Next Article in Journal
Editorial for the Special Issue: Pathophysiology of Chronic Kidney Disease and Its Complications
Next Article in Special Issue
Predictive Value and Therapeutic Significance of Somatic BRCA Mutation in Solid Tumors
Previous Article in Journal
Mitochondria-Targeted Catalase Does Not Suppress Development of Cellular Senescence during Aging
Previous Article in Special Issue
The Potential of Extracellular Matrix- and Integrin Adhesion Complex-Related Molecules for Prostate Cancer Biomarker Discovery
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current and Emerging Diagnostic, Prognostic, and Predictive Biomarkers in Head and Neck Cancer

by
Hänel W. Eberly
1,
Bao Y. Sciscent
1,
F. Jeffrey Lorenz
1,
Eleni M. Rettig
2 and
Neerav Goyal
1,*
1
Department of Otolaryngology Head and Neck Surgery, Penn State Milton S. Hershey Medical Center, Penn State College of Medicine, Hershey, PA 17033, USA
2
Department of Otolaryngology Head and Neck Surgery, Brigham and Women’s Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02108, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(2), 415; https://doi.org/10.3390/biomedicines12020415
Submission received: 8 January 2024 / Revised: 31 January 2024 / Accepted: 2 February 2024 / Published: 10 February 2024
(This article belongs to the Special Issue Feature Reviews in Cancer Biomarkers)

Abstract

:
Head and neck cancers (HNC) are a biologically diverse set of cancers that are responsible for over 660,000 new diagnoses each year. Current therapies for HNC require a comprehensive, multimodal approach encompassing resection, radiation therapy, and systemic therapy. With an increased understanding of the mechanisms behind HNC, there has been growing interest in more accurate prognostic indicators of disease, effective post-treatment surveillance, and individualized treatments. This chapter will highlight the commonly used and studied biomarkers in head and neck squamous cell carcinoma.

1. Introduction

Head and neck cancer (HNC) ranks as the seventh most common cancer worldwide and is responsible for over 660,000 new diagnoses each year [1]. Squamous cell carcinomas, which arise from the epithelial lining of the oral cavity, pharynx, and larynx, comprise approximately 90% of HNCs [1,2,3,4]. The epidemiology of HNC is changing worldwide due to decreasing smoking rates and the rise of HPV-positive tumors, which are primarily of oropharyngeal origin [5,6,7,8]. Despite advances in treatment, there has been a gradual increase in overall mortality, with five-year survival rates of approximately 50–85%, depending on the type and location of the tumor [1,5,6]. Treatment of HNC confers significant morbidity and impacts the quality of life [9,10,11,12].
Current therapies for HNC require a multimodal approach that may include surgical resection, radiation therapy, and/or systemic therapy [2,9,13]. With the growing understanding of the mechanisms behind HNC, there has been interest in more accurate prognostic indicators and effective post-treatment surveillance, as well as alternative individualized treatments. In this chapter, we review the existing literature regarding molecular biomarkers for various types of HNC.
This article reviews common diagnostic, prognostic, and predictive biomarkers, with an emphasis on viral-mediated vs. non-viral mediated disease. The role of human papillomavirus (HPV) has been increasingly recognized in the pathophysiology of oropharyngeal squamous cell carcinoma (OPSCC), such that two distinct disease entities are now recognized: HPV-negative disease associated with risk factors such as tobacco and alcohol use, and HPV-positive disease [2,14]. In addition to HPV, the Epstein–Barr virus (EBV) has also been recognized as playing a role in various head and neck cancers, most notably nasopharyngeal carcinoma [15,16].
Diagnostic biomarkers can confirm the presence of a disease or identify patients with subtypes of a disease [17]. Prognostic biomarkers predict a cancer’s outcomes irrespective of any specific treatment administered and reflect the intrinsic aggressiveness of the malignancy. In contrast to predictive biomarkers, the presence of a prognostic biomarker does not affect the treatment benefit [18]. Predictive biomarkers predict the outcome of a particular treatment, often through a comparison between two treatment approaches [18]. These biomarkers may be especially relevant when designing clinical trials, as they offer insights into identifying patient cohorts that may benefit most from a treatment [19]. There is also overlap between prognostic and predictive biomarkers, so the relevant biomarkers in these categories have been combined. A list of the mentioned biomarkers is available in Table 1.

2. HPV-Driven HNCs

Human papillomavirus (HPV) is a primary cause for most (70–90%) OPSCCs and some non-OPSCCs in the United States [20,21]. HPV-positive disease has been shown to have a more favorable response to treatment and improved survival when compared to HPV-negative disease [2,22]. HPV16 accounts for most cases [2]. Methods for discerning HPV-driven oncogenesis in tumor tissue are varied, with emerging evidence that the prognosis differs according to how HPV-positivity is defined. In addition, several HPV-specific minimally invasive biomarkers have recently been identified, leading to an increased interest in early diagnosis and screening [2].

2.1. Direct vs. Indirect HPV Testing in Tumor Tissue

HNCs may be evaluated for the presence of HPV-mediated disease via direct methods, e.g., the detection of HPV DNA or mRNA expressed in tumor tissues, or via indirect methods, e.g., the identification of p16Ink4a (p16) expression via immunohistochemistry.
P16 is a known biomarker of HPV oncogenesis that is considered an indirect marker of HPV-positive disease in OPSCC [23]. It is a tumor-suppressor protein and cell-cycle regulator that is overexpressed as a result of HPV-driven oncogenesis and detected via immunohistochemical evaluation, whereby at least 70% nuclear and cytoplasmic expression is considered p16-positive. Importantly, p16 is also overexpressed as a result of other, HPV-independent processes and thus is only considered a surrogate for HPV positivity in the oropharynx, where the prevalence of HPV is high and thus the positive predictive value is high. Guidelines by the College of American Pathologists recommend that p16 testing should be performed on oropharyngeal tumor tissue specimens based on previous literature on this biomarker’s utility as an independent predictor of OPSCC prognosis, its widespread availability, and exemplary performance on specimen samples. However, this recommendation only extends to OPSCC, and p16 testing is not recommended for other tumor types including neuroendocrine or salivary gland tumors [24].
Direct methods of HPV detection in tumor tissues include ISH or PCR for HPV DNA or mRNA. The detection of ‘transcriptionally active’ HPV refers to ISH for HPV E6 or E7 mRNA; ISH for HPV DNA is not recommended [24]. These methods are HPV genotype-specific and are less readily available than p16 IHC. Importantly, some evidence suggests that the prognosis for HPV+ OPSCC varies by HPV genotype, suggesting that type-specific methods of HPV detection may be preferred in the future [25].
Up to 20% of patients with p16-positive tumors test negative for HPV DNA or RNA [21]. Other examples of discordant cases exist, where p16-positive patients are HPV-negative and p16-negative patients are HPV-positive. A recent large study of 7654 patients found that discordance between p16 and HPV status negatively impacts prognosis, leading the authors to recommend expanding indications for direct HPV testing [22]. Overexpression of p16 in HPV- tumors appears to happen due to differing mechanisms, necessitating the use of various techniques for detection and the consideration of factors unrelated to HPV status. Mehanna et al. found that when using p16 IHC alone for HPV status determination, approximately 8% of p16-positive patients would be incorrectly classified as having an HPV+ tumor [22]. The authors concluded that while routine HPV and p16 evaluation should be conducted in OPSCC clinical trials and in clinical settings where more accurate counseling is wanted, the classification of patients with OPSCC based on p16 status alone is still inadequate in routine clinical practice [22].

2.2. Blood- and Saliva-Based HPV Biomarkers

Antibodies to HPV E6 antigens, especially the E6 protein, are linked to an increased risk of developing oropharyngeal cancer and appear in blood collected up to several decades before the onset of disease [20,26]. At the time of diagnosis, HPV16-E6 seropositivity is a highly sensitive [27,28] and specific [26,27] marker for HPV-positive OPSCC. HPV16-E6 seropositivity is generally stable, and does not have a strong association with the response to treatment or recurrence [20].
Circulating tumor HPV DNA (ctHPVDNA), referring to fragments of DNA shed from tumor cells into the blood, has been identified using highly sensitive PCR or next-generation sequencing techniques in around 90% of patients with HPV-positive OPSCC [29,30]. It is highly specific for malignancy, and is also dynamic, varying with the burden of disease and the response to treatment due to its short half-life, and is under study for use in the post-treatment surveillance of HPV+ OPC [28]. These properties make ctHPVDNA a promising clinical tool that is under study for use in the diagnosis, treatment, and surveillance of HPV-positive OPSCC.
ctHPVDNA has been most extensively studied in the post-treatment surveillance setting, where it has a high (~95–100%) positive and negative predictive value for disease recurrence and may complement PET/CT to improve the accuracy of surveillance compared with the current standard of care [28,31,32,33,34,35,36]. During treatment, dynamic changes in ctHPVDNA are being studied to guide therapy in real-time, including dose adjustment during definitive radiation (e.g., NCT04900623). Among patients treated with surgery, postoperatively detectable ctHPVDNA is correlated with a risk of residual disease and extra-nodal extension, suggesting it is a high-risk feature that warrants consideration of adjuvant therapy [35,37]. In the prediagnostic setting, ctHPVDNA has been detected In blood collected several years prior to the diagnosis of HPV+ OPSCC in a subset of patients [38]. Ongoing research will elucidate whether the incorporation of ctHPVDNA into HPV-positive OPSCC diagnosis, treatment and surveillance paradigms will measurably improve patient outcomes.
Oncogenic HPV DNA found in oral rinses has also been associated with treatment response and recurrence following treatment but has lower sensitivity compared to ctHPVDNA and is subject to more variability [39,40]. Bystander infections or a patient’s inability to clear viral cells following infection are potential confounders, making this method less specific compared to measuring circulating tumor HPV DNA [40,41].

2.3. HPV Tumor Status in Non-Oropharynx HNC Sites

HPV is detected in a subset of HNCs outside of the oropharynx (larynx, oral cavity, hypopharynx, nasopharynx); however, the prevalence is much lower than in the oropharynx and the clinical significance is unclear [42]. While the prevalence of HPV positivity has been increasing in laryngeal and oral cavity cancer, there does not seem to be an association between HPV status and survival [43,44]. At this time, testing for HPV in non-oropharynx HNCs is not currently recommended [45]. However, recent research has highlighted the utility of high-risk HPV in understanding the prognosis of sinonasal cancer, with high-risk HPV being associated with a better prognosis in these patients [43,46,47]. Several recent papers have found that the prevalence of HPV positivity in sinonasal cancer has been increasing, with favorable survival being associated with HPV status [48,49].

3. Prognostic and Predictive Molecular Markers in HPV-Positive Tumors

3.1. HPV Viral Integration

A more recent area of focus in HPV-positive head and neck cancer is HPV viral integration into the host cellular genome. Viral integration is associated with higher transcription rates of E6 and E7 and the progression of carcinoma in cervical cancers [50,51], and has been appreciated in head and neck carcinoma. However, the association between viral integration and clinical outcomes is not well understood [52], and current evidence is often conflicting, with variability in methods for detecting viral integration. While there is some evidence for the epigenetic upregulation of regions around the integrated HPV genome in the host, the clinical significance of these findings is under investigation [53].

3.2. HPV Subtype

The HPV genotype is not a widely used measure of risk stratification for head and neck squamous cell carcinoma. This is largely due to the relative homogeneity of the HPV genotype in North America, with only around 8–14% of oropharyngeal cancers having genotypes other than HPV-16 [54,55]. However, emerging literature suggests that tumors associated with HPV-16 tend to have better prognoses compared to other genotypes, with a five-year overall survival of 83% versus 69% in HPV16 and HPV-non16, respectively, in a recent meta-analysis of 1310 HPV16 and 219 HPV-non16 patients [25,56,57,58]. Further studies should be carried out to examine the HPV genotype and its role in prognosis and treatment decision-making.

3.3. Estrogen Receptor Positivity

Recent studies have examined tumor estrogen receptor alpha (ERa) positivity as a biomarker for improved overall survival and recurrence-free survival in HPV-positive oropharyngeal cancer [52,53]. While ERa is a biomarker and therapeutic target for breast cancer, its utility in head and neck squamous cell carcinoma has only recently been investigated. Several studies have found an improved prognosis in patients with Era-positive oropharyngeal squamous cell carcinoma treated with chemoradiation, even after accounting for various clinical risk factors [59,60,61]. While there are no current studies investigating ERa as a therapeutic target in head and neck cancer, its presence in tumors may help guide treatment selection and de-intensification [59].

3.4. Apolipoprotein B mRNA Editing Enzyme, Catalytic Polypeptide

The apolipoprotein B mRNA editing enzyme catalytic polypeptide-like (APOBEC) family of cytidine deaminases is made up of aberrantly-acting DNA-modifying enzymes that have been linked to DNA mutations and tumor formation [62,63]. APOBEC activation has a role in the immune system’s response to viruses including HPV and EBV [64,65]. Recent studies have shown higher APOBEC activity in HPV-positive HNC [66,67], as well as a correlation between the mutational burden of HPV-positive head and neck cancer with APOBEC enrichment [14,68,69]. Patients with HNC have an improved treatment response to chemotherapy and immunotherapy, with a higher expression of certain A3H or A3G proteins being correlated with increased survival [67,69,70,71].

4. Role of EBV in Nasopharyngeal Cancer

The Epstein–Barr virus (EBV) is closely linked to the pathogenesis of nasopharyngeal cancer, a distinct form of HNC endemic to the Asian population. EBV is a ubiquitous virus that infects a majority of the global population, usually establishing a lifelong latent infection in B lymphocytes. In the context of NPC, EBV plays a pivotal role in oncogenesis by promoting genetic alterations, immune evasion, and the dysregulation of cellular signaling pathways [72,73]. The complex interplay of viral and host factors involved in EBV-associated nasopharyngeal carcinoma allows for the use of diagnostic, prognostic, and predictive biomarkers.

