Next Article in Journal
The Influence of Autoimmune Thyroid Diseases on Viral Pneumonia Development, Including COVID-19: A Two-Sample Mendelian Randomization Study
Next Article in Special Issue
The Intricate Interplay between APOBEC3 Proteins and DNA Tumour Viruses
Previous Article in Journal
The Trimeric Autotransporter Adhesin EmaA and Infective Endocarditis
Previous Article in Special Issue
The Clinical Utility of Circulating HPV DNA Biomarker in Oropharyngeal, Cervical, Anal, and Skin HPV-Related Cancers: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

p16 Expression in Laryngeal Squamous Cell Carcinoma: A Surrogate or Independent Prognostic Marker?

by
Roberto Gallus
1,
Davide Rizzo
2,3,
Giorgia Rossi
4,
Luca Mureddu
2,*,
Jacopo Galli
4,5,
Alberto Artuso
1 and
Francesco Bussu
2,3
1
Otolaryngology, Mater Olbia Hospital, 07026 Olbia, Italy
2
U.O.C. Otorinolaringoiatria, Azienda Ospedaliero Universitaria di Sassari, Viale San Pietro, 43, 07100 Sassari, Italy
3
Otolaryngology Division, Department of Medicine, Surgery and Pharmacology, University of Sassari, Viale San Pietro, 43, 07100 Sassari, Italy
4
Unit of Otorhinolaryngology and Head-Neck Surgery, “A. Gemelli” Hospital Foundation IRCCS, 00168 Rome, Italy
5
Department of Head-Neck and Sensory Organs, Catholic University of Sacred Heart, 00168 Rome, Italy
*
Author to whom correspondence should be addressed.
Pathogens 2024, 13(2), 100; https://doi.org/10.3390/pathogens13020100
Submission received: 7 January 2024 / Revised: 21 January 2024 / Accepted: 22 January 2024 / Published: 24 January 2024

Abstract

:
Laryngeal squamous cell carcinoma (LSCC) is a common malignancy that, despite scientific advancements, has not seen an improvement in its prognosis in the last decades. Few promising predictive markers have been found and none are relevant in clinical practice. p16ink4a, an oncosuppressor protein involved in cell cycle arrest, with a prognostic impact on other cancers, has been widely used in the head and neck region as a surrogate marker of HPV infection. Published papers and recent meta-analyses seem to minimize the biological role of HPV in the context of LSCC’s cancerogenesis, and to disprove the reliability of p16ink4a as a surrogate prognostic marker in this context, while still highlighting its potential role as an independent predictor of survival. Unfortunately, the available literature, in particular during the last two decades, is often not focused on its potential role as an independent biomarker and few relevant data are found in papers mainly focused on HPV. The available data suggest that future research should focus specifically on p16ink4a, taking into account both its potential inactivation and overexpression, different patterns of staining, and immunohistochemistry cutoffs, and should focus not on its potential role as a surrogate marker but on its independent role as a predictor of survival.

1. Introduction

The discovery and study of the role of high-risk Human Papillomaviruses (HPVs) in oropharyngeal squamous cell carcinomas (OPSCCs) during the last 20 years have completely changed our understanding of a subset of these tumors and have supported the identification of HPV-positive OPSCC as a distinct clinical entity [1,2,3,4,5,6]. The knowledge accumulated during this timeframe has led to the recent change in the American Joint Committee on Cancer (AJCC) TNM (tumor, node, metastasis) staging system and is supposed to ultimately pave the way to deintensification protocols in a selected cohort of patients [7]. An unintended byproduct of the rise in HPV-positive OPSCCs has been the shedding of light on the elected surrogate marker p16INK4a [8], which is well known to oncologists and clinicians dealing with cervical cancers and less known to otolaryngologists and researchers in the field of head and neck cancers. In fact, in both the aforementioned eighth edition of the AJCC TNM staging system and current major guidelines, p16INK4a immunohistochemistry (p16 IHC) is considered the reference technique for the detection of HPV in the oropharynx [9,10]. However, this technique has some relevant limitations that have been extensively discussed elsewhere [11,12].
After first confirming the role of HPV in the cancerogenesis of a subset of OPSCCs, efforts were made to seek its impact on other head and neck cancers, with mixed and sometimes disappointing results, especially when it came to its impact on prognosis [13]. Many of the studies that focus on non-oropharyngeal cancers have often relied, probably inappropriately [14], on p16INK4a as a surrogate marker of HPV involvement, thereby gifting us with a relevant amount of data regarding its overexpression in squamous cellular carcinomas (SCCs) from different subsites of the head and neck. These same studies have sometimes offered data on its eventual independent prognostic significance in these subsites. The aims of this narrative review are to discuss such evidence, often feeble, in the setting of laryngeal squamous cell carcinomas, and to put the attention front and center on p16INK4a, which is often a misunderstood marker that is discussed prevalently in relation to HPV in this particular setting.

2. p16ink4a

p16 is a tumor suppressor gene located within the INK4a/ARF locus on the short arm of chromosome 9p21.3 that encodes for the homonym protein p16INK4a and three other transcriptional variants: p14ARF (alternative reading frame), p12, and p16γ [15]. Other names of the same gene include MTS-1 (multiple tumor suppressor-1), INK4A (inhibitor of cyclin-dependent kinase 4a), and CDKN2A (cyclin-dependent kinase inhibitor 2a) [16,17,18]. p16INK4a belongs to the INK4 family of CDK inhibitors that also includes p15INK4B, p18INK4C, and p19INK4D. All these proteins are involved in tumor growth inhibition and tumor suppression [19,20]. The p16 gene function in particular is related to the regulation of the p53 and pRB cell cycle pathways [15]. Its location (9p21.3) is the site of the loss of heterozygosity in many malignancies [19]. The gene includes five exons (E1β, E1α, E2, E2γ, and E3), and three of them (E1α, E2, and E3), each encoding for 156 amino acids, are responsible for the p16INK4a transcript. The structure of p16INK4a includes four ankyrin repeats arranged in a helix involved in interactions with target proteins [21].
The p16INK4a involvement in cell cycle regulation is focused on the inhibition of the S phase. While its function is still not fully understood, what is known is that p16INK4a is able to bind CDK4/6 and to consequently inhibit D–CDK4/6 complex formation and the phosphorylation of Rb family members. Hypophosphorylated Rb proteins bind E2F1, preventing the transcription of the S-phase genes and leading to cycle arrest [19]. p16INK4a and pRb seem to be bound by a feedback loop in which hypophosphorylated pRb downregulates p16INK4a, while phosphorylated pRb induces p16 expression [22]. p16INK4a overexpression is at least partially involved in cellular senescence and cell cycle arrest in nonsenescent cells [23], a growth arrest mechanism meant to protect the cell from hyperproliferative mechanisms and cellular stress (such as exposure to toxic substances, oxidative stress, DNA damage) [24]. It should be noted that other mechanisms can regulate p16INK4a expression, and that its oncosuppressor role can be mediated via different mechanisms not limited to the pRb–E2F pathway [25,26,27,28]. p16INK4a also seems to be involved in other cellular mechanisms potentially relevant for tumorigenesis, acting as a tumor suppressor on various fronts. In fact, in vivo and in vitro data suggest a role in tumor invasion, angiogenesis, and apoptosis in various cancers [29]. Other reported functions of p16INK4a exerted through an accurate balance between its overexpression and downregulation include a potential role in the healing processes [30,31,32], stem cell maintenance and cell differentiation [33], glucose homeostasis and fat metabolism [34,35], and embryonic development [36].
Both the downregulation and overexpression of p16INK4a are phenomena seen in the context of cancer [29]. Available data show that p16INK4a is inactivated via various mechanisms (point mutations, loss of heterozygosity, methylation of the promoter region) in 50% of human cancers, including head and neck cancers [19]. Mutations at the level of the INK4a/ARF locus are seen in some inherited cancer syndromes [37,38,39]. Available evidence shows that the inactivation of p16INK4a could be both an early event leading to preneoplastic lesions or a late event during tumor progression [40,41,42]. While the inactivation of p16INK4a is usually seen in premalignant and malignant lesions and has been interpreted as a step to overcome the natural protection mechanism of the cell, overexpression too can be seen in benign lesions and in the very early stages of transformation, and sometimes also in malignant lesions. Overexpression in benign and premalignant lesions has been interpreted in the context of its natural role as an oncosuppressor and mediator of oncogene-induced senescence [19], protecting the cell from malignant transformation, while its accumulation in the later phases of malignant transformation could be related to anomalies of the p16Ink4a–Rb pathway and to accumulation through a positive-feedback mechanism [43]. This interpretation is consistent with the fact that Rb loss is an event frequently seen in tumor progression. Unfortunately, little is known about p16INK4a overexpression in cancer, a phenomenon seen especially in HPV-related cancers, such as cervical cancer, anal cancer, or oropharyngeal squamous cell carcinoma, albeit sometimes described in non-HPV-related cancers [44,45,46,47]. Interestingly, while the main known function of p16INK4a takes place in the nucleus, its overexpression is often seen at the cytoplasmic level, possibly with different functions. While the exact mechanisms behind cytoplasmic accumulation are poorly understood, both CDK4 sequestration and anion exchanger 1 (AE1)-mediated accumulation have been hypothesized [48,49]. A prevalent cytoplasmic or concomitant nuclear/cytoplasmic expression has been seen in the contexts of breast cancer, colorectal cancer, and some non-epithelial tumors [44,45,48,50,51,52,53]. Interestingly, a high cytoplasmic and low nuclear expression has been linked with a poorer prognosis in head and neck cancers [54].

