Oral Oncology
Oral Oncology
                                                                              Oral Oncology
                                                       journal homepage: www.elsevier.com/locate/oraloncology
Review
From infection to immortality: The role of HPV and telomerase in head and
neck cancer☆
Silvia Giunco a,b, Annarosa Del Mistro b , Marzia Morello b, Jacopo Lidonnici a, Helena Frayle b,
Silvia Gori b , Anita De Rossi a,* , Paolo Boscolo-Rizzo c
a
    Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova, 35128 Padova, Italy
b
    Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV – IRCCS, 35128 Padova, Italy
c
    Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
A R T I C L E I N F O A B S T R A C T
Keywords:                                                   Head and neck squamous cell carcinomas (HNSCCs) represent a heterogeneous group of malignancies with
HPV                                                         multifactorial aetiologies. High-risk human papillomavirus (hrHPV) infections, particularly HPV16, and the
Telomerase                                                  dysregulation of telomerase activity, specifically through its catalytic subunit, telomerase reverse transcriptase
TERT
                                                            (TERT) are among the key contributors to HNSCC development and progression. HPV promotes oncogenesis via
HNSCC
Carcinogenesis
                                                            the E6 and E7 oncoproteins, which inactivate tumour suppressors TP53 and RB1, leading to unchecked cellular
Tumour progression                                          proliferation. Concurrently, telomerase activation plays a critical role in HNSCC by maintaining telomere length,
Therapeutic targets                                         thus enabling cellular immortality, and facilitating tumour development and progression. The interplay between
                                                            HPV and telomerase is significant; HPV oncoprotein E6 enhances telomerase activity through multiple regulatory
                                                            mechanisms, including upregulating TERT expression. Beyond telomere maintenance, TERT influences signalling
                                                            pathways, cellular metabolism, and the tumour microenvironment, contributing to aggressive tumour behaviour
                                                            and poor prognosis. This review integrates the roles of HPV and telomerase in HNSCC, focusing on their mo
                                                            lecular mechanisms and interactions that drive carcinogenesis and influence disease progression. Understanding
                                                            the synergistic effects of HPV and TERT in HNSCC may be crucial for risk stratification, prognostic assessment,
                                                            and the development of novel therapeutic strategies targeting these specific molecular pathways.
HPV and telomerase: Key players in HNSCC                                                         have been observed in the incidence of oropharyngeal squamous cell
                                                                                                 carcinoma (OPSCC), primarily due to the rise in HPV-associated carci
    Globally, in 2022 head and neck cancer represented 4 % of all can                           nomas in many countries [6]. HPV-associated OPSCC constitutes a
cers, ranking as the 7th most prevalent cancers worldwide [1]. Partic                           distinct entity with a different clinical presentation, a unique genetic
ularly, head and neck squamous cell carcinomas (HNSCCs) encompass a                              profile [7], a distinct tumour immune environment [8], and increased
heterogeneous group of malignancies (i.e. cancer of the oral cavity,                             sensitivity to chemotherapy and radiotherapy [9]. HPV-associated
oropharynx, larynx and hypopharynx) with diverse drivers contributing                            OPSCC is characterized by a significantly better prognosis than HPV-
to its development and progression [2]. Among these, high risk human                             independent counterpart [10,11] with HPV status being the single
papillomavirus (hrHPV) infection and dysregulated telomerase activity                            most important determinant of outcome [10,11].
have emerged as key players [3,4].                                                                   In parallel, the reactivation of telomerase, an enzymatic complex
    HPV infection, particularly by the oncogenic type 16, promotes                               consisting of the catalytic subunit telomerase reverse transcriptase
neoplastic transformation through the action of the viral oncoproteins                           (TERT) and the telomerase RNA component (TERC), enables immor
E6 and E7, which inactivate the major tumour suppressor proteins,                                talization of cancer cells by maintaining telomere length [12]. Telo
tumour protein p53 (TP53) and RB transcriptional corepressor 1 (RB1),                            merase is upregulated in most HNSCCs through increased expression of
respectively [5]. In the last decades, significant time-trend variations                         TERT enzyme and the RNA component TERC. The interplay between
    This article is part of a special issue entitled: ‘HPV and Head and Neck Cancers’ published in Oral Oncology.
    ☆
 * Corresponding author at: Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova, Via Gattamelata
64, 35128 Padova, Italy.
    E-mail address: anita.derossi@unipd.it (A. De Rossi).
https://doi.org/10.1016/j.oraloncology.2024.107169
Received 14 August 2024; Received in revised form 19 December 2024; Accepted 25 December 2024
Available online 3 January 2025
1368-8375/© 2024 The Author(s). Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-
nc-nd/4.0/).
S. Giunco et al.                                                                                                               Oral Oncology 161 (2025) 107169
HPV and TERT is significant in HNSCC. HPV oncoprotein E6 enhances                 secondary genetic alterations that confer a more aggressive phenotype
telomerase activity through various regulatory mechanisms, aiding in              [24].
cell immortalization and cancer development and progression [13].                     HPV-associated OPSCC typically develops from the reticulated
Besides telomere maintenance, TERT influences signalling pathways,                epithelium covering the crypts of the palatine tonsils and the base of the
cellular metabolism, and the tumour immune microenvironment                       tongue, which represent immunologically privileged sites for HPV-
[14,15], contributing to tumour progression, metastasis, and poor                 transforming infections [25]. Consequently, only 3 % of OPSCCs
prognosis [16,17].                                                                arising from non-tonsillar sites harbour hrHPV [26]. Due to its function
    Understanding the roles of HPV and TERT in HNSCC and their                    in capturing inhaled and ingested antigens and transporting them into
interaction is crucial for risk stratification, prognosis prediction, and         the subepithelial spaces, the reticulated epithelium features a porous
development of novel therapeutic strategies tailored to target specific           basal membrane, consisting of a network of specific collagen types
molecular pathways implicated in HNSCC, such as inhibiting HPV E6-                which facilitate close interaction between the stationary epithelial cells
mediated TERT upregulation and telomerase activity in tumours. This               and the migratory lymphoid cell population [27]. Transformed cells can
review aims to provide an in-depth analysis of the roles played by HPV            thus rapidly and easily spread through efferent lymphatic vessels to
and TERT in HNSCC, as well as to explore the potential interactions               regional cervical lymph nodes. Furthermore, HPV16 E6 and E7 onco
between these two factors in driving carcinogenesis and influencing               gene expression can modify the phenotype of infected tissue stem cells,
disease progression.                                                              thereby increasing the number of migratory cancer stem cells [28].
                                                                                  These metastases often exhibit cystic degeneration and may undergo
From infection to malignancy                                                      rapid changes in size, attributed to the neoplastic cells attempting to
                                                                                  recapitulate the morphology of the originating crypts within the lymph
Molecular mechanisms of HPV-associated carcinogenesis                             nodes [29]. Clinically, this implies that carcinoma in situ is somewhat
                                                                                  elusive in these cases [30], and the presentation of a relatively early-
    Many different HPV types have been characterized so far; all infect           stage tumour with a large cystic lateral cervical lymph node package
the epithelial cells, and distinct types are involved in mucosal and              is highly indicative of a HPV-associated OPSCC [31], and in many cases
cutaneous infections [18]. Among the mucosal HPV types, only a subset             the primary tumour remains occult [32,33]. Compared to the cervix,
is associated with cancer, and constitute the hrHPV group, that includes          specific features of the oropharynx include a much higher frequency of
types HPV16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, whereas other            HPV16, the low prevalence of productive HPV infections, and the
HPV types are associated with asymptomatic productive infections and              absence of dysplastic lesions preceding the invasive phase [34].
benign lesions [19]. The HPV genome is a circular double-stranded DNA
of approximately 8000 base pairs, containing a regulatory region (LCR,            Identifying HPV-associated carcinomas
long control region) and open reading frames (ORF). HPV ORFs are
between 7–9 depending on the genotype and are divided in Early and                    Understanding the good prognosis and better response to chemo-
Late regions. The early (E) region encodes proteins involved in virus             radiotherapy of the HNSCC causally related to HPV, compared to the
replication (E1, E2) and cellular transformation (E6, E7, E5), while the          HPV-independent carcinomas, poses new challenges to the diagnostic
late (L) proteins constitute the viral capsid. The cellular transformation        process. It is necessary to accurately identify HPV-associated tumours to
process represents a state of abortive infection, where the pathway of            provide appropriate patient counselling and management [35]. Several
viral gene expression leading to virus particles production is interrupted,       methodologies have been employed to detect HPV (DNA or RNA) se
resulting in abnormal and deregulated expression of the E6 and E7 on             quences and the expression of the p16INK4A protein in the neoplastic
cogenes [20].                                                                     tissue. These assays differ in sensitivity and specificity; HPV-DNA PCR
    Knowledge on the natural history of HPV infection and the HPV                 and/or p16INK4A overexpression by immunohistochemistry (IHC) are the
neoplastic transformation process primarily derives from studies per             most widely utilized diagnostic tools and there is a consensus on
formed in the uterine cervix, and the HPV carcinogenetic pathways in              defining as HPV-associated HNSCCs only those positive for both HPV-
the other body sites in which HPV plays a carcinogenetic role are                 DNA and p16INK4A protein [36,37].
inferred from these studies. Common features across all body sites                    More recently, besides tissues, other less invasive sample types for
include persistent infection by a hrHPV, the involvement of cells highly          investigating HPV involvement in HNSCCs have been used. These
susceptible to transformation (such as those present in the squamo-               include cytological samples obtained by fine needle aspiration or
columnar junction of the cervix, the dentate line of the anal canal, and          mucosal brushing/rinsing specimens, and body fluids (mainly blood)
the tonsillar crypts of the oropharynx), and the overexpression of the            [36,38]. Cytological samples have been primarily employed as means to
viral E6 and E7 oncoproteins. These proteins interact with key regula            investigate neck squamous cell carcinomas of unknown primary
tors of the cell cycle and apoptosis, leading to up-regulation of the             (NSCCUP), a sizable proportion of which are HPV-associated [33,39].
cellular protein cyclin dependent kinase inhibitor 2A (CDKN2A, also               On the other hand, the use of blood as a source of circulating tumour
known as p16INK4A) and degradation of the tumour-suppressor proteins              HPV DNA (ctHPVDNA) has been mostly investigated in patients with
TP53 and RB1. Additionally, E6 and E7 interfere with the immune                   HPV-associated OPSCC for monitoring the response to therapy and/or
response by neutralizing key transcription factors involved in the innate         for detecting tumour recurrence [40], particularly in case of indeter
immunity and by delaying the activation of the adaptive immune                    minate imaging findings or as a stand-alone tool.
response [21]. The E6 protein also promotes TERT expression [22].
