported by increasing number of studies.

CXC chemokine receptor 2

In the squamous cell carcinoma of the larynx, the expression of CXC chemokine receptor 2 (CXCR2) has been observed to be substantially higher in tumor tissue than that in the paraneoplastic tissue. The increased expression has been reported to be significantly related with lymph node metastasis, histological grade and 5-year survival of these patients. Thus, expression of CXCR2 can be considered as a potent prognostic marker for laryngeal squamous cell carcinoma[13].

CXCR4

The importance of CXCR4 in tumour progression and organ-specific metastasis in patients with HNSCC has been reported by a number of authors[14,15]. Wang et al studied the expression of CXCR4 in nasopharyngeal carcinoma tissues and found an increased CXCR4 expression in tumor tissues. Besides this, they also suggested that the increased expression of CXCR4 may be correlated with increased metastatic rates and poor overall survival of the patients[16]. This finding was consistent with another study which also reported significantly elevated CXCR4 mRNA in HNSCC tissues as compared to paraneoplastic tissues and that the increased expression was associated with increased risk of lymph node metastasis and distant metastasis[17]. Therefore, CXCR4 expression can also be used as a marker to predict prognosis and metastasis in patients with HNSCC.

CC chemokine receptor 7

CC chemokine receptor 7 (CCR7) is another CC chemokine receptor, which has been demonstrated to play a significant role in the migration of activated dendritic cells to regional lymph nodes. Its expression has also been reported to be elevated in HNSCC tumor tissues as compared to paraneoplastic tissues. Furthermore, the elevated expression of CCR7 has been found to correlate with lymph node metastasis and tumor tissue histological differentiation status[17]. Similar findings have been reported by another study which analyzed the expression of CCR7 in primary and metastatic tumor cell lines and also in biopsy material from both primary and metastatic lesions. They reported that CCR7 expression was increased in metastatic cells and tissues[18]. On the basis of these reports, an important role may be conferred to CCR7 in predicting the metastasis and prognosis in HNSCC patients.

HUMAN PAPILLOMA VIRUS

Human papilloma virus (HPV), especially HPV16, is considered one of the causing factors for HNSCC. HPV DNA has been found in 15% to 25% of HNSCC and the association differs depending on the site of the tumor[19]. HPV DNA is detected in 45%-67% of cases of cancers of the tonsil, in 13%-25% of hypopharyngeal cancer, in 12%-18% of the cancers of oral cavity and in 3%-7% of carcinoma larynx and it may be associated with prognosis of disease, especially in tonsillar cancers[20]. There are reports in literature suggesting that HNSCC with HPV has a favorable prognosis and that, it in fact, is a distinct clinicopathological entity[21]. HPV16 and HPV18 are considered to be the high risk HPVs, which produce E6 and E7 oncoproteins, implicated in transformation of cell and altering the control of cell cycle. Oncoprotein E7 binds to and induces the proteolysis of pRb while E6 inactivates p53 by accelerating its ubiquitin mediated degradation[22]. Thus, HPV DNA may act as a diagnostic and prognostic marker in patients of HNSCC.

It is of interest to know that adding p16INK4A immunostaining to HPV DNA detection may prove to be very useful in diagnosing HPV-related oral squamous cell carcinoma and it has been observed that HPV(+) and p16INK4A(+) types of tumors have better prognosis[23]. As reported by Danish Head and Neck Cancer Group 5 trial, p16INK4A(+) tumors appeared to be associated more strongly with poor histopathologic differentiation as compared to the p16INK4A(-) ones, but the difference was not statistically significant, indicating that p16INK4A alone is not an adequate marker[24]. In the study of panitumamub efficacy in patients with recurrent and/or metastatic head and neck cancer (SPECTRUM), the authors reported that the p16INK4A status of the tumor might have significant bearings in designing future trials in cases of recurrent or metastatic HNSCC[25].

MICROSATELLITE INSTABILITY

Microsatellite instability (MSI) may be analyzed using different markers. Researchers have detected loss of heterozygosity (LOH) in tumor cell derived DNA (deoxyribonucleic acid) from mouth washing or lesion brushing samples in patients with T2N0M0 and T1N0M0 tumors[26]. MSI analysis in tumor cell DNA is of value in detection of pre-malignant conditions like erythroplakia and leukoplakia[1]. It has also been reported that LOH of 9p21 may be an initial event in HNSCC and may be associated with preneoplastic lesions as well as 30% of cases of squamous cell carcinoma[27]. Loss of chromosomal region 9p21 is seen in > 70% of cases, making it the most frequent genetic alteration seen in squamous cell dysplasia and HNSCC[27,28]. Some of the studies in which MSI was analyzed using different set of markers in patients with HNSCC have also reported MSI in 12.5%-35% of the cases while microsatellite alteration rate was detected to be 75%-95%[29-31]. Instability frequency has been reported to be related to the repeat unit length and overall size of the short tandem repeat (STR) affecting the probability of error during DNA replication. STR characteristics vary in different populations and those with longer average repeat size are more prone to instability than the ones having smaller repeat size[31].

MSI analyses have the disadvantage of lack of uniformity in selection of different methods or the type and number of markers evaluated[32]. A standard approach is yet to be developed for this marker to be useful as an early diagnostic marker in HNSCC patients.

METHYLATION

Gene activation due to hypermethylation of cytosine-phosphate-guanine (CpG)-rich promoter regions has been reported in early stages of HNSCC[30]. A specificity of 96% in salivary specimens for methylation specific polymerase chain reaction has been reported for detection of HNSCC[33]. Whereas it was observed to be 90% in salivary samples and 72% for serum samples in yet another study[34]. The lower rate of promoter hypermethylation may be due to dilution with normal, non-methylated DNA from normal mucosal areas[34]. It has also been reported that promoter hypermethylation may be associated with age and ethnicity of the patient or with history of chronic tobacco or alcohol consumption[35,36].

The disadvantages of methylation markers include lack of sensitivity, specificity, complexity and inconvenience in HNSCC detection in body fluids.

METALLOPROTEINASES

These include a large number of zinc and calcium dependent endopeptidases. These enzymes are implicated in extracellular matrix degradation leading to spread of the tumor cells out of the tissue of origin[37-39]. Besides migration of tumor cells, metalloproteinases (MMPs) play a significant role in providing a microenvironment conducive for the growth and angiogenesis of tumors. These also help in cellular differentiation, proliferation and apoptosis in tumor tissues[38]. Several types of MMPs, e.g., MMP-1, the gelatinases (MMP-2 and MMP-9) and the stromelysins (MMP-3 and MMP-10) play a role in tissue invasion by cancer cells and metastasis[40,41]. Elevated levels of MMP-2 or MMP-9 have been observed in many types of cancers including HNSCC, lung, breast, colorectal and ovarian carcinoma indicating an association with tumor progression[42-45].

