
Precision oncolytic viral therapy in colorectal cancer: Genetic targeting and immune modulation for personalized treatment (Review)
- Authors:
- Published online on: May 6, 2025 https://doi.org/10.3892/ijmm.2025.5545
- Article Number: 104
-
Copyright: © Sultan et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Colorectal cancer (CRC) is one of the most prevalent cancer types worldwide, and was observed to affect 1.9 million individuals in 2020 and led to ~930,000 deaths in the same year, constituting ~10% of cancer-related deaths (1,2). In particular, CRC incidence seems to be increasing in the younger population; early-onset CRC cases have been growing and doubling between 1990 and 2019, which emphasizes the importance of increasing CRC screening and implementing culturally appropriate interventions (3,4). CRC incidence is most elevated in Oceania, including Australia and New Zealand, Europe and in at least some African areas (2).
At present, CRC treatments are based on the disease stage and include surgery, systemic chemotherapy, targeted therapy and radiotherapy. The most distinctive cases of CRC are those that present with hepatic, pulmonary or peritoneal metastases, for which radical surgery, hepatic/pulmonary resections and cytoreductive surgery associated with hyperthermic intraperitoneal chemotherapy are helpful (5-7). Furthermore, bevacizumab and cetuximab as systemic therapies add only 3-6 months of survival to patients with metastatic CRC, and high rates of relapse are still observed (8,9). Single-agent kinase inhibitors selective for pathways such as PI3K/Akt/mTOR have proven suboptimal due to cross-talk between these pathways and poor safety profiles (10). The specificity of traditional treatments, such as chemotherapy and radiotherapy, is very low; they cause significant side effects, and despite treatment with neoadjuvant therapy, there is a chance of recurrence in 54% of patients (11-16). Such issues indicate the importance of a new approach, such as oncolytic viral therapy (OVT). OVs specifically target and destroy tumor cells and stimulate strong antitumor immune responses (17-19). One of the most promising developments is the Food and Drug Administration-approved oncolytic herpes simplex virus type 1 (HSV-1) for treating melanoma. Additionally, there is growing interest in the consistent and clinically significant data reported for the use of vaccinia virus in treating solid carcinomas (20).
The ability to directly lyse tumors and modulate the immune response makes OVT worthwhile. For instance, recent advancements in OVT have shown significant promise in treating CRC. Notably, the oncolytic vaccinia virus, Pexa-Vec, has been evaluated in clinical trials specifically targeting patients with refractory metastatic CRC (21). Early detection through saliva biomarkers can enhance patient selection for OVT, allowing timely intervention and improved treatment success. Understanding oral microbiota may also help optimize OVT protocols for personalized care (22).
An improved matrix stiffness stress in the TME restrains immune cell infiltration and immunotherapy responsiveness; however, it is poorly understood in CRC (23-26). Increasing the understanding of the mechanisms behind extracellular matrix (ECM) biomechanics could improve the effectiveness of immune checkpoint blockade (ICB) therapy and adoptive cell therapy (ACT) (27,28). OVT boosts the upregulation of tumor-associated antigens (TAAs) and cytokines, thereby enhancing the effectiveness of ICB. This is especially valuable in tumors with challenging immune microenvironments, such as microsatellite-stable (MSS) CRC, which typically shows limited response to immunotherapy (28-30). Engineered OVs, such as OH2, demonstrate enhanced specificity and oncolytic potency due to targeted gene manipulations, particularly when compared with unmodified or naturally occurring OVs (29). Together with immunotherapy, OVT has been reported to produce only mild side effects and high rates of CD8+ T-cell infiltration in other cancer types such as melanoma and epithelial tumors, including colorectal, bladder and renal cancer (30,31). Recent research has revealed new approaches to treating CRC by teasing out the potential interactions between OVT, immunotherapy and the mechanical properties of tumors (32). More specifically, it has shed light on several general strategies to possibly optimize ICBs, ACTs and tumor cell vaccines for the treatment of CRC, including the deconstruction of ECM-related hurdles that prevent OVT strategies from reaching target cancer cells and improving the existing OVT strategies tailored to each patient (33).
