Next Article in Journal
Infectious Spleen and Kidney Necrosis Virus Triggers Ferroptosis in CPB Cells to Enhance Virus Replication
Previous Article in Journal
Genetic Diversity in the Capsid Protein-Coding Region of HIV-1 Circulating in Benguela, Angola: Implications for Primary Resistance to the Novel Capsid Inhibitor Lenacapavir
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Update: Immunotherapeutic Strategies in HPV-Associated Head and Neck Squamous Cell Carcinoma

Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
*
Author to whom correspondence should be addressed.
Viruses 2025, 17(5), 712; https://doi.org/10.3390/v17050712
Submission received: 24 April 2025 / Revised: 14 May 2025 / Accepted: 15 May 2025 / Published: 16 May 2025
(This article belongs to the Special Issue Advancements in Immunotherapy for Human Papillomavirus)

Abstract

:
The incidence of human papillomavirus (HPV)-associated oropharyngeal squamous cell carcinoma (OPSCC) has increased substantially over the past three decades, and since 2017, it has been recognized in the AJCC staging system as distinct from its HPV-negative counterpart. The underlying mechanisms of HPV-associated carcinogenesis, tumor microenvironment, and host immune response represent opportunities for therapeutic development. While anti-PD-1 immunotherapy is now part of standard treatment for recurrent or metastatic head and neck squamous cell carcinoma (HNSCC) in general, there are no established immunotherapeutic strategies specifically for HPV-related HNSCC. In this context, multiple emerging approaches are being actively studied—among these are therapeutic vaccines with or without anti-PD-(L)1 adjuvants, peptide–HLA-based immunotherapeutic platforms, and adoptive cell therapies including tumor-infiltrating lymphocytes (TILs), T-cell receptor (TCR) therapy, and chimeric antigen receptor (CAR) T-cell therapy. Beyond further maturation of these novel immunotherapeutic strategies, additional work is needed to delineate the optimal disease state of application (localized versus recurrent/metastatic), as well as in the development of small molecule inhibitors targeting HPV-specific mechanisms of viral oncogenesis.

1. Introduction

In 2022, cancers of the head and neck accounted for 950,000 new cases and 480,000 cancer deaths globally, making up approximately 5% of both new cases and deaths worldwide [1]. Malignancies of epithelial origin, specifically squamous cell carcinoma (SCC), comprise the majority of cancers identified at the major anatomical sites: oral cavity, pharynx (including the oropharynx, hypopharynx, and nasopharynx), larynx, and nasal cavity and paranasal sinuses (also called the sinonasal tract) [2,3,4]. For the purposes of clarity, most cancers of the nasopharynx are Ebstein–Barr virus (EBV)-associated, which is considered a distinct entity from most head and neck SCC (HNSCC) [5]. In the United States, nearly two-thirds of HNSCC cases are locally advanced or metastatic at diagnosis [6].
Traditionally, HNSCC has been considered a disease of older men, associated with major risk factors of heavy tobacco and alcohol use. The incidence of tobacco-associated cancers, including human papillomavirus (HPV)-negative HNSCC, has been decreasing in the United States, Europe, and other developed countries, whereas it continues to rise in developing countries, including much of Asia [4,7]. By contrast, particularly in developed countries, rates of human papillomavirus (HPV)-associated HNSCC have increased substantially over the last three decades. While HPV has been recognized as a potential etiologic factor in HNSCC since the 1980s, increased provider recognition, directed testing, and potential changes in oral sexual practices have led to a substantial increase in the proportion of head and neck cancers that are HPV-associated over this period [8,9,10]. Though a vaccine-preventable disease, public awareness of HPV as a risk factor for oropharyngeal cancer remains poor [11].
In this review, we will discuss HPV-associated HNSCC, focusing on the underlying mechanisms of HPV-associated carcinogenesis, the tumor microenvironment, and the host immune response as potential susceptibilities for therapeutic development. We will review the current standard of checkpoint inhibitor immunotherapy in the recurrent or metastatic setting, focusing on outcomes in HPV-associated disease. While there are presently no established immunotherapeutic strategies specifically for HPV-related SCC, we will discuss several emerging immunotherapeutic approaches (including vaccines and adoptive T-cell therapies) in clinical testing and end with commentary on future directions.

2. HPV-Associated Oropharyngeal Squamous Cell Carcinoma (OPSCC)

Infection with HPV accounted for approximately 30% of new oropharyngeal carcinoma cases worldwide in 2018 [12]. In recent decades, this HPV-attributable fraction has been consistently higher among high-income countries, including much of Europe and North America, compared to lower-income regions [12,13]. HPV detection in oropharyngeal cancers increased from 16 to 72% between the 1980s and early 2000s in the United States, and was reported at 52% in the United Kingdom between 2002 and 2011 [10,14]. While HNSCC of non-oropharyngeal sites can be associated with HPV infection at reported rates ranging from 2 to 24%, co-testing for p16 expression by immunohistochemistry (IHC) is less commonly performed, making causality and thus functional significance harder to assess [12,15,16]. Though there are over 230 fully characterized types of HPV, 14 are considered the highest risk for HPV-associated carcinogenesis [17,18,19]. HPV-16 is the highest risk mucosal subtype, identified in 85 to 90% of HPV-associated oropharyngeal squamous cell carcinomas (OPSCCs). Other high-risk HPV subtypes, including HPV-18 and HPV-33, are identified much less commonly, with typical frequencies of less than 1 or 2% [20]. Notably, this HPV distribution differs from cervical cancer, where approximately half of cases are attributed to HPV-16 [21].
The recognition of HPV as a key etiologic factor in OPSCC led to the observation of substantially better clinical outcomes among patients with HPV-associated disease compared to those whose cancers were HPV-negative [22,23]. Therefore, in 2017, the American Joint Committee on Cancer (AJCC) updated its eighth edition staging manual to include a separate staging system for HPV-associated OPSCC. This resulted in an overall downstaging of HPV-associated disease according to the TNM criteria relative to the previous seventh edition and relative to HPV-negative OPSCC [24,25]. HPV status does not affect staging or treatment decisions at non-oropharyngeal sites and may not be routinely assessed. Most patients with clinically localized HPV-associated OPSCC are treated with definitive concurrent chemoradiotherapy, though surgery or radiation alone are often used for small (cT1-2) tumors, particularly those that are node-negative (cN0) [2]. In aggregates, approximately two-thirds of patients with localized HNSCC will develop disease recurrence, typically within two years [3]. While outcomes are significantly better for HPV-associated disease, around 25 to 30% of patients will still recur [26,27]. For these cases of metastatic disease or recurrence which cannot be managed with salvage local therapies, the preferred first-line regimen is either pembrolizumab monotherapy (for PD-L1 CPS ≥ 1) or pembrolizumab with platinum-doublet chemotherapy (any PD-L1) based on KEYNOTE-048 [28]. While a small minority of patients experience prolonged disease control with checkpoint inhibitor immunotherapy, median PFS remains approximately one year, and options for subsequent systemic treatment remain an area of unmet need [29].

3. Molecular Biology of HPV-Associated Carcinogenesis

HPVs are non-enveloped, double-stranded circular deoxyribonucleic acid (DNA) viruses that infect the human squamous epithelium. The viral genome consists of approximately 8000 base pairs within which are three functional regions—a cluster of “early” viral genes (E1–E7) required for replication, a set of “late” genes (L1, L2) required for viral capsid formation, and a large non-coding long control region (LCR) [30,31]. Within the infected host cell, HPV E6 and E7 viral proteins are primarily implicated in carcinogenesis. Via ubiquitin-mediated processes, E6 and E7 function chiefly to induce degradation of the tumor suppressor proteins p53 and retinoblastoma (pRb), respectively [32]. Downstream of pRb degradation, E2F promotes the transcription of CDKN2A and consequently increases p16INK4a expression [33]. Overall, this leads to triggering cell cycle synthesis (S) phase re-entry on top of loss of p53-dependent cell cycle arrest, thereby promoting further viral DNA replication [34]. This simplified version of HPV pathogenesis illustrates the diagnostic tests utilized in clinical practice. IHC for p16 expression is most common due to its low cost and rapid turnaround, and the eighth edition of the AJCC staging system defines HPV association based on p16 status [24]. While highly sensitive for HPV-associated OPSCC, p16 IHC has only moderate specificity, leading to the recommendation that p16 IHC be combined with HPV DNA PCR to improve this [35]. While detection of E6/E7 mRNA is considered the gold standard diagnostic assay, it is infrequently used due to limited availability and cost [36].
In addition to the described mechanism via p53 and pRb, the E6 and E7 oncoproteins have other cellular protein targets which promote carcinogenesis. For example, E6 can lead to the degradation of PDZ domain-containing proteins, many of which function as tumor suppressors and have critical roles in signal transduction [37]. E6 has also been shown to promote NFX1-91 degradation, removing it from the hTERT promoter and leading to increased telomerase expression [38,39]. Class I histone deacetylases (HDACs) may be downstream effectors of E7 via E2F-induced transcription, leading to changes in chromatin remodeling and gene expression [37,40,41].
The E1 and E2 viral proteins are involved in viral replication and gene expression. E1 is the only HPV viral protein product with direct enzymatic function and acts as a DNA helicase [42]. E2 is critical for the regulation of viral gene transcription, as well as the segregation of viral plasmids during cell division. In HPV-associated malignancies, the viral genome may be episomal and/or integrated into the host cell genome. Integration often occurs in the E1 or E2 region and can lead to loss of negative feedback from the E2 protein [43]. HPV integration may be associated with inferior prognosis and more aggressive clinical phenotypes in HNSCC; however, conflicting studies have also been reported, perhaps a consequence of variable detection methods to identify integration events [43,44,45,46].
The overall genomic landscape of HPV-associated HNSCC has been compared to that of its HPV-negative counterpart. HPV-associated tumors are consistently associated with mutations in PIK3CA, with mutations in DDX3X, FGFR2/3, and KRAS variably reported. In contrast, HPV-negative tumors are enriched for mutations in TP53 and CDKN2A and more frequently exhibit amplifications in EGFR, CCND1, and FGFR1 [47,48,49,50]. Overall tumor mutational burden (TMB) is comparable between HPV-associated and HPV-negative HNSCC, though TMB is higher among HPV-associated tumors in patients with significant smoking history (≥10 pack-years) [48,50].

4. Immune System Evasion and Tumor Microenvironment in HPV-Associated HNSCC

HPVs have developed a number of mechanisms mediating immune evasion by HPV-associated malignancies. T-cell activation relies on intact antigen presentation by antigen-presenting cells (APCs) complexed with major histocompatibility complex (MHC) class I and II molecules, the human version of which is the human leukocyte antigen (HLA) system [51]. A large-scale genome-wide association study (GWAS) has identified multiple HLA loci associated with oropharyngeal cancer risk, with statistical significance among only the HPV-associated cancers [52]. In vitro, both the HPV-16 and HPV-18 E7 proteins lead to repression of the MHC class I heavy chain promoter [53]. The HPV E5 protein has been shown to downregulate peptide-bound MHC molecules via the alkalinization of late endosomes, thereby limiting the activation of antitumor T-cells [54,55]. In particular, HPV selectively downregulates HLA-A, HLB-B, and HLA-C molecules but spares the nonclassical HLA-E molecule, functioning as an inhibitory signal for cytotoxic natural killer (NK) cell activity [54].
The cGAS-STING pathway is a conserved component of the innate immune system that senses cytosolic DNA and serves as a critical defense mechanism against DNA viruses [56]. The activation of this pathway by viral antigens leads to a cytokine milieu and tumor microenvironment (TME) favoring a cytotoxic T-cell response and viral clearance [57]. Though much of the data remain preclinical, HPV E7 has been shown to downregulate the cGAS-STING pathway though various mechanisms [58,59].
Expression of the PD-L1 immune checkpoint protein has been reported at higher frequencies in patients with HPV-associated compared to HPV-negative tumors [60,61]. However, large cohort analyses have demonstrated that HPV association predicts better response to checkpoint inhibitor immunotherapy independent of PD-L1 status [62]. These data, combined with other studies looking at concomitant tumor-infiltrating lymphocyte (TIL) infiltrate and gene expression profiles, suggests that PD-L1 expression may be a proxy for a generally inflamed TME rather than a direct effect of HPV [57,60].
In spite of these mechanisms of immune evasion, there are a number of HPV-specific immune responses, many of which are associated with prognostic significance. HPV-16 E6- and E7-specific T-cells have been identified in both tumor and blood samples of patients with HPV-16-associated HNSCC [63,64]. These TILs are skewed towards CD4+ T-cells with an antitumor T helper (TH)1/TH17 profile [63]. More generally, higher levels of TILs have been demonstrated in HPV-associated disease compared with HPV-negative HNSCC and are correlated with better clinical response [65,66,67]. Furthermore, high numbers of CD4+ FOXP3+ regulatory T-cells have been identified in HNSCC and are associated with inferior outcomes, though the relative prevalence in HPV-associated versus HPV-negative disease is presently unclear [64]. Factors involving other immune cell types, including lower CD20+ B-cells, lower dendritic cell (DC)-like macrophages, higher tumor associated macrophages (TAMs), and higher myeloid-derived suppressor cells (MDSCs), have also been associated with inferior clinical outcomes but are not necessarily HPV-specific [63,68,69,70,71,72,73].

