Next Article in Journal
Interaction of Arsenic Exposure and Transcriptomic Profile in Basal Cell Carcinoma
Next Article in Special Issue
Neutrophil to Lymphocyte Ratio in Oropharyngeal Squamous Cell Carcinoma: A Systematic Review and Meta-Analysis
Previous Article in Journal
The NAMPT Inhibitor FK866 Increases Metformin Sensitivity in Pancreatic Cancer Cells
Previous Article in Special Issue
Insight into Classification and Risk Stratification of Head and Neck Squamous Cell Carcinoma in Era of Emerging Biomarkers with Focus on Histopathologic Parameters
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Biomarkers of Malignant Transformation in Head and Neck Dysplasia

by
Kushi Ranganath
1,
Allen L. Feng
2,
Ramon A. Franco
2,
Mark A. Varvares
2,
William C. Faquin
2,
Matthew R. Naunheim
2,* and
Srinivas Vinod Saladi
2,3,*
1
Department of Otolaryngology-Head & Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
2
Department of Otolaryngology-Head & Neck Surgery, Massachusetts Eye & Ear, Boston, MA 02114, USA
3
Broad Institute of MIT and Harvard, Boston, MA 02114, USA
*
Authors to whom correspondence should be addressed.
Cancers 2022, 14(22), 5581; https://doi.org/10.3390/cancers14225581
Submission received: 5 October 2022 / Revised: 1 November 2022 / Accepted: 7 November 2022 / Published: 15 November 2022
(This article belongs to the Special Issue The Biomarkers and Detection of Head and Neck Cancer)

Abstract

:

Simple Summary

Head and neck cancer patients face significant morbidity and mortality. Early detection and diagnosis of disease followed by timely intervention is necessary for improving clinical management for these patients. There remains a need to be able to identify whether an early pre-cancerous lesion at the dysplasia stage will progress to invasive cancer. Biomarkers are biological molecules found in blood or tissue that are measurable at early stages of disease and can be applied to predict the progression of such lesions. The aim of this review is to comprehensively present the available evidence on the most frequently altered tumor molecular biomarkers present in head and neck dysplasia as well as their potential clinical applications.

Abstract

Head and neck squamous cell carcinoma (HNSCC) and its treatments are associated with substantial morbidity, often resulting in cosmetic deformity and loss of physiologic functions including speech and swallowing. Despite advancements in treatment, 5-year survival rates for mucosal malignancies remain below 70%. Effective prevention of HNSCC demands an understanding of the molecular pathways of carcinogenesis. Specifically, defining features of pre-cancerous dysplastic lesions that indicate a better or worse prognosis is necessary to help identify patients who are likely to develop a carcinoma and allow a more aggressive approach to management. There remains a need for identification of biomarkers that can provide both early prognostic and predictive value in clinical decision-making by serving as both therapeutic targets as well as predictors of therapy response. Here, we comprehensively review the most frequently altered molecular biomarkers of malignant transformation in head and neck dysplasia. These markers are involved in a wide range of cellular processes in head and neck carcinogenesis, including extracellular matrix degradation, cell motility and invasion, cell–cell adhesion, solute transport, immortalization, metabolism, the cell cycle and apoptosis, transcription, and cell signaling.

1. Introduction

Head and neck squamous cell carcinoma (HNSCC) is the fifth most common cancer, accounting for approximately 5% of all malignancies worldwide [1,2]. Head and neck cancer encompasses cutaneous malignancies of the face and scalp as well as tumors arising from mucosal surfaces of the upper aerodigestive tract, namely the oral cavity, nasopharynx, oropharynx, hypopharynx, and larynx [3]. When diagnosis is delayed, which is the case in over half of HNSCC patients, HNSCC carries a poor prognosis and is associated with substantial mortality [4]. Despite advancements in treatment, 5-year survival rates for mucosal malignancies remain below 70% [5]. HNSCC and its treatments are also associated with substantial morbidity, often resulting in cosmetic deformity and loss of physiologic functions including speech and swallowing [3].
Effective prevention of HNSCC demands an understanding of the molecular pathways of carcinogenesis. The most important risk factors for HNSCC include tobacco and alcohol consumption and Human Papillomavirus (HPV) infection [6]. Prolonged exposure to these carcinogens drives genetic and epigenetic alterations that result in progression from dysplasia to carcinoma in situ to invasive carcinoma [7]. The current clinical practice for assessing at-risk lesions within the head and neck is histopathologic examination for dysplasia and carcinoma [8]. The current grading of dysplasia has several limitations, notably that the stepwise nature of dysplasia classification does not reflect the more continuous evolution of this disease process [9,10]. Furthermore, dysplasia refers to a heterogeneous group of cytologic and architectural changes, with not all cases progressing to invasive carcinoma or following the classic ordered histologic progression model [11,12]. At this time, histologic characteristics cannot reliably determine whether a dysplastic lesion will progress. Defining features of dysplastic lesions that indicate a better or worse prognosis is necessary to refine clinical management [12]. Such prognostic biomarkers may help identify patients who are likely to develop a carcinoma and allow a more aggressive approach to management.
There remains a need for the identification and validation of novel biomarkers that can predict the progression of pre-malignant lesions [13]. However, biological markers in HNSCC are not as well-studied as they are in other tumor types, such as breast or pancreatic cancer. To that end, we summarize the genetic progression model of HNSCC followed by a comprehensive review of molecular biomarkers of malignant transformation in head and neck dysplasia with an emphasis on oral cavity and laryngeal dysplasia (Figure 1).

2. Genetic Mechanisms Underlying Progression of Dysplasia to Carcinoma

In the classic histologic progression model of HNSCC, lesions proceed from normal mucosa to epithelial hyperplasia to low- or high-grade dysplasia to carcinoma in situ (CIS) to invasive carcinoma. Of note, not all dysplasia progress in a histologically recognizable stepwise fashion through this model. The risk of developing invasive carcinoma increases with progression of histologic grade.
A subset of the genetic events that drive the progression from one stage to the next is outlined here. Accumulation of loss of heterozygosity (LOH) at multiple genetic loci leads to histopathologic progression [14]. LOH at 9p21 occurs early as normal mucosa becomes proliferative and develops the histologic appearance of hyperplasia. This results in inactivation of the associated tumor suppressor genes cyclin-dependent kinase inhibitor 2A (CDKN2A), which encodes the CDK inhibitor p16INK4A, and ARF, which encodes the p53 stabilizer p14. LOH at the 3p and 17p chromosome regions occurs at the subsequent transition from hyperplasia to dysplasia. The most common loci that are lost include 3p14, 3p21, and 17p13, and their associated tumor suppressor genes fragile histidine triad gene (FIHT), Ras association domain family 1 isoform A (RASSF1A), and TP53, respectively. Amplification of 11q13 and resultant overexpression of cyclin D1 as well as LOH of 13q21 and resultant inactivation of the retinoblastoma gene occurs at the transition from dysplasia to CIS. LOH at 6p, 8, 4q27, and 10q23 leads to the inactivation of tumor suppressors including phosphatase and tensin homolog (PTEN) and marks the transition from CIS to invasive carcinoma [14,15,16,17,18] (Figure 2). Notably, this genetic progression occurs within the context of a field effect, where tissue adjacent to lesions share genetic alterations and undergo histopathologic changes that may result in synchronous or metachronous tumors [14].

3. Extracellular and Cell Surface Biomarkers

3.1. Matrix Metalloproteinases

Matrix metalloproteinases (MMP) are extracellular proteinases which degrade extracellular matrix and basement membrane. They play roles in regulating tissue homeostasis, wound healing, immunity, inflammation, and angiogenesis. Alterations in MMP activity are implicated in chronic inflammatory diseases and cancer [19]. MMP-2 and MMP-9 are type IV collagenases [20] that dissolve collagen IV, the main structural component of basement membrane [21]. Breach of this barrier between epithelium and underlying mesenchyme is a key event in the progression from dysplasia to invasive carcinoma [22]. Studies have reported overexpression of MMP-2 and MMP-9 in oral epithelial dysplasia (OED) [23], with MMP-9 expression correlating significantly with OED grade in patient samples [24,25]. Levels of MMP-1 and MMP-9 mRNA are specifically elevated in OED cases that progress to cancer compared to those which do not [26]. MMPs also regulate the early tumor microenvironment, with findings of high MMP expression in both tumor and stromal cells which function as “initiators” and “promoters” of carcinogenesis, respectively [27]. Stromal cells include fibroblasts, macrophages, and vascular endothelial cells that secrete MMPs into the extracellular space [28]. The extracellular MMP inducer (EMMPRIN) stimulates MMP synthesis in stromal fibroblasts at early stages of tumorigenesis, and EMMPRIN expression also correlates with the grade of OED patient samples [29]. MMP-9 expression and tumor vascularity progressively increase from normal mucosa to OED, indicating an additional pro-angiogenic role of MMPs in early tumor formation [24]. Ultimately, MMP overexpression mediates early tumor invasion, epithelial to mesenchymal transition (EMT), and angiogenesis.

3.2. Podoplanin

Podoplanin (PDPN) is a mucin-like transmembrane glycoprotein that plays roles in the development of the lungs, lymphatic system, and heart as well as the biology of immune cells and platelet activation [30]. PDPN overexpression has been reported in various human squamous cell carcinomas, particularly oral cavity, larynx, skin, cervical, and lung cancers [31]. PDPN is expressed in 90% of oral squamous cell carcinomas (OSCC) [32] and 37% of oral leukoplakias (OL) [33]. PDPN expression has been found to correlate with the degree of OED in patient samples and predicts a higher rate of progression to oral cancer [33,34,35,36]. In tumor cells, PDPN enables migration and invasion by modulating the actin cytoskeleton. PDPN induces tumor invasion by mediating single-cell migration following EMT or collective cell migration in the absence of EMT [37]. It is present at the invasive peripheral edge of approximately 80% of human squamous cell carcinomas [38,39]. Acquisition of PDPN expression at the tumor front may therefore drive invasion into surrounding tissue during tumor initiation.