2.1. p16ink4a as a Surrogate Marker

In the last decades, immunostaining for p16INK4a has seen a great deal of success as a diagnostic technique in cervical cancer and OPSCC, which are both HPV-related tumors [55]. In fact, in this particular setting, p16INK4a behaves as a surrogate marker not only of HPV infection but also of its active role in the cancerogenic process [56]. The reason behind the overexpression of p16INK4a in HPV-related cancers has been extensively discussed in the literature. In brief, after the integration of the high-risk HPV genome into the host cell, three main oncoproteins, E5, E6, and E7, are expressed. E7 is able to associate with pRb and the related proteins p107 and p130, leading to their degradation, ultimately unlocking cell cycle progression by allowing E2F-regulated transcription [57]. This event leads to p16INK4a overexpression via a positive-feedback loop. Part of the reason that p16 immunohistochemistry (IHC) has been extensively used lies in the fact that it is a simple, broadly available, and relatively cheap technique [11], and despite some relevant limitations, as mentioned before, it is now considered the reference technique for the identification of HPV-related OPSCCs by the major guidelines, albeit with some caveats [58]. In fact, while it is acceptable to use p16 ICH in populations characterized by high fractions of HPV-positive OPSCCs, such as those of North America and Northern Europe, the same is not true elsewhere. While p16 IHC is characterized by a high sensibility, it lacks specificity, which means that in populations with a low prevalence of HPV-positive OPSCCs, higher numbers of false-positive cases will be seen. This has a potential detrimental effect on patient stratification and selection for deintensification protocols in these populations, and it could, theoretically, also affect HPV detection in subsites other than the oropharynx, where the HPV reported prevalence is often low. In fact, an ever-growing amount of evidence discourages its use as a surrogate marker of HPV infection outside of the oropharyngeal site and, in particular, in LSCCs, as well as under specific conditions (a low prevalence of HPV-related oropharyngeal squamous cell carcinoma), even for the oropharyngeal site [5,14,59,60,61,62]. The last decade of research on the topic seems to also minimize its potential biologic role in the cancerogenic process of LSCCs, probably impacting only a small subgroup of patients. This subset is quantifiable in around 11% of LSCCs according to a recent analysis of the National Cancer Database that notably included data coming from studies that sometimes used p16IHC as the only detection method, thereby probably overestimating the true prevalence [63]. Notably, a recent meta-analysis by Sahovaler and colleagues also found a prognostic impact only when evaluating p16ink4a positivity but not HPV DNA positivity [13], reflecting the potential different biological meanings of the two markers.

2.2. Prognostic Role of p16ink4a

As seen, under different circumstances, both p16INK4a overexpression and downregulation can be seen in different cancers in both the early and advanced stages. When p16INK4a is downregulated as a step towards cancerogenesis, lower expression levels can be a marker of both malignant transformation and a poor prognosis. This is the case with melanoma, in which lower expression levels are associated with metastases [64] and osteosarcoma, where it predicts a poor response to chemotherapy [65]. In the setting of pulmonary squamous cell carcinoma, p16 methylation is not only an early-stage event and potential diagnostic biomarker but also serves to differentiate a primary pulmonary lesion from a metastasis from a p16INK4a -overexpressing cancer, such as cervical squamous cell carcinoma [66]. p16INK4a downregulation has also been identified as a potential prognostic biomarker of a poor prognosis in non-small-cell lung carcinoma [66,67] and glioblastoma [68]. In tumors that overexpress p16INK4a after malignant transformation, detecting this event could be of prognostic value. In fact, some studies suggest that p16INK4a overexpression could be related to unfavorable tumor characteristics in a subset of tumors, including adenocarcinoma, breast cancer, astrocytoma, and gastrointestinal stromal tumors [44,45,47,69].