5. Checkpoint Inhibitor Immunotherapy

Given this background, there is biologic rationale for checkpoint inhibitor immunotherapy in HPV-associated HNSCC. In reality, PD-(L)1 inhibitors have largely been studied in HPV-agnostic cohorts, with only secondary analyses of efficacy by HPV status. KEYNOTE-048 was the practice changing, randomized phase III trial investigating pembrolizumab alone or with chemotherapy versus the established EXTREME regimen of cetuximab plus chemotherapy in the front-line, recurrent, or metastatic settings. Overall survival (OS), but not PFS or objective response rate (ORR), was improved with pembrolizumab alone in the PD-L1 CPS ≥ 1 population (12.3 vs. 10.3 months, p = 0.009) and for pembrolizumab plus chemotherapy in all comers (13.0 vs. 10.7 months, p = 0.003), both in comparison to cetuximab plus chemotherapy. Of the 447 patients with oropharyngeal primary tumors, 56% were p16-positive by IHC [28]. In the four-year follow-up, the magnitude of OS benefit appeared similar between the p16-positive and p16-negative subgroups [29].
A number of studies investigating PD-(L)1 inhibition in recurrent or metastatic HPV-associated OPSCC after progression on platinum-based regimens report numerically improved response rates or overall survival for HPV-associated cancers compared to HPV-negative cancers [74,75,76,77,78]. Other studies have not reported such differences [79,80]. However, these trends should be interpreted cautiously, as many of these analyses report point estimates from small subgroups, leading to wide confidence intervals and an inability to demonstrate statistical significance. Study design varied across trials, with specific inclusion/exclusion criteria and variation in PD-L1 expression status potentially leading to differential treatment response. Furthermore, a pooled analysis of nine clinical trials of PD-(L)1 inhibitor monotherapy in advanced HNSCC demonstrated numerically higher but not statistically significant ORR (18.8 vs. 12.2%) and DCR (42.8 vs. 34.4%) for HPV-associated disease compared to HPV-negative disease [81]. Ultimately, PD-(L)1 inhibition has demonstrated benefit in both entities, which are biologically and molecularly distinct, as previously emphasized.
Antibodies targeting immune checkpoints other than PD-(L)1 have been studied in basket trials of advanced solid tumors and dedicated cohorts of HNSCC. The PD-L1 inhibitor durvalumab in combination with the CTLA-4 inhibitor tremelimumab has been studied in both the first-(CONDOR, EAGLE) and subsequent-line settings (KESTREL) for advanced HNSCC, with no differences in efficacy compared to standard therapy and no evident difference by HPV status [82,83,84]. Other immune checkpoints including TIM-3, TIGIT, LAG-3, and VISTA are areas of active investigation, recently reviewed by Struckmeier et al. [85]. Data from these studies remain preliminary, oftentimes with monoclonal antibodies given in combination with PD-(L)1 inhibitor immunotherapy or as bispecific antibodies that target both PD-(L)1 and the other immune checkpoint [85]. Whether these novel checkpoint inhibition strategies have differential efficacy by HPV status has yet to be determined.
EGFR inhibitors (namely cetuximab) have also been studied in combination with the PD-1 inhibitors pembrolizumab or nivolumab. A recent meta-analysis included 118 patients with advanced HNSCC receiving combination therapy and 684 patients receiving PD-1 inhibitor monotherapy. Pooled ORR and OS were significantly improved for combination cetuximab and PD-1 inhibition compared to PD-1 inhibitor monotherapy in the HPV-negative subgroup (ORR 46 vs. 15%, p < 0.001; 1-year OS rate 59 vs. 36%, p < 0.001), but not in those with HPV-associated disease (ORR 18 vs. 17%, p = 0.686; 1-year OS rate 55 vs. 40%, p = 0.252) [86]. While these results require further validation in prospective studies, the aforementioned enrichment of EGFR pathway activation in HPV-negative HNSCC lends biological credence to this observation [48,87].

6. Therapeutic HPV Vaccines

While prophylactic vaccines derived from viral L1 proteins are widely recommended for the prevention of HPV-associated malignancy, different targets are needed for therapeutic vaccination [88,89]. In general, the goal of therapeutic vaccination is to stimulate an adaptive immune response capable of inducing cytotoxic activity with sufficient sensitivity and specificity to allow for tumor control while minimizing adverse effects. HPV oncoproteins E6 and E7 have long been considered ideal target “non-self” antigens because they are exclusively produced by cancer cells, constitutively expressed, and persistently necessary for oncogenesis [90,91,92]. Several cancer vaccine platforms have emerged in the HPV-associated HNSCC space, broadly divided into molecular vaccines (DNA-, RNA-, or peptide-based) and viral or bacterial vector-based vaccines [92]. All of these strategies are designed to induce a host immune response against the target antigen and thus the cancer cell, but do not directly interfere with E6 or E7 function.
DNA vaccines on their own are poorly immunogenic in humans; therefore, many constructs incorporate cytokine coding genes or are co-administered with anti-PD-(L)1 immunotherapy [93]. MEDI0457 is a DNA vaccine encoding the E6 and E7 antigens from HPV-16 and 18, administered together with a plasmid encoding an IL-12 adjuvant. As a monotherapy, MEDI0457 was studied in locally advanced, p16-positive HNSCC. This study demonstrated antigen-specific T-cell activity and cellular responses exceeding one year but was not designed to evaluate clinical efficacy [94]. MEDI0457 was subsequently studied in combination with the PD-L1 inhibitor durvalumab in a Phase Ib/IIa trial, including 29 evaluable patients with previously treated HPV-associated HNSCC who were immunotherapy-naïve. ORR was 27.6% and median PFS 3.5 months; however, the study did not reach its primary efficacy endpoint of ORR (lower bound of 95% confidence interval excludes ORR H0 ≤ 15%), likely affected by poor enrollment [95]. AMV002 is a DNA vaccine composed of two plasmids encoding two variants of a fusion protein of E6 and E7, demonstrating enhanced E6- or E7-specific cell-mediated responses in the phase I dose escalation study [96]. In the subsequent phase Ib study in recurrent/metastatic HPV-associated OPSCC, AMV002 with durvalumab was well-tolerated, with no grade 3 or higher vaccine-related adverse events (AEs). There was a single complete response (ORR 8%); however, most patients (83%) in this study were immunotherapy-naïve and the prior treatments and PD-L1 status of the single responding patient were not reported [97]. At present, there are no ongoing studies of either MEDI0457 or AMV002 in HPV-associated HNSCC.
Technologic advancements improving mRNA delivery and stability have made therapeutic RNA vaccines an area of developing interest. BNT113 is a liposome-encapsulated mRNA vaccine encoding for HPV-16 E6 and E7. In the phase I/II study of BNT113 monotherapy across HPV-16-positive cancers of multiple subtypes (including three HNSCCs), disease control was observed in 5/7 and cellular immune responses against E6 or E7 in 3/7 evaluable patients with advanced disease. No objective responses were reported [98]. Preliminary results from the safety run-in phase of the phase II BNT111-01 trial, studying BNT113 plus pembrolizumab as a first-line treatment for recurrent/metastatic HPV-16-positive, PD-L1-positive (CPS ≥ 1) HNSCC have been reported. Among 15 patients, ORR was 40% and median PFS 3.9 months, with treatment-related AEs mainly grade 1–3 flu-like symptoms. The randomized portion of this study (AHEAD-MERIT) is ongoing, comparing the combination to pembrolizumab monotherapy (NCT04534205) [99].
Peptide-based vaccines consist of amino acid sequences containing immunogenic epitopes of the target of interest, typically delivered with a concomitant adjuvant. PDS0101 is a peptide vaccine containing six epitopes of HPV-16 E6 and E7, co-administered with the immune-activating cationic lipid R-DOTAP [100]. In a single-arm study of PDS0101 and pembrolizumab in the first-line setting for PD-L1-positive, recurrent/metastatic HPV-16-positive HNSCC (n = 53), ORR was 34% and median PFS 6.3 months [101]. In a separate cohort of checkpoint inhibitor-refractory patients (n = 21), no responses were observed [102]. The phase III, randomized VERSATILE-003 study is ongoing, comparing PDS0101 plus pembrolizumab to pembrolizumab monotherapy in the first-line, recurrent/metastatic, PD-L1-positive population (NCT06790966). PDS0101 is also being studied in combination with M9241 (an immunocytokine composed of IL-12 heterodimers) and bintrafusp alfa (a TGF-β “trap”/anti-PD-L1 fusion protein) in advanced HPV-16-positive malignancies (including 13 OPSCC). Interim results show promise, with ORR 88% (7/8) in checkpoint inhibitor-naïve disease and ORR 27% (6/22) in checkpoint-refractory disease [103,104]. ISA101 is composed of overlapping peptide sequences covering the complete HPV-16 E6 and E7 proteins [105]. Building upon single-agent data in cervical cancer, the phase II study of ISA101 with nivolumab enrolled 24 patients with advanced HPV-16-positive malignancy, 22 of whom had OPSCC. ORR was 36% and median PFS 2.5 months, noting that all patients were immunotherapy-naïve [106]. ISA101b is the subsequent iteration of this vaccine, which was studied in the phase II OpcemISA study, randomizing patients with recurrent/metastatic HPV-16- and PD-L1-positive OPSCC (n = 198) to either ISA101b plus cemiplimab or cemiplimab monotherapy. The primary outcome of ORR was not different between the arms (25% for combination vs. 23% for control), though an exploratory per protocol analysis among patients with PD-L1 CPS ≥ 20 suggested potential benefit among this subgroup [107].
While live vector-based vaccines are highly immunogenic, the potential safety risks in immunocompromised patients and the induction of neutralizing vector-specific antibodies are common areas of concern [108]. ADSX11-001 is a live Listeria monocytogenes vector vaccine engineered to secrete the HPV-16 E7 protein fused to an attenuated Listeria virulence factor [109]. A phase I trial of ADSX11-001 in patients with locally advanced HPV-16-positive OPSCC (NCT01598792) was complicated by a case of systemic listeriosis in the second patient enrolled, occurring within 24 h of the first vaccine administration [110]. The trial was paused and ultimately terminated following this severe AE. A separate study in OPSCC prior to robotic surgery reported a few HPV-specific T-cell responses, but results were limited by poor accrual (NCT02002182). HB-200 is a viral vector vaccine composed of two replication-attenuated arenavirus vectors derived from the lymphocytic choriomeningitis (HB-201) and Pichinde viruses (HB-202). It expresses an HPV-16 E7E6 fusion protein and as monotherapy has been shown to induce HPV-16 tumor-specific T-cell responses in heavily pretreated patients [111]. HB-200 has been studied in combination with pembrolizumab as a first-line treatment for HPV-16- and PD-L1-positive recurrent/metastatic HNSCC (n = 42), most cases of which were checkpoint inhibitor-naïve (95%). ORR was 43% overall and 59% among PD-L1 CPS ≥ 20 [112]. This study is ongoing and includes a separate cohort for subsequent-line treatment (NCT04180215). PRGN-2009 is a gorilla adenovirus vector vaccine containing multiple epitopes of HPV-16 and 18 E6 and E7, as well as additional T-cell enhancer agonist epitopes [113]. In advanced HPV-16 and 18 cancers, the combination of PRGN-2009 and bintrafusp alfa led to objective response in 3 of 10 evaluable patients (ORR 30%), two of whom were previously refractory to checkpoint inhibitors [114]. PRGN-2009 is also being studied in the neoadjuvant/induction setting, either in combination with chemotherapy or with checkpoint inhibitor immunotherapy (NCT06223568, NCT05996523).
Despite different vaccine platforms, all the aforementioned strategies target HPV E6 and/or E7. Multiple independent groups have demonstrated HPV-specific T-cell responses against epitopes derived from HPV E2 and E5 proteins (in addition to E6 and E7), providing other candidate targets for therapeutic HPV vaccination [115,116,117]. Furthermore, whether personalized vaccines directed against a patient’s tumor-specific antigens could be more effective is unknown, though the appeal of such a strategy may be higher in HPV-negative HNSCC where conserved viral targets do not exist [118]. Table 1 summarizes ongoing studies of therapeutic vaccines in HPV-associated HNSCC.

