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

REVIEW article

Front. Immunol., 08 July 2025

Sec. Cancer Immunity and Immunotherapy

Volume 16 - 2025 | https://doi.org/10.3389/fimmu.2025.1610858

This article is part of the Research TopicBreakthroughs in Immune Checkpoint Therapy: Overcoming Resistance with Novel TechniquesView all 6 articles

Synergy of oncolytic adenovirus and immune checkpoint inhibitors: transforming cancer immunotherapy paradigms

  • Department of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China

Oncolytic viruses (OVs) offer a promising antitumor strategy by selectively lysing tumor cells and simultaneously activating innate and adaptive immune responses. Recent studies have shed light on the immunostimulatory mechanisms of OVs, particularly oncolytic adenovirus (OAds), which are emerging as leading candidates due to their favorable safety profile, genomic stability, and efficient transduction capacity. Despite the significant progress made by immune checkpoint inhibitors (ICIs) in antitumor therapy, treatment resistance continues to be a major barrier to their clinical effectiveness. OVs and ICIs work synergistically: OVs reprogram the immunosuppressive tumor microenvironment (TME) through immune cell recruitment and pro-inflammatory cytokine production, potentially overcoming ICI resistance. In turn, ICIs enhance T cell function by blocking inhibitory signaling pathways. This review highlights recent preclinical and clinical advancements in the therapeutic potential of combining OAds with ICIs, while also addressing critical translational challenges. We propose a strategic framework for optimizing the development and clinical trial design of these combination therapies to advance precision immunotherapy.

1 Introduction

Over the past decade, Immune checkpoint inhibitors (ICIs) have significantly improved the treatment of melanoma and non-small cell lung cancer (NSCLC). Despite their success, challenges such as primary and acquired resistance continue to persist in clinical practice (14). The primary resistance mechanisms are largely attributed to the development of an immunosuppressive tumor microenvironment (TME), which is characterized by limited T cell infiltration in “cold” tumors, compensatory activation of immune checkpoints, and metabolic reprogramming of tumor cells (5). To overcome these challenges, research efforts have increasingly centered on the combination of ICIs with other immune-modulatory therapies.

Genetically engineered oncolytic viruses (OVs) have emerged as promising candidates for cancer therapy due to their dual mechanism of action. These viruses selectively target tumor cells, induce immunogenic cell death (ICD), release tumor-associated antigens (TAAs), and activate systemic antitumor immune responses (6). Oncolytic adenovirus (OAds), in particular, have demonstrated significant clinical potential. The recombinant adenovirus H101 was approved in China in 2005 for the treatment of nasopharyngeal carcinoma (7). Second-generation OAds, such as DNX-2401 (Delta-24-RGD), ONCOS-102 (Ad5/3-D24-GM-CSF), and Enadenotucirev (formerly Colad1), have shown promising therapeutic effects in clinical trials (810). Despite these advances, the clinical application of OVs faces several challenges, including limitations in the route of administration and the antiviral immune response, which can diminish their efficacy. Intratumoral (IT) administration remain the primary method in clinical trials, while intravenous (IV) administration, which is more relevant to clinical practice, is hindered by neutralizing factors in the blood (11). Most studies have concluded that the antiviral response negatively impacts the antitumor response; however, there are also opposing viewpoints that challenge this notion. Therefore, finding a way to balance both responses is an important aspect that requires further consideration. These challenges have also spurred the development of various OV modifications and novel delivery systems to improve clinical translation.

A growing body of preclinical and clinical evidence indicates that combination therapies outperform monotherapies in terms of tumor control and survival rates (1215). This advantage stems from the complementary mechanisms of OVs, which remodel the immunosuppressive TME, and ICIs, which alleviate T cell exhaustion and amplify the antitumor immune response. In the treatment with ICIs, the polymorphism of Fcγ receptors can modify the antibody-mediated immune response, thereby influencing the extent of immune system activation (16). It may also result in the immune system attacking normal tissues, especially in the presence of Fcγ receptor variations, which can exacerbate the occurrence of immune-related adverse events (irAEs) (17). OVs have the ability to deliver immunomodulators directly into the TME. Xie et al. engineered OAds to carry transgenes encoding the extracellular domains of SIRPα or Siglec10 on the Fc scaffold to specifically target macrophages, or to encode the extracellular domain of TIGIT to target T cells (18). This method precisely activates specific immune cells. Moreover, OVs can also deliver an anti-PD-1 antibody or a CTLA4-specific single-chain variable fragment (ScFv), thereby further boosting antitumor immune responses with minimal systemic toxicity (19, 20). Currently, two primary strategies for combining OVs and ICIs are being explored: one approach administers OVs and ICIs as separate agents, while the other involves encoding immunomodulatory molecules into the OV genome for local expression. A systematic comparison of these strategies and an in-depth discussion are provided in the study by Wan et al. (21).

This review aims to provide a comprehensive evaluation of the molecular mechanisms, clinical advancements, and challenges associated with OAd-ICI combination therapies. Our focus will be on three primary areas: the immunological basis of their synergistic effects, the key findings from current clinical trials, and the factors that limit the efficacy of these therapies, along with potential solutions. We intend for this review to establish a solid foundation and offer valuable insights to guide future research and optimize clinical trial designs.

2 OAds

OVs target tumor cells through receptor-mediated entry. Once inside the cells, the viral genome is transported to the nucleus, where it initiates replication and transcription. As the viral progeny accumulates, they trigger cell lysis and death, a process known as direct oncolysis. This cytolytic action leads to two main outcomes: the release of progeny viruses, which propagate the infection to neighboring tumor cells and the release of TAAs, which enhance antigen presentation by dendritic cells (DCs). This subsequently activates tumor-specific T cell responses, thereby establishing systemic antitumor immunity. Genetically modified OVs, such as those incorporating cytokine genes or modulating immune checkpoints, can further augment these therapeutic effects.

2.1 Direct oncolysis

OAds are double-stranded DNA viruses that replicate within the nucleus of host cells. Tumor-specific regulatory elements, such as modified E1A promoters, are employed to restrict viral replication to tumor cells, thereby enhancing the safety of high dose administrations (2224). The mechanism of viral entry varies among different OAd serotypes. Most OAds utilize the coxsackievirus-adenovirus receptor (CAR), while certain subgroups, such as subgroup B and specific D variants, employ CD46 for cell attachment (25). After internalization through receptor-mediated endocytosis, uncoating proteins disrupt the endosomal membrane, enabling the virus to escape into the cytoplasm. The viral genomes are then transported along microtubules to the nuclear pore complex, where they undergo nuclear translocation (26).

The E1A and E1B genes are essential for initiating viral replication. Specifically, the conserved region 2 (CR2) of E1A interacts with the retinoblastoma (Rb) protein, leading to the release of the E2F transcription factor, which drives the progression of the cell cycle into the S phase—a key event for viral replication in tumor cells (27, 28). Additionally, the E1B-encoded proteins, including the 19 kDa and 55 kDa isoforms, inhibit apoptosis and facilitate viral DNA replication (29, 30). These molecular interactions enhance the tumor specificity of the virus while minimizing off-target toxicity in preclinical models.

ONYX-015, the first tumor-selective adenovirus to undergo clinical evaluation, carries a deletion in the E1B-55K gene, which limits replication to p53-deficient tumor cells while sparing normal cells with intact p53 (31). H101, a derivative of ONYX-015 that also contains a deletion in the E1B/E3 region, was approved by the Chinese FDA as the first commercially available OV (32, 33). DNX-2401 employs an RGD peptide-modified fibronectin to facilitate viral entry through αvβ3/αvβ5 integrins, effectively bypassing the need for CAR receptors in tumor cells (34, 35). Enadenotucirev utilizes a chimeric Ad11p/Ad3 backbone to overcome pre-existing humoral immunity, featuring a unique serotype profile that enhances tumor-selective lysis and fosters immune cell infiltration, establishing it as a highly adaptable therapeutic platform (3638). A summary of other genetically modified adenoviruses is provided in Table 1.

Table 1
www.frontiersin.org

Table 1. Genetic modifications in OAds.

2.2 Activation of antitumor immune response

The therapeutic efficacy of OVs extends beyond their ability to replicate, with a robust antitumor immune response being integral to their success. Remarkably, even replication-incompetent or inactivated OVs can trigger immunogenic responses. Similar to radiotherapy and chemotherapy, OVs can induce ICD, a form of regulated cell death (RCD) that activates adaptive immunity (39). Different OVs activate distinct ICD pathways. For instance, adenoviruses primarily induce ICD through autophagy, necrosis, and pyroptosis, processes driven by the release of damage-associated molecular patterns (DAMPs), pathogen-associated molecular patterns (PAMPs), and TAAs (40, 41). DAMPs consist of intracellular signaling molecules such as calreticulin (CRT), heat shock proteins (HSPs), and high-mobility group box 1 (HMGB1), while PAMPs comprise viral nucleic acids, capsid components, and structural proteins (4244). These immunostimulatory signals recruit dendritic cells, activate natural killer (NK) cells, and promote the infiltration of antigen-presenting cell (APC) into the TME. As a result, cytokines and chemokines are secreted, triggering inflammatory responses. DCs subsequently prime CD4+ and CD8+ T cells via MHC II/I antigen presentation, which in turn activate adaptive immune response (45).

In order to improve efficacy, engineered OVs are strategically designed to express a range of immune-modulatory factors that effectively counteract the immunosuppressive TME. ONCOS-102, a chimeric OAd encoding granulocyte-macrophage colony-stimulating factor (GM-CSF), enhances the maturation of APCs while sustaining intratumoral viral replication and promoting cellular immunity (9). Likewise, CG0070 (cretostimogene grenadenorepvec), CGTG-102, and CGTG-602 leverage GM-CSF expression to potentiate their antitumor activity (4649). TILT-123 simultaneously expresses tumor necrosis factor-alpha (TNF-α) and interleukin-2 (IL-2), driving T cell activation while reprogramming the immunosuppressive microenvironment (50). Ad5-PC is a novel platform engineered to express a bispecific PD-1/CD137L fusion protein, promoting sustained cytotoxic T lymphocytes (CTLs) persistence and heightened immune activation, ultimately resulting in durable tumor control in hepatocellular carcinoma (HCC) (51). Multiple other immunomodulatory factors, including IL-12, CXCL10, and CD40L, have been successfully integrated into the oncolytic viral vector platform (Table 2).

Table 2
www.frontiersin.org

Table 2. Encoding of immunostimulatory transgenes by OAds.

2.3 Progress in clinical studies

Clinical trials investigating the use of OVs for the treatment of head and neck tumors have demonstrated significant therapeutic potential. In a Phase I trial of DNX-2401 monotherapy for patients with recurrent glioblastoma, 20% of participants survived for over three years. Notably, three patients experienced up to a 95% reduction in tumor volume, with histopathological analysis confirming substantial immune cell infiltration (52). A subsequent Phase I dose-escalation study in patients with diffuse intrinsic pontine glioma (DIPG) reported a median overall survival (mOS) of 17.8 months, with three patients surviving beyond 24 months. Remarkably, one patient with wild-type H3 and IDH1 mutations achieved sustained remission (53). Ongoing clinical trials evaluating DNX-2401 include NCT01956734 and NCT03896568, continue to explore its therapeutic potential.

In advanced HCC, monotherapy with OBP-301 has been shown to enhance CD8+ T cell infiltration, although tumor regression remains limited, indicating its potential for synergistic use with ICIs (54). A Phase I trial (NCT03172819) is currently assessing the efficacy of OBP-301 in combination with pembrolizumab for the treatment of advanced solid tumors. Similarly, ONCOS-102 has been demonstrated to elicit both localized and systemic CD8+ T cell responses, while also upregulating PD-L1 expression in a Phase I trial (9).

OAds have demonstrated significant potential not only in solid tumors but also in other malignancies. A retrospective study highlighted the efficacy of H101 in treating malignant ascites, reporting an ascites response rate (ARR) of 40% and an ascites control rate (ACR) of 75% (55). Phase II clinical trials have confirmed the safety of intraperitoneal H101 injection and revealed enhanced immune checkpoint interactions between CD8+ T cells and myeloid cells in long-term responders, as evidenced by CellPhoneDB analysis (56). The therapeutic effect may be attributed to the unique characteristics of the ascites microenvironment, which lacks a physical barrier, thereby facilitating viral spread. In addition, other OAds, such as enadenotucirev, AdAPT-001, and YSCH-01, have shown clinical promise in the treatment of malignant tumors (Table 3).

Table 3
www.frontiersin.org

Table 3. The clinical trials exploring the efficacy of OAds monotherapy.

Despite their potential, the clinical application of OVs in treating “cold tumors”, which are characterized by limited immune infiltration, faces several challenges. These include uncertainties surrounding long-term efficacy and the impact of tumor heterogeneity on the success of monotherapy treatments.

2.4 Safety

Wild-type adenoviruses often cause off-target infections in normal tissues due to their lack of tumor specificity. In contrast, engineered OAds exhibit improved tumor selectivity through various genetic modifications. For example, ONYX-015 enables tumor-specific replication in p53-deficient cells through a deletion of the E1B55K gene, while H101 enhances selectivity by introducing double deletions of the E3 and E1B55K genes. The ZD55 platform combines the E1B55K deletion with a transgene insertion site, creating a bifunctional design (32, 57, 58). Moreover, OVs such as CG7870, CG7060, OBP-301/401, GD55, and CRAd-S.pk7 restrict viral replication to tumor cells by utilizing tissue-specific promoters, including prostate-specific antigen (PSA) and human telomerase (5965).

In addition to optimizing targeting, viral tropism plays a crucial role in OAd therapy. While natural adenoviruses primarily infect CAR receptor-positive cells through fibronectin binding, genetically engineered OAds are capable of precisely targeting CAR-negative tumor cells, thereby reducing the risk of infecting normal tissues. Specifically, serotype 5 human adenovirus exhibits a strong hepatic tropism when administered intravenously, as it is captured by Kupffer cells and primarily localizes to hepatocytes. This pronounced hepatic targeting poses a significant challenge for clinical application (66, 67). Surface charge modifications, such as the HAdV-5-HexPos3_ΔCAR variant, have shown substantial potential in reducing non-target organ tropism (67).

Genetically modified OVs typically exhibit a favorable safety profile, with a low incidence of severe adverse events or the need for therapy discontinuation. Common side effects include transient systemic symptoms such as fever and fatigue, as well as localized inflammation at the injection site. However, a therapeutic paradox remains: achieving optimal intratumoral viral titers while maintaining systemic safety is a significant challenge due to the dose-dependent nature of the clinical response.

2.5 Delivery methods and immunological clearance

OVs are predominantly delivered through two routes: IT injection and IV injection. Alternative methods, such as intraperitoneal (IP) injection, subcutaneous (SC) injection, and intravesical (IVE) injection, are less commonly used (6870). In preclinical studies, the choice of delivery route is typically determined by the specific experimental objectives, whereas in clinical practice, patient safety is the foremost consideration. IT injection is the standard method for treating accessible tumors, though it has limitations when targeting deep or metastatic lesions (71). Additionally, IT injection carries risks, including bleeding, infection, and tumor seeding, which require specialized clinical expertise to manage effectively.

IV administration offers systemic delivery, thereby overcoming the limitations of IT injection and enhancing clinical feasibility. Nevertheless, pre-existing antiviral antibodies and serum protein binding can significantly reduce the bioavailability of the virus (72). Hepatic sequestration by Kupffer cells poses another significant challenge, as these macrophages rapidly clear circulating adenoviral particles, thereby limiting the effective delivery of therapeutic payloads (73, 74). To address these challenges, researchers have developed several strategies, including capsid modifications, liposomal formulations, cell-based delivery systems (e.g., neural stem cells, mesenchymal stem cells, T cells, tumor-infiltrating lymphocytes [TILs], NK cells, DCs, and human dental pulp stem cells), extracellular vesicles, gelatin hydrogels, and nanomaterials (65, 7585).

The immunogenicity of OVs plays a critical role in determining the appropriate route of administration. Highly immunogenic variants are better suited for localized IT delivery, while strains with lower immunogenicity may be more effective for prolonged systemic circulation through IV administration (86). Additionally, it is essential to consider the interaction between delivery platforms and the tumor immune microenvironment to mitigate potential immunosuppressive effects. Developing advanced delivery systems that enhance viral bioavailability while ensuring a strong safety profile is critical for achieving therapeutic concentrations at the tumor site.

