Hut 2025 Laryngeal CA in The Modern Era
Hut 2025 Laryngeal CA in The Modern Era
                                          1   Department of Doctoral Studies, “Victor Babes” University of Medicine and Pharmacy Timisoara,
                                              Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; [email protected] (A.-R.H.);
                                              [email protected] (D.P.); [email protected] (A.C.M.)
                                          2   ENT Department, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square
                                              No. 2, 300041 Timisoara, Romania; [email protected] (G.I.); [email protected] (D.H.);
                                              [email protected] (L.M.); [email protected] (M.C.); [email protected] (N.C.B.)
                                          3   ENT Department, Emergency City Hospital, 300254 Timisoara, Romania
                                          4   Hôpital Européen Georges-Pompidou, Assistance Publique-Hôpitaux de Paris, Université Paris Cité,
                                              20 Rue Leblanc, 75015 Paris, France; [email protected]
                                          5   Center for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes”
                                              University of Medicine and Pharmacy, 300041 Timisoara, Romania
                                          *   Correspondence: [email protected]
                               upper aerodigestive tract [1]. According to global cancer statistics for 2022, laryngeal
                               cancer (LC) is among the most prevalent cancers, with 188,960 new cases and 103,216
                               deaths reported [2]. The vast majority of these—between 85% and 95%—are classified as
                               squamous cell carcinomas (SCC) [3]. The epidemiology of LC reveals a substantial burden.
                               In 2022, global incidence rates reached over 165,598 cases in men and over 23,362 in women,
                               with associated deaths exceeding 90,256 and 12,960, respectively [2].
                                     Geographically, Cuba (7.8 per 100,000) and Montenegro (7.0 per 100,000) report the
                               highest incidence rates, while Eswatini (0.18 per 100,000) and Cameroon (0.31 per 100,000)
                               have the lowest. Asia is the most prevalent continent for LC, followed by Europe, whilst
                               the epidemiologic burden in Africa remains low [4]. Within Europe, a high-prevalence
                               region, incidence varies considerably; Spain reports rates above 12 per 100,000, while the
                               UK’s rates are below 5 per 100,000 [5]. In Romania, oral malignant tumors are prevalent,
                               with approximately 2388 new cases annually [6].
                                     Regarding LC mortality, Cuba (3.9 per 100,000) and Montenegro (3.5 per 100,000) also
                               register high mortality rates, in contrast to Iceland (0.08 per 100,000) and Martinique (0.11
                               per 100,000), which have the lowest [4]. These variations are linked to socioeconomic factors,
                               with higher incidence in areas with lower average incomes and education [7]. Regarding
                               gender differences, in 2021, the age-standardized DALY rate of LC in males was approx-
                               imately 7.13 times higher than that in females, with males experiencing 282.12 cases per
                               100,000 individuals, compared to 39.59 cases per 100,000 individuals in females [8]. Higher
                               Human Development Index (HDI) regions tend to have higher incidence but lower mortal-
                               ity, reflecting disparities in healthcare access and quality, while lower Sociodemographic
                               Index (SDI) regions experience higher mortality rates [9,10]. Key etiological factors include
                               tobacco smoking [11,12], alcohol consumption [13–15], high-risk human papillomavirus
                               (HPV) infection [16], and occupational exposures such as asbestos [17,18].
                                     Characteristic clinical manifestations often prompt the diagnosis of LC. These symp-
                               toms, which can vary significantly depending on the tumour’s location and size, commonly
                               include a persistent lump or non-healing sore, ongoing throat pain, difficulty swallowing
                               (dysphagia), and alterations in voice quality, such as hoarseness [19]. Despite significant
                               advancements in instruments like flexible laryngoscopes, surgical techniques, and chemora-
                               diation therapy, the mortality rate remains high; the 5-year survival rate is 64% because
                               approximately two-thirds of patients are diagnosed with advanced cancer, leading to a
                               poor prognosis [20]. Furthermore, even with improvements in treatment modalities, the
                               American Cancer Society has reported a tendency for the 5-year survival rate for laryngeal
                               cancer patients to decline [21].
                                     Treatment options for LC include surgery, radiation therapy, and chemotherapy, used
                               alone or in combination [22,23]. Recent advancements in immunotherapy and targeted
                               therapy have significantly impacted laryngeal cancer treatment, offering hope for patients
                               with recurrent or advanced disease. These therapies improve progression-free and over-
                               all survival rates, particularly in combination. The integration of immunotherapy with
                               targeted therapies like anti-EGFR has shown promising results in tumor response, and is
                               increasingly considered for managing recurrent locoregionally advanced squamous cell
                               carcinoma of the head and neck, including laryngeal cancer [24]. A complex interplay
                               of factors determines a patient’s prognosis in LC, broadly categorized as relating to the
                               host, the tumour itself, or the treatment strategy [25]. Host factors encompass individual
                               characteristics such as age, sex, nutritional status, general health and physical condition,
                               coexisting medical conditions’ presence, and the immune response’s robustness. Tumour-
                               related factors include the primary location of cancer, its TNM stage (which reflects the
                               size, involvement of lymph nodes, and presence of distant metastasis), the microscopic
                               grade (degree of abnormality), and whether any other primary cancers are present con-
J. Clin. Med. 2025, 14, 3367                                                                                          3 of 32
                               3. Results
                               3.1. Risk Factors of LC
                                     A combination of lifestyle and environmental factors significantly influences LC
                               development (Figure 1).
                                     Undoubtedly, tobacco use is the most prominent risk factor [27]. The risk of LC
                               increases substantially with both the duration and intensity of smoking, exhibiting a dose-
                               response relationship. However, this relationship may not be perfectly linear, with a
                               possible “saturation effect” at very high levels of consumption (more than 20 years and
                               over 30 cigarettes per day) [11,12]. Smoking cessation reduces risk, but it remains elevated
                               for up to 15 years after quitting [28]. The synergistic effect of tobacco’s carcinogens is
                               amplified by human papillomavirus [16]. Cigarette smoking correlates with a sevenfold
                               risk increase of LC [11]. Passive smoking also contributes to LC deaths [28]. Smokers have
J. Clin. Med. 2025, 14, 3367                                                                                                     4 of 32
J. Clin. Med. 2025, 14, x FOR PEER REVIEW likelihood of dying from LC [29]. Black smokers demonstrate a higher risk4level
                                    a higher                                                                         of 35
                                        Excessive   alcohol
                                         Undoubtedly,       consumption
                                                         tobacco  use is the is most
                                                                                a significant independent
                                                                                      prominent    risk factorrisk factor
                                                                                                                [27]. The for
                                                                                                                           riskLC,
                                                                                                                                 of ex-
                                                                                                                                    LC
                                  hibiting  a clear dose–response   relationship   where  both  the amount    and  duration  of
                                   increases substantially with both the duration and intensity of smoking, exhibiting a dose-  intake
                                  proportionally    elevate the
                                   response relationship.       risk [13–15].
                                                             However,           Alcohol accounts
                                                                         this relationship    may notfor abesubstantial portionwith
                                                                                                             perfectly linear,   of la-a
                                  ryngeal cancer-related mortality, especially in regions with high alcohol consumption
                                   possible “saturation effect” at very high levels of consumption (more than 20 years and
                                  levels [31]. In Europe, approximately 30% of individuals who died from LC were identified
                                   over 30 cigarettes per day) [11,12]. Smoking cessation reduces risk, but it remains elevated
                                  as alcoholics [31]. Moreover, the global contribution of alcohol to the incidence of LC may
                                   for up to 15 years after quitting [28]. The synergistic effect of tobacco s carcinogens is
                                  be on the rise [32].
                                   amplified by human papillomavirus [16]. Cigarette smoking correlates with a sevenfold
                                        While traditional risk factors like tobacco and alcohol remain significant, emerging con-
                                   risk increase of LC [11]. Passive smoking also contributes to LC deaths [28]. Smokers have
                                  cerns and socioeconomic disparities require further attention. Although initially marketed
                                   a higher likelihood of dying from LC [29]. Black smokers demonstrate a higher risk level
                                  as a safer alternative, e-cigarettes have raised concerns due to the presence of potentially
                                   than white smokers [30].
                                  carcinogenic substances in some e-liquids [33]. A Korean study found formaldehyde and
                                         Excessive alcohol consumption is a significant independent risk factor for LC,
                                  acetaldehyde in all 225 tested e-liquids [34]. Both of the compounds are classified as Group
                                   exhibiting a clear dose–response relationship where both the amount and duration of
                                  1 carcinogens with links to head and neck cancers [35]. N′ -nitrosonornicotine (NNN), a
                                   intake proportionally elevate the risk [13–15]. Alcohol accounts for a substantial portion
                                  TSNA, has been shown to induce head and neck tumours in animal studies [36]. PAHs, like
                                   of laryngeal cancer-related mortality, especially in regions with high alcohol consumption
                                  1-hydroxypyrene (1-HOP) and benzopyrene, have demonstrated carcinogenic effects on
                                   levels [31]. In Europe, approximately 30% of individuals who died from LC were
                                  the upper respiratory tract in animal models [36,37]. While in vivo studies on e-cigarettes
                                   identified as alcoholics [31]. Moreover, the global contribution of alcohol to the incidence
                                  and laryngeal mucosa are limited, one study in rats showed non-statistically significant
                                   of LC may be on the rise [32].
                                  hyperplasia and metaplasia after four weeks of exposure to e-cigarette aerosols [38].
                                         While traditional risk factors like tobacco and alcohol remain significant, emerging
                                        Opium, classified as a human carcinogen by the IARC (International Agency for
                                   concerns and socioeconomic disparities require further attention. Although initially
                                  Research on Cancer), has shown preliminary links to increased head and neck squamous
                                   marketed as a safer alternative, e-cigarettes have raised concerns due to the presence of
                                  cell carcinoma (HNSCC) risk, including LC, although more research is needed to solidify
                                   potentially carcinogenic substances in some e-liquids [33]. A Korean study found
                                  this connection. The mechanism likely involves the carcinogenic alkaloids present in opium,
                                   formaldehyde and acetaldehyde in all 225 tested e-liquids [34]. Both of the compounds
                                  which can induce DNA damage and mutations upon metabolic activation [39,40].
                                   are classified as Group 1 carcinogens with links to head and neck cancers [35]. N′-
                                        Beyond tobacco and alcohol, certain viral infections, notably high-risk strains of human
                                   nitrosonornicotine (NNN), a TSNA, has been shown to induce head and neck tumours in
                                  papillomavirus (HPV), have been implicated in LC development [41].
                                   animal studies [36]. PAHs, like 1-hydroxypyrene (1-HOP) and benzopyrene, have
                                        Over the past few decades, HPV has been recognized as a key etiological agent, espe-
                                   demonstrated carcinogenic effects on the upper respiratory tract in animal models [36,37].
                                  cially in oropharyngeal squamous cell carcinoma (OPSCC), where HPV-positive tumors
                                   While in vivo studies on e-cigarettes and laryngeal mucosa are limited, one study in rats
                                   showed non-statistically significant hyperplasia and metaplasia after four weeks of
                                   exposure to e-cigarette aerosols [38].
J. Clin. Med. 2025, 14, 3367                                                                                           5 of 32
                               form a distinct clinical and molecular subgroup compared with their HPV-negative coun-
                               terparts [42]. However, the etiological role of HPV in LC appears to vary significantly by
                               geographic region, with a lower prevalence in some areas, suggesting it is not always a
                               primary cause [43–45]. Moreover, EBV has been suggested as a risk factor for LC. EBV’s
                               genome and latent protein EBNA have been found in malignant laryngeal cells, suggesting
                               a potential role as a risk factor or cofactor, though its presence in LSCC can be inconsis-
                               tent [46]. EBV, like HPV, can produce oncoproteins that disrupt cell cycle control and
                               promote uncontrolled cell growth, contributing to carcinogenesis [47].
                                     Other identified risk factors include occupational exposure to asbestos [17,18] and
                               possibly nickel or ionizing radiation [48]. Chewing betel, particularly in combination with
                               tobacco and alcohol, substantially elevates risk, specifically in specific populations like
                               Taiwan [49,50]. Helicobacter pylori infection has also been linked to an increased risk of
                               LC [51], as has gastroesophageal reflux disease (GERD) and laryngopharyngeal reflux
                               (LPR), likely due to chronic inflammation. Chronic inflammation leads to the release of
                               reactive oxygen species (ROS) and inflammatory mediators, which can damage DNA,
                               promote cell proliferation, and create a microenvironment conducive to tumour develop-
                               ment [52–54]. Moreover, metabolic syndrome and its associated components (high blood
                               glucose, increased waist circumference, elevated triglycerides, high blood pressure, and
                               low HDL cholesterol) have also been identified as independent risk factors [55,56].
                                     The oral and throat microbiome is emerging as a significant factor. Differences in bac-
                               terial composition, particularly elevated levels of genera like Fusobacterium, Prevotella, and
                               Streptococcus, have been observed in LC patients compared to healthy individuals [57,58].
                               Fusobacterium, an invasive anaerobe, may contribute to chronic inflammation and carcino-
                               genesis, and shifts in the oral microbiome could serve as an early marker of LC [58,59]. The
                               microbiome’s influence extends to immune system modulation, metabolic regulation, and
                               even cancer promotion, with interactions with alcohol and tobacco further contributing
                               to LSCC development. Certain bacteria can produce carcinogenic metabolites, exacerbate
                               inflammation, and suppress anti-tumor immune responses [60–63].
                                     LC outcomes are significantly worsened by delays in diagnosis and treatment. Socioe-
                               conomic disparities such as lack of health insurance and limited healthcare access delaying
                               cancer screening contribute to these delays and to the number of deaths from modifiable
                               risk factors such as tobacco, alcohol and occupational exposures [64,65]. Men tend to have
                               an increased risk of LC [66–68] and have a poor prognosis in comparison to women [69].
                               Preventive measures, particularly reducing tobacco and alcohol consumption and im-
                               proving access to screening, are crucial for mitigating the burden of this disease [70,71].
                               Furthermore, in 2021, attributable deaths due to tobacco, occupational risks, and alcohol
                               were 66.46%, 5.92%, and 12.4%, respectively. While deaths attributable to these factors have
                               generally decreased since 1990, deaths due to occupational risks have increased in females.
                               The proportion of LC deaths attributable to tobacco was highest in high-middle sociodemo-
                               graphic index (SDI) regions (76.15%), while high SDI regions had the highest proportions
                               attributable to occupational risks (11.56%) and alcohol (19.31%) [72]. Country-specific data
                               reveals that Armenia (middle SDI) had the highest proportion of LC deaths attributable to
                               tobacco (83.86%), while the UK (high SDI) had the highest proportion linked to occupational
                               risks (20.11%), and Czechia (high SDI) had the highest proportion attributable to alcohol
                               (27.35%). These disparities highlight the complex interplay between individual behaviours,
                               environmental exposures, and access to preventative care and treatment, underscoring the
                               need for targeted interventions [72].
J. Clin. Med. 2025, 14, 3367                                                                                                                                        6 of 32
                               3.3. Diagnosis
                                     Accurate diagnosis of LC relies on a thorough patient history, a comprehensive phys-
                               ical examination, and appropriate diagnostic procedures. The patient’s history should
                               include detailed inquiries about risk factors like tobacco and alcohol use, current medica-
                               tions, and any coexisting medical conditions that might influence treatment decisions [82].
                               The clinical presentation of LC varies considerably depending on the tumour’s location and
                               size. Glottic tumours often present early with hoarseness, while supraglottic tumours may
                               manifest later with symptoms such as pain, persistent hoarseness, or dysphagia (difficulty
                               swallowing) [82]. The diagnostic modalities of LC are summarized in Table 2. Direct
                               visualization of the larynx is essential, typically achieved through indirect laryngoscopy,
                               flexible fiberoptic laryngoscopy, or video stroboscopy. Videostroboscopy, in particular,
                               demonstrates high sensitivity (96.8%) and specificity (92.8%) in predicting the invasiveness
                               of laryngeal lesions [83]. Tissue confirmation through biopsy of the primary tumour or
                               fine-needle aspiration of suspicious lymph nodes is crucial for definitive diagnosis. Imag-
                               ing studies play a vital role in staging cancer. Computed tomography (CT) is valuable
                               for assessing bone involvement, while positron emission tomography combined with CT
                               (PET/CT) helps detect recurrences, local and nodal spread, and distant metastases [84].
                               Magnetic resonance imaging (MRI) offers superior sensitivity (80%) and specificity (92.9%)
                               compared to CT (60% sensitivity, 85.7% specificity) in evaluating cartilage and soft tissue
                               invasion [85]. Narrow-band imaging (NBI) has also emerged as a highly sensitive (97%)
                               and specific (92.5%) technique for identifying both LC and its precursor lesions [86].
                                    Traditional diagnostic approaches for LC, such as imaging and tissue biopsy, are
                               fundamental but can be limited in detecting early-stage disease. Over the last decade, there
                               has been increasing interest in using liquid biopsies to detect cancer-specific biomarkers
                               in patients’ body fluids [87,88]. Liquid biopsy has been reported to play roles in early
                               malignancy detection in diverse tumor types [89,90]. As a rapid and noninvasive approach,
J. Clin. Med. 2025, 14, 3367                                                                                            8 of 32
                                regulators, oncogenes and tumour suppressors, growth factor pathway components, angio-
                                genic factors, structural proteins, sex hormone signalling components, and immunological
                                markers [116]. Table 3 summarize the important biomarkers and their function in laryngeal
                                cell cancer.
Table 3. Molecular Biomarkers in Laryngeal SCC: Biological Roles and Clinical Implications.