7. Adoptive Cell Therapies

The principle of adoptive cell therapy (ACT) has existed since the 1980s, when the first trials of TILs were first conducted in melanoma [119]. Broadly, the principle of these therapies involves the extraction and expansion of immune cells ex vivo, preparative lymphodepleting chemotherapy to facilitate engraftment, and finally adoptive cell transfer into the patient, with the intent of subsequent in vivo persistence [120,121]. Thus far, most of these therapies involve the manipulation of autologous T lymphocytes, consisting of genetically modified T-cell receptors (TCRs), chimeric antigen receptor (CAR) T-cell therapy, and TILs.
Engineered TCRs involve the modification of a patient’s T-cells to express a TCR recognizing a specific target antigen on cancer cells. As the TCR is engineered to recognize tumor antigens presented by specific MHC molecules, these therapies are HLA-specific, with development thus far largely restricted to HLA-A*02:01. Genetically modified TCRs can be used for both intracellular and cell surface targets, whereas CAR T-cell therapy is restricted to surface antigens because of direct antigen recognition and binding [122]. As with previously discussed vaccination strategies, the intracellular HPV E6 and E7 proteins have been the most common candidate targets, specifically for engineered TCRs. In contrast, the identification of specific and recurrent cell surface antigens for CAR T-cell targeting has thus far limited the development of these therapies specifically for HPV-associated malignancies [57]. Lastly, autologous TIL therapy involves the ex vivo expansion of a heterogenous lymphocyte population, typically derived from a surgical resection specimen. While additional ex vivo selection and genetic modification can be performed, TILs are inherently multi-targeted and therefore less suitable for ubiquitously expressed viral antigens [123].
Only a few clinical studies of ACT in HPV-associated HNSCC have thus far been reported. A phase I/II trial enrolled 12 patients with metastatic HPV-16-positive solid tumors, prior platinum-based chemotherapy, and HLA-A*02:01, including one patient with OPSCC. Patients received lymphodepleting chemotherapy followed by autologous engineered TCRs against HPV-16 E6 with adjuvant high-dose IL-2. Two objective responses were observed at the highest dose level, and no dose-limiting toxicities (DLTs) were reported in the phase I portion. The loss of HLA-A*02:01 and frameshift deletion of the interferon gamma receptor 1 were demonstrated as two potential resistance mechanisms in patients who did not respond to this therapy [124]. Potentially more promising was a phase I study of E7 TCR T-cells in a similar patient population, conducted by the same group. This product resulted in objective responses in 6 of 12 patients, including four of eight with anti-PD-1 refractory disease and two of four with primary head and neck tumors [125]. The phase II component of this study is ongoing (NCT02858310). In general, most ongoing TCR studies focus on E7 TCR T-cells, which in preclinical models have demonstrated higher functional avidity and improved effector function compared to E6 TCR T-cells [126]. Multiple studies of CAR T-cell therapy (and at least one study of CAR NK-cell therapy) are ongoing in HNSCC, though they are not HPV-specific. A phase II trial of the TIL therapy lifileucel (LN-145) has been completed though not published, with an ORR of only 11% among 53 evaluable patients with recurrent or metastatic HNSCC (NCT03083873). The combination of lifileucel with pembrolizumab appears more promising based on preliminary results (ORR 44% among nine patients with HNSCC); however, this study is ongoing (NCT03645928) [127]. Another autologous TIL (TBio-4101) was previously studied with pembrolizumab in advanced solid tumors, including HNSCC with only piecemeal reporting of results; however, as of early 2025, it is no longer being developed (NCT05576077) [128,129,130]. Table 2 summarizes the ongoing studies of ACT in HPV-associated HNSCC.

8. Peptide–HLA-Based Immunotherapeutic Platform

A number of manufacturing, safety, and logistical considerations represent potential limitations to current ACTs. In this context, the Immuno-STATTM (Selective Targeting and Alteration of T-cells) framework is an off-the-shelf, peptide–HLA-based immunotherapeutic platform designed to activate antitumor T-cells in vivo in an APC-independent manner. These constructs consist of a covalent fusion of a MHC class I allele, target peptide epitope, co-modulator(s), and an Fc domain [131]. Specifically, CUE-101 is an Immuno-STATTM construct being actively investigated in HPV-16-positive HNSCC, consisting of an HLA complex (HLA-A*0201), an HPV-16 E7 peptide epitope, and four attenuated IL-2 molecules [132]. Interim results from an ongoing phase I study in HPV-16-positive recurrent/metastatic HNSCC demonstrated an ORR of 47% and median PFS of 5.8 months for the combination of CUE-101 and pembrolizumab among 19 evaluable PD-L1-positive patients in the first-line setting. Among 19 evaluable patients receiving CUE-101 monotherapy in the subsequent-line setting after previous first-line platinum-based chemotherapy and/or pembrolizumab, there was a single partial response (ORR 5%). CUE-101 appeared well tolerated both as a monotherapy and in combination with pembrolizumab, with 92% of AEs of grade 2 or less [133].

9. Conclusions and Future Directions

The underlying molecular biology, TME, and host immune response in HPV-associated HNSCC offers several potential vulnerabilities for therapeutic targeting. Checkpoint inhibitor immunotherapy, specifically PD-1 inhibition, has been standardly applied to advanced HNSCC regardless of HPV status. Yet only a subset of patients derives durable benefit, and there is strong preclinical evidence to support immune evasion and local immunosuppression in HPV-related oncogenesis. Therapeutic vaccines, TCR T-cell therapies, and peptide–HLA-based constructs represent emerging immunotherapeutic strategies in this space, nearly all of which target HPV E6 or E7 oncoproteins. Response rates to these therapies have been modest thus far, though they appear more promising when combined with checkpoint inhibitor immunotherapy. The restriction of TCR T-cell therapies and peptide–HLA-based constructs to a few HLA alleles limits the candidate population, and further understanding of resistance via genetic alterations in antigen processing is needed.
Much of our understanding of oncogenesis in HPV-associated HNSCC is derived from primary tumors in patients with localized disease. Several recent studies have identified molecular alterations characteristic of recurrent but not primary tumors, some of which overlap with HPV-negative HNSCC [134,135,136]. Nearly all the aforementioned novel immunotherapies are primarily being studied in the recurrent or metastatic setting. Whether recurrent disease relies upon the same viral oncogenic machinery and immune environment as in localized disease requires further study, which may help guide the appropriate setting in which to apply these therapies [137].
Despite our detailed understanding of the essential HPV-associated oncoproteins, no small molecule inhibitors have thus far emerged as potential therapies in the clinic. Limitations to this development have included the small molecular size of these proteins, their structural similarity to functional domains of proteins required for normal function, and the lack of enzymatic activity (other than E1) targeted by currently available antiviral agents [90,138]. Emerging strategies with early in vitro data from cervical cancer cell lines include the prevention of downstream tumor suppressor degradation via targeted disruption of viral oncoprotein binding (e.g., between E6/p53 or E7/PTPN14) and targeted HPV oncoprotein degradation via proteolysis-targeting chimera (PROTAC) technology [138,139,140,141]. Other candidate strategies with preclinical evidence includes siRNA and short hairpin RNA (shRNA) to silence E6, E7, or the LCR of the HPV genome, CRISPR/Cas9 engineering to target HPV E6 or E7, and a small molecule compound capable of inhibiting the ATPase activity of the HPV E1 helicase [142,143,144,145,146,147,148]. Despite these emerging avenues of development, further translational efforts are needed to therapeutically exploit the viral machinery of HPV-associated HNSCC, as well as other virus-driven malignancies.

Author Contributions

Conceptualization, F.S. and A.D.C.; writing—original draft preparation, F.S.; writing—review and editing, F.S. and A.D.C.; supervision, A.D.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

F.S. has received honoraria from MJH Life Sciences and served on advisory boards for OncoHost and Tempus. A.D.C. has received institutional clinical trial support from Coherus.

Abbreviations

The following abbreviations are used in this manuscript:
ACTAdoptive cell therapy
AEAdverse event
AJCCAmerican Joint Committee on Cancer
APCAntigen-presenting cell
CARChimeric antigen receptor
CPSCombined positive score
CTLA-4Cytotoxic T-lymphocyte-associated protein 4
DCDendritic cell
DLTDose-limiting toxicity
DNADeoxyribonucleic acid
EBVEbstein–Barr virus
EGFREpidermal growth factor receptor
FcFragment crystallizable
GWASGenome-wide association study
HDACHistone deacetylase
HLAHuman leukocyte antigen
HNSCCHead and neck squamous cell carcinoma
HPVHuman papillomavirus
IHCImmunohistochemistry
IL-2Interleukin-2
LAG-3Lymphocyte activation gene-3
LCRLong control region
MAGEMelanoma antigen gene
MDPIMultidisciplinary Digital Publishing Institute
MDSCMyeloid-derived suppressor cell
MHCMajor histocompatibility complex
mRNAMessenger ribonucleic acid
NKNatural killer
NSCLCNon-small cell lung cancer
OPSCCOropharyngeal squamous cell carcinoma
ORRObjective response rate
OSOverall survival
pCRPathologic complete response
PD-1Programmed cell death protein 1
PD-L1Programmed cell death ligand 1
PFSProgression-free survival
PRAMEPreferentially expressed antigen in melanoma
PROTACProteolysis-targeting chimera
RbRetinoblastoma
RNARibonucleic acid
RP2DRecommended phase II dose
SCCSquamous cell carcinoma
shRNAShort hairpin ribonucleic acid
siRNASmall interfering ribonucleic acid
STATSelective targeting and alteration of T-cells
TAMTumor-associated macrophage
TCRT-cell receptor
TGF-βTransforming growth factor-beta
THT helper
TIGITT-cell immunoreceptor with immunoglobulin and ITIM domain
TILTumor-infiltrating lymphocyte
TIM-3T-cell immunoglobulin and mucin-domain containing-3
TMBTumor mutational burden
VISTAV-domain immunoglobulin suppressor of T-cell activation

