Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Immunol., 29 August 2025

Sec. Cancer Immunity and Immunotherapy

Volume 16 - 2025 | https://doi.org/10.3389/fimmu.2025.1631258

This article is part of the Research TopicOvercoming Resistance to Immune Checkpoint Inhibitors: Mechanisms and Strategies in Cancer TherapyView all 5 articles

Targeting archetypes of viral-driven cancers with immunotherapy: a perspective on immunogenicity within the tumor microenvironment

  • 1Department of Otolaryngology—Head and Neck Surgery, Stanford Cancer Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
  • 2Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
  • 3A*STAR Skin Research Lab, Agency for Science Technology and Research, Singapore, Singapore
  • 4Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore

Viral etiologies of cancers have been widely studied for tumorigenesis and in recent years, widely recognized for their potential influence on immune regulation and response to immune checkpoint blockade (ICB). Here, we review the current understanding of how various oncogenic viruses are related to tumor immunogenicity and the tumor immune microenvironment. The present work also highlights the distinct features of these viral-driven cancers, that can be largely prognostic for better patient survival and response to ICB. On the other hand, there are also several commonalities in which these cancers acquire resistance against conventional immunotherapy. Finally, we discuss our perspectives to address the existing conundrums to gain clearer insights on how the interplay between anti-viral and anti-tumor immunity can be exploited to develop novel therapeutic interventions.

1 Introduction

Across different types of cancers, some of the key determinants of successful cancer immune checkpoint blockade (ICB) therapy often include tumor immunogenicity and the tumor microenvironment (TME) (1). Immunogenicity refers to the ability to induce adaptive immune responses influenced by the presence of tumor-derived antigens, and among these, neoantigens have gained increasing attention for its potential in enhancing anti-tumor immunity (2, 3). Neoantigens are novel peptides arising from somatic mutations such as single-nucleotide variants (SNVs), base insertions and deletions (INDELs) and gene fusions (4). Importantly, these unique peptides are highly immunogenic as they are not present in normal cells and thus are not subjected to central immune tolerance (5). A high tumor mutational burden (TMB) is generally associated with increased likelihood of generating immunogenic neoantigens, which is well-recognized as a strong predictor for response to ICB across cancer types (68). However, this relationship does not hold true across all cancer types. Cancers such as the viral-driven Merkel cell carcinoma (MCC) have low to moderate TMB yet reported to have high objective response rates (ORRs) to ICB (1, 911). In addition, the patient-specific nature of neoantigens results in a variable pre-existing T cell landscape which may also influence the eventual responses to ICBs (12). Characteristics of the T cell receptor (TCR) repertoire, such as clonal diversity, expansion and convergence may also serve as potential predictors of ICB treatment outcomes (13). It is noteworthy that TCRs generally have a much higher affinity for viral antigens than for tumor-related antigens (14). This critical finding could explain the inadequate efficacies of tumor antigen vaccines in priming T cells within the tumor-bearing host while at the same time providing a strong motivation for engineering TCR-based therapies for viral-associated antigens in cancers.

At the same time, understanding how the immune landscape is shaped within the tumor microenvironment (TME) plays a critical role in guiding immune intervention and developing innovative therapeutic strategies to target different tumor types. The immunological state of the TME can be broadly classified as “hot” or “cold”, which further modulates immunogenicity over the course of tumor progression. “Hot” tumors are highly inflamed and usually characterized by high infiltration of immune cells, particularly cytotoxic T cells, and increased expression of inflammatory markers such as IFN-γ and TNF-α (15). Typically, “hot” tumors are also associated with a higher TMB leading to elevated neoantigen presentation and better responses to ICBs (16). Moreover, certain cancers with “hot” TMEs can contain ectopic lymphoid aggregates commonly known as tertiary lymphoid structures (TLS) (17). These TLS are typically characterized by a central B-cell zone surrounded by a T-cell rich region, along with dendritic cells (DCs) and high endothelial venules (18). Depending on its maturation state, TLS can also contain activated B cells capable of differentiating into plasma cells that secrete high-affinity antibodies, which can enhance the anti-tumor immune response (19). In contrast, “cold” tumors are characterized by poor immune infiltration, lower TMB and PD-L1 expression. A “cold” TME is dominated by immunosuppressive cytokines such as IL-10 and TGF‐β, rendering them more unresponsive to existing immunotherapies (20). While this phenotype may vary across different cancer types, “cold” tumors generally represent an immune desert which do not respond well to ICBs. As such, various strategies are being explored to alter the immunological “temperature” of cold tumors and improve their response to immunotherapies (21).

It is important to note that not all high TMB tumors are immunogenic or “immune-hot”. Consequently, such associations can only be applied to specific tumor types (22). Of interest, a recent meta-analysis reported cancers associated with HPV and HBV/HCV can be prognostic for better overall survival and even higher ORRs to ICBs (23). However, there is limited understanding on how viral factors modulate anti-tumor responses within the tumor immune landscape. As such, the present review discussed the underpinning research to identify how oncogenic viral factors play an integral role in influencing tumor immunogenicity and TME. Here, we focused on several cancer types that show great potential with ICB therapies and outlined common distinct traits across viral-driven cancers that impact anti-tumor immunity (Summarized in Figure 1). Ultimately, we deem that there is great potential to advance the immune oncology field towards harnessing anti-viral immunity across multiple types of viral-driven solid tumors.

Figure 1
Diagram comparing viral-associated and non-viral cancers. The left side illustrates viral-specific CD8 T cell response, persistence of viral antigens, and viral integration into the host's genome. It also shows tertiary lymphoid structures and viral-specific humoral response. The right side depicts tumor-specific CD8 T cell response, tumor neoantigens, tumor mutational burden, and microsatellite instability. Both sides include common processes such as the dichotomy of TAMs, PD-L1 expression and downregulation of antigen presentation machinery.

Figure 1. Summary of the key similarities and differences of the tumor immune microenvironment between viral and non-viral cancers. Factors generally more strongly associated with viral-associated cancers are depicted in yellow (Left), and those more strongly associated with non-viral cancers are depicted in green (Right). Similarities are shown in pink. In brief, viral-specific CD8 T cells are robust to mount a satisfactory anti-tumor immune response. Viral integration into host genome can influence immunogenicity and at the same time, potentially implicates the development of intrinsic tumor immune evasive mechanisms. While the TME can be favorably conditioned by the presence of TLS, there is a functional dichotomy of immune cells. In particular, tumor-associated macrophages (TAMs) and B cells can acquire either anti- or pro-tumoral immunoregulatory phenotypes to shape the immune landscape. Schematic illustration created with BioRender.com.

2 Merkel cell carcinoma as a unique viral-driven neuroendocrine tumor

Merkel cell polyomavirus (MCPyV) has recently emerged as an oncogenic virus, accounting for at least 80% of the Merkel cell carcinoma (MCCs) cases worldwide. MCC is a rare and aggressive type of skin cancer with an exponentially increasing incidence rate between 2000 and 2013, highlighting the growing clinical importance of this disease (24). MCC is divided into 2 distinct molecular subtypes. Virus-negative MCC (MCPyV- MCC) is characterized by a higher mutational burden, with most of them resembling UV-induced mutations, while MCPyV+ MCC expresses primary oncogenic drivers small T (sT) and long T (LT) antigens (25). TMB was reported to be higher in MCPyV- MCC tumors as compared to MCPyV+ tumors (26). In fact, MCPyV-MCC tumors can harbor more tumor neoantigens than cutaneous melanoma or NSCLC (27). In a case study of a particular MCPyV-MCC patient, the presence of neoantigen-specific Th1 cells were detected after anti-PD-L1 therapy but intriguingly, no neoantigen-specific CD8 T cells were detected (28).

2.1 Immunogenicity of viral associated Merkel cell carcinoma does not correlate with tumor mutational burden

The high immunogenicity of MCPyV+ MCC tumors could perhaps be better explained by the presence of MCPyV antigens and viral-reactive T cells in ICB responders. The presence of KLL- (dominant epitope of the MCPyV common T-ag) specific intratumoral T cells favored better disease-specific survival and lower risk of metastatic disease (29). Frequencies of MCPyV-specific CD8 T cells also correlated with T-Ag antibody titers alongside increasing tumor burden (30, 31). Of interest, these viral specific CD8 T cells also highly expressed both PD-1 and TIM-3 which can be inhibited to improve T cell activation as demonstrated in vitro (31). Moreover, a traceable increase in T antigen-specific CD8 T cells was detected in the peripheral blood of ICB responders, which was associated with improved progression-free survival. The authors also further demonstrated that these viral reactive T cells can be expanded with robust killing capacity to target MCC tumor cells in vitro (32, 33). Another study critically uncovered that high baseline frequencies of circulating viral-specific CD8 T cells, but not intratumoral CD8 T cells, was associated with beneficial responses to ICBs, which may represent a potential application as a predictive biomarker for immunotherapy. Amongst MCPyV specific T cells, further CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing) analysis revealed intratumoral CD8 T cells to be terminally exhausted while circulating CD8 T cells highly expressed TCF7, CD62L and LEF1, resembling a more functional stem-like memory phenotype (34). Notably, tumor regression was achieved in a case reported where MCPyV-specific T cells were adoptively transferred (35). Collectively, these findings highlighted the potential of using other TCR-dependent therapies such as adoptive T cell therapy and tumor vaccines to complement the existing success of ICBs in treating MCCs.

Alongside the presence of these viral antigens, MCPyV+ MCC can promote immune evasion via multiple mechanisms. Like many other cancers, modulation in expression levels of MHC and other molecules of the antigen presentation machinery (APM) are widely implicated in the escape of immune surveillance in MCPyV+ MCC (36). Ritter et al. demonstrated that APM genes are epigenetically silenced by histone hypoacetylation, highlighting the potential use of HDAC inhibitors as therapeutic primers for response to ICBs (37). Likewise, the STING pathway (innate immune regulator stimulator of IFN genes) was also revealed to be largely silenced in MCC, implicating the downregulation of NF-κB signaling. The reactivation of STING could reconfigure the “cold” TME of MCC to enhance immune infiltration and surveillance (38, 39). Additionally, MCPyV sT was uncovered to interfere with type I IFN signaling by either direct transcriptional repression or histone modifications (40). Overall, it is evident that MCPyV can enhance immunogenicity and at the same time, drive immune tolerance within the TME of MCC.

2.2 Exploiting the favorable immune landscape of MCPyV+ MCCs for immunotherapy

Interestingly, MCC can be well-infiltrated with both effector and central memory T cells (41). In fact, MCPyV positivity in MCC is associated with greater infiltration of T cells and macrophages within the tumor microenvironment, which consequently contributes to favorable survival outcomes (42). Additionally, a gene signature derived from a comprehensive characterization of pro-inflammatory γδ T cells was found to be a potential predictor of improved survival and responses to ICBs (43). While most immunological studies on MCC were heavily dependent on phenotypic profiling of patient tissues, the engraftment of MCC tumor fragments into immuno-deficient NSG mice has been explored as a functional patient-derived xenograft (PDX) model. Comparing the phenotype of tumor-infiltrating lymphocytes (TILs) before engraftment to those 5 days post implantation into the mice, there is an increase in activated effector T cells and a reduction of FOXP3+ regulatory T cells (Tregs). The selective depletion of CD25+ T cells intriguingly enhances tumor growth in vivo, suggesting the presence of active T cell-mediated anti-tumor immunity that limited the initial growth of the engraft in the PDX model (41). However, there remains no evidence of Treg mediated impedance of anti-tumor immunity within MCC. Likewise, there could be potential involvement of humoral immunity in MCC. While antibody reactivity to MCPyV capsid protein VP1 is seemingly ubiquitous within the general population, the abundance of antibodies recognizing MCPyV tumor-associated oncoproteins (T antigens) was found to correlate with tumor burden. Importantly, the increase in titers of such antibodies precedes recurrence and metastatic progression, highlighting their potential as biomarkers for clinical utility (30). Future studies should be focused on further dissecting the understanding of B cells within MCC using deep immune-profiling to provide potential insights on how humoral immunity can be further exploited to improve conventional therapies.

2.3 Future prospects for immunotherapy in MCPyV+ MCC

Although ORRs reported with ICB treatment of MCPyV+ MCC is undeniably promising (44, 45), the direct interplay between MCPyV and the host’s immune system is not clearly understood. Moreover, treatment responses are greatly influenced by immune evasion mechanisms and the composition of the TME (46). Hence, further investigation into these factors is essential to uncover the novel biological insights and translatable therapeutic options, particularly for treatment-resistant patients. Interestingly, PD-1 expressed on MCC tumor cells was also demonstrated to be a growth promoter driving mitochondrial respiration and tumor cell proliferation, which can be reversed by conventional inhibition of the PD-1/PD-L1 axis (47). Considering the rare demographics of MCC, addressing further complex biological questions could be impacted by the limited motivation of scientists and oncologists. The fact that PD-L1 correlated with both MCPyV positivity and the infiltration of TILs (48) regardless highlights the value of leveraging MCC as a suitable model to further dissect new mechanisms underlying the cross talk between anti-viral and anti-tumor immunity in future studies.

At the same time, there are also some interests in developing tumor vaccines to target MCPyV+ MCC, though the current progress from such studies are largely preclinical. Early studies from more than a decade ago have already demonstrated the effectiveness of using DNA vaccines to generate LT-specific CD8 T cell responses in syngeneic tumor mice models (49, 50). Truncated LT can also be incorporated into monocyte-derived dendritic cells, which act as antigen presenting cells for the stimulation of autologous T cells (51). A more recent study further improvised a fusion design that integrates LT to LAMP1 (lysosomal-associated membrane protein 1), enhancing antigen presentation to potentiate antigen-specific CD4 T cell responses and humoral responses in vivo (52). Likewise, there are similar studies to evaluate the efficacy of DNA vaccines encoding sT, mounting antigen-specific CD8 T cell responses (53). However, it should be emphasized that the majority of these vaccine studies were heavily reliant on the overexpression of T-ag in cutaneous B16F10 melanoma cells prior to inoculation into immunocompetent mice. The use of alternative transgenic mouse models for MCC has been recently developed, which should be explored for future immunological studies (54). To delve deeper into the TME for new discoveries, one can also start with interrogating publicly available single cell sequencing of MCC tumors or perform immune gene deconvolution within bulk transcriptomics datasets (55, 56). With clear evidence of MCPyV-associated humoral immunity in MCC (30), another plausible hypothesis could be the potential involvement of B cells in the co-stimulation of T cells within the tumor-bearing host. However, to further dive deeper into understanding such immune regulation would require an immunocompetent mouse model – one that is currently lacking in the field.

3 Distinct viral-associated immune landscapes in hepatocellular carcinoma

Hepatocellular carcinoma (HCC) is the leading primary malignancy of the liver and the third most common cause of cancer-related mortality globally (57). 5-year survival is at a dismal 10-20% despite treatment. It typically arises in the background of chronic liver disease and cirrhosis. Risk factors of HCC include chronic viral (HBV/HCV) hepatitis, aflatoxin exposure, alcoholism, and metabolic syndrome, which features metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH). Globally, HBV is responsible for more than 50% of HCC cases, especially in endemic regions such as Asia and sub-Saharan Africa (58). Notably, MASLD/MASH accounts for half of the new HCC cases in the US and, with the rollout of universal HBV vaccination, this trend is expected to take predominance in both industrialized and developing societies alike (59).

3.1 Etiology of HCC impacts immunogenicity and immune responses in HCC

HBV is a partially double-stranded DNA virus from the Hepadnaviridae family. It integrates into the host genome and expresses viral proteins such as HBsAg and HBx, contributing to oncogenesis via TERT or MLL4 (60) and to the modulation of immune surveillance (61). In contrast, HCV is a positive-sense RNA virus from the Flaviviridae family that does not integrate but causes HCC through sustained inflammation, fibrosis, and immune perturbation (62). Both viruses can shape distinct immune microenvironments, affecting tumor development and treatment response. As such, the immunogenicity of HCC can vary with its underlying cause. In HBV-related HCC, the presence of viral antigens, including HBsAg, HBcAg, and HBx, theoretically provides targets for immune recognition of the tumor; but it is often HBx that remains as the only HBV protein detectable in tumor cells (63). Notably, HBx promotes tumor invasion and metastasis in a mechanism unrelated to diminished immunogenicity (63). Virus-specific CD8+ T cells are detectable in peripheral blood and TILs in HBV-HCC patients (6466). However, these T cells are often functionally exhausted due to persistent antigen stimulation, high expression of inhibitory receptors (e.g., PD-1, LAG-3), and metabolic dysregulation (67). Similarly, another inhibitory receptor, TIGIT was also upregulated by TILs and the co-inhibition with PD-1 could restore immune activation ex vivo (68). Together, these evidence offers an explanation towards the limited efficacy of HBV-specific T cell therapy in HCC control (6972). While HCV-related HCCs also present viral antigens capable of stimulating T cell responses (73), there is limited literature regarding the presence of HCV-specific T cells in the TILs of HCV-HCC patients. MASLD-HCC, however, is more reliant on neoantigen-driven immunity. Notably, TMB is often higher in non-viral HCCs (especially MASLD-HCC), potentially increasing the number of neoantigens presented by tumor cells (74). Despite a higher TMB, MASLD-HCCs do not uniformly exhibit robust immune activation, suggesting that antigen presentation or immune cell priming may be impaired.

3.2 Influence of the tumor microenvironment may play a larger role than immunogenicity during treatment of HCC

The composition and functional state of the TME in HCC are shaped by its etiology and play a crucial role in determining prognosis and response to immunotherapy. A detailed analysis of lymphoid and myeloid populations reveals both conserved and aetiology-specific features. In HBV-related HCC, the TME is enriched with immunosuppressive populations, especially Tregs, tumor associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) (67, 7577). Tregs can contribute to local immunosuppression and correlated with worse prognosis in HCC patients (78). CD8+ T cells are often excluded from the tumor core, a phenomenon correlated with TGF-β signaling and the presence of activated hepatic stellate cells (79). TAMs, on the other hand, are highly heterogeneous in origin or phenotype in HBV-HCC but typically possess immunomodulatory characteristics (80). In addition, IL-10-expressing B cells, present in HBV-HCC, suppresses CD4+ T cell activity (81). Lesser is understood about the tumor microenvironment of HCV-HCC tumors. A prominent feature uncovered is that chronic viral antigen stimulation drives CD8+ T cell exhaustion with elevated PD-1 and TIM-3 expressions (67, 82). These T cells often exhibit a downregulation of T cell activation signatures compared to those in HBV-HCC, which correlates with a reduced immune infiltration in HCV-HCC tumors (83). Of note, in chronic HCV-infected patients, NK cell expression of TIM-3 and CD38 may be an early sign of impending HCC (84). While there are studies that did not find differences in the proportion and phenotypes of TILs in HCV-HCC compared to other etiologies (85, 86), further investigations would be warranted, particularly in the understanding of immunoregulatory cell types within the TME.

Unlike viral-driven HCCs, the immune landscape of MASLD-HCC is markedly distinct. Early-stage MASLD is accompanied by increased macrophage and lymphocyte infiltration, where these macrophages are predominantly proinflammatory, engaging in TNFα and IL-6-based acute inflammatory responses (87). Importantly, a recent seminal paper showed that the activated PD-1+ CD8+ T cells in MASH do direct damage and resulted in impaired immune surveillance and HCC development (88). Prophylactic anti-PD1 ICI treatment in MASH mice resulted in increased, rather than decreased, HCC tumorigenesis (88). Back-to-back published, it was also shown that these activated T cells exhibit resident, effector, and exhausted characteristics, and perform killing functions independent of antigen presentation, resulting in liver damage commonly seen in MASH (89). In addition, there may be dominance of TREM2+, MARCO+, and CD206+ macrophages within tumor and peritumoral regions in MASLD-HCC (90). These macrophages have high lipid, impaired phagocytosis, and produce cytokines such as IL-6, IL-10 and TGF-β (91, 92). Moreover, CD8 T cells are often sparse and localized to the tumor margin, co-expressing PD-1 and CD39 with an exhausted phenotype (88, 93). Interestingly, cancer-associated fibroblasts (CAFs) are also enriched in inflammatory gene signatures and produce IL-34, which may promote Treg infiltration and suppress CD8+ T cell activity, especially in non-viral HCC (94, 95). These findings have highlighted the importance of inflammation in MASLD-HCC transition, and the distinct temporal roles of different immune cell populations in the liver during disease development and progression.