3 Other OVs under investigation

3.1 Herpes simplex virus

Herpes Simplex Virus (HSV) is an enveloped double-stranded DNA virus that possesses both a nucleocapsid and an outer membrane (87). It exists in two serotypes: HSV-1 and HSV-2. HSV-1 holds considerable antitumor potential due to its large genome, which can accommodate multiple exogenous genes, as well as its ability to evade immune responses (88). Engineered HSV-1-based oncolytic viruses, such as Talimogene laherparepvec (T-VEC) and G47Δ, have been approved for the treatment of malignant melanoma and gliomas, respectively (89, 90).

3.2 Vaccinia virus

Vaccinia virus (VV) is a double-stranded DNA virus of approximately 190kb in size, belonging to the poxvirus family (91). Known for its excellent safety profile and rapid replication cycle, VV has emerged as a leading platform for oncolytic virotherapy (92, 93). Genetically modified strains, such as JX-594 and Pexa-Vec, are currently undergoing clinical trials. Notably, JX-594 has shown promising results for IV administration and demonstrated resistance to neutralization by antibodies and complement (94, 95).

3.3 Reovirus

Reovirus (RV) is a non-enveloped double-stranded RNA virus that primarily causes mild upper respiratory or gastrointestinal infections (96, 97). Pelareorep, a type 3 reovirus, has shown suitability for IV administration and boasts an excellent safety profile. It has demonstrated antitumor efficacy in several clinical trials, positioning it as one of the most advanced oncolytic RNA virus therapeutics (98).

3.4 Measles virus

Measles virus (MeV) is an enveloped, single-stranded negative-sense RNA virus that naturally targets tumors (99). Its excellent safety profile and the absence of dose-limiting toxicity make it a promising candidate for oncolytic virotherapy (100).

3.5 Newcastle disease virus

Newcastle disease virus (NDV) is an enveloped, single-stranded negative-sense RNA virus with a substantial capacity for exogenous gene insertion. Its P/M gene intergenic region serves as an ideal site for genetic modification (101, 102). Early clinical trials using the wild-type NDV strain have demonstrated good patient tolerance (103).

Other viruses, including coxsackievirus, seneca valley virus, poliovirus, and vesicular stomatitis virus (VSV), have also shown promise in antitumor therapy (104107). Each of these viruses possesses unique biological properties, providing diverse therapeutic options for tumor treatment and broadening the potential applications of oncolytic virotherapy.

4 Immune checkpoint inhibitors

4.1 Mechanisms

4.1.1 PD-1/PD-L1 and CTLA-4/B7 pathways

ICIs play a pivotal role in enabling tumors to escape immune surveillance. By blocking these inhibitory pathways, ICIs enhance T cell responses targeting tumor cells. Cytotoxic T lymphocyte-associated protein 4 (CTLA-4), a homolog of the co-stimulatory receptor CD28, negatively regulates T cell activation. While CD28 promotes T cell activation by binding B7 molecules (CD80/CD86) on DCs, CTLA-4 competes with CD28 for binding to B7 with a higher affinity, thereby inhibiting T cell activation (108, 109). Furthermore, inhibition of CTLA-4 disrupts the function of Tregs, which inherently express high levels of CTLA-4 and are essential in mediating immune suppression within the TME (110, 111).

PD-1, a key member of the CD28 superfamily, interacts with two ligands: PD-L1 (B7-H1) and PD-L2 (B7-DC) (112, 113). Upon binding, PD-1 triggers inhibitory signaling by recruiting SHP-2, a phosphatase that dephosphorylates critical signaling molecules in the T cell receptor (TCR) pathway, including ZAP-70 and CD3ζ (114116). This cascade of signaling events impairs T cell activation, proliferation, and cytokine production, thereby diminishing the antitumor immune response. As second-generation ICIs, PD-1/PD-L1 inhibitor have shown clinical efficacy and are approved for various cancers, including melanoma, NSCLC, and renal cell carcinoma (RCC). However, despite enhancing T cell activation, targeting CTLA-4 or PD-1 alone is insufficient to fully control tumor progression.

4.1.2 Novel immune checkpoint inhibitors

To enhance therapeutic outcomes, research has increasingly focused on identifying additional immune checkpoint targets. Novel co-inhibitory receptors currently under investigation include lymphocyte activation gene-3 (LAG-3), T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT), T cell immunoglobulin and mucin-domain containing-3 (TIM-3), and signal regulatory protein alpha (SIRPα). Concurrently, co-stimulatory receptors such as inducible T cell costimulator (ICOS), members of the TNF receptor superfamily (e.g., OX40 and 4-1BB), and Toll-like receptors (TLRs) have emerged as promising candidates for therapeutic targeting.

LAG-3, also known as CD223, is a transmembrane glycoprotein structurally similar to CD4 (117). The first FDA-approved LAG-3 inhibitor, Opdualag, combines the anti-LAG-3 antibody relatlimab-rmbw with the anti-PD-1 antibody nivolumab. Approved in 2022, Opdualag is used for the treatment advanced melanoma treatment (118).

TIGIT, predominantly expressed on T cells, NK cells, Tregs, and tumor-infiltrating lymphocytes (TILs), binds to the ligands CD155 and CD112 (119, 120). By competing with CD226 for binding to CD155, TIGIT suppresses NK cell-mediated cytotoxicity. Blocking both TIGIT and PD-1 can restore CD226 signaling and enhance CD8+ T cell responses (121123). Clinical trials with agents such as tiragolumab, vibostolimab, and tislelizumab have demonstrated promising results.

TIM-3 is expressed on exhausted T cells and interacts with several ligands, including phosphatidylserine and galectin-9, to promote immune tolerance (124). The binding of TIM-3 to MHC II directly suppresses T cell proliferation. While preclinical studies have demonstrated therapeutic potential, a bispecific antibody targeting TIM-3 and PD-L1, known as LY3415244, exhibited unexpected immunogenicity, leading to the discontinuation of clinical trials (124126).

SIRPα binds to CD47, sending a “don’t eat me” signal to macrophages, which inhibits phagocytosis (127, 128). Blocking the CD47-SIRPα axis can promote tumor clearance by macrophages.

4.1.3 Novel immune checkpoint stimulators

ICOS, a member of the CD28 family, is expressed on activated T cells and enhances T cell responses by binding to ICOS-L and activating the PI3K signaling pathway (129). Preclinical studies have demonstrated that blockade of CTLA-4 upregulates ICOS on CD4+ T cells, prompting the ongoing Phase II clinical trial of the ICOS agonist vopratelimab in combination with ipilimumab for PD-1/PD-L1-resistant NSCLC (NCT03989362).

The 4-1BB receptor (CD137), which is expressed on activated T and NK cells, has emerged as a promising target for treating solid malignancies (130, 131). However, systemic administration of 4-1BB agonists has been associated with dose-limiting hepatotoxicity (132, 133). TLRs within TME promote tumor progression by activating the NF-κB pathway and inducing immunosuppressive cytokines. In contrast, TLR7/8 agonists have been shown to enhance antitumor immunity when used in combination with immune checkpoint blockade (ICB) (134, 135).

ICB has become a key therapeutic strategy for advanced malignancies, with current research focusing on three main objectives: improving clinical outcomes by enhancing efficacy and reducing irAEs, developing predictive biomarkers for stratify treatment responses, and advancing combinatorial therapeutic approaches. Despite these advancements, some translational barriers remain, particularly in maintaining immunological equilibrium and addressing intratumoral heterogeneity.

4.2 Resistance to treatment with ICIs

The introduction of ICIs has significantly transformed the treatment landscape for various cancer types; however, ongoing challenges such as treatment resistance and the optimization of therapeutic protocols remain critical areas for research. Resistance to ICIs can be categorized into two main types: primary resistance, which is characterized by the absence of an initial therapeutic response due to inherent tumor characteristic, and acquired resistance, which occurs after an initial clinical response followed by disease progression (4).

Mechanistically, resistance to ICIs arises from both tumor-intrinsic and microenvironmental factors. Intrinsic mechanisms encompass a low burden of neoantigens, dysregulation of oncogenic signaling and metabolic pathways, impaired type I interferon (IFN-I) signaling, defective antigen processing and presentation, and epigenetic modifications (136). Extrinsic mechanisms involve insufficient T cell infiltration, the expansion of immunosuppressive cell populations (such as myeloid-derived suppressor cells [MDSCs] and Tregs), the induction of alternative immune checkpoints (e.g., TIM-3, LAG-3), epithelial-mesenchymal transition (EMT), pro-angiogenic signaling, and dysbiosis of the gut microbiome (137).

Tumors are commonly categorized as immunologically “hot” (inflamed) or “cold” (non-inflamed) based on TME characteristics, including profiles of inflammatory cytokines and the infiltration of CD8+ T cells. Primary resistance is typically observed in “cold” tumors, which exhibit minimal immune cell infiltration, while acquired resistance often develops in initially responsive “hot” tumors through adaptive immune-editing processes (138140). This classification has clinical significance, as “cold” tumors typically demonstrate low PD-L1 expression and poor responses to immunotherapy. A key mechanism of resistance is T cell exhaustion, which results from prolonged antigen exposure. Exhausted T cells are characterized by: 1) upregulation of co-inhibitory receptors (PD-1, CTLA-4, LAG-3), 2) metabolic dysfunction, and 3) reduced secretion of effector cytokines (IFN-γ, TNF-α, IL-2) (141). Although PD-1 blockade can temporarily restore T cell functionality through epigenetic modifications, complete recovery is often not achieved, as terminally exhausted T cell clones persist even after ICI therapy.

IFN-γ illustrates the dual role of immune regulation, as it promotes antitumor immunity by upregulating MHC I upregulation while simultaneously facilitating immune escape through the expression of PD-L1 and the recruitment of regulatory T cells (Tregs) (142, 143). Current therapeutic strategies are increasingly focused on rationally designed combination therapies, with promising synergistic effects observed between ICIs and OAds. OAds enhance the immune response by reconfiguring immunosuppressive networks within the TME.

5 Combination strategy reverses drug resistance

5.1 Combination therapy optimizes antitumor efficacy

Current methodologies for the integration of OVs and ICIs are primarily categorized into two distinct strategies: 1) transgene delivery systems using engineered oncolytic adenoviral vectors (OAds expressing ICIs), and 2) spatiotemporally separated delivery of OAds and ICIs (OAds and ICIs). The first strategy presents several advantages compared to conventional approaches, and certain mechanisms underlying this combination therapy are depicted in Figure 1. we conducted a search of the PubMed database for promising preclinical and clinical studies examining the integration of OVs and ICIs over the past five years, as summarized in Table 4.

Figure 1
Illustration depicting different approaches to using oncolytic adenoviruses (OAds) in cancer therapy. The top section shows OAds monotherapy upregulating PD-L1 expression in tumors, contributing to immune resistance. The bottom section depicts combination therapies with immune checkpoint inhibitors (ICIs), activating the tumor microenvironment and blocking PD-L1, reversing resistance. Various ICIs like aPD-L1 scFv and TIGIT-Fc are shown. Benefits include mitigating immune-related adverse effects, delivering immunomodulators, and overcoming viral neutralization, while maintaining potency.

Figure 1. Synergy of oncolytic adenovirus (OAds) and immune checkpoint inhibitors (ICIs). OVs lyse tumor cells and induce immunogenic cell death (ICD) in tumor cells, releasing damage-associated molecular patterns (DAMPs), pathogen-associated molecular patterns (PAMPs) and soluble tumor-associated antigens (TAAs), which stimulate innate and adaptive immune responses, as well as inducing upregulation of PD-L1 expression in tumor cells, leading to T cell exhaustion. However, PD-L1 expression can be blocked by OAds in combination with ICIs (mainly OAds and ICIs, OAds expressing ICIs), thereby reversing resistance. In addition, OVs expressing ICIs have the following advantages over separate individual dosing: reducing immune-related adverse events (irAEs) associated with systemic ICI therapy; flexible delivery of cytokines, immune checkpoint molecules, and other immunomodulators to further activate the immune microenvironment; diminishing virus-induced neutralization and ensuring effective viral load. Created with BioRender.com.

Table 4
www.frontiersin.org

Table 4. Preclinical and clinical trials combining OAds with ICIs (2019-2024).

5.2 Progress in preclinical and clinical research

5.2.1 OAds and ICIs

Initial validation of the effectiveness of combining the two immunotherapeutic agents has been carried out using various preclinical animal models. OBP-301 is a novel attenuated type 5 adenovirus that utilizes the hTERT promoter to enhance the expression of the adenovirus early in areas linked to an internal ribosome entry site (IRES) sequence. This genetic construct facilitates tumor-specific viral replication and induces lytic cell death in a variety of cancer cell types (54). Its derivative, OBP-502, has been shown to induce ICD in models of CRC and pancreatic ductal adenocarcinoma (PDAC), functioning synergistically with PD-1 blockade to promote CD8+ T cell infiltration and enhance systemic antitumor immunity (144). Additionally, the gemcitabine-resistant variant OBP-702 has been found to counteract GM-CSF-mediated immunosuppression, thereby further improving the efficacy of PD-L1 blockade in PDAC (138). Engineered oncolytic vectors facilitate multimodal immunomodulation through various mechanisms. The combination of ONCOS-102 with pembrolizumab has been shown to significantly reduce tumor burden, while cytokine-armed viral constructs effectively reprogram the immunosuppressive TME (145). Adv-CXCL10, a recombinant adenovirus encoding CXCL10, enhances the effectiveness of PD-1 inhibitors by promoting the expansion of CXCR3+ T cells (146). Furthermore, the bifunctional agent TILT-123, which expresses TNFα and IL-2, alters the immune landscape by the ratios of CD8+ to CD4+ T cells and promoting the maturation of DC across various cancer models, including the induction of tertiary lymphoid structures in head and neck cancers (14, 140, 147151). Similarly, ZD55-IL24 has been shown to mitigate immune exclusion in non-inflamed tumors (152). The use of multi-agent combinations may further enhance therapeutic effectiveness. In a model of triple-negative breast cancer (TNBC), OAds demonstrated a synergistic effect when combined with anti-PD-L1 and anti-CTLA-4 immunotherapies, resulting in improved tumor control and extended survival, with 20% of the subjects exhibiting complete suppression of metastasis (12).

Clinical trials provide additional evidence supporting the potential of integrating the two approaches to facilitate clinical translation. Substantial advancements have been achieved in the research pertaining to cancer of the gastrointestinal tumors. In a Phase I clinical trial, the administration of enadenotucirev as a monotherapy resulted in the infiltration of CD8+ T cells within microsatellite stable (MSS) colorectal cancer (CRC) tumors, indicating the potential for virus-induced immunogenicity (10). A subsequent investigation that combined enadenotucirev with nivolumab demonstrated improvements in OS and T cell activation in MSS CRC patients (38). In patients with refractory HCC, the combination of H101 and nivolumab yielded an objective response rate (ORR) of 11.1% and a mOS of 15.04 months (15). Importantly, certain individuals classified as having stable disease (SD) experienced extended OS with ongoing treatment, highlighting the need for further investigation of combination therapies in larger clinical trials.

In the treatment of head and neck tumors, DNX-2401 has shown efficacy in reversing PD-1-mediated T cell exhaustion, leading to sustained remission in patients with glioma (153). A Phase 1/2 clinical trial indicated a 12-month overall survival rate of 52.7% for IDH1 wild-type gliomas, thereby reinforcing the potential of this therapeutic strategy for treating refractory tumor types (154). In the context of urological tumors, the Phase 2 CORE-001 trial demonstrated that the combination of CG0070 and pembrolizumab in patients with Bacillus Calmette-Guérin (BCG)-unresponsive bladder cancer yielded favorable risk-benefit profiles, with a complete remission (CR) rate of 57.1% at 12 months and 51.4% at 24 months (155). Additionally, ongoing prospective trials are assessing the efficacy of H101 in conjunction with PD-1 inhibitors for patients with advanced malignant pleural mesothelioma or non-muscle invasive bladder cancer who have not responded to previous treatments (NCT06031636, NCT05564897). Furthermore, investigations into triple-drug combinations involving OVs, immunotherapies, and anti-angiogenic agents are currently in progress (NCT05303090).