Overexpression of p16INK4A and TERT proteins can be considered useful             Telomere and telomerase in carcinogenesis
biomarkers for diagnosis and/or prognosis of HPV-associated carci
nomas [4,17,23]. Overall, persistent infection with hrHPV types estab            Guardians of the genome: Understanding telomeres
lishes a replication-competent and immune-compromised environment
that can eventually lead to cancer.                                                  Telomeres are specialized DNA structures located at the ends of
    In the HPV-associated carcinomas, HPV sequences are often inte               chromosomes, crucial for maintaining genomic integrity [41]. They
grated into the cellular genome [20]. Viral genome integration has been           consist of variable tandem repeats of a short double-stranded DNA
reported to occur in 65–75 % of HPV16-positive HNSCCs, and in seven               sequence (5′-TTAGGG-3′) associated with a set of specialized telomeric
out of eight cell lines derived from tissues of HPV-associated primary            DNA binding proteins, i.e. the shelterin complex, which has specific
HNSCCs; the cellular integration site varies widely, and it has been              capping functions and protects telomere end from being misrecognized
postulated that HPV integration into cancer-related genes may induce              as DNA damage [42].
                                                                              2
S. Giunco et al.                                                                                                                 Oral Oncology 161 (2025) 107169
    In normal somatic cells, telomeres progressively shorten with each             protein level, thus supporting MYC-driven oncogenesis [64–66].
cell division. When they reach a critical length, they lose their protective       Notably, MYC may be overexpressed in HNSCC and is associated with a
shelterin complex and its function, becoming perceived as double-strand            poor prognosis [67].
breaks (DSBs) which activate the DNA damage response. This leads to                    As illustrated in Fig. 1, accumulating evidence suggests the existence
cellular replicative senescence, mediated by checkpoint signalling                 of feed-forward regulatory loops between TERT and various transcrip
pathways involving proteins such as TP53 and p16INK4A [42], and cells              tional factors, including WNT/β-catenin, NF-κB, and MYC, whereby the
cease to proliferate and undergo senescence or apoptosis, depending on             transcriptional factors regulate TERT transcription (see below) and
the cell type [43].                                                                TERT, in turn, regulates their transcriptional levels and/or their cellular
    Further erosion of telomeres, which may occur in checkpoint-                   compartmentalization and stability. These regulatory loops, once acti
compromised cells, results in genomic instability, a key event in carci           vated, can contribute to tumour progression through the regulation of
nogenesis initiation [44]. The activation of telomere maintenance                  multiple hallmarks of cancer [55]. Specifically, TERT’s non-canonical
mechanisms allows cells harbouring tumour-promoting mutations and                  functions within these loops may significantly influence relevant path
genomic instability to evade this crisis, conferring unlimited prolifera          ways in HNSCC, promoting persistent high TERT expression, autono
tive potential. In the majority of tumours (85–90 %), including HNSCC,             mous cancer cell proliferation, and tumour progression.
telomere maintenance is achieved through the activation of telomerase                  Given telomerase’s extensive involvement in the vast majority of
enzyme [45,46], while a minority (10–15 %) utilize a recombinant-                  tumours, and the requirement of telomere length maintenance for un
based mechanism, i.e. the Alternative Lengthening of Telomeres (ALT)               limited cell proliferation, TERT and telomere length have been proposed
[47].                                                                              as prognostic biomarkers [68]. Indeed, high TERT expression in cancer
                                                                                   cells have been associated with disease aggressiveness, advanced clin
The enzyme of immortality: Telomerase and its roles                                ical stage, and poor overall survival (OS) and disease-free survival (DFS)
                                                                                   in several cancer types, including HNSCC [4,16,17,68–70]. As above
    Telomerase is a ribonucleoprotein complex containing a catalytic               mentioned, it is conceivable that this aggressive behaviour may be
protein with telomere-specific reverse transcriptase activity, TERT,               attributable to telomerase’s non-canonical functions.
which synthesizes telomeric sequences de novo using an internal RNA as                 It has been suggested that telomere length may also have a clinical
a template, TERC [48]. While TERC has a broad tissue distribution,                 significance, as telomere shortening is a critical early event in carcino
TERT is the rate-limiting component of the complex. TERT is physio                genesis in many tumour types [68]. Several studies demonstrated that
logically expressed during embryogenesis to support the high rate of               approximately half [16,17] of patients with HNSCC have short telo
proliferation, but its expression is inhibited in most somatic adult cells         meres in the normal tissue adjacent to the cancer. Short telomeres in
[49]. When telomerase is downregulated, telomeric sequences lost at                these surrounding tissues are strongly and independently associated
each cell division are not restored, leading to progressive telomere               with mucosal failure, supporting the concept that short telomeres may
erosion and subsequent cellular replicative senescence, a pivotal event            favour genetic instability, thus increasing the potential for trans
in the aging process. TERT (re)activation that inappropriately occurs in           formation. Consequently, telomere shortening can serve as a marker for
the vast majority of cancers [45,46], prevents cellular replicative                field cancerization, aiding in identifying patients at risk for local re
senescence and leads to unlimited cellular proliferative potential,                currences [16,17].
thereby promoting tumour formation and progression [50].
    Two main strategies are currently employed to estimate telomerase              Mechanisms of TERT activation in tumours
levels from fresh or frozen tissues: quantification of TERT mRNA levels
using PCR-based techniques, and measurement of telomerase activity,                    In cancers, TERT reactivation includes transcriptional, post-
via the TRAP (Telomeric Repeat Amplification Protocol) assay [51].                 transcriptional and epigenetic regulation, as well as multiple changes
Additionally, in clinical settings, immunohistochemistry with specific             at the TERT promoter, including chromosomal rearrangements and
anti-TERT antibodies offers a practical method for directly assessing              mutations [71]. The transcription of the TERT gene is the key determi
TERT expression in tumour tissues. [52,53].                                        nant to regulating telomerase activity. The TERT promoter contains
                                                                                   binding sites for numerous transcriptional activators, such as SP1, MYC,
Beyond telomeres: Non-canonical functions of telomerase                            HIF-1α, transcription factor AP-2α, β-catenin, NF-κB, E-twenty-six (ETS)
                                                                                   family members, as well as transcriptional repressors, including Wilms’
    Although telomere maintenance is the primary canonical function of             tumour, TP53, NFX1, MXD1 (also known as MAD) and CTCF [72].
telomerase, several studies indicate that its expression is also associated            The wild-type TERT promoter is often silenced by the trimethylation
with many telomere-length independent functions that contribute to                 of histone H3 Lys27 (H3K27me3) [73]. TERT promoter also contains a
tumour development, including enhancement of proliferation, resis                 cluster of CpG sites that contributes to transcriptional regulation
tance to apoptosis, inflammation, invasion and metastasis                          through DNA methylation [71]. Additionally, somatic mutations in the
[14,15,54,55]. TERT significantly influences several key signalling                TERT core promoter can create de novo ETS binding sites, thus
pathways crucial for cancer progression. TERT binds to promoters                   enhancing TERT transcription [74,75]. In particular, TERT promoter
responsive to WNT signalling to regulate WNT target genes [56]. This               mutations mainly occur at nucleotides 1,295,228 (-124C > T) and
regulation affects cell survival, proliferation, cell polarity, differentia       1,295,250 (-146C > T) within the TERT core promoter and are among
tion during embryonic development, and carcinogenesis [56–59].                     the most common non-coding mutations in solid tumours, with a broad
Notably, overexpression of TERT in primary human oral epithelial cells             spectrum of prevalence [76–79].
induces epithelial-mesenchymal transition (EMT) by activating the                      In HNSCC, the overall prevalence of -124C > T and -146C > T TERT
WNT/β-catenin pathway, enhancing epithelial cells invasiveness.                    promoter mutations is 21 %, with the -124C > T mutation being more
Conversely, TERT silencing inhibits WNT/β-catenin signalling and sup              common than the -146C > T mutation. These mutations are more
presses EMT in oral cancer [60]. TERT also contributes to tumour-                  prevalent in OCSCCs compared to other head and neck regions; indeed,
promoting processes by inducing the transcription of the NF-κB target              nearly 50 % of OCSCC harboured TERT promoter mutations, whereas
genes, including interleukin 6 (IL6), interleukin 8 (IL8), matrix metal           only 1 % of SCCs from the oropharynx and 12 % from the larynx/hy
lopeptidase 9 (MMP9), and tumour necrosis factor (TNFα), commonly                  popharynx exhibited these mutations [80].
over expressed in HNSCC [61–63]. Moreover, TERT interacts with the
MYC pathway by regulating MYC transcription through binding MYC
transcription factor NME2 or by directly interacting with MYC at the
                                                                               3
S. Giunco et al.                                                                                                                      Oral Oncology 161 (2025) 107169
Fig. 1. HPV and TERT interplay in HNSCC: In both HPV-associated and HPV-independent HNSCC, telomerase activation is an essential process for maintaining
cellular replicative capacity through the maintenance of telomere length. The HPV E6 protein is the main driver of TERT/telomerase activation in infected cells. In
addition to providing cells with unlimited proliferation potential, telomerase interacts with other cancer-related signalling pathways, such as WNT/β-catenin, NF-kB,
and MYC. These non-canonical roles of telomerase can impact cancer progression by activating cellular programs that enhance tumour cell proliferation, motility,
migration, invasion, and epithelial-to-mesenchymal transition within the framework of feed-forward loops. In addition, telomerase non-canonical roles may influence
viral DNA maintenance and HPV persistence. HPV: human papilloma virus; TERT: telomerase reverse transcriptase; TERC: telomerase RNA component; TP53:
tumour protein p53; RB1: RB transcriptional corepressor 1; WNT: WNT/β-catenin signalling; NF-κB: NF-κB signalling; MYC: MYC pathway. Created in BioR
ender.com.