3.3 Understanding viral/non-viral immune evasion mechanisms for future therapeutic directions

Immune escape is central to HCC progression and varies according to etiology. In HBV-related HCC, viral proteins like HBx can downregulate MHC class I molecules and IFN-γ expression, impairing CD8+ T cell recognition and inducing their apoptosis (96, 97). Silencing HBx with a 5’-triphosphate siRNA can reduce the differentiation of Tregs and MDSCs (98). In HCV-related HCC, immune evasion arises from T cell exhaustion and altered antigen-presenting cell (APC) function (99). DCs in HCV-HCC also exhibit impaired IL-12 production, diminishing effective T cell priming (100). As such, engineering CAR-T cells and CAR-NK cells against known HCC targets like GPC3 might represent promising strategies to overcome these potential immune evasion mechanisms within the TME (101105). In MASLD-HCC, immune suppression is contrastingly driven by metabolic dysfunction. Lipid accumulation in hepatocytes and immune cells impairs immunogenicity (106). TAMs, (including peritumoral macrophages and monocytes), upregulate PD-L1 (107, 108). IL-6 secreted by TAMs contributes to STAT3 activation in tumor cells, reinforcing immune resistance (109). Still, anti-PD-1/PD-L1 ICB therapies have transformed the treatment landscape for advanced HCC (110, 111). The profound clinical question is, therefore, what predicts the response to ICBs. While a recent study shows that poor Atezo/Bev response is associated with high glypican-3 (GPC3) or alpha-fetoprotein (AFP) expression, no correlation was seen between the expression of PD-L1 and Atezo/Bev response – contrary to the data in non-small cell lung cancer or melanoma (112). Notably, etiology may impact therapeutic outcomes. A recent meta-analysis of 3,739 patients shows that non-viral HCC does not seem to benefit, or benefit less, from ICBs compared to HBV-HCC, yet the heterogeneity of the trials is high (113, 114). This underscores the need to dissect the immunological nuances across HCC etiologies.

Indeed, targeting the immunosuppressive TME also represents a promising avenue for improving outcomes in HCC. Combination therapies that integrate ICB with multi-kinase inhibitors (e.g., cabozantinib) have shown promise in clinical trials, especially for the HBV-HCC subgroup (115, 116). Furthermore, several trials also investigated the potential of an oncolytic viral vaccine, Pexa-Vec as means to completement conventional HCC therapies (117119). These regimens may leverage immunogenic cell death and inflammation to enhance ICB response. Myeloid-targeted therapies such as anti-TREM2, anti-MARCO, and CSF1R inhibitors are also under preclinical investigations, which may synergize with conventional ICBs (120122). However, inhibiting TREM2 or depleting TREM2+ macrophages may have repercussions for their protective functions particularly at the earlier stages of liver damage (123125). Similar dual protective-immunosuppressive functions also hold in MARCO (126129). Therefore, different studies are disconnected in terms of the temporal progression of myeloid functions, where the proinflammatory and anti-inflammatory roles of myeloid cells may both promote HCC tumorigenesis and progression, depending on the rather heterogeneous temporal sequence of events. Importantly, new platforms including organoids can incorporate autologous immune cells and stromal components to offer powerful translational tools for biomarker discovery and drug screening in HCC (130). Their ability to retain patient-specific TME features, including lipid dysregulation and immune cell crosstalk, makes them ideal for preclinical testing of aetiology-specific immunotherapies (131). Future work should focus on refining preclinical models, validating findings in clinical cohorts, and integrating spatial and multi-omics approaches to fully map the immunologic heterogeneity of HCC (132). Stratifying patients by etiology and immune profile may enable more effective, personalized immunotherapeutic interventions.

4 Role of human papillomaviruses in carcinogenesis

Human papillomaviruses (HPVs) are a subclass of papillomaviruses, which are non-enveloped, icosahedral, double-stranded DNA viruses (133, 134). Although over 200 strains of HPV have now been identified, they can be generally stratified into high and low-risk HPVs – representing two subgroups of HPVs that are either overrepresented or rarely present in HPV-positive (HPV+) cancers (135). Of note are HPV16 and HPV18, the two most common high-risk strains of HPV in HPV+ cancers (136, 137). While most HPV infections are eventually cleared, chronic infections, particularly with high-risk strains, can result in the development of a range of anogenital and oropharyngeal cancers (OPCs). HPV is by far the leading cause of most anogenital cancers, accounting for 40-95% of vulvar, vaginal, penile and anal cancers and virtually all cervical cancers (137, 138). Furthermore, it accounts for around 70% of all OPCs and around 20% of other head and neck squamous cell carcinomas (HNSCCs) (139). HPV viral oncogenes E6 and E7 have been shown to be necessary and sufficient for cellular immortalization and transformation (140142). Briefly, E6 acts to degrade p53 by associating with the E6-associating protein (E6-AP) – a canonical E3 ubiquitin ligase, coordinating with E7, which blocks binding of key cell cycle checkpoint proteins including pRb, p21 and p27, together causing deregulated cell cycling and genomic instability. This instability has been thought to be the main contributor to the slow kinetics of carcinogenesis, through gradual accumulation of genomic aberrations coupled with uncontrolled cell cycling.

4.1 HPV-associated immunogenicity may vary with host genome integration

Our discussion here will focus on HNSCCs, in which responses to immunotherapy have met much more variability. This is in contrast to cervical cancers – which are consistently reported to have high ORRs to ICBs (143146). An important difference to note is that while virtually all cervical cancers are viral associated, only a subset of HNSCCs are HPV positive. HPV+ HNSCCs are generally associated with better prognoses compared to HPV- HNSCC, even after controlling for confounding factors such as tumor stage, smoking status and alcohol usage (147150). Furthermore, they demonstrate enhanced radio and chemotherapy responsiveness compared to HPV- HNSCC (150). However, there are mixed results when comparing the efficacy of immune checkpoint blockade (ICB) between HPV+ and HPV- HNSCC cases, with some studies demonstrating enhanced response amongst HPV+ cases (149, 151, 152), and some demonstrating no difference (153). This discrepancy might be attributed to how the levels of PD-1/PD-L1 expression cannot be trivially disentangled from the level of immune infiltrate. Notably, there are also contradicting reports of PD-L1 and PD-1 levels and viral status, with some reporting increased levels in HPV+ HNSCC (154157) and others reporting no correlation (151, 158160). This is probably indicative of heterogenous cohorts and implies that PD-L1 levels are also influenced by other tumor intrinsic/extrinsic factors not related to viral status. Nevertheless, the expression of PD-L1/PD-1 stratifies responders to ICBs in HNSCC, which can be, in part, causally linked to viral status and immune infiltration (153, 155, 161).

It is important to note that in HPV+ cancers, the HPV genome can exist as an episome, integrated into the host genome or a mixture of both, although in most HPV+ cervical cancers (~80%), they are stably integrated into the host genome (162). This aberrant integration event often occurs within the coding region of the E2 gene, which codes for an important transcription factor that is essential in the careful regulation of the expression of the oncogenic proteins E6 and E7 (163, 164). Sustained E6 and E7 expression are essentially required for the establishment and persistence of HPV+ cancers (165, 166) and consequently, the genomic landscapes of HPV positive and negative cancers are vastly different (167171). There are contrasting studies on the impact of viral status on mutational loads, with some demonstrating no difference (169), and some showing greater mutational burden in HPV- HNSCCs (172, 173). Despite this, the mutational landscape of HPV+ and HPV- HNSCCs vary widely due to the origins of these genomic aberrations.

Due to the presence of viral antigens and increased immune cell infiltration into the tumors, HPV+ HNSCCs are generally thought to be more immunogenic (174). Interestingly, the viral genome is often maintained in episomes or a mixture of integrated and episomal genomes in HPV+ HNSCCs (175, 176). A recent study looking at integration events in HPV+ HNSCCs revealed that integration-negative tumors correlated with an increased immune signature, specifically T, B and NK cells compared with integration-positive HPV+ HNSCCs (177). Furthermore, emerging evidence has demonstrated constant expression of many early viral genes when the genome is maintained episomally, drawing the link between integration-negative HPV+ HNSCC and increased viral antigenic presence to influence viral-specific immune responses (178, 179). Integration events also play a role in modulating the genomic landscape, gene expression profiles and even epigenetic signatures (180) within HPV+ HNSCCs, driving differential responses to various therapies and correlating with prognosis (181). Such events presumably also contribute to divergent TMEs between integration positive and negative HPV+ cancers (182). It is noteworthy that HPV+ HNSCCs rarely exhibit oncogenic/tumor suppressor driver mutations (183, 184), and instead are completely dependent on the E6/E7 viral oncogenes (165, 166). In the context of non-viral OPCs, tobacco and alcohol overuse are strong contributors of overall TMB and immunogenicity (168, 173, 185). Notably, immunogenicity and immune cell infiltration of HPV- OPCs vary widely, depending on neoantigen load and driver mutations accumulated (161, 186). Nevertheless, a higher TMB in HNSCC, independent of viral status, is linked to superior immunotherapy responses (161, 171, 187).

4.2 HPV-associated HNSCCs are largely influenced by their microenvironment

Similar to HCCs, the role of the tumor microenvironment of HNSCCs appears to be a stronger deterministic factor for ICB responses rather than immunogenicity. A high degree of intratumoral immune cell infiltrate may be a key factor in HPV+ individuals’ improved response to conventional treatment and favorable clinical outcome. In HNSCCs, the immune landscape of HPV+ tumors had considerably more infiltrating IFNγ+ CD8+ T lymphocytes, DCs and more proinflammatory cytokines within the milieu (157). Multiplex immunofluorescence coupled with immune-related gene expression profiling revealed that compared to HPV- OPCs, HPV+ lesions were more heavily infiltrated by CD8+ T cells, with an increase in various subsets of T cells including cytotoxic and exhausted cells. Spatially, these T cells appeared in much closer proximity to tumor cells, CD163+ macrophages and FOXP3+ Tregs (154). This overall suggests a stronger activation of immune pathways and an inflamed TME. Additionally, Eberhardt et al. identified CD8 T cell clones specific to a range of E proteins, and further characterized a subset of HPV-specific PD-1+ stem-like population capable of proliferating upon exposure to antigen in vitro (188). This study presents evidence of the ability of these T cell clones to maintain cytotoxic responses under persistent antigenic exposure and ultimately, alludes to the amenability of HPV+ HNSCC to respond to PD-1 checkpoint blockade.

While the differentiation trajectories of CD8 T cells are relatively similar between HPV+ and HPV- HNSCCs, there could be viral-driven divergence in the polarization of CD4 T cells and B cell subsets (189). Leveraging TCGA datasets, it was reported that HPV positivity correlated with increased levels of CD4 T follicular helper (Tfh) and Tregs (190). The CD4 T cell compartment in HPV+ samples also showed skewing towards an inflammatory Th1 response. Furthermore, these T cells presented with a higher expression of a range of exhaustion-related molecules including LAG3, PD1, TIGIT and TIM3, which counterintuitively correlated with improved survival, presumably suggesting an active T cell response. Importantly, this correlation was not seen in HPV- samples, indicating a viral-specific T cell response. This ‘T-cell-inflamed’ phenotype points towards the potential of immune checkpoint inhibitor blockade as a HPV+HNSCC-specific treatment. While inflammation could drive immune tolerance within the TME, it still remains unclear if Tregs are significantly enriched in HPV+ HNSCC compared to HPV- HNSCC. It appears that there is heterogeneity in localization of these Tregs, which some studies observing an increased Treg infiltrate in the stromal compartment (191), while others demonstrated enriched Treg signatures within the intraepithelial compartment (192). Studies have also reported correlation of a higher level of Treg infiltration with better prognosis in certain HNSCC subsets (193, 194). While Treg-dependent immune suppression is associated with poor prognosis in some cancers, the paradoxical opposite observed in HPV+ HNSCCs is thought to be reflective of an overall pro-inflammatory immune microenvironment, promoting general CD4/8 T cell infiltration (190). This immune ‘hot’ environment is speculated to be a virus-dependent phenomenon.

Shifting focus to viral-driven humoral immunity differences, while CD20+ B cells were enriched in HPV+ HNSCC, studies did not find correlation between B cells and patient survival (192, 195). However, the TME of HPV+ HNSCC is characterized by active HPV-specific intertumoral B cell responses and antibody production (196). While antibodies against viral proteins such as E2, E6 and E7 were detected, E2-specific responses appeared most dominant, based off IgG titers. The authors also demonstrated the preferential localization of these B cells (and antibody-producing cells) to the tumor stroma, where they form germinal center-like clusters indicative of an activated B cell phenotype. While the link between HPV-specific antibodies and enhanced anti-tumor immunity is unclear, studies have evidenced a correlation between anti-HPV antibodies and survival benefit (197, 198). Another study revealed that germinal center B cells were enriched in HPV+ HNSCC, while HPV- HNSCC had fewer total B cells and presented in a non-germinal center state (189). Further TCGA analysis also demonstrated enriched signatures of plasma and memory B cells in HPV+ HNSCC, which correlated with higher CXCL13 production from CD4+ T cells (199). This correlated with better prognosis, indicating viral-specific mechanisms driving preferential induction of TLS that presumably contributes to enhanced antitumor responses as a peripheral consequence of persistent HPV infections. Using a murine model of HPV+ HNSCC, Kim et al. demonstrated an expansion of memory B cells, plasma cells and antigen-specific B cells upon radiotherapy or PD-1 blockade. Furthermore, IgM and IgG serum levels were elevated post PD-1 treatment in a cohort of HPV+ HNSCC patients that showed positive clinical response, strongly suggesting that a similar mechanism of B cell expansion correlated with better response to PD-1 blockade (200). Collectively, the notion that B cells may play a key role in the immune-mediated eradication of HPV-driven HNSCCs is promising. It is therefore imperative to gain a better mechanistic understanding of this link, perhaps through established murine mouse models of HPV+ HNSCC, informing of potential vaccination strategies to enhance B cell responses against HPV.

Focusing on the innate lymphocyte compartment, particularly NK cells due to their heavy involvement in early anti-tumor and metastatic responses (201), a pan-cancer analysis uncovered HNSCCs to have one of the highest mean CD56dim NK cell infiltration particularly in HPV+ HNSCCs compared to HPV- HNSCC (202, 203). This phenomenon could perhaps explain why lower MHC class I expression is correlated with favorable prognosis in HPV+ HNSCC but a poor prognosis in HPV- HNSCC (204, 205). NK cell activity is also modulated by the balance between activating and inhibitory ligands present within the TME (206). Of interest, HPV+ HNSCC trended towards a higher HLA-G expression than HPV- samples (192). HLA-G is a known inhibitory MHC molecule is recognized by KIRs and LILRB1/2 expressed on NK cells, which represents a possible NK inhibitory axis that is differentially modulated based on viral status of HNSCC (207). Interestingly, an intraepithelial ILC1-like NK state was also described in HNSCC independent of HPV status. These CD49a+CD103+ cells represented a tissue-resident (trNK) phenotype that co-expressed key cytotoxic signatures indicative of its ability to kill tumor cells (208). It would be of interest to dissect the impact of viral presence on the tissue-residency status of NK cells, since these trNKs represent a potentially immunomodulatory subset of NKs (209) that maintain the activation status of various adaptive immune cells, including CD4+ and CD8+ T cells (210).

While HNSCCs possess favorable features of adaptive immunity, the regulatory role of myeloid cells may play a critical influence as well. Increased infiltration of CD68+ macrophages is associated with poorer prognoses in HNSCC (211). Notably, there is an increased density of CD68+ macrophages in transcriptionally active HPV+ HNSCC compared to HPV- HNSCC patients. M1-like macrophage inflammatory signatures were also enriched in HPV+ HNSCC alluding to unique viral-driven mechanisms of modulating monocyte infiltration and macrophage polarization (190, 212). Similarly, the functionality of DCs within the TME can be influenced by viral-driven factors. Despite showing no correlation between the abundance of infiltrating plasmacytoid dendritic cells (pDCs) and viral status in HNSCC, pDCs have a reduced capacity to produce IFNα upon toll-like receptor activation in HPV-negative samples but remain uncompromised in HPV+ tumors. This effect was dependent on differential levels of TNFα and IL-10 between viral and non-viral cases (213). While no direct link between viral status and TNFα levels has been discovered yet, we speculate that this difference in cytokine milieu is attributed to the different forms of immune evasion mechanisms that the HPV virus takes during chronic infections and carcinogenesis of HNSCC compared to non-viral-induced mechanisms.

4.3 Addressing immune evasion and uncertainties in HPV-associated HNSCCs

There is clear evidence of viral-specific regulation within the TME of HNSCC. HPV-specific mechanisms of immune evasion in HNSCCs have been widely studied and previously discussed (214216). In general, HPV early proteins (particularly E5, E6, E7) are central in the downregulation of host immune responses against the virus. Some common nodes of immunomodulation include modulation of the NF-κB pathway (217220), inhibition of inflammatory cytokine production (221, 222), interferon and pattern recognition receptor signaling (217, 223, 224) and the disruption of antigen presentation processes (225230). These mechanisms aim to downregulate the recognition and activation of innate and adaptive immunity against the virus. It is important to note that these evasion mechanisms are not exclusive to viruses capable of inducing carcinogenesis. These processes are, however, seen as factors that exacerbate carcinogenesis by inducing chronic inflammation due to persistence of HPV infections, since immunosurveillance is widely disrupted and viral clearance is impaired (231). Further mechanistic studies are needed to elucidate the relative contributions of viral evasion mechanisms to the suppression of anti-tumor immunity in these HPV-driven cancers.

Nevertheless, it appears that targeting these viral mechanisms could, in theory, be complementary to immunotherapeutic options (such as immune checkpoint blockade) against HPV+ cancers, since eliminating the virus would target the main vulnerability of these malignancies. Antigen-primed DCs were explored to mount HPV-specific responses to complement CAR-T cell therapy (232). Even in the absence of known antigens, such DC vaccines could also be prepared by the fusion of tumor and dendritic cells ex vivo (233). Importantly, robust pre-clinical and clinical responses to TCR-T cell therapy targeting HPV-E7 in HNSCC and cervical cancers were observed (234, 235). Still, prophylactic vaccinations against HPV strains is the current best option to prevent cervical cancers (236), which are almost always caused by a persistent HPV infection. While efforts are underway to determine the efficacy of therapeutic HPV vaccines against HPV+ HNSCC (237241), it is imperative to better understand the consequences of chronic HPV infection on immunomodulation within the TME to determine if targeting the virus is a viable option as a therapeutic.

5 Epstein-Barr virus amongst the earliest known viruses for oncogenesis

Epstein-Barr virus (EBV), the first human tumorigenic virus discovered in 1964 in Burkitt’s Lymphoma cells, has since been implicated in various malignancies including epithelial cancers like gastric cancers (GCs) and nasopharyngeal carcinomas (NPCs) (242). Unlike the positive findings reported on HPV and HBV/HCV-associated cancers, the presence of EBV may not necessarily contribute to better OS or ORRs across EBV-driven cancers (23, 243). Our current understanding is that the involvement of EBV in the modulation of the tumor immune landscape is much more complex in contrast to other oncogenic viruses, which could explain the highly variable therapeutic responses to ICBs as reported in EBV-associated cancers.

Of which, EBVaGCs (EBV-associated gastric cancers) seem to yield better clinical outcomes despite being a small minority of EBV-driven cancers. EBVaGCs is a molecularly and clinically distinct subtype of gastric cancer accounting for about 10% of gastric cancers world-wide (244). It is largely driven by extensive viral epigenetic modifications, mainly DNA hypermethylation, unlike other gastric cancers, which are driven by mutational burden or genomic instability such as high microsatellite (MSI) or chromosomal instability (CI) (245). EBV promotes oncogenesis through BART miRNAs and BARF1 (BamHI-A rightward frame 1a), inducing methylation and altering gene expression. EBVaGC is not only characterized by high DNA hypermethylation but also frequent PIK3CA mutations and overexpression of JAK2, PD-L1 and PD-L2 (246). Paradoxically, despite its immune-rich TME, it exhibits reduced sensitivity to conventional chemotherapy (docetaxel and 5-fluorouracil) (247), highlighting the necessity for further investigation on its immunogenic profile and alternative therapeutic strategies such as immunotherapeutics for better patient outcomes.