However, in MSS CRC, the aforementioned improvements have not yielded long-lasting responses, and the highly immunosuppressive TME presents unique obstacles that warrant novel solutions. OVT has been identified as a promising strategy as the oncolytic capabilities are congruent with the immunomodulatory properties. However, there are several obstacles to improve virus delivery, tumor escape mechanisms and the incorporation of OVT into combined treatment strategies (34). The present review fills the gap regarding the lack of a comprehensive synthesis of how recent innovations in genetic engineering, biomarkers and immune modulation are being translated into clinical applications for OVT in CRC. This includes addressing barriers such as tumor heterogeneity, delivery limitations and the integration of OVT with other therapies, which have hindered broader adoption and optimization of this treatment modality. A graphical overview of precision OVT in CRC is provided in Fig. 1. The present review will also address key questions to develop a roadmap for precision OVT in CRC, highlighting technological advancements that could transform the current treatment landscape. The present review discusses not only the application of engineered OVs in the treatment of immune suppression and drug resistance but also underscores the potential of OVT to transform cancer therapy by investigating its value in becoming a fundamental anticancer weapon.
Genetic targeting of OVs in CRC
Strategies for identifying optimal OVs
The selection of optimal OVs relies on key criteria including tumor specificity, replication efficiency and immune-stimulating capabilities. Genetic engineering approaches, such as receptor targeting and promoter-driven tropism, enhance viral selectivity for cancer cells while sparing healthy tissues. Preclinical screening further refines candidates based on safety profiles, oncolytic potency and synergy with existing therapies (35).
Selection criteria for OVs in CRC
For precision OVT in CRC, a virus is designed to infect and kill cancer cells but not healthy cells. One of the criteria for selection is replication preference in tumor cells. For example, TG6002, which was manufactured using the genetically modified vaccinia virus, has selected replication in cancer cells due to viral gene deletion. TG6002 converts 5-fluorocytosine to 5-fluorouracil at the tumor site, improving localized therapeutic effects (36). Similarly, different types of adenoviruses, such as Ad-PE, int and ins-GCV, with fabricated integrin-targeting peptides have improved transduction efficiency and therapeutic activity compared with controls (37).
In addition to replication preference, other critical criteria should be considered when selecting OVs for CRC. First, tumor specificity is paramount; OVs must selectively target cancer cells while sparing normal tissues to minimize off-target effects (38). This can be achieved through genetic modifications that exploit unique TME features, such as hypoxia or altered receptor expression (39). Second, the ability of OVs to modulate the immune system is essential. Beyond direct oncolysis, OVs can stimulate both innate and adaptive immune responses, generating long-term antitumor immunity (40). Third, safety remains a key concern, particularly for systemic delivery, where risks of neutralization by circulating antibodies or off-target infection must be mitigated (41). Finally, the potential for combination therapies, such as pairing OVs with immune checkpoint inhibitors (ICIs) or chemotherapy, should be evaluated to enhance therapeutic efficacy and overcome resistance mechanisms (42).
Mechanisms of action: Oncolysis and immunomodulation
OVs cause tumor cell shedding and immunogenic death, releasing TAAs into the immune system. This immune activation combines synergistically with checkpoint inhibitors or T-cell engagers, as illustrated in preclinical models of CRC (43). The gut microbiome plays a significant role in CRC progression, and its modulation can influence the efficacy of immunotherapies, including OVs. Emerging evidence suggests that specific gut microbiota compositions may predict treatment response and modulate the anticancer immune response, although the exact mechanisms remain an active area of research (44).
Implementation approaches for augmenting effectiveness
OVT has emerged as a promising modality for cancer treatment, particularly in CRC, where OVs selectively infect and lyse tumor cells while sparing normal tissues (45). To enhance the efficacy of OVT, localized delivery systems such as imaging-guided or catheter-based approaches can be employed to achieve optimal viral density within the TME while minimizing systemic toxicity (46). Carrier cells, including mesenchymal stem cells and cytotoxic immune cells such as T cells, have been explored as 'Trojan horse' delivery vehicles to transport OVs to tumor sites, leveraging their innate homing capabilities (47). However, challenges such as rapid clearance by the reticuloendothelial system and neutralization by circulating antibodies highlight the need for innovative strategies, such as engineering OVs to regulate abnormalities in the TME (such as neovascularization and ECM stiffness) or combining them with ICIs to amplify antitumor immunity (21). Recent advances in genetic editing, viral retargeting and nanotechnology platforms further underscore the potential of OVs to overcome barriers to systemic delivery and improve therapeutic outcomes (48). Non-invasive imaging techniques also play a pivotal role in monitoring viral kinetics and ensuring safety during treatment (49). Together, these strategies provide a comprehensive framework for advancing OVT in CRC.