In HNSCC, patients have been found to have increased levels of MMP-3, MMP-8 and MMP-9[46] while MMP-1 and MMP-10 have been reported to be useful for detection of cancer of oral cavity and gingiva[47]. In another study, MMP-9 has been reported to be able to detect stage I HNSCC disease with 80% positivity[48]. The disadvantage with MMP-9 lies in its poor specificity to discriminate cancer with benign disease[49].

INTERLEUKINS

Interleukin (IL)-6 and IL-8 have been linked with tumor progression and metastasis along with playing a role in the process of carcinogenesis[12]. IL-8 holds potential for acting as an early biomarker in salivary samples while IL-6 in serum samples for detection of oral cavity or oropharynx squamous cell carcinoma (OSCC)[50,51]. In some other studies, increased levels of IL-6 and IL-8 have been reported in a variety of specimens like cell line supernatants, tumor tissues and serum of patients with HNSCC[52,53].

Zimmermann et al[54] reported that four mRNAs (OAZ, SAT, IL-8 and IL-1β) in salivary samples have a collective sensitivity and specificity of 91% in detection of cancer of oral cavity. On the other hand, the levels of salivary IL-8 were found to be raised in patients of OSCC as compared to controls but the difference was not statistically significant[55]. Thus, further studies are required to establish the sensitivity and specificity of IL-8 and IL-6 as biomarkers in patients of OSCC.

MICRO RNA

These are small non-coding RNA (ribonucleic acid) sequences playing a role in regulation of gene expression affecting a variety of physiological processes[56]. miRNAs, by virtue of their vast range of consequences may act both as oncogenes and tumor supressor genes[57]. In many types of cancers, dysregulation of genes for miRNAs has been reported and these can be used for detection and classification of different solid tumors[58]. The change in micro RNAs (miRNAs) in cancer cells as compared to normal cells has been reported to be many folds than the extent of change in mRNA[59].

It has been proposed that miR-106b-25 cluster and miR-375 may be involved in development and progression of HNSCC and that miR-451 could act as a prospective prognostic marker for recurrence in HNSCC patients. The same authors also observed one third of the miRNAs to be dysregulated in HNSCC[60]. Park et al[61] reported significantly lower levels of miR-125a and miR-200a in the saliva of OSCC patients as compared to controls. miR-205 has been found to have a variable expression in a number of tumor cells and, particularly, to be highly overexpressed in HNSCC cell lines and may prove helpful in detecting occult metastatic tumor deposits[62,63]. Deregulation of miR-138 has been commonly found in HNSCC and other types of cancer. A number of functional targets for miR-38 have been reported which include genes involved in initiation and progression of HNSCC[64]. It has also been demonstrated that restoration of transfected miR-34a mimics significantly inhibits the capability for epithelial-mesenchymal transition of cancer stem cell-phenotype and functionally decreases clonogenic and invasive capability in HNSCC cell lines[65].

Micro RNA biomarkers are superior to their mRNA counterparts. Because of their robust profiling and better stability in routine clinical samples, they may prove to be more suitable for analysis in some tissue samples[64]. Thus, miRNA may prove to be promising early biomarkers in detection of HNSCC but further research is needed to substantiate their role as screening tools.

MELANOMA-ASSOCIATED GENE

Melanoma-associated gene (MAGE) participates in the process of carcinogenesis by suppressing apoptosis[66]. Other similar tumor-specific shared antigen families like G antigen gene, B melanoma antigen gene and L antigen family 3 gene have been categorized at molecular levels[67-69]. These antigens, usually peptides in nature, may be significantly associated with tumor immunology as their expression has been found specific to tumor cells, e.g., HNSCC, melanoma, carcinoma ovary, bladder cancer, carcinoma lung and colorectal cancer[70-73]. Expression of MAGE A3 and A4 has been found to be positive in early invasive carcinoma (by excisional biopsy) where brush and incisional biopsy was negative in a suspicious looking leukoplakic lesion[74]. Expression of MAGE has also been shown in the sputum samples of patients with HNSCC[75]. Therefore, it may be used as an early biomarker for HNSCC detection as it has not been observed to be expressed in normal healthy tissues with exception of testis[76]. Other studies have also reported 85.5%-90% expression rate of MAGE in HNSCC tissue[77,78]. It may help in initiating target specific immunotherapy in these patients[79]. According to a recent report by Lee et al[80] expression of MAGE-A1-6 in sputum predicts poor oncologic outcome in patients with squamous cell carcinoma of the larynx and hypopharynx. MAGE-A expression has been reported to be associated with poorer five year survival rate, thus, indicating its potential as a prognostic marker also[81].

CENTROSOME ABNORMALITIES

Centrosome abnormalities have also been observed in HNSCC. It has been reported that 17 out of 18 tumor samples analyzed from patients with HNSCC demonstrated centrosome hyperamplification. Based on these findings, it has been suggested by authors that centrosomal hyperamplification could be used as a marker for HNSCC[82]. Furthermore, the p53 suppressor gene, the most commonly mutated gene found in human cancers, has been reported to correlate with centrosome hyperamplification in HNSCC. Centrosome hyperamplification is either observed in tumors with mutated p53 or in tumours that retain wild type p53 but with an overexpressed Mdm2, an oncogene which is responsible for inhibiting the transactivation function of p53[83]. Increased frequency of centrosomal abnormalities has also been seen in OSCC in cells with spindle checkpoint protein CDC20 overexpression[84]. This may be because of the fact that in cancer cells, genes that encode for proteins involved in mitotic checkpoints/mitotic regulations are generally found mutated or overexpressed.

ACTIN AND MYOSIN

These are cytoskeletal proteins responsible for cell motility and invasion which are important components of epithelial tumorigenesis[85]. Increased expression of actin and myosin has been observed in exfoliated cells present in soluble saliva in patients with malignancy as compared to those with pre-cancerous lesions[86]. Increased actin isoforms have been observed in invasive basal cell carcinoma[87], squamous cell carcinoma of cervix[88] and esophagus[89] and invasive OSCC[90]. Increase in myosin abundance has been observed in proteomics of tissue from OSCC region[91]. However, Turhani et al[92] have reported a lesser expression of myosin light chain in HNSCC contradicting the existing findings.

Thus, actin and myosin need rigorous research with larger sample groups including issues like sensitivity and specificity for their establishment as HNSCC biomarkers.

CYTOKERATINS

Cytokeratins (CKs) are one of the major components of intracellular filament network found in different tissues[93]. CKs are expressed in a number of combinations depending on the type of epithelial cell of origin[94]. They are further divided into two subtypes (I and II) that are generally coexpressed[95,96]. These are found to be overexpressed in OSCC tissue as compared to normal mucosa[93]. Overexpression of cytokeratins has been related with tumor progression and prognosis[97]. Constitutive expression of cytokeratin-17 (CK-17) in the lungs is only found in the normal basal cells[98]. It is now emerging as a tissue-specific immunohistochemical biomarker in squamous cell carcinoma of larynx[99]. CK-17 mRNA overexpression has been demonstrated in OSCC by few authors[96,99,100]. These studies were mainly performed in cancer tissue and not in saliva or serum samples. Increased expression of CK-17 has been demonstrated in respiratory syncitial virus infected epithelial cells also[95].

