Next Article in Journal
Ensuring Reproducibility and Deploying Models with the Image2Radiomics Framework: An Evaluation of Image Processing on PanNET Model Performance
Previous Article in Journal
Integration of an OS-Based Machine Learning Score (AS Score) and Immunoscore as Ancillary Tools for Predicting Immunotherapy Response in Sarcomas
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Oncolytic Therapies for Glioblastoma: Advances, Challenges, and Future Perspectives

1
Hamidiye International School of Medicine, University of Health Sciences, 34668 Istanbul, Türkiye
2
Faculty of Medicine, Baskent University, 06790 Ankara, Türkiye
3
Department of Medical Oncology, Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, 34865 Istanbul, Turkey
4
School of Medicine, Yarmouk University, Irbid 21163, Jordan
5
Department of Neurosurgery, University of Wisconsin School of Medicine, Madison, WI 53706, USA
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(15), 2550; https://doi.org/10.3390/cancers17152550 (registering DOI)
Submission received: 26 June 2025 / Revised: 25 July 2025 / Accepted: 28 July 2025 / Published: 1 August 2025
(This article belongs to the Section Cancer Immunology and Immunotherapy)

Simple Summary

Glioblastoma (GBM) is a highly aggressive brain tumor with limited treatment options, prompting interest in oncolytic therapies, particularly oncolytic viruses (OVs), that can destroy tumor cells and activate immune responses. Promising viral candidates include modified herpes simplex virus, adenovirus, poliovirus, and Newcastle disease virus, each offering tumor-selective advantages. While some OV therapies like G207 and Delytact™ have shown clinical potential, challenges such as immune resistance, delivery obstacles, and patient variability remain. Combining OVs with other treatments, such as immune checkpoint inhibitors or chemotherapy, offers potential but needs further validation. Additionally, non-viral oncolytic agents, like tumor-targeting bacteria, have been emerging, yet they are underexplored. To move forward, research must focus on improving OV delivery, tailoring treatments using biomarkers, and addressing the immunosuppressive tumor environment.

Abstract

Glioblastoma (GBM) remains one of the most aggressive and treatment-resistant brain tumors, necessitating novel therapeutic approaches. Oncolytic treatments, particularly oncolytic viruses (OVs), have emerged as promising candidates by selectively infecting and lysing tumor cells while stimulating anti-tumor immunity. Various virus-based therapies are under investigation, including genetically engineered herpes simplex virus (HSV), adenovirus, poliovirus, reovirus, vaccinia virus, measles virus, and Newcastle disease virus, each exploiting unique tumor-selective mechanisms. While some, such as HSV-based therapies including G207 and DelytactTM, have demonstrated clinical progress, significant challenges persist, including immune evasion, heterogeneity in patient response, and delivery barriers due to the blood–brain barrier. Moreover, combination strategies integrating OVs with immune checkpoint inhibitors, chemotherapy, and radiation are promising but require further clinical validation. Non-viral oncolytic approaches, such as tumor-targeting bacteria and synthetic peptides, remain underexplored. This review highlights current advancements while addressing critical gaps in the literature, including the need for optimized delivery methods, better biomarker-based patient stratification, and a deeper understanding of GBM’s immunosuppressive microenvironment. Future research should focus on enhancing OV specificity, engineering viruses to deliver therapeutic genes, and integrating OVs with precision medicine strategies. By identifying these gaps, this review provides a framework for advancing oncolytic therapies in GBM treatment.

1. Introduction

Glioblastoma (GBM) is the most frequent primary malignant brain tumor in adults, representing nearly half of all primary central nervous system (CNS) cancers. Its estimated annual incidence is around 3 cases per 100,000 individuals [1]. GBMs usually arise de novo and are characterized by aggressive local invasion into surrounding brain tissue, though they rarely metastasize to other organs [2]. To date, the only well-established GBM risk factor is exposure of the CNS to ionizing radiation [3]. The current standard of care includes maximal safe surgical resection, followed by radiotherapy in combination with concurrent and adjuvant temozolomide chemotherapy [4]. Despite current treatments, the prognosis for GBM patients remains extremely poor, with fewer than 3% surviving five years post-diagnosis, with older age being the most consistent predictor of unfavorable outcomes [5].
Vascular endothelial growth factor (VEGF) is markedly overexpressed in GBM, driving abnormal neovascularization and aggressive tumor growth. Consequently, anti-VEGF therapies have emerged as important adjuncts in GBM management by targeting the tumor’s angiogenic dependency [6]. Among these, bevacizumab received accelerated approval from the FDA in 2009 for the treatment of recurrent GBM following prior therapy, supported by phase II trial data demonstrating meaningful clinical benefits, including improved 6-month progression-free survival, enhanced radiographic and neurologic responses, and reduced cerebral edema, despite no significant improvement in overall survival [7]. Additionally, in one cohort, bevacizumab significantly reduced corticosteroid requirements in patients receiving steroids at baseline, with 83.3% achieving more than a 50% dose reduction, reflecting improved peritumoral edema control and contributing to better quality of life [8]. Overall, bevacizumab remains a valuable adjunct in GBM management for improving disease control and symptom burden, despite the absence of an overall survival advantage.
Even with first-line therapies, GBM almost invariably recurs, with median overall survival after recurrence ranging from 2 to 9 months [9]. This dismal outlook is largely attributable to several challenges, including significant intertumoral and intratumoral heterogeneity that promotes the emergence of resistant cell populations; tumors in difficult anatomical locations shielded by the blood–brain barrier (BBB), thus limiting effective drug delivery; and an immunosuppressive tumor microenvironment [10]. Additionally, GBM is classified as a “cold tumor” because of its immunosuppressive environment and weak T-cell response. As a result, immune checkpoint immunotherapy and chimeric antigen receptor (CAR) T-cell therapy have shown limited success for GBM treatment [5,10].
Traditional treatments are unable to surmount these obstacles, underscoring the urgent need for innovative therapies [11]. Oncolytic virotherapy (OVT) represents a promising new personalized approach for addressing these GBM treatment challenges.
Viruses and bacteria, once viewed as harmful, are now recognized as powerful allies for GBM therapy. Their diversity, selectivity, and manipulability enable them to replicate within to destroy tumor cells while leaving normal cells undamaged, achieving what conventional treatments often fail to accomplish [4,12]. Oncolytic viruses (OVs) have been the focus of many experimental GMB studies due to their compatibility with the brain’s unique microenvironment, the absence of distant metastasis with GBM, and the tumor’s rapid cell division, which facilitates viral replication [10,13,14,15,16] (Figure 1).
OVs exert antitumor effects through both direct cytolytic mechanisms and immune-mediated responses. Direct oncolysis involves the selective replication of viruses within tumor cells, leading to cell rupture and local inflammation [17]. More critically, OVs initiate a robust antitumor immune response by releasing damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) during tumor cell lysis [18,19]. These molecular signals stimulate the production of type I interferons (IFNs) and other proinflammatory cytokines, activating dendritic cells, macrophages, and other antigen-presenting cells (APCs), which in turn prime tumor- and virus-specific CD4+ and CD8+ T cells [19]. This dual stimulation of innate and adaptive immunity is believed to overcome the immunologic tolerance that characterizes GBM, effectively transforming it from a “cold” to a “hot” tumor [19]. This pleiotropic immune activation has been associated with improved survival in subsets of GBM patients.
These mechanisms highlight the novel and promising potential of OVT for GBM treatment by effectively addressing many of the limitations of traditional therapies. By overcoming obstacles including the tumor’s complex anatomical location, extensive heterogeneity, BBB infiltration, microscopic invasion, and immunosuppressive microenvironment, OVT offers a new therapeutic avenue [10].
Encouraging outcomes from both preclinical and clinical studies, most notably the conditional approval of the oncolytic virus G47∆ (Delytact) in Japan for brain tumors, further reinforce the promise of this approach, making it more acceptable among physicians [16]. This growing success of OVT marks a potentially game-changing development for GBM treatment, bringing renewed optimism in the fight against this highly aggressive and lethal disease.
This review summarizes the current landscape of oncolytic therapies for glioblastoma, encompassing both viral and non-viral approaches. It examines the underlying mechanisms and clinical progress of key oncolytic viruses, including Herpes Simplex Virus [HSV], Adenovirus, Poliovirus, and Reovirus. We also highlight combination therapies that are designed to enhance OVT efficacy. And finally, we explore emerging non-viral alternatives and outline future directions in the field by highlighting innovations in delivery systems, biomarker-guided patient selection, and the integration of oncolytic agents with targeted therapies.

2. Virus-Based Oncolytic Therapies

Multiple viral platforms, including adenovirus, HSV, reovirus, and measles virus, have been investigated for their unique mechanisms of action and tumor-targeting capabilities in preclinical GBM models. These approaches leverage both direct oncolysis and the modulation of the tumor microenvironment to reduce tumor burden and enhance radiosensitivity, providing multifaceted in vivo treatment strategies as illustrated in Figure 2.

2.1. Herpes Simplex Virus

Herpesviruses are double-stranded DNA viruses, with nine types known to infect humans, most notably herpes simplex virus 1 (HSV-1) and HSV-2. These viruses can establish lifelong latent infections, particularly in neural tissues, and may reactivate under immunosuppressive conditions, leading to lytic infections and associated clinical complications [20]. During lytic infection, numerous viral gene products are expressed, whereas latent infection is characterized by limited gene expression [21,22]. HSV encodes over 70 genes, and its interaction with host cells plays a crucial role in controlling both latency and reactivation. Since HSV can fuse with cell membranes and enter host cells using specific receptor-ligand interactions and selectively lyse tumor cells following tumor cell-specific virus replication, it holds strong promise for oncolytic therapy.
Herpes simplex virus type 1 presents a highly favorable candidate for oncolytic virotherapy, owing to its unique biological features and well-established clinical profile. With a large genome (~152 kb), HSV-1 contains non-essential sequences that can be safely removed to accommodate therapeutic genes without compromising viral replication. Importantly, since the viral DNA does not integrate into the host genome, the risk of insertional mutagenesis is significantly minimized [23]. Although HSV-1 is extensively characterized as a human pathogen from a clinical perspective, its use as an oncolytic virus required substantial genetic engineering to attenuate its neurovirulence and enhance tumor selectivity. These modifications have enabled HSV-1 derivatives, such as G207 and M032, to safely and effectively target malignant cells while sparing healthy tissue. The availability of approved antiviral therapies, such as acyclovir, offers a crucial safety net to manage potential off-target viral activity [24]. Preclinical mouse studies have shown that oncolytic HSV-1 selectively infects and destroys tumor cells without damaging healthy tissues. In addition to direct tumor cell lysis, this process also triggers an antitumor immune response, which promotes anti-tumor immunity (immunovirotherapy) [25]. This dual effect of HSV-1—oncolysis and immune activation—makes it a promising candidate for oncolytic virotherapy. To date, numerous viruses have been genetically modified as oncolytic agents and tested in clinical trials against various cancer types, including GBM. Among these, HSV-1-based Talimogene laherparepvec (T-VEC) remains the only oncolytic virotherapy agent approved for the treatment of advanced melanoma by both the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) [15].
T-VEC is a product of the deletion of the γ34.5 virulence gene, which normally blocks the PKR pathway, and the US12 gene, which inhibits antigen presentation in infected cells. HSV-1 preserves protein synthesis during infection by preventing the PKR-mediated phosphorylation of eIF2α through its ICP34.5 protein, thereby counteracting the host antiviral response, while ICP 47 suppresses immune response by preventing antigen presentation via MHC class I molecules [26,27,28]. Additionally, the US 11 protein counteracts host cell protein synthesis shutdown, thereby overcoming viral suppression. This simultaneous alteration of gene dynamics finely adjusts the virulence of oHSV-1, enabling it to replicate in cancer cells that are moderately resistant.
In the face of these challenges, GBM stands out as a promising candidate for oncolytic virotherapy. The neuro-attenuated oHSV1 vector is currently being tested in both human and non-human studies.

2.1.1. Herpes Simplex Virus Oncolysis in Lab and Animal Studies

A broad array of genetically modified oncolytic herpes simplex viruses (oHSVs) has demonstrated promising antitumor efficacy in preclinical GBM models (Table 1) [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56]. Variants engineered to express immune-modulating cytokines (e.g., IL-2, IL-12) or tumor suppressors (e.g., PTEN isoforms) enhanced immune cell infiltration, reversed immunosuppressive signaling pathways such as IL6/STAT3 and PI3K/AKT, and overcame resistance to radiotherapy and checkpoint inhibition. Tumor-targeted constructs, such as HER2-, EGFRvIII-, and CXCR4-specific oHSVs, showed selective cytotoxicity against glioma stem-like cells (GSCs) and improved survival, particularly in early-stage disease. Several approaches addressed the intrinsic resistance of GSCs to viral replication by targeting pathways like TNF-α, TGF-β, JNK, and STAT3 using pharmacologic inhibitors or adjunctive methods such as fasting. Combinatorial strategies, including oHSV with CAR-T or NK cells, MEK inhibitors, bortezomib, or extracellular matrix-degrading enzymes (e.g., ChaseM), yielded synergistic effects, enhancing viral spread, immune activation, and tumor regression. Additionally, modifying the tumor microenvironment (such as blocking HMGB1 to reduce edema or degrading chondroitin sulfate proteoglycans to improve penetration) was crucial for therapeutic success. These findings suggest that future efforts should focus on personalized oHSV design based on tumor profiles, rational combinations with immunotherapies or targeted agents, and innovative delivery strategies to sustain viral activity and remodel the tumor milieu. Translation into early-phase clinical trials with biomarker-driven stratification will be essential to realize the full potential of oHSV-based therapies in GBM.

2.1.2. Herpes Simplex Virus Oncolysis in Clinical Studies

Recent clinical trials exploring oHSV therapy for GBM have produced encouraging early-phase results, highlighting both safety and therapeutic potential (Table 2) [57,58,59,60,61]. Various engineered HSV strains, including G207, G47∆, M032, and CAN-3110, were tested across adult and pediatric populations with relapsed or treatment-resistant GBM. These studies demonstrated that oHSV therapy is generally well tolerated, with manageable side effects and evidence of immune activation within the tumor microenvironment. In several cases, radiographic and histological improvements were observed, along with extended survival in select patients.
Importantly, these trials revealed that oHSV can convert immunologically “cold” tumors into “hot” ones by enhancing lymphocyte infiltration and triggering immune-related gene expression. Pre-existing HSV immunity also appeared to enhance therapeutic response in some patients, suggesting a possible role for personalized approaches in future applications. Despite variability in survival outcomes and adverse events, the overall safety and immunostimulatory effects of oHSV support its continued investigation.
Future trials should focus on optimizing viral engineering, improving delivery methods, and combining oHSV with treatments such as radiotherapy or immunotherapy. Larger, controlled studies are needed to confirm efficacy and guide its integration into clinical practice. Ultimately, oHSV represents a promising frontier in the treatment of GBM, with potential to overcome many limitations of current therapeutic options.

2.2. Adenovirus

Adenoviruses (AdVs) are non-enveloped, double-stranded DNA viruses from the Adenoviridae family, with over 100 identified serotypes, 49 of which infect humans [62,63]. These viruses are highly prevalent worldwide and can infect various organs including the respiratory tract, gastrointestinal system, kidneys, and conjunctiva [62]. While most infections are asymptomatic or mild, AdV can cause severe complications including hemorrhagic cystitis, pancreatitis, and meningoencephalitis, especially in immunocompromised individuals. Transmission occurs via respiratory droplets, direct contact, or the fecal-oral route [64]. AdVs display organ-specific tropism based on serotype, with infection severity and location often influenced by the entry route. These viruses can remain latent in tonsils, lymphocytes, and other tissues. Their 26 to 45 kbp genome contains early, intermediate, and late transcription regions responsible for replication and viral gene expression [65,66]. With an icosahedral capsid structure and significant dependence on host cells for replication, AdVs have both lytic and latent phases and possess oncogenic potential, particularly through E1A-mediated transformation in experimental models.
Adenoviruses have recently gained attention as oncolytic agents for cancer immunotherapy due to their large gene-carrying capacity and tumor-selective replication. Oncolytic adenoviruses (oAds) destroy tumor cells directly through lysis and indirectly by stimulating immune responses, without harming healthy cells [67]. They can be genetically engineered to carry immunostimulatory genes or to target tumors with specific mutations such as p53 deficiency, as seen in Onyx-015 and H101 (Oncorine), which is approved in China for head and neck cancers [68]. These engineered viruses improve T cell activation, antigen presentation, and immune modulation. In addition, oAds show promise by disrupting the immunosuppressive tumor microenvironment and triggering immune-mediated GBM tumor cell destruction. By enhancing antigen release and activating cytotoxic T cells via DAMPs and PAMPs, oAds may offer a much-needed therapeutic GBM strategy, with ongoing studies exploring their clinical potential.

2.2.1. Adenovirus Oncolysis in Lab and Animal Studies

The summarized studies in Table 3 present evidence that genetically modified oAdVs hold significant promise for GBM treatment [69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105]. One of the most consistent findings across these models is that replication-competent oAdVs elicit stronger immune responses and better tumor control compared to non-replicating vectors. Key genetic modifications such as capsid engineering (e.g., Ad5/Ad37 fiber swaps), incorporation of immunostimulatory genes like IL-15, or the addition of tumor-targeting promoters (e.g., Ki67, hTERT) enable selective targeting of tumor cells and enhance antitumor immune activity. Some studies demonstrated that combining oAdVs with immune checkpoint inhibitors (e.g., anti-PD-1/PD-L1) or standard therapies (e.g., temozolomide or olaparib) further amplify therapeutic outcomes. Notably, carriers like mesenchymal stem cells improved viral delivery and immune evasion, and novel delivery routes such as intranasal administration increased survival in animal models, underscoring the importance of optimizing both vector design and delivery strategies.
Collectively, these findings suggest that future therapeutic directions should prioritize multi-modal strategies that integrate oAdVs with immunotherapy, targeted delivery systems, and tumor-specific promoters to enhance efficacy while minimizing off-target effects. The development of third-generation vectors (e.g., TS-2021) and exploration of underutilized serotypes (e.g., Ad6) illustrate ongoing innovations. Future studies should also explore personalized oAdV therapies based on patient-specific tumor profiles and immune landscapes.

2.2.2. Adenovirus Oncolysis in Clinical Studies

Published clinical studies highlight the growing promise of oAdvs as a therapeutic GBM modality (Table 4) [106,107,108,109,110]. Early-phase trials, such as Fares et al. using NSC-CRAd-S-pk7, demonstrated not only a manageable safety profile but also a median survival of 18.4 months which is considered an encouraging outcome for newly diagnosed patients [100]. Similarly, Delta24-RGD (DNX-2401), delivered via convection-enhanced delivery, showed complete tumor regression in one patient who survived for eight years, reflecting the potential for durable responses. Importantly, the addition of RGD motifs allowed viral entry despite the presence of neutralizing antibodies, broadening the applicability of these therapies even in pre-immunized individuals. Combinatorial approaches involving immune checkpoint inhibitors like pembrolizumab have also yielded promising results, with improved median survival (12.5 months) and no dose-limiting toxicities, suggesting synergy between viral oncolysis and immunotherapy.
Despite these advances, study sample sizes were small, limiting the generalizability of findings. Median survival varied widely between trials, likely reflecting differences in patient populations (e.g., newly diagnosed vs. recurrent GBM), delivery methods, and virus design. Furthermore, while oAdv therapy has shown immune activation and tumor responses, the variability in infusion area and delivery precision noticed in current studies underscores the need for optimized delivery protocols. Future studies should focus on larger, randomized trials that compare oAdv monotherapy and combination regimens with standard-of-care treatments. Moreover, integrating molecular profiling to select patients, most likely to benefit and understand mechanisms of resistance, will be crucial for advancing oAdv-based strategies into mainstream neuro-oncology practice.

2.3. Measles Viruses

The measles virus (MeV), a highly contagious negative-strand RNA virus from the Paramyxoviridae family, has shown promising oncolytic potential due to its natural tropism for tumor cells, ability to induce syncytia formation, and capacity to stimulate strong virus-specific immune responses [111,112]. The attenuated Edmonston strain (MV-Edm) has been genetically engineered and demonstrated to have durable responses in preclinical GBM models as well as good safety profiles in early-phase clinical trials (Table 5) [113,114,115,116,117,118]. However, antiviral resistance mechanisms in tumor cells remain a barrier. In one recent study, they concluded that MeV encoding miR-122 may have the potential to reduce target protein levels by 40%, though Drosha-dependent nuclear retention limited miRNA production [114]. Another study using patient-derived xenograft (PDX) models identified the JAK1 pathway as a key regulator of MeV replication, and combining MeV with the JAK1 inhibitor ruxolitinib improved viral replication and treatment efficacy, highlighting the need for further integration with immune-modulating strategies such as T-cell monitoring [115].

2.4. Newcastle Disease Virus

Newcastle Disease Virus (NDV), an avian paramyxovirus, has shown strong potential as a GBM oncolytic agent due to its ability to selectively replicate in tumor cells [119]. NDV induces cancer cell death through intrinsic and extrinsic caspase pathways, ER stress responses, and the release of TNF-α, while triggering robust inflammatory and interferon responses via its HN and F proteins [120]. GBM cells often lack effective type I interferon signaling, making them especially vulnerable to NDV-mediated oncolysis [121]. Recombinant NDV (rNDV) strains further enhance this effect, although preexisting immunity remains a challenge. Current strategies focus on improving efficacy through molecular modifications and combination therapies [122].
The preclinical studies indicate that NDV is a promising oncolytic virotherapy agent for GBM (Table 6) [121,123,124,125,126,127,128,129]. Although more research is needed for clinical application, combining NDV with Temozolomide enhances its oncolytic effects. The interferon pathway deficiency in glioblastoma cells allows for greater viral replication, which helps suppress tumor growth. Animal studies have also shown increased efficacy of recombinant NDV-PTEN. Additionally, using an antiviral serum against interferon viruses alongside NDV may still be effective in patients with existing virus immunity. Though clinical research on NDV in GBM is limited, existing studies suggest notable potential (Table 6). Further investigation is needed to optimize dosing, delivery timing (especially before anti-NDV antibodies develop), and to identify tumor profiles most responsive to therapy. NDV-infected tumor vaccines show strong peripheral immune activation, but understanding their effect within the tumor microenvironment is crucial, particularly in overcoming GBM’s immunosuppressive mechanisms. While no active clinical trials currently explore NDV for GBM, preclinical research remains active, supporting the need for future clinical studies to evaluate its safety and therapeutic impact.
NDV is well-known as a highly contagious and pathogenic virus in avian species, raising understandable regulatory, ecological, and zoonotic safety concerns regarding its use in human clinical settings. Despite this, extensive preclinical and clinical investigations have demonstrated that attenuated and recombinant NDV strains can be safely administered to patients, with no serious adverse events reported to date (Table 6). The use of NDV oncolytic virotherapy in several clinical trials has been presented as a safety strategy for treating various solid tumors [121,130], including melanoma [131], renal cell carcinoma [132], and ovarian cancer [133], among others. These studies collectively support a favorable safety profile under controlled conditions and adherence to strict biosafety measures and dosing protocols. Furthermore, the lack of preexisting immunity to NDV in humans and the virus’s selective replication in tumor cells deficient in type I interferon signaling provide a compelling rationale for its continued development as an oncolytic agent. Nevertheless, ongoing vigilance is necessary to monitor potential environmental and regulatory risks, and careful patient selection based on tumor interferon pathway status may enhance therapeutic efficacy while minimizing safety concerns.