References

  1. 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. [Google Scholar] [CrossRef] [PubMed]
  2. Chow, L.Q.M. Head and Neck Cancer. N. Engl. J. Med. 2020, 382, 60–72. [Google Scholar] [CrossRef] [PubMed]
  3. Argiris, A.; Karamouzis, M.V.; Raben, D.; Ferris, R.L. Head and neck cancer. Lancet 2008, 371, 1695–1709. [Google Scholar] [CrossRef] [PubMed]
  4. Cohen, N.; Fedewa, S.; Chen, A.Y. Epidemiology and Demographics of the Head and Neck Cancer Population. Oral. Maxillofac. Surg. Clin. N. Am. 2018, 30, 381–395. [Google Scholar] [CrossRef]
  5. Wei, W.I.; Sham, J.S. Nasopharyngeal carcinoma. Lancet 2005, 365, 2041–2054. [Google Scholar] [CrossRef]
  6. Miller, K.D.; Nogueira, L.; Mariotto, A.B.; Rowland, J.H.; Yabroff, K.R.; Alfano, C.M.; Jemal, A.; Kramer, J.L.; Siegel, R.L. Cancer treatment and survivorship statistics, 2019. CA Cancer J. Clin. 2019, 69, 363–385. [Google Scholar] [CrossRef]
  7. Barsouk, A.; Aluru, J.S.; Rawla, P.; Saginala, K.; Barsouk, A. Epidemiology, Risk Factors, and Prevention of Head and Neck Squamous Cell Carcinoma. Med. Sci. 2023, 11, 42. [Google Scholar] [CrossRef]
  8. Syrjanen, K.; Syrjanen, S.; Lamberg, M.; Pyrhonen, S.; Nuutinen, J. Morphological and immunohistochemical evidence suggesting human papillomavirus (HPV) involvement in oral squamous cell carcinogenesis. Int. J. Oral. Surg. 1983, 12, 418–424. [Google Scholar] [CrossRef]
  9. Gillison, M.L.; Chaturvedi, A.K.; Anderson, W.F.; Fakhry, C. Epidemiology of Human Papillomavirus-Positive Head and Neck Squamous Cell Carcinoma. J. Clin. Oncol. 2015, 33, 3235–3242. [Google Scholar] [CrossRef]
  10. Chaturvedi, A.K.; Engels, E.A.; Pfeiffer, R.M.; Hernandez, B.Y.; Xiao, W.; Kim, E.; Jiang, B.; Goodman, M.T.; Sibug-Saber, M.; Cozen, W.; et al. Human papillomavirus and rising oropharyngeal cancer incidence in the United States. J. Clin. Oncol. 2011, 29, 4294–4301. [Google Scholar] [CrossRef]
  11. Lechner, M.; Masterson, L.; Mermelstein, S.; Liu, J.; Rehman, U.; Chen, M.; O’Mahoney, J.; Holsinger, F.C. Oropharyngeal cancer: Lack of human papillomavirus awareness and economic burden in the United States. Clin. Transl. Med. 2024, 14, e70062. [Google Scholar] [CrossRef] [PubMed]
  12. de Martel, C.; Georges, D.; Bray, F.; Ferlay, J.; Clifford, G.M. Global burden of cancer attributable to infections in 2018: A worldwide incidence analysis. Lancet Glob. Health 2020, 8, e180–e190. [Google Scholar] [CrossRef] [PubMed]
  13. de Martel, C.; Ferlay, J.; Franceschi, S.; Vignat, J.; Bray, F.; Forman, D.; Plummer, M. Global burden of cancers attributable to infections in 2008: A review and synthetic analysis. Lancet Oncol. 2012, 13, 607–615. [Google Scholar] [CrossRef] [PubMed]
  14. Schache, A.G.; Powell, N.G.; Cuschieri, K.S.; Robinson, M.; Leary, S.; Mehanna, H.; Rapozo, D.; Long, A.; Cubie, H.; Junor, E.; et al. HPV-Related Oropharynx Cancer in the United Kingdom: An Evolution in the Understanding of Disease Etiology. Cancer Res. 2016, 76, 6598–6606. [Google Scholar] [CrossRef]
  15. Abogunrin, S.; Di Tanna, G.L.; Keeping, S.; Carroll, S.; Iheanacho, I. Prevalence of human papillomavirus in head and neck cancers in European populations: A meta-analysis. BMC Cancer 2014, 14, 968. [Google Scholar] [CrossRef]
  16. Mehanna, H.; Beech, T.; Nicholson, T.; El-Hariry, I.; McConkey, C.; Paleri, V.; Roberts, S. Prevalence of human papillomavirus in oropharyngeal and nonoropharyngeal head and neck cancer—Systematic review and meta-analysis of trends by time and region. Head Neck 2013, 35, 747–755. [Google Scholar] [CrossRef]
  17. Abate, A.; Munshea, A.; Nibret, E.; Alemayehu, D.H.; Alemu, A.; Abdissa, A.; Mihret, A.; Abebe, M.; Mulu, A. Characterization of human papillomavirus genotypes and their coverage in vaccine delivered to Ethiopian women. Sci. Rep. 2024, 14, 7976. [Google Scholar] [CrossRef]
  18. Correa, R.M.; Baena, A.; Valls, J.; Colucci, M.C.; Mendoza, L.; Rol, M.; Wiesner, C.; Ferrera, A.; Fellner, M.D.; Gonzalez, J.V.; et al. Distribution of human papillomavirus genotypes by severity of cervical lesions in HPV screened positive women from the ESTAMPA study in Latin America. PLoS ONE 2022, 17, e0272205. [Google Scholar] [CrossRef]
  19. IARC. Working Group on the Evaluation of Carcinogenic Risks to Humans: Human Papillomaviruses; World Health Organization: Geneva, Switzerland, 2007. [Google Scholar]
  20. Kreimer, A.R.; Clifford, G.M.; Boyle, P.; Franceschi, S. Human papillomavirus types in head and neck squamous cell carcinomas worldwide: A systematic review. Cancer Epidemiol. Biomarkers Prev. 2005, 14, 467–475. [Google Scholar] [CrossRef]
  21. Clifford, G.M.; Smith, J.S.; Plummer, M.; Munoz, N.; Franceschi, S. Human papillomavirus types in invasive cervical cancer worldwide: A meta-analysis. Br. J. Cancer 2003, 88, 63–73. [Google Scholar] [CrossRef]
  22. Nguyen-Tan, P.F.; Zhang, Q.; Ang, K.K.; Weber, R.S.; Rosenthal, D.I.; Soulieres, D.; Kim, H.; Silverman, C.; Raben, A.; Galloway, T.J.; et al. Randomized phase III trial to test accelerated versus standard fractionation in combination with concurrent cisplatin for head and neck carcinomas in the Radiation Therapy Oncology Group 0129 trial: Long-term report of efficacy and toxicity. J. Clin. Oncol. 2014, 32, 3858–3866. [Google Scholar] [CrossRef] [PubMed]
  23. Huang, S.H.; Xu, W.; Waldron, J.; Siu, L.; Shen, X.; Tong, L.; Ringash, J.; Bayley, A.; Kim, J.; Hope, A.; et al. Refining American Joint Committee on Cancer/Union for International Cancer Control TNM stage and prognostic groups for human papillomavirus-related oropharyngeal carcinomas. J. Clin. Oncol. 2015, 33, 836–845. [Google Scholar] [CrossRef] [PubMed]
  24. Amin, M.B.; Greene, F.L.; Edge, S.B.; Compton, C.C.; Gershenwald, J.E.; Brookland, R.K.; Meyer, L.; Gress, D.M.; Byrd, D.R.; Winchester, D.P. The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more “personalized” approach to cancer staging. CA Cancer J. Clin. 2017, 67, 93–99. [Google Scholar] [CrossRef] [PubMed]
  25. Lydiatt, W.M.; Patel, S.G.; O’Sullivan, B.; Brandwein, M.S.; Ridge, J.A.; Migliacci, J.C.; Loomis, A.M.; Shah, J.P. Head and Neck cancers-major changes in the American Joint Committee on cancer eighth edition cancer staging manual. CA Cancer J. Clin. 2017, 67, 122–137. [Google Scholar] [CrossRef]
  26. Ang, K.K.; Harris, J.; Wheeler, R.; Weber, R.; Rosenthal, D.I.; Nguyen-Tan, P.F.; Westra, W.H.; Chung, C.H.; Jordan, R.C.; Lu, C.; et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N. Engl. J. Med. 2010, 363, 24–35. [Google Scholar] [CrossRef]
  27. Ang, K.K.; Zhang, Q.; Rosenthal, D.I.; Nguyen-Tan, P.F.; Sherman, E.J.; Weber, R.S.; Galvin, J.M.; Bonner, J.A.; Harris, J.; El-Naggar, A.K.; et al. Randomized phase III trial of concurrent accelerated radiation plus cisplatin with or without cetuximab for stage III to IV head and neck carcinoma: RTOG 0522. J. Clin. Oncol. 2014, 32, 2940–2950. [Google Scholar] [CrossRef]
  28. Burtness, B.; Harrington, K.J.; Greil, R.; Soulieres, D.; Tahara, M.; de Castro, G., Jr.; Psyrri, A.; Baste, N.; Neupane, P.; Bratland, A.; et al. 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. [Google Scholar] [CrossRef]
  29. Harrington, K.J.; Burtness, B.; Greil, R.; Soulieres, D.; Tahara, M.; de Castro, G., Jr.; Psyrri, A.; Brana, I.; Baste, N.; Neupane, P.; et al. Pembrolizumab with or Without Chemotherapy in Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma: Updated Results of the Phase III KEYNOTE-048 Study. J. Clin. Oncol. 2023, 41, 790–802. [Google Scholar] [CrossRef]
  30. Rampias, T.; Sasaki, C.; Psyrri, A. Molecular mechanisms of HPV induced carcinogenesis in head and neck. Oral. Oncol. 2014, 50, 356–363. [Google Scholar] [CrossRef]
  31. Fehrmann, F.; Laimins, L.A. Human papillomaviruses: Targeting differentiating epithelial cells for malignant transformation. Oncogene 2003, 22, 5201–5207. [Google Scholar] [CrossRef]
  32. Doorbar, J. The papillomavirus life cycle. J. Clin. Virol. 2005, 32 (Suppl. S1), S7–S15. [Google Scholar] [CrossRef] [PubMed]
  33. Sherr, C.J.; Roberts, J.M. CDK inhibitors: Positive and negative regulators of G1-phase progression. Genes. Dev. 1999, 13, 1501–1512. [Google Scholar] [CrossRef] [PubMed]
  34. Hebner, C.M.; Laimins, L.A. Human papillomaviruses: Basic mechanisms of pathogenesis and oncogenicity. Rev. Med. Virol. 2006, 16, 83–97. [Google Scholar] [CrossRef] [PubMed]
  35. Prigge, E.S.; Arbyn, M.; von Knebel Doeberitz, M.; Reuschenbach, M. Diagnostic accuracy of p16(INK4a) immunohistochemistry in oropharyngeal squamous cell carcinomas: A systematic review and meta-analysis. Int. J. Cancer 2017, 140, 1186–1198. [Google Scholar] [CrossRef]
  36. Schache, A.G.; Liloglou, T.; Risk, J.M.; Jones, T.M.; Ma, X.J.; Wang, H.; Bui, S.; Luo, Y.; Sloan, P.; Shaw, R.J.; et al. Validation of a novel diagnostic standard in HPV-positive oropharyngeal squamous cell carcinoma. Br. J. Cancer 2013, 108, 1332–1339. [Google Scholar] [CrossRef]
  37. Wise-Draper, T.M.; Wells, S.I. Papillomavirus E6 and E7 proteins and their cellular targets. Front. Biosci. 2008, 13, 1003–1017. [Google Scholar] [CrossRef]
  38. Gewin, L.; Myers, H.; Kiyono, T.; Galloway, D.A. Identification of a novel telomerase repressor that interacts with the human papillomavirus type-16 E6/E6-AP complex. Genes Dev. 2004, 18, 2269–2282. [Google Scholar] [CrossRef]
  39. Katzenellenbogen, R. Telomerase Induction in HPV Infection and Oncogenesis. Viruses 2017, 9, 180. [Google Scholar] [CrossRef]
  40. Longworth, M.S.; Wilson, R.; Laimins, L.A. HPV31 E7 facilitates replication by activating E2F2 transcription through its interaction with HDACs. EMBO J. 2005, 24, 1821–1830. [Google Scholar] [CrossRef]
  41. Banerjee, N.S.; Moore, D.W.; Broker, T.R.; Chow, L.T. Vorinostat, a pan-HDAC inhibitor, abrogates productive HPV-18 DNA amplification. Proc. Natl. Acad. Sci. USA 2018, 115, E11138–E11147. [Google Scholar] [CrossRef]
  42. Titolo, S.; Pelletier, A.; Pulichino, A.M.; Brault, K.; Wardrop, E.; White, P.W.; Cordingley, M.G.; Archambault, J. Identification of domains of the human papillomavirus type 11 E1 helicase involved in oligomerization and binding to the viral origin. J. Virol. 2000, 74, 7349–7361. [Google Scholar] [CrossRef] [PubMed]