                                                                                  4
S. Giunco et al.                                                                                                                 Oral Oncology 161 (2025) 107169
gene transcription, cell proliferation, which could have important roles           differential telomerase dynamics between HPV-associated and HPV-
in tumour progression as well as in the viral life cycle. In this regard, it       independent cases. What has emerged from several investigations is
has been demonstrated that TERT can promote the induction of the EMT               that HPV-associated HNSCCs express higher levels of TERT [17,117]
phenotype in E6/E7-expressing keratinocytes [90]. Thus, it is likely that          and that TERT promoter mutations are very rare in HPV-associated
high levels of TERT expression induced by HPV could also enhance the               oropharyngeal cancer [69,109]. This is likely because there is no se
non-telomeric functions of telomerase, contributing to the metastatic              lective pressure for clones to acquire promoter mutations, as the viral
behaviour observed for some of the HPV-associated malignancies                     infection itself is sufficient to maintain TERT levels necessary for sus
[33,91].                                                                           taining the unlimited tumour cell replication. However, HPV-associated
    Miller et al. demonstrated that during the immortalization of kera            HNSCC have a markedly better prognosis compared to their HPV-
tinocytes expressing the HPV protein E7, TERT induces the expression of            independent counterparts [10]. Considering the non-canonical func
BMI1, a key component of the polycomb group complex 1 involved in                  tions of TERT that are potentially associated with enhanced cancer
chromatin remodeling and implicated in tumorigenesis and cancer                    aggressiveness [55], this observation may present a paradox given also
progression, including HNSCC [92]. BMI1 is crucial in cancer stem cell             the capability of E6 to activate TERT. A possible explanation for this
biology and is expressed in head and neck cancer stem cells, regardless            contradiction lies in the immune microenvironment linked to TERT
of HPV-association, potentially contributing to chemoresistance, recur            expression. This microenvironment is characterized by robust B-
rence, and metastasis [93,94]. Its expression may arise early in head and          lymphocyte infiltration, and cases with high TERT expression and sig
neck cancer development [95], underscoring the potential role of TERT              nificant B-cell infiltration are associated with improved progression-free
in BMI1 regulation during HPV-associated oncogenesis.                              survival [117]. Despite the beneficial impact of high TERT levels in the
    It has been suggested that the primary intent of HPV in increasing             presence of B-cell infiltration occurred regardless of HPV status [117],
TERT protein and telomerase activity is unlikely to be cellular immor             conditions resulting in very high TERT levels, which can trigger an
talization or oncogenic transformation, given that tumorigenic cells are           immune response and a “hot” immunological tumour microenvironment
not permissive to HPV replication. Instead, because TERT expression is             are significantly more common in HPV-associated cancers compared to
characteristic of stem cells, E6 might induce keratinocytes to acquire a           HPV-independent ones [8,17]. In HPV-independent cancers, elevated
stem-like phenotype, thereby facilitating HPV persistence or latency in            TERT levels in a “cold” immunological context are more likely to facil
the squamous epithelium [22]. On this ground, it is significant that               itate the non-canonical functions of telomerase, thereby driving tumour
TERT can transcriptionally activate cellular gene sets of WNT signalling           progression. Furthermore, genomic instability, a hallmark of cancer
pathway [22,56], tuning a stem-like state potentially promoting HPV                progression [119], emerges as another critical factor that may influence
persistence (Fig. 1).                                                              the dual nature of TERT’s role in these tumours. HPV-associated tu
    Additionally, TERT induction and the consequent increase in telo              mours are indeed characterized by a lower mutational burden, whereas
merase activity might play a role in viral DNA replication, as TERT ac            HPV-independent HNSCC tend to have higher mutational load, which
tivates the HPV LCR [96]. The LCR is a noncoding segment constituting              often correlate with more aggressive behaviour. Interestingly, in
about 10 % of the HPV genome and contains elements critical for viral              recurrent and metastatic HPV-associated tumours, genetic alterations
transcription and replication. This finding highlights an interaction be          such as TP53 mutations (15 %), 3p deletions (55 %), and whole-genome
tween HPV and telomerase in host cells, suggesting that TERT may                   duplications (25 %) become more frequent, aligning their molecular
regulate HPV-DNA maintenance or replication independently of its                   profiles more closely with those of HPV-independent HNSCC. These
telomerase activity.                                                               changes might reflect tumour adaptation during progression or treat
                                                                                   ment, distinguishing recurrent and metastatic HPV-associated HNSCCs
Clinical and therapeutic implications of TERT in HNSCC                             from their primary counterparts [120]. Taken together, these observa
                                                                                   tions highlight the complex and dynamic role of TERT in HNSCC. The
    TERT plays a pivotal role in HNSCC, with potential implications for            prognostic impact of TERT expression appears to be modulated not only
clinical practice and treatment strategies [97]. A comprehensive sum              by its levels but also by the characteristics of the tumour microenvi
mary of TERT-related studies, including HPV data, is provided in                   ronment and genomic instability, underscoring the importance of inte
Table 1.                                                                           grating molecular and immune landscape analyses to better understand
    The high levels of telomerase expression detected in the vast ma              its role in HNSCC outcomes.
jority of HNSCC cases strongly correlate with aggressive tumour
behaviour and poor patient outcomes [4]. The impact of TERT in HNSCC               TERT/telomerase inhibition strategies
extends beyond its canonical role in telomere maintenance, contributing
to various cancer hallmarks, including cell proliferation, angiogenesis,               The requirement for TERT overexpression to sustain the immortal
invasion, and metastasis [14,96]. TERT’s interaction with key signalling           phenotype, a hallmark of cancer, highlights its potential as a therapeutic
pathways such as WNT/β-catenin and NF-κB amplifies its influence on                target in both HPV-associated and HPV-independent HNSCC [97]. Tar
tumour progression, creating a complex network of pro-oncogenic ef                geting telomerase in HNSCC could offer significant benefits due to its
fects [56,61,63].                                                                  selective upregulation in cancer cells, while being inactive in most
    The clinical significance of TERT in HNSCC is evident in its associ           normal somatic cells. This specificity reduces the likelihood of off-target
ation with several adverse prognostic factors. Increased TERT expres              effects, a significant advantage over many current chemotherapeutics
sion and telomerase activity are linked to advanced tumour stage, poor             which affect both cancerous and normal cells. Inhibiting telomerase
differentiation, lymph node involvement, and extracapsular extension               could effectively limit the proliferation of cancer cells, leading to their
[17]. These factors contribute to higher rates of regional and distant             eventual senescence and apoptosis.
metastases, reduced treatment response, and ultimately, poor OS [118].                 Although telomerase has many attractive qualities as a target for
Notably, telomerase activity has emerged as an independent predictor of            cancer treatment, creating effective clinical therapies has been difficult
poor survival in HNSCC patients, even when accounting for other clin              due to several challenges. These include the limitations of preclinical
ical and pathological factors [16,113]. This underscores its potential as a        models, the absence of detailed high-resolution structures of human
valuable prognostic marker in clinical practice.                                   telomerase, and issues with adaptive drug resistance [121]. Telomerase
    Despite the well-established role of the HPV oncoprotein E6 in                 inhibitors work by gradually shortening telomeres, which can take
inducing TERT expression, it is surprising that very few studies have              several cell divisions, potentially allowing cancer cells to develop
evaluated TERT in relation to HPV status (Table 1). While data specific            resistance mechanisms before the treatment becomes effective, making
to HPV-associated HNSCC are limited, some evidence suggests                        them less suitable as first-line treatments [121]. Additionally, cancer
                                                                               5
S. Giunco et al.                                                                                                                             Oral Oncology 161 (2025) 107169
Table 1
Key studies on TERT/telomerase in HNSCC, including data addressing HPV-associated OPSCC.
  Authors [Ref]     Year   Number of cases                  OPSCC HPV-        Detection of TERT/             Main findings
                                                            associated        telomerase (assay)
                                                            cases
  Ogawa Y et al.    1998   25 patients with oral and        not available     TA (TRAP)                      Lower levels of TA correlated with better response to radiation
    [98]                   oropharyngeal SCC                                                                 therapy (P = 0.025).
  Patel MM et al    2002   110 HNSCC + matched              not available     TA (TRAP)                      TA in adjacent mucosa was associated with poor 2-year DFS (P
    [99]                   adjacent mucosa                                                                   < 0.05).
  Koscielny S       2004   80 HNSCC + matched adjacent      not available     TA (TRAP)                      No correlation between TA and local and regional recurrences
    et al. [100]           mucosa                                                                            and OS.
  Liao CT et al.    2004   217 HNSCC + matched              not available     TA (TRAP)                      No correlation between TA and local and regional recurrences
    [101]                  adjacent mucosa                                                                   and OS.
  El Samny T        2005   35 patients with laryngeal SCC   not applicable    TERT mRNA (RT-PCR)             TERT levels in tumour edges significantly correlated with OS (P
    et al. [102]                                                                                             = 0.04).
  Luzar B et al.    2005   40 laryngeal and 16              not applicable    TERT mRNA (relative            No correlation between level of TERT mRNA and OS.
    [103]                  hypopharyngeal SCC                                 quantification by PCR based
                                                                              kit)
  Freier K et al.   2007   352 HNSCC                        not available     TERT (FISH); TERT (IHC)        No differences in OS and DFS in HNSCC with increased TERT
    [104]                                                                                                    expression.
  Pannone G         2007   42 oral SCC + matched            not available     TERT mRNA (RT-PCR) TERT        Stage I patients with higher TERT expression had a lower OS (P
    et al. [105]           adjacent mucosa                                    (IHC)                          = 0.04).
  Chen HH et al.    2007   82 oral SCC + 116 OED + 21       not available     TERT (IHC)                     Increased expression of TERT is an early event in oral
    [106]                  normal oral mucosa                                                                carcinogenesis. Oral SCC patients with nuclear TERT labelling
                                                                                                             scores > 100 % had significantly shorter OS than those with
                                                                                                             nuclear TERT labelling scores < 100 % (P = 0.011).
  Fabricius EM      2009   40 HNSCC + 38 tumor-free         not available     TA (TRAP) TERT (IHC)           No significative correlation was found between high TERT
    et al. [107]           surgical margins + 18 tumor-                                                      expression and OS.
                           free distant from tumor
  Qu Y et al.       2014   235 laryngeal SCC                not applicable    TERT promoter mutations        TERT promoter mutation -146C > T was associated with worse
   [108]                                                                      (pyrosequencing)               OS of laryngeal cancer patients (P = 0.01).
  Boscolo-Rizzo     2015   139 HNSCC + matched              9/30 cases (30    TERT mRNA (RT-PCR)             Higher TERT levels in cancer tissues significantly correlated
    P et al. [17]          adjacent mucosa                  %)                                               with higher risk of regional failure (P = 0.045), distant failure
                                                                                                             (P = 0.067), and worse DSF (P = 0.037). HPV-associated
                                                                                                             tumours had higher TERT levels than HPV-independent (P =
                                                                                                             0.020).
  Annunziata C      2018   24 HNSCC                         5/9 cases (55.6   TERT promoter mutations        TERT promoter mutations were frequent in oral SCC (60 %) but
   et al. [109]                                             %)                (Sanger sequencing)            absent in oropharyngeal SCC (P = 0.007).
  Mundi N et al.    2019   135 patients with SCC of the     not applicable    TERT promoter mutations        TERT promoter was mutated in 0.4 % of the samples. No
   [110]                   oral cavity                                        (Illumina Ampliseq platform)   association with OS (HR: 0.592, P = 0.131).
  Boscolo-Rizzo     2019   101 HNSCC + matched              10/22 cases       TERT mRNA (RT-PCR)             Higher TERT levels in cancer tissues significantly correlated
    P et al. [16]          adjacent mucosa + plasma         (45.5 %)                                         with higher risk of regional failure (P = 0.032), progression (P
                                                                                                             = 0.049), and death (P = 0.005).
  Arantes LMRB      2020   88 HNSCC                         not available     TERT promoter mutations        27 % of cases harboured TERT promoter mutations (92 % in
    et al. [111]                                                              (pyrosequencing)               oral cavity site). Patients with -124C > T TERT promoter
                                                                                                             mutation showed a significant decrease in DFS (20.0 vs. 63.0
                                                                                                             %, P = 0.017) and in OS (16.7 vs. 45.1 %, P = 0.017).
  Yilmaz I et al.   2020   189 HNSCC                        not available     TERT promoter mutations        TERT promoter mutations were detected in the oral cavity (75
    [112]                                                                     (PCR-based direct              %); larynx (8.4 %), hypopharynx (16.6 %), and none in
                                                                              sequencing)                    oropharynx. No significant association between TERT promoter
                                                                                                             mutations and OS was found.
  Boscolo-Rizzo     2020   101 HNSCC + matched              11/23 cases (48   TERT promoter mutations        TERT promoter harboured mutations in 12 tumours (11.9 %;
    P et al. [69]          adjacent mucosa                  %)                (Sanger sequencing)            37 % in oral cavity site). No significant association between
                                                                                                             TERT promoter status and OS. No TERT-promoter mutations
                                                                                                             were detected in HPV-associated tumours.