                                     Alterations that enhance the function of the epidermal growth factor receptor (EGFR)
                                axis and defects that diminish the function of the transforming growth factor-β receptor
                                (TGF-βR) axis collectively promote unregulated cellular proliferation, reduced apoptosis,
                                and increased cell survival. In the context of EGFR, overexpression or activating muta-
                                tions result in the receptor being persistently active, thereby continuously stimulating
                                the RAS–RAF–MEK–ERK and PI3K–AKT signaling pathways. This activation leads to
                                the upregulation of pro-proliferative markers such as Cyclin D1 and Ki-67, as well as
                                anti-apoptotic proteins like Bcl-2, while concurrently downregulating cell-cycle inhibitors
                                and increased cell survival. In the context of EGFR, overexpression or activating muta-
                                tions result in the receptor being persistently active, thereby continuously stimulating the
J. Clin. Med. 2025, 14, 3367    RAS–RAF–MEK–ERK and PI3K–AKT signaling pathways. This activation leads to the                        up-
                                                                                                                                 10 of 32
                                regulation of pro-proliferative markers such as Cyclin D1 and Ki-67, as well as anti-apop-
                                totic proteins like Bcl-2, while concurrently downregulating cell-cycle inhibitors such as
                                p27. as
                               such   Conversely,     the loss
                                        p27. Conversely,     theofloss
                                                                   TGF-βR     signaling,
                                                                       of TGF-βR          due todue
                                                                                     signaling,   receptor   mutations
                                                                                                     to receptor        or disruptions
                                                                                                                  mutations  or disrup-
                                in theinSMAD
                               tions     the SMADpathway,     impairs
                                                       pathway,         its normal
                                                                   impairs            growth-inhibitory
                                                                             its normal   growth-inhibitoryfunctions,  resulting
                                                                                                                functions,        in de-
                                                                                                                           resulting   in
                                creased levels
                               decreased         of of
                                            levels  p53p53andand
                                                               p27. ThisThis
                                                                   p27.   lossloss
                                                                                effectively  removes
                                                                                    effectively       critical
                                                                                                removes        regulatory
                                                                                                           critical        mechanisms
                                                                                                                    regulatory  mecha-
                                that inhibit
                               nisms          cell-cycle
                                       that inhibit        progression
                                                      cell-cycle          and apoptosis.
                                                                  progression                TheseThese
                                                                                 and apoptosis.     opposing    molecular
                                                                                                          opposing          events
                                                                                                                     molecular      con-
                                                                                                                                 events
                                verge within
                               converge         the the
                                           within    nucleus,   driving
                                                         nucleus,         DNA
                                                                    driving   DNAreplication,  promoting
                                                                                      replication, promotingsurvival  under
                                                                                                                 survival    stress
                                                                                                                          under     con-
                                                                                                                                  stress
                                ditions, and
                               conditions,     ultimately
                                             and  ultimately contributing
                                                               contributingtotothethedevelopment
                                                                                       developmentandand aggressiveness       laryngeal
                                                                                                          aggressiveness of laryngeal
                                cancer(Figure
                               cancer   (Figure2).2).
                               Figure2.2.Core
                               Figure    CoreMolecular
                                              MolecularPathways
                                                       PathwaysDriving
                                                                DrivingLaryngeal
                                                                       LaryngealSquamous
                                                                                 SquamousCell
                                                                                         CellCarcinoma
                                                                                              CarcinomaPathogenesis.
                                                                                                        Pathogenesis.
                                     Biomarkers
                                      Biomarkers in in laryngeal
                                                        laryngeal squamous
                                                                   squamous cell
                                                                               cell carcinoma
                                                                                     carcinoma (LSCC)
                                                                                                   (LSCC) encompass
                                                                                                            encompass aawidewiderange
                                                                                                                                    range
                               of
                                of molecules
                                   molecules that
                                               that modulate
                                                     modulate keykey cellular
                                                                      cellular processes
                                                                               processes andand may
                                                                                                  may impact
                                                                                                        impact tumor
                                                                                                                 tumor behavior
                                                                                                                         behavior and and
                               clinical
                                clinicaloutcomes.
                                         outcomes.
                                     Long
                                      Long noncoding
                                            noncoding RNAs RNAs (lncRNAs)
                                                                 (lncRNAs) areare RNA
                                                                                  RNA molecules
                                                                                          molecules that
                                                                                                       that do
                                                                                                             do not
                                                                                                                 notcode
                                                                                                                     codefor
                                                                                                                           forproteins
                                                                                                                                proteins
                               but
                                butregulate
                                    regulategene
                                              geneexpression
                                                     expressionandandvarious
                                                                       variouscellular
                                                                                cellularfunctions.
                                                                                          functions.Their
                                                                                                       Theirdysregulation
                                                                                                              dysregulationisislinked
                                                                                                                                   linked
                               to
                                tocancer
                                   cancerdevelopment,
                                           development,including
                                                            includingLSCC,
                                                                       LSCC, where
                                                                               wheremanymanylncRNAs
                                                                                                lncRNAs are are upregulated
                                                                                                                upregulatedin  in tumor
                                                                                                                                   tumor
                               tissues.   Thisoverexpression
                                tissues. This   overexpressionoftenoftenpromotes
                                                                          promotestumortumor    progression,
                                                                                             progression,       enhances
                                                                                                             enhances       metastasis,
                                                                                                                        metastasis,    re-
                               reduces   radiosensitivity,and
                                duces radiosensitivity,     andworsens
                                                                 worsens overall
                                                                           overall survival,
                                                                                    survival, partly by activating
                                                                                                           activating pathways
                                                                                                                       pathways such such
                               as
                                as Wnt,
                                   Wnt, Sox-2, andandTGF-β1
                                                        TGF-β1[145–147,157].
                                                                 [145–147,157].Owing
                                                                                   Owing  to to  their
                                                                                             their     stability
                                                                                                    stability     in body
                                                                                                              in body       fluids
                                                                                                                       fluids  and andease
                               ease  of detection
                                of detection       by non-invasive
                                              by non-invasive         methods,
                                                                 methods,        lncRNAs
                                                                            lncRNAs           are considered
                                                                                        are considered          promising
                                                                                                           promising        biomarker
                                                                                                                       biomarker      can-
                               candidates   [158].
                                didates [158].
                                     The cell cycle, which is frequently disrupted in cancer, offers several potential biomark-
                               ers. Ki-67, a nuclear protein expressed during all active phases of the cell cycle but absent
                               in resting cells, serves as a direct marker of proliferation. In LSCC, elevated Ki-67 levels are
                               associated with poorly differentiated tumors, advanced TNM stages, increased recurrence
                               risk, and shorter disease-free survival [133–136]. However, its relationship with radiother-
                               apy response is complex; while high levels might imply increased radiosensitivity in some
                               studies [159,160], other reports suggest that low Ki-67 can be linked to better local control in
                               early-stage tumors, possibly due to factors like DNA repair capacity and hypoxia [161,162].
                               Cyclin D1, which drives the G1 to S phase transition by complexing with CDKs 4 and 6, is
                               commonly overexpressed in LSCC and correlates with lymph node metastasis and poorer
                               survival; its overexpression may also occur alongside TP53 mutations [131,139,140]. In
                               contrast, the cyclin-dependent inhibitor p27 is frequently underexpressed in LSCC, with
J. Clin. Med. 2025, 14, 3367                                                                                          11 of 32
                               low levels linked to tumor recurrence and poorer outcomes [140,163]. An inverse relation-
                               ship between cyclin D1 and p27 is evident, with patients displaying cyclin D1 positivity
                               alongside p27 negativity often faring worse [116]. In addition, p16—a CDKI closely related
                               to the HPV 16 status frequently seen in head and neck cancers—is typically downregulated
                               in tumors with high cyclin D1, and its presence tends to indicate better treatment response
                               and survival [164].
                                     Deregulation of apoptotic pathways is another hallmark of LSCC. Bcl-2, an intra-
                               cellular membrane protein that inhibits apoptosis by modulating cytochrome C release
                               and interacting with pro-apoptotic proteins, shows conflicting associations. While some
                               studies report no correlation between Bcl-2 expression and clinical outcomes [117–121],
                               other investigations have linked its overexpression to lymph node metastasis, advanced
                               stage, poor differentiation, increased recurrence, and radioresistance [165]. A meta-analysis
                               by Silva et al., (2023) even suggests that Bcl-2 overexpression may be associated with poorer
                               lymph node metastasis, overall survival (OS), and disease-free survival (DFS), although
                               these findings should be interpreted with caution because of study variability and potential
                               bias [166].
                                     Tumor suppressor and oncogenes also play crucial roles in LSCC. The tumor sup-
                               pressor p53, which activates cell cycle arrest, DNA repair, senescence, or apoptosis in
                               response to stress, is mutated in approximately 60–80% of LSCC cases [167–169]. These
                               mutations are frequently linked to more aggressive disease features; however, consensus on
                               the prognostic role of p53 remains elusive due to heterogeneous and sometimes conflicting
                               study results [170–173].
                                     Growth factor signaling contributes further to LSCC progression. Epidermal growth
                               factor receptor (EGFR), a transmembrane receptor tyrosine kinase that binds ligands such
                               as EGF and TGF-α, triggers intracellular pathways leading to increased cell proliferation,
                               angiogenesis, and survival. EGFR overexpression in LSCC is associated with progression
                               to malignancy, a higher risk of metastasis, and reduced survival, particularly when found
                               together with high levels of cyclin D1 and Ki-67 [173–177]. Similarly, alterations in the TGF-
                               β pathway—most notably the loss of TGF-β receptor II expression in lesions progressing to
                               invasive carcinoma—reduce the growth-suppressive effects of TGF-β and promote tumor
                               advancement [169–178].
                                     Angiogenesis, a process vital for tumor growth and metastasis, is reflected by several
                               markers in LSCC. Vascular endothelial growth factor (VEGF) increases vascular permeabil-
                               ity and stimulates endothelial cell proliferation and migration, and its upregulation in LSCC
                               correlates with dysplasia progression, local recurrence, metastasis, and shorter disease-free
                               survival, though some studies find no significant association [126,150,179–182]. Additional
                               angiogenic markers such as angiogenin and CD105 (endoglin) are also implicated, with
                               their elevated expression correlating with recurrence, advanced disease, and poorer clinical
                               outcomes [126,148–150,183,184].
                                     Structural proteins that maintain cell adhesion and tissue integrity are also signifi-
                               cant. E-cadherin, a transmembrane glycoprotein essential for cell-cell adhesion, is often
                               downregulated in LSCC, correlating with poorer tumor differentiation, increased risk of
                               nodal metastasis, and advanced tumor stage; reduced expression may also be linked to
                               shorter disease-free survival [125–127,185]. The cell surface glycoprotein CD44, a known
                               marker of cancer stem cells, is similarly associated with higher tumor grade and poorer
                               5-year survival [122–124]. Overexpression of focal adhesion kinase (FAK) and cortactin—an
                               actin-binding protein involved in cell motility and invasion—has been linked to aggressive
                               behavior and recurrence in LSCC [126,151–154].
                                     The role of sex hormone signaling in LSCC remains debated. Estrogen receptors
                               (ERα and ERβ, including variants such as ERα66 and ERα36) display variable expression
J. Clin. Med. 2025, 14, 3367                                                                                         12 of 32
                               patterns; while early-stage LSCC tumors may show increased levels compared to normal
                               tissue, advanced stages often exhibit a shift in receptor subtype expression that correlates
                               with a more unfavorable prognosis [155,186,187]. Likewise, progesterone receptor (PR)
                               expression is higher in poorly differentiated LSCC and those with nodal metastasis, whereas
                               androgen receptor (AR) expression is typically reduced in invasive tumors. Elevated
                               prolactin receptor (PRLR) expression has also been noted in LSCC and correlates with
                               poorer survival outcomes [155,156].
                                     Immunological biomarkers reflect the critical role of the host immune response in
                               modulating tumor growth. Tumor-infiltrating lymphocytes (TILs), particularly cytotoxic
                               CD8+ T cells, are associated with improved survival in LSCC—even in cases of tobacco-
                               related disease [188–190]. LSCC continues to have a poor prognosis, with a 5-year survival
                               rate of 50 to 60% and suboptimal functional outcomes. The expression of PD-L1 and the
                               tumor microenvironment markers (CD4, CD8, CD68, and CD163) were examined in LSCC
                               using immunohistochemistry. PD-L1 expression demonstrated a statistically significant
                               positive correlation with all the tumor microenvironment cells studied. Higher expressions
                               of CD68 and CD163 were significantly associated with worse clinical outcomes in LSCC
                               patients [191]. To determine which LSCC patients might benefit from immunomodulation
                               therapies, it is crucial to understand the relationship between PD-L1 expression, immune
                               cell distribution, and prognosis. Conversely, the expression of programmed death-ligand 1
                               (PD-L1) on tumor and immune cells, which aids tumors in evading immune attacks, has
                               been linked to clinical outcomes and may make tumors amenable to immune checkpoint
                               inhibitor therapy [140,192–195].
                                     Emerging biomarkers in LSCC continue to broaden our understanding of tumor biol-
                               ogy. Among these, heat shock proteins (HSPs) have diverse roles: HSP27 and HSP70 are
                               linked to advanced tumor stage, chemoresistance (particularly to cisplatin), and radiother-
                               apy resistance [196–201] whereas HSP47 appears to function as a tumor suppressor, with
                               higher expression correlating with better prognosis and longer overall survival [202]. Met-
                               allothioneins (MTs), especially the MT1 and MT2 isoforms, are found at higher levels in ma-
                               lignant laryngeal lesions than in benign or dysplastic ones, and genetic variations in MT2A
                               may increase LSCC risk, making them potential early biomarkers [203–206]. Components of
                               the oxidative stress response, such as nuclear factor erythroid 2-related factor 2 (Nrf2) and
                               heme oxygenase-1 (HO-1), are also implicated; increased nuclear Nrf2 expression correlates
                               with cisplatin resistance, while HO-1 may counteract cisplatin-induced apoptosis, although
                               its prognostic role in LSCC is still under investigation [207–212]. Cyclooxygenase-2 (COX-2)
                               is another emerging marker; its elevated expression in LSCC is linked to enhanced an-
                               giogenesis, inflammatory signaling, and resistance to radiotherapy, and it is associated
                               with poorer clinical outcomes [213–219]. Lastly, several microRNAs (miRNAs)—such as
                               Hs_miR-21_5p, Hs_miR-218_3p, and Hs_miR-210_3p, which are expressed exclusively in
                               malignant laryngeal lesions—are implicated in regulating tumor cell migration, invasion,
                               and treatment resistance, further supporting their utility as precise biomarkers [220–227].
                               IB) and submental (level IA) nodes are seldom involved. Approximately 55% of patients
                               with supraglottic carcinoma present with clinically involved lymph nodes at diagnosis,
                               with 16% showing bilateral involvement.
                                     Furthermore, up to half of clinically node-negative patients may harbour occult nodal
                               metastases. The risk of lymph node involvement increases with tumour size and grade,
                               reaching approximately 40% for T1 and T2 tumours and 60% for T3 and T4 lesions [228–230].
                               In contrast, the subglottis has a less developed lymphatic network, resulting in a lower
                               incidence of lymph node metastasis (20% to 50%). Subglottic lymphatic channels drain
                               anteriorly through the cricothyroid membrane to the middle and lower jugular or Delphian
                               nodes and posterolaterally to the paratracheal nodes. The true vocal cords are almost
                               devoid of lymphatics, leading to a very low incidence of nodal metastasis in early-stage (T1)
                               glottic cancers, approaching zero. This incidence increases with stage: 2% for T2, 15–20% for
                               T3, and 20–30% for T4 lesions. Occult nodal involvement is found in approximately 16% of
                               clinically node-negative T3 and T4 glottic cancers [229,230]. Glottic cancer spread typically
                               involves extension into the supraglottis or subglottis, leveraging their more prosperous
                               lymphatic supply. Distant metastasis is relatively uncommon in LC, occurring in 10–20% of
                               patients, predominantly those with supraglottic and subglottic primaries, although autopsy
                               studies reveal a higher rate of subclinical metastases. The lung is the most frequent site
                               of distant metastasis, followed by bone (20% of patients with distant disease) and liver
                               (10% clinically). Brain metastases are rare [229–231]. Because of the high risk of second
                               primary cancers in these patients, tissue confirmation of suspected metastases is essential.
                               Factors increasing the risk of distant metastasis include lymph node involvement, low
                               neck metastases, advanced stage, and extranodal extension (ENE), with ENE potentially
                               increasing the risk tenfold [19,232].
                               3.7. Treatment
                                    The treatment of LC is a multifaceted endeavor, with approaches tailored to the
                               cancer’s stage, location, and the patient’s overall condition. For early-stage (T1-T2, N0,
                               M0) LCs, treatments often aim to preserve the larynx. These can involve laryngeal-sparing
                               surgeries, which selectively remove cancerous tissue, or definitive radiotherapy (RT),
                               employing high-energy beams to eradicate cancer cells [236]. Table 4 summarizes the
                               treatment modalities of LC.
J. Clin. Med. 2025, 14, 3367                                                                                                                     14 of 32
                                         Currently, the standard treatment for locally advanced LC, when aiming for lar-
                                    ynx preservation, is concurrent chemoradiation therapy (CRT). This approach combines
                                    high-dose cisplatin chemotherapy with radiation therapy, delivered simultaneously. The
                                    effectiveness of this concurrent approach was established by the RTOG 91-11 trial, which
                                    demonstrated superior larynx preservation rates compared to induction chemotherapy fol-
                                    lowed by radiation or radiation alone [237]. Intensity-modulated radiation therapy (IMRT)
                                    is a widely used technique that allows for highly conformal treatment plans, minimizing
                                    radiation dose to sensitive structures like the spinal cord, oesophagus, and salivary glands,
                                    thereby reducing toxicities [238].
J. Clin. Med. 2025, 14, 3367                                                                                         15 of 32
                                     Beyond the standard CRT approach, several other therapies and considerations are
                               relevant in the treatment of LC. For early-stage disease, definitive radiotherapy alone or
                               laryngeal-sparing surgery may be sufficient [236].
                                     Alternative systemic therapies may be considered for patients who are not candidates
                               for cisplatin. Although cetuximab, an EGFR inhibitor, can enhance radiation’s effects, it
                               has shown inferior overall survival compared to cisplatin in certain head and neck can-
                               cers [239]. Thus, for platinum-ineligible cases, carboplatin and fluorouracil, or cetuximab
                               alone, might be considered based on improvements in survival compared to radiation
                               alone in clinical trials.
                                     Immunotherapy, specifically with checkpoint inhibitors like pembrolizumab and
                               nivolumab, is currently approved for recurrent or metastatic head and neck squamous
                               cell carcinoma (HNSCC) [240]. Pembrolizumab and nivolumab received FDA approval in
                               2016 [240,241]. In the pivotal CheckMate 141 Asian subset, nivolumab conferred a median
                               overall survival of 12.1 months vs. 6.2 months with investigator’s choice therapy, and the
                               estimated 2-year OS rates were 22.7% vs. 0%, respectively; notably, patients who devel-
                               oped any treatment-related adverse events particularly skin-related disorders experienced
                               superior survival, suggesting that early immune-related toxicity may predict benefit [242].
                               Multiple clinical trials are actively investigating the role of these agents in combination
                               with radiation therapy for locally advanced LC, exploring their potential as alternative
                               radiosensitizers [243,244]. For example, the multinational, Phase III, double-blind, placebo-
                               controlled JAVELIN Head and Neck 100 trial (NCT02952586) is assessing whether adding
                               the PD-L1 inhibitor avelumab to standard cisplatin-based chemoradiotherapy improves
                               progression-free and overall survival compared with chemoradiotherapy plus placebo in
                               patients with high-risk, nonmetastatic, locoregionally advanced HNSCC [245].