5.2.2 OAds expressing ICIs

The existence of a functional Fc region can be advantageous yet problematic, as immune checkpoints are extensively distributed throughout the organism, leading to irAEs following the systemic administration of antibodies. To mitigate this issue, Hamdan et al. developed an Fc-fusion peptide aimed at PD-L1, which included a chimeric constant region composed of IgG1 and IgA1 (IgGA) (156). This construct was subsequently incorporated into an OAd (Ad-Cab), thereby facilitating the activation of neutrophil effector functions mediated by both IgG1 and IgA1 (156). Moreover, RCAd-LTH-shPD-L1, a dual-transgene OAd, facilitates the localized administration of anti-VEGF antibodies and PD-L1-targeting short hairpin RNA (shRNA), thereby enhancing the secretion of IFN-γ, interleukin-6 (IL-6), and interleukin-12 (IL-12), while also promoting T cell infiltration (157). This strategy contributes to the normalization of tumor vasculature and the reprogramming of immunosuppressive TME networks. The anti-PD-L1 scFv-expressing OAd, ZD55-apd-L1-scFv, demonstrated superior antitumor efficacy compared to both the parental ZD55 virus and systemic anti-PD-L1 therapies (158). CAV2-AU-M2, an anti-PD-1 single-domain antibody (sdAb)-armed OAd, synergistically integrates multiple immunotherapeutic approaches to address the challenges associated with osteosarcoma treatment (159). Furthermore, the bispecific CD137 agonist/PD-L1 blocker Ad5-PC enhances CTL activity through the simultaneous activation of the CD137 pathway and blockade of the PD-1/PD-L1 interaction (51). Several combination trials utilizing novel checkpoint inhibitors have yielded encouraging results. OAds expressing TIM-3, 4-1BBL, (SIRPα)-IgG1 Fc, ICOSL, CD40L, and OX40L demonstrated antitumor effects across various preclinical tumor models (130, 160163). This emerging strategy capitalizes on the characteristic immune cell infiltration within the TME to create recombinant OAds for targeted therapy. This approach encompasses three distinct types of recombinant OAds: OAd-SIRPα-Fc, OAd-Siglec10-Fc, and OAd-TIGIT-Fc. OAd-SIRPα-Fc and OAd-Siglec10-Fc have been shown to significantly suppress tumor growth in macrophage-rich tumor microenvironments, while OAd-TIGIT-Fc primarily enhances T cell activation (18). The targeted methodology facilitates tumor-selective immunotherapy. Other preclinical and clinical evidence supports the combination of OAd-encoded ICIs with systemic checkpoint blockade (164166).

The localized production of antibodies by OVs enhances the specificity of therapeutic interventions while reducing the adverse effects typically associated with ICIs alone (21). Furthermore, ICIs can regulate the balance of the immune response, thereby reducing the clearance of the virus. In contrast, the independent administration of OVs and ICIs offers greater flexibility but may elevate the potential for toxicity. Therefore, the selection of the most appropriate treatment regimen should be tailored to the particular clinical context.

6 The integration of OAds with other treatment modalities

Beyond combination with ICIs, OAds also have demonstrated synergistic therapeutic effects when integrated with chemotherapy, radiotherapy, adoptive cell therapy (ACT), targeted therapy and other treatment modalities.

Chemotherapeutic agents have the potential to augment the effectiveness of OV therapy by attenuating the host’s antiviral immune response. For instance, the adenovirus Ad5/3-pCDX2, which is regulated by the CDX2 promoter, when administered in conjunction with 5-fluorouracil (5-FU), leads to an upregulation of CDX2 expression in tumors, thereby significantly inhibiting the proliferation of CDX2-negative CRC (167). Clinical evidence has also indicated that the combination of LOAd703 with standard chemotherapy demonstrates favorable safety profiles and preliminary antitumor activity in patients diagnosed with advanced PDAC (168). Similarly, the integration of ONCOS-102 with pemetrexed and platinum-based chemotherapeutics in the treatment of malignant pleural mesothelioma has resulted in an extended mOS of 20.3 months, markedly enhancing patient outcomes compared to chemotherapy alone, while also facilitating T cell infiltration and the expression of pro-inflammatory cytokines (169). Additional combination therapies, such as DNX2401 with temozolomide (NCT01956734) and H101 with FOLFOX (NCT05124002), are currently under investigation in clinical trials.

The integration of OVs with radiotherapy has demonstrated a distinctive synergistic effect. The release of antigens induced by radiation enhances the immune response initiated by OVs, while the viral infection simultaneously impairs the tumor cells’ capacity to repair DNA damage caused by radiation. Research has indicated that adenoviral proteins can directly disrupt the DNA damage response (DDR) pathway, thereby modulating critical processes involved in the recognition and repair of DNA damage (170). Furthermore, the recently developed PEG-coated intravenous RadioOnco formulation has effectively inhibited DNA damage repair mechanisms, resulting in the activation of durable antitumor immune responses and offering substantial benefits in the management of tumor metastasis and recurrence (171).

The synergistic effects of OVs and targeted therapy primarily rely on the precise regulation of critical signaling pathways. The JAK-STAT pathway is particularly significant in determining tumor sensitivity to OAds, as impairments within this pathway markedly increase viral susceptibility (172). JAK inhibitors, such as ruxolitinib, have demonstrated potential in augmenting the effectiveness of OV therapies; however, the outcomes are contingent upon the specific experimental models employed. For instance, in the context of VSV-IFNβ treatment, ruxolitinib was found to enhance viral activity in resistant cell lines, yet it did not yield a substantial improvement in survival rates within immune-competent models of NSCLC (173, 174). In the case of CRC, the combination of OVs with PI3K-γ inhibitors may address ICI resistance associated with the PI3K/AKT/mTOR signaling pathway. Nevertheless, challenges persist due to the potential for cross-resistance arising from compensatory mechanisms within tumor signaling pathways and the remodeling of the TME (175).

ACT, which incorporates the use of effector cells such as CAR-T cells, NK cells, DCs, or TILs, has encountered several obstacles in the treatment of solid tumors. These challenges include inadequate tumor infiltration, the presence of immunosuppressive microenvironments, T cell exhaustion, and limited cell persistence (176). The integration of genetically modified OVs with ACT has shown potential in enhancing the efficacy of CAR-T cells. For instance, CAR-T cells infected with the TS-2021 virus exhibit sustained activity through autocrine interleukin-15 (IL-15), thereby overcoming resistance in glioblastoma therapy (177). OAds that express specific chemokines can facilitate the infiltration of CAR-T cells into tumors and modify the immune microenvironment (178). Furthermore, the combination of OVs with NK cells or DCs has demonstrated notable synergistic antitumor effects (179, 180).

Innovative approaches, including photodynamic immunotherapy and high-dose vitamin C, exhibit potential for future integration with OV (181, 182). While combination therapies present considerable benefits, additional research is essential to refine safety evaluations, dosing protocols, and other relevant factors. Among these, the integration of ICIs with OVs is regarded as one of the most promising clinical strategies, supported by numerous trials that confirm its therapeutic efficacy.

7 Clinical challenges and future directions

While the integration of OVs and ICIs demonstrates promise for antitumor treatment, several challenges remain to be resolved. Key considerations include establishing the optimal timing for administration, achieving a balance between the antiviral and antitumor immune responses, and identifying effective predictive tumor biomarkers. Furthermore, the interplay between OVs and microbiome is currently under investigation, which may offer insights for the advancement of novel therapeutic strategies.

7.1 Appropriate timing for treatment

The integration of OVs and ICIs necessitates meticulous timing, especially in the synchronization of various administration approaches, including alternating, sequential, or concurrent delivery methods. Nguyen et al. established a classification framework that delineates five distinct paradigms of administration: (i) anti-PD-1 priming→OV; (ii)concurrent administration; (iii)OV priming→anti-PD-1;(iv) concurrent therapy priming→anti-PD-1; and (v)OV priming→concurrent therapy (183). Preclinical findings indicate that optimal synergy is attained when OV priming is succeeded by simultaneous dual therapy. From a mechanistic perspective, the initial administration of the OV facilitates the recruitment of TILs and creates an inflamed TME. Nevertheless, the compensatory upregulation of PD-L1 on tumor cells may diminish this therapeutic effect. The occurrence of adaptive resistance underscores the importance of implementing a sequential approach to the administration of OV in conjunction with ICI. The inhibition of PD-1 serves to alleviate T-cell exhaustion while simultaneously augmenting the antitumor immune response elicited by OV, thus preserving the integrity of the cancer-immunity cycle. In the HaP-T1 PDAC models, the “OV priming→concurrent therapy” sequence achieved pathologic complete remission, whereas the sequence of “anti-PD-1→OV” sequencing only enhanced tumor control (69). Tumor-free survivors exhibited durable immune memory, indicating the possibility of neoadjuvant applications through preoperative intratumoral delivery of OV.

The clinical development of T-VEC, a HSV engineered for tumor-selective replication, exemplifies both the potential and the challenges associated with combination immunotherapy. A Phase II clinical trial demonstrated that the combination of T-VEC with ipilimumab resulted in a higher ORR compared to ipilimumab administered alone (odds ratio, 2.9, P =0.002), while maintaining a similar safety profile (184). Conversely, a Phase III clinical trial did not reveal a significant advantage in PFS or OS when T-VEC was combined with pembrolizumab, as opposed to pembrolizumab monotherapy (185). In both studies, OV was initially employed to elicit an early anti-cancer immune response, followed by the introduction of ICIs to enhance the immune effect synergistically. However, the differing outcomes may be attributed to insufficient time in the Phase III study to fully activate the TME. Given that factors such as tumor biology, treatment design, and pharmacological parameters influence therapeutic responses, the timing of intervention is crucial for achieving efficacy and must be meticulously calibrated across various cancer types. The intricate relationship between T cell activation and the heterogeneous TME presents potential risks, including the potential for premature or excessive immunostimulation, which can lead to irAEs and accelerated T cell exhaustion.

7.2 The balance between the antitumor response and antiviral response

Enhancing the efficacy of OV therapy necessitates a careful equilibrium between the antitumor response and the host’s antiviral immune response. This issue is notably exemplified by wild-type adenoviruses, which face obstacles posed by pre-existing neutralizing antibodies that impede their spread (72). Tumor-intrinsic defense mechanisms, such as the phosphorylation of PKR mediated by IFN, induce cell cycle arrest and apoptosis, further restricting the propagation of the virus. Furthermore, the innate immune response accelerates the clearance of OV via the activation of NK cells and the production of IFN-γ (186, 187). Simultaneously, DCs present viral antigens to CD4+ T cells, thereby initiating an antiviral immune response, while neutralizing antibodies produced by B cells further augment antiviral immunity (188). Although this antiviral response is effective in regulating viral dissemination and mitigating toxicity, it concurrently reduces the efficacy of OV-mediated tumor destruction. Contemporary approaches aimed at diminishing the clearance of viruses are diverse, including polymer encapsulation, the replacement of protein coronas, the utilization of nanovesicle shielding to circumvent neutralizing antibodies, serotype switching, and the implementation of cell-based delivery systems that extend viral activity for the advancement of antitumor immunity (85, 189, 190).

An alternative perspective posits that the antiviral response may yield beneficial effects. Gujar et al. illustrated that antiviral CD4+ T cells enhance the responses of tumor-specific CD8+ T cells (188). These CD4+ T cells facilitate the maturation of DCs through interactions involving CD40-CD40L and MHC II/epitope-TCR, which allows for the cross-presentation of tumor antigens to CD8+ T cells. Consequently, these CD8+ T cells are able to target and eliminate OV-infected tumor cells (191, 192). Furthermore, Zamarin et al. demonstrated that pre-existing immunity to NDV enhance its therapeutic efficacy by augmenting systemic antitumor immunity (193).

The dual characteristics of antiviral immunity, which both inhibit OV replication and enhance antitumor immunity, present a therapeutic paradox. Addressing this issue necessitates a comprehensive understanding of the interactions among viruses, tumors, and the immune system in order to optimize therapeutic opportunities.

7.3 Potential biomarkers

A significant obstacle in the translation of OV therapy from laboratory settings to clinical application is the absence of predictive biomarkers that are grounded in the viral mechanisms of action. In contrast to well-established biomarkers, such as PD-L1 expression and elevated tumor mutational burden (TMB), which are utilized in ICI therapy, research on OV biomarkers remains in its nascent phase. There exists an urgent requirement for systematic biomarkers capable of elucidating the intricate interactions among viruses, tumors, and the immune system.

In preclinical studies, a recombinant HSV-1 vector that expresses hPD-1scFv has been shown to upregulate CTLA-4 and TIM-3 on exhausted CD8+ T cells (194). An immunohistochemical analysis conducted on 19 biopsy samples indicated that TIM-3 expression was significantly elevated in patients with a poor prognosis (P = 0.006) (195). Conversely, a clinical investigation involving 15 patients with various cancer types revealed that 60% of these patients exhibited downregulation of TIM-3 expression, which was associated with markedly improved clinical outcomes. Mechanistically, the downregulation of TIM-3 facilitated the redistribution and infiltration of CD8+ T cells into the tumor core, thereby increasing TILs (196). These findings suggest that TIM-3 may represent a potential biomarker for OV therapy; however, further validation through additional clinical trials is warranted. Notably, investigations into YST-OVH have revealed that tumors exhibiting elevated immune activation at baseline are more prone to demonstrate immune suppression. Initial studies have identified several factors, including B cell activation, complement activity, tumor-associated macrophages (TAM), and IFN signaling pathways, as potential prognostic and predictive biomarkers for OV therapy (194).

Deficiencies in host antiviral mechanisms are increasingly recognized as potential predictive biomarkers for OV therapy. For instance, indicators associated with the IFN pathway, such as the characteristics of interferon-stimulated genes (ISGs) including MX1, EPSTI1, XAF1, and GBP1, have been correlated with tumor sensitivity to VSV (197). In a departure from conventional paradigms, Ishino et al. demonstrated that oncolytic HSV-1 represents a promising therapeutic approach for hematological malignancies. The expression of nectin-1, rather than deficiencies in the cellular antiviral mechanisms, is a critical determinant of tumor cell susceptibility to HSV-1 and may serve as a predictor of therapeutic efficacy (198). Additionally, another investigation revealed that D2HG, a metabolite produced as a result of IDH1 mutations, impedes the IFN antiviral response in glioma cells, thereby increasing their sensitivity to OV therapy (199). Moreover, immunoglobulin-like transcript 2 (ILT2), a significant inhibitor of T cell responses, may function as a potential biomarker for assessing clinical responses in melanoma patients undergoing treatment with VV (200). Moreover, fluctuations in the expression levels of viral receptors, processing enzymes, and genes critical for viral infection may lead to varying degrees of susceptibility among cancer cells to particular viral agents.

Alterations in peripheral blood counts present a more straightforward and expedited predictive approach. For example, a Phase Ia/Ib clinical trial demonstrated that baseline neutrophil levels could serve as a predictor for the response to OH2, an oncolytic virus derived from HSV-2, in patients with advanced melanoma (201). Furthermore, the TUNIMO Phase I trial revealed that a decrease in acute lymphocyte levels following TILT-123 therapy is associated with therapeutic efficacy in a cohort of 20 patients with advanced solid tumors (202). These findings suggest a practical and cost-effective method for monitoring the efficacy of OV. Future investigations should aim to further validate the significance of these biomarkers in relation to other OVs to facilitate broader applicability. Additionally, a clinical study involving 202 cancer patients treated with an OAd identified low baseline serum levels of high mobility group box 1 protein (HMGB1) as an independent positive prognostic and predictive factor for oncolytic immunotherapy in individuals with advanced cancer (203).

The advancement of biomarkers for OV therapy encounters distinct challenges, particularly due to the significant tumor heterogeneity and the variability in immune pathway activation induced by different viral strains. Existing research has predominantly involved diverse cancer patient cohorts, and predictive models tailored to specific tumor types are still rare. To enhance the precision and clinical efficacy of OV therapy, it is imperative that future investigations prioritize the design of clinical trials within more homogeneous patient populations.

7.4 The Interaction between OVs and microbiome

The interplay between OVs and microbiome represents a burgeoning area of investigation within the realm of cancer immunotherapy. This interdisciplinary domain integrates aspects of virology, microbiome research, and tumor immunology, thereby providing novel insights for the treatment of cancer.

The gut microbiome, recognized as one of the most intricate microbial communities within the human body, is instrumental in modulating antitumor immune responses. Relevant research indicates that specific compositions of the gut microbiota are significantly associated with the effectiveness of OV therapy. A healthy gut microbiome facilitates T cell recognition of tumor antigens, promoting the activation of cytotoxic CD8+ T cells. Probiotic bacteria including Bacteroides fragilis, Akkermansia muciniphila, and Bifidobacterium have been linked to enhanced responses to immunotherapy (204206). In the Delta-24-RGDOX model, a high prevalence of Bifidobacterium was associated with improved survival outcomes, and the antitumor effects of Ad5D24-CpG were found to be partially reliant on the modulation of the gut microbiome (207). These observations imply that strategically altering the gut microbiome to favor a more “beneficial” bacterial composition may represent a novel therapeutic approach to augment and predict clinical outcomes for cancer patients receiving immunotherapy. Interventions such as dietary modifications, probiotic supplementation, or fecal microbiota transplantation enhance patient responses and increase therapeutic efficacy (208).