3.3. Claudin, JAM-A, and E-Cadherin

Cell–cell adhesion is critical to maintain a barrier against tumor invasion. Epithelial tight junctions are comprised of claudins (CLDN-1), occludins, and junctional adhesion molecules (JAM-A) [40]. Overexpression of CLDN-1 and JAM-A has been reported in OED and OSCC patient samples and correlates with histologic grade. The mechanism by which upregulation of these proteins leads to tumorigenesis is not fully elucidated. However, findings of CLDN-1 and JAM-A delocalization from the cell membrane to cytoplasm during malignant transformation suggests their possible role in cell signaling [40]. Studies in other tumor types posit that CLDN-1 promotes MMP activity leading to cell migration, and JAM-A induces EMT [40,41,42]. Adherens junctions are another key component of epithelial tissue architecture and are comprised of E-cadherins, calcium-dependent adhesion proteins [43]. In addition to stabilizing cell–cell interactions [44], E-cadherin mediates signaling pathways involved in cellular proliferation, differentiation, and apoptosis [43,45]. E-cadherin expression has been found to decrease with increasing grades of dysplasia in OL patient samples [43,46,47]. Loss of the “invasion suppressor” E-cadherin is therefore a critical step in EMT and tumor invasion in oral carcinogenesis [43,48].

3.4. CD44 and CD133

CD44 is a cell surface transmembrane glycoprotein involved in cell migration, adhesion, leukocyte and lymphocyte activation, myelopoiesis and lymphopoiesis, and angiogenesis [49]. Alternative splicing of the CD44 gene results in standard (CD44s) and variant (e.g., CD44v3, CD44v6) isoforms which interact with ligands including hyaluronate, osteopontin, collagens, and metalloproteinases [49,50]. CD44s is expressed in most human tissues, whereas variant isoforms are more limited in distribution and often expressed in response to oncogenic signals [49,51]. CD44 is a well-known cancer stem cell (CSC) marker [51]. While some studies report CD44s overexpression in OED [52] and dysplastic oral lichen planus (OLP) [53], others found reduced CD44v6 expression in OED patient samples [54]. Decreased CD44 expression was also observed in non-tumor epithelium adjacent to oral and laryngeal cancers, suggesting an early role for CD44 in carcinogenesis [55,56,57]. Taken together, there is no consistent pattern of CD44 expression in HNSCC, with observations of both CD44 gain and loss [49,58]. Nonetheless, dysregulation of CD44 expression alters the early proliferative status and adhesive properties of tumor cells [49].
CD133, also known as Prominin-1, is a cell surface transmembrane glycoprotein and the most frequently used CSC marker [59]. The physiologic function of CD133 is unclear, although it may be involved in membrane organization, autophagy, signal transduction (IL-8, mTOR, PI3K, MAPK), cellular scaffolding, and glucose metabolism [60]. In CSCs, it plays critical roles in self-renewal, cell growth, differentiation, and metabolism [59,60]. Increased CD133 expression has been reported in OED and OSCC relative to normal mucosa in patient samples [61,62]. CD133 overexpression is also significantly associated with malignant progression to carcinoma in 80% of OLP cases [63] and 59% of OL cases [64]. Furthermore, CD133 delocalization from the cell membrane to the nucleus or cytoplasm also correlates with malignant transformation [65].

3.5. Glucose Transporters

Glucose transporters (GLUT) facilitate glucose and fructose transport across the cell membrane, thereby regulating cellular energy metabolism [66,67]. Rapidly proliferating cancer cells have a high ATP and glucose requirement, resulting in the upregulation of GLUT and glycolytic enzymes to meet this demand [66]. Glycolysis is especially advantageous to promote tumor survival under hypoxic conditions and when energy supply is limited, which is often the case in carcinogenesis [67,68]. In addition to functioning as an insulin-dependent glucose transporter, GLUT-4 is an activator of glucose-independent signaling pathways that promote migration and invasion [69]. Studies have reported overexpression of GLUT-1 and GLUT-4 in OED relative to OSCC [68,70], and increases in GLUT-1 expression also correlate with degree of dysplasia in patient samples [67]. Increased expression in dysplastic over carcinomatous lesions suggests GLUT upregulation occurs early in oral tumorigenesis [68,71].

4. Cytosolic Biomarkers

4.1. Aldehyde Dehydrogenases

Metabolic dysregulation is a consistent feature of tumor cells [72]. Aldehyde dehydrogenase (ALDH) is a cytosolic enzyme involved in the oxidation of aldehydes to carboxylic acids, essential for reduction of oxidative stress [73]. Certain isoforms of ALDH (e.g., ALDH1A1, ALDH1A2, ALDH1A3, ALDH8A1) oxidize retinol to retinoic acid (RA), resulting in RA cell signaling that induces the “stemness” of CSCs [74]. ALDH1 is a well-known CSC marker in HNSCC [75]. Its expression correlates with the number of cells undergoing EMT and therefore reflects the invasive potential of a tumor [76]. Levels of ALDH1 expression have been found to increase significantly from dysplasia to OSCC in association with histologic grade in patient samples [73,76]. ALDH1 expression is associated with a 4.17-fold increase in the risk of malignant transformation of OL, with 48.1% of ALDH1-positive OL clinical cases progressing to invasive carcinoma [64].

4.2. Molecular Chaperones

Heat shock proteins (HSP) are molecular chaperones that regulate protein folding, refolding of misfolded proteins, protein repair and degradation, and intracellular protein transport [77,78]. Cellular stressors such as low nutrient supply, oxidative stress, hypoxia, injury, and apoptotic signals can induce HSP synthesis [77]. Of the five major families of HSPs, HSP70, and HSP90 are the most frequently identified to play a role in OSCC. HSP70 and HSP90 expression have been found to increase with increasing grades of OED and OSCC in patient samples [79,80,81].

4.3. Mitosis and Apoptosis Regulators

The microtubular cytoskeleton is critical to the integrity of mitosis. Stathmin is a cytosolic phosphoprotein that is involved in the regulation of the microtubule cytoskeleton. Depending on its phosphorylation state, stathmin regulates both entry into and exit from mitosis via modulation of mitotic spindle assembly [82]. Increased stathmin expression has been reported in various tumor types [83,84] including nasopharyngeal carcinoma and OSCC [85,86]. Stathmin expression increases with increasing grades of OED in patient samples, suggesting an early role in tumorigenesis [87,88].
Apoptosis is essential to the physiologic turnover of human tissue [89]. Regulators of apoptosis can be divided into the Bcl-2 family and inhibitors of apoptosis (IAP) family [90]. Bcl-2 anti-apoptotic proteins inhibit mitochondrial outer membrane permeabilization, thereby limiting the release of cytochrome c and resultant caspase activation [91,92]. Increased Bcl-2 expression is an early event in carcinogenesis and is observed in 30–60% of OED [89,93]. Detection of Bcl-2 has also been used to discriminate OL from OLP with high specificity in clinical samples, suggesting its potential role as a marker of malignant transformation [94]. IAPs regulate apoptosis downstream of Bcl-2 by functioning as endogenous inhibitors of caspases [92]. Survivin is one member of the IAP family that is expressed in 40–70% of OL patient cases [95,96]. Survivin positivity has also been reported in 94% of dysplasia patient cases that transformed to OSCC [97].

5. Nuclear Biomarkers

5.1. Cell Cycle Regulators

The p53 tumor suppressor is encoded by the TP53 gene on chromosome 17 [98]. Mutations in p53 are detectable in 50% of human cancers [99]. The p53 family of transcription factors induces the expression of genes needed for cell cycle arrest, DNA repair, apoptosis, cellular senescence, and metabolism. This classically occurs in response to DNA damage, oxidative stress, and hypoxia [100]. P53 acts at the G1/S cell cycle checkpoint where it activates the p21-Rb-E2F pathway. In G1, hyperphosphorylation of retinoblastoma protein (Rb) by cyclin-dependent kinases (CDK) releases E2F transcription factors from the Rb-E2F repressor complex and drives cells into S phase [101]. At this point, cell cycle arrest by p53 is primarily mediated by activation of the CDK inhibitors p21, p15, and p16 [92,98,102]. P53 target genes such as 14-3-3σ also mediate arrest at the subsequent G2/M transition [103]. MDM2 is a negative regulator of p53 that functions as a E3 ubiquitin ligase that marks p53 for proteasomal degradation [104].
Levels of p53 expression increase in association with histologic grade of OED [105]. In fact, p53 expression is present in 80% of oral epithelial hyperplasia [106] and increases significantly from OL to OSCC in patient samples [81]. P53 expression is also associated with progression to malignancy in laryngeal dysplasia [107]. Expression of the p53 homologs, p63 and p73, is increased in OED relative to normal mucosa [108,109,110]. Overexpression of other cell cycle regulators including cyclin D1, Rb, and MDM2 have been reported in OL relative to normal mucosa [111]. Of the G1/S regulators, loss of p16 is the earliest event, and subsequent loss of pRb, gain of p53, and gain of cyclin D1 are associated with malignant progression [112,113] (Figure 3).

5.2. Transcriptional Regulators

C-Jun is a transcriptional activator of the activator protein 1 (AP-1) transcription factor family [114] that mediates proliferation, apoptosis, DNA repair, and differentiation [114,115]. C-Jun expression levels have been found to correlate with the severity of oral dysplasia [116,117]. Observations of increased C-Jun expression in dysplasia cases which progressed to carcinoma suggest its early role in carcinogenesis [118]. SOX-2 and OCT-4 are transcription factors that induce the self-renewal and pluripotency properties of embryonic stem cells [119]. Levels of SOX-2 and OCT-4 expression also increase with progression from dysplasia to OSCC in patient samples [34,120]. SOX-2 and OCT-4 overexpression may be necessary to prevent apoptosis [121,122] and regulate EMT in tumor cells, respectively [122] (Figure 4).