                                     Similarly, the Phase III, double-blind, placebo-controlled KEYNOTE-412 trial
                               (NCT03040999) evaluated the addition of pembrolizumab (200 mg q3w) to standard
                               cisplatin-based chemoradiotherapy (70 Gy in 35 fractions) in 804 patients with newly
                               diagnosed, high-risk, unresected, locoregionally advanced HNSCC. Pembrolizumab was
                               given one dose before CRT, two doses during CRT, and up to 14 maintenance doses there-
                               after. After a median follow-up of 47.7 months, median event-free survival was not reached
                               in the pembrolizumab arm versus 46.6 months in the placebo arm (hazard ratio 0.83 [95%
                               CI 0.68–1.03]; log-rank p = 0.043, above the prespecified threshold of p ≤ 0.024), and median
                               overall survival was not reached in either arm. Grade ≥3 adverse events occurred in
                               92% of pembrolizumab-treated versus 88% of placebo-treated patients, with similar safety
                               profiles and no new signals [246]. This trial highlights the challenge of integrating PD-1
                               inhibitors into definitive CRT for locally advanced HNSCC and underscores the need for
                               biomarker-driven strategies.
                                     Pembrolizumab, a highly selective humanized monoclonal antibody, is designed to
                               disrupt the PD-1 immune checkpoint pathway. It works by binding to the PD-1 receptor on
                               T cells, preventing it from interacting with its ligands, PD-L1 and PD-L2, which are often
                               expressed on tumour cells and other cells in the tumour microenvironment. This blockade
                               releases the “brake” on the immune system, allowing T cells to become activated and attack
                               cancer cells. In the context of recurrent or metastatic head and neck squamous cell carci-
                               noma (HNSCC), including LSCC, the Keynote 040 study investigated the effectiveness of
                               pembrolizumab against standard treatments. In this trial, 247 participants were randomly
                               assigned to receive either pembrolizumab or one of the following standard-of-care thera-
                               pies: methotrexate (40–60 mg/m2 ), docetaxel (30–40 mg/m2 ), or cetuximab (250 mg/m2
                               weekly after an initial 400 mg/m2 loading dose). The median overall survival for patients
                               treated with pembrolizumab was 8.4 months, compared to 6.9 months for those receiving
                               standard therapy. This difference represented a statistically significant improvement in
J. Clin. Med. 2025, 14, 3367                                                                                           16 of 32
                               survival for the pembrolizumab group, with a hazard ratio (HR) of 0.80 (95% CI 0.65–0.98;
                               p = 0.0161) [247]. The benefit of pembrolizumab was even more pronounced in patients
                               whose tumours had a high proportion of cells expressing PD-L1, as indicated by a tumour
                               proportion score (TPS) of 50% or greater. In this subgroup, the median overall survival was
                               11.6 months with pembrolizumab versus 6.6 months with standard care, demonstrating
                               a more substantial and statistically significant improvement (HR: 0.53, 95% CI 0.35–0.81;
                               p = 0.014) [247].
                                     CTLA-4 inhibitors, such as ipilimumab and tremelimumab, block the CTLA-4 immune
                               checkpoint on T cells preventing its engagement with CD 80/86—and thereby sustain T-cell
                               activation to enhance antitumor immunity. In CheckMate 651 (NCT02741570), 947 patients
                               with previously untreated R/M SCCHN were randomized to first-line nivolumab + ipili-
                               mumab versus the EXTREME regimen. Neither the overall population (median OS 13.9 vs.
                               13.5 months; HR 0.95; p = 0.4951) nor the PD-L1 CPS ≥ 20 subgroup (17.6 vs 14.6 months;
                               HR 0.78; p = 0.0469) met the primary OS endpoint, although nivolumab + ipilimumab had
                               fewer grade 3–4 treatment-related adverse events (28.2% vs. 70.7%) [248]. In the phase III
                               KESTREL trial (NCT02551159), neither tumor PD-L1 expression (TC ≥ 50%/IC ≥ 25% or
                               TC ≥ 25%) nor a low neutrophil-to-lymphocyte ratio (≤7) enriched for improved outcomes
                               with first-line durvalumab (D) or durvalumab + tremelimumab (D + T) versus EXTREME.
                               However, in patients whose blood tumor mutational burden was ≥16 mut/Mb, D + T
                               achieved a median OS hazard ratio of 0.69 (95% CI 0.39–1.25) and a complete response rate
                               of 8.6% versus 4.3% with EXTREME, suggesting bTMB may help identify those most likely
                               to benefit from checkpoint blockade [249].
                                     In the phase III KESTREL trial (NCT02551159), first-line durvalumab monother-
                               apy or durvalumab plus tremelimumab was compared with EXTREME in recur-
                               rent/metastatic HNSCC, and archival tumors or blood were assayed for PD-L1, blood
                               tumor mutational burden (bTMB), and neutrophil-to-lymphocyte ratio (NLR). PD-L1
                               expression (TC ≥ 50%/IC ≥ 25% or TC ≥ 25%) and NLR ≤ 7 failed to enrich for either
                               overall survival or response rates, whereas in the bTMB ≥ 16 mut/Mb subgroup, durval-
                               umab plus tremelimumab achieved an OS hazard ratio of 0.69 (95% CI 0.39–1.25) versus
                               EXTREME and doubled the complete response rate (8.6% vs. 4.3%), suggesting high
                               bTMB may identify patients most likely to benefit from PD-L1/CTLA-4 blockade.
                                     In the phase III CheckMate 651 trial, first-line nivolumab plus ipilimumab did not
                               improve overall survival compared with the EXTREME regimen in patients with recurrent
                               or metastatic SCCHN, yielding a median OS of 13.9 versus 13.5 months in the intent-to-
                               treat population (HR 0.95, p = 0.495) and 17.6 versus 14.6 months in those with PD-L1
                               CPS ≥ 20 (HR 0.78, p = 0.047), despite a substantially lower rate of grade 3–4 treatment-
                               related adverse events (28.2% vs. 70.7%). Among responders, the duration of response was
                               markedly longer with nivolumab/ipilimumab (32.6 vs. 7.0 months), but progression-free
                               survival and response rates were similar between arms.
                                     Proton therapy represents another evolving radiation modality. It offers the potential
                               for a more favourable therapeutic window compared to photons due to the Bragg Peak
                               phenomenon, where the majority of the radiation dose is deposited at a specific depth,
                               minimizing the dose beyond the target. However, access to proton therapy is limited,
                               and prospective, multi-institutional data comparing it to IMRT are still emerging [250].
                               Stereotactic body radiation therapy (SBRT), which delivers very high doses of radiation in a
                               few fractions, is being investigated for early-stage glottic cancers but is not yet established
                               for laryngeal preservation and should only be used within a clinical trial [251].
                                     While many treatments focus on larynx preservation, total laryngectomy (TL) remains
                               a crucial option, particularly for patients with extensive tumour invasion, significant
                               pre-existing swallowing dysfunction, or those who are not candidates for or have failed
J. Clin. Med. 2025, 14, 3367                                                                                            17 of 32
                               3.8. Survival
                                     The prognosis for patients with LC is strongly correlated with the disease stage at
                               initial diagnosis and treatment. Other factors that play a role in survival are the patient’s
                               general health and smoking cessation. In the United States, the overall 5-year survival
                               rate for LC is 61% [268]. Survival rates decline significantly with the advancing stage. For
                               diseases confined to the larynx (stages I and II), the 5-year survival rate is 78%. However,
                               regional lymph node metastases (stage III) reduce the 5-year survival to 46%, and distant
                               metastases lower it to 34% [269]. Survival outcomes also vary by laryngeal subsite. Glottic
                               cancer generally has the most favourable prognosis, followed by supraglottic cancer, with
                               subglottic tumours having the least favourable outlook [113]. Specifically, the overall 5-year
J. Clin. Med. 2025, 14, 3367                                                                                           18 of 32
                               survival for glottic SCC is 77%, reaching 84% for localized disease (stages I and II) but
                               decreasing to 52% with nodal involvement (stage III) and 45% with distant spread [270].
                               For supraglottic SCC, the overall 5-year survival is 45%, with rates of 61% for localized
                               disease, 46% for stage III, and 30% for metastatic disease [271,272]. Subglottic SCC has an
                               overall 5-year survival rate of 49%, 59% for stages I and II, 38% for stage III, and 44% for
                               metastatic disease [273,274].
                                     The accurate prediction of prognosis in LC is crucial for tailoring treatment strategies.
                               While the TNM staging system provides a foundation, it does not fully capture the het-
                               erogeneity of the disease, particularly regarding the impact of lymph node involvement.
                               Therefore, integrating both clinical factors and biomarkers into comprehensive prognostic
                               models is an area of active investigation. Recent efforts have focused on developing nomo-
                               grams which is a statistical tools that combine multiple variables to provide individualized
                               risk assessments. For instance, Juan et al., (2024) [275] developed and validated nomo-
                               grams specifically for early-stage LSCC to predict postoperative recurrence-free survival
                               (RFS) and overall survival (OS). Their models incorporated readily available preoperative
                               blood markers (platelet counts, fibrinogen, platelet-to-lymphocyte ratio, systemic immune-
                               inflammation index, and hemoglobin) alongside clinicopathological characteristics (tumor
                               diameter and differentiation degree). These nomograms demonstrated superior predictive
                               accuracy compared to T staging alone [275].
                                     Similarly, Shi et al., (2017) [276] developed nomograms for LSCC patients undergoing
                               neck dissection, incorporating the lymph node ratio (LNR)—a factor reflecting both the
                               extent of nodal disease and the thoroughness of the dissection—along with a compre-
                               hensive set of clinical and pathological factors, including T stage, N stage, tumor size
                               and demographics. Their models, validated both internally and externally, also showed
                               improved predictive performance for both OS and cancer-specific survival (CSS) compared
                               to models without LNR and to traditional TNM staging [276]. These studies highlight that
                               readily available clinical parameters and biomarkers can significantly enhance prognos-
                               tication in LSCC, there is a need for further investigations into developing new scoring
                               systems. Further research that include novel biomarkers discussed in the review may
                               improve current scoring system and therefore, be used by clinicians to decide the optimal
                               personalized treatment.
                                     Quality-of-life (QoL) outcomes are paramount in the treatment of LC, particularly
                               when comparing organ-preserving approaches versus TL. For early-stage (T1-T2) glottic
                               cancers, both TLM and RT aim for larynx preservation, but their impact on voice and QoL
                               can differ. A meta-analysis by Qasem et al., (2024) [277] found no significant difference
                               between TLM and RT in overall voice-related QoL measures, including the Voice Handicap
                               Index-30 (VHI-30). However, TLM was associated with significantly better outcomes
                               in specific acoustic parameters like jitter and shimmer, suggesting potentially improved
                               voice stability [277]. In more advanced T3 glottic cancers, TLM can still be an option,
                               and Chien et al., (2020) [278] showed that carefully selected T3 glottic SCC patients could
                               achieve satisfactory QoL and larynx preservation rates after CO2 TLM. Even after total
                               laryngectomy, where the larynx is removed, voice rehabilitation is crucial for QoL [278].
                               However, Wulff et al., (2020) [279], in a systematic review, found that individuals who
                               underwent TL generally reported worse health-related quality of life (HRQoL) compared
                               to a male normative reference population, though the reported symptom burden was often
                               mild. The heterogeneity and generally low quality of existing studies highlight a need
                               for more rigorous research in this area [279]. These studies, overall, emphasize a need for
                               individualized assessment when choosing a treatment and obtaining a high level of QoL.
                               Table 5 shows a summary of survival data provided within this section.
J. Clin. Med. 2025, 14, 3367                                                                                            19 of 32
                               4. Conclusions
                                    LC, a complex and heterogeneous disease, presents significant diagnosis, treatment,
                               and prognostication challenges. This review has highlighted the crucial role of accurate stag-
                               ing in guiding treatment decisions, incorporating both clinical and pathological findings.
                               The distinct histological variants of LSCC, each with unique characteristics and behaviors,
                               necessitate careful pathological evaluation. The expanding knowledge of biomarkers offers
                               the potential for improved risk stratification and personalized treatment approaches, al-
                               though further validation is needed for many of these markers. While treatment strategies
J. Clin. Med. 2025, 14, 3367                                                                                                           20 of 32
                                   have evolved, with a growing emphasis on organ preservation, the advanced-stage disease
                                   continues to pose a significant challenge.
                                         Recognizing that no single specialist can address every nuance of LC care, it is essential
                                   to integrate multidisciplinary team (MDT) decision-making throughout the patient journey.
                                   Regular MDT reviews, which bring together head and neck surgeons, radiation and medical
                                   oncologists, radiologists, pathologists, and allied health professionals, ensure that emerging
                                   biomarkers and novel therapies are interpreted within a comprehensive clinical context.
                                         Future research in LC must address several critical gaps to advance the field. A pri-
                                   mary focus should be the rigorous validation of promising biomarkers, moving beyond
                                   exploratory studies to establish their clinical utility in diverse patient populations and treat-
                                   ment settings. This includes standardizing assays and determining optimal cut-off values
                                   for clinical decision-making. Simultaneously, the development of novel targeted therapies
                                   is crucial, particularly those addressing specific molecular alterations driving tumor growth
                                   and resistance. Emphasis should also be on personalized medicine. This means creating
                                   strategies that are individualized and focus on the combination of specific tumoral factors
                                   and host factors. This personalized approach necessitates integrating comprehensive ge-
                                   nomic and proteomic profiling with detailed clinical data to tailor treatment selection and
                                   predict response to therapies. Finally, optimizing treatment strategies, including refining
                                   radiation techniques, exploring immunotherapy combinations, and developing strategies
                                   to overcome resistance, is essential to improve patient outcomes and quality of life.
                                   Author Contributions: Conceptualization, A.-R.H. and E.R.B.; methodology, A.-R.H.; software, D.P.;
                                   validation, G.I., D.H. and L.M.; formal analysis, G.I.; investigation, D.H. and N.C.B.; resources,
                                   R.G.; data curation, D.P.; writing—original draft preparation, A.-R.H.; writing—review and editing,
                                   A.C.M. and N.C.B.; visualization, M.C.; supervision, A.C.M.; project administration, E.R.B.; funding
                                   acquisition, E.R.B. All authors have read and agreed to the published version of the manuscript.
References
1.    Mody, M.D.; Rocco, J.W.; Yom, S.S.; Haddad, R.I.; Saba, N.F. Head and Neck Cancer. Lancet 2021, 398, 2289–2299. [CrossRef]
      [PubMed]
2.    Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global Cancer Statistics 2022: GLOBOCAN
      Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2024, 74, 229–263. [CrossRef]
      [PubMed]
3.    Ciolofan, M.S.; Vlăescu, A.N.; Mogoantă, C.-A.; Ionit, ă, E.; Ionit, ă, I.; Căpitănescu, A.-N.; Mitroi, M.-R.; Anghelina, F. Clinical,
      Histological and Immunohistochemical Evaluation of Larynx Cancer. Curr. Health Sci. J. 2017, 43, 367–375. [CrossRef]
4.    Ferlay, J.; Ervik, M.; Lam, F.; Laversanne, M.; Colombet, M.; Mery, L.; Piñeros, M.; Znaor, A.; Soerjomataram, I.; Bray, F.; et al. Lyon,
      France: International Agency for Research on Cancer. Available online: https://gco.iarc.who.int/media/globocan/factsheets/
      cancers/14-larynx-fact-sheet.pdf (accessed on 14 March 2025).
5.    Zuo, J.-J.; Tao, Z.-Z.; Chen, C.; Hu, Z.-W.; Xu, Y.-X.; Zheng, A.-Y.; Guo, Y. Characteristics of Cigarette Smoking without Alcohol
      Consumption and Laryngeal Cancer: Overall and Time-Risk Relation. A Meta-Analysis of Observational Studies. Eur. Arch.
      Oto-Rhino-Laryngol. 2017, 274, 1617–1631. [CrossRef] [PubMed]
6.    Kis, A.M.; Watz, C.G.; Motofelea, A.C.; Chiriac, S.; Poenaru, M.; Dehelean, C.A.; Borza, C.; Ionita, I. Challenges of Pharyngeal
      Cancer Screening in Lower-Income Countries during Economic and Social Transitions: A Population-Based Analysis. Eur. J.
      Investig. Health Psychol. Educ. 2023, 13, 2226–2237. [CrossRef]
7.    Tang, J.A.; Lango, M.N. Diverging Incidence Trends for Larynx and Tonsil Cancer in Low Socioeconomic Regions of the US. Oral.
      Oncol. 2019, 91, 65–68. [CrossRef]
J. Clin. Med. 2025, 14, 3367                                                                                                               21 of 32
8.    Liao, L.; Wang, H.; Cui, W.; Zhang, Q.; He, X.; Wang, L.; Xiong, Y.; Jiang, L.; Xie, Y. Global, Regional and National Burden and
      Trends of Larynx Cancer among Adults Aged 55 and Older from 1990 to 2021: Results from the Global Burden of Disease Study
      2021. BMC Public. Health 2025, 25, 906. [CrossRef]
9.    Wang, J.-Y.; Zhang, Q.-W.; Wen, K.; Wang, C.; Ji, X.; Zhang, L. Temporal Trends in Incidence and Mortality Rates of Laryngeal
      Cancer at the Global, Regional and National Levels, 1990–2017. BMJ Open 2021, 11, e050387. [CrossRef]
10.   Balogun, O.; Rodin, D.; Ngwa, W.; Grover, S.; Longo, J. Challenges and Prospects for Providing Radiation Oncology Services in
      Africa. Semin. Radiat. Oncol. 2017, 27, 184–188. [CrossRef]
11.   Chang, C.-P.; Chang, S.-C.; Chuang, S.-C.; Berthiller, J.; Ferro, G.; Matsuo, K.; Wünsch-Filho, V.; Toporcov, T.N.; de Carvalho, M.B.;
      La Vecchia, C. Age at Start of Using Tobacco on the Risk of Head and Neck Cancer: Pooled Analysis in the International Head
      and Neck Cancer Epidemiology Consortium (INHANCE). Cancer Epidemiol. 2019, 63, 101615. [CrossRef]
12.   Di Credico, G.; Edefonti, V.; Polesel, J.; Pauli, F.; Torelli, N.; Serraino, D.; Negri, E.; Luce, D.; Stucker, I.; Matsuo, K. Joint Effects of
      Intensity and Duration of Cigarette Smoking on the Risk of Head and Neck Cancer: A Bivariate Spline Model Approach. Oral.