2.3. p16ink4a in the Context of LSCC

Laryngeal squamous cell carcinoma is a common head and neck cancer. It affects men more commonly than women. Its known risk factors are mainly tobacco and alcohol consumption, but dietary and environmental factors may also be involved, and the potential role of laryngopharyngeal reflux has been discussed [70]. LSCC is one of the rare cancers with a decreased 5-year survival rate during the last decades [63], and, for this reason, prognostic and predictive markers for LSCC are particularly sought-after. While no prognostic markers are routinely used or recommended in clinical practice for LSCC, a number of potential prognostic markers have been proposed over time, and a comprehensive review has recently been published by Cavaliere and colleagues [71]. The role of HPV infection in cancerogenesis and as a prognostic biomarker is much more controversial, and although a small subset of LSCC may be related to HPV, its clinical and prognostic relevance is probably limited [13,63]. However, most of the available data on p16INK4a expression in LSCC derive from the use of p16IHC as a common technique of choice to demonstrate HPV involvement, often dismissing the possibility of overexpression related to different mechanisms.
By analyzing the literature, two distinct phases of the research effort covering the role of p16ink4a in laryngeal cancer can be identified. The first one, between the 1990s and the turn of 2000, was more focused on the role of P16 inactivation as a step toward cancer progression, and the second phase, which started around the mid-2000s, was more focused on the role of p16ink4a as a surrogate marker of HPV infection and on the role of its overexpression. In fact, almost 30 years ago, in the wake of works defining the inactivation of p16ink4a as a potential step towards cancerogenesis in some head and neck cancers [72,73], the first evidence of the loss of heterozygosity (LOH) at the 9p arm in a subset of laryngeal cancers in a variable percentage of LSCCs emerged [74,75]. Later, along with allelic deletions, both point mutations and promoter hypermethylation were described as relatively frequent events involved in a subset of LSCCs, and LOH in particular seemed to be associated with more advanced and metastatic cases [76]. During this first wave of relevant papers, enough evidence was building around the role of the inactivation of p16ink4a in head and neck cancers to suggest gene therapy as a potential treatment to be considered [77,78]. Notably, p16ink4a inactivation seemed to be present in more than 50% of patients [79]. The role of the LOH and mutations in head and neck cancers was later confirmed via next-generation sequencing analysis [80,81]. It has been suggested that the length of exposure to tobacco and alcohol (but not the intensity) is associated with the homozygous deletion of p16 [82], and that a lack of dietary folates is associated with p16 methylation [83]. An association between weak p16ink4a expression and advanced disease was later confirmed by other authors [79,84], while p16 point mutations were found to be independently associated with the risk of relapse and death in advanced LSCCs, albeit only in a small subset of them [85]. In contemporary papers, p16 anomalies were also found to be potentially associated with the tumor grade [86,87] and invasiveness and regional lymph node metastasis [56,88,89,90]. However, not all research groups looking for a prognostic impact of p16 inactivation found the same results, sometimes confirming a higher frequency of genetic anomalies in more advanced cases [79,91,92,93]. In later years, characterized by papers focused on the potential role of HPV in non-oropharyngeal head and neck cancers, some papers started to point to the fact that while a subset of laryngeal cancers indeed overexpressed p16ink4a (range: 4.7% [94]–39.02% [95], according to the papers cited in this review), this did not seem to reflect relevant HPV involvement in the cancerogenic process, but it was still associated with a trend towards better survival [94,96,97,98] and progression-free survival [94,97]. Relevant exceptions exist at both extremes. In a series of 123 glottic LSCCs, p16-positive cases had a significantly better 2-year disease-free survival and fewer nodal relapses [95]. Allegra et al. found a positive impact on the 5-year overall survival (OS) and disease-specific survival in primarily operated cases, along with fewer nodal metastases [99]. A similar result was found by our group analyzing 95 consecutive LSCCs treated with different modalities. We found a positive impact on the relapse-free survival (RFS) for the whole series and a positive impact on the OS in primarily operated cases [100]. In a study involving 812 patients, Zhu and colleagues found that p16ink4a-positive patients had better OS, disease-specific survival, and RFS [101]. Other studies failed to find any trends or significant correlations between the p16ink4a status and prognosis. Young and colleagues did not find any impact on the 2-year OS or RFS in a cohort of 307 patients, and other groups reported similar results for smaller cohorts [102,103]. Lastly, the paper by Larque and colleagues is a relevant outlier, as they found a better prognosis in patients with a negative p16ink4a status. Notably, they also looked for p16INK4a mRNA expression and gene mutations that did not correlate well with p16INK4a protein expression [104]. Two recent papers, one a propensity-scored analysis of the National Cancer Database for survival outcomes by high-risk Human Papillomavirus status in non-oropharyngeal head and neck squamous cell carcinomas [63], and the other a systematic review and meta-analysis of the survival outcomes in Human Papillomavirus-associated non-oropharyngeal squamous cell carcinomas [13], offer us a broad view of the issue and some interesting insights. Tian and colleagues included in their analysis a total of 4804 LSCC patients, and an HPV+ status was associated with better survival at 1, 2, and 5 years of follow-up. A relevant issue with this paper is that it is focused on the HPV status, meaning that some papers, albeit a minority, did not use p16 to assess the HPV status and thus their analyses do not fairly reflect the impact of p16INK4a on the prognosis. This limitation is overcome in the systematic review and meta-analysis by Sahovaler and colleagues, as they decided to group studies according to their detection techniques. Their paper included 24 studies and 9793 laryngeal cancer patients, and the subgroup analysis showed a significant survival improvement for p16INK4a-positive patients but not for HPV-DNA-positive patients.
What has been discussed up to this point has directly and indirectly highlighted some relevant problems that prevent us from fully understanding the impact of p16INK4a on laryngeal cancer, and especially on its prognosis. As seen, few papers have been published on the matter, and they are often not focused on p16INK4a but prevalently or solely on the role of HPV in laryngeal cancer [105,106]. Moreover, different “non oropharyngeal tumors” are often grouped together, determining a loss of precious data. The discussed papers analyze the impact of p16INK4a on tumors of different stages and treated with different treatment modalities. This could be relevant, as all the papers discussing a significant impact on the prognosis were mainly based on patients treated with surgery as their primary option [95,99,100,101], and the papers that stratified according to the treatment modality found no impact [101] or a negative one [100] on patients treated with nonsurgical modalities. As seen, there are two strikingly distinct phases of the research efforts, one focused on p16INK4a inactivation as a step of LSCC cancerogenesis with a potential negative impact on prognosis, and the other focused on p16INK4a overexpression both as a marker of HPV involvement in LSCC cancerogenesis and as a potential positive prognostic marker in and of itself. The two conditions might very well coexist, and this fact should be reflected properly in future research efforts, as both possibilities should be sought while looking for the prognostic impact of p16INK4a expression. One problem that is tightly linked to the last one is the cutoff chosen to determine p16INK4a expression. The most relevant papers discussed in this review use a wide range of cutoffs, including the following: undefined [97,98]; expression (undefined level) in both the nucleus and cytoplasm [94]; >70% diffuse staining (nuclear and cytoplasmic) [95,101,107]; nuclear staining scored with the intensity reactivity score (IRS) with various cutoffs [99,100]; strong and diffuse (>25%) cytoplasmic and/or nuclear staining [102]; an intensity score of 2 (moderate) or 3 (strong) in ≥30% of tumor cells [103]; nuclear and cytoplasmic staining in >50% of cells [96]; nuclear staining in >50% of cells [108]; strong and diffuse cytoplasmic and nuclear staining in all basal and suprabasal cells [104]. This variability is detrimental to any attempt at a coherent analysis and interpretation of the data. It is not possible to tell by the state of the art how these different cutoffs affected the findings of the papers, or whether any of them correlate better with the clinicopathological characteristics or survival trends. Moreover, one thing to consider is the selection of a cutoff that reflects HPV positivity (a 70% cutoff with nuclear and cytoplasmic expression with at least moderate-to-strong intensity is recommended by the NCCN citing the guideline from the College of American Pathologists [109]), and another is a cutoff that reflects p16INK4a overexpression or inactivation when considering it as an independent marker. The matter is further complicated by early evidence that the staining pattern of p16INK4a might, in and of itself, be predictive of certain clinicopathological characteristics of the tumor [54,110]. Lazăr and colleagues [110] analyzed 88 cases of LSCCs looking for different patterns of the distribution/intensity of the staining and their respective correlations with the clinicopathological characteristics. They found that different patterns were associated with different levels of nodal involvement. A similar observation was made by Zhao and colleagues [54]. Future research focusing on p16INK4a as a prognostic marker will need to properly assess the ideal cutoffs and analyze different staining patterns and their respective associations with the clinicopathological characteristics and survival outcomes.

3. Conclusions

The current body of research is limited and often out of focus. However, especially as clinicians that deal everyday with patients and challenging decisions, we are in desperate need of a prognostic marker, or, even better, a marker that is predictive of the treatment response, capable of mirroring in LSCCs the role of HPV in OPSCCs. The detection of p16INK4a expression via IHC is a good candidate, as it is relatively cheap and accessible and can be applied on paraffin-embedded and formalin-fixed tissues, and it is only limited by its dependance on an experienced anatomopathologist for its interpretation. While its role as a surrogate marker of HPV infection has been largely re-evaluated in the context of LSCCs, and the role of HPV in the cancerogenesis of this particular subsite is probably marginal, it is our opinion that p16 may have some potential as a standalone prognostic marker. While, at the moment, this is only a hypothesis, we have seen that single studies, such as those published by Allegra and colleagues [99,110] and our research group [100], and a recent meta-analysis by Sahovaler and colleagues [13], point in this direction. This hypothesis could be better investigated in the setting of vast and multicentric prospective studies, accounting for both its under- or overexpression and different patterns of expression, to clarify once and for all its eventual prognostic significance and utility in clinical practice. Considering the potential relevance, the latter objective is worth a common scientific effort from the research groups most involved in the field.

Author Contributions

Conceptualization, R.G., L.M. and F.B.; methodology, R.G.; investigation, R.G., L.M. and G.R.; resources, F.B., J.G. and A.A.; writing—original draft preparation, R.G., D.R. and L.M.; writing—review and editing, A.A., J.G. and F.B.; supervision, A.A., J.G. and F.B.; project administration, A.A. and F.B.; funding acquisition, A.A. and F.B. 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