As CK-6 and CK-16 are found to be constitutively expressed in mucosal stratified squamous epithelia, they may be ragarded as markers of cellular hyperproliferation. CK-6 may also be considered as an additional squamous differentiation biomarker in poorly-differentiated cancers. Though CK-17 was also detected in most of the cases, its expression is not found to be uniform[101].

These markers need stringent workup with larger samples, including body fluids and also on issues regarding its specificity before their validation as biomarkers for HNSCC.

p53

It is the most frequently studied molecular marker in HNSCC[102]. The p53 pathway is activated when cells become old or damaged. The p53, a 53 kd protein, may then arrest cell cycle for DNA to be repaired or lead to apoptosis if damage is irreparable[103]. Alteration in function of p53 may be seen as a result of mutation or sequestration by other cellular proteins. Mutations of p53 gene are the most commonly encountered mutations in carcinomas including HNSCC[102,104]. p53 is associated with maintenance of cellular integrity and is regarded as guardian of genome[105]. Mutations in p53 in HNSCC patients have been reported by a number of researchers with an expression range of 50%-60% of the tumor cells[103,105-108]. Its expression can be conveniently studied with immunohistochemistry techniques for detection of cancers but complete role of p53 in pathogenesis of HNSCC is still not clear[109,110]. Survival rate has also been reported to be higher in p53 negative patients as compared to those who are positive for p53 mutations[102]. Thus, this marker has a fair potential for diagnostic and prognostic use in patients with HNSCC. Another interesting finding is that HPV infection rarely coexists with p53 mutation as both of them can independently lead to p53 inactivation implicated in HNSCC tumor[111].

EUKARYOTIC TRANSLATION FACTOR 4E

It is a protein involved in the initiation of protein synthesis[112]. Overexpression of eukaryotic translation factor 4E (eIF4E) has been found associated with different stages of carcinogenesis including metastasis. It is related with transformation of fibroblasts and primary epithelial cells[113,114]. Overexpression of this protein in mice has been found to be associated with a number of malignancies like lymphomas, angiosarcomas, hepatomas and carcinoma lung[115]. An expression of 100% in HNSCC has been reported in some studies[116,117]. Overexpression of eIF4E in cancers like breast, bladder, lung and HNSCC has been found to correlate with an increased risk of disease progression and poor prognosis[113,118-121]. Another study has reported overexpression of eIF4E in tumor free surgical margins to be related to loco-regional recurrence in patients of HNSCC[122].

Therefore, eIF4E may prove to be a significant independent prognostic predictor in terms of recurrence and survival in patients of HNSCC.

LOSS OF FUNCTION OF DNA REPAIR GENES

Effective DNA repair may be considered as a major determinant of cancer-free survival. Various mutations in DNA repair genes, especially, of the nucleotide excision repair (NER) group (XP genes in xeroderma pigmentosum patients), DNA crosslink repair (Fanconi anemia genes) mutations affecting the mismatch repair genes, and a number of others are the cause of several hereditary cancerous syndromes[123]. The dominant moderator of mismatch repair in HNSCC is promoter hypermethylation rather than direct mutation[124]. There is also limited data in HNSCC demonstrating a link between poly-(ADP-Ribose) polymerase overexpression and cisplatin resistance suggesting a possible role for chemoresistant tumours. Hyperphosphorylation of replication protein A, a single-strand DNA binding protein, that is, integral to HR, has also been implicated as a mechanism for cisplatin resistance in HNSCC cell lines[124]. Multiple (5-7) risk NER genotypes have been associated with a 2.4-fold increased relative risk of second primary HNSCC[124]. The inactivation of these DNA repair genes may be linked to carcinogenesis by decreasing genomic stability and producing certain genetic alterations[125] (Table 1).

Table 1 Comparison of different biomarkers of head neck squamous cell carcinoma.
MarkerMechanismType of specimenRoleLimitations
Chemokine receptorsIncreased expression in tumor tissueBiopsy specimenPrognosis and metastasisClinical validation by further research required
HPVDNA associated HNSCC, oncoprotein productionTumor tissueDiagnosis and prognosisLack of sensitivity and specificity
MSILOH in tumor derived DNAMouth washings/lesion brushingsDetection of pre-malignant lesionLack of uniformity of method
Methylation markersGene inactivation following hypermethylation in promoter regionSaliva/serumEarly detectionLack of sensitivity/specificity, complex methodology
MMPsProvide conducive microenvironment for tumor growth, degrade ECM promoting tumor migrationTumor tissue/salivaEarly detectionPoor specificity
InterleukinsParticipate in process of tumor growth and metastasisTumor tissue/cell line supernatants/saliva/serumEarly and convenient biomarkerLack of sensitivity and specificity
miRNARole in regulation of gene expressionTumor tissue/salivaEarly detectionClinical validation required
MAGESuppresses apoptosisBiopsy specimen/salivaPrognosis and in selecting targeted immunotherapyClinical validation required
Centrosome abnormalitiesMutation due to hyperamplificationTumor tissueEarly detectionFurther research required to understand molecular mechanism
Actin and myosinIncreased expression leading to greater invasivenessTumor tissue/salivaEarly detectionLack of sensitivity and specificity
CytokeratinsOver-expression associated with tumor progressionTumor tissue/saliva/serumEarly detection and prognosisClinical validation required
p53Mutation affects apoptosis/repair of malignant cellsTumor tissueDiagnosis, prognosis, convenient markerComplete role in HNSCC yet to be deciphered
eIF4EOverexpression associated with tranformation of fibroblasts and epithelial cellsTumor tissuePrognostic indicatorLack of sensitivity and specificity
ROLE OF IMAGING BIOMARKES

Besides biochemical and pathological biomarkers, a significant role is being played by imaging biomarkers in the early detection of head and neck oncology. To go into the details of these biomarkers is beyond the scope of the present article, but these are proving to be vital in initial staging, treatment planning, monitoring and follow-up of the patients with HNSCC non-invasively. 18F-fluoro-2-deoxyglucose positron emission tomography/computerized tomography (PET/CT) proves to be more sensitive and specific as compared to magnetic resonance imaging (MRI) or CT alone[126,127]. Recently introduced regional PET/Gd (gadolinium-enhanced T1-weighted)-MRI combined with whole-body PET/MRI appears to be quite promising in detecting early lesions[128]. There is a need for further refinement and a concerted approach regarding imaging and molecular biomarkers for HNSCC which may help in early detection, targeted therapy and improved monitoring.