Recent investigations have elucidated a complex interplay among the gut microbiome, OVs, and the IFN system. IFN is the first line of defense against pathogens and functions as a potent immunostimulant. It possesses various roles, including antiviral activity, immune regulation, and antitumor effects. Research conducted by Yi et al. indicates that the IFN-I response, which is stimulated by the microbiota, can enhance antiviral immunity; however, excessive activation of this response may accelerate the clearance of OVs (209). Achieving a balance among these elements is essential for optimizing treatment strategies, particularly in the context of CRC. Unlike conventional viral delivery methods, the oral administration of RV not only engaged with the host immune system but also resulted in the secretion of IgA+ antibodies in the Peyer’s patch of the terminal ileum (210).

In contrast to the gut microbiome, the tumor microbiome (TM), encompassing the bacteria, fungi, and viruses found within tumor tissue, remains inadequately characterized. A recent review has outlined various strategies aimed at modulating the TM to improve cancer treatment outcomes (211). Wu and colleagues discovered that infection with Fusobacterium nucleatum in gastric cancer cells can attract tumor-associated neutrophils, which subsequently enhance the expression of PD-L1. This mechanism facilitates immune evasion and sensitizes the tumor to ICB, while potentially diminishing the immune-activating effects of OVs (212).

A growing body of research has indicated that the diversity and composition of the host gut microbiota are correlated with the effectiveness of immunotherapy and the occurrence of irAEs. This suggests the potential for utilizing microbiome as innovative biomarkers to predict patient responses to immunotherapy, as well as targeting microbiome as prospective anticancer agents, either independently or as adjuncts. An in-depth investigation into the mechanisms by which microbiome function will not only deepen our comprehension of tumorigenesis and its progression but will also elucidate the interactions between OVs and the microbiome. This enhanced understanding will serve as a basis for the formulation of more targeted and effective therapeutic approaches.

8 Discussion

OVs have emerged as a promising category of immunotherapeutic agents, exhibiting notable safety profiles and the capacity to ameliorate the immunosuppressive characteristics of the TME. OAds are particularly distinguished by their unique benefits, with over fifty clinical trials currently registered on ClinicalTrials.gov (refer to Table 5). A significant advantage of OVs, in contrast to conventional therapies, lies in their reduced systemic toxicity and enhanced tumor selectivity. Engineered OVs induce direct cytotoxic effects on neoplastic cells through targeted oncolysis, while simultaneously augmenting systemic antitumor immune responses. This process involves the activation of DCs and the proliferation of antigen-specific T cells, which contribute to the establishment of long-term immune memory. While the clinical effectiveness of OV monotherapy may be limited, the combination of OVs with ICIs has the potential to address both primary and acquired resistance, thereby improving therapeutic outcomes. Moreover, the exclusive use of ICIs may result in significant off-target organ damage and irAEs, including immune-mediated pneumonia, myocarditis, and thyroid dysfunction, which can lead to treatment cessation or even mortality. The localized expression of checkpoint modulators via OVs may mitigate systemic toxicity while preserving antitumor efficacy.

Table 5
www.frontiersin.org

Table 5. Summary of the OAds has been completed or recruiting in ClinicalTrials.gov (excluded suspend, withdraw, terminated study status).

Contemporary clinical research predominantly centers on assessing the therapeutic efficacy of OVs in individuals diagnosed with advanced or metastatic cancer. Subgroup analyses indicate that prior treatment history significantly influences prognosis; specifically, patients who have previously received sorafenib or surgical interventions tend to experience less favorable clinical outcomes, whereas those who have undergone ablation therapy may derive potential survival advantages (15). In light of these observations, the incorporation of OV combination therapy could be contemplated as a first-line treatment strategy for certain malignant tumor (213). Nonetheless, the precise efficacy and safety of this approach require validation through comprehensive clinical trials.

IT administration continues to be the primary approach for therapeutic administration; however, the difficulties associated with targeting diffuse metastases have prompted the exploration of alternative delivery methods. IV administration expands the potential applications of OV therapy, yet it faces challenges related to the presence of neutralizing antibodies. The previously mentioned oral formulation utilizing VSV has shown preclinical safety and efficacy in murine models of colon cancer and melanoma. These results have been linked to the modulation of the gut microbiome and the activation of T cells, mechanisms that do not directly oncolysis (210). This method of administration presents several advantages, including the simplification of procedural requirements, increased clinical feasibility, and improved patient recruitment and adherence to treatment protocols. Furthermore, Additionally, the combination of VSV with αPD-1 (L1) and/or αCTLA-4 antibodies shows potential for inducing durable protective immunity and enhancing treatment tolerance (210).

In the future, critical elements for the progression of OV therapy involve the implementation of multicenter clinical trials to substantiate its antitumor efficacy, the development of engineered viruses with multifaceted functionalities, and the investigation of enhanced combinatorial strategies with ICIs. These synergistic initiatives are essential for repositioning OV from a secondary option for late-stage patients to a primary modality in cancer treatment, thereby establishing it as a significant contributor to the field of tumor immunology.

Author contributions

CC: Investigation, Writing – original draft. QW: Conceptualization, Investigation, Writing – review & editing. SZ: Conceptualization, Funding acquisition, Supervision, Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Conflict of interest

The authors declare that the research 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.

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.

Glossary

OVs: Oncolytic viruses

OAds: Oncolytic adenoviruses

ICIs: Immune checkpoint inhibitors

ICD: Immunogenic cell death

TME: Tumor microenvironment

TAAs: Tumor-associated antigens

VV: Vaccinia virus

HSV: Herpes simplex virus

RV: Reovirus

MeV: Measles virus

NSCLC: Non-small cell lung cancer

PD-1/PD-L1: Programmed cell death receptor/ligand 1

CTLA-4: Cytotoxic T lymphocyte-associated protein 4

CAR: Coxsackievirus-adenovirus receptor

CR2: Conserved region 2

Rb: Retinoblastoma

RCD: Regulated cell death

DAMPs: Damage-associated molecular patterns

PAMPs: Pathogen-associated molecular patterns

TAAs: Tumor-associated antigens

CRT: Calreticulin

HSPs: Heat shock proteins

HMGB1: High-mobility group box 1

DC: Dendritic

NK: Natural killer

APC: Antigen-presenting cell

CTLs: Cytotoxic T lymphocytes

GM-CSF: Granulocyte-macrophage colony-stimulating factor

TNF-α: Tumor necrosis factor-alpha

IL-2: Interleukin-2

DIPG: Diffuse intrinsic pontine glioma

HCC: Hepatocellular carcinoma

mOS: Median overall survival

ARR: Ascites response rate

ACR: Ascites control rate

PSA: Prostate-specific antigen

IT: Intratumoral

IV: Intravenous

IP: Intraperitoneal

SC: Subcutaneous

IVE: Intravesical

IM: Intramuscular

NSCs: Neural stem cells

MSCs: Mesenchymal stem cells

RCC: Renal cell carcinoma

LAG-3: Lymphocyte activation gene-3

TIGIT: T cell immunoreceptor with immunoglobulin and ITIM domain

TIM-3: T cell immunoglobulin and mucin-domain containing-3

SIRPα: Signal regulatory protein alpha

ICOS: Inducible T cell costimulator

TLRs: Toll-like receptors

irAEs: immune-related adverse events

ICB: Immune checkpoint blockade

EMT: Epithelial-mesenchymal transition

TMB: High tumor mutational burden

PDAC: Pancreatic ductal adenocarcinoma

TNBC: Triple-negative breast cancer

MSS: Microsatellite stable

ORR: Objective response rate

SD: Stable disease

CR: Complete remission

NDV: Newcastle disease virus

T-VEC: Talimogene laherparepvec

PFS: Progression-free survival

VSV: Vesicular stomatitis virus

TILs: Tumor-infiltrating lymphocytes

CTLs: Cytotoxic T lymphocytes

PSA: Prostate-specific antigen

Tregs: Regulatory T cells

TCR: T cell receptor

IFN: Interferon

CRC: Colorectal cancer

ACT: Adoptive cell therapy

DDR: DNA damage response

TAM: Tumor-associated macrophages

ILT2: Immunoglobulin-like transcript 2

TM: Tumor microbiome

T-VEC: Talimogene laherparepvec

MDSCs: Myeloid-derived suppressor cells

IRES: Internal ribosome entry site

BCG: Bacillus Calmette-Guérin

shRNA: Short hairpin RNA

IL-6: Interleukin-6

IL-12: Interleukin-12

IL-15: Interleukin-15

scFv: Single-chain variable fragment

sdAb: Single-domain antibody

5-FU: 5-fluorouracil

ISGs: Interferon-stimulated genes

HMGB1: High mobility group box 1 protein.

References

1. Henick BS, Koch PD, Gainor JF, Awad MM, Chiuzan C, Izard S, et al. Neoadjuvant atezolizumab + chemotherapy for resectable NSCLC: 3-year clinical update of phase II clinical trial results and translational findings. J Immunother Cancer. (2024) 12:e009301. doi: 10.1136/jitc-2024-009301

PubMed Abstract | Crossref Full Text | Google Scholar

2. Robert C, Long GV, Larkin J, Wolchok JD, Hassel JC, Schadendorf D, et al. Long-term outcomes among patients who respond within the first year to nivolumab plus ipilimumab or nivolumab monotherapy: A pooled analysis in 935 patients. Eur J Cancer. (2025) 214:115119. doi: 10.1016/j.ejca.2024.115119

PubMed Abstract | Crossref Full Text | Google Scholar

3. Sun Q, Hong Z, Zhang C, Wang L, Han Z, and Ma D. Immune checkpoint therapy for solid tumours: clinical dilemmas and future trends. Signal Transduct Target Ther. (2023) 8:320. doi: 10.1038/s41392-023-01522-4

PubMed Abstract | Crossref Full Text | Google Scholar

4. Sharma P, Hu-Lieskovan S, Wargo JA, and Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. (2017) 168:707–23. doi: 10.1016/j.cell.2017.01.017

PubMed Abstract | Crossref Full Text | Google Scholar

5. Galon J and Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov. (2019) 18:197–218. doi: 10.1038/s41573-018-0007-y

PubMed Abstract | Crossref Full Text | Google Scholar

6. Gujar S, Bell J, and Diallo JS. SnapShot: cancer immunotherapy with oncolytic viruses. Cell. (2019) 176:1240–1240.e1. doi: 10.1016/j.cell.2019.01.051

PubMed Abstract | Crossref Full Text | Google Scholar

7. Niemann J and Kuhnel F. Oncolytic viruses: adenoviruses. Virus Genes. (2017) 53:700–6. doi: 10.1007/s11262-017-1488-1

PubMed Abstract | Crossref Full Text | Google Scholar

8. The oncolytic adenovirus DNX-2401 has antitumor activity in glioblastoma. Cancer Discov. (2018) 8:382. doi: 10.1158/2159-8290.CD-RW2018-031

PubMed Abstract | Crossref Full Text | Google Scholar

9. Ranki T, Pesonen S, Hemminki A, Partanen K, Kairemo K, Alanko T, et al. Phase I study with ONCOS-102 for the treatment of solid tumors - an evaluation of clinical response and exploratory analyses of immune markers. J Immunother Cancer. (2016) 4:17. doi: 10.1186/s40425-016-0121-5

PubMed Abstract | Crossref Full Text | Google Scholar

10. Machiels JP, Salazar R, Rottey S, Duran I, Dirix L, Geboes K, et al. A phase 1 dose escalation study of the oncolytic adenovirus enadenotucirev, administered intravenously to patients with epithelial solid tumors (EVOLVE). J Immunother Cancer. (2019) 7:20. doi: 10.1186/s40425-019-0510-7

PubMed Abstract | Crossref Full Text | Google Scholar

11. Atasheva S, Emerson CC, Yao J, Young C, Stewart PL, and Shayakhmetov DM. Systemic cancer therapy with engineered adenovirus that evades innate immunity. Sci Transl Med. (2020) 12:eabc6659. doi: 10.1126/scitranslmed.abc6659

PubMed Abstract | Crossref Full Text | Google Scholar

12. Zhang H, Xie W, Zhang Y, Dong X, Liu C, Yi J, et al. Oncolytic adenoviruses synergistically enhance anti-PD-L1 and anti-CTLA-4 immunotherapy by modulating the tumour microenvironment in a 4T1 orthotopic mouse model. Cancer Gene Ther. (2022) 29:456–65. doi: 10.1038/s41417-021-00389-3

PubMed Abstract | Crossref Full Text | Google Scholar

13. Klawitter M, El-Ayoubi A, Buch J, Rüttinger J, Ehrenfeld M, Lichtenegger E, et al. The oncolytic adenovirus XVir-N-31, in combination with the blockade of the PD-1/PD-L1 axis, conveys abscopal effects in a humanized glioblastoma mouse model. Int J Mol Sci. (2022) 23:9965. doi: 10.3390/ijms23179965

PubMed Abstract | Crossref Full Text | Google Scholar

14. Heiniö C, Clubb J, Kudling T, Quixabeira D, Cervera-Carrascon V, Havunen R, et al. Effective combination immunotherapy with oncolytic adenovirus and anti-PD-1 for treatment of human and murine ovarian cancers. Diseases. (2022) 10:52. doi: 10.3390/diseases10030052

PubMed Abstract | Crossref Full Text | Google Scholar

15. Yi L, Ning Z, Xu L, Shen Y, Zhu X, Yu W, et al. The combination treatment of oncolytic adenovirus H101 with nivolumab for refractory advanced hepatocellular carcinoma: an open-label, single-arm, pilot study. ESMO Open. (2024) 9:102239. doi: 10.1016/j.esmoop.2024.102239

PubMed Abstract | Crossref Full Text | Google Scholar

16. Nimmerjahn F and Ravetch JV. Fcgamma receptors as regulators of immune responses. Nat Rev Immunol. (2008) 8:34–47. doi: 10.1038/nri2206

PubMed Abstract | Crossref Full Text | Google Scholar

17. Kinder M, Greenplate AR, Strohl WR, Jordan RE, and Brezski RJ. An Fc engineering approach that modulates antibody-dependent cytokine release without altering cell-killing functions. MAbs. (2015) 7:494–504. doi: 10.1080/19420862.2015.1022692

PubMed Abstract | Crossref Full Text | Google Scholar

18. Xie D, Tian Y, Hu D, Wang Y, Yang Y, Zhou B, et al. Oncolytic adenoviruses expressing checkpoint inhibitors for cancer therapy. Signal Transduct Target Ther. (2023) 8:436. doi: 10.1038/s41392-023-01683-2

PubMed Abstract | Crossref Full Text | Google Scholar

19. Tian C, Liu J, Zhou H, Li J, Sun C, Zhu W, et al. Enhanced anti-tumor response elicited by a novel oncolytic HSV-1 engineered with an anti-PD-1 antibody. Cancer Lett. (2021) 518:49–58. doi: 10.1016/j.canlet.2021.06.005

PubMed Abstract | Crossref Full Text | Google Scholar

20. Lei GL, Wang LP, Dong SH, Sun F, Cheng JX, Yang XL, et al. A recombinant influenza virus with a CTLA4-specific scFv inhibits tumor growth in a mouse model. Cell Biol Int. (2021) 45:1202–10. doi: 10.1002/cbin.11559

PubMed Abstract | Crossref Full Text | Google Scholar

21. Wan PK, Fernandes RA, and Seymour LW. Oncolytic viruses and antibodies: are they more successful when delivered separately or when engineered as a single agent? J Immunother Cancer. (2023) 11:e006518. doi: 10.1136/jitc-2022-006518

PubMed Abstract | Crossref Full Text | Google Scholar

22. Farrera-Sal M, Moya-Borrego L, Bazan-Peregrino M, and Alemany R. Evolving status of clinical immunotherapy with oncolytic adenovirus. Clin Cancer Res. (2021) 27:2979–88. doi: 10.1158/1078-0432.CCR-20-1565

PubMed Abstract | Crossref Full Text | Google Scholar

23. Rojas JJ, Guedan S, Searle PF, Martinez-Quintanilla J, Gil-Hoyos R, Alcayaga-Miranda F, et al. Minimal RB-responsive E1A promoter modification to attain potency, selectivity, and transgene-arming capacity in oncolytic adenoviruses. Mol Ther. (2010) 18:1960–71. doi: 10.1038/mt.2010.173

PubMed Abstract | Crossref Full Text | Google Scholar

24. Bommareddy PK, Shettigar M, and Kaufman HL. Integrating oncolytic viruses in combination cancer immunotherapy. Nat Rev Immunol. (2018) 18:498–513. doi: 10.1038/s41577-018-0014-6