  Giunco S et al.   2021   144 oral SCC and 57 normal       not available     TERT promoter mutations        31 % of cases harboured TERT promoter mutations. Patients
    [113]                  adjacent mucosal                                   (Sanger sequencing)            with -124C > T TERT promoter mutation showed higher risk of
                                                                                                             local recurrence (HR: 2.75, P = 0.0143), disease progression
                                                                                                             (HR: 2.71, P = 0.0024), and death (HR: 2.71, P = 0.0079).
  Kim H et al.      2021   80 tonsillar carcinomas          64/80 cases (80   TERT promoter mutations        7.5 % of cases harboured TERT promoter mutations. TERT
    [114]                                                   %)                (NAClamp™ TERT mutation        promoter mutations statistically associated with shorter DFS (P
                                                                              detection kit)                 = 0.007).
  Yu Y et al.       2021   117 HPV-negative HNSCC                            NGS panel                      TERT promoter mutations were associated with a higher
    [115]                                                                                                    cumulative incidence of locoregional failure (P < 0.001).
                                                                                   6
S. Giunco et al.                                                                                                                        Oral Oncology 161 (2025) 107169
Table 1 (continued )
  Authors [Ref]    Year    Number of cases                OPSCC HPV-        Detection of TERT/            Main findings
                                                          associated        telomerase (assay)
                                                          cases
  Haraguchi K      2022    53 oral SCC                    not available     TERT (IHC)                    Patients with high TERT expression showed higher risk of
   et al. [116]                                                                                           disease progression (HR: 5.763, P = 0.008) and death (HR:
                                                                                                          10.075, P = 0.034).
  Xian S et al.    2023    156 HNSCC                      46/156 (49 %)     Transcriptomic data from      Significant positive correlation was found between TERT
    [117]                                                                   The Cancer Genome Atlas       expression and HPV status (P < 0.001). High TERT expression,
                                                                            (TGCA)                        combined with elevated B-cell infiltration in tumours, was
                                                                                                          associated with improved PFS (P = 0.0048), irrespective of HPV
                                                                                                          status.
TERT, telomerase reverse transcriptase; Ref, reference; OPSCC, oropharyngeal squamous cell carcinoma; SCC, squamous cell carcinoma; HNSCC head and neck
squamous cell carcinoma; TA, telomerase activity; TRAP, telomeric repeat amplification protocol; DFS, disease-free survival; PFS, progression-free survival; RT-PCR,
reverse transcription polymerase chain reaction; OS, overall survival; FISH, Fluorescence in situ hybridization; IHC, immunohistochemistry; OED, oral epithelial
dysplasia; HR, hazard ratio; NGS, next generation sequencing.
cells may activate ALT mechanism to maintain telomere length in the                      As stated above, studies have consistently shown that the presence of
absence of telomerase activity [47,122]. The ALT, though less common,                HPV-DNA in tumour cells is associated with a significant reduction in
could become more prevalent as a resistance strategy, necessitating                  the risk of death compared to HPV-independent tumours [10] with these
combination therapies to effectively target both telomerase and ALT                  neoplasm being more sensitive to radiation therapy and chemothera
pathways [123].                                                                      peutic agents [134]. The enhanced treatment responsiveness has led to
    While telomerase is inactive in most somatic cells, it is active in              discussions about de-escalating therapy in HPV-associated patients to
certain stem cell populations [124]. Inhibiting telomerase could                     reduce treatment-related morbidity without compromising survival
adversely affect these cells, leading to side effects such as bone marrow            outcomes [135,136]. However, it is increasingly evident that HPV-
suppression and impaired tissue regeneration. Therefore, careful                     associated tumours represent a heterogeneous group, with some tu
consideration and monitoring of side effects are essential in the clinical           mours exhibiting unexpectedly aggressive behaviour [137,138]. This
application of telomerase inhibitors.                                                heterogeneity challenges the current paradigm of uniform treatment de-
    Despite numerous challenges, several strategies to target telomerase             escalation for all HPV-associated cases [139]. Among the most impli
function have been developed, some of which have entered clinical trials             cated factors in modulating prognosis in patients with HPV-associated
[121]. Imetelstat, a direct telomerase inhibitor, shortens telomeres and             OPSCC are cigarette smoking [10] and extranodal extension (ENE)
shows efficacy in hematologic malignancies, though it has dose-limiting              [140], as well as molecular biomarkers [141,142]. Though still debated
toxicities in solid tumour trials [125,126]. Indirect inhibitors, like G-            [29,143], some recent studies suggest that the extent of ENE may
quadruplex stabilizers (e.g., CX-5461), disrupt telomerase function by               significantly influence prognosis [140]. This has led some authors to
blocking the resolution of telomeric G-quadruplex DNA inducing DNA                   reconsider the role of ENE in staging, suggesting that every radiological
damage and cell death [127]. Additionally, suicide gene therapies like               ENE + should be reclassified into the cN3b category [144].
Telomelysin utilize a replication-competent oncolytic adenovirus engi                   In this scenario, TERT expression emerges as a promising quantita
neered with a TERT promoter, enabling tumour-specific replication and                tive biomarker to address this clinical challenge. High TERT expression,
showing promising antitumor effects [128,129]. Telomelysin has been                  coupled with significant B-cell tumour infiltration, may suggest
proposed for a Phase II clinical trial to assess its efficacy in patients with       immune-mediated tumour control [117], identifying patients who could
inoperable, recurrent, or progressive HNSCC (NCT04685499).                           be ideal candidates for de-escalation therapies. Conversely, high TERT
    Immunotherapies, including peptide vaccines (such as GV1001,                     expression without substantial immune infiltration, even in HPV-
GX301, UV1, Vx-001, and UCPVax) and the DNA vaccine INVAC-1, offer                   associated tumours, may indicate a more aggressive disease, poten
immunogenic TERT epitopes that activate immune responses against                     tially identifying a subset of HPV-associated HNSCCs that behave more
cancer cells expressing telomerase. Immunotherapies against TERT                     like their HPV-independent counterparts, particularly in cases with
display potential when combined with immune checkpoint inhibitors                    altered genetic and molecular profiles [120]. This insight is crucial for
[130–132]. The ongoing VolATIL study, a Phase II trial, evaluates the                patient stratification: HPV-associated tumours with high TERT expres
combination of atezolizumab (an immune checkpoint inhibitor) and                     sion but poor immune infiltration might require more aggressive treat
UCPVax, a therapeutic vaccine based on telomerase-derived peptides, in               ment approaches and should be excluded from de-intensification trials
HPV-associated cancers, including head and neck cancer                               to avoid undertreatment. On the other hand, patients with low TERT
(NCT03946358) [133]. The recent findings that high TERT expression,                  expression might be ideal candidates for de-escalation therapies. Thus,
combined with elevated B-cell infiltration in tumours, is associated with            by integrating TERT expression analysis into clinical assessment, on
improved progression-free survival, irrespective of HPV status [117],                cologists could more accurately predict tumour behaviour, optimize
position TERT as a key antigen in mediating local antitumor immunity in              treatment strategies, and improve patient outcomes. This approach
HNSCC. Collectively, these data strongly suggest that innovative                     aligns with the growing trend towards precision medicine in oncology,
immunotherapeutic strategies targeting TERT could be effective in both               offering a more nuanced and personalized approach to managing HPV-
HPV-associated and independent HNSCC, potentially expanding treat                   associated HNSCC [145].
ment options for these challenging malignancies.                                         Understanding how HPV E6 activates TERT opens avenues for novel
                                                                                     therapeutic strategies, potentially leading to drugs that target this spe
Potential clinical implication of the interaction between HPV                        cific interaction in HPV-associated HNSCCs while sparing normal cells
and telomerase                                                                       [84]. Moreover, this relationship might elucidate mechanisms of treat
                                                                                     ment resistance, such as how HPV’s ability to maintain high TERT
    The interaction between TERT and HPV in HNSCC carries potential                  expression could contribute to radiotherapy or chemotherapy resistance
significant clinical implications, spanning diagnosis, prognosis, and                [146].
treatment strategies. Understanding this relationship may be crucial for                 The potential role of TERT in HPV-associated HNSCC may extend
advancing patient care and outcomes.                                                 beyond initial diagnosis and treatment planning to post-treatment
                                                                                 7
S. Giunco et al.                                                                                                                          Oral Oncology 161 (2025) 107169
surveillance. However, while tumours with a high prevalence of TERT               Visualization, Investigation, Conceptualization.
promoter mutations, such as hepatocellular carcinoma, demonstrate
promising potential for utilizing cell-free DNA quantification in treat          Funding
ment and post-treatment surveillance [147], this approach may have
limited utility in HPV-associated OPSCCs. In these cases, the prevalence             This research did not receive any specific grant from funding
of TERT promoter mutations is marginal [79,113], suggesting that                  agencies in the public, commercial, or not-for-profit sectors.
alternative strategies may be necessary to provide reliable predictive
insights for treatment responses in this specific patient population.
    Overall, the integration of TERT expression analysis in the clinical          Declaration of competing interest
assessment of HPV-associated HNSCC presents a promising avenue for
more precise risk stratification and personalized treatment strategies.               The authors declare that they have no known competing financial
Further research into the HPV-TERT interaction may not only refine our            interests or personal relationships that could have appeared to influence
approach to managing these tumours but also pave the way for novel                the work reported in this paper.
targeted therapies, potentially transforming the treatment landscape for
HNSCC patients.                                                                   References
                                                                                    [1] Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global
Conclusion                                                                              cancer statistics 2022: GLOBOCAN estimates of incidence and mortality
                                                                                        worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2024;74:229–63.
    The complex interplay between HPV and telomerase in HNSCC                           https://doi.org/10.3322/caac.21834.
                                                                                    [2] Leemans CR, Snijders PJF, Brakenhoff RH. The molecular landscape of head and
represents a critical area of research with significant implications for                neck cancer. Nat Rev Cancer 2018;18:269–82. https://doi.org/10.1038/
understanding disease pathogenesis, improving diagnostic approaches,                    nrc.2018.11.
and developing targeted therapies. This review has highlighted the                  [3] Lechner M, Liu J, Masterson L, Fenton TR. HPV-associated oropharyngeal cancer:
                                                                                        epidemiology, molecular biology and clinical management. Nat Rev Clin Oncol
crucial role of HPV, particularly HPV16, in a subset of HNSCCs, espe                   2022;19:306–27. https://doi.org/10.1038/s41571-022-00603-7.
cially OPSCC. HPV-associated tumours exhibit distinct molecular and                 [4] Boscolo-Rizzo P, Da Mosto MC, Rampazzo E, Giunco S, Del Mistro A,
clinical characteristics, including better prognosis and treatment                      Menegaldo A, et al. Telomeres and telomerase in head and neck squamous cell
                                                                                        carcinoma: from pathogenesis to clinical implications. Cancer Metastasis Rev
response. Telomerase reactivation, primarily through increased
                                                                                        2016;35:457–74. https://doi.org/10.1007/s10555-016-9633-1.
expression of TERT, is a hallmark of most cancers, including HNSCC,                 [5] Ghittoni R, Accardi R, Hasan U, Gheit T, Sylla B, Tommasino M. The biological
with high TERT expression associated with advanced tumour stage,                        properties of E6 and E7 oncoproteins from human papillomaviruses. Virus Genes
increased metastasis, and poor prognosis.                                               2010;40:1–13. https://doi.org/10.1007/s11262-009-0412-8.