5. Biomarkers for the Screening and Diagnosis of EBV-Positive Nasopharyngeal Cancer

Serologic testing for antibodies to EBV antigens has been used to evaluate patients with suspected nasopharyngeal carcinoma, and for the screening of endemic populations [73,74]. In particular, the EBNA1 peptide has been shown to have optimal performance characteristics when detecting asymptomatic nasopharyngeal carcinoma. Using optimized threshold values, the sensitivity of EBNA1 IgA has been shown to be 80–85.7% [73,74], with a specificity of 51.2% in one study [73]. A novel biomarker, anti-BNLF2b total antibody (P85Ab), is a promising biomarker found to have a higher sensitivity, specificity, and positive predictive value compared to the standard two-antibody-based screening method (EBV nuclear antigen 1 IgA and EBV viral capsid antigen IgA). The positive predictive value was also increased, which may improve the cost-effectiveness of screening [75]. While promising, further prospective studies and randomized controlled trials are necessary to further guide the use of these biomarkers [76].
The detection of cancer-related EBV DNA in the bloodstream has been confirmed as a reliable indicator for nasopharyngeal carcinoma [72]. EBV DNA has been detected in 96% of patients with nasopharyngeal carcinoma using quantitative real-time polymerase chain reaction and has shown utility in determining the prognosis and detecting residual disease [77,78]. Several studies have also examined the utility of using EBV DNA in plasma to screen for asymptomatic cases of nasopharyngeal carcinoma at an early disease stage, with promising results [72,79,80,81]. In a prospective study of asymptomatic patients, a screening test that marked patients as positive if they had tested positive for EBV DNA twice, four weeks apart, showed a greater than 97% sensitivity, specificity, and negative predictive value. Of those with detected nasopharyngeal carcinoma, 70% had stage 1 or 2 disease, which is significantly higher than historical cohorts that showed a greater percentage of late-stage disease [74].

6. Prognostic and Predictive Molecular Markers in EBV Positive Nasopharyngeal Cancer

Patients with nasopharyngeal carcinoma have been shown to have high serologic titers against viral antigens, as discussed above. Recent studies have focused on circulating EBV DNA as a non-invasive and clinically useful biomarker for the prognosis of EBV, with studies showing a higher risk of mortality, recurrence, and metastasis associated with higher levels of pre-treatment EBV DNA compared to those with low levels [82,83,84]. However, many of these studies are limited to single-institution studies or smaller cohorts, with little variation in ethnic variety that can influence prognosis. Furthermore, patient management was not standardized between studies, which may influence the results.

7. Prognostic and Predictive Molecular Markers in Virus-Negative HNCs

Many HNCs are not driven by viral oncogenesis, but rather chemical carcinogens, most commonly tobacco [85]. The clinical and molecular profiles of these tumors are distinct from those of virally driven cancers, and they are considered to be separate disease entities. Clinically, HNCs that are not associated with HPV or EBV occur in all subsites of the head and neck, most commonly in middle-aged and older men who smoke. While the incidence of this form of HNC is decreasing, it tends to have a poorer prognosis. There are a number of biomarkers that are utilized in virus-negative tumors with diagnostic, prognostic, and predictive significance. These are presented beginning with those that are currently used in clinical practice followed by those that are still under investigation.

7.1. TP53 and P53

The expression of the tumor-suppressor protein p53 is altered by mutations in the TP53 gene, which are prevalent in various cancers [85]. Previous studies have found that aberrant forms of p53 proteins, in conjunction with TP53 mutations, can confer increased resistance of HNC to chemotherapy [86,87]. The p53 tumor-suppressor protein has oncoprotective functions, including the modulation of reactive oxygen species, the regulation of the G1/S phase of the cell cycle, and the induction of apoptosis [86,88]. Levels of p53 overexpression in HNC vary due to differing risk factors and pathogenesis across the world [89,90,91]. The loss of p53 expression or the overexpression of a mutant p53 protein are associated with a poor prognosis, increased extranodal extension, higher tumor grades, and a higher rate of recurrence in HNC [88,92]. Treatment strategies to restore the wild-type function of p53 or induce the degradation of mutant p53 proteins are challenging because they require knowledge of a specific p53 mutation to be effective [87].

7.2. EGFR

Epidermal growth factor receptor (EGFR) is overexpressed in a variety of solid tumors, including approximately 80% of HNC [93]. Heightened EGFR expression is associated with a poorer prognosis and a more aggressive tumor presentation [93,94,95]. Multiple treatment approaches targeting EGFR have been explored, including the extracellular domain, intracellular domain, and at the genetic level [96]. For example, EGFR-specific monoclonal antibodies bind to the receptor and block ligand binding, resulting in the blockade of EGFR phosphorylation and downstream signaling. The use of cetuximab in combination with radiotherapy in patients with locoregionally advanced HNC demonstrated an improvement in the duration of locoregional control [97]. Various combinations have been attempted, including the use of cetuximab as a monotherapy [98] or in combination with cisplatin and radiotherapy [99]. Tyrosine kinase inhibitors are another class of drugs that blocks tyrosine kinase enzymes, thus reducing downstream signaling in this pathway [100]. Various treatment methods and combinations with chemo- and radiotherapy have been attempted, similarly to EGFR-specific monoclonal antibodies [100].

7.3. PD-L1 Expression

Programmed death-ligand 1 (PD-L1) is a pivotal immune checkpoint regulator expressed in various immune cells [101]. In HNC, the interaction of PD-L1 and its receptor, programmed cell death protein 1 (PD-1), leads to the suppression of immune responses, and particularly the inhibition of T cell activation. This interaction effectively hampers the body’s natural ability to recognize and eliminate cancer cells. The upregulation of PD-L1 in the tumor microenvironment creates an immunosuppressive shield, allowing cancer cells to evade detection and destruction by the immune system. Consequently, this molecular interplay promotes tumor progression [102].
In the context of therapeutic interventions, efforts have been directed toward disrupting the PD-1/PD-L1 axis to restore effective antitumor immune responses. Anti-PD-1 antibodies, such as nivolumab and pembrolizumab, block the interaction between PD-L1 and PD-1, thereby unleashing the immune system to mount a more robust attack against cancer cells. This targeted immunotherapy demonstrated improved survival outcomes for patients with HNC [103,104]. Consequently, immunotherapy now plays a central role in the treatment of many HNCs, marking a significant shift in the treatment landscape.
However, the nuanced role of PD-L1 in HNC treatment extends beyond being a therapeutic target. Other studies have examined the prognostic value of PD-L1, and have found it to be an independent risk factor for oral squamous cell carcinoma, with increased expression being associated with distant metastasis and poor overall survival [105,106]. However, other studies found that the increased expression of PD-L1 was associated with longer disease-free survival in high-risk head and neck squamous cell carcinoma [107]. Limitations to the use of PD-L1 include its unknown significance in head and neck squamous cell carcinoma apart from oral cavity cancer, and the lack of data on the PD-L1 ligand in head and neck squamous cell carcinoma [108].

7.4. Beta 2-Microglobulin

β 2-microglobulin ( β 2M) is a component of the major histocompatibility complex (MHC) class I and holds significance in assessing tumor status in various cancers [109,110,111]. While β 2M is typically present at low physiological levels, elevated levels have been observed in conditions like renal failure and certain malignancies, including oral SCC. This suggests that elevated β 2M could serve as an effective diagnostic marker [112]. Levels may be assessed in the blood as well as saliva, making fast and cost-effective screening possible [113,114]. β 2M levels have been studied not only for diagnostic purposes but also as a potential marker for tumor progression, metastasis, and survival in patients with oral cavity SCC [110,115]. Most studies examining β 2M are limited by small sample sizes, and β 2M has not been sufficiently validated for use as a biomarker in various clinical settings [113].

7.5. Hypoxia Markers

Hypoxia, defined as a mismatch between oxygen supply and demand [116] has been studied in the context of solid tumors including head and neck squamous cell carcinoma [117]. Tumor growth intensifies the requirement for sufficient cellular oxygenation, which often exacerbates hypoxia [118]. Hypoxia is linked to tumor progression, as it modifies the free radical chemistry of tumors and contributes to a more aggressive phenotype. As a result, there is a decreased sensitivity to radiotherapy and some forms of chemotherapy [116,119]. In addition, hypoxia can lead to increased genetic instability, mutations, an antitumor immune response, and the evolution of genetically hypoxia-resistant phenotypes [111,120,121].
The most important endogenous biomarkers for hypoxia include the hypoxia-inducible factor (HIF)-1a and -2a pathways, glucose transporter (GLUT)-1, CA-IX, and osteopontin (OPN), among others. In a systematic review, the expression of endogenous hypoxia biomarkers was commonly observed and associated with poorer survival and locoregional control in most studies, along with unfavorable clinicopathological tumor characteristics [116].

7.6. Neutrophil to Lymphocyte Ratio

Chronic inflammation is implicated in tumorigenesis across various cancers, including HNC [122,123,124]. The neutrophil-to-lymphocyte ratio (NLR) characterizes the inflammatory response to cancer and has emerged as predictive of outcomes in solid tumors [125]. Studies consistently demonstrate that a high pre-treatment NLR is associated with poor overall and progression-free survival as well as an increased likelihood of tumor recurrence [126,127]. Interestingly, a recent study showed that a very low pre-NLR seemed to have a similar effect on prognosis [128]. Post-treatment NLR has also been utilized in a similar fashion to pre-treatment values [122,129]. Interestingly, a recent study showed that a very low pre-operative NLR may yield a similar negative effect on prognosis [105]. Given the easy availability, objectivity, and cost-effectiveness of NLR, it may be advantageous to other biomarkers [128].

7.7. PTEN

PTEN is a tumor suppressor that negatively regulates the PI3K–AKT–mTOR pathway and various processes related to cell growth and proliferation [130]. PTEN mutations have been detected in numerous tumor types, including breast, blood/lymph, central nervous system, and thyroid, among others [131,132]. Assessments of the effect of PTEN expression on the prognosis of HNC yield mixed results, with some demonstrating that increased PTEN expression is associated with a favorable outcome following treatment [130,133], while others suggest that heightened expression is linked to poor outcomes [134]. The current literature highlights the challenge of making robust comparisons between groups due to the diverse methodologies employed for IHC scoring of PTEN.

7.8. Cyclin D1

The G1 checkpoint, a crucial determinant governing the progression of cell division [62], is tightly regulated by a group of proteins including cyclins, cyclin-dependent kinases (CDKs), and CDK inhibitors [135]. Cyclin D1 has been implicated in the development of tumors of the esophagus, ovary, breast, colon, lung, and head and neck [136]. Previous studies have established associations between the overexpression of cyclin D1 and the development of regional lymph node metastases in HNC [137,138,139], as well as decreased disease-free and overall survival [104,140,141,142].

7.9. ERCC1

Excision repair cross-complementation group 1 (ERCC1) plays an important role in the nucleotide excision repair pathway of DNA repair [143]. ERCC1 gene polymorphisms have been associated with an increased risk of certain cancers, including nasopharyngeal carcinoma [144], lung cancer [145], melanoma [146], and pancreatic cancer [147]. Increased expressions of ERCC1 and related polymorphisms are associated with decreased progression-free survival [148], increased susceptibility to nasopharyngeal carcinoma [144], increased rates of recurrence in patients treated with concurrent chemoradiotherapy [149], and increased resistance to radiotherapy [150]. These studies highlight a possible role for more aggressive treatment in these patients and may allow clinicians to predict which patients might benefit from platinum-based chemotherapy [151].

7.10. Cathepsin D

Cathepsin D is a lysosomal enzyme found throughout the body’s cells. It has been found to be overexpressed and/or abnormally processed in various cancer cells and is thought to have a role in local tumor invasion and metastasis [152]. While cathepsin D has been studied mostly in the context of breast cancer patients, several authors have examined its role in head and neck squamous cell carcinoma [152,153,154,155]. Several studies have found that cathepsin D gene expression was associated with metastasis in head and neck squamous cell carcinoma [154,155,156].

7.11. Bcl-2

Bcl-2 is a key mitochondrial protein that regulates apoptosis, with overexpression of the protein neutralizing pro-apoptotic proteins and inhibiting apoptosis [157]. In HNC, the role of Bcl-2 as a prognostic and predictive biomarker remains equivocal, with conflicting evidence. Several studies have found Bcl-2 overexpression to be an independent risk factor for HNC [158,159], as well as an independent poor prognostic factor [157,160]. However, others have contrarily reported that a negative Bcl-2 expression correlates with a worse prognosis [161,162] or indicated no association between Bcl-2 expression and prognosis or tumor aggressiveness [162,163,164].

7.12. Tumor Budding and Epithelial–Mesenchymal Transition

Tumor budding, defined as the presence of single tumor cells or groups of tumor cells at the tumor margin [165], has been established as a prognostic factor in colorectal cancer, pancreatic cancer, esophageal cancer, and breast cancer [166,167,168]. In HNCs, tumor budding has been described as a prognostic factor for lymph node metastasis and early-stage OPSCC [169,170]. Tumor budding has also been studied in association with epithelial–mesenchymal transition, which involves the transition of an epithelial cell to a mesenchymal cell phenotype [171]. This transition, thought by some to play a role in the initiation of tumor budding, is associated with the conversion of cancer cells into a more invasive and metastatic phenotype. This enhanced cellular state enables cancer migration to other regions of the body. Tumor budding cells have been found to express various molecules such as ZEB1, ZEB2, E-cadherin, and SNA1, which are characteristic of epithelial–mesenchymal transition [167,172]. Some studies suggest that tumor budding and epithelial–mesenchymal transition are independent processes in OPSCC progression [173], and others have found significant associations between the two processes [174,175].

7.13. DNA Methylation

Known contributors to OPSCC such as tobacco, alcohol abuse, and HPV positivity confer increased risk through epigenetic changes such as DNA methylation [176,177,178]. DNA methylation occurs through the addition of a methyl group to a carbon of cytosine, forming 5-methylcytosine [177]. This change in DNA structure leads to a change in the activity of transcription factors and the mobility of various proteins [179]. Tobacco and alcohol use have been associated with hypomethylation in HNC, while HPV-positive tumors have been associated with hypermethylation [179]. Furthermore, differing degrees of methylation have been linked to different tumor locations, and various sites of methylation have been shown to be correlated with more aggressive disease progression [177,180,181]. There is a need for further studies on this topic, given the rapidly expanding identification of new biomarkers and the need to transfer this knowledge to clinical practice.