PubMed Abstract | Crossref Full Text | Google Scholar

25. Jayawardena N, Burga LN, Poirier JT, and Bostina M. Virus-receptor interactions: structural insights for oncolytic virus development. Oncol Virother. (2019) 8:39–56. doi: 10.2147/OV.S218494

PubMed Abstract | Crossref Full Text | Google Scholar

26. Charman M, Herrmann C, and Weitzman MD. Viral and cellular interactions during adenovirus DNA replication. FEBS Lett. (2019) 593:3531–50. doi: 10.1002/1873-3468.13695

PubMed Abstract | Crossref Full Text | Google Scholar

27. Heise C, Hermiston T, Johnson L, Brooks G, Sampson-Johannes A, Williams A, et al. An adenovirus E1A mutant that demonstrates potent and selective systemic anti-tumoral efficacy. Nat Med. (2000) 6:1134–9. doi: 10.1038/80474

PubMed Abstract | Crossref Full Text | Google Scholar

28. Abudoureyimu M, Lai Y, Tian C, Wang T, Wang R, and Chu X. Oncolytic adenovirus-A nova for gene-targeted oncolytic viral therapy in HCC. Front Oncol. (2019) 9:1182. doi: 10.3389/fonc.2019.01182

PubMed Abstract | Crossref Full Text | Google Scholar

29. Hidalgo P, Ip WH, Dobner T, and Gonzalez RA. The biology of the adenovirus E1B 55K protein. FEBS Lett. (2019) 593:3504–17. doi: 10.1002/1873-3468.13694

PubMed Abstract | Crossref Full Text | Google Scholar

30. Liu TC, Hallden G, Wang Y, Brooks G, Francis J, Lemoine N, et al. An E1B-19 kDa gene deletion mutant adenovirus demonstrates tumor necrosis factor-enhanced cancer selectivity and enhanced oncolytic potency. Mol Ther. (2004) 9:786–803. doi: 10.1016/j.ymthe.2004.03.017

PubMed Abstract | Crossref Full Text | Google Scholar

31. Wiman KG. Strategies for therapeutic targeting of the p53 pathway in cancer. Cell Death Differ. (2006) 13:921–6. doi: 10.1038/sj.cdd.4401921

PubMed Abstract | Crossref Full Text | Google Scholar

32. Bischoff JR, Kirn DH, Williams A, Heise C, Horn S, Muna M, et al. An adenovirus mutant that replicates selectively in p53-deficient human tumor cells. Science. (1996) 274:373–6. doi: 10.1126/science.274.5286.373

PubMed Abstract | Crossref Full Text | Google Scholar

33. Garber K. China approves world’s first oncolytic virus therapy for cancer treatment. J Natl Cancer Inst. (2006) 98:298–300. doi: 10.1093/jnci/djj111

PubMed Abstract | Crossref Full Text | Google Scholar

34. Fueyo J, Alemany R, Gomez-Manzano C, Fuller GN, Khan A, Conrad CA, et al. Preclinical characterization of the antiglioma activity of a tropism-enhanced adenovirus targeted to the retinoblastoma pathway. J Natl Cancer Inst. (2003) 95:652–60. doi: 10.1093/jnci/95.9.652

PubMed Abstract | Crossref Full Text | Google Scholar

35. Fueyo J, Gomez-Manzano C, Alemany R, Lee PS, McDonnell TJ, Mitlianga P, et al. A mutant oncolytic adenovirus targeting the Rb pathway produces anti-glioma effect in vivo. Oncogene. (2000) 19:2–12. doi: 10.1038/sj.onc.1203251

PubMed Abstract | Crossref Full Text | Google Scholar

36. Kuhn I, Harden P, Bauzon M, Chartier C, Nye J, Thorne S, et al. Directed evolution generates a novel oncolytic virus for the treatment of colon cancer. PloS One. (2008) 3:e2409. doi: 10.1371/journal.pone.0002409

PubMed Abstract | Crossref Full Text | Google Scholar

37. Holterman L, Vogels R, van der Vlugt R, Sieuwerts M, Grimbergen J, Kaspers J, et al. Novel replication-incompetent vector derived from adenovirus type 11 (Ad11) for vaccination and gene therapy: low seroprevalence and non-cross-reactivity with Ad5. J Virol. (2004) 78:13207–15. doi: 10.1128/JVI.78.23.13207-13215.2004

PubMed Abstract | Crossref Full Text | Google Scholar

38. Fakih M, Harb W, Mahadevan D, Babiker H, Berlin J, Lillie T, et al. Safety and efficacy of the tumor-selective adenovirus enadenotucirev, in combination with nivolumab, in patients with advanced/metastatic epithelial cancer: a phase I clinical trial (SPICE). J Immunother Cancer. (2023) 11:e006561. doi: 10.1136/jitc-2022-006561

PubMed Abstract | Crossref Full Text | Google Scholar

39. Galluzzi L, Buque A, Kepp O, Zitvogel L, and Kroemer G. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol. (2017) 17:97–111. doi: 10.1038/nri.2016.107

PubMed Abstract | Crossref Full Text | Google Scholar

40. Ma J, Ramachandran M, Jin C, Quijano-Rubio C, Martikainen M, Yu D, et al. Characterization of virus-mediated immunogenic cancer cell death and the consequences for oncolytic virus-based immunotherapy of cancer. Cell Death Dis. (2020) 11:48. doi: 10.1038/s41419-020-2236-3

PubMed Abstract | Crossref Full Text | Google Scholar

41. Galluzzi L, Vitale I, Warren S, Adjemian S, Agostinis P, Martinez AB, et al. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J Immunother Cancer. (2020) 8:e000337corr1. doi: 10.1136/jitc-2019-000337corr1

PubMed Abstract | Crossref Full Text | Google Scholar

42. Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P, et al. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer. (2012) 12:860–75. doi: 10.1038/nrc3380

PubMed Abstract | Crossref Full Text | Google Scholar

43. Garg AD, Dudek-Peric AM, Romano E, and Agostinis P. Immunogenic cell death. Int J Dev Biol. (2015) 59:131–40. doi: 10.1387/ijdb.150061pa

PubMed Abstract | Crossref Full Text | Google Scholar

44. Tang D, Kang R, Coyne CB, Zeh HJ, and Lotze MT. PAMPs and DAMPs: signal 0s that spur autophagy and immunity. Immunol Rev. (2012) 249:158–75. doi: 10.1111/j.1600-065X.2012.01146.x

PubMed Abstract | Crossref Full Text | Google Scholar

45. Kim Y, Clements DR, Sterea AM, Jang HW, Gujar SA, and Lee PW. Dendritic cells in oncolytic virus-based anti-cancer therapy. Viruses. (2015) 7:6506–25. doi: 10.3390/v7122953

PubMed Abstract | Crossref Full Text | Google Scholar

46. Ramesh N, Ge Y, Ennist DL, Zhu M, Mina M, Ganesh S, et al. CG0070, a conditionally replicating granulocyte macrophage colony-stimulating factor–armed oncolytic adenovirus for the treatment of bladder cancer. Clin Cancer Res. (2006) 12:305–13. doi: 10.1158/1078-0432.CCR-05-1059

PubMed Abstract | Crossref Full Text | Google Scholar

47. Bramante S, Koski A, Kipar A, Diaconu I, Liikanen I, Hemminki O, et al. Serotype chimeric oncolytic adenovirus coding for GM-CSF for treatment of sarcoma in rodents and humans. Int J Cancer. (2014) 135:720–30. doi: 10.1002/ijc.28696

PubMed Abstract | Crossref Full Text | Google Scholar

48. Hemminki O, Parviainen S, Juhila J, Turkki R, Linder N, Lundin J, et al. Immunological data from cancer patients treated with Ad5/3-E2F-Δ24-GMCSF suggests utility for tumor immunotherapy. Oncotarget. (2015) 6:4467–81. doi: 10.18632/oncotarget.2901

PubMed Abstract | Crossref Full Text | Google Scholar

49. Li R, Villa NY, Yu X, Johnson JO, Borjas G, Dhillon J, et al. Oncolytic immunotherapy with nivolumab in muscle-invasive bladder cancer: a phase 1b trial. Nat Med. (2024) 31(1):176–88. doi: 10.1038/s41591-024-03324-9

PubMed Abstract | Crossref Full Text | Google Scholar

50. Pakola SA, Peltola KJ, Clubb JHA, Jirovec E, Haybout L, Kudling TV, et al. Safety, efficacy, and biological data of T-cell-enabling oncolytic adenovirus TILT-123 in advanced solid cancers from the TUNIMO monotherapy phase I trial. Clin Cancer Res. (2024) 30:3715–25. doi: 10.1158/1078-0432.CCR-23-3874

PubMed Abstract | Crossref Full Text | Google Scholar

51. Zhang Y, Zhang H, Wei M, Mou T, Shi T, Ma Y, et al. Recombinant adenovirus expressing a soluble fusion protein PD-1/CD137L subverts the suppression of CD8(+) T cells in HCC. Mol Ther. (2019) 27:1906–18. doi: 10.1016/j.ymthe.2019.07.019

PubMed Abstract | Crossref Full Text | Google Scholar

52. Lang FF, Conrad C, Gomez-Manzano C, Yung WKA, Sawaya R, Weinberg JS, et al. Phase I study of DNX-2401 (Delta-24-RGD) oncolytic adenovirus: replication and immunotherapeutic effects in recurrent Malignant glioma. J Clin Oncol. (2018) 36:1419–27. doi: 10.1200/JCO.2017.75.8219

PubMed Abstract | Crossref Full Text | Google Scholar

53. Gallego Perez-Larraya J, Garcia-Moure M, Labiano S, Patino-Garcia A, Dobbs J, Gonzalez-Huarriz M, et al. Oncolytic DNX-2401 virus for pediatric diffuse intrinsic pontine glioma. N Engl J Med. (2022) 386:2471–81. doi: 10.1056/NEJMoa2202028

PubMed Abstract | Crossref Full Text | Google Scholar

54. Heo J, Liang JD, Kim CW, Woo HY, Shih IL, Su TH, et al. Safety and dose escalation of the targeted oncolytic adenovirus OBP-301 for refractory advanced liver cancer: Phase I clinical trial. Mol Ther. (2023) 31:2077–88. doi: 10.1016/j.ymthe.2023.04.006

PubMed Abstract | Crossref Full Text | Google Scholar

55. Zhang Y, Qian L, Chen K, Gu S, Wang J, Meng Z, et al. Intraperitoneal oncolytic virotherapy for patients with Malignant ascites: Characterization of clinical efficacy and antitumor immune response. Mol Ther Oncol. (2022) 25:31–42. doi: 10.1016/j.omto.2022.03.003

PubMed Abstract | Crossref Full Text | Google Scholar

56. Zhang Y, Qian L, Chen K, Gu S, Meng Z, Wang J, et al. Oncolytic adenovirus in treating Malignant ascites: A phase II trial and longitudinal single-cell study. Mol Ther. (2024) 32:2000–20. doi: 10.1016/j.ymthe.2024.04.029

PubMed Abstract | Crossref Full Text | Google Scholar

57. McCormick F. ONYX-015 selectivity and the p14ARF pathway. Oncogene. (2000) 19:6670–2. doi: 10.1038/sj.onc.1204096

PubMed Abstract | Crossref Full Text | Google Scholar

58. Zhang ZL, Zou WG, Luo CX, Li BH, Wang JH, Sun LY, et al. An armed oncolytic adenovirus system, ZD55-gene, demonstrating potent antitumoral efficacy. Cell Res. (2003) 13:481–9. doi: 10.1038/sj.cr.7290191

PubMed Abstract | Crossref Full Text | Google Scholar

59. Yu DC, Chen Y, Seng M, Dilley J, and Henderson DR. The addition of adenovirus type 5 region E3 enables calydon virus 787 to eliminate distant prostate tumor xenografts. Cancer Res. (1999) 59:4200–3.

PubMed Abstract | Google Scholar

60. DeWeese TL, van der Poel H, Li S, Mikhak B, Drew R, Goemann M, et al. A phase I trial of CV706, a replication-competent, PSA selective oncolytic adenovirus, for the treatment of locally recurrent prostate cancer following radiation therapy. Cancer Res. (2001) 61:7464–72.

PubMed Abstract | Google Scholar

61. Kawashima T, Kagawa S, Kobayashi N, Shirakiya Y, Umeoka T, Teraishi F, et al. Telomerase-specific replication-selective virotherapy for human cancer. Clin Cancer Res. (2004) 10:285–92. doi: 10.1158/1078-0432.CCR-1075-3

PubMed Abstract | Crossref Full Text | Google Scholar

62. Hashimoto Y, Watanabe Y, Shirakiya Y, Uno F, Kagawa S, Kawamura H, et al. Establishment of biological and pharmacokinetic assays of telomerase-specific replication-selective adenovirus. Cancer Sci. (2008) 99:385–90. doi: 10.1111/j.1349-7006.2007.00665.x

PubMed Abstract | Crossref Full Text | Google Scholar

63. Yano S, Takehara K, Kishimoto H, Tazawa H, Urata Y, Kagawa S, et al. Tumor-targeting adenovirus OBP-401 inhibits primary and metastatic tumor growth of triple-negative breast cancer in orthotopic nude-mouse models. Oncotarget. (2016) 7:85273–82. doi: 10.18632/oncotarget.13296

PubMed Abstract | Crossref Full Text | Google Scholar

64. Ying C, Xiao BD, Qin Y, Wang BR, Liu XY, Wang RW, et al. GOLPH2-regulated oncolytic adenovirus, GD55, exerts strong killing effect on human prostate cancer stem-like cells in vitro and in vivo. Acta Pharmacol Sin. (2018) 39:405–14. doi: 10.1038/aps.2017.91

PubMed Abstract | Crossref Full Text | Google Scholar

65. Fares J, Ahmed AU, Ulasov IV, Sonabend AM, Miska J, Lee-Chang C, et al. Neural stem cell delivery of an oncolytic adenovirus in newly diagnosed Malignant glioma: a first-in-human, phase 1, dose-escalation trial. Lancet Oncol. (2021) 22:1103–14. doi: 10.1016/S1470-2045(21)00245-X

PubMed Abstract | Crossref Full Text | Google Scholar

66. Naumenko VA, Vishnevskiy DA, Stepanenko AA, Sosnovtseva AO, Chernysheva AA, Abakumova TO, et al. In vivo tracking for oncolytic adenovirus interactions with liver cells. Biomedicines. (2022) 10:1697. doi: 10.3390/biomedicines10071697

PubMed Abstract | Crossref Full Text | Google Scholar

67. Wienen F, Nilson R, Allmendinger E, Peters S, Barth TFE, Kochanek S, et al. An oncolytic HAdV-5 with reduced surface charge combines diminished toxicity and improved tumor targeting. Mol Ther Oncol. (2024) 32:200909. doi: 10.1016/j.omton.2024.200909

PubMed Abstract | Crossref Full Text | Google Scholar

68. Wang P, Li X, Wang J, Gao D, Li Y, Li H, et al. Re-designing Interleukin-12 to enhance its safety and potential as an anti-tumor immunotherapeutic agent. Nat Commun. (2017) 8:1395. doi: 10.1038/s41467-017-01385-8

PubMed Abstract | Crossref Full Text | Google Scholar

69. Jung BK, Ko HY, Kang H, Hong J, Ahn HM, Na Y, et al. Relaxin-expressing oncolytic adenovirus induces remodeling of physical and immunological aspects of cold tumor to potentiate PD-1 blockade. J Immunother Cancer. (2020) 8:e000763. doi: 10.1136/jitc-2020-000763

PubMed Abstract | Crossref Full Text | Google Scholar

70. Boorjian SA, Alemozaffar M, Konety BR, Shore ND, Gomella LG, Kamat AM, et al. Intravesical nadofaragene firadenovec gene therapy for BCG-unresponsive non-muscle-invasive bladder cancer: a single-arm, open-label, repeat-dose clinical trial. Lancet Oncol. (2021) 22:107–17. doi: 10.1016/S1470-2045(20)30540-4

PubMed Abstract | Crossref Full Text | Google Scholar

71. Zheng M, Huang J, Tong A, and Yang H. Oncolytic viruses for cancer therapy: barriers and recent advances. Mol Ther Oncol. (2019) 15:234–47. doi: 10.1016/j.omto.2019.10.007

PubMed Abstract | Crossref Full Text | Google Scholar

72. Chen L, Zuo M, Zhou Q, and Wang Y. Oncolytic virotherapy in cancer treatment: challenges and optimization prospects. Front Immunol. (2023) 14:1308890. doi: 10.3389/fimmu.2023.1308890