                                                                                    [6] Zumsteg ZS, Luu M, Rosenberg PS, Elrod JK, Bray F, Vaccarella S, et al. Global
    The HPV E6 oncoprotein emerges as a potent activator of TERT                        epidemiologic patterns of oropharyngeal cancer incidence trends. J Natl Cancer
expression and telomerase activity, operating through multiple mecha                   Inst 2023;115:1544–54. https://doi.org/10.1093/jnci/djad169.
nisms at transcriptional, epigenetic, and post-transcriptional levels               [7] Lohavanichbutr P, Houck J, Fan W, Yueh B, Mendez E, Futran N, et al.
                                                                                        Genomewide gene expression profiles of HPV-positive and HPV-negative
[22,148]. The interaction between HPV and TERT extends telomerase                       oropharyngeal cancer: potential implications for treatment choices. Archives of
functions, beyond its canonical role in telomere maintenance. TERT                      Otolaryngology–Head & Neck Surgery 2009;135:180–8. https://doi.org/
exhibits non-canonical functions that contribute to various aspects of                  10.1001/archoto.2008.540.
                                                                                    [8] Tosi A, Parisatto B, Menegaldo A, Spinato G, Guido M, Del Mistro A, et al. The
cancer behaviour, including cell proliferation, angiogenesis, invasion,                 immune microenvironment of HPV-positive and HPV-negative oropharyngeal
and metastasis [96]. Additionally, TERT potentially plays roles in                      squamous cell carcinoma: a multiparametric quantitative and spatial analysis
facilitating HPV persistence, viral DNA replication, and promoting a                    unveils a rationale to target treatment-naïve tumors with immune checkpoint
                                                                                        inhibitors. J Exp Clin Cancer Res 2022;41:279. https://doi.org/10.1186/s13046-
stem-like phenotype in infected cells [20,56].                                          022-02481-4.
    Targeting telomerase in HNSCC presents a promising therapeutic                  [9] Antonioli M, Pagni B, Vescovo T, Ellis R, Cosway B, Rollo F, et al. HPV sensitizes
avenue, though challenges remain in developing effective clinical stra                 OPSCC cells to cisplatin-induced apoptosis by inhibiting autophagy through E7-
                                                                                        mediated degradation of AMBRA1. Autophagy 2021;17:2842–55. https://doi.
tegies [121]. Understanding these intricate relationships provides
                                                                                        org/10.1080/15548627.2020.1847444.
valuable insights into the molecular mechanisms underlying HPV-                    [10] Ang KK, Harris J, Wheeler R, Weber R, Rosenthal DI, Nguyen-Tan PF, et al.
associated HNSCC, offering potential for improving diagnostic accu                     Human papillomavirus and survival of patients with oropharyngeal cancer.
racy, refining prognostic assessments, and developing novel targeted                    N Engl J Med 2010;363:24–35. https://doi.org/10.1056/NEJMoa0912217.
                                                                                   [11] Licitra L, Perrone F, Bossi P, Suardi S, Mariani L, Artusi R, et al. High-risk human
therapies.                                                                              papillomavirus affects prognosis in patients with surgically treated oropharyngeal
    Future research should focus on elucidating the full spectrum of                    squamous cell carcinoma. J Clin Oncol 2006;24:5630–6. https://doi.org/
TERT’s non-canonical functions in HPV-associated carcinogenesis and                     10.1200/JCO.2005.04.6136.
                                                                                   [12] Blackburn EH, Epel ES, Lin J. Human telomere biology: a contributory and
exploring innovative approaches to target the telomerase pathway in                     interactive factor in aging, disease risks, and protection. Science 2015;350:
HNSCC. As our knowledge of the HPV-telomerase axis in HNSCC con                        1193–8. https://doi.org/10.1126/science.aab3389.
tinues to expand, it holds promise for personalized medicine ap                   [13] Wallace NA, Galloway DA. Novel functions of the human papillomavirus E6
                                                                                        oncoproteins. Annu Rev Virol 2015;2:403–23. https://doi.org/10.1146/annurev-
proaches, potentially leading to more effective treatments and improved                 virology-100114-055021.
outcomes for patients with this challenging malignancy. Continued                  [14] Ségal-Bendirdjian E, Geli V. Non-canonical roles of telomerase: unraveling the
investigation in this field is crucial for advancing our understanding and              imbroglio. Front Cell Dev Biol 2019;7:332. https://doi.org/10.3389/
                                                                                        fcell.2019.00332.
management of HNSCC in the coming years.                                           [15] Martínez P, Blasco MA. Telomeric and extra-telomeric roles for telomerase and
                                                                                        the telomere-binding proteins. Nat Rev Cancer 2011;11:161–76. https://doi.org/
CRediT authorship contribution statement                                                10.1038/nrc3025.
                                                                                   [16] Boscolo-Rizzo P, Rampazzo E, Polesel J, Giunco S, Menegaldo A, Mantovani M,
                                                                                        et al. Predictive and prognostic significance of telomerase levels/telomere length
    Silvia Giunco: Writing – review & editing, Writing – original draft,                in tissues and peripheral blood in head and neck squamous cell carcinoma. Sci
Visualization, Investigation. Annarosa Del Mistro: Writing – review &                   Rep 2019;9. https://doi.org/10.1038/s41598-019-54028-x.
                                                                                   [17] Boscolo-Rizzo P, Rampazzo E, Perissinotto E, Piano MA, Giunco S, Baboci L, et al.
editing, Writing – original draft, Investigation. Marzia Morello: Writing
                                                                                        Telomere shortening in mucosa surrounding the tumor: Biosensor of field
– original draft, Investigation. Jacopo Lidonnici: Writing – original                   cancerization and prognostic marker of mucosal failure in head and neck
draft, Investigation. Helena Frayle: Writing – original draft, Investiga               squamous cell carcinoma. Oral Oncol 2015;51:500–7. https://doi.org/10.1016/j.
tion. Silvia Gori: Writing – original draft, Investigation. Anita de Rossi:             oraloncology.2015.02.100.
                                                                                   [18] Béziat V. Human genetic dissection of papillomavirus-driven diseases: new
Writing – review & editing, Supervision, Conceptualization. Paolo                       insight into their pathogenesis. Hum Genet 2020;139:919–39. https://doi.org/
Boscolo-Rizzo: Writing – review & editing, Writing – original draft,                    10.1007/s00439-020-02183-x.
                                                                              8
S. Giunco et al.                                                                                                                                       Oral Oncology 161 (2025) 107169
 [19] Bouvard V, Baan R, Straif K, Grosse Y, Secretan B, El Ghissassi F, et al. A review of       [44] Hackett JA, Greider CW. Balancing instability: dual roles for telomerase and
      human carcinogens–Part B: biological agents. Lancet Oncol 2009;10:321–2.                         telomere dysfunction in tumorigenesis. Oncogene 2002;21:619–26. https://doi.
      https://doi.org/10.1016/s1470-2045(09)70096-8.                                                   org/10.1038/sj.onc.1205061.
 [20] Schiffman M, Doorbar J, Wentzensen N, de Sanjosé S, Fakhry C, Monk BJ, et al.              [45] Blackburn EH, Greider CW, Szostak JW. Telomeres and telomerase: the path from
      Carcinogenic human papillomavirus infection. Nat Rev Dis Primers 2016;2:                         maize, tetrahymena and yeast to human cancer and aging. Nat Med 2006;12:
      16086. https://doi.org/10.1038/nrdp.2016.86.                                                     1133–8. https://doi.org/10.1038/nm1006-1133.
 [21] Lo Cigno I, Calati F, Girone C, Catozzo M, Gariglio M. High-risk HPV oncoproteins           [46] Shay JW, Wright WE. Role of telomeres and telomerase in cancer. Semin Cancer
      E6 and E7 and their interplay with the innate immune response: uncovering                        Biol 2011;21:349–53. https://doi.org/10.1016/j.semcancer.2011.10.001.
      mechanisms of immune evasion and therapeutic prospects. J Med Virol 2024;96:                [47] Shay JW, Wright WE. Telomeres and telomerase: three decades of progress. Nat
      e29685. https://doi.org/10.1002/jmv.29685.                                                       Rev Genet 2019;20:299–309. https://doi.org/10.1038/s41576-019-0099-1.
 [22] Liu X, Dakic A, Zhang Y, Dai Y, Chen R, Schlegel R. HPV E6 protein interacts                [48] Roake CM, Artandi SE. Regulation of human telomerase in homeostasis and
      physically and functionally with the cellular telomerase complex. Proc Natl Acad                 disease. Nat Rev Mol Cell Biol 2020;21:384–97. https://doi.org/10.1038/s41580-
      Sci 2009;106:18780–5. https://doi.org/10.1073/pnas.0906357106.                                   020-0234-z.
 [23] Pańczyszyn A, Boniewska-Bernacka E, Głąb G. Telomeres and telomerase during                [49] Schmidt JC, Cech TR. Human telomerase: biogenesis, trafficking, recruitment,
      human papillomavirus-induced carcinogenesis. Mol Diagn Ther 2018;22:421–30.                      and activation. Genes Dev 2015;29:1095–105. https://doi.org/10.1101/
      https://doi.org/10.1007/s40291-018-0336-x.                                                       gad.263863.115.
 [24] Walline HM, Goudsmit CM, McHugh JB, Tang AL, Owen JH, Teh BT, et al.                        [50] Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;
      Integration of high-risk human papillomavirus into cellular cancer-related genes                 144:646–74. https://doi.org/10.1016/j.cell.2011.02.013.
      in head and neck cancer cell lines. Head Neck 2017;39:840–52. https://doi.org/              [51] Zhou X, Xing D. Assays for human telomerase activity: progress and prospects.
      10.1002/hed.24729.                                                                               Chem Soc Rev 2012;41:4643–56. https://doi.org/10.1039/c2cs35045a.
 [25] Lyford-Pike S, Peng S, Young GD, Taube JM, Westra WH, Akpeng B, et al.                      [52] Hiyama E, Hiyama K, Yokoyama T, Shay JW. Immunohistochemical detection of
      Evidence for a role of the PD-1:PD-L1 pathway in immune resistance of HPV-                       telomerase (hTERT) protein in human cancer tissues and a subset of cells in
      associated head and neck squamous cell carcinoma. Cancer Res 2013;73:                            normal tissues. Neoplasia 2001;3(1):17–26. https://doi.org/10.1038/sj.
      1733–41. https://doi.org/10.1158/0008-5472.CAN-12-2384.                                          neo.7900134.
 [26] Gelwan E, Malm I-J, Khararjian A, Fakhry C, Bishop JA, Westra WH. Nonuniform                [53] Yan P, Benhattar J, Seelentag W, Stehle JC, Bosman FT. Immunohistochemical
      distribution of high-risk human papillomavirus in squamous cell carcinomas of                    localization of hTERT protein in human tissues. Histochem Cell Biol 2004;121:
      the oropharynx: rethinking the anatomic boundaries of oral and oropharyngeal                     391–7. https://doi.org/10.1007/s00418-004-0645-5.
      carcinoma from an oncologic HPV perspective. Am J Surg Pathol 2017;41:                      [54] Li Y, Tergaonkar V. Noncanonical functions of telomerase: implications in
      1722–8. https://doi.org/10.1097/PAS.0000000000000929.                                            telomerase-targeted cancer therapies. Cancer Res 2014;74:1639–44. https://doi.