5.3. DNA Replication and Repair Regulators

Dysregulation of DNA replication and repair processes contributes to the genomic instability of tumor cells. Upregulation of telomerase, an RNA-dependent DNA polymerase that maintains telomere length, has been observed in many tumor types. The catalytic protein component of telomerase is known as human telomerase reverse transcriptase (hTERT). Findings of increased hTERT expression with progression from normal mucosa to OSCC suggests it may promote immortalization of tumor cells [123]. Minichromosome maintenance complex component 2 (MCM2) is a member of the MCM family of proteins which function as replication initiation factors and promote DNA elongation [124]. MCM2 expression is also known to increase progressively in association with the severity of OED in clinical samples [125].
DNA mismatch repair genes, notably MutSα and MutLα, play a similarly important role in preserving genomic integrity and are reduced in oral dysplasia and carcinoma compared to normal mucosa [126]. Ataxia telangiectasia mutated (ATM) and RAD-3 related (ATR) proteins and their downstream checkpoint kinases Chk-2 and Chk-1, respectively, play a similarly critical role in regulating the response to DNA damage. ATM-Chk2 and ATR-Chk1 signaling pathways activate cell cycle checkpoint pathways which induce cell cycle arrest and DNA repair [127]. ATM expression has been found to increase with progression from OL to OSCC and is independently associated with a significantly increased risk of malignant transformation. However, levels of Chk-2 expression do not correlate with the degree of OED, suggesting that it may be regulated differently [128]. Significantly reduced levels of expression of phosphorylated ATR and Chk-1 have been observed in patients with OED progressing to OSCC [129]. Taken together, there are limited studies suggesting an inconsistent pattern of both upregulation and downregulation of DNA repair molecules. Nonetheless, it appears that dysregulation of DNA mismatch repair processes occurs early in oral carcinogenesis.

5.4. Hippo Pathway

The transcriptional co-regulators Yes-associated protein (YAP) and WW domain-containing transcription regulator protein 1 (WWTR1), hereafter referred to as TAZ, are major effectors of the Hippo pathway that interact with the transcriptional enhanced associate domain (TEAD) family of transcription factors. When the Hippo pathway is off, YAP/TAZ are unphosphorylated and bind TEAD to induce transcriptional programs for cellular proliferation, survival, and migration. When the Hippo pathway is on, inhibitory phosphorylation of YAP/TAZ results in their retention and degradation in the cytoplasm [130] (Figure 5). Somatic alterations of the Hippo pathway have been reported in nearly 50% of HNSCC. FAT1 is an upstream YAP1 inhibitor that is mutated in nearly 30% of HNSCC, and FAT1 deletion has been proposed to contribute to a hybrid EMT state, tumor stemness, and metastasis [131]. We have also recently shown that YAP1 maintains an active chromatin state in HNSCC and promotes tumorigenesis through cooperation with BRD4 [132]. In HPV-related HNSCC, HPV oncoproteins E6 and E7 also activate YAP nuclear localization, promote YAP, TAZ, and TEAD as well as their transcriptional targets, and inhibit upstream YAP1 inhibitors (e.g., PTPN14, MST 1/2) [133,134]. YAP has been found to be elevated in correlation with histologic grade of OED in clinical samples and has been recognized as a potent driver of tumorigenesis in a mouse model [109,135].

6. Conclusions and Clinical Applications

In the dysplasia-to-carcinoma sequence of HNSCC, the presence of dysplasia significantly increases the risk cancer. However, not all dysplastic lesions are the same. Histologic grade cannot reflect all precancerous changes present in a lesion and therefore is not a robust predictor of malignant transformation [136]. For this reason, identification of molecular biomarkers that can discriminate lesions at increased risk of progression is paramount. Most studies have focused on identifying biomarkers in cancer, with comparatively fewer studies of biomarkers altered at earlier stages. We have provided a comprehensive review of the most frequently altered tumor markers present in head and neck dysplasia. These markers are involved in a wide range of cellular processes in head and neck carcinogenesis, including extracellular matrix degradation, cell motility and invasion, cell-cell adhesion, solute transport, immortalization, metabolism, the cell cycle and apoptosis, transcription, and signaling pathways [137] (Table 1).
The findings of this review suggest that molecules involved in early head and neck tumorigenesis are detectable and measurable, and they may have applications as diagnostic biomarkers for the early detection of cancer. There is not yet enough evidence to support an ordered model of molecular progression in head and neck cancer. Future studies performing molecular phenotyping of cancer cells at each stage of progression are necessary. Molecular biomarkers of high-risk precancerous lesions that correlate with transformation to SCC can provide prognostic value in clinical decision-making. The use of these molecular markers in conjunction with other early clinical and histologic indicators may reliably predict disease progression [138]. The validation of prognostic models which significantly predict malignant transformation and recurrence based on specific histologic biomarkers (e.g., basal cell hyperplasia, loss of epithelial cohesion) has been successful and is ongoing [139]. Ultimately, validation of a prognostic scoring system incorporating molecular markers, clinical risk factors, and histologic grading will provide the greatest clinical utility.
As the field advances toward personalized cancer therapy, there has been an increasing emphasis on identifying predictive biomarkers that can serve as both therapeutic targets as well as predictors of therapy response. For example, we have recently shown that in a subpopulation of HNSCC patients with FAT1 mutations, YAP1, and bromodomain-containing protein 4 (BRD4) can be therapeutically targeted with BET bromodomain inhibitors [132]. Similarly, studies have shown that deficiencies in DNA mismatch repair predict a positive response to immune checkpoint inhibition as well as platinum-based chemotherapies in HNSCC [140,141]. This is also true of cell cycle regulators, as amplification of cyclin D1 predicts cisplatin resistance, TP53 mutations predict susceptibility to G2-M cell cycle inhibitors [142], and Bcl-2 overexpression predicts radiotherapy failure with 71% accuracy [139]. Various therapeutic biomarkers, ranging from viral oncoproteins (e.g., HPV, EBV) and receptor tyrosine kinases (e.g., EGFR, PIK3CA) to immune checkpoint markers (e.g., PD-L1, PD-L2) and tumor suppressor proteins (e.g., TP53) have been identified as therapeutic targets in HNSCC [142]. This work has expanded treatment options in HNSCC to include surgery, radio- and chemotherapy, immune checkpoint inhibition, and molecular targeted therapy [143]. However, there are comparatively fewer studies testing the vulnerability of premalignant cells to these therapies [144]. Therapeutic interventions for HNSCC may not be equally efficacious in pre-cancerous head and neck lesions. For example, EGFR targeted therapy, which has been approved for HNSCC treatment, was not found to have comparable efficacy when used in patients with oral premalignant lesions [145,146]. Identifying molecular based strategies specifically for precancerous lesions is paramount for prevention of HNSCC, and the biomarkers reviewed here provide a future direction as potential therapeutic targets.
Despite their promising clinical applications, as few as 0.1% of biomarkers are successfully translated into routine clinical practice due to several limitations in current studies: (1) variation in methodology for measurement of marker expression; (2) heterogeneity of cancer samples and study populations; (3) small sample sizes; (4) limited sensitivity, specificity, and predictive value of markers; (5) absence of rigorous clinical validation of markers [147,148]. Future standardized studies addressing these limitations are needed before biomarker tests can be recommended for clinical use. Ultimately, elucidating the cellular, molecular, and genetic events in head and neck carcinogenesis and associated tumor markers is critical to improving the management of HNSCC.

Author Contributions

Conceptualization, K.R., M.R.N. and S.V.S.; writing—original draft preparation, K.R.; writing—review and editing, K.R., A.L.F., R.A.F., M.A.V., W.C.F., M.R.N. and S.V.S. All authors have read and agreed to the published version of the manuscript.