PubMed Abstract | Crossref Full Text | Google Scholar

73. Ye X, Jerebtsova M, and Ray PE. Liver bypass significantly increases the transduction efficiency of recombinant adenoviral vectors in the lung, intestine, and kidney. Hum Gene Ther. (2000) 11:621–7. doi: 10.1089/10430340050015806

PubMed Abstract | Crossref Full Text | Google Scholar

74. Worgall S, Wolff G, Falck-Pedersen E, and Crystal RG. Innate immune mechanisms dominate elimination of adenoviral vectors following in vivo administration. Hum Gene Ther. (1997) 8:37–44. doi: 10.1089/hum.1997.8.1-37

PubMed Abstract | Crossref Full Text | Google Scholar

75. Wang Z, Yu B, Wang B, Yan J, Feng X, Wang Z, et al. A novel capsid-modified oncolytic recombinant adenovirus type 5 for tumor-targeting gene therapy by intravenous route. Oncotarget. (2016) 7:47287–301. doi: 10.18632/oncotarget.10075

PubMed Abstract | Crossref Full Text | Google Scholar

76. Zhao Y, Le TMD, Hong J, Jiao A, Yoon AR, and Yun CO. Smart accumulating dual-targeting lipid envelopes equipping oncolytic adenovirus for enhancing cancer gene therapeutic efficacy. ACS Nano. (2024) 18:27869–90. doi: 10.1021/acsnano.4c02165

PubMed Abstract | Crossref Full Text | Google Scholar

77. Rincón E, Cejalvo T, Kanojia D, Alfranca A, Rodríguez-Milla M, Gil Hoyos RA, et al. Mesenchymal stem cell carriers enhance antitumor efficacy of oncolytic adenoviruses in an immunocompetent mouse model. Oncotarget. (2017) 8:45415–31. doi: 10.18632/oncotarget.17557

PubMed Abstract | Crossref Full Text | Google Scholar

78. Chen Y, Chen X, Bao W, Liu G, Wei W, and Ping Y. An oncolytic virus-T cell chimera for cancer immunotherapy. Nat Biotechnol. (2024) 42:1876–87. doi: 10.1038/s41587-023-02118-7

PubMed Abstract | Crossref Full Text | Google Scholar

79. Santos J, Heiniö C, Quixabeira D, Zafar S, Clubb J, Pakola S, et al. Systemic delivery of oncolytic adenovirus to tumors using tumor-infiltrating lymphocytes as carriers. Cells. (2021) 10:978. doi: 10.3390/cells10050978

PubMed Abstract | Crossref Full Text | Google Scholar

80. Ding L, Gao Q, Xu Z, Cai L, Chen S, Zhang X, et al. An inter-supplementary biohybrid system based on natural killer cells for the combinational immunotherapy and virotherapy of cancer. Adv Sci (Weinh). (2022) 9:e2103470. doi: 10.1002/advs.202103470

PubMed Abstract | Crossref Full Text | Google Scholar

81. Li ZL, Liang X, Li HC, Wang ZM, and Chong T. Dendritic cells serve as a “Trojan horse” for oncolytic adenovirus delivery in the treatment of mouse prostate cancer. Acta Pharmacol Sin. (2016) 37:1121–8. doi: 10.1038/aps.2016.59

PubMed Abstract | Crossref Full Text | Google Scholar

82. He X, Yao W, Zhu JD, Jin X, Liu XY, Zhang KJ, et al. Potent antitumor efficacy of human dental pulp stem cells armed with YSCH-01 oncolytic adenovirus. J Transl Med. (2023) 21:688. doi: 10.1186/s12967-023-04539-z

PubMed Abstract | Crossref Full Text | Google Scholar

83. Saari H, Turunen T, Lõhmus A, Turunen M, Jalasvuori M, Butcher SJ, et al. Extracellular vesicles provide a capsid-free vector for oncolytic adenoviral DNA delivery. J Extracell Vesicles. (2020) 9:1747206. doi: 10.1080/20013078.2020.1747206

PubMed Abstract | Crossref Full Text | Google Scholar

84. Zhang W, Zhang J, Zhang J, Chu J, and Zhang Z. Novel combination therapy using recombinant oncolytic adenovirus silk hydrogel and PD-L1 inhibitor for bladder cancer treatment. J Nanobiotechnol. (2024) 22:638. doi: 10.1186/s12951-024-02903-9

PubMed Abstract | Crossref Full Text | Google Scholar

85. Lv P, Chen X, Fu S, Ren E, Liu C, Liu X, et al. Surface engineering of oncolytic adenovirus for a combination of immune checkpoint blockade and virotherapy. Biomater Sci. (2021) 9:7392–401. doi: 10.1039/D1BM00928A

PubMed Abstract | Crossref Full Text | Google Scholar

86. Cejalvo JM, Falato C, Villanueva L, Tolosa P, González X, Pascal M, et al. Oncolytic viruses: A new immunotherapeutic approach for breast cancer treatment? Cancer Treat Rev. (2022) 106:102392. doi: 10.1016/j.ctrv.2022.102392

PubMed Abstract | Crossref Full Text | Google Scholar

87. Yuan S, Wang J, Zhu D, Wang N, Gao Q, Chen W, et al. Cryo-EM structure of a herpesvirus capsid at 3.1 Å. Science. (2018) 360:eaao7283. doi: 10.1126/science.aao7283

PubMed Abstract | Crossref Full Text | Google Scholar

88. Mozzi A, Cagliani R, Pontremoli C, Forni D, Saulle I, Saresella M, et al. Simplexviruses successfully adapt to their host by fine-tuning immune responses. Mol Biol Evol. (2022) 39:msac142. doi: 10.1093/molbev/msac142

PubMed Abstract | Crossref Full Text | Google Scholar

89. Conry RM, Westbrook B, McKee S, and Norwood TG. Talimogene laherparepvec: First in class oncolytic virotherapy. Hum Vaccin Immunother. (2018) 14:839–46. doi: 10.1080/21645515.2017.1412896

PubMed Abstract | Crossref Full Text | Google Scholar

90. Todo T, Ito H, Ino Y, Ohtsu H, Ota Y, Shibahara J, et al. Intratumoral oncolytic herpes virus G47Δ for residual or recurrent glioblastoma: a phase 2 trial. Nat Med. (2022) 28:1630–9. doi: 10.1038/s41591-022-01897-x

PubMed Abstract | Crossref Full Text | Google Scholar

91. Guo ZS, Lu B, Guo Z, Giehl E, Feist M, Dai E, et al. Vaccinia virus-mediated cancer immunotherapy: cancer vaccines and oncolytics. J Immunother Cancer. (2019) 7:6. doi: 10.1186/s40425-018-0495-7

PubMed Abstract | Crossref Full Text | Google Scholar

92. Katsafanas GC and Moss B. Colocalization of transcription and translation within cytoplasmic poxvirus factories coordinates viral expression and subjugates host functions. Cell Host Microbe. (2007) 2:221–8. doi: 10.1016/j.chom.2007.08.005

PubMed Abstract | Crossref Full Text | Google Scholar

93. Salzman NP. The rate of formation of vaccinia deoxyribonucleic acid and vaccinia virus. Virology. (1960) 10:150–2. doi: 10.1016/0042-6822(60)90015-5

PubMed Abstract | Crossref Full Text | Google Scholar

94. Alemany R. A smart move against cancer for vaccinia virus. Lancet Oncol. (2008) 9:507–8. doi: 10.1016/S1470-2045(08)70136-0

PubMed Abstract | Crossref Full Text | Google Scholar

95. Samson A, West EJ, Carmichael J, Scott KJ, Turnbull S, Kuszlewicz B, et al. Neoadjuvant intravenous oncolytic vaccinia virus therapy promotes anticancer immunity in patients. Cancer Immunol Res. (2022) 10:745–56. doi: 10.1158/2326-6066.CIR-21-0171

PubMed Abstract | Crossref Full Text | Google Scholar

96. Kelly K, Nawrocki S, Mita A, Coffey M, Giles FJ, and Mita M. Reovirus-based therapy for cancer. Expert Opin Biol Ther. (2009) 9:817–30. doi: 10.1517/14712590903002039

PubMed Abstract | Crossref Full Text | Google Scholar

97. Kim M, Chung YH, and Johnston RN. Reovirus and tumor oncolysis. J Microbiol. (2007) 45:187–92.

Google Scholar

98. Clark AS, Zhao F, Klein P, Montero AJ, Falkson C, Krill-Jackson E, et al. A Phase II Randomized Study of Paclitaxel alone or combined with Pelareorep with or without Avelumab in Metastatic Hormone Receptor Positive Breast Cancer: the BRACELET-01/PrE0113 study. Clin Cancer Res. (2025). doi: 10.1158/1078-0432.CCR-24-2701

PubMed Abstract | Crossref Full Text | Google Scholar

99. Horikami SM and Moyer SA. Structure, transcription, and replication of measles virus. Curr Top Microbiol Immunol. (1995) 191:35–50. doi: 10.1007/978-3-642-78621-1_3

PubMed Abstract | Crossref Full Text | Google Scholar

100. Mühlebach MD. Measles virus in cancer therapy. Curr Opin Virol. (2020) 41:85–97. doi: 10.1016/j.coviro.2020.07.016

PubMed Abstract | Crossref Full Text | Google Scholar

101. Schirrmacher V, van Gool S, and Stuecker W. Breaking therapy resistance: an update on oncolytic Newcastle disease virus for improvements of cancer therapy. Biomedicines. (2019) 7:66. doi: 10.3390/biomedicines7030066

PubMed Abstract | Crossref Full Text | Google Scholar

102. Tian Y, Xie D, and Yang L. Engineering strategies to enhance oncolytic viruses in cancer immunotherapy. Signal Transduct Target Ther. (2022) 7:117. doi: 10.1038/s41392-022-00951-x

PubMed Abstract | Crossref Full Text | Google Scholar

103. Li M, Zhu Y, and Li M. Newcastle disease virus induces pyroptosis in canine mammary tumour CMT-U27 cells via the TNFα/NF-κB/NLRP3 signalling pathway. Vet Comp Oncol. (2025) 23:224–35. doi: 10.1111/vco.13048

PubMed Abstract | Crossref Full Text | Google Scholar

104. Dummer R, Robert C, Scolyer RA, Taube JM, Tetzlaff MT, Menzies AM, et al. Neoadjuvant anti-PD-1 alone or in combination with anti-TIGIT or an oncolytic virus in resectable stage IIIB-D melanoma: a phase 1/2 trial. Nat Med. (2025) 31:144–51. doi: 10.1038/s41591-024-03411-x

PubMed Abstract | Crossref Full Text | Google Scholar

105. Bryant JD, Lee JS, De Almeida A, Jacques J, Chang CH, Fassler W, et al. Seneca Valley virus replicons are packaged in trans and have the capacity to overcome the limitations of viral transgene expression. Mol Ther Oncol. (2023) 28:321–33. doi: 10.1016/j.omto.2023.02.005

PubMed Abstract | Crossref Full Text | Google Scholar

106. Vazeh H, Behboudi E, Hashemzadeh-Omran A, and Moradi A. Live-attenuated poliovirus-induced extrinsic apoptosis through Caspase 8 within breast cancer cell lines expressing CD155. Breast Cancer. (2022) 29:899–907. doi: 10.1007/s12282-022-01372-y

PubMed Abstract | Crossref Full Text | Google Scholar

107. Mullins-Dansereau V, Myre ML, Bardoul A, Geoffroy K, Rallo Pita MJ, Béland D, et al. Oncolytic VSV-IL-2 has enhanced anticancer vaccination adjuvant abilities. J Immunother Cancer. (2025) 13:e010570. doi: 10.1136/jitc-2024-010570

PubMed Abstract | Crossref Full Text | Google Scholar

108. Walunas TL, Lenschow DJ, Bakker CY, Linsley PS, Freeman GJ, Green JM, et al. CTLA-4 can function as a negative regulator of T cell activation. Immunity. (1994) 1:405–13. doi: 10.1016/1074-7613(94)90071-X

PubMed Abstract | Crossref Full Text | Google Scholar

109. Krummel MF and Allison JP. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med. (1995) 182:459–65. doi: 10.1084/jem.182.2.459

PubMed Abstract | Crossref Full Text | Google Scholar

110. Wei SC, Duffy CR, and Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. (2018) 8:1069–86. doi: 10.1158/2159-8290.CD-18-0367

PubMed Abstract | Crossref Full Text | Google Scholar

111. Leach DR, Krummel MF, and Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. (1996) 271:1734–6. doi: 10.1126/science.271.5256.1734

PubMed Abstract | Crossref Full Text | Google Scholar

112. Dong H, Zhu G, Tamada K, and Chen L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat Med. (1999) 5:1365–9. doi: 10.1038/70932

PubMed Abstract | Crossref Full Text | Google Scholar

113. Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. (2001) 2:261–8. doi: 10.1038/85330

PubMed Abstract | Crossref Full Text | Google Scholar

114. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. (2000) 192:1027–34. doi: 10.1084/jem.192.7.1027

PubMed Abstract | Crossref Full Text | Google Scholar

115. Okazaki T, Maeda A, Nishimura H, Kurosaki T, and Honjo T. PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc Natl Acad Sci. (2001) 98:13866–71. doi: 10.1073/pnas.231486598

PubMed Abstract | Crossref Full Text | Google Scholar

116. Sheppard KA, Fitz LJ, Lee JM, Benander C, George JA, Wooters J, et al. PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3zeta signalosome and downstream signaling to PKCtheta. FEBS Lett. (2004) 574:37–41. doi: 10.1016/j.febslet.2004.07.083

PubMed Abstract | Crossref Full Text | Google Scholar

117. Triebel F, Jitsukawa S, Baixeras E, Roman-Roman S, Genevee C, Viegas-Pequignot E, et al. LAG-3, a novel lymphocyte activation gene closely related to CD4. J Exp Med. (1990) 171:1393–405. doi: 10.1084/jem.171.5.1393

PubMed Abstract | Crossref Full Text | Google Scholar

118. Paik J. Nivolumab plus relatlimab: first approval. Drugs. (2022) 82:925–31. doi: 10.1007/s40265-022-01723-1

PubMed Abstract | Crossref Full Text | Google Scholar

119. Zhang P, Liu X, Gu Z, Jiang Z, Zhao S, Song Y, et al. Targeting TIGIT for cancer immunotherapy: recent advances and future directions. biomark Res. (2024) 12:7. doi: 10.1186/s40364-023-00543-z

PubMed Abstract | Crossref Full Text | Google Scholar

120. Ge Z, Peppelenbosch MP, Sprengers D, and Kwekkeboom J. TIGIT, the next step towards successful combination immune checkpoint therapy in cancer. Front Immunol. (2021) 12:699895. doi: 10.3389/fimmu.2021.699895

PubMed Abstract | Crossref Full Text | Google Scholar

121. Pauken KE and Wherry EJ. TIGIT and CD226: tipping the balance between costimulatory and coinhibitory molecules to augment the cancer immunotherapy toolkit. Cancer Cell. (2014) 26:785–7. doi: 10.1016/j.ccell.2014.11.016

PubMed Abstract | Crossref Full Text | Google Scholar

122. Chauvin JM, Ka M, Pagliano O, Menna C, Ding Q, DeBlasio R, et al. IL15 stimulation with TIGIT blockade reverses CD155-mediated NK-cell dysfunction in melanoma. Clin Cancer Res. (2020) 26:5520–33. doi: 10.1158/1078-0432.CCR-20-0575

PubMed Abstract | Crossref Full Text | Google Scholar

123. Johnston RJ, Comps-Agrar L, Hackney J, Yu X, Huseni M, Yang Y, et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. Cancer Cell. (2014) 26:923–37. doi: 10.1016/j.ccell.2014.10.018

PubMed Abstract | Crossref Full Text | Google Scholar

124. Zhang J, Wang L, Guo H, Kong S, Li W, He Q, et al. The role of Tim-3 blockade in the tumor immune microenvironment beyond T cells. Pharmacol Res. (2024) 209:107458. doi: 10.1016/j.phrs.2024.107458

PubMed Abstract | Crossref Full Text | Google Scholar

125. Hellmann MD, Bivi N, Calderon B, Shimizu T, Delafontaine B, Liu ZT, et al. Safety and immunogenicity of LY3415244, a bispecific antibody against TIM-3 and PD-L1, in patients with advanced solid tumors. Clin Cancer Res. (2021) 27:2773–81. doi: 10.1158/1078-0432.CCR-20-3716