 [27] Miniggio HD. Biological significance of palatine tonsillar epithelium:                           org/10.1158/0008-5472.CAN-13-3568.
      microstructure and disease. S Afr Dent J 2016;71:496–9.                                     [55] Kumar N, Sethi G. Telomerase and hallmarks of cancer: an intricate interplay
 [28] Hufbauer M, Maltseva M, Meinrath J, Lechner A, Beutner D, Huebbers CU, et al.                    governing cancer cell evolution. Cancer Lett 2023;578:216459. https://doi.org/
      HPV16 increases the number of migratory cancer stem cells and modulates their                    10.1016/j.canlet.2023.216459.
      miRNA expression profile in oropharyngeal cancer. Int J Cancer 2018;143:                    [56] Park J-I, Venteicher AS, Hong JY, Choi J, Jun S, Shkreli M, et al. Telomerase
      1426–39. https://doi.org/10.1002/ijc.31538.                                                      modulates Wnt signalling by association with target gene chromatin. Nature
 [29] Ferris RL, Westra W. Oropharyngeal carcinoma with a special focus on HPV-                        2009;460:66–72. https://doi.org/10.1038/nature08137.
      related squamous cell carcinoma. Annu Rev Pathol 2023;18:515–35. https://doi.               [57] Logan CY, Nusse R. The Wnt signaling pathway in development and disease.
      org/10.1146/annurev-pathmechdis-031521-041424.                                                   Annu Rev Cell Dev Biol 2004;20:781–810. https://doi.org/10.1146/annurev.
 [30] Huang SH, O’Sullivan B. Overview of the 8th edition TNM classification for head                  cellbio.20.010403.113126.
      and neck cancer. Curr Treat Options Oncol 2017;18(40). https://doi.org/                     [58] Yang F, Zeng Q, Yu G, Li S, Wang C-Y. Wnt/beta-catenin signaling inhibits death
      10.1007/s11864-017-0484-y.                                                                       receptor-mediated apoptosis and promotes invasive growth of HNSCC. Cell Signal
 [31] Liederbach E, Kyrillos A, Wang C-H, Liu JC, Sturgis EM, Bhayani MK. The                          2006;18:679–87. https://doi.org/10.1016/j.cellsig.2005.06.015.
      national landscape of human papillomavirus-associated oropharynx squamous                   [59] Iwai S, Yonekawa A, Harada C, Hamada M, Katagiri W, Nakazawa M, et al.
      cell carcinoma. Int J Cancer 2017;140:504–12. https://doi.org/10.1002/                           Involvement of the Wnt-β-catenin pathway in invasion and migration of oral
      ijc.30442.                                                                                       squamous carcinoma cells. Int J Oncol 2010;37:1095–103. https://doi.org/
 [32] Boscolo-Rizzo P, Schroeder L, Romeo S, Pawlita M. The prevalence of human                        10.3892/ijo_00000761.
      papillomavirus in squamous cell carcinoma of unknown primary site metastatic                [60] Zhao T, Hu F, Qiao B, Chen Z, Tao Q. Telomerase reverse transcriptase potentially
      to neck lymph nodes: a systematic review. Clin Exp Metastasis 2015;32:835–45.                    promotes the progression of oral squamous cell carcinoma through induction of
      https://doi.org/10.1007/s10585-015-9744-z.                                                       epithelial-mesenchymal transition. Int J Oncol 2015;46:2205–15. https://doi.
 [33] Schroeder L, Boscolo-Rizzo P, Dal Cin E, Romeo S, Baboci L, Dyckhoff G, et al.                   org/10.3892/ijo.2015.2927.
      Human papillomavirus as prognostic marker with rising prevalence in neck                    [61] Ghosh A, Saginc G, Leow SC, Khattar E, Shin EM, Yan TD, et al. Telomerase
      squamous cell carcinoma of unknown primary: a retrospective multicentre study.                   directly regulates NF-κB-dependent transcription. Nat Cell Biol 2012;14:1270–81.
      Eur J Cancer 2017;74:73–81. https://doi.org/10.1016/j.ejca.2016.12.020.                          https://doi.org/10.1038/ncb2621.
 [34] Lim YX, D’Silva NJ. HPV-associated oropharyngeal cancer: in search of surrogate             [62] Ding D, Xi P, Zhou J, Wang M, Cong Y-S. Human telomerase reverse transcriptase
      biomarkers for early lesions. Oncogene 2024;43:543–54. https://doi.org/                          regulates MMP expression independently of telomerase activity via NF-κB-
      10.1038/s41388-023-02927-9.                                                                      dependent transcription. FASEB J 2013;27:4375–83. https://doi.org/10.1096/
 [35] Boscolo-Rizzo P, Pawlita M, Holzinger D. From HPV-positive towards HPV-driven                    fj.13-230904.
      oropharyngeal squamous cell carcinomas. Cancer Treat Rev 2016;42:24–9.                      [63] Allen CT, Ricker JL, Chen Z, Van Waes C. Role of activated nuclear factor-κB in
      https://doi.org/10.1016/j.ctrv.2015.10.009.                                                      the pathogenesis and therapy of squamous cell carcinoma of the head and neck.
 [36] Mehanna H, Alemany L, von Buchwald C. Advances in testing for human                              Head Neck 2007;29:959–71. https://doi.org/10.1002/hed.20615.
      papillomavirus -mediated head and neck cancer. Curr Opin Oncol 2024;36:                     [64] Kar A, Saha D, Purohit G, Singh A, Kumar P, Yadav VK, et al. Metastases
      143–6. https://doi.org/10.1097/CCO.0000000000001029.                                             suppressor NME2 associates with telomere ends and telomerase and reduces
 [37] Mena M, Wang X, Tous S, Quiros B, Clavero O, Alejo M, et al. Concordance of                      telomerase activity within cells. Nucleic Acids Res 2012;40:2554–65. https://doi.
      p16INK4a and E6*I mRNA among HPV-DNA-Positive oropharyngeal, laryngeal,                          org/10.1093/nar/gkr1109.
      and oral cavity carcinomas from the ICO international study. Cancers (Basel)                [65] Koh CM, Khattar E, Leow SC, Liu CY, Muller J, Ang WX, et al. Telomerase
      2022;14:3787. https://doi.org/10.3390/cancers14153787.                                           regulates MYC-driven oncogenesis independent of its reverse transcriptase
 [38] Boscolo-Rizzo P, Polesel J, Menegaldo A, Sia E, Stellin M, Tirelli G. Human                      activity. J Clin Invest 2015;125:2109–22. https://doi.org/10.1172/JCI79134.
      papillomaviruses 16 and 18 E6 oncoprotein detection test in primary                         [66] Tang B, Xie R, Qin Y, Xiao Y-F, Yong X, Zheng L, et al. Human telomerase reverse
      oropharyngeal carcinomas and metastatic lymph nodes: a cross-sectional study.                    transcriptase (hTERT) promotes gastric cancer invasion through cooperating with
      Head Neck Pathol 2024;18:101. https://doi.org/10.1007/s12105-024-01713-7.                        c-Myc to upregulate heparanase expression. Oncotarget 2016;7:11364–79.
 [39] Menegaldo A, Schroeder L, Holzinger D, Tirelli G, Dal Cin E, Tofanelli M, et al.                 https://doi.org/10.18632/oncotarget.6575.
      Detection of HPV16/18 E6 oncoproteins in head and neck squamous cell                        [67] Liu S, Qin Z, Mao Y, Zhang W, Wang Y, Jia L, et al. Therapeutic targeting of MYC
      carcinoma using a protein immunochromatographic assay. Laryngoscope 2021;                        in head and neck squamous cell carcinoma. Oncoimmunology 2022;11:2130583.
      131:1042–8. https://doi.org/10.1002/lary.29184.                                                  https://doi.org/10.1080/2162402X.2022.2130583.
 [40] Campo F, Iocca O, Paolini F, Manciocco V, Moretto S, De Virgilio A, et al. The              [68] Giunco S, Rampazzo E, Celeghin A, Petrara MR, De Rossi A. Telomere and
      landscape of circulating tumor HPV DNA and TTMV-HPVDNA for surveillance of                       telomerase in carcinogenesis: their role as prognostic biomarkers. Curr Pathobiol
      HPV-oropharyngeal carcinoma: systematic review and meta-analysis. J Exp Clin                     Rep 2015;3:315–28. https://doi.org/10.1007/s40139-015-0087-x.
      Cancer Res 2024;43:215. https://doi.org/10.1186/s13046-024-03137-1.                         [69] Boscolo-Rizzo P, Giunco S, Rampazzo E, Brutti M, Spinato G, Menegaldo A, et al.
 [41] Blackburn EH. The end of the (DNA) line. Nat Struct Biol 2000;7:847–50. https://                 TERT promoter hotspot mutations and their relationship with TERT levels and
      doi.org/10.1038/79594.                                                                           telomere erosion in patients with head and neck squamous cell carcinoma.
 [42] Palm W, de Lange T. How shelterin protects mammalian telomeres. Annu Rev                         J Cancer Res Clin Oncol 2020;146:381–9. https://doi.org/10.1007/s00432-020-
      Genet 2008;42:301–34. https://doi.org/10.1146/annurev.                                           03130-z.
      genet.41.110306.130350.                                                                     [70] González-Moles MÁ, Moya-González E, García-Ferrera A, Nieto-Casado P, Ramos-
 [43] Maciejowski J, de Lange T. Telomeres in cancer: tumour suppression and genome                    García P. Prognostic and clinicopathological significance of telomerase reverse
      instability. Nat Rev Mol Cell Biol 2017;18:175–86. https://doi.org/10.1038/                      transcriptase upregulation in oral cancer: a systematic review and meta-analysis.
      nrm.2016.171.                                                                                    Cancers (Basel) 2022;14:3673. https://doi.org/10.3390/cancers14153673.
                                                                                              9
S. Giunco et al.                                                                                                                                        Oral Oncology 161 (2025) 107169
 [71] Akincilar SC, Unal B, Tergaonkar V. Reactivation of telomerase in cancer. Cell             [97] McCaul JA, Gordon KE, Clark LJ, Parkinson EK. Telomerase inhibition and the
      Mol Life Sci 2016;73:1659–70. https://doi.org/10.1007/s00018-016-2146-9.                        future management of head-and-neck cancer. Lancet Oncol 2002;3:280–8.
 [72] Hafezi F, Perez BD. The solo play of TERT promoter mutations. Cells 2020;9:749.                 https://doi.org/10.1016/s1470-2045(02)00729-5.
      https://doi.org/10.3390/cells9030749.                                                      [98] Ogawa Y, Nishioka A, Hamada N, Terashima M, Inomata T, Yoshida S, et al.