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Gillison, M.L.; Koch, W.M.; Capone, R.B.; Spafford, M.; Westra, W.H.; Wu, L.; Zahurak, M.L.; Daniel, R.W.; Viglione, M.; Symer, D.E.; et al. Evidence for a Causal Association between Human Papillomavirus and a Subset of Head and Neck Cancers. J. Natl. Cancer Inst. 2000, 92, 709–720. [Google Scholar] [CrossRef] [PubMed]
  2. Ragin, C.C.R.; Taioli, E. Survival of Squamous Cell Carcinoma of the Head and Neck in Relation to Human Papillomavirus Infection: Review and Meta-Analysis. Int. J. Cancer 2007, 121, 1813–1820. [Google Scholar] [CrossRef] [PubMed]
  3. Gillison, M.L. HPV and Prognosis for Patients with Oropharynx Cancer. Eur. J. Cancer 2009, 45 (Suppl. 1), 383–385. [Google Scholar] [CrossRef] [PubMed]
  4. 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] [PubMed]
  5. Bussu, F.; Sali, M.; Gallus, R.; Petrone, G.; Zannoni, G.F.; Autorino, R.; Dinapoli, N.; Santangelo, R.; Vellone, V.G.; Graziani, C.; et al. Human Papillomavirus (HPV) Infection in Squamous Cell Carcinomas Arising from the Oropharynx: Detection of HPV DNA and p16 Immunohistochemistry as Diagnostic and Prognostic Indicators—A Pilot Study. Int. J. Radiat. Oncol. Biol. Phys. 2014, 89, 1115–1120. [Google Scholar] [CrossRef]
  6. Lindquist, D.; Romanitan, M.; Hammarstedt, L.; Näsman, A.; Dahlstrand, H.; Lindholm, J.; Onelöv, L.; Ramqvist, T.; Ye, W.; Munck-Wikland, E.; et al. Human Papillomavirus Is a Favourable Prognostic Factor in Tonsillar Cancer and Its Oncogenic Role Is Supported by the Expression of E6 and E7. Mol. Oncol. 2007, 1, 350–355. [Google Scholar] [CrossRef] [PubMed]
  7. Petrelli, F.; Luciani, A.; Ghidini, A.; Cherri, S.; Gamba, P.; Maddalo, M.; Bossi, P.; Zaniboni, A. Treatment de-Escalation for HPV+ Oropharyngeal Cancer: A Systematic Review and Meta-Analysis. Head Neck 2022, 44, 1255–1266. [Google Scholar] [CrossRef]
  8. Klussmann, J.P.; Gültekin, E.; Weissenborn, S.J.; Wieland, U.; Dries, V.; Dienes, H.P.; Eckel, H.E.; Pfister, H.J.; Fuchs, P.G. Expression of p16 Protein Identifies a Distinct Entity of Tonsillar Carcinomas Associated with Human Papillomavirus. Am. J. Pathol. 2003, 162, 747–753. [Google Scholar] [CrossRef]
  9. National Comprehensive Cancer Network. Treatment by Cancer Type. Available online: https://www.nccn.org/guidelines/category_1 (accessed on 27 January 2023).
  10. Amin, M.B.; Greene, F.L.; Edge, S.B.; Compton, C.C.; Gershenwald, J.E.; Brookland, R.K.; Meyer, L.; Gress, D.M.; Byrd, D.R.; Winchester, D.P. The Eighth Edition AJCC Cancer Staging Manual: Continuing to Build a Bridge from a Population-Based to a More “Personalized” Approach to Cancer Staging. CA Cancer J. Clin. 2017, 67, 93–99. [Google Scholar] [CrossRef]
  11. El-Naggar, A.K.; Westra, W.H. p16 Expression as a Surrogate Marker for HPV-Related Oropharyngeal Carcinoma: A Guide for Interpretative Relevance and Consistency. Head Neck 2012, 34, 459–461. [Google Scholar] [CrossRef]
  12. Bussu, F.; Ragin, C.; Boscolo-Rizzo, P.; Rizzo, D.; Gallus, R.; Delogu, G.; Morbini, P.; Tommasino, M. HPV as a Marker for Molecular Characterization in Head and Neck Oncology: Looking for a Standardization of Clinical Use and of Detection Method(s) in Clinical Practice. Head Neck 2019, 41, 1104–1111. [Google Scholar] [CrossRef]
  13. Sahovaler, A.; Kim, M.H.; Mendez, A.; Palma, D.; Fung, K.; Yoo, J.; Nichols, A.C.; MacNeil, S.D. Survival Outcomes in Human Papillomavirus-Associated Nonoropharyngeal Squamous Cell Carcinomas: A Systematic Review and Meta-Analysis. JAMA Otolaryngol. Head Neck Surg. 2020, 146, 1158–1166. [Google Scholar] [CrossRef] [PubMed]
  14. Bussu, F.; Sali, M.; Gallus, R.; Vellone, V.G.; Zannoni, G.F.; Autorino, R.; Dinapoli, N.; Santangelo, R.; Martucci, R.; Graziani, C.; et al. HPV Infection in Squamous Cell Carcinomas Arising from Different Mucosal Sites of the Head and Neck Region. Is p16 Immunohistochemistry a Reliable Surrogate Marker? Br. J. Cancer 2013, 108, 1157–1162. [Google Scholar] [CrossRef] [PubMed]
  15. Serra, S.; Chetty, R.  p16. J. Clin. Pathol. 2018, 71, 853–858. [Google Scholar] [CrossRef] [PubMed]
  16. Stone, S.; Jiang, P.; Dayananth, P.; Tavtigian, S.V.; Katcher, H.; Parry, D.; Peters, G.; Kamb, A. Complex Structure and Regulation of the P16 (MTS1) Locus. Cancer Res. 1995, 55, 2988–2994. [Google Scholar] [PubMed]
  17. Mao, L.; Merlo, A.; Bedi, G.; Shapiro, G.I.; Edwards, C.D.; Rollins, B.J.; Sidransky, D. A Novel p16INK4A Transcript. Cancer Res. 1995, 55, 2995–2997. [Google Scholar] [PubMed]
  18. Serrano, M.; Hannon, G.J.; Beach, D. A New Regulatory Motif in Cell-Cycle Control Causing Specific Inhibition of Cyclin D/CDK4. Nature 1993, 366, 704–707. [Google Scholar] [CrossRef]
  19. Serrano, M. The Tumor Suppressor Protein p16INK4a. Exp. Cell Res. 1997, 237, 7–13. [Google Scholar] [CrossRef]
  20. Komata, T.; Kanzawa, T.; Takeuchi, H.; Germano, I.M.; Schreiber, M.; Kondo, Y.; Kondo, S. Antitumour Effect of Cyclin-Dependent Kinase Inhibitors (p16(INK4A), p18(INK4C), p19(INK4D), p21(WAF1/CIP1) and p27(KIP1)) on Malignant Glioma Cells. Br. J. Cancer 2003, 88, 1277–1280. [Google Scholar] [CrossRef]
  21. Li, J.; Poi, M.J.; Tsai, M.-D. Regulatory Mechanisms of Tumor Suppressor P16(INK4A) and Their Relevance to Cancer. Biochemistry 2011, 50, 5566–5582. [Google Scholar] [CrossRef]
  22. Li, Y.; Nichols, M.A.; Shay, J.W.; Xiong, Y. Transcriptional Repression of the D-Type Cyclin-Dependent Kinase Inhibitor p16 by the Retinoblastoma Susceptibility Gene Product pRb. Cancer Res. 1994, 54, 6078–6082. [Google Scholar] [PubMed]
  23. Schmitt, C.A.; Fridman, J.S.; Yang, M.; Lee, S.; Baranov, E.; Hoffman, R.M.; Lowe, S.W. A Senescence Program Controlled by p53 and p16INK4a Contributes to the Outcome of Cancer Therapy. Cell 2002, 109, 335–346. [Google Scholar] [CrossRef]
  24. Bringold, F.; Serrano, M. Tumor Suppressors and Oncogenes in Cellular Senescence. Exp. Gerontol. 2000, 35, 317–329. [Google Scholar] [CrossRef] [PubMed]