5.1 EBV drives distinct immune profiles in gastric and nasopharyngeal carcinomas

Like other viral-driven cancers, the immunogenicity of EBVaGCs is also neither driven by mutational burden nor genomic instability. EBVaGCs were found to be mutually exclusive from MSI-H GCs with high amplification of PD-L1 expression (248251). Conflictingly, PD-L1 expression can be either associated with both poor or better patient survival (250, 252, 253). While MSI-H tumors typically respond well to conventional ICBs, a case study reported a late stage metastatic EBV-GC patient that also showed beneficial response to PD-L1 blockade (avelumab). Of note, the patient’s tumor did not show high mutational burden or any mismatch repair defect. The authors then interrogated TCGA cohort showing that EBV-GC are microsatellite stable with low mutational burden but are well infiltrated by immune cells (254). Another study reported a patient with EBVaGC that displayed durable complete response to ICB, overcoming resistance to trastuzumab plus chemotherapy (255). While MSI-H tumors are associated with B2M (beta-2 microglobulin) mutation, which is a form of acquired resistance to immunotherapy, durable responses to ICBs can still be observed in MSI-H tumors within EBV-negative GCs (256). Furthermore, though not significant, a considerably high numbers of MSI-high GCs were either negative for HLA-A/B/C (22/37 cases) or B2M (21/37 cases) (257). Conversely, EBVaGCs highly express both MHCI and MHCII molecules, which is likely a potential consequence of being highly infiltrated by activated immune cells into the TME (257259). In fact, the expression of HLA-DR was shown to be prognostic for better five-year overall survival (260). Importantly, EBVaGC represents a distinct clinicopathological entity with low incidence of lymph node invasion (249, 261). Following up by the same authors, EBVaGC tumors were reported to be better infiltrated with CD8 T cells and mature DCs (262). Both higher infiltrates of CD8+ and FOXP3+ cells were also found to be prognostic for better five-year overall survival (253). Likewise, the TME of EBVaGCs can contain high density of DCs, and interestingly, the maturation of these DCs can also be suppressed by exosomes derived from EBVaGC tumor cells (263).

Unlike gastric cancers, a great majority of NPCs are EBV+, which are also much more extensively studied given the considerably higher occurrences particularly across Asian countries (139, 264). Notably, United States was the second most common study sites for immunotherapy trials of NPCs despite much lower incidence rates across the world (243). Various EBV encoded nuclear antigens (EBNAs) and latent membrane proteins (LMPs) can be expressed in NPCs (265). Of which, EBNA1 can be overexpressed in NPCs, associated with metastasis (266) and immunosuppression within the TME (267). Mechanistically, EBNA1 contributes to TGFβ-mediated Treg formation and the production of Treg chemoattractants, CCL20 and CXCL12 (267, 268). Even though EBNA1 can be considered an EBV antigen, it is poorly immunogenic in cancer (269). Several other EBV-derived molecules were also found to play an integral role in tumor immune escape in NPCs. Like EBVaGCs, reports revealed mutations and downregulation of MHCI and MHCII molecules in EBVaNPCs (270, 271). Another EBV-encoded protein, BNLF2a, also inhibits TAP (transporter associated with antigen processing) to reduce antigen presentation and evade EBV-specific CD8 T cells (272). At the epigenetic level, LMP2A mediates hypermethylation of the HLA-ABC promoter. It was further demonstrated that the use of 5’-azacytidine as a demethylation agent was able to restore the expression of HLA-ABC in epithelial-origin tumor cell lines in vitro (273). In addition, EBV-encoded microRNAs (miRNAs) have profound immune suppressive effects against viral-specific T cells through means of downregulating of TAP1, TAP2 and HLA-ABC (274). Likewise, these EBV miRNAs also suppress the differentiation of naïve CD4 T cells into Th1 cells and the subsequent release of pro-inflammatory cytokines (275). Other non-coding RNAs such as circular RNAs (circRNAs) could also be involved in the immune modulation of the TME. EBV-encoded circBART2.2 was demonstrated to upregulate PD-L1 in NPC by promoting RIG-I signaling and the activation of IRF3 and NF-kB, causing T cell suppression (276).

5.2 Roles of tertiary lymphoid structures and B cells within the TME of NPCs

NPCs can be a promising target for ICBs considering the likely formation of TLS within the TME. Distinct TLS formation has been profiled in EBVaNPC, identifying a unique population of CXCL13-producing CD4+ T cells which can contribute to the recruitment of B cells and the maturation of TLS (277). Importantly, only B and plasma cells correlated with tumor mutational load in NPCs (278). Furthermore, in a recent study by Helmink et al., the enriched presence of B cells within TLS correlated with better outcomes in patients treated with ICB (279). While TLS can be common within NPC tumors, EBV-encoded LMP1 can suppress the maturation of antibody secreting cells and germinal center B cells. At the same time, LMP1-expressing B cells can act as regulatory B cells with high expression of IDO-1 (indoleamine 2,3-dioxygenase 1) (280). NPC-derived LMP1 was found to be non-immunogenic, in contrast to B cell-derived LMP1 which is capable of eliciting immune rejection in vivo (281, 282). Nevertheless, there is a rationale for combining ICB with strategies that promote TLS development. The presence of memory B cells and plasma cells may contribute to both T cell activation and antigen presentation, amplifying the local anti-tumor immunity through carefully coordinated B-T cell interactions.

5.3 Promising prospects in targeting EBV-driven cancers with immunotherapy

Satisfactory responses to ICBs have been reported in metastatic NPCs (243, 283, 284). From an experimental perspective, perhaps the way forward is to study immune responses to novel treatments such as EBV-targeted cell therapies and cancer vaccines. A case report highlighted the potential synergy of ICBs and the adoptive transfer of EBV-specific T cells resulting in the patient showing complete resolution of metastatic disease without any signs of relapse. More interestingly, the combinatory treatment resulted in the emergence of novel T cell clonotypes alongside the maintenance of dominant clones, indicating potential epitope spreading and TCR diversification (285). It was also demonstrated that the use of CRISPR/Cas9 to delete PD-1 could enhance in vitro and in vivo killing of GCs by cytotoxic T lymphocytes (CTLs) specific for the viral antigen LMP2A (latent membrane protein 2A) (251). Prior immunization with BARF1, an EBV antigen presented on tumor cells, may represent a potential tumor vaccine target, eliciting both humoral and T cell-mediated immune responses in vivo (286). Additionally, the targeting of alternative immune checkpoints such as TIM-3 and LAG-3 on tumor-specific CTL clones can further enhance eradication of tumor cells as demonstrated in GCs (287).

Alternatively, one could also consider exploiting the fact that EBV-infected targets are highly susceptible to NK cell-mediated killing (288, 289). NK cells incubated in EBV seropositive serum were demonstrated to be highly activated in vitro, suggesting an interplay of the humoral immunity or other upregulated cytokine factors within the viral-infected host (290). Adoptive transfer of NK cells in combination with anti-PD1 therapy also showed promising efficacy in a GC xenograft model (291). Furthermore, mesothelin-targeting CAR-NK92 cells were demonstrated to specifically eradicate GCs both in vitro and in vivo (292). Still, it is well known that adoptive NK cell therapies in general do not penetrate well into solid tumors (293, 294). It may perhaps be more effective to reinvigorate intratumoral NK cells within the TME, but our current understanding of tumor-infiltrating NK cells in GCs and NPCs are limited. The upregulation of EBV-encoded BZLF1 (BamHI Z fragment leftward open reading frame 1) during the early lytic cycle sensitizes viral-infected cells to NK cell-mediating killing by the upregulation of NKG2D ligands. However, such BZLF1-dependent sensitization could be counteracted by BHRF1, a viral homologue for BCL-2 acting as a potent anti-apoptotic protein also known to drive chemoresistance in EBV-associated cancers (295, 296). Apart from BHRF1-conferred resistance, other immune evasion pathways were characterized particularly in EBVaNPC. EBV-encoded microRNA BART7 (miR-BART7) indirectly represses the expression of NKG2D ligand, MIC-A (major histocompatibility complex class I chain-related peptide A) to desensitize NPC tumors from NK cell-mediated killing (297). The EBV gene, BCRF1, also encodes an IL-10 homologue that was demonstrated to impair NK cell and CD4 T cell activity (272). Another study reported lower infiltrates of granzyme B-positive NK cells in EBVaNPCs and further demonstrated that LMP2A upregulates F3 (Coagulation factor III), which in turn triggers platelet aggregation that suppresses NK cell cytotoxicity (298). Despite EBVaGCs being less extensively studied than NPCs, an immune deconvolution on the TCGA dataset for bulk GC tumor transcriptomics data putatively uncovered EBVaGCs to have higher infiltration of NK cells and T cells, but unexpectedly not B cells, as compared to EBV-negative GCs. Compared to both adjacent normal and EBV-negative tissues, EBVaGCs expressed higher levels of CD155 (encoding PVR) which is a ligand for either inhibitory receptors (TIGIT and CD96) or activating receptor, DNAM-1 (299, 300). Thus, future studies can focus on characterizing the immune profile of intratumoral NK cells to evaluate the potential of existing anti-TIGIT blockade as an alternative ICB (301, 302), acting to potently reinvigorate NK cell activity by enhancing PVR-DNAM1 binding.

Looking forward, one can also leverage on the wealth of publicly accessible NPC datasets to study novel cellular interactions that may play a critical role within the tumor immune landscape. Large datasets can be interrogated using immune deconvolution approaches such as CIBERSORT to uncover representative TME features that are prognostic to patient survival (303, 304). Single cell transcriptomics also revealed a unique population of Clec9a+ DCs, though its functions and relevance in NPCs remained unelucidated (278). In addition, the use of the CellPhoneDb algorithm further revealed putative cell-cell interactions that is unique between LMP+ NPC tumor cells and immune cells, driven by the chemokine CX3CL1 (305, 306). Contrastingly, there are lesser data resources available to understand EBVaGC. One can explore the use of a transplantable strain of EBVaGC known as “KT” in a humanized mice xenograft (307). Taken together, there are indeed several EBV-derived molecules that are of druggable potential but yet, a research gap remains to address if these targets can be feasibly combined with ICBs to yield better therapeutic responses in the clinics. We also render that EBVaNPCs may seem to appear more immune-tolerant for the fact that they are much more well-studied than EBVaGCs, which are of much lower incidence rates. Although not yet elucidated, it is highly plausible that EBV-driven mechanisms of immune evasion could occur in EBVaGCs as similarly observed in NPCs.

6 Concluding remarks

In general, it appears that cancers of viral etiologies tend not to rely on tumor mutational burden or neo/tumor antigens to prime immune responses. Evidently in these viral-driven cancers, ICB treatments are widely evaluated in the clinics while viral-associated immunity can be exploited in various forms of tumor vaccines or adoptive cell therapies (Summarized in Table 1). Taken together, we should agree here that there is substantial knowledge to exploit viral-associated immunogenicity and the highly dynamic TME for translation into direct applications within the oncology space. Moreover, the utilization of artificial intelligence (AI) has enabled precise prediction of ICB response (309), tumor progression or recurrence (132, 310), as well as immunogenic neoantigens (311, 312) for novel immunotherapies such as neoantigen vaccines. With a multi-omics, AI-powered analysis of the TME heterogeneity (313), it is hoped that immune signatures of different viral-associated cancers can be further delineated, ultimately improving therapeutic efficacy and patient outcomes with precision immuno-oncology. To conclude, we envision that clinicians can recognize and leverage these factors as powerful biomarkers for patients’ responses to ICBs, and at the same time, inspire future science to revisit rare cancers such as MCPyV+ MCCs and EBVaGCs for more novel and critical discoveries to improve conventional ICBs.

Table 1
www.frontiersin.org

Table 1. Non-exhaustive examples highlighted for various immunotherapies targeting viral-associated cancers.

Author contributions

KL: Writing – original draft, Writing – review & editing. SK: Writing – original draft, Writing – review & editing. JZ: Writing – original draft, Writing – review & editing. SN: Conceptualization, Funding acquisition, Supervision, Writing – review & editing.

Funding

The author(s) declare financial support was received for the research and/or publication of this article. This work is supported by the national medical research council (OFYIRG24jul-0034 and OFIRG24jan-0111), A*STAR CDF (C243512007) and core funds of Singapore Immunology Network (SIgN), A*STAR.

Acknowledgments

The authors thank all funding agencies for supporting their research.

Conflict of interest

All authors declare that the present work was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Lee JS and Ruppin E. Multiomics prediction of response rates to therapies to inhibit programmed cell death 1 and programmed cell death 1 ligand 1. JAMA Oncol. (2019) 5:1614–8. doi: 10.1001/jamaoncol.2019.2311

PubMed Abstract | Crossref Full Text | Google Scholar

2. Schumacher TN and Schreiber RD. Neoantigens in cancer immunotherapy. Science. (2015) 348:69–74. doi: 10.1126/science.aaa4971

PubMed Abstract | Crossref Full Text | Google Scholar

3. Yarchoan M, Johnson BA 3rd, Lutz ER, Laheru DA, and Jaffee EM. Targeting neoantigens to augment antitumour immunity. Nat Rev Cancer. (2017) 17:209–22. doi: 10.1038/nrc.2016.154

PubMed Abstract | Crossref Full Text | Google Scholar

4. Xie N, Shen G, Gao W, Huang Z, Huang C, and Fu L. Neoantigens: promising targets for cancer therapy. Signal Transduct Target Ther. (2023) 8:9. doi: 10.1038/s41392-022-01270-x

PubMed Abstract | Crossref Full Text | Google Scholar

5. Riaz N, Morris L, Havel JJ, Makarov V, Desrichard A, and Chan TA. The role of neoantigens in response to immune checkpoint blockade. Int Immunol. (2016) 28:411–9. doi: 10.1093/intimm/dxw019

PubMed Abstract | Crossref Full Text | Google Scholar

6. Alban TJ, Riaz N, Parthasarathy P, Makarov V, Kendall S, Yoo SK, et al. Neoantigen immunogenicity landscapes and evolution of tumor ecosystems during immunotherapy with nivolumab. Nat Med. (2024) 30:3209–22. doi: 10.1038/s41591-024-03240-y

PubMed Abstract | Crossref Full Text | Google Scholar

7. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. (2015) 348:124–8. doi: 10.1126/science.aaa1348

PubMed Abstract | Crossref Full Text | Google Scholar

8. Ma W, Pham B, and Li T. Cancer neoantigens as potential targets for immunotherapy. Clin Exp Metastasis. (2022) 39:51–60. doi: 10.1007/s10585-021-10091-1

PubMed Abstract | Crossref Full Text | Google Scholar

9. Yarchoan M, Hopkins A, and Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med. (2017) 377:2500–1. doi: 10.1056/NEJMc1713444

PubMed Abstract | Crossref Full Text | Google Scholar

10. Osipov A, Lim SJ, Popovic A, Azad NS, Laheru DA, Zheng L, et al. Tumor mutational burden, toxicity, and response of immune checkpoint inhibitors targeting PD(L)1, CTLA-4, and combination: A meta-regression analysis. Clin Cancer Res. (2020) 26:4842–51. doi: 10.1158/1078-0432.CCR-20-0458

PubMed Abstract | Crossref Full Text | Google Scholar

11. Ahmed J, Das B, Shin S, and Chen A. Challenges and future directions in the management of tumor mutational burden-high (TMB-H) advanced solid Malignancies. Cancers (Basel). (2023) 15. doi: 10.3390/cancers15245841

PubMed Abstract | Crossref Full Text | Google Scholar

12. Lybaert L, Thielemans K, Feldman SA, van der Burg SH, Bogaert C, and Ott PA. Neoantigen-directed therapeutics in the clinic: where are we? Trends Cancer. (2023) 9:503–19. doi: 10.1016/j.trecan.2023.02.004

PubMed Abstract | Crossref Full Text | Google Scholar

13. Kidman J, Principe N, Watson M, Lassmann T, Holt RA, Nowak AK, et al. Characteristics of TCR repertoire associated with successful immune checkpoint therapy responses. Front Immunol. (2020) 11:587014. doi: 10.3389/fimmu.2020.587014

PubMed Abstract | Crossref Full Text | Google Scholar

14. Aleksic M, Liddy N, Molloy PE, Pumphrey N, Vuidepot A, Chang KM, et al. Different affinity windows for virus and cancer-specific T-cell receptors: implications for therapeutic strategies. Eur J Immunol. (2012) 42:3174–9. doi: 10.1002/eji.201242606

PubMed Abstract | Crossref Full Text | Google Scholar

15. Wang L, Geng H, Liu Y, Liu L, Chen Y, Wu F, et al. Hot and cold tumors: Immunological features and the therapeutic strategies. MedComm (2020). (2023) 4:e343. doi: 10.1002/mco2.v4.5

Crossref Full Text | Google Scholar

16. Maleki Vareki S. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J Immunother Cancer. (2018) 6:157. doi: 10.1186/s40425-018-0479-7

PubMed Abstract | Crossref Full Text | Google Scholar

17. Chen Y, Wu Y, Yan G, and Zhang G. Tertiary lymphoid structures in cancer: maturation and induction. Front Immunol. (2024) 15:1369626. doi: 10.3389/fimmu.2024.1369626

PubMed Abstract | Crossref Full Text | Google Scholar

18. Munoz-Erazo L, Rhodes JL, Marion VC, and Kemp RA. Tertiary lymphoid structures in cancer - considerations for patient prognosis. Cell Mol Immunol. (2020) 17:570–5. doi: 10.1038/s41423-020-0457-0

PubMed Abstract | Crossref Full Text | Google Scholar

19. Sautes-Fridman C, Petitprez F, Calderaro J, and Fridman WH. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer. (2019) 19:307–25. doi: 10.1038/s41568-019-0144-6

PubMed Abstract | Crossref Full Text | Google Scholar

20. Yang L, Pang Y, and Moses HL. TGF-beta and immune cells: an important regulatory axis in the tumor microenvironment and progression. Trends Immunol. (2010) 31:220–7. doi: 10.1016/j.it.2010.04.002

PubMed Abstract | Crossref Full Text | Google Scholar

21. Ouyang P, Wang L, Wu J, Tian Y, Chen C, Li D, et al. Overcoming cold tumors: a combination strategy of immune checkpoint inhibitors. Front Immunol. (2024) 15:1344272. doi: 10.3389/fimmu.2024.1344272

PubMed Abstract | Crossref Full Text | Google Scholar

22. Qian ZY, Pan YQ, Li XX, Chen YX, Wu HX, Liu ZX, et al. Modulator of TMB-associated immune infiltration (MOTIF) predicts immunotherapy response and guides combination therapy. Sci Bull (Beijing). (2024) 69:803–22. doi: 10.1016/j.scib.2024.01.025

PubMed Abstract | Crossref Full Text | Google Scholar

23. Wu C, Ke Y, Wan L, and Xie X. Efficacy of immune checkpoint inhibitors differs in various status of carcinoma: a study based on 29 cohorts with 3255 participants. Cancer Immunol Immunother. (2024) 73:79. doi: 10.1007/s00262-024-03663-z

PubMed Abstract | Crossref Full Text | Google Scholar

24. Siqueira SOM, Campos-do-Carmo G, Dos Santos ALS, Martins C, and de Melo AC. Merkel cell carcinoma: epidemiology, clinical features, diagnosis and treatment of a rare disease. Bras Dermatol. (2023) 98:277–86. doi: 10.1016/j.abd.2022.09.003

PubMed Abstract | Crossref Full Text | Google Scholar

25. Loke ASW, Lambert PF, and Spurgeon ME. Current in vitro and in vivo models to study MCPyV-associated MCC. Viruses. (2022) 14. doi: 10.3390/v14102204

PubMed Abstract | Crossref Full Text | Google Scholar

26. Harms PW, Vats P, Verhaegen ME, Robinson DR, Wu YM, Dhanasekaran SM, et al. The distinctive mutational spectra of polyomavirus-negative merkel cell carcinoma. Cancer Res. (2015) 75:3720–7. doi: 10.1158/0008-5472.CAN-15-0702

PubMed Abstract | Crossref Full Text | Google Scholar

27. Goh G, Walradt T, Markarov V, Blom A, Riaz N, Doumani R, et al. Mutational landscape of MCPyV-positive and MCPyV-negative Merkel cell carcinomas with implications for immunotherapy. Oncotarget. (2016) 7:3403–15. doi: 10.18632/oncotarget.6494