7.14. MicroRNAs

MicroRNAs (miRNA), molecules of non-coding DNA that regulate gene expression and dysregulation [182], have been associated with the initiation and progression of malignancy [183]. Studies have shown them to be prognostic biomarkers in B-cell lymphomas [184], lung cancers [185], and hepatocellular carcinoma [186]. This is a biomarker of interest in HNC because the miRNA expression profiles have been found to differ between HPV-positive and -negative HNCs [187,188]. Different miRNA signatures have been found to improve the risk stratification of HPV-positive and -negative tumors [189,190]. One study of HPV-negative HNC patients who received chemoradiotherapy reported that a five-miRNA signature (hsa-let-7g-3p, hsa-miR-6508-5p, hsa-miR-210-5p, hsa-miR-4306, and hsa-miR-7161-3p) was a strong prognostic indictor for recurrence and survival. Other studies are also investigating its utility in the evaluation of treatment response in HNCs [189,191].

8. Emerging Biomarkers

Several other emerging biomarkers have been studied in recent decades regarding their role as prognostic biomarkers in HNC. These include beta tubulin isotypes, the proteasomal degradation protein PSMD14, the T cell regulator SSP1, and the matrix metalloproteinase (MMP) family of enzymes that degrade the extracellular matrix [192,193,194,195]. Beta tubulin II and III have been implicated in predicting outcomes of taxane and cisplatin-based chemotherapy in HNC with promising results [151,193,196]. However, their utility in a clinical setting has yet to be established and further research needs to be conducted. PSMD14 is another protein that is associated with poor progression and an advanced tumor stage [194,197], with some research being conducted on its potential role as a therapeutic target in the Akt pathway [197]. SSP1 has been studied in terms of its ability to modulate the immunosuppressive mechanisms of tumor cells, especially in patients treated with Nivolumab [195]. The MMP family of enzymes has also been shown to be involved in immune cell infiltration in HNC, specifically MMP14, MMP16, and MMP19 [198]. Future studies should be conducted to standardize the measurement of these biomarkers, verify their expression levels, and further establish their clinical significance.

9. Conclusions

Biomarkers provide an opportunity to advance head and neck cancer care by potentially leading to earlier diagnosis, improving prognostication, aiding treatment decision making, and helping identify recurrences post-treatment. Several such biomarkers are now routinely utilized in HNC care. These include p16, HPV, and ctHPVDNA in the HPV-positive population; EBV and anti-EBV antibodies in the EBV-positive population; and p53, EGFR, and PDL1 in the non-virally induced cancer population; with a myriad of other biomarkers being studied.
As medicine evolves to become more individualized, there arises a heightened demand for reliable and clinically applicable biomarkers to contribute to the effective management of patients suffering from HNC.

Author Contributions

Conceptualization, H.W.E. and N.G.; investigation, H.W.E.; writing—original draft preparation, H.W.E.; writing—review and editing, B.Y.S., E.M.R., N.G. and F.J.L.; supervision, N.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