CONCLUSION

Understanding the molecular mechanisms of HNSCC is important to identify its biomarkers. Finding genetic alterations can lead to early detection of the disease. These can be detected in tumor tissue, saliva/body fluids washing the affected tissue or in the serum. A variety of molecular markers have been explained in literature. There may be a tremendous role of these markers in affecting the outcome of the disease by aiding in timely diagnosis and even in selecting specific therapy. Many of these have already shown their potential in this field like interleukins, MAGE, MSI, etc., but still there are issues of specificity, sensitivity and clinical validation with some of these. With more standardised and uniform platform for sample selection, processing and data analysis along with stringent workup of the cases, these biomarkers may prove to be indispensable investigative tools in patients with HNSCC and may even help in better understanding of the pathogenesis of the disease. Thus, there is a strong hope that these molecular biomarkers or patterns of markers, alone or in co-ordination with imaging markers, could, in the future, be utilized for early detection of HNSCC, tumor metastasis and may aid in determining the best therapeutic modality for patient care.

Footnotes

P- Reviewer: Zaravinos A S- Editor: Qiu S L- Editor: A E- Editor: Liu SQ

References
1.  Kang H, Kiess A, Chung CH. Emerging biomarkers in head and neck cancer in the era of genomics. Nat Rev Clin Oncol. 2015;12:11-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 201]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
2.  Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin. 2005;55:74-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13286]  [Cited by in F6Publishing: 13416]  [Article Influence: 706.1]  [Reference Citation Analysis (1)]
3.  Chin D, Boyle GM, Theile DR, Parsons PG, Coman WB. Molecular introduction to head and neck cancer (HNSCC) carcinogenesis. Br J Plast Surg. 2004;57:595-602.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, Thun MJ. Cancer statistics, 2008. CA Cancer J Clin. 2008;58:71-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8283]  [Cited by in F6Publishing: 8159]  [Article Influence: 509.9]  [Reference Citation Analysis (0)]
5.  Jemal A, Thun MJ, Ries LA, Howe HL, Weir HK, Center MM, Ward E, Wu XC, Eheman C, Anderson R, Ajani UA, Kohler B, Edwards BK. Annual report to the nation on the status of cancer, 1975-2005, featuring trends in lung cancer, tobacco use, and tobacco control. J Natl Cancer Inst. 2008;100:1672-1694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 680]  [Cited by in F6Publishing: 827]  [Article Influence: 51.7]  [Reference Citation Analysis (0)]
6.  Mao L, Hong WK, Papadimitrakopoulou VA. Focus on head and neck cancer. Cancer Cell. 2004;5:311-316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 273]  [Cited by in F6Publishing: 280]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
7.  Forastiere A, Koch W, Trotti A, Sidransky D. Head and neck cancer. N Engl J Med. 2001;345:1890-1900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 974]  [Cited by in F6Publishing: 1009]  [Article Influence: 43.9]  [Reference Citation Analysis (0)]
8.  Miyazaki H, Patel V, Wang H, Edmunds RK, Gutkind JS, Yeudall WA. Down-regulation of CXCL5 inhibits squamous carcinogenesis. Cancer Res. 2006;66:4279-4284.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Mydlarz WK, Hennessey PT, Califano JA. Advances and Perspectives in the Molecular Diagnosis of Head and Neck Cancer. Expert Opin Med Diagn. 2010;4:53-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
10.  Schaaij-Visser TB, Brakenhoff RH, Leemans CR, Heck AJ, Slijper M. Protein biomarker discovery for head and neck cancer. J Proteomics. 2010;73:1790-1803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 55]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
11.  Tran N, O’Brien CJ, Clark J, Rose B. Potential role of micro-RNAs in head and neck tumorigenesis. Head Neck. 2010;32:1099-1111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 46]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
12.  Lee KD, Lee HS, Jeon CH. Body fluid biomarkers for early detection of head and neck squamous cell carcinomas. Anticancer Res. 2011;31:1161-1167.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Bektas-Kayhan K, Unur M, Boy-Metin Z, Cakmakoglu B. MCP-1 and CCR2 gene variants in oral squamous cell carcinoma. Oral Dis. 2012;18:55-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
14.  Tan CT, Chu CY, Lu YC, Chang CC, Lin BR, Wu HH, Liu HL, Cha ST, Prakash E, Ko JY. CXCL12/CXCR4 promotes laryngeal and hypopharyngeal squamous cell carcinoma metastasis through MMP-13-dependent invasion via the ERK1/2/AP-1 pathway. Carcinogenesis. 2008;29:1519-1527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 79]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
15.  Rehman AO, Wang CY. CXCL12/SDF-1 alpha activates NF-kappaB and promotes oral cancer invasion through the Carma3/Bcl10/Malt1 complex. Int J Oral Sci. 2009;1:105-118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 77]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
16.  Wang N, Wu QL, Fang Y, Mai HQ, Zeng MS, Shen GP, Hou JH, Zeng YX. Expression of chemokine receptor CXCR4 in nasopharyngeal carcinoma: pattern of expression and correlation with clinical outcome. J Transl Med. 2005;3:26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 47]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
17.  Ueda M, Shimada T, Goto Y, Tei K, Nakai S, Hisa Y, Kannagi R. Expression of CC-chemokine receptor 7 (CCR7) and CXC-chemokine receptor 4 (CXCR4) in head and neck squamous cell carcinoma. Auris Nasus Larynx. 2010;37:488-495.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 25]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
18.  Wang J, Xi L, Hunt JL, Gooding W, Whiteside TL, Chen Z, Godfrey TE, Ferris RL. Expression pattern of chemokine receptor 6 (CCR6) and CCR7 in squamous cell carcinoma of the head and neck identifies a novel metastatic phenotype. Cancer Res. 2004;64:1861-1866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 129]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
19.  Betiol J, Villa LL, Sichero L. Impact of HPV infection on the development of head and neck cancer. Braz J Med Biol Res. 2013;46:217-226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
20.  Perez-Ordoñez B, Beauchemin M, Jordan RC. Molecular biology of squamous cell carcinoma of the head and neck. J Clin Pathol. 2006;59:445-453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 190]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
21.  Licitra L, Perrone F, Bossi P, Suardi S, Mariani L, Artusi R, Oggionni M, Rossini C, Cantù G, Squadrelli M. High-risk human papillomavirus affects prognosis in patients with surgically treated oropharyngeal squamous cell carcinoma. J Clin Oncol. 2006;24:5630-5636.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 511]  [Cited by in F6Publishing: 496]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
22.  Vietía D, Liuzzi J, Avila M, De Guglielmo Z, Prado Y, Correnti M. Human papillomavirus detection in head and neck squamous cell carcinoma. Ecancermedicalscience. 2014;8:475.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 7]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