      Oncol. 2019, 94, 47–57. [CrossRef] [PubMed]
13.   LoConte, N.K.; Brewster, A.M.; Kaur, J.S.; Merrill, J.K.; Alberg, A.J. Alcohol and Cancer: A Statement of the American Society of
      Clinical Oncology. J. Clin. Oncol. 2018, 36, 83–93. [CrossRef] [PubMed]
14.   Berdzuli, N.; Ferreira-Borges, C.; Gual, A.; Rehm, J. Alcohol Control Policy in Europe: Overview and Exemplary Countries. Int. J.
      Environ. Res. Public Health 2020, 17, 8162. [CrossRef] [PubMed]
15.   Bagnardi, V.; Rota, M.; Botteri, E.; Tramacere, I.; Islami, F.; Fedirko, V.; Scotti, L.; Jenab, M.; Turati, F.; Pasquali, E. Alcohol
      Consumption and Site-Specific Cancer Risk: A Comprehensive Dose–Response Meta-Analysis. Br. J. Cancer 2015, 112, 580–593.
      [CrossRef]
16.   He, J.; Meng, H.; Yao, G.; Fu, Y.; Geng, J. New Research Progress of HPV and Larynx Cancer. Progress. Mod. Biomed. 2017, 17,
      5989–5992.
17.   Peng, W.; Mi, J.; Jiang, Y. Asbestos Exposure and Laryngeal Cancer Mortality. Laryngoscope 2016, 126, 1169–1174. [CrossRef]
18.   Hall, A.L.; Kromhout, H.; Schüz, J.; Peters, S.; Portengen, L.; Vermeulen, R.; Agudo, A.; Ahrens, W.; Boffetta, P.; Brennan, P.
      Laryngeal Cancer Risks in Workers Exposed to Lung Carcinogens: Exposure–Effect Analyses Using a Quantitative Job Exposure
      Matrix. Epidemiology 2020, 31, 145–154. [CrossRef]
19.   Haddad, G.; Sataloff, R.T.; Hamdan, A.-L. Laryngeal Metastatic Lesions: A Literature Review. J. Voice 2024, 38, 1458–1464.
      [CrossRef]
20.   Santos, A.; Santos, I.C.; dos Reis, P.F.; Rodrigues, V.D.; Peres, W.A.F. Impact of Nutritional Status on Survival in Head and Neck
      Cancer Patients After Total Laryngectomy. Nutr. Cancer 2022, 74, 1252–1260. [CrossRef]
21.   Siegel, R.; Ma, J.; Zou, Z.; Jemal, A. Cancer Statistics, 2014. CA Cancer J. Clin. 2014, 64, 9–29. [CrossRef]
22.   Hoffmann, T.K. Systemic therapy strategies for head-neck carcinomas: Current status. Laryngo Rhino Otol. 2012, 91 (Suppl. 1),
      S23–S143. [CrossRef]
23.   Baird, B.J.; Sung, C.K.; Beadle, B.M.; Divi, V. Treatment of Early-Stage Laryngeal Cancer: A Comparison of Treatment Options.
      Oral. Oncol. 2018, 87, 8–16. [CrossRef]
24.   Zhang, Y.; Li, Y.; Fu, Q.; Han, Z.; Wang, D.; Umar Shinge, S.A.; Muluh, T.A.; Lu, X. Combined Immunotherapy and Targeted
      Therapies for Cancer Treatment: Recent Advances and Future Perspectives. Curr. Cancer Drug Targets 2023, 23, 251–264. [CrossRef]
      [PubMed]
25.   Bradford, C.R.; Ferlito, A.; Devaney, K.O.; Mäkitie, A.A.; Rinaldo, A. Prognostic Factors in Laryngeal Squamous Cell Carcinoma.
      Laryngoscope Investig. Otolaryngol. 2020, 5, 74–81. [CrossRef] [PubMed]
26.   Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global Cancer Statistics 2018: GLOBOCAN Estimates of
      Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2018, 68, 394–424. [CrossRef]
27.   Lortet-Tieulent, J.; Sauer, A.G.; Siegel, R.L.; Miller, K.D.; Islami, F.; Fedewa, S.A.; Jacobs, E.J.; Jemal, A. State-Level Cancer
      Mortality Attributable to Cigarette Smoking in the United States. JAMA Intern. Med. 2016, 176, 1792–1798. [CrossRef]
28.   Permitasari, A.L.; Satibi, S.; Kristina, S.A. National Burden of Cancers Attributable to Secondhand Smoking in Indonesia. Asian
      Pac. J. Cancer Prev. 2018, 19, 1951–1955.
29.   Allegra, E.; Bianco, M.R.; Ralli, M.; Greco, A.; Angeletti, D.; De Vincentiis, M. Role of Clinical-Demographic Data in Survival
      Rates of Advanced Laryngeal Cancer. Medicina 2021, 57, 267. [CrossRef]
30.   Voltzke, K.J.; Lee, Y.-C.A.; Zhang, Z.-F.; Zevallos, J.P.; Yu, G.-P.; Winn, D.M.; Vaughan, T.L.; Sturgis, E.M.; Smith, E.; Schwartz, S.M.
      Racial Differences in the Relationship between Tobacco, Alcohol, and the Risk of Head and Neck Cancer: Pooled Analysis of US
      Studies in the INHANCE Consortium. Cancer Causes Control 2018, 29, 619–630. [CrossRef]
31.   Manthey, J.; Shield, K.D.; Rylett, M.; Hasan, O.S.M.; Probst, C.; Rehm, J. Global Alcohol Exposure between 1990 and 2017 and
      Forecasts until 2030: A Modelling Study. Lancet 2019, 393, 2493–2502. [CrossRef]
J. Clin. Med. 2025, 14, 3367                                                                                                             22 of 32
32.   Grevers, X.; Ruan, Y.; Poirier, A.E.; Walter, S.D.; Villeneuve, P.J.; Friedenreich, C.M.; Brenner, D.R.; Team, C.S. Estimates of the
      Current and Future Burden of Cancer Attributable to Alcohol Consumption in Canada. Prev. Med. 2019, 122, 40–48. [CrossRef]
      [PubMed]
33.   Szukalska, M.; Szyfter, K.; Florek, E.; Rodrigo, J.P.; Rinaldo, A.; Mäkitie, A.A.; Strojan, P.; Takes, R.P.; Suárez, C.; Saba, N.F.; et al.
      Electronic Cigarettes and Head and Neck Cancer Risk-Current State of Art. Cancers 2020, 12, 3274. [CrossRef] [PubMed]
34.   Lim, H.-H.; Shin, H.-S. Measurement of Aldehydes in Replacement Liquids of Electronic Cigarettes by Headspace Gas
      Chromatography-Mass Spectrometry. Bull. Korean Chem. Soc. 2013, 34, 2691–2696. [CrossRef]
35.   IARC Working Group on the Evaluation of Carcinogenic Risks to Humans; World Health Organization; International Agency for
      Research on Cancer. Tobacco Smoke and Involuntary Smoking; IARC: Lyon, France, 2004; ISBN 9283212835.
36.   Jethwa, A.R.; Khariwala, S.S. Tobacco-Related Carcinogenesis in Head and Neck Cancer. Cancer Metastasis Rev. 2017, 36, 411–423.
      [CrossRef]
37.   Khariwala, S.S.; Carmella, S.G.; Stepanov, I.; Fernandes, P.; Lassig, A.A.; Yueh, B.; Hatsukami, D.; Hecht, S.S. Elevated Levels of
      1-hydroxypyrene and N′ -nitrosonornicotine in Smokers with Head and Neck Cancer: A Matched Control Study. Head Neck 2013,
      35, 1096–1100. [CrossRef]
38.   Salturk, Z.; Çakır, Ç.; Sünnetçi, G.; Atar, Y.; Kumral, T.L.; Yıldırım, G.; Berkiten, G.; Uyar, Y. Effects of Electronic Nicotine Delivery
      System on Larynx: Experimental Study. J. Voice 2015, 29, 560–563. [CrossRef]
39.   Sheikh, M.; Shakeri, R.; Poustchi, H.; Pourshams, A.; Etemadi, A.; Islami, F.; Khoshnia, M.; Gharavi, A.; Roshandel, G.; Khademi,
      H. Opium Use and Subsequent Incidence of Cancer: Results from the Golestan Cohort Study. Lancet Glob. Health 2020, 8,
      e649–e660. [CrossRef]
40.   Mohebbi, E.; Hadji, M.; Rashidian, H.; Rezaianzadeh, A.; Marzban, M.; Haghdoost, A.A.; Naghibzadeh Tahami, A.; Moradi,
      A.; Gholipour, M.; Najafi, F. Opium Use and the Risk of Head and Neck Squamous Cell Carcinoma. Int. J. Cancer 2021, 148,
      1066–1076. [CrossRef]
41.   Tsimplaki, E.; Argyri, E.; Sakellaridis, A.; Kyrodimos, E.; Xesfyngi, D.; Panotopoulou, E. Oropharyngeal and Laryngeal but
      Not Oral Cancers Are Strongly Associated with High-risk Human Papillomavirus in 172 Greek Patients. J. Med. Virol. 2017, 89,
      170–176. [CrossRef]
42.   Sabatini, M.E.; Chiocca, S. Human Papillomavirus as a Driver of Head and Neck Cancers. Br. J. Cancer 2020, 122, 306–314.
      [CrossRef]
43.   Nahet, A.; Boublenza, L.; Hassaine, H.; Masdoua, N.; Pretet, J.-L.; Belglaiaa, E.; Mougin, C. HPV DNA Genotyping: A Study of
      Anogenital, Head and Neck and Skin Cancers in a Population from West Algerian. HPV Detection in Different Cancers from an
      Algerian Population. Bull. Cancer 2016, 103, 455–460. [CrossRef] [PubMed]
44.   Kariche, N.; Hortal, M.T.; Benyahia, S.; Alemany, L.; Moulaï, N.; Clavero, O.; Muñoz, M.; Ouahioune, W.; Djennaoui, D.; Touil-
      Boukoffa, C. Comparative Assessment of HPV, Alcohol and Tobacco Etiological Fractions in Algerian Patients with Laryngeal
      Squamous Cell Carcinoma. Infect. Agents Cancer 2018, 13, 1–8. [CrossRef] [PubMed]
45.   Chen, W.-C.; Chuang, H.-C.; Lin, Y.-T.; Huang, C.-C.; Chien, C.-Y. Clinical Impact of Human Papillomavirus in Laryngeal
      Squamous Cell Carcinoma: A Retrospective Study. PeerJ 2017, 5, e3395. [CrossRef]
46.   De Lima, M.A.P.; Silva, Á.D.L.; do Nascimento Filho, A.C.S.; Cordeiro, T.L.; Bezerra, J.P.d.S.; Rocha, M.A.B.; Pinheiro, S.d.F.L.;
      Pinheiro, R.F.F., Jr.; Gadelha, M.d.S.V.; da Silva, C.G.L. Epstein-Barr Virus-Associated Carcinoma of the Larynx: A Systematic
      Review with Meta-Analysis. Pathogens 2021, 10, 1429. [CrossRef]
47.   Vazquez-Guillen, J.M.; Palacios-Saucedo, G.C.; Alanis-Valdez, A.Y.; Huerta-Escobedo, A.; Zavala-Pompa, A.; Rivera-Morales, L.G.;
      Martinez-Torres, A.C.; Gonzalez-Villasana, V.; Serna-Hernandez, J.C.; Hernandez-Martinez, S.J. p16INK4a and pRb Expression in
      Laryngeal Squamous Cell Carcinoma with and without Infection by EBV or Different Genotypes of HPV: A Retrospective Study.
      Infect. Agents Cancer 2023, 18, 43. [CrossRef]
48.   Igissin, N.; Zatonskikh, V.; Telmanova, Z.; Tulebaev, R.; Moore, M. Laryngeal Cancer: Epidemiology, Etiology, and Prevention: A
      Narrative Review. Iran. J. Public Health 2023, 52, 2248–2259. [CrossRef]
49.   Wu, Y.-H.; Yen, C.-J.; Hsiao, J.-R.; Ou, C.-Y.; Huang, J.-S.; Wong, T.-Y.; Tsai, S.-T.; Huang, C.-C.; Lee, W.-T.; Chen, K.-C. A
      Comprehensive Analysis on the Association between Tobacco-Free Betel Quid and Risk of Head and Neck Cancer in Taiwanese
      Men. PLoS ONE 2016, 11, e0164937. [CrossRef]
50.   Chang, C.; Siwakoti, B.; Sapkota, A.; Gautam, D.K.; Lee, Y.A.; Monroe, M.; Hashibe, M. Tobacco Smoking, Chewing Habits,
      Alcohol Drinking and the Risk of Head and Neck Cancer in Nepal. Int. J. Cancer 2020, 147, 866–875. [CrossRef]
51.   Zhou, J.; Zhang, D.; Yang, Y.; Zhou, L.; Tao, L. Association between Helicobacter Pylori Infection and Carcinoma of the Larynx or
      Pharynx. Head Neck 2016, 38, E2291–E2296. [CrossRef]
52.   Eells, A.C.; Mackintosh, C.; Marks, L.; Marino, M.J. Gastroesophageal Reflux Disease and Head and Neck Cancers: A Systematic
      Review and Meta-Analysis. Am. J. Otolaryngol. 2020, 41, 102653. [CrossRef]
53.   Parsel, S.M.; Wu, E.L.; Riley, C.A.; McCoul, E.D. Gastroesophageal and Laryngopharyngeal Reflux Associated with Laryngeal
      Malignancy: A Systematic Review and Meta-Analysis. Clin. Gastroenterol. Hepatol. 2019, 17, 1253–1264. [CrossRef] [PubMed]
J. Clin. Med. 2025, 14, 3367                                                                                                             23 of 32
54.   Sereg-Bahar, M.; Jerin, A.; Hocevar-Boltezar, I. Higher Levels of Total Pepsin and Bile Acids in the Saliva as a Possible Risk Factor
      for Early Laryngeal Cancer. Radiol. Oncol. 2015, 49, 59–64. [CrossRef] [PubMed]
55.   Kim, S.-Y.; Han, K.; Joo, Y.-H. Metabolic Syndrome and Incidence of Laryngeal Cancer: A Nationwide Cohort Study. Sci. Rep.
      2019, 9, 667. [CrossRef]
56.   Kim, H.-B.; Kim, G.-J.; Han, K.; Joo, Y.-H. Changes in Metabolic Syndrome Status and Risk of Laryngeal Cancer: A Nationwide
      Cohort Study. PLoS ONE 2021, 16, e0252872. [CrossRef]
57.   Gong, H.-L.; Shi, Y.; Zhou, L.; Wu, C.-P.; Cao, P.-Y.; Tao, L.; Xu, C.; Hou, D.-S.; Wang, Y.-Z. The Composition of Microbiome in
      Larynx and the Throat Biodiversity between Laryngeal Squamous Cell Carcinoma Patients and Control Population. PLoS ONE
      2013, 8, e66476. [CrossRef]
58.   Yu, S.; Chen, J.; Zhao, Y.; Yan, F.; Fan, Y.; Xia, X.; Shan, G.; Zhang, P.; Chen, X. Oral-Microbiome-Derived Signatures Enable
      Non-Invasive Diagnosis of Laryngeal Cancers. J. Transl. Med. 2023, 21, 438. [CrossRef]
59.   Li, Y.; Tan, X.; Zhao, X.; Xu, Z.; Dai, W.; Duan, W.; Huang, S.; Zhang, E.; Liu, J.; Zhang, S.; et al. Composition and Function of Oral
      Microbiota between Gingival Squamous Cell Carcinoma and Periodontitis. Oral. Oncol. 2020, 107, 104710. [CrossRef]
60.   Kau, A.L.; Ahern, P.P.; Griffin, N.W.; Goodman, A.L.; Gordon, J.I. Human Nutrition, the Gut Microbiome and the Immune System.
      Nature 2011, 474, 327–336. [CrossRef]
61.   Kelly, D.; Mulder, I.E. Microbiome and Immunological Interactions. Nutr. Rev. 2012, 70, S18–S30. [CrossRef]
62.   Arthur, J.C.; Perez-Chanona, E.; Mühlbauer, M.; Tomkovich, S.; Uronis, J.M.; Fan, T.-J.; Campbell, B.J.; Abujamel, T.; Dogan,
      B.; Rogers, A.B.; et al. Intestinal Inflammation Targets Cancer-Inducing Activity of the Microbiota. Science 2012, 338, 120–123.
      [CrossRef]
63.   Hayes, R.B.; Ahn, J.; Fan, X.; Peters, B.A.; Ma, Y.; Yang, L.; Agalliu, I.; Burk, R.D.; Ganly, I.; Purdue, M.P.; et al. Association of Oral
      Microbiome With Risk for Incident Head and Neck Squamous Cell Cancer. JAMA Oncol. 2018, 4, 358–365. [CrossRef] [PubMed]
64.   McMaughan, D.J.; Oloruntoba, O.; Smith, M.L. Socioeconomic Status and Access to Healthcare: Interrelated Drivers for Healthy
      Aging. Front. Public Health 2020, 8, 231. [CrossRef] [PubMed]
65.   Fullmer, T.; Wilde, D.C.; Shi, J.W.; Wagner, T.; Skinner, H.; Eicher, S.A.; Sandulache, V.C.; Hernandez, D.J. Demographic and
      Tumor Characteristic Impact on Laryngeal Cancer Outcomes in a Minority Underserved Patient Population. Otolaryngol. Head
      Neck Surg. 2020, 162, 888–896. [CrossRef]
66.   Lu, Y.; Li, P.; Luo, G.; Liu, D.; Zou, H. Cancer Attributable to Human Papillomavirus Infection in China: Burden and Trends.
      Cancer 2020, 126, 3719–3732. [CrossRef]
67.   Buttmann-Schweiger, N.; Deleré, Y.; Klug, S.J.; Kraywinkel, K. Cancer Incidence in Germany Attributable to Human Papillo-
      mavirus in 2013. BMC Cancer 2017, 17, 1–8. [CrossRef]
68.   Senkomago, V. Human Papillomavirus–Attributable Cancers—United States, 2012–2016. MMWR. Morb. Mortal. Wkly. Rep. 2019,
      68, 724–728. [CrossRef]
69.   Wang, N.; Lv, H.; Huang, M. Impact of Gender on Survival in Patients with Laryngeal Squamous Cell Carcinoma: A Propensity
      Score Matching Analysis. Int. J. Clin. Exp. Pathol. 2020, 13, 573–581.