We would like to acknowledge Caia Hypatia for support in manuscript preparation and submission.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Gormley, M.; Creaney, G.; Schache, A.; Ingarfield, K.; Conway, D.I. Reviewing the Epidemiology of Head and Neck Cancer: Definitions, Trends and Risk Factors. Br. Dent. J. 2022, 233, 780–786. [Google Scholar] [CrossRef]
  2. Rettig, E.M.; Sethi, R.K.V. Cancer of the Oropharynx and the Association with Human Papillomavirus. Hematol./Oncol. Clin. N. Am. 2021, 35, 913–931. [Google Scholar] [CrossRef] [PubMed]
  3. Dive, A.M.; Bodhade, A.S.; Mishra, M.S.; Upadhyaya, N. Histological Patterns of Head and Neck Tumors: An Insight to Tumor Histology. J. Oral Maxillofac. Pathol. 2014, 18, 58–68. [Google Scholar] [CrossRef] [PubMed]
  4. Pai, S.I.; Westra, W.H. Molecular Pathology of Head and Neck Cancer: Implications for Diagnosis, Prognosis, and Treatment. Annu. Rev. Pathol. Mech. Dis. 2009, 4, 49–70. [Google Scholar] [CrossRef] [PubMed]
  5. Bosetti, C.; Carioli, G.; Santucci, C.; Bertuccio, P.; Gallus, S.; Garavello, W.; Negri, E.; La Vecchia, C. Global Trends in Oral and Pharyngeal Cancer Incidence and Mortality. Int. J. Cancer 2020, 147, 1040–1049. [Google Scholar] [CrossRef]
  6. Warnakulasuriya, S. Global Epidemiology of Oral and Oropharyngeal Cancer. Oral Oncol. 2009, 45, 309–316. [Google Scholar] [CrossRef]
  7. Conway, D.I.; Purkayastha, M.; Chestnutt, I.G. The Changing Epidemiology of Oral Cancer: Definitions, Trends, and Risk Factors. Br. Dent. J. 2018, 225, 867–873. [Google Scholar] [CrossRef]
  8. Fakhry, C.; Krapcho, M.; Eisele, D.W.; D’Souza, G. Head and Neck Squamous Cell Cancers in the United States Are Rare and the Risk Now Is Higher among White Individuals Compared with Black Individuals. Cancer 2018, 124, 2125–2133. [Google Scholar] [CrossRef]
  9. van Rooij, J.A.F.; Roubos, J.; Vrancken Peeters, N.J.M.C.; Rijken, B.F.M.; Corten, E.M.L.; Mureau, M.A.M. Long-Term Patient-Reported Outcomes after Reconstructive Surgery for Head and Neck Cancer: A Systematic Review. Head Neck 2023, 45, 2469–2477. [Google Scholar] [CrossRef]
  10. Dahill, A.; Al-Nakishbandi, H.; Cunningham, K.B.; Humphris, G.M.; Lowe, D.; Rogers, S.N. Loneliness and Quality of Life after Head and Neck Cancer. Br. J. Oral Maxillofac. Surg. 2020, 58, 959–965. [Google Scholar] [CrossRef]
  11. Buga, S.; Banerjee, C.; Salman, J.; Cangin, M.; Zachariah, F.; Freeman, B. Supportive Care for the Head and Neck Cancer Patient. Cancer Treat. Res. 2018, 174, 249–270. [Google Scholar] [CrossRef]
  12. Lane, C.; Higgins, R.C.; Goyal, N. Psychological Survivorship in Head and Neck Cancer. Semin. Plast. Surg. 2023, 37, 46–52. [Google Scholar] [CrossRef]
  13. Johnson, D.E.; Burtness, B.; Leemans, C.R.; Lui, V.W.Y.; Bauman, J.E.; Grandis, J.R. Head and Neck Squamous Cell Carcinoma. Nat. Rev. Dis. Primers 2020, 6, 92. [Google Scholar] [CrossRef] [PubMed]
  14. Gillison, M.L.; D’Souza, G.; Westra, W.; Sugar, E.; Xiao, W.; Begum, S.; Viscidi, R. Distinct Risk Factor Profiles for Human Papillomavirus Type 16-Positive and Human Papillomavirus Type 16-Negative Head and Neck Cancers. J. Natl. Cancer Inst. 2008, 100, 407–420. [Google Scholar] [CrossRef] [PubMed]
  15. Raab-Traub, N. Epstein-Barr Virus in the Pathogenesis of NPC. Semin. Cancer Biol. 2002, 12, 431–441. [Google Scholar] [CrossRef] [PubMed]
  16. Raab-Traub, N. Novel Mechanisms of EBV-Induced Oncogenesis. Curr. Opin. Virol. 2012, 2, 453–458. [Google Scholar] [CrossRef] [PubMed]
  17. Califf, R.M. Biomarker Definitions and Their Applications. Exp. Biol. Med. 2018, 243, 213–221. [Google Scholar] [CrossRef] [PubMed]
  18. Rahimy, E.; Gensheimer, M.F.; Beadle, B.; Le, Q.-T. Lessons and Opportunities for Biomarker-Driven Radiation Personalization in Head and Neck Cancer. Semin. Radiat. Oncol. 2023, 33, 336–347. [Google Scholar] [CrossRef]
  19. Hsieh, J.C.-H.; Wang, H.-M.; Wu, M.-H.; Chang, K.-P.; Chang, P.-H.; Liao, C.-T.; Liau, C.-T. Review of Emerging Biomarkers in Head and Neck Squamous Cell Carcinoma in the Era of Immunotherapy and Targeted Therapy. Head Neck 2019, 41 (Suppl. S1), 19–45. [Google Scholar] [CrossRef]
  20. Rettig, E.M.; Waterboer, T.; Sim, E.; Faden, D.L.; Butt, J.; Hanna, G.J.; Del Vecchio Fitz, C.; Kuperwasser, C.; Sroussi, H. Relationship of HPV16 E6 Seropositivity with Circulating Tumor Tissue Modified HPV16 DNA before Head and Neck Cancer Diagnosis. Oral Oncol. 2023, 141, 106417. [Google Scholar] [CrossRef]
  21. Chaturvedi, A.K.; Anderson, W.F.; Lortet-Tieulent, J.; Curado, M.P.; Ferlay, J.; Franceschi, S.; Rosenberg, P.S.; Bray, F.; Gillison, M.L. Worldwide Trends in Incidence Rates for Oral Cavity and Oropharyngeal Cancers. J. Clin. Oncol. 2013, 31, 4550–4559. [Google Scholar] [CrossRef]
  22. Mehanna, H.; Taberna, M.; von Buchwald, C.; Tous, S.; Brooks, J.; Mena, M.; Morey, F.; Grønhøj, C.; Rasmussen, J.H.; Garset-Zamani, M.; et al. Prognostic Implications of P16 and HPV Discordance in Oropharyngeal Cancer (HNCIG-EPIC-OPC): A Multicentre, Multinational, Individual Patient Data Analysis. Lancet Oncol. 2023, 24, 239–251. [Google Scholar] [CrossRef]
  23. Ang, K.K.; Harris, J.; Wheeler, R.; Weber, R.; Rosenthal, D.I.; Nguyen-Tân, P.F.; Westra, W.H.; Chung, C.H.; Jordan, R.C.; Lu, C.; et al. Human Papillomavirus and Survival of Patients with Oropharyngeal Cancer. N. Engl. J. Med. 2010, 363, 24–35. [Google Scholar] [CrossRef]
  24. Lewis, J.S.; Beadle, B.; Bishop, J.A.; Chernock, R.D.; Colasacco, C.; Lacchetti, C.; Moncur, J.T.; Rocco, J.W.; Schwartz, M.R.; Seethala, R.R.; et al. Human Papillomavirus Testing in Head and Neck Carcinomas: Guideline from the College of American Pathologists. Arch. Pathol. Lab. Med. 2018, 142, 559–597. [Google Scholar] [CrossRef]
  25. Shenker, R.F.; Razavian, N.B.; D’Agostino, R.B.; Mowery, Y.M.; Brizel, D.M.; Hughes, R.T. Clinical Outcomes of Oropharyngeal Squamous Cell Carcinoma Stratified by Human Papillomavirus Subtype: A Systematic Review and Meta-Analysis. Oral Oncol. 2023, 148, 106644. [Google Scholar] [CrossRef]
  26. Kreimer, A.R.; Ferreiro-Iglesias, A.; Nygard, M.; Bender, N.; Schroeder, L.; Hildesheim, A.; Robbins, H.A.; Pawlita, M.; Langseth, H.; Schlecht, N.F.; et al. Timing of HPV16-E6 Antibody Seroconversion before OPSCC: Findings from the HPVC3 Consortium. Ann. Oncol. 2019, 30, 1335–1343. [Google Scholar] [CrossRef]
  27. Holzinger, D.; Wichmann, G.; Baboci, L.; Michel, A.; Höfler, D.; Wiesenfarth, M.; Schroeder, L.; Boscolo-Rizzo, P.; Herold-Mende, C.; Dyckhoff, G.; et al. Sensitivity and Specificity of Antibodies against HPV16 E6 and Other Early Proteins for the Detection of HPV16-Driven Oropharyngeal Squamous Cell Carcinoma. Int. J. Cancer 2017, 140, 2748–2757. [Google Scholar] [CrossRef]
  28. Lang Kuhs, K.A.; Kreimer, A.R.; Trivedi, S.; Holzinger, D.; Pawlita, M.; Pfeiffer, R.M.; Gibson, S.P.; Schmitt, N.C.; Hildesheim, A.; Waterboer, T.; et al. Human Papillomavirus 16 E6 Antibodies Are Sensitive for Human Papillomavirus-Driven Oropharyngeal Cancer and Are Associated with Recurrence. Cancer 2017, 123, 4382–4390. [Google Scholar] [CrossRef] [PubMed]
  29. Hanna, G.J.; Supplee, J.G.; Kuang, Y.; Mahmood, U.; Lau, C.J.; Haddad, R.I.; Jänne, P.A.; Paweletz, C.P. Plasma HPV Cell-Free DNA Monitoring in Advanced HPV-Associated Oropharyngeal Cancer. Ann. Oncol. 2018, 29, 1980–1986. [Google Scholar] [CrossRef] [PubMed]
  30. Siravegna, G.; O’Boyle, C.J.; Varmeh, S.; Queenan, N.; Michel, A.; Stein, J.; Thierauf, J.; Sadow, P.M.; Faquin, W.C.; Perry, S.K.; et al. Cell-Free HPV DNA Provides an Accurate and Rapid Diagnosis of HPV-Associated Head and Neck Cancer. Clin. Cancer Res. 2022, 28, 719–727. [Google Scholar] [CrossRef] [PubMed]
  31. Ferrandino, R.M.; Chen, S.; Kappauf, C.; Barlow, J.; Gold, B.S.; Berger, M.H.; Westra, W.H.; Teng, M.S.; Khan, M.N.; Posner, M.R.; et al. Performance of Liquid Biopsy for Diagnosis and Surveillance of Human Papillomavirus–Associated Oropharyngeal Cancer. JAMA Otolaryngol. Head Neck Surg. 2023, 149, 971. [Google Scholar] [CrossRef]
  32. Berger, B.M.; Hanna, G.J.; Posner, M.R.; Genden, E.M.; Lautersztain, J.; Naber, S.P.; Del Vecchio Fitz, C.; Kuperwasser, C. Detection of Occult Recurrence Using Circulating Tumor Tissue Modified Viral HPV DNA among Patients Treated for HPV-Driven Oropharyngeal Carcinoma. Clin. Cancer Res. 2022, 28, 4292–4301. [Google Scholar] [CrossRef] [PubMed]
  33. Warlow, S.J.; Adamowicz, M.; Thomson, J.P.; Wescott, R.A.; Robert, C.; Carey, L.M.; Thain, H.; Cuschieri, K.; Li, L.Q.; Conn, B.; et al. Longitudinal Measurement of HPV Copy Number in Cell-Free DNA Is Associated with Patient Outcomes in HPV-Positive Oropharyngeal Cancer. Eur. J. Surg. Oncol. 2022, 48, 1224–1234. [Google Scholar] [CrossRef]
  34. Chera, B.S.; Kumar, S.; Shen, C.; Amdur, R.; Dagan, R.; Green, R.; Goldman, E.; Weiss, J.; Grilley-Olson, J.; Patel, S.; et al. Plasma Circulating Tumor HPV DNA for the Surveillance of Cancer Recurrence in HPV-Associated Oropharyngeal Cancer. J. Clin. Oncol. 2020, 38, 1050–1058. [Google Scholar] [CrossRef]
  35. O’Boyle, C.J.; Siravegna, G.; Varmeh, S.; Queenan, N.; Michel, A.; Pang, K.C.S.; Stein, J.; Thierauf, J.C.; Sadow, P.M.; Faquin, W.C.; et al. Cell-Free Human Papillomavirus DNA Kinetics after Surgery for Human Papillomavirus-Associated Oropharyngeal Cancer. Cancer 2022, 128, 2193–2204. [Google Scholar] [CrossRef]
  36. Tanaka, H.; Takemoto, N.; Horie, M.; Takai, E.; Fukusumi, T.; Suzuki, M.; Eguchi, H.; Komukai, S.; Tatsumi, M.; Isohashi, F.; et al. Circulating Tumor HPV DNA Complements PET-CT in Guiding Management after Radiotherapy in HPV-Related Squamous Cell Carcinoma of the Head and Neck. Int. J. Cancer 2021, 148, 995–1005. [Google Scholar] [CrossRef] [PubMed]
  37. Routman, D.M.; Kumar, S.; Chera, B.S.; Jethwa, K.R.; Van Abel, K.M.; Frechette, K.; DeWees, T.; Golafshar, M.; Garcia, J.J.; Price, D.L.; et al. Detectable Postoperative Circulating Tumor Human Papillomavirus DNA and Association with Recurrence in Patients With HPV-Associated Oropharyngeal Squamous Cell Carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2022, 113, 530–538. [Google Scholar] [CrossRef]
  38. Rettig, E.M.; Faden, D.L.; Sandhu, S.; Wong, K.; Faquin, W.C.; Warinner, C.; Stephens, P.; Kumar, S.; Kuperwasser, C.; Richmon, J.D.; et al. Detection of Circulating Tumor Human Papillomavirus DNA before Diagnosis of HPV-Positive Head and Neck Cancer. Int. J. Cancer 2022, 151, 1081–1085. [Google Scholar] [CrossRef] [PubMed]
  39. Fakhry, C.; Westra, W.H.; Li, S.; Cmelak, A.; Ridge, J.A.; Pinto, H.; Forastiere, A.; Gillison, M.L. Improved Survival of Patients with Human Papillomavirus-Positive Head and Neck Squamous Cell Carcinoma in a Prospective Clinical Trial. J. Natl. Cancer Inst. 2008, 100, 261–269. [Google Scholar] [CrossRef]
  40. Rettig, E.M.; Wentz, A.; Posner, M.R.; Gross, N.D.; Haddad, R.I.; Gillison, M.L.; Fakhry, C.; Quon, H.; Sikora, A.G.; Stott, W.J.; et al. Prognostic Implication of Persistent Human Papillomavirus Type 16 DNA Detection in Oral Rinses for Human Papillomavirus-Related Oropharyngeal Carcinoma. JAMA Oncol. 2015, 1, 907–915. [Google Scholar] [CrossRef]
  41. Ahn, S.M.; Chan, J.Y.K.; Zhang, Z.; Wang, H.; Khan, Z.; Bishop, J.A.; Westra, W.; Koch, W.M.; Califano, J.A. Saliva and Plasma Quantitative Polymerase Chain Reaction-Based Detection and Surveillance of Human Papillomavirus-Related Head and Neck Cancer. JAMA Otolaryngol. Head Neck Surg. 2014, 140, 846–854. [Google Scholar] [CrossRef]
  42. Castellsagué, X.; Alemany, L.; Quer, M.; Halec, G.; Quirós, B.; Tous, S.; Clavero, O.; Alòs, L.; Biegner, T.; Szafarowski, T.; et al. HPV Involvement in Head and Neck Cancers: Comprehensive Assessment of Biomarkers in 3680 Patients. J. Natl. Cancer Inst. 2016, 108, djv403. [Google Scholar] [CrossRef] [PubMed]
  43. Elgart, K.; Faden, D.L. Sinonasal Squamous Cell Carcinoma: Etiology, Pathogenesis, and the Role of Human Papilloma Virus. Curr. Otorhinolaryngol. Rep. 2020, 8, 111–119. [Google Scholar] [CrossRef]