PubMed Abstract | Crossref Full Text | Google Scholar

28. Church C, Pulliam T, Longino N, Park SY, Smythe KS, Makarov V, et al. Transcriptional and functional analyses of neoantigen-specific CD4 T cells during a profound response to anti-PD-L1 in metastatic Merkel cell carcinoma. J Immunother Cancer. (2022) 10. doi: 10.1136/jitc-2022-005328

PubMed Abstract | Crossref Full Text | Google Scholar

29. Miller NJ, Church CD, Dong L, Crispin D, Fitzgibbon MP, Lachance K, et al. Tumor-infiltrating merkel cell polyomavirus-specific T cells are diverse and associated with improved patient survival. Cancer Immunol Res. (2017) 5:137–47. doi: 10.1158/2326-6066.CIR-16-0210

PubMed Abstract | Crossref Full Text | Google Scholar

30. Paulson KG, Carter JJ, Johnson LG, Cahill KW, Iyer JG, Schrama D, et al. Antibodies to merkel cell polyomavirus T antigen oncoproteins reflect tumor burden in merkel cell carcinoma patients. Cancer Res. (2010) 70:8388–97. doi: 10.1158/0008-5472.CAN-10-2128

PubMed Abstract | Crossref Full Text | Google Scholar

31. Afanasiev OK, Yelistratova L, Miller N, Nagase K, Paulson K, Iyer JG, et al. Merkel polyomavirus-specific T cells fluctuate with merkel cell carcinoma burden and express therapeutically targetable PD-1 and Tim-3 exhaustion markers. Clin Cancer Res. (2013) 19:5351–60. doi: 10.1158/1078-0432.CCR-13-0035

PubMed Abstract | Crossref Full Text | Google Scholar

32. Wong MK and Yee C. Polyomavirus-positive Merkel cell carcinoma: the beginning of the beginning. J Clin Invest. (2024) 134. doi: 10.1172/JCI179749

PubMed Abstract | Crossref Full Text | Google Scholar

33. Hansen UK, Church CD, Carnaz Simoes AM, Frej MS, Bentzen AK, Tvingsholm SA, et al. T antigen-specific CD8+ T cells associate with PD-1 blockade response in virus-positive Merkel cell carcinoma. J Clin Invest. (2024) 134. doi: 10.1172/JCI177082

PubMed Abstract | Crossref Full Text | Google Scholar

34. Pulliam T, Jani S, Jing L, Ryu H, Jojic A, Shasha C, et al. Circulating cancer-specific CD8 T cell frequency is associated with response to PD-1 blockade in Merkel cell carcinoma. Cell Rep Med. (2024) 5:101412. doi: 10.1016/j.xcrm.2024.101412

PubMed Abstract | Crossref Full Text | Google Scholar

35. Chapuis AG, Afanasiev OK, Iyer JG, Paulson KG, Parvathaneni U, Hwang JH, et al. Regression of metastatic Merkel cell carcinoma following transfer of polyomavirus-specific T cells and therapies capable of re-inducing HLA class-I. Cancer Immunol Res. (2014) 2:27–36. doi: 10.1158/2326-6066.CIR-13-0087

PubMed Abstract | Crossref Full Text | Google Scholar

36. Paulson KG, Tegeder A, Willmes C, Iyer JG, Afanasiev OK, Schrama D, et al. Downregulation of MHC-I expression is prevalent but reversible in Merkel cell carcinoma. Cancer Immunol Res. (2014) 2:1071–9. doi: 10.1158/2326-6066.CIR-14-0005

PubMed Abstract | Crossref Full Text | Google Scholar

37. Ritter C, Fan K, Paschen A, Reker Hardrup S, Ferrone S, Nghiem P, et al. Epigenetic priming restores the HLA class-I antigen processing machinery expression in Merkel cell carcinoma. Sci Rep. (2017) 7:2290. doi: 10.1038/s41598-017-02608-0

PubMed Abstract | Crossref Full Text | Google Scholar

38. Liu W, Kim GB, Krump NA, Zhou Y, Riley JL, and You J. Selective reactivation of STING signaling to target Merkel cell carcinoma. Proc Natl Acad Sci U S A. (2020) 117:13730–9. doi: 10.1073/pnas.1919690117

PubMed Abstract | Crossref Full Text | Google Scholar

39. Abe T and Barber GN. Cytosolic-DNA-mediated, STING-dependent proinflammatory gene induction necessitates canonical NF-kappaB activation through TBK1. J Virol. (2014) 88:5328–41. doi: 10.1128/JVI.00037-14

PubMed Abstract | Crossref Full Text | Google Scholar

40. Ohnezeit D, Huang J, Westerkamp U, Brinschwitz V, Schmidt C, Gunther T, et al. Merkel cell polyomavirus small tumor antigen contributes to immune evasion by interfering with type I interferon signaling. PloS Pathog. (2024) 20:e1012426. doi: 10.1371/journal.ppat.1012426

PubMed Abstract | Crossref Full Text | Google Scholar

41. Dowlatshahi M, Huang V, Gehad AE, Jiang Y, Calarese A, Teague JE, et al. Tumor-specific T cells in human Merkel cell carcinomas: a possible role for Tregs and T-cell exhaustion in reducing T-cell responses. J Invest Dermatol. (2013) 133:1879–89. doi: 10.1038/jid.2013.75

PubMed Abstract | Crossref Full Text | Google Scholar

42. Sihto H, Bohling T, Kavola H, Koljonen V, Salmi M, Jalkanen S, et al. Tumor infiltrating immune cells and outcome of Merkel cell carcinoma: a population-based study. Clin Cancer Res. (2012) 18:2872–81. doi: 10.1158/1078-0432.CCR-11-3020

PubMed Abstract | Crossref Full Text | Google Scholar

43. Gherardin NA, Waldeck K, Caneborg A, Martelotto LG, Balachander S, Zethoven M, et al. gammadelta T cells in merkel cell carcinomas have a proinflammatory profile prognostic of patient survival. Cancer Immunol Res. (2021) 9:612–23. doi: 10.1158/2326-6066.CIR-20-0817

PubMed Abstract | Crossref Full Text | Google Scholar

44. Nghiem PT, Bhatia S, Lipson EJ, KudChadkar RR, Miller NJ, Annamalai L, et al. PD-1 blockade with pembrolizumab in advanced merkel-cell carcinoma. N Engl J Med. (2016) 374:2542–52. doi: 10.1056/NEJMoa1603702

PubMed Abstract | Crossref Full Text | Google Scholar

45. de Moraes FCA, Kreuz M, de Lara ICA, Lobo AOM, and Burbano RMR. Efficacy and safety of PD-1/PD-L1 inhibitors in patients with Merkel Cell Carcinoma: a systematic review and Meta-analysis. BMC Cancer. (2024) 24:1357. doi: 10.1186/s12885-024-13129-1

PubMed Abstract | Crossref Full Text | Google Scholar

46. Spassova I, Ugurel S, Kubat L, Zimmer L, Terheyden P, Mohr A, et al. Clinical and molecular characteristics associated with response to therapeutic PD-1/PD-L1 inhibition in advanced Merkel cell carcinoma. J Immunother Cancer. (2022) 10. doi: 10.1136/jitc-2021-003198

PubMed Abstract | Crossref Full Text | Google Scholar

47. Martins C, Rasbach E, Heppt MV, Singh P, Kulcsar Z, Holzgruber J, et al. Tumor cell-intrinsic PD-1 promotes Merkel cell carcinoma growth by activating downstream mTOR-mitochondrial ROS signaling. Sci Adv. (2024) 10:eadi2012. doi: 10.1126/sciadv.adi2012

PubMed Abstract | Crossref Full Text | Google Scholar

48. Lipson EJ, Vincent JG, Loyo M, Kagohara LT, Luber BS, Wang H, et al. PD-L1 expression in the Merkel cell carcinoma microenvironment: association with inflammation, Merkel cell polyomavirus and overall survival. Cancer Immunol Res. (2013) 1:54–63. doi: 10.1158/2326-6066.CIR-13-0034

PubMed Abstract | Crossref Full Text | Google Scholar

49. Gomez BP, Wang C, Viscidi RP, Peng S, He L, Wu TC, et al. Strategy for eliciting antigen-specific CD8+ T cell-mediated immune response against a cryptic CTL epitope of merkel cell polyomavirus large T antigen. Cell Biosci. (2012) 2:36. doi: 10.1186/2045-3701-2-36

PubMed Abstract | Crossref Full Text | Google Scholar

50. Zeng Q, Gomez BP, Viscidi RP, Peng S, He L, Ma B, et al. Development of a DNA vaccine targeting Merkel cell polyomavirus. Vaccine. (2012) 30:1322–9. doi: 10.1016/j.vaccine.2011.12.072

PubMed Abstract | Crossref Full Text | Google Scholar

51. Gerer KF, Erdmann M, Hadrup SR, Lyngaa R, Martin LM, Voll RE, et al. Preclinical evaluation of NF-kappaB-triggered dendritic cells expressing the viral oncogenic driver of Merkel cell carcinoma for therapeutic vaccination. Ther Adv Med Oncol. (2017) 9:451–64. doi: 10.1177/1758834017712630

PubMed Abstract | Crossref Full Text | Google Scholar

52. Buchta Rosean C, Leyder EC, Hamilton J, Carter JJ, Galloway DA, Koelle DM, et al. LAMP1 targeting of the large T antigen of Merkel cell polyomavirus results in potent CD4 T cell responses and tumor inhibition. Front Immunol. (2023) 14:1253568. doi: 10.3389/fimmu.2023.1253568

PubMed Abstract | Crossref Full Text | Google Scholar

53. Gomez B, He L, Tsai YC, Wu TC, Viscidi RP, and Hung CF. Creation of a Merkel cell polyomavirus small T antigen-expressing murine tumor model and a DNA vaccine targeting small T antigen. Cell Biosci. (2013) 3:29. doi: 10.1186/2045-3701-3-29

PubMed Abstract | Crossref Full Text | Google Scholar

54. Verhaegen ME, Harms PW, Van Goor JJ, Arche J, Patrick MT, Wilbert D, et al. Direct cellular reprogramming enables development of viral T antigen-driven Merkel cell carcinoma in mice. J Clin Invest. (2022) 132. doi: 10.1172/JCI152069

PubMed Abstract | Crossref Full Text | Google Scholar

55. Das BK, Kannan A, Velasco GJ, Kunika MD, Lambrecht N, Nguyen Q, et al. Single-cell dissection of Merkel cell carcinoma heterogeneity unveils transcriptomic plasticity and therapeutic vulnerabilities. Cell Rep Med. (2023) 4:101101. doi: 10.1016/j.xcrm.2023.101101

PubMed Abstract | Crossref Full Text | Google Scholar

56. Sundqvist B, Kilpinen S, Bohling T, Koljonen V, and Sihto H. Activation of oncogenic and immune-response pathways is linked to disease-specific survival in merkel cell carcinoma. Cancers (Basel). (2022) 14. doi: 10.3390/cancers14153591

PubMed Abstract | Crossref Full Text | Google Scholar

57. Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. (2024) 74:229–63. doi: 10.3322/caac.21834

PubMed Abstract | Crossref Full Text | Google Scholar

58. Ringelhan M, McKeating JA, and Protzer U. Viral hepatitis and liver cancer. Philos Trans R Soc Lond B Biol Sci. (2017) 372. doi: 10.1038/s41575-020-00381-6

PubMed Abstract | Crossref Full Text | Google Scholar

59. Huang DQ, El-Serag HB, and Loomba R. Global epidemiology of NAFLD-related HCC: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol. (2021) 18:223–38. doi: 10.1038/s41575-020-00381-6

PubMed Abstract | Crossref Full Text | Google Scholar

60. Yeh SH, Li CL, Lin YY, Ho MC, Wang YC, Tseng ST, et al. Hepatitis B virus DNA integration drives carcinogenesis and provides a new biomarker for HBV-related HCC. Cell Mol Gastroenterol Hepatol. (2023) 15:921–9. doi: 10.1016/j.jcmgh.2023.01.001

PubMed Abstract | Crossref Full Text | Google Scholar

61. Zhao K, Liu A, and Xia Y. Insights into Hepatitis B Virus DNA Integration-55 Years after Virus Discovery. Innovation (Camb). (2020) 1:100034. doi: 10.1016/j.xinn.2020.100034

PubMed Abstract | Crossref Full Text | Google Scholar

62. Milosevic I, Todorovic N, Filipovic A, Simic J, Markovic M, Stevanovic O, et al. HCV and HCC tango-deciphering the intricate dance of disease: A review article. Int J Mol Sci. (2023) 24. doi: 10.3390/ijms242216048

PubMed Abstract | Crossref Full Text | Google Scholar

63. Lara-Pezzi E, Gomez-Gaviro MV, Galvez BG, Mira E, Iniguez MA, Fresno M, et al. The hepatitis B virus X protein promotes tumor cell invasion by inducing membrane-type matrix metalloproteinase-1 and cyclooxygenase-2 expression. J Clin Invest. (2002) 110:1831–8. doi: 10.1172/JCI200215887

PubMed Abstract | Crossref Full Text | Google Scholar

64. Gehring AJ, Ho ZZ, Tan AT, Aung MO, Lee KH, Tan KC, et al. Profile of tumor antigen-specific CD8 T cells in patients with hepatitis B virus-related hepatocellular carcinoma. Gastroenterology. (2009) 137:682–90. doi: 10.1053/j.gastro.2009.04.045

PubMed Abstract | Crossref Full Text | Google Scholar

65. Cheng Y, Gunasegaran B, Singh HD, Dutertre CA, Loh CY, Lim JQ, et al. Non-terminally exhausted tumor-resident memory HBV-specific T cell responses correlate with relapse-free survival in hepatocellular carcinoma. Immunity. (2021) 54:1825–40 e7. doi: 10.1016/j.immuni.2021.06.013

PubMed Abstract | Crossref Full Text | Google Scholar

66. You M, Gao Y, Fu J, Xie R, Zhu Z, Hong Z, et al. Epigenetic regulation of HBV-specific tumor-infiltrating T cells in HBV-related HCC. Hepatology. (2023) 78:943–58. doi: 10.1097/HEP.0000000000000369

PubMed Abstract | Crossref Full Text | Google Scholar

67. Lim CJ, Lee YH, Pan L, Lai L, Chua C, Wasser M, et al. Multidimensional analyses reveal distinct immune microenvironment in hepatitis B virus-related hepatocellular carcinoma. Gut. (2019) 68:916–27. doi: 10.1136/gutjnl-2018-316510

PubMed Abstract | Crossref Full Text | Google Scholar

68. Ge Z, Zhou G, Campos Carrascosa L, Gausvik E, Boor PPC, Noordam L, et al. TIGIT and PD1 Co-blockade Restores ex vivo Functions of Human Tumor-Infiltrating CD8(+) T Cells in Hepatocellular Carcinoma. Cell Mol Gastroenterol Hepatol. (2021) 12:443–64. doi: 10.1016/j.jcmgh.2021.03.003

PubMed Abstract | Crossref Full Text | Google Scholar

69. Bertoletti A, Brunetto M, Maini MK, Bonino F, Qasim W, and Stauss H. T cell receptor-therapy in HBV-related hepatocellularcarcinoma. Oncoimmunology. (2015) 4:e1008354. doi: 10.1080/2162402X.2015.1008354

PubMed Abstract | Crossref Full Text | Google Scholar

70. Meng F, Zhao J, Tan AT, Hu W, Wang SY, Jin J, et al. Immunotherapy of HBV-related advanced hepatocellular carcinoma with short-term HBV-specific TCR expressed T cells: results of dose escalation, phase I trial. Hepatol Int. (2021) 15:1402–12. doi: 10.1007/s12072-021-10250-2

PubMed Abstract | Crossref Full Text | Google Scholar

71. Qasim W, Brunetto M, Gehring AJ, Xue SA, Schurich A, Khakpoor A, et al. Immunotherapy of HCC metastases with autologous T cell receptor redirected T cells, targeting HBsAg in a liver transplant patient. J Hepatol. (2015) 62:486–91. doi: 10.1016/j.jhep.2014.10.001

PubMed Abstract | Crossref Full Text | Google Scholar

72. Wan X, Wisskirchen K, Jin T, Yang L, Wang X, Wu X, et al. Genetically-modified, redirected T cells target hepatitis B surface antigen-positive hepatocytes and hepatocellular carcinoma lesions in a clinical setting. Clin Mol Hepatol. (2024) 30:735–55. doi: 10.3350/cmh.2024.0058

PubMed Abstract | Crossref Full Text | Google Scholar

73. Wolski D, Foote PK, Chen DY, Lewis-Ximenez LL, Fauvelle C, Aneja J, et al. Early transcriptional divergence marks virus-specific primary human CD8(+) T cells in chronic versus acute infection. Immunity. (2017) 47:648–63 e8. doi: 10.1016/j.immuni.2017.09.006

PubMed Abstract | Crossref Full Text | Google Scholar

74. Harding JJ, Nandakumar S, Armenia J, Khalil DN, Albano M, Ly M, et al. Prospective genotyping of hepatocellular carcinoma: clinical implications of next-generation sequencing for matching patients to targeted and immune therapies. Clin Cancer Res. (2019) 25:2116–26. doi: 10.1158/1078-0432.CCR-18-2293

PubMed Abstract | Crossref Full Text | Google Scholar

75. Lee WC, Wang YC, Cheng CH, Wu TH, Lee CF, Wu TJ, et al. Myeloid-derived suppressor cells in the patients with liver resection for hepatitis B virus-related hepatocellular carcinoma. Sci Rep. (2019) 9:2269. doi: 10.1038/s41598-019-38785-3

PubMed Abstract | Crossref Full Text | Google Scholar

76. Song G, Shi Y, Zhang M, Goswami S, Afridi S, Meng L, et al. Global immune characterization of HBV/HCV-related hepatocellular carcinoma identifies macrophage and T-cell subsets associated with disease progression. Cell Discov. (2020) 6:90. doi: 10.1038/s41421-020-00214-5

PubMed Abstract | Crossref Full Text | Google Scholar

77. Liu Y, Kim ES, and Guo H. Hepatitis B virus-related hepatocellular carcinoma exhibits distinct intratumoral microbiota and immune microenvironment signatures. J Med Virol. (2024) 96:e29485. doi: 10.1002/jmv.29485

PubMed Abstract | Crossref Full Text | Google Scholar

78. Fu J, Xu D, Liu Z, Shi M, Zhao P, Fu B, et al. Increased regulatory T cells correlate with CD8 T-cell impairment and poor survival in hepatocellular carcinoma patients. Gastroenterology. (2007) 132:2328–39. doi: 10.1053/j.gastro.2007.03.102

PubMed Abstract | Crossref Full Text | Google Scholar

79. Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. (2018) 554:544–8. doi: 10.1038/nature25501

PubMed Abstract | Crossref Full Text | Google Scholar

80. Zheng H, Peng X, Yang S, Li X, Huang M, Wei S, et al. Targeting tumor-associated macrophages in hepatocellular carcinoma: biology, strategy, and immunotherapy. Cell Death Discov. (2023) 9:65. doi: 10.1038/s41420-023-01356-7

PubMed Abstract | Crossref Full Text | Google Scholar

81. Xue H, Lin F, Tan H, Zhu ZQ, Zhang ZY, and Zhao L. Overrepresentation of IL-10-expressing B cells suppresses cytotoxic CD4+ T cell activity in HBV-induced hepatocellular carcinoma. PloS One. (2016) 11:e0154815. doi: 10.1371/journal.pone.0154815

PubMed Abstract | Crossref Full Text | Google Scholar

82. Nakamoto N, Cho H, Shaked A, Olthoff K, Valiga ME, Kaminski M, et al. Synergistic reversal of intrahepatic HCV-specific CD8 T cell exhaustion by combined PD-1/CTLA-4 blockade. PloS Pathog. (2009) 5:e1000313. doi: 10.1371/journal.ppat.1000313

PubMed Abstract | Crossref Full Text | Google Scholar

83. De Battista D, Zamboni F, Gerstein H, Sato S, Markowitz TE, Lack J, et al. Molecular signature and immune landscape of HCV-associated hepatocellular carcinoma (HCC): differences and similarities with HBV-HCC. J Hepatocell Carcinoma. (2021) 8:1399–413. doi: 10.2147/JHC.S325959