Funding

S.V.S. received Mike Toth head and neck cancer funds.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Aupérin, A. Epidemiology of head and neck cancers: An update. Curr. Opin. Oncol. 2020, 32, 178–186. [Google Scholar] [CrossRef]
  2. Marcu, L.G.; Yeoh, E. A review of risk factors and genetic alterations in head and neck carcinogenesis and implications for current and future approaches to treatment. J. Cancer Res. Clin. Oncol. 2009, 135, 1303–1314. [Google Scholar] [CrossRef]
  3. Stepnick, D.; Gilpin, D. Head and Neck Cancer: An Overview. Semin. Plast. Surg. 2010, 24, 107–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Seiwert, T.Y.; Cohen, E.E.W. State-of-the-art management of locally advanced head and neck cancer. Br. J. Cancer 2005, 92, 1341–1348. [Google Scholar] [CrossRef] [PubMed]
  5. Pulte, D.; Brenner, H. Changes in Survival in Head and Neck Cancers in the Late 20th and Early 21st Century: A Period Analysis. Oncologist 2010, 15, 994–1001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Miranda-Galvis, M.; Loveless, R.; Kowalski, L.; Teng, Y. Impacts of Environmental Factors on Head and Neck Cancer Pathogenesis and Progression. Cells 2021, 10, 389. [Google Scholar] [CrossRef] [PubMed]
  7. Klein, J.D.; Grandis, J.R. The Molecular Pathogenesis of Head and Neck Cancer. Cancer Biol. Ther. 2010, 9, 1–7. [Google Scholar] [CrossRef] [Green Version]
  8. Helliwell, T. ‘Risky’ epithelium in the larynx—A practical diagnosis? Histopathology 1999, 34, 262–265. [Google Scholar] [CrossRef]
  9. Bosman, F.T. Dysplasia classification: Pathology in disgrace? J. Pathol. 2001, 194, 143–144. [Google Scholar] [CrossRef]
  10. Fleskens, S.; Slootweg, P. Grading systems in head and neck dysplasia: Their prognostic value, weaknesses and utility. Head Neck Oncol. 2009, 1, 11. [Google Scholar] [CrossRef]
  11. Kujan, O.; Oliver, R.J.; Khattab, A.; Roberts, S.A.; Thakker, N.; Sloan, P. Evaluation of a new binary system of grading oral epithelial dysplasia for prediction of malignant transformation. Oral Oncol. 2006, 42, 987–993. [Google Scholar] [CrossRef]
  12. Garnis, C.; Chari, R.; Buys, T.P.H.; Zhang, L.; Ng, R.T.; Rosin, M.P.; Lam, W.L. Genomic imbalances in precancerous tissues signal oral cancer risk. Mol. Cancer 2009, 8, 50. [Google Scholar] [CrossRef] [Green Version]
  13. Chin, D.; Boyle, G.M.; Williams, R.M.; Ferguson, K.; Pandeya, N.; Pedley, J.; Campbell, C.M.; Theile, D.R.; Parsons, P.G.; Coman, W.B. Novel markers for poor prognosis in head and neck cancer. Int. J. Cancer 2005, 113, 789–797. [Google Scholar] [CrossRef] [PubMed]
  14. Califano, J.; van der Riet, P.; Westra, W.; Nawroz, H.; Clayman, G.; Piantadosi, S.; Corio, R.; Lee, D.; Greenberg, B.; Koch, W.; et al. Genetic progression model for head and neck cancer: Implications for field cancerization. Cancer Res. 1996, 56, 2488–2492. [Google Scholar] [CrossRef]
  15. Haddad, R.I.; Shin, D.M. Recent Advances in Head and Neck Cancer. N. Engl. J. Med. 2008, 359, 1143–1154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Perez-Ordonez, B.; Beauchemin, M.; Jordan, R.C.K. Molecular biology of squamous cell carcinoma of the head and neck. J. Clin. Pathol. 2006, 59, 445–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Johnson, D.E.; Burtness, B.; Leemans, C.R.; Lui, V.W.Y.; Bauman, J.E.; Grandis, J.R. Head and neck squamous cell carcinoma. Nat. Rev. Dis. Primers 2020, 6, 92. [Google Scholar] [CrossRef]
  18. Argiris, A.; Karamouzis, M.V.; Raben, D.; Ferris, R.L. Head and neck cancer. Lancet 2008, 371, 1695–1709. [Google Scholar] [CrossRef]
  19. Loffek, S.; Schilling, O.; Franzke, C.-W. Biological role of matrix metalloproteinases: A critical balance. Eur. Respir. J. 2011, 38, 191–208. [Google Scholar] [CrossRef] [Green Version]
  20. Stamenkovic, I. Matrix metalloproteinases in tumor invasion and metastasis. Semin. Cancer Biol. 2000, 10, 415–433. [Google Scholar] [CrossRef]
  21. Kadler, K.E.; Hill, A.; Canty-Laird, E.G. Collagen fibrillogenesis: Fibronectin, integrins, and minor collagens as organizers and nucleators. Curr. Opin. Cell Biol. 2008, 20, 495–501. [Google Scholar] [CrossRef]
  22. Bosman, F.T. The borderline: Basement membranes and the transition from premalignant to malignant neoplasia. Microsc. Res. Tech. 1994, 28, 216–225. [Google Scholar] [CrossRef] [Green Version]
  23. Fan, H.-X.; Li, H.-X.; Chen, D.; Gao, Z.-X.; Zheng, J.-H. Changes in the expression of MMP2, MMP9, and ColIV in stromal cells in oral squamous tongue cell carcinoma: Relationships and prognostic implications. J. Exp. Clin. Cancer Res. 2012, 31, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Jose, D.; Mane, D.R. Correlation of matrix metalloproteinase-9 expression with morphometric analysis of mucosal vasculature in oral squamous cell carcinoma, oral epithelial dysplasia, and normal oral mucosa. Int. J. Health Sci. 2018, 12, 36–43. [Google Scholar]
  25. Fraga, C.A.d.C.; Farias, L.C.; de Oliveira, M.V.M.; Domingos, P.L.B.; Pereira, C.S.; Silva, T.F.; Roy, A.; Gomez, R.S.; de Paula, A.M.B.; Guimarães, A.L.S. Increased VEGFR2 and MMP9 protein levels are associated with epithelial dysplasia grading. Pathol. Res. Pract. 2014, 210, 959–964. [Google Scholar] [CrossRef] [PubMed]
  26. Jordan, R.C.K.; Macabeo-Ong, M.; Shiboski, C.H.; Dekker, N.; Ginzinger, D.G.; Wong, D.T.W.; Schmidt, B.L. Overexpression of Matrix Metalloproteinase-1 and -9 mRNA Is Associated with Progression of Oral Dysplasia to Cancer. Clin. Cancer Res. 2004, 10, 6460–6465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Zhang, Z.; Guo, W.; Zhang, Y.; Wang, X.; Liu, H.; Xu, S.; Zhao, Z.; Chen, D. Changes in the expression of Col IV, gelatinase and TIMP-1 in oral leukoplakia. Int. J. Clin. Exp. Pathol. 2017, 10, 8535–8543. [Google Scholar] [PubMed]
  28. Kessenbrock, K.; Plaks, V.; Werb, Z. Matrix metalloproteinases: Regulators of the tumor microenvironment. Cell 2010, 141, 52–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Mane, D.R.; Arora, M. Immunohistochemical expression of extracellular matrix metalloproteinase inducer (EMMPRIN) in normal oral mucosa, oral epithelial dysplasia and oral squamous cell carcinoma. J. Oral Maxillofac. Pathol. 2018, 22, 279–280. [Google Scholar] [CrossRef]
  30. Astarita, J.L.; Acton, S.E.; Turley, S.J. Podoplanin: Emerging functions in development, the immune system, and cancer. Front. Immunol. 2012, 3, 283. [Google Scholar] [CrossRef] [Green Version]
  31. Ugorski, M.; Dziegiel, P.; Suchanski, J. Podoplanin—A small glycoprotein with many faces. Am. J. Cancer Res. 2016, 6, 370–386. [Google Scholar] [PubMed]
  32. Yuan, P.; Temam, S.; El-Naggar, A.; Zhou, X.; Liu, D.D.; Lee, J.J.; Mao, L. Overexpression of podoplanin in oral cancer and its association with poor clinical outcome. Cancer 2006, 107, 563–569. [Google Scholar] [CrossRef] [PubMed]
  33. Kawaguchi, H.; El-Naggar, A.K.; Papadimitrakopoulou, V.; Ren, H.; Fan, Y.-H.; Feng, L.; Lee, J.J.; Kim, E.; Hong, W.K.; Lippman, S.M.; et al. Podoplanin: A Novel Marker for Oral Cancer Risk in Patients with Oral Premalignancy. J. Clin. Oncol. 2008, 26, 354–360. [Google Scholar] [CrossRef] [PubMed]
  34. Verma, V.; Chandrashekar, C. Evaluation of SOX2 and podoplanin expression in oral epithelial dysplasia and its correlation with malignant transformation. J. Investig. Clin. Dent. 2019, 10, e12450. [Google Scholar] [CrossRef] [PubMed]
  35. D’Souza, B.; Nayak, R.; Kotrashetti, V.S. Immunohistochemical Expression of Podoplanin in Clinical Variants of Oral Leukoplakia and Its Correlation with Epithelial Dysplasia. Appl. Immunohistochem. Mol. Morphol. 2018, 26, 132–139. [Google Scholar] [CrossRef] [PubMed]
  36. Grochau, K.J.; Safi, A.-F.; Drebber, U.; Grandoch, A.; Zöller, J.E.; Kreppel, M. Podoplanin expression in oral leukoplakia─a prospective study. J. Cranio-Maxillofac. Surg. 2019, 47, 505–509. [Google Scholar] [CrossRef] [PubMed]
  37. Wicki, A.; Christofori, G. The potential role of podoplanin in tumour invasion. Br. J. Cancer 2007, 96, 1–5. [Google Scholar] [CrossRef]
  38. Wicki, A.; Lehembre, F.; Wick, N.; Hantusch, B.; Kerjaschki, D.; Christofori, G. Tumor invasion in the absence of epithelial-mesenchymal transition: Podoplanin-mediated remodeling of the actin cytoskeleton. Cancer Cell 2006, 9, 261–272. [Google Scholar] [CrossRef]