  44. Vazquez-Guillen, J.M.; Palacios-Saucedo, G.C.; Alanis-Valdez, A.Y.; Huerta-Escobedo, A.; Zavala-Pompa, A.; Rivera-Morales, L.G.; Martinez-Torres, A.C.; Gonzalez-Villasana, V.; Serna-Hernandez, J.C.; Hernandez-Martinez, S.J.; et al. p16INK4a and pRb Expression in Laryngeal Squamous Cell Carcinoma with and without Infection by EBV or Different Genotypes of HPV: A Retrospective Study. Infect. Agents Cancer 2023, 18, 43. [Google Scholar] [CrossRef]
  45. Sirohi, D.; Schmidt, R.L.; Aisner, D.L.; Behdad, A.; Betz, B.L.; Brown, N.; Coleman, J.F.; Corless, C.L.; Deftereos, G.; Ewalt, M.D.; et al. Multi-Institutional Evaluation of Interrater Agreement of Variant Classification Based on the 2017 Association for Molecular Pathology, American Society of Clinical Oncology, and College of American Pathologists Standards and Guidelines for the Interpretation and Reporting of Sequence Variants in Cancer. J. Mol. Diagn 2020, 22, 284–293. [Google Scholar] [CrossRef]
  46. Holliday, D.; Mehrad, M.; Ely, K.A.; Tong, F.; Wang, X.; Hang, J.-F.; Kuo, Y.-J.; Velez-Torres, J.M.; Lott-Limbach, A.; Lewis, J.S. Sinonasal Adenosquamous Carcinoma—Morphology and Genetic Drivers Including Low- and High-Risk Human Papillomavirus mRNA, DEK::AFF2 Fusion, and MAML2 Rearrangement. Head Neck Pathol. 2023, 17, 487–497. [Google Scholar] [CrossRef] [PubMed]
  47. Bishop, J.A.; Guo, T.W.; Smith, D.F.; Wang, H.; Ogawa, T.; Pai, S.I.; Westra, W.H. Human Papillomavirus-Related Carcinomas of the Sinonasal Tract. Am. J. Surg. Pathol. 2013, 37, 185–192. [Google Scholar] [CrossRef] [PubMed]
  48. Chang Sing Pang, K.J.W.; Mur, T.; Collins, L.; Rao, S.R.; Faden, D.L. Human Papillomavirus in Sinonasal Squamous Cell Carcinoma: A Systematic Review and Meta-Analysis. Cancers 2020, 13, 45. [Google Scholar] [CrossRef]
  49. London, N.R.; Windon, M.J.; Amanian, A.; Zamuner, F.T.; Bishop, J.; Fakhry, C.; Rooper, L.M. Evaluation of the Incidence of Human Papillomavirus-Associated Squamous Cell Carcinoma of the Sinonasal Tract Among US Adults. JAMA Netw. Open 2023, 6, e2255971. [Google Scholar] [CrossRef]
  50. Cricca, M.; Morselli-Labate, A.M.; Venturoli, S.; Ambretti, S.; Gentilomi, G.A.; Gallinella, G.; Costa, S.; Musiani, M.; Zerbini, M. Viral DNA Load, Physical Status and E2/E6 Ratio as Markers to Grade HPV16 Positive Women for High-Grade Cervical Lesions. Gynecol. Oncol. 2007, 106, 549–557. [Google Scholar] [CrossRef]
  51. Ho, C.-M.; Lee, B.-H.; Chang, S.-F.; Chien, T.-Y.; Huang, S.-H.; Yan, C.-C.; Cheng, W.-F. Integration of Human Papillomavirus Correlates with High Levels of Viral Oncogene Transcripts in Cervical Carcinogenesis. Virus Res. 2011, 161, 124–130. [Google Scholar] [CrossRef]
  52. Walline, H.M.; Komarck, C.M.; McHugh, J.B.; Bellile, E.L.; Brenner, J.C.; Prince, M.E.; McKean, E.L.; Chepeha, D.B.; Wolf, G.T.; Worden, F.P.; et al. Genomic Integration of High-Risk HPV Alters Gene Expression in Oropharyngeal Squamous Cell Carcinoma. Mol. Cancer Res. 2016, 14, 941–952. [Google Scholar] [CrossRef]
  53. Mima, M.; Okabe, A.; Hoshii, T.; Nakagawa, T.; Kurokawa, T.; Kondo, S.; Mizokami, H.; Fukuyo, M.; Fujiki, R.; Rahmutulla, B.; et al. Tumorigenic Activation around HPV Integrated Sites in Head and Neck Squamous Cell Carcinoma. Int. J. Cancer 2023, 152, 1847–1862. [Google Scholar] [CrossRef]
  54. Ndiaye, C.; Mena, M.; Alemany, L.; Arbyn, M.; Castellsagué, X.; Laporte, L.; Bosch, F.X.; de Sanjosé, S.; Trottier, H. HPV DNA, E6/E7 mRNA, and p16INK4a Detection in Head and Neck Cancers: A Systematic Review and Meta-Analysis. Lancet Oncol. 2014, 15, 1319–1331. [Google Scholar] [CrossRef]
  55. Shi, W.; Kato, H.; Perez-Ordonez, B.; Pintilie, M.; Huang, S.; Hui, A.; O’Sullivan, B.; Waldron, J.; Cummings, B.; Kim, J.; et al. Comparative Prognostic Value of HPV16 E6 mRNA Compared with in Situ Hybridization for Human Oropharyngeal Squamous Carcinoma. J. Clin. Oncol. 2009, 27, 6213–6221. [Google Scholar] [CrossRef]
  56. Bratman, S.V.; Bruce, J.P.; O’Sullivan, B.; Pugh, T.J.; Xu, W.; Yip, K.W.; Liu, F.-F. Human Papillomavirus Genotype Association with Survival in Head and Neck Squamous Cell Carcinoma. JAMA Oncol. 2016, 2, 823–826. [Google Scholar] [CrossRef]
  57. No, J.H.; Sung, M.-W.; Hah, J.H.; Choi, S.H.; Lee, M.-C.; Kim, H.S.; Song, Y.-S. Prevalence and Prognostic Value of Human Papillomavirus Genotypes in Tonsillar Squamous Cell Carcinoma: A Korean Multicenter Study. Cancer 2015, 121, 535–544. [Google Scholar] [CrossRef] [PubMed]
  58. Lassen, P.; Eriksen, J.G.; 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. [Google Scholar] [CrossRef]
  59. Koenigs, M.B.; Lefranc-Torres, A.; Bonilla-Velez, J.; Patel, K.B.; Hayes, D.N.; Glomski, K.; Busse, P.M.; Chan, A.W.; Clark, J.R.; Deschler, D.G.; et al. Association of Estrogen Receptor Alpha Expression with Survival in Oropharyngeal Cancer Following Chemoradiation Therapy. J. Natl. Cancer Inst. 2019, 111, 933–942. [Google Scholar] [CrossRef] [PubMed]
  60. Patel, K.B.; Mroz, E.A.; Faquin, W.C.; Rocco, J.W. A Combination of Intra-Tumor Genetic Heterogeneity, Estrogen Receptor Alpha and Human Papillomavirus Status Predicts Outcomes in Head and Neck Squamous Cell Carcinoma Following Chemoradiotherapy. Oral Oncol. 2021, 120, 105421. [Google Scholar] [CrossRef] [PubMed]
  61. DE Oliveira Neto, C.P.; Brito, H.O.; DA Costa, R.M.G.; Brito, L.M.O. Is There a Role for Sex Hormone Receptors in Head-and-Neck Cancer? Links with HPV Infection and Prognosis. Anticancer Res. 2021, 41, 3707–3716. [Google Scholar] [CrossRef] [PubMed]
  62. Sherr, C.J.; Roberts, J.M. CDK Inhibitors: Positive and Negative Regulators of G1-Phase Progression. Genes Dev. 1999, 13, 1501–1512. [Google Scholar] [CrossRef]
  63. Kuong, K.J.; Loeb, L.A. APOBEC3B Mutagenesis in Cancer. Nat. Genet. 2013, 45, 964–965. [Google Scholar] [CrossRef] [PubMed]
  64. Pautasso, S.; Galitska, G.; Dell’Oste, V.; Biolatti, M.; Cagliani, R.; Forni, D.; De Andrea, M.; Gariglio, M.; Sironi, M.; Landolfo, S. Strategy of Human Cytomegalovirus to Escape Interferon Beta-Induced APOBEC3G Editing Activity. J. Virol. 2018, 92, e01224-18. [Google Scholar] [CrossRef]
  65. Ahasan, M.M.; Wakae, K.; Wang, Z.; Kitamura, K.; Liu, G.; Koura, M.; Imayasu, M.; Sakamoto, N.; Hanaoka, K.; Nakamura, M.; et al. APOBEC3A and 3C Decrease Human Papillomavirus 16 Pseudovirion Infectivity. Biochem. Biophys. Res. Commun. 2015, 457, 295–299. [Google Scholar] [CrossRef] [PubMed]
  66. Cannataro, V.L.; Gaffney, S.G.; Sasaki, T.; Issaeva, N.; Grewal, N.K.S.; Grandis, J.R.; Yarbrough, W.G.; Burtness, B.; Anderson, K.S.; Townsend, J.P. APOBEC-Induced Mutations and Their Cancer Effect Size in Head and Neck Squamous Cell Carcinoma. Oncogene 2019, 38, 3475–3487. [Google Scholar] [CrossRef]
  67. Riva, G.; Pecorari, G.; Biolatti, M.; Pautasso, S.; Lo Cigno, I.; Garzaro, M.; Dell’Oste, V.; Landolfo, S. PYHIN Genes as Potential Biomarkers for Prognosis of Human Papillomavirus-Positive or -Negative Head and Neck Squamous Cell Carcinomas. Mol. Biol. Rep. 2019, 46, 3333–3347. [Google Scholar] [CrossRef]
  68. Faden, D.L.; Thomas, S.; Cantalupo, P.G.; Agrawal, N.; Myers, J.; DeRisi, J. Multi-Modality Analysis Supports APOBEC as a Major Source of Mutations in Head and Neck Squamous Cell Carcinoma. Oral Oncol. 2017, 74, 8–14. [Google Scholar] [CrossRef]
  69. Riva, G.; Albano, C.; Gugliesi, F.; Pasquero, S.; Pacheco, S.F.C.; Pecorari, G.; Landolfo, S.; Biolatti, M.; Dell’Oste, V. HPV Meets APOBEC: New Players in Head and Neck Cancer. Int. J. Mol. Sci. 2021, 22, 1402. [Google Scholar] [CrossRef]
  70. Liu, Q.; Luo, Y.-W.; Cao, R.-Y.; Pan, X.; Chen, X.-J.; Zhang, S.-Y.; Zhang, W.-L.; Zhou, J.-Y.; Cheng, B.; Ren, X.-Y. Association between APOBEC3H-Mediated Demethylation and Immune Landscape in Head and Neck Squamous Carcinoma. BioMed Res. Int. 2020, 2020, 4612375. [Google Scholar] [CrossRef]
  71. Conner, K.L.; Shaik, A.N.; Ekinci, E.; Kim, S.; Ruterbusch, J.J.; Cote, M.L.; Patrick, S.M. HPV Induction of APOBEC3 Enzymes Mediate Overall Survival and Response to Cisplatin in Head and Neck Cancer. DNA Repair 2020, 87, 102802. [Google Scholar] [CrossRef] [PubMed]
  72. Tsao, S.W.; Tsang, C.M.; Lo, K.W. Epstein–Barr Virus Infection and Nasopharyngeal Carcinoma. Philos. Trans. R. Soc. B 2017, 372, 20160270. [Google Scholar] [CrossRef] [PubMed]
  73. Su, Z.Y.; Siak, P.Y.; Leong, C.-O.; Cheah, S.-C. The Role of Epstein–Barr Virus in Nasopharyngeal Carcinoma. Front. Microbiol. 2023, 14, 1116143. [Google Scholar] [CrossRef] [PubMed]
  74. Chan, K.C.A.; Woo, J.K.S.; King, A.; Zee, B.C.Y.; Lam, W.K.J.; Chan, S.L.; Chu, S.W.I.; Mak, C.; Tse, I.O.L.; Leung, S.Y.M.; et al. Analysis of Plasma Epstein-Barr Virus DNA to Screen for Nasopharyngeal Cancer. N. Engl. J. Med. 2017, 377, 513–522. [Google Scholar] [CrossRef] [PubMed]
  75. Li, T.; Li, F.; Guo, X.; Hong, C.; Yu, X.; Wu, B.; Lian, S.; Song, L.; Tang, J.; Wen, S.; et al. Anti–Epstein–Barr Virus BNLF2b for Mass Screening for Nasopharyngeal Cancer. N. Engl. J. Med. 2023, 389, 808–819. [Google Scholar] [CrossRef]
  76. Lam, W.K.J.; King, A.D.; Miller, J.A.; Liu, Z.; Yu, K.J.; Chua, M.L.K.; Ma, B.B.Y.; Chen, M.Y.; Pinsky, B.A.; Lou, P.-J.; et al. Recommendations for Epstein-Barr Virus–Based Screening for Nasopharyngeal Cancer in High- and Intermediate-Risk Regions. JNCI J. Natl. Cancer Inst. 2023, 115, 355–364. [Google Scholar] [CrossRef]
  77. Wong, L.P.; Lai, K.T.W.; Tsui, E.; Kwong, K.H.; Tsang, R.H.N.; Ma, E.S.K. Plasma Epstein-Barr Virus (EBV) DNA: Role as a Screening Test for Nasopharyngeal Carcinoma (NPC)? Int. J. Cancer 2005, 117, 515–516. [Google Scholar] [CrossRef]
  78. Wang, H.-Y.; Hsieh, C.-H.; Wen, C.-N.; Wen, Y.-H.; Chen, C.-H.; Lu, J.-J. Cancers Screening in an Asymptomatic Population by Using Multiple Tumour Markers. PLoS ONE 2016, 11, e0158285. [Google Scholar] [CrossRef]
  79. Yu, K.J.; Hsu, W.-L.; Pfeiffer, R.M.; Chiang, C.-J.; Wang, C.-P.; Lou, P.-J.; Cheng, Y.-J.; Gravitt, P.; Diehl, S.R.; Goldstein, A.M.; et al. Prognostic Utility of Anti-EBV Antibody Testing for Defining NPC Risk among Individuals from High-Risk NPC Families. Clin. Cancer Res. 2011, 17, 1906–1914. [Google Scholar] [CrossRef] [PubMed]
  80. Coghill, A.E.; Hsu, W.-L.; Pfeiffer, R.M.; Juwana, H.; Yu, K.J.; Lou, P.-J.; Wang, C.-P.; Chen, J.-Y.; Chen, C.-J.; Middeldorp, J.M.; et al. Epstein-Barr Virus Serology as a Potential Screening Marker for Nasopharyngeal Carcinoma among High-Risk Individuals from Multiplex Families in Taiwan. Cancer Epidemiol. Biomark. Prev. 2014, 23, 1213–1219. [Google Scholar] [CrossRef] [PubMed]
  81. Chan, K.C.A.; Hung, E.C.W.; Woo, J.K.S.; Chan, P.K.S.; Leung, S.-F.; Lai, F.P.T.; Cheng, A.S.M.; Yeung, S.W.; Chan, Y.W.; Tsui, T.K.C.; et al. Early Detection of Nasopharyngeal Carcinoma by Plasma Epstein-Barr Virus DNA Analysis in a Surveillance Program. Cancer 2013, 119, 1838–1844. [Google Scholar] [CrossRef]
  82. Alami, I.E.; Gihbid, A.; Charoute, H.; Khaali, W.; Brahim, S.M.; Tawfiq, N.; Cadi, R.; Belghmi, K.; El Mzibri, M.; Khyatti, M. Prognostic Value of Epstein-Barr Virus DNA Load in Nasopharyngeal Carcinoma: A Meta-Analysis. Pan Afr. Med. J. 2022, 41, 6. [Google Scholar] [CrossRef]
  83. Weng, J.; Wei, J.; Si, J.; Qin, Y.; Li, M.; Liu, F.; Si, Y.; Su, J. Clinical Outcomes of Residual or Recurrent Nasopharyngeal Carcinoma Treated with Endoscopic Nasopharyngectomy plus Chemoradiotherapy or with Chemoradiotherapy Alone: A Retrospective Study. PeerJ 2017, 5, e3912. [Google Scholar] [CrossRef] [PubMed]
  84. Alfieri, S.; Iacovelli, N.A.; Marceglia, S.; Lasorsa, I.; Resteghini, C.; Taverna, F.; Mazzocchi, A.; Orlandi, E.; Guzzo, M.; Bianchi, R.; et al. Circulating Pre-Treatment Epstein-Barr Virus DNA as Prognostic Factor in Locally-Advanced Nasopharyngeal Cancer in a Non-Endemic Area. Oncotarget 2017, 8, 47780–47789. [Google Scholar] [CrossRef]
  85. Naumov, S.S.; Kulbakin, D.E.; Krakhmal, N.V.; Vtorushin, S.V. Molecular and Biological Factors in the Prognosis of Head and Neck Squamous Cell Cancer. Mol. Biol. Rep. 2023, 50, 7839–7849. [Google Scholar] [CrossRef] [PubMed]
  86. Redman-Rivera, L.N.; Shaver, T.M.; Jin, H.; Marshall, C.B.; Schafer, J.M.; Sheng, Q.; Hongo, R.A.; Beckermann, K.E.; Wheeler, F.C.; Lehmann, B.D.; et al. Acquisition of Aneuploidy Drives Mutant P53-Associated Gain-of-Function Phenotypes. Nat. Commun. 2021, 12, 5184. [Google Scholar] [CrossRef] [PubMed]