  25. Sanchez-Cespedes, M.; Reed, A.L.; Buta, M.; Wu, L.; Westra, W.H.; Herman, J.G.; Yang, S.C.; Jen, J.; Sidransky, D. Inactivation of the INK4A/ARF Locus Frequently Coexists with TP53 Mutations in Non-Small Cell Lung Cancer. Oncogene 1999, 18, 5843–5849. [Google Scholar] [CrossRef] [PubMed]
  26. Nishiwaki, E.; Turner, S.L.; Harju, S.; Miyazaki, S.; Kashiwagi, M.; Koh, J.; Serizawa, H. Regulation of CDK7-Carboxyl-Terminal Domain Kinase Activity by the Tumor Suppressor p16(INK4A) Contributes to Cell Cycle Regulation. Mol. Cell. Biol. 2000, 20, 7726–7734. [Google Scholar] [CrossRef] [PubMed]
  27. Choi, B.Y.; Choi, H.S.; Ko, K.; Cho, Y.-Y.; Zhu, F.; Kang, B.S.; Ermakova, S.P.; Ma, W.-Y.; Bode, A.M.; Dong, Z. The Tumor Suppressor p16(INK4a) Prevents Cell Transformation through Inhibition of c-Jun Phosphorylation and AP-1 Activity. Nat. Struct. Mol. Biol. 2005, 12, 699–707. [Google Scholar] [CrossRef] [PubMed]
  28. Serizawa, H. Cyclin-Dependent Kinase Inhibitor p16INK4A Inhibits Phosphorylation of RNA Polymerase II by General Transcription Factor TFIIH. J. Biol. Chem. 1998, 273, 5427–5430. [Google Scholar] [CrossRef]
  29. Romagosa, C.; Simonetti, S.; López-Vicente, L.; Mazo, A.; Lleonart, M.E.; Castellvi, J.; Ramon y Cajal, S. p16(Ink4a) Overexpression in Cancer: A Tumor Suppressor Gene Associated with Senescence and High-Grade Tumors. Oncogene 2011, 30, 2087–2097. [Google Scholar] [CrossRef]
  30. Demaria, M.; Ohtani, N.; Youssef, S.A.; Rodier, F.; Toussaint, W.; Mitchell, J.R.; Laberge, R.-M.; Vijg, J.; Van Steeg, H.; Dollé, M.E.T.; et al. An Essential Role for Senescent Cells in Optimal Wound Healing through Secretion of PDGF-AA. Dev. Cell 2014, 31, 722–733. [Google Scholar] [CrossRef]
  31. Ding, Q.; Liu, H.; Liu, L.; Ma, C.; Qin, H.; Wei, Y.; Ren, Y. Deletion of p16 Accelerates Fracture Healing in Geriatric Mice. Am. J. Transl. Res. 2021, 13, 11107–11125. [Google Scholar]
  32. Shi, J.; Sun, J.; Liu, L.; Shan, T.; Meng, H.; Yang, T.; Wang, S.; Wei, T.; Chen, B.; Ma, Y.; et al. P16ink4a Overexpression Ameliorates Cardiac Remodeling of Mouse Following Myocardial Infarction via CDK4/pRb Pathway. Biochem. Biophys. Res. Commun. 2022, 595, 62–68. [Google Scholar] [CrossRef] [PubMed]
  33. Adam, R.C.; Fuchs, E. The Yin and Yang of Chromatin Dynamics In Stem Cell Fate Selection. Trends Genet. 2016, 32, 89–100. [Google Scholar] [CrossRef] [PubMed]
  34. Bantubungi, K.; Hannou, S.-A.; Caron-Houde, S.; Vallez, E.; Baron, M.; Lucas, A.; Bouchaert, E.; Paumelle, R.; Tailleux, A.; Staels, B. Cdkn2a/p16Ink4a Regulates Fasting-Induced Hepatic Gluconeogenesis through the PKA-CREB-PGC1α Pathway. Diabetes 2014, 63, 3199–3209. [Google Scholar] [CrossRef] [PubMed]
  35. Deleye, Y.; Cotte, A.K.; Hannou, S.A.; Hennuyer, N.; Bernard, L.; Derudas, B.; Caron, S.; Legry, V.; Vallez, E.; Dorchies, E.; et al. CDKN2A/p16INK4a Suppresses Hepatic Fatty Acid Oxidation through the AMPKα2-SIRT1-PPARα Signaling Pathway. J. Biol. Chem. 2020, 295, 17310–17322. [Google Scholar] [CrossRef] [PubMed]
  36. Safwan-Zaiter, H.; Wagner, N.; Wagner, K.-D. P16INK4A-More Than a Senescence Marker. Life 2022, 12, 1332. [Google Scholar] [CrossRef]
  37. Orlow, I.; Begg, C.B.; Cotignola, J.; Roy, P.; Hummer, A.J.; Clas, B.A.; Mujumdar, U.; Canchola, R.; Armstrong, B.K.; Kricker, A.; et al. CDKN2A Germline Mutations in Individuals with Cutaneous Malignant Melanoma. J. Investig. Dermatol. 2007, 127, 1234–1243. [Google Scholar] [CrossRef] [PubMed]
  38. Magnusson, S.; Borg, A.; Kristoffersson, U.; Nilbert, M.; Wiebe, T.; Olsson, H. Higher Occurrence of Childhood Cancer in Families with Germline Mutations in BRCA2, MMR and CDKN2A Genes. Fam. Cancer 2008, 7, 331–337. [Google Scholar] [CrossRef]
  39. Monnerat, C.; Chompret, A.; Kannengiesser, C.; Avril, M.-F.; Janin, N.; Spatz, A.; Guinebretière, J.-M.; Marian, C.; Barrois, M.; Boitier, F.; et al. BRCA1, BRCA2, TP53, and CDKN2A Germline Mutations in Patients with Breast Cancer and Cutaneous Melanoma. Fam. Cancer 2007, 6, 453–461. [Google Scholar] [CrossRef]
  40. Fukushima, N.; Sato, N.; Ueki, T.; Rosty, C.; Walter, K.M.; Wilentz, R.E.; Yeo, C.J.; Hruban, R.H.; Goggins, M. Aberrant Methylation of Preproenkephalin and p16 Genes in Pancreatic Intraepithelial Neoplasia and Pancreatic Ductal Adenocarcinoma. Am. J. Pathol. 2002, 160, 1573–1581. [Google Scholar] [CrossRef]
  41. Paulson, T.G.; Galipeau, P.C.; Xu, L.; Kissel, H.D.; Li, X.; Blount, P.L.; Sanchez, C.A.; Odze, R.D.; Reid, B.J. p16 Mutation Spectrum in the Premalignant Condition Barrett’s Esophagus. PLoS ONE 2008, 3, e3809. [Google Scholar] [CrossRef]
  42. Guida, M.; Sanguedolce, F.; Bufo, P.; Di Spiezio Sardo, A.; Bifulco, G.; Nappi, C.; Pannone, G. Aberrant DNA Hypermethylation of hMLH-1 and CDKN2A/p16 Genes in Benign, Premalignant and Malignant Endometrial Lesions. Eur. J. Gynaecol. Oncol. 2009, 30, 267–270. [Google Scholar] [PubMed]
  43. Schwartz, B.; Avivi-Green, C.; Polak-Charcon, S. Sodium Butyrate Induces Retinoblastoma Protein Dephosphorylation, p16 Expression and Growth Arrest of Colon Cancer Cells. Mol. Cell. Biochem. 1998, 188, 21–30. [Google Scholar] [CrossRef] [PubMed]
  44. Milde-Langosch, K.; Bamberger, A.M.; Rieck, G.; Kelp, B.; Löning, T. Overexpression of the p16 Cell Cycle Inhibitor in Breast Cancer Is Associated with a More Malignant Phenotype. Breast Cancer Res. Treat. 2001, 67, 61–70. [Google Scholar] [CrossRef] [PubMed]
  45. Arifin, M.T.; Hama, S.; Kajiwara, Y.; Sugiyama, K.; Saito, T.; Matsuura, S.; Yamasaki, F.; Arita, K.; Kurisu, K. Cytoplasmic, but Not Nuclear, p16 Expression May Signal Poor Prognosis in High-Grade Astrocytomas. J. Neurooncol. 2006, 77, 273–277. [Google Scholar] [CrossRef]
  46. O’Neill, C.J.; McBride, H.A.; Connolly, L.E.; McCluggage, W.G. Uterine Leiomyosarcomas Are Characterized by High p16, p53 and MIB1 Expression in Comparison with Usual Leiomyomas, Leiomyoma Variants and Smooth Muscle Tumours of Uncertain Malignant Potential. Histopathology 2007, 50, 851–858. [Google Scholar] [CrossRef]
  47. Lam, A.K.-Y.; Ong, K.; Giv, M.J.; Ho, Y.-H. p16 Expression in Colorectal Adenocarcinoma: Marker of Aggressiveness and Morphological Types. Pathology 2008, 40, 580–585. [Google Scholar] [CrossRef]