 [73] Stern JL, Theodorescu D, Vogelstein B, Papadopoulos N, Cech TR. Mutation of the                 Changes in telomerase activity of advanced cancers of oral cavity and oropharynx
      TERT promoter, switch to active chromatin, and monoallelic TERT expression in                   during radiation therapy: correlation with clinical outcome. Int J Mol Med 1998;
      multiple cancers. Genes Dev 2015;29:2219–24. https://doi.org/10.1101/                           2:301–7. https://doi.org/10.3892/ijmm.2.3.301.
      gad.269498.115.                                                                            [99] Patel MM, Parekh LJ, Jha FP, Sainger RN, Patel JB, Patel DD, et al. Clinical
 [74] Horn S, Figl A, Rachakonda PS, Fischer C, Sucker A, Gast A, et al. TERT promoter                usefulness of telomerase activation and telomere length in head and neck cancer.
      mutations in familial and sporadic melanoma. Science 2013;339:959–61. https://                  Head Neck 2002;24:1060–7. https://doi.org/10.1002/hed.10169.
      doi.org/10.1126/science.1230062.                                                          [100] Koscielny S, Fv E, Dahse R, Fiedler W. The influence of reactivation of the
 [75] Huang FW, Hodis E, Xu MJ, Kryukov GV, Chin L, Garraway LA. Highly recurrent                     telomerase in tumour tissue on the prognosis of squamous cell carcinomas in the
      TERT promoter mutations in human melanoma. Science 2013;339:957–9.                              head and neck. J Oral Pathol Med 2004;33:538–42. https://doi.org/10.1111/
      https://doi.org/10.1126/science.1229259.                                                        j.1600-0714.2004.00232.x.
 [76] Vinagre J, Almeida A, Pópulo H, Batista R, Lyra J, Pinto V, et al. Frequency of          [101] Liao CT, Tung-Chieh Chang J, Wang HM, Chen IH, Lin CY, et al. Telomerase as an
      TERT promoter mutations in human cancers. Nat Commun 2013;4:2185. https://                      independent prognostic factor in head and neck squamous cell carcinoma. Head
      doi.org/10.1038/ncomms3185.                                                                     Neck 2004;26:504–12. https://doi.org/10.1002/hed.20007.
 [77] Killela PJ, Reitman ZJ, Jiao Y, Bettegowda C, Agrawal N, Diaz LA, et al. TERT             [102] El Samny T, El Halaby H, El Fiky L, Nassar M, El Makhzangy A, Eissa S. Prognostic
      promoter mutations occur frequently in gliomas and a subset of tumors derived                   value of telomerase and DNA ploidy in laryngeal carcinoma. Eur Arch
      from cells with low rates of self-renewal. Proc Natl Acad Sci 2013;110:6021–6.                  Otorhinolaryngol 2005;262:799–803. https://doi.org/10.1007/s00405-004-
      https://doi.org/10.1073/pnas.1303607110.                                                        0904-z.
 [78] Tornesello ML, Cerasuolo A, Starita N, Amiranda S, Bonelli P, Tuccillo FM, et al.         [103] Luzar B, Poljak M, Fischinger J, Klopcic U, Gale N. Telomerase catalytic subunit
      Reactivation of telomerase reverse transcriptase expression in cancer: the role of              gene expression does not influence survival of patients with squamous cell
      TERT promoter mutations. Front Cell Dev Biol 2023;11:1286683. https://doi.                      carcinoma of the larynx and hypopharynx: a case-control study. J Laryngol Otol
      org/10.3389/fcell.2023.1286683.                                                                 2005;119:917–21. https://doi.org/10.1258/002221505774783403.
 [79] Bell RJA, Rube HT, Xavier-Magalhães A, Costa BM, Mancini A, Song JS, et al.              [104] Freier K, Pungs S, Flechtenmacher C, Bosch FX, Lichter P, Joos S, et al. Frequent
      Understanding TERT promoter mutations: a common path to immortality. Mol                        high telomerase reverse transcriptase expression in primary oral squamous cell
      Cancer Res 2016;14:315–23. https://doi.org/10.1158/1541-7786.MCR-16-0003.                       carcinoma. J Oral Pathol Med 2007;36:267–72. https://doi.org/10.1111/j.1600-
 [80] Boscolo-Rizzo P, Tirelli G, Polesel J, Sia E, Phillips V, Borsetto D, et al. TERT               0714.2007.00531.x.
      promoter mutations in head and neck squamous cell carcinoma: A systematic                 [105] Pannone G, De Maria S, Zamparese R, Metafora S, Serpico R, Morelli F, et al.
      review and meta-analysis on prevalence and prognostic significance. Oral Oncol                  Prognostic value of human telomerase reverse transcriptase gene expression in
      2023;140:106398. https://doi.org/10.1016/j.oraloncology.2023.106398.                            oral carcinogenesis. Int J Oncol 2007;30:1349–57. https://doi.org/10.3892/
 [81] Katzenellenbogen RA. Activation of telomerase by HPVs. Virus Res 2017;231:                      ijo.30.6.1349.
      50–5. https://doi.org/10.1016/j.virusres.2016.11.003.                                     [106] Chen HH, Yu CH, Wang JT, Liu BY, Wang YP, Sun A, et al. Expression of human
 [82] Veldman T, Horikawa I, Barrett JC, Schlegel R. Transcriptional activation of the                telomerase reverse transcriptase (hTERT) protein is significantly associated with
      telomerase hTERT gene by human papillomavirus type 16 E6 oncoprotein. J Virol                   the progression, recurrence and prognosis of oral squamous cell carcinoma in
      2001;75:4467–72. https://doi.org/10.1128/JVI.75.9.4467-4472.2001.                               Taiwan. Oral Oncol 2007;43:122–9. https://doi.org/10.1016/j.
 [83] Oh ST, Kyo S, Laimins LA. Telomerase activation by human papillomavirus type                    oraloncology.2006.01.011.
      16 E6 protein: induction of human telomerase reverse transcriptase expression             [107] Fabricius EM, Kruse-Boitschenko U, Khoury R, Wildner GP, Raguse JD, Klein M.
      through Myc and GC-rich Sp1 binding sites. J Virol 2001;75:5559–66. https://                    Immunohistochemical determination of the appropriate anti-hTERT antibodies
      doi.org/10.1128/JVI.75.12.5559-5566.2001.                                                       for in situ detection of telomerase activity in frozen sections of head and neck
 [84] McMurray HR, McCance DJ. Human papillomavirus type 16 E6 activates TERT                         squamous cell carcinomas and tumor margin tissues. Int J Oncol 2009;34:
      gene transcription through induction of c-Myc and release of USF-mediated                       1257–79.
      repression. J Virol 2003;77:9852–61. https://doi.org/10.1128/jvi.77.18.9852-              [108] Qu Y, Dang S, Wu K, Shao Y, Yang Q, Ji M, et al. TERT promoter mutations
      9861.2003.                                                                                      predict worse survival in laryngeal cancer patients. Int J Cancer 2014;135:
 [85] Veldman T, Liu X, Yuan H, Schlegel R. Human papillomavirus E6 and Myc                           1008–10. https://doi.org/10.1002/ijc.28728.
      proteins associate in vivo and bind to and cooperatively activate the telomerase          [109] Annunziata C, Pezzuto F, Greggi S, Ionna F, Losito S, Botti G, et al. Distinct
      reverse transcriptase promoter. Proc Natl Acad Sci U S A 2003;100:8211–6.                       profiles of TERT promoter mutations and telomerase expression in head and neck
      https://doi.org/10.1073/pnas.1435900100.                                                        cancer and cervical carcinoma. Int J Cancer 2018;143:1153–61. https://doi.org/
 [86] Rajagopalan D, Pandey AK, Xiuzhen MC, Lee KK, Hora S, Zhang Y, et al. TIP60                     10.1002/ijc.31412.
      represses telomerase expression by inhibiting Sp1 binding to the TERT promoter.           [110] Mundi N, Prokopec SD, Ghasemi F, Warner A, Patel K, MacNeil D, et al. Genomic
      PLoS Pathog 2017;13:e1006681. https://doi.org/10.1371/journal.ppat.1006681.                     and human papillomavirus profiling of an oral cancer cohort identifies TP53 as a
 [87] Gewin L, Myers H, Kiyono T, Galloway DA. Identification of a novel telomerase                   predictor of overall survival. Cancers Head Neck 2019;4:5. https://doi.org/
      repressor that interacts with the human papillomavirus type-16 E6/E6-AP                         10.1186/s41199-019-0045-0.
      complex. Genes Dev 2004;18:2269–82. https://doi.org/10.1101/gad.1214704.                  [111] Arantes LMRB, Cruvinel-Carloni A, de Carvalho AC, Sorroche BP, Carvalho AL,
 [88] Xu M, Luo W, Elzi DJ, Grandori C, Galloway DA. NFX1 interacts with mSin3A/                      Scapulatempo-Neto C, et al. TERT promoter mutation C228T increases risk for
      histone deacetylase to repress hTERT transcription in keratinocytes. Mol Cell Biol              tumor recurrence and death in head and neck cancer patients. Front Oncol 2020;
      2008;28:4819–28. https://doi.org/10.1128/MCB.01969-07.                                          10:1275. https://doi.org/10.3389/fonc.2020.01275.
 [89] Billingsley CL, Chintala S, Katzenellenbogen RA. Post-Transcriptional Gene                [112] Yilmaz I, Erkul BE, Ozturk Sari S, Issin G, Tural E, Terzi Kaya Terzi N, et al.
      Regulation by HPV 16E6 and Its Host Protein Partners. Viruses 2022;14:1483.                     Promoter region mutations of the telomerase reverse transcriptase (TERT) gene in
      https://doi.org/10.3390/v14071483.                                                              head and neck squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral
 [90] Wang A, Zhou D, Krawczyk E, Li T, Simic V, Lu J, et al. Overexpression of the                   Radiol 2020;130:63–70. https://doi.org/10.1016/j.oooo.2020.02.015.
      telomerase holoenzyme induces EMT and tumorigenesis of HPV-immortalized                   [113] Giunco S, Boscolo-Rizzo P, Rampazzo E, Tirelli G, Alessandrini L, Di Carlo R, et al.
      keratinocytes. J Med Virol 2023;95:e28681. https://doi.org/10.1002/jmv.28681.                   TERT promoter mutations and rs2853669 polymorphism: useful markers for
 [91] Tribius S, Hoffmann AS, Bastrop S, Gorogh T, Haag J, Rocken C, et al. HPV status                clinical outcome stratification of patients with oral cavity squamous cell
      in patients with head and neck of carcinoma of unknown primary site: HPV,                       carcinoma. Front Oncol 2021;11. https://doi.org/10.3389/fonc.2021.782658.
      tobacco smoking, and outcome. Oral Oncol 2012;48:1178–84. https://doi.org/                [114] Kim H, Kwon MJ, Park B, Choi HG, Nam ES, Cho SJ, et al. Negative prognostic
      10.1016/j.oraloncology.2012.05.022.                                                             implication of TERT promoter mutations in human papillomavirus-negative
 [92] Miller J, Dakic A, Chen R, Palechor-Ceron N, Dai Y, Kallakury B, et al. HPV16 E7                tonsillar squamous cell carcinoma under the new 8th AJCC staging system. Indian
      protein and hTERT proteins defective for telomere maintenance cooperate to                      J Surg Oncol 2021;12(Suppl 1):134–43. https://doi.org/10.1007/s13193-020-
      immortalize human keratinocytes. PLoS Pathog 2013;9:e1003284. https://doi.                      01200-9.
      org/10.1371/journal.ppat.1003284.                                                         [115] Yu Y, Fan D, Song X, Zakeri K, Chen L, Kang J, et al. TERT promoter mutations are
 [93] Herzog AE, Somayaji R, Nör JE. Bmi-1: A master regulator of head and neck                      enriched in oral cavity cancers and associated with locoregional recurrence. JCO
      cancer stemness. Front Oral Health 2023 Jan;16(4):1080255. https://doi.org/                     Precis Oncol 2021;5:2000515. https://doi.org/10.1200/PO.20.00515.