  87. Zhao, D.; Tahaney, W.M.; Mazumdar, A.; Savage, M.I.; Brown, P.H. Molecularly Targeted Therapies for P53-Mutant Cancers. Cell. Mol. Life Sci. 2017, 74, 4171–4187. [Google Scholar] [CrossRef] [PubMed]
  88. Hashmi, A.A.; Hussain, Z.F.; Hashmi, S.K.; Irfan, M.; Khan, E.Y.; Faridi, N.; Khan, A.; Edhi, M.M. Immunohistochemical over Expression of P53 in Head and Neck Squamous Cell Carcinoma: Clinical and Prognostic Significance. BMC Res. Notes 2018, 11, 433. [Google Scholar] [CrossRef] [PubMed]
  89. Kerdpon, D.; Sriplung, H.; Kietthubthew, S. Expression of P53 in Oral Squamous Cell Carcinoma and Its Association with Risk Habits in Southern Thailand. Oral Oncol. 2001, 37, 553–557. [Google Scholar] [CrossRef] [PubMed]
  90. Dragomir, L.P.; Simionescu, C.; Mărgăritescu, C.; Stepan, A.; Dragomir, I.M.; Popescu, M.R. P53, P16 and Ki67 Immunoexpression in Oral Squamous Carcinomas. Rom. J. Morphol. Embryol. 2012, 53, 89–93. [Google Scholar]
  91. Etemad-Moghadam, S.; Keyhani, A.; Yazdani, K.; Alaeddini, M. Status of P53 and P27(KIP1) in Iranian Patients with Oral Squamous Cell Carcinoma. Iran. Red. Crescent. Med. J. 2015, 17, e19359. [Google Scholar] [CrossRef]
  92. Caponio, V.C.A.; Troiano, G.; Adipietro, I.; Zhurakivska, K.; Arena, C.; Mangieri, D.; Mascitti, M.; Cirillo, N.; Lo Muzio, L. Computational Analysis of TP53 Mutational Landscape Unveils Key Prognostic Signatures and Distinct Pathobiological Pathways in Head and Neck Squamous Cell Cancer. Br. J. Cancer 2020, 123, 1302–1314. [Google Scholar] [CrossRef]
  93. Temam, S.; Kawaguchi, H.; El-Naggar, A.K.; Jelinek, J.; Tang, H.; Liu, D.D.; Lang, W.; Issa, J.-P.; Lee, J.J.; Mao, L. Epidermal Growth Factor Receptor Copy Number Alterations Correlate with Poor Clinical Outcome in Patients with Head and Neck Squamous Cancer. J. Clin. Oncol. 2007, 25, 2164–2170. [Google Scholar] [CrossRef]
  94. Rubin Grandis, J.; Melhem, M.F.; Gooding, W.E.; Day, R.; Holst, V.A.; Wagener, M.M.; Drenning, S.D.; Tweardy, D.J. Levels of TGF-Alpha and EGFR Protein in Head and Neck Squamous Cell Carcinoma and Patient Survival. J. Natl. Cancer Inst. 1998, 90, 824–832. [Google Scholar] [CrossRef] [PubMed]
  95. Morrison, L.E.; Jacobson, K.K.; Friedman, M.; Schroeder, J.W.; Coon, J.S. Aberrant EGFR and Chromosome 7 Associate with Outcome in Laryngeal Cancer. Laryngoscope 2005, 115, 1212–1218. [Google Scholar] [CrossRef] [PubMed]
  96. Pomerantz, R.G.; Grandis, J.R. The Epidermal Growth Factor Receptor Signaling Network in Head and Neck Carcinogenesis and Implications for Targeted Therapy. Semin. Oncol. 2004, 31, 734–743. [Google Scholar] [CrossRef]
  97. Hsieh, C.-H.; Chang, J.W.C.; Hsieh, J.-J.; Hsu, T.; Huang, S.-F.; Liao, C.-T.; Wang, H.-M. Epidermal Growth Factor Receptor Mutations in Patients with Oral Cavity Cancer in a Betel Nut Chewing-Prevalent Area. Head Neck 2011, 33, 1758–1764. [Google Scholar] [CrossRef] [PubMed]
  98. Cunningham, D.; Humblet, Y.; Siena, S.; Khayat, D.; Bleiberg, H.; Santoro, A.; Bets, D.; Mueser, M.; Harstrick, A.; Verslype, C.; et al. Cetuximab Monotherapy and Cetuximab plus Irinotecan in Irinotecan-Refractory Metastatic Colorectal Cancer. N. Engl. J. Med. 2004, 351, 337–345. [Google Scholar] [CrossRef]
  99. Pfister, D.G.; Su, Y.B.; Kraus, D.H.; Wolden, S.L.; Lis, E.; Aliff, T.B.; Zahalsky, A.J.; Lake, S.; Needle, M.N.; Shaha, A.R.; et al. Concurrent Cetuximab, Cisplatin, and Concomitant Boost Radiotherapy for Locoregionally Advanced, Squamous Cell Head and Neck Cancer: A Pilot Phase II Study of a New Combined-Modality Paradigm. J. Clin. Oncol. 2006, 24, 1072–1078. [Google Scholar] [CrossRef] [PubMed]
  100. Astsaturov, I.; Cohen, R.B.; Harari, P. Targeting Epidermal Growth Factor Receptor Signaling in the Treatment of Head and Neck Cancer. Expert Rev. Anticancer Ther. 2006, 6, 1179–1193. [Google Scholar] [CrossRef] [PubMed]
  101. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and Its Ligands in Tolerance and Immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef]
  102. Qiao, X.; Jiang, J.; Pang, X.; Huang, M.; Tang, Y.; Liang, X.; Tang, Y. The Evolving Landscape of PD-1/PD-L1 Pathway in Head and Neck Cancer. Front. Immunol. 2020, 11, 1721. [Google Scholar] [CrossRef] [PubMed]
  103. Seiwert, T.Y.; Burtness, B.; Mehra, R.; Weiss, J.; Berger, R.; Eder, J.P.; Heath, K.; McClanahan, T.; Lunceford, J.; Gause, C.; et al. Safety and Clinical Activity of Pembrolizumab for Treatment of Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck (KEYNOTE-012): An Open-Label, Multicentre, Phase 1b Trial. Lancet Oncol. 2016, 17, 956–965. [Google Scholar] [CrossRef] [PubMed]
  104. Miyamoto, R.; Uzawa, N.; Nagaoka, S.; Hirata, Y.; Amagasa, T. Prognostic Significance of Cyclin D1 Amplification and Overexpression in Oral Squamous Cell Carcinomas. Oral Oncol. 2003, 39, 610–618. [Google Scholar] [CrossRef] [PubMed]
  105. Lin, Y.-M.; Sung, W.-W.; Hsieh, M.-J.; Tsai, S.-C.; Lai, H.-W.; Yang, S.-M.; Shen, K.-H.; Chen, M.-K.; Lee, H.; Yeh, K.-T.; et al. High PD-L1 Expression Correlates with Metastasis and Poor Prognosis in Oral Squamous Cell Carcinoma. PLoS ONE 2015, 10, e0142656. [Google Scholar] [CrossRef] [PubMed]
  106. Ngamphaiboon, N.; Chureemas, T.; Siripoon, T.; Arsa, L.; Trachu, N.; Jiarpinitnun, C.; Pattaranutaporn, P.; Sirachainan, E.; Larbcharoensub, N. Characteristics and Impact of Programmed Death-Ligand 1 Expression, CD8+ Tumor-Infiltrating Lymphocytes, and P16 Status in Head and Neck Squamous Cell Carcinoma. Med. Oncol. 2019, 36, 21. [Google Scholar] [CrossRef]
  107. Roper, E.; Lum, T.; Palme, C.E.; Ashford, B.; Ch’ng, S.; Ranson, M.; Boyer, M.; Clark, J.; Gupta, R. PD-L1 Expression Predicts Longer Disease Free Survival in High Risk Head and Neck Cutaneous Squamous Cell Carcinoma. Pathology 2017, 49, 499–505. [Google Scholar] [CrossRef] [PubMed]
  108. Müller, T.; Braun, M.; Dietrich, D.; Aktekin, S.; Höft, S.; Kristiansen, G.; Göke, F.; Schröck, A.; Brägelmann, J.; Held, S.A.E.; et al. PD-L1: A Novel Prognostic Biomarker in Head and Neck Squamous Cell Carcinoma. Oncotarget 2017, 8, 52889–52900. [Google Scholar] [CrossRef]
  109. Margalit, A.; Sheikhet, H.M.; Carmi, Y.; Berko, D.; Tzehoval, E.; Eisenbach, L.; Gross, G. Induction of Antitumor Immunity by CTL Epitopes Genetically Linked to Membrane-Anchored Beta2-Microglobulin. J. Immunol. 2006, 176, 217–224. [Google Scholar] [CrossRef]
  110. Chen, C.-H.; Su, C.-Y.; Chien, C.-Y.; Huang, C.-C.; Chuang, H.-C.; Fang, F.-M.; Huang, H.-Y.; Chen, C.-M.; Chiou, S.-J. Overexpression of Beta2-Microglobulin Is Associated with Poor Survival in Patients with Oral Cavity Squamous Cell Carcinoma and Contributes to Oral Cancer Cell Migration and Invasion. Br. J. Cancer 2008, 99, 1453–1461. [Google Scholar] [CrossRef]
  111. Klein, T.; Levin, I.; Niska, A.; Koren, R.; Gal, R.; Schachter, J.; Kfir, B.; Narinski, R.; Warchaizer, S.; Klein, B. Correlation between Tumour and Serum Beta 2m Expression in Patients with Breast Cancer. Eur. J. Immunogenet. 1996, 23, 417–423. [Google Scholar] [CrossRef] [PubMed]
  112. Diwan, N.N.; Chavan, M.S.; Motgi, A.A. Evaluation of Serum Beta-2 Microglobulin as a Diagnostic and Prognostic Marker in Oral Squamous Cell Carcinoma and Leukoplakia. Arch. Can. Res. 2016, 4, 120. [Google Scholar] [CrossRef]
  113. Nosratzehi, F.; Nosratzehi, T.; Alijani, E.; Rad, S.S. Salivary Β2-Microglobulin Levels in Patients with Erosive Oral Lichen Planus and Squamous Cell Carcinoma. BMC Res. Notes 2020, 13, 294. [Google Scholar] [CrossRef]
  114. Rasool, M.; Khan, S.R.; Malik, A.; Khan, K.M.; Zahid, S.; Manan, A.; Qazi, M.H.; Naseer, M.I. Comparative Studies of Salivary and Blood Sialic Acid, Lipid Peroxidation and Antioxidative Status in Oral Squamous Cell Carcinoma (OSCC). Pak. J. Med. Sci. 2014, 30, 466–471. [Google Scholar] [CrossRef]
  115. Jiang, Q.; Patima, S.; Ye, D.-X.; Pan, H.-Y.; Zhang, P.; Zhang, Z.-Y. Upregulation of Β2-Microglobulin Expression in Progressive Human Oral Squamous Cell Carcinoma. Oncol. Rep. 2012, 27, 1058–1064. [Google Scholar] [CrossRef]
  116. Swartz, J.E.; Pothen, A.J.; Stegeman, I.; Willems, S.M.; Grolman, W. Clinical Implications of Hypoxia Biomarker Expression in Head and Neck Squamous Cell Carcinoma: A Systematic Review. Cancer Med. 2015, 4, 1101–1116. [Google Scholar] [CrossRef] [PubMed]
  117. Höckel, M.; Vaupel, P. Tumor Hypoxia: Definitions and Current Clinical, Biologic, and Molecular Aspects. J. Natl. Cancer Inst. 2001, 93, 266–276. [Google Scholar] [CrossRef]
  118. Denko, N.C. Hypoxia, HIF1 and Glucose Metabolism in the Solid Tumour. Nat. Rev. Cancer 2008, 8, 705–713. [Google Scholar] [CrossRef]
  119. Mayer, A.; Vaupel, P. Hypoxia, Lactate Accumulation, and Acidosis: Siblings or Accomplices Driving Tumor Progression and Resistance to Therapy? Adv. Exp. Med. Biol. 2013, 789, 203–209. [Google Scholar] [CrossRef]
  120. Movahedi, K.; Laoui, D.; Gysemans, C.; Baeten, M.; Stangé, G.; Van den Bossche, J.; Mack, M.; Pipeleers, D.; In’t Veld, P.; De Baetselier, P.; et al. Different Tumor Microenvironments Contain Functionally Distinct Subsets of Macrophages Derived from Ly6C(High) Monocytes. Cancer Res. 2010, 70, 5728–5739. [Google Scholar] [CrossRef]
  121. Graeber, T.G.; Osmanian, C.; Jacks, T.; Housman, D.E.; Koch, C.J.; Lowe, S.W.; Giaccia, A.J. Hypoxia-Mediated Selection of Cells with Diminished Apoptotic Potential in Solid Tumours. Nature 1996, 379, 88–91. [Google Scholar] [CrossRef] [PubMed]
  122. Lin, A.J.; Gang, M.; Rao, Y.J.; Campian, J.; Daly, M.; Gay, H.; Oppelt, P.; Jackson, R.S.; Rich, J.; Paniello, R.; et al. Association of Posttreatment Lymphopenia and Elevated Neutrophil-to-Lymphocyte Ratio with Poor Clinical Outcomes in Patients With Human Papillomavirus-Negative Oropharyngeal Cancers. JAMA Otolaryngol. Head Neck Surg. 2019, 145, 413–421. [Google Scholar] [CrossRef] [PubMed]
  123. Lorente, D.; Mateo, J.; Templeton, A.J.; Zafeiriou, Z.; Bianchini, D.; Ferraldeschi, R.; Bahl, A.; Shen, L.; Su, Z.; Sartor, O.; et al. Baseline Neutrophil-Lymphocyte Ratio (NLR) Is Associated with Survival and Response to Treatment with Second-Line Chemotherapy for Advanced Prostate Cancer Independent of Baseline Steroid Use. Ann. Oncol. 2015, 26, 750–755. [Google Scholar] [CrossRef] [PubMed]
  124. Scilla, K.A.; Bentzen, S.M.; Lam, V.K.; Mohindra, P.; Nichols, E.M.; Vyfhuis, M.A.; Bhooshan, N.; Feigenberg, S.J.; Edelman, M.J.; Feliciano, J.L. Neutrophil-Lymphocyte Ratio Is a Prognostic Marker in Patients with Locally Advanced (Stage IIIA and IIIB) Non-Small Cell Lung Cancer Treated with Combined Modality Therapy. Oncologist 2017, 22, 737–742. [Google Scholar] [CrossRef] [PubMed]
  125. Kaźmierska, J.; Bajon, T.; Winiecki, T.; Borowczak, D.; Bandurska-Luque, A.; Jankowska, M.; Żmijewska-Tomczak, M. Significance of Neutrophil to Lymphocyte Ratio as a Predictor of Outcome in Head and Neck Cancer Treated with Definitive Chemoradiation. Rep. Pract. Oncol. Radiother. 2023, 28, 389–398. [Google Scholar] [CrossRef] [PubMed]
  126. Hu, X.; Tian, T.; Zhang, X.; Sun, Q.; Chen, Y.; Jiang, W. Neutrophil-to-Lymphocyte and Hypopharyngeal Cancer Prognosis: System Review and Meta-Analysis. Head Neck 2023, 45, 492–502. [Google Scholar] [CrossRef]
  127. Cho, J.-K.; Kim, M.W.; Choi, I.S.; Moon, U.Y.; Kim, M.-J.; Sohn, I.; Kim, S.; Jeong, H.-S. Optimal Cutoff of Pretreatment Neutrophil-to-Lymphocyte Ratio in Head and Neck Cancer Patients: A Meta-Analysis and Validation Study. BMC Cancer 2018, 18, 969. [Google Scholar] [CrossRef]
  128. Mattavelli, D.; Lombardi, D.; Missale, F.; Calza, S.; Battocchio, S.; Paderno, A.; Bozzola, A.; Bossi, P.; Vermi, W.; Piazza, C.; et al. Prognostic Nomograms in Oral Squamous Cell Carcinoma: The Negative Impact of Low Neutrophil to Lymphocyte Ratio. Front. Oncol. 2019, 9, 339. [Google Scholar] [CrossRef]
  129. Kim, D.Y.; Kim, I.S.; Park, S.G.; Kim, H.; Choi, Y.J.; Seol, Y.M. Prognostic Value of Posttreatment Neutrophil-Lymphocyte Ratio in Head and Neck Squamous Cell Carcinoma Treated by Chemoradiotherapy. Auris Nasus Larynx 2017, 44, 199–204. [Google Scholar] [CrossRef]