PubMed Abstract | Crossref Full Text | Google Scholar

126. de Spéville BD and Moreno V. Antidrug antibodies and drug development: challenges in the immunotherapy era. Clin Cancer Res. (2021) 27:2669–71. doi: 10.1158/1078-0432.CCR-21-0168

PubMed Abstract | Crossref Full Text | Google Scholar

127. Liu Y, Weng L, Wang Y, Zhang J, Wu Q, Zhao P, et al. Deciphering the role of CD47 in cancer immunotherapy. J Adv Res. (2024) 63:129–58. doi: 10.1016/j.jare.2023.10.009

PubMed Abstract | Crossref Full Text | Google Scholar

128. Timms JF, Swanson KD, Marie-Cardine A, Raab M, Rudd CE, Schraven B, et al. SHPS-1 is a scaffold for assembling distinct adhesion-regulated multi-protein complexes in macrophages. Curr Biol. (1999) 9:927–30. doi: 10.1016/S0960-9822(99)80401-1

PubMed Abstract | Crossref Full Text | Google Scholar

129. Gaikwad S, Agrawal MY, Kaushik I, Ramachandran S, and Srivastava SK. Immune checkpoint proteins: Signaling mechanisms and molecular interactions in cancer immunotherapy. Semin Cancer Biol. (2022) 86:137–50. doi: 10.1016/j.semcancer.2022.03.014

PubMed Abstract | Crossref Full Text | Google Scholar

130. Laspidea V, Puigdelloses M, Labiano S, Marrodán L, Garcia-Moure M, Zalacain M, et al. Exploiting 4-1BB immune checkpoint to enhance the efficacy of oncolytic virotherapy for diffuse intrinsic pontine gliomas. JCI Insight. (2022) 7:e154812. doi: 10.1172/jci.insight.154812

PubMed Abstract | Crossref Full Text | Google Scholar

131. Croft M. Co-stimulatory members of the TNFR family: keys to effective T-cell immunity? Nat Rev Immunol. (2003) 3:609–20. doi: 10.1038/nri1148

PubMed Abstract | Crossref Full Text | Google Scholar

132. Yonezawa A, Dutt S, Chester C, Kim J, and Kohrt HE. Boosting cancer immunotherapy with anti-CD137 antibody therapy. Clin Cancer Res. (2015) 21:3113–20. doi: 10.1158/1078-0432.CCR-15-0263

PubMed Abstract | Crossref Full Text | Google Scholar

133. Chester C, Sanmamed MF, Wang J, and Melero I. Immunotherapy targeting 4-1BB: mechanistic rationale, clinical results, and future strategies. Blood. (2018) 131:49–57. doi: 10.1182/blood-2017-06-741041

PubMed Abstract | Crossref Full Text | Google Scholar

134. Sato Y, Goto Y, Narita N, and Hoon DS. Cancer cells expressing toll-like receptors and the tumor microenvironment. Cancer Microenviron. (2009) 2 Suppl 1:205–14. doi: 10.1007/s12307-009-0022-y

PubMed Abstract | Crossref Full Text | Google Scholar

135. Ito H, Ando T, Arioka Y, Saito K, and Seishima M. Inhibition of indoleamine 2,3-dioxygenase activity enhances the anti-tumour effects of a Toll-like receptor 7 agonist in an established cancer model. Immunology. (2015) 144:621–30. doi: 10.1111/imm.12413

PubMed Abstract | Crossref Full Text | Google Scholar

136. Karasarides M, Cogdill AP, Robbins PB, Bowden M, Burton EM, Butterfield LH, et al. Hallmarks of resistance to immune-checkpoint inhibitors. Cancer Immunol Res. (2022) 10:372–83. doi: 10.1158/2326-6066.CIR-20-0586

PubMed Abstract | Crossref Full Text | Google Scholar

137. Vesely MD, Zhang T, and Chen L. Resistance mechanisms to anti-PD cancer immunotherapy. Annu Rev Immunol. (2022) 40:45–74. doi: 10.1146/annurev-immunol-070621-030155

PubMed Abstract | Crossref Full Text | Google Scholar

138. Kajiwara Y, Tazawa H, Yamada M, Kanaya N, Fushimi T, Kikuchi S, et al. Oncolytic virus-mediated reducing of myeloid-derived suppressor cells enhances the efficacy of PD-L1 blockade in gemcitabine-resistant pancreatic cancer. Cancer Immunol Immunother. (2023) 72:1285–300. doi: 10.1007/s00262-022-03334-x

PubMed Abstract | Crossref Full Text | Google Scholar

139. Esposito A, Agostini A, Quero G, Piro G, Priori L, Caggiano A, et al. Colorectal cancer patients-derived immunity-organoid platform unveils cancer-specific tissue markers associated with immunotherapy resistance. Cell Death Dis. (2024) 15:878. doi: 10.1038/s41419-024-07266-5

PubMed Abstract | Crossref Full Text | Google Scholar

140. Clubb JHA, Kudling TV, Heiniö C, Basnet S, Pakola S, Cervera Carrascón V, et al. Adenovirus encoding tumor necrosis factor alpha and interleukin 2 induces a tertiary lymphoid structure signature in immune checkpoint inhibitor refractory head and neck cancer. Front Immunol. (2022) 13:794251. doi: 10.3389/fimmu.2022.794251

PubMed Abstract | Crossref Full Text | Google Scholar

141. Chow A, Perica K, Klebanoff CA, and Wolchok JD. Clinical implications of T cell exhaustion for cancer immunotherapy. Nat Rev Clin Oncol. (2022) 19:775–90. doi: 10.1038/s41571-022-00689-z

PubMed Abstract | Crossref Full Text | Google Scholar

142. Gao J, Shi LZ, Zhao H, Chen J, Xiong L, He Q, et al. Loss of IFN-γ Pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell. (2016) 167:397–404.e9. doi: 10.1016/j.cell.2016.08.069

PubMed Abstract | Crossref Full Text | Google Scholar

143. Platanias LC. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev Immunol. (2005) 5:375–86. doi: 10.1038/nri1604

PubMed Abstract | Crossref Full Text | Google Scholar

144. Kanaya N, Kuroda S, Kakiuchi Y, Kumon K, Tsumura T, Hashimoto M, et al. Immune modulation by telomerase-specific oncolytic adenovirus synergistically enhances antitumor efficacy with anti-PD1 antibody. Mol Ther. (2020) 28:794–804. doi: 10.1016/j.ymthe.2020.01.003

PubMed Abstract | Crossref Full Text | Google Scholar

145. Kuryk L, Moller AW, and Jaderberg M. Combination of immunogenic oncolytic adenovirus ONCOS-102 with anti-PD-1 pembrolizumab exhibits synergistic antitumor effect in humanized A2058 melanoma huNOG mouse model. Oncoimmunology. (2019) 8:e1532763. doi: 10.1080/2162402X.2018.1532763

PubMed Abstract | Crossref Full Text | Google Scholar

146. Li X, Lu M, Yuan M, Ye J, Zhang W, Xu L, et al. CXCL10-armed oncolytic adenovirus promotes tumor-infiltrating T-cell chemotaxis to enhance anti-PD-1 therapy. Oncoimmunology. (2022) 11:2118210. doi: 10.1080/2162402X.2022.2118210

PubMed Abstract | Crossref Full Text | Google Scholar

147. Cervera-Carrascon V, Quixabeira DCA, Santos JM, Havunen R, Milenova I, Verhoeff J, et al. Adenovirus Armed With TNFa and IL2 Added to aPD-1 Regimen Mediates Antitumor Efficacy in Tumors Refractory to aPD-1. Front Immunol. (2021) 12:706517. doi: 10.3389/fimmu.2021.706517

PubMed Abstract | Crossref Full Text | Google Scholar

148. Kudling TV, Clubb JHA, Pakola S, Quixabeira DCA, Lähdeniemi IAK, Heiniö C, et al. Effective intravenous delivery of adenovirus armed with TNFα and IL-2 improves anti-PD-1 checkpoint blockade in non-small cell lung cancer. Oncoimmunology. (2023) 12:2241710. doi: 10.1080/2162402X.2023.2241710

PubMed Abstract | Crossref Full Text | Google Scholar

149. Cervera-Carrascon V, Quixabeira DCA, Santos JM, Havunen R, Zafar S, Hemminki O, et al. Tumor microenvironment remodeling by an engineered oncolytic adenovirus results in improved outcome from PD-L1 inhibition. Oncoimmunology. (2020) 9:1761229. doi: 10.1080/2162402X.2020.1761229

PubMed Abstract | Crossref Full Text | Google Scholar

150. Cervera-Carrascon V, Siurala M, Santos JM, Havunen R, Tahtinen S, Karell P, et al. TNFa and IL-2 armed adenoviruses enable complete responses by anti-PD-1 checkpoint blockade. Oncoimmunology. (2018) 7:e1412902. doi: 10.1080/2162402X.2017.1412902

PubMed Abstract | Crossref Full Text | Google Scholar

151. Clubb JHA, Kudling TV, Girych M, Haybout L, Pakola S, Hamdan F, et al. Development of a Syrian hamster anti-PD-L1 monoclonal antibody enables oncolytic adenoviral immunotherapy modelling in an immunocompetent virus replication permissive setting. Front Immunol. (2023) 14:1060540. doi: 10.3389/fimmu.2023.1060540

PubMed Abstract | Crossref Full Text | Google Scholar

152. Hu HJ, Liang X, Li HL, Wang HY, Gu JF, Sun LY, et al. Enhanced anti-melanoma efficacy through a combination of the armed oncolytic adenovirus ZD55-IL-24 and immune checkpoint blockade in B16-bearing immunocompetent mouse model. Cancer Immunol Immunother. (2021) 70:3541–55. doi: 10.1007/s00262-021-02946-z

PubMed Abstract | Crossref Full Text | Google Scholar

153. Belcaid Z, Berrevoets C, Choi J, van Beelen E, Stavrakaki E, Pierson T, et al. Low-dose oncolytic adenovirus therapy overcomes tumor-induced immune suppression and sensitizes intracranial gliomas to anti-PD-1 therapy. Neurooncol Adv. (2020) 2:vdaa011. doi: 10.1093/noajnl/vdaa011

PubMed Abstract | Crossref Full Text | Google Scholar

154. Nassiri F, Patil V, Yefet LS, Singh O, Liu J, Dang RMA, et al. Oncolytic DNX-2401 virotherapy plus pembrolizumab in recurrent glioblastoma: a phase 1/2 trial. Nat Med. (2023) 29:1370–8. doi: 10.1038/s41591-023-02347-y

PubMed Abstract | Crossref Full Text | Google Scholar

155. Li R, Shah PH, Stewart TF, Nam JK, Bivalacqua TJ, Lamm DL, et al. Oncolytic adenoviral therapy plus pembrolizumab in BCG-unresponsive non-muscle-invasive bladder cancer: the phase 2 CORE-001 trial. Nat Med. (2024) 30:2216–23. doi: 10.1038/s41591-024-03025-3

PubMed Abstract | Crossref Full Text | Google Scholar

156. Hamdan F, Ylosmaki E, Chiaro J, Giannoula Y, Long M, Fusciello M, et al. Novel oncolytic adenovirus expressing enhanced cross-hybrid IgGA Fc PD-L1 inhibitor activates multiple immune effector populations leading to enhanced tumor killing in vitro, in vivo and with patient-derived tumor organoids. J Immunother Cancer. (2021) 9:e003000. doi: 10.1136/jitc-2021-003000

PubMed Abstract | Crossref Full Text | Google Scholar

157. Meng Y, Liu H, Zhu H, Zhang W, Sun D, Han X, et al. RCAd-LTH-shPD-L1, a double-gene recombinant oncolytic adenovirus with enhanced antitumor immunity, increases lymphocyte infiltration and reshapes the tumor microenvironment. J Immunother Cancer. (2024) 12:e007171. doi: 10.1136/jitc-2023-007171

PubMed Abstract | Crossref Full Text | Google Scholar

158. Mei S, Peng S, Vong EG, and Zhan J. A dual-functional oncolytic adenovirus ZD55-aPD-L1 scFv armed with PD-L1 inhibitor potentiates its antitumor activity. Int Immunopharmacol. (2024) 128:111579. doi: 10.1016/j.intimp.2024.111579

PubMed Abstract | Crossref Full Text | Google Scholar

159. Higgins TA, Patton DJ, Shimko-Lofano IM, Eller TL, Molinari R, Sandey M, et al. The development and characterization of a next-generation oncolytic virus armed with an anti-PD-1 sdAb for osteosarcoma treatment in vitro. Cells. (2024) 13:351. doi: 10.3390/cells13040351

PubMed Abstract | Crossref Full Text | Google Scholar

160. Qiang L, Huili Z, Leilei Z, Xiaoyan W, Hui W, Biao H, et al. Intratumoral delivery of a Tim-3 antibody-encoding oncolytic adenovirus engages an effective antitumor immune response in liver cancer. J Cancer Res Clin Oncol. (2023) 149:18201–13. doi: 10.1007/s00432-023-05501-8

PubMed Abstract | Crossref Full Text | Google Scholar

161. Huang Y, Lv SQ, Liu PY, Ye ZL, Yang H, Li LF, et al. A SIRPα-Fc fusion protein enhances the antitumor effect of oncolytic adenovirus against ovarian cancer. Mol Oncol. (2020) 14:657–68. doi: 10.1002/1878-0261.12628

PubMed Abstract | Crossref Full Text | Google Scholar

162. Saffarzadeh N, Foord E, O'Leary E, Mahmoun R, Birkballe Hansen T, Levitsky V, et al. Inducing expression of ICOS-L by oncolytic adenovirus to enhance tumor-specific bi-specific antibody efficacy. J Transl Med. (2024) 22:250. doi: 10.1186/s12967-024-05049-2

PubMed Abstract | Crossref Full Text | Google Scholar

163. Ylösmäki E, Ylösmäki L, Fusciello M, Martins B, Ahokas P, Cojoc H, et al. Characterization of a novel OX40 ligand and CD40 ligand-expressing oncolytic adenovirus used in the PeptiCRAd cancer vaccine platform. Mol Ther Oncol. (2021) 20:459–69. doi: 10.1016/j.omto.2021.02.006

PubMed Abstract | Crossref Full Text | Google Scholar

164. Kuryk L, Mathlouthi S, Wieczorek M, Gad B, Rinner B, Malfanti A, et al. Priming with oncolytic adenovirus followed by anti-PD-1 and paclitaxel treatment leads to improved anti-cancer efficacy in the 3D TNBC model. Eur J Pharm Biopharm. (2024) 199:114300. doi: 10.1016/j.ejpb.2024.114300

PubMed Abstract | Crossref Full Text | Google Scholar

165. Garofalo M, Wieczorek M, Anders I, Staniszewska M, Lazniewski M, Prygiel M, et al. Novel combinatorial therapy of oncolytic adenovirus AdV5/3-D24-ICOSL-CD40L with anti PD-1 exhibits enhanced anti-cancer efficacy through promotion of intratumoral T-cell infiltration and modulation of tumour microenvironment in mesothelioma mouse model. Front Oncol. (2023) 13:1259314. doi: 10.3389/fonc.2023.1259314

PubMed Abstract | Crossref Full Text | Google Scholar

166. Wenthe J, Naseri S, Hellström AC, Moreno R, Ullenhag G, Alemany R, et al. Immune priming using DC- and T cell-targeting gene therapy sensitizes both treated and distant B16 tumors to checkpoint inhibition. Mol Ther Oncol. (2022) 24:429–42. doi: 10.1016/j.omto.2022.01.003

PubMed Abstract | Crossref Full Text | Google Scholar

167. Nakamura N, Sato-Dahlman M, Travis E, Jacobsen K, and Yamamoto M. CDX2 promoter-controlled oncolytic adenovirus suppresses tumor growth and liver metastasis of colorectal cancer. Cancer Sci. (2025). doi: 10.1111/cas.70063

PubMed Abstract | Crossref Full Text | Google Scholar

168. Musher BL, Rowinsky EK, Smaglo BG, Abidi W, Othman M, Patel K, et al. LOAd703, an oncolytic virus-based immunostimulatory gene therapy, combined with chemotherapy for unresectable or metastatic pancreatic cancer (LOKON001): results from arm 1 of a non-randomised, single-centre, phase 1/2 study. Lancet Oncol. (2024) 25:488–500. doi: 10.1016/S1470-2045(24)00079-2