  48. Zhao, P.; Mao, X.; Talbot, I.C. Aberrant Cytological Localization of p16 and CDK4 in Colorectal Epithelia in the Normal Adenoma Carcinoma Sequence. World J. Gastroenterol. 2006, 12, 6391–6396. [Google Scholar] [CrossRef]
  49. Shen, W.-W.; Wu, J.; Cai, L.; Liu, B.-Y.; Gao, Y.; Chen, G.-Q.; Fu, G.-H. Expression of Anion Exchanger 1 Sequestrates p16 in the Cytoplasm in Gastric and Colonic Adenocarcinoma. Neoplasia 2007, 9, 812–819. [Google Scholar] [CrossRef]
  50. Di Vinci, A.; Perdelli, L.; Banelli, B.; Salvi, S.; Casciano, I.; Gelvi, I.; Allemanni, G.; Margallo, E.; Gatteschi, B.; Romani, M. p16(INK4a) Promoter Methylation and Protein Expression in Breast Fibroadenoma and Carcinoma. Int. J. Cancer 2005, 114, 414–421. [Google Scholar] [CrossRef]
  51. Dai, C.Y.; Furth, E.E.; Mick, R.; Koh, J.; Takayama, T.; Niitsu, Y.; Enders, G.H. p16(INK4a) Expression Begins Early in Human Colon Neoplasia and Correlates Inversely with Markers of Cell Proliferation. Gastroenterology 2000, 119, 929–942. [Google Scholar] [CrossRef]
  52. Beasley, M.B.; Lantuejoul, S.; Abbondanzo, S.; Chu, W.-S.; Hasleton, P.S.; Travis, W.D.; Brambilla, E. The P16/cyclin D1/Rb Pathway in Neuroendocrine Tumors of the Lung. Hum. Pathol. 2003, 34, 136–142. [Google Scholar] [CrossRef] [PubMed]
  53. Dabbs, D.J. Diagnostic Immunohistochemistry E-Book: Theranostic and Genomic Applications, 6th ed.; Elsevier Health Sciences: Amsterdam, The Netherlands, 2017. [Google Scholar]
  54. Zhao, N.; Ang, M.-K.; Yin, X.-Y.; Patel, M.R.; Fritchie, K.; Thorne, L.; Muldrew, K.L.; Hayward, M.C.; Sun, W.; Wilkerson, M.D.; et al. Different Cellular p16(INK4a) Localisation May Signal Different Survival Outcomes in Head and Neck Cancer. Br. J. Cancer 2012, 107, 482–490. [Google Scholar] [CrossRef] [PubMed]
  55. DeVita, V.T.; Lawrence, T.S.; Rosenberg, S.A. DeVita, Hellman, and Rosenberg’s Cancer: Principles & Practice of Oncology, 11th ed.; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2008. [Google Scholar]
  56. Fu, Z.-J.; Ma, Z.-Y.; Wang, Q.-R.; Lei, D.-P.; Wang, R.; Liu, C.-X.; Pan, X.-L. Overexpression of CyclinD1 and Underexpression of p16 Correlate with Lymph Node Metastases in Laryngeal Squamous Cell Carcinoma in Chinese Patients. Clin. Exp. Metastasis 2008, 25, 887–892. [Google Scholar] [CrossRef] [PubMed]
  57. Tommasino, M. The Human Papillomavirus Family and Its Role in Carcinogenesis. Semin. Cancer Biol. 2014, 26, 13–21. [Google Scholar] [CrossRef] [PubMed]
  58. Nauta, I.H.; Rietbergen, M.M.; van Bokhoven, A.A.J.D.; Bloemena, E.; Lissenberg-Witte, B.I.; Heideman, D.A.M.; Baatenburg de Jong, R.J.; Brakenhoff, R.H.; Leemans, C.R. Evaluation of the eighth TNM classification on p16-positive oropharyngeal squamous cell carcinomas in the Netherlands and the importance of additional HPV DNA testing. Ann. Oncol. 2018, 29, 1273–1279. [Google Scholar] [CrossRef] [PubMed]
  59. Wang, H.; Zhang, Y.; Bai, W.; Wang, B.; Wei, J.; Ji, R.; Xin, Y.; Dong, L.; Jiang, X. Feasibility of Immunohistochemical p16 Staining in the Diagnosis of Human Papillomavirus Infection in Patients With Squamous Cell Carcinoma of the Head and Neck: A Systematic Review and Meta-Analysis. Front. Oncol. 2020, 10, 524928. [Google Scholar] [CrossRef]
  60. Mena, M.; Wang, X.; Tous, S.; Quiros, B.; Clavero, O.; Alejo, M.; Morey, F.; Taberna, M.; Leon Vintro, X.; Lloveras Rubio, B.; et al. Concordance of p16 and E6*I mRNA among HPV-DNA-Positive Oropharyngeal, Laryngeal, and Oral Cavity Carcinomas from the ICO International Study. Cancers 2022, 14, 3787. [Google Scholar] [CrossRef]
  61. Bussu, F.; Muresu, N.; Crescio, C.; Gallus, R.; Rizzo, D.; Cossu, A.; Sechi, I.; Fedeli, M.; Cossu, A.; Delogu, G.; et al. Low Prevalence of HPV Related Oropharyngeal Carcinogenesis in Northern Sardinia. Cancers 2022, 14, 4205. [Google Scholar] [CrossRef]
  62. Gallus, R.; Nauta, I.H.; Marklund, L.; Rizzo, D.; Crescio, C.; Mureddu, L.; Tropiano, P.; Delogu, G.; Bussu, F. Accuracy of p16 IHC in Classifying HPV-Driven OPSCC in Different Populations. Cancers 2023, 15, 656. [Google Scholar] [CrossRef]
  63. Tian, S.; Switchenko, J.M.; Jhaveri, J.; Cassidy, R.J.; Ferris, M.J.; Press, R.H.; Pfister, N.T.; Patel, M.R.; Saba, N.F.; McDonald, M.W.; et al. Survival Outcomes by High-Risk Human Papillomavirus Status in Nonoropharyngeal Head and Neck Squamous Cell Carcinomas: A Propensity-Scored Analysis of the National Cancer Data Base. Cancer 2019, 125, 2782–2793. [Google Scholar] [CrossRef]
  64. Straume, O.; Sviland, L.; Akslen, L.A. Loss of Nuclear p16 Protein Expression Correlates with Increased Tumor Cell Proliferation (Ki-67) and Poor Prognosis in Patients with Vertical Growth Phase Melanoma. Clin. Cancer Res. 2000, 6, 1845–1853. [Google Scholar] [PubMed]
  65. Chakravarti, A.; DeSilvio, M.; Zhang, M.; Grignon, D.; Rosenthal, S.; Asbell, S.O.; Hanks, G.; Sandler, H.M.; Khor, L.-Y.; Pollack, A.; et al. Prognostic Value of p16 in Locally Advanced Prostate Cancer: A Study Based on Radiation Therapy Oncology Group Protocol 9202. J. Clin. Oncol. 2007, 25, 3082–3089. [Google Scholar] [CrossRef] [PubMed]
  66. Wang, C.-W.; Wu, T.-I.; Yu, C.-T.; Wu, Y.-C.; Teng, Y.-H.; Chin, S.-Y.; Lai, C.-H.; Chen, T.-C. Usefulness of p16 for Differentiating Primary Pulmonary Squamous Cell Carcinoma From Cervical Squamous Cell Carcinoma Metastatic to the Lung. Am. J. Clin. Pathol. 2009, 131, 715–722. [Google Scholar] [CrossRef] [PubMed]
  67. Tong, J.; Sun, X.; Cheng, H.; Zhao, D.; Ma, J.; Zhen, Q.; Cao, Y.; Zhu, H.; Bai, J. Expression of p16 in Non-Small Cell Lung Cancer and Its Prognostic Significance: A Meta-Analysis of Published Literatures. Lung Cancer 2011, 74, 155–163. [Google Scholar] [CrossRef] [PubMed]
  68. Park, J.W.; Kang, J.; Lim, K.Y.; Kim, H.; Kim, S.-I.; Won, J.K.; Park, C.-K.; Park, S.-H. The Prognostic Significance of p16 Expression Pattern in Diffuse Gliomas. J. Pathol. Transl. Med. 2020, 55, 102–111. [Google Scholar] [CrossRef] [PubMed]
  69. Steigen, S.E.; Bjerkehagen, B.; Haugland, H.K.; Nordrum, I.S.; Løberg, E.M.; Isaksen, V.; Eide, T.J.; Nielsen, T.O. Diagnostic and Prognostic Markers for Gastrointestinal Stromal Tumors in Norway. Mod. Pathol. 2008, 21, 46–53. [Google Scholar] [CrossRef] [PubMed]