      10.3389/froh.2023.1080255.                                                                [116] Haraguchi K, Habu M, Yada N, Sasaguri M, Yoshioka I, Tominaga K. Human
 [94] Sharaf K, Lechner A, Haider SP, Wiebringhaus R, Walz C, Kranz G, et al.                         telomerase reverse transcriptase protein expression is associated with survival in
      Discrimination of cancer stem cell markers ALDH1A1, BCL11B, BMI-1, and CD44                     patients with oral squamous cell carcinoma. Int J Clin Exp Pathol 2022;15:29–37.
      in different tissues of HNSCC patients. Curr Oncol 2021;28:2763–74. https://doi.                PMID: 35145581.
      org/10.3390/curroncol28040241.                                                            [117] Xian S, Dosset M, Castro A, Carter H, Zanetti M. Transcriptional analysis links B
 [95] Mori T. Involvement of the p53-p16/RB pathway control mechanism in early-                       cells and TERT expression to favorable prognosis in head and neck cancer. PNAS
      stage carcinogenesis in head and neck squamous cell carcinoma. Pathol Int 2022;                 Nexus 2023;2:pgad046. https://doi.org/10.1093/pnasnexus/pgad046.
      72:577–88. https://doi.org/10.1111/pin.13279.                                             [118] Johnson DE, Burtness B, Leemans CR, Lui VWY, Bauman JE, Grandis JR. Head
 [96] Rasouli S, Dakic A, Wang Q-E, Mitchell D, Blakaj DM, Putluri N, et al.                          and neck squamous cell carcinoma. Nat Rev Dis Primers 2020;6:92. https://doi.
      Noncanonical functions of telomerase and telomeres in viruses-associated cancer.                org/10.1038/s41572-020-00224-3.
      J Med Virol 2024;96:e29665. https://doi.org/10.1002/jmv.29665.                            [119] Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov 2022;12[15:
                                                                                                      31–46. https://doi.org/10.1158/2159-8290.CD-21-1059.
                                                                                           10
S. Giunco et al.                                                                                                                                        Oral Oncology 161 (2025) 107169
[120] Morris LGT, Chandramohan R, West L, Zehir A, Chakravarty D, Pfister DG, et al.             [135] Sandulache VC, Kirby RP, Lai SY. Moving from conventional to adaptive risk
      The molecular landscape of recurrent and metastatic head and neck cancers:                       stratification for oropharyngeal cancer. Front Oncol 2024;14:1287010. https://
      insights from a precision oncology sequencing platform. JAMA Oncol 2017;3:                       doi.org/10.3389/fonc.2024.1287010.
      244–55. https://doi.org/10.1001/jamaoncol.2016.1790.                                       [136] Bates JE, Steuer CE. HPV as a carcinomic driver in head and neck cancer: a de-
[121] Guterres AN, Villanueva J. Targeting telomerase for cancer therapy. Oncogene                     escalated future? Curr Treat Options Oncol 2022;23:325–32. https://doi.org/
      2020;39:5811–24. https://doi.org/10.1038/s41388-020-01405-w.                                     10.1007/s11864-022-00950-8.
[122] MacKenzie D, Watters AK, To JT, Young MW, Muratori J, Wilkoff MH, et al. ALT               [137] Lim YX, Mierzwa ML, Sartor MA, D’Silva NJ. Clinical, morphologic and molecular
      positivity in human cancers: prevalence and clinical insights. Cancers (Basel)                   heterogeneity of HPV-associated oropharyngeal cancer. Oncogene 2023;42:
      2021;13:2384. https://doi.org/10.3390/cancers13102384.                                           2939–55. https://doi.org/10.1038/s41388-023-02819-y.
[123] Sohn EJ, Goralsky JA, Shay JW, Min J. The molecular mechanisms and                         [138] Kaka AS, Kumar B, Kumar P, Wakely PE, Kirsch CM, Old MO, et al. Highly
      therapeutic prospects of alternative lengthening of telomeres (ALT). Cancers                     aggressive HPV-related oropharyngeal cancer: clinical, radiologic, and pathologic
      (Basel) 2023;15:1945. https://doi.org/10.3390/cancers15071945.                                   characteristics. Oral Surg Oral Med Oral Pathol Oral Radiol 2013;116:327–35.
[124] Lupatov AY, Yarygin KN. Telomeres and telomerase in the control of stem cells.                   https://doi.org/10.1016/j.oooo.2013.04.011.
      Biomedicines 2022;10:2335. https://doi.org/10.3390/biomedicines10102335.                   [139] Chen AM. De-escalated radiation for human papillomavirus virus-related
[125] Salloum R, Hummel TR, Kumar SS, Dorris K, Li S, Lin T, et al. A molecular biology                oropharyngeal cancer: Who, why, what, where, when, how, how much…and
      and phase II study of imetelstat (GRN163L) in children with recurrent or                         what next? Radiother Oncol 2024:110373. https://doi.org/10.1016/j.
      refractory central nervous system malignancies: a pediatric brain tumor                          radonc.2024.110373.
      consortium study. J Neurooncol 2016;129:443–51. https://doi.org/10.1007/                   [140] Benchetrit L, Torabi SJ, Givi B, Haughey B, Judson BL. Prognostic significance of
      s11060-016-2189-7.                                                                               extranodal extension in HPV-mediated oropharyngeal carcinoma: a systematic
[126] Mascarenhas J, Harrison CN, Kiladjian JJ, Komrokji RS, Koschmieder S,                            review and meta-analysis. Otolaryngol Head Neck Surg 2021;164:720–32.
      Vannucchi AM, et al. Imetelstat in intermediate-2 or high-risk myelofibrosis                     https://doi.org/10.1177/0194599820951176.
      refractory to JAK inhibitor: IMpactMF phase III study design. Future Oncol 2022;           [141] Furlan C, Polesel J, Barzan L, Franchin G, Sulfaro S, Romeo S, et al. Prognostic
      18:2393–402. https://doi.org/10.2217/fon-2022-0235.                                              significance of LINE-1 hypomethylation in oropharyngeal squamous cell
[127] Xu H, Di Antonio M, McKinney S, Mathew V, Ho B, O’Neil NJ, et al. CX-5461 is a                   carcinoma. Clin Epigenetics 2017;9:58. https://doi.org/10.1186/s13148-017-
      DNA G-quadruplex stabilizer with selective lethality in BRCA1/2 deficient                        0357-z.
      tumours. Nat Commun 2017;8:14432. https://doi.org/10.1038/ncomms14432.                     [142] Casarotto M, Lupato V, Giurato G, Guerrieri R, Sulfaro S, Salvati A, et al. LINE-1
[128] Fujita K, Kimura M, Kondo N, Sakakibara A, Sano D, Ishiguro Y, et al. Anti-tumor                 hypomethylation is associated with poor outcomes in locoregionally advanced
      effects of telomelysin for head and neck squamous cell carcinoma. Oncol Rep                      oropharyngeal cancer. Clin Epigenetics 2022;14:171. https://doi.org/10.1186/
      2008;20:1363–8. https://doi.org/10.3892/or_00000153.                                             s13148-022-01386-5.
[129] Kondo N, Tsukuda M, Kimura M, Fujita K, Sakakibara A, Takahashi H, et al.                  [143] Freitag J, Wald T, Kuhnt T, Gradistanac T, Kolb M, Dietz A, et al. Extracapsular
      Antitumor effects of telomelysin in combination with paclitaxel or cisplatin on                  extension of neck nodes and absence of human papillomavirus 16-DNA are
      head and neck squamous cell carcinoma. Oncol Rep 2010;23:355–63. https://doi.                    predictors of impaired survival in p16-positive oropharyngeal squamous cell
      org/10.3892/or_00000643.                                                                         carcinoma. Cancer 2020;126:1856–72. https://doi.org/10.1002/cncr.32667.
[130] Wang Y, Zhang X, Chen G, Shao M. Clinical research progress of telomerase                  [144] Huang SH, O’Sullivan B, Su J, Bartlett E, Kim J, Waldron JN, et al. Prognostic
      targeted cancer immunotherapy: a literature review. Transl Cancer Res 2024;13:                   importance of radiologic extranodal extension in HPV-positive oropharyngeal
      3904–21. https://doi.org/10.21037/tcr-24-196.                                                    carcinoma and its potential role in refining TNM-8 cN-classification. Radiother
[131] Ellingsen EB, Mangsbo SM, Hovig E, Gaudernack G. Telomerase as a target for                      Oncol 2020;144:13–22. https://doi.org/10.1016/j.radonc.2019.10.011.
      therapeutic cancer vaccines and considerations for optimizing their clinical               [145] Krsek A, Baticic L, Sotosek V, Braut T. The role of biomarkers in HPV-positive
      potential. Front Immunol 2021;12:682492. https://doi.org/10.3389/                                head and neck squamous cell carcinoma: towards precision medicine. Diagnostics
      fimmu.2021.682492.                                                                               (Basel) 2024;14:1448. https://doi.org/10.3390/diagnostics14131448.
[132] Sugarman ET, Zhang G, Shay JW. In perspective: an update on telomere targeting             [146] Hutchinson M-K-N-D, Mierzwa M. D’Silva NJ. Radiation resistance in head and
      in cancer. Mol Carcinog 2019;58:1581–8. https://doi.org/10.1002/mc.23035.                        neck squamous cell carcinoma: dire need for an appropriate sensitizer. Oncogene
[133] Rebucci-Peixoto M, Vienot A, Adotevi O, Jacquin M, Ghiringhelli F, de la                         2020;39:3638–49. https://doi.org/10.1038/s41388-020-1250-3.
      Fouchardière C, et al. A Phase II study evaluating the interest to combine UCPVax,        [147] Lehrich BM, Zhang J, Monga SP, Dhanasekaran R. Battle of the biopsies: Role of
      a telomerase CD4 TH1-inducer cancer vaccine, and atezolizumab for the                            tissue and liquid biopsy in hepatocellular carcinoma. J Hepatol 2024;80:515–30.
      treatment of HPV positive cancers: VolATIL study. Front Oncol 2022;12:957580.                    https://doi.org/10.1016/j.jhep.2023.11.030.
      https://doi.org/10.3389/fonc.2022.957580.                                                  [148] Zhang Y, Dakic A, Chen R, Dai Y, Schlegel R, Liu X. Direct HPV E6/Myc
[134] Zhang M, Hong AM. The human papillomavirus confers radiosensitivity in                           interactions induce histone modifications, Pol II phosphorylation, and hTERT
      oropharyngeal cancer cells by enhancing DNA double strand break. Oncotarget                      promoter activation. Oncotarget 2017;8:96323–39. https://doi.org/10.18632/
      2020;11:1417–26. https://doi.org/10.18632/oncotarget.27535.                                      oncotarget.22036.
11