  43. Balaji, H.; Demers, I.; Wuerdemann, N.; Schrijnder, J.; Kremer, B.; Klussmann, J.P.; Huebbers, C.U.; Speel, E.M. Causes and Consequences of HPV Integration in Head and Neck Squamous Cell Carcinomas: State of the Art. Cancers 2021, 13, 4089. [Google Scholar] [CrossRef] [PubMed]
  44. Koneva, L.A.; Zhang, Y.; Virani, S.; Hall, P.B.; McHugh, J.B.; Chepeha, D.B.; Wolf, G.T.; Carey, T.E.; Rozek, L.S.; Sartor, M.A. HPV Integration in HNSCC Correlates with Survival Outcomes, Immune Response Signatures, and Candidate Drivers. Mol. Cancer Res. 2018, 16, 90–102. [Google Scholar] [CrossRef] [PubMed]
  45. Locati, L.D.; Serafini, M.S.; Ianno, M.F.; Carenzo, A.; Orlandi, E.; Resteghin, C.; Cavalieri, S.; Bossi, P.; Canevari, S.; Licitra, L.; et al. Mining of Self-Organizing Map Gene-Expression Portraits Reveals Prognostic Stratification of HPV-Positive Head and Neck Squamous Cell Carcinoma. Cancers 2019, 11, 1057. [Google Scholar] [CrossRef]
  46. Pinatti, L.M.; Sinha, H.N.; Brummel, C.V.; Goudsmit, C.M.; Geddes, T.J.; Wilson, G.D.; Akervall, J.A.; Brenner, C.J.; Walline, H.M.; Carey, T.E. Association of human papillomavirus integration with better patient outcomes in oropharyngeal squamous cell carcinoma. Head Neck 2021, 43, 544–557. [Google Scholar] [CrossRef]
  47. Chung, C.H.; Guthrie, V.B.; Masica, D.L.; Tokheim, C.; Kang, H.; Richmon, J.; Agrawal, N.; Fakhry, C.; Quon, H.; Subramaniam, R.M.; et al. Genomic alterations in head and neck squamous cell carcinoma determined by cancer gene-targeted sequencing. Ann. Oncol. 2015, 26, 1216–1223. [Google Scholar] [CrossRef]
  48. Seiwert, T.Y.; Zuo, Z.; Keck, M.K.; Khattri, A.; Pedamallu, C.S.; Stricker, T.; Brown, C.; Pugh, T.J.; Stojanov, P.; Cho, J.; et al. Integrative and comparative genomic analysis of HPV-positive and HPV-negative head and neck squamous cell carcinomas. Clin. Cancer Res. 2015, 21, 632–641. [Google Scholar] [CrossRef]
  49. Cancer Genome Atlas, N. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 2015, 517, 576–582. [Google Scholar] [CrossRef]
  50. Doerstling, S.; Winski, D.; Katsoulakis, E.; Agarwal, P.; Poonnen, P.J.; Snowdon, J.L.; Jackson, G.P.; Weeraratne, D.; Kelley, M.J.; Vashistha, V. Mutational profiles of head and neck squamous cell carcinomas based upon human papillomavirus status in the Veterans Affairs National Precision Oncology Program. J. Cancer Res. Clin. Oncol. 2023, 149, 69–77. [Google Scholar] [CrossRef]
  51. Krensky, A.M. The HLA system, antigen processing and presentation. Kidney Int. Suppl. 1997, 58, S2–S7. [Google Scholar]
  52. Lesseur, C.; Diergaarde, B.; Olshan, A.F.; Wunsch-Filho, V.; Ness, A.R.; Liu, G.; Lacko, M.; Eluf-Neto, J.; Franceschi, S.; Lagiou, P.; et al. Genome-wide association analyses identify new susceptibility loci for oral cavity and pharyngeal cancer. Nat. Genet. 2016, 48, 1544–1550. [Google Scholar] [CrossRef] [PubMed]
  53. Georgopoulos, N.T.; Proffitt, J.L.; Blair, G.E. Transcriptional regulation of the major histocompatibility complex (MHC) class I heavy chain, TAP1 and LMP2 genes by the human papillomavirus (HPV) type 6b, 16 and 18 E7 oncoproteins. Oncogene 2000, 19, 4930–4935. [Google Scholar] [CrossRef]
  54. Ashrafi, G.H.; Haghshenas, M.R.; Marchetti, B.; O’Brien, P.M.; Campo, M.S. E5 protein of human papillomavirus type 16 selectively downregulates surface HLA class I. Int. J. Cancer 2005, 113, 276–283. [Google Scholar] [CrossRef] [PubMed]
  55. Hemmat, N.; Baghi, H.B. Human papillomavirus E5 protein, the undercover culprit of tumorigenesis. Infect. Agents Cancer 2018, 13, 31. [Google Scholar] [CrossRef] [PubMed]
  56. Ablasser, A.; Chen, Z.J. cGAS in action: Expanding roles in immunity and inflammation. Science 2019, 363, eaat8657. [Google Scholar] [CrossRef]
  57. Shamseddine, A.A.; Burman, B.; Lee, N.Y.; Zamarin, D.; Riaz, N. Tumor Immunity and Immunotherapy for HPV-Related Cancers. Cancer Discov. 2021, 11, 1896–1912. [Google Scholar] [CrossRef]
  58. Lau, L.; Gray, E.E.; Brunette, R.L.; Stetson, D.B. DNA tumor virus oncogenes antagonize the cGAS-STING DNA-sensing pathway. Science 2015, 350, 568–571. [Google Scholar] [CrossRef]
  59. Shaikh, M.H.; Bortnik, V.; McMillan, N.A.; Idris, A. cGAS-STING responses are dampened in high-risk HPV type 16 positive head and neck squamous cell carcinoma cells. Microb. Pathog. 2019, 132, 162–165. [Google Scholar] [CrossRef]
  60. Lyford-Pike, S.; Peng, S.; Young, G.D.; Taube, J.M.; Westra, W.H.; Akpeng, B.; Bruno, T.C.; Richmon, J.D.; Wang, H.; Bishop, J.A.; et al. Evidence for a role of the PD-1:PD-L1 pathway in immune resistance of HPV-associated head and neck squamous cell carcinoma. Cancer Res. 2013, 73, 1733–1741. [Google Scholar] [CrossRef]
  61. Hong, A.M.; Ferguson, P.; Dodds, T.; Jones, D.; Li, M.; Yang, J.; Scolyer, R.A. Significant association of PD-L1 expression with human papillomavirus positivity and its prognostic impact in oropharyngeal cancer. Oral. Oncol. 2019, 92, 33–39. [Google Scholar] [CrossRef]
  62. Wang, J.; Sun, H.; Zeng, Q.; Guo, X.J.; Wang, H.; Liu, H.H.; Dong, Z.Y. HPV-positive status associated with inflamed immune microenvironment and improved response to anti-PD-1 therapy in head and neck squamous cell carcinoma. Sci. Rep. 2019, 9, 13404. [Google Scholar] [CrossRef] [PubMed]
  63. Welters, M.J.P.; Ma, W.; Santegoets, S.; Goedemans, R.; Ehsan, I.; Jordanova, E.S.; van Ham, V.J.; van Unen, V.; Koning, F.; van Egmond, S.I.; et al. Intratumoral HPV16-Specific T Cells Constitute a Type I-Oriented Tumor Microenvironment to Improve Survival in HPV16-Driven Oropharyngeal Cancer. Clin. Cancer Res. 2018, 24, 634–647. [Google Scholar] [CrossRef] [PubMed]
  64. Heusinkveld, M.; Goedemans, R.; Briet, R.J.; Gelderblom, H.; Nortier, J.W.; Gorter, A.; Smit, V.T.; Langeveld, A.P.; Jansen, J.C.; van der Burg, S.H. Systemic and local human papillomavirus 16-specific T-cell immunity in patients with head and neck cancer. Int. J. Cancer 2012, 131, E74–E85. [Google Scholar] [CrossRef] [PubMed]
  65. Jung, A.C.; Guihard, S.; Krugell, S.; Ledrappier, S.; Brochot, A.; Dalstein, V.; Job, S.; de Reynies, A.; Noel, G.; Wasylyk, B.; et al. CD8-alpha T-cell infiltration in human papillomavirus-related oropharyngeal carcinoma correlates with improved patient prognosis. Int. J. Cancer 2013, 132, E26–E36. [Google Scholar] [CrossRef]
  66. Nordfors, C.; Grun, N.; Tertipis, N.; Ahrlund-Richter, A.; Haeggblom, L.; Sivars, L.; Du, J.; Nyberg, T.; Marklund, L.; Munck-Wikland, E.; et al. CD8+ and CD4+ tumour infiltrating lymphocytes in relation to human papillomavirus status and clinical outcome in tonsillar and base of tongue squamous cell carcinoma. Eur. J. Cancer 2013, 49, 2522–2530. [Google Scholar] [CrossRef]
  67. Punt, S.; Dronkers, E.A.; Welters, M.J.; Goedemans, R.; Koljenovic, S.; Bloemena, E.; Snijders, P.J.; Gorter, A.; van der Burg, S.H.; Baatenburg de Jong, R.J.; et al. A beneficial tumor microenvironment in oropharyngeal squamous cell carcinoma is characterized by a high T cell and low IL-17(+) cell frequency. Cancer Immunol. Immunother. 2016, 65, 393–403. [Google Scholar] [CrossRef]
  68. Hladikova, K.; Koucky, V.; Boucek, J.; Laco, J.; Grega, M.; Hodek, M.; Zabrodsky, M.; Vosmik, M.; Rozkosova, K.; Vosmikova, H.; et al. Tumor-infiltrating B cells affect the progression of oropharyngeal squamous cell carcinoma via cell-to-cell interactions with CD8(+) T cells. J. Immunother. Cancer 2019, 7, 261. [Google Scholar] [CrossRef]
  69. Wood, O.; Woo, J.; Seumois, G.; Savelyeva, N.; McCann, K.J.; Singh, D.; Jones, T.; Peel, L.; Breen, M.S.; Ward, M.; et al. Gene expression analysis of TIL rich HPV-driven head and neck tumors reveals a distinct B-cell signature when compared to HPV independent tumors. Oncotarget 2016, 7, 56781–56797. [Google Scholar] [CrossRef]
  70. Pretscher, D.; Distel, L.V.; Grabenbauer, G.G.; Wittlinger, M.; Buettner, M.; Niedobitek, G. Distribution of immune cells in head and neck cancer: CD8+ T-cells and CD20+ B-cells in metastatic lymph nodes are associated with favourable outcome in patients with oro- and hypopharyngeal carcinoma. BMC Cancer 2009, 9, 292. [Google Scholar] [CrossRef]
  71. Ou, D.; Adam, J.; Garberis, I.; Blanchard, P.; Nguyen, F.; Levy, A.; Casiraghi, O.; Gorphe, P.; Breuskin, I.; Janot, F.; et al. Influence of tumor-associated macrophages and HLA class I expression according to HPV status in head and neck cancer patients receiving chemo/bioradiotherapy. Radiother. Oncol. 2019, 130, 89–96. [Google Scholar] [CrossRef]
  72. Nakamura, K.; Smyth, M.J. Myeloid immunosuppression and immune checkpoints in the tumor microenvironment. Cell Mol. Immunol. 2020, 17, 1–12. [Google Scholar] [CrossRef] [PubMed]
  73. Lang, S.; Bruderek, K.; Kaspar, C.; Hoing, B.; Kanaan, O.; Dominas, N.; Hussain, T.; Droege, F.; Eyth, C.; Hadaschik, B.; et al. Clinical Relevance and Suppressive Capacity of Human Myeloid-Derived Suppressor Cell Subsets. Clin. Cancer Res. 2018, 24, 4834–4844. [Google Scholar] [CrossRef] [PubMed]
  74. Ferris, R.L.; Blumenschein, G., Jr.; Fayette, J.; Guigay, J.; Colevas, A.D.; Licitra, L.; Harrington, K.; Kasper, S.; Vokes, E.E.; Even, C.; et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2016, 375, 1856–1867. [Google Scholar] [CrossRef] [PubMed]
  75. Ferris, R.L.; Blumenschein, G., Jr.; Fayette, J.; Guigay, J.; Colevas, A.D.; Licitra, L.; Harrington, K.J.; Kasper, S.; Vokes, E.E.; Even, C.; et al. Nivolumab vs investigator’s choice in recurrent or metastatic squamous cell carcinoma of the head and neck: 2-year long-term survival update of CheckMate 141 with analyses by tumor PD-L1 expression. Oral. Oncol. 2018, 81, 45–51. [Google Scholar] [CrossRef]
  76. Zandberg, D.P.; Algazi, A.P.; Jimeno, A.; Good, J.S.; Fayette, J.; Bouganim, N.; Ready, N.E.; Clement, P.M.; Even, C.; Jang, R.W.; et al. Durvalumab for recurrent or metastatic head and neck squamous cell carcinoma: Results from a single-arm, phase II study in patients with >/=25% tumour cell PD-L1 expression who have progressed on platinum-based chemotherapy. Eur. J. Cancer 2019, 107, 142–152. [Google Scholar] [CrossRef]
  77. Seiwert, T.Y.; Burtness, B.; Mehra, R.; Weiss, J.; Berger, R.; Eder, J.P.; Heath, K.; McClanahan, T.; Lunceford, J.; Gause, C.; et al. Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): An open-label, multicentre, phase 1b trial. Lancet Oncol. 2016, 17, 956–965. [Google Scholar] [CrossRef]