PubMed Abstract | Crossref Full Text | Google Scholar

84. Engelskircher SA, Chen PC, Strunz B, Oltmanns C, Ristic T, Owusu Sekyere S, et al. Impending HCC diagnosis in patients with cirrhosis after HCV cure features a natural killer cell signature. Hepatology. (2024) 80:202–22. doi: 10.1097/HEP.0000000000000804

PubMed Abstract | Crossref Full Text | Google Scholar

85. Cancer Genome Atlas Research Network. Electronic address wbe, Cancer Genome Atlas Research N. Comprehensive and Integrative Genomic Characterization of Hepatocellular Carcinoma. Cell. (2017) 169:1327–41 e23. doi: 10.1016/j.cell.2017.05.046

PubMed Abstract | Crossref Full Text | Google Scholar

86. Montironi C, Castet F, Haber PK, Pinyol R, Torres-Martin M, Torrens L, et al. Inflamed and non-inflamed classes of HCC: a revised immunogenomic classification. Gut. (2023) 72:129–40. doi: 10.1136/gutjnl-2021-325918

PubMed Abstract | Crossref Full Text | Google Scholar

87. Kazankov K, Jorgensen SMD, Thomsen KL, Moller HJ, Vilstrup H, George J, et al. The role of macrophages in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Nat Rev Gastroenterol Hepatol. (2019) 16:145–59. doi: 10.1038/s41575-018-0082-x

PubMed Abstract | Crossref Full Text | Google Scholar

88. Pfister D, Nunez NG, Pinyol R, Govaere O, Pinter M, Szydlowska M, et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature. (2021) 592:450–6. doi: 10.1038/s41586-021-03362-0

PubMed Abstract | Crossref Full Text | Google Scholar

89. Dudek M, Pfister D, Donakonda S, Filpe P, Schneider A, Laschinger M, et al. Auto-aggressive CXCR6(+) CD8 T cells cause liver immune pathology in NASH. Nature. (2021) 592:444–9. doi: 10.1038/s41586-021-03233-8

PubMed Abstract | Crossref Full Text | Google Scholar

90. Ramirez CFA and Akkari L. Myeloid cell path to Malignancy: insights into liver cancer. Trends Cancer. (2025) 11(6):591–610. doi: 10.1016/j.trecan.2025.02.006

PubMed Abstract | Crossref Full Text | Google Scholar

91. Wan S, Zhao E, Kryczek I, Vatan L, Sadovskaya A, Ludema G, et al. Tumor-associated macrophages produce interleukin 6 and signal via STAT3 to promote expansion of human hepatocellular carcinoma stem cells. Gastroenterology. (2014) 147:1393–404. doi: 10.1053/j.gastro.2014.08.039

PubMed Abstract | Crossref Full Text | Google Scholar

92. Yu W, Zhang Y, Sun L, Huang W, Li X, Xia N, et al. Myeloid Trem2 ameliorates the progression of metabolic dysfunction-associated steatotic liver disease by regulating macrophage pyroptosis and inflammation resolution. Metabolism. (2024) 155:155911. doi: 10.1016/j.metabol.2024.155911

PubMed Abstract | Crossref Full Text | Google Scholar

93. Ma J, Zheng B, Goswami S, Meng L, Zhang D, Cao C, et al. PD1(Hi) CD8(+) T cells correlate with exhausted signature and poor clinical outcome in hepatocellular carcinoma. J Immunother Cancer. (2019) 7:331. doi: 10.1186/s40425-019-0814-7

PubMed Abstract | Crossref Full Text | Google Scholar

94. Wang G, Zhou Z, Jin W, Zhang X, Zhang H, and Wang X. Single-cell transcriptome sequencing reveals spatial distribution of IL34(+) cancer-associated fibroblasts in hepatocellular carcinoma tumor microenvironment. NPJ Precis Oncol. (2023) 7:133. doi: 10.1038/s41698-023-00483-9

PubMed Abstract | Crossref Full Text | Google Scholar

95. Noda Y, Kawaguchi T, Korenaga M, Yoshio S, Komukai S, Nakano M, et al. High serum interleukin-34 level is a predictor of poor prognosis in patients with non-viral hepatocellular carcinoma. Hepatol Res. (2019) 49:1046–53. doi: 10.1111/hepr.13350

PubMed Abstract | Crossref Full Text | Google Scholar

96. Lee MJ, Jin YH, Kim K, Choi Y, Kim HC, and Park S. Expression of hepatitis B virus x protein in hepatocytes suppresses CD8 T cell activity. Immune Netw. (2010) 10:126–34. doi: 10.4110/in.2010.10.4.126

PubMed Abstract | Crossref Full Text | Google Scholar

97. Chen Y, Cheng M, and Tian Z. Hepatitis B virus down-regulates expressions of MHC class I molecules on hepatoplastoma cell line. Cell Mol Immunol. (2006) 3:373–8.

PubMed Abstract | Google Scholar

98. Han Q, Hou Z, Yin C, Zhang C, and Zhang J. 5'-triphosphate siRNA targeting HBx elicits a potent anti-HBV immune response in pAAV-HBV transfected mice. Antiviral Res. (2019) 161:36–45. doi: 10.1016/j.antiviral.2018.11.006

PubMed Abstract | Crossref Full Text | Google Scholar

99. Hofmann M, Tauber C, Hensel N, and Thimme R. CD8(+) T cell responses during HCV infection and HCC. J Clin Med. (2021) 10. doi: 10.3390/jcm10050991

PubMed Abstract | Crossref Full Text | Google Scholar

100. Kanto T, Inoue M, Miyatake H, Sato A, Sakakibara M, Yakushijin T, et al. Reduced numbers and impaired ability of myeloid and plasmacytoid dendritic cells to polarize T helper cells in chronic hepatitis C virus infection. J Infect Dis. (2004) 190:1919–26. doi: 10.1086/jid.2004.190.issue-11

PubMed Abstract | Crossref Full Text | Google Scholar

101. Li W, Guo L, Rathi P, Marinova E, Gao X, Wu MF, et al. Redirecting T cells to glypican-3 with 4-1BB zeta chimeric antigen receptors results in th1 polarization and potent antitumor activity. Hum Gene Ther. (2017) 28:437–48. doi: 10.1089/hum.2016.025

PubMed Abstract | Crossref Full Text | Google Scholar

102. Gao H, Li K, Tu H, Pan X, Jiang H, Shi B, et al. Development of T cells redirected to glypican-3 for the treatment of hepatocellular carcinoma. Clin Cancer Res. (2014) 20:6418–28. doi: 10.1158/1078-0432.CCR-14-1170

PubMed Abstract | Crossref Full Text | Google Scholar

103. Li D, Qin J, Zhou T, Li Y, Cheng X, Chen Z, et al. Bispecific GPC3/PD−1 CAR−T cells for the treatment of HCC. Int J Oncol. (2023) 62. doi: 10.3892/ijo.2023.5501

PubMed Abstract | Crossref Full Text | Google Scholar

104. Lin X, Liu Z, Dong X, Wang K, Sun Y, Zhang H, et al. Radiotherapy enhances the anti-tumor effect of CAR-NK cells for hepatocellular carcinoma. J Transl Med. (2024) 22:929. doi: 10.1186/s12967-024-05724-4

PubMed Abstract | Crossref Full Text | Google Scholar

105. Yang L, Pham K, Xi Y, Wu Q, Liu D, Robertson KD, et al. Exploring Glypican-3 targeted CAR-NK treatment and potential therapy resistance in hepatocellular carcinoma. PloS One. (2025) 20:e0317401. doi: 10.1371/journal.pone.0317401

PubMed Abstract | Crossref Full Text | Google Scholar

106. Ma C, Kesarwala AH, Eggert T, Medina-Echeverz J, Kleiner DE, Jin P, et al. NAFLD causes selective CD4(+) T lymphocyte loss and promotes hepatocarcinogenesis. Nature. (2016) 531:253–7. doi: 10.1038/nature16969

PubMed Abstract | Crossref Full Text | Google Scholar

107. Kuang DM, Zhao Q, Peng C, Xu J, Zhang JP, Wu C, et al. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med. (2009) 206:1327–37. doi: 10.1084/jem.20082173

PubMed Abstract | Crossref Full Text | Google Scholar

108. Petty AJ, Dai R, Lapalombella R, Baiocchi RA, Benson DM, Li Z, et al. Hedgehog-induced PD-L1 on tumor-associated macrophages is critical for suppression of tumor-infiltrating CD8+ T cell function. JCI Insight. (2021) 6. doi: 10.1172/jci.insight.146707

PubMed Abstract | Crossref Full Text | Google Scholar

109. He G and Karin M. NF-kappaB and STAT3 - key players in liver inflammation and cancer. Cell Res. (2011) 21:159–68. doi: 10.1038/cr.2010.183

PubMed Abstract | Crossref Full Text | Google Scholar

110. Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med. (2020) 382:1894–905. doi: 10.1056/NEJMoa1915745

PubMed Abstract | Crossref Full Text | Google Scholar

111. Abou-Alfa GK, Lau G, Kudo M, Chan SL, Kelley RK, Furuse J, et al. Tremelimumab plus durvalumab in unresectable hepatocellular carcinoma. NEJM Evid. (2022) 1:EVIDoa2100070. doi: 10.1056/EVIDoa2100070

PubMed Abstract | Crossref Full Text | Google Scholar

112. Zhu AX, Abbas AR, de Galarreta MR, Guan Y, Lu S, Koeppen H, et al. Molecular correlates of clinical response and resistance to atezolizumab in combination with bevacizumab in advanced hepatocellular carcinoma. Nat Med. (2022) 28:1599–611. doi: 10.1038/s41591-022-01868-2

PubMed Abstract | Crossref Full Text | Google Scholar

113. Haber PK, Puigvehi M, Castet F, Lourdusamy V, Montal R, Tabrizian P, et al. Evidence-based management of hepatocellular carcinoma: systematic review and meta-analysis of randomized controlled trials (2002-2020). Gastroenterology. (2021) 161:879–98. doi: 10.1053/j.gastro.2021.06.008

PubMed Abstract | Crossref Full Text | Google Scholar

114. Pinter M, Pinato DJ, Ramadori P, and Heikenwalder M. NASH and hepatocellular carcinoma: immunology and immunotherapy. Clin Cancer Res. (2023) 29:513–20. doi: 10.1158/1078-0432.CCR-21-1258

PubMed Abstract | Crossref Full Text | Google Scholar

115. Kelley RK, Rimassa L, Cheng AL, Kaseb A, Qin S, Zhu AX, et al. Cabozantinib plus atezolizumab versus sorafenib for advanced hepatocellular carcinoma (COSMIC-312): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. (2022) 23:995–1008. doi: 10.1016/S1470-2045(22)00326-6

PubMed Abstract | Crossref Full Text | Google Scholar

116. Llovet JM, Kudo M, Merle P, Meyer T, Qin S, Ikeda M, et al. Lenvatinib plus pembrolizumab versus lenvatinib plus placebo for advanced hepatocellular carcinoma (LEAP-002): a randomised, double-blind, phase 3 trial. Lancet Oncol. (2023) 24:1399–410. doi: 10.1016/S1470-2045(23)00469-2

PubMed Abstract | Crossref Full Text | Google Scholar

117. Moehler M, Heo J, Lee HC, Tak WY, Chao Y, Paik SW, et al. Vaccinia-based oncolytic immunotherapy Pexastimogene Devacirepvec in patients with advanced hepatocellular carcinoma after sorafenib failure: a randomized multicenter Phase IIb trial (TRAVERSE). Oncoimmunology. (2019) 8:1615817. doi: 10.1080/2162402X.2019.1615817

PubMed Abstract | Crossref Full Text | Google Scholar

118. Abou-Alfa GK, Galle PR, Chao Y, Erinjeri J, Heo J, Borad MJ, et al. PHOCUS: A phase 3, randomized, open-label study of sequential treatment with pexa-vec (JX-594) and sorafenib in patients with advanced hepatocellular carcinoma. Liver Cancer. (2024) 13:248–64. doi: 10.1159/000533650

PubMed Abstract | Crossref Full Text | Google Scholar

119. Breitbach CJ, Moon A, Burke J, Hwang TH, and Kirn DH. A phase 2, open-label, randomized study of pexa-vec (JX-594) administered by intratumoral injection in patients with unresectable primary hepatocellular carcinoma. Methods Mol Biol. (2015) 1317:343–57. doi: 10.1007/978-1-4939-2727-2_19

PubMed Abstract | Crossref Full Text | Google Scholar

120. Molgora M, Esaulova E, Vermi W, Hou J, Chen Y, Luo J, et al. TREM2 modulation remodels the tumor myeloid landscape enhancing anti-PD-1 immunotherapy. Cell. (2020) 182:886–900 e17. doi: 10.1016/j.cell.2020.07.013

PubMed Abstract | Crossref Full Text | Google Scholar

121. Binnewies M, Pollack JL, Rudolph J, Dash S, Abushawish M, Lee T, et al. Targeting TREM2 on tumor-associated macrophages enhances immunotherapy. Cell Rep. (2021) 37:109844. doi: 10.1016/j.celrep.2021.109844

PubMed Abstract | Crossref Full Text | Google Scholar

122. Guo H, Wang M, Ni C, Yang C, Fu C, Zhang X, et al. TREM2 promotes the formation of a tumor-supportive microenvironment in hepatocellular carcinoma. J Exp Clin Cancer Res. (2025) 44:20. doi: 10.1186/s13046-025-03287-w

PubMed Abstract | Crossref Full Text | Google Scholar

123. Hou J, Zhang J, Cui P, Zhou Y, Liu C, Wu X, et al. TREM2 sustains macrophage-hepatocyte metabolic coordination in nonalcoholic fatty liver disease and sepsis. J Clin Invest. (2021) 131. doi: 10.1172/JCI135197

PubMed Abstract | Crossref Full Text | Google Scholar

124. Hendrikx T, Porsch F, Kiss MG, Rajcic D, Papac-Milicevic N, Hoebinger C, et al. Soluble TREM2 levels reflect the recruitment and expansion of TREM2(+) macrophages that localize to fibrotic areas and limit NASH. J Hepatol. (2022) 77:1373–85. doi: 10.1016/j.jhep.2022.06.004

PubMed Abstract | Crossref Full Text | Google Scholar

125. De Ponti FF, Bujko A, Liu Z, Collins PJ, Schuermans S, Maueroder C, et al. Spatially restricted and ontogenically distinct hepatic macrophages are required for tissue repair. Immunity. (2025) 58:362–80 e10. doi: 10.1016/j.immuni.2025.01.002

PubMed Abstract | Crossref Full Text | Google Scholar

126. Sun H, Song J, Weng C, Xu J, Huang M, Huang Q, et al. Association of decreased expression of the macrophage scavenger receptor MARCO with tumor progression and poor prognosis in human hepatocellular carcinoma. J Gastroenterol Hepatol. (2017) 32:1107–14. doi: 10.1111/jgh.2017.32.issue-5

PubMed Abstract | Crossref Full Text | Google Scholar

127. Eisinger S, Sarhan D, Boura VF, Ibarlucea-Benitez I, Tyystjarvi S, Oliynyk G, et al. Targeting a scavenger receptor on tumor-associated macrophages activates tumor cell killing by natural killer cells. Proc Natl Acad Sci U S A. (2020) 117:32005–16. doi: 10.1073/pnas.2015343117

PubMed Abstract | Crossref Full Text | Google Scholar

128. La Fleur L, Botling J, He F, Pelicano C, Zhou C, He C, et al. Targeting MARCO and IL37R on immunosuppressive macrophages in lung cancer blocks regulatory T cells and supports cytotoxic lymphocyte function. Cancer Res. (2021) 81:956–67. doi: 10.1158/0008-5472.CAN-20-1885

PubMed Abstract | Crossref Full Text | Google Scholar

129. Miyamoto Y, Kikuta J, Matsui T, Hasegawa T, Fujii K, Okuzaki D, et al. Periportal macrophages protect against commensal-driven liver inflammation. Nature. (2024) 629:901–9. doi: 10.1038/s41586-024-07372-6

PubMed Abstract | Crossref Full Text | Google Scholar

130. Yuki K, Cheng N, Nakano M, and Kuo CJ. Organoid models of tumor immunology. Trends Immunol. (2020) 41:652–64. doi: 10.1016/j.it.2020.06.010

PubMed Abstract | Crossref Full Text | Google Scholar

131. Recaldin T, Steinacher L, Gjeta B, Harter MF, Adam L, Kromer K, et al. Human organoids with an autologous tissue-resident immune compartment. Nature. (2024) 633:165–73. doi: 10.1038/s41586-024-07791-5

PubMed Abstract | Crossref Full Text | Google Scholar

132. Jia G, He P, Dai T, Goh D, Wang J, Sun M, et al. Spatial immune scoring system predicts hepatocellular carcinoma recurrence. Nature. (2025) 640:1031–41. doi: 10.1038/s41586-025-08668-x

PubMed Abstract | Crossref Full Text | Google Scholar

133. Rector A and Van Ranst M. Animal papillomaviruses. Virology. (2013) 445:213–23. doi: 10.1016/j.virol.2013.05.007

PubMed Abstract | Crossref Full Text | Google Scholar

134. Doorbar J, Egawa N, Griffin H, Kranjec C, and Murakami I. Human papillomavirus molecular biology and disease association. Rev Med Virol. (2015) 25 Suppl 1:2–23. doi: 10.1002/rmv.1822

PubMed Abstract | Crossref Full Text | Google Scholar

135. zur Hausen H. Papillomaviruses in the causation of human cancers - a brief historical account. Virology. (2009) 384:260–5. doi: 10.1016/j.virol.2008.11.046

PubMed Abstract | Crossref Full Text | Google Scholar

136. Tao X, Zhang H, Li J, Zhang H, Xiao J, Zhang L, et al. Prevalence of HPV-16/18 genotypes and immediate histopathologic correlation results in a Chinese population with negative cytology and positive high-risk HPV testing. Cancer Cytopathol. (2019) 127:650–7. doi: 10.1002/cncy.v127.10

PubMed Abstract | Crossref Full Text | Google Scholar

137. Schwarz E, Freese UK, Gissmann L, Mayer W, Roggenbuck B, Stremlau A, et al. Structure and transcription of human papillomavirus sequences in cervical carcinoma cells. Nature. (1985) 314:111–4. doi: 10.1038/314111a0

PubMed Abstract | Crossref Full Text | Google Scholar

138. Sofiani VH, Veisi P, Rukerd MRZ, Ghazi R, and Nakhaie M. The complexity of human papilloma virus in cancers: a narrative review. Infect Agent Cancer. (2023) 18:13. doi: 10.1186/s13027-023-00488-w

PubMed Abstract | Crossref Full Text | Google Scholar

139. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, and 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. doi: 10.3322/caac.21492

PubMed Abstract | Crossref Full Text | Google Scholar

140. Yim EK and Park JS. The role of HPV E6 and E7 oncoproteins in HPV-associated cervical carcinogenesis. Cancer Res Treat. (2005) 37:319–24. doi: 10.4143/crt.2005.37.6.319

PubMed Abstract | Crossref Full Text | Google Scholar

141. Munger K, Baldwin A, Edwards KM, Hayakawa H, Nguyen CL, Owens M, et al. Mechanisms of human papillomavirus-induced oncogenesis. J Virol. (2004) 78:11451–60. doi: 10.1128/JVI.78.21.11451-11460.2004

PubMed Abstract | Crossref Full Text | Google Scholar

142. Hoppe-Seyler K, Bossler F, Braun JA, Herrmann AL, and Hoppe-Seyler F. The HPV E6/E7 oncogenes: key factors for viral carcinogenesis and therapeutic targets. Trends Microbiol. (2018) 26:158–68. doi: 10.1016/j.tim.2017.07.007

PubMed Abstract | Crossref Full Text | Google Scholar

143. Chung HC, Ros W, Delord JP, Perets R, Italiano A, Shapira-Frommer R, et al. Efficacy and safety of pembrolizumab in previously treated advanced cervical cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol. (2019) 37:1470–8. doi: 10.1200/JCO.18.01265

PubMed Abstract | Crossref Full Text | Google Scholar

144. Frenel JS, Le Tourneau C, O'Neil B, Ott PA, Piha-Paul SA, Gomez-Roca C, et al. Safety and efficacy of pembrolizumab in advanced, programmed death ligand 1-positive cervical cancer: results from the phase ib KEYNOTE-028 trial. J Clin Oncol. (2017) 35:4035–41. doi: 10.1200/JCO.2017.74.5471