70.   Hashim, D.; Genden, E.; Posner, M.; Hashibe, M.; Boffetta, P. Head and Neck Cancer Prevention: From Primary Prevention to
      Impact of Clinicians on Reducing Burden. Ann. Oncol. 2019, 30, 744–756. [CrossRef]
71.   Andersson, T.M.L.; Engholm, G.; Pukkala, E.; Stenbeck, M.; Tryggvadottir, L.; Storm, H.; Weiderpass, E. Avoidable Cancers in the
      Nordic Countries—The Impact of Alcohol Consumption. Eur. J. Cancer 2018, 103, 299–307. [CrossRef]
72.   Zhou, T.; Wang, X.; Zhu, Q.; Zhou, E.; Zhang, J.; Song, F.; Xu, C.; Shen, Y.; Zou, J.; Zhu, H.; et al. Global Trends and Risk Factors
      of Laryngeal Cancer: A Systematic Analysis for the Global Burden of Disease Study (1990–2021). BMC Cancer 2025, 25, 296.
      [CrossRef]
73.   Liberale, C.; Soloperto, D.; Marchioni, A.; Monzani, D.; Sacchetto, L. Updates on Larynx Cancer: Risk Factors and Oncogenesis.
      Int. J. Mol. Sci. 2023, 24, 12913. [CrossRef] [PubMed]
74.   Dutta, R.; Husain, Q.; Kam, D.; Dubal, P.M.; Baredes, S.; Eloy, J.A. Laryngeal Papillary Squamous Cell Carcinoma. Otolaryngol.
      Head Neck Surg. 2015, 153, 54–59. [CrossRef] [PubMed]
75.   MacNeil, S.D. Non-Squamous Laryngeal Cancer. Otolaryngol. Clin. N. Am. 2023, 56, 345–359. [CrossRef] [PubMed]
76.   Dubal, P.M.; Marchiano, E.; Kam, D.; Dutta, R.; Kalyoussef, E.; Baredes, S.; Eloy, J.A. Laryngeal Spindle Cell Carcinoma: A
      Population-Based Analysis of Incidence and Survival. Laryngoscope 2015, 125, 2709–2714. [CrossRef]
77.   Gerry, D.; Fritsch, V.A.; Lentsch, E.J. Spindle Cell Carcinoma of the Upper Aerodigestive Tract. Ann. Otol. Rhinol. Laryngol. 2014,
      123, 576–583. [CrossRef]
78.   Fritsch, V.A.; Lentsch, E.J. Basaloid Squamous Cell Carcinoma of the Larynx: Analysis of 145 Cases with Comparison to
      Conventional Squamous Cell Carcinoma. Head Neck 2013, 36, 164–170. [CrossRef]
79.   Kass, J.I.; Lee, S.C.; Abberbock, S.; Seethala, R.R.; Duvvuri, U. Adenosquamous Carcinoma of the Head and Neck: Molecular
      Analysis Using CRTC-MAML FISH and Survival Comparison with Paired Conventional Squamous Cell Carcinoma. Laryngoscope
      2015, 125, E371–E376. [CrossRef]
J. Clin. Med. 2025, 14, 3367                                                                                                             24 of 32
80.  Kusafuka, K.; Muramatsu, K.; Iida, Y.; Mori, K.; Miki, T.; Suda, T.; Fuke, T.; Kamijo, T.; Onitsuka, T.; Nakajima, T. MUC Expression
     in Adenosquamous Carcinoma of the Head and Neck Regions of Japanese Patients: Immunohistochemical Analysis. Pathol. Int.
     2014, 64, 104–114. [CrossRef]
81. Schick, U.; Pusztaszeri, M.; Betz, M.; Ghadjar, P.; Demiroz, C.; Kaanders, J.H.A.M.; Ozsahin, M. Adenosquamous Carcinoma of
     the Head and Neck: Report of 20 Cases and Review of the Literature. Oral. Surg. Oral. Med. Oral. Pathol. Oral. Radiol. 2013, 116,
     313–320. [CrossRef]
82. Jones, T.M.; De, M.; Foran, B.; Harrington, K.; Mortimore, S. Laryngeal Cancer: United Kingdom National Multidisciplinary
     Guidelines. J. Laryngol. Otol. 2016, 130, S75–S82. [CrossRef]
83. El-Demerdash, A.; Fawaz, S.A.; Sabri, S.M.; Sweed, A.; Rabie, H. Sensitivity and Specificity of Stroboscopy in Preoperative
     Differentiation of Dysplasia from Early Invasive Glottic Carcinoma. Eur. Arch. Oto-Rhino-Laryngol. 2015, 272, 1189–1193.
     [CrossRef] [PubMed]
84. Gupta, T.; Master, Z.; Kannan, S.; Agarwal, J.P.; Ghsoh-Laskar, S.; Rangarajan, V.; Murthy, V.; Budrukkar, A. Diagnostic
     Performance of Post-Treatment FDG PET or FDG PET/CT Imaging in Head and Neck Cancer: A Systematic Review and
     Meta-Analysis. Eur. J. Nucl. Med. Mol. Imaging 2011, 38, 2083–2095. [CrossRef] [PubMed]
85. Sarkar, A.; Sharma, N.; Raghavan, A. Role of Computed Tomography, Magnetic Resonance Imaging, and Endoscopy in Prether-
     apeutic Evaluation of Laryngeal Tumors. Astrocyte 2016, 2, 172. Available online: https://go.gale.com/ps/i.do?id=GALE%
     7CA466753779&sid=googleScholar&v=2.1&it=r&linkaccess=abs&issn=23490977&p=AONE&sw=w&userGroupName=anon~
     c07fdcb8&aty=open-web-entry (accessed on 14 March 2025). [CrossRef]
86. De Vito, A.; Meccariello, G.; Vicini, C. Narrow Band Imaging as Screening Test for Early Detection of Laryngeal Cancer: A
     Prospective Study. Clin. Otolaryngol. 2016, 42, 347–353. [CrossRef] [PubMed]
87. Siravegna, G.; Marsoni, S.; Siena, S.; Bardelli, A. Integrating Liquid Biopsies into the Management of Cancer. Nat. Rev. Clin. Oncol.
     2017, 14, 531–548. [CrossRef]
88. Arantes, L.M.R.B.; De Carvalho, A.C.; Melendez, M.E.; Lopes Carvalho, A. Serum, Plasma and Saliva Biomarkers for Head and
     Neck Cancer. Expert. Rev. Mol. Diagn. 2018, 18, 85–112. [CrossRef]
89. Cohen, J.D.; Li, L.; Wang, Y.; Thoburn, C.; Afsari, B.; Danilova, L.; Douville, C.; Javed, A.A.; Wong, F.; Mattox, A.; et al. Detection
     and Localization of Surgically Resectable Cancers with a Multi-Analyte Blood Test. Science 2018, 359, 926–930. [CrossRef]
90. Cohen, J.D.; Javed, A.A.; Thoburn, C.; Wong, F.; Tie, J.; Gibbs, P.; Schmidt, C.M.; Yip-Schneider, M.T.; Allen, P.J.; Schattner, M.;
     et al. Combined Circulating Tumor DNA and Protein Biomarker-Based Liquid Biopsy for the Earlier Detection of Pancreatic
     Cancers. Proc. Natl. Acad. Sci. USA 2017, 114, 10202–10207. [CrossRef]
91. Russano, M.; Napolitano, A.; Ribelli, G.; Iuliani, M.; Simonetti, S.; Citarella, F.; Pantano, F.; Dell’Aquila, E.; Anesi, C.; Silvestris, N.;
     et al. Correction to: Liquid Biopsy and Tumor Heterogeneity in Metastatic Solid Tumors: The Potentiality of Blood Samples. J.
     Exp. Clin. Cancer Res. 2020, 39, 120. [CrossRef]
92. Guthrie, G.J.K.; Charles, K.A.; Roxburgh, C.S.D.; Horgan, P.G.; McMillan, D.C.; Clarke, S.J. The Systemic Inflammation-Based
     Neutrophil–Lymphocyte Ratio: Experience in Patients with Cancer. Crit. Rev. Oncol./Hematol. 2013, 88, 218–230. [CrossRef]
93. Iacob, R.; Mandea, M.; Iacob, S.; Pietrosanu, C.; Paul, D.; Hainarosie, R.; Gheorghe, C. Liquid Biopsy in Squamous Cell Carcinoma
     of the Esophagus and of the Head and Neck. Front. Med. 2022, 9, 827297. [CrossRef] [PubMed]
94. Yang, W.-Y.; Feng, L.-F.; Meng, X.; Chen, R.; Xu, W.-H.; Hou, J.; Xu, T.; Zhang, L. Liquid Biopsy in Head and Neck Squamous
     Cell Carcinoma: Circulating Tumor Cells, Circulating Tumor DNA, and Exosomes. Expert. Rev. Mol. Diagn. 2020, 20, 1213–1227.
     [CrossRef] [PubMed]
95. Payne, K.; Spruce, R.; Beggs, A.; Sharma, N.; Kong, A.; Martin, T.; Parmar, S.; Praveen, P.; Nankivell, P.; Mehanna, H. Circulating
     Tumor DNA as a Biomarker and Liquid Biopsy in Head and Neck Squamous Cell Carcinoma. Head Neck 2018, 40, 1598–1604.
     [CrossRef] [PubMed]
96. Vrba, L.; Oshiro, M.M.; Kim, S.S.; Garland, L.L.; Placencia, C.; Mahadevan, D.; Nelson, M.A.; Futscher, B.W. DNA Methylation
     Biomarkers Discovered in Silico Detect Cancer in Liquid Biopsies from Non-Small Cell Lung Cancer Patients. Epigenetics 2020, 15,
     419–430. [CrossRef]
97. Ooki, A.; Maleki, Z.; Tsay, J.-C.J.; Goparaju, C.; Brait, M.; Turaga, N.; Nam, H.-S.; Rom, W.N.; Pass, H.I.; Sidransky, D. A Panel
     of Novel Detection and Prognostic Methylated DNA Markers in Primary Non–Small Cell Lung Cancer and Serum DNA. Clin.
     Cancer Res. 2017, 23, 7141–7152. [CrossRef]
98. Schröck, A.; Leisse, A.; de Vos, L.; Gevensleben, H.; Dröge, F.; Franzen, A.; Wachendörfer, M.; Schröck, F.; Ellinger, J.; Teschke, M.
     Free-Circulating Methylated DNA in Blood for Diagnosis, Staging, Prognosis, and Monitoring of Head and Neck Squamous Cell
     Carcinoma Patients: An Observational Prospective Cohort Study. Clin. Chem. 2017, 63, 1288–1296. [CrossRef]
99. Sanchez-Cespedes, M.; Esteller, M.; Wu, L.; Nawroz-Danish, H.; Yoo, G.H.; Koch, W.M.; Jen, J.; Herman, J.G.; Sidransky, D. Gene
     Promoter Hypermethylation in Tumors and Serum of Head and Neck Cancer Patients. Cancer Res. 2000, 60, 892–895.
100. Kawada, T.; Takahashi, H.; Sakakura, K.; Ida, S.; Mito, I.; Toyoda, M.; Chikamatsu, K. Circulating Tumor Cells in Patients with
     Head and Neck Squamous Cell Carcinoma: Feasibility of Detection and Quantitation. Head Neck 2017, 39, 2180–2186. [CrossRef]
J. Clin. Med. 2025, 14, 3367                                                                                                        25 of 32
101. Nichols, A.C.; Lowes, L.E.; Szeto, C.C.T.; Basmaji, J.; Dhaliwal, S.; Chapeskie, C.; Todorovic, B.; Read, N.; Venkatesan, V.;
     Hammond, A.; et al. Detection of Circulating Tumor Cells in Advanced Head and Neck Cancer Using the Cellsearch System.
     Head Neck 2011, 34, 1440–1444. [CrossRef]
102. Rizzo, M.I.; Ralli, M.; Nicolazzo, C.; Gradilone, A.; Carletti, R.; Di Gioia, C.; De Vincentiis, M.; Greco, A. Detection of Circulating
     Tumor Cells in Patients with Laryngeal Cancer Using ScreenCell: Comparative Pre- and Post-Operative Analysis and Association
     with Prognosis. Oncol. Lett. 2020, 19, 4183–4188. [CrossRef]
103. Powrózek, T.; Mlak, R.; Brzozowska, A.; Mazurek, M.; Goł˛ebiowski, P.; Małecka-Massalska, T. miRNA-130a Significantly Improves
     Accuracy of SGA Nutritional Assessment Tool in Prediction of Malnutrition and Cachexia in Radiotherapy-Treated Head and
     Neck Cancer Patients. Cancers 2018, 10, 294. [CrossRef] [PubMed]
104. Cao, Y.-C.; Song, L.-Q.; Xu, W.-W.; Qi, J.-J.; Wang, X.-Y.; Su, Y. Serum miR-632 Is a Potential Marker for the Diagnosis and
     Prognosis in Laryngeal Squamous Cell Carcinoma. Acta Oto-Laryngol. 2020, 140, 418–421. [CrossRef] [PubMed]
105. Wang, J.L.; Wang, X.; Yang, D.; Shi, W.J. The Expression of MicroRNA-155 in Plasma and Tissue Is Matched in Human Laryngeal
     Squamous Cell Carcinoma. Yonsei Med. J. 2016, 57, 298–305. [CrossRef]
106. Wang, J.; Zhou, Y.; Lu, J.; Sun, Y.; Xiao, H.; Liu, M.; Tian, L. Combined Detection of Serum Exosomal miR-21 and HOTAIR as
     Diagnostic and Prognostic Biomarkers for Laryngeal Squamous Cell Carcinoma. Med. Oncol. 2014, 31, 148. [CrossRef]
107. Cabanero, M.; Tsao, M.S. Circulating Tumour DNA in EGFR-Mutant Non-Small-Cell Lung Cancer. Curr. Oncol. 2018, 25, 38–44.
     [CrossRef]
108. Mahmood, H.; Shaban, M.; Rajpoot, N.; Khurram, S.A. Artificial Intelligence-Based Methods in Head and Neck Cancer Diagnosis:
     An Overview. Br. J. Cancer 2021, 124, 1934–1940. [CrossRef]
109. Zhang, L.; Wu, Y.; Zheng, B.; Su, L.; Chen, Y.; Ma, S.; Hu, Q.; Zou, X.; Yao, L.; Yang, Y.; et al. Rapid Histology of Laryngeal
     Squamous Cell Carcinoma with Deep-Learning Based Stimulated Raman Scattering Microscopy. Theranostics 2019, 9, 2541–2554.
     [CrossRef]
110. Moccia, S.; De Momi, E.; Guarnaschelli, M.; Savazzi, M.; Laborai, A.; Guastini, L.; Peretti, G.; Mattos, L.S. Confident Texture-Based
     Laryngeal Tissue Classification for Early Stage Diagnosis Support. J. Med. Imaging (Bellingham Wash.) 2017, 4, 34502. [CrossRef]
111. Choi, N.; Kim, J.; Yi, H.; Kim, H.; Kim, T.H.; Chung, M.J.; Ji, M.; Kim, Z.; Son, Y.-I. The Use of Artificial Intelligence Models to
     Predict Survival in Patients with Laryngeal Squamous Cell Carcinoma. Sci. Rep. 2023, 13, 9734. [CrossRef]
112. Brandstorp-Boesen, J.; Sørum Falk, R.; Boysen, M.; Brøndbo, K. Impact of Stage, Management and Recurrence on Survival Rates
     in Laryngeal Cancer. PLoS ONE 2017, 12, e0179371. [CrossRef]
113. Mulcahy, C.F.; Mohamed, A.S.R.; Kanwar, A.; Hutcheson, K.A.; Ghosh, A.; Vock, D.; Weber, R.S.; Lai, S.Y.; Gunn, G.B. Age-adjusted
     Comorbidity and Survival in Locally Advanced Laryngeal Cancer. Head Neck 2018, 40, 2060–2069. [PubMed]
114. Egelmeer, A.G.T.M.; Velazquez, E.R.; de Jong, J.M.A.; Oberije, C.; Geussens, Y.; Nuyts, S.; Kremer, B.; Rietveld, D.; René Leemans,
     C.; de Jong, M.C.; et al. Development and Validation of a Nomogram for Prediction of Survival and Local Control in Laryngeal
     Carcinoma Patients Treated with Radiotherapy Alone: A Cohort Study Based on 994 Patients. Radiother. Oncol. 2011, 100, 108–115.
     [CrossRef] [PubMed]
115. Datema, F.R.; Ferrier, M.B.; Vergouwe, Y.; Moya, A.; Molenaar, J.; Piccirillo, J.F.; Baatenburg de Jong, R.J. Update and External
     Validation of a Head and Neck Cancer Prognostic Model. Head Neck 2012, 35, 1232–1237. [CrossRef] [PubMed]
116. Cavaliere, M.; Bisogno, A.; Scarpa, A.; D’Urso, A.; Marra, P.; Colacurcio, V.; De Luca, P.; Ralli, M.; Cassandro, E.; Cassandro, C.