  78. Mehra, R.; Seiwert, T.Y.; Gupta, S.; Weiss, J.; Gluck, I.; Eder, J.P.; Burtness, B.; Tahara, M.; Keam, B.; Kang, H.; et al. Efficacy and safety of pembrolizumab in recurrent/metastatic head and neck squamous cell carcinoma: Pooled analyses after long-term follow-up in KEYNOTE-012. Br. J. Cancer 2018, 119, 153–159. [Google Scholar] [CrossRef]
  79. Bauml, J.; Seiwert, T.Y.; Pfister, D.G.; Worden, F.; Liu, S.V.; Gilbert, J.; Saba, N.F.; Weiss, J.; Wirth, L.; Sukari, A.; et al. Pembrolizumab for Platinum- and Cetuximab-Refractory Head and Neck Cancer: Results From a Single-Arm, Phase II Study. J. Clin. Oncol. 2017, 35, 1542–1549. [Google Scholar] [CrossRef]
  80. Colevas, A.D.; Bahleda, R.; Braiteh, F.; Balmanoukian, A.; Brana, I.; Chau, N.G.; Sarkar, I.; Molinero, L.; Grossman, W.; Kabbinavar, F.; et al. Safety and clinical activity of atezolizumab in head and neck cancer: Results from a phase I trial. Ann. Oncol. 2018, 29, 2247–2253. [Google Scholar] [CrossRef]
  81. Wang, B.C.; Cao, R.B.; Li, P.D.; Fu, C. The effects and safety of PD-1/PD-L1 inhibitors on head and neck cancer: A systematic review and meta-analysis. Cancer Med. 2019, 8, 5969–5978. [Google Scholar] [CrossRef]
  82. Ferris, R.L.; Haddad, R.; Even, C.; Tahara, M.; Dvorkin, M.; Ciuleanu, T.E.; Clement, P.M.; Mesia, R.; Kutukova, S.; Zholudeva, L.; et al. Durvalumab with or without tremelimumab in patients with recurrent or metastatic head and neck squamous cell carcinoma: EAGLE, a randomized, open-label phase III study. Ann. Oncol. 2020, 31, 942–950. [Google Scholar] [CrossRef] [PubMed]
  83. Psyrri, A.; Fayette, J.; Harrington, K.; Gillison, M.; Ahn, M.J.; Takahashi, S.; Weiss, J.; Machiels, J.P.; Baxi, S.; Vasilyev, A.; et al. Durvalumab with or without tremelimumab versus the EXTREME regimen as first-line treatment for recurrent or metastatic squamous cell carcinoma of the head and neck: KESTREL, a randomized, open-label, phase III study. Ann. Oncol. 2023, 34, 262–274. [Google Scholar] [CrossRef] [PubMed]
  84. Siu, L.L.; Even, C.; Mesia, R.; Remenar, E.; Daste, A.; Delord, J.P.; Krauss, J.; Saba, N.F.; Nabell, L.; Ready, N.E.; et al. Safety and Efficacy of Durvalumab With or Without Tremelimumab in Patients With PD-L1-Low/Negative Recurrent or Metastatic HNSCC: The Phase 2 CONDOR Randomized Clinical Trial. JAMA Oncol. 2019, 5, 195–203. [Google Scholar] [CrossRef] [PubMed]
  85. Struckmeier, A.K.; Gosau, M.; Smeets, R. Immunotherapeutic strategies beyond the PD-1/PD-L1 pathway in head and neck squamous cell carcinoma—A scoping review on current developments in agents targeting TIM-3, TIGIT, LAG-3, and VISTA. Oral. Oncol. 2025, 161, 107145. [Google Scholar] [CrossRef]
  86. Zhang, S.; Zheng, M.; Nie, D.; Xu, L.; Tian, H.; Wang, M.; Liu, W.; Feng, Z.; Han, F. Efficacy of cetuximab plus PD-1 inhibitor differs by HPV status in head and neck squamous cell carcinoma: A systematic review and meta-analysis. J. Immunother. Cancer 2022, 10, e005158. [Google Scholar] [CrossRef]
  87. Mock, A.; Plath, M.; Moratin, J.; Tapken, M.J.; Jager, D.; Krauss, J.; Frohling, S.; Hess, J.; Zaoui, K. EGFR and PI3K Pathway Activities Might Guide Drug Repurposing in HPV-Negative Head and Neck Cancers. Front. Oncol. 2021, 11, 678966. [Google Scholar] [CrossRef]
  88. Chaturvedi, A.K.; Graubard, B.I.; Broutian, T.; Pickard, R.K.L.; Tong, Z.Y.; Xiao, W.; Kahle, L.; Gillison, M.L. Effect of Prophylactic Human Papillomavirus (HPV) Vaccination on Oral HPV Infections Among Young Adults in the United States. J. Clin. Oncol. 2018, 36, 262–267. [Google Scholar] [CrossRef]
  89. Wang, C.; Dickie, J.; Sutavani, R.V.; Pointer, C.; Thomas, G.J.; Savelyeva, N. Targeting Head and Neck Cancer by Vaccination. Front. Immunol. 2018, 9, 830. [Google Scholar] [CrossRef]
  90. D’Abramo, C.M.; Archambault, J. Small molecule inhibitors of human papillomavirus protein—Protein interactions. Open Virol. J. 2011, 5, 80–95. [Google Scholar] [CrossRef]
  91. Devaraja, K.; Aggarwal, S.; Singh, M. Therapeutic Vaccination in Head and Neck Squamous Cell Carcinoma-A Review. Vaccines 2023, 11, 634. [Google Scholar] [CrossRef]
  92. Filippini, D.M.; Broseghini, E.; Liberale, C.; Gallerani, G.; Siepe, G.; Nobili, E.; Ferracin, M.; Molteni, G. Vaccine-Based Immunotherapy for Oropharyngeal and Nasopharyngeal Cancers. J. Clin. Med. 2025, 14, 1170. [Google Scholar] [CrossRef] [PubMed]
  93. Lopes, A.; Vandermeulen, G.; Preat, V. Cancer DNA vaccines: Current preclinical and clinical developments and future perspectives. J. Exp. Clin. Cancer Res. 2019, 38, 146. [Google Scholar] [CrossRef] [PubMed]
  94. Aggarwal, C.; Cohen, R.B.; Morrow, M.P.; Kraynyak, K.A.; Sylvester, A.J.; Knoblock, D.M.; Bauml, J.M.; Weinstein, G.S.; Lin, A.; Boyer, J.; et al. Immunotherapy Targeting HPV16/18 Generates Potent Immune Responses in HPV-Associated Head and Neck Cancer. Clin. Cancer Res. 2019, 25, 110–124. [Google Scholar] [CrossRef] [PubMed]
  95. Aggarwal, C.; Saba, N.F.; Algazi, A.; Sukari, A.; Seiwert, T.Y.; Haigentz, M.; Porosnicu, M.; Bonomi, M.; Boyer, J.; Esser, M.T.; et al. Safety and Efficacy of MEDI0457 plus Durvalumab in Patients with Human Papillomavirus-Associated Recurrent/Metastatic Head and Neck Squamous Cell Carcinoma. Clin. Cancer Res. 2023, 29, 560–570. [Google Scholar] [CrossRef]
  96. Chandra, J.; Woo, W.P.; Finlayson, N.; Liu, H.Y.; McGrath, M.; Ladwa, R.; Brauer, M.; Xu, Y.; Hanson, S.; Panizza, B.; et al. A phase 1, single centre, open label, escalating dose study to assess the safety, tolerability and immunogenicity of a therapeutic human papillomavirus (HPV) DNA vaccine (AMV002) for HPV-associated head and neck cancer (HNC). Cancer Immunol. Immunother. 2021, 70, 743–753. [Google Scholar] [CrossRef]
  97. Ladwa, R.; Chandra, J.; Woo, W.P.; Finlayson, N.; Liu, H.; McGrath, M.; See, A.; Hughes, B.G.; Cooper, C.L.; Jackson, J.E.; et al. A phase Ib study to assess the safety of the human papillomavirus DNA vaccine (AMV002) in combination with durvalumab for HPV-associated oropharyngeal squamous cell carcinoma. Front. Oncol. 2024, 14, 1419258. [Google Scholar] [CrossRef]
  98. Ottensmeier, C.; King, E.; Crabb, S.; Karydis, I.; Graham, D.M.; Martin, K.; Eberhart, I.; Ewings, S.; Lee, P.; McCann, K.; et al. A phase I/II trial of therapeutic HPV vaccine (BNT113) in patients with HPV16 driven carcinoma. Ann. Oncol. 2024, 35, S680. [Google Scholar] [CrossRef]
  99. Saba, N.F.; Klinghammer, K.; Castelluci, E.; Colevas, A.D.; Rutkowski, T.; Thurner, D.; Muller-Richter, U.; Maio, M.; Grewal, J.S.; Ottensmeier, C.; et al. Exploratory efficacy and translational results from the safety run in of AHEAD-MERIT, a phase II trial of first line pembrolizumab plus the fixed-antigen cancer vaccine BNT113 in advanced HPV16+ HNSCC. Ann. Oncol. 2024, 35, S627. [Google Scholar] [CrossRef]
  100. Smalley Rumfield, C.; Pellom, S.T.; Morillon Ii, Y.M.; Schlom, J.; Jochems, C. Immunomodulation to enhance the efficacy of an HPV therapeutic vaccine. J. Immunother. Cancer 2020, 8, e000612. [Google Scholar] [CrossRef]
  101. Weiss, J.; Kaczmar, J.; Harrington, K.J.; Mehra, R.; Worden, F.; Zinner, R.; Giri, A.; Kang, H.; Najeeb, S.; Panella, T.; et al. VERSATILE-002: Survival with first-line treatment with PDS0101 therapeutic vaccine and pembrolizumab in HPV16-positive recurrent/metastatic head and neck squamous cell carcinoma (HNSCC). Ann. Oncol. 2024, 35, S628. [Google Scholar] [CrossRef]
  102. PDSB, PDS Biotech Announces Interim 24-Month Survival Rate of 74% in Immune Checkpoint Inhibitor Naïve Head and Neck Cancer Patients Treated with PDS0101 in Combination with KEYTRUDA® (pembrolizumab). 2023. Available online: https://www.globenewswire.com/news-release/2023/10/03/2753452/37149/en/PDS-Biotech-Announces-Interim-24-Month-Survival-Rate-of-74-in-Immune-Checkpoint-Inhibitor-Na%C3%AFve-Head-and-Neck-Cancer-Patients-Treated-with-PDS0101-in-Combination-with-KEYTRUDA-pemb.html (accessed on 14 May 2025).
  103. Strauss, J.; Floudas, C.S.; Pastor, D.M.; Manu, M.; Lamping, E.; Francis, D.C.; Cordes, L.M.; Marte, J.; Donahue, R.N.; Jochems, C.; et al. Phase II evaluation of the combination of PDS0101, M9241, and bintrafusp alfa in patients with HPV 16+ malignancies. J. Clin. Oncol. 2022, 40, 2518. [Google Scholar] [CrossRef]
  104. Fallon, J.; Tighe, R.; Kradjian, G.; Guzman, W.; Bernhardt, A.; Neuteboom, B.; Lan, Y.; Sabzevari, H.; Schlom, J.; Greiner, J.W. The immunocytokine NHS-IL12 as a potential cancer therapeutic. Oncotarget 2014, 5, 1869–1884. [Google Scholar] [CrossRef] [PubMed]
  105. Kenter, G.G.; Welters, M.J.; Valentijn, A.R.; Lowik, M.J.; Berends-van der Meer, D.M.; Vloon, A.P.; Essahsah, F.; Fathers, L.M.; Offringa, R.; Drijfhout, J.W.; et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N. Engl. J. Med. 2009, 361, 1838–1847. [Google Scholar] [CrossRef] [PubMed]
  106. Massarelli, E.; William, W.; Johnson, F.; Kies, M.; Ferrarotto, R.; Guo, M.; Feng, L.; Lee, J.J.; Tran, H.; Kim, Y.U.; et al. Combining Immune Checkpoint Blockade and Tumor-Specific Vaccine for Patients With Incurable Human Papillomavirus 16-Related Cancer: A Phase 2 Clinical Trial. JAMA Oncol. 2019, 5, 67–73. [Google Scholar] [CrossRef]
  107. Even, C.; Harrington, K.J.; Massarelli, E.; Hesselink, M.K.; Visscher, S.; Fury, M.G.; Sanders, F.; Laban, S.; Fayette, J.; Oliva, M.; et al. Results of a randomized, double-blind, placebo-controlled, phase 2 study (OpcemISA) of the combination of ISA101b and cemiplimab versus cemiplimab for recurrent/metastatic (R/M) HPV16-positive oropharyngeal cancer (OPC). J. Clin. Oncol. 2024, 42, 6003. [Google Scholar] [CrossRef]
  108. Yang, A.; Jeang, J.; Cheng, K.; Cheng, T.; Yang, B.; Wu, T.C.; Hung, C.F. Current state in the development of candidate therapeutic HPV vaccines. Expert. Rev. Vaccines 2016, 15, 989–1007. [Google Scholar] [CrossRef]
  109. Cory, L.; Chu, C. ADXS-HPV: A therapeutic Listeria vaccination targeting cervical cancers expressing the HPV E7 antigen. Hum. Vaccin. Immunother. 2014, 10, 3190–3195. [Google Scholar] [CrossRef]
  110. Sacco, J.J.; Evans, M.; Harrington, K.J.; Man, S.; Powell, N.; Shaw, R.J.; Jones, T.M. Systemic listeriosis following vaccination with the attenuated Listeria monocytogenes therapeutic vaccine, ADXS11-001. Hum. Vaccin. Immunother. 2016, 12, 1085–1086. [Google Scholar] [CrossRef]