  39. Atsumi, N.; Ishii, G.; Kojima, M.; Sanada, M.; Fujii, S.; Ochiai, A. Podoplanin, a novel marker of tumor-initiating cells in human squamous cell carcinoma A431. Biochem. Biophys. Res. Commun. 2008, 373, 36–41. [Google Scholar] [CrossRef] [PubMed]
  40. Upadhaya, P.; Barhoi, D.; Giri, A.; Bhattacharjee, A.; Giri, S. Joint detection of claudin-1 and junctional adhesion molecule-A as a therapeutic target in oral epithelial dysplasia and oral squamous cell carcinoma. J. Cell. Biochem. 2019, 120, 18117–18127. [Google Scholar] [CrossRef]
  41. Tian, Y.; Tian, Y.; Zhang, W.; Wei, F.; Yang, J.; Luo, X.; Zhou, T.; Hou, B.; Qian, S.; Deng, X.; et al. Junctional adhesion molecule-A, an epithelial–mesenchymal transition inducer, correlates with metastasis and poor prognosis in human nasopharyngeal cancer. Carcinogenesis 2015, 36, 41–48. [Google Scholar] [CrossRef]
  42. Dhawan, P.; Singh, A.B.; Deane, N.G.; No, Y.; Shiou, S.-R.; Schmidt, C.; Neff, J.; Washington, M.K.; Beauchamp, R.D. Claudin-1 regulates cellular transformation and metastatic behavior in colon cancer. J. Clin. Investig. 2005, 115, 1765–1776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Von Zeidler, S.V.; Botelho, T.d.S.; Mendonça, E.F.; Batista, A.C. E-cadherin as a potential biomarker of malignant transformation in oral leukoplakia: A retrospective cohort study. BMC Cancer 2014, 14, 972. [Google Scholar] [CrossRef] [Green Version]
  44. Adams, C.L.; Chen, Y.-T.; Smith, S.J.; Nelson, W.J. Mechanisms of Epithelial Cell–Cell Adhesion and Cell Compaction Revealed by High-resolution Tracking of E-Cadherin–Green Fluorescent Protein. J. Cell Biol. 1998, 142, 1105–1119. [Google Scholar] [CrossRef]
  45. Halbleib, J.M.; Nelson, W.J. Cadherins in development: Cell adhesion, sorting, and tissue morphogenesis. Genes Dev. 2006, 20, 3199–3214. [Google Scholar] [CrossRef] [Green Version]
  46. Sathish, I.I.; Asokan, K.; Krithika, C.L.; Ramanathan, A. Expression of E- Cadherin and Levels of Dysplasia in Oral Leukoplakia—A Prospective Cohort Study. Asian Pac. J. Cancer Prev. 2020, 21, 405–410. [Google Scholar] [CrossRef] [PubMed]
  47. Santos García, A.; Abad Hernández, M.M.; Fonseca Sánchez, E.; Gonzalez, R.J.; Galindo Villardón, P.; Cruz Hernández, J.J.; Bullon-Sopelana, A. E-cadherin, laminin and collagen IV expression in the evolution from dysplasia to oral squamous cell carcinoma. Med. Oral Patol. Oral Cir. Bucal. 2006, 11, E100–E105. [Google Scholar] [PubMed]
  48. Zhang, W.; Alt-Holland, A.; Margulis, A.; Shamis, Y.; Fusenig, N.E.; Rodeck, U.; Garlick, J.A. E-cadherin loss promotes the initiation of squamous cell carcinoma invasion through modulation of integrin-mediated adhesion. J. Cell Sci. 2006, 119, 283–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Sneath, R.J.; Mangham, D.C. The normal structure and function of CD44 and its role in neoplasia. J. Clin. Pathol. Mol. Pathol. 1998, 51, 191–200. [Google Scholar] [CrossRef] [Green Version]
  50. Thapa, R.; Wilson, G.D. The Importance of CD44 as a Stem Cell Biomarker and Therapeutic Target in Cancer. Stem Cells Int. 2016, 2016, 2087204. [Google Scholar] [CrossRef] [Green Version]
  51. Wang, L.; Zuo, X.; Xie, K.; Wei, D. The Role of CD44 and Cancer Stem Cells. Methods Mol. Biol. 2018, 1692, 31–42. [Google Scholar] [CrossRef] [PubMed]
  52. Saghravanian, N.; Mirhashemi, M.; Ghazi, N.; Taghipour, A.; Mohajertehran, F. Evaluation of CD24 and CD44 as cancer stem cell markers in squamous cell carcinoma and epithelial dysplasia of the oral cavity by q-RT-PCR. Dent. Res. J. 2020, 17, 208. [Google Scholar] [CrossRef]
  53. Ghazi, N.; Saghravanian, N.; Ghazi, A.; Shakeri, M.T.; Khajehbahrami, H. CD44 Expression in Dysplastic and Non-Dysplastic Oral Lichen Planus. Int. J. Cancer Manag. 2020, 13, e98061. [Google Scholar] [CrossRef]
  54. Godge, P.; Poonja, L.; Py, G.; Ls, P. Quantitative assessment of expression of cell adhesion molecule (CD44) splice variants: CD44 standard (CD44s) and v5, v6 isoforms in oral leukoplakias: An immunohistochemical study. Indian J. Dent. Res. 2011, 22, 493. [Google Scholar] [CrossRef] [PubMed]
  55. González-Moles, M.A.; Bravo, M.; Ruiz-Ávila, I.; Esteban, F.; Bascones-Martínez, A.; González-Moles, S. Adhesion molecule CD44 expression in non-tumour epithelium adjacent to tongue cancer. Oral Oncol. 2004, 40, 281–286. [Google Scholar] [CrossRef] [PubMed]
  56. Hirvikoski, P.; Tammi, R.; Kumpulainen, E.; Virtaniemi, J.; Parkkinen, J.J.; Tammi, M.; Johansson, R.; Ågren, U.; Karhunen, J.; Kosma, V.-M. Irregular expression of hyaluronan and its CD44 receptor is associated with metastatic phenotype in laryngeal squamous cell carcinoma. Virchows Arch. 1999, 434, 37–44. [Google Scholar] [CrossRef]
  57. Soukka, T.; Salmi, M.; Joensuu, H.; Häkkinen, L.; Sointu, P.; Koulu, L.; Kalimo, K.; Klemi, P.; Grenman, R.; Jalkanen, S. Regulation of CD44v6-containing isoforms during proliferation of normal and malignant epithelial cells. Cancer Res. 1997, 57, 2281–2289. [Google Scholar]
  58. Mesrati, M.H.; Syafruddin, S.E.; Mohtar, M.A.; Syahir, A. CD44: A Multifunctional Mediator of Cancer Progression. Biomolecules 2021, 11, 1850. [Google Scholar] [CrossRef]
  59. Glumac, P.M.; Lebeau, A.M. The role of CD133 in cancer: A concise review. Clin. Transl. Med. 2018, 7, 18. [Google Scholar] [CrossRef]
  60. Li, Z. CD133: A stem cell biomarker and beyond. Exp. Hematol. Oncol. 2013, 2, 17. [Google Scholar] [CrossRef] [Green Version]
  61. Ravindran, G.; Devaraj, H. Aberrant expression of CD133 and musashi-1 in preneoplastic and neoplastic human oral squamous epithelium and their correlation with clinicopathological factors. Head Neck 2012, 34, 1129–1135. [Google Scholar] [CrossRef] [PubMed]
  62. Jiajia, Q.; Yan, S.; Changqing, Y.; Wenjing, J.; Han, Z.; Yuanpan, C.; Qiuyan, L. Clinical significance of CD44 and CD133 expression in oral potentially malignant disorder and oral squamous cell carcinoma. Hua Xi Kou Qiang Yi Xue Za Zhi 2017, 35, 311–316. [Google Scholar] [CrossRef] [PubMed]
  63. Sun, L.; Feng, J.; Ma, L.; Liu, W.; Zhou, Z. CD133 expression in oral lichen planus correlated with the risk for progression to oral squamous cell carcinoma. Ann. Diagn. Pathol. 2013, 17, 486–489. [Google Scholar] [CrossRef] [PubMed]
  64. Liu, W.; Wu, L.; Shen, X.-M.; Shi, L.-J.; Zhang, C.-P.; Xu, L.-Q.; Zhou, Z.-T. Expression patterns of cancer stem cell markers ALDH1 and CD133 correlate with a high risk of malignant transformation of oral leukoplakia. Int. J. Cancer 2012, 132, 868–874. [Google Scholar] [CrossRef] [PubMed]
  65. Luna, E.C.M.; Bezerra, T.M.M.; de Barros Silva, P.G.; Cavalcante, R.B.; Costa, F.W.G.; Alves, A.P.N.N.; Chaves, F.N.; Pereira, K.M.A. CD133 Role in Oral Carcinogenesis. Asian Pac. J. Cancer Prev. 2020, 21, 2501–2506. [Google Scholar] [CrossRef]
  66. Botha, H.; Farah, C.; Koo, K.; Cirillo, N.; McCullough, M.; Paolini, R.; Celentano, A. The Role of Glucose Transporters in Oral Squamous Cell Carcinoma. Biomolecules 2021, 11, 1070. [Google Scholar] [CrossRef]
  67. Angadi, V.C.; Angadi, P.V. GLUT-1 immunoexpression in oral epithelial dysplasia, oral squamous cell carcinoma, and verrucous carcinoma. J. Oral Sci. 2015, 57, 115–122. [Google Scholar] [CrossRef] [Green Version]
  68. Feitosa, S.G.; Viana, K.F.; Luna, E.C.M.; Costa, F.W.G.; Cavalcante, R.B.; Chaves, F.N.; Chaves, H.V.; Pereira, K.M.A. Immunohistochemical Evaluation of GLUT-3 and GLUT-4 in Oral Epithelial Dysplasia and Oral Squamous Cell Carcinoma. Asian Pac. J. Cancer Prev. 2018, 19, 1779–1783. [Google Scholar] [CrossRef]
  69. Chang, Y.-C.; Chi, T.; Chang, W.-M.; Su, C.-Y.; Lin, Y.-F.; Chen, C.-L.; Chen, M.-H.; Chang, P.M.-H.; Wu, A.T.H.; Hsiao, M. Glucose transporter 4 promotes head and neck squamous cell carcinoma metastasis through the TRIM24-DDX58 axis. J. Hematol. Oncol. 2017, 10, 11. [Google Scholar] [CrossRef] [Green Version]
  70. Pereira, K.M.A.; Feitosa, S.G.; Lima, A.T.T.; Luna, E.C.M.; Cavalcante, R.B.; de Lima, K.C.; Chaves, F.N.; Costa, F.W.G. Immunohistochemical Evaluation of Glucose Transporter Type 1 in Epithelial Dysplasia and Oral Squamous Cell Carcinoma. Asian Pac. J. Cancer Prev. 2016, 17, 147–151. [Google Scholar] [CrossRef] [Green Version]
  71. Reisser, C.; Eichhorn, K.; Herold-Mende, C.; Born, A.I.; Bannasch, P. Expression of facilitative glucose transport proteins during development of squamous cell carcinomas of the head and neck. Int. J. Cancer. 1999, 80, 194–198. [Google Scholar] [CrossRef]