PubMed Abstract | Crossref Full Text | Google Scholar

145. Santin AD, Deng W, Frumovitz M, Buza N, Bellone S, Huh W, et al. Phase II evaluation of nivolumab in the treatment of persistent or recurrent cervical cancer (NCT02257528/NRG-GY002). Gynecol Oncol. (2020) 157:161–6. doi: 10.1016/j.ygyno.2019.12.034

PubMed Abstract | Crossref Full Text | Google Scholar

146. Tamura K, Hasegawa K, Katsumata N, Matsumoto K, Mukai H, Takahashi S, et al. Efficacy and safety of nivolumab in Japanese patients with uterine cervical cancer, uterine corpus cancer, or soft tissue sarcoma: Multicenter, open-label phase 2 trial. Cancer Sci. (2019) 110:2894–904. doi: 10.1111/cas.v110.9

Crossref Full Text | Google Scholar

147. Fakhry C, Westra WH, Li S, Cmelak A, Ridge JA, Pinto H, et al. Improved survival of patients with human papillomavirus-positive head and neck squamous cell carcinoma in a prospective clinical trial. J Natl Cancer Inst. (2008) 100:261–9. doi: 10.1093/jnci/djn011

PubMed Abstract | Crossref Full Text | Google Scholar

148. Chaturvedi AK, Engels EA, Pfeiffer RM, Hernandez BY, Xiao W, Kim E, et al. Human papillomavirus and rising oropharyngeal cancer incidence in the United States. J Clin Oncol. (2011) 29:4294–301. doi: 10.1200/JCO.2011.36.4596

PubMed Abstract | Crossref Full Text | Google Scholar

149. Xu Y, Zhu G, Maroun CA, Wu IXY, Huang D, Seiwert TY, et al. Programmed death-1/programmed death-ligand 1-axis blockade in recurrent or metastatic head and neck squamous cell carcinoma stratified by human papillomavirus status: A systematic review and meta-analysis. Front Immunol. (2021) 12:645170. doi: 10.3389/fimmu.2021.645170

PubMed Abstract | Crossref Full Text | Google Scholar

150. Fakhry C, Zhang Q, Nguyen-Tan PF, Rosenthal D, El-Naggar A, Garden AS, et al. Human papillomavirus and overall survival after progression of oropharyngeal squamous cell carcinoma. J Clin Oncol. (2014) 32:3365–73. doi: 10.1200/JCO.2014.55.1937

PubMed Abstract | Crossref Full Text | Google Scholar

151. Wang J, Sun H, Zeng Q, Guo XJ, Wang H, Liu HH, et al. 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. doi: 10.1038/s41598-019-49771-0

PubMed Abstract | Crossref Full Text | Google Scholar

152. Badoual C, Hans S, Merillon N, Van Ryswick C, Ravel P, Benhamouda N, et al. PD-1-expressing tumor-infiltrating T cells are a favorable prognostic biomarker in HPV-associated head and neck cancer. Cancer Res. (2013) 73:128–38. doi: 10.1158/0008-5472.CAN-12-2606

PubMed Abstract | Crossref Full Text | Google Scholar

153. Patel JJ, Levy DA, Nguyen SA, Knochelmann HM, and Day TA. Impact of PD-L1 expression and human papillomavirus status in anti-PD1/PDL1 immunotherapy for head and neck squamous cell carcinoma-Systematic review and meta-analysis. Head Neck. (2020) 42:774–86. doi: 10.1002/hed.26036

PubMed Abstract | Crossref Full Text | Google Scholar

154. Tosi A, Parisatto B, Menegaldo A, Spinato G, Guido M, Del Mistro A, et al. The immune microenvironment of HPV-positive and HPV-negative oropharyngeal squamous cell carcinoma: a multiparametric quantitative and spatial analysis unveils a rationale to target treatment-naive tumors with immune checkpoint inhibitors. J Exp Clin Cancer Res. (2022) 41:279. doi: 10.1186/s13046-022-02481-4

PubMed Abstract | Crossref Full Text | Google Scholar

155. Lyford-Pike S, Peng S, Young GD, Taube JM, Westra WH, Akpeng B, 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) 73:1733–41. doi: 10.1158/0008-5472.CAN-12-2384

PubMed Abstract | Crossref Full Text | Google Scholar

156. Balermpas P, Rodel F, Krause M, Linge A, Lohaus F, Baumann M, et al. The PD-1/PD-L1 axis and human papilloma virus in patients with head and neck cancer after adjuvant chemoradiotherapy: A multicentre study of the German Cancer Consortium Radiation Oncology Group (DKTK-ROG). Int J Cancer. (2017) 141:594–603. doi: 10.1002/ijc.30770

PubMed Abstract | Crossref Full Text | Google Scholar

157. Partlova S, Boucek J, Kloudova K, Lukesova E, Zabrodsky M, Grega M, et al. Distinct patterns of intratumoral immune cell infiltrates in patients with HPV-associated compared to non-virally induced head and neck squamous cell carcinoma. Oncoimmunology. (2015) 4:e965570. doi: 10.4161/21624011.2014.965570

PubMed Abstract | Crossref Full Text | Google Scholar

158. Kim HS, Lee JY, Lim SH, Park K, Sun JM, Ko YH, et al. Association between PD-L1 and HPV status and the prognostic value of PD-L1 in oropharyngeal squamous cell carcinoma. Cancer Res Treat. (2016) 48:527–36. doi: 10.4143/crt.2015.249

PubMed Abstract | Crossref Full Text | Google Scholar

159. Ou D, Adam J, Garberis I, Blanchard P, Nguyen F, Levy A, 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. doi: 10.1080/2162402X.2017.1341030

PubMed Abstract | Crossref Full Text | Google Scholar

160. Poropatich K, Fontanarosa J, Swaminathan S, Dittmann D, Chen S, Samant S, et al. Comprehensive T-cell immunophenotyping and next-generation sequencing of human papillomavirus (HPV)-positive and HPV-negative head and neck squamous cell carcinomas. J Pathol. (2017) 243:354–65. doi: 10.1002/path.2017.243.issue-3

PubMed Abstract | Crossref Full Text | Google Scholar

161. Haddad RI, Seiwert TY, Chow LQM, Gupta S, Weiss J, Gluck I, et al. Influence of tumor mutational burden, inflammatory gene expression profile, and PD-L1 expression on response to pembrolizumab in head and neck squamous cell carcinoma. J Immunother Cancer. (2022) 10. doi: 10.1136/jitc-2021-003026

PubMed Abstract | Crossref Full Text | Google Scholar

162. Zhou L, Qiu Q, Zhou Q, Li J, Yu M, Li K, et al. Long-read sequencing unveils high-resolution HPV integration and its oncogenic progression in cervical cancer. Nat Commun. (2022) 13:2563. doi: 10.1038/s41467-022-30190-1

PubMed Abstract | Crossref Full Text | Google Scholar

163. Tsakogiannis D, Gortsilas P, Kyriakopoulou Z, Ruether IG, Dimitriou TG, Orfanoudakis G, et al. Sites of disruption within E1 and E2 genes of HPV16 and association with cervical dysplasia. J Med Virol. (2015) 87:1973–80. doi: 10.1002/jmv.v87.11

PubMed Abstract | Crossref Full Text | Google Scholar

164. Shirasawa H, Tomita Y, Fuse A, Yamamoto T, Tanzawa H, Sekiya S, et al. Structure and expression of an integrated human papillomavirus type 16 genome amplified in a cervical carcinoma cell line. J Gen Virol. (1989) 70:1913–9. doi: 10.1099/0022-1317-70-7-1913

PubMed Abstract | Crossref Full Text | Google Scholar

165. Yamato K, Yamada T, Kizaki M, Ui-Tei K, Natori Y, Fujino M, et al. New highly potent and specific E6 and E7 siRNAs for treatment of HPV16 positive cervical cancer. Cancer Gene Ther. (2008) 15:140–53. doi: 10.1038/sj.cgt.7701118

PubMed Abstract | Crossref Full Text | Google Scholar

166. Jabbar SF, Abrams L, Glick A, and Lambert PF. Persistence of high-grade cervical dysplasia and cervical cancer requires the continuous expression of the human papillomavirus type 16 E7 oncogene. Cancer Res. (2009) 69:4407–14. doi: 10.1158/0008-5472.CAN-09-0023

PubMed Abstract | Crossref Full Text | Google Scholar

167. Hayes DN, Van Waes C, and Seiwert TY. Genetic landscape of human papillomavirus-associated head and neck cancer and comparison to tobacco-related tumors. J Clin Oncol. (2015) 33:3227–34. doi: 10.1200/JCO.2015.62.1086

PubMed Abstract | Crossref Full Text | Google Scholar

168. Leemans CR, Snijders PJF, and Brakenhoff RH. The molecular landscape of head and neck cancer. Nat Rev Cancer. (2018) 18:269–82. doi: 10.1038/nrc.2018.11

PubMed Abstract | Crossref Full Text | Google Scholar

169. Seiwert TY, Zuo Z, Keck MK, Khattri A, Pedamallu CS, Stricker T, et al. Integrative and comparative genomic analysis of HPV-positive and HPV-negative head and neck squamous cell carcinomas. Clin Cancer Res. (2015) 21:632–41. doi: 10.1158/1078-0432.CCR-13-3310

PubMed Abstract | Crossref Full Text | Google Scholar

170. Morris LGT, Chandramohan R, West L, Zehir A, Chakravarty D, Pfister DG, et al. The molecular landscape of recurrent and metastatic head and neck cancers: insights from a precision oncology sequencing platform. JAMA Oncol. (2017) 3:244–55. doi: 10.1001/jamaoncol.2016.1790

PubMed Abstract | Crossref Full Text | Google Scholar

171. Hanna GJ, Lizotte P, Cavanaugh M, Kuo FC, Shivdasani P, Frieden A, et al. Frameshift events predict anti-PD-1/L1 response in head and neck cancer. JCI Insight. (2018) 3. doi: 10.1172/jci.insight.98811

PubMed Abstract | Crossref Full Text | Google Scholar

172. Stransky N, Egloff AM, Tward AD, Kostic AD, Cibulskis K, Sivachenko A, et al. The mutational landscape of head and neck squamous cell carcinoma. Science. (2011) 333:1157–60. doi: 10.1126/science.1208130

PubMed Abstract | Crossref Full Text | Google Scholar

173. Agrawal N, Frederick MJ, Pickering CR, Bettegowda C, Chang K, Li RJ, et al. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science. (2011) 333:1154–7. doi: 10.1126/science.1206923

PubMed Abstract | Crossref Full Text | Google Scholar

174. Conarty JP and Wieland A. The tumor-specific immune landscape in HPV+ Head and neck cancer. Viruses. (2023) 15. doi: 10.3390/v15061296

PubMed Abstract | Crossref Full Text | Google Scholar

175. Morgan IM, DiNardo LJ, and Windle B. Integration of human papillomavirus genomes in head and neck cancer: is it time to consider a paradigm shift? Viruses. (2017) 9. doi: 10.3390/v9080208

PubMed Abstract | Crossref Full Text | Google Scholar

176. Nulton TJ, Olex AL, Dozmorov M, Morgan IM, and Windle B. Analysis of The Cancer Genome Atlas sequencing data reveals novel properties of the human papillomavirus 16 genome in head and neck squamous cell carcinoma. Oncotarget. (2017) 8:17684–99. doi: 10.18632/oncotarget.15179

PubMed Abstract | Crossref Full Text | Google Scholar

177. Koneva LA, Zhang Y, Virani S, Hall PB, McHugh JB, Chepeha DB, et al. HPV integration in HNSCC correlates with survival outcomes, immune response signatures, and candidate drivers. Mol Cancer Res. (2018) 16:90–102. doi: 10.1158/1541-7786.MCR-17-0153

PubMed Abstract | Crossref Full Text | Google Scholar

178. Williams VM, Filippova M, Soto U, and Duerksen-Hughes PJ. HPV-DNA integration and carcinogenesis: putative roles for inflammation and oxidative stress. Future Virol. (2011) 6:45–57. doi: 10.2217/fvl.10.73

PubMed Abstract | Crossref Full Text | Google Scholar

179. Graham SV. Human papillomavirus: gene expression, regulation and prospects for novel diagnostic methods and antiviral therapies. Future Microbiol. (2010) 5:1493–506. doi: 10.2217/fmb.10.107

PubMed Abstract | Crossref Full Text | Google Scholar

180. Parfenov M, Pedamallu CS, Gehlenborg N, Freeman SS, Danilova L, Bristow CA, et al. Characterization of HPV and host genome interactions in primary head and neck cancers. Proc Natl Acad Sci U S A. (2014) 111:15544–9. doi: 10.1073/pnas.1416074111

PubMed Abstract | Crossref Full Text | Google Scholar

181. Nulton TJ, Kim NK, DiNardo LJ, Morgan IM, and Windle B. Patients with integrated HPV16 in head and neck cancer show poor survival. Oral Oncol. (2018) 80:52–5. doi: 10.1016/j.oraloncology.2018.03.015

PubMed Abstract | Crossref Full Text | Google Scholar

182. Zhang Y, Koneva LA, Virani S, Arthur AE, Virani A, Hall PB, et al. Subtypes of HPV-positive head and neck cancers are associated with HPV characteristics, copy number alterations, PIK3CA mutation, and pathway signatures. Clin Cancer Res. (2016) 22:4735–45. doi: 10.1158/1078-0432.CCR-16-0323

PubMed Abstract | Crossref Full Text | Google Scholar

183. Cancer Genome Atlas N. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. (2015) 517:576–82. doi: 10.1038/nature14129

PubMed Abstract | Crossref Full Text | Google Scholar

184. Chung CH, Guthrie VB, Masica DL, Tokheim C, Kang H, Richmon J, et al. Genomic alterations in head and neck squamous cell carcinoma determined by cancer gene-targeted sequencing. Ann Oncol. (2015) 26:1216–23. doi: 10.1093/annonc/mdv109

PubMed Abstract | Crossref Full Text | Google Scholar

185. Muijlwijk T, Nauta IH, van der Lee A, Grunewald KJT, Brink A, Ganzevles SH, et al. Hallmarks of a genomically distinct subclass of head and neck cancer. Nat Commun. (2024) 15:9060. doi: 10.1038/s41467-024-53390-3

PubMed Abstract | Crossref Full Text | Google Scholar

186. Schutt CR, Sun H, Pradhan JS, Saenger Y, Ley J, Adkins D, et al. Genomic and neoantigen evolution from primary tumor to first metastases in head and neck squamous cell carcinoma. Oncotarget. (2021) 12:534–48. doi: 10.18632/oncotarget.27907

PubMed Abstract | Crossref Full Text | Google Scholar

187. Rodrigo JP, Sanchez-Canteli M, Otero-Rosales M, Martinez-Camblor P, Hermida-Prado F, and Garcia-Pedrero JM. Tumor mutational burden predictability in head and neck squamous cell carcinoma patients treated with immunotherapy: systematic review and meta-analysis. J Transl Med. (2024) 22:135. doi: 10.1186/s12967-024-04937-x

PubMed Abstract | Crossref Full Text | Google Scholar

188. Eberhardt CS, Kissick HT, Patel MR, Cardenas MA, Prokhnevska N, Obeng RC, et al. Functional HPV-specific PD-1(+) stem-like CD8 T cells in head and neck cancer. Nature. (2021) 597:279–84. doi: 10.1038/s41586-021-03862-z

PubMed Abstract | Crossref Full Text | Google Scholar

189. Cillo AR, Kurten CHL, Tabib T, Qi Z, Onkar S, Wang T, et al. Immune landscape of viral- and carcinogen-driven head and neck cancer. Immunity. (2020) 52:183–99 e9. doi: 10.1016/j.immuni.2019.11.014

PubMed Abstract | Crossref Full Text | Google Scholar

190. Gameiro SF, Ghasemi F, Barrett JW, Koropatnick J, Nichols AC, Mymryk JS, et al. Treatment-naive HPV+ head and neck cancers display a T-cell-inflamed phenotype distinct from their HPV- counterparts that has implications for immunotherapy. Oncoimmunology. (2018) 7:e1498439. doi: 10.1080/2162402X.2018.1498439

PubMed Abstract | Crossref Full Text | Google Scholar

191. Kindt N, Descamps G, Seminerio I, Bellier J, Lechien JR, Mat Q, et al. High stromal Foxp3-positive T cell number combined to tumor stage improved prognosis in head and neck squamous cell carcinoma. Oral Oncol. (2017) 67:183–91. doi: 10.1016/j.oraloncology.2017.02.023

PubMed Abstract | Crossref Full Text | Google Scholar

192. Russell S, Angell T, Lechner M, Liebertz D, Correa A, Sinha U, et al. Immune cell infiltration patterns and survival in head and neck squamous cell carcinoma. Head Neck Oncol. (2013) 5:24.

PubMed Abstract | Google Scholar

193. Lukesova E, Boucek J, Rotnaglova E, Salakova M, Koslabova E, Grega M, et al. High level of Tregs is a positive prognostic marker in patients with HPV-positive oral and oropharyngeal squamous cell carcinomas. BioMed Res Int. (2014) 2014:303929. doi: 10.1155/2014/303929

PubMed Abstract | Crossref Full Text | Google Scholar

194. Nasman A, Romanitan M, Nordfors C, Grun N, Johansson H, Hammarstedt L, et al. Tumor infiltrating CD8+ and Foxp3+ lymphocytes correlate to clinical outcome and human papillomavirus (HPV) status in tonsillar cancer. PloS One. (2012) 7:e38711. doi: 10.1371/journal.pone.0038711

PubMed Abstract | Crossref Full Text | Google Scholar

195. Wood O, Woo J, Seumois G, Savelyeva N, McCann KJ, Singh D, 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) 7:56781–97. doi: 10.18632/oncotarget.10788

PubMed Abstract | Crossref Full Text | Google Scholar

196. Wieland A, Patel MR, Cardenas MA, Eberhardt CS, Hudson WH, Obeng RC, et al. Defining HPV-specific B cell responses in patients with head and neck cancer. Nature. (2021) 597:274–8. doi: 10.1038/s41586-020-2931-3

PubMed Abstract | Crossref Full Text | Google Scholar

197. Lang Kuhs KA, Kreimer AR, Trivedi S, Holzinger D, Pawlita M, Pfeiffer RM, et al. Human papillomavirus 16 E6 antibodies are sensitive for human papillomavirus-driven oropharyngeal cancer and are associated with recurrence. Cancer. (2017) 123:4382–90. doi: 10.1002/cncr.v123.22

PubMed Abstract | Crossref Full Text | Google Scholar

198. Dahlstrom KR, Anderson KS, Cheng JN, Chowell D, Li G, Posner M, et al. HPV serum antibodies as predictors of survival and disease progression in patients with HPV-positive squamous cell carcinoma of the oropharynx. Clin Cancer Res. (2015) 21:2861–9. doi: 10.1158/1078-0432.CCR-14-3323

PubMed Abstract | Crossref Full Text | Google Scholar

199. Zhang S, Wang B, Ma F, Tong F, Yan B, Liu T, et al. Characteristics of B lymphocyte infiltration in HPV(+) head and neck squamous cell carcinoma. Cancer Sci. (2021) 112:1402–16. doi: 10.1111/cas.v112.4

PubMed Abstract | Crossref Full Text | Google Scholar

200. Kim SS, Shen S, Miyauchi S, Sanders PD, Franiak-Pietryga I, Mell L, et al. B cells improve overall survival in HPV-associated squamous cell carcinomas and are activated by radiation and PD-1 blockade. Clin Cancer Res. (2020) 26:3345–59. doi: 10.1158/1078-0432.CCR-19-3211

PubMed Abstract | Crossref Full Text | Google Scholar

201. Coenon L, Geindreau M, Ghiringhelli F, Villalba M, and Bruchard M. Natural Killer cells at the frontline in the fight against cancer. Cell Death Dis. (2024) 15:614. doi: 10.1038/s41419-024-06976-0

PubMed Abstract | Crossref Full Text | Google Scholar

202. Mandal R, Senbabaoglu Y, Desrichard A, Havel JJ, Dalin MG, Riaz N, et al. The head and neck cancer immune landscape and its immunotherapeutic implications. JCI Insight. (2016) 1:e89829. doi: 10.1172/jci.insight.89829