  111. Ho, A.L.; Pearson, A.T.; Fu, S.; Posner, M.R.; Adkins, D.; Nabell, L.; Niu, J.; Richardson, D.L.; Leidner, R.S.; Chung, K.; et al. Characterization of tumor specific CD8+ T cell responses in patients with recurrent/metastatic HPV16-positive head and neck cancer receiving HB-200 monotherapy as second or later line treatment in a phase 1 study. J. Immunother. Cancer 2023, 11, 770. [Google Scholar]
  112. Ho, A.L.; Nabell, L.; Neupane, P.C.; Posner, M.R.; Yilmaz, E.; Niu, J.; Naqash, A.R.; Pearson, A.T.; Wong, S.J.; Nieva, J.J.; et al. HB-200 arenavirus-based immunotherapy plus pembrolizumab as first-line treatment of patients with recurrent/metastatic HPV16-positive head and neck cancer: Updated results. J. Clin. Oncol. 2024, 42, 6005. [Google Scholar] [CrossRef]
  113. Pellom, S.T.; Smalley Rumfield, C.; Morillon, Y.M., 2nd; Roller, N.; Poppe, L.K.; Brough, D.E.; Sabzevari, H.; Schlom, J.; Jochems, C. Characterization of recombinant gorilla adenovirus HPV therapeutic vaccine PRGN-2009. JCI Insight 2021, 6, e141912. [Google Scholar] [CrossRef] [PubMed]
  114. Floudas, C.S.; Strauss, J.; Redman, J.M.; Pastor, D.M.; Turkbey, E.B.; Donahue, R.N.; Jochems, C.; McMahon, S.; Lamping, E.; Cordes, L.M.; et al. Phase I evaluation of PRGN-2009 alone and in combination with bintrafusp alfa in patients (pts) with recurrent/metastatic (R/M) HPV-associated cancers (HPV-C). J. Clin. Oncol. 2023, 41, 2628. [Google Scholar] [CrossRef]
  115. Bhatt, K.H.; Neller, M.A.; Srihari, S.; Crooks, P.; Lekieffre, L.; Aftab, B.T.; Liu, H.; Smith, C.; Kenny, L.; Porceddu, S.; et al. Profiling HPV-16-specific T cell responses reveals broad antigen reactivities in oropharyngeal cancer patients. J. Exp. Med. 2020, 217, e20200389. [Google Scholar] [CrossRef] [PubMed]
  116. Eberhardt, C.S.; Kissick, H.T.; Patel, M.R.; Cardenas, M.A.; Prokhnevska, N.; Obeng, R.C.; Nasti, T.H.; Griffith, C.C.; Im, S.J.; Wang, X.; et al. Functional HPV-specific PD-1(+) stem-like CD8 T cells in head and neck cancer. Nature 2021, 597, 279–284. [Google Scholar] [CrossRef]
  117. Krishna, S.; Ulrich, P.; Wilson, E.; Parikh, F.; Narang, P.; Yang, S.; Read, A.K.; Kim-Schulze, S.; Park, J.G.; Posner, M.; et al. Human Papilloma Virus Specific Immunogenicity and Dysfunction of CD8(+) T Cells in Head and Neck Cancer. Cancer Res. 2018, 78, 6159–6170. [Google Scholar] [CrossRef]
  118. Le Tourneau, C.; Delord, J.P.; Lalanne, A.; Jamet, C.; Spring-Giusti, C.; Tavernaro, A.; Bastien, B.; Brandely-Talbot, M.; Quemeneur, E.; Onoue, K.; et al. Randomized phase I trial of adjuvant individualized TG4050 vaccine in patients with locally advanced resected HPV-negative head and neck squamous cell carcinoma (HNSCC). J. Immunother. Cancer 2024, 12. [Google Scholar]
  119. Topalian, S.L.; Solomon, D.; Avis, F.P.; Chang, A.E.; Freerksen, D.L.; Linehan, W.M.; Lotze, M.T.; Robertson, C.N.; Seipp, C.A.; Simon, P.; et al. Immunotherapy of patients with advanced cancer using tumor-infiltrating lymphocytes and recombinant interleukin-2: A pilot study. J. Clin. Oncol. 1988, 6, 839–853. [Google Scholar] [CrossRef]
  120. Morotti, M.; Albukhari, A.; Alsaadi, A.; Artibani, M.; Brenton, J.D.; Curbishley, S.M.; Dong, T.; Dustin, M.L.; Hu, Z.; McGranahan, N.; et al. Promises and challenges of adoptive T-cell therapies for solid tumours. Br. J. Cancer 2021, 124, 1759–1776. [Google Scholar] [CrossRef]
  121. Canelo-Vilaseca, M.; Sabbah, M.; Di Blasi, R.; Cristinelli, C.; Sureda, A.; Caillat-Zucman, S.; Thieblemont, C. Lymphodepletion chemotherapy in chimeric antigen receptor-engineered T (CAR-T) cell therapy in lymphoma. Bone Marrow Transplant. 2025, 60, 559–567. [Google Scholar] [CrossRef]
  122. Albarran, V.; San Roman, M.; Pozas, J.; Chamorro, J.; Rosero, D.I.; Guerrero, P.; Calvo, J.C.; Gonzalez, C.; Garcia de Quevedo, C.; Perez de Aguado, P.; et al. Adoptive T cell therapy for solid tumors: Current landscape and future challenges. Front. Immunol. 2024, 15, 1352805. [Google Scholar] [CrossRef]
  123. Zhang, P.; Zhang, G.; Wan, X. Challenges and new technologies in adoptive cell therapy. J. Hematol. Oncol. 2023, 16, 97. [Google Scholar] [CrossRef] [PubMed]
  124. Doran, S.L.; Stevanovic, S.; Adhikary, S.; Gartner, J.J.; Jia, L.; Kwong, M.L.M.; Faquin, W.C.; Hewitt, S.M.; Sherry, R.M.; Yang, J.C.; et al. T-Cell Receptor Gene Therapy for Human Papillomavirus-Associated Epithelial Cancers: A First-in-Human, Phase I/II Study. J. Clin. Oncol. 2019, 37, 2759–2768. [Google Scholar] [CrossRef] [PubMed]
  125. Nagarsheth, N.B.; Norberg, S.M.; Sinkoe, A.L.; Adhikary, S.; Meyer, T.J.; Lack, J.B.; Warner, A.C.; Schweitzer, C.; Doran, S.L.; Korrapati, S.; et al. TCR-engineered T cells targeting E7 for patients with metastatic HPV-associated epithelial cancers. Nat. Med. 2021, 27, 419–425. [Google Scholar] [CrossRef] [PubMed]
  126. Jin, B.Y.; Campbell, T.E.; Draper, L.M.; Stevanovic, S.; Weissbrich, B.; Yu, Z.; Restifo, N.P.; Rosenberg, S.A.; Trimble, C.L.; Hinrichs, C.S. Engineered T cells targeting E7 mediate regression of human papillomavirus cancers in a murine model. JCI Insight 2018, 3, e99488. [Google Scholar] [CrossRef]
  127. Jimeno, A.; Papa, S.; Haigentz, M.; Rodriguez-Moreno, J.; Schardt, J.; Fardis, M.; Finckenstein, G.; Fiaz, R.; Chen, G.; Cacovean, A.; et al. Safety and efficacy of tumor infiltrating lymphocytes (TIL, LN-145) in combination with pembrolizumab for advanced, recurrent or metastatic HNSCC. J. Immunother. Cancer 2020, 8, A378. [Google Scholar]
  128. Biologics, T. Turnstone Biologics Corp. Reports Positive Initial Data from Phase 1 Trial of TIDAL-01 in Metastatic Colorectal Cancer; Biospace: West Des Moines, IA, USA, 2024. [Google Scholar]
  129. Pikor, L.A.; Bernard, A.; Brassard, N.; Fritzsche, A.; Kluew, A.; Jilesen, Z.K.; Nikota, J.; Bareja, R.; Laing, C.; Stojdl, D.F.; et al. TIDAL-01: A selected TIL process that enriches for neoantigen reactive TIL in solid tumors. Cancer Res. 2023, 83, 4049. [Google Scholar] [CrossRef]
  130. Biologics, T. Turnstone Biologics Announces Plans to Explore Strategic Alternatives; Turnstone Biologics Corp.: La Jolla, CA, USA, 2025. [Google Scholar]
  131. Seidel, R.D.; Merazga, Z.; Thapa, D.R.; Soriano, J.; Spaulding, E.; Vakkasoglu, A.S.; Ruthardt, P.; Bautista, W.; Quayle, S.N.; Kiener, P.A.; et al. Peptide-HLA-based immunotherapeutics platforms for direct modulation of antigen-specific T cells. Sci. Rep. 2021, 11, 19220. [Google Scholar] [CrossRef]
  132. Quayle, S.N.; Girgis, N.; Thapa, D.R.; Merazga, Z.; Kemp, M.M.; Histed, A.; Zhao, F.; Moreta, M.; Ruthardt, P.; Hulot, S.; et al. CUE-101, a Novel E7-pHLA-IL2-Fc Fusion Protein, Enhances Tumor Antigen-Specific T-Cell Activation for the Treatment of HPV16-Driven Malignancies. Clin. Cancer Res. 2020, 26, 1953–1964. [Google Scholar] [CrossRef]
  133. Colevas, A.D.; Chung, C.H.; Adkins, D.; Rodriguez, C.P.; Park, J.C.; Gibson, M.K.; Sukari, A.; Worden, F.P.; Johnson, F.M.; Saba, N.F.; et al. A phase 1 dose-escalation and expansion study of CUE-101, given as monotherapy and in combination with pembrolizumab, in patients with recurrent/metastatic HPV16+ head and neck squamous cell cancer (R/M HNSCC). J. Clin. Oncol. 2024, 42, 6004. [Google Scholar] [CrossRef]
  134. Harbison, R.A.; Kubik, M.; Konnick, E.Q.; Zhang, Q.; Lee, S.G.; Park, H.; Zhang, J.; Carlson, C.S.; Chen, C.; Schwartz, S.M.; et al. The mutational landscape of recurrent versus nonrecurrent human papillomavirus-related oropharyngeal cancer. JCI Insight 2018, 3, e99327. [Google Scholar] [CrossRef]
  135. Gleber-Netto, F.O.; Rao, X.; Guo, T.; Xi, Y.; Gao, M.; Shen, L.; Erikson, K.; Kalu, N.N.; Ren, S.; Xu, G.; et al. Variations in HPV function are associated with survival in squamous cell carcinoma. JCI Insight 2019, 4, e124762. [Google Scholar] [CrossRef] [PubMed]
  136. Ahrlund-Richter, A.; Holzhauser, S.; Dalianis, T.; Nasman, A.; Mints, M. Whole-Exome Sequencing of HPV Positive Tonsillar and Base of Tongue Squamous Cell Carcinomas Reveals a Global Mutational Pattern along with Relapse-Specific Somatic Variants. Cancers 2021, 14, 77. [Google Scholar] [CrossRef] [PubMed]
  137. Lechner, M.; Liu, J.; Masterson, L.; Fenton, T.R. HPV-associated oropharyngeal cancer: Epidemiology, molecular biology and clinical management. Nat. Rev. Clin. Oncol. 2022, 19, 306–327. [Google Scholar] [CrossRef] [PubMed]
  138. Bertagnin, C.; Messa, L.; Pavan, M.; Celegato, M.; Sturlese, M.; Mercorelli, B.; Moro, S.; Loregian, A. A small molecule targeting the interaction between human papillomavirus E7 oncoprotein and cellular phosphatase PTPN14 exerts antitumoral activity in cervical cancer cells. Cancer Lett. 2023, 571, 216331. [Google Scholar] [CrossRef]
  139. Celegato, M.; Messa, L.; Bertagnin, C.; Mercorelli, B.; Loregian, A. Targeted Disruption of E6/p53 Binding Exerts Broad Activity and Synergism with Paclitaxel and Topotecan against HPV-Transformed Cancer Cells. Cancers 2021, 14, 193. [Google Scholar] [CrossRef]
  140. Bekes, M.; Langley, D.R.; Crews, C.M. PROTAC targeted protein degraders: The past is prologue. Nat. Rev. Drug Discov. 2022, 21, 181–200. [Google Scholar] [CrossRef]
  141. Mukerjee, N.; Maitra, S.; Gorai, S.; Ghosh, A.; Alexiou, A.; Thorat, N.D. Revolutionizing Human papillomavirus (HPV)-related cancer therapies: Unveiling the promise of Proteolysis Targeting Chimeras (PROTACs) and Proteolysis Targeting Antibodies (PROTABs) in cancer nano-vaccines. J. Med. Virol. 2023, 95, e29135. [Google Scholar] [CrossRef]
  142. Nishida, H.; Matsumoto, Y.; Kawana, K.; Christie, R.J.; Naito, M.; Kim, B.S.; Toh, K.; Min, H.S.; Yi, Y.; Matsumoto, Y.; et al. Systemic delivery of siRNA by actively targeted polyion complex micelles for silencing the E6 and E7 human papillomavirus oncogenes. J. Control. Release 2016, 231, 29–37. [Google Scholar] [CrossRef]
  143. Sato, N.; Saga, Y.; Uchibori, R.; Tsukahara, T.; Urabe, M.; Kume, A.; Fujiwara, H.; Suzuki, M.; Ozawa, K.; Mizukami, H. Eradication of cervical cancer in vivo by an AAV vector that encodes shRNA targeting human papillomavirus type 16 E6/E7. Int. J. Oncol. 2018, 52, 687–696. [Google Scholar]
  144. Zhou, J.; Li, B.; Peng, C.; Wang, F.; Fu, Z.; Zhou, C.; Hong, D.; Ye, F.; Lu, W.; Xie, X. Inhibition of cervical cancer cell growth in vitro and in vivo by lentiviral-vector mediated shRNA targeting the common promoter of HPV16 E6 and E7 oncogenes. Antiviral Res. 2013, 98, 305–313. [Google Scholar] [CrossRef]