23.  Zaravinos A. An updated overview of HPV-associated head and neck carcinomas. Oncotarget. 2014;5:3956-3969.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 81]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
24.  Lassen P, Eriksen JG, Hamilton-Dutoit S, Tramm T, Alsner J, Overgaard J. Effect of HPV-associated p16INK4A expression on response to radiotherapy and survival in squamous cell carcinoma of the head and neck. J Clin Oncol. 2009;27:1992-1998.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 475]  [Cited by in F6Publishing: 457]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
25.  Vermorken JB, Stöhlmacher-Williams J, Davidenko I, Licitra L, Winquist E, Villanueva C, Foa P, Rottey S, Skladowski K, Tahara M. Cisplatin and fluorouracil with or without panitumumab in patients with recurrent or metastatic squamous-cell carcinoma of the head and neck (SPECTRUM): an open-label phase 3 randomised trial. Lancet Oncol. 2013;14:697-710.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Nunes DN, Kowalski LP, Simpson AJ. Detection of oral and oropharyngeal cancer by microsatellite analysis in mouth washes and lesion brushings. Oral Oncol. 2000;36:525-528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 37]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
27.  García Martínez J, Pérez-Escuredo J, García-Carracedo D, Alonso-Guervós M, Suárez-Nieto C, Llorente-Pendás JL, Alvarez-Marcos C, Hermsen M. [Analysis of microsatellite instability in laryngeal squamous cell carcinoma]. Acta Otorrinolaringol Esp. 2012;63:79-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
28.  Yalniz Z, Demokan S, Suoglu Y, Ulusan M, Dalay N. Assessment of microsatellite instability in head and neck cancer using consensus markers. Mol Biol Rep. 2010;37:3541-3545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
29.  Rózańska-Kudelska M, Walenczak I, Pepiński W, Sieśkiewicz A, Skawrońska M, Rogowski M. Evaluation of tumor microsatellite instability in laryngeal cancer using five quasimonomorphic mononucleotide repeats and pentaplex PCR. Adv Med Sci. 2008;53:59-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
30.  Lin SC, Chang MF, Chung MY, Chang CS, Kao SY, Liu CJ, Chang KW. Frequent microsatellite alterations of chromosome locus 4q13.1 in oral squamous cell carcinomas. J Oral Pathol Med. 2005;34:209-213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
31.  Lampri ES, Chondrogiannis G, Ioachim E, Varouktsi A, Mitselou A, Galani A, Briassoulis E, Kanavaros P, Galani V. Biomarkers of head and neck cancer, tools or a gordian knot? Int J Clin Exp Med. 2015;8:10340-10357.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Maehara Y, Oda S, Sugimachi K. The instability within: problems in current analyses of microsatellite instability. Mutat Res. 2001;461:249-263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
33.  Sanchez-Cespedes M, Esteller M, Wu L, Nawroz-Danish H, Yoo GH, Koch WM, Jen J, Herman JG, Sidransky D. Gene promoter hypermethylation in tumors and serum of head and neck cancer patients. Cancer Res. 2000;60:892-895.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Carvalho AL, Jeronimo C, Kim MM, Henrique R, Zhang Z, Hoque MO, Chang S, Brait M, Nayak CS, Jiang WW. Evaluation of promoter hypermethylation detection in body fluids as a screening/diagnosis tool for head and neck squamous cell carcinoma. Clin Cancer Res. 2008;14:97-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 396]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
35.  Chang HW, Ling GS, Wei WI, Yuen AP. Smoking and drinking can induce p15 methylation in the upper aerodigestive tract of healthy individuals and patients with head and neck squamous cell carcinoma. Cancer. 2004;101:125-132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 60]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
36.  Belinsky SA, Palmisano WA, Gilliland FD, Crooks LA, Divine KK, Winters SA, Grimes MJ, Harms HJ, Tellez CS, Smith TM. Aberrant promoter methylation in bronchial epithelium and sputum from current and former smokers. Cancer Res. 2002;62:2370-2377.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Rodrigo JP, Ferlito A, Suárez C, Shaha AR, Silver CE, Devaney KO, Bradley PJ, Bocker JM, McLaren KM, Grénman R. New molecular diagnostic methods in head and neck cancer. Head Neck. 2005;27:995-1003.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
38.  Nelson AR, Fingleton B, Rothenberg ML, Matrisian LM. Matrix metalloproteinases: biologic activity and clinical implications. J Clin Oncol. 2000;18:1135-1149.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Ravanti L, Kähäri VM. Matrix metalloproteinases in wound repair (review). Int J Mol Med. 2000;6:391-407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
40.  Cathcart J, Pulkoski-Gross A, Cao J. Targeting Matrix Metalloproteinases in Cancer: Bringing New Life to Old Ideas. Genes Dis. 2015;2:26-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
41.  Marcos CA, Martínez DA, de Los Toyos JR, Domínguez Iglesias F, Hermsen M, Guervós MA, Pendás JL. The usefulness of new serum tumor markers in head and neck squamous cell carcinoma. Otolaryngol Head Neck Surg. 2009;140:375-380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
42.  Linkov F, Lisovich A, Yurkovetsky Z, Marrangoni A, Velikokhatnaya L, Nolen B, Winans M, Bigbee W, Siegfried J, Lokshin A. Early detection of head and neck cancer: development of a novel screening tool using multiplexed immunobead-based biomarker profiling. Cancer Epidemiol Biomarkers Prev. 2007;16:102-107.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Zucker S, Lysik RM, Zarrabi MH, Moll U. M(r) 92,000 type IV collagenase is increased in plasma of patients with colon cancer and breast cancer. Cancer Res. 1993;53:140-146.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Ali-Labib R, Louka ML, Galal IH, Tarek M. Evaluation of matrix metalloproteinase-2 in lung cancer. Proteomics Clin Appl. 2014;8:251-257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
45.  Garzetti GG, Ciavattini A, Lucarini G, Goteri G, Romanini C, Biagini G. Increased serum 72 KDa metalloproteinase in serous ovarian tumors: comparison with CA 125. Anticancer Res. 1996;16:2123-2127.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Kuropkat C, Plehn S, Herz U, Dünne AA, Renz H, Werner JA. Tumor marker potential of serum matrix metalloproteinases in patients with head and neck cancer. Anticancer Res. 2002;22:2221-2227.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Yen CY, Chen CH, Chang CH, Tseng HF, Liu SY, Chuang LY, Wen CH, Chang HW. Matrix metalloproteinases (MMP) 1 and MMP10 but not MMP12 are potential oral cancer markers. Biomarkers. 2009;14:244-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 48]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