2.5. Reovirus

Reovirus (Pelareorep), a wild-type Type 3 Dearing strain, has emerged as a promising oncolytic virotherapy for GBM treatment by exploiting the dysregulated Ras signaling pathway commonly found in malignant cells [13]. In normal cells, reovirus replication is blocked by an innate antiviral response mediated by protein kinase R (PKR), which inhibits viral protein synthesis [134]. However, in GBM cells with constitutively active Ras signaling, this PKR-mediated defense is impaired, allowing efficient viral replication, selective tumor cell lysis, and sparing of healthy tissue. Beyond direct oncolysis, reovirus infection promotes immunogenic cell death, releasing tumor-associated antigens and proinflammatory cytokines that stimulate systemic antitumor immunity. This dual mechanism (targeted cytotoxicity and immune activation) positions Pelareorep as a promising candidate against GBM, especially when combined with other immunotherapies or standard treatments to enhance efficacy and overcome resistance.
The current published literature demonstrates that Pelareorep-based reovirus therapy, alone or combined with GM-CSF, is generally safe and well tolerated across diverse glioma patient populations, including recurrent malignant gliomas, newly diagnosed GBM, and pediatric high-grade gliomas (Table 7) [135,136,137,138]. Viral delivery to tumor sites was consistently confirmed, with evidence of blood–brain barrier penetration and immunogenic effects including increased T cell activity and tumor cell death. However, while early biological and immunological engagement is promising, clinical efficacy remains limited, especially for pediatric high-grade gliomas where rapid tumor progression and antiviral antibody neutralization likely diminished therapeutic benefit. These findings suggest that future research should focus on strategies to enhance sustained viral activity and overcome immune neutralization, potentially through combination therapies or immune modulation, to translate these encouraging biological effects into meaningful clinical outcomes.

2.6. Retroviruses

Retroviruses, particularly replication-competent retroviruses (RCRs) and retroviral replicating vectors (RRVs), have emerged as promising agents in oncolytic virotherapy for GBM due to their ability to stably integrate therapeutic genes into tumor genomes. Preclinical studies have demonstrated the efficacy of various engineered retroviral vectors such as MLV-based RCRs, Toca 511, and modified foamy viruses in delivering suicide genes like cytosine deaminase or inducible caspase systems, leading to tumor-selective replication, sustained gene expression, and enhanced survival in animal models (Table 8) [139,140,141,142]. Clinical trials, especially those investigating Toca 511 in combination with the prodrug Toca FC, have shown favorable safety profiles and encouraging survival outcomes, with some patients achieving durable complete responses [142,143,144,145,146]. Molecular and immunologic analyses suggest immune-related mechanisms contribute to therapeutic benefit, reinforcing the potential of retrovirus-based strategies in personalized cancer therapy. Despite challenges such as variability in patient response and vector behavior, the accumulated evidence supports further development of retroviral platforms for effective, tumor-selective gene therapy in GBM.

2.7. Vaccinia Virus

Recent advancements in vaccinia virus (VV)-based oncolytic therapies highlight promising breakthroughs for GBM treatment. Several innovative VV constructs have demonstrated strong tumor selectivity, BBB penetration, and the ability to reshape the immunosuppressive tumor microenvironment. A new generation VV armed with interleukin-21 (IL-21) synergized with anti-PD1 therapy promoted durable immune memory to achieve long-term tumor control in murine models [147]. Another construct, ΔF4LΔJ2R VV, showed enhanced efficacy when combined with radiotherapy, curing the majority of treated mice and improving CD8+/Treg ratios [148]. VV-GMCSF-Lact, engineered to express GM-CSF and lactaptin, showed cytotoxicity against patient-derived GBM cells and successful tumor targeting after intravenous delivery [149]. Additionally, a BMP-4-expressing VV demonstrated strong activity against glioma stem cells, preventing tumor recurrence and significantly improving survival in preclinical models [150]. Although another IL-12-expressing VV (VVL-m12) was developed primarily for lung cancer, it also demonstrated potent antitumor activity, strong immunomodulation, and synergy with α-PD1 therapy—findings with potential translational relevance for GBM treatment [151]. Collectively, these studies underscore the therapeutic potential of VV-based virotherapy in GBM, especially when combined with immune checkpoint inhibitors or radiation. Future research should focus on clinical translation and optimizing combinations that overcome GBM’s immunoresistance and recurrence.

2.8. Parvovirus H-1

Parvovirus H-1 (H-1PV), a rodent-derived protoparvovirus from the *Parvoviridae* family, exhibits strong tumor tropism due to dysregulated DNA damage responses and interferon signaling in cancer cells, while remaining non-pathogenic to normal cells [152]. Its oncolytic effects are primarily mediated by the NS1 protein, which induces replication stress, caspase-dependent apoptosis, and immunogenic cell death marked by DAMP release and subsequent immune activation [153,154]. Because productive parvovirus infection requires cell division and is enhanced by oncogenic transformation, certain parvoviruses show promise as oncolytic agents. Mechanistic studies revealed that H-1PV induces apoptosis in tumor cells by activating the CPP32 ICE-like cysteine protease, leading to PARP cleavage and TNF-alpha-like morphological changes, with selective toxicity toward tumor cells over normal astrocytes [155,156]. In vitro study aimed to screen of 12 parvoviruses against human glioblastomas, LuIII, H-1, MVMp, and MVM-G52 showed the highest oncolytic activity at high multiplicity of infection [157]. At low multiplicity of infection, only LuIII effectively infected and killed all five glioblastoma cell lines tested, while H-1 was effective in two [157]. LuIII’s enhanced oncolysis was linked to its capsid and full genome, which enabled selective infection of glioma cells over normal glia in vitro and in vivo [157]. In the used mouse models at the same study, LuIII significantly reduced tumor growth without adverse effects, even after intravenous or intracranial administration [157]. In a Phase I/IIa clinical trial (NCT01301430) involving 18 patients with progressive or recurrent GBM, ParvOryx was administered either intratumorally or intravenously at various doses [158]. The treatment was well tolerated with no dose-limiting toxicities and achieved a median overall survival of 15.5 months [158,159]. Viral transcripts were detected in tumors regardless of dose or administration route, indicating that ParvOryx can cross the blood–brain barrier (BBB) [158]. Moreover, treatment enhanced tumor infiltration by CD4+ and CD8+ T cells and increased pro-inflammatory cytokines in six patients [159]. Despite these promising results, no further clinical trials in GBM are currently underway.

2.9. Poliovirus

Poliovirus recombinant Sabin-rhinovirus IRES (PVSRIPO) is an engineered oncolytic poliovirus derived from the Sabin type 1 strain, modified with a human rhinovirus type 2 IRES to reduce neurovirulence while preserving its ability to target tumors overexpressing CD155, a receptor highly expressed in GBM [160]. In GBM, CD155 found to be consistently expressed across a wide range of models, including laboratory and primary GBM cell lines, patient-derived explant cells, GBM biopsy-derived xenografts, and surgically resected tumor tissues [161,162], underscoring its relevance as a potential therapeutic target in this aggressive brain tumor. PVSRIPO replicates in CD155+ tumor and APCs, leading to direct oncolysis and innate immune activation through MDA-mediated type I interferon signaling, which enhances dendritic cell maturation and CD8+ T cell priming [163,164].
In vivo, PVSRIPO reduced tumor burden and prolonged survival in murine GBM models, with systemic immune responses and no signs of neurovirulence in primate studies [165,166]. Clinically, a notable result from the Phase I trial (NCT01491893) reported a complete and durable radiologic remission in a patient with recurrent GBM, who remained disease-free for 57 months following treatment—one of the longest survivals reported with virotherapy in GBM [166]. Moreover, combining PVSRIPO with anti–PD-1 therapy may lead to enhanced T cell infiltration and better outcomes as reported in other malignancies such as melanoma [167].

2.10. Zika Virus

Zika virus (ZIKV), a neurotropic, mosquito-borne Flavivirus, demonstrates promising oncolytic potential against GBM, particularly by targeting glioma stem cells (GSCs), which are critical drivers of tumor growth and recurrence [168,169]. ZIKV exploits the overexpression of AXL and its ligand Gas6 in GBM cells for cellular entry, preferentially infecting and killing GSCs while sparing differentiated glioma and normal neural cells [170,171]. Its selective cytotoxicity is mediated through factors like elevated SOX2 and integrin αvβ5, alongside suppressed innate immune responses in GSCs [172]. In vitro studies show ZIKV induces rapid cytopathic effects and apoptosis in GBM cells, possibly through digoxin elevation and TNF-α upregulation, with NS5 identified as the primary inhibitory viral protein [173,174,175]. In vivo, ZIKV monotherapy improved survival by targeting GSCs and eliciting robust CD8+ T cell responses [176], with combination therapies such as with anti–PD-1 or irradiation, yielding synergistic benefits [177]. Although no clinical trials have tested ZIKV in GBM, a compelling case report described a GBM patient experiencing long-term remission following natural ZIKV infection, suggesting a potential real-world therapeutic effect worth further investigation [178].

3. Combination Oncolytic Therapies

There is no doubt that OVT offers powerful and innovative avenues for treating GBM, leveraging therapeutic pathways inaccessible to conventional modalities. These viruses reshape the tumor microenvironment (TME) by “being the matches that light the fire” to bring immunological heat into otherwise cold tumors, as quoted from Webb et al. (2023) [179]. However, even promising approaches like OVT encounter limitations when used as a monotherapy. For instance, intratumoral injection of OVs triggers the recruitment of tumor-associated macrophages (TAMs), which cluster around the injection site, and restrict viral spread into other tumor areas [180]. TAMs may also secrete TNF-α, which suppresses viral replication by inducing apoptosis in infected glioma cells [180]. Furthermore, GBM’s increased stiffness extracellular matrix creates an additional structural barrier that restricts the infiltration of OVs into the tumor [181]. Tumor heterogeneity further complicates treatment efficacy; Sottoriva et al. demonstrated that different regions within the same GBM tumor harbor genetically distinct subclones belonging to separate cellular lineages, making it unlikely that a single therapy could effectively target all tumor populations [182]. Additionally, glioma-initiating cells (GICs) are known for their robust DNA repair mechanisms and self-renewal capacity that exhibit resistance to OVs [183].
The aforementioned limitations of OVT as a standalone therapy along with the limitations of other monotherapies underscore the need for combination therapies, each targeting different facets of the tumor to fully ignite a durable antitumor response. In the following subsections, we will explore key combination strategies involving OVs for GBM. This includes conventional modalities such as surgical resection, chemotherapy, and radiotherapy, as well as widely utilized immunotherapies, including immune checkpoint inhibitors and transgene-armed OVs.

3.1. Combination of OVs with Surgical Resection Strategies

Surgical resection remains the cornerstone of standard-of-care treatment for GBM [184]. A greater extent of tumor removal has been consistently associated with improved survival outcomes [184]. However, the infiltrative nature of GBM, characterized by microscopic, finger-like extensions into surrounding brain tissue, poses significant challenges to achieving gross total resection (GTR), even with advanced preoperative and intraoperative imaging modalities [185]. To address the risk of recurrence from residual, migrating tumor cells, one promising strategy involves delivering OVs directly into the post-resection cavity. These viruses will then selectively infect and replicate within residual tumor cells, potentially reducing the likelihood of recurrence as shown in many studies [186]. This approach was clinically demonstrated in a first-in-human phase I trial by Fares et al., in which an engineered oncolytic adenovirus (NSC-CRAd-S-pk7) was administered into the resection margins. The treatment was well-tolerated, induced immune-mediated anti-glioma activity, and showed encouraging survival outcomes in newly diagnosed patients, particularly those with MGMT-unmethylated tumors [106].

3.2. Combination of OVs with Chemotherapeutic Agents

Chemotherapy remains a cornerstone for GBM first-line treatment, with temozolomide being the most commonly employed agent [187]. As an alkylating compound, TMZ exerts its cytotoxic effect by introducing O6-methylguanine lesions through methylation of DNA purine bases [188]. However, its clinical efficacy is often compromised by the elevated expression of the DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT), which reverses the damage by removing methyl groups from the O6 position of guanine, thereby promoting resistance to TMZ and limiting its potential [187]. To overcome this resistance, combining chemotherapy with OVs has emerged as a promising strategy. Chemotherapeutic agents can sensitize tumor cells to viral oncolysis by inducing DNA damage or impairing cellular defense mechanisms. Conversely, OVs can enhance the effectiveness of chemotherapy by disrupting DNA repair pathways and selectively lysing therapy-resistant tumor cells. This bidirectional enhancement can augment tumor cell death more than would any modality alone [189].

3.3. Combination of OVs with Radiotherapy

Radiotherapy (RT) remains a key component of standard GBM treatment. It induces DNA damage, such as strand breaks and base modifications, and contributes to immune activation by promoting the release of proinflammatory cytokines and recruiting effector T cells to the tumor microenvironment [190]. However, the therapeutic efficacy of RT is often limited by the hypoxic nature of GBM. Oxygen plays a critical role in amplifying the effects of low linear energy transfer radiation by stabilizing DNA radicals into permanent damage. In hypoxic conditions, the absence of sufficient oxygen allows tumor cells more time to repair radiation-induced DNA lesions, thereby diminishing treatment effectiveness [191]. Emerging evidence supports the synergistic potential of combining RT with OVs. Studies have shown that ionizing radiation might enhance the replication of oHSV, while the virus, in turn, interferes with DNA double-strand break repair in irradiated GBM cells [59,189]. Furthermore, a recent in vitro study demonstrated that the oncolytic vaccinia virus ΔF4LΔJ2R effectively replicates in and kills irradiated brain tumor–initiating cells [148]. When used in combination with RT in immune-competent orthotopic CT2A-luc mouse models, this approach significantly prolonged survival and achieved complete tumor regression in the majority of treated animals—outperforming each therapy alone [148].

3.4. Combination of OVs with Immune Checkpoint Inhibitors

Immune checkpoints such as PD-1, CTLA-4, LAG3, and TIM3 are essential regulators of T cell responses, maintaining immune tolerance and protecting against autoimmunity. However, many tumors, including GBM, hijack these inhibitory pathways to evade immune attack and promote tumor progression [192]. In GBM, immune evasion is facilitated by the widespread expression of PD-L1, which is found in up to 90% of tumor cells. This contributes to T cell exhaustion and suppression of effective immune responses [193]. Such findings have driven the development of immune checkpoint inhibitors (ICIs) which are antibodies designed to block these negative signals and reactivate T cells, enabling them to recognize and eliminate tumor cells [194].
Despite their clinical success in several malignancies, ICIs have shown limited benefit for GBM. The phase III CheckMate 143 trial, for example, reported no significant survival improvement with the PD-1 blocker nivolumab compared to bevacizumab in patients with recurrent GBM [195]. According to Arrieta et al., this resistance is largely attributed to GBM’s low immunogenicity, limited infiltration by effector T cells, and an immunosuppressive TME. Additional obstacles include impaired antigen presentation due to low MHC I and II expression, and the limited permeability of the BBB [196]. To enhance ICI efficacy in GBM, combination strategies with OVs are being explored. OVs can induce immunogenic cell death, promote infiltration of immune cells into the tumor, and increase the expression of immune checkpoint ligands such as PD-L1 that may sensitize tumors to ICIs [137]. This synergistic interaction between OVs and ICIs may help overcome the resistance seen with GBM monotherapy. For instance, in a preclinical study by Sugawara et al. (2021), the combined use of intratumoral G47Δ, a modified HSV, and systemic anti-CTLA-4 antibody significantly depleted regulatory T cells and enhanced the activity of other immune cells highlighting the potential of such combination approaches in reprogramming the GBM immune environment [197].

3.5. Arming OVs with Therapeutic Transgenes

The genes for cytokines (GM-CSF, IL-12), immunomodulatory factors (anti-PD1), anti-angiogenic factors, TME inhibitors/degraders, and cytotoxic proteins can now be incorporated into OVs, allowing them to induce potent antitumor immune responses in addition to their traditional role of direct tumor cell lysis [198]. Arming OVs with these transgenes offers two major advantages over unmodified (unarmed) OVs. First, while unarmed OVs primarily rely on direct oncolysis and passive stimulation of the immune system, transgene-armed OVs actively reshape the highly immunosuppressive TME of GBM in multiple targeted mechanisms, leading to more enhanced antitumor immune responses [199]. For example, a modified herpes simplex virus (G47-mIL12) engineered to express IL-12 significantly prolonged survival in mice with intracerebral glioma by targeting glioma stem cells, increasing IFN-γ production, inhibiting angiogenesis, and reducing regulatory T cell infiltration into the tumor [200]. Secondly, arming OVs with cytokines such as IL-12 enables direct delivery of these potent immunostimulatory agents into the tumor, reducing the systemic toxicity typically seen when administered systematically without OVs carriers. This approach boosts the local immune attack on tumor cells while enhancing safety [201,202].
GM-CSF is another key cytokine used to enhance the immunogenic potential of OVs. It recruits and activates dendritic cells, macrophages, and other antigen-presenting cells, ultimately enhancing tumor antigen presentation and priming of T cells [199]. The effectiveness of incorporating GM-CSF into OVs is well demonstrated by talimogene laherparepvec (T-VEC), a genetically engineered HSV-1 designed to express GM-CSF [203]. Clinical trials showed that T-VEC achieved higher response rates than treatment with GM-CSF alone, emphasizing the added therapeutic benefit of coupling OVT with transgene therapy [203].

4. Non-Viral Oncolytic Strategies

4.1. Oncolytic Bacteria

The GBM is notoriously resistant to treatment due to its hypoxic, necrotic microenvironment, which limits the effectiveness of chemotherapy and radiotherapy [204,205]. However, this same environment makes it an ideal target for anaerobic bacteria like Clostridium novyi-NT, a genetically modified strain engineered to lack its lethal toxin gene [206]. Non-viral oncolytic strategies have garnered attention as alternative therapeutic avenues for GBM, particularly due to their ability to exploit the tumor’s hypoxic microenvironment and stimulate anti-tumor immunity. Among these, Clostridium novyi-NT (C. novyi-NT), which is an attenuated strain of an obligate anaerobic bacterium, has shown remarkable tumor specificity and therapeutic efficacy in preclinical GBM models.
When injected directly into tumors, C. novyi-NT spores germinate only in hypoxic regions, selectively destroying tumor tissue while sparing healthy brain (Figure 3) [161]. In rat glioma models, intratumoral injection led to rapid tumor reduction and significantly improved survival. Importantly, the bacteria could also target invasive tumor cells beyond the resection margin, which are areas typically inaccessible to conventional therapies. Beyond direct tumor lysis, bacterial germination triggers strong local inflammation and may stimulate systemic antitumor immune responses [206,207]. Compared to systemic delivery, direct injection ensures better tumor targeting with fewer off-target effects [208].
These findings were mirrored in a veterinary trial: dogs with soft tissue sarcomas responded well to C. novyi-NT, with a 37.5% objective response rate and manageable local side effects [209]. These encouraging findings justified compassionate use in a human patient with leiomyosarcoma, where a single intratumoral injection significantly reduced tumor volume, supporting the feasibility of bacterial-mediated tumor lysis in solid malignancies [206]. Given the infiltrative growth, hypoxia, and resistance to standard therapies seen in high-grade gliomas, localized bacterial therapy such as C. novyi-NT may offer a targeted and effective strategy, albeit with potential risks like brain edema or abscess formation that require clinical monitoring and surgical readiness.
Another promising approach involves VXM01, an oral DNA vaccine based on Salmonella typhi Ty21a engineered to carry a plasmid encoding vascular endothelial growth factor receptor 2 (VEGFR-2) (Figure 3). This strategy aims to elicit systemic T-cell responses targeting tumor vasculature. In a clinical trial involving patients with progressive GBM who had failed standard radiochemotherapy, VXM01 was well tolerated and induced VEGFR-2–specific T-cell responses in over half of treated individuals [210]. Notably, enhanced CD8+/Treg ratios and reduced intratumoral PD-L1 expression were observed post-treatment, correlating with prolonged survival in some patients. These findings support further development of VXM01, especially in combination with immune checkpoint inhibitors such as anti-PD-L1 antibodies, to enhance immune activation in an otherwise immunosuppressive GBM microenvironment.
Recent research has highlighted the promising role of bacteria as vectors for therapeutic gene delivery or to secrete anti-tumor agents locally, minimizing systemic toxicity [211]. In addition, bacterial components such as lipopolysaccharides or flagellin can stimulate innate immune responses and reverse the immunosuppressive tumor microenvironment, enhancing the efficacy of immunotherapies [212]. Some studies have even explored the use of bacteria to alter the gut–brain axis, indirectly influencing the brain’s immune surveillance and tumor response. Taken together, these findings suggest that with further refinement and clinical translation, bacteria-based strategies could offer a novel and complementary approach to conventional GBM therapies.
Gram-negative bacteria, such as attenuated Salmonella typhimurium (VNP20009), can stimulate innate immunity through Toll-like and NOD-like receptors on phagocytes, reactivating antitumor responses in the post-surgical tumor bed [213]. These bacteria promote immune cell infiltration, overcome immune “coldness,” and present tumor-associated antigens to T cells, bridging innate and adaptive immunity [214,215]. Intriguingly, retrospective studies suggest improved GBM patient outcomes when localized Gram-negative infections occur postoperatively, indicating a therapeutic window for bacterial intervention [216,217,218]. Building on this, engineered bacteria, such as immunostimulatory autolysing Salmonella-nanocapsule delivery systems (IASNDS), can safely colonize hypoxic tumor regions and release immunogenic bacterial components through controlled lysis, inducing pyroptosis and antigen release [211]. These events enhance antigen presentation, cytokine release, and T cell activation, culminating in robust antitumor immunity. Coupled with ATP-responsive hydrogels that sustain local immune activation via CpG oligonucleotide release, this innovative bacteriotherapy strategy demonstrates significant promise in preventing GBM recurrence by reshaping the immune microenvironment, mobilizing both innate and adaptive immunity, and offering a transformative post-surgical adjunct in GBM management.
Looking forward, the integration of CRISPR-Cas9 genome editing offers a transformative avenue to enhance the precision, safety, and efficacy of oncolytic bacterial therapies. CRISPR can be employed to knock out virulence genes, insert safety switches, or engineer bacterial payloads that secrete immune-stimulating cytokines (e.g., IL-12, GM-CSF), tumor-selective toxins, or checkpoint inhibitors directly within the tumor microenvironment [219]. For example, Clostridium or Salmonella strains could be modified to include inducible suicide genes or biosensors responsive to tumor-specific signals, allowing for precise control over bacterial replication and therapeutic payloads. Additionally, CRISPR-based engineering enables the creation of multi-functional bacterial vectors that combine tumor targeting, immune modulation, and biosafety; all essential for clinical translation for GBM, where immune privilege and anatomical constraints pose significant therapeutic barriers.