  70. Steuer, C.E.; El-Deiry, M.; Parks, J.R.; Higgins, K.A.; Saba, N.F. An Update on Larynx Cancer. CA Cancer J. Clin. 2017, 67, 31–50. [Google Scholar] [CrossRef] [PubMed]
  71. Cavaliere, M.; Bisogno, A.; Scarpa, A.; D’Urso, A.; Marra, P.; Colacurcio, V.; De Luca, P.; Ralli, M.; Cassandro, E.; Cassandro, C. Biomarkers of Laryngeal Squamous Cell Carcinoma: A Review. Ann. Diagn. Pathol. 2021, 54, 151787. [Google Scholar] [CrossRef]
  72. Zhang, S.Y.; Klein-Szanto, A.J.; Sauter, E.R.; Shafarenko, M.; Mitsunaga, S.; Nobori, T.; Carson, D.A.; Ridge, J.A.; Goodrow, T.L. Higher Frequency of Alterations in the p16/CDKN2 Gene in Squamous Cell Carcinoma Cell Lines than in Primary Tumors of the Head and Neck. Cancer Res. 1994, 54, 5050–5053. [Google Scholar]
  73. Califano, J.; van der Riet, P.; Westra, W.; Nawroz, H.; Clayman, G.; Piantadosi, S.; Corio, R.; Lee, D.; Greenberg, B.; Koch, W.; et al. Genetic Progression Model for Head and Neck Cancer: Implications for Field Cancerization. Cancer Res. 1996, 56, 2488–2492. [Google Scholar] [CrossRef]
  74. Kiaris, H.; Spanakis, N.; Ergazaki, M.; Sourvinos, G.; Spandidos, D.A. Loss of Heterozygosity at 9p and 17q in Human Laryngeal Tumors. Cancer Lett. 1995, 97, 129–134. [Google Scholar] [CrossRef] [PubMed]
  75. Louhelainen, J.; Szyfter, K.; Szyfter, W.; Hemminki, K. Loss of Heterozygosity and Microsatellite Instability in Larynx Cancer. Int. J. Oncol. 1997, 10, 247–252. [Google Scholar] [CrossRef] [PubMed]
  76. Jares, P.; Fernández, P.L.; Nadal, A.; Cazorla, M.; Hernández, L.; Pinyol, M.; Hernández, S.; Traserra, J.; Cardesa, A.; Campo, E. p16MTS1/CDK4I Mutations and Concomitant Loss of Heterozygosity at 9p21–23 Are Frequent Events in Squamous Cell Carcinoma of the Larynx. Oncogene 1997, 15, 1445–1453. [Google Scholar] [CrossRef] [PubMed]
  77. Rocco, J.W.; Li, D.; Liggett, W.H., Jr.; Duan, L.; Saunders, J.K., Jr.; Sidransky, D.; O’Malley, B.W., Jr. p16INK4A Adenovirus-Mediated Gene Therapy for Human Head and Neck Squamous Cell Cancer. Clin. Cancer Res. 1998, 4, 1697–1704. [Google Scholar] [PubMed]
  78. Rhee, J.G.; Li, D.; O’Malley, B.W., Jr.; Suntharalingam, M. Combination Radiation and Adenovirus-Mediated P16(INK4A) Gene Therapy in a Murine Model for Head and Neck Cancer. ORL J. Otorhinolaryngol. Relat. Spec. 2003, 65, 144–154. [Google Scholar] [CrossRef] [PubMed]
  79. Yuen, P.W.; Man, M.; Lam, K.Y.; Kwong, Y.L. Clinicopathological Significance of p16 Gene Expression in the Surgical Treatment of Head and Neck Squamous Cell Carcinomas. J. Clin. Pathol. 2002, 55, 58–60. [Google Scholar] [CrossRef] [PubMed]
  80. Loyo, M.; Li, R.J.; Bettegowda, C.; Pickering, C.R.; Frederick, M.J.; Myers, J.N.; Agrawal, N. Lessons Learned from next-Generation Sequencing in Head and Neck Cancer. Head Neck 2013, 35, 454–463. [Google Scholar] [CrossRef]
  81. Nichols, A.C.; Yoo, J.; Palma, D.A.; Fung, K.; Franklin, J.H.; Koropatnick, J.; Mymryk, J.S.; Batada, N.N.; Barrett, J.W. Frequent Mutations in TP53 and CDKN2A Found by next-Generation Sequencing of Head and Neck Cancer Cell Lines. Arch. Otolaryngol. Head Neck Surg. 2012, 138, 732–739. [Google Scholar] [CrossRef]
  82. Kraunz, K.S.; McClean, M.D.; Nelson, H.H.; Peters, E.; Calderon, H.; Kelsey, K.T. Duration but Not Intensity of Alcohol and Tobacco Exposure Predicts p16INK4A Homozygous Deletion in Head and Neck Squamous Cell Carcinoma. Cancer Res. 2006, 66, 4512–4515. [Google Scholar] [CrossRef]
  83. Kraunz, K.S.; Hsiung, D.; McClean, M.D.; Liu, M.; Osanyingbemi, J.; Nelson, H.H.; Kelsey, K.T. Dietary Folate Is Associated with p16(INK4A) Methylation in Head and Neck Squamous Cell Carcinoma. Int. J. Cancer 2006, 119, 1553–1557. [Google Scholar] [CrossRef]
  84. Krecicki, T.; Smigiel, R.; Fraczek, M.; Kowalczyk, M.; Sasiadek, M.M. Studies of the Cell Cycle Regulatory Proteins P16, Cyclin D1 and Retinoblastoma Protein in Laryngeal Carcinoma Tissue. J. Laryngol. Otol. 2004, 118, 676–680. [Google Scholar] [CrossRef] [PubMed]
  85. Bazan, V.; Zanna, I.; Migliavacca, M.; Sanz-Casla, M.T.; Maestro, M.L.; Corsale, S.; Macaluso, M.; Dardanoni, G.; Restivo, S.; Quintela, P.L.; et al. Prognostic Significance of p16INK4a Alterations and 9p21 Loss of Heterozygosity in Locally Advanced Laryngeal Squamous Cell Carcinoma. J. Cell. Physiol. 2002, 192, 286–293. [Google Scholar] [CrossRef] [PubMed]
  86. Smigiel, R.; Sasiadek, M.; Krecicki, T.; Ramsey, D.; Jagielski, J.; Blin, N. Inactivation of the Cyclin-Dependent Kinase Inhibitor 2A (CDKN2A) Gene in Squamous Cell Carcinoma of the Larynx. Mol. Carcinog. 2004, 39, 147–154. [Google Scholar] [CrossRef]
  87. Sasiadek, M.M.; Stembalska-Kozlowska, A.; Smigiel, R.; Ramsey, D.; Kayademir, T.; Blin, N. Impairment of MLH1 and CDKN2A in Oncogenesis of Laryngeal Cancer. Br. J. Cancer 2004, 90, 1594–1599. [Google Scholar] [CrossRef] [PubMed]
  88. Huang, H.; Cui, Y.; Tang, D.; Tao, Y.; Liu, Q. Correlation of p16 mutation and biological behavior in Chinese laryngeal cancer. Lin Chuang Er Bi Yan Hou Ke Za Zhi = J. Clin. Otorhinolaryngol. 2001, 15, 253–254. [Google Scholar]
  89. Allegra, E.; Caltabiano, R.; Amorosi, A.; Vasquez, E.; Garozzo, A.; Puzzo, L. Expression of BMI1 and p16 in Laryngeal Squamous Cell Carcinoma. Head Neck 2013, 35, 847–851. [Google Scholar] [CrossRef]
  90. Pierini, S.; Jordanov, S.H.; Mitkova, A.V.; Chalakov, I.J.; Melnicharov, M.B.; Kunev, K.V.; Mitev, V.I.; Kaneva, R.P.; Goranova, T.E. Promoter Hypermethylation of CDKN2A, MGMT, MLH1, and DAPK Genes in Laryngeal Squamous Cell Carcinoma and Their Associations with Clinical Profiles of the Patients. Head Neck 2014, 36, 1103–1108. [Google Scholar] [CrossRef]
  91. El-Salahy, E.; Abou-Ghalia, A.H.; Adli, A.; Kassim, S.K. The Cell Cycle Regulators and Cyclin D1: Relationship to Clinicopathological Parameters and Disease-Free Survival in Laryngeal Carcinoma Patients. Cancer Genom. Proteom. 2005, 2, 239–245. [Google Scholar]
  92. Koscielny, S.; Dahse, R.; Ernst, G.; von Eggeling, F. The Prognostic Relevance of p16 Inactivation in Head and Neck Cancer. ORL J. Otorhinolaryngol. Relat. Spec. 2007, 69, 30–36. [Google Scholar] [CrossRef]