PubMed Abstract | Crossref Full Text | Google Scholar

203. Wagner S, Wittekindt C, Reuschenbach M, Hennig B, Thevarajah M, Wurdemann N, et al. CD56-positive lymphocyte infiltration in relation to human papillomavirus association and prognostic significance in oropharyngeal squamous cell carcinoma. Int J Cancer. (2016) 138:2263–73. doi: 10.1002/ijc.v138.9

PubMed Abstract | Crossref Full Text | Google Scholar

204. Nasman A, Andersson E, Marklund L, Tertipis N, Hammarstedt-Nordenvall L, Attner P, et al. HLA class I and II expression in oropharyngeal squamous cell carcinoma in relation to tumor HPV status and clinical outcome. PloS One. (2013) 8:e77025. doi: 10.1371/journal.pone.0077025

PubMed Abstract | Crossref Full Text | Google Scholar

205. Nasman A, Andersson E, Nordfors C, Grun N, Johansson H, Munck-Wikland E, et al. MHC class I expression in HPV positive and negative tonsillar squamous cell carcinoma in correlation to clinical outcome. Int J Cancer. (2013) 132:72–81. doi: 10.1002/ijc.v132.1

Crossref Full Text | Google Scholar

206. Lanier LL. NK cell recognition. Annu Rev Immunol. (2005) 23:225–74. doi: 10.1146/annurev.immunol.23.021704.115526

PubMed Abstract | Crossref Full Text | Google Scholar

207. Rouas-Freiss N, Goncalves RM, Menier C, Dausset J, and Carosella ED. Direct evidence to support the role of HLA-G in protecting the fetus from maternal uterine natural killer cytolysis. Proc Natl Acad Sci U S A. (1997) 94:11520–5. doi: 10.1073/pnas.94.21.11520

PubMed Abstract | Crossref Full Text | Google Scholar

208. Moreno-Nieves UY, Tay JK, Saumyaa S, Horowitz NB, Shin JH, Mohammad IA, et al. Landscape of innate lymphoid cells in human head and neck cancer reveals divergent NK cell states in the tumor microenvironment. Proc Natl Acad Sci U.S.A. (2021) 118. doi: 10.1073/pnas.2101169118

PubMed Abstract | Crossref Full Text | Google Scholar

209. Torcellan T, Friedrich C, Doucet-Ladeveze R, Ossner T, Sole VV, Riedmann S, et al. Circulating NK cells establish tissue residency upon acute infection of skin and mediate accelerated effector responses to secondary infection. Immunity. (2024) 57:124–40 e7. doi: 10.1016/j.immuni.2023.11.018

PubMed Abstract | Crossref Full Text | Google Scholar

210. Schuster IS, Sng XYX, Lau CM, Powell DR, Weizman OE, Fleming P, et al. Infection induces tissue-resident memory NK cells that safeguard tissue health. Immunity. (2023) 56:531–46 e6. doi: 10.1016/j.immuni.2023.01.016

PubMed Abstract | Crossref Full Text | Google Scholar

211. Seminerio I, Kindt N, Descamps G, Bellier J, Lechien JR, Mat Q, et al. High infiltration of CD68+ macrophages is associated with poor prognoses of head and neck squamous cell carcinoma patients and is influenced by human papillomavirus. Oncotarget. (2018) 9:11046–59. doi: 10.18632/oncotarget.24306

PubMed Abstract | Crossref Full Text | Google Scholar

212. Chen X, Yan B, Lou H, Shen Z, Tong F, Zhai A, et al. Immunological network analysis in HPV associated head and neck squamous cancer and implications for disease prognosis. Mol Immunol. (2018) 96:28–36. doi: 10.1016/j.molimm.2018.02.005

PubMed Abstract | Crossref Full Text | Google Scholar

213. Koucky V, Hladikova K, Taborska E, Boucek J, Grega M, Spisek R, et al. The cytokine milieu compromises functional capacity of tumor-infiltrating plasmacytoid dendritic cells in HPV-negative but not in HPV-positive HNSCC. Cancer Immunol Immunother. (2021) 70:2545–57. doi: 10.1007/s00262-021-02874-y

PubMed Abstract | Crossref Full Text | Google Scholar

214. Zhou C, Tuong ZK, and Frazer IH. Papillomavirus immune evasion strategies target the infected cell and the local immune system. Front Oncol. (2019) 9:682. doi: 10.3389/fonc.2019.00682

PubMed Abstract | Crossref Full Text | Google Scholar

215. Miyauchi S, Roy S, Boutros N, and Sharabi AB. Virus-mediated immunosuppression in head and neck cancer. Oncogene. (2025) 44:933–43. doi: 10.1038/s41388-025-03295-2

PubMed Abstract | Crossref Full Text | Google Scholar

216. Westrich JA, Warren CJ, and Pyeon D. Evasion of host immune defenses by human papillomavirus. Virus Res. (2017) 231:21–33. doi: 10.1016/j.virusres.2016.11.023

PubMed Abstract | Crossref Full Text | Google Scholar

217. Nees M, Geoghegan JM, Hyman T, Frank S, Miller L, and Woodworth CD. Papillomavirus type 16 oncogenes downregulate expression of interferon-responsive genes and upregulate proliferation-associated and NF-kappaB-responsive genes in cervical keratinocytes. J Virol. (2001) 75:4283–96. doi: 10.1128/JVI.75.9.4283-4296.2001

PubMed Abstract | Crossref Full Text | Google Scholar

218. Tilborghs S, Corthouts J, Verhoeven Y, Arias D, Rolfo C, Trinh XB, et al. The role of Nuclear Factor-kappa B signaling in human cervical cancer. Crit Rev Oncol Hematol. (2017) 120:141–50. doi: 10.1016/j.critrevonc.2017.11.001

PubMed Abstract | Crossref Full Text | Google Scholar

219. Huang SM and McCance DJ. Down regulation of the interleukin-8 promoter by human papillomavirus type 16 E6 and E7 through effects on CREB binding protein/p300 and P/CAF. J Virol. (2002) 76:8710–21. doi: 10.1128/JVI.76.17.8710-8721.2002

PubMed Abstract | Crossref Full Text | Google Scholar

220. Zimmermann H, Degenkolbe R, Bernard HU, and O'Connor MJ. The human papillomavirus type 16 E6 oncoprotein can down-regulate p53 activity by targeting the transcriptional coactivator CBP/p300. J Virol. (1999) 73:6209–19. doi: 10.1128/JVI.73.8.6209-6219.1999

PubMed Abstract | Crossref Full Text | Google Scholar

221. Ronco LV, Karpova AY, Vidal M, and Howley PM. Human papillomavirus 16 E6 oncoprotein binds to interferon regulatory factor-3 and inhibits its transcriptional activity. Genes Dev. (1998) 12:2061–72. doi: 10.1101/gad.12.13.2061

PubMed Abstract | Crossref Full Text | Google Scholar

222. Park JS, Kim EJ, Kwon HJ, Hwang ES, Namkoong SE, and Um SJ. Inactivation of interferon regulatory factor-1 tumor suppressor protein by HPV E7 oncoprotein. Implication for the E7-mediated immune evasion mechanism in cervical carcinogenesis. J Biol Chem. (2000) 275:6764–9. doi: 10.1074/jbc.275.10.6764

PubMed Abstract | Crossref Full Text | Google Scholar

223. Chang YE and Laimins LA. Microarray analysis identifies interferon-inducible genes and Stat-1 as major transcriptional targets of human papillomavirus type 31. J Virol. (2000) 74:4174–82. doi: 10.1128/JVI.74.9.4174-4182.2000

PubMed Abstract | Crossref Full Text | Google Scholar

224. Hong S, Mehta KP, and Laimins LA. Suppression of STAT-1 expression by human papillomaviruses is necessary for differentiation-dependent genome amplification and plasmid maintenance. J Virol. (2011) 85:9486–94. doi: 10.1128/JVI.05007-11

PubMed Abstract | Crossref Full Text | Google Scholar

225. Georgopoulos NT, Proffitt JL, and Blair GE. 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) 19:4930–5. doi: 10.1038/sj.onc.1203860

PubMed Abstract | Crossref Full Text | Google Scholar

226. Heller C, Weisser T, Mueller-Schickert A, Rufer E, Hoh A, Leonhardt RM, et al. Identification of key amino acid residues that determine the ability of high risk HPV16-E7 to dysregulate major histocompatibility complex class I expression. J Biol Chem. (2011) 286:10983–97. doi: 10.1074/jbc.M110.199190

PubMed Abstract | Crossref Full Text | Google Scholar

227. Li H, Ou X, Xiong J, and Wang T. HPV16E7 mediates HADC chromatin repression and downregulation of MHC class I genes in HPV16 tumorigenic cells through interaction with an MHC class I promoter. Biochem Biophys Res Commun. (2006) 349:1315–21. doi: 10.1016/j.bbrc.2006.08.182

PubMed Abstract | Crossref Full Text | Google Scholar

228. Li H, Zhan T, Li C, Liu M, and Wang QK. Repression of MHC class I transcription by HPV16E7 through interaction with a putative RXRbeta motif and NF-kappaB cytoplasmic sequestration. Biochem Biophys Res Commun. (2009) 388:383–8. doi: 10.1016/j.bbrc.2009.08.019

PubMed Abstract | Crossref Full Text | Google Scholar

229. Ashrafi GH, Haghshenas MR, Marchetti B, O'Brien PM, and Campo MS. E5 protein of human papillomavirus type 16 selectively downregulates surface HLA class I. Int J Cancer. (2005) 113:276–83. doi: 10.1002/ijc.v113:2

PubMed Abstract | Crossref Full Text | Google Scholar

230. Ashrafi GH, Haghshenas M, Marchetti B, and Campo MS. E5 protein of human papillomavirus 16 downregulates HLA class I and interacts with the heavy chain via its first hydrophobic domain. Int J Cancer. (2006) 119:2105–12. doi: 10.1002/ijc.v119:9

PubMed Abstract | Crossref Full Text | Google Scholar

231. Senba M and Mori N. Mechanisms of virus immune evasion lead to development from chronic inflammation to cancer formation associated with human papillomavirus infection. Oncol Rev. (2012) 6:e17. doi: 10.4081/oncol.2012.e17

PubMed Abstract | Crossref Full Text | Google Scholar

232. Zheng J, Huang J, Ma W, Yang W, and Hu B. The antitumor activity of CAR-T-PD1 cells enhanced by HPV16mE7-pulsed and SOCS1-silenced DCs in cervical cancer models. Cancer Manag Res. (2021) 13:6045–53. doi: 10.2147/CMAR.S321402

PubMed Abstract | Crossref Full Text | Google Scholar

233. Lee WT, Tamai H, Cohen P, Teker AM, and Shu S. Immunotherapy of established murine squamous cell carcinoma using fused dendritic-tumor cell hybrids. Arch Otolaryngol Head Neck Surg. (2008) 134:608–13. doi: 10.1001/archotol.134.6.608

PubMed Abstract | Crossref Full Text | Google Scholar

234. Nagarsheth NB, Norberg SM, Sinkoe AL, Adhikary S, Meyer TJ, Lack JB, et al. TCR-engineered T cells targeting E7 for patients with metastatic HPV-associated epithelial cancers. Nat Med. (2021) 27:419–25. doi: 10.1038/s41591-020-01225-1

PubMed Abstract | Crossref Full Text | Google Scholar

235. Jin BY, Campbell TE, Draper LM, Stevanovic S, Weissbrich B, Yu Z, et al. Engineered T cells targeting E7 mediate regression of human papillomavirus cancers in a murine model. JCI Insight. (2018) 3. doi: 10.1172/jci.insight.99488

PubMed Abstract | Crossref Full Text | Google Scholar

236. Wang R, Pan W, Jin L, Huang W, Li Y, Wu D, et al. Human papillomavirus vaccine against cervical cancer: Opportunity and challenge. Cancer Lett. (2020) 471:88–102. doi: 10.1016/j.canlet.2019.11.039

PubMed Abstract | Crossref Full Text | Google Scholar

237. Miles B, Safran HP, and Monk BJ. Therapeutic options for treatment of human papillomavirus-associated cancers - novel immunologic vaccines: ADXS11-001. Gynecol Oncol Res Pract. (2017) 4:10. doi: 10.1186/s40661-017-0047-8

PubMed Abstract | Crossref Full Text | Google Scholar

238. Toes RE, Blom RJ, Offringa R, Kast WM, and Melief CJ. Enhanced tumor outgrowth after peptide vaccination. Functional deletion of tumor-specific CTL induced by peptide vaccination can lead to the inability to reject tumors. J Immunol. (1996) 156:3911–8. doi: 10.4049/jimmunol.156.10.3911

PubMed Abstract | Crossref Full Text | Google Scholar

239. Miles BA, Monk BJ, and Safran HP. Mechanistic insights into ADXS11–001 human papillomavirus-associated cancer immunotherapy. Gynecol Oncol Res Pract. (2017) 4:9. doi: 10.1186/s40661-017-0046-9

PubMed Abstract | Crossref Full Text | Google Scholar

240. Galicia-Carmona T, Arango-Bravo E, Serrano-Olvera JA, Flores-de La Torre C, Cruz-Esquivel I, Villalobos-Valencia R, et al. ADXS11–001 LM-LLO as specific immunotherapy in cervical cancer. Hum Vaccin Immunother. (2021) 17:2617–25. doi: 10.1080/21645515.2021.1893036

PubMed Abstract | Crossref Full Text | Google Scholar

241. Basu P, Mehta A, Jain M, Gupta S, Nagarkar RV, John S, et al. A randomized phase 2 study of ADXS11–001 listeria monocytogenes-listeriolysin O immunotherapy with or without cisplatin in treatment of advanced cervical cancer. Int J Gynecol Cancer. (2018) 28:764–72. doi: 10.1097/IGC.0000000000001235

PubMed Abstract | Crossref Full Text | Google Scholar

242. Baumforth KR, Young LS, Flavell KJ, Constandinou C, and Murray PG. The Epstein-Barr virus and its association with human cancers. Mol Pathol. (1999) 52:307–22. doi: 10.1136/mp.52.6.307

PubMed Abstract | Crossref Full Text | Google Scholar

243. Huang H, Yao Y, Deng X, Weng H, Chen Z, Yu L, et al. Characteristics of immunotherapy trials for nasopharyngeal carcinoma over a 15-year period. Front Immunol. (2023) 14:1195659. doi: 10.3389/fimmu.2023.1195659

PubMed Abstract | Crossref Full Text | Google Scholar

244. Naseem M, Barzi A, Brezden-Masley C, Puccini A, Berger MD, Tokunaga R, et al. Outlooks on Epstein-Barr virus associated gastric cancer. Cancer Treat Rev. (2018) 66:15–22. doi: 10.1016/j.ctrv.2018.03.006

PubMed Abstract | Crossref Full Text | Google Scholar

245. Nemtsova MV, Kuznetsova EB, and Bure IV. Chromosomal instability in gastric cancer: role in tumor development, progression, and therapy. Int J Mol Sci. (2023) 24. doi: 10.3390/ijms242316961

PubMed Abstract | Crossref Full Text | Google Scholar

246. Nishikawa J, Iizasa H, Yoshiyama H, Nakamura M, Saito M, Sasaki S, et al. The role of epigenetic regulation in epstein-barr virus-associated gastric cancer. Int J Mol Sci. (2017) 18. doi: 10.3390/ijms18081606

PubMed Abstract | Crossref Full Text | Google Scholar

247. Shin HJ, Kim DN, and Lee SK. Association between Epstein-Barr virus infection and chemoresistance to docetaxel in gastric carcinoma. Mol Cells. (2011) 32:173–9. doi: 10.1007/s10059-011-0066-y

PubMed Abstract | Crossref Full Text | Google Scholar

248. Cancer Genome Atlas Research N. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. (2014) 513:202–9. doi: 10.1038/nature13480

PubMed Abstract | Crossref Full Text | Google Scholar

249. Park JH, Kim EK, Kim YH, Kim JH, Bae YS, Lee YC, et al. Epstein-Barr virus positivity, not mismatch repair-deficiency, is a favorable risk factor for lymph node metastasis in submucosa-invasive early gastric cancer. Gastric Cancer. (2016) 19:1041–51. doi: 10.1007/s10120-015-0565-1

PubMed Abstract | Crossref Full Text | Google Scholar

250. Xing X, Guo J, Ding G, Li B, Dong B, Feng Q, et al. Analysis of PD1, PDL1, PDL2 expression and T cells infiltration in 1014 gastric cancer patients. Oncoimmunology. (2018) 7:e1356144. doi: 10.1080/2162402X.2017.1356144

PubMed Abstract | Crossref Full Text | Google Scholar

251. Su S, Zou Z, Chen F, Ding N, Du J, Shao J, et al. CRISPR-Cas9-mediated disruption of PD-1 on human T cells for adoptive cellular therapies of EBV positive gastric cancer. Oncoimmunology. (2017) 6:e1249558. doi: 10.1080/2162402X.2016.1249558

PubMed Abstract | Crossref Full Text | Google Scholar

252. Seo AN, Kang BW, Kwon OK, Park KB, Lee SS, Chung HY, et al. Intratumoural PD-L1 expression is associated with worse survival of patients with Epstein-Barr virus-associated gastric cancer. Br J Cancer. (2017) 117:1753–60. doi: 10.1038/bjc.2017.369

PubMed Abstract | Crossref Full Text | Google Scholar

253. Ma J, Li J, Hao Y, Nie Y, Li Z, Qian M, et al. Differentiated tumor immune microenvironment of Epstein-Barr virus-associated and negative gastric cancer: implication in prognosis and immunotherapy. Oncotarget. (2017) 8:67094–103. doi: 10.18632/oncotarget.17945

PubMed Abstract | Crossref Full Text | Google Scholar

254. Panda A, Mehnert JM, Hirshfield KM, Riedlinger G, Damare S, Saunders T, et al. Immune activation and benefit from avelumab in EBV-positive gastric cancer. J Natl Cancer Inst. (2018) 110:316–20. doi: 10.1093/jnci/djx213

PubMed Abstract | Crossref Full Text | Google Scholar

255. Pan Y, Lu L, Liu H, Chen D, Han N, Yao R, et al. Case report: Long response to PD-1 blockade after failure of trastuzumab plus chemotherapy in advanced Epstein-Barr virus-associated gastric cancer. Front Immunol. (2022) 13:1003859. doi: 10.3389/fimmu.2022.1003859

PubMed Abstract | Crossref Full Text | Google Scholar

256. Janjigian YY, Sanchez-Vega F, Jonsson P, Chatila WK, Hechtman JF, Ku GY, et al. Genetic predictors of response to systemic therapy in esophagogastric cancer. Cancer Discov. (2018) 8:49–58. doi: 10.1158/2159-8290.CD-17-0787

PubMed Abstract | Crossref Full Text | Google Scholar

257. Park Y, Koh J, Kwak Y, Ahn SH, Park DJ, Kim HH, et al. Clinicopathologic significance of human leukocyte antigen class I expression in patients with stage II and III gastric cancer. Cancer Immunol Immunother. (2019) 68:1779–90. doi: 10.1007/s00262-019-02410-z

PubMed Abstract | Crossref Full Text | Google Scholar

258. Ghasemi F, Gameiro SF, Tessier TM, Maciver AH, and Mymryk JS. High levels of class I major histocompatibility complex mRNA are present in epstein-barr virus-associated gastric adenocarcinomas. Cells. (2020) 9. doi: 10.3390/cells9020499

PubMed Abstract | Crossref Full Text | Google Scholar

259. Ghasemi F, Tessier TM, Gameiro SF, Maciver AH, Cecchini MJ, and Mymryk JS. High MHC-II expression in Epstein-Barr virus-associated gastric cancers suggests that tumor cells serve an important role in antigen presentation. Sci Rep. (2020) 10:14786. doi: 10.1038/s41598-020-71775-4

PubMed Abstract | Crossref Full Text | Google Scholar

260. Ma XC, Hattori T, Kushima R, Terata N, and Kodama M. Expression of HLA-class II antigen in gastric carcinomas. Its relationship to histopathological grade, lymphocyte infiltration and five-year survival rate. Acta Oncol. (1994) 33:187–90. doi: 10.3109/02841869409098403