4.2. Oncolytic Peptides

In recent years, peptide-based therapies have emerged as a promising alternative for GBM as a non-viral oncolytic strategy. Peptides demonstrate high specificity in targeted therapies due to their ability to bind with high affinity to receptors that are overexpressed or tumor-specific in cancer cells. Thanks to these properties, they can selectively target tumor cells while exerting minimal toxic effects on healthy tissues. Additionally, certain peptides have been reported in the literature to exhibit intrinsic antineoplastic activity and may be used as pharmacological agents in drug delivery systems or for modulating intracellular signaling cascades. Among these strategies, p28, a peptide with high permeability across cellular and blood–brain barriers, stands out for its therapeutic efficacy against GBM as well as its potential role as a carrier for diagnostic imaging agents. In GBM animal models, p28 has been shown to enhance the cytotoxic activity of temozolomide, a DNA-damaging agent known to be effective against brain tumors. Furthermore, the chemical conjugate of p28 with the near-infrared (NIR) dye indocyanine green (ICG), referred to as ICG-p28, has been demonstrated to selectively localize to tumors in GBM animal models, with accumulation observed in both the cytoplasmic space and the nucleus. Most notably, p28 has exhibited a significant anti-tumor effect on GBM [220]. Collectively, these findings suggest that p28 possesses strong potential as a dual-function agent, capable of both therapeutic impact and precision imaging for GBM management.
The tumor microenvironment plays a critical role in mediating resistance to GBM therapies. As a result, recent therapeutic strategies have focused on targeting components of the extracellular matrix (ECM), which contributes to this resistance. Brevican (Bcan), an ECM protein highly expressed in GBM tissue, has a specific isoform called dg-Bcan that was recently identified as a promising molecular target for GBM. In this context, a newly designed 8-amino-acid D-peptide, termed BTP-7, demonstrates strong binding affinity and high specificity toward dg-Bcan. It is selectively taken up by GBM cells, allowing for precise tumor targeting. To demonstrate its diagnostic potential, BTP-7 was radiolabeled with 18F and used in PET imaging in mouse models bearing intracranial GBM. The imaging results confirmed that BTP-7 effectively localizes to tumor regions, supporting its use in non-invasive tumor detection. Furthermore, BTP-7’s structure, which is resistant to enzymatic degradation, allows it to cross the blood–brain barrier and accumulate within tumor tissue [221]. These properties suggest that BTP-7 may serve as a promising tool for both diagnostic imaging and targeted therapy in GBM, offering a minimally invasive approach to managing this highly aggressive cancer.
In addition to preclinical studies investigating the effects of oncolytic peptides in GBM treatment, clinical trials have also assessed the efficacy of peptide-based vaccines in GBM patients. A recent study followed 173 patients with GBM who were treated with a personalized peptide vaccine targeting tumor-specific neoantigens and evaluated the feasibility, immunogenicity, and therapeutic potential of this individualized vaccine approach [222]. Among newly diagnosed and recurrent patients, the median overall survival from the time of diagnosis was reported as 31.9 months. The peptide vaccine was well tolerated, even when administered alongside standard therapies. Serious adverse events were rare, and side effects were mostly mild to moderate in severity. Blood sample analysis revealed that the vaccine elicited strong T-cell responses in a large proportion of patients. Furthermore, these vaccine-induced T-cell responses were found to be durable in most individuals. Patients who developed strong T-cell responses against the peptides demonstrated significantly longer survival [222]. In conclusion, personalized neoantigen-based peptide vaccines emerge as an effective and well-tolerated immunotherapeutic strategy for patients with glioblastoma.

5. Concerns and Future Directions in Oncolytic Therapies

Several challenges have been proposed to explain the limited success of OVs in the treatment of GBMs. These include suboptimal viral delivery methods, issues related to prodrug selection and compliance, and an insufficient induction of durable antitumor immune responses [179]. Most studies to date have employed intratumoral injection of viral particles following surgical resection. Although this strategy enables direct delivery to the tumor site, it remains difficult to ascertain the extent to which residual microscopic tumor cells are exposed to the virus, particularly in the presence of normal brain parenchyma or necrotic tissue, which may act as physical barriers [223]. Furthermore, the efficiency of viral uptake by tumor cells and the likelihood of successful replication remain uncertain. Enhancing viral tropism for tumor cells and optimizing delivery strategies such as through convection-enhanced delivery may help address these limitations [224]. Although techniques such as convection-enhanced delivery may improve distribution, inconsistent prodrug dosing and timing, as observed in the Toca 511/Toca FC trial, where patients received fewer cycles than preclinical models suggested were necessary, further limit therapeutic efficacy [223].
The profound intertumoral and intratumoral heterogeneity of GBM (well-characterized by TCGA into proneural, mesenchymal, and classical subtypes) complicates treatment, as phenotypic plasticity and subtype transitions can lead to resistance and failure to target all tumor cell populations [225,226,227]. Moreover, the TME, composed of the extracellular matrix, immune cells, and stem cells, significantly influences tumor progression and therapeutic response, and must be considered both as a barrier and a potential modulator of efficacy [196]. Perhaps the greatest obstacle is the profoundly immunosuppressive nature of GBM, characterized by a low mutational burden, secretion of immunosuppressive factors [228], sparse and exhausted tumor-infiltrating lymphocytes [229], and systemic T cell sequestration [228]. Therefore, achieving lasting efficacy with OVs will likely require combination strategies, such as pairing OVs with immune checkpoint inhibitors or cytokine-based therapies to boost peritumoral CD8+ lymphocyte infiltration [228,229].
Future directions in oncolytic therapies of GBM are increasingly focused on improving the specificity, delivery, and overall therapeutic efficacy of these agents. One promising approach involves nanoparticle-based delivery systems, which can encapsulate oncolytic viruses or agents to help them bypass biological barriers such as the blood–brain barrier. Another area of development includes the use of armed oncolytic viruses that are genetically modified to deliver therapeutic payloads, such as cytokines like IL-12 or IFN-beta, or other immune-modulating factors. Additionally, combining oncolytic viruses with targeted therapies such as agents against EGFR, VEGF, or IDH mutations may enhance antitumor effects through complementary mechanisms. Efforts are also underway to identify reliable biomarkers to guide patient selection, aiming to match specific molecular profiles with the most effective therapeutic strategies.

6. Conclusions

Oncolytic therapies represent a promising frontier in the fight against GBM, offering a novel approach that combines targeted tumor lysis with potent immune activation. Despite significant strides such as the clinical progress of HSV-based therapies, challenges remain, including immune evasion, tumor heterogeneity, and delivery limitations imposed by the blood–brain barrier. Integrating oncolytic viruses with immune checkpoint inhibitors, chemotherapy, and radiation has demonstrated synergistic potential, but these strategies require further clinical validation. Moreover, non-viral oncolytic agents, such as tumor-targeting bacteria and synthetic peptides, remain underexplored, but they may expand the therapeutic arsenal. Future research must prioritize refining delivery techniques, enhancing tumor selectivity, and leveraging biomarkers for patient stratification. By addressing these gaps and aligning OVT with precision medicine strategies, there is a real opportunity to overcome the therapeutic stagnation in GBM and offer patients more effective, durable treatment options.

Funding

This research received no specific grants from any funding agency in the public, commercial, or non-profit sectors.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

Conflicts of Interest

The authors declare no conflicts of interest in preparing this paper.

List of Abbreviations

Ad5: Adenovirus Serotype 5, Ad6: Adenovirus Serotype 6, AdV: Adenovirus, AKT: Protein Kinase B, BBB: Blood–Brain Barrier, Bcan: Brevican, BTP-7: Brevican Targeting Peptide 7, C. novyi-NT: Clostridium novyi—Non-Toxic strain, CAR: Chimeric Antigen Receptor, CAR-T: Chimeric Antigen Receptor T Cells, ChaseM: Chondroitinase ABC, CNS: Central Nervous System, CRISPR: Clustered Regularly Interspaced Short Palindromic Repeats, CT2A-luc: A luciferase-expressing variant of the CT-2A mouse glioma cell line, CTLA-4: Cytotoxic T-Lymphocyte Antigen 4, CXCR4: C-X-C Chemokine Receptor Type 4, DAMPs: Damage-Associated Molecular Patterns, dg-Bcan: Deglycosylated Brevican, ECM: Extracellular Matrix, EGFRvIII: Epidermal Growth Factor Receptor Variant III, EMA: European Medicines Agency, ER: Endoplasmic Reticulum, FDA: U.S. Food and Drug Administration, G47Δ: Genetically engineered HSV-1 lacking ICP47, γ34.5, and ICP6 genes, GBM: Glioblastoma, GICs: Glioma-Initiating Cells, GM-CSF: Granulocyte-Macrophage Colony-Stimulating Factor, GSCs: Glioma Stem-like Cells, GTR: Gross Total Resection, HER2: Human Epidermal Growth Factor Receptor 2, HMGB1: High Mobility Group Box 1, HSV: Herpes Simplex Virus, HSV-1: Herpes Simplex Virus Type 1, HSV-2: Herpes Simplex Virus Type 2, ICG: Indocyanine Green, ICIs: Immune Checkpoint Inhibitors, ICP: Infected Cell Protein, IFN-γ: Interferon Gamma, IL-12: Interleukin-12, IL-2: Interleukin-2, IL-21: Interleukin-21, IL-6: Interleukin-6, JAK1: Janus Kinase 1, JNK: c-Jun N-terminal Kinase, LAG3: Lymphocyte Activation Gene 3, MEK: Mitogen-Activated Protein Kinase Kinase, MeV: Measles Virus, MGMT: O6-methylguanine-DNA Methyltransferase, MHC: Major Histocompatibility Complex, miR-122: MicroRNA-122, MV-Edm: Measles Virus Edmonston Strain, NDV: Newcastle Disease Virus, NIR: Near-Infrared, NK cells: Natural Killer Cells, NSC-CRAd-S-pk7: Neural Stem Cell–Carrier Conditionally Replicating Adenovirus with Polylysine Modification, oAd: Oncolytic Adenovirus, oAdV: Oncolytic Adenoviral Vector, oHSV: Oncolytic Herpes Simplex Virus, oHSV-1: Oncolytic Herpes Simplex Virus Type 1, OVs: Oncolytic Viruses, OVT: Oncolytic Virotherapy, PAMPs: Pathogen-Associated Molecular Patterns, PD-1: Programmed Death-1, PD-L1: Programmed Death-Ligand 1, PDX: Patient-Derived Xenograft, PET: Positron Emission Tomography, PI3K: Phosphoinositide 3-Kinase, PKR: Protein Kinase R, PTEN: Phosphatase and Tensin Homolog, RGD: Arginine-Glycine-Aspartate (motif), rNDV: Recombinant Newcastle Disease Virus, RT: Radiotherapy, STAT3: Signal Transducer and Activator of Transcription 3, TAMs: Tumor-Associated Macrophages, TGF-β: Transforming Growth Factor Beta, TIM3: T cell immunoglobulin and mucin-domain containing-3, TME: Tumor Microenvironment, TNF-α: Tumor Necrosis Factor Alpha, TS-2021: Third-generation Oncolytic Adenovirus Vector, T-VEC: Talimogene Laherparepvec, VV: Vaccinia Virus, VEGFR-2: Vascular Endothelial Growth Factor Receptor 2, VXM01: Oral DNA vaccine targeting VEGFR-2, ΔF4LΔJ2R: Double-deletion Vaccinia Virus lacking F4L and J2R genes.