48.  Ranuncolo SM, Matos E, Loria D, Vilensky M, Rojo R, Bal de Kier Joffé E, Inés Puricelli L. Circulating 92-kilodalton matrix metalloproteinase (MMP-9) activity is enhanced in the euglobulin plasma fraction of head and neck squamous cell carcinoma. Cancer. 2002;94:1483-1491.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Somiari SB, Shriver CD, Heckman C, Olsen C, Hu H, Jordan R, Arciero C, Russell S, Garguilo G, Hooke J. Plasma concentration and activity of matrix metalloproteinase 2 and 9 in patients with breast disease, breast cancer and at risk of developing breast cancer. Cancer Lett. 2006;233:98-107.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Li Y, St John MA, Zhou X, Kim Y, Sinha U, Jordan RC, Eisele D, Abemayor E, Elashoff D, Park NH. Salivary transcriptome diagnostics for oral cancer detection. Clin Cancer Res. 2004;10:8442-8450.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 395]  [Cited by in F6Publishing: 381]  [Article Influence: 19.1]  [Reference Citation Analysis (0)]
51.  St John MA, Li Y, Zhou X, Denny P, Ho CM, Montemagno C, Shi W, Qi F, Wu B, Sinha U. Interleukin 6 and interleukin 8 as potential biomarkers for oral cavity and oropharyngeal squamous cell carcinoma. Arch Otolaryngol Head Neck Surg. 2004;130:929-935.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
52.  Cohen AN, Veena MS, Srivatsan ES, Wang MB. Suppression of interleukin 6 and 8 production in head and neck cancer cells with curcumin via inhibition of Ikappa beta kinase. Arch Otolaryngol Head Neck Surg. 2009;135:190-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 47]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
53.  Chen Z, Malhotra PS, Thomas GR, Ondrey FG, Duffey DC, Smith CW, Enamorado I, Yeh NT, Kroog GS, Rudy S. Expression of proinflammatory and proangiogenic cytokines in patients with head and neck cancer. Clin Cancer Res. 1999;5:1369-1379.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Zimmermann BG, Park NJ, Wong DT. Genomic targets in saliva. Ann N Y Acad Sci. 2007;1098:184-191.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  SahebJamee M, Eslami M, AtarbashiMoghadam F, Sarafnejad A. Salivary concentration of TNFalpha, IL1 alpha, IL6, and IL8 in oral squamous cell carcinoma. Med Oral Patol Oral Cir Bucal. 2008;13:E292-E295.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Ke XS, Liu CM, Liu DP, Liang CC. MicroRNAs: key participants in gene regulatory networks. Curr Opin Chem Biol. 2003;7:516-523.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Kent OA, Mendell JT. A small piece in the cancer puzzle: microRNAs as tumor suppressors and oncogenes. Oncogene. 2006;25:6188-6196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 514]  [Cited by in F6Publishing: 560]  [Article Influence: 31.1]  [Reference Citation Analysis (0)]
58.  Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA. MicroRNA expression profiles classify human cancers. Nature. 2005;435:834-838.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7124]  [Cited by in F6Publishing: 7179]  [Article Influence: 377.8]  [Reference Citation Analysis (0)]
59.  Jiang J, Lee EJ, Gusev Y, Schmittgen TD. Real-time expression profiling of microRNA precursors in human cancer cell lines. Nucleic Acids Res. 2005;33:5394-5403.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Hui AB, Lenarduzzi M, Krushel T, Waldron L, Pintilie M, Shi W, Perez-Ordonez B, Jurisica I, O’Sullivan B, Waldron J. Comprehensive MicroRNA profiling for head and neck squamous cell carcinomas. Clin Cancer Res. 2010;16:1129-1139.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Park NJ, Zhou H, Elashoff D, Henson BS, Kastratovic DA, Abemayor E, Wong DT. Salivary microRNA: discovery, characterization, and clinical utility for oral cancer detection. Clin Cancer Res. 2009;15:5473-5477.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Tran N, McLean T, Zhang X, Zhao CJ, Thomson JM, O’Brien C, Rose B. MicroRNA expression profiles in head and neck cancer cell lines. Biochem Biophys Res Commun. 2007;358:12-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 183]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
63.  Feber A, Xi L, Luketich JD, Pennathur A, Landreneau RJ, Wu M, Swanson SJ, Godfrey TE, Litle VR. MicroRNA expression profiles of esophageal cancer. J Thorac Cardiovasc Surg. 2008;135:255-60; discussion 260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 282]  [Cited by in F6Publishing: 301]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
64.  Jin Y, Chen D, Cabay RJ, Wang A, Crowe DL, Zhou X. Role of microRNA-138 as a potential tumor suppressor in head and neck squamous cell carcinoma. Int Rev Cell Mol Biol. 2013;303:357-385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
65.  Sun Z, Hu W, Xu J, Kaufmann AM, Albers AE. MicroRNA-34a regulates epithelial-mesenchymal transition and cancer stem cell phenotype of head and neck squamous cell carcinoma in vitro. Int J Oncol. 2015;47:1339-1350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
66.  Yang B, O’Herrin SM, Wu J, Reagan-Shaw S, Ma Y, Bhat KM, Gravekamp C, Setaluri V, Peters N, Hoffmann FM. MAGE-A, mMage-b, and MAGE-C proteins form complexes with KAP1 and suppress p53-dependent apoptosis in MAGE-positive cell lines. Cancer Res. 2007;67:9954-9962.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 213]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
67.  Akers SN, Odunsi K, Karpf AR. Regulation of cancer germline antigen gene expression: implications for cancer immunotherapy. Future Oncol. 2010;6:717-732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 78]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
68.  Cuffel C, Rivals JP, Zaugg Y, Salvi S, Seelentag W, Speiser DE, Liénard D, Monnier P, Romero P, Bron L. Pattern and clinical significance of cancer-testis gene expression in head and neck squamous cell carcinoma. Int J Cancer. 2011;128:2625-2634.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 56]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
69.  Cesson V, Rivals JP, Escher A, Piotet E, Thielemans K, Posevitz V, Dojcinovic D, Monnier P, Speiser D, Bron L. MAGE-A3 and MAGE-A4 specific CD4(+) T cells in head and neck cancer patients: detection of naturally acquired responses and identification of new epitopes. Cancer Immunol Immunother. 2011;60:23-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
70.  Kienstra MA, Neel HB, Strome SE, Roche P. Identification of NY-ESO-1, MAGE-1, and MAGE-3 in head and neck squamous cell carcinoma. Head Neck. 2003;25:457-463.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Yakirevich E, Sabo E, Lavie O, Mazareb S, Spagnoli GC, Resnick MB. Expression of the MAGE-A4 and NY-ESO-1 cancer-testis antigens in serous ovarian neoplasms. Clin Cancer Res. 2003;9:6453-6460.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Kocher T, Zheng M, Bolli M, Simon R, Forster T, Schultz-Thater E, Remmel E, Noppen C, Schmid U, Ackermann D. Prognostic relevance of MAGE-A4 tumor antigen expression in transitional cell carcinoma of the urinary bladder: a tissue microarray study. Int J Cancer. 2002;100:702-705.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 54]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
73.  Jheon S, Hyun DS, Lee SC, Yoon GS, Jeon CH, Park JW, Park CK, Jung MH, Lee KD, Chang HK. Lung cancer detection by a RT-nested PCR using MAGE A1--6 common primers. Lung Cancer. 2004;43:29-37.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Metzler P, Mollaoglu N, Schwarz S, Neukam FW, Nkenke E, Ries J. MAGE-A as a novel approach in the diagnostic accuracy of oral squamous cell cancer: a case report. Head Neck Oncol. 2009;1:39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
75.  Lee KD, Lee HH, Joo HB, Lee HS, Yu TH, Chang HK, Jeon CH, Park JW. Expression of MAGE A 1-6 mRNA in sputa of head and neck cancer patients--a preliminary report. Anticancer Res. 2006;26:1513-1518.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Jungbluth AA, Busam KJ, Kolb D, Iversen K, Coplan K, Chen YT, Spagnoli GC, Old LJ. Expression of MAGE-antigens in normal tissues and cancer. Int J Cancer. 2000;85:460-465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
77.  Ries J, Vairaktaris E, Mollaoglu N, Wiltfang J, Neukam FW, Nkenke E. Expression of melanoma-associated antigens in oral squamous cell carcinoma. J Oral Pathol Med. 2008;37:88-93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
78.  Song DW, Shin SJ, Kim DE, Jung SG, Park JW, Lee KD. Detection of MAGE and SSX gene expressions by RT-nested PCR using common primers in head and neck cancer. Clin Exp Otorhinolaryngol. 2008;1:97-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
79.  Ries J, Schultze-Mosgau S, Neukam F, Diebel E, Wiltfang J. Investigation of the expression of melanoma antigen-encoding genes (MAGE-A1 to -A6) in oral squamous cell carcinomas to determine potential targets for gene-based cancer immunotherapy. Int J Oncol. 2005;26:817-824.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Lee KD, Lee HS, Kim SW, Park T, Hong JC, Chang HK, Jung SB, Jeon CH, Park JW. Clinical significance of melanoma-associated antigen A1-6 expression in sputum of patients with squamous cell carcinoma of the larynx and hypopharynx. Head Neck. 2015;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
81.  Pastorcic-Grgic M, Sarcevic B, Dosen D, Juretic A, Spagnoli GC, Grgic M. Prognostic value of MAGE-A and NY-ESO-1 expression in pharyngeal cancer. Head Neck. 2010;32:1178-1184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
82.  Gustafson LM, Gleich LL, Fukasawa K, Chadwell J, Miller MA, Stambrook PJ, Gluckman JL. Centrosome hyperamplification in head and neck squamous cell carcinoma: a potential phenotypic marker of tumor aggressiveness. Laryngoscope. 2000;110:1798-1801.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Tarapore P, Fukasawa K. Loss of p53 and centrosome hyperamplification. Oncogene. 2002;21:6234-6240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 149]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
84.  Thirthagiri E, Robinson CM, Huntley S, Davies M, Yap LF, Prime SS, Paterson IC. Spindle assembly checkpoint and centrosome abnormalities in oral cancer. Cancer Lett. 2007;258:276-285.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Friedl P, Gilmour D. Collective cell migration in morphogenesis, regeneration and cancer. Nat Rev Mol Cell Biol. 2009;10:445-457.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1783]  [Cited by in F6Publishing: 1832]  [Article Influence: 122.1]  [Reference Citation Analysis (0)]
86.  de Jong EP, Xie H, Onsongo G, Stone MD, Chen XB, Kooren JA, Refsland EW, Griffin RJ, Ondrey FG, Wu B. Quantitative proteomics reveals myosin and actin as promising saliva biomarkers for distinguishing pre-malignant and malignant oral lesions. PLoS One. 2010;5:e11148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 82]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
87.  Uzquiano MC, Prieto VG, Nash JW, Ivan DS, Gong Y, Lazar AJ, Diwan AH. Metastatic basal cell carcinoma exhibits reduced actin expression. Mod Pathol. 2008;21:540-543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 32]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
88.  Li Q, Huang W, Zhou X. Expression of CD34, alpha-smooth muscle actin and transforming growth factor-beta1 in squamous intraepithelial lesions and squamous cell carcinoma of the cervix. J Int Med Res. 2009;37:446-454.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Qi Y, Chiu JF, Wang L, Kwong DL, He QY. Comparative proteomic analysis of esophageal squamous cell carcinoma. Proteomics. 2005;5:2960-2971.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 117]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
90.  Shi L, Sun SZ, Wang ZG. [Expression of stromal CD34 and alpha-smooth muscle actin in oral invasive cancer]. Zhonghua Kou Qiang Yi Xue Za Zhi. 2006;41:106-107.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Lo WY, Tsai MH, Tsai Y, Hua CH, Tsai FJ, Huang SY, Tsai CH, Lai CC. Identification of over-expressed proteins in oral squamous cell carcinoma (OSCC) patients by clinical proteomic analysis. Clin Chim Acta. 2007;376:101-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 105]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
92.  Turhani D, Krapfenbauer K, Thurnher D, Langen H, Fountoulakis M. Identification of differentially expressed, tumor-associated proteins in oral squamous cell carcinoma by proteomic analysis. Electrophoresis. 2006;27:1417-1423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 106]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
93.  Barak V, Goike H, Panaretakis KW, Einarsson R. Clinical utility of cytokeratins as tumor markers. Clin Biochem. 2004;37:529-540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 226]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
94.  Moll R. Cytokeratins as markers of differentiation in the diagnosis of epithelial tumors. Subcell Biochem. 1998;31:205-262.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Chu P, Wu E, Weiss LM. Cytokeratin 7 and cytokeratin 20 expression in epithelial neoplasms: a survey of 435 cases. Mod Pathol. 2000;13:962-972.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Toyoshima T, Vairaktaris E, Nkenke E, Schlegel KA, Neukam FW, Ries J. Cytokeratin 17 mRNA expression has potential for diagnostic marker of oral squamous cell carcinoma. J Cancer Res Clin Oncol. 2008;134:515-521.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Domachowske JB, Bonville CA, Rosenberg HF. Cytokeratin 17 is expressed in cells infected with respiratory syncytial virus via NF-kappaB activation and is associated with the formation of cytopathic syncytia. J Infect Dis. 2000;182:1022-1028.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
98.  Cohen-Kerem R, Madah W, Sabo E, Rahat MA, Greenberg E, Elmalah I. Cytokeratin-17 as a potential marker for squamous cell carcinoma of the larynx. Ann Otol Rhinol Laryngol. 2004;113:821-827.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Toyoshima T, Koch F, Kaemmerer P, Vairaktaris E, Al-Nawas B, Wagner W. Expression of cytokeratin 17 mRNA in oral squamous cell carcinoma cells obtained by brush biopsy: preliminary results. J Oral Pathol Med. 2009;38:530-534.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
100.  Wei KJ, Zhang L, Yang X, Zhong LP, Zhou XJ, Pan HY, Li J, Chen WT, Zhang ZY. Overexpression of cytokeratin 17 protein in oral squamous cell carcinoma in vitro and in vivo. Oral Dis. 2009;15:111-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
101.  Sesterhenn AM, Mandic R, Dünne AA, Werner JA. Cytokeratins 6 and 16 are frequently expressed in head and neck squamous cell carcinoma cell lines and fresh biopsies. Anticancer Res. 2005;25:2675-2680.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Jalali MM, Heidarzadeh A, Zavarei MJ, Sarmast H. p53 overexpression impacts on the prognosis of laryngeal squamous cell carcinomas. Asian Pac J Cancer Prev. 2011;12:1731-1734.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  van Oijen MG, Slootweg PJ. Gain-of-function mutations in the tumor suppressor gene p53. Clin Cancer Res. 2000;6:2138-2145.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Hardisson D. Molecular pathogenesis of head and neck squamous cell carcinoma. Eur Arch Otorhinolaryngol. 2003;260:502-508.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Bradford CR, Kumar B, Bellile E, Lee J, Taylor J, D’Silva N, Cordell K, Kleer C, Kupfer R, Kumar P. Biomarkers in advanced larynx cancer. Laryngoscope. 2014;124:179-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 35]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
106.  Nathan CO, Sanders K, Abreo FW, Nassar R, Glass J. Correlation of p53 and the proto-oncogene eIF4E in larynx cancers: prognostic implications. Cancer Res. 2000;60:3599-3604.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Poeta ML, Manola J, Goldwasser MA, Forastiere A, Benoit N, Califano JA, Ridge JA, Goodwin J, Kenady D, Saunders J. TP53 mutations and survival in squamous-cell carcinoma of the head and neck. N Engl J Med. 2007;357:2552-2561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 568]  [Cited by in F6Publishing: 541]  [Article Influence: 31.8]  [Reference Citation Analysis (0)]
108.  van Houten VM, Tabor MP, van den Brekel MW, Kummer JA, Denkers F, Dijkstra J, Leemans R, van der Waal I, Snow GB, Brakenhoff RH. Mutated p53 as a molecular marker for the diagnosis of head and neck cancer. J Pathol. 2002;198:476-486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 109]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
109.  Gasco M, Crook T. The p53 network in head and neck cancer. Oral Oncol. 2003;39:222-231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 108]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
110.  Song HS, Do YR, Kang SH, Jeong KY, Kim YS. Prognostic significance of immunohistochemical expression of p53 gene product in operable breast cancer. Cancer Res Treat. 2006;38:218-223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 10]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
111.  Mitra S, Banerjee S, Misra C, Singh RK, Roy A, Sengupta A, Panda CK, Roychoudhury S. Interplay between human papilloma virus infection and p53 gene alterations in head and neck squamous cell carcinoma of an Indian patient population. J Clin Pathol. 2007;60:1040-1047.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 32]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
112.  Chen CN, Hsieh FJ, Cheng YM, Lee PH, Chang KJ. Expression of eukaryotic initiation factor 4E in gastric adenocarcinoma and its association with clinical outcome. J Surg Oncol. 2004;86:22-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
113.  De Benedetti A, Graff JR. eIF-4E expression and its role in malignancies and metastases. Oncogene. 2004;23:3189-3199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 570]  [Cited by in F6Publishing: 595]  [Article Influence: 29.8]  [Reference Citation Analysis (0)]
114.  Avdulov S, Li S, Michalek V, Burrichter D, Peterson M, Perlman DM, Manivel JC, Sonenberg N, Yee D, Bitterman PB. Activation of translation complex eIF4F is essential for the genesis and maintenance of the malignant phenotype in human mammary epithelial cells. Cancer Cell. 2004;5:553-563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
115.  Ruggero D, Montanaro L, Ma L, Xu W, Londei P, Cordon-Cardo C, Pandolfi PP. The translation factor eIF-4E promotes tumor formation and cooperates with c-Myc in lymphomagenesis. Nat Med. 2004;10:484-486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 454]  [Cited by in F6Publishing: 474]  [Article Influence: 23.7]  [Reference Citation Analysis (0)]
116.  Cardesa A, Nadal A. Carcinoma of the head and neck in the HPV era. Acta Dermatovenerol Alp Pannonica Adriat. 2011;20:161-173.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Nathan CO, Liu L, Li BD, Abreo FW, Nandy I, De Benedetti A. Detection of the proto-oncogene eIF4E in surgical margins may predict recurrence in head and neck cancer. Oncogene. 1997;15:579-584.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 148]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
118.  Crew JP, Fuggle S, Bicknell R, Cranston DW, de Benedetti A, Harris AL. Eukaryotic initiation factor-4E in superficial and muscle invasive bladder cancer and its correlation with vascular endothelial growth factor expression and tumour progression. Br J Cancer. 2000;82:161-166.  [PubMed]  [DOI]  [Cited in This Article: ]
119.  Mamane Y, Petroulakis E, Rong L, Yoshida K, Ler LW, Sonenberg N. eIF4E--from translation to transformation. Oncogene. 2004;23:3172-3179.  [PubMed]  [DOI]  [Cited in This Article: ]
120.  Wang R, Geng J, Wang JH, Chu XY, Geng HC, Chen LB. Overexpression of eukaryotic initiation factor 4E (eIF4E) and its clinical significance in lung adenocarcinoma. Lung Cancer. 2009;66:237-244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 32]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
121.  Holm N, Byrnes K, Johnson L, Abreo F, Sehon K, Alley J, Meschonat C, Md QC, Li BD. A prospective trial on initiation factor 4E (eIF4E) overexpression and cancer recurrence in node-negative breast cancer. Ann Surg Oncol. 2008;15:3207-3215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
122.  Chakraborty S, Mohiyuddin SM, Gopinath KS, Kumar A. Involvement of TSC genes and differential expression of other members of the mTOR signaling pathway in oral squamous cell carcinoma. BMC Cancer. 2008;8:163.  [PubMed]  [DOI]  [Cited in This Article: ]
123.  Lahtz C, Pfeifer GP. Epigenetic changes of DNA repair genes in cancer. J Mol Cell Biol. 2011;3:51-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 129]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
124.  Riaz N, Morris LG, Lee W, Chan TA. Unraveling the molecular genetics of head and neck cancer through genome-wide approaches. Genes Dis. 2014;1:75-86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 60]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
125.  Jin B, Robertson KD. DNA methyltransferases, DNA damage repair, and cancer. Adv Exp Med Biol. 2013;754:3-29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 307]  [Article Influence: 27.9]  [Reference Citation Analysis (0)]
126.  Paidpally V, Chirindel A, Lam S, Agrawal N, Quon H, Subramaniam RM. FDG-PET/CT imaging biomarkers in head and neck squamous cell carcinoma. Imaging Med. 2012;4:633-647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 89]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
127.  Differding S, Hanin FX, Grégoire V. PET imaging biomarkers in head and neck cancer. Eur J Nucl Med Mol Imaging. 2015;42:613-622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
128.  Lee SJ, Seo HJ, Cheon GJ, Kim JH, Kim EE, Kang KW, Paeng JC, Chung JK, Lee DS. Usefulness of Integrated PET/MRI in Head and Neck Cancer: A Preliminary Study. Nucl Med Mol Imaging. 2014;48:98-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]