PubMed Abstract | Crossref Full Text | Google Scholar

169. Ponce S, Cedrés S, Ricordel C, Isambert N, Viteri S, Herrera-Juarez M, et al. ONCOS-102 plus pemetrexed and platinum chemotherapy in Malignant pleural mesothelioma: a randomized phase 2 study investigating clinical outcomes and the tumor microenvironment. J Immunother Cancer. (2023) 11:e007552. doi: 10.1136/jitc-2023-007552

PubMed Abstract | Crossref Full Text | Google Scholar

170. Pokrovska TD, Jacobus EJ, Puliyadi R, Prevo R, Frost S, Dyer A, et al. External beam radiation therapy and enadenotucirev: inhibition of the DDR and mechanisms of radiation-mediated virus increase. Cancers (Basel). (2020) 12:798. doi: 10.3390/cancers12040798

PubMed Abstract | Crossref Full Text | Google Scholar

171. Xu C, Chen L, Liu G, Xu J, Lv W, Gao X, et al. Tailoring an intravenously injectable oncolytic virus for augmenting radiotherapy. Cell Rep Med. (2025) 6:102078. doi: 10.1016/j.xcrm.2025.102078

PubMed Abstract | Crossref Full Text | Google Scholar

172. Yan D, Li G, Yuan Y, Li H, Cao H, Dai Y, et al. SOCS3 inhibiting JAK-STAT pathway enhances oncolytic adenovirus efficacy by potentiating viral replication and T-cell activation. Cancer Gene Ther. (2024) 31:397–409. doi: 10.1038/s41417-023-00710-2

PubMed Abstract | Crossref Full Text | Google Scholar

173. Patel MR, Jacobson BA, Ji Y, Drees J, Tang S, Xiong K, et al. Vesicular stomatitis virus expressing interferon-β is oncolytic and promotes antitumor immune responses in a syngeneic murine model of non-small cell lung cancer. Oncotarget. (2015) 6:33165–77. doi: 10.18632/oncotarget.5320

PubMed Abstract | Crossref Full Text | Google Scholar

174. Patel MR, Dash A, Jacobson BA, Ji Y, Baumann D, Ismail K, et al. JAK/STAT inhibition with ruxolitinib enhances oncolytic virotherapy in non-small cell lung cancer models. Cancer Gene Ther. (2019) 26:411–8. doi: 10.1038/s41417-018-0074-6

PubMed Abstract | Crossref Full Text | Google Scholar

175. Stefani C, Miricescu D, Stanescu S, Nica RI, Greabu M, Totan AR, et al. Growth factors, PI3K/AKT/mTOR and MAPK signaling pathways in colorectal cancer pathogenesis: where are we now? Int J Mol Sci. (2021) 22:10260. doi: 10.3390/ijms221910260

PubMed Abstract | Crossref Full Text | Google Scholar

176. Fang L, Tian W, Zhang C, Wang X, Li W, Zhang Q, et al. Oncolytic adenovirus-mediated expression of CCL5 and IL12 facilitates CA9-targeting CAR-T therapy against renal cell carcinoma. Pharmacol Res. (2023) 189:106701. doi: 10.1016/j.phrs.2023.106701

PubMed Abstract | Crossref Full Text | Google Scholar

177. Fang S, Wu J, Liu Y, Wang P, Yuan G, Gao J, et al. CD70 CAR-T cells empowered by TS-2021 through ex vivo transduction show potent antitumor efficacy against glioblastoma. J Exp Clin Cancer Res. (2025) 44:173. doi: 10.1186/s13046-025-03431-6

PubMed Abstract | Crossref Full Text | Google Scholar

178. Wang G, Zhang Z, Zhong K, Wang Z, Yang N, Tang X, et al. CXCL11-armed oncolytic adenoviruses enhance CAR-T cell therapeutic efficacy and reprogram tumor microenvironment in glioblastoma. Mol Ther. (2023) 31:134–53. doi: 10.1016/j.ymthe.2022.08.021

PubMed Abstract | Crossref Full Text | Google Scholar

179. Li X, Zhang Y, Mao Z, Zhao H, Cao H, Wang J, et al. Decorin-armed oncolytic adenovirus promotes natural killers (NKs) activation and infiltration to enhance NK therapy in CRC model. Mol BioMed. (2024) 5:48. doi: 10.1186/s43556-024-00212-z

PubMed Abstract | Crossref Full Text | Google Scholar

180. Zafar S, Sorsa S, Siurala M, Hemminki O, Havunen R, Cervera-Carrascon V, et al. CD40L coding oncolytic adenovirus allows long-term survival of humanized mice receiving dendritic cell therapy. Oncoimmunology. (2018) 7:e1490856. doi: 10.1080/2162402X.2018.1490856

PubMed Abstract | Crossref Full Text | Google Scholar

181. Wang J, Zhu Y, Chen Y, Huang Y, Guo Q, Wang Y, et al. Three-in-one oncolytic adenovirus system initiates a synergetic photodynamic immunotherapy in immune-suppressive cholangiocarcinoma. Small. (2023) 19:e2207668. doi: 10.1002/smll.202207668

PubMed Abstract | Crossref Full Text | Google Scholar

182. Ma J, Zhang C, Shi G, Yue D, Shu Y, Hu S, et al. High-dose VitC plus oncolytic adenoviruses enhance immunogenic tumor cell death and reprogram tumor immune microenvironment. Mol Ther. (2022) 30:644–61. doi: 10.1016/j.ymthe.2021.09.015

PubMed Abstract | Crossref Full Text | Google Scholar

183. Nguyen H-M, Bommareddy PK, Silk AW, and Saha D. Optimal timing of PD-1 blockade in combination with oncolytic virus therapy. Semin Cancer Biol. (2022) 86:971–80. doi: 10.1016/j.semcancer.2021.05.019

PubMed Abstract | Crossref Full Text | Google Scholar

184. Chesney J, Puzanov I, Collichio F, Singh P, Milhem MM, Glaspy J, et al. Randomized, open-label phase II study evaluating the efficacy and safety of talimogene laherparepvec in combination with ipilimumab versus ipilimumab alone in patients with advanced, unresectable melanoma. J Clin Oncol. (2018) 36:1658–67. doi: 10.1200/JCO.2017.73.7379

PubMed Abstract | Crossref Full Text | Google Scholar

185. Chesney JA, Ribas A, Long GV, Kirkwood JM, Dummer R, Puzanov I, et al. Randomized, double-blind, placebo-controlled, global phase III trial of talimogene laherparepvec combined with pembrolizumab for advanced melanoma. J Clin Oncol. (2023) 41:528–40. doi: 10.1200/JCO.22.00343

PubMed Abstract | Crossref Full Text | Google Scholar

186. Barber GN. Host defense, viruses and apoptosis. Cell Death Differ. (2001) 8:113–26. doi: 10.1038/sj.cdd.4400823

PubMed Abstract | Crossref Full Text | Google Scholar

187. Parker BS, Rautela J, and Hertzog PJ. Antitumour actions of interferons: implications for cancer therapy. Nat Rev Cancer. (2016) 16:131–44. doi: 10.1038/nrc.2016.14

PubMed Abstract | Crossref Full Text | Google Scholar

188. Gujar S, Pol JG, Kim Y, Lee PW, and Kroemer G. Antitumor benefits of antiviral immunity: an underappreciated aspect of oncolytic virotherapies. Trends Immunol. (2018) 39:209–21. doi: 10.1016/j.it.2017.11.006

PubMed Abstract | Crossref Full Text | Google Scholar

189. Wang S, Yang X, Ma YY, Wu J, Jin K, Zhao R, et al. An engineered self-biomineralized oncolytic adenovirus induces effective antitumor immunity and synergizes with immune checkpoint blockade. Cancer Immunol Res. (2024) 12:1640–54. doi: 10.1158/2326-6066.CIR-23-0957

PubMed Abstract | Crossref Full Text | Google Scholar

190. Huang H, Liu M, Sun M, Duan S, Pan S, Liu P, et al. Virus-protein corona replacement strategy to improve the antitumor efficacy of intravenously injected oncolytic adenovirus. ACS Nano. (2023) 17:14461–74. doi: 10.1021/acsnano.3c00847

PubMed Abstract | Crossref Full Text | Google Scholar

191. Brown MC, Holl EK, Boczkowski D, Dobrikova E, Mosaheb M, Chandramohan V, et al. Cancer immunotherapy with recombinant poliovirus induces IFN-dominant activation of dendritic cells and tumor antigen-specific CTLs. Sci Transl Med. (2017) 9:eaan4220. doi: 10.1126/scitranslmed.aan4220

PubMed Abstract | Crossref Full Text | Google Scholar

192. Swain SL, McKinstry KK, and Strutt TM. Expanding roles for CD4+ T cells in immunity to viruses. Nat Rev Immunol. (2012) 12:136–48. doi: 10.1038/nri3152

PubMed Abstract | Crossref Full Text | Google Scholar

193. Ricca JM, Oseledchyk A, Walther T, Liu C, Mangarin L, Merghoub T, et al. Pre-existing immunity to oncolytic virus potentiates its immunotherapeutic efficacy. Mol Ther. (2018) 26:1008–19. doi: 10.1016/j.ymthe.2018.01.019

PubMed Abstract | Crossref Full Text | Google Scholar

194. Ju F, Luo Y, Lin C, Jia X, Xu Z, Tian R, et al. Oncolytic virus expressing PD-1 inhibitors activates a collaborative intratumoral immune response to control tumor and synergizes with CTLA-4 or TIM-3 blockade. J Immunother Cancer. (2022) 10:e004762. doi: 10.1136/jitc-2022-004762

PubMed Abstract | Crossref Full Text | Google Scholar

195. Taipale K, Liikanen I, Juhila J, Turkki R, Tähtinen S, Kankainen M, et al. Chronic activation of innate immunity correlates with poor prognosis in cancer patients treated with oncolytic adenovirus. Mol Ther. (2016) 24:175–83. doi: 10.1038/mt.2015.143

PubMed Abstract | Crossref Full Text | Google Scholar

196. Liikanen I, Basnet S, Quixabeira DCA, Taipale K, Hemminki O, Oksanen M, et al. Oncolytic adenovirus decreases the proportion of TIM-3(+) subset of tumor-infiltrating CD8(+) T cells with correlation to improved survival in patients with cancer. J Immunother Cancer. (2022) 10:e003490. doi: 10.1136/jitc-2021-003490

PubMed Abstract | Crossref Full Text | Google Scholar

197. Hastie E, Cataldi M, Moerdyk-Schauwecker MJ, Felt SA, Steuerwald N, and Grdzelishvili VZ. Novel biomarkers of resistance of pancreatic cancer cells to oncolytic vesicular stomatitis virus. Oncotarget. (2016) 7:61601–18. doi: 10.18632/oncotarget.11202

PubMed Abstract | Crossref Full Text | Google Scholar

198. Ishino R, Kawase Y, Kitawaki T, Sugimoto N, Oku M, Uchida S, et al. Oncolytic virus therapy with HSV-1 for hematological Malignancies. Mol Ther. (2021) 29:762–74. doi: 10.1016/j.ymthe.2020.09.041

PubMed Abstract | Crossref Full Text | Google Scholar

199. Chen X, Liu J, Li Y, Zeng Y, Wang F, Cheng Z, et al. IDH1 mutation impairs antiviral response and potentiates oncolytic virotherapy in glioma. Nat Commun. (2023) 14:6781. doi: 10.1038/s41467-023-42545-3

PubMed Abstract | Crossref Full Text | Google Scholar

200. Zloza A, Kim DW, Kim-Schulze S, Jagoda MC, Monsurro V, Marincola FM, et al. Immunoglobulin-like transcript 2 (ILT2) is a biomarker of therapeutic response to oncolytic immunotherapy with vaccinia viruses. J Immunother Cancer. (2014) 2:1. doi: 10.1186/2051-1426-2-1

PubMed Abstract | Crossref Full Text | Google Scholar

201. Wang X, Tian H, Chi Z, Si L, Sheng X, Hu H, et al. Oncolytic virus OH2 extends survival in patients with PD-1 pretreated melanoma: phase Ia/Ib trial results and biomarker insights. J Immunother Cancer. (2025) 13:e010662. doi: 10.1136/jitc-2024-010662

PubMed Abstract | Crossref Full Text | Google Scholar

202. Pakola SA, Clubb JHA, Kudling TV, van der Heijden M, Jirovec E, Arias V, et al. Transient lymphocyte count decrease correlates with oncolytic adenovirus efficacy in humans: mechanistic and biomarker findings from TUNIMO phase I trial. J Immunother Cancer. (2025) 13:e010493. doi: 10.1136/jitc-2024-010493

PubMed Abstract | Crossref Full Text | Google Scholar

203. Liikanen I, Koski A, Merisalo-Soikkeli M, Hemminki O, Oksanen M, Kairemo K, et al. Serum HMGB1 is a predictive and prognostic biomarker for oncolytic immunotherapy. Oncoimmunology. (2015) 4:e989771. doi: 10.4161/2162402X.2014.989771

PubMed Abstract | Crossref Full Text | Google Scholar

204. Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. (2015) 350:1079–84. doi: 10.1126/science.aad1329

PubMed Abstract | Crossref Full Text | Google Scholar

205. Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. (2018) 359:91–7. doi: 10.1126/science.aan3706

PubMed Abstract | Crossref Full Text | Google Scholar

206. Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. (2018) 359:104–8. doi: 10.1126/science.aao3290

PubMed Abstract | Crossref Full Text | Google Scholar

207. Meléndez-Vázquez NM, Nguyen TT, Fan X, López-Rivas AR, Fueyo J, Gomez-Manzano C, et al. Gut microbiota composition is associated with the efficacy of Delta-24-RGDOX in Malignant gliomas. Mol Ther Oncol. (2024) 32:200787. doi: 10.1016/j.omton.2024.200787

PubMed Abstract | Crossref Full Text | Google Scholar

208. Xie J, Liu M, Deng X, Tang Y, Zheng S, Ou X, et al. Gut microbiota reshapes cancer immunotherapy efficacy: Mechanisms and therapeutic strategies. Imeta. (2024) 3:e156. doi: 10.1002/imt2.156

PubMed Abstract | Crossref Full Text | Google Scholar

209. Yi J, Lin P, Li Q, Zhang A, and Kong X. A new strategy for treating colorectal cancer: Regulating the influence of intestinal flora and oncolytic virus on interferon. Mol Ther Oncol. (2023) 30:254–74. doi: 10.1016/j.omto.2023.08.010

PubMed Abstract | Crossref Full Text | Google Scholar

210. Lee WS, Lee SJ, Lee HJ, Yang H, Go EJ, Gansukh E, et al. Oral reovirus reshapes the gut microbiome and enhances antitumor immunity in colon cancer. Nat Commun. (2024) 15:9092. doi: 10.1038/s41467-024-53347-6

PubMed Abstract | Crossref Full Text | Google Scholar

211. Zhang H, Fu L, Leiliang X, Qu C, Wu W, Wen R, et al. Beyond the Gut: The intratumoral microbiome’s influence on tumorigenesis and treatment response. Cancer Commun (Lond). (2024) 44:1130–67. doi: 10.1002/cac2.12597

PubMed Abstract | Crossref Full Text | Google Scholar

212. Zhang T, Li Y, Zhai E, Zhao R, Qian Y, Huang Z, et al. Intratumoral fusobacterium nucleatum recruits tumor-associated neutrophils to promote gastric cancer progression and immune evasion. Cancer Res. (2025) 85:1819–41. doi: 10.1158/0008-5472.CAN-24-2580

PubMed Abstract | Crossref Full Text | Google Scholar

213. Bourgeois-Daigneault MC, Roy DG, Aitken AS, El Sayes N, Martin NT, Varette O, et al. Neoadjuvant oncolytic virotherapy before surgery sensitizes triple-negative breast cancer to immune checkpoint therapy. Sci Transl Med. (2018) 10:eaao1641. doi: 10.1126/scitranslmed.aao1641

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: oncolytic adenovirus, immune checkpoint inhibitors, drug resistance, tumor microenvironment, immunotherapy

Citation: Cheng C, Wang Q and Zhang S (2025) Synergy of oncolytic adenovirus and immune checkpoint inhibitors: transforming cancer immunotherapy paradigms. Front. Immunol. 16:1610858. doi: 10.3389/fimmu.2025.1610858

Received: 13 April 2025; Accepted: 23 June 2025;
Published: 08 July 2025.

Edited by:

Xing-xing Fan, Macau University of Science and Technology, Macao, China

Reviewed by:

Fabian Benencia, Ohio University, United States
Santenna Chenchula, All India Institute of Medical Sciences, Bhopal, India

Copyright © 2025 Cheng, Wang and Zhang. 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: Shuang Zhang, c2h1YW5nLnpoYW5nQHNjdS5lZHUuY24=

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.