     Biomarkers of Laryngeal Squamous Cell Carcinoma: A Review. Ann. Diagn. Pathol. 2021, 54, 151787. [CrossRef]
117. Gioacchini, F.M.; Alicandri-Ciufelli, M.; Rubini, C.; Magliulo, G.; Re, M. Prognostic Value of Bcl-2 Expression in Squamous Cell
     Carcinoma of the Larynx: A Systematic Review. Int. J. Biol. Markers 2015, 30, 155–160. [CrossRef]
118. Rashad, U.M.; Hussein, M.R.; Algizawy, S.M. Alterations of P53 and Bcl-2 Protein Expression in the Recurrent Laryngeal and
     Pharyngeal Squamous Cell Carcinoma. Am. J. Otolaryngol. 2011, 32, 210–214. [CrossRef]
119. Yildirim, S.; Cermik, H.; Işitmangil, T.; Baloglu, H.; Gungor, A.; Pekkafali, Z. Significance of P53 and Bcl-2 Immunoexpression in
     the Prognosis of Laryngeal Squamous Cell Carcinoma. J. Int. Med. Res. 2002, 30, 597–600. [CrossRef]
120. Ozdek, A.; Sarac, S.; Akyol, M.U.; Sungur, A.; Yilmaz, T. C-Myc and Bcl-2 Expression In Supraglottic Squamous Cell Carcinoma
     of the Larynx. Otolaryngol. Head Neck Surg. 2004, 131, 77–83. [CrossRef]
121. Jakstas, T.; Bartnykaite, A.; Padervinskis, E.; Vegiene, A.; Juozaityte, E.; Uloza, V.; Ugenskiene, R. The Association of TP53, BCL2,
     BAX and NOXA SNPs and Laryngeal Squamous Cell Carcinoma Development. Int. J. Mol. Sci. 2024, 25, 11849. [CrossRef]
122. Chen, J.; Zhou, J.; Lu, J.; Xiong, H.; Shi, X.; Gong, L. Significance of CD44 Expression in Head and Neck Cancer: A Systemic
     Review and Meta-Analysis. BMC Cancer 2014, 14, 15. [CrossRef]
123. Trapasso, S.; Allegra, E. Role of CD44 as a Marker of Cancer Stem Cells in Head and Neck Cancer. Biol. Targets Ther. 2012, 6,
     379–383. [CrossRef]
124. Joshua, B.; Kaplan, M.J.; Doweck, I.; Pai, R.; Weissman, I.L.; Prince, M.E.; Ailles, L.E. Frequency of Cells Expressing CD44, a Head
     and Neck Cancer Stem Cell Marker: Correlation with Tumor Aggressiveness. Head Neck 2011, 34, 42–49. [CrossRef] [PubMed]
J. Clin. Med. 2025, 14, 3367                                                                                                              26 of 32
125. Re, M.; Gioacchini, F.M.; Scarpa, A.; Cassandro, C.; Tulli, M.; Cassandro, E. The Prognostic Significance of E-Cadherin Expression
     in Laryngeal Squamous-Cell Carcinoma: A Systematic Review. Acta Otorhinolaryngol. Ital. 2018, 38, 504–510. [CrossRef] [PubMed]
126. Li, J.; Zhang, G.; Yang, X.; Li, S.; Liu, X.; Yang, Q.; Li, Y.; Ye, J. Reduced E-Cadherin Expression Is Associated with Lymph Node
     Metastases in Laryngeal Squamous Cell Carcinoma. Auris Nasus Larynx 2012, 39, 186–192. [CrossRef]
127. Akdeniz, O.; Akduman, D.; Haksever, M.; Ozkarakas, H.; Müezzinoglu, B. Relationships between Clinical Behavior of Laryngeal
     Squamous Cell Carcinomas and Expression of VEGF, MMP-9 and E-Cadherin. Asian Pac. J. Cancer Prev. 2013, 14, 5301–5310.
     [CrossRef]
128. Cappellesso, R.; Marioni, G.; Crescenzi, M.; Giacomelli, L.; Guzzardo, V.; Mussato, A.; Staffieri, A.; Martini, A.; Blandamura, S.;
     Fassina, A. The Prognostic Role of the Epithelial-Mesenchymal Transition Markers E-Cadherin and Slug in Laryngeal Squamous
     Cell Carcinoma. Histopathology 2015, 67, 491–500. [CrossRef]
129. CDKN2A Gene: MedlinePlus Genetics. Available online: https://medlineplus.gov/genetics/gene/cdkn2a/ (accessed on 8
     May 2025).
130. Nowosad, A.; Besson, A. CDKN1B/P27 Regulates Autophagy via the Control of Ragulator and MTOR Activity in Amino
     Acid-Deprived Cells. Autophagy 2020, 16, 2297–2298. [CrossRef]
131. Gioacchini, F.M.; Alicandri-Ciufelli, M.; Kaleci, S.; Magliulo, G.; Presutti, L.; Re, M. The Prognostic Value of Cyclin D1 Expression
     in Head and Neck Squamous Cell Carcinoma. Eur. Arch. Oto-Rhino-Laryngol. 2014, 273, 801–809. [CrossRef]
132. Zhang, Z.; Tang, H.; Lin, J.; Hu, Y.; Luo, G.; Luo, Z.; Cheng, C.; Wang, P. Clinicopathologic and Prognostic Significance of Human
     Epidermal Growth Factor Receptor in Patients with Gastric Cancer: An Updated Meta-Analysis. Oncotarget 2017, 8, 17202–17215.
     [CrossRef]
133. Gioacchini, F.M.; Alicandri-Ciufelli, M.; Magliulo, G.; Rubini, C.; Presutti, L.; Re, M. The Clinical Relevance of Ki-67 Expression in
     Laryngeal Squamous Cell Carcinoma. Eur. Arch. Oto-Rhino-Laryngol. 2014, 272, 1569–1576. [CrossRef]
134. Mirza, S.; Jeannon, J.P.; Soames, J.; Wilson, J.A. Is Ki67 a Marker for the Transformation of Laryngeal Dysplasia to Carcinoma?
     Acta Oto-Laryngol. 2006, 126, 418–421. [CrossRef] [PubMed]
135. Acikalin, M.F.; Öner, Ü.; Tel, N.; Paşaoğlu, Ö.; Çakli, H.; Çolak, E. Prognostic Significance of Ki-67 Expression for Patients with
     Laryngeal Squamous Cell Carcinoma Primarily Treated by Total Laryngectomy. Eur. Arch. Oto-Rhino-Laryngol. 2003, 261, 376–380.
     [CrossRef] [PubMed]
136. Re, M.; Zizzi, A.; Ferrante, L.; Stramazzotti, D.; Goteri, G.; Gioacchini, F.M.; Olivieri, F.; Magliulo, G.; Rubini, C. P63 and Ki-67
     Immunostainings in Laryngeal Squamous Cell Carcinoma Are Related to Survival. Eur. Arch. Oto-Rhino-Laryngol. 2014, 271,
     1641–1651. [CrossRef] [PubMed]
137. Boehm, E.M.; Gildenberg, M.S.; Washington, M.T. The Many Roles of PCNA in Eukaryotic DNA Replication. In The Enzymes;
     Academic Press: Cambridge, MA, USA, 2016; Volume 39, pp. 231–254.
138. Zhao, H.; Chen, Q.; Alam, A.; Cui, J.; Suen, K.C.; Soo, A.P.; Eguchi, S.; Gu, J.; Ma, D. The Role of Osteopontin in the Progression of
     Solid Organ Tumour. Cell Death Dis. 2018, 9, 356. [CrossRef]
139. Leemans, C.R.; Snijders, P.J.F.; Brakenhoff, R.H. The Molecular Landscape of Head and Neck Cancer. Nat. Rev. Cancer 2018, 18,
     269–282. [CrossRef]
140. Bradford, C.R.; Kumar, B.; Bellile, E.; Lee, J.; Taylor, J.; D’Silva, N.; Cordell, K.; Kleer, C.; Kupfer, R.; Kumar, P.; et al. Biomarkers in
     Advanced Larynx Cancer. Laryngoscope 2014, 124, 179–187. [CrossRef]
141. Shibuya, M. Vascular Endothelial Growth Factor (VEGF) and Its Receptor (VEGFR) Signaling in Angiogenesis. Genes. Cancer
     2011, 2, 1097–1105. [CrossRef] [PubMed]
142. Teknos, T.N.; Cox, C.; Yoo, S.; Chepeha, D.B.; Wolf, G.T.; Bradford, C.R.; Carey, T.E.; Fisher, S.G. Elevated Serum Vascular
     Endothelial Growth Factor and Decreased Survival in Advanced Laryngeal Carcinoma. Head Neck 2002, 24, 1004–1011. [CrossRef]
143. Parikh, R.R.; Yang, Q.; Haffty, B.G. Prognostic Significance of Vascular Endothelial Growth Factor Protein Levels in T1-2 N0
     Laryngeal Cancer Treated with Primary Radiation Therapy. Cancer 2007, 109, 566–573. [CrossRef]
144. Neuchrist, C.; Quint, C.; Pammer, A.; Burian, M. Vascular Endothelial Growth Factor (VEGF) and Microvessel Density in
     Squamous Cell Carcinomas of the Larynx: An Immunohistochemical Study. Acta Otolaryngol. 1999, 119, 732–738. [CrossRef]
145. Cossu, A.M.; Mosca, L.; Zappavigna, S.; Misso, G.; Bocchetti, M.; De Micco, F.; Quagliuolo, L.; Porcelli, M.; Caraglia, M.; Boccellino,
     M. Long Non-Coding RNAs as Important Biomarkers in Laryngeal Cancer and Other Head and Neck Tumours. Int. J. Mol. Sci.
     2019, 20, 3444. [CrossRef] [PubMed]
146. Balas, M.M.; Johnson, A.M. Exploring the Mechanisms behind Long Noncoding RNAs and Cancer. Non-Coding RNA Res. 2018, 3,
     108–117. [CrossRef] [PubMed]
147. Zhou, R.; Chen, K.K.; Zhang, J.; Xiao, B.; Huang, Z.; Ju, C.; Sun, J.; Zhang, F.; Lv, X.-B.; Huang, G. The Decade of Exosomal Long
     RNA Species: An Emerging Cancer Antagonist. Mol. Cancer 2018, 17, 75. [CrossRef]
148. Zvrko, E.; Mikic, A.; Vuckovic, L. CD105 Expression as a Measure of Microvessel Density in Supraglottic Laryngeal Squamous
     Cell Carcinoma. Eur. Arch. Oto-Rhino-Laryngol. 2009, 266, 1971–1976. [CrossRef]
J. Clin. Med. 2025, 14, 3367                                                                                                         27 of 32
149. Zvrko, E.; Mikic, A.; Vuckovic, L. Clinicopathologic Significance of CD105-Assessed Microvessel Density in Glottic Laryngeal
     Squamous Cell Carcinoma. Auris Nasus Larynx 2010, 37, 77–83. [CrossRef]
150. Marioni, G.; Blandamura, S.; Loreggian, L.; Koussis, H.; Lionello, M.; Giacomelli, L.; Fasanaro, E.; Lovato, A.; Staffieri, A.
     Laryngeal Carcinoma Prognosis after Postoperative Radiotherapy Correlates with CD105 Expression, but Not with Angiogenin
     or EGFR Expression. Eur. Arch. Oto-Rhino-Laryngol. 2011, 268, 1779–1787. [CrossRef]
151. Villaronga, M.Á.; Hermida-Prado, F.; Granda-Díaz, R.; Menéndez, S.T.; Álvarez-Teijeiro, S.; Quer, M.; Vilaseca, I.; Allonca, E.;
     Garzón-Arango, M.; Sanz-Moreno, V.; et al. Immunohistochemical Expression of Cortactin and Focal Adhesion Kinase Predicts
     Recurrence Risk and Laryngeal Cancer Risk Beyond Histologic Grading. Cancer Epidemiol. Biomark. Prev. 2018, 27, 805–813.
     [CrossRef]
152. Rodrigo, J.P.; Álvarez-Alija, G.; Menéndez, S.T.; Mancebo, G.; Allonca, E.; García-Carracedo, D.; Fresno, M.F.; Suárez, C.; García-
     Pedrero, J.M. Cortactin and Focal Adhesion Kinase as Predictors of Cancer Risk in Patients with Laryngeal Premalignancy. Cancer
     Prev. Res. 2011, 4, 1333–1341. [CrossRef]
153. Marioni, G.; Lionello, M.; Marchese-Ragona, R.; Fasanaro, E.; Valentini, E.; Zanoletti, E.; Stritoni, P.; Ramacciotti, G.; Guzzardo, V.;
     Giacomelli, L.; et al. Cortactin and Phosphorylated Cortactin Tyr421 and Tyr466 Expression in Supraglottic Laryngeal Carcinomas
     and Lymph Node Metastases. Int. J. Biol. Markers 2017, 33, 79–86. [CrossRef] [PubMed]
154. Gibcus, J.H.; Mastik, M.F.; Menkema, L.; de Bock, G.H.; Kluin, P.M.; Schuuring, E.; van der Wal, J.E. Cortactin Expression Predicts
     Poor Survival in Laryngeal Carcinoma. Br. J. Cancer 2008, 98, 950–955. [CrossRef]
155. Fei, M.; Zhang, J.; Zhou, J.; Xu, Y.; Wang, J. Sex-Related Hormone Receptor in Laryngeal Squamous Cell Carcinoma: Correlation
     with Androgen Estrogen-α and Prolactin Receptor Expression and Influence of Prognosis. Acta Oto-Laryngol. 2017, 138, 66–72.
     [CrossRef] [PubMed]
156. Kalyi, V.V.; Goncharuk-Khomyn, M.Y. Analysis of the Role and Impact of Sex Hormones in Diagnostics and Assessment of
     Clinical Progress of Laryngeal Cancer. Morphologia 2016, 10, 41–45. [CrossRef]
157. Ulitsky, I.; Bartel, D.P. lincRNAs: Genomics, Evolution, and Mechanisms. Cell 2013, 154, 26–46. [CrossRef] [PubMed]
158. Ziegler, C.; Kretz, M. The More the Merrier-Complexity in Long Non-Coding RNA Loci. Front. Endocrinol. 2017, 8, 90. [CrossRef]
159. Ahmed, W.A.; Suzuki, K.; Imaeda, Y.; Horibe, Y. Ki-67, P53 and Epidermal Growth Factor Receptor Expression in Early Glottic
     Cancer Involving the Anterior Commissure Treated with Radiotherapy. Auris Nasus Larynx 2008, 35, 213–219. [CrossRef]
160. Aras, S.; Ozkanli, S.; Erdem, E.; Gokalp, S.; Erdogan, C.E. Investigation of Low and High Dose Rate X-Ray Effects on Histopatho-
     logical Changes and Prognostic Importance of Ki-67 in Laryngeal Cancer Radiotherapy. Appl. Radiat. Isot. 2023, 197, 110823.
     [CrossRef]
161. Sakata, K.; Oouchi, A.; Nagakura, H.; Akiba, H.; Tamakawa, M.; Koito, K.; Hareyama, M.; Asakura, K.; Satoh, M.; Ohtani, S.
     Accelerated Radiotherapy for T1, 2 Glottic Carcinoma: Analysis of Results with KI-67 Index. Int. J. Radiat. Oncol. Biol. Phys. 2000,
     47, 81–88. [CrossRef]
162. Liu, X.-Y.; Hanjing, S.; Zhang, B.-H.; Luo, X.-Y. Expression of P16 and Ki67 in Laryngeal Squamous Cell Carcinoma and Their
     Clinical Significance. Front. Oncol. 2024, 14, 1430830. [CrossRef]
163. DE Almeida, M.R.; Pérez-Sayáns, M.; Suárez-Peñaranda, J.M.; Somoza-Martín, J.M.; García-García, A. P27(Kip1) Expression as a
     Prognostic Marker for Squamous Cell Carcinoma of the Head and Neck. Oncol. Lett. 2015, 10, 2675–2682. [CrossRef]
164. Allegra, E.; Bianco, M.R.; Mignogna, C.; Caltabiano, R.; Grasso, M.; Puzzo, L. Role of P16 Expression in the Prognosis of Patients
     with Laryngeal Cancer: A Single Retrospective Analysis. Cancer Control 2021, 28, 10732748211033544. [CrossRef]
165. Lee, L.-A.; Fang, T.-J.; Li, H.-Y.; Chuang, H.-H.; Kang, C.-J.; Chang, K.-P.; Liao, C.-T.; Chen, T.-C.; Huang, C.-G.; Yen, T.-C. Effects
     of Epstein-Barr Virus Infection on the Risk and Prognosis of Primary Laryngeal Squamous Cell Carcinoma: A Hospital-Based
     Case-Control Study in Taiwan. Cancers 2021, 13, 1741. [CrossRef] [PubMed]
166. Silva, F.F.V.e.; Caponio, V.C.A.; Camolesi, G.C.V.; Padín-Iruegas, M.E.; Lorenzo-Pouso, A.I.; Lima, K.C.; Vieira, S.L.S.; Chamorro-
     Petronacci, C.M.; Suaréz-Peñaranda, J.M.; Pérez-Sayáns, M. Correlation of Bcl-2 Expression with Prognosis and Survival in
     Patients with Head and Neck Cancer: A Systematic Review and Meta-Analysis. Crit. Rev. Oncol./Hematol. 2023, 187, 104021.
     [CrossRef] [PubMed]
167. Chan, J.Y.K.; Zhen, G.; Agrawal, N. The Role of Tumor DNA as a Diagnostic Tool for Head and Neck Squamous Cell Carcinoma.
     Semin. Cancer Biol. 2019, 55, 1–7. [CrossRef] [PubMed]
168. Petitjean, A.; Mathe, E.; Kato, S.; Ishioka, C.; Tavtigian, S.V.; Hainaut, P.; Olivier, M. Impact of Mutant P53 Functional Properties
     on TP53 Mutation Patterns and Tumor Phenotype: Lessons from Recent Developments in the IARC TP53 Database. Human.
     Mutat. 2007, 28, 622–629. [CrossRef]
169. Leemans, C.R.; Braakhuis, B.J.M.; Brakenhoff, R.H. The Molecular Biology of Head and Neck Cancer. Nat. Rev. Cancer 2010, 11,
     9–22. [CrossRef]
170. Poeta, M.L.; Manola, J.; Goldwasser, M.A.; Forastiere, A.; Benoit, N.; Califano, J.A.; Ridge, J.A.; Goodwin, J.; Kenady, D.; Saunders,
     J.; et al. TP53 Mutations and Survival in Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2007, 357, 2552–2561.
     [CrossRef]
J. Clin. Med. 2025, 14, 3367                                                                                                            28 of 32
171. Ioachim, E. Expression Patterns of Cyclins D1, E and Cyclin-Dependent Kinase Inhibitors P21waf1/Cip1, P27kip1 in Colorectal
     Carcinoma: Correlation with Other Cell Cycle Regulators (pRb, P53 and Ki-67 and PCNA) and Clinicopathological Features. Int.
     J. Clin. Pract. 2008, 62, 1736–1743. [CrossRef]
172. Basyuni, S.; Nugent, G.; Ferro, A.; Barker, E.; Reddin, I.; Jones, O.; Lechner, M.; O’Leary, B.; Jones, T.; Masterson, L.; et al. Value of
     P53 Sequencing in the Prognostication of Head and Neck Cancer: A Systematic Review and Meta-Analysis. Sci. Rep. 2022, 12,
     20776. [CrossRef]
173. Tan, A.; Eskiizmir, G.; Kamiloglu, U.; Sarioglu, S. P53 and PTEN Expression Evaluation with Molecular Evident Recent Criteria in
     Laryngeal Carcinoma. Medicine 2023, 102, e33676. [CrossRef]
174. Chong, C.R.; Jänne, P.A. The Quest to Overcome Resistance to EGFR-Targeted Therapies in Cancer. Nat. Med. 2013, 19, 1389–1400.