TME remodeling
OVs alter the tumor-associated landscape to enhance the infiltration of immune cells and reduce the presence of immunosuppressive cells. This promotes antitumor immune responses and, more specifically, boosts the effects when used alongside toxic therapies such as chemotherapy or radiotherapy. For example, the engineered vaccinia virus, coxsackievirus B3, plus FOLFOXIRI enhance immunogenicity and CRC survival (50-52).
Enhancing tumor selectivity and safety
The selectivity and safety of OVs are critical to CRC management. Viruses are designed to interact with receptor molecules upregulated in CRC cells, including CD46 and intercellular adhesion molecule-1, improving the affinity and viral replication (53-55). Other sites in CRC tumors, including specific metabolic pathways and immune checkpoint suppression, enhance viral survival and replication within an immunosuppressive environment (56). Genetic modifications make the OVs more selective, allowing them to target tumor cells instead of normal cells, thereby enhancing the therapeutic ratio (21).
Genetic engineering approaches for enhanced specificity in CRC
Genetic engineering approaches have enabled the fine-tuning of OVs to improve tumor specificity, replication efficiency and immunogenicity. For example, talimogene laherparepvec (T-VEC) is an engineered oncolytic HSV-1 designed to preferentially replicate in tumor cells while inducing antitumor immune responses (57).
Enhancing tumor tropism and selectivity
The addition of a heterologous receptor binding domain) to OVs improves their ability to recognize a greater number of receptors. This modification significantly enhances the tropism of these viruses, concentrating on increasing their effectiveness against tumor cells (58). When it comes to therapeutic applications, engineered viruses with an expanded ability to recognize receptors can target and eliminate a wider range of cancer cells, enhancing their capacity to kill tumors that might be less susceptible to viral-mediated cell destruction; it also reduces the dissemination of the OV to the tumor site and enhances the overall effectiveness of the treatment process in combating cancer (59). The regulation of transcription factors has also been studied to selectively enhance viral replication in cancerous tissues (60).
Incorporating immunomodulatory and anti-angiogenic genes
Anti-angiogenic genes can prevent tumor angiogenesis, limiting tumor growth and metastasis (61,62). Additionally, immunomodulatory genes such as interleukin-12 (IL-12) and C-X-C motif chemokine ligand 11 (CXCL11) notably enhance the effect of the immune system on the tumor, improving the therapeutic index of OVs. IL-12 promotes T cells producing interferon-γ and improves the recognition of tumor cells by cytotoxic T lymphocytes (CTLs) and the intrinsic cytotoxic activity of CTLs. Furthermore, CXCL11 is a potent chemokine that induces immune cells, including CTLs and T helper type 1 (Th1), to infiltrate the TME and strengthen the effective antitumor immune response (63). By combining immunomodulatory tactics with anti-angiogenic methods that disrupt tumor angiogenesis and deprive tumors of their blood supply, it is possible to design a cohesive treatment strategy that addresses both tumor growth and enhances the immune defense against cancer (64,65).
CRISPR/Cas9 applications in OVT
CRISPR/Cas9 technology offers a precise and efficient method for editing viral genomes to improve tumor selectivity (66). For example, a tissue-specific HSV-1 has been designed by knocking in the murine IL-12 and CXCL11 cassettes using the ICP34.5 coding region and knocking out the immunomodulatory gene, ICP47. This double alteration improves the selectivity of viral replication in tumor cells and boosts antitumor immunity against CRC. Additionally, the armed HSV-1 features enhanced tumor selective replication and initiates an immune response, resulting in potent antitumor effects. IL-12 favors the Th1 cell development and CTL response, and CXCL11 has been credited for attracting effector T cells and natural killer (NK) cells to the tumor site (67). This coaction greatly optimizes the functional capabilities of immune effector cells and enhances therapeutic efficacy against tumor cells. This approach may be considered a promising strategy for improving OVT in the treatment of CRC (67,68).