References

  1. Miller, K.D.; Ostrom, Q.T.; Kruchko, C.; Patil, N.; Tihan, T.; Cioffi, G.; Brat, D.J.; Barnholtz-Sloan, J.S. Brain and other central nervous system tumor statistics, 2021. CA Cancer J. Clin. 2021, 71, 381–406. [Google Scholar] [CrossRef] [PubMed]
  2. Ohgaki, H.; Kleihues, P. The definition of primary and secondary glioblastoma. Clin. Cancer Res. 2013, 19, 764–772. [Google Scholar] [CrossRef] [PubMed]
  3. Hanif, F.; Muzaffar, K.; Perveen, K.; Malhi, S.M.; Simjee, S.U. Glioblastoma multiforme: A review of its epidemiology and pathogenesis through clinical presentation and treatment. Asian Pac. J. Cancer Prev. 2017, 18, 3–9. [Google Scholar] [CrossRef] [PubMed]
  4. Angom, R.S.; Nakka, N.M.R.; Bhattacharya, S. Advances in glioblastoma therapy: An update on current approaches. Brain Sci. 2023, 13, 1536. [Google Scholar] [CrossRef] [PubMed]
  5. Ohgaki, H.; Dessen, P.; Jourde, B.; Horstmann, S.; Nishikawa, T.; Di Patre, P.L.; Burkhard, C.; Schuler, D.; Probst-Hensch, N.M.; Maiorka, P.C.; et al. Genetic pathways to glioblastoma: A population-based study. Cancer Res. 2004, 64, 6892–6899. [Google Scholar] [CrossRef] [PubMed]
  6. Wu, W.; Klockow, J.L.; Zhang, M.; Lafortune, F.; Chang, E.; Jin, L.; Wu, Y.; Daldrup-Link, H.E. Glioblastoma multiforme (GBM): An overview of current therapies and mechanisms of resistance. Front. Oncol. 2021, 11, 769872. [Google Scholar] [CrossRef]
  7. Kreisl, T.N.; Kim, L.; Moore, K.; Duic, P.; Royce, C.; Stroud, I.; Garren, N.; Mackey, M.; Butman, J.A.; Camphausen, K.; et al. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J. Clin. Oncol. 2009, 27, 740–745. [Google Scholar] [CrossRef] [PubMed]
  8. Gramatzki, D.; Roth, P.; Rushing, E.J.; Weller, J.; Andratschke, N.; Hofer, S.; Korol, D.; Regli, L.; Pangalu, A.; Pless, M.; et al. Bevacizumab may improve quality of life, but not overall survival in glioblastoma: An epidemiological study. Ann. Oncol. 2018, 29, 1431–1436. [Google Scholar] [CrossRef] [PubMed]
  9. Weller, M.; Le Rhun, E. How did lomustine become standard of care in recurrent glioblastoma? Cancer Treat. Rev. 2020, 87, 102029. [Google Scholar] [CrossRef] [PubMed]
  10. Salvato, I.; Marchini, A. Immunotherapeutic strategies for the treatment of glioblastoma: Current challenges and future perspectives. Cancers 2024, 16, 1276. [Google Scholar] [CrossRef] [PubMed]
  11. Liu, Y.; Zhou, F.; Ali, H.; Lathia, J.D.; Chen, P. Immunotherapy for glioblastoma: Current state, challenges, and future perspectives. Cell Mol. Immunol. 2024, 21, 1354–1375. [Google Scholar] [CrossRef] [PubMed]
  12. Wang, J.; Liu, Y.; Zhang, A.; Yu, W.; Lei, Q.; Xiao, B.; Luo, Z. Investigational microbiological therapy for glioma. Cancers 2022, 14, 5977. [Google Scholar] [CrossRef] [PubMed]
  13. Hamad, A.; Yusubalieva, G.M.; Lipatova, A.V. Recent developments in glioblastoma therapy: Oncolytic viruses and emerging future strategies. Viruses 2023, 15, 547. [Google Scholar] [CrossRef] [PubMed]
  14. Asija, S.; Chatterjee, A.; Goda, J.S.; Yadav, S.; Chekuri, G.; Purwar, R. Oncolytic immunovirotherapy for high-grade gliomas: A novel and an evolving therapeutic option. Front. Immunol. 2023, 14, 1118246. [Google Scholar] [CrossRef] [PubMed]
  15. Todo, T.; Ito, H.; Ino, Y.; Ohtsu, H.; Ota, Y.; Shibahara, J.; Tanaka, M. Intratumoral oncolytic herpes virus G47∆ for residual or recurrent glioblastoma: A phase 2 trial. Nat. Med. 2022, 28, 1630–1639. [Google Scholar] [CrossRef] [PubMed]
  16. Shoaf, M.L.; Desjardins, A. Oncolytic viral therapy for malignant glioma and their application in clinical practice. Neurotherapeutics 2022, 19, 1732–1750. [Google Scholar] [CrossRef] [PubMed]
  17. Kaufman, H.L.; Kohlhapp, F.J.; Zloza, A. Oncolytic viruses: A new class of immunotherapy drugs. Nat. Rev. Drug Discov. 2015, 14, 642–662. [Google Scholar] [CrossRef]
  18. Mihelson, N.; McGavern, D.B. Viral Control of Glioblastoma. Viruses 2021, 13, 1264. [Google Scholar] [CrossRef]
  19. Lin, D.; Shen, Y.; Liang, T. Oncolytic virotherapy: Basic principles, recent advances and future directions. Signal Transduct. Target. Ther. 2023, 8, 156. [Google Scholar] [CrossRef]
  20. Suzich, J.B.; Cliffe, A.R. Strength in diversity: Understanding the pathways to herpes simplex virus reactivation. Virology 2018, 522, 81–91. [Google Scholar] [CrossRef]
  21. Zhu, S.; Viejo-Borbolla, A. Pathogenesis and virulence of herpes simplex virus. Virulence 2021, 12, 2670–2702. [Google Scholar] [CrossRef]
  22. Ma, W.; He, H.; Wang, H. Oncolytic herpes simplex virus and immunotherapy. BMC Immunol. 2018, 19, 40. [Google Scholar] [CrossRef]
  23. Kardani, K.; Sanchez Gil, J.; Rabkin, S.D. Oncolytic herpes simplex viruses for the treatment of glioma and targeting glioblastoma stem-like cells. Front. Cell. Infect. Microbiol. 2023, 13, 1206111. [Google Scholar] [CrossRef]
  24. Miest, T.S.; Cattaneo, R. New viruses for cancer therapy: Meeting clinical needs. Nat. Rev. Microbiol. 2014, 12, 23–34. [Google Scholar] [CrossRef]
  25. Zhang, S.; Rabkin, S.D. The discovery and development of oncolytic viruses: Are they the future of cancer immunotherapy? Expert Opin. Drug Discov. 2021, 16, 391–410. [Google Scholar] [CrossRef]
  26. Conry, R.M.; Westbrook, B.; McKee, S.; Norwood, T.G. Talimogene laherparepvec: First in class oncolytic virotherapy. Hum. Vaccines Immunother. 2018, 14, 839–846. [Google Scholar] [CrossRef] [PubMed]
  27. Kohlhapp, F.J.; Kaufman, H.L. Molecular Pathways: Mechanism of Action for Talimogene Laherparepvec, a New Oncolytic Virus Immunotherapy. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 1048–1054. [Google Scholar] [CrossRef] [PubMed]
  28. Russell, S.J.; Peng, K.W. Oncolytic Virotherapy: A Contest between Apples and Oranges. Mol. Ther. J. Am. Soc. Gene Ther. 2017, 25, 1107–1116. [Google Scholar] [CrossRef]
  29. MartuzaR, L.; Malick, A.; Markert, J.M.; Ruffner, K.L.; Coen, D.M. Experimental therapy of human glioma by means of a genetically engineered virus mutant. Science 1991, 252, 854–856. [Google Scholar] [CrossRef]
  30. Sahu, U.; Mullarkey, M.P.; Pei, G.; Zhao, Z.; Hong, B.; Kaur, B. oHSV-P10 reduces glioma stem cell enrichment after oncolytic HSV therapy. Mol. Ther. Oncolytics 2023, 29, 30–41. [Google Scholar] [CrossRef] [PubMed]
  31. Russell, L.; Swanner, J.; Jaime-Ramirez, A.C.; Wang, Y.; Sprague, A.; Banasavadi-Siddegowda, Y.; Yoo, J.Y.; Sizemore, G.M.; Kladney, R.; Zhang, J.; et al. PTEN expression by an oncolytic herpesvirus directs T-cell mediated tumor clearance. Nat. Commun. 2018, 9, 5006. [Google Scholar] [CrossRef]
  32. Rivas, T.; Goodrich, J.A.; Kugel, J.F. The Herpes Simplex Virus 1 Protein ICP4 Acts as both an Activator and a Repressor of Host Genome Transcription during Infection. Mol. Cell. Biol. 2021, 41, e0017121. [Google Scholar] [CrossRef] [PubMed]
  33. Reale, A.; Gatta, A.; Shaik, A.K.B.; Shallak, M.; Chiaravalli, A.M.; Cerati, M.; Zaccaria, M.; La Rosa, S.; Calistri, A.; Accolla, R.S.; et al. An oncolytic HSV-1 vector induces a therapeutic adaptive immune response against glioblastoma. J. Transl. Med. 2024, 22, 862. [Google Scholar] [CrossRef]
  34. Wang, L.; Zhou, X.; Chen, X.; Liu, Y.; Huang, Y.; Cheng, Y.; Ren, P.; Zhao, J.; Zhou, G.G. Enhanced therapeutic efficacy for glioblastoma immunotherapy with an oncolytic herpes simplex virus armed with anti-PD-1 antibody and IL-12. Mol. Ther. Oncol. 2024, 32, 200799. [Google Scholar] [CrossRef]
  35. Bommareddy, P.K.; Wakimoto, H.; Martuza, R.L.; Kaufman, H.L.; Rabkin, S.D.; Saha, D. Oncolytic herpes simplex virus expressing IL-2 controls glioblastoma growth and improves survival. J. Immunother. Cancer 2024, 12, e008880. [Google Scholar] [CrossRef]
  36. Saha, D.; Martuza, R.L.; Rabkin, S.D. Macrophage Polarization Contributes to Glioblastoma Eradication by Combination Immunovirotherapy and Immune Checkpoint Blockade. Cancer Cell 2017, 32, 253–267.e5. [Google Scholar] [CrossRef]
  37. Alessandrini, F.; Menotti, L.; Avitabile, E.; Appolloni, I.; Ceresa, D.; Marubbi, D.; Campadelli-Fiume, G.; Malatesta, P. Eradication of glioblastoma by immuno-virotherapy with a retargeted oncolytic HSV in a preclinical model. Oncogene 2019, 38, 4467–4479. [Google Scholar] [CrossRef] [PubMed]
  38. Appolloni, I.; Alessandrini, F.; Menotti, L.; Avitabile, E.; Marubbi, D.; Piga, N.; Ceresa, D.; Piaggio, F.; Campadelli-Fiume, G.; Malatesta, P. Specificity, Safety, Efficacy of EGFRvIII-Retargeted Oncolytic HSV for Xenotransplanted Human Glioblastoma. Viruses 2021, 13, 1677. [Google Scholar] [CrossRef] [PubMed]
  39. Zheng, Y.; Wang, X.; Ji, Q.; Fang, A.; Song, L.; Xu, X.; Lin, Y.; Peng, Y.; Yu, J.; Xie, L.; et al. OH2 oncolytic virus: A novel approach to glioblastoma intervention through direct targeting of tumor cells and augmentation of anti-tumor immune responses. Cancer Lett. 2024, 589, 216834. [Google Scholar] [CrossRef]
  40. Nakashima, H.; Nguyen, T.; Kasai, K.; Passaro, C.; Ito, H.; Goins, W.F.; Shaikh, I.; Erdelyi, R.; Nishihara, R.; Nakano, I.; et al. Toxicity and Efficacy of a Novel GADD34-expressing Oncolytic HSV-1 for the Treatment of Experimental Glioblastoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2018, 24, 2574–2584. [Google Scholar] [CrossRef]
  41. Nehama, D.; Di Ianni, N.; Musio, S.; Du, H.; Patané, M.; Pollo, B.; Finocchiaro, G.; Park, J.J.H.; Dunn, D.E.; Edwards, D.S.; et al. B7-H3-redirected chimeric antigen receptor T cells target glioblastoma and neurospheres. EBioMedicine 2019, 47, 33–43. [Google Scholar] [CrossRef] [PubMed]
  42. Chalise, L.; Kato, A.; Ohno, M.; Maeda, S.; Yamamichi, A.; Kuramitsu, S.; Shiina, S.; Takahashi, H.; Ozone, S.; Yamaguchi, J.; et al. Efficacy of cancer-specific anti-podoplanin CAR-T cells and oncolytic herpes virus G47Δ combination therapy against glioblastoma. Mol. Ther. Oncolytics 2022, 26, 265–274. [Google Scholar] [CrossRef]
  43. Meisen, W.H.; Wohleb, E.S.; Jaime-Ramirez, A.C.; Bolyard, C.; Yoo, J.Y.; Russell, L.; Hardcastle, J.; Dubin, S.; Muili, K.; Yu, J.; et al. The Impact of Macrophage- and Microglia-Secreted TNFα on Oncolytic HSV-1 Therapy in the Glioblastoma Tumor Microenvironment. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2015, 21, 3274–3285. [Google Scholar] [CrossRef]
  44. Yoo, J.Y.; Swanner, J.; Otani, Y.; Nair, M.; Park, F.; Banasavadi-Siddegowda, Y.; Liu, J.; Jaime-Ramirez, A.C.; Hong, B.; Geng, F.; et al. Oncolytic HSV therapy increases trametinib access to brain tumors and sensitizes them in vivo. Neuro-Oncol. 2019, 21, 1131–1140. [Google Scholar] [CrossRef]
  45. Otani, Y.; Yoo, J.Y.; Chao, S.; Liu, J.; Jaime-Ramirez, A.C.; Lee, T.J.; Hurwitz, B.; Yan, Y.; Dai, H.; Glorioso, J.C.; et al. Oncolytic HSV-Infected Glioma Cells Activate NOTCH in Adjacent Tumor Cells Sensitizing Tumors to Gamma Secretase Inhibition. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 2381–2392. [Google Scholar] [CrossRef]
  46. Esaki, S.; Rabkin, S.D.; Martuza, R.L.; Wakimoto, H. Transient fasting enhances replication of oncolytic herpes simplex virus in glioblastoma. Am. J. Cancer Res. 2016, 6, 300–311. [Google Scholar]
  47. Peters, C.; Paget, M.; Tshilenge, K.T.; Saha, D.; Antoszczyk, S.; Baars, A.; Frost, T.; Martuza, R.L.; Wakimoto, H.; Rabkin, S.D. Restriction of Replication of Oncolytic Herpes Simplex Virus with a Deletion of γ34.5 in Glioblastoma Stem-Like Cells. J. Virol. 2018, 92, e00246-18. [Google Scholar] [CrossRef]
  48. Yoo, J.Y.; Jaime-Ramirez, A.C.; Bolyard, C.; Dai, H.; Nallanagulagari, T.; Wojton, J.; Hurwitz, B.S.; Relation, T.; Lee, T.J.; Lotze, M.T.; et al. Bortezomib Treatment Sensitizes Oncolytic HSV-1-Treated Tumors to NK Cell Immunotherapy. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 5265–5276. [Google Scholar] [CrossRef]
  49. Monie, D.D.; Correia, C.; Zhang, C.; Ung, C.Y.; Vile, R.G.; Li, H. Modular network mechanism of CCN1-associated resistance to HSV-1-derived oncolytic immunovirotherapies for glioblastomas. Sci. Rep. 2021, 11, 11198. [Google Scholar] [CrossRef] [PubMed]
  50. Esaki, S.; Nigim, F.; Moon, E.; Luk, S.; Kiyokawa, J.; Curry, W., Jr.; Cahill, D.P.; Chi, A.S.; Iafrate, A.J.; Martuza, R.L.; et al. Blockade of transforming growth factor-β signaling enhances oncolytic herpes simplex virus efficacy in patient-derived recurrent glioblastoma models. Int. J. Cancer 2017, 141, 2348–2358. [Google Scholar] [CrossRef] [PubMed]
  51. Delwar, Z.M.; Kuo, Y.; Wen, Y.H.; Rennie, P.S.; Jia, W. Oncolytic Virotherapy Blockade by Microglia and Macrophages Requires STAT1/3. Cancer Res. 2018, 78, 718–730. [Google Scholar] [CrossRef] [PubMed]
  52. Jaime-Ramirez, A.C.; Dmitrieva, N.; Yoo, J.Y.; Banasavadi-Siddegowda, Y.; Zhang, J.; Relation, T.; Bolyard, C.; Wojton, J.; Kaur, B. Humanized chondroitinase ABC sensitizes glioblastoma cells to temozolomide. J. Gene Med. 2017, 19, e2942. [Google Scholar] [CrossRef] [PubMed]
  53. Hong, B.; Muili, K.; Bolyard, C.; Russell, L.; Lee, T.J.; Banasavadi-Siddegowda, Y.; Yoo, J.Y.; Yan, Y.; Ballester, L.Y.; Bockhorst, K.H.; et al. Suppression of HMGB1 Released in the Glioblastoma Tumor Microenvironment Reduces Tumoral Edema. Mol. Ther. Oncolytics 2018, 12, 93–102. [Google Scholar] [CrossRef] [PubMed]
  54. Jackson, J.W.; Hall, B.L.; Marzulli, M.; Shah, V.K.; Bailey, L.; Chiocca, E.A.; Goins, W.F.; Kohanbash, G.; Cohen, J.B.; Glorioso, J.C. Treatment of glioblastoma with current oHSV variants reveals differences in efficacy and immune cell recruitment. Mol. Ther. Oncolytics 2021, 22, 444–453. [Google Scholar] [CrossRef]
  55. Sanchez Gil, J.; Dubois, M.; Neirinckx, V.; Lombard, A.; Coppieters, N.; D’Arrigo, P.; Isci, D.; Aldenhoff, T.; Brouwers, B.; Lassence, C.; et al. Nanobody-based retargeting of an oncolytic herpesvirus for eliminating CXCR4+ GBM cells: A proof of principle. Mol. Ther. Oncolytics 2022, 26, 35–48. [Google Scholar] [CrossRef]
  56. Alayo, Q.A.; Ito, H.; Passaro, C.; Zdioruk, M.; Mahmoud, A.B.; Grauwet, K.; Zhang, X.; Lawler, S.E.; Reardon, D.A.; Goins, W.F.; et al. Glioblastoma infiltration of both tumor- and virus-antigen specific cytotoxic T cells correlates with experimental virotherapy responses. Sci. Rep. 2020, 10, 5095. [Google Scholar] [CrossRef]
  57. Markert, J.M.; Razdan, S.N.; Kuo, H.C.; Cantor, A.; Knoll, A.; Karrasch, M.; Nabors, L.B.; Markiewicz, M.; Agee, B.S.; Coleman, J.M.; et al. A phase 1 trial of oncolytic HSV-1, G207, given in combination with radiation for recurrent GBM demonstrates safety and radiographic responses. Mol. Ther. J. Am. Soc. Gene Ther. 2014, 22, 1048–1055. [Google Scholar] [CrossRef]
  58. Friedman, G.K.; Johnston, J.M.; Bag, A.K.; Bernstock, J.D.; Li, R.; Aban, I.; Kachurak, K.; Nan, L.; Kang, K.D.; Totsch, S.; et al. Oncolytic HSV-1 G207 Immunovirotherapy for Pediatric High-Grade Gliomas. N. Engl. J. Med. 2021, 384, 1613–1622. [Google Scholar] [CrossRef]
  59. Todo, T.; Ino, Y.; Ohtsu, H.; Shibahara, J.; Tanaka, M. A phase I/II study of triple-mutated oncolytic herpes virus G47∆ in patients with progressive glioblastoma. Nat. Commun. 2022, 13, 4119. [Google Scholar] [CrossRef]
  60. Estevez-Ordonez, D.; Stein, J.; Maleknia, P.; Gallegos, C.; Atchley, T.; Laskay, N.; Clements, J.; Lobbous, M.; Leavenworth, J.; Riley, K.; et al. CTIM-13. Phase I Clinical Trial of Oncolytic Hsv-1 M032, A Second-Generation Virus Armed to Expressed Il-12, for the Treatment of Adult Patients with Recurrent or Progressive Malignant Glioma. Neuro-Oncol. 2023, 25 (Suppl. S5), v64. [Google Scholar] [CrossRef]
  61. Ling, A.L.; Solomon, I.H.; Landivar, A.M.; Nakashima, H.; Woods, J.K.; Santos, A.; Masud, N.; Fell, G.; Mo, X.; Yilmaz, A.S.; et al. Clinical trial links oncolytic immunoactivation to survival in glioblastoma. Nature 2023, 623, 157–166. [Google Scholar] [CrossRef]
  62. Usman, N.; Suarez, M. Adenoviruses. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  63. Binder, A.M.; Biggs, H.M.; Haynes, A.K.; Chommanard, C.; Lu, X.; Erdman, D.D.; Watson, J.T.; Gerber, S.I. Human Adenovirus Surveillance—United States, 2003–2016. MMWR. Morb. Mortal. Wkly. Rep. 2017, 66, 1039–1042. [Google Scholar] [CrossRef]
  64. Lynch, J.P., 3rd; Kajon, A.E. Adenovirus: Epidemiology, Global Spread of Novel Serotypes, and Advances in Treatment and Prevention. Semin. Respir. Crit. Care Med. 2016, 37, 586–602. [Google Scholar] [CrossRef] [PubMed]
  65. Lee, C.S.; Bishop, E.S.; Zhang, R.; Yu, X.; Farina, E.M.; Yan, S.; Zhao, C.; Zheng, Z.; Shu, Y.; Wu, X.; et al. Adenovirus-Mediated Gene Delivery: Potential Applications for Gene and Cell-Based Therapies in the New Era of Personalized Medicine. Genes Dis. 2017, 4, 43–63. [Google Scholar] [CrossRef] [PubMed]
  66. Westergren Jakobsson, A.; Segerman, B.; Wallerman, O.; Lind, S.B.; Zhao, H.; Rubin, C.J.; Pettersson, U.; Akusjärvi, G. The Human Adenovirus Type 2 Transcriptome: An Amazing Complexity of Alternatively Spliced mRNAs. J. Virol. 2021, 95, e01869-20. [Google Scholar] [CrossRef]
  67. Matsunaga, W.; Gotoh, A. Adenovirus as a Vector and Oncolytic Virus. Curr. Issues Mol. Biol. 2023, 45, 4826–4840. [Google Scholar] [CrossRef]
  68. Tian, Y.; Xie, D.; Yang, L. Engineering strategies to enhance oncolytic viruses in cancer immunotherapy. Signal Transduct. Target. Ther. 2022, 7, 117. [Google Scholar] [CrossRef] [PubMed]
  69. Kim, J.W.; Miska, J.; Young, J.S.; Rashidi, A.; Kane, J.R.; Panek, W.K.; Kanojia, D.; Han, Y.; Balyasnikova, I.V.; Lesniak, M.S. A Comparative Study of Replication-Incompetent and -Competent Adenoviral Therapy-Mediated Immune Response in a Murine Glioma Model. Mol. Ther. Oncolytics 2017, 5, 97–104. [Google Scholar] [CrossRef]
  70. Wang, L.; Liu, W.; Li, Z.; Wang, X.; Feng, X.; Wang, Z.; Wu, J.; Zhang, H.; Wu, H.; Kong, W.; et al. A tropism-transformed Oncolytic Adenovirus with Dual Capsid Modifications for enhanced Glioblastoma Therapy. J. Cancer 2020, 11, 5713–5726. [Google Scholar] [CrossRef]
  71. Wang, S.; Yan, W.; Kong, L.; Zuo, S.; Wu, J.; Zhu, C.; Huang, H.; He, B.; Dong, J.; Wei, J. Oncolytic viruses engineered to enforce cholesterol efflux restore tumor-associated macrophage phagocytosis and anti-tumor immunity in glioblastoma. Nat. Commun. 2023, 14, 4367. [Google Scholar] [CrossRef]
  72. Kiyokawa, J.; Kawamura, Y.; Ghouse, S.M.; Acar, S.; Barçın, E.; Martínez-Quintanilla, J.; Martuza, R.L.; Alemany, R.; Rabkin, S.D.; Shah, K.; et al. Modification of Extracellular Matrix Enhances Oncolytic Adenovirus Immunotherapy in Glioblastoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 889–902. [Google Scholar] [CrossRef] [PubMed]
  73. Li, M.; Li, G.; Kiyokawa, J.; Tirmizi, Z.; Richardson, L.G.; Ning, J.; Das, S.; Martuza, R.L.; Stemmer-Rachamimov, A.; Rabkin, S.D.; et al. Characterization and oncolytic virus targeting of FAP-expressing tumor-associated pericytes in glioblastoma. Acta Neuropathol. Commun. 2020, 8, 221. [Google Scholar] [CrossRef]
  74. Chen, Y.; Chen, X.; Bao, W.; Liu, G.; Wei, W.; Ping, Y. An oncolytic virus-T cell chimera for cancer immunotherapy. Nat. Biotechnol. 2024, 42, 1876–1887. [Google Scholar] [CrossRef]
  75. Zhang, Q.; Zhang, J.; Tian, Y.; Wang, J.; Jin, G.; Liu, F. Ki67-targeted oncolytic adenovirus expressing IL-15 improves intratumoral T cell infiltration and PD-L1 expression in glioblastoma. Virology 2023, 587, 109885. [Google Scholar] [CrossRef]
  76. Wang, P.; Zhang, J.; Zhang, Q.; Liu, F. Mesenchymal stem cells loaded with Ad5-Ki67/IL-15 enhance oncolytic adenovirotherapy in experimental glioblastoma. Biomed. Pharmacother. 2023, 157, 114035. [Google Scholar] [CrossRef]
  77. Wang, J.; Zhang, J.; Zhang, Q.; Zhang, W.; Zhang, Q.; Jin, G.; Liu, F. TS-2021, a third-generation oncolytic adenovirus that carried Ki67 promoter, TGF-β2 5’UTR, and IL-15 against experimental glioblastoma. J. Med. Virol. 2024, 96, e29335. [Google Scholar] [CrossRef] [PubMed]
  78. El-Ayoubi, A.; Klawitter, M.; Rüttinger, J.; Wellhäusser, G.; Holm, P.S.; Danielyan, L.; Naumann, U. Intranasal Delivery of Oncolytic Adenovirus XVir-N-31 via Optimized Shuttle Cells Significantly Extends Survival of Glioblastoma-Bearing Mice. Cancers 2023, 15, 4912. [Google Scholar] [CrossRef]
  79. Liu, Y.; Fang, S.; Wang, P.; Zhang, J.; Liu, F. Olaparib Enhances the Efficacy of Third-Generation Oncolytic Adenoviruses Against Glioblastoma by Modulating DNA Damage Response and p66shc-Induced Apoptosis. CNS Neurosci. Ther. 2024, 30, e70124. [Google Scholar] [CrossRef]
  80. Klawitter, M.; El-Ayoubi, A.; Buch, J.; Rüttinger, J.; Ehrenfeld, M.; Lichtenegger, E.; Krüger, M.A.; Mantwill, K.; Koll, F.J.; Kowarik, M.C.; 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. [Google Scholar] [CrossRef] [PubMed]
  81. Jiang, S.; Chai, H.H.; Fang, X.L.; Xu, H.S.; Li, T.W.; Tang, Q.S.; Gu, J.F.; Zhang, K.J.; Liu, X.Y.; Shi, Z.F.; et al. Double-modified oncolytic adenovirus armed with a recombinant interferon-like gene enhanced abscopal effects against malignant glioma. Neuro-Oncol. Adv. 2023, 5, vdad117. [Google Scholar] [CrossRef]
  82. Oh, E.; Hong, J.; Kwon, O.J.; Yun, C.O. A hypoxia- and telomerase-responsive oncolytic adenovirus expressing secretable trimeric TRAIL triggers tumor-specific apoptosis and promotes viral dispersion in TRAIL-resistant glioblastoma. Sci. Rep. 2018, 8, 1420. [Google Scholar] [CrossRef] [PubMed]
  83. Romanenko, M.V.; Dolgova, E.V.; Osipov, I.D.; Ritter, G.S.; Sizova, M.S.; Proskurina, A.S.; Efremov, Y.R.; Bayborodin, S.I.; Potter, E.A.; Taranov, O.S.; et al. Oncolytic Effect of Adenoviruses Serotypes 5 and 6 Against U87 Glioblastoma Cancer Stem Cells. Anticancer Res. 2019, 39, 6073–6086. [Google Scholar] [CrossRef]
  84. Bates, E.A.; Lovatt, C.; Plein, A.R.; Davies, J.A.; Siebzehnrubl, F.A.; Parker, A.L. Engineering Adenoviral Vectors with Improved GBM Selectivity. Viruses 2023, 15, 1086. [Google Scholar] [CrossRef]
  85. González-Morales, A.; Zabaleta, A.; García-Moure, M.; Alonso, M.M.; Fernández-Irigoyen, J.; Santamaría, E. Oncolytic adenovirus Delta-24-RGD induces a widespread glioma proteotype remodeling during autophagy. J. Proteom. 2019, 194, 168–178. [Google Scholar] [CrossRef] [PubMed]
  86. Bhoopathi, P.; Mannangatti, P.; Pradhan, A.K.; Kumar, A.; Maji, S.; Lang, F.F.; Klibanov, A.L.; Madan, E.; Cavenee, W.K.; Keoprasert, T.; et al. Noninvasive therapy of brain cancer using a unique systemic delivery methodology with a cancer terminator virus. J. Cell. Physiol. 2024, 239, e31302. [Google Scholar] [CrossRef]
  87. Shimizu, Y.; Gumin, J.; Gao, F.; Hossain, A.; Shpall, E.J.; Kondo, A.; Parker Kerrigan, B.C.; Yang, J.; Ledbetter, D.; Fueyo, J.; et al. Characterization of patient-derived bone marrow human mesenchymal stem cells as oncolytic virus carriers for the treatment of glioblastoma. J. Neurosurg. 2021, 136, 757–767. [Google Scholar] [CrossRef]
  88. Meléndez-Vázquez, N.M.; Nguyen, T.T.; Fan, X.; López-Rivas, A.R.; Fueyo, J.; Gomez-Manzano, C.; Godoy-Vitorino, F. Gut microbiota composition is associated with the efficacy of Delta-24-RGDOX in malignant gliomas. Molecular therapy. Oncology 2024, 32, 200787. [Google Scholar] [CrossRef] [PubMed]
  89. Rivera-Molina, Y.; Jiang, H.; Fueyo, J.; Nguyen, T.; Shin, D.H.; Youssef, G.; Fan, X.; Gumin, J.; Alonso, M.M.; Phadnis, S.; et al. GITRL-armed Delta-24-RGD oncolytic adenovirus prolongs survival and induces anti-glioma immune memory. Neuro-Oncol. Adv. 2019, 1, vdz009. [Google Scholar] [CrossRef]
  90. Berghauser Pont, L.M.; Kleijn, A.; Kloezeman, J.J.; van den Bossche, W.; Kaufmann, J.K.; de Vrij, J.; Leenstra, S.; Dirven, C.M.; Lamfers, M.L. The HDAC Inhibitors Scriptaid and LBH589 Combined with the Oncolytic Virus Delta24-RGD Exert Enhanced Anti-Tumor Efficacy in Patient-Derived Glioblastoma Cells. PLoS ONE 2015, 10, e0127058. [Google Scholar] [CrossRef]