     [CrossRef]
175. Lionello, M.; Staffieri, A.; Marioni, G. Potential Prognostic and Therapeutic Role for Angiogenesis Markers in Laryngeal
     Carcinoma. Acta Oto-Laryngol. 2012, 132, 574–582. [CrossRef] [PubMed]
176. Kontić, M.; Čolović, Z.; Paladin, I.; Gabelica, M.; Barić, A.; Pešutić-Pisac, V. Association between EGFR Expression and Clinical
     Outcome of Laryngeal HPV Squamous Cell Carcinoma. Acta Oto-Laryngol. 2019, 139, 913–917. [CrossRef] [PubMed]
177. Marijić, B.; Braut, T.; Babarović, E.; Krstulja, M.; Maržić, D.; Avirović, M.; Kujundžić, M.; Hadžisejdić, I. Nuclear EGFR Expression
     Is Associated with Poor Survival in Laryngeal Carcinoma. Appl. Immunohistochem. Mol. Morphol. 2021, 29, 576–584. [CrossRef]
     [PubMed]
178. Kapral, M.; Strzalka, B.; Kowalczyk, M.; Jurzak, M.; Mazurek, U.; Gierek, T.; Paluch, J.; Markowski, J.; Swiatkowska, L.; Weglarz,
     L. Transforming Growth Factor Beta Isoforms (TGF-Beta1, TGF-Beta2, TGF-Beta3) Messenger RNA Expression in Laryngeal
     Cancer. Am. J. Otolaryngol. 2008, 29, 233–237. [CrossRef]
179. Mosch, B.; Reissenweber, B.; Neuber, C.; Pietzsch, J. Eph Receptors and Ephrin Ligands: Important Players in Angiogenesis and
     Tumor Angiogenesis. J. Oncol. 2010, 2010, 135285. [CrossRef]
180. Shigyo, H.; Nonaka, S.; Katada, A.; Bandoh, N.; Ogino, T.; Katayama, A.; Takahara, M.; Hayashi, T.; Harabuchi, Y. Inducible Nitric
     Oxide Synthase Expression in Various Laryngeal Lesions in Relation to Carcinogenesis, Angiogenesis, and Patients’ Prognosis.
     Acta Oto-Laryngol. 2007, 127, 970–979. [CrossRef]
181. Rueda, A.; Cazorla, O.; Pérez, L.; Álvarez, M.; Redondo, M.; Gallego, E.; Sáez, M.; Medina, J.A.; Solano, J.; Matilla, A. Vascular
     Endothelial Growth Factor and Vascular Endothelial Growth Factor Receptor-2 Tumor Expression in Patients with Advanced
     Laryngeal Cancer after Induction Chemotherapy for Organ Preservation. Head Neck 2010, 33, 808–816. [CrossRef]
182. Bonhin, R.G.; Rocha, V.B.C.; Carvalho, G.M.d.; Guimarães, A.C.; Crespo, A.N.; Chone, C.T.; Amstalden, E.M.I. Correlation
     between Vascular Endothelial Growth Factor Expression and Presence of Lymph Node Metastasis in Advanced Squamous Cell
     Carcinoma of the Larynx. Braz. J. Otorhinolaryngol. 2015, 81, 58–62. [CrossRef]
183. Yoshioka, N.; Wang, L.; Kishimoto, K.; Tsuji, T.; Hu, G. A Therapeutic Target for Prostate Cancer Based on Angiogenin-Stimulated
     Angiogenesis and Cancer Cell Proliferation. Proc. Natl. Acad. Sci. USA 2006, 103, 14519–14524. [CrossRef]
184. Marioni, G.; Marino, F.; Blandamura, S.; D’Alessandro, E.; Giacomelli, L.; Guzzardo, V.; Lionello, M.; De Filippis, C.; Staffieri, A.
     Neoangiogenesis in Laryngeal Carcinoma: Angiogenin and CD105 Expression Is Related to Carcinoma Recurrence Rate and
     Disease-free Survival. Histopathology 2010, 57, 535–543. [CrossRef]
185. Carico, E.; Radici, M.; Bucci, B.; Firrisi, L.; Fabiano, A.; Salerno, G.; Giovagnoli, M.R.; Vecchione, A. p16INK4/Ki-67 Dual-Staining
     Expression as a Prognostic Indicator in Laryngeal Cancer. J. Cancer Prev. Curr. Res. 2014, 1, 68–72. [CrossRef]
186. Verma, A.; Schwartz, N.; Cohen, D.J.; Boyan, B.D.; Schwartz, Z. Estrogen Signaling and Estrogen Receptors as Prognostic
     Indicators in Laryngeal Cancer. Steroids 2019, 152, 108498. [CrossRef] [PubMed]
187. Schwartz, N.; Verma, A.; Muktipaty, C.; Bivens, C.; Schwartz, Z.; Boyan, B.D. Estradiol Receptor Profile and Estrogen Responsive-
     ness in Laryngeal Cancer and Clinical Outcomes. Steroids 2019, 142, 34–42. [CrossRef]
188. Ou, D.; Adam, J.; Garberis, I.; Blanchard, P.; Nguyen, F.; Levy, A.; Casiraghi, O.; Gorphe, P.; Breuskin, I.; Janot, F.; et al. Clinical
     Relevance of Tumor Infiltrating Lymphocytes, PD-L1 Expression and Correlation with HPV/P16 in Head and Neck Cancer
     Treated with Bio- or Chemo-Radiotherapy. Oncoimmunology 2017, 6, e1341030. [CrossRef]
189. Vassilakopoulou, M.; Avgeris, M.; Velcheti, V.; Kotoula, V.; Rampias, T.; Chatzopoulos, K.; Perisanidis, C.; Kontos, C.K.; Giotakis,
     A.I.; Scorilas, A.; et al. Evaluation of PD-L1 Expression and Associated Tumor-Infiltrating Lymphocytes in Laryngeal Squamous
     Cell Carcinoma. Clin. Cancer Res. 2016, 22, 704–713. [CrossRef]
190. Wang, J.; Wang, S.; Song, X.; Zeng, W.; Wang, S.; Chen, F.; Ding, H. The Prognostic Value of Systemic and Local Inflammation in
     Patients with Laryngeal Squamous Cell Carcinoma. OncoTargets Ther. 2016, 9, 7177–7185. [CrossRef]
191. Tudor, F.; Marijić, B.; Babarović, E.; Hadžisejdić, I. Significance of PD-L1 and Tumor Microenvironment in Laryngeal Squamous
     Cell Cancer. Cancers 2024, 16, 2645. [CrossRef]
192. Economopoulou, P.; de Bree, R.; Kotsantis, I.; Psyrri, A. Diagnostic Tumor Markers in Head and Neck Squamous Cell Carcinoma
     (HNSCC) in the Clinical Setting. Front. Oncol. 2019, 9, 827. [CrossRef]
J. Clin. Med. 2025, 14, 3367                                                                                                       29 of 32
193. Syn, N.L.; Teng, M.W.L.; Mok, T.S.K.; Soo, R.A. De-Novo and Acquired Resistance to Immune Checkpoint Targeting. Lancet Oncol.
     2017, 18, e731–e741. [CrossRef]
194. Francisco, L.M.; Sage, P.T.; Sharpe, A.H. The PD-1 Pathway in Tolerance and Autoimmunity. Immunol. Rev. 2010, 236, 219–242.
     [CrossRef]
195. Ritprajak, P.; Azuma, M. Intrinsic and Extrinsic Control of Expression of the Immunoregulatory Molecule PD-L1 in Epithelial
     Cells and Squamous Cell Carcinoma. Oral. Oncol. 2015, 51, 221–228. [CrossRef] [PubMed]
196. Karam, J.; Fadous-Khalifé, M.C.; Tannous, R.; Fakhreddine, S.; Massoud, M.; Hadchity, J.; Aftimos, G.; Hadchity, E. Role of
     Krüppel-like Factor 4 and Heat Shock Protein 27 in Cancer of the Larynx. Mol. Clin. Oncol. 2017, 7, 808–814. [CrossRef] [PubMed]
197. Kaigorodova, E.V.; Zavyalova, M.V.; Bychkov, V.A.; Perelmuter, V.M.; Choynzonov, E.L. Functional State of the Hsp27 Chaperone
     as a Molecular Marker of an Unfavorable Course of Larynx Cancer. Cancer Biomark. 2016, 17, 145–153. [CrossRef]
198. Lee, J.-H.; Sun, D.; Cho, K.-J.; Kim, M.-S.; Hong, M.-H.; Kim, I.-K.; Lee, J.-S.; Lee, J.-H. Overexpression of Human 27 kDa Heat
     Shock Protein in Laryngeal Cancer Cells Confers Chemoresistance Associated with Cell Growth Delay. J. Cancer Res. Clin. Oncol.
     2006, 133, 37–46. [CrossRef] [PubMed]
199. Rappa, F.; Pitruzzella, A.; Marino Gammazza, A.; Barone, R.; Mocciaro, E.; Tomasello, G.; Carini, F.; Farina, F.; Zummo, G.;
     Conway de Macario, E.; et al. Quantitative Patterns of Hsps in Tubular Adenoma Compared with Normal and Tumor Tissues
     Reveal the Value of Hsp10 and Hsp60 in Early Diagnosis of Large Bowel Cancer. Cell Stress Chaperones 2016, 21, 927–933.
     [CrossRef]
200. Xu, J.; Wang, K.; Zhang, X.; Qiu, Y.; Huang, D.; Li, W.; Xiao, X.; Tian, Y. HSP70: A Promising Target for Laryngeal Carcinoma
     Radiaotherapy by Inhibiting Cleavage and Degradation of Nucleolin. J. Exp. Clin. Cancer Res. 2010, 29, 106. [CrossRef]
201. Orasan, A.; Negru, M.-C.; Morgovan, A.I.; Fleser, R.C.; Sandu, D.; Sitaru, A.M.; Motofelea, A.-C.; Balica, N.C. Strategies to
     Mitigate Cisplatin-Induced Ototoxicity: A Literature Review of Protective Agents, Mechanisms, and Clinical Gaps. Audiol. Res.
     2025, 15, 22. [CrossRef]
202. Song, X.; Liao, Z.; Zhou, C.; Lin, R.; Lu, J.; Cai, L.; Tan, X.; Zeng, W.; Lu, X.; Zheng, W.; et al. HSP47 Is Associated with the
     Prognosis of Laryngeal Squamous Cell Carcinoma by Inhibiting Cell Viability and Invasion and Promoting Apoptosis. Oncol. Rep.
     2017, 38, 2444–2452. [CrossRef]
203. Si, M.; Lang, J. The Roles of Metallothioneins in Carcinogenesis. J. Hematol. Oncol. 2018, 11, 107. [CrossRef]
204. Ioachim, E.; Assimakopoulos, D.; Peschos, D.; Zissi, A.; Skevas, A.; Agnantis, N.J. Immunohistochemical Expression of Metalloth-
     ionein in Benign Premalignant and Malignant Epithelium of the Larynx: Correlation with P53 and Proliferative Cell Nuclear
     Antigen. Pathol.—Res. Pract. 1999, 195, 809–814. [CrossRef]
205. Nowinska, K.; Chmielewska, M.; Piotrowska, A.; Pula, B.; Pastuszewski, W.; Krecicki, T.; Podhorska-Okołow, M.; Zabel, M.;
     Dziegiel, P. Correlation between Levels of Expression of Minichromosome Maintenance Proteins, Ki-67 Proliferation Antigen and
     Metallothionein I/II in Laryngeal Squamous Cell Cancer. Int. J. Oncol. 2015, 48, 635–645. [CrossRef] [PubMed]
206. Starska, K.; Krześlak, A.; Forma, E.; Olszewski, J.; Lewy-Trenda, I.; Osuch-Wójcikiewicz, E.; Bryś, M. Genetic Polymorphism of
     Metallothionein 2A and Risk of Laryngeal Cancer in a Polish Population. Med. Oncol. 2014, 31, 75. [CrossRef] [PubMed]
207. Emanuele, S.; Celesia, A.; D’Anneo, A.; Lauricella, M.; Carlisi, D.; De Blasio, A.; Giuliano, M. The Good and Bad of Nrf2: An
     Update in Cancer and New Perspectives in COVID-19. Int. J. Mol. Sci. 2021, 22, 7963. [CrossRef]
208. Namani, A.; Matiur Rahaman, M.; Chen, M.; Tang, X. Gene-Expression Signature Regulated by the KEAP1-NRF2-CUL3 Axis Is
     Associated with a Poor Prognosis in Head and Neck Squamous Cell Cancer. BMC Cancer 2018, 18, 46. [CrossRef] [PubMed]
209. Xia, D.; Zhang, X.-R.; Ma, Y.-L.; Zhao, Z.-J.; Zhao, R.; Wang, Y.-Y. Nrf2 Promotes Esophageal Squamous Cell Carcinoma (ESCC)
     Resistance to Radiotherapy through the CaMKIIα-Associated Activation of Autophagy. Cell Biosci. 2020, 10, 90. [CrossRef]
210. Sheth, S.; Farquhar, D.R.; Schrank, T.P.; Stepp, W.; Mazul, A.; Hayward, M.; Lenze, N.; Little, P.; Jo, H.; Major, M.B.; et al.
     Correlation of Alterations in the KEAP1/CUL3/NFE2L2 Pathway with Radiation Failure in Larynx Squamous Cell Carcinoma.
     Laryngoscope Investig. Otolaryngol. 2021, 6, 699–707. [CrossRef]
211. Li, C.; Wu, H.; Wang, S.; Zhu, J. Expression and Correlation of NRF2, KEAP1, NQO-1 and HO-1 in Advanced Squamous Cell
     Carcinoma of the Larynx and Their Association with Clinicopathologic Features. Mol. Med. Rep. 2016, 14, 5171–5179. [CrossRef]
212. Zhou, Z.; Xu, J.; Bao, X.; Shi, J.; Liu, B.; Chen, Y.; Li, J. Nuclear Nrf2 Activity in Laryngeal Carcinoma Is Regulated by SENP3 After
     Cisplatin-Induced Reactive Oxygen Species Stress. J. Cancer 2019, 10, 3427–3434. [CrossRef]
213. Cui, J.; Jia, J. Natural COX-2 Inhibitors as Promising Anti-Inflammatory Agents: An Update. Curr. Med. Chem. 2021, 28, 3622–3646.
     [CrossRef]
214. Li, M.; Li, M.; Wei, Y.; Xu, H. Prognostic and Clinical Significance of Cyclooxygenase-2 Overexpression in Endometrial Cancer: A
     Meta-Analysis. Front. Oncol. 2020, 10, 1202. [CrossRef]
215. Kourelis, K.; Vandoros, G.; Kourelis, T.; Papadas, T.; Goumas, P.; Sotiropoulou-Bonikou, G. Low COX2 in Tumor and Upregulation
     in Stroma Mark Laryngeal Squamous Cell Carcinoma Progression. Laryngoscope 2009, 119, 1723–1729. [CrossRef] [PubMed]
J. Clin. Med. 2025, 14, 3367                                                                                                          30 of 32
216. Chen, Y.-F.; Luo, R.-Z.; Li, Y.; Cui, B.-K.; Song, M.; Yang, A.-K.; Chen, W.-K. High Expression Levels of COX-2 and P300 Are
     Associated with Unfavorable Survival in Laryngeal Squamous Cell Carcinoma. Eur. Arch. Otorhinolaryngol. 2013, 270, 1009–1017.
     [CrossRef] [PubMed]
217. Du, J.; Feng, J.; Luo, D.; Peng, L. Prognostic and Clinical Significance of COX-2 Overexpression in Laryngeal Cancer: A
     Meta-Analysis. Front. Oncol. 2022, 12, 854946. [CrossRef] [PubMed]
218. Sackett, M.K.; Bairati, I.; Meyer, F.; Jobin, E.; Lussier, S.; Fortin, A.; Gélinas, M.; Nabid, A.; Brochet, F.; Têtu, B. Prognostic
     Significance of Cyclooxygenase-2 Overexpression in Glottic Cancer. Clin. Cancer Res. 2008, 14, 67–73. [CrossRef]
219. Klatka, J.; Grywalska, E.; Hymos, A.; Guz, M.; Polberg, K.; Roliński, J.; Stepulak, A. Cyclooxygenase-2 Inhibition Enhances
     Proliferation of NKT Cells Derived from Patients with Laryngeal Cancer. Anticancer Res. 2017, 37, 4059–4066. [CrossRef]
220. Kinoshita, T.; Hanazawa, T.; Nohata, N.; Kikkawa, N.; Enokida, H.; Yoshino, H.; Yamasaki, T.; Hidaka, H.; Nakagawa, M.;
     Okamoto, Y.; et al. Tumor Suppressive microRNA-218 Inhibits Cancer Cell Migration and Invasion through Targeting Laminin-332
     in Head and Neck Squamous Cell Carcinoma. Oncotarget 2012, 3, 1386–1400. [CrossRef]
221. Bruzgielewicz, A.; Osuch-Wojcikiewicz, E.; Niemczyk, K.; Sieniawska-Buccella, O.; Siwak, M.; Walczak, A.; Nowak, A.; Majsterek,
     I. Altered Expression of miRNAs Is Related to Larynx Cancer TNM Stage and Patients’ Smoking Status. DNA Cell Biol. 2017, 36,
     581–588. [CrossRef]
222. Xu, Y.; Lin, Y.-P.; Yang, D.; Zhang, G.; Zhou, H.-F. Clinical Significance of miR-149 in the Survival of Patients with Laryngeal
     Squamous Cell Carcinoma. BioMed Res. Int. 2016, 2016, 8561251. [CrossRef]
223. Karatas, O.F. Antiproliferative Potential of miR-33a in Laryngeal Cancer Hep-2 Cells via Targeting PIM1. Head Neck 2018, 40,
     2455–2461. [CrossRef]
224. Chen, H.; Cai, X.; Du, B.; Cai, J.; Luo, Z. MicroRNA-150-5p Inhibits the Proliferation and Invasion of Human Larynx Epidermiod
     Cancer Cells Though Regulating Peptidyl-Prolyl Cis/Trans Isomerase. Braz. J. Otorhinolaryngol. 2023, 89, 383–392. [CrossRef]
225. Luo, M.; Sun, G.; Sun, J. MiR-196b Affects the Progression and Prognosis of Human LSCC through Targeting PCDH-17. Auris
     Nasus Larynx 2019, 46, 583–592. [CrossRef] [PubMed]
226. Zhang, F.; Cao, H. MicroRNA-143-3p Suppresses Cell Growth and Invasion in Laryngeal Squamous Cell Carcinoma via Targeting
     the k-Ras/Raf/MEK/ERK Signaling Pathway. Int. J. Oncol. 2018, 54, 689–701. [CrossRef] [PubMed]
227. Maia, D.; de Carvalho, A.C.; Horst, M.A.; Carvalho, A.L.; Scapulatempo-Neto, C.; Vettore, A.L. Expression of miR-296-5p as
     Predictive Marker for Radiotherapy Resistance in Early-Stage Laryngeal Carcinoma. J. Transl. Med. 2015, 13, 262. [CrossRef]
     [PubMed]
228. Oztürkcan, S.; Katilmiş, H.; Ozdemir, I.; Tuna, B.; Güvenç, I.A.; Dündar, R. Occult Contralateral Nodal Metastases in Supraglottic
     Laryngeal Cancer Crossing the Midline. Eur. Arch. Oto-Rhino-Laryngol. 2009, 266, 117–120. [CrossRef]
229. Koroulakis, A.; Agarwal, M. Laryngeal Cancer. In StatPearls [Internet]; StatPearls Publishing: St. Petersburg, FL, USA, 2024.