  130. Snietura, M.; Jaworska, M.; Mlynarczyk-Liszka, J.; Goraj-Zajac, A.; Piglowski, W.; Lange, D.; Wozniak, G.; Nowara, E.; Suwinski, R. PTEN as a Prognostic and Predictive Marker in Postoperative Radiotherapy for Squamous Cell Cancer of the Head and Neck. PLoS ONE 2012, 7, e33396. [Google Scholar] [CrossRef]
  131. Pérez-Tenorio, G.; Alkhori, L.; Olsson, B.; Waltersson, M.A.; Nordenskjöld, B.; Rutqvist, L.E.; Skoog, L.; Stål, O. PIK3CA Mutations and PTEN Loss Correlate with Similar Prognostic Factors and Are Not Mutually Exclusive in Breast Cancer. Clin. Cancer Res. 2007, 13, 3577–3584. [Google Scholar] [CrossRef]
  132. Jotta, P.Y.; Ganazza, M.A.; Silva, A.; Viana, M.B.; da Silva, M.J.; Zambaldi, L.J.G.; Barata, J.T.; Brandalise, S.R.; Yunes, J.A. Negative Prognostic Impact of PTEN Mutation in Pediatric T-Cell Acute Lymphoblastic Leukemia. Leukemia 2010, 24, 239–242. [Google Scholar] [CrossRef]
  133. da Costa, A.A.B.A.; D’Almeida Costa, F.; Ribeiro, A.R.; Guimarães, A.P.; Chinen, L.T.; Lopes, C.A.P.; de Lima, V.C.C. Low PTEN Expression Is Associated with Worse Overall Survival in Head and Neck Squamous Cell Carcinoma Patients Treated with Chemotherapy and Cetuximab. Int. J. Clin. Oncol. 2015, 20, 282–289. [Google Scholar] [CrossRef]
  134. Pattje, W.J.; Schuuring, E.; Mastik, M.F.; Slagter-Menkema, L.; Schrijvers, M.L.; Alessi, S.; van der Laan, B.F.a.M.; Roodenburg, J.L.N.; Langendijk, J.A.; van der Wal, J.E. The Phosphatase and Tensin Homologue Deleted on Chromosome 10 Mediates Radiosensitivity in Head and Neck Cancer. Br. J. Cancer 2010, 102, 1778–1785. [Google Scholar] [CrossRef]
  135. Weinberg, R.A. The Retinoblastoma Protein and Cell Cycle Control. Cell 1995, 81, 323–330. [Google Scholar] [CrossRef]
  136. Bartkova, J.; Lukas, J.; Strauss, M.; Bartek, J. Cyclin D1 Oncoprotein Aberrantly Accumulates in Malignancies of Diverse Histogenesis. Oncogene 1995, 10, 775–778. [Google Scholar]
  137. Dong, Y.; Sui, L.; Sugimoto, K.; Tai, Y.; Tokuda, M. Cyclin D1-CDK4 Complex, a Possible Critical Factor for Cell Proliferation and Prognosis in Laryngeal Squamous Cell Carcinomas. Int. J. Cancer 2001, 95, 209–215. [Google Scholar] [CrossRef]
  138. Carlos de Vicente, J.; Herrero-Zapatero, A.; Fresno, M.F.; López-Arranz, J.S. Expression of Cyclin D1 and Ki-67 in Squamous Cell Carcinoma of the Oral Cavity: Clinicopathological and Prognostic Significance. Oral Oncol. 2002, 38, 301–308. [Google Scholar] [CrossRef] [PubMed]
  139. Capaccio, P.; Pruneri, G.; Carboni, N.; Pagliari, A.V.; Quatela, M.; Cesana, B.M.; Pignataro, L. Cyclin D1 Expression Is Predictive of Occult Metastases in Head and Neck Cancer Patients with Clinically Negative Cervical Lymph Nodes. Head Neck 2000, 22, 234–240. [Google Scholar] [CrossRef]
  140. Bova, R.J.; Quinn, D.I.; Nankervis, J.S.; Cole, I.E.; Sheridan, B.F.; Jensen, M.J.; Morgan, G.J.; Hughes, C.J.; Sutherland, R.L. Cyclin D1 and p16INK4A Expression Predict Reduced Survival in Carcinoma of the Anterior Tongue. Clin. Cancer Res. 1999, 5, 2810–2819. [Google Scholar] [PubMed]
  141. Mineta, H.; Miura, K.; Takebayashi, S.; Ueda, Y.; Misawa, K.; Harada, H.; Wennerberg, J.; Dictor, M. Cyclin D1 Overexpression Correlates with Poor Prognosis in Patients with Tongue Squamous Cell Carcinoma. Oral Oncol. 2000, 36, 194–198. [Google Scholar] [CrossRef]
  142. Huang, S.-F.; Cheng, S.-D.; Chuang, W.-Y.; Chen, I.-H.; Liao, C.-T.; Wang, H.-M.; Hsieh, L.-L. Cyclin D1 Overexpression and Poor Clinical Outcomes in Taiwanese Oral Cavity Squamous Cell Carcinoma. World J. Surg. Oncol. 2012, 10, 40. [Google Scholar] [CrossRef]
  143. Wang, C.; Gan, N.; Liu, P.; Chen, H.; Li, Y.; Li, X. Expression and Genetic Polymorphisms of ERCC1 in Chinese Han Patients with Oral Squamous Cell Carcinoma. BioMed Res. Int. 2020, 2020, 1207809. [Google Scholar] [CrossRef]
  144. Yang, Z.-H.; Dai, Q.; Kong, X.-L.; Yang, W.-L.; Zhang, L. Association of ERCC1 Polymorphisms and Susceptibility to Nasopharyngeal Carcinoma. Mol. Carcinog. 2009, 48, 196–201. [Google Scholar] [CrossRef]
  145. Zhu, J.; Hua, R.-X.; Jiang, J.; Zhao, L.-Q.; Sun, X.; Luan, J.; Lang, Y.; Sun, Y.; Shang, K.; Peng, S.; et al. Association Studies of ERCC1 Polymorphisms with Lung Cancer Susceptibility: A Systematic Review and Meta-Analysis. PLoS ONE 2014, 9, e97616. [Google Scholar] [CrossRef] [PubMed]
  146. Gao, R.; Reece, K.M.; Sissung, T.; Fu, S.H.; Venzon, D.J.; Reed, E.; Spencer, S.D.; Price, D.K.; Figg, W.D. Are Race-Specific ERCC1 Haplotypes in Melanoma Cases versus Controls Related to the Predictive and Prognostic Value of ERCC1 N118N? BMJ Open 2013, 3, e002030. [Google Scholar] [CrossRef] [PubMed]
  147. He, M.G.; Zheng, K.; Tan, D.; Wang, Z.X. Association between ERCC1 and ERCC2 Gene Polymorphisms and Susceptibility to Pancreatic Cancer. Genet. Mol. Res. 2016, 15, 5. [Google Scholar] [CrossRef] [PubMed]
  148. Chen, C.; Wang, F.; Wang, Z.; Li, C.; Luo, H.; Liang, Y.; An, X.; Shao, J.; Li, Y. Polymorphisms in ERCC1 C8092A Predict Progression-Free Survival in Metastatic/Recurrent Nasopharyngeal Carcinoma Treated with Cisplatin-Based Chemotherapy. Cancer Chemother. Pharmacol. 2013, 72, 315–322. [Google Scholar] [CrossRef]
  149. Senghore, T.; Chien, H.-T.; Wang, W.-C.; Chen, Y.-X.; Young, C.-K.; Huang, S.-F.; Yeh, C.-C. Polymorphisms in ERCC5 Rs17655 and ERCC1 Rs735482 Genes Associated with the Survival of Male Patients with Postoperative Oral Squamous Cell Carcinoma Treated with Adjuvant Concurrent Chemoradiotherapy. J. Clin. Med. 2019, 8, 33. [Google Scholar] [CrossRef] [PubMed]
  150. Benzeid, R.; Gihbid, A.; Tawfiq, N.; Benchakroun, N.; Bendahhou, K.; Benider, A.; Guensi, A.; El Benna, N.; Filali Maltouf, A.; Attaleb, M.; et al. Genetic Polymorphisms in ERCC1 Gene and Their Association with Response to Radiotherapy in Moroccan Patients with Nasopharyngeal Carcinoma. Asian Pac. J. Cancer Prev. 2023, 24, 93–99. [Google Scholar] [CrossRef] [PubMed]
  151. Langer, C.J. Exploring Biomarkers in Head and Neck Cancer. Cancer 2012, 118, 3882–3892. [Google Scholar] [CrossRef] [PubMed]
  152. Strojan, P.; Budihna, M.; Smid, L.; Vrhovec, I.; Skrk, J. Cathepsin D in Tissue and Serum of Patients with Squamous Cell Carcinoma of the Head and Neck. Cancer Lett. 1998, 130, 49–56. [Google Scholar] [CrossRef] [PubMed]
  153. Ferrandina, G.; Scambia, G.; Benedetti Panici, P.; Almadori, G.; Paludetti, G.; Cadoni, G.; Distefano, M.; Maurizi, M.; Mancuso, S. Cathepsin D in Primary Squamous Laryngeal Tumors: Correlation with Clinico-Pathological Parameters and Receptor Status. Cancer Lett. 1992, 67, 133–138. [Google Scholar] [CrossRef] [PubMed]
  154. Gandour-Edwards, R.; Trock, B.; Donald, P.J. Predictive Value of Cathepsin-D for Cervical Lymph Node Metastasis in Head and Neck Squamous Cell Carcinoma. Head Neck 1999, 21, 718–722. [Google Scholar] [CrossRef]
  155. Zeillinger, R.; Swoboda, H.; Machacek, E.; Nekahm, D.; Sliutz, G.; Knogler, W.; Speiser, P.; Swoboda, E.; Kubista, E. Expression of Cathepsin D in Head and Neck Cancer. Eur. J. Cancer 1992, 28A, 1413–1415. [Google Scholar] [CrossRef]
  156. Brysk, M.M.; Lei, G.; Adler-Storthz, K.; Chen, Z.; Horikoshi, T.; Brysk, H.; Tyring, S.K.; Arany, I. Differentiation and Cathepsin D Expression in Human Oral Tumors. Laryngoscope 1998, 108, 1234–1237. [Google Scholar] [CrossRef]
  157. Silva, F.F.V.E.; Caponio, V.C.A.; Camolesi, G.C.V.; Padín-Iruegas, M.E.; Lorenzo-Pouso, A.I.; Lima, K.C.; Vieira, S.L.S.; Chamorro-Petronacci, C.M.; Suaréz-Peñaranda, J.M.; Pérez-Sayáns, M. Correlation of Bcl-2 Expression with Prognosis and Survival in Patients with Head and Neck Cancer: A Systematic Review and Meta-Analysis. Crit. Rev. Oncol. Hematol. 2023, 187, 104021. [Google Scholar] [CrossRef]
  158. Gomatos, I.P.; Georgiou, A.; Giotakis, J.; Manolopoulos, L.; Apostolou, K.; Chatzigianni, E.; Albanopoulos, K.; Ferekidou, E. The Role of Host Immune Response and Apoptosis in Patients with Laryngeal Squamous Cell Carcinoma. ORL J. Otorhinolaryngol. Relat. Spec. 2007, 69, 159–166. [Google Scholar] [CrossRef]
  159. Lee, L.-A.; Fang, T.-J.; Li, H.-Y.; Chuang, H.-H.; Kang, C.-J.; Chang, K.-P.; Liao, C.-T.; Chen, T.-C.; Huang, C.-G.; Yen, T.-C. Effects of Epstein-Barr Virus Infection on the Risk and Prognosis of Primary Laryngeal Squamous Cell Carcinoma: A Hospital-Based Case-Control Study in Taiwan. Cancers 2021, 13, 1741. [Google Scholar] [CrossRef]
  160. Michaud, W.A.; Nichols, A.C.; Mroz, E.A.; Faquin, W.C.; Clark, J.R.; Begum, S.; Westra, W.H.; Wada, H.; Busse, P.M.; Ellisen, L.W.; et al. Bcl-2 Blocks Cisplatin-Induced Apoptosis and Predicts Poor Outcome Following Chemoradiation Treatment in Advanced Oropharyngeal Squamous Cell Carcinoma. Clin. Cancer Res. 2009, 15, 1645–1654. [Google Scholar] [CrossRef] [PubMed]
  161. Lo Muzio, L.; Falaschini, S.; Farina, A.; Rubini, C.; Pezzetti, F.; Campisi, G.; De Rosa, G.; Capogreco, M.; Carinci, F. Bcl-2 as Prognostic Factor in Head and Neck Squamous Cell Carcinoma. Oncol. Res. 2005, 15, 249–255. [Google Scholar] [CrossRef]
  162. Ito, T.; Fujieda, S.; Tsuzuki, H.; Sunaga, H.; Fan, G.; Sugimoto, C.; Fukuda, M.; Saito, H. Decreased Expression of Bax Is Correlated with Poor Prognosis in Oral and Oropharyngeal Carcinoma. Cancer Lett. 1999, 140, 81–91. [Google Scholar] [CrossRef] [PubMed]
  163. Lovato, A.; Franz, L.; Carraro, V.; Bandolin, L.; Contro, G.; Ottaviano, G.; de Filippis, C.; Blandamura, S.; Alessandrini, L.; Marioni, G. Maspin Expression and Anti-Apoptotic Pathway Regulation by Bcl2 in Laryngeal Cancer. Ann. Diagn. Pathol. 2020, 45, 151471. [Google Scholar] [CrossRef] [PubMed]
  164. Giotakis, A.I.; Lazaris, A.C.; Kataki, A.; Kontos, C.K.; Giotakis, E.I. Positive BCL2L12 Expression Predicts Favorable Prognosis in Patients with Laryngeal Squamous Cell Carcinoma. Cancer Biomark. 2019, 25, 141–149. [Google Scholar] [CrossRef]
  165. Ueno, H.; Murphy, J.; Jass, J.R.; Mochizuki, H.; Talbot, I.C. Tumour “budding” as an Index to Estimate the Potential of Aggressiveness in Rectal Cancer. Histopathology 2002, 40, 127–132. [Google Scholar] [CrossRef] [PubMed]
  166. Liang, F.; Cao, W.; Wang, Y.; Li, L.; Zhang, G.; Wang, Z. The Prognostic Value of Tumor Budding in Invasive Breast Cancer. Pathol. Res. Pract. 2013, 209, 269–275. [Google Scholar] [CrossRef] [PubMed]
  167. Galván, J.A.; Zlobec, I.; Wartenberg, M.; Lugli, A.; Gloor, B.; Perren, A.; Karamitopoulou, E. Expression of E-Cadherin Repressors SNAIL, ZEB1 and ZEB2 by Tumour and Stromal Cells Influences Tumour-Budding Phenotype and Suggests Heterogeneity of Stromal Cells in Pancreatic Cancer. Br. J. Cancer 2015, 112, 1944–1950. [Google Scholar] [CrossRef]
  168. Niwa, Y.; Yamada, S.; Koike, M.; Kanda, M.; Fujii, T.; Nakayama, G.; Sugimoto, H.; Nomoto, S.; Fujiwara, M.; Kodera, Y. Epithelial to Mesenchymal Transition Correlates with Tumor Budding and Predicts Prognosis in Esophageal Squamous Cell Carcinoma. J. Surg. Oncol. 2014, 110, 764–769. [Google Scholar] [CrossRef]
  169. Angadi, P.V.; Patil, P.V.; Hallikeri, K.; Mallapur, M.D.; Hallikerimath, S.; Kale, A.D. Tumor Budding Is an Independent Prognostic Factor for Prediction of Lymph Node Metastasis in Oral Squamous Cell Carcinoma. Int. J. Surg. Pathol. 2015, 23, 102–110. [Google Scholar] [CrossRef]
  170. Bronsert, P.; Enderle-Ammour, K.; Bader, M.; Timme, S.; Kuehs, M.; Csanadi, A.; Kayser, G.; Kohler, I.; Bausch, D.; Hoeppner, J.; et al. Cancer Cell Invasion and EMT Marker Expression: A Three-Dimensional Study of the Human Cancer-Host Interface. J. Pathol. 2014, 234, 410–422. [Google Scholar] [CrossRef]
  171. Kalluri, R.; Weinberg, R.A. The Basics of Epithelial-Mesenchymal Transition. J. Clin. Investig. 2009, 119, 1420–1428. [Google Scholar] [CrossRef]
  172. Yusra; Semba, S.; Yokozaki, H. Biological Significance of Tumor Budding at the Invasive Front of Human Colorectal Carcinoma Cells. Int. J. Oncol. 2012, 41, 201–210. [Google Scholar] [CrossRef]
  173. Jensen, D.H.; Reibel, J.; Mackenzie, I.C.; Dabelsteen, E. Single Cell Migration in Oral Squamous Cell Carcinoma-Possible Evidence of Epithelial-Mesenchymal Transition in Vivo. J. Oral Pathol. Med. 2015, 44, 674–679. [Google Scholar] [CrossRef]