  91. Qiao, J.; Dey, M.; Chang, A.L.; Kim, J.W.; Miska, J.; Ling, A.M.; Nettlebeck, D.; Han, Y.; Zhang, L.; Lesniak, M.S.; et al. Intratumoral oncolytic adenoviral treatment modulates the glioma microenvironment and facilitates systemic tumor-antigen-specific T cell therapy. Oncoimmunology 2015, 4, e1022302. [Google Scholar] [CrossRef]
  92. Srinivasan, V.M.; Gumin, J.; Camstra, K.M.; Collins, D.E.; Chen, M.M.; Shpall, E.J.; Parker Kerrigan, B.C.; Johnson, J.N.; Chen, S.R.; Fueyo, J.; et al. Endovascular Selective Intra-Arterial Infusion of Mesenchymal Stem Cells Loaded with Delta-24 in a Canine Model. Neurosurgery 2020, 88, E102–E113. [Google Scholar] [CrossRef] [PubMed]
  93. Stepanenko, A.A.; Sosnovtseva, A.O.; Valikhov, M.P.; Chernysheva, A.A.; Cherepanov, S.A.; Yusubalieva, G.M.; Ruzsics, Z.; Lipatova, A.V.; Chekhonin, V.P. Superior infectivity of the fiber chimeric oncolytic adenoviruses Ad5/35 and Ad5/3 over Ad5-delta-24-RGD in primary glioma cultures. Mol. Ther. Oncolytics 2021, 24, 230–248. [Google Scholar] [CrossRef] [PubMed]
  94. Koch, M.S.; Zdioruk, M.; Nowicki, M.O.; Griffith, A.M.; Aguilar, E.; Aguilar, L.K.; Guzik, B.W.; Barone, F.; Tak, P.P.; Tabatabai, G.; et al. Systemic high-dose dexamethasone treatment may modulate the efficacy of intratumoral viral oncolytic immunotherapy in glioblastoma models. J. Immunother. Cancer 2022, 10, e003368. [Google Scholar] [CrossRef]
  95. Belcaid, Z.; Berrevoets, C.; Choi, J.; van Beelen, E.; Stavrakaki, E.; Pierson, T.; Kloezeman, J.; Routkevitch, D.; van der Kaaij, M.; van der Ploeg, A.; et al. Low-dose oncolytic adenovirus therapy overcomes tumor-induced immune suppression and sensitizes intracranial gliomas to anti-PD-1 therapy. Neuro-Oncol. Adv. 2020, 2, vdaa011. [Google Scholar] [CrossRef]
  96. Qiao, C.; Xu, Y.; He, Y.; Cai, Z.; Wang, H. Oncolytic adenovirus H101 enhances the anti-tumor effects of PD-1 blockade via CD47 downregulation in tumor cells. Oncol. Res. 2025, 33, 1161–1172. [Google Scholar] [CrossRef] [PubMed]
  97. Koch, M.S.; Zdioruk, M.; Nowicki, M.O.; Griffith, A.M.; Aguilar-Cordova, E.; Aguilar, L.K.; Guzik, B.W.; Barone, F.; Tak, P.P.; Schregel, K.; et al. Perturbing DDR signaling enhances cytotoxic effects of local oncolytic virotherapy and modulates the immune environment in glioma. Mol. Ther. Oncolytics 2022, 26, 275–288. [Google Scholar] [CrossRef]
  98. Niittykoski, M.; von Und Zu Fraunberg, M.; Martikainen, M.; Rauramaa, T.; Immonen, A.; Koponen, S.; Leinonen, V.; Vähä-Koskela, M.; Zhang, Q.; Kühnel, F.; et al. Immunohistochemical Characterization and Sensitivity to Human Adenovirus Serotypes 3, 5, and 11p of New Cell Lines Derived from Human Diffuse Grade II to IV Gliomas. Transl. Oncol. 2017, 10, 772–779. [Google Scholar] [CrossRef]
  99. Kleijn, A.; van den Bossche, W.; Haefner, E.S.; Belcaid, Z.; Burghoorn-Maas, C.; Kloezeman, J.J.; Pas, S.D.; Leenstra, S.; Debets, R.; de Vrij, J.; et al. The Sequence of Delta24-RGD and TMZ Administration in Malignant Glioma Affects the Role of CD8+T Cell Anti-tumor Activity. Mol. Ther. Oncolytics 2017, 5, 11–19. [Google Scholar] [CrossRef]
  100. Stepanenko, A.A.; Sosnovtseva, A.O.; Valikhov, M.P.; Vasiukova, A.A.; Abramova, O.V.; Lipatova, A.V.; Yusubalieva, G.M.; Chekhonin, V.P. Comparison of the L3-23K and L5-Fiber Regions for Arming the Oncolytic Adenovirus Ad5-Delta-24-RGD with Reporter and Therapeutic Transgenes. Int. J. Mol. Sci. 2025, 26, 3700. [Google Scholar] [CrossRef]
  101. Wang, G.; Zhang, Z.; Zhong, K.; Wang, Z.; Yang, N.; Tang, X.; Li, H.; Lu, Q.; Wu, Z.; Yuan, B.; et al. CXCL11-armed oncolytic adenoviruses enhance CAR-T cell therapeutic efficacy and reprogram tumor microenvironment in glioblastoma. Mol. Ther. J. Am. Soc. Gene Ther. 2023, 31, 134–153. [Google Scholar] [CrossRef]
  102. Huang, J.; Zheng, M.; Zhang, Z.; Tang, X.; Chen, Y.; Peng, A.; Peng, X.; Tong, A.; Zhou, L. Interleukin-7-loaded oncolytic adenovirus improves CAR-T cell therapy for glioblastoma. Cancer Immunol. Immunother. 2021, 70, 2453–2465. [Google Scholar] [CrossRef] [PubMed]
  103. Wang, G.; Mu, M.; Zhang, Z.; Chen, Y.; Yang, N.; Zhong, K.; Li, Y.; Lu, F.; Guo, G.; Tong, A. Systemic delivery of tannic acid-ferric-masked oncolytic adenovirus reprograms tumor microenvironment for improved therapeutic efficacy in glioblastoma. Cancer Gene Ther. 2024, 31, 1804–1817. [Google Scholar] [CrossRef]
  104. Batalla-Covello, J.; Ngai, H.W.; Flores, L.; McDonald, M.; Hyde, C.; Gonzaga, J.; Hammad, M.; Gutova, M.; Portnow, J.; Synold, T.; et al. Multiple Treatment Cycles of Neural Stem Cell Delivered Oncolytic Adenovirus for the Treatment of Glioblastoma. Cancers 2021, 13, 6320. [Google Scholar] [CrossRef] [PubMed]
  105. Koch, M.S.; Zdioruk, M.; Nowicki, M.O.; Hoetker, M.S.; Herbert, Z.T.; Barone, F.; Tak, P.P.; Chiocca, E.A.; Tabatabai, G.; Lawler, S.E. Uncovering transcriptomic landscape alterations of CAN-2409 in in vitro and in vivo glioma models. Front. Med. 2023, 10, 1140352. [Google Scholar] [CrossRef]
  106. Fares, J.; Ahmed, A.U.; Ulasov, I.V.; Sonabend, A.M.; Miska, J.; Lee-Chang, C.; Balyasnikova, I.V.; Chandler, J.P.; Portnow, J.; Tate, M.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. The Lancet. Oncology 2021, 22, 1103–1114. [Google Scholar] [CrossRef]
  107. van Putten, E.H.P.; Kleijn, A.; van Beusechem, V.W.; Noske, D.; Lamers, C.H.J.; de Goede, A.L.; Idema, S.; Hoefnagel, D.; Kloezeman, J.J.; Fueyo, J.; et al. Convection Enhanced Delivery of the Oncolytic Adenovirus Delta24-RGD in Patients with Recurrent GBM: A Phase I Clinical Trial Including Correlative Studies. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2022, 28, 1572–1585. [Google Scholar] [CrossRef]
  108. van der Meulen-Muileman, I.H.; Amado-Azevedo, J.; Lamfers, M.L.M.; Kleijn, A.; Idema, S.; Noske, D.P.; Dirven, C.M.F.; van Beusechem, V.W. Adenovirus-Neutralizing and Infection-Promoting Activities Measured in Serum of Human Brain Cancer Patients Treated with Oncolytic Adenovirus Ad5-∆24.RGD. Int. J. Mol. Sci. 2025, 26, 854. [Google Scholar] [CrossRef]
  109. Peña Pino, I.; Darrow, D.P.; Chen, C.C. Magnetic Resonance Imaging-Aided SmartFlow Convection Delivery of DNX-2401: A Pilot, Prospective Case Series. World Neurosurg. 2024, 181, e833–e840. [Google Scholar] [CrossRef]
  110. Nassiri, F.; Patil, V.; Yefet, L.S.; Singh, O.; Liu, J.; Dang, R.M.A.; Yamaguchi, T.N.; Daras, M.; Cloughesy, T.F.; Colman, H.; et al. Oncolytic DNX-2401 virotherapy plus pembrolizumab in recurrent glioblastoma: A phase 1/2 trial. Nat. Med. 2023, 29, 1370–1378. [Google Scholar] [CrossRef]
  111. Taylor, O.G.; Brzozowski, J.S.; Skelding, K.A. Glioblastoma multiforme: An overview of emerging therapeutic targets. Front. Oncol. 2019, 9, 963. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  112. Bhattacharjee, S.; Yadava, P.K. Measles virus: Background and oncolytic virotherapy. Biochem. Biophys. Rep. 2018, 13, 58–62. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  113. Allen, C.; Paraskevakou, G.; Liu, C.; Iankov, I.D.; Msaouel, P.; Zollman, P.; Myers, R.; Peng, K.W.; Russell, S.J.; Galanis, E. Oncolytic measles virus strains in the treatment of gliomas. Expert Opin. Biol. Ther. 2008, 8, 213–220. [Google Scholar] [CrossRef]
  114. Anker, S.C.; Szczeponik, M.G.; Dessila, J.; Dittus, K.; Engeland, C.E.; Jäger, D.; Ungerechts, G.; Leber, M.F. Oncolytic measles virus encoding microRNA for targeted RNA interference. Viruses 2023, 15, 308. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  115. Forsyth, P.A.; Abate-Daga, D. Viral therapy gets personal: A potential gene signature to predict susceptibility to measles virus oncolysis. J. Natl. Cancer Inst. 2018, 110, 1139–1140. [Google Scholar] [CrossRef] [PubMed]
  116. Muñoz-Alía, M.Á.; Nace, R.A.; Tischer, A.; Zhang, L.; Bah, E.S.; Auton, M.; Russell, S.J. MeV-Stealth: A CD46-specific oncolytic measles virus resistant to neutralization by measles-immune human serum. PLoS Pathog. 2021, 17, e1009283. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  117. Ammour, Y.; Susova, O.; Krasnov, G.; Nikolaeva, E.; Varachev, V.; Schetinina, Y.; Gavrilova, M.; Mitrofanov, A.; Poletaeva, A.; Bekyashev, A.; et al. Transcriptome analysis of human glioblastoma cells susceptible to infection with the Leningrad-16 vaccine strain of measles virus. Viruses 2022, 14, 2433. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  118. Galanis, E.; Dooley, K.E.; Keith Anderson, S.; Kurokawa, C.B.; Carrero, X.W.; Uhm, J.H.; Federspiel, M.J.; Leontovich, A.A.; Aderca, I.; Viker, K.B.; et al. Carcinoembryonic antigen-expressing oncolytic measles virus derivative in recurrent glioblastoma: A phase 1 trial. Nat. Commun. 2024, 15, 493. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  119. Abdullah, J.M.; Mustafa, Z.; Ideris, A. Newcastle disease virus interaction in targeted therapy against proliferation and invasion pathways of glioblastoma multiforme. Biomed. Res. Int. 2014, 2014, 386470. [Google Scholar] [CrossRef] [PubMed]
  120. Kim, S.H.; Samal, S.K. Innovation in Newcastle Disease Virus Vectored Avian Influenza Vaccines. Viruses 2019, 11, 300. [Google Scholar] [CrossRef]
  121. García-Romero, N.; Palacín-Aliana, I.; Esteban-Rubio, S.; Madurga, R.; Rius-Rocabert, S.; Carrión-Navarro, J.; Presa, J.; Cuadrado-Castano, S.; Sánchez-Gómez, P.; García-Sastre, A.; et al. Newcastle disease virus (NDV) oncolytic activity in human glioma tumors is dependent on CDKN2A-Type I IFN gene cluster codeletion. Cells 2020, 9, 1405. [Google Scholar] [CrossRef]
  122. Yang, H.; Tian, J.; Zhao, J.; Zhao, Y.; Zhang, G. The Application of Newcastle Disease Virus (NDV): Vaccine Vectors and Tumor Therapy. Viruses 2024, 16, 886. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  123. Soliman, R.M.; Nishioka, K.; Daidoji, T.; Noyori, O.; Nakaya, T. Chimeric Newcastle Disease Virus Vectors Expressing Human IFN-γ Mediate Target Immune Responses and Enable Multifaceted Treatments. Biomedicines 2023, 11, 455. [Google Scholar] [CrossRef]
  124. Kadhim, Z.A.; Sulaiman, G.M.; Al-Shammari, A.M.; Khan, R.A.; Al Rugaie, O.; Mohammed, H.A. Oncolytic Newcastle Disease Virus Co-Delivered with Modified PLGA Nanoparticles Encapsulating Temozolomide against Glioblastoma Cells: Developing an Effective Treatment Strategy. Molecules 2022, 27, 5757. [Google Scholar] [CrossRef]
  125. Jang, S.H.; Jung, B.K.; An, Y.H.; Jang, H. The phosphatase and tensin homolog gene inserted between NP and P gene of recombinant Newcastle disease virus oncolytic effect test to glioblastoma cell and xenograft mouse model. Virol. J. 2022, 19, 21. [Google Scholar] [CrossRef] [PubMed]
  126. Freeman, A.I.; Zakay-Rones, Z.; Gomori, J.M.; Linetsky, E.; Rasooly, L.; Greenbaum, E.; Rozenman-Yair, S.; Panet, A.; Libson, E.; Irving, C.S.; et al. Phase I/II trial of intravenous NDV-HUJ oncolytic virus in recurrent glioblastoma multiforme. Mol. Ther. J. Am. Soc. Gene Ther. 2006, 13, 221–228. [Google Scholar] [CrossRef] [PubMed]
  127. Csatary, L.K.; Gosztonyi, G.; Szeberenyi, J.; Fabian, Z.; Liszka, V.; Bodey, B.; Csatary, C.M. MTH-68/H oncolytic viral treatment in human high-grade gliomas. J. Neuro-Oncol. 2004, 67, 83–93. [Google Scholar] [CrossRef] [PubMed]
  128. Schneider, T.; Gerhards, R.; Kirches, E.; Firsching, R. Preliminary results of active specific immunization with modified tumor cell vaccine in glioblastoma multiforme. J. Neuro-Oncol. 2001, 53, 39–46. [Google Scholar] [CrossRef]
  129. Steiner, H.H.; Bonsanto, M.M.; Beckhove, P.; Brysch, M.; Geletneky, K.; Ahmadi, R.; Schuele-Freyer, R.; Kremer, P.; Ranaie, G.; Matejic, D.; et al. Antitumor vaccination of patients with glioblastoma multiforme: A pilot study to assess feasibility, safety, and clinical benefit. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2004, 22, 4272–4281. [Google Scholar] [CrossRef]
  130. Keshavarz, M.; Nejad, A.S.M.; Esghaei, M.; Bokharaei-Salim, F.; Dianat-Moghadam, H.; Keyvani, H.; Ghaemi, A. Oncolytic Newcastle disease virus reduces growth of cervical cancer cell by inducing apoptosis. Saudi J. Biol. Sci. 2020, 27, 47–52. [Google Scholar] [CrossRef]
  131. Cassel, W.A.; Murray, D.R. A ten-year follow-up on stage II malignant melanoma patients treated postsurgically with Newcastle disease virus oncolysate. Med. Oncol. Tumor Pharmacother. 1992, 9, 169–171. [Google Scholar] [CrossRef]
  132. Pomer, S.; Schirrmacher, V.; Thiele, R.; Lohrke, H.; Brkovic, D.; Staehler, G. Tumor response and 4 year survival-data of patients with advanced renal-cell carcinoma treated with autologous tumor vaccine and subcutaneous R-IL-2 and IFN-alpha(2b). Int. J. Oncol. 1995, 6, 947–954. [Google Scholar] [CrossRef] [PubMed]
  133. Möbus, V.; Horn, S.; Stöck, M.; Schirrmacher, V. Tumor cell vaccination for gynecological tumors. Hybridoma 1993, 12, 543–547. [Google Scholar] [CrossRef] [PubMed]
  134. Müller, L.; Berkeley, R.; Barr, T.; Ilett, E.; Errington-Mais, F. Past, Present and Future of Oncolytic Reovirus. Cancers 2020, 12, 3219. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  135. Kicielinski, K.P.; Chiocca, E.A.; Yu, J.S.; Gill, G.M.; Coffey, M.; Markert, J.M. Phase 1 clinical trial of intratumoral reovirus infusion for the treatment of recurrent malignant gliomas in adults. Mol. Ther. 2014, 22, 1056–1062. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  136. Kendall, J.; Chalmers, A.; McBain, C.; Melcher, A.; Samson, A.; Phillip, R.; Brown, S.; Short, S. CTIM-14. Pelareorep and Granulocyte-Macrophage Colony-Stimulating Factor (Gm-Csf) with Standard Chemoradiotherapy/Adjuvant Temozolomide for Glioblastoma Multiforme (Gbm) Patients: Reoglio Phase I Trial Results. Neuro Oncol. 2020, 22 (Suppl. S2), ii35–ii36. [Google Scholar] [CrossRef] [PubMed Central]
  137. Samson, A.; Scott, K.J.; Taggart, D.; West, E.J.; Wilson, E.; Nuovo, G.J.; Thomson, S.; Corns, R.; Mathew, R.K.; Fuller, M.J.; et al. Intravenous delivery of oncolytic reovirus to brain tumor patients immunologically primes for subsequent checkpoint blockade. Sci. Transl. Med. 2018, 10, eaam7577. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  138. Schuelke, M.R.; Gundelach, J.H.; Coffey, M.; West, E.; Scott, K.; Johnson, D.R.; Samson, A.; Melcher, A.; Vile, R.G.; Bram, R.J. Phase I trial of sargramostim/pelareorep therapy in pediatric patients with recurrent or refractory high-grade brain tumors. Neuro-Oncol. Adv. 2022, 4, vdac085. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  139. Tai, C.K.; Wang, W.J.; Chen, T.C.; Kasahara, N. Single-shot, multicycle suicide gene therapy by replication-competent retrovirus vectors achieves long-term survival benefit in experimental glioma. Mol. Ther. J. Am. Soc. Gene Ther. 2005, 12, 842–851. [Google Scholar] [CrossRef]
  140. Perez, O.D.; Logg, C.R.; Hiraoka, K.; Diago, O.; Burnett, R.; Inagaki, A.; Jolson, D.; Amundson, K.; Buckley, T.; Lohse, D.; et al. Design and selection of Toca 511 for clinical use: Modified retroviral replicating vector with improved stability and gene expression. Mol. Ther. J. Am. Soc. Gene Ther. 2012, 20, 1689–1698. [Google Scholar] [CrossRef]
  141. Budzik, K.M.; Nace, R.A.; Ikeda, Y.; Russell, S.J. Oncolytic Foamy Virus—Generation and properties of a nonpathogenic replicating retroviral vector system that targets chronically proliferating cancer cells. J. Virol. 2021, 95, e00015-21. [Google Scholar] [CrossRef]
  142. Budzik, K.M.; Nace, R.A.; Ikeda, Y.; Russell, S.J. Evaluation of the stability and intratumoral delivery of foreign transgenes encoded by an oncolytic Foamy Virus vector. Cancer Gene Ther. 2022, 29, 1240–1251. [Google Scholar] [CrossRef]
  143. Cloughesy, T.F.; Landolfi, J.; Hogan, D.J.; Bloomfield, S.; Carter, B.; Chen, C.C.; Elder, J.B.; Kalkanis, S.N.; Kesari, S.; Lai, A.; et al. Phase 1 trial of vocimagene amiretrorepvec and 5-fluorocytosine for recurrent high-grade glioma. Sci. Transl. Med. 2016, 8, 341ra75. [Google Scholar] [CrossRef]
  144. Cloughesy, T.F.; Landolfi, J.; Vogelbaum, M.A.; Ostertag, D.; Elder, J.B.; Bloomfield, S.; Carter, B.; Chen, C.C.; Kalkanis, S.N.; Kesari, S.; et al. Durable complete responses in some recurrent high-grade glioma patients treated with Toca 511 + Toca FC. Neuro-Oncol. 2018, 20, 1383–1392. [Google Scholar] [CrossRef] [PubMed]
  145. Accomando, W.P.; Rao, A.R.; Hogan, D.J.; Newman, A.M.; Nakao, A.; Alizadeh, A.A.; Diehn, M.; Diago, O.R.; Gammon, D.; Haghighi, A.; et al. Molecular and Immunologic Signatures are Related to Clinical Benefit from Treatment with Vocimagene Amiretrorepvec (Toca 511) and 5-Fluorocytosine (Toca FC) in Patients with Glioma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 6176–6186. [Google Scholar] [CrossRef] [PubMed]
  146. Hogan, D.J.; Zhu, J.J.; Diago, O.R.; Gammon, D.; Haghighi, A.; Lu, G.; Das, A.; Gruber, H.E.; Jolly, D.J.; Ostertag, D. Molecular Analyses Support the Safety and Activity of Retroviral Replicating Vector Toca 511 in Patients. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2018, 24, 4680–4693. [Google Scholar] [CrossRef] [PubMed]
  147. Sun, Y.; Zhang, Z.; Zhang, C.; Zhang, N.; Wang, P.; Chu, Y.; Chard Dunmall, L.S.; Lemoine, N.R.; Wang, Y. An effective therapeutic regime for treatment of glioma using oncolytic vaccinia virus expressing IL-21 in combination with immune checkpoint inhibition. Mol. Ther. Oncolytics 2022, 26, 105–119. [Google Scholar] [CrossRef] [PubMed]
  148. Storozynsky, Q.T.; Agopsowicz, K.C.; Noyce, R.S.; Bukhari, A.B.; Han, X.; Snyder, N.; Umer, B.A.; Gamper, A.M.; Godbout, R.; Evans, D.H.; et al. Radiation combined with oncolytic vaccinia virus provides pronounced antitumor efficacy and induces immune protection in an aggressive glioblastoma model. Cancer Lett. 2023, 562, 216169. [Google Scholar] [CrossRef]
  149. Vasileva, N.; Ageenko, A.; Dmitrieva, M.; Nushtaeva, A.; Mishinov, S.; Kochneva, G.; Richter, V.; Kuligina, E. Double Recombinant Vaccinia Virus: A Candidate Drug against Human Glioblastoma. Life 2021, 11, 1084. [Google Scholar] [CrossRef]
  150. Chen, L.; Wang, P.; Di Gioia, C.; Yuan, M.; Zhang, Z.; Miao, J.; Yan, W.; Zhao, G.; Jia, Y.; Wang, N.; et al. A novel oncolytic Vaccinia virus armed with IL-12 augments antitumor immune responses leading to durable regression in murine models of lung cancer. Front. Immunol. 2025, 15, 1492464. [Google Scholar] [CrossRef]
  151. Duggal, R.; Geissinger, U.; Zhang, Q.; Aguilar, J.; Chen, N.G.; Binda, E.; Vescovi, A.L.; Szalay, A.A. Vaccinia virus expressing bone morphogenetic protein-4 in novel glioblastoma orthotopic models facilitates enhanced tumor regression and long-term survival. J. Transl. Med. 2013, 11, 155. [Google Scholar] [CrossRef]
  152. Cotmore, S.F.; Agbandje-McKenna, M.; Chiorini, J.A.; Mukha, D.V.; Pintel, D.J.; Qiu, J.; Soderlund-Venermo, M. The family Parvoviridae. Arch. Virol. 2019, 164, 1237–1247. [Google Scholar] [CrossRef] [PubMed]
  153. Raykov, Z.; Grekova, S.; Leuchs, B.; Aprahamian, M.; Rommelaere, J. Arming parvoviruses with CpG motifs to improve their oncosuppressive capacity. Int. J. Cancer 2008, 122, 2880–2884. [Google Scholar] [CrossRef] [PubMed]
  154. Grekova, S.P.; Aprahamian, M.; Daeffler, L.; Leuchs, B.; Angelova, A.L.; Giese, N.A.; Rommelaere, J.; Raykov, Z. Interferon γ improves the vaccination potential of oncolytic H-1 parvovirus by modulating the tumor microenvironment. Cancer Gene Ther. 2012, 19, 589–602. [Google Scholar] [CrossRef] [PubMed]
  155. Rayet, B.; Lopez-Guerrero, J.A.; Rommelaere, J.; Dinsart, C. Induction of programmed cell death by parvovirus H-1 in U937 cells: Connection with the tumor necrosis factor alpha signalling pathway. J Virol. 1998, 72, 8893–8903. [Google Scholar] [CrossRef]
  156. Angelova, A.L.; Barf, M.; Geletneky, K.; Unterberg, A.; and Rommelaere, J. Immunotherapeutic Potential of Oncolytic H-1 Parvovirus: Hints of Glioblastoma Microenvironment Conversion towards Immunogenicity. Viruses 2017, 9, 382. [Google Scholar] [CrossRef]
  157. Paglino, J.C.; Ozduman, K.; van den Pol, A.N. LuIII parvovirus selectively and efficiently targets, replicates in, and kills human glioma cells. J. Virol. 2012, 86, 7280–7291. [Google Scholar] [CrossRef] [PubMed]
  158. Geletneky, K.; Hajda, J.; Angelova, A.L.; Leuchs, B.; Capper, D.; Bartsch, A.J.; Neumann, J.-O.; Schöning, T.; Hüsing, J.; Beelte, B.; et al. Oncolytic H-1 Parvovirus Shows Safety and Signs of Immunogenic Activity in a First Phase I/IIa Glioblastoma Trial. Mol. Ther. 2017, 25, 2620–2634. [Google Scholar] [CrossRef]
  159. Geletneky, K.; Huesing, J.; Rommelaere, J.; Schlehofer, J.R.; Leuchs, B.; Dahm, M.; Krebs, O.; Doeberitz, M.V.K.; Huber, B.; Hajda, J. Phase I/IIa study of intratumoral/intracerebral or intravenous/intracerebral administration of Parvovirus H-1 (ParvOryx) in patients with progressive primary or recurrent glioblastoma multiforme: ParvOryx01 protocol. BMC Cancer 2012, 12, 99. [Google Scholar] [CrossRef] [PubMed]
  160. Gromeier, M.; Alexander, L.; Wimmer, E. Internal ribosomal entry site substitution eliminates neurovirulence in intergeneric poliovirus recombinants. Proc. Natl. Acad. Sci. USA 1996, 93, 2370–2375. [Google Scholar] [CrossRef]
  161. Chandramohan, V.; Bryant, J.D.; Piao, H.; Keir, S.T.; Lipp, E.S.; Lefaivre, M.; Perkinson, K.; Bigner, D.D.; Gromeier, M.; McLendon, R.E. Validation of an Immunohistochemistry Assay for Detection of CD155, the Poliovirus Receptor, in Malignant Gliomas. Arch. Pathol. Lab. Med. 2017, 141, 1697–1704. [Google Scholar] [CrossRef]
  162. Liu, F.; Huang, J.; Xiong, Y.; Li, S.; Liu, Z. Large-scale analysis reveals the specific clinical and immune features of CD155 in glioma. Aging 2019, 11, 5463–5482. [Google Scholar] [CrossRef]
  163. Merrill, M.K.; Bernhardt, G.; Sampson, J.H.; Wikstrand, C.J.; Bigner, D.D.; Gromeier, M. Poliovirus receptor CD155-targeted oncolysis of glioma. Neuro-Oncology 2004, 6, 208–217. [Google Scholar] [CrossRef]
  164. Brown, M.C.; Holl, E.K.; Boczkowski, D.; Dobrikova, E.; Mosaheb, M.; Chandramohan, V.; Bigner, D.D.; Gromeier, M.; Nair, S.K. Cancer immunotherapy with recombinant poliovirus induces IFN-dominant activation of dendritic cells and tumor antigen–specific CTLs. Sci. Transl. Med. 2017, 9. [Google Scholar] [CrossRef]
  165. Desjardins, A.; Sampson, J.H.; Peters, K.B.; Ranjan, T.; Vlahovic, G.; Threatt, S.; Herndon, J.E.; Boulton, S.; Lally-Goss, D.; McSherry, F.; et al. Oncolytic Polio/Rhinovirus Recombinant (PVSRIPO) in Recurrent Glioblastoma (GBM): First Phase I Clinical Trial Evaluating the Intratumoral Administration. Neuro-Oncology 2014, 16, iii43. [Google Scholar] [CrossRef]
  166. Desjardins, A.; Gromeier, M.; Herndon, J.E.; Beaubier, N.; Bolognesi, D.P.; Friedman, A.H.; Friedman, H.S.; McSherry, F.; Muscat, A.M.; Nair, S.; et al. Recurrent Glioblastoma Treated with Recombinant Poliovirus. N. Engl. J. Med. 2018, 379, 150–161. [Google Scholar] [CrossRef]
  167. Beasley, G.M.; Nair, S.K.; E Farrow, N.; Landa, K.; Selim, M.A.; Wiggs, C.A.; Jung, S.-H.; Bigner, D.D.; Kelly, A.T.; Gromeier, M.; et al. Phase I trial of intratumoral PVSRIPO in patients with unresectable, treatment-refractory melanoma. J. Immunother. Cancer 2021, 9, e002203. [Google Scholar] [CrossRef]
  168. Bhagat, R.; Kaur, G.; Seth, P. Molecular mechanisms of zika virus pathogenesis: An update. Indian J. Med. Res. 2021, 154, 433–445. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  169. Bhardwaj, U.; Pandey, N.; Rastogi, M.; Singh, S.K. Gist of Zika Virus pathogenesis. Virology 2021, 560, 86–95. [Google Scholar] [CrossRef] [PubMed]
  170. Meertens, L.; Labeau, A.; Dejarnac, O.; Cipriani, S.; Sinigaglia, L.; Bonnet-Madin, L.; Le Charpentier, T.; Hafirassou, M.L.; Zamborlini, A.; Cao-Lormeau, V.M.; et al. Axl Mediates ZIKA Virus Entry in Human Glial Cells and Modulates Innate Immune Responses. Cell Rep. 2017, 18, 324–333. [Google Scholar] [CrossRef] [PubMed]