  145. Hong, D.; Lu, W.; Ye, F.; Hu, Y.; Xie, X. Gene silencing of HPV16 E6/E7 induced by promoter-targeting siRNA in SiHa cells. Br. J. Cancer 2009, 101, 1798–1804. [Google Scholar] [CrossRef] [PubMed]
  146. Jubair, L.; Fallaha, S.; McMillan, N.A.J. Systemic Delivery of CRISPR/Cas9 Targeting HPV Oncogenes Is Effective at Eliminating Established Tumors. Mol. Ther. 2019, 27, 2091–2099. [Google Scholar] [CrossRef] [PubMed]
  147. Zhen, S.; Qiang, R.; Lu, J.; Tuo, X.; Yang, X.; Li, X. CRISPR/Cas9-HPV-liposome enhances antitumor immunity and treatment of HPV infection-associated cervical cancer. J. Med. Virol. 2023, 95, e28144. [Google Scholar] [CrossRef] [PubMed]
  148. Faucher, A.M.; White, P.W.; Brochu, C.; Grand-Maitre, C.; Rancourt, J.; Fazal, G. Discovery of small-molecule inhibitors of the ATPase activity of human papillomavirus E1 helicase. J. Med. Chem. 2004, 47, 18–21. [Google Scholar] [CrossRef]
Table 1. Ongoing clinical trials of therapeutic vaccines in HPV-associated HNSCC.
Table 1. Ongoing clinical trials of therapeutic vaccines in HPV-associated HNSCC.
NCT NumberDesignPopulationTreatmentVaccine
Target
Primary
Outcome
Status *
DNA-based vaccines
NCT06016920Phase I/II, multi-center, non-randomized, n = 51Previously untreated recurrent/metastatic HPV-16-positive HNSCC with PD-L1 CPS ≥ 1VB10.16 with pembrolizumabHPV-16 E6/E7Phase I: DLT, AEs
Phase II: ORR, immune response
Recruiting
RNA-based vaccines
NCT04534205
(AHEAD-MERIT)
Phase II/III, multi-center, randomized, n = 350Previously untreated recurrent/metastatic HPV-16-positive HNSCC with PD-L1 CPS ≥ 1BNT113 with pembrolizumab vs. pembrolizumab monotherapyHPV-16 E6/E7Part A: AEs
Part B: OS, PFS
Recruiting
Peptide-based vaccines
NCT04260126
(VERSATILE-002)
Phase II, multi-center, multi-cohort, non-randomized, n = 95Recurrent/metastatic HPV-16-positive HNSCC, PD-L1 CPS ≥ 1 if checkpoint inhibitor-naïve, any PD-L1 CPS if checkpoint inhibitor-experiencedPDS0101 with pembrolizumabHPV-16 E6/E7ORRActive, not recruiting
NCT06790966
(VERSATILE-003)
Phase III, multi-center, randomized, n = 351Previously untreated recurrent/metastatic HPV-16-positive HNSCC with PD-L1 CPS ≥ 1PDS0101 with pembrolizumab vs. pembrolizumab monotherapyHPV-16 E6/E7OSRecruiting
NCT05232851Phase I/II, single-center, multi-arm, non-randomized, n = 24Locally advanced OPSCC with high-risk HPV-specific testing and at least one risk factorPDS0101 alone or with pembrolizumabHPV-16 E6/E7Pathologic and HPV cell-free tumor DNA responseRecruiting
NCT03669718
(OpcemISA)
Phase II, multi-center, randomized, n = 198Recurrent/metastatic HPV-16-positive OPSCC with PD-L1 CPS ≥ 1, anti-PD-1 therapy-naïve, first- or second-line settingISA101b with cemiplimab vs. cemiplimab monotherapyHPV-16 E6/E7ORR, AEsActive, not recruiting
NCT04398524Phase II, multi-center, single-arm, n = 65Recurrent/metastatic HPV-16-positive OPSCC with progression on prior anti-PD-1 therapyISA101b with cemiplimabHPV-16 E6/E7ORRActive, not recruiting
Viral vector-based vaccines
NCT04180215Phase I/II, multi-center, multi-cohort, non-randomized, n = 200Recurrent/metastatic HPV-16-positive HNSCC eligible to receive pembrolizumab as standard of careHB-201 alone or HB-201 and HB-202 combination, with standard-of-care regimen including pembrolizumabHPV-16 E6/E7Phase I: DLT, RP2D
Phase II: ORR
Active, not recruiting
NCT05108870Phase I/II, single-center, randomized, n = 98Locally advanced HPV-16-positive OPSCC, cT3-T4 (any N), or cN1-N3Neoadjuvant HB-201 alone vs. HB-201 and HB-202 combination, both with carboplatin and paclitaxel chemotherapy prior to definitive therapyHPV-16 E6/E7Phase I: DLT, AEs
Phase II: Deep response rates
Recruiting
NCT04432597Phase I/II, single-center, multi-arm, non-randomized, n = 70Locally advanced or metastatic HPV-associated cancer (Phase I); stage II–III p16-postive OPSCC (phase II)PRGN-2009 alone or with bintrafusp alfa (M7824)HPV-16 and HPV-18 E6/E7Phase I: RP2D, safety
Phase II: Increase in CD3+ tumor-infiltrating T-cells
Active, not recruiting
NCT05996523Phase II, single-center, single-arm, n = 29Stage I-II p16-positive OPSCC planned for definitive surgery or chemoradiationPRGN-2009 with pembrolizumab prior to definitive therapyHPV-16 and HPV-18 E6/E7Increase in CD3+ tumor-infiltrating T-cellsRecruiting
NCT06223568Phase II, single-center, multi-arm, randomized, n = 70Resectable stage I-II OPSCC, HPV-associated with any high-risk serotypeNeoadjuvant cisplatin and docetaxel chemotherapy alone or with PRGN-2009 prior to surgeryHPV-16 and HPV-18 E6/E7pCR rateRecruiting
NCT06319963Phase I/II, multi-center, multi-cohort, non-randomized, n = 72Arm A: Recurrent/metastatic HPV-associated OPSCC or cervical cancer
Arm B: Untreated, locally advanced HPV-associated OPSCC (cT1-2N2-3, cT3-4N0-3) or cervical cancer (Stage IB-IVA)
Lenti-HPV-07HPV-16 and HPV-18 E6/E7AEs, optimal biological doseRecruiting
* Status as of 18 April 2025. Abbreviations: AE, adverse event; DLT, dose-limiting toxicity; DNA, deoxyribonucleic acid; HNSCC, head and neck squamous cell carcinoma; HPV, human papillomavirus; OPSCC, oropharyngeal squamous cell carcinoma; ORR, objective response rate; OS, overall survival; PD-1, programmed cell death protein 1; PD-L1, programmed cell death ligand 1; pCR, pathologic complete response; PFS, progression-free survival; RNA, ribonucleic acid; RP2D, recommended phase II dose.
Table 2. Ongoing clinical trials of adoptive cell therapies in HPV-associated HNSCC.
Table 2. Ongoing clinical trials of adoptive cell therapies in HPV-associated HNSCC.
NCT NumberDesignPopulationTreatmentPrimary
Outcome
Status *
Genetically modified TCR therapies (all with HPV- and HLA-specific enrollment criteria)
NCT05639972Phase I/II, single-center, single-arm, feasibility, n = 15Locoregionally advanced HPV-16-positive cancer and HLA-A*02:01 alleleE7 TCR T-cellsFeasibility of administration Recruiting
NCT05686226Phase II, single-center, single-arm, n = 20Recurrent/metastatic HPV-16-positive cancer and HLA-A*02:01 alleleE7 TCR T-cellsORRRecruiting
NCT05973487Phase I, multi-center, non-randomized, multi-cohort, n = 840Unresectable/metastatic solids tumors; HPV-16-positive with HLA-A*02:01 allele in cohort C; includes additional combination cohorts with a second target antigen E7 TCR T-cells as monotherapy and in combination with other TCRs targeting MAGE-A1, MAGE-A4, MAGE-C2, PRAMESafety, RP2DRecruiting
NCT02858310Phase I/II, multi-center, single-arm, n = 180Recurrent/metastatic HPV-16-positive cancers with HLA-A*02:01 alleleE7 TCR T-cellsPhase I: Safety
Phase II: ORR
Recruiting
NCT05787535Phase I, single-center, single-arm, n = 17 Recurrent/metastatic HPV-18-positive solid tumors and HLA-DRB1*09:01 alleleHRYZ-T101 TCR T-cellsDLT, AEsRecruiting
NCT05952947Phase I, multi-center, single-arm, n = 32Recurrent/metastatic HPV-18-positive solid tumors and HLA-DRB1*09:01 alleleHRYZ-T101 TCR T-cellsDLT, AEs Recruiting
CAR-T or CAR-NK cell therapies
NCT06096038Phase I/II, single-center, single-arm, n = 33Recurrent/metastatic HNSCCAutologous CAR-T against CSPG4AEsRecruiting
NCT04119024Phase I, multi-center, single-arm, n = 18Solid tumors expressing IL13Ralpha2Autologous CAR-T against IL13Ralpha2DLT, AEsRecruiting
NCT05239143Phase I, multi-center, multi-cohort, non-randomized, n = 180Advanced/metastatic epithelial-derived cancersAllogeneic CAR-T against MUC1-CDLT, AEs, ORRActive, not recruiting
NCT06682793
(DENALI-1)
Phase I/II, multi-center, single-arm, n = 240Unresectable/metastatic solid tumors expressing EGFR and with loss of HLA-A*02 expression; requires germline HLA-A*02 heterozygosityAllogeneic logic-gated TmodTM CAR-T (A2B395)Phase I: DLT, AEs, RP2D
Phase II: ORR
Not yet recruiting
NCT06383507Phase I, single-center, single-arm, n = 18Relapsed/refractory CD70 positive solid tumorsAllogeneic CAR-T against CD70AEsRecruiting
NCT04847466Phase II, single-center, single-arm, n = 55Recurrent/metastatic gastric or head and neck cancer with prior chemotherapy and anti-PD-1 therapy Allogeneic CAR-NK cells against PD-L1, combined with pembrolizumabORRRecruiting
TIL therapies
NCT03645928
(IOV-COM-202)
Phase II, multi-center, multi-cohort, non-randomized, n = 245Advanced melanoma, HNSCC, and NSCLC; recurrent/metastatic HNSCC in cohort 2A without prior checkpoint inhibitorsAutologous TIL (LN-145, lifileucel) combined with pembrolizumabORR, grade ≥ 3 AEsRecruiting
NCT05902520Phase I, single-center, multi-arm, randomized, n = 18Unresectable/metastatic solid tumorsAutologous double-positive (CD39+, CD103+) CD8+ TIL (AGX148) with or without siRNA PD-1 modulationAEsRecruiting
NCT06236425Phase I, single-center, multi-arm, non-randomized, n = 15Recurrent/metastatic HNSCC with previous progression on pembrolizumab or pembrolizumab/platinum chemotherapyAutologous TIL leveraging single-cell sorting of patient-specific neoepitope-reactive T-cells (TBio-4101), combined with pembrolizumabAEsRecruiting
* Status as of 18 April 2025. Abbreviations: AE, adverse event; CAR, chimeric antigen receptor; DLT, dose-limiting toxicity; EGFR, epidermal growth factor receptor; HLA, human leukocyte antigen; HNSCC, head and neck squamous cell carcinoma; HPV, human papillomavirus; MAGE, melanoma antigen gene; NSCLC, non-small cell lung cancer; ORR, objective response rate; PD-1, programmed cell death protein 1; PD-L1, programmed cell death ligand 1; PRAME, preferentially expressed antigen in melanoma; RP2D, recommended phase 2 dose; siRNA, small interfering ribonucleic acid; TCR, T-cell receptor.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sun, F.; Colevas, A.D. Update: Immunotherapeutic Strategies in HPV-Associated Head and Neck Squamous Cell Carcinoma. Viruses 2025, 17, 712. https://doi.org/10.3390/v17050712

AMA Style

Sun F, Colevas AD. Update: Immunotherapeutic Strategies in HPV-Associated Head and Neck Squamous Cell Carcinoma. Viruses. 2025; 17(5):712. https://doi.org/10.3390/v17050712

Chicago/Turabian Style

Sun, Fangdi, and A. Dimitrios Colevas. 2025. "Update: Immunotherapeutic Strategies in HPV-Associated Head and Neck Squamous Cell Carcinoma" Viruses 17, no. 5: 712. https://doi.org/10.3390/v17050712

APA Style

Sun, F., & Colevas, A. D. (2025). Update: Immunotherapeutic Strategies in HPV-Associated Head and Neck Squamous Cell Carcinoma. Viruses, 17(5), 712. https://doi.org/10.3390/v17050712

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

Article Metrics

Back to TopTop