Tumor-selective promoters
The incorporation of telomerase and CEA gene promoters into OVs ensures targeted viral replication and therapeutic gene expression exclusively in cancer cells (69). This strategy significantly reduces the likelihood of side effects by sparing normal tissues, thereby improving the safety and efficacy of OVT for CRC (54). As a tumor marker with significantly increased expression in colon cancer, CEA serves as an ideal target for constructing oncolytic adenoviruses (OAVs) (70). These viruses are designed to selectively replicate in and lyse tumor cells while sparing normal tissues. The CEA promoter has been successfully used to regulate the expression of therapeutic genes in OAVs, enhancing their specificity and efficacy against CRC (70,71,72).
Advances in safety and delivery mechanisms
OVs represent a novel antitumor strategy that selectively targets CRC cells while leaving surrounding healthy tissue intact. However, their clinical application is limited by challenges related to the safety and efficacy of delivering OVs to the target tumor tissue. To address these issues, researchers have developed innovative methods. One approach involves engineering OVs to carry natural microRNAs (miRNAs), which are potent regulators of gene expression. By incorporating sequences upregulated in CRC, miRNAs guide OVs specifically toward cancer cells, minimizing side effects and enhancing safety (73). Another promising strategy is the virosomal administration of interferon, where viral particles are encapsulated in liposomes to protect them from destruction by antibodies and complement proteins. The lipid membrane of these liposomes can also be modified with targeting molecules, such as antibodies or ligands, to direct the OVs to the tumor site. This improvement significantly enhances the efficiency and effectiveness of OV delivery (74) Key strategies for engineering OVs are illustrated in Fig. 2. Although those strategies have shown promising outcomes in preclinical studies, a number of improvements and clinical trials are required to confirm their safety and effectiveness for treating CRC (75,76).
Comparison of viral platforms
Several types of OVs for CRC treatment are in use today, which includes the adenoviruses, oHSV2, vaccinia viruses, reoviruses and the Newcastle disease virus. Adenoviruses have been highlighted for their biosafety and potential to selectively infect and lyse cancer cells with therapeutic protein expression, augmented by immunomodulation when combined with checkpoint inhibitors (77). Other OVs include oHSV2, derived from HSV-2, which has exhibited broad potency in a study inducing CRC cell necrosis and enhancing adaptive immune responses, improving the survival time of tumor-bearing mice (78). The vaccinia virus and reovirus exhibit unique characteristics, including a heightened ability to infect tumor tissues and selectively replicate within cancer cells. These properties make them promising candidates for advanced virotherapy strategies aimed at improving tumor targeting and therapeutic outcomes (79,80). Newcastle disease virus is pinpointed for its enhanced effects when applied in conjunction with other methods, thereby proving the capability of OVs in improving the treatment of CRC (81,82).
The benefits of adeno-derived oncolytic therapy in CRC arise from the selective replication of the virus in cancer cells, sparing normal cells. This selectivity is achieved by incorporating cancer cell-specific promoters, such as cyclooxygenase-2, which drive viral replication specifically in cancerous cells. Consequently, OAVs can selectively infect and kill cancer cells, with the relative sparing of normal neighboring tissues (83). Moreover, OAVs may recode the TME and induce an intensive immune reaction, which may cooperate with immunotherapies such as anti-programmed cell death protein 1 (PD-1) (84). Nevertheless, drawbacks include difficulties in applying high local concentrations to distant metastatic tumors since conventional methods may not always be sufficient. In addition, OAVs can prompt immunogenic cell death (ICD), but the immune responses can sometimes hinder their effectiveness. Thus, immune clearance strategies are needed (85).
The safety of HSV, particularly the genetically modified T-VEC, is favorable. T-VEC has been shown to have low toxicity in a clinical trial, and side effects, if any, are controllable. Risks and complications are frequent, but severe adverse events are rare. The HSV OVT has been shown to explicitly target tumor cells without affecting normal cells, which has greatly enhanced the safety of the virus (86). HSV can be administered for patients with advanced cancer, such as CRC (87). The vaccinia virus, including modified strains such as JX-594, also demonstrates fairly good safety indicators. In certain co-housing investigations, it has been administered with fewer reported serious side effects. JX-594 does not cause severe local reactions even when used with other chemotherapeutic agents and is compatible with standard clinical treatment regimens (88,89). Certain research has shown that HSV can produce systemic tumor regression in CRC-bearing animals (90). Current clinical trials are testing its utility when combined with other immunotherapies and when administered as a monotherapy. Initial findings indicate that HSV immunomodulatory capabilities can increase antitumor immunity and have implied the effective treatment of patients with CRC (86). Since the antitumor activity is well-known, the vaccinia virus has shown promising results in phase I clinical trials. For instance, Pexa-Vec has been demonstrated to be effective in decreasing tumor size and improving immune effects on CRC cells. Its potential in stimulating the immune system and directly lysing cancer cells provides enough merit to warrant more developments regarding CRC therapy (88,89).
Continually propelling the research effort on CRC vaccines, Ad5GUCY2C-PADRE is a non-replicating plasmid-based adenoviral vector encoding the GUCY2C antigen fused with the helper T-cell epitope, PADRE. This adenoviral vector has been shown to elicit vigorous cytotoxic and humoral immune responses, directly targeting CRC cells with upregulated GUCY2C expression with little side effects (91). However, it was revealed that antibodies targeting the core adenoviral vector could neutralize adenoviral vectors, which may be an issue in the immunology of adenoviral vaccines (91). Furthermore, although the adenoviral vector is known to elicit preexisting immunity in the patient population, a significant drawback, the oncolytic adenovirus Ad5 [E1-, E2b]-CEA(6D) holds promise due its ability to induce immunogenicity against CEA (92). Intratumor influenza vaccines that increase CD8+ T cells in proficient mismatch repair (pMMR) CRC are being launched to treat patients (NCT04591379) (93). Recombinant adenoviruses can transduce antigens and present oncolytic properties that activate CTLs (94). There is also evidence that the Sendai virus and vesicular stomatitis virus have oncolytic abilities in other preclinical CRC models, preferring to infect and kill cancer cells (95). Table I summarizes various OVs, including vaccinia, adenovirus, HSV and Newcastle disease virus, detailing their engineered constructs, immune responses and efficacy in preclinical CRC models. These viruses demonstrate potent anti-tumor effects through mechanisms such as immune activation, cytokine induction and targeted cytotoxicity.
Targeting key genetic mutations in CRC for enhanced efficacy
The advancement of CRC and the continual alteration of its state depend on mutations disrupting or impairing critical genes that regulate cell turnover. For instance, KRAS mutations result in the constitutive activation of RAS proteins, driving enhanced cell division and survival, thereby promoting tumorigenesis (96). However, studies have also shown that APC mutations affect the regulation of WNT signaling and hence encourage increased division of tumor cells. Both tropism mutation types are highly associated with CRC pathogenesis (97) and the alteration is applicable irrespective of the stage or location of the tumor. Additionally, loss of p53 function through mutational inactivation of the tumor-suppressor gene TP53 hampers facets of the CRC cell's ability to respond to DNA damage, thereby causing the cells to continue to proliferate (96). SMAD4 loss increases tumor invasiveness, enhances metastatic properties, induces chemo-resistance and is associated with a poorer survival, decreasing the overall survival of patients (98,99).
KRAS mutations as targets for OVT
Due to its high incidence rate in CRC (estimated at 45%), KRAS mutations are considered suitable target candidates for OVT (100). These mutations are often associated with resistance to chemotherapy and standard EGFR-directed therapies, the ability of cancer cells to adapt to treatment and a poor prognosis (101). KRAS mutations play a critical role in reprogramming cancer cell metabolism to support rapid growth and survival. These mutations increase the dependency of cancer cells on glutamine, an essential nutrient for energy production and biosynthesis (102). Additionally, KRAS mutations upregulate nuclear factor erythroid 2-related factor 2 (Nrf2) signaling, a pathway that regulates antioxidant responses and metabolic adaptations. By enhancing Nrf2 activity, cancer cells can manage oxidative stress, maintain redox balance and sustain metabolic pathways such as glycolysis and glutaminolysis. Targeting these metabolic vulnerabilities, including glutamine metabolism and Nrf2 signaling, offers potential therapeutic strategies to combat KRAS-driven cancer (103,104). For example, Pelareorep (also known as REOLYSIN), an OVT, can target and kill KRAS-mutated cells, or any tumor cell. Furthermore, these viruses can induce autophagy in cancer cells, which may enhance the therapeutic efficacy of this approach compared with other treatments (105).
APC and TP53 genetic alterations in CRC and OVT
Genetic changes in APC and TP53 are significant in CRC progression. Most of the APC gene abnormalities are primary to CRC tumorigenesis, whereby the overproduction of β-catenin stimulates cell proliferation. During the later phases of multistep carcinogenesis, alterations in TP53 affect critical cellular functions such as the cell cycle and apoptosis and thus promote malignancy (106). These genetic changes elucidate the molecular pathogenesis of CRC and highlight their potential as effective targets for OVT (107). Adenoviruses with deletions in the E1B 55kDa gene are OAVs that selectively infect cancer cells with mutations or deletions in the TP53 gene. The E1B 55kDa protein, under normal circumstances, binds to and inactivates the p53 tumor suppressor to enhance replication. The loss of this gene means OAVs are able to replicate only in cells where p53 function is compromised, an attribute of numerous types of cancer (71,108,109).
BRAF mutations in deficient mismatch repair (dMMR)/microsatellite instability-high (MSI-H) CRC
Another critical target is the BRAFV600E mutation, prevalent in dMMR or MSI-H CRC. While BRAF and MEK inhibitors have shown success in melanoma (105) and non-small cell lung cancer (110), CRC requires additional EGFR blockade due to compensatory EGFR upregulation (111). Integrating targeted therapies for BRAF mutations with OVT has the potential to improve outcomes in these subsets of patients with CRC (112).
HER2 amplification and upregulation
HER2 (also known as erbB2) amplification is frequently detected in CRC and can serve as a prognostic and therapeutic gene; its upregulation is related to tumor progression and poor prognosis, as well as in breast cancer (113-115). The synergy between HER2-targeted treatments with OVT has been noted, especially in HER2-positive CRC subtypes (116).
MSI-H, dMMR and high tumor mutational burden (TMB)
Since MSI-H, dMMR and TMB are related to sensitivity and prognosis, these biomarkers are essential for identifying patients appropriate for immunotherapy. Patients with such features show improved outcomes when administered ICIs common for CRC (117). For instance, it has been identified that CRC tumors with high TMB show increased responsiveness to immunotherapy, as higher mutational loads generate more neoantigens, enhancing the immune system's ability to recognize and attack cancer cells (118). Future research on OVT biomarkers could contribute to developing individualized therapeutic strategies, potentially leading to higher effectiveness in treatment outcomes.
Inflammatory microenvironment and PIK3CA mutations
The pro-inflammatory microenvironment of CRC also yields potential targets for OVT. At present, options for molecular prognostic markers for CRC, including serum IL-6, IL-8, programmed cell death ligand 1 (PD-L1), CEA (71,119), CA19-9 and MMP-9, are still being explored regarding their potential use in immunotherapy (120). Additionally, PIK3CA mutations are associated with the growth and survival of CRC, making them effective targets for OVT. These mutations can improve the vulnerabilities of the tumor to viral infection, which provides a new direction for the treatment of CRC (121).
Immune modulation strategies in OVT for CRC
Mechanisms of immune evasion in CRC
In a number of cases, the CRC TME is marked by the infiltration of immunosuppressive immune cells, including regulatory T cells (Tregs), MDSCs and M2 macrophages. These cells can negatively regulate effector T cells and NK cells, reducing the antitumor immune response (122). Tumor hypoxia, or low oxygen tension, is a recurrent phenomenon in the TME of CRC. Hypoxia can stimulate the release of hypoxia-inducible factor 1-α (HIF-1α), which is involved in the formation of immunosuppressive factors and therapy resistance (123-125). ECM components in the TME can be degraded by enzymes such as MMPs, and this degradation can prevent immune cells from infiltrating the tumor and promote immunosuppressive activity (126). CRC cells can also release PD-L1, which binds to PD-1 on the surface of T cells, thus suppressing their functioning. The mechanism of this interaction is one of the primary significant ways the tumor can escape the immune response in the TME (127,128). The CRC TME can secrete cytokines such as TGF-β, IL-10 and VEGF, which promote cancer progression by supporting tumor growth, facilitating the formation of new blood vessels and enabling immune system evasion. Furthermore, metabolic changes, including glycolysis and glutaminolysis, produce immunosuppressive metabolites and consume nutrients required for immune cells (129).
In CRC, immune cells such as Tregs and MDSCs are particularly relevant for immune evasion and avoidance (130). Tregs can secrete adenosine and transfer cAMP to effector T cells, thereby suppressing their activity. Tregs also outcompete effector T cells for IL-2, metabolize IL-2 into its biologically inactive form, and inhibit the production of IL-2 by dendritic cells, which are crucial for activating effector T cells. Additionally, Tregs can suppress effector T cells through the induction of apoptosis (131). Both Tregs and MDSCs enhance each other's proliferation, creating a symbiotic circuit of immunosuppression within the TME (132,133). Immune cells, such as Tregs and MDSCs, in the microenvironment of CRC decrease the immune response needed for the virus to act upon the cancer cells and destroy them during OVT (134). These cells can help neutralize the virus and suppress the initiation of antitumor immunity, lowering the effectiveness of the therapy (32).
Synergy between OVT and ICIs
ICIs disrupt the binding between the immune checkpoint proteins, such as PD-1/PD-L1, CTLA-4 and sTim-3 (135) and their receptors on the tumor or immune cell surface (134). Cancer cells typically use these interactions to subvert the immune response system in the body. ICIs function by blocking inhibitory pathways, such as PD-1/PD-L1 or CTLA-4 interactions, thereby releasing the suppressive signals on T cells. This enhances T-cell activation and enables them to more effectively recognize and eliminate cancer cells (136).
Combining OVT and ICIs, the immune response to CRC is improved via several mechanisms. OVT employs viruses capable of infecting and killing malignant cells and, at the same time, disseminating TAAs and stimulating antitumor immunity (55). This can enhance the visibility of the tumor to the immune system, thus increasing its immunogenicity (137). Furthermore, OVT can increase the levels of immune checkpoint proteins on cancer cells, allowing ICIs to bind and enhance the immune response. In addition, OVT can alter the tumor-related stroma and make it less immunosuppressive to cancer cells, thus enabling improved immune cell infiltration and effector functions (55). This pro-additive synergy increases the possibility of a more profound and longer lasting immunological response to CRC (137).
Experimental and clinical data have demonstrated the synergism between OVT and anti-PD-1/PD-L1 and CTLA-4 immunotherapies in CRC. Previous in vivo investigations have revealed that OVT may enhance the immunogenicity of tumors, making tumors more conspicuous to the immune system, and raising the levels of immune checkpoint proteins that can be modulated by ICIs (138). According to clinical trials, these combinations have resulted in objective responses and disease control with reasonable toxicity (139,140). CTLA-4 and PD-1/PD-L1, together with the blockade, upregulate T cell activation and treatment effectiveness, improving the anti-tumor results (141,142).
To maintain the effectiveness of OVT, studies have highlighted the importance of engineering OVs with immune-modulatory genes. For instance, incorporating genes for cytokines such as granulocyte-macrophage colony-stimulating factor (GM-CSF) or checkpoint inhibitors such as anti-PD-1 can enhance antitumor immunity and extend the duration of viral activity (143). Recent findings suggest that OVs equipped with costimulatory molecules, such as VALO-D102 encoding CD40L and OX40L, can significantly improve tumor growth control and boost the infiltration of tumor-specific CD8+ effector T cells, as demonstrated in melanoma models (54). Enhancing the efficacy of OVT can also be achieved by targeting the TME through strategies such as remodeling the ECM or inhibiting immunosuppressive pathways, particularly those involving PI3Kγ in macrophages (144). Additionally, combining OVT with targeted therapies, such as anti-angiogenic agents (including regorafenib) (145) or EGFR inhibitors (including cetuximab), enhances viral delivery and counteracts resistance mechanisms in the TME (146). The synergistic mechanism of OVT and ICIs is depicted in Fig. 3.
CRC develops resistance to ICIs through mechanisms such as altered PI3K/AKT/mTOR signaling, often due to PIK3CA mutations, and interactions within an immunosuppressive TME (147). To overcome this resistance, combining OVT with PI3K-γ inhibitors such as copanlisib or histone deacetylase inhibitors (HDIs) can effectively suppress oncogenic pathways, enhance viral replication and synergistically improve T-cell infiltration alongside ICIs (148). Nevertheless, there are problems with this approach. Despite ICI therapy, 30-50% of patients with MSI-H CRC develop drug resistance, while single-agent ICIs have been reported to have little impact in patients with pMMR or MSS metastatic CRC (149,150). Furthermore, the effectiveness and safety of PD-1/PD-L1 and CTLA-4 ICIs on advanced CRC are still inconclusive in clinical studies, and the outcomes are paradoxical (139,151). By contrast, previous findings suggest that NK-1R antagonists induce apoptosis in CRC cells through endoplasmic reticulum stress and calcium release, activating the PERK/eIF2α/ATF4/CHOP pathway, which enhances chemotherapy sensitivity and reveals potential biomarkers and therapeutic targets (152). Moreover, the heterogeneity of CRC or the TME can modulate the response to therapeutic intervention, and there may be shortcomings in identifying patients likely to benefit from this combined treatment strategy (153).
Enhancing viral immunogenicity for an improved immune response
Viral immunogenicity is essential in the mechanism of action of OVT for CRC since it establishes the capacity of the virus to elicit an immune response against cancer cells (154). When OVs infect and lyse cancer cells, they display antigens that the immune system can see and thus stimulate T cells and other immune factors (155). This process is termed ICD and is crucial for priming and reinforcing antitumor responses (57). OVs can be engineered genetically to boost this immunogenicity. These changes may concern the deletion of viral genes that inhibit the host immune response, the addition of other genes that encode immune-stimulating products, such as cytokines (IL-12 and IL-15) or antibodies (such as against PD-1), and the presence of molecules enhancing viral replication in tumor cells and their spread (156,157). For example, oncolytic HSV-1 containing the humanized anti-PD-1 antibody gene has been reported to strengthen immune response and suppress tumor progression in models of CRC (158). Moreover, cytokine preconditioning parental cells can increase the release or the antitumor activity of exosomes originating from such parent cells, thus strengthening the immune response (159). These genetic changes seek to transform the 'cold', difficult-to-treat tumors that are less sensitive to ICIs and enhance their ability to stimulate an immune response (154).
Immune adjuvants play a pivotal role in enhancing the effectiveness of OVT in CRC by amplifying the immune response. They stimulate the innate immune system, promoting the recruitment and maturation of antigen-presenting cells, which is crucial for initiating adaptive immunity against cancer cells (160). For example, T-VEC produces GM-CSF, which improves antigen-presenting cell function and elicits a copious T-cell response at the tumor site (161,162). However, genetic modifications can be made to increase immunogenicity by endowing OVs with 'micrometals', CD40L and OX40L, which have been found to improve tumor control and CD8+ effector T cell functions (163).
New strategies to enhance viral immunogenicity for OVT include the use of OVs with cytokine-gene or immune-checkpoint inhibitors (54). For instance, VALO-D102, an adenovirus that encodes CD40L and OX40L, has a more robust efficacy on tumor suppression if used with an anti-PD-1 antibody (163). Additionally, there is evidence of the employment of bispecific T-cell activators and other antibody formats to increase oncolytic viral productivity (164,165). These strategies aim to counteract the suppressive immunological characteristics of tumors in OVT, thereby enhancing therapeutic effects on CRC.
Furthermore, the emerging therapeutic applications of exosomes are gaining attention due to their ability to modulate epithelial-mesenchymal transition (EMT) and other tumor-promoting processes. Recent advances highlight their potential as effective delivery platforms for miRNAs or drugs to reverse EMT or enhance treatment efficacy (166). By integrating exosome technology with existing OVT strategies there is potential for improved treatment outcomes in CRC. Additionally, exosomes play a crucial role as essential mediators of intercellular communication, regulating EMT. This enables tumor cells to acquire invasive and metastatic properties, which poses challenges for effective treatment strategies (167).
Role of the TME in modulating OVT efficacy
The TME in CRC is replete with immunosuppressive cells such as Tregs and tumor-associated macrophages (TAMs) that form a massive hurdle to immune effector cells (161). OVT can change this TME by using ICD, which will release other TAAs and recruit immune cells, including macrophages and T cells. These immune