PubMed Abstract | Crossref Full Text | Google Scholar

261. van Beek J, zur Hausen A, Klein Kranenbarg E, van de Velde CJ, Middeldorp JM, van den Brule AJ, et al. EBV-positive gastric adenocarcinomas: a distinct clinicopathologic entity with a low frequency of lymph node involvement. J Clin Oncol. (2004) 22:664–70. doi: 10.1200/JCO.2004.08.061

PubMed Abstract | Crossref Full Text | Google Scholar

262. van Beek J, zur Hausen A, Snel SN, Berkhof J, Kranenbarg EK, van de Velde CJ, et al. Morphological evidence of an activated cytotoxic T-cell infiltrate in EBV-positive gastric carcinoma preventing lymph node metastases. Am J Surg Pathol. (2006) 30:59–65. doi: 10.1097/01.pas.0000176428.06629.1e

PubMed Abstract | Crossref Full Text | Google Scholar

263. Hinata M, Kunita A, Abe H, Morishita Y, Sakuma K, Yamashita H, et al. Exosomes of epstein-barr virus-associated gastric carcinoma suppress dendritic cell maturation. Microorganisms. (2020) 8. doi: 10.3390/microorganisms8111776

PubMed Abstract | Crossref Full Text | Google Scholar

264. Su ZY, Siak PY, Leong CO, and Cheah SC. The role of Epstein-Barr virus in nasopharyngeal carcinoma. Front Microbiol. (2023) 14:1116143. doi: 10.3389/fmicb.2023.1116143

PubMed Abstract | Crossref Full Text | Google Scholar

265. Fahraeus R, Fu HL, Ernberg I, Finke J, Rowe M, Klein G, et al. Expression of Epstein-Barr virus-encoded proteins in nasopharyngeal carcinoma. Int J Cancer. (1988) 42:329–38. doi: 10.1002/ijc.2910420305

PubMed Abstract | Crossref Full Text | Google Scholar

266. Wang L, Tian WD, Xu X, Nie B, Lu J, Liu X, et al. Epstein-Barr virus nuclear antigen 1 (EBNA1) protein induction of epithelial-mesenchymal transition in nasopharyngeal carcinoma cells. Cancer. (2014) 120:363–72. doi: 10.1002/cncr.v120.3

PubMed Abstract | Crossref Full Text | Google Scholar

267. Huo S, Luo Y, Deng R, Liu X, Wang J, Wang L, et al. EBV-EBNA1 constructs an immunosuppressive microenvironment for nasopharyngeal carcinoma by promoting the chemoattraction of Treg cells. J Immunother Cancer. (2020) 8. doi: 10.1136/jitc-2020-001588

PubMed Abstract | Crossref Full Text | Google Scholar

268. Wang J, Luo Y, Bi P, Lu J, Wang F, Liu X, et al. Mechanisms of Epstein-Barr virus nuclear antigen 1 favor Tregs accumulation in nasopharyngeal carcinoma. Cancer Med. (2020) 9:5598–608. doi: 10.1002/cam4.v9.15

PubMed Abstract | Crossref Full Text | Google Scholar

269. Trivedi P, Winberg G, and Klein G. Differential immunogenicity of Epstein-Barr virus (EBV) encoded growth transformation-associated antigens in a murine model system. Eur J Cancer. (1997) 33:912–7. doi: 10.1016/S0959-8049(96)00514-X

PubMed Abstract | Crossref Full Text | Google Scholar

270. Li YY, Chung GT, Lui VW, To KF, Ma BB, Chow C, et al. Exome and genome sequencing of nasopharynx cancer identifies NF-kappaB pathway activating mutations. Nat Commun. (2017) 8:14121. doi: 10.1038/ncomms14121

PubMed Abstract | Crossref Full Text | Google Scholar

271. Yao Y, Minter HA, Chen X, Reynolds GM, Bromley M, and Arrand JR. Heterogeneity of HLA and EBER expression in Epstein-Barr virus-associated nasopharyngeal carcinoma. Int J Cancer. (2000) 88:949–55. doi: 10.1002/1097-0215(20001215)88:6<949::AID-IJC18>3.0.CO;2-6

PubMed Abstract | Crossref Full Text | Google Scholar

272. Jochum S, Moosmann A, Lang S, Hammerschmidt W, and Zeidler R. The EBV immunoevasins vIL-10 and BNLF2a protect newly infected B cells from immune recognition and elimination. PloS Pathog. (2012) 8:e1002704. doi: 10.1371/journal.ppat.1002704

PubMed Abstract | Crossref Full Text | Google Scholar

273. Singh S and Banerjee S. Downregulation of HLA-ABC expression through promoter hypermethylation and downmodulation of MIC-A/B surface expression in LMP2A-positive epithelial carcinoma cell lines. Sci Rep. (2020) 10:5415. doi: 10.1038/s41598-020-62081-0

PubMed Abstract | Crossref Full Text | Google Scholar

274. Albanese M, Tagawa T, Bouvet M, Maliqi L, Lutter D, Hoser J, et al. Epstein-Barr virus microRNAs reduce immune surveillance by virus-specific CD8+ T cells. Proc Natl Acad Sci U S A. (2016) 113:E6467–E75. doi: 10.1073/pnas.1605884113

PubMed Abstract | Crossref Full Text | Google Scholar

275. Tagawa T, Albanese M, Bouvet M, Moosmann A, Mautner J, Heissmeyer V, et al. Epstein-Barr viral miRNAs inhibit antiviral CD4+ T cell responses targeting IL-12 and peptide processing. J Exp Med. (2016) 213:2065–80. doi: 10.1084/jem.20160248

PubMed Abstract | Crossref Full Text | Google Scholar

276. Ge J, Wang J, Xiong F, Jiang X, Zhu K, Wang Y, et al. Epstein-barr virus-encoded circular RNA circBART2.2 promotes immune escape of nasopharyngeal carcinoma by regulating PD-L1. Cancer Res. (2021) 81:5074–88. doi: 10.1158/0008-5472.CAN-20-4321

PubMed Abstract | Crossref Full Text | Google Scholar

277. Li JP, Wu CY, Chen MY, Liu SX, Yan SM, Kang YF, et al. PD-1(+)CXCR5(-)CD4(+) Th-CXCL13 cell subset drives B cells into tertiary lymphoid structures of nasopharyngeal carcinoma. J Immunother Cancer. (2021) 9. doi: 10.1136/jitc-2020-002101

PubMed Abstract | Crossref Full Text | Google Scholar

278. Chen YP, Yin JH, Li WF, Li HJ, Chen DP, Zhang CJ, et al. Single-cell transcriptomics reveals regulators underlying immune cell diversity and immune subtypes associated with prognosis in nasopharyngeal carcinoma. Cell Res. (2020) 30:1024–42. doi: 10.1038/s41422-020-0374-x

PubMed Abstract | Crossref Full Text | Google Scholar

279. Helmink BA, Reddy SM, Gao J, Zhang S, Basar R, Thakur R, et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature. (2020) 577:549–55. doi: 10.1038/s41586-019-1922-8

PubMed Abstract | Crossref Full Text | Google Scholar

280. Tsai CY, Sakakibara S, Yasui T, Minamitani T, Okuzaki D, and Kikutani H. Bystander inhibition of humoral immune responses by Epstein-Barr virus LMP1. Int Immunol. (2018) 30:579–90. doi: 10.1093/intimm/dxy053

PubMed Abstract | Crossref Full Text | Google Scholar

281. Trivedi P, Hu LF, Chen F, Christensson B, Masucci MG, Klein G, et al. Epstein-Barr virus (EBV)-encoded membrane protein LMP1 from a nasopharyngeal carcinoma is non-immunogenic in a murine model system, in contrast to a B cell-derived homologue. Eur J Cancer. (1994) 30A:84–8. doi: 10.1016/S0959-8049(05)80024-3

PubMed Abstract | Crossref Full Text | Google Scholar

282. Hu L, Troyanovsky B, Zhang X, Trivedi P, Ernberg I, and Klein G. Differences in the immunogenicity of latent membrane protein 1 (LMP1) encoded by Epstein-Barr virus genomes derived from LMP1-positive and -negative nasopharyngeal carcinoma. Cancer Res. (2000) 60:5589–93.

PubMed Abstract | Google Scholar

283. Ma BBY, Lim WT, Goh BC, Hui EP, Lo KW, Pettinger A, et al. Antitumor activity of nivolumab in recurrent and metastatic nasopharyngeal carcinoma: an international, multicenter study of the mayo clinic phase 2 consortium (NCI-9742). J Clin Oncol. (2018) 36:1412–8. doi: 10.1200/JCO.2017.77.0388

PubMed Abstract | Crossref Full Text | Google Scholar

284. Lim DW, Kao HF, Suteja L, Li CH, Quah HS, Tan DS, et al. Clinical efficacy and biomarker analysis of dual PD-1/CTLA-4 blockade in recurrent/metastatic EBV-associated nasopharyngeal carcinoma. Nat Commun. (2023) 14:2781. doi: 10.1038/s41467-023-38407-7

PubMed Abstract | Crossref Full Text | Google Scholar

285. Smith C, McGrath M, Neller MA, Matthews KK, Crooks P, Le Texier L, et al. Complete response to PD-1 blockade following EBV-specific T-cell therapy in metastatic nasopharyngeal carcinoma. NPJ Precis Oncol. (2021) 5:24. doi: 10.1038/s41698-021-00162-7

PubMed Abstract | Crossref Full Text | Google Scholar

286. Zhu X, Perales-Puchalt A, Wojtak K, Xu Z, Yun K, Bhojnagarwala PS, et al. DNA immunotherapy targeting BARF1 induces potent anti-tumor responses against Epstein-Barr-virus-associated carcinomas. Mol Ther Oncolytics. (2022) 24:218–29. doi: 10.1016/j.omto.2021.12.017

PubMed Abstract | Crossref Full Text | Google Scholar

287. Mimura K, Kua LF, Xiao JF, Asuncion BR, Nakayama Y, Syn N, et al. Combined inhibition of PD-1/PD-L1, Lag-3, and Tim-3 axes augments antitumor immunity in gastric cancer-T cell coculture models. Gastric Cancer. (2021) 24:611–23. doi: 10.1007/s10120-020-01151-8

PubMed Abstract | Crossref Full Text | Google Scholar

288. Azzi T, Lunemann A, Murer A, Ueda S, Beziat V, Malmberg KJ, et al. Role for early-differentiated natural killer cells in infectious mononucleosis. Blood. (2014) 124:2533–43. doi: 10.1182/blood-2014-01-553024

PubMed Abstract | Crossref Full Text | Google Scholar

289. Desimio MG, Covino DA, Cancrini C, and Doria M. Entry into the lytic cycle exposes EBV-infected cells to NK cell killing via upregulation of the MICB ligand for NKG2D and activation of the CD56(bright) and NKG2A(+)KIR(+)CD56(dim) subsets. Front Immunol. (2024) 15:1467304. doi: 10.3389/fimmu.2024.1467304

PubMed Abstract | Crossref Full Text | Google Scholar

290. Lopez-Montanes M, Alari-Pahissa E, Sintes J, Martinez-Rodriguez JE, Muntasell A, and Lopez-Botet M. Antibody-dependent NK cell activation differentially targets EBV-infected cells in lytic cycle and bystander B lymphocytes bound to viral antigen-containing particles. J Immunol. (2017) 199:656–65. doi: 10.4049/jimmunol.1601574

PubMed Abstract | Crossref Full Text | Google Scholar

291. Abdolahi S, Ghazvinian Z, Muhammadnejad S, Ahmadvand M, Aghdaei HA, Ebrahimi-Barough S, et al. Adaptive NK cell therapy modulated by anti-PD-1 antibody in gastric cancer model. Front Pharmacol. (2021) 12:733075. doi: 10.3389/fphar.2021.733075

PubMed Abstract | Crossref Full Text | Google Scholar

292. Cao B, Liu M, Huang J, Zhou J, Li J, Lian H, et al. Development of mesothelin-specific CAR NK-92 cells for the treatment of gastric cancer. Int J Biol Sci. (2021) 17:3850–61. doi: 10.7150/ijbs.64630

PubMed Abstract | Crossref Full Text | Google Scholar

293. Tong L, Jimenez-Cortegana C, Tay AHM, Wickstrom S, Galluzzi L, and Lundqvist A. NK cells and solid tumors: therapeutic potential and persisting obstacles. Mol Cancer. (2022) 21:206. doi: 10.1186/s12943-022-01672-z

PubMed Abstract | Crossref Full Text | Google Scholar

294. Neo SY, Xu S, Chong J, Lam KP, and Wu J. Harnessing novel strategies and cell types to overcome immune tolerance during adoptive cell therapy in cancer. J Immunother Cancer. (2023) 11. doi: 10.1136/jitc-2022-006434

PubMed Abstract | Crossref Full Text | Google Scholar

295. Williams LR, Quinn LL, Rowe M, and Zuo J. Induction of the lytic cycle sensitizes epstein-barr virus-infected B cells to NK cell killing that is counteracted by virus-mediated NK cell evasion mechanisms in the late lytic cycle. J Virol. (2016) 90:947–58. doi: 10.1128/JVI.01932-15

PubMed Abstract | Crossref Full Text | Google Scholar

296. Fitzsimmons L, Cartlidge R, Chang C, Sejic N, Galbraith LCA, Suraweera CD, et al. EBV BCL-2 homologue BHRF1 drives chemoresistance and lymphomagenesis by inhibiting multiple cellular pro-apoptotic proteins. Cell Death Differ. (2020) 27:1554–68. doi: 10.1038/s41418-019-0435-1

PubMed Abstract | Crossref Full Text | Google Scholar

297. Wong TS, Chen S, Zhang MJ, Chan JY, and Gao W. Epstein-Barr virus-encoded microRNA BART7 downregulates major histocompatibility complex class I chain-related peptide A and reduces the cytotoxicity of natural killer cells to nasopharyngeal carcinoma. Oncol Lett. (2018) 16:2887–92. doi: 10.3892/ol.2018.9041

PubMed Abstract | Crossref Full Text | Google Scholar

298. Duan X, Chen H, Zhou X, Liu P, Zhang X, Zhu Q, et al. EBV infection in epithelial Malignancies induces resistance to antitumor natural killer cells via F3-mediated platelet aggregation. Cancer Res. (2022) 82:1070–83. doi: 10.1158/0008-5472.CAN-21-2292

PubMed Abstract | Crossref Full Text | Google Scholar

299. Salnikov M, Prusinkiewicz MA, Lin S, Ghasemi F, Cecchini MJ, and Mymryk JS. Tumor-infiltrating T cells in EBV-associated gastric carcinomas exhibit high levels of multiple markers of activation, effector gene expression, and exhaustion. Viruses. (2023) 15. doi: 10.3390/v15010176

PubMed Abstract | Crossref Full Text | Google Scholar

300. Tong L, Kremer V, Neo SY, Liu Y, Chen Y, Wagner AK, et al. Renal cell carcinoma escapes NK cell-mediated immune surveillance through the downregulation of DNAM-1. Cancer Commun (Lond). (2023) 43:855–9. doi: 10.1002/cac2.12446

PubMed Abstract | Crossref Full Text | Google Scholar

301. Cho BC, Abreu DR, Hussein M, Cobo M, Patel AJ, Secen N, et al. Tiragolumab plus atezolizumab versus placebo plus atezolizumab as a first-line treatment for PD-L1-selected non-small-cell lung cancer (CITYSCAPE): primary and follow-up analyses of a randomised, double-blind, phase 2 study. Lancet Oncol. (2022) 23:781–92. doi: 10.1016/S1470-2045(22)00226-1

PubMed Abstract | Crossref Full Text | Google Scholar

302. Guan X, Hu R, Choi Y, Srivats S, Nabet BY, Silva J, et al. Anti-TIGIT antibody improves PD-L1 blockade through myeloid and T(reg) cells. Nature. (2024) 627:646–55. doi: 10.1038/s41586-024-07121-9

PubMed Abstract | Crossref Full Text | Google Scholar

303. Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. (2015) 12:453–7. doi: 10.1038/nmeth.3337

PubMed Abstract | Crossref Full Text | Google Scholar

304. Gong L, Kwong DL, Dai W, Wu P, Li S, Yan Q, et al. Comprehensive single-cell sequencing reveals the stromal dynamics and tumor-specific characteristics in the microenvironment of nasopharyngeal carcinoma. Nat Commun. (2021) 12:1540. doi: 10.1038/s41467-021-21795-z

PubMed Abstract | Crossref Full Text | Google Scholar

305. Efremova M, Vento-Tormo M, Teichmann SA, and Vento-Tormo R. CellPhoneDB: inferring cell-cell communication from combined expression of multi-subunit ligand-receptor complexes. Nat Protoc. (2020) 15:1484–506. doi: 10.1038/s41596-020-0292-x

PubMed Abstract | Crossref Full Text | Google Scholar

306. Liu Y, He S, Wang XL, Peng W, Chen QY, Chi DM, et al. Tumour heterogeneity and intercellular networks of nasopharyngeal carcinoma at single cell resolution. Nat Commun. (2021) 12:741. doi: 10.1038/s41467-021-21043-4

PubMed Abstract | Crossref Full Text | Google Scholar

307. Chong JM, Sakuma K, Sudo M, Osawa T, Ohara E, Uozaki H, et al. Interleukin-1beta expression in human gastric carcinoma with Epstein-Barr virus infection. J Virol. (2002) 76:6825–31. doi: 10.1128/JVI.76.13.6825-6831.2002

PubMed Abstract | Crossref Full Text | Google Scholar

308. Park YP, Jin L, Bennett KB, Wang D, Fredenburg KM, Tseng JE, et al. CD70 as a target for chimeric antigen receptor T cells in head and neck squamous cell carcinoma. Oral Oncol. (2018) 78:145–50. doi: 10.1016/j.oraloncology.2018.01.024

PubMed Abstract | Crossref Full Text | Google Scholar

309. Prelaj A, Miskovic V, Zanitti M, Trovo F, Genova C, Viscardi G, et al. Artificial intelligence for predictive biomarker discovery in immuno-oncology: a systematic review. Ann Oncol. (2024) 35:29–65. doi: 10.1016/j.annonc.2023.10.125

PubMed Abstract | Crossref Full Text | Google Scholar

310. Sorin M, Rezanejad M, Karimi E, Fiset B, Desharnais L, Perus LJM, et al. Single-cell spatial landscapes of the lung tumour immune microenvironment. Nature. (2023) 614:548–54. doi: 10.1038/s41586-022-05672-3

PubMed Abstract | Crossref Full Text | Google Scholar

311. Bulik-Sullivan B, Busby J, Palmer CD, Davis MJ, Murphy T, Clark A, et al. Deep learning using tumor HLA peptide mass spectrometry datasets improves neoantigen identification. Nat Biotechnol. (2018) 37:55–63. doi: 10.1038/nbt.4313

PubMed Abstract | Crossref Full Text | Google Scholar

312. Chen B, Khodadoust MS, Olsson N, Wagar LE, Fast E, Liu CL, et al. Predicting HLA class II antigen presentation through integrated deep learning. Nat Biotechnol. (2019) 37:1332–43. doi: 10.1038/s41587-019-0280-2

PubMed Abstract | Crossref Full Text | Google Scholar

313. Yan Y, Sun D, Hu J, Chen Y, Sun L, Yu H, et al. Multi-omic profiling highlights factors associated with resistance to immuno-chemotherapy in non-small-cell lung cancer. Nat Genet. (2025) 57:126–39. doi: 10.1038/s41588-024-01998-y

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: viral-driven cancer, immunotherapy, immunogenicity, immune check inhibitor (ICI), tumor microenvironment (TME)

Citation: Lee K, Kim S, Zhao J and Neo SY (2025) Targeting archetypes of viral-driven cancers with immunotherapy: a perspective on immunogenicity within the tumor microenvironment. Front. Immunol. 16:1631258. doi: 10.3389/fimmu.2025.1631258

Received: 19 May 2025; Accepted: 18 August 2025;
Published: 29 August 2025.

Edited by:

Stefano Cavalieri, Fondazione IRCCS Istituto Nazionale dei Tumori, Italy

Reviewed by:

Wei Wang, Michigan State University, United States
Daniel T. Mytych, Amgen, United States
Zhaokai Zhou, Central South University, China

Copyright © 2025 Lee, Kim, Zhao and Neo. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Shi Yong Neo, bmVvX3NoaV95b25nQGEtc3Rhci5lZHUuc2c=

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.