  171. Zhu, Z.; Gorman, M.J.; McKenzie, L.D.; Chai, J.N.; Hubert, C.G.; Prager, B.C.; Fernandez, E.; Richner, J.M.; Zhang, R.; Shan, C.; et al. Zika virus has oncolytic activity against glioblastoma stem cells. J. Exp. Med. 2017, 214, 2843–2857, Erratum in J. Exp. Med. 2017, 214, 3145. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  172. Zhu, Z.; Mesci, P.; Bernatchez, J.A.; Gimple, R.C.; Wang, X.; Schafer, S.T.; Wettersten, H.I.; Beck, S.; Clark, A.E.; Wu, Q.; et al. Zika Virus Targets Glioblastoma Stem Cells through a SOX2-Integrin αvβ5 Axis. Cell Stem Cell. 2020, 26, 187–204.e10. [Google Scholar] [CrossRef] [PubMed]
  173. Lima, E.O.; Guerreiro, T.M.; Melo, C.F.O.; de Oliveira, D.N.; Machado, D.; Lancelloti, M.; Catharino, R.R. MALDI imaging detects endogenous digoxin in glioblastoma cells infected by Zika virus-Would it be the oncolytic key? J. Mass Spectrom. 2018, 53, 257–263. [Google Scholar] [CrossRef] [PubMed]
  174. Dabaja, M.Z.; Lima, E.O.; de Oliveira, D.N.; Guerreiro, T.M.; Melo, C.F.O.R.; Morishita, K.N.; Lancellotti, M.; Gois Ruiz, A.L.T.; Goulart, G.; Duarte, D.A.; et al. Metabolic alterations induced by attenuated Zika virus in glioblastoma cells. Cell Biosci. 2018, 8, 47. [Google Scholar] [CrossRef] [PubMed]
  175. Li, H.; Hu, Y.; Huang, J.; Feng, Y.; Zhang, Z.; Zhong, K.; Chen, Y.; Wang, Z.; Huang, C.; Yang, H.; et al. Zika virus NS5 protein inhibits cell growth and invasion of glioma. Biochim. Biophys. Acta Mol. Cell Res. 2020, 1867, 118655. [Google Scholar] [CrossRef] [PubMed]
  176. Nair, S.; Mazzoccoli, L.; Jash, A.; Govero, J.; Bais, S.S.; Hu, T.; Fontes-Garfias, C.R.; Shan, C.; Okada, H.; Shresta, S.; et al. Zika virus oncolytic activity requires CD8+ T cells and is boosted by immune checkpoint blockade. JCI Insight 2021, 6, e144619. [Google Scholar] [CrossRef]
  177. Crane, A.T.; Chrostek, M.R.; Krishna, V.D.; Shiao, M.; Toman, N.G.; Pearce, C.M.; Tran, S.K.; Sipe, C.J.; Guo, W.; Voth, J.P.; et al. Zika virus-based immunotherapy enhances long-term survival of rodents with brain tumors through upregulation of memory T-cells. PLoS ONE 2020, 15, e0232858. [Google Scholar] [CrossRef]
  178. Garcez, P.P.; Guasti, A.; Ventura, N.; Higa, L.M.; Andreiuolo, F.; de Freitas, G.P.A.; Ribeiro, L.J.; Maia, R.A.; de Lima, S.M.B.; de Souza Azevedo, A.; et al. Case report: Regression of Glioblastoma after flavivirus infection. Front. Med. 2023, 10, 1192070. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  179. Webb, M.J.; Sener, U.; Vile, R.G. Current status and challenges of oncolytic virotherapy for the treatment of glioblastoma. Pharmaceuticals 2023, 16, 793. [Google Scholar] [CrossRef]
  180. Liu, J.; Piranlioglu, R.; Ye, F.; Shu, K.; Lei, T.; Nakashima, H. Immunosuppressive cells in oncolytic virotherapy for glioma: Challenges and solutions. Front Cell Infect Microbiol. 2023, 13, 1141034. [Google Scholar] [CrossRef]
  181. Khoonkari, M.; Liang, D.; Kamperman, M.; van Rijn, P. The unfolded protein response sensor PERK mediates stiffness adaptation in glioblastoma cells. Int. J. Mol. Sci. 2022, 23, 6520. [Google Scholar] [CrossRef] [PubMed]
  182. Sottoriva, A.; Spiteri, I.; Piccirillo, S.G.M.; Touloumis, A.; Collins, V.P.; Marioni, J.C.; Curtis, C.; Watts, C.; Tavaré, S. Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc. Natl. Acad. Sci. USA. 2013, 110, 4009–4014. [Google Scholar] [CrossRef]
  183. Tamura, K.; Wakimoto, H.; Agarwal, A.S.; Rabkin, S.D.; Bhere, D.; Martuza, R.L.; Kuroda, T.; Kasmieh, R.; Shah, K. Multimechanistic tumor-targeted oncolytic virus overcomes resistance in brain tumors. Mol. Ther. 2013, 21, 68–77. [Google Scholar] [CrossRef]
  184. Ohka, F.; Natsume, A.; Wakabayashi, T. Current trends in targeted therapies for glioblastoma multiforme. Neurol. Res. Int. 2012, 2012, 878425. [Google Scholar] [CrossRef]
  185. Kircher, M.F.; de la Zerda, A.; Jokerst, J.V.; Zavaleta, C.L.; Kempen, P.J.; Mittra, E.; Pitter, K.; Huang, R.; Campos, C.; Habte, F.; et al. A brain tumor molecular imaging strategy using a new triple-modality MRI-photoacoustic-Raman nanoparticle. Nat. Med. 2012, 18, 829–834. [Google Scholar] [CrossRef]
  186. Ageenko, A.; Vasileva, N.; Richter, V.; Kuligina, E. Combination of oncolytic virotherapy with different antitumor approaches against glioblastoma. Int. J. Mol. Sci. 2024, 25, 2042. [Google Scholar] [CrossRef]
  187. Han, X.; Abdallah, M.O.E.; Breuer, P.; Evert, B. Downregulation of MGMT expression by targeted editing of DNA methylation enhances temozolomide sensitivity in glioblastoma. Cell Oncol. 2023, 46, 697–710. [Google Scholar] [CrossRef]
  188. Yu, W.; Zhang, L.; Wei, Q.; Shao, A. O6-methylguanine-DNA methyltransferase (MGMT): Challenges and new opportunities in glioma chemotherapy. Front. Oncol. 2020, 9, 1547. [Google Scholar] [CrossRef]
  189. Kanai, R.; Rabkin, S.D.; Yip, S.; Martuza, R.L. Oncolytic virus-mediated manipulation of DNA damage responses: Synergy with chemotherapy in killing glioblastoma stem cells. J. Natl. Cancer Inst. 2012, 104, 42–55. [Google Scholar] [CrossRef]
  190. Awada, H.; Paris, F.; Pecqueur, C. Exploiting radiation immunostimulatory effects to improve glioblastoma outcome. Neuro Oncol. 2023, 25, 433–446. [Google Scholar] [CrossRef]
  191. Chédeville, A.; Madureira, P.A. The role of hypoxia in glioblastoma radiotherapy resistance. Cancers 2021, 13, 542. [Google Scholar] [CrossRef]
  192. Rodriguez, S.M.B.; Tataranu, L.G.; Kamel, A.; Turliuc, S.; Rizea, R.E.; Dricu, A. Glioblastoma and Immune Checkpoint Inhibitors: A Glance at Available Treatment Options and Future Directions. Int. J. Mol. Sci. 2024, 25, 10765. [Google Scholar] [CrossRef]
  193. Berghoff, A.S.; Kiesel, B.; Widhalm, G.; Rajky, O.; Ricken, G.; Wöhrer, A.; Dieckmann, K.; Filipits, M.; Brandstetter, A.; Weller, M.; et al. Programmed death ligand 1 expression and tumor-infiltrating lymphocytes in glioblastoma. Neuro Oncol. 2015, 17, 1064–1075. [Google Scholar] [CrossRef]
  194. Bagchi, S.; Yuan, R.; Engleman, E.G. Immune checkpoint inhibitors for the treatment of cancer: Clinical impact and mechanisms of response and resistance. Annu. Rev. Pathol. 2021, 16, 223–249. [Google Scholar] [CrossRef]
  195. Reardon, D.A.; Brandes, A.A.; Omuro, A.; Mulholland, P.; Lim, M.; Wick, A.; Baehring, J.; Ahluwalia, M.S.; Roth, P.; Bähr, O.; et al. Effect of nivolumab vs bevacizumab in patients with recurrent glioblastoma: The CheckMate 143 phase 3 randomized clinical trial. JAMA Oncol. 2020, 6, 1003–1010. [Google Scholar] [CrossRef]
  196. Arrieta, V.A.; Dmello, C.; McGrail, D.J.; Brat, D.J.; Lee-Chang, C.; Heimberger, A.B.; Chand, D.; Stupp, R.; Sonabend, A.M. Immune checkpoint blockade in glioblastoma: From tumor heterogeneity to personalized treatment. J. Clin. Investig. 2023, 133, e163447. [Google Scholar] [CrossRef]
  197. Sugawara, K.; Iwai, M.; Yajima, S.; Tanaka, M.; Yanagihara, K.; Seto, Y.; Todo, T. Oncolytic herpes virus G47Δ works synergistically with CTLA-4 inhibition through dynamic intratumoral immune modulation. Mol. Ther. Oncolytics 2021, 21, 132–145. [Google Scholar] [CrossRef]
  198. Zhang, Q.; Liu, F. Advances and potential pitfalls of oncolytic viruses expressing immunomodulatory transgene therapy for malignant gliomas. Cell Death Dis. 2020, 11, 485. [Google Scholar] [CrossRef]
  199. Vasileva, N.; Ageenko, A.; Byvakina, A.; Sen’kova, A.; Kochneva, G.; Mishinov, S.; Richter, V.; Kuligina, E. The recombinant oncolytic virus VV-GMCSF-Lact and chemotherapy drugs against human glioma. Int. J. Mol. Sci. 2024, 25, 4244. [Google Scholar] [CrossRef]
  200. Cheema, T.A.; Wakimoto, H.; Fecci, P.E.; Ning, J.; Kuroda, T.; Jeyaretna, D.S.; Martuza, R.L.; Rabkin, S.D. Multifaceted oncolytic virus therapy for glioblastoma in an immunocompetent cancer stem cell model. Proc. Natl. Acad. Sci. USA. 2013, 110, 12006–12011. [Google Scholar] [CrossRef]
  201. Jiang, H.; Shin, D.H.; Nguyen, T.T.; Fueyo, J.; Fan, X.; Henry, V.; Carrillo, C.C.; Yi, Y.; Alonso, M.M.; Collier, T.L.; et al. Localized treatment with oncolytic adenovirus Delta-24-RGDOX induces systemic immunity against disseminated subcutaneous and intracranial melanomas. Clin. Cancer Res. 2019, 25, 6801–6814. [Google Scholar] [CrossRef]
  202. Roberts, N.J.; Zhang, L.; Janku, F.; Collins, A.; Bai, R.Y.; Staedtke, V.; Rusk, A.W.; Tung, D.; Miller, M.; Roix, J.; et al. Intratumoral injection of Clostridium novyi-NT spores induces antitumor responses. Sci. Transl. Med. 2014, 6, 249ra111. [Google Scholar] [CrossRef]
  203. Andtbacka, R.H.; Kaufman, H.L.; Collichio, F.; Amatruda, T.; Senzer, N.; Chesney, J.; Delman, K.A.; Spitler, L.E.; Puzanov, I.; Agarwala, S.S.; et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J. Clin. Oncol. 2015, 33, 2780–2788. [Google Scholar] [CrossRef]
  204. Wilson, W.R.; Hay, M.P. Targeting hypoxia in cancer therapy. Nature reviews. Cancer 2011, 11, 393–410. [Google Scholar] [CrossRef]
  205. Horsman, M.R.; Mortensen, L.S.; Petersen, J.B.; Busk, M.; Overgaard, J. Imaging hypoxia to improve radiotherapy outcome. Nat. Rev. Clin. Oncol. 2012, 9, 674–687. [Google Scholar] [CrossRef]
  206. Agrawal, N.; Bettegowda, C.; Cheong, I.; Geschwind, J.F.; Drake, C.G.; Hipkiss, E.L.; Tatsumi, M.; Dang, L.H.; Diaz, L.A., Jr.; Pomper, M.; et al. Bacteriolytic therapy can generate a potent immune response against experimental tumors. Proc. Natl. Acad. Sci. USA 2004, 101, 15172–15177. [Google Scholar] [CrossRef]
  207. Krick, E.L.; Sorenmo, K.U.; Rankin, S.C.; Cheong, I.; Kobrin, B.; Thornton, K.; Kinzler, K.W.; Vogelstein, B.; Zhou, S.; Diaz, L.A., Jr. Evaluation of Clostridium novyi-NT spores in dogs with naturally occurring tumors. Am. J. Vet. Res. 2012, 73, 112–118. [Google Scholar] [CrossRef]
  208. Diaz, L.A., Jr.; Cheong, I.; Foss, C.A.; Zhang, X.; Peters, B.A.; Agrawal, N.; Bettegowda, C.; Karim, B.; Liu, G.; Khan, K.; et al. Pharmacologic and toxicologic evaluation of C. novyi-NT spores. Toxicol. Sci. Off. J. Soc. Toxicol. 2005, 88, 562–575. [Google Scholar] [CrossRef]
  209. Paoloni, M.; Khanna, C. Translation of new cancer treatments from pet dogs to humans. Nat. Rev. Cancer 2008, 8, 147–156. [Google Scholar] [CrossRef]
  210. Wick, W.; Wick, A.; Sahm, F.; Riehl, D.; von Deimling, A.; Bendszus, M.; Kickingereder, P.; Beckhove, P.; Schmitz-Winnenthal, F.H.; Jungk, C.; et al. VXM01 phase I study in patients with progressive glioblastoma: Final results. JCO 2018, 36, 2017. [Google Scholar] [CrossRef]
  211. Sun, R.; Liu, M.; Lu, J.; Chu, B.; Yang, Y.; Song, B.; Wang, H.; He, Y. Bacteria loaded with glucose polymer and photosensitive ICG silicon-nanoparticles for glioblastoma photothermal immunotherapy. Nat. Commun. 2022, 13, 5127. [Google Scholar] [CrossRef]
  212. Zhou, M.; Tang, Y.; Xu, W.; Hao, X.; Li, Y.; Huang, S.; Xiang, D.; Wu, J. Bacteria-based immunotherapy for cancer: A systematic review of preclinical studies. Front. Immunol. 2023, 14, 1140463. [Google Scholar] [CrossRef]
  213. Zhang, Y.; Xi, K.; Fu, Z.; Zhang, Y.; Cheng, B.; Feng, F.; Dong, Y.; Fang, Z.; Zhang, Y.; Shen, J.; et al. Stimulation of tumoricidal immunity via bacteriotherapy inhibits glioblastoma relapse. Nat. Commun. 2024, 15, 4241. [Google Scholar] [CrossRef]
  214. Banerjee, A.; Kim, B.J.; Carmona, E.M.; Cutting, A.S.; Gurney, M.A.; Carlos, C.; Feuer, R.; Prasadarao, N.V.; Doran, K.S. Bacterial Pili exploit integrin machinery to promote immune activation and efficient blood-brain barrier penetration. Nat. Commun. 2011, 2, 462. [Google Scholar] [CrossRef]
  215. Naghavian, R.; Faigle, W.; Oldrati, P.; Wang, J.; Toussaint, N.C.; Qiu, Y.; Medici, G.; Wacker, M.; Freudenmann, L.K.; Bonté, P.E.; et al. Microbial peptides activate tumour-infiltrating lymphocytes in glioblastoma. Nature 2023, 617, 807–817. [Google Scholar] [CrossRef]
  216. De Bonis, P.; Albanese, A.; Lofrese, G.; de Waure, C.; Mangiola, A.; Pettorini, B.L.; Pompucci, A.; Balducci, M.; Fiorentino, A.; Lauriola, L.; et al. Postoperative infection may influence survival in patients with glioblastoma: Simply a myth? Neurosurgery 2011, 69, 864–869. [Google Scholar] [CrossRef]
  217. Bowles, A.P., Jr.; Perkins, E. Long-term remission of malignant brain tumors after intracranial infection: A report of four cases. Neurosurgery 1999, 44, 636–643. [Google Scholar] [CrossRef]
  218. Solár, P.; Mackerle, Z.; Hendrych, M.; Pospisil, P.; Lakomy, R.; Valekova, H.; Hermanova, M.; Jancalek, R. Prolonged survival in patients with local chronic infection after high-grade glioma treatment: Two case reports. Front. Oncol. 2022, 12, 1073036. [Google Scholar] [CrossRef]
  219. Sari-Ak, D.; Alomari, O.; Shomali, R.A.; Lim, J.; Thimiri Govinda Raj, D.B. Advances in CRISPR-Cas9 for the Baculovirus Vector System: A Systematic Review. Viruses 2023, 15, 54. [Google Scholar] [CrossRef]
  220. Mander, S.; Naffouje, S.A.; Gao, J.; Li, W.; Christov, K.; Green, A.; Bongarzone, E.R.; Das Gupta, T.K.; Yamada, T. Tumor-targeting cell-penetrating peptide, p28, for glioblastoma imaging and therapy. Front. Oncol. 2022, 12, 940001. [Google Scholar] [CrossRef]
  221. von Spreckelsen, N.; Fadzen, C.M.; Hartrampf, N.; Ghotmi, Y.; Wolfe, J.M.; Dubey, S.; Yang, B.Y.; Kijewski, M.F.; Wang, S.; Farquhar, C.; et al. Targeting glioblastoma using a novel peptide specific to a deglycosylated isoform of brevican. Adv. Ther. 2021, 4, 2000244. [Google Scholar] [CrossRef]
  222. Latzer, P.; Zelba, H.; Battke, F.; Reinhardt, A.; Shao, B.; Bartsch, O.; Rabsteyn, A.; Harter, J.; Schulze, M.; Okech, T.; et al. A real-world observation of patients with glioblastoma treated with a personalized peptide vaccine. Nat. Commun. 2024, 15, 6870. [Google Scholar] [CrossRef]
  223. Collins, S.A.; Shah, A.H.; Ostertag, D.; Kasahara, N.; Jolly, D.J. Clinical development of retroviral replicating vector Toca 511 for gene therapy of cancer. Expert Opin. Biol. Ther. 2021, 21, 1199–1214. [Google Scholar] [CrossRef]
  224. D’Amico, R.S.; Aghi, M.K.; Vogelbaum, M.A.; Bruce, J.N. Convection-enhanced drug delivery for glioblastoma: A review. J. Neuro-Oncol. 2021, 151, 415–427. [Google Scholar] [CrossRef]
  225. Friedmann-Morvinski, D. Glioblastoma heterogeneity and cancer cell plasticity. Crit. Rev. Oncog. 2014, 19, 327–336. [Google Scholar] [CrossRef]
  226. Becker, A.P.; Sells, B.E.; Haque, S.J.; Chakravarti, A. Tumor Heterogeneity in Glioblastomas: From Light Microscopy to Molecular Pathology. Cancers 2021, 13, 761. [Google Scholar] [CrossRef]
  227. Zhang, P.; Xia, Q.; Liu, L.; Li, S.; Dong, L. Current Opinion on Molecular Characterization for GBM Classification in Guiding Clinical Diagnosis, Prognosis, and Therapy. Front. Mol. Biosci. 2020, 7, 562798. [Google Scholar] [CrossRef]
  228. Chongsathidkiet, P.; Jackson, C.; Koyama, S.; Loebel, F.; Cui, X.; Farber, S.H.; Woroniecka, K.; Elsamadicy, A.A.; Dechant, C.A.; Kemeny, H.R.; et al. Sequestration of T cells in bone marrow in the setting of glioblastoma and other intracranial tumors. Nat. Med. 2018, 24, 1459–1468. [Google Scholar] [CrossRef]
  229. Campian, J.L.; Ghosh, S.; Kapoor, V.; Yan, R.; Thotala, S.; Jash, A.; Hu, T.; Mahadevan, A.; Rifai, K.; Page, L.; et al. Long-Acting Recombinant Human Interleukin-7, NT-I7, Increases Cytotoxic CD8 T Cells and Enhances Survival in Mouse Glioma Models. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2022, 28, 1229–1239. [Google Scholar] [CrossRef]
Figure 1. This schematic illustrates the immunologic and cytolytic mechanisms of oncolytic virotherapy against glioblastoma. Following intratumoral administration, oncolytic viruses selectively infect tumor cells (1), replicate and induce oncolysis with release of transgenic and viral products (2), and spread to neighboring tumor cells (3). Viral infection triggers the release of chemokines that attract immune cells (4), leading to antigen presentation by macrophages and dendritic cells. This promotes activation of tumor- and virus-specific CD4+ and CD8+ T cells, while anti–PD-L1 checkpoint blockade enhances cytotoxic T-cell function. Several viral platforms, including poliovirus (PVS-RIPO), adenovirus (DNX-2401), herpes simplex virus (M032), and measles virus, are under clinical investigation for glioblastoma therapy [created with BioRender.com].
Figure 1. This schematic illustrates the immunologic and cytolytic mechanisms of oncolytic virotherapy against glioblastoma. Following intratumoral administration, oncolytic viruses selectively infect tumor cells (1), replicate and induce oncolysis with release of transgenic and viral products (2), and spread to neighboring tumor cells (3). Viral infection triggers the release of chemokines that attract immune cells (4), leading to antigen presentation by macrophages and dendritic cells. This promotes activation of tumor- and virus-specific CD4+ and CD8+ T cells, while anti–PD-L1 checkpoint blockade enhances cytotoxic T-cell function. Several viral platforms, including poliovirus (PVS-RIPO), adenovirus (DNX-2401), herpes simplex virus (M032), and measles virus, are under clinical investigation for glioblastoma therapy [created with BioRender.com].
Cancers 17 02550 g001
Figure 2. This figure illustrates the distinct mechanisms by which various oncolytic viruses modulate the glioblastoma (GBM) microenvironment in vivo. (A) Adenovirus (ICOVIR17) promotes extracellular matrix degradation and T-cell recruitment, reducing tumor burden. (B) Herpes simplex virus (HSV-P10) downregulates the AKT/IL-6/STAT3 pathway, sensitizing GBM cells to radiotherapy. (C) Reovirus selectively replicates in RAS-activated tumor cells by suppressing PKR, activating NK cells, CD8+ T cells, macrophages, and dendritic cells. (D) Measles virus infects CD46-high tumor cells, releasing PAMPs and enhancing phagocytosis by dendritic cells, particularly promoting maturation. Viruses are delivered intratumorally or systemically in GBM-bearing mice in all models, offering insight into their therapeutic and immunomodulatory potential. [Created with BioRender.com].
Figure 2. This figure illustrates the distinct mechanisms by which various oncolytic viruses modulate the glioblastoma (GBM) microenvironment in vivo. (A) Adenovirus (ICOVIR17) promotes extracellular matrix degradation and T-cell recruitment, reducing tumor burden. (B) Herpes simplex virus (HSV-P10) downregulates the AKT/IL-6/STAT3 pathway, sensitizing GBM cells to radiotherapy. (C) Reovirus selectively replicates in RAS-activated tumor cells by suppressing PKR, activating NK cells, CD8+ T cells, macrophages, and dendritic cells. (D) Measles virus infects CD46-high tumor cells, releasing PAMPs and enhancing phagocytosis by dendritic cells, particularly promoting maturation. Viruses are delivered intratumorally or systemically in GBM-bearing mice in all models, offering insight into their therapeutic and immunomodulatory potential. [Created with BioRender.com].
Cancers 17 02550 g002
Figure 3. Schematic representation of engineered oncolytic bacteria targeting glioblastoma. VXM01, an oral Salmonella-based VEGFR-2 DNA vaccine, enhances CD8+ T-cell responses and reduces PD-L1 expression. Locally injected C. novyi-NT germinates in hypoxic tumor cores, inducing direct oncolysis and immune activation. Both approaches remodel the tumor microenvironment and elicit systemic antitumor immunity. Preclinical models and early clinical data support their translational potential in GBM therapy. [Created with BioRender.com].
Figure 3. Schematic representation of engineered oncolytic bacteria targeting glioblastoma. VXM01, an oral Salmonella-based VEGFR-2 DNA vaccine, enhances CD8+ T-cell responses and reduces PD-L1 expression. Locally injected C. novyi-NT germinates in hypoxic tumor cores, inducing direct oncolysis and immune activation. Both approaches remodel the tumor microenvironment and elicit systemic antitumor immunity. Preclinical models and early clinical data support their translational potential in GBM therapy. [Created with BioRender.com].
Cancers 17 02550 g003
Table 1. Overview of Oncolytic Herpes Simplex Virus (oHSV) Variants and Therapeutic Strategies in Glioblastoma Models.
Table 1. Overview of Oncolytic Herpes Simplex Virus (oHSV) Variants and Therapeutic Strategies in Glioblastoma Models.
oHSV VariantGenetic Modifications/AdditionsStudy ModelKey FindingsReference
zLacks thymidine kinase (TK); attenuated neurovirulenceU87 glioma cells (in vitro, mice)Cytotoxic to human glioma cells; improved survival and reduced tumor burden in mice[29]
oHSV-P10Expresses PTEN-LHuman GBM12 cells and miceEliminated GSCs, blocked IL6/JAK/STAT3 signaling; overcame radiotherapy resistance[30]
HSV-P10Expresses PTEN-αGBM cells, immune-competent miceSuppressed PI3K/AKT pathway, reduced PD-L1, enhanced adaptive immune response[31]
oHSV-1 (ICP4+)Deleted γ34.5 and ICP47; retains ICP4Glioma cell lines and animal modelsReduced invasion via downregulation of Sp1; high anti-tumor activity with less immune activation[32]
EGFP-oHSV-1Δγ34.5/ΔUS12; includes EGFP reporterGL261 glioma mouse modelImproved survival and immune response; greater tumor control vs. other oHSV-1 variants[33]
C5252Deleted γ34.5 and 15 kb repeat; expresses IL-12 and anti-PD-1 fragmentHuman GBM cells and miceStrong cytotoxicity despite low replication; induced apoptosis, immune activation (IFN-γ, TNF-α)[34]
G47Δ-mIL2Expresses IL-2Orthotopic glioma mouse modelEnhanced CD4+/CD8+ T-cell infiltration; prolonged survival; no synergy with PD-1 blockade[35]
G47Δ-mIL12Expresses IL-12; combined with anti-PD-1/CTLA-4Glioma mouse modelImproved survival; increased M1 macrophages and effector T cells; enhanced immune response[36]
R-115Targets HER2; expresses IL-12HER2+ glioma in BALB/c miceLong-term remission, antibody generation, HER2-independent protection; no CD4/CD8 increase noted[37]
R-613Targets EGFRvIIIhGICs and EGFRvIII+ glioma in miceEffective in early-stage tumors; less effective in advanced disease; suitable for combination therapy[38]
OH2 (HSV-2 based)HSV-2-derived oncolytic virus with tumor-selective replicationIn vitro, xenograft miceSelective tumor replication, DNA damage, suppressed tumor cells and M2 TAMs, enhanced macrophage and T cell infiltration, slowed GBM, prolonged survival[39]
NG34 (HSV-1 with GADD34)HSV-1 engineered to express GADD34 under nestin promoterIn vitro, mouse modelsSimilar efficacy to rQNestin34.5, reduced neurotoxicity and brain damage[40]
HSV + CAR-T cells (B7-H3-directed)CAR-T cells engineered to express B7-H3-targeted CAR and carry HSVOrthotopic GBM miceCAR-T cells deliver HSV to distant tumor sites, enhanced T cell infiltration, prolonged survival[41]
CAR-T + HSV-1 G47ΔHSV-1 G47Δ combined with CAR-T cellsMouse modelsTumor regression, extended survival[42]
oHSV-1Wild-type or engineered oHSV-1Murine models, in vitroTNFα from M1 macrophages/microglia inhibits viral replication; blocking TNFα increases viral spread and survival[43]
MEK inhibitor + oHSV-1Pharmacological MEK inhibitor combined with oHSV-1In vitro, murine gliomaTrametinib reduces TNFα production, enhances oHSV replication and survival[44]
oHSV + Gamma Secretase Inhibitor (GSI)Wild-type oHSV combined with NOTCH pathway inhibitorIn vitro, orthotopic miceInhibition of NOTCH signaling boosts therapy efficacy with maintained safety[45]
oHSV G47ΔHSV-1 G47ΔPatient-derived cultures, miceFasting boosts viral replication and cytotoxicity by suppressing JNK pathway[46]
γ34.5-deficient oHSVDeletion of γ34.5 gene in oHSV; Us11 protein expressionGSCs, ScGCs cultureGSCs resist virus replication due to translational blockade; Us11 protein restores replication[47]
Bortezomib + oHSV1 + NK cellsoHSV + proteasome inhibitor + NK cellsIn vitro, miceNecroptosis induction, increased NK activation, improved tumor suppression[48]