  72. Brooks Robey, R.; Weisz, J.; Kuemmerle, N.B.; Salzberg, A.C.; Berg, A.; Brown, D.G.; Kubik, L.; Palorini, R.; Al-Mulla, F.; Al-Temaimi, R.; et al. Metabolic reprogramming and dysregulated metabolism: Cause, consequence and/or enabler of environmental carcinogenesis? Carcinogenesis 2015, 36, S203–S231. [Google Scholar] [CrossRef] [PubMed]
  73. Rao, R.S.; Raju, K.L.; Augustine, D.; Patil, S. Prognostic Significance of ALDH1, Bmi1, and OCT4 Expression in Oral Epithelial Dysplasia and Oral Squamous Cell Carcinoma. Cancer Control 2020, 27, 1073274820904959. [Google Scholar] [CrossRef] [PubMed]
  74. Marcato, P.; Dean, C.A.; Giacomantonio, C.A.; Lee, P.W. Aldehyde dehydrogenase: Its role as a cancer stem cell marker comes down to the specific isoform. Cell Cycle 2011, 10, 1378–1384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Chen, Y.-C.; Chen, Y.-W.; Hsu, H.-S.; Tseng, L.-M.; Huang, P.-I.; Lu, K.-H.; Chen, D.-T.; Tai, L.-K.; Yung, M.-C.; Chang, S.-C.; et al. Aldehyde dehydrogenase 1 is a putative marker for cancer stem cells in head and neck squamous cancer. Biochem. Biophys. Res. Commun. 2009, 385, 307–313. [Google Scholar] [CrossRef]
  76. Dhumal, S.N.; Choudhari, S.K.; Patankar, S.; Ghule, S.S.; Jadhav, Y.B.; Masne, S. Cancer Stem Cell Markers, CD44 and ALDH1, for Assessment of Cancer Risk in OPMDs and Lymph Node Metastasis in Oral Squamous Cell Carcinoma. Head Neck Pathol. 2021, 16, 453–465. [Google Scholar] [CrossRef]
  77. Dubey, A.; Prajapati, K.S.; Swamy, M.; Pachauri, V. Heat shock proteins: A therapeutic target worth to consider. Vet. World 2015, 8, 46–51. [Google Scholar] [CrossRef] [Green Version]
  78. Hendrick, J.P.; Hartl, F.-U. Molecular chaperone functions of heat-shock proteins. Annu. Rev. Biochem. 1993, 62, 349–384. [Google Scholar] [CrossRef]
  79. Priyanka, K.P.; Majumdar, S.; Kotina, S.; Uppala, D.; Balla, H. Expression of Heat Shock Protein 70 in Oral Epithelial Dysplasia and Oral Squamous Cell Carcinoma: An Immunohistochemical Study. Contemp. Clin. Dent. 2019, 10, 185–190. [Google Scholar] [CrossRef]
  80. Patil, P.; Nandimath, K.; Prabhu, S.; Naikmasur, V.G. Heat shock protein (HSP70) as a marker of epithelial dysplasia in oral dysplastic lesions: A clinicopathological study. J. Oral Maxillofac. Pathol. 2015, 19, 53–57. [Google Scholar] [CrossRef] [Green Version]
  81. Bar, J.K.; Cierpikowski, P.; Lis-Nawara, A.; Duc, P.; Hałoń, A.; Radwan-Oczko, M. Comparison of p53, HSP90, E-cadherin and HPV in oral lichen planus and oral squamous cell carcinoma. Acta Otorhinolaryngol. Ital. 2021, 41, 514–522. [Google Scholar] [CrossRef] [PubMed]
  82. Sobel, A. Stathmin: A relay phosphoprotein for multiple signal transduction? Trends Biochem. Sci. 1991, 16, 301–305. [Google Scholar] [CrossRef]
  83. Kang, W.; Tong, J.H.M.; Chan, A.; Lung, R.W.M.; Chau, S.L.; Wong, Q.W.L.; Wong, N.; Yu, J.; Cheng, A.; To, K.F. Stathmin1 plays oncogenic role and is a target of MicroRNA-223 in gastric cancer. PLoS ONE 2012, 7, e33919. [Google Scholar] [CrossRef] [PubMed]
  84. Curmi, P.A.; Noguès, C.; Lachkar, S.; Carelle, N.; Gonthier, M.P.; Sobel, A.; Lidereau, R.; Bièche, I. Overexpression of stathmin in breast carcinomas points out to highly proliferative tumours. Br. J. Cancer 2000, 82, 142–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Cheng, A.-L.; Huang, W.-G.; Chen, Z.-C.; Peng, F.; Zhang, P.-F.; Li, M.-Y.; Li, F.; Li, J.-L.; Li, C.; Yi, H.; et al. Identification of Novel Nasopharyngeal Carcinoma Biomarkers by Laser Capture Microdissection and Proteomic Analysis. Clin. Cancer Res. 2008, 14, 435–445. [Google Scholar] [CrossRef] [Green Version]
  86. Kouzu, Y.; Uzawa, K.; Koike, H.; Saito, K.; Nakashima, D.; Higo, M.; Endo, Y.; Kasamatsu, A.; Shiiba, M.; Bukawa, H.; et al. Overexpression of stathmin in oral squamous-cell carcinoma: Correlation with tumour progression and poor prognosis. Br. J. Cancer 2006, 94, 717–723. [Google Scholar] [CrossRef] [Green Version]
  87. Vadla, P.; Deepthi, G.; Arun Kumar, C.; Bashamalla, R.; Syeda, N.; Naramala, S. Immunohistochemical expression of stathmin in oral dysplasia: An original study with an insight of its action on microtubules. J. Oral Maxillofac. Pathol. 2021, 25, 247–252. [Google Scholar] [CrossRef]
  88. Vadla, P.; Yeluri, S.; Deepthi, G.; Guttikonda, V.R.; Taneeru, S.; Naramala, S. Stathmin! An immunohistochemical analysis of the novel marker in Oral Squamous Cell Carcinoma and Oral Leukoplakia. Asian Pac. J. Cancer Prev. 2020, 21, 3317–3323. [Google Scholar] [CrossRef]
  89. Pallavi, N.; Nalabolu, G.R.K.; Hiremath, S.K.S. Bcl-2 and c-Myc expression in oral dysplasia and oral squamous cell carcinoma: An immunohistochemical study to assess tumor progression. J. Oral Maxillofac. Pathol. 2018, 22, 325. [Google Scholar] [CrossRef]
  90. Altieri, D.C. Survivin in apoptosis control and cell cycle regulation in cancer. Prog. Cell Cycle Res. 2003, 5, 447–452. [Google Scholar]
  91. Chipuk, J.E.; Green, D.R. How do BCL-2 proteins induce mitochondrial outer membrane permeabilization? Trends Cell Biol. 2008, 18, 157–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Hardwick, J.M.; Soane, L. Multiple Functions of BCL-2 Family Proteins. Cold Spring Harb. Perspect. Biol. 2013, 5, a008722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Juneja, S.; Chaitanya, N.B.; Agarwal, M. Immunohistochemical expression of Bcl-2 in oral epithelial dysplasia and oral squamous cell carcinoma. Indian J. Cancer 2015, 52, 505. [Google Scholar] [CrossRef] [PubMed]
  94. Chamorro-Petronacci, C.M.; de Mendoza, I.L.-I.; Suarez-Peñaranda, J.M.; Padin-Iruegas, E.; Blanco-Carrion, A.; Lorenzo-Pouso, A.I.; Ortega, K.L.; Pérez-Sayáns, M. Immunohistochemical Characterization of Bcl-2 in Oral Potentially Malignant Disorders. Appl. Immunohistochem. Mol. Morphol. 2021, 29, 706–712. [Google Scholar] [CrossRef]
  95. Tanaka, C.; Uzawa, K.; Shibahara, T.; Yokoe, H.; Noma, H.; Tanzawa, H. Expression of an Inhibitor of Apoptosis, Survivin, in Oral Carcinogenesis. J. Dent. Res. 2003, 82, 607–611. [Google Scholar] [CrossRef] [PubMed]
  96. Angelin, D.; Nair, B.J. Comparative evaluation of survivin expression in leukoplakia, lichen planus, and oral squamous cell carcinoma: An immunohistochemical study. J. Cancer Res. Ther. 2020, 16, 569. [Google Scholar] [CrossRef]
  97. Muzio, L.L.; Pannone, G.; Leonardi, R.; Staibano, S.; Mignogna, M.; de Rosa, G.; Kudo, Y.; Takata, T.; Altieri, D. Survivin, a Potential Early Predictor of Tumor Progression in the Oral Mucosa. J. Dent. Res. 2003, 82, 923–928. [Google Scholar] [CrossRef]
  98. Pietsch, E.C.; Sykes, S.M.; McMahon, S.B.; E Murphy, M. The p53 family and programmed cell death. Oncogene 2008, 27, 6507–6521. [Google Scholar] [CrossRef] [Green Version]
  99. Levine, A.J.; Oren, M. The first 30 years of p53: Growing ever more complex. Nat. Rev. Cancer 2009, 9, 749–758. [Google Scholar] [CrossRef] [Green Version]
  100. Marei, H.E.; Althani, A.; Afifi, N.; Hasan, A.; Caceci, T.; Pozzoli, G.; Morrione, A.; Giordano, A.; Cenciarelli, C. p53 signaling in cancer progression and therapy. Cancer Cell Int. 2021, 21, 703. [Google Scholar] [CrossRef]
  101. Gordon, E.M.; Ravicz, J.R.; Liu, S.; Chawla, S.P.; Hall, F.L. Cell cycle checkpoint control: The cyclin G1/Mdm2/p53 axis emerges as a strategic target for broad-spectrum cancer gene therapy—A review of molecular mechanisms for oncologists. Mol. Clin. Oncol. 2018, 9, 115–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Harris, S.L.; Levine, A.J. The p53 pathway: Positive and negative feedback loops. Oncogene 2005, 24, 2899–2908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Chen, J. The Cell-Cycle Arrest and Apoptotic Functions of p53 in Tumor Initiation and Progression. Cold Spring Harb. Perspect. Med. 2016, 6, a026104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Nag, S.; Qin, J.; Srivenugopal, K.S.; Wang, M.; Zhang, R. The MDM2-p53 pathway revisited. J. Biomed. Res. 2013, 27, 254–271. [Google Scholar] [CrossRef]
  105. Boaz, K.; Pandya, J.A.; Natarajan, S.; Manaktala, N.; Nandita, K.; Lewis, A.J. A correlation of immunohistochemical expression of TP53 and CDKN1A in oral epithelial dysplasia and oral squamous cell carcinoma. J. Cancer Res. Ther. 2018, 14, 666–670. [Google Scholar] [CrossRef]