  174. Attramadal, C.G.; Kumar, S.; Boysen, M.E.; Dhakal, H.P.; Nesland, J.M.; Bryne, M. Tumor Budding, EMT and Cancer Stem Cells in T1-2/N0 Oral Squamous Cell Carcinomas. Anticancer Res. 2015, 35, 6111–6120. [Google Scholar]
  175. Wang, C.; Huang, H.; Huang, Z.; Wang, A.; Chen, X.; Huang, L.; Zhou, X.; Liu, X. Tumor Budding Correlates with Poor Prognosis and Epithelial-Mesenchymal Transition in Tongue Squamous Cell Carcinoma. J. Oral Pathol. Med. 2011, 40, 545–551. [Google Scholar] [CrossRef]
  176. Hattori, N.; Ushijima, T. Epigenetic Impact of Infection on Carcinogenesis: Mechanisms and Applications. Genome Med. 2016, 8, 10. [Google Scholar] [CrossRef] [PubMed]
  177. Liouta, G.; Adamaki, M.; Tsintarakis, A.; Zoumpourlis, P.; Liouta, A.; Agelaki, S.; Zoumpourlis, V. DNA Methylation as a Diagnostic, Prognostic, and Predictive Biomarker in Head and Neck Cancer. Int. J. Mol. Sci. 2023, 24, 2996. [Google Scholar] [CrossRef] [PubMed]
  178. Wang, T.-H.; Hsia, S.-M.; Shih, Y.-H.; Shieh, T.-M. Association of Smoking, Alcohol Use, and Betel Quid Chewing with Epigenetic Aberrations in Cancers. Int. J. Mol. Sci. 2017, 18, 1210. [Google Scholar] [CrossRef] [PubMed]
  179. Camuzi, D.; de Almeida Simão, T.; Dias, F.; Ribeiro Pinto, L.F.; Soares-Lima, S.C. Head and Neck Cancers Are Not Alike When Tarred with the Same Brush: An Epigenetic Perspective from the Cancerization Field to Prognosis. Cancers 2021, 13, 5630. [Google Scholar] [CrossRef] [PubMed]
  180. Starzer, A.M.; Heller, G.; Tomasich, E.; Melchardt, T.; Feldmann, K.; Hatziioannou, T.; Traint, S.; Minichsdorfer, C.; Schwarz-Nemec, U.; Nackenhorst, M.; et al. DNA Methylation Profiles Differ in Responders versus Non-Responders to Anti-PD-1 Immune Checkpoint Inhibitors in Patients with Advanced and Metastatic Head and Neck Squamous Cell Carcinoma. J. Immunother. Cancer 2022, 10, e003420. [Google Scholar] [CrossRef] [PubMed]
  181. Goltz, D.; Gevensleben, H.; Dietrich, J.; Schroeck, F.; de Vos, L.; Droege, F.; Kristiansen, G.; Schroeck, A.; Landsberg, J.; Bootz, F.; et al. PDCD1 (PD-1) Promoter Methylation Predicts Outcome in Head and Neck Squamous Cell Carcinoma Patients. Oncotarget 2017, 8, 41011–41020. [Google Scholar] [CrossRef] [PubMed]
  182. Hostetter, M.K. The Third Component of Complement: New Functions for an Old Friend. J. Lab. Clin. Med. 1993, 122, 491–496. [Google Scholar] [PubMed]
  183. Peng, Y.; Croce, C.M. The Role of MicroRNAs in Human Cancer. Signal Transduct. Target. Ther. 2016, 1, 15004. [Google Scholar] [CrossRef] [PubMed]
  184. Lawrie, C.H.; Gal, S.; Dunlop, H.M.; Pushkaran, B.; Liggins, A.P.; Pulford, K.; Banham, A.H.; Pezzella, F.; Boultwood, J.; Wainscoat, J.S.; et al. Detection of Elevated Levels of Tumour-Associated microRNAs in Serum of Patients with Diffuse Large B-Cell Lymphoma. Br. J. Haematol. 2008, 141, 672–675. [Google Scholar] [CrossRef] [PubMed]
  185. Hu, Z.; Chen, X.; Zhao, Y.; Tian, T.; Jin, G.; Shu, Y.; Chen, Y.; Xu, L.; Zen, K.; Zhang, C.; et al. Serum microRNA Signatures Identified in a Genome-Wide Serum microRNA Expression Profiling Predict Survival of Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2010, 28, 1721–1726. [Google Scholar] [CrossRef] [PubMed]
  186. Ji, J.; Shi, J.; Budhu, A.; Yu, Z.; Forgues, M.; Roessler, S.; Ambs, S.; Chen, Y.; Meltzer, P.S.; Croce, C.M.; et al. MicroRNA Expression, Survival, and Response to Interferon in Liver Cancer. N. Engl. J. Med. 2009, 361, 1437–1447. [Google Scholar] [CrossRef] [PubMed]
  187. Ludwig, S.; Sharma, P.; Wise, P.; Sposto, R.; Hollingshead, D.; Lamb, J.; Lang, S.; Fabbri, M.; Whiteside, T.L. mRNA and miRNA Profiles of Exosomes from Cultured Tumor Cells Reveal Biomarkers Specific for HPV16-Positive and HPV16-Negative Head and Neck Cancer. Int. J. Mol. Sci. 2020, 21, 8570. [Google Scholar] [CrossRef] [PubMed]
  188. Lajer, C.B.; Nielsen, F.C.; Friis-Hansen, L.; Norrild, B.; Borup, R.; Garnæs, E.; Rossing, M.; Specht, L.; Therkildsen, M.H.; Nauntofte, B.; et al. Different miRNA Signatures of Oral and Pharyngeal Squamous Cell Carcinomas: A Prospective Translational Study. Br. J. Cancer 2011, 104, 830–840. [Google Scholar] [CrossRef]
  189. Hess, J.; Unger, K.; Maihoefer, C.; Schüttrumpf, L.; Wintergerst, L.; Heider, T.; Weber, P.; Marschner, S.; Braselmann, H.; Samaga, D.; et al. A Five-MicroRNA Signature Predicts Survival and Disease Control of Patients with Head and Neck Cancer Negative for HPV Infection. Clin. Cancer Res. 2019, 25, 1505–1516. [Google Scholar] [CrossRef]
  190. Hess, J.; Unger, K.; Maihoefer, C.; Schüttrumpf, L.; Weber, P.; Marschner, S.; Wintergerst, L.; Pflugradt, U.; Baumeister, P.; Walch, A.; et al. Integration of P16/HPV DNA Status with a 24-miRNA-Defined Molecular Phenotype Improves Clinically Relevant Stratification of Head and Neck Cancer Patients. Cancers 2022, 14, 3745. [Google Scholar] [CrossRef]
  191. Hess, A.-K.; Müer, A.; Mairinger, F.D.; Weichert, W.; Stenzinger, A.; Hummel, M.; Budach, V.; Tinhofer, I. MiR-200b and miR-155 as Predictive Biomarkers for the Efficacy of Chemoradiation in Locally Advanced Head and Neck Squamous Cell Carcinoma. Eur. J. Cancer 2017, 77, 3–12. [Google Scholar] [CrossRef]
  192. Liu, Z.; Tian, Z.; Zhang, C.; Sun, J.; Zhang, Z.; He, Y. Microvascular Reconstruction in Elderly Oral Cancer Patients: Does Diabetes Status Have a Predictive Role in Free Flap Complications? J. Oral Maxillofac. Surg. 2015, 73, 357–369. [Google Scholar] [CrossRef]
  193. Koh, Y.; Kim, T.M.; Jeon, Y.K.; Kwon, T.-K.; Hah, J.H.; Lee, S.-H.; Kim, D.-W.; Wu, H.-G.; Rhee, C.-S.; Sung, M.-W.; et al. Class III Beta-Tubulin, but Not ERCC1, Is a Strong Predictive and Prognostic Marker in Locally Advanced Head and Neck Squamous Cell Carcinoma. Ann. Oncol. 2009, 20, 1414–1419. [Google Scholar] [CrossRef]
  194. Schnoell, J.; Scheiflinger, A.; Al-Gboore, S.; Kadletz-Wanke, L.; Kenner, L.; Heiduschka, G.; Jank, B.J. The Prognostic Role of PSMD14 in Head and Neck Squamous Cell Carcinoma. J. Cancer Res. Clin. Oncol. 2023, 149, 2483–2490. [Google Scholar] [CrossRef]
  195. Johansson, A.-C.; Ansell, A.; Jerhammar, F.; Lindh, M.B.; Grénman, R.; Munck-Wikland, E.; Östman, A.; Roberg, K. Cancer-Associated Fibroblasts Induce Matrix Metalloproteinase-Mediated Cetuximab Resistance in Head and Neck Squamous Cell Carcinoma Cells. Mol. Cancer Res. 2012, 10, 1158–1168. [Google Scholar] [CrossRef]
  196. Cullen, K.J.; Schumaker, L.; Nikitakis, N.; Goloubeva, O.; Tan, M.; Sarlis, N.J.; Haddad, R.I.; Posner, M.R. Beta-Tubulin-II Expression Strongly Predicts Outcome in Patients Receiving Induction Chemotherapy for Locally Advanced Squamous Carcinoma of the Head and Neck: A Companion Analysis of the TAX 324 Trial. J. Clin. Oncol. 2009, 27, 6222–6228. [Google Scholar] [CrossRef] [PubMed]
  197. Jing, C.; Duan, Y.; Zhou, M.; Yue, K.; Zhuo, S.; Li, X.; Liu, D.; Ye, B.; Lai, Q.; Li, L.; et al. Blockade of Deubiquitinating Enzyme PSMD14 Overcomes Chemoresistance in Head and Neck Squamous Cell Carcinoma by Antagonizing E2F1/Akt/SOX2-Mediated Stemness. Theranostics 2021, 11, 2655–2669. [Google Scholar] [CrossRef] [PubMed]
  198. Liu, M.; Huang, L.; Liu, Y.; Yang, S.; Rao, Y.; Chen, X.; Nie, M.; Liu, X. Identification of the MMP Family as Therapeutic Targets and Prognostic Biomarkers in the Microenvironment of Head and Neck Squamous Cell Carcinoma. J. Transl. Med. 2023, 21, 208. [Google Scholar] [CrossRef] [PubMed]
Table 1. List of biomarkers mentioned in this review.
Table 1. List of biomarkers mentioned in this review.
BiomarkerDescription
Diagnosis
EBV statusSerologic tests examining antibodies to EBV IgA are performed to evaluate patients with suspected nasopharyngeal carcinoma.
Beta 2-microglobulinA component of the major histocompatibility complex (MHC) class I. It is associated with tumor status in various cancers.
Diagnostic/Prognostic
p16Ink4aAn indirect marker of HPV-positive disease in oropharyngeal squamous cell carcinomas.
HPV-E6 seropositivityAntibodies to the HPV virus E6 antigen. Linked to an increased risk of developing oropharyngeal cancer.
Circulating tumor HPV DNAFragments of HPV DNA shed from tumor cells into the blood; highly specific for malignancy.
Oral HPV DNAOncogenic HPV DNA found in oral rinses. Has been associated with treatment response and recurrence following treatment but has been shown to have less sensitivity compared to ctHPVDNA.
Prognostic
Estrogen receptor positivityTumor estrogen receptor alpha (Era) positivity, being studied as a biomarker for improved overall survival and recurrence-free survival in HPV-positive oropharyngeal cancer.
Hypoxia markersHypoxia is associated with tumor progression, contributing to a more aggressive phenotype and modifying the free radical chemistry of tumors.
TP53 and P53A tumor-suppressor gene that regulates gene overexpression in head and neck squamous cell carcinoma.
Cyclin D1Regulator of the G1 checkpoint of cell division. D1 has been implicated in the development of tumors of the esophagus, ovary, breast, colon, lung, and the head and neck.
Cathepsin-DA lysosomal enzyme found throughout the body’s cells. It has been found to be overexpressed and/or abnormally processed in various cancer cells and is thought to have a role in local tumor invasion and metastasis.
Bcl-2A key mitochondrial protein that regulates apoptosis, with overexpression of the protein neutralizing pro-apoptotic proteins and inhibiting apoptosis in the cell.
Prognostic/predictive
Apolipoprotein B mRNA Editing Enzyme, Catalytic Polypeptide (APOBEC)Aberrantly acting DNA-modifying enzymes that have been linked to DNA mutations and tumor formation.
Neutrophil to Lymphocyte RatioThe balance of neutrophils and lymphocytes, expressed as a neutrophil-to-lymphocyte ratio (NLR), has been shown to predict outcomes in solid tumors.
PTENA tumor suppressor that negatively regulates the PI3K–AKT–mTOR pathway and regulates various processes related to cell growth and proliferation.
ERCC1Excision repair cross-complementation group 1 (ERCC1) is an important part of the nucleotide excision repair pathway of DNA repair.
PD-L1 ExpressionPD-L1 is an immune checkpoint regulator and immunomodulatory protein expressed in various immune cells.
EGFREpidermal growth factor receptor, overexpressed in a variety of solid tumors, with significant representation in head and neck squamous cell carcinoma.
Tumor budding and epithelial–mesenchymal transitionTumor budding is defined as the presence of single tumor cells or groups of tumor cells at the tumor margin. Epithelial–mesenchymal transition involves the transformation of an epithelial cell into a mesenchymal cell phenotype.
DNA methylationDNA methylation occurs through the addition of a methyl group to a carbon of cytosine, forming 5-methylcytosine. This leads to a change in the activity of transcription factors and the mobility of various proteins.
MicroRNAsMicroRNAs are molecules of non-coding DNA that regulate gene expression and dysregulation.
Beta-tubulin isotypesBeta-tubulin isotypes regulate the structure of microtubules.
PSMD14PSMD14 is a proteasomal degradation protein that removes ubiquitin from proteins.
SSP1SSP1 prevents the proliferation of effector T cells.
Matrix metalloproteinase family of enzymesMatrix metalloproteinases participate in a variety of biological processes including the degradation of tissue components, angiogenesis, and neurogenesis.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Eberly, H.W.; Sciscent, B.Y.; Lorenz, F.J.; Rettig, E.M.; Goyal, N. Current and Emerging Diagnostic, Prognostic, and Predictive Biomarkers in Head and Neck Cancer. Biomedicines 2024, 12, 415. https://doi.org/10.3390/biomedicines12020415

AMA Style

Eberly HW, Sciscent BY, Lorenz FJ, Rettig EM, Goyal N. Current and Emerging Diagnostic, Prognostic, and Predictive Biomarkers in Head and Neck Cancer. Biomedicines. 2024; 12(2):415. https://doi.org/10.3390/biomedicines12020415

Chicago/Turabian Style

Eberly, Hänel W., Bao Y. Sciscent, F. Jeffrey Lorenz, Eleni M. Rettig, and Neerav Goyal. 2024. "Current and Emerging Diagnostic, Prognostic, and Predictive Biomarkers in Head and Neck Cancer" Biomedicines 12, no. 2: 415. https://doi.org/10.3390/biomedicines12020415

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