CCN1 and HSV resistanceWild-type HSV-1; CCN1 expression modulates resistanceGBM cell linesCCN1 mediates early viral resistance by innate immune activation[49]
oHSV + TGF-β receptor inhibitoroHSV combined with TGF-β pathway inhibitorGSCs, animal modelsBlocking TGF-β boosts viral replication and survival via JNK-MAPK pathway[50]
oHSV + C16 inhibitorPharmacologic STAT1/3 inhibitor combined with oHSVU87 xenograft, cell culturesInhibiting STAT1/3 in microglia/macrophages promotes oHSV replication and tumor regression[51]
oHSV releasing ChaseM enzyme + temozolomideoHSV engineered to release ChaseM enzyme (degrades CSPGs)Preclinical mouse modelsDegrades CSPGs, improves tumor penetration and apoptotic death, extends survival[52]
oHSV + anti-HMGB1 antibodiesWild-type oHSV combined with HMGB1 neutralizing antibodiesIn vitro, mouse brain tumorsBlocking HMGB1 reduces edema and improves survival with oHSV therapy[53]
KG4:T124HSV-1 derivative (specific modifications not detailed)GL261N4 and CT2A murine glioma modelsCleared quickly in CT2A; limited immune response and therapeutic effect.[54]
rQNestin34.5v.1ICP34.5 under nestin promoterGL261N4 and CT2A murine glioma modelsHigher viral load; persisted longer in GL261N4, enhancing immune infiltration and survival.[55]
CXCR4-targeted oHSVGlycoprotein D modified with CXCR4-specific nanobody in attenuated HSV-1GSC xenograft mouse modelsTargeted CXCR4+ GSCs; reduced tumor growth and improved survival.[56]
oHSV: Oncolytic Herpes Simplex Virus, TK: Thymidine Kinase, GBM: Glioblastoma, GSCs: Glioma Stem Cells, IL: Interleukin, JAK: Janus Kinase, STAT: Signal Transducer and Activator of Transcription, PI3K: Phosphoinositide 3-Kinase, AKT: Protein Kinase B, PD-L1: Programmed Death-Ligand 1, Sp1: Specificity Protein 1, EGFP: Enhanced Green Fluorescent Protein, IFN-γ: Interferon gamma, TNF-α: Tumor Necrosis Factor alpha, CTLA-4: Cytotoxic T-Lymphocyte Antigen 4, HER2: Human Epidermal Growth Factor Receptor 2, EGFRvIII: Epidermal Growth Factor Receptor variant III, TAMs: Tumor-Associated Macrophages, CAR-T: Chimeric Antigen Receptor T-cells, JNK: c-Jun N-terminal Kinase, STAT1/3: STAT1 and STAT3 Proteins, CSPGs: Chondroitin Sulfate Proteoglycans, HMGB1: High Mobility Group Box 1.
Table 2. Summary of Clinical Studies Evaluating oHSV-Based Treatments in Glioblastoma Patients.
Table 2. Summary of Clinical Studies Evaluating oHSV-Based Treatments in Glioblastoma Patients.
Virus UsedPatient PopulationDesign and
Intervention
Adverse EventsMedian
Survival
Notable FindingsReference
G207 (γ134.5-deleted HSV-1)9 adults with recurrent malignant gliomaSingle intratumoral G207 injection + 24 h later 5 Gy radiation; 2 patients had a second injectionWell tolerated; no severe side effects7.5 monthsSafe combination with radiotherapy; 3 patients showed marked radiologic response[57]
G20712 pediatric patients (7–18 y/o), mostly GBMIntratumoral G207 ± radiationHigh rate of AEs (e.g., diarrhea, bradycardia, seizures), but manageable12.2 months11/12 had clinical or radiological improvement; increased lymphocyte infiltration; 4 survived > 18 months[58]
G47∆ (triple-mutated oHSV)13 adults with recurrent/advanced GBMUp to 6 intratumoral injections over 2 weeksCommon: nausea, fever, headache; manageable with corticosteroidsNot reported; 3 > 46 moCD4+/CD8+ T-cell infiltration; 1 patient survived > 11 years[59]
M032 (IL-12 expressing HSV-1)21 adults with recurrent gliomaSingle intratumoral injectionGrade 3–4 AEs in only 1 patient at high dose; no severe toxicity at max dose9.38 monthsGenerally well tolerated; individualized response suggests need for personalized dosing[60]
CAN-3110 (ICP34.5 under nestin promoter)41 patients with recurrent GBMSingle intratumoral injectionNo serious AEs at highest dosesNot stated clearlyHSV-seropositive patients had better survival; T-cell activation and immune gene upregulation observed[61]
oHSV: Oncolytic Herpes Simplex Virus, GBM: Glioblastoma, IL: Interleukin, AE(s): Adverse Event(s), CTLA-4: Cytotoxic T-Lymphocyte Antigen 4, CD4+/CD8+: T-cell Surface Markers (Helper/Cytotoxic), MRI: Magnetic Resonance Imaging, Gy: Gray (Unit of Radiation Dose).
Table 3. Overview of Oncolytic Adenovirus Variants and Therapeutic Strategies in Glioblastoma Models.
Table 3. Overview of Oncolytic Adenovirus Variants and Therapeutic Strategies in Glioblastoma Models.
oADV VariantGenetic Modifications/AdditionsStudy ModelKey FindingsRef.
Replicating vs. Non-replicating oAdVsReplication-competent vs. incompetent adenovirusesCell lines, mouse modelsReplicating oAdVs enhanced immune cell infiltration and survival[69]
Ad5-pIX-Ad37IX capsid protein with dimerization domain; fiber knob from Ad37In vitro, in vivoEnhanced cell entry and oncolytic activity[70]
oAdV-ApoA1Carries apolipoprotein A1Cell lines, mouse modelsReduced 7-KC, activated TNF signaling, improved immune response[71]
ICOVIR17Carries hyaluronidase enzymeGBM mouse modelsIncreased macrophages, CD8+ T cells, and survival with anti-PD-1[72]
ICOVIR15∆24-E1A, RGD-modified fiberGBM cells, mouse modelsTargeted FAP+ pericytes and tumor cells; induced apoptosis[73]
ONCOTECHT cell–associated, PD-L1 targetingCancer mouse modelsReduced PD-L1, enhanced survival[74]
Ad5-Ki67/IL-15Ki67 promoter; IL-15 expressionGlioma cells, mouse modelsReduced PD-L1, boosted T cell infiltration[75]
MSC-Ad5-Ki67/IL-15MSC-carried virus with IL-15 and Ki67 promoterIn vitro, in vivoEnhanced macrophage infiltration and efficacy[76]
TS-2021 (Ad5 KT-E1A-IL-15)Ki67 promoter, TGF-β2 5′UTR, IL-15In vitro, in vivoReduced tumor burden, improved survival[77]
TS-2021 + OlaparibSame as TS-2021; combined with PARP inhibitorGBM cells, mouse modelsSynergistic tumor apoptosis and survival benefit[78]
XVir-N-31 (Intranasal)Carrier-cell optimized oAdVGBM-bearing miceNon-invasive delivery reduced tumor burden, improved survival[79]
XVir-N-31 + ICICombined with anti-PD-1/PD-L1In vitro, humanized mouse modelsEnhanced immune cell infiltration, tumor regression[80]
YSCH-01Recombinant interferon-like geneGlioma cells, hamster modelsStrong local and distant tumor suppression[81]
H5CmTERT-Ad/TRAILhTERT promoter, secretable trimeric TRAILIn vitro, in vivoEffective in TRAIL-resistant tumors, induced death in hypoxia[82]
Ad6Native Ad6 serotypeGBM cells, mouse modelsCytotoxicity, reduced GBM stem cells[83]
Ad5 (hTERT/survivin promoters)GBM-specific promoters (hTERT, survivin)GBM cell linesSelective cytotoxicity in GBM cells[84]
Delta-24-RGD (Proteomic analysis)∆24-E1A, RGD-modified fiberPhase I clinical trial samplesAltered kinase/cytokine profiles, immune activation[85]
CTV (Ad3 fiber + Ad5 capsid)Produces MDA-7/IL-24In vivo, GBM modelsExtended survival, enhanced with Temozolomide[86]
PD-BM-MSC-D24Delta-24-RGD loaded in chemo-treated BM-hMSCIn vitro, in vivoEffective delivery and tumor suppression[87]
Delta-24-RGDOXOncolytic adenovirus with RGD motif and OX40LMouse modelsProlonged survival; changes in gut microbiota with dominance of Bifidobacterium. Microbiota may influence therapeutic efficacy.[88]
Delta-24-GREATDelta-24 modified with GITRL geneHuman and mouse glioma cell lines; mouse modelsEnhanced immune response, increased memory T cells, tumor rejection after re-challenge.[89]
Delta-24-RGD + HDAC inhibitorsCombined with scriptaid and LBH589Patient-derived glioblastoma linesSynergistic antitumor effects; scriptaid ↑ caspase-3/7 and apoptosis; LBH589 ↑ LDH and phospho-p70S6K.[90]
AdCMVdelta24Delta-24 under CMV promoterMouse GBM modelsReduced Tregs, increased IFNγ+ CD8+ T cells; reprogrammed Tregs into stimulatory phenotype.[91]
hMSC-D24Delta-24-RGD loaded into human MSCsDog GBM model (large animal); intra-arterial delivery (ESIA)ESIA safe in anterior cerebral circulation; stroke risk in posterior; proof-of-concept for large-animal delivery method.[92]
Ad5/35-delta-24, Ad5/3-delta-24Fiber region modifiedHuman and rodent glioma lines; mouse modelsAd5/35-delta-24: strong immune-mediated tumor suppression; induced immune memory; superior to Ad5-delta-24-RGD.[93]
CAN-2409 + dexamethasoneSimultaneous administration with corticosteroidIn vitro and in vivo experimentsdecreased immune activation; decreased tumor response; decreased median survival; dexamethasone suppresses CAN-2409 efficacy.[94]
Delta-24-RGD + anti-PD-1Combination with immune checkpoint blockadeIn vivo and in vitro modelsSynergistic effect; Increased CD8+ T cells and IFNγ production; improved survival over monotherapies.[95]
H101 + anti-PD-1H101 suppresses CD47; anti-PD-1 immunotherapyHuman glioblastoma lines (U87-MG)Increased T cell infiltration, macrophage phagocytosis, cytokines; enhanced anti-tumor effect.[96]
CAN-2409 + ATR inhibitor (AZD6738)Combination with DNA damage repair inhibitorIn vitro and in vivo GBM modelsIncreased γH2AX; decreased PD-L1; improved survival; enhanced DNA damage and immune response.[97]
Oncolytic Ad (receptor sensitivity study)N/AHuman glioma cell lines (Grade II–IV)Receptor expression (CAR, CD46, DSG-2) not predictive of infectivity or efficacy.[98]
Delta-24-RGD + TMZCombination with temozolomide (standard chemo)Murine glioma lines; mouse modelsSynergy when Delta-24-RGD precedes TMZ; reversed effects if sequence is changed; Increased CD8+ T cells.[99]
Ad5-Delta-24-RGD with L3-23K vs. L5-Fiber gene additionGene insertion at different viral regionsGBM cell lines; mouse modelsGene expression higher at L3-23K; insertion site affected oncolytic activity; no major difference in cytotoxicity.[100]
CXCL11-carrying oAdoAd encoding CXCL11 chemokineOrthotopic GBM mouse models; cell culturesIncreased CD8+ T cell activation; Increased Tregs; improved CAR-T therapy efficacy in GBM.[101]
oAd-IL7 + CAR-T (B7H3)Oncolytic adenovirus expressing IL-7In vivo and in vitro modelsIncreased intratumoral T cells; Increased median survival; synergistic effect with CAR-T therapy.[102]
OA@TA-Fe3+-CXCL11 oAdCXCL11 oAd coated with tannic acid & Fe3+ ionsIn vivo and in vitro GBM modelsIncreased retention and oncolytic activity; Fe3+ reduced hypoxia via O2 generation; immune stimulation.[103]
NSC.CRAd-S-pk7CRAd-Survivin-pk7 delivered via neural stem cellsMice with competent immune systemMultiple doses at high levels effective; immune system did not hinder therapy; shown to be safe in Phase I trials.[104]
CAN-2409Expresses HSV-TK (thymidine kinase)Glioma stem-like cells; mouse modelsEnriched p53/cell cycle pathways; regulated MYC, CCNB1, PLK1, CDC20; Increased IL-12, Increased T cell activation.[105]
oAdV: Oncolytic Adenovirus, GBM: Glioblastoma, IL: Interleukin, PD-L1: Programmed Death-Ligand 1, MSC: Mesenchymal Stem Cell, TGF-β: Transforming Growth Factor beta, TRAIL: TNF-Related Apoptosis-Inducing Ligand, TMZ: Temozolomide, CAR-T: Chimeric Antigen Receptor T-cells, IFNγ: Interferon gamma, CD8+: Cytotoxic T-cell Marker, FAP: Fibroblast Activation Protein, RGD: Arginine-Glycine-Aspartic Acid Motif, CMV: Cytomegalovirus, MYC: MYC Proto-Oncogene, PLK1: Polo-Like Kinase 1, CDC20: Cell Division Cycle Protein 20.
Table 4. Summary of Clinical Studies Evaluating Adenovirus-Based Treatments in Glioblastoma Patients.
Table 4. Summary of Clinical Studies Evaluating Adenovirus-Based Treatments in Glioblastoma Patients.
Virus UsedPatient PopulationDesign and InterventionAdverse EventsMedian SurvivalNotable FindingsRef.
NSC-CRAd-S-pk711 newly diagnosed glioblastoma patientsPhase 1; oAdv delivered via neural stem cells (NSC) after tumor resection; followed by standard chemo-radiotherapyDecreased lymphocytes, headache, anemia, fatigue, nausea, hypoalbuminemia; NSC-CRAd-S-pk7-related meningitis (1 pt), subdural fluid (1 pt)18.4 monthsSafe; did not delay standard therapy; promising survival outcomes; supports Phase 2 progression[106]
Delta24-RGD (DNX-2401)Patients with recurrent glioblastomaPhase 1; oAdv administered via convection-enhanced delivery (CED) to tumor and peritumoral areasDose-limiting: increased intracranial pressure, temporary viral meningitis129 days1 complete responder (8-year survival), 1 partial response (2.5 years); ↑ NK, T cells, proinflammatory cytokines; immune activation observed[107]
Ad5-Δ24.RGDGBM patients with pre-existing adenovirus antibodiesObservational immune monitoring studyNot specifiedNot specifiedDespite neutralizing antibodies, RGD modification enabled efficacy by alternative cellular entry; supports oAdv utility in seropositive patients[108]
DNX-2401 + pembrolizumab3 recurrent glioblastoma patientsPhase 2 pilot; DNX-2401 delivered via SmartFlow catheter under real-time MRI guidance; followed by pembrolizumabNone reportedNot reported2 partial responders with infusion area >1 cm; 1 non-responder with <1 cm infusion; safe and feasible approach for image-guided intratumoral administration[109]
DNX-2401 + pembrolizumab49 patients with recurrent glioblastomaPhase 1/2; DNX-2401 combined with anti-PD-1 (pembrolizumab)No dose-limiting toxicities reported12.5 monthsCombination was safe; MRI response associated with long-term survival; supports benefit over monotherapy[110]
GBM: Glioblastoma, oAdV: Oncolytic Adenovirus, NSC: Neural Stem Cell, CED: Convection-Enhanced Delivery, AE(s): Adverse Event(s), MRI: Magnetic Resonance Imaging, PD-1: Programmed Death-1, IL: Interleukin, CR: Complete Response, PR: Partial Response.
Table 5. Therapeutic Development of oMeV for Glioblastoma: Genetic Engineering, Preclinical Efficacy, and Clinical Safety.
Table 5. Therapeutic Development of oMeV for Glioblastoma: Genetic Engineering, Preclinical Efficacy, and Clinical Safety.
oMeV VariantGenetic Modifications/AdditionsStudy ModelKey FindingsRef.
Preclinical
MV-CEAInsertion of the soluble N-terminal extracellular domain of human CEA into MV-NSe backbone (MV-Edm lineage); CEA gene placed before N geneHuman glioma cell lines (primary and GBM-derived); orthotopic glioma mouse models (e.g., U87, GBM14, GBM39, GBM6)Enabled noninvasive monitoring of viral gene expression via serum CEA levels; induced selective cytopathic effects in glioma cells; promoted apoptosis; spared normal astrocytes and fibroblasts; prolonged survival in glioma models[113]
MV-miR-122Engineered to encode microRNA-122 (miR-122); tested with luciferase reporter systemIn vitro glioma cell modelDemonstrated that MV may have the potential to deliver functional miRNA and leads to a reduced target protein expression by 40%; limited by Drosha-mediated miRNA processing inefficiency in cytoplasm[114]
MeV + RuxolitinibCombination of wild-type Edmonston-lineage MeV with JAK1 pathway inhibitor ruxolitinibPatient-derived xenograft (PDX) glioblastoma models; 22-gene expression analysisJAK1 inhibition increased MeV replication and therapeutic sensitivity; highlighted potential for combination virotherapy and immune modulation[115]
MeV-stealthMeV glycoproteins replaced with CDV glycoproteins; CDV-H fused with CD46-targeting scFv; CDV-F signal peptide modified to redirect tropismMultiple tumor xenografts in immunodeficient miceImmune-evasive tumor targeting; specific lysis of CD46-overexpressing tumors; reduced off-target effects[116]
MV-L16 (live attenuated measles virus strain)GBM-derived primary cell lines (Gbl7n, Gbl11n, etc.)Preclinical translational study (ex vivo analysis of patient-derived glioma lines)MV-L16 induced significant caspase-3/7 activation, confirming apoptosis. mRNA-seq showed predictive immunologic gene expression changes in virus-sensitive GBM cells.[117]
Clinical Studies
Virus UsedDesign and InterventionAdverse EventsNotable FindingsRef.
MV-CEA (Edmonston strain expressing carcinoembryonic antigen)22 patients with recurrent glioblastoma (GBM)
Phase I, First-in-human trial (NCT00390299). Group A: MV-CEA injected into resection cavity. Group B: Intratumoral injection Day 1, followed by resection cavity injection Day 5.
No dose-limiting toxicity, even at max dose. Minor, manageable adverse events (not specified in detail).Repeated intratumoral MV-CEA is safe. CEA levels correlate with viral replication. Dual-Labeling Diagnostic Assay (DLDA) may predict viral response.[118]
oMeV: Oncolytic Measles Virus, GBM: Glioblastoma, CEA: Carcinoembryonic Antigen, MV: Measles Virus, miR: MicroRNA, JAK: Janus Kinase, PDX: Patient-Derived Xenograft, CDV: Canine Distemper Virus, scFv: Single-Chain Variable Fragment, CD46: Complement Decay-Accelerating Factor, mRNA-seq: Messenger RNA Sequencing, DLDA: Dual-Labeling Diagnostic Assay, NS: Not Specified.
Table 6. Preclinical and Clinical Evaluation of Oncolytic Newcastle Disease Virus (oNDV) Variants in Glioma Therapy.
Table 6. Preclinical and Clinical Evaluation of Oncolytic Newcastle Disease Virus (oNDV) Variants in Glioma Therapy.
oNDV VariantGenetic Modifications/AdditionsStudy ModelKey FindingsRef.
Preclinical Studies
Wild-type NDVNoneHuman GBM cell lines (GBM18, GBM27, etc.) in immunodeficient miceIFN-I gene cluster deletion enhanced NDV replication and tumor lethality; NS1-expressing NDV overcame IFN-I resistance[121]
NDV-2F/2HN-IFNγIncorporated F and HN genes from APMV-2; added human IFN-γ geneHuman PBMCs, chicken embryo fibroblasts (CEFs), Caco-2 (colon cancer) cell lineEnhanced IFN-γ expression and immune response; increased cancer cell death; evaded NDV-specific antibodies[123]
NDV + TMZ-PLGA-NPsCombination of wild-type NDV with Temozolomide-loaded PLGA nanoparticlesHuman GBM cell lines (in vitro)Combination therapy improved cytotoxicity over individual agents; TMZ-NPs enhanced drug stability and delivery[124]
rNDV-PTEN (Pos. 1 vs. 2)PTEN gene inserted between NP-P (Pos. 1) or P-M (Pos. 2) genes in NDV genomeT98G GBM cells in immunodeficient mice (in vitro and in vivo)PTEN overexpression (especially in Pos. 1) suppressed cancer growth markers (P-Akt, hTERT); intratumoral injection more effective than intravenous delivery[125]
Clinical Studies
Virus UsedDesign and InterventionAdverse EventsNotable FindingsRef.
NDV-HUJPhase I/II, open-label trial (n = 11); phase I: escalating weekly cycles (0.1 to 55 BIU); phase II: 3 weekly cycles of 5 days at 11 BIU, then maintenance 2x/weekNo adverse events related to NDV-HUJNDV found in blood, urine, CSF, saliva, tumor; 1 pt had complete remission but relapsed in 3 months; anti-NDV antibodies plateaued at 8 weeks[126]
MTH-68/HCase series (n = 4); daily to twice daily dosing ranging from 2 × 107 to 2.5 × 108 PFUNot reportedTumor regression, neurological improvement, steroid withdrawal; long-term survival 5–9 years from diagnosis[127]
UlsterNonrandomized controlled study (n = 22); ASI vaccine (NDV + cisplatin-inactivated autologous tumor) via cutaneous injection every 2 wks × 5; CG had chemo (nimustine + VM-26)No significant adverse events with ASIASI induced immune response via DTH; no survival difference, but TG (ASI) had mean survival of 46 weeks vs. 48 weeks for CG[128]
Ulster (ATV-NDV)Nonrandomized controlled study (n = 111); NDV-inactivated autologous tumor cells injected q3–4 wks up to 8 doses post-RTNo significant adverse events with ATV-NDVMedian OS: TG 100 wks, CG-NS 49 wks, CG-S 88 wks; elevated CD8+ TILs, increased memory T cells in long-term survivors; durable immune memory[129]
oNDV: Oncolytic Newcastle Disease Virus, GBM: Glioblastoma, IFN: Interferon, NDV: Newcastle Disease Virus, PBMCs: Peripheral Blood Mononuclear Cells, CEFs: Chicken Embryo Fibroblasts, TMZ: Temozolomide, PLGA: Poly (lactic-co-glycolic acid), NP: Nucleoprotein, P: Phosphoprotein, M: Matrix Protein, DTH: Delayed-Type Hypersensitivity, OS: Overall Survival, TILs: Tumor-Infiltrating Lymphocytes.
Table 7. Clinical Evaluation of Reovirus (Pelareorep) Therapy in Adult and Pediatric Gliomas: Delivery, Safety, and Immune Responses.
Table 7. Clinical Evaluation of Reovirus (Pelareorep) Therapy in Adult and Pediatric Gliomas: Delivery, Safety, and Immune Responses.
Virus UsedPatient PopulationDesign and InterventionAdverse EventsNotable FindingsRef.
Reovirus (Pelareorep)Recurrent malignant gliomasPhase I trial; intratumoral catheter-based infusion for 72 hrsOne transient grade 3 seizure; otherwise well toleratedVirus reached tumor; histology showed cell death and immune activation; safe at high doses[135]
Reovirus + GM-CSFNewly diagnosed GBMReoGlio (2017–2020): IV pelareorep + GM-CSF during concurrent chemoradiation and adjuvant TMZFlu-like symptoms, reversible hypotension; no major safety concerns or treatment delaysVirus delivery to tumor confirmed; some T cell activity observed in resected tumors; early signals of immunological engagement[136]
ReovirusNewly diagnosed GBMWindow-of-Opportunity trial: single IV dose prior to surgeryWell toleratedViral RNA/proteins detected in tumor tissue; increased CD8+ T cell activity; proof of BBB crossing and immunogenic modulation[137]
Reovirus + GM-CSFPediatric high-grade gliomas (incl. GBM)Small trial (IV Pelareorep + GM-CSF)Grade 3 hyponatremia (1 patient); overall well toleratedAll patients progressed within weeks; early antibody neutralization likely impacted efficacy; confirms tumor entry via IV but poor clinical outcome[138]
GBM: Glioblastoma, IV: Intravenous, TMZ: Temozolomide, GM-CSF: Granulocyte-Macrophage Colony-Stimulating Factor, BBB: Blood–Brain Barrier, CD8+: Cytotoxic T-Cells, TILs: Tumor-Infiltrating Lymphocytes, RNA: Ribonucleic Acid, PFU: Plaque-Forming Units.
Table 8. Preclinical and Clinical Evaluation of Genetically Modified Oncolytic Retroviruses in Glioblastoma Therapy.
Table 8. Preclinical and Clinical Evaluation of Genetically Modified Oncolytic Retroviruses in Glioblastoma Therapy.
Virus VariantGenetic Modifications/AdditionsStudy ModelKey FindingsRef.
Pre-clinical Studies
MLV-based RCRRCR vector expressing yeast cytosine deaminase gene with 5-FC prodrugCell lines, mouse modelsSingle injection led to widespread tumor infection and enhanced survival; tumor-selective expression of suicide gene.[139]
Toca 511Amphotropic MLV encoding modified yeast cytosine deaminase 2 (yCD2)In vitro, in vivoTumor-targeted delivery; converts 5-FC to 5-FU; safe replication; efficient and stable gene delivery in glioblastoma.[140]
oFV-GFPoFV vector with chimpanzee virus PAN1/PAN2 genes and green fluorescent protein (GFP) transgeneGBM cells, mouse modelsSafe, stable, and tumor-persistent; spreads well even in slow-growing tumors; prolongs survival; can reactivate after dormancy.[141]
oFV-GFP, oFV-TK, oFV-iCasp9GFP, thymidine kinase (TK), and inducible caspase 9 (iCasp9) inserted into oFV vectorsHuman GBM cells, mouse modelsEffective long-term gene expression; GFP expression persisted for 66 days; larger genes like iCasp9 lost over time during replication in tumors.[142]
Clinical Studies
Virus UsedDesign and InterventionAdverse EventsNotable FindingsRef.
Toca 51145 patients with glioblastomaPhase 1 trial; virus injected after surgery followed by oral Toca FC and standard chemoradiotherapyGrade 3 asthenia, hydrocephalus, thrombocytopenia, procedural pain[143]
Toca 51156 patients with recurrent high-grade gliomaPhase 1 dose-escalation; intratumoral virus injection post-surgery + Toca FC oral prodrugGrade 2 skin rash, mucositis, facial swelling; Grade 3 hemorrhagic enteritis and colitis[144]
Toca 51156 high-grade glioma patientsPhase 1; integrated omics analysis (WES, RNA-seq, ELISA) on tumor and blood to assess responseNot disclosed[145]
Toca 511127 high-grade glioma patientsLarge study using extended-release Toca FC and molecular monitoring (RNA/DNA tracking, integration site analysis, mutation profiling)Hematologic adverse events including lymphoma[146]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alomari, O.; Eyvazova, H.; Güney, B.; Al Juhmani, R.; Odabasi, H.; Al-Rawabdeh, L.; Mokresh, M.E.; Erginoglu, U.; Keles, A.; Baskaya, M.K. Oncolytic Therapies for Glioblastoma: Advances, Challenges, and Future Perspectives. Cancers 2025, 17, 2550. https://doi.org/10.3390/cancers17152550

AMA Style

Alomari O, Eyvazova H, Güney B, Al Juhmani R, Odabasi H, Al-Rawabdeh L, Mokresh ME, Erginoglu U, Keles A, Baskaya MK. Oncolytic Therapies for Glioblastoma: Advances, Challenges, and Future Perspectives. Cancers. 2025; 17(15):2550. https://doi.org/10.3390/cancers17152550

Chicago/Turabian Style

Alomari, Omar, Habiba Eyvazova, Beyzanur Güney, Rana Al Juhmani, Hatice Odabasi, Lubna Al-Rawabdeh, Muhammed Edib Mokresh, Ufuk Erginoglu, Abdullah Keles, and Mustafa K. Baskaya. 2025. "Oncolytic Therapies for Glioblastoma: Advances, Challenges, and Future Perspectives" Cancers 17, no. 15: 2550. https://doi.org/10.3390/cancers17152550

APA Style

Alomari, O., Eyvazova, H., Güney, B., Al Juhmani, R., Odabasi, H., Al-Rawabdeh, L., Mokresh, M. E., Erginoglu, U., Keles, A., & Baskaya, M. K. (2025). Oncolytic Therapies for Glioblastoma: Advances, Challenges, and Future Perspectives. Cancers, 17(15), 2550. https://doi.org/10.3390/cancers17152550

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

Article Metrics

Back to TopTop