  106. Abrahao, A.; Bonelli, B.V.; Nunes, F.D.; Dias, E.P.; Cabral, M.G. Immunohistochemical expression of p53, p16 and hTERT in oral squamous cell carcinoma and potentially malignant disorders. Braz. Oral Res. 2011, 25, 34–41. [Google Scholar] [CrossRef]
  107. Rodrigo, J.P.; García-Pedrero, J.M.; Suárez, C.; Takes, R.P.; Thompson, L.; Slootweg, P.J.; Woolgar, J.A.; Westra, W.H.; Brakenhoff, R.H.; Rinaldo, A.; et al. Biomarkers predicting malignant progression of laryngeal epithelial precursor lesions: A systematic review. Eur. Arch. Oto-Rhino-Laryngol. 2012, 269, 1073–1083. [Google Scholar] [CrossRef]
  108. Kawano, S.; Matsubara, R.; Kiyosue, T.; Goto, Y.; Hirano, M.; Jinno, T.; Toyoshima, T.; Kitamura, R.; Oobu, K.; Nakamura, S. Increased ΔNp63 expression is predictive of malignant transformation in oral epithelial dysplasia and poor prognosis in oral squamous cell carcinoma. Int. J. Oncol. 2011, 39, 1391–1399. [Google Scholar] [CrossRef]
  109. Ono, S.; Nakano, K.; Takabatake, K.; Kawai, H.; Nagatsuka, H. Immunohistochemistry of YAP and dNp63 and survival analysis of patients bearing precancerous lesion and oral squamous cell carcinoma. Int. J. Med. Sci. 2019, 16, 766–773. [Google Scholar] [CrossRef] [Green Version]
  110. Chen, Y.-K.; Ms, S.-S.H.; Lin, L.-M. p73 expression for human buccal epithelial dysplasia and squamous cell carcinoma: Does it correlate with nodal status of carcinoma and is there a relationship with malignant change of epithelial dysplasia? Head Neck 2004, 26, 945–952. [Google Scholar] [CrossRef]
  111. Bascones-Martínez, A.; López-Durán, M.; Cano-Sánchez, J.; Sánchez-Verde, L.; Díez-Rodríguez, A.; Aguirre-Echebarría, P.; Álvarez-Fernández, E.; González-Moles, M.A.; Bascones-Ilundain, J.; Muzio, L.L.; et al. Differences in the expression of five senescence markers in oral cancer, oral leukoplakia and control samples in humans. Oncol. Lett. 2012, 3, 1319–1325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Bradley, K.T.; Budnick, S.D.; Logani, S. Immunohistochemical detection of p16INK4a in dysplastic lesions of the oral cavity. Mod. Pathol. 2006, 19, 1310–1316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Soni, S.; Kaur, J.; Kumar, A.; Chakravarti, N.; Mathur, M.; Bahadur, S.; Shukla, N.K.; Deo, S.V.; Ralhan, R. Alterations of Rb Pathway Components Are Frequent Events in Patients with Oral Epithelial Dysplasia and Predict Clinical Outcome in Patients with Squamous Cell Carcinoma. Oncology 2005, 68, 314–325. [Google Scholar] [CrossRef] [PubMed]
  114. Garces de los Fayos Alonso, I.; Liang, H.-C.; Turner, S.D.; Lagger, S.; Merkel, O.; Kenner, L. The Role of Activator Protein-1 (AP-1) Family Members in CD30-Positive Lymphomas. Cancers 2018, 10, 93. [Google Scholar] [CrossRef] [Green Version]
  115. Meng, Q.; Xia, Y. c-Jun, at the crossroad of the signaling network. Protein Cell 2011, 2, 889–898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Turatti, E.; Neves, A.d.C.; de Magalhães, M.H.C.G.; de Sousa, S.O.M. Assessment of c-Jun, c-Fos and cyclin D1 in premalignant and malignant oral lesions. J. Oral Sci. 2005, 47, 71–76. [Google Scholar] [CrossRef] [Green Version]
  117. Shraddha, K.; Niranjan, K.; Hallikeri, K. Immunolocalization of c-Jun in normal mucosa, oral submucous fibrosis, epithelial dysplasia, and oral squamous cell carcinoma: A comparative study. J. Cancer Res. Ther. 2018, 14, 1180–1183. [Google Scholar] [CrossRef]
  118. Lima, J.S.; Correa, L.; Klingbeil, M.F.G.; de Sousa, S.C.O.M. C-Jun, pc-Jun, and p27 are differently expressed in oral leukoplakias in smokers and never-smokers. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2016, 121, 73–80. [Google Scholar] [CrossRef]
  119. Rizzino, A. Concise Review: The Sox2-Oct4 Connection: Critical Players in a Much Larger Interdependent Network Integrated at Multiple Levels. Stem Cells 2013, 31, 1033–1039. [Google Scholar] [CrossRef] [Green Version]
  120. Ghazi, N.; Aali, N.; Shahrokhi, V.-R.; Mohajertehran, F.; Saghravanian, N. Relative Expression of SOX2 and OCT4 in Oral Squamous Cell Carcinoma and Oral Epithelial Dysplasia. Rep. Biochem. Mol. Biol. 2020, 9, 171–179. [Google Scholar] [CrossRef]
  121. Hussenet, T.; Du Manoir, S. SOX2 in squamous cell carcinoma: Amplifying a pleiotropic oncogene along carcinogenesis. Cell Cycle 2010, 9, 1480–1486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Tsai, L.-L.; Hu, F.-W.; Lee, S.-S.; Yu, C.-H.; Yu, C.-C.; Chang, Y.-C. Oct4 Mediates Tumor Initiating Properties in Oral Squamous Cell Carcinomas through the Regulation of Epithelial-Mesenchymal Transition. PLoS ONE 2014, 9, e87207. [Google Scholar] [CrossRef] [PubMed]
  123. Raghunandan, B.N.; Sanjai, K.; Kumaraswamy, J.; Papaiah, L.; Pandey, B.; Jyothi, B.M. Expression of human telomerase reverse transcriptase protein in oral epithelial dysplasia and oral squamous cell carcinoma: An immunohistochemical study. J. Oral Maxillofac. Pathol. 2016, 20, 96–101. [Google Scholar] [CrossRef] [PubMed]
  124. Tye, B.K. MCM Proteins in DNA Replication. Annu. Rev. Biochem. 1999, 68, 649–686. [Google Scholar] [CrossRef]
  125. Zakaria, S.H.; Farag, H.A.; Khater, D.S. Immunohistochemical Expression of MCM-2 in Oral Epithelial Dysplasias. Appl. Immunohistochem. Mol. Morphol. 2018, 26, 509–513. [Google Scholar] [CrossRef] [PubMed]
  126. Jessri, M.; Dalley, A.; Farah, C. MutSα and MutLα immunoexpression analysis in diagnostic grading of oral epithelial dysplasia and squamous cell carcinoma. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2015, 119, 74–82. [Google Scholar] [CrossRef] [Green Version]
  127. Awasthi, P.; Foiani, M.; Kumar, A. ATM and ATR signaling at a glance. J. Cell Sci. 2015, 128, 4255–4262. [Google Scholar] [CrossRef] [Green Version]
  128. Zhu, M.; Liu, W.; Shi, L.; Xiao, X.; Wu, W.; Wu, L.; Zhou, Z. Expression of DNA doublestrand repair proteins in oral leukoplakia and the risk of malignant transformation. Oncol. Lett. 2018, 15, 9827–9835. [Google Scholar] [CrossRef]
  129. Ho, M.W.; Ryan, M.P.; Gupta, J.; Triantafyllou, A.; Risk, J.M.; Shaw, R.J.; Wilson, J.B. Loss of FANCD2 and related proteins may predict malignant transformation in oral epithelial dysplasia. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2022, 133, 377–387. [Google Scholar] [CrossRef]
  130. Ma, S.; Meng, Z.; Chen, R.; Guan, K.-L. The Hippo Pathway: Biology and Pathophysiology. Annu. Rev. Biochem. 2019, 88, 577–604. [Google Scholar] [CrossRef] [Green Version]
  131. Pastushenko, I.; Mauri, F.; Song, Y.; de Cock, F.; Meeusen, B.; Swedlund, B.; Impens, F.; van Haver, D.; Opitz, M.; Thery, M.; et al. Fat1 deletion promotes hybrid EMT state, tumour stemness and metastasis. Nature 2021, 589, 448–455. [Google Scholar] [CrossRef] [PubMed]
  132. Chen, N.; Golczer, G.; Ghose, S.; Lin, B.; Langenbucher, A.; Webb, J.; Bhanot, H.; Abt, N.B.; Lin, D.; Varvares, M.; et al. YAP1 maintains active chromatin state in head and neck squamous cell carcinomas that promotes tumorigenesis through cooperation with BRD4. Cell Rep. 2022, 39, 110970. [Google Scholar] [CrossRef] [PubMed]
  133. Hatterschide, J.; Castagnino, P.; Kim, H.W.; Sperry, S.M.; Montone, K.T.; Basu, D.; A White, E. YAP1 activation by human papillomavirus E7 promotes basal cell identity in squamous epithelia. eLife 2022, 11, e75466. [Google Scholar] [CrossRef]
  134. Olmedo-Nieva, L.; Muñoz-Bello, J.O.; Manzo-Merino, J.; Lizano, M. New insights in Hippo signalling alteration in human papillomavirus-related cancers. Cell. Signal. 2020, 76, 109815. [Google Scholar] [CrossRef]
  135. Omori, H.; Nishio, M.; Masuda, M.; Miyachi, Y.; Ueda, F.; Nakano, T.; Sato, K.; Mimori, K.; Taguchi, K.; Hikasa, H.; et al. YAP1 is a potent driver of the onset and progression of oral squamous cell carcinoma. Sci. Adv. 2020, 6, eaay3324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Schaaij-Visser, T.B.; Brakenhoff, R.H.; Leemans, C.R.; Heck, A.J.; Slijper, M. Protein biomarker discovery for head and neck cancer. J. Proteom. 2010, 73, 1790–1803. [Google Scholar] [CrossRef]
  137. Hunter, K.D.; Parkinson, E.K.; Harrison, P.R. Profiling early head and neck cancer. Nat. Rev. Cancer 2005, 5, 127–135. [Google Scholar] [CrossRef]
  138. Ranganathan, K.; Kavitha, L. Oral epithelial dysplasia: Classifications and clinical relevance in risk assessment of oral potentially malignant disorders. J. Oral Maxillofac. Pathol. 2019, 23, 19–27. [Google Scholar] [CrossRef]