230. Sharbel, D.D.; Abkemeier, M.; Groves, M.W.; Albergotti, W.G.; Byrd, J.K.; Reyes-Gelves, C. Occult Metastasis in Laryngeal
     Squamous Cell Carcinoma: A Systematic Review and Meta-Analysis. Ann. Otol. Rhinol. Laryngol. 2021, 130, 67–77. [CrossRef]
231. Khan, U.; MacKay, C.; Rigby, M.; Trites, J.; Corsten, M.; Taylor, S.M. Management of Positive Resection Margins Following
     Transoral Laser Microsurgery for Glottic Cancer. Laryngoscope Investig. Otolaryngol. 2023, 8, 1579–1583. [CrossRef]
232. Kjems, J.; Zukauskaite, R.; Johansen, J.; Eriksen, J.G.; Lassen, P.; Andersen, E.; Andersen, M.; Farhadi, M.; Overgaard, J.; Vogelius,
     I.R. Distant Metastases in Squamous Cell Carcinoma of the Pharynx and Larynx: A Population-Based DAHANCA Study. Acta
     Oncol. 2021, 60, 1472–1480. [CrossRef]
233. Van den Bovenkamp, K.; van der Vegt, B.; Halmos, G.B.; Slagter-Menkema, L.; Langendijk, J.A.; van Dijk, B.A.C.; Schuuring, E.;
     van der Laan, B.F.A.M. The Relation between Hypoxia and Proliferation Biomarkers with Radiosensitivity in Locally Advanced
     Laryngeal Cancer. Eur. Arch. Oto-Rhino-Laryngol. 2023, 280, 3801–3809. [CrossRef]
234. Jotic, A.; Milovanovic, J.; Savic-Vujovic, K.; Radin, Z.; Medic, B.; Folic, M.; Pavlovic, B.; Vujovic, A.; Dundjerovic, D. Immune Cell
     and Biochemical Biomarkers in Advanced Laryngeal Cancer. Dose Response 2022, 20, 15593258221115537. [CrossRef]
235. Koca, T.; Cetmi, D.A.; Aksoy, R.; Korcum, A.F. The Predictive Role of Inflammatory Biomarkers in Patients with Larynx Cancer
     Undergoing Definitive Radiotherapy. Technol. Cancer Res. Treat. 2024, 23, 15330338241280433. [CrossRef]
236. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [CrossRef] [PubMed]
237. Forastiere, A.A.; Zhang, Q.; Weber, R.S.; Maor, M.H.; Goepfert, H.; Pajak, T.F.; Morrison, W.; Glisson, B.; Trotti, A.; Ridge, J.A.;
     et al. Long-Term Results of RTOG 91-11: A Comparison of Three Nonsurgical Treatment Strategies to Preserve the Larynx in
     Patients with Locally Advanced Larynx Cancer. JCO 2013, 31, 845–852. [CrossRef] [PubMed]
238. Eisbruch, A.; Harris, J.; Garden, A.S.; Chao, C.K.S.; Straube, W.; Harari, P.M. Multi-Institutional Trial of Accelerated Hypofraction-
     ated Intensity-Modulated Radiation Therapy for Early-Stage Oropharyngeal Cancer (RTOG 00-22). Int. J. Radiat. Oncol. Biol. Phys.
     2010, 76, 1333–1338. [CrossRef] [PubMed]
239. Gillison, M.L.; Trotti, A.M.; Harris, J.; Eisbruch, A.; Harari, P.M.; Adelstein, D.J.; Jordan, R.C.K.; Zhao, W.; Sturgis, E.M.; Burtness,
     B.; et al. Radiotherapy plus Cetuximab or Cisplatin in Human Papillomavirus-Positive Oropharyngeal Cancer (NRG Oncology
     RTOG 1016): A Randomised, Multicentre, Non-Inferiority Trial. Lancet 2019, 393, 40–50. [CrossRef]
J. Clin. Med. 2025, 14, 3367                                                                                                           31 of 32
240. Burtness, B.; Harrington, K.J.; Greil, R.; Soulières, D.; Tahara, M.; Castro, G. Pembrolizumab Alone or with Chemotherapy versus
     Cetuximab with Chemotherapy for Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck (KEYNOTE-048): A
     Randomised, Open-Label, Phase 3 Study. Lancet 2019, 394, 1915–1928. [CrossRef]
241. NCI. FDA Approves Nivolumab for Head and Neck Cancer. Available online: https://www.cancer.gov/news-events/cancer-
     currents-blog/2016/fda-nivolumab-scchn (accessed on 4 May 2025).
242. Yen, C.-J.; Kiyota, N.; Hanai, N.; Takahashi, S.; Yokota, T.; Iwae, S.; Shimizu, Y.; Hong, R.-L.; Goto, M.; Kang, J.-H.; et al. Two-Year
     Follow-up of a Randomized Phase III Clinical Trial of Nivolumab vs. the Investigator’s Choice of Therapy in the Asian Population
     for Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck (CheckMate 141). Head Neck 2020, 42, 2852–2862.
     [CrossRef]
243. Lee, N.Y.; Ferris, R.L.; Psyrri, A.; Haddad, R.I.; Tahara, M.; Bourhis, J.; Harrington, K.; Chang, P.M.H.; Lin, J.C.; Razaq, M.A.;
     et al. Avelumab plus Standard-of-Care Chemoradiotherapy versus Chemoradiotherapy Alone in Patients with Locally Advanced
     Squamous Cell Carcinoma of the Head and Neck: A Randomised, Double-Blind, Placebo-Controlled, Multicentre, Phase 3 Trial.
     Lancet Oncol. 2021, 22, 450–462. [CrossRef]
244. Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Yokoi, T.; Chiappori, A.; Lee, K.H.; de Wit, M.; et al.
     Durvalumab after Chemoradiotherapy in Stage III Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2017, 377, 1919–1929. [CrossRef]
245. Yu, Y.; Lee, N.Y. JAVELIN Head and Neck 100: A Phase III Trial of Avelumab and Chemoradiation for Locally Advanced Head
     and Neck Cancer. Future Oncol. 2019, 15, 687–694. [CrossRef]
246. Machiels, J.-P.; Tao, Y.; Licitra, L.; Burtness, B.; Tahara, M.; Rischin, D.; Alves, G.; Lima, I.P.F.; Hughes, B.G.M.; Pointreau, Y.;
     et al. Pembrolizumab plus Concurrent Chemoradiotherapy versus Placebo plus Concurrent Chemoradiotherapy in Patients with
     Locally Advanced Squamous Cell Carcinoma of the Head and Neck (KEYNOTE-412): A Randomised, Double-Blind, Phase 3
     Trial. Lancet Oncol. 2024, 25, 572–587. [CrossRef]
247. Cohen, E.E.W.; Soulières, D.; Le Tourneau, C.; Dinis, J.; Licitra, L.; Ahn, M.-J.; Soria, A.; Machiels, J.-P.; Mach, N.; Mehra, R.
     Pembrolizumab versus Methotrexate, Docetaxel, or Cetuximab for Recurrent or Metastatic Head-and-Neck Squamous Cell
     Carcinoma (KEYNOTE-040): A Randomised, Open-Label, Phase 3 Study. Lancet 2019, 393, 156–167. [CrossRef] [PubMed]
248. Haddad, R.I.; Harrington, K.; Tahara, M.; Ferris, R.L.; Gillison, M.; Fayette, J.; Daste, A.; Koralewski, P.; Zurawski, B.; Taberna, M.;
     et al. Nivolumab Plus Ipilimumab Versus EXTREME Regimen as First-Line Treatment for Recurrent/Metastatic Squamous Cell
     Carcinoma of the Head and Neck: The Final Results of CheckMate 651. J. Clin. Oncol. 2023, 41, 2166–2180. [CrossRef] [PubMed]
249. Seiwert, T.Y.; Wildsmith, S.; Fayette, J.; Harrington, K.; Gillison, M.; Ahn, M.-J.; Takahashi, S.; Weiss, J.; Machiels, J.-P.; Baxi, S.;
     et al. Outcomes in Biomarker-Selected Subgroups from the KESTREL Study of Durvalumab and Tremelimumab in Recurrent or
     Metastatic Head and Neck Squamous Cell Carcinoma. Cancer Immunol. Immunother. 2024, 73, 70. [CrossRef] [PubMed]
250. Li, X.; Lee, A.; Cohen, M.A.; Sherman, E.J.; Lee, N.Y. Past, Present and Future of Proton Therapy for Head and Neck Cancer. Oral.
     Oncol. 2020, 110, 104879. [CrossRef]
251. Kang, B.-H.; Yu, T.; Kim, J.H.; Park, J.M.; Kim, J.-I.; Chung, E.-J.; Kwon, S.K.; Kim, J.H.; Wu, H.G. Early Closure of a Phase 1
     Clinical Trial for SABR in Early-Stage Glottic Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2019, 105, 104–109. [CrossRef]
252. Chotipanich, A. Total Laryngectomy: A Review of Surgical Techniques. Cureus 2021, 13, e18181. [CrossRef]
253. Campbell, G.; Glazer, T.A.; Kimple, R.J.; Bruce, J.Y. Advances in Organ Preservation for Laryngeal Cancer. Curr. Treat. Options
     Oncol. 2022, 23, 594–608. [CrossRef]
254. Remacle, M.; Van Haverbeke, C.; Eckel, H.; Bradley, P.; Chevalier, D.; Djukic, V.; de Vicentiis, M.; Friedrich, G.; Olofsson, J.; Peretti,
     G.; et al. Proposal for Revision of the European Laryngological Society Classification of Endoscopic Cordectomies. Eur. Arch.
     Oto-Rhino-Laryngol. 2007, 264, 709. [CrossRef]
255. Canis, M.; Ihler, F.; Martin, A.; Wolff, H.A.; Matthias, C.; Steiner, W. Organ Preservation in T4a Laryngeal Cancer: Is Transoral
     Laser Microsurgery an Option? Eur. Arch. Oto-Rhino-Laryngol. 2013, 270, 2719–2727. [CrossRef]
256. Peretti, G.; Piazza, C.; Mora, F.; Garofolo, S.; Guastini, L. Reasonable Limits for Transoral Laser Microsurgery in Laryngeal Cancer.
     Curr. Opin. Otolaryngol. Head Neck Surg. 2016, 24, 135–139. [CrossRef]
257. Day, A.T.; Sinha, P.; Nussenbaum, B.; Kallogjeri, D.; Haughey, B.H. Management of Primary T1–T4 Glottic Squamous Cell
     Carcinoma by Transoral Laser Microsurgery. Laryngoscope 2016, 127, 597–604. [CrossRef] [PubMed]
258. Weiss, B.G.; Ihler, F.; Wolff, H.A.; Schneider, S.; Canis, M.; Steiner, W.; Welz, C. Transoral Laser Microsurgery for Treatment for
     Hypopharyngeal Cancer in 211 Patients. Head Neck 2017, 39, 1631–1638. [CrossRef] [PubMed]
259. Canis, M.; Martin, A.; Ihler, F.; Wolff, H.A.; Kron, M.; Matthias, C.; Steiner, W. Results of Transoral Laser Microsurgery for
     Supraglottic Carcinoma in 277 Patients. Eur. Arch. Oto-Rhino-Laryngol. 2013, 270, 2315–2326. [CrossRef] [PubMed]
260. Hoffmann, C.; Cornu, N.; Hans, S.; Sadoughi, B.; Badoual, C.; Brasnu, D. Early Glottic Cancer Involving the Anterior Commissure
     Treated by Transoral Laser Cordectomy. Laryngoscope 2015, 126, 1817–1822. [CrossRef]
261. Breda, E.; Catarino, R.; Monteiro, E. Transoral Laser Microsurgery for Laryngeal Carcinoma: Survival Analysis in a Hospital-based
     Population. Head Neck 2014, 37, 1181–1186. [CrossRef]
J. Clin. Med. 2025, 14, 3367                                                                                                         32 of 32
262. Vilaseca, I.; Aviles-Jurado, F.X.; Valduvieco, I.; Berenguer, J.; Grau, J.J.; Baste, N.; Muxí, Á.; Castillo, P.; Lehrer, E.; Jordana,
     M. Transoral Laser Microsurgery in Locally Advanced Laryngeal Cancer: Prognostic Impact of Anterior versus Posterior
     Compartments. Head Neck 2021, 43, 3832–3842. [CrossRef]
263. Hans, S.; Chekkoury-Idrissi, Y.; Circiu, M.P.; Distinguin, L.; Crevier-Buchman, L.; Lechien, J.R. Surgical, Oncological, and
     Functional Outcomes of Transoral Robotic Supraglottic Laryngectomy. Laryngoscope 2020, 131, 1060–1065. [CrossRef]
264. Lechien, J.R.; Fakhry, N.; Saussez, S.; Chiesa-Estomba, C.-M.; Chekkoury-Idrissi, Y.; Cammaroto, G.; Melkane, A.E.; Barillari, M.R.;
     Crevier-Buchman, L.; Ayad, T.; et al. Surgical, Clinical and Functional Outcomes of Transoral Robotic Surgery for Supraglottic
     Laryngeal Cancers: A Systematic Review. Oral. Oncol. 2020, 109, 104848. [CrossRef]
265. Lechien, J.R.; Baudouin, R.; Circiu, M.P.; Chiesa-Estomba, C.M.; Crevier-Buchman, L.; Hans, S. Transoral Robotic Cordectomy for
     Glottic Carcinoma: A Rapid Review. Eur. Arch. Oto-Rhino-Laryngol. 2022, 279, 5449–5456. [CrossRef]
266. Remacle, M.; Prasad, V.M.N. Preliminary Experience in Transoral Laryngeal Surgery with a Flexible Robotic System for Benign
     Lesions of the Vocal Folds. Eur. Arch. Oto-Rhino-Laryngol. 2018, 275, 761–765. [CrossRef]
267. Hans, S.; Chebib, E.; Lisan, Q.; Chekkoury-Idrissi, Y.; Distinguin, L.; Circiu, M.P.; Crevier-Buchman, L.; Lechien, J.R. Oncological,
     Surgical and Functional Outcomes of Transoral Robotic Cordectomy for Early Glottic Carcinoma. J. Voice 2023, 37, 801.e3–801.e7.
     [CrossRef] [PubMed]
268. Steuer, C.E.; El-Deiry, M.; Parks, J.R.; Higgins, K.A.; Saba, N.F. An Update on Larynx Cancer. CA Cancer J. Clin. 2017, 67, 31–50.
     [CrossRef] [PubMed]
269. Li, M.M.; Zhao, S.; Eskander, A.; Rygalski, C.; Brock, G.; Parikh, A.S.; Haring, C.T.; Swendseid, B.; Zhan, K.Y.; Bradford, C.R.
     Stage Migration and Survival Trends in Laryngeal Cancer. Ann. Surg. Oncol. 2021, 28, 7300–7309. [CrossRef]
270. Patel, T.R.; Eggerstedt, M.; Toor, J.; Tajudeen, B.A.; Husain, I.; Stenson, K.; Al-Khudari, S. Occult Lymph Node Metastasis in
     Early-stage Glottic Cancer in the National Cancer Database. Laryngoscope 2021, 131, E1139–E1146. [CrossRef]
271. Nahavandipour, A.; Jakobsen, K.K.; Grønhøj, C.; Hebbelstrup Jensen, D.; Kim Schmidt Karnov, K.; Klitmøller Agander, T.; Specht,
     L.; von Buchwald, C. Incidence and Survival of Laryngeal Cancer in Denmark: A Nation-Wide Study from 1980 to 2014. Acta
     Oncol. 2019, 58, 977–982. [CrossRef]
272. Petrakos, I.; Kontzoglou, K.; Nikolopoulos, T.P.; Papadopoulos, O.; Kostakis, A. Glottic and Supraglottic Laryngeal Cancer:
     Epidemiology, Treatment Patterns and Survival in 164 Patients. J. Buon 2012, 17, 700–705.
273. Yang, F.; He, L.; Rao, Y.; Feng, Y.; Wang, J. Survival Analysis of Patients with Subglottic Squamous Cell Carcinoma Based on the
     SEER Database. Braz. J. Otorhinolaryngol. 2023, 88, S70–S80. [CrossRef]
274. MacNeil, S.D.; Patel, K.; Liu, K.; Shariff, S.; Yoo, J.; Nichols, A.; Fung, K.; Garg, A.X. Survival of Patients with Subglottic Squamous
     Cell Carcinoma. Curr. Oncol. 2018, 25, e569. [CrossRef]
275. Juan, X.; Jiali, H.; Ziqi, L.; Liqing, Z.; Han, Z. Development and Validation of Nomogram Models for Predicting Postoperative
     Prognosis of Early-Stage Laryngeal Squamous Cell Carcinoma. Curr. Probl. Cancer 2024, 49, 101079. [CrossRef]
276. Shi, X.; Hu, W.-P.; Ji, Q.-H. Development of Comprehensive Nomograms for Evaluating Overall and Cancer-Specific Survival of
     Laryngeal Squamous Cell Carcinoma Patients Treated with Neck Dissection. Oncotarget 2017, 8, 29722–29740. [CrossRef]
277. Qasem, M.; Qasem, N.; Kinshuck, A.; Milinis, K. Long-Term Laryngeal Function and Quality of Life Following Treatment of Early
     Glottic Cancer: A Meta-Analysis. Otolaryngol. Head Neck Surg. 2025, 172, 375–385. [CrossRef] [PubMed]
278. Chien, P.-J.; Hung, L.-T.; Wang, L.-W.; Yang, M.-H.; Chu, P.-Y. Oncologic Results and Quality of Life in Patients with T3 Glottic
     Cancer after Transoral Laser Microsurgery. Eur. Arch. Oto-Rhino-Laryngol. 2021, 278, 2983–2992. [CrossRef] [PubMed]
279. Wulff, N.B.; Højager, A.; Wessel, I.; Dalton, S.O.; Homøe, P. Health-related Quality of Life Following Total Laryngectomy: A
     Systematic Review. Laryngoscope 2021, 131, 820–831. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.