  93. Swellam, M.; El-Arab, L.R.E.; Adly, A. Prognostic Value of Cell-Cycle Regulators and Cellular Biomarkers in Laryngeal Squamous Cell Carcinoma. Clin. Biochem. 2008, 41, 1059–1066. [Google Scholar] [CrossRef]
  94. Murakami, N.; Mori, T.; Machida, R.; Kodaira, T.; Ito, Y.; Shikama, N.; Konishi, K.; Matsumoto, Y.; Murakami, Y.; Nakamura, N.; et al. Prognostic Value of Epithelial Cell Adhesion Molecules in T1-2N0M0 Glottic Cancer. Laryngoscope 2021, 131, 1522–1527. [Google Scholar] [CrossRef] [PubMed]
  95. Sánchez Barrueco, A.; González Galán, F.; Villacampa Aubá, J.M.; Díaz Tapia, G.; Fernández Hernández, S.; Martín-Arriscado Arroba, C.; Cenjor Español, C.; Almodóvar Álvarez, C. p16 Influence on Laryngeal Squamous Cell Carcinoma Relapse and Survival. Otolaryngol. Head Neck Surg. 2019, 160, 1042–1047. [Google Scholar] [CrossRef] [PubMed]
  96. Chernock, R.D.; Wang, X.; Gao, G.; Lewis, J.S., Jr.; Zhang, Q.; Thorstad, W.L.; El-Mofty, S.K. Detection and Significance of Human Papillomavirus, CDKN2A(p16) and CDKN1A(p21) Expression in Squamous Cell Carcinoma of the Larynx. Mod. Pathol. 2013, 26, 223–231. [Google Scholar] [CrossRef] [PubMed]
  97. Chung, C.H.; Zhang, Q.; Kong, C.S.; Harris, J.; Fertig, E.J.; Harari, P.M.; Wang, D.; Redmond, K.P.; Shenouda, G.; Trotti, A.; et al. p16 Protein Expression and Human Papillomavirus Status as Prognostic Biomarkers of Nonoropharyngeal Head and Neck Squamous Cell Carcinoma. J. Clin. Oncol. 2014, 32, 3930–3938. [Google Scholar] [CrossRef] [PubMed]
  98. Dogantemur, S.; Ozdemir, S.; Uguz, A.; Surmelioglu, O.; Dagkiran, M.; Tarkan, O.; Tuncer, U. Assessment of HPV 16, HPV 18, p16 Expression in Advanced Stage Laryngeal Cancer Patients and Prognostic Significance. Braz. J. Otorhinolaryngol. 2020, 86, 351–357. [Google Scholar] [CrossRef]
  99. Allegra, E.; Bianco, M.R.; Mignogna, C.; Caltabiano, R.; Grasso, M.; Puzzo, L. Role of P16 Expression in the Prognosis of Patients With Laryngeal Cancer: A Single Retrospective Analysis. Cancer Control 2021, 28, 10732748211033544. [Google Scholar] [CrossRef]
  100. Gallus, R.; Gheit, T.; Holzinger, D.; Petrillo, M.; Rizzo, D.; Petrone, G.; Miccichè, F.; Mattiucci, G.C.; Arciuolo, D.; Capobianco, G.; et al. Prevalence of HPV Infection and p16 Overexpression in Surgically Treated Laryngeal Squamous Cell Carcinoma. Vaccines 2022, 10, 204. [Google Scholar] [CrossRef]
  101. Zhu, Y.; Xia, X.; Gross, N.; Dahlstrom, K.R.; Gao, L.; Liang, Z.; Gao, Z.; Wei, P.; Liu, C.; Li, G.; et al. Prognostic Implications of Human Papillomavirus Status and p16 Expression in Laryngeal Squamous Cell Carcinoma. Head Neck 2019, 41, 4151–4163. [Google Scholar] [CrossRef]
  102. Lam, E.W.H.; Chan, M.M.H.; Wai, C.K.C.; Ngai, C.M.; Chen, Z.; Wong, M.C.S.; Yeung, A.C.M.; Tong, J.H.M.; Chan, A.B.W.; To, K.-F.; et al. The Role of Human Papillomavirus in Laryngeal Cancer in Southern China. J. Med. Virol. 2018, 90, 1150–1159. [Google Scholar] [CrossRef]
  103. Young, R.J.; Urban, D.; Angel, C.; Corry, J.; Lyons, B.; Vallance, N.; Kleid, S.; Iseli, T.A.; Solomon, B.; Rischin, D. Frequency and Prognostic Significance of p16(INK4A) Protein Overexpression and Transcriptionally Active Human Papillomavirus Infection in Laryngeal Squamous Cell Carcinoma. Br. J. Cancer 2015, 112, 1098–1104. [Google Scholar] [CrossRef]
  104. Larque, A.B.; Conde, L.; Hakim, S.; Alos, L.; Jares, P.; Vilaseca, I.; Cardesa, A.; Nadal, A. P16(INK4a) Overexpression Is Associated with CDKN2A Mutation and Worse Prognosis in HPV-Negative Laryngeal Squamous Cell Carcinomas. Virchows Arch. 2015, 466, 375–382. [Google Scholar] [CrossRef] [PubMed]
  105. Hernandez, B.Y.; Goodman, M.T.; Lynch, C.F.; Cozen, W.; Unger, E.R.; Steinau, M.; Thompson, T.; Saber, M.S.; Altekruse, S.F.; Lyu, C.; et al. Human Papillomavirus Prevalence in Invasive Laryngeal Cancer in the United States. PLoS ONE 2014, 9, e115931. [Google Scholar] [CrossRef] [PubMed]
  106. Chen, W.-C.; Chuang, H.-C.; Lin, Y.-T.; Huang, C.-C.; Chien, C.-Y. Clinical Impact of Human Papillomavirus in Laryngeal Squamous Cell Carcinoma: A Retrospective Study. PeerJ 2017, 5, e3395. [Google Scholar] [CrossRef] [PubMed]
  107. Dahm, V.; Haitel, A.; Kaider, A.; Stanisz, I.; Beer, A.; Lill, C. Cancer Stage and Pack-Years, but Not p16 or HPV, Are Relevant for Survival in Hypopharyngeal and Laryngeal Squamous Cell Carcinomas. Eur. Arch. Otorhinolaryngol. 2018, 275, 1837–1843. [Google Scholar] [CrossRef]
  108. Salazar, C.R.; Anayannis, N.; Smith, R.V.; Wang, Y.; Haigentz, M., Jr.; Garg, M.; Schiff, B.A.; Kawachi, N.; Elman, J.; Belbin, T.J.; et al. Combined P16 and Human Papillomavirus Testing Predicts Head and Neck Cancer Survival. Int. J. Cancer 2014, 135, 2404–2412. [Google Scholar] [CrossRef]
  109. Lewis, J.S., Jr.; 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]
  110. Lazăr, C.S.; Şovrea, A.S.; Georgiu, C.; Crişan, D.; Mirescu, Ş.C.; Cosgarea, M. Different Patterns of p16INK4a Immunohistochemical Expression and Their Biological Implications in Laryngeal Squamous Cell Carcinoma. Rom. J. Morphol. Embryol. 2020, 61, 697–706. [Google Scholar] [CrossRef]
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

Gallus, R.; Rizzo, D.; Rossi, G.; Mureddu, L.; Galli, J.; Artuso, A.; Bussu, F. p16 Expression in Laryngeal Squamous Cell Carcinoma: A Surrogate or Independent Prognostic Marker? Pathogens 2024, 13, 100. https://doi.org/10.3390/pathogens13020100

AMA Style

Gallus R, Rizzo D, Rossi G, Mureddu L, Galli J, Artuso A, Bussu F. p16 Expression in Laryngeal Squamous Cell Carcinoma: A Surrogate or Independent Prognostic Marker? Pathogens. 2024; 13(2):100. https://doi.org/10.3390/pathogens13020100

Chicago/Turabian Style

Gallus, Roberto, Davide Rizzo, Giorgia Rossi, Luca Mureddu, Jacopo Galli, Alberto Artuso, and Francesco Bussu. 2024. "p16 Expression in Laryngeal Squamous Cell Carcinoma: A Surrogate or Independent Prognostic Marker?" Pathogens 13, no. 2: 100. https://doi.org/10.3390/pathogens13020100

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