  139. Mahmood, H.; Bradburn, M.; Rajpoot, N.; Islam, N.M.; Kujan, O.; Khurram, S.A. Prediction of malignant transformation and recurrence of oral epithelial dysplasia using architectural and cytological feature specific prognostic models. Mod. Pathol. 2022, 35, 1151–1159. [Google Scholar] [CrossRef]
  140. Cilona, M.; Locatello, L.G.; Novelli, L.; Gallo, O. The Mismatch Repair System (MMR) in Head and Neck Carcinogenesis and Its Role in Modulating the Response to Immunotherapy: A Critical Review. Cancers 2020, 12, 3006. [Google Scholar] [CrossRef]
  141. Ameri, A.; Mortazavi, N.; Ahmadi, H.K.; Novin, K. ERCC1 Expression Can Predict Response to Platinum-Based Induction Chemotherapy in Head and Neck Cancer Cases. Asian Pac. J. Cancer Prev. 2016, 17, 87–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Eze, N.; Lo, Y.-C.; Burtness, B. Biomarker driven treatment of head and neck squamous cell cancer. Cancers Head Neck 2017, 2, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Rieke, D.T.; Klinghammer, K.; Keilholz, U. Targeted Therapy of Head and Neck Cancer. Oncol. Res. Treat. 2016, 39, 780–786. [Google Scholar] [CrossRef] [PubMed]
  144. Van Harten, A.; Brakenhoff, R. Targeted Treatment of Head and Neck (Pre)Cancer: Preclinical Target Identification and Development of Novel Therapeutic Applications. Cancers 2021, 13, 2774. [Google Scholar] [CrossRef] [PubMed]
  145. Khan, Z.; Epstein, J.B.; Marur, S.; Gillespie, M.B.; Feldman, L.; Tsai, H.-L.; Zhang, Z.; Wang, H.; Sciubba, J.; Ferris, R.L.; et al. Cetuximab activity in dysplastic lesions of the upper aerodigestive tract. Oral Oncol. 2016, 56, 60–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. William, W.N., Jr.; Papadimitrakopoulou, V.; Lee, J.J.; Mao, L.; Cohen, E.E.W.; Lin, H.Y.; Gillenwater, A.M.; Martin, J.W.; Lingen, M.W.; Boyle, J.O.; et al. Erlotinib and the Risk of Oral Cancer: The Erlotinib Prevention of Oral Cancer (EPOC) Randomized Clinical Trial. JAMA Oncol. 2016, 2, 209–216. [Google Scholar] [CrossRef] [Green Version]
  147. Mishra, A.; Verma, M. Cancer Biomarkers: Are We Ready for the Prime Time? Cancers 2010, 2, 190–208. [Google Scholar] [CrossRef]
  148. Negm, R.S.; Verma, M.; Srivastava, S. The promise of biomarkers in cancer screening and detection. Trends Mol. Med. 2002, 8, 288–293. [Google Scholar] [CrossRef]
Figure 1. Molecular biomarkers in head and neck dysplasia. Biomarkers include matrix metalloproteinases, cell surface receptors, cancer stem cell markers, cell adhesion molecules, transcriptional regulators, DNA replication and repair proteins, and cell cycle regulators. Created with BioRender.
Figure 1. Molecular biomarkers in head and neck dysplasia. Biomarkers include matrix metalloproteinases, cell surface receptors, cancer stem cell markers, cell adhesion molecules, transcriptional regulators, DNA replication and repair proteins, and cell cycle regulators. Created with BioRender.
Cancers 14 05581 g001
Figure 2. Genetic progression model of head and neck squamous cell carcinoma. Accumulation of loss of heterozygosity or amplification at various genetic loci occurs as lesions proceed from normal mucosa to hyperplasia to dysplasia to carcinoma in situ to invasive carcinoma. Created with BioRender.
Figure 2. Genetic progression model of head and neck squamous cell carcinoma. Accumulation of loss of heterozygosity or amplification at various genetic loci occurs as lesions proceed from normal mucosa to hyperplasia to dysplasia to carcinoma in situ to invasive carcinoma. Created with BioRender.
Cancers 14 05581 g002
Figure 3. p53 tumor suppressor pathway. p53 acts on the G1/S checkpoint of the cell cycle to mediate cell cycle arrest via activation of CDK inhibitors. Arrows represent alterations in head and neck dysplasia. Adapted from “G1/S Checkpoint” and “The p53-Mediated Response,” by BioRender.com (2022) (https://app.biorender.com/biorender-templates (accessed on 10 August 2022)).
Figure 3. p53 tumor suppressor pathway. p53 acts on the G1/S checkpoint of the cell cycle to mediate cell cycle arrest via activation of CDK inhibitors. Arrows represent alterations in head and neck dysplasia. Adapted from “G1/S Checkpoint” and “The p53-Mediated Response,” by BioRender.com (2022) (https://app.biorender.com/biorender-templates (accessed on 10 August 2022)).
Cancers 14 05581 g003
Figure 4. Transcriptional regulators in head and neck dysplasia: (A) p53, p63, and p73; (B) SOX-2 and OCT-4; (C) AP-1 and C-Jun; (D) BRD-4, YAP, TAZ, and TEAD. Created with BioRender.
Figure 4. Transcriptional regulators in head and neck dysplasia: (A) p53, p63, and p73; (B) SOX-2 and OCT-4; (C) AP-1 and C-Jun; (D) BRD-4, YAP, TAZ, and TEAD. Created with BioRender.
Cancers 14 05581 g004
Figure 5. Hippo pathway with Human Papilloma Virus E6/E7 Oncoproteins. Adapted from “The Hippo Tumor Suppressor Pathway” and “Hippo Pathway in Mammals”, by BioRender.com (2022) (https://app.biorender.com/biorender-templates (accessed on 10 August 2022)).
Figure 5. Hippo pathway with Human Papilloma Virus E6/E7 Oncoproteins. Adapted from “The Hippo Tumor Suppressor Pathway” and “Hippo Pathway in Mammals”, by BioRender.com (2022) (https://app.biorender.com/biorender-templates (accessed on 10 August 2022)).
Cancers 14 05581 g005
Table 1. Histopathologic markers of malignant transformation in head and neck dysplasia.
Table 1. Histopathologic markers of malignant transformation in head and neck dysplasia.
Biomarker CategoryMember (s)Physiologic FunctionRole in Tumorigenesis
Extracellular and Cell-Surface
Extracellular DegradationMMP-2, MMP-9Type IV Collagenase
  • Tumor invasion
Cell MotilityPodoplaninTransmembrane Glycoprotein
  • Tumor invasion
Cell-Cell AdhesionClaudinTight Junction Protein
  • Migration and MMP induction
JAM-ATight Junction Protein
  • EMT
E-CadherinAdherens Junction Protein
  • EMT
Solute TransportGLUT-1, GLUT-4Glucose Transporter
  • Cellular energy supply
Cancer Stem Cell MarkersCD44Transmembrane Glycoprotein
  • Proliferation
  • Tumor invasion
CD133Transmembrane Glycoprotein
  • Membrane organization
  • Signal transduction
  • Glucose metabolism
  • MMP induction
Cytosolic Markers
Metabolic Regulators ALDH-1Phase-I Oxidase
  • Induction of cell “stemness”
  • EMT
Molecular Chaperones HSP70, HSP90Heat Shock Proteins
  • Regulation of protein folding, transport, and repair
Mitosis RegulatorsStathminCytoskeleton Phosphoprotein
  • Regulation of entry into and exit from mitosis
Apoptosis RegulatorsBcl-2Inhibitor of Apoptosis
  • Inhibition of mitochondrial outer membrane permeabilization and apoptosis
SurvivinInhibitor of Apoptosis
  • Inhibition of caspases
Nuclear Markers
Cell Cycle Regulatorsp53
p63
p16
Cyclin D1
pRb
MDM2
Tumor Suppressor
Tumor Suppressor
Tumor Suppressor
Inhibitor of pRb
Tumor Suppressor
E3 Ubiquitin Ligase
  • Uncontrolled proliferation
Transcriptional RegulatorsC-JunAP-1 Transcription Factor
  • Cell proliferation
SOX-2/OCT-4Reprogramming Transcription Factors
  • Self-renewal, de-differentiation
YAP/TAZHippo Pathway Mediator
  • Cell proliferation and migration
DNA Replication and
Repair Regulators
hTERTTelomerase Protein
  • Maintains telomere length
  • Immortalization of tumor cells
MCM2Replication Initiation Factor
  • Immortalization of tumor cells
MutSα
MutLα
DNA Mismatch Repair Protein
  • Reduced genomic integrity
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ranganath, K.; Feng, A.L.; Franco, R.A.; Varvares, M.A.; Faquin, W.C.; Naunheim, M.R.; Saladi, S.V. Molecular Biomarkers of Malignant Transformation in Head and Neck Dysplasia. Cancers 2022, 14, 5581. https://doi.org/10.3390/cancers14225581

AMA Style

Ranganath K, Feng AL, Franco RA, Varvares MA, Faquin WC, Naunheim MR, Saladi SV. Molecular Biomarkers of Malignant Transformation in Head and Neck Dysplasia. Cancers. 2022; 14(22):5581. https://doi.org/10.3390/cancers14225581

Chicago/Turabian Style

Ranganath, Kushi, Allen L. Feng, Ramon A. Franco, Mark A. Varvares, William C. Faquin, Matthew R. Naunheim, and Srinivas Vinod Saladi. 2022. "Molecular Biomarkers of Malignant Transformation in Head and Neck Dysplasia" Cancers 14, no. 22: 5581. https://doi.org/10.3390/cancers14225581

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop