Next Article in Journal
Single-Port Versus Multi-Port Robotic Radical Prostatectomy in Elderly Patients
Previous Article in Journal
Beneficial Outcomes of Immunoenhancing Nutritional Interventions in Perioperative Care for Oral Cancer: A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Oncolytic Viruses as a Novel Therapeutic Approach for Colorectal Cancer: Mechanisms, Current Advances, and Future Directions

by
Francisco Pérez-Domínguez
1,2,
Claudia Quezada-Monrás
3,4,
Leonardo Cárcamo
5,
Juan P. Muñoz
6,* and
Diego Carrillo-Beltrán
1,4,*
1
Laboratorio Oncovirología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5090000, Chile
2
Servicio Medicina Interna, Hospital Base Valdivia, Valdivia 5090000, Chile
3
Laboratorio de Biología Tumoral, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5090000, Chile
4
Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
5
Servicio de Cirugía, Hospital Base Valdivia, Valdivia 5090000, Chile
6
Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile
*
Authors to whom correspondence should be addressed.
Cancers 2025, 17(11), 1854; https://doi.org/10.3390/cancers17111854
Submission received: 13 April 2025 / Revised: 15 May 2025 / Accepted: 20 May 2025 / Published: 31 May 2025
(This article belongs to the Section Cancer Therapy)

Simple Summary

Colorectal cancer (CRC) remains one of the most prevalent and deadly malignancies worldwide, often diagnosed at advanced stages and showing limited response to conventional therapies. Oncolytic virotherapy has emerged as a novel and promising approach that combines direct tumor cell killing with immune system activation. This review summarizes the current state of oncolytic viruses (OVs) under investigation for CRC, including adenovirus, herpes simplex virus (HSV), reovirus, vesicular stomatitis virus (VSV), vaccinia virus (VV), and measles virus (MV). Each viral platform is discussed in terms of its mechanism of action, preclinical and clinical data, limitations, and future directions. Overall, oncolytic viruses represent a dynamic and evolving therapeutic class with the potential to address unmet needs in CRC treatment and improve patient outcomes.

Abstract

This review provides an updated overview of oncolytic virotherapy as a promising therapeutic strategy for colorectal cancer (CRC), focusing on six key viral platforms: adenovirus, herpes simplex virus (HSV), reovirus, vesicular stomatitis virus (VSV), vaccinia virus (VV), and measles virus (MV). These viruses exhibit tumor-selective replication and exert their effects through mechanisms such as direct oncolysis, the delivery of immunostimulatory genes (e.g., IL-12, IL-15, GM-CSF), the activation of innate and adaptive immune responses, and the remodeling of the tumor microenvironment. Preclinical and early clinical studies suggest that oncolytic viruses can enhance the efficacy of existing treatments, particularly in immunologically “cold” tumors such as microsatellite stable CRC, when used in combination with chemotherapy or immune checkpoint inhibitors. Despite encouraging results, several challenges remain, including antiviral immune clearance, tumor heterogeneity, and limitations in systemic delivery. Current research focuses on improving viral engineering, enhancing tumor targeting, and designing combinatorial strategies to overcome resistance and maximize clinical benefits. Overall, oncolytic viruses represent a versatile and evolving therapeutic class with the potential to address unmet clinical needs in CRC.

1. Introduction

Colorectal cancer (CRC) was the fourth most commonly diagnosed cancer and the second leading cause of cancer-related deaths in the United States in 2024, following lung and bronchial cancer [1]. Due to widespread screening programs, CRC incidence has declined over recent decades. However, since the mid-1980s, incidence rates have been increasing among adults aged 20–39, with a similar upward trend observed in individuals aged 40–54 since the mid-1990s. Between 2011 and 2019, the incidence rate increased by approximately 2% annually in those under 50, including individuals aged 50–54 [2]. Interestingly, despite a significant annual decline of 4.3% in the incidence of localized-stage disease from 2006 to 2019, there has been an increase in advanced-stage diagnoses [2]. The transition to later-stage diagnoses can be attributed to the saturation of screening programs, the tendency to disproportionately detect and remove slower-growing adenomas rather than more aggressive ones, and the rising prevalence of early-onset CRC, which is more frequently diagnosed at an advanced stage [3]. The first-line treatment of advanced or metastatic CRC (mCRC) typically consists of a chemotherapy combination, often associated with a biologic agent, based on the specific molecular profile of the patient. For this reason, patients with advanced CRC should undergo genetic tumor analysis to detect KRAS/NRAS and BRAF mutations, HER2 amplifications, and microsatellite instability (MSI)/mismatch repair (MMR) status, among other biomarkers [4]. The chemotherapy drugs utilized in first- and second-line treatment for mCRC include 5-Fluorouracil (5-FU), capecitabine, oxaliplatin, and irinotecan, which may be combined with biologic agents targeting the vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR) pathways, as well as immunotherapy options [5]. Bevacizumab is a humanized monoclonal antibody that inhibits VEGF, a key factor in tumor angiogenesis. Combined data from multiple randomized phase II trials have demonstrated that incorporating bevacizumab into first-line 5-FU therapy enhances overall survival (OS) in patients with unresectable mCRC compared to those treated with these regimens without bevacizumab [6,7]. Similarly, cetuximab and panitumumab, monoclonal antibodies targeting EGFR and blocking its downstream signaling pathways, offer significant clinical benefits in treating patients with RAS wild-type mCRC [8,9].
Despite these advancements, several limitations have been identified in the existing first-line treatments. One of the most relevant challenges is the diverse molecular nature of colorectal tumors, which plays a crucial role in treatment resistance, prompting the exploration of combination therapies designed to bypass both intrinsic and acquired resistance by simultaneously targeting multiple carcinogenic pathways. However, a significant drawback of using numerous targeted agents is the increased risk of cumulative toxicity [10,11]. These limitations have driven the development of novel therapeutic approaches, among which virotherapy has emerged as a promising option. Oncolytic virotherapy refers to genetically engineered or naturally occurring viruses that selectively infect, replicate, and destroy cancer cells while sparing normal tissues. This selectivity is often attributed to the impaired antiviral responses and dysregulated signaling pathways within tumor cells [12]. Unlike traditional therapies such as chemotherapy and radiotherapy, which often lack specificity and can lead to systemic toxicity, oncolytic viruses (OVs) provide both direct tumor lysis and a robust activation of anti-tumor immune responses, offering a dual therapeutic effect [13]. Several viral platforms have been explored for colorectal cancer (CRC), including adenovirus, herpes simplex virus (HSV), reovirus, vesicular stomatitis virus (VSV), vaccinia virus (VV), and measles virus, each with distinct mechanisms of action, safety profiles, and clinical progress [14]. Given these properties, OVs represent a compelling and versatile strategy within the expanding arsenal of cancer immunotherapies (Figure 1) [15].
In this review, we describe the mechanisms of action of OVs, analyze the most recent preclinical and clinical studies in CRC, and discuss the limitations, challenges, and future perspectives in this field of study.

2. Oncolytic Viruses

OVs are self-replicating microorganisms, either naturally occurring or genetically modified, that selectively infect and multiply within tumor cells [16]. Their ability to preferentially target cancer cells arises from the molecular abnormalities that drive tumor progression, such as defects in antiviral signaling pathways and dysregulated cell cycle control [17]. Tumorigenesis is a multistep process involving gene mutations, which collectively lead to malignant transformation. OVs exploit these vulnerabilities, allowing for selective replication in tumor cells while sparing normal tissues.
The anti-oncogenic effects of OVs have been recognized since the mid-20th century, as shown in studies by Moore [18,19]. However, it was not until recent advances were made in molecular oncology that the development of OV-based therapies became feasible on a clinical scale [20]. A major breakthrough occurred with a Phase III randomized clinical trial evaluating a genetically engineered herpes simplex virus type 1 (HSV-1) expressing granulocyte–macrophage colony-stimulating factor (GM-CSF). This trial demonstrated a significantly higher durable response rate in patients with advanced melanoma, leading to the approval of this virus (talimogene laherparepvec (T-VEC)) by the U.S. Food and Drug Administration (FDA) as the first oncolytic virus-based immunotherapy for cancer [21].
OVs eliminate tumor cells through multiple mechanisms. First, they induce direct oncolysis by replicating within tumor cells, leading to cell lysis and the release of new viral particles that spread the infection. This process is facilitated by tumor-specific genetic mutations that allow for viral replication while restricting infection in healthy tissues [20]. Second, OVs trigger immunogenic cell death, releasing tumor-associated antigens (TAAs) and danger-associated molecular patterns (DAMPs), which stimulate anti-tumor immunity. This activation enhances dendritic cell function and primes cytotoxic T lymphocytes to target cancer cells beyond the initial infection site [20,22]. Third, OVs reshape the tumor microenvironment (TME) by counteracting immunosuppressive signals. They inhibit regulatory T cells and myeloid-derived suppressor cells while promoting the release of proinflammatory cytokines and chemokines that strengthen immune responses [23].
Additionally, some OVs are engineered to express therapeutic genes that enhance their anti-cancer efficacy. These modifications may include the expression of cytokines, prodrug-converting enzymes, or immune checkpoint inhibitors (ICIs) that further amplify immune activation [24,25]. Together, these mechanisms create a robust anti-tumor response, positioning OVs as promising agents in cancer therapy, particularly in combination with immunotherapies and conventional treatments.
Among emerging cancer treatments, combining oncolytic virotherapy with other immunotherapies—particularly ICIs and chimeric antigen receptor T-cell (CAR-T) therapy—has shown significant potential [26,27]. OVs play a crucial role in converting immunologically “cold” tumors into “hot” ones, thereby increasing their sensitivity to ICIs [28]. Additionally, OVs can function as carriers for CAR-T cells, facilitating their infiltration into the tumor and overcoming the immunosuppressive microenvironment, thus enhancing their cytotoxic activity against solid tumors [29]. This dual approach capitalizes on the immune-modulating properties of OVs to improve the efficacy of ICIs and CAR-T therapies, making it a promising strategy in cancer treatment.

3. Types of OVs Studied for CRC

3.1. Adenovirus

3.1.1. Mechanism of Action

Adenoviruses are non-enveloped, double-stranded DNA viruses with a well-characterized genome, making them highly amenable to genetic engineering [30,31]. Their high transduction efficiency allows for effective gene delivery to various cell types. Additionally, their ability to induce strong immune responses makes them valuable for vaccine development and oncolytic therapy [32].
Oncolytic adenoviruses exhibit selective replication in CRC cells, leading to the direct lysis of tumor cells and the release of tumor-associated antigens [33]. Gene-silencing strategies have also been explored, such as an shRNA-expressing adenovirus targeting DNA-PKcs, a key enzyme in DNA repair. Adenoviral vectors have modulated the TME, facilitating systemic anti-tumor effects such as the abscopal phenomenon [34]. The telomerase-specific oncolytic adenovirus OBP-301 (Telomelysin) utilizes extracellular vesicles (EVs) to deliver viral components to distant metastatic sites [35]. These EVs induce apoptosis and autophagy while circumventing immune suppression, thereby increasing tumor-specific targeting and reducing off-target effects [36]. This mechanism supports combination with ICIs for enhanced immunotherapeutic outcomes [36]. Genetic engineering has enabled the expression of immune-stimulatory molecules by oncolytic adenoviruses, amplifying their therapeutic impact. A prominent example is the development of IL-15-expressing adenoviruses, which enhance the activity of natural killer (NK) cells, CD8+ T cells, and other immune effectors [37,38]. In CRC models, combining IL-15-expressing oncolytic adenoviruses with cytotoxic T lymphocytes (CTLs) has demonstrated superior tumor suppression, reinforcing the potential of virotherapy in immuno-oncology [37]. Another innovation is the construction of a bicistronic oncolytic adenovirus (Ad-CD-GMCSF), encoding cytosine deaminase (CD) and granulocyte-macrophage colony-stimulating factor (GM-CSF). CD converts the prodrug 5-fluorocytosine (5-FC) into the chemotherapeutic agent 5-FU, inducing tumor cell death, while GM-CSF recruits antigen-presenting cells, such as dendritic cells and macrophages, to enhance T-cell priming [39,40]. This dual-function approach improved tumor clearance, enhanced survival, and modulated the TME by reducing immunosuppressive cytokines [39].

3.1.2. Clinical and Preclinical Evidence

Studies demonstrate improved efficacy via co-administration with chemotherapy or gene silencing vectors. Several adenoviral constructs show tumor suppression in xenograft models and patient-derived xenografts. This oncolytic activity promotes T-cell infiltration and activation, thereby enhancing the efficacy of ICIs [41]. In the same way, enhancements such as L-carnosine loading improve viral transduction and apoptosis, boosting therapeutic efficacy [42]. A notable example is the chimpanzee-derived adenovirus AdC7-SP/E1A-ΔE3, which is engineered to bypass pre-existing immunity to human adenoviruses. This construct selectively replicates in CRC cells and induces p53-independent mitochondrial apoptosis [43]. Further strategies involve combination therapies to enhance efficacy. For instance, a conditionally replicative adenovirus (CRAd) co-administered with valproic acid (VPA), a histone deacetylase inhibitor, significantly increased tumor suppression relative to either agent alone. Mechanistically, this combination reduced proliferation, triggered DNA damage, increased polyploidy, and induced H2AX phosphorylation, which is indicative of DNA double-strand breaks, while downregulating DNA repair proteins [44].
In adenovirus gene-silencing strategies, this vector increased radiosensitivity; its effect was limited in xenograft models [45,46,47,48]. However, combining it with a CRAd improved gene delivery and radiosensitization, underscoring the promise of adenovirus-based radiosensitizing approaches for CRC [48]. Another study designed a conditionally replicative oncolytic adenovirus, Ad312-E1A, to target CRC cells with IGF2 imprinting (LOI) loss, a common epigenetic abnormality in CRC and other malignancies [49,50]. The virus selectively replicates in IGF2 LOI-positive tumor cells, reducing cell viability and inducing apoptosis, while sparing normal cells. In xenograft models, Ad312-E1A effectively suppressed tumor growth and prolonged survival, demonstrating its potential as a targeted gene therapy approach [50]. Another strategy involved an adenovirus engineered to deliver TRAIL (tumor-necrosis-factor-related apoptosis-inducing ligand), which induced apoptosis with minimal impact on healthy cells. In patient-derived xenograft models, the TRAIL-expressing construct (Ad/TRAIL-E1) provided superior tumor suppression and survival benefits compared to conventional virotherapy [51].
Another telomerase-specific oncolytic adenovirus, OBP-502, enhances ICI efficacy by promoting immunogenic cell death (ICD). It induces the release of ATP and HMGB1, leading to CD8+ T-cell infiltration and Foxp3+ T-cell suppression, thereby improving anti-tumor immunity. When combined with PD-1 blockade, OBP-502 exhibits synergistic effects, including tumor suppression and an abscopal effect in CRC models [52]. Furthermore, a synergy between oncolytic adenovirus dl1520 (ONYX-015) and chemotherapy (melphalan) has demonstrated enhanced tumor cell death in colorectal adenocarcinoma models [53]. ONYX-015 selectively replicates in p53-deficient tumors, constituting over 50% of all cancers, making it a promising targeted therapy [54]. Combining ONYX-015 with chemotherapy significantly increases apoptosis and inhibits proliferation, highlighting the potential of virus–chemotherapy combinations [53]. These findings suggest that oncolytic virotherapy can enhance conventional treatments, improving cancer cell selectivity and reducing resistance.

3.2. Herpes Simplex Virus (HSV)

3.2.1. Mechanism of Action

HSV is a large double-stranded DNA virus with a 152 kb genome enclosed in an icosahedral capsid and a lipid envelope [55]. These structural and genomic features make HSV an attractive platform for genetic modification to improve tumor specificity and patient safety. Unlike retroviruses, HSV replicates within the host cell nucleus without integrating into the host genome, thus avoiding insertional mutagenesis. Furthermore, the virus remains sensitive to standard antiviral agents, adding a layer of safety for clinical use [56]. Oncolytic HSVs (oHSVs) selectively replicate in tumor cells, promoting direct lysis and the release of tumor-associated antigens. This cytolytic activity triggers immunogenic cell death and activates innate and adaptive immune responses. Additionally, oHSVs can be engineered to express immunostimulatory genes, further enhancing anti-tumor immunity and therapeutic efficacy [57,58].
In summary, oHSVs selectively replicate in tumor cells, causing oncolysis and antigen release. They modulate the tumor microenvironment and can be engineered to express immunostimulatory transgenes.

3.2.2. Clinical and Preclinical Evidence

One of the primary therapeutic mechanisms of oHSV is direct tumor cell lysis. NV1020 is a well-studied oHSV that selectively replicates in cancer cells, inducing cell death while sparing normal tissue [59,60]. Preclinical studies have shown that NV1020, when combined with chemotherapeutic agents such as 5-FU, SN38, or oxaliplatin, leads to enhanced tumor suppression through additive or synergistic interactions. In murine CRC models, this combination therapy significantly reduced tumor volume and prolonged survival [61]. Another notable example is NV1066, an oHSV variant capable of selectively targeting and eliminating tumor-initiating cells (TICs) in CRC. TICs, identified through serum-free culture, exhibit high tumorigenic potential and overexpress Akt1, a protein linked to cancer cell survival and stemness [62,63]. NV1066 efficiently infects and replicates within TICs, inducing oncolysis and reducing tumor growth in vivo [63]. Targeting cancer stem-like cells (CSCs) further, Signal-Smart 2 (SS2), an engineered oHSV-1, was developed to selectively eliminate CD133+ cells, a subpopulation associated with tumor aggressiveness and therapeutic resistance. SS2 exhibited high specificity and reduced tumor invasiveness and progression in both in vitro and in vivo models [64]. Similarly, oHSV-2 has demonstrated potent anti-tumor effects by selectively targeting CSCs and bulk tumor cells. In CRC models, oHSV-2 treatment resulted in tumor necrosis and reduced cell invasion, and it synergized with 5-FU to enhance therapeutic efficacy [65,66].
One approach by which OHVs transform the TME involves High-Mobility Group Box 1 (HMGB1), which is a ubiquitous and highly conserved protein involved in gene regulation, immune response, and apoptosis [67,68]. Previous studies suggest that HMGB1 can inhibit aerobic respiration and alter mitochondrial metabolism, potentially leading to tumor cell death under low-oxygen conditions [69,70]. Hence, oHSV-1 expressing the HMGB1 protein (HSV-HMGB1) has shown enhanced cytotoxicity in normoxia but, paradoxically, increased colon cancer cell viability in hypoxia by inducing autophagy rather than apoptosis. A mechanistic analysis revealed that HSV-HMGB1 modulates the subcellular localization of HMGB1 and p53, influencing cell survival pathways [71]. Likewise, oHSV2 effectively targets murine colon carcinoma by altering the immune microenvironment and inducing anti-tumor immunity [72]. The virus reduces immunosuppressive cells while increasing CD8+ T cells, NK cells, and dendritic cells, thereby enhancing systemic immune responses. In vivo, oHSV-2 treatment eradicated primary tumors, prevented recurrence, and improved survival without notable toxicity, underscoring its potential as a safe and effective therapeutic agent [72].
oHSVs can be further modified to express immunostimulatory molecules, amplifying host anti-tumor immunity. One such construct, oHSV1-IL15B, encodes the IL-15/IL-15Rα complex to enhance cytotoxic T-cell responses. Combined with oHSV1-aPD1 expressing an anti-PD-1 antibody, this dual therapy elicited robust CD8+ T-cell activation and increased tumor apoptosis [73,74,75]. Another attenuated HSV-1 vector, HSV-G47Δ, has shown efficacy in an orthotopic rectal cancer mouse model by reducing tumor size and improving survival without systemic toxicity. The study also revealed that inhibition of the protein kinase R (PKR) antiviral pathway enhanced viral replication and oncolytic activity [76]. Further advancements include the development of O-HSV1211, an oHSV-1 engineered via CRISPR/Cas9 to express IL-12 and the chemokine CXCL11. This construct promoted immune cell infiltration, stimulated interferon-gamma (IFN-γ) production, and significantly improved tumor control and survival in CRC models [77].

3.3. Reovirus

3.3.1. Mechanism of Action

Reovirus is a non-enveloped, double-stranded RNA virus with a segmented genome and an icosahedral capsid [78]. It selectively replicates in cancer cells harboring activated Ras signaling pathways, leading to direct tumor cell lysis and the activation of anti-tumor immune responses [79]. In addition to inducing innate and adaptive immunity, reovirus enhances T-cell and NK cell activity. Its capacity for systemic delivery and compatibility with chemotherapy and immunotherapy strategies highlights its potential as a promising oncolytic platform [80]. The following sections summarize the latest findings regarding the mechanisms of action of reoviruses in colon tumors.
One of the principal mechanisms of reovirus-mediated oncolysis involves modulating intracellular signaling pathways. Glycogen synthase kinase-3β (GSK-3β), a central regulator of the Wnt/β-catenin signaling pathway, is known to phosphorylate β-catenin, thereby promoting its degradation and attenuating oncogenic signaling [81]. The inhibition of GSK-3β has been shown to enhance reovirus-induced apoptosis in colon cancer cells. Although the upregulation of β-catenin does not increase viral replication, GSK-3β inhibition suppresses NF-κB activity, accelerating apoptosis via caspase-8 activation. This co-treatment strategy significantly amplifies cancer cell death compared to reovirus alone [82]. Another signaling pathway that has been evaluated is KRAS, a proto-oncogene that encodes a small GTPase involved in cell signaling pathways that regulate tumor growth, differentiation, and survival [83]. Reovirus can also reshape the TME, enhancing immune surveillance and systemic anti-tumor effects [84].
Reovirus has been shown to directly stimulate immune effector cells, particularly NK cells. Viral exposure increases NK cell cytotoxicity in a dose-dependent manner by activating Toll-like receptor 3 (TLR3) signaling. This activation leads to the upregulation of cytotoxic proteins such as perforin and granzymes, along with proinflammatory cytokines including TNF-α and IFN-γ [85]. Furthermore, when reovirus is combined with cetuximab, an anti-EGFR monoclonal antibody, it significantly enhances antibody-dependent cell-mediated cytotoxicity (ADCC) against CRC cells, regardless of the KRAS mutation status. Compared to monotherapies, this combinatorial approach leads to tumor suppression in preclinical models [86].
Another immune-enhancing strategy involves inhibiting transforming growth factor-beta (TGF-β), a cytokine that promotes tumor progression by facilitating epithelial-to-mesenchymal transition and immune evasion. TGF-β blockade significantly boosts reovirus-induced T-cell infiltration. Combined with CD3-bispecific antibody therapy, this regimen achieves complete tumor regression in CRC models [87,88].
In summary, reovirus preferentially targets KRAS-mutant cells, inducing oncolysis and activating innate immune pathways, particularly via TLR3.

3.3.2. Clinical and Preclinical Evidence

Oncolytic reovirus preferentially induces apoptosis in KRAS-mutant CRC cells and synergizes with irinotecan, a topoisomerase I inhibitor [89]. The oral administration of reovirus RC402 has also been tested, which activates immune responses in Peyer’s patches of the terminal ileum. This stimulation promotes antigen presentation and T-cell activation in distant tumor sites, even without direct viral infection of the tumors. Notably, this effect depends on the gut microbiome, as its depletion abolishes the therapeutic benefit. When combined with immune checkpoint inhibitors (anti–PD-1 and anti–CTLA-4), oral reovirus administration achieves complete tumor regression and establishes durable immune memory, highlighting its promise as a non-invasive immunotherapy [90]. Likewise, Augustine et al. (2022) [84] have shown that oncolytic reovirus enhances immune responses in microsatellite-stable CRC (MSS CRC), making it more sensitive to anti-PD-1 therapy. Reovirus induces direct tumor cell apoptosis, activates innate immune pathways, increases PD-L1 expression, and promotes T-cell infiltration, reducing immunosuppressive macrophages. In murine models, the combination of reovirus and anti-PD-1 significantly reduces tumor growth and improves survival, which suggests that reovirus can convert immune-resistant MSS tumors into immunotherapy-responsive cancers [84]. In a clinical setting, reovirus administration in metastatic CRC patients harboring KRAS mutations has demonstrated substantial immunomodulatory effects. Treatment increased anti-tumor cytokines (GM-CSF, IL-12, IFN-γ), decreased pro-tumorigenic cytokines (IL-8, VEGF), and activated antigen-presenting cells and CD8+ T cells. In addition, reovirus suppressed the expression of miR-29a-3p, a microRNA associated with CRC progression. Transcriptomic profiling revealed an upregulation of immune-related gene expression, reinforcing the dual role of reovirus as both a cytolytic agent and an immune stimulator [91].

3.4. Vesicular Stomatitis Virus (VSV)

3.4.1. Mechanism of Action

VSV is an RNA virus with a bullet-shaped morphology, a rapid replication cycle, and broad cellular tropism, making it an ideal oncolytic virus [92]. VSV selectively infects and kills cancer cells with defective IFN responses, while sparing normal cells. It induces direct oncolysis and triggers immune responses, enhancing anti-tumor immunity [93]. Due to its low pre-existing immunity in humans, ease of genetic modification, and ability to deliver therapeutic transgenes, VSV is a promising candidate for oncolytic virotherapy in CRC [94]. A key mechanism of VSV-mediated tumor suppression is the induction of apoptosis via its matrix (M) protein. Mutant forms of the M protein, such as ΔM51 and ΔM51R, have been studied for their improved safety and efficacy profiles. Both variants have demonstrated the ability to trigger caspase-dependent apoptosis in CRC cells, with ΔM51 offering enhanced biosafety due to a lower risk of reversion to a pathogenic phenotype [95]. VSV also exerts immunomodulatory effects that reshape the TME.

3.4.2. Clinical and Preclinical Evidence

In a murine model of the peritoneal surface dissemination (PSD) of CRC, the intraperitoneal administration of ΔM51R VSV significantly inhibited tumor progression, prolonged survival, and reprogrammed the TME. Notably, this intervention increased CD4+ T cells and peritoneal immune cell populations, while reducing immunosuppressive MDSCs and proinflammatory cytokines such as IL-6 and MCP-1 [96]. To further enhance immunotherapeutic efficacy, VSV has been genetically engineered to express interleukin-15 (IL-15), a potent cytokine that stimulates NK cells and CD8+ cytotoxic T cells. In murine models of metastatic colon adenocarcinoma, localized IL-15 expression from VSV significantly improved survival and promoted tumor clearance [97,98]. This effect was notably superior to systemic IL-15 administration, emphasizing the therapeutic potential of oncolytic viruses armed with immune-stimulating transgenes [98]. VSV is a promising cancer therapy; however, antiviral immune responses must be considered [99,100]. To address this limitation, Alluqmani et al. evaluated the combination of VSVΔ51 with the immunomodulatory enhancer vanadyl sulfate (VS) in colon cancer models, demonstrating that VS enhances viral replication and shifts immune signaling from type I to type II interferon responses [101,102]. The combined therapy significantly increased proinflammatory cytokine secretion, improved tumor antigen-specific T-cell responses, and enhanced overall anti-tumor immunity [101]. Similarly, octyl itaconate (4-OI) is a macrophage inflammatory response suppressor that modulates the transcriptional regulator NRF2 by alkylating its repressor KEAP1, thereby reducing inflammation and inhibiting type I IFN responses [103,104]. When used with VSVΔ51, 4-OI significantly enhanced viral replication and oncolysis, particularly in IFN-resistant colon cancer cells. Mechanistic studies showed that 4-OI alters critical components of the MAVS, NF-κB, and IFN pathways, resulting in improved therapeutic outcomes. Both in vitro and in vivo experiments, including studies on patient-derived tumoroids, confirmed the increased efficacy of this combination in resistant CRC models [105].

3.5. Vaccinia Virus (VV)

3.5.1. Mechanism of Action

VV is a large double-stranded DNA virus that replicates in the cytoplasm, avoiding host genome integration, making it a safe and stable oncolytic agent [106]. It has natural tumor tropism and a rapid replication cycle, and it can infect cancer cells under hypoxic conditions, which are common in tumors. Its large genome (~190 kb) allows for genetic modifications to express therapeutic genes (e.g., cytokines, immune checkpoint inhibitors), boosting anti-tumor immunity [107]. These properties make VV a versatile and practical oncolytic virus, capable of direct oncolysis, vascular disruption, and immune activation, improving its potential for combination cancer therapies. VV infection has also been shown to increase the infiltration of iNOS+ myeloid-derived suppressor cells (MDSCs), which contributed to nitric oxide (NO)-mediated tumor regression rather than suppressing viral spread [108]. Reprogramming the TME using chemokine-modulating (CKM) therapies has shown the potential to improve VV-based immunotherapy. VV can also be used as a vector to deliver immunostimulatory cytokines, amplifying systemic anti-tumor responses [109]. In summary, VV exerts cytolytic effects and expresses transgenes such as IL-12, IL-9, and checkpoint inhibitors. It prevents genome integration and operates in hypoxia.

3.5.2. Clinical and Preclinical Evidence

Several engineered VV constructs have demonstrated potent cytolytic activity in CRC models. One example is a conditionally replicative VV (VV-FCU1), which encodes the suicide gene FCU1. This gene converts the prodrug 5-fluorocytosine (5-FC) into the chemotherapeutic agent 5-FU, resulting in significant tumor reduction in both subcutaneous and orthotopic liver metastasis models of human CRC [110].
Jeong et al. (2020) [111] developed a novel VV construct (NOV) co-expressing TRAIL and angiopoietin-1 (Ang1), replacing viral thymidine kinase (vTK) and vaccinia growth factor (VGF) to enhance tumor selectivity and reduce off-target effects. NOV exhibited superior oncolytic activity, apoptosis induction, and therapeutic efficacy compared to conventional VV strains. In syngeneic CRC mouse models, NOV significantly suppressed tumor growth, increased CD8+ T cell infiltration, and prolonged survival [111]. Additionally, VVs engineered with deletions of immunomodulatory genes such as N1L, K1L, K3L, A46R, and A52R have shown enhanced tumor selectivity and anti-tumor efficacy. These deletions reduce the virus’s ability to evade host interferon responses, thereby decreasing replication in normal tissues while maintaining infectivity in tumor cells. This approach improved CRC models’ tumor lysis, immune infiltration, and survival outcomes [112].
Histone deacetylase inhibitors (HDIs) are epigenetic modulators that suppress antiviral responses, particularly IFN signaling, making cancer cells more susceptible to viral infection [113,114]. Trichostatin A (TSA), a potent HDI, was found to significantly improve vaccinia virus (VV) replication and spread in cancer cells, while having minimal effects on normal cells [115]. Deleting the B18R gene, which encodes a soluble IFN receptor, enhances VV safety by increasing viral clearance from normal tissues, although it may also reduce viral replication in tumors [116,117]. In a colon cancer xenograft model, combining TSA and a B18R-deleted VV delayed tumor progression and increased survival compared to either treatment alone [118]. An oncolytic VV expressing an anti-CD47 nanobody (OVV-αCD47nb) blocks the CD47-SIRPα “don’t eat me” signal, enhancing macrophage-mediated phagocytosis and promoting CD8+ T cell activation. OVV-αCD47nb outperformed unmodified VV in tumor regression and survival, and reprogrammed tumor-associated macrophages from an M2 immunosuppressive phenotype to an M1 proinflammatory state [119]. On the other hand, combining vvDD-CXCL11 with CKM cocktails increased Th1-associated chemokines (CXCL9, CXCL10) and reduced Treg-attracting chemokines (CCL22, CXCL12), enhancing immune cell recruitment and survival in CRC models [120]. Similarly, VV engineered to express the chemokine CCL19 promoted the infiltration of dendritic cells and CD4+ T cells while maintaining effective viral oncolysis [121].
Chen et al. (2019) [122] developed a VV encoding interleukin-23 (IL-23), which enhanced immune cell infiltration, promoted Th1 chemokine production, and transformed immunologically “cold” tumors into “hot” tumors. These effects were mediated through IL-10 upregulation and required CD4+ and CD8+ T cells and IFN-γ for therapeutic efficacy. In CRC models, IL-9 expression increased viral persistence, modulated the TME by reducing MDSCs, and increased the infiltration of both CD4+ and CD8+ T cells. Combined with anti–CTLA–4 therapy, this strategy significantly improved anti-tumor immunity and survival [123,124]. Combinatorial therapies involving VV and immune checkpoint blockade have also shown promise. Co-administration of VV with anti–PD-L1 therapy led to stronger CD8+ T cell activation, reduced T cell exhaustion, and improved immune responses. This combination reduced metastases and prolonged survival in colon tumor-bearing mice compared to monotherapy [125].
Further enhancement was observed using recombinant VV expressing interleukin-15 (IL-15) and its receptor subunit IL-15Rα. This construct increased CD8+ T cell activity and cytokine release, leading to durable tumor regression. Combined with anti-PD-1 therapy, complete tumor eradication and long-term survival were achieved in all treated mice [126]. Similarly, VV expressing tethered IL-12 has been shown to promote CD8+ T cell infiltration, reduce Tregs, and transform cold tumors into hot tumors, improving survival. Combined with PD-1 blockade, it resulted in complete tumor regression in late-stage tumors [127].

3.6. Measles Virus (MV)

3.6.1. Mechanism of Action

Oncolytic MV, derived from the attenuated Edmonston B strain, has shown promise in preclinical models of CRC due to its natural tumor tropism and immunogenicity [128]. Recent genetic modifications have further enhanced its tumor selectivity, immune-stimulating capacity, and safety profile, making MV an attractive candidate for virotherapy in CRC. MV selectively infects cells overexpressing CD133 or uPAR and induces strong immune responses, especially when armed with IL-12 [129].

3.6.2. Clinical and Preclinical Evidence

Wang et al. (2021) explored the anti-tumor efficacy of an oncolytic measles virus encoding interleukin-12 (MeVac FmIL-12) in colon cancer [130]. In both in vivo and ex vivo models, MeVac FmIL-12 significantly enhanced anti-tumor immunity by upregulating proinflammatory cytokines, promoting apoptosis, and reducing cancer cell viability. This immune activation translated into systemic tumor rejection and prolonged survival in a rat model, demonstrating the potential of IL-12-armed MV as an immunovirotherapy platform for CRC [130].
Jing et al. (2009) [131] developed a retargeted MV that explicitly recognizes the urokinase-type plasminogen activator receptor (uPAR), a surface protein commonly overexpressed in solid tumors, including colon cancer. The modified virus efficiently infected and lysed uPAR-expressing tumor cells, inhibited tumor progression, and extended survival in preclinical models. Systemic administration facilitated targeted tumor tissue and vasculature infection, supporting its use as a tumor-specific oncolytic vector [131]. Further safety and efficacy evaluations of this uPAR-targeted measles virus (MV-m-uPA) were conducted by Jing et al. (2014) in syngeneic colon and mammary tumor models [132]. MV-m-uPA selectively infected uPAR-overexpressing CT-26 colon cancer cells, induced apoptosis, and suppressed tumor growth while prolonging survival. Notably, the virus exhibited preferential tumor accumulation without causing systemic toxicity, reinforcing its safety for future translational applications in CRC [132].
In another approach targeting tumor-initiating cells (TICs), Bach et al. (2013) [133] engineered CD133-specific oncolytic measles viruses to eliminate this stem-like subpopulation in solid tumors, including CRC. These CD133-targeted viruses exhibited enhanced infection and cytolytic activity against CD133+ tumor cells, outperforming non-targeted MV strains. Table 1 summarizes the clinical trials conducted with each oncolytic virus in CRC. Table 2 is a comparative table of the OVs used in CRC, considering the genome, tumor selectivity, immunostimulatory potential, clinical safety, clinical maturity, key advantages, and critical limitations.

4. Selection Criteria for CRC Patients for Trials with OVs

In clinical trials evaluating oncolytic adenovirus, participant selection exhibits similarities across the different studies. First, most trials include patients with a good performance status, as these individuals are more likely to tolerate immunotherapy interventions [134,135,136,137,138,139,140,141,142,143]. Additionally, eligibility generally requires adequate hematologic, hepatic, and renal function to minimize the potential risk of treatment-related toxicity. Conversely, patients who are commonly excluded are immunocompromised patients or those with active autoimmune disease, and pregnant or breastfeeding women. Secondly, some differences observed in the studies are related to disease status. While some studies enroll patients with metastatic or active tumors, others are restricted to individuals with resected tumors or patients with no evidence of disease after resection (NED). Interestingly, some trials exclude patients harboring molecular biomarkers such as MMR-D/MSI-H, who would respond better to standard immunotherapy [137]. In contrast, some studies include tumors with antigen overexpression, such as CEA, MUC1, and GCC, which represent potential vaccine immune target proteins [138,139,140,141]. In terms of the population characteristics, the Ad5-hGCC trial includes Caucasian and African American colon cancer patients in stages I and II as participants, unlike the rest of the trials that do not specify ethnic origin [140]. Furthermore, studies such as the IL-2 trial incorporate tumor burden as part of their criteria; however, other metastatic CRC trials do not include tumor volume as a selection factor [139].
In the context of oncolytic herpes virotherapy, participation criteria share several similarities with those observed in adenovirus-based therapy. However, unlike adenovirus trials, most herpes-based studies are focused on advanced, metastatic, and refractory tumors [144,145,146,147,148]. Regarding the tumor burden, both the RP2/RP3 and ONCR-177 trials require the presence of hepatic metastasis and at least one injectable tumor(s) ≥1 cm [144,145,146,147,148]. The OH2 trial, in contrast, includes stable post-chemotherapy and accessible lesions [147]; on the other hand, the NV1020 trial excludes patients with extrahepatic disease [146]. In terms of molecular biomarkers, RP2/RP3 is the only trial among those reviewed that excludes MSI-H and BRAF V600E mutations as part of the selection criteria [144]. Furthermore, most trials require at least four weeks to have elapsed since the last immune and chemotherapy treatment as a restriction on previous therapies [144,145,146,147,148]. Finally, both NV1020 and ONCR-177 protocols prohibit the concomitant use of antiviral therapy against HSV, due to the potential interference with virus replication [146,148].
Regarding vaccinia virotherapy, the clinical status of cancer at the time of participant selection varies across trials. Most enrolled patients exhibited tumoral progression despite multiple lines of treatment, typically characterized by disseminated and unresectable disease [149,150,151,152,153,154,155,156,157]. In contrast, studies such as CV301 + nivolumab or vaccinia-CEA-TRICOM + docetaxel include patients with resectable metastases, who are generally treated with curative intention and have no evident disease [150,156]. Another difference between trials is the selection of specific biomarkers. For instance, the PEXA-VEC + ICI (JX-594) trial actively includes MSS tumors (and excludes those with MSI-H) because these tumors are unlikely to respond to standard ICI therapy [155]. Similarly, the p53MVA and p53MVA-plus-pembrolizumab trials include p53-overexpressing cancers as part of the target protein of the viral vector [151,152]. Additionally, vaccinia-CEA-TRICOM + docetaxel and vaccinia-CEA-MUC-1-TRICOM incorporate the biomarker CEA, a tumor-associated receptor recognized by TRICOM-based vaccines [156,157]. Notably, the vaccinia-CEA-TRICOM + docetaxel trial specifies that at least 6 of 10 patients in each treatment arm must be HLA-A2 positive [156]. Lastly, one of the JX-594 trials uses KRAS/EGFR status to determine eligibility after anti-EGFR treatment failure (if KRAS WT) [154].

5. Limitations, Challenges, and Future Perspectives

5.1. Mechanisms of Resistance

Despite substantial advances in oncolytic virotherapy, multiple resistance mechanisms that limit viral replication, tumor cell killing, and systemic immune activation in CRC models have been identified. One such mechanism involves host-derived antimicrobial peptides. Human α-defensin 5 (HD5), secreted by epithelial cells, has been shown to interfere with oncolytic adenovirus therapy by binding to viral capsid proteins, thereby blocking endosomal escape and viral replication [158,159,160]. Interestingly, human adenovirus serotype 3 (HAdV3) can partially evade this inhibition by producing penton-dodecahedral (PtDd) particles that neutralize HD5, allowing for continued viral dissemination [160]. These findings suggest that tumor-expressed HD5 represents a significant barrier to adenovirus-based therapies, necessitating the design of viral vectors capable of overcoming such innate defenses.
Interactions between chemotherapeutics and virotherapy can also compromise viral replication. Kulu et al. (2013) [161] demonstrated that 5-FU and irinotecan (CPT-11) inhibit HSV-1 replication in colon cancer cells by disrupting NF-κB signaling and phosphorylating eIF-2α. Since NF-κB activation is essential for HSV-1 propagation, chemotherapy-induced signaling events may create a cellular environment that is unfavorable for viral replication, highlighting the importance of optimizing therapeutic combinations [161].
Resistance to oHSV-1 therapy has also been linked to gene expression changes that impair viral entry, replication, and induction of apoptosis. Studies using HSV-1-resistant CRC cell lines revealed alterations in surface receptors, metabolic pathways, and immune signaling components contributing to therapy failure [162].
Reovirus therapy, similarly, faces limitations. Vanhoudt et al. (2008) [163] reported that intact reovirus T3D virions failed to infect freshly isolated human colorectal tumor cells, regardless of KRAS mutation status. Only intermediate subviral particles achieved transient infection, producing viral proteins without generating infectious progeny or inducing cytolysis. This resistance was attributed to the abnormal localization of the junctional adhesion molecule 1 (JAM1), the primary reovirus receptor, and cellular responses that abort viral replication [163].
Resistance to VSV is frequently driven by the chronic activation of innate immune pathways. Transcriptomic profiling identified the elevated expression of interferon-stimulated genes (ISGs), including Mx2, Oasl, and Irf7, which conferred cross-resistance to both VSV and Sindbis viruses [164]. These resistance signatures were conserved across diverse tumor cell lines, indicating a reproducible antiviral state. Additional studies in CT26 colon carcinoma cells revealed heterogeneous resistance mechanisms, including one clone characterized by immune activation and ISG expression and another defined by cytoskeletal remodeling and altered intracellular signaling [99]. Together, these outcomes underline the need for strategies to counteract innate immune activation to enhance the efficacy of oncolytic virotherapy.
The inhibition of viral replication also represents a critical challenge. The c-Jun N-terminal kinase (JNK) pathway, known to regulate stress responses and apoptosis, can limit viral propagation. JNK-deficient cells showed a 100-fold increase in VV replication and virus-induced apoptosis, suggesting that JNK acts as a viral restriction factor, likely by activating the double-stranded RNA-dependent PKR pathway [165,166,167,168,169]. Therefore, modulating stress-related kinases may improve the replication and efficacy of oncolytic viruses.

5.2. Personalized Strategies and Combined Therapies

Numerous innovative strategies are being developed to overcome the limitations of single-agent virotherapy, including personalized vaccines, combinatorial virus designs, and the modulation of host immune responses.
Feola et al. (2022) [170] developed a personalized immunopeptidomics-based approach to generate oncolytic vaccines tailored to colorectal tumors. Their platform, PeptiCRAd, integrates oncolytic adenoviruses with tumor-specific MHC-I-restricted peptides, enhancing antigen-specific immune responses and tumor suppression in preclinical models [170]. The same group created PeptiCab, a novel adenovirus-based cancer vaccine targeting Fcγ and Fcα receptors to amplify adaptive immunity. This construct incorporates tumor antigens and a PD-L1 inhibitor, promoting neutrophil polarization toward an antigen-presenting phenotype and boosting T-cell responses [171]. A second approach involves enhancing the fusogenic potential of oncolytic viruses. VSV-p14, a recombinant VSV expressing the reovirus-derived fusion-associated small transmembrane (FAST) protein p14, demonstrated improved tumor cell fusion, viral spread, and immune activation. In CT26 tumor models, VSV-p14 significantly increased CD4+, CD8+, NK, and NKT cell infiltration, resulting in superior tumor control and prolonged survival [172].
Another strategy is the inactivation of key tumor resistance factors. As mentioned above, VSV is a promising oncolytic therapy for colon cancer, yet some tumors develop resistance [99,164]. Because of this, a study has identified Cancer Upregulated Gene 2 (CUG2) as a factor whereby tumor cells resist VSV infection through STAT1 activation and OASL2 upregulation, enhancing antiviral defenses. Suppressing STAT1 or OASL2 restored VSV susceptibility, increasing viral replication and apoptosis in resistant tumor cells. These results suggest that targeting CUG2-mediated pathways could improve VSV-based virotherapy’s efficacy in resistant CRC [173].
Another promising strategy involves engineering viruses with enhanced fusogenicity. Fusogenic vaccinia virus (FUVAC), which carries a mutation in the K2L gene to promote syncytia formation, demonstrated superior tumor cell killing, immunogenic cell death, and CD8+ T cell recruitment. In bilateral colon tumor models, FUVAC induced the regression of both treated and distant tumors. Combined with PD-1 blockade, FUVAC achieved complete tumor regression and long-term immune memory [174,175,176].
Additionally, inducing ferroptosis—a form of iron-dependent cell death—may potentiate oncolytic virotherapy. Co-treatment with elastin and VV led to reduced tumor growth, extended survival, and enhanced immune memory. Mechanistically, elastin promoted dendritic cell maturation and CD8+ T cell activation, increasing IFN-γ+ and PD-1+ T cells within the TME [177]. These results suggest that ferroptosis induction can synergize with VV to augment anti-tumor immunity in CRC.

5.3. Virotherapy’s Clinical Limitations in Colon Cancers

Despite the significant advances in virotherapy in CRC, several limitations are observed when put into clinical practice. One limitation in the clinical application is the acquisition of viral resistance. Cancer proteins responsible for suppressing cytokine signaling and members of the integrin class of cell surface receptors may develop resistance that reduces viral attachment and increases acute-phase responses against viral infection [162]. In the same way, antiviral antibodies are more likely to be found after repeated oncolytic measles virus exposure, suggesting a neutralizing activity under long-term treatments [132,178]. Physical barriers caused by cytoskeletal reorganization and poor vascular perfusion may impede viral diffusion to cancer cells and reduce therapeutic efficacy [99].
Another concern in the application of virotherapy is the risk of toxicity in healthy tissues. One example of this is the administration of vaccinia virus, which has shown no dose-limiting toxicities. However, researchers express doubts regarding patients without prior vaccinia vaccination and immunosuppressed patients, who were excluded from their studies [179]. Intriguingly, the persistent stimulation of the T cell immune response by viral therapy may turn into a dysfunctional or “exhausted” state, which alters treatment outcomes; however, combined strategies could synergistically enhance the anti-tumor adaptive immune response and reverse T cell exhaustion within the TME [84].
The systemic administration of viral therapy has also shown limitations, such as healthcare problems and immune sensitivity, resulting in undesirable side effects [180,181]. Therefore, researchers have developed local approaches, such as nanomaterials, to deliver viruses to the site of interest [182]. The intravenous infusion of virotherapy is a safe treatment delivery modality in metastatic CRC; however, this may also be challenging because it results in a less effective response than locoregional delivery due to systemic viral clearance [178].
Lastly, the use of virotherapy as monotherapy in colon cancer has shown limited clinical results due to the production of immunosuppressive factors for tumor protection. Thus, most current efforts focus on combining different strategies to enhance therapy efficacy while avoiding cell toxicity [183]. For instance, oncolytic reovirus that sensitizes microsatellite stable (MSS) CRC to immune checkpoint inhibition, such as anti-PD-1 treatment, increases cell death among MSS cells in mouse models. Although these findings shed light on promising combined immunological mechanisms in animal models, the authors also mention that translating preclinical findings to the clinic remains a significant challenge [84].

6. Conclusions

OVs represent a promising therapeutic approach for CRC that takes advantage of genetically modified viruses’ ability to selectively target and lyse tumor cells while stimulating anti-tumor immune responses. The efficacy of virotherapy is based on multiple mechanisms, including direct oncolysis, the modulation of the TME, and the enhancement of systemic immune responses. Despite these advances, several challenges hinder the widespread clinical application of OVs in CRC.
One major limitation is the development of resistance mechanisms that impair viral infection, replication, and immune activation. Tumor cells can evade virotherapy by secreting immunosuppressive factors, modifying cell surface receptors to prevent viral entry, and activating antiviral signaling pathways, including interferon-stimulated genes. Additionally, the TME can promote resistance by increasing regulatory T cells, myeloid-derived suppressor cells, and hypoxia, inhibiting the efficacy of OVs.
To overcome these challenges, novel strategies are being explored, including the genetic engineering of OVs to enhance tumor selectivity and immune stimulation. Combining virotherapy with immune checkpoint inhibitors, CAR-T cell therapy, and personalized cancer vaccines has significant potential to improve treatment outcomes. Additionally, fusogenic viruses, the modulation of tumor metabolic pathways, and the inhibition of antiviral defenses are emerging approaches for enhancing OV efficacy.
Future research should optimize OV delivery, reduce resistance mechanisms, and improve patient stratification to maximize therapeutic benefits. By addressing these limitations, oncolytic virotherapy could become a key component of multimodal treatment strategies for CRC, providing a more effective and personalized approach to cancer therapy.

Author Contributions

Conceptualization, D.C.-B., F.P.-D., C.Q.-M. and J.P.M.; resources, D.C.-B. and J.P.M.; writing—review and editing, D.C.-B., F.P.-D., L.C. and J.P.M.; visualization, D.C.-B., F.P.-D., L.C. and J.P.M.; supervision, D.C.-B. and J.P.M.; funding acquisition, D.C.-B. and J.P.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Proyecto Fondecyt de iniciación en investigación 2025 No. 11250378 ANID (D.C.-B.). Millennium Science Initiative Program—ICN09_016/ICN 2021_045 (D.C.-B.) and by the Universidad de Tarapacá under the Programa Fortalecimiento Grupos de Investigación UTA 4769-24 (J.P.M).

Acknowledgments

The authors would like to thank Diego Soto-Jiménez from Universidad de Tarapacá for his valuable assistance in managing the references while preparing this manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Glossary

OVOncolytic Virus
CRCColorectal Cancer
HSVHerpes Simplex Virus
VSVVesicular Stomatitis Virus
VVVaccinia Virus
MVMeasles Virus
TMETumor Microenvironment
ILInterleukin
GM-CSFGranulocyte-Macrophage Colony-Stimulating Factor
TRAILTNF-Related Apoptosis-Inducing Ligand
aPD-1Anti-Programmed Death-1
CXCL11C-X-C Motif Chemokine Ligand 11
TLR3Toll-Like Receptor 3
NKNatural Killer (cell)
MDSCMyeloid-Derived Suppressor Cell
TregRegulatory T Cell
ICIsImmune Checkpoint Inhibitors
HDACHistone Deacetylase
CSCCancer Stem Cell
CAR-TChimeric Antigen Receptor T Cell

References

  1. Siegel, R.L.; Kratzer, T.B.; Giaquinto, A.N.; Sung, H.; Jemal, A. Cancer statistics, 2025. CA Cancer J. Clin. 2025, 75, 10–45. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Wagle, N.S.; Cercek, A.; Smith, R.A.; Jemal, A. Colorectal cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 233–254. [Google Scholar] [CrossRef] [PubMed]
  3. Sawicki, T.; Ruszkowska, M.; Danielewicz, A.; Niedźwiedzka, E.; Arłukowicz, T.; Przybyłowicz, K.E. A review of colorectal cancer in terms of epidemiology, risk factors, development, symptoms and diagnosis. Cancers 2021, 13, 2025. [Google Scholar] [CrossRef] [PubMed]
  4. Benson, A.B.; Venook, A.P.; Adam, M.; Chang, G.; Chen, Y.J.; Ciombor, K.K.; Cohen, S.A.; Cooper, H.S.; Deming, D.; Garrido-Laguna, I.; et al. Colon Cancer, Version 3.2024, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Canc Netw. 2024, 22, e240029. [Google Scholar] [CrossRef]
  5. Chen, W.; Shi, K.; Yu, Y.; Yang, P.; Bei, Z.; Mo, D.; Yuan, L.; Pan, M.; Chen, Y.; Qian, Z. Drug delivery systems for colorectal cancer chemotherapy. Chin. Chem. Lett. 2024, 35, 109159. [Google Scholar] [CrossRef]
  6. Kabbinavar, F.; Hurwitz, H.I.; Fehrenbacher, L.; Meropol, N.J.; Novotny, W.F.; Lieberman, G.; Griffing, S.; Bergsland, E. Phase II, randomized trial comparing bevacizumab plus fluorouracil (FU)/leucovorin (LV) with FU/LV alone in patients with metastatic colorectal cancer. J. Clin. Oncol. 2003, 21, 60–65. [Google Scholar] [CrossRef]
  7. Hurwitz, H.; Fehrenbacher, L.; Novotny, W.; Cartwright, T.; Hainsworth, J.; Heim, W.; Berlin, J.; Baron, A.; Griffing, S.; Holmgren, E.; et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N. Engl. J. Med. 2004, 350, 2335–2342. [Google Scholar] [CrossRef]
  8. Pietrantonio, F.; Cremolini, C.; Petrelli, F.; Di Bartolomeo, M.; Loupakis, F.; Maggi, C.; Antoniotti, C.; de Braud, F.; Falcone, A.; Iacovelli, R. First-line anti-EGFR monoclonal antibodies in panRAS wild-type metastatic colorectal cancer: A systematic review and meta-analysis. Crit. Rev. Oncol. Hematol. 2015, 96, 156–166. [Google Scholar] [CrossRef]
  9. Sorich, M.J.; Wiese, M.D.; Rowland, A.; Kichenadasse, G.; McKinnon, R.A.; Karapetis, C.S. Extended RAS mutations and anti-EGFR monoclonal antibody survival benefit in metastatic colorectal cancer: A meta-analysis of randomized, controlled trials. Ann. Oncol. 2015, 26, 13–21. [Google Scholar] [CrossRef]
  10. Al Bitar, S.; El-Sabban, M.; Doughan, S.; Abou-Kheir, W. Molecular mechanisms targeting drug-resistance and metastasis in colorectal cancer: Updates and beyond. World J. Gastroenterol. 2023, 29, 1395–1426. [Google Scholar] [CrossRef]
  11. Cherri, S.; Oneda, E.; Zanotti, L.; Zaniboni, A. Optimizing the first-line treatment for metastatic colorectal cancer. Front. Oncol. 2023, 13, 1246716. [Google Scholar] [CrossRef] [PubMed]
  12. Yan, Z.; Zhang, Z.; Chen, Y.; Xu, J.; Wang, J.; Wang, Z. Enhancing cancer therapy: The integration of oncolytic virus therapy with diverse treatments. Cancer Cell Int. 2024, 24, 242. [Google Scholar] [CrossRef] [PubMed]
  13. Ma, R.; Li, Z.; Chiocca, E.A.; Caligiuri, M.A.; Yu, J. The emerging field of oncolytic virus-based cancer immunotherapy. Trends Cancer 2023, 9, 122–139. [Google Scholar] [CrossRef]
  14. Ren, Y.; Miao, J.M.; Wang, Y.Y.; Fan, Z.; Kong, X.B.; Yang, L.; Cheng, G. Oncolytic viruses combined with immune checkpoint therapy for colorectal cancer is a promising treatment option. Front. Immunol. 2022, 13, 961796. [Google Scholar] [CrossRef]
  15. 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]
  16. Maroun, J.; Miguel, M.-A.; Arun, A.; Autumn, S.; Kah-Whye, P.; Russell, S. Designing and Building Oncolytic Viruses. Future Virol. 2017, 12, 193–213. [Google Scholar] [CrossRef]
  17. Parato, K.A.; Senger, D.; Forsyth, P.A.; Bell, J.C. Recent progress in the battle between oncolytic viruses and tumours. Nat. Rev. Cancer 2005, 5, 965–976. [Google Scholar] [CrossRef]
  18. Moore, A.E. Effect of inoculation of the viruses of influenza A and herpes simplex on the growth of transplantable tumors in mice. Cancer 1949, 2, 516–524. [Google Scholar] [CrossRef]
  19. Moore, A.E. The destructive effect of the virus of Russian Far East encephalitis on the transplantable mouse sarcoma 180. Cancer 1949, 2, 525–534. [Google Scholar] [CrossRef]
  20. 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]
  21. 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] [PubMed]
  22. Zamarin, D.; Holmgaard, R.B.; Subudhi, S.K.; Park, J.S.; Mansour, M.; Palese, P.; Merghoub, T.; Wolchok, J.D.; Allison, J.P. Localized oncolytic virotherapy overcomes systemic tumor resistance to immune checkpoint blockade immunotherapy. Sci. Transl. Med. 2014, 6, 226ra232. [Google Scholar] [CrossRef] [PubMed]
  23. Di Paolo, N.C.; Miao, E.A.; Iwakura, Y.; Murali-Krishna, K.; Aderem, A.; Flavell, R.A.; Papayannopoulou, T.; Shayakhmetov, D.M. Virus binding to a plasma membrane receptor triggers interleukin-1 alpha-mediated proinflammatory macrophage response in vivo. Immunity 2009, 31, 110–121. [Google Scholar] [CrossRef] [PubMed]
  24. Liu, B.L.; Robinson, M.; Han, Z.Q.; Branston, R.H.; English, C.; Reay, P.; McGrath, Y.; Thomas, S.K.; Thornton, M.; Bullock, P.; et al. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther. 2003, 10, 292–303. [Google Scholar] [CrossRef]
  25. Rojas, J.J.; Sampath, P.; Hou, W.; Thorne, S.H. Defining Effective Combinations of Immune Checkpoint Blockade and Oncolytic Virotherapy. Clin. Cancer Res. 2015, 21, 5543–5551. [Google Scholar] [CrossRef]
  26. Ajina, A.; Maher, J. Prospects for combined use of oncolytic viruses and CAR T-cells. J. Immunother. Cancer 2017, 5, 90. [Google Scholar] [CrossRef]
  27. Chen, C.Y.; Hutzen, B.; Wedekind, M.F.; Cripe, T.P. Oncolytic virus and PD-1/PD-L1 blockade combination therapy. Oncolytic Virother. 2018, 7, 65–77. [Google Scholar] [CrossRef]
  28. Achard, C.; Surendran, A.; Wedge, M.E.; Ungerechts, G.; Bell, J.; Ilkow, C.S. Lighting a Fire in the Tumor Microenvironment Using Oncolytic Immunotherapy. EBioMedicine 2018, 31, 17–24. [Google Scholar] [CrossRef]
  29. Lan, Q.; Xia, S.; Wang, Q.; Xu, W.; Huang, H.; Jiang, S.; Lu, L. Development of oncolytic virotherapy: From genetic modification to combination therapy. Front. Med. 2020, 14, 160–184. [Google Scholar] [CrossRef]
  30. Stasiak, A.C.; Stehle, T. Human adenovirus binding to host cell receptors: A structural view. Med. Microbiol. Immunol. 2020, 209, 325–333. [Google Scholar] [CrossRef]
  31. Kulanayake, S.; Tikoo, S.K. Adenovirus Core Proteins: Structure and Function. Viruses 2021, 13, 388. [Google Scholar] [CrossRef] [PubMed]
  32. Yang, K.; Feng, S.; Luo, Z. Oncolytic Adenovirus, a New Treatment Strategy for Prostate Cancer. Biomedicines 2022, 10, 3262. [Google Scholar] [CrossRef] [PubMed]
  33. Tripodi, L.; Vitale, M.; Cerullo, V.; Pastore, L. Oncolytic adenoviruses for cancer therapy. Int. J. Mol. Sci. 2021, 22, 2517. [Google Scholar] [CrossRef]
  34. Gao, J.; Zhang, W.; Ehrhardt, A. Expanding the Spectrum of Adenoviral Vectors for Cancer Therapy. Cancers 2020, 12, 1139. [Google Scholar] [CrossRef]
  35. Fujiwara, T. Multidisciplinary oncolytic virotherapy for gastrointestinal cancer. Ann. Gastroenterol. Surg. 2019, 3, 396–404. [Google Scholar] [CrossRef]
  36. Kakiuchi, Y.; Kuroda, S.; Kanaya, N.; Kumon, K.; Tsumura, T.; Hashimoto, M.; Yagi, C.; Sugimoto, R.; Hamada, Y.; Kikuchi, S.; et al. Local oncolytic adenovirotherapy produces an abscopal effect via tumor-derived extracellular vesicles. Mol. Ther. 2021, 29, 2920–2930. [Google Scholar] [CrossRef]
  37. Yan, Y.; Li, S.; Jia, T.; Du, X.; Xu, Y.; Zhao, Y.; Li, L.; Liang, K.; Liang, W.; Sun, H.; et al. Combined therapy with CTL cells and oncolytic adenovirus expressing IL-15-induced enhanced antitumor activity. Tumour Biol. 2015, 36, 4535–4543. [Google Scholar] [CrossRef]
  38. Yan, Y.; Xu, Y.; Zhao, Y.; Li, L.; Sun, P.; Liu, H.; Fan, Q.; Liang, K.; Liang, W.; Sun, H.; et al. Combination of E2F-1 promoter-regulated oncolytic adenovirus and cytokine-induced killer cells enhances the antitumor effects in an orthotopic rectal cancer model. Tumour Biol. 2014, 35, 1113–1122. [Google Scholar] [CrossRef]
  39. Akbulut, H.; Coleri, A.; Sahin, G.; Tang, Y.; Icli, F. A bicistronic adenoviral vector carrying cytosine deaminase and granulocyte-macrophage colony-stimulating factor increases the therapeutic efficacy of cancer gene therapy. Hum. Gene Ther. 2019, 30, 999–1007. [Google Scholar] [CrossRef]
  40. Bommareddy, P.K.; Shettigar, M.; Kaufman, H.L. Integrating oncolytic viruses in combination cancer immunotherapy. Nat. Rev. Immunol. 2018, 18, 498–513. [Google Scholar] [CrossRef]
  41. Shi, G.; Yang, Q.; Zhang, Y.; Jiang, Q.; Lin, Y.; Yang, S.; Wang, H.; Cheng, L.; Zhang, X.; Li, Y.; et al. Modulating the Tumor Microenvironment via Oncolytic Viruses and CSF-1R Inhibition Synergistically Enhances Anti-PD-1 Immunotherapy. Mol. Ther. 2019, 27, 244–260. [Google Scholar] [CrossRef] [PubMed]
  42. Garofalo, M.; Iovine, B.; Kuryk, L.; Capasso, C.; Hirvinen, M.; Vitale, A.; Yliperttula, M.; Bevilacqua, M.A.; Cerullo, V. Oncolytic Adenovirus Loaded with L-carnosine as Novel Strategy to Enhance the Antitumor Activity. Mol. Cancer Ther. 2016, 15, 651–660. [Google Scholar] [CrossRef] [PubMed]
  43. Cheng, T.; Song, Y.; Zhang, Y.; Zhang, C.; Yin, J.; Chi, Y.; Zhou, D. A novel oncolytic adenovirus based on simian adenovirus serotype 24. Oncotarget 2017, 8, 26871–26885. [Google Scholar] [CrossRef]
  44. Bressy, C.; Majhen, D.; Raddi, N.; Jdey, W.; Cornilleau, G.; Zig, L.; Guirouilh-Barbat, J.; Lopez, B.S.; Bawa, O.; Opolon, P.; et al. Combined therapy of colon carcinomas with an oncolytic adenovirus and valproic acid. Oncotarget 2017, 8, 97344–97360. [Google Scholar] [CrossRef]
  45. Kurimasa, A.; Ouyang, H.; Dong, L.J.; Wang, S.; Li, X.; Cordon-Cardo, C.; Chen, D.J.; Li, G.C. Catalytic subunit of DNA-dependent protein kinase: Impact on lymphocyte development and tumorigenesis. Proc. Natl. Acad. Sci. USA 1999, 96, 1403–1408. [Google Scholar] [CrossRef]
  46. Taccioli, G.E.; Amatucci, A.G.; Beamish, H.J.; Gell, D.; Xiang, X.H.; Torres Arzayus, M.I.; Priestley, A.; Jackson, S.P.; Marshak Rothstein, A.; Jeggo, P.A.; et al. Targeted disruption of the catalytic subunit of the DNA-PK gene in mice confers severe combined immunodeficiency and radiosensitivity. Immunity 1998, 9, 355–366. [Google Scholar] [CrossRef]
  47. Thompson, L.H.; Brookman, K.W.; Jones, N.J.; Allen, S.A.; Carrano, A.V. Molecular cloning of the human XRCC1 gene, which corrects defective DNA strand break repair and sister chromatid exchange. Mol. Cell Biol. 1990, 10, 6160–6171. [Google Scholar]
  48. Kon, T.; Zhang, X.; Huang, Q.; Yang, Z.; Liu, S.; Yan, B.; Li, F.; Wang, H.; Li, C.Y. Oncolytic virus-mediated tumor radiosensitization in mice through DNA-PKcs-specific shRNA. Transl. Cancer Res. 2012, 1, 4–14. [Google Scholar]
  49. Kaneda, A.; Feinberg, A.P. Loss of imprinting of IGF2: A common epigenetic modifier of intestinal tumor risk. Cancer Res. 2005, 65, 11236–11240. [Google Scholar] [CrossRef]
  50. Sun, H.; Pan, Y.; He, B.; Deng, Q.; Li, R.; Xu, Y.; Chen, J.; Gao, T.; Ying, H.; Wang, F.; et al. Gene therapy for human colorectal cancer cell lines with recombinant adenovirus 5 based on loss of the insulin-like growth factor 2 imprinting. Int. J. Oncol. 2015, 46, 1759–1767. [Google Scholar] [CrossRef]
  51. Zhou, W.; Zhu, H.; Chen, W.; Hu, X.; Pang, X.; Zhang, J.; Huang, X.; Fang, B.; He, C. Treatment of patient tumor-derived colon cancer xenografts by a TRAIL gene-armed oncolytic adenovirus. Cancer Gene Ther. 2011, 18, 336–345. [Google Scholar] [CrossRef] [PubMed]
  52. Kanaya, N.; Kuroda, S.; Kakiuchi, Y.; Kumon, K.; Tsumura, T.; Hashimoto, M.; Morihiro, T.; Kubota, T.; Aoyama, K.; Kikuchi, S.; et al. Immune Modulation by Telomerase-Specific Oncolytic Adenovirus Synergistically Enhances Antitumor Efficacy with Anti-PD1 Antibody. Mol. Ther. 2020, 28, 794–804. [Google Scholar] [CrossRef] [PubMed]
  53. Ferguson, P.J.; Sykelyk, A.; Figueredo, R.; Koropatnick, J. Synergistic cytotoxicity against human tumor cell lines by oncolytic adenovirus dl1520 (ONYX-015) and melphalan. Tumori 2016, 102, 31–39. [Google Scholar] [CrossRef]
  54. Bischoff, J.R.; Kirn, D.H.; Williams, A.; Heise, C.; Horn, S.; Muna, M.; Ng, L.; Nye, J.A.; Sampson-Johannes, A.; Fattaey, A.; et al. An adenovirus mutant that replicates selectively in p53-deficient human tumor cells. Science 1996, 274, 373–376. [Google Scholar] [CrossRef]
  55. Shen, Y.; Nemunaitis, J. Herpes simplex virus 1 (HSV-1) for cancer treatment. Cancer Gene Ther. 2006, 13, 975–992. [Google Scholar] [CrossRef]
  56. Yin, J.; Markert, J.M.; Leavenworth, J.W. Modulation of the Intratumoral Immune Landscape by Oncolytic Herpes Simplex Virus Virotherapy. Front. Oncol. 2017, 7, 136. [Google Scholar] [CrossRef]
  57. Aldrak, N.; Alsaab, S.; Algethami, A.; Bhere, D.; Wakimoto, H.; Shah, K.; Alomary, M.N.; Zaidan, N. Oncolytic herpes simplex virus-based therapies for cancer. Cells 2021, 10, 1541. [Google Scholar] [CrossRef]
  58. Ghouse, S.M.; Nguyen, H.-M.; Bommareddy, P.K.; Guz-Montgomery, K.; Saha, D. Oncolytic herpes simplex virus encoding IL12 controls triple-negative breast cancer growth and metastasis. Front. Oncol. 2020, 10, 384. [Google Scholar] [CrossRef]
  59. Bennett, J.J.; Delman, K.A.; Burt, B.M.; Mariotti, A.; Malhotra, S.; Zager, J.; Petrowsky, H.; Mastorides, S.; Federoff, H.; Fong, Y. Comparison of safety, delivery, and efficacy of two oncolytic herpes viruses (G207 and NV1020) for peritoneal cancer. Cancer Gene Ther. 2002, 9, 935–945. [Google Scholar] [CrossRef]
  60. McAuliffe, P.F.; Jarnagin, W.R.; Johnson, P.; Delman, K.A.; Federoff, H.; Fong, Y. Effective treatment of pancreatic tumors with two multimutated herpes simplex oncolytic viruses. J. Gastrointest. Surg. 2000, 4, 580–588. [Google Scholar] [CrossRef]
  61. Gutermann, A.; Mayer, E.; Dehn-Rothfelser, K.V.; Breidenstein, C.; Weber, M.; Muench, M.; Gungor, D.; Suehnel, J.; Moebius, U.; Lechmann, M. Efficacy of oncolytic herpesvirus NV1020 can be enhanced by combination with chemotherapeutics in colon carcinoma cells. Hum. Gene Ther. 2006, 17, 1241–1253. [Google Scholar] [CrossRef] [PubMed]
  62. Kooby, D.A.; Carew, J.F.; Halterman, M.W.; Mack, J.E.; Bertino, J.R.; Blumgart, L.H.; Federoff, H.J.; Fong, Y. Oncolytic viral therapy for human colorectal cancer and liver metastases using a multi-mutated herpes simplex virus type-1 (G207). FASEB J. 1999, 13, 1325–1334. [Google Scholar] [CrossRef] [PubMed]
  63. Warner, S.G.; Haddad, D.; Au, J.; Carson, J.S.; O’Leary, M.P.; Lewis, C.; Monette, S.; Fong, Y. Oncolytic herpes simplex virus kills stem-like tumor-initiating colon cancer cells. Mol. Ther. -Oncolytics 2016, 3, 16013. [Google Scholar] [CrossRef]
  64. Terai, K.; Bi, D.; Liu, Z.; Kimura, K.; Sanaat, Z.; Dolatkhah, R.; Soleimani, M.; Jones, C.; Bright, A.; Esfandyari, T. A novel oncolytic herpes capable of cell-specific transcriptional targeting of CD133±cancer cells induces significant tumor regression. Stem Cells 2018, 36, 1154–1169. [Google Scholar] [CrossRef]
  65. Yang, H.; Peng, T.; Li, J.; Wang, Y.; Zhang, W.; Zhang, P.; Peng, S.; Du, T.; Li, Y.; Yan, Q. Treatment of colon cancer with oncolytic herpes simplex virus in preclinical models. Gene Ther. 2016, 23, 450–459. [Google Scholar] [CrossRef]
  66. Zhao, Q.; Zhang, W.; Ning, Z.; Zhuang, X.; Lu, H.; Liang, J.; Li, J.; Zhang, Y.; Dong, Y.; Zhang, Y.; et al. A novel oncolytic herpes simplex virus type 2 has potent anti-tumor activity. PLoS ONE 2014, 9, e93103. [Google Scholar] [CrossRef]
  67. Cheng, K.J.; Alshawsh, M.A.; Mejia Mohamed, E.H.; Thavagnanam, S.; Sinniah, A.; Ibrahim, Z.A. HMGB1: An overview of its versatile roles in the pathogenesis of colorectal cancer. Cell Oncol. 2020, 43, 177–193. [Google Scholar] [CrossRef]
  68. Rapoport, B.L.; Steel, H.C.; Theron, A.J.; Heyman, L.; Smit, T.; Ramdas, Y.; Anderson, R. High Mobility Group Box 1 in Human Cancer. Cells 2020, 9, 1664. [Google Scholar] [CrossRef]
  69. Cerwenka, A.; Kopitz, J.; Schirmacher, P.; Roth, W.; Gdynia, G. HMGB1: The metabolic weapon in the arsenal of NK cells. Mol. Cell. Oncol. 2016, 3, e1175538. [Google Scholar] [CrossRef]
  70. Gdynia, G.; Sauer, S.W.; Kopitz, J.; Fuchs, D.; Duglova, K.; Ruppert, T.; Miller, M.; Pahl, J.; Cerwenka, A.; Enders, M.; et al. The HMGB1 protein induces a metabolic type of tumour cell death by blocking aerobic respiration. Nat. Commun. 2016, 7, 10764. [Google Scholar] [CrossRef]
  71. Shayan, S.; Arashkia, A.; Bahramali, G.; Abdoli, A.; Nosrati, M.S.S.; Azadmanesh, K. Cell type-specific response of colon cancer tumor cell lines to oncolytic HSV-1 virotherapy in hypoxia. Cancer Cell Int. 2022, 22, 164. [Google Scholar] [CrossRef]
  72. Zhang, W.; Hu, X.; Liang, J.; Zhu, Y.; Zeng, B.; Feng, L.; Zhao, C.; Liu, S.; Liu, B.; Zhang, K. oHSV2 can target murine colon carcinoma by altering the immune status of the tumor microenvironment and inducing antitumor immunity. Mol. Ther. Oncolytics 2020, 16, 158–171. [Google Scholar] [CrossRef] [PubMed]
  73. Goins, W.F.; Hall, B.; Cohen, J.B.; Glorioso, J.C. Retargeting of herpes simplex virus (HSV) vectors. Curr. Opin. Virol. 2016, 21, 93–101. [Google Scholar] [CrossRef] [PubMed]
  74. 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.e255. [Google Scholar] [CrossRef]
  75. Hu, Z.; Li, Y.; Yang, J.; Liu, J.; Zhou, H.; Sun, C.; Tian, C.; Zhu, C.; Shao, M.; Wang, S. Improved antitumor effectiveness of oncolytic HSV-1 viruses engineered with IL-15/IL-15Rα complex combined with oncolytic HSV-1-aPD1 targets colon cancer. Sci. Rep. 2024, 14, 23671. [Google Scholar] [CrossRef]
  76. Kolodkin-Gal, D.; Edden, Y.; Hartshtark, Z.; Ilan, L.; Khalaileh, A.; Pikarsky, A.; Pikarsky, E.; Rabkin, S.; Panet, A.; Zamir, G. Herpes simplex virus delivery to orthotopic rectal carcinoma results in an efficient and selective antitumor effect. Gene Ther. 2009, 16, 905–915. [Google Scholar] [CrossRef]
  77. Zhang, N.; Li, J.; Yu, J.; Wan, Y.; Zhang, C.; Zhang, H.; Cao, Y. Construction of an IL12 and CXCL11 armed oncolytic herpes simplex virus using the CRISPR/Cas9 system for colon cancer treatment. Virus Res. 2023, 323, 198979. [Google Scholar] [CrossRef]
  78. DeAntoneo, C.; Danthi, P.; Balachandran, S. Reovirus Activated Cell Death Pathways. Cells 2022, 11, 1757. [Google Scholar] [CrossRef]
  79. Kemp, V.; Hoeben, R.C.; Van den Wollenberg, D.J. Exploring reovirus plasticity for improving its use as oncolytic virus. Viruses 2015, 8, 4. [Google Scholar] [CrossRef]
  80. 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]
  81. Shakoori, A.; Ougolkov, A.; Yu, Z.W.; Zhang, B.; Modarressi, M.H.; Billadeau, D.D.; Mai, M.; Takahashi, Y.; Minamoto, T. Deregulated GSK3beta activity in colorectal cancer: Its association with tumor cell survival and proliferation. Biochem. Biophys. Res. Commun. 2005, 334, 1365–1373. [Google Scholar] [CrossRef] [PubMed]
  82. Min, H.J.; Koh, S.S.; Cho, I.R.; Srisuttee, R.; Park, E.H.; Jhun, B.H.; Kim, Y.G.; Oh, S.; Kwak, J.E.; Johnston, R.N.; et al. Inhibition of GSK-3beta enhances reovirus-induced apoptosis in colon cancer cells. Int. J. Oncol. 2009, 35, 617–624. [Google Scholar] [PubMed]
  83. Negri, F.; Bottarelli, L.; de’Angelis, G.L.; Gnetti, L. KRAS: A druggable target in colon cancer patients. Int. J. Mol. Sci. 2022, 23, 4120. [Google Scholar] [CrossRef]
  84. Augustine, T.; John, P.; Friedman, T.; Jiffry, J.; Guzik, H.; Mannan, R.; Gupta, R.; Delano, C.; Mariadason, J.M.; Zang, X.; et al. Potentiating effect of reovirus on immune checkpoint inhibition in microsatellite stable colorectal cancer. Front. Oncol. 2022, 12, 1018767. [Google Scholar] [CrossRef]
  85. Müller, L.M.E.; Migneco, G.; Scott, G.B.; Down, J.; King, S.; Askar, B.; Jennings, V.; Oyajobi, B.; Scott, K.; West, E.; et al. Reovirus-induced cell-mediated immunity for the treatment of multiple myeloma within the resistant bone marrow niche. J. Immunother. Cancer 2021, 9, e001803. [Google Scholar] [CrossRef]
  86. Zhang, H.; Ren, Y.; Wang, F.; Tu, X.; Tong, Z.; Liu, L.; Zheng, Y.; Zhao, P.; Cheng, J.; Li, J. The long-term effectiveness and mechanism of oncolytic virotherapy combined with anti-PD-L1 antibody in colorectal cancer patient. Cancer Gene Ther. 2024, 31, 1412–1426. [Google Scholar] [CrossRef]
  87. Angioni, R.; Sánchez-Rodríguez, R.; Viola, A.; Molon, B. TGF-β in Cancer: Metabolic Driver of the Tolerogenic Crosstalk in the Tumor Microenvironment. Cancers 2021, 13, 401. [Google Scholar] [CrossRef]
  88. Groeneveldt, C.; van Ginkel, J.Q.; Kinderman, P.; Sluijter, M.; Griffioen, L.; Labrie, C.; van den Wollenberg, D.J.; Hoeben, R.C.; van der Burg, S.H.; Ten Dijke, P. Intertumoral differences dictate the outcome of TGF-β blockade on the efficacy of viro-immunotherapy. Cancer Res. Commun. 2023, 3, 325–337. [Google Scholar] [CrossRef]
  89. Maitra, R.; Seetharam, R.; Tesfa, L.; Augustine, T.A.; Klampfer, L.; Coffey, M.C.; Mariadason, J.M.; Goel, S. Oncolytic reovirus preferentially induces apoptosis in KRAS mutant colorectal cancer cells, and synergizes with irinotecan. Oncotarget 2014, 5, 2807. [Google Scholar] [CrossRef]
  90. Lee, W.S.; Lee, S.J.; Lee, H.J.; Yang, H.; Go, E.-J.; Gansukh, E.; Song, K.-H.; Xiang, X.; Park, D.G.; Alain, T. Oral reovirus reshapes the gut microbiome and enhances antitumor immunity in colon cancer. Nat. Commun. 2024, 15, 9092. [Google Scholar] [CrossRef]
  91. Parakrama, R.; Fogel, E.; Chandy, C.; Augustine, T.; Coffey, M.; Tesfa, L.; Goel, S.; Maitra, R. Immune characterization of metastatic colorectal cancer patients post reovirus administration. BMC Cancer 2020, 20, 1–11. [Google Scholar] [CrossRef] [PubMed]
  92. Hastie, E.; Grdzelishvili, V.Z. Vesicular stomatitis virus as a flexible platform for oncolytic virotherapy against cancer. J. Gen. Virol. 2012, 93, 2529–2545. [Google Scholar] [CrossRef] [PubMed]
  93. Abdelmageed, A.A.; Dewhurst, S.; Ferran, M.C. Employing the Oncolytic Vesicular Stomatitis Virus in Cancer Virotherapy: Resistance and Clinical Considerations. Viruses 2024, 17, 16. [Google Scholar] [CrossRef]
  94. Ahmed, M.M.; Okesanya, O.J.; Ukoaka, B.M.; Ibrahim, A.M.; Lucero-Prisno, D.E., III. Vesicular Stomatitis Virus: Insights into Pathogenesis, Immune Evasion, and Technological Innovations in Oncolytic and Vaccine Development. Viruses 2024, 16, 1933. [Google Scholar] [CrossRef]
  95. Gray, Z.; Tabarraei, A.; Moradi, A.; Kalani, M.R. M51R and Delta-M51 matrix protein of the vesicular stomatitis virus induce apoptosis in colorectal cancer cells. Mol. Biol. Rep. 2019, 46, 3371–3379. [Google Scholar] [CrossRef]
  96. Day, G.L.; Bryan, M.L.; Northrup, S.A.; Lyles, D.S.; Westcott, M.M.; Stewart, J.H., IV. Immune effects of M51R vesicular stomatitis virus treatment of carcinomatosis from colon cancer. J. Surg. Res. 2020, 245, 127–135. [Google Scholar] [CrossRef]
  97. Ohteki, T. Critical role for IL-15 in innate immunity. Curr. Mol. Med. 2002, 2, 371–380. [Google Scholar] [CrossRef]
  98. Stephenson, K.; Barra, N.; Davies, E.; Ashkar, A.; Lichty, B. Expressing human interleukin-15 from oncolytic vesicular stomatitis virus improves survival in a murine metastatic colon adenocarcinoma model through the enhancement of anti-tumor immunity. Cancer Gene Ther. 2012, 19, 238–246. [Google Scholar] [CrossRef]
  99. Larrieux, A.; Sanjuán, R. Murine colon cancer derived cells exhibit heterogeneous resistance profiles against an oncolytic virus. Sci. Rep. 2024, 14, 27209. [Google Scholar] [CrossRef]
  100. Selman, M.; Rousso, C.; Bergeron, A.; Son, H.H.; Krishnan, R.; El-Sayes, N.A.; Varette, O.; Chen, A.; Le Boeuf, F.; Tzelepis, F. Multi-modal potentiation of oncolytic virotherapy by vanadium compounds. Mol. Ther. 2018, 26, 56–69. [Google Scholar] [CrossRef]
  101. Alluqmani, N.; Jirovec, A.; Taha, Z.; Varette, O.; Chen, A.; Serrano, D.; Maznyi, G.; Khan, S.; Forbes, N.E.; Arulanandam, R. Vanadyl sulfate-enhanced oncolytic virus immunotherapy mediates the antitumor immune response by upregulating the secretion of pro-inflammatory cytokines and chemokines. Front. Immunol. 2022, 13, 1032356. [Google Scholar] [CrossRef] [PubMed]
  102. McAusland, T.M.; van Vloten, J.P.; Santry, L.A.; Guilleman, M.M.; Rghei, A.D.; Ferreira, E.M.; Ingrao, J.C.; Arulanandam, R.; Major, P.P.; Susta, L. Combining vanadyl sulfate with Newcastle disease virus potentiates rapid innate immune-mediated regression with curative potential in murine cancer models. Mol. Ther. Oncolytics 2021, 20, 306–324. [Google Scholar] [CrossRef] [PubMed]
  103. Bambouskova, M.; Gorvel, L.; Lampropoulou, V.; Sergushichev, A.; Loginicheva, E.; Johnson, K.; Korenfeld, D.; Mathyer, M.E.; Kim, H.; Huang, L.-H. Electrophilic properties of itaconate and derivatives regulate the IκBζ–ATF3 inflammatory axis. Nature 2018, 556, 501–504. [Google Scholar] [CrossRef] [PubMed]
  104. Mills, E.L.; Ryan, D.G.; Prag, H.A.; Dikovskaya, D.; Menon, D.; Zaslona, Z.; Jedrychowski, M.P.; Costa, A.S.; Higgins, M.; Hams, E. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature 2018, 556, 113–117. [Google Scholar] [CrossRef]
  105. Kurmasheva, N.; Said, A.; Wong, B.; Kinderman, P.; Han, X.; Rahimic, A.H.; Kress, A.; Carter-Timofte, M.E.; Holm, E.; van der Horst, D. Octyl itaconate enhances VSVΔ51 oncolytic virotherapy by multitarget inhibition of antiviral and inflammatory pathways. Nat. Commun. 2024, 15, 4096. [Google Scholar] [CrossRef]
  106. Li, M.; Zhang, M.; Ye, Q.; Liu, Y.; Qian, W. Preclinical and clinical trials of oncolytic vaccinia virus in cancer immunotherapy: A comprehensive review. Cancer Biol. Med. 2023, 20, 646–661. [Google Scholar] [CrossRef]
  107. Inoue, T.; Byrne, T.; Inoue, M.; Tait, M.E.; Wall, P.; Wang, A.; Dermyer, M.R.; Laklai, H.; Binder, J.J.; Lees, C.; et al. Oncolytic Vaccinia Virus Gene Modification and Cytokine Expression Effects on Tumor Infection, Immune Response, and Killing. Mol. Cancer Ther. 2021, 20, 1481–1494. [Google Scholar] [CrossRef]
  108. Kilinc, M.O.; Ehrig, K.; Pessian, M.; Minev, B.R.; Szalay, A.A. Colonization of xenograft tumors by oncolytic vaccinia virus (VACV) results in enhanced tumor killing due to the involvement of myeloid cells. J. Transl. Med. 2016, 14, 1–12. [Google Scholar] [CrossRef]
  109. Guo, Z.S.; Lu, B.; Guo, Z.; Giehl, E.; Feist, M.; Dai, E.; Liu, W.; Storkus, W.J.; He, Y.; Liu, Z.; et al. Vaccinia virus-mediated cancer immunotherapy: Cancer vaccines and oncolytics. J. Immunother. Cancer 2019, 7, 6. [Google Scholar] [CrossRef]
  110. Foloppe, J.; Kintz, J.; Futin, N.; Findeli, A.; Cordier, P.; Schlesinger, Y.; Hoffmann, C.; Tosch, C.; Balloul, J.; Erbs, P. Targeted delivery of a suicide gene to human colorectal tumors by a conditionally replicating vaccinia virus. Gene Ther. 2008, 15, 1361–1371. [Google Scholar] [CrossRef]
  111. Jeong, S.N.; Yoo, S.Y. Novel Oncolytic Virus Armed with Cancer Suicide Gene and Normal Vasculogenic Gene for Improved Anti-Tumor Activity. Cancers 2020, 12, 1070. [Google Scholar] [CrossRef] [PubMed]
  112. Ho, T.Y.; Mealiea, D.; Okamoto, L.; Stojdl, D.F.; McCart, J.A. Deletion of immunomodulatory genes as a novel approach to oncolytic vaccinia virus development. Mol. Ther. Oncolytics 2021, 22, 85–97. [Google Scholar] [CrossRef] [PubMed]
  113. Génin, P.; Morin, P.; Civas, A. Impairment of interferon-induced IRF-7 gene expression due to inhibition of ISGF3 formation by trichostatin A. J. Virol. 2003, 77, 7113–7119. [Google Scholar] [CrossRef]
  114. Suh, H.-S.; Choi, S.; Khattar, P.; Choi, N.; Lee, S.C. Histone deacetylase inhibitors suppress the expression of inflammatory and innate immune response genes in human microglia and astrocytes. J. Neuroimmune Pharmacol. 2010, 5, 521–532. [Google Scholar] [CrossRef]
  115. Vanhaecke, T.; Papeleu, P.; Elaut, G.; Rogiers, V. Trichostatin A-like hydroxamate histone deacetylase inhibitors as therapeutic agents: Toxicological point of view. Curr. Med. Chem. 2004, 11, 1629–1643. [Google Scholar] [CrossRef]
  116. Colamonici, O.R.; Domanski, P.; Sweitzer, S.M.; Larner, A.; Buller, R.M.L. Vaccinia virus B18R gene encodes a type I interferon-binding protein that blocks interferon α transmembrane signaling (∗). J. Biol. Chem. 1995, 270, 15974–15978. [Google Scholar] [CrossRef]
  117. Kirn, D.H.; Wang, Y.; Le Boeuf, F.; Bell, J.; Thorne, S.H. Targeting of interferon-beta to produce a specific, multi-mechanistic oncolytic vaccinia virus. PLoS Med. 2007, 4, e353. [Google Scholar] [CrossRef]
  118. MacTavish, H.; Diallo, J.S.; Huang, B.; Stanford, M.; Le Boeuf, F.; De Silva, N.; Cox, J.; Simmons, J.G.; Guimond, T.; Falls, T.; et al. Enhancement of vaccinia virus based oncolysis with histone deacetylase inhibitors. PLoS ONE 2010, 5, e14462. [Google Scholar] [CrossRef]
  119. Li, Z.; Li, M.; Yang, L.; Chen, J.; Ye, Q.; Qian, W.; Wang, S. Oncolytic vaccinia virus armed with anti-CD47 nanobody elicit potent antitumor effects on multiple tumor models via enhancing innate and adoptive immunity. J. Immunother. Cancer 2024, 12, e009473. [Google Scholar] [CrossRef]
  120. Francis, L.; Guo, Z.S.; Liu, Z.; Ravindranathan, R.; Urban, J.A.; Sathaiah, M.; Magge, D.; Kalinski, P.; Bartlett, D.L. Modulation of chemokines in the tumor microenvironment enhances oncolytic virotherapy for colorectal cancer. Oncotarget 2016, 7, 22174. [Google Scholar] [CrossRef]
  121. Li, J.; O’Malley, M.; Sampath, P.; Kalinski, P.; Bartlett, D.L.; Thorne, S.H. Expression of CCL19 from oncolytic vaccinia enhances immunotherapeutic potential while maintaining oncolytic activity. Neoplasia 2012, 14, 1115–1121. [Google Scholar] [CrossRef] [PubMed]
  122. Chen, L.; Chen, H.; Ye, J.; Ge, Y.; Wang, H.; Dai, E.; Ren, J.; Liu, W.; Ma, C.; Ju, S. Intratumoral expression of interleukin 23 variants using oncolytic vaccinia virus elicit potent antitumor effects on multiple tumor models via tumor microenvironment modulation. Theranostics 2021, 11, 6668. [Google Scholar] [CrossRef] [PubMed]
  123. Wang, J.; Sun, M.; Zhao, H.; Huang, Y.; Li, D.; Mao, D.; Zhang, Z.; Zhu, X.; Dong, X.; Zhao, X. IL-9 exerts antitumor effects in colon cancer and transforms the tumor microenvironment in vivo. Technol. Cancer Res. Treat. 2019, 18, 1533033819857737. [Google Scholar] [CrossRef]
  124. Ye, J.; Chen, L.; Waltermire, J.; Zhao, J.; Ren, J.; Guo, Z.; Bartlett, D.L.; Liu, Z. Intratumoral Delivery of Interleukin 9 via Oncolytic Vaccinia Virus Elicits Potent Antitumor Effects in Tumor Models. Cancers 2024, 16, 1021. [Google Scholar] [CrossRef]
  125. Wu, R.; Tong, S.; Yin, J.; Zhu, Z.; Mao, Z.; Xu, L. Oncolytic vaccinia virus acts synergistically with anti-PD-L1 antibody to enhance the killing of colon cancer cells by CD8(+) T cells. Pathol. Res. Pr. 2023, 247, 154535. [Google Scholar] [CrossRef]
  126. Shakiba, Y.; Vorobyev, P.O.; Yusubalieva, G.M.; Kochetkov, D.V.; Zajtseva, K.V.; Valikhov, M.P.; Kalsin, V.A.; Zabozlaev, F.G.; Semkina, A.S.; Troitskiy, A.V.; et al. Oncolytic therapy with recombinant vaccinia viruses targeting the interleukin-15 pathway elicits a synergistic response. Mol. Ther. Oncolytics 2023, 29, 158–168. [Google Scholar] [CrossRef]
  127. Ge, Y.; Wang, H.; Ren, J.; Liu, W.; Chen, L.; Chen, H.; Ye, J.; Dai, E.; Ma, C.; Ju, S.; et al. Oncolytic vaccinia virus delivering tethered IL-12 enhances antitumor effects with improved safety. J. Immunother. Cancer 2020, 8, e000710. [Google Scholar] [CrossRef]
  128. Engeland, C.E.; Ungerechts, G. Measles virus as an oncolytic immunotherapy. Cancers 2021, 13, 544. [Google Scholar] [CrossRef]
  129. Kleinlützum, D.; Hanauer, J.D.; Muik, A.; Hanschmann, K.-M.; Kays, S.-K.; Ayala-Breton, C.; Peng, K.-W.; Mühlebach, M.D.; Abel, T.; Buchholz, C.J. Enhancing the oncolytic activity of CD133-targeted measles virus: Receptor extension or chimerism with vesicular stomatitis virus are most effective. Front. Oncol. 2017, 7, 127. [Google Scholar] [CrossRef]
  130. Wang, J.; Liu, T.; Chen, J. Oncolytic Measles Virus Encoding Interleukin-12 Mediated Antitumor Activity and Immunologic Control of Colon Cancer In Vivo and Ex Vivo. Cancer Biother. Radiopharm. 2021, 36, 774–782. [Google Scholar]
  131. Jing, Y.; Tong, C.; Zhang, J.; Nakamura, T.; Iankov, I.; Russell, S.J.; Merchan, J.R. Tumor and vascular targeting of a novel oncolytic measles virus retargeted against the urokinase receptor. Cancer Res. 2009, 69, 1459–1468. [Google Scholar] [CrossRef] [PubMed]
  132. Jing, Y.; Zaias, J.; Duncan, R.; Russell, S.J.; Merchan, J.R. In vivo safety, biodistribution and antitumor effects of uPAR retargeted oncolytic measles virus in syngeneic cancer models. Gene Ther. 2014, 21, 289–297. [Google Scholar] [CrossRef] [PubMed]
  133. Bach, P.; Abel, T.; Hoffmann, C.; Gal, Z.; Braun, G.; Voelker, I.; Ball, C.R.; Johnston, I.C.D.; Lauer, U.M.; Herold-Mende, C.; et al. Specific Elimination of CD133+ Tumor Cells with Targeted Oncolytic Measles Virus. Cancer Res. 2013, 73, 865–874. [Google Scholar] [CrossRef] [PubMed]
  134. U.S. National Library of Medicine. Mechanism of Action Trial of ColoAd1 (MOA). 2013. Available online: https://www.clinicaltrials.gov/study/NCT02053220 (accessed on 13 May 2025).
  135. U.S. National Library of Medicine. A Clinical Study of BioTTT001 in Combination with Toripalimab and Regorafenib in Patients with Colorectal Cancer. 2024. Available online: https://clinicaltrials.gov/study/NCT06283134 (accessed on 13 May 2025).
  136. U.S. National Library of Medicine. Pilot Study of GVAX in Colorectal Cancer Cells. 2023. Available online: https://clinicaltrials.gov/study/NCT01952730 (accessed on 13 May 2025).
  137. U.S. National Library of Medicine. Standard of Care Alone or in Combination with Ad-CEA Vaccine and Avelumab in People with Previously Untreated Metastatic Colorectal Cancer QUILT-2.004. 2021. Available online: https://clinicaltrials.gov/study/NCT03050814 (accessed on 13 May 2025).
  138. U.S. National Library of Medicine. Multi-Targeted Recombinant Ad5 (CEA/MUC1/Brachyury) Based Immunotherapy Vaccine Regimen in People with Advanced Cancer. 2020. Available online: https://clinicaltrials.gov/study/NCT03384316 (accessed on 13 May 2025).
  139. U.S. National Library of Medicine. Interleukin-12 Gene in Treating Patients with Liver Metastases Secondary to Colorectal Cancer. 2017. Available online: https://clinicaltrials.gov/study/NCT00072098 (accessed on 13 May 2025).
  140. U.S. National Library of Medicine. Phase I Study of Ad5-hGCC (Human Guanylyl Cyclase C)-PADRE in Stage I/II Colon Cancer. 2025. Available online: https://clinicaltrials.gov/study/NCT01972737 (accessed on 13 May 2025).
  141. U.S. National Library of Medicine. A Vaccine (Ad5.F35-hGCC-PADRE) for the Treatment of Gastrointestinal Adenocarcinoma. 2025. Available online: https://clinicaltrials.gov/study/NCT04111172 (accessed on 13 May 2025).
  142. U.S. National Library of Medicine. Safety Study of Human MUC-1 (Mucin-1) Adenoviral Vector Vaccine for Immunotherapy of Epithelial Cancers (MUC-1). 2016. Available online: https://clinicaltrials.gov/study/NCT02140996 (accessed on 13 May 2025).
  143. U.S. National Library of Medicine. VB-111 in Combination with Nivolumab in People with Metastatic Colorectal Cancer (mCRC). 2023. Available online: https://clinicaltrials.gov/study/NCT04166383 (accessed on 13 May 2025).
  144. U.S. National Library of Medicine. RP2/RP3 in Combination with Atezolizumab and Bevacizumab for the Treatment of Patients with CRC. 2025. Available online: https://clinicaltrials.gov/study/NCT05733611 (accessed on 13 May 2025).
  145. U.S. National Library of Medicine. A Clinical Study of T3011 in Combination with Toripalimab and Regorafenib in Patients with Colorectal Cancer. 2024. Available online: https://clinicaltrials.gov/study/NCT06283303 (accessed on 13 May 2025).
  146. U.S. National Library of Medicine. Gene Therapy in Treating Patients with Colon Cancer That Has Spread to the Liver. 2013. Available online: https://clinicaltrials.gov/study/NCT00012155 (accessed on 13 May 2025).
  147. U.S. National Library of Medicine. First-line Maintenance of OH2 Injection for Advanced Colorectal Cancer. 2024. Available online: https://clinicaltrials.gov/study/NCT05648006 (accessed on 13 May 2025).
  148. U.S. National Library of Medicine. Study of ONCR-177 Alone and in Combination with PD-1 Blockade in Adult Subjects with Advanced and/or Refractory Cutaneous, Subcutaneous or Metastatic Nodal Solid Tumors or with Liver Metastases of Solid Tumors. 2023. Available online: https://clinicaltrials.gov/study/NCT04348916 (accessed on 13 May 2025).
  149. U.S. National Library of Medicine. A Phase I Clinical Study of Intratumoral Injection Oncolytic Vaccinia Virus GC001 in Patient with Advanced Solid Tumors. 2024. Available online: https://clinicaltrials.gov/study/NCT06508307 (accessed on 13 May 2025).
  150. U.S. National Library of Medicine. A Trial of Perioperative CV301 Vaccination in Combination with Nivolumab and Systemic Chemotherapy for Metastatic CRC. 2024. Available online: https://clinicaltrials.gov/study/NCT03547999 (accessed on 13 May 2025).
  151. U.S. National Library of Medicine. Vaccine Therapy in Treating Patients with Colorectal, Stomach, or Pancreatic Cancer. 2017. Available online: https://clinicaltrials.gov/study/NCT01191684 (accessed on 13 May 2025).
  152. U.S. National Library of Medicine. Vaccine Therapy and Pembrolizumab in Treating Patients with Solid Tumors That Have Failed Prior Therapy. 2025. Available online: https://clinicaltrials.gov/study/NCT02432963 (accessed on 13 May 2025).
  153. U.S. National Library of Medicine. Safety Study of Recombinant Vaccinia Virus Administered Intravenously in Patients with Metastatic, Refractory Colorectal Carcinoma. 2016. Available online: https://clinicaltrials.gov/study/NCT01380600 (accessed on 13 May 2025).
  154. U.S. National Library of Medicine. Recombinant Vaccinia Virus Administered Intravenously in Patients with Metastatic, Refractory Colorectal Carcinoma. 2021. Available online: https://clinicaltrials.gov/study/NCT01394939 (accessed on 13 May 2025).
  155. U.S. National Library of Medicine. A Phase I/II Study of Pexa-Vec Oncolytic Virus in Combination with Immune Checkpoint Inhibition in Refractory Colorectal Cancer. 2023. Available online: https://clinicaltrials.gov/study/NCT03206073 (accessed on 13 May 2025).
  156. U.S. National Library of Medicine. Vaccine Therapy and Sargramostim with or Without Docetaxel in Treating Patients with Metastatic Lung Cancer or Metastatic Colorectal Cancer. 2014. Available online: https://clinicaltrials.gov/study/NCT00088933 (accessed on 13 May 2025).
  157. U.S. National Library of Medicine. Vaccine Therapy in Treating Patients with Liver or Lung Metastases From Colorectal Cancer. 2015. Available online: https://clinicaltrials.gov/study/NCT00103142 (accessed on 13 May 2025).
  158. Flatt, J.W.; Kim, R.; Smith, J.G.; Nemerow, G.R.; Stewart, P.L. An intrinsically disordered region of the adenovirus capsid is implicated in neutralization by human alpha defensin 5. PLoS ONE 2013, 8, e61571. [Google Scholar] [CrossRef]
  159. Smith, J.G.; Silvestry, M.; Lindert, S.; Lu, W.; Nemerow, G.R.; Stewart, P.L. Insight into the mechanisms of adenovirus capsid disassembly from studies of defensin neutralization. PLoS Pathog. 2010, 6, e1000959. [Google Scholar] [CrossRef]
  160. Vragniau, C.; Hübner, J.-M.; Beidler, P.; Gil, S.; Saydaminova, K.; Lu, Z.-Z.; Yumul, R.; Wang, H.; Richter, M.; Sova, P. Studies on the interaction of tumor-derived HD5 alpha defensins with adenoviruses and implications for oncolytic adenovirus therapy. J. Virol. 2017, 91, e02185-18. [Google Scholar] [CrossRef]
  161. Kulu, Y.; Kawasaki, H.; Donahue, J.M.; Kasuya, H.; Cusack, J.C.; Choi, E.W.; Kuruppu, D.K.; Fuchs, B.C.; Tanabe, K.K. Concurrent chemotherapy inhibits herpes simplex virus-1 replication and oncolysis. Cancer Gene Ther. 2013, 20, 133–140. [Google Scholar] [CrossRef]
  162. Song, T.; Haddad, D.; Adusumilli, P.; Kim, T.; Stiles, B.; Hezel, M.; Socci, N.; Gönen, M.; Fong, Y. Molecular network pathways and functional analysis of tumor signatures associated with development of resistance to viral gene therapy. Cancer Gene Ther. 2012, 19, 38–48. [Google Scholar] [CrossRef]
  163. Van Houdt, W.; Smakman, N.; van Den Wollenberg, D.; Emmink, B.; Veenendaal, L.; Van Diest, P.; Hoeben, R.; Borel Rinkes, I.; Kranenburg, O. Transient infection of freshly isolated human colorectal tumor cells by reovirus T3D intermediate subviral particles. Cancer Gene Ther. 2008, 15, 284–292. [Google Scholar] [CrossRef]
  164. Larrieux, A.; Sanjuán, R. Cellular resistance to an oncolytic virus is driven by chronic activation of innate immunity. Iscience 2023, 26, 105749. [Google Scholar] [CrossRef]
  165. Clarke, P.; Meintzer, S.M.; Wang, Y.; Moffitt, L.A.; Richardson-Burns, S.M.; Johnson, G.L.; Tyler, K.L. JNK regulates the release of proapoptotic mitochondrial factors in reovirus-infected cells. J. Virol. 2004, 78, 13132–13138. [Google Scholar] [CrossRef] [PubMed]
  166. Hirasawa, K.; Kim, A.; Han, H.-S.; Han, J.; Jun, H.-S.; Yoon, J.-W. Effect of p38 mitogen-activated protein kinase on the replication of encephalomyocarditis virus. J. Virol. 2003, 77, 5649–5656. [Google Scholar] [CrossRef] [PubMed]
  167. Holloway, G.; Coulson, B.S. Rotavirus activates JNK and p38 signaling pathways in intestinal cells, leading to AP-1-driven transcriptional responses and enhanced virus replication. J. Virol. 2006, 80, 10624–10633. [Google Scholar] [CrossRef]
  168. Rahaus, M.; Desloges, N.; Wolff, M.H. Replication of varicella-zoster virus is influenced by the levels of JNK/SAPK and p38/MAPK activation. J. Gen. Virol. 2004, 85, 3529–3540. [Google Scholar] [CrossRef]
  169. Hu, W.; Hofstetter, W.; Guo, W.; Li, H.; Pataer, A.; Peng, H.H.; Guo, Z.S.; Bartlett, D.L.; Lin, A.; Swisher, S.G. JNK-deficiency enhanced oncolytic vaccinia virus replication and blocked activation of double-stranded RNA-dependent protein kinase. Cancer Gene Ther. 2008, 15, 616–624. [Google Scholar] [CrossRef]
  170. Feola, S.; Chiaro, J.; Martins, B.; Russo, S.; Fusciello, M.; Ylösmäki, E.; Bonini, C.; Ruggiero, E.; Hamdan, F.; Feodoroff, M. A novel immunopeptidomic-based pipeline for the generation of personalized oncolytic cancer vaccines. Elife 2022, 11, e71156. [Google Scholar] [CrossRef]
  171. Feola, S.; Hamdan, F.; Russo, S.; Chiaro, J.; Fusciello, M.; Feodoroff, M.; Antignani, G.; D’Alessio, F.; Mölsä, R.; Stigzelius, V. Novel peptide-based oncolytic vaccine for enhancement of adaptive antitumor immune response via co-engagement of innate Fcγ and Fcα receptors. J. Immunother. Cancer 2024, 12, e008342. [Google Scholar] [CrossRef]
  172. Le Boeuf, F.; Gebremeskel, S.; McMullen, N.; He, H.; Greenshields, A.L.; Hoskin, D.W.; Bell, J.C.; Johnston, B.; Pan, C.; Duncan, R. Reovirus FAST protein enhances vesicular stomatitis virus oncolytic virotherapy in primary and metastatic tumor models. Mol. Ther. Oncolytics 2017, 6, 80–89. [Google Scholar] [CrossRef]
  173. Malilas, W.; Koh, S.S.; Srisuttee, R.; Boonying, W.; Cho, I.; Jeong, C.; Johnston, R.; Chung, Y. Cancer upregulated gene 2, a novel oncogene, confers resistance to oncolytic vesicular stomatitis virus through STAT1-OASL2 signaling. Cancer Gene Ther. 2013, 20, 125–132. [Google Scholar] [CrossRef]
  174. Nakatake, M.; Kurosaki, H.; Kuwano, N.; Horita, K.; Ito, M.; Kono, H.; Okamura, T.; Hasegawa, K.; Yasutomi, Y.; Nakamura, T. Partial deletion of glycoprotein B5R enhances vaccinia virus neutralization escape while preserving oncolytic function. Mol. Ther. Oncolytics 2019, 14, 159–171. [Google Scholar] [CrossRef]
  175. Burton, C.; Bartee, E. Syncytia formation in oncolytic virotherapy. Mol. Ther. Oncolytics 2019, 15, 131–139. [Google Scholar] [CrossRef] [PubMed]
  176. Nakatake, M.; Kuwano, N.; Kaitsurumaru, E.; Kurosaki, H.; Nakamura, T. Fusogenic oncolytic vaccinia virus enhances systemic antitumor immune response by modulating the tumor microenvironment. Mol. Ther. 2021, 29, 1782–1793. [Google Scholar] [CrossRef] [PubMed]
  177. Liu, W.; Chen, H.; Zhu, Z.; Liu, Z.; Ma, C.; Lee, Y.J.; Bartlett, D.L.; Guo, Z.-S. Ferroptosis inducer improves the efficacy of oncolytic virus-mediated cancer immunotherapy. Biomedicines 2022, 10, 1425. [Google Scholar] [CrossRef]
  178. Yamaki, M.; Shinozaki, K.; Sakaguchi, T.; Meseck, M.; Ebert, O.; Ohdan, H.; Woo, S.L. The potential of recombinant vesicular stomatitis virus-mediated virotherapy against metastatic colon cancer. Int. J. Mol. Med. 2013, 31, 299–306. [Google Scholar] [CrossRef]
  179. Zeh, H.J.; Downs-Canner, S.; McCart, J.A.; Guo, Z.S.; Rao, U.N.; Ramalingam, L.; Thorne, S.H.; Jones, H.L.; Kalinski, P.; Wieckowski, E.; et al. First-in-man study of western reserve strain oncolytic vaccinia virus: Safety, systemic spread, and antitumor activity. Mol. Ther. 2015, 23, 202–214. [Google Scholar] [CrossRef]
  180. Goossens, M.; Pauwels, K.; Willemarck, N.; Breyer, D. Environmental risk assessment of clinical trials involving modified vaccinia virus Ankara (MVA)-based vectors. Curr. Gene Ther. 2013, 13, 413–420. [Google Scholar] [CrossRef]
  181. Petersen, B.W.; Harms, T.J.; Reynolds, M.G.; Harrison, L.H. Use of Vaccinia Virus Smallpox Vaccine in Laboratory and Health Care Personnel at Risk for Occupational Exposure to Orthopoxviruses—Recommendations of the Advisory Committee on Immunization Practices (ACIP), 2015. MMWR Morb. Mortal. Wkly. Rep. 2016, 65, 257–262. [Google Scholar] [CrossRef]
  182. Badrinath, N.; Jeong, Y.I.; Woo, H.Y.; Bang, S.Y.; Kim, C.; Heo, J.; Kang, D.H.; Yoo, S.Y. Local delivery of a cancer-favoring oncolytic vaccinia virus via poly (lactic-co-glycolic acid) nanofiber for theranostic purposes. Int. J. Pharm. 2018, 552, 437–442. [Google Scholar] [CrossRef]
  183. Liu, Z.; Ravindranathan, R.; Kalinski, P.; Guo, Z.S.; Bartlett, D.L. Rational combination of oncolytic vaccinia virus and PD-L1 blockade works synergistically to enhance therapeutic efficacy. Nat. Commun. 2017, 8, 14754. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the leading oncolytic virus platforms investigated for colorectal cancer (CRC) and their mechanisms of action. Adenoviruses (AD) selectively replicate in tumor cells and deliver therapeutic genes such as IL-15, GM-CSF, and TRAIL, promoting direct lysis and immune activation. Herpes simplex viruses (HSV) induce tumor cell death and express immunomodulatory molecules such as IL-12, CXCL11, and anti–PD–1 antibodies, facilitating CD8+ T cell recruitment and the remodeling of the tumor microenvironment (TME). Reoviruses preferentially infect KRAS-mutant cells and activate innate immunity via TLR3, leading to apoptotic tumor cell death. Vesicular stomatitis viruses (VSV) trigger apoptosis through their matrix protein and are armed with IL-15 to enhance CD8+ and NK cell responses while modulating interferon signaling. Vaccinia viruses (VV) act effectively in hypoxic tumor areas and express a range of cytokines, including IL-12, IL-9, IL-23, and TRAIL, contributing to immune cell infiltration and the suppression of tumor-promoting signals. Measles viruses (MV) target tumor-initiating cells expressing uPAR or CD133 and are often engineered to express IL-12, inducing durable anti-tumor immune responses and long-term memory. (Created in https://BioRender.com. Accessed on 13 May 2025).
Figure 1. Schematic representation of the leading oncolytic virus platforms investigated for colorectal cancer (CRC) and their mechanisms of action. Adenoviruses (AD) selectively replicate in tumor cells and deliver therapeutic genes such as IL-15, GM-CSF, and TRAIL, promoting direct lysis and immune activation. Herpes simplex viruses (HSV) induce tumor cell death and express immunomodulatory molecules such as IL-12, CXCL11, and anti–PD–1 antibodies, facilitating CD8+ T cell recruitment and the remodeling of the tumor microenvironment (TME). Reoviruses preferentially infect KRAS-mutant cells and activate innate immunity via TLR3, leading to apoptotic tumor cell death. Vesicular stomatitis viruses (VSV) trigger apoptosis through their matrix protein and are armed with IL-15 to enhance CD8+ and NK cell responses while modulating interferon signaling. Vaccinia viruses (VV) act effectively in hypoxic tumor areas and express a range of cytokines, including IL-12, IL-9, IL-23, and TRAIL, contributing to immune cell infiltration and the suppression of tumor-promoting signals. Measles viruses (MV) target tumor-initiating cells expressing uPAR or CD133 and are often engineered to express IL-12, inducing durable anti-tumor immune responses and long-term memory. (Created in https://BioRender.com. Accessed on 13 May 2025).
Cancers 17 01854 g001
Table 1. Clinical trials of oncolytic virus therapy in patients with CRC *.
Table 1. Clinical trials of oncolytic virus therapy in patients with CRC *.
Virus TypeTherapyCancer StatusStudy StatusPhase ParticipantsClinical Trial ID
AdenovirusColoAd1ResectableCompleted1 17NCT02053220
BioTTT001 + Toraplizumab and RegorafenibLiver MetastasisNot yet recruiting1 40NCT06283134
GVAXLiver MetastasisTerminated1 (Pilot study)1NCT01952730
Ad-CEA + avelumabMetastaticTerminated2 30NCT03050814
ETBX-011, ETBX-061, ETBX-051AdvancedCompleted1 11NCT03384316
IL-12MetastaticTerminated1 22NCT00072098
Ad5-hGCC-PADREStage I/IICompleted1 1NCT01972737
Ad5.F35-hGCC-PADREStage III/IVActive, not recruiting2 81NCT04111172
Ad-sig-hMUC-1/ecdCD40LRecurrent or MetastaticUnknown status1 24NCT02140996
VB-111 + NivolumabMetastaticCompleted2 14NCT04166383
Herpes Simplex VirusRP2/RP3 + Atezolizumab and BevacizumabAdvancedActive, not recruiting2 4NCT05733611
T3011 + Toripalimab and RegorafenibLiver MetastasisNot yet recruiting1 8NCT06283303
NV1020Liver MetastasisCompleted1 No dataNCT00012155
OH2 + CapecitabineAdvancedTerminated2 7NCT05648006
ONCR-177Refractory, MetastaticTerminated1 66NCT04348916
ReovirusREOLYSIN + FOLFIRI and bevacizumabMetastaticCompleted1 36NCT01274624
Vaccinia VirusGC001AdvancedRecruiting1 21NCT06508307
CV301 + Nivolumab and Systemic ChemotherapyMetastatic Active, not recruiting2 78NCT03547999
p53MVAUnresectable and chemotherapy resistantCompleted1 12NCT01191684
p53MVA + pembrolizumabAdvancedActive, not recruiting1 11NCT02432963
JX-594RefractoryCompleted1 15NCT01469611
JX-594Liver MetastasisTerminated2a 2NCT01329809
JX-594Metastatic, RefractoryCompleted1b 15NCT01380600
JX-594Metastatic, RefractoryCompleted1/2a 52NCT01394939
JX-594 + Tremelimumab/DurvalumabRefractoryCompleted1/2 34NCT03206073
vaccinia-CEA-TRICOM + docetaxelMetastaticTerminated1 60NCT00088933
vaccinia-CEA-MUC-1-TRICOMMetastaticCompleted2 74NCT00103142
Measles VirusMVF-HER-2 (266–296/597–626)MetastaticCompleted1 65NCT01376505
PD1-VaxxOperable high MSI a Not yet recruiting2 44NCT06692959
* Data taken from clinicaltrials.gov; neither suspended nor withdrawn from trials. Results already published. a Microsatellite instability—high.
Table 2. Comparative analysis of OVs used in CRC.
Table 2. Comparative analysis of OVs used in CRC.
VirusGenome/TypeTumor SelectivityImmunostimulatory PotentialClinical SafetyClinical Maturity *Key AdvantagesCritical Limitations
AdenovirusdsDNA, non-envelopedHigh (e.g., LOI, p53 loss)High (e.g., IL-15, GM-CSF)Well characterizedPhase I–II in CRCGenetically tractable, scalable productionPre-existing immunity, blocked by HD5, limited systemic delivery
Herpes Simplex Virus (HSV)dsDNA, envelopedHigh (solid tumors)Very highHigh (e.g., T-VEC approved)Phase I–II in CRCLarge transgene capacity, antiviral control optionsComplex manufacturing, latency potential
ReovirusdsRNA, non-envelopedModerate (RAS-mutant preference)ModerateExcellentPhase I in CRCOral delivery, native tropism, minimal engineeringJAM1 receptor localization limits infectivity in primary tumors
Vesicular Stomatitis Virus (VSV)ssRNA, envelopedHigh (IFN-defective cells)HighModeratePreclinical/Phase IRapid replication, fusogenic potential, and low seroprevalenceSystemic toxicity, fast immune clearance
Vaccinia Virus (VV)dsDNA, envelopedHigh (hypoxia adapted)HighHigh (extensive safety data)Phase I–II in CRCCytoplasmic replication, large genome for transgenesManufacturing burden, proinflammatory, and immune clearance
Measles Virus (MV)ssRNA, envelopedHigh (via uPAR/CD133 targeting)HighHigh (attenuated strains)Phase I in CRCTumor tropism, engineered targeting, systemic administrationRisk of neutralizing antibodies, less clinical experience in CRC
* Data taken from: clinicaltrials.gov.
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

Pérez-Domínguez, F.; Quezada-Monrás, C.; Cárcamo, L.; Muñoz, J.P.; Carrillo-Beltrán, D. Oncolytic Viruses as a Novel Therapeutic Approach for Colorectal Cancer: Mechanisms, Current Advances, and Future Directions. Cancers 2025, 17, 1854. https://doi.org/10.3390/cancers17111854

AMA Style

Pérez-Domínguez F, Quezada-Monrás C, Cárcamo L, Muñoz JP, Carrillo-Beltrán D. Oncolytic Viruses as a Novel Therapeutic Approach for Colorectal Cancer: Mechanisms, Current Advances, and Future Directions. Cancers. 2025; 17(11):1854. https://doi.org/10.3390/cancers17111854

Chicago/Turabian Style

Pérez-Domínguez, Francisco, Claudia Quezada-Monrás, Leonardo Cárcamo, Juan P. Muñoz, and Diego Carrillo-Beltrán. 2025. "Oncolytic Viruses as a Novel Therapeutic Approach for Colorectal Cancer: Mechanisms, Current Advances, and Future Directions" Cancers 17, no. 11: 1854. https://doi.org/10.3390/cancers17111854

APA Style

Pérez-Domínguez, F., Quezada-Monrás, C., Cárcamo, L., Muñoz, J. P., & Carrillo-Beltrán, D. (2025). Oncolytic Viruses as a Novel Therapeutic Approach for Colorectal Cancer: Mechanisms, Current Advances, and Future Directions. Cancers, 17(11), 1854. https://doi.org/10.3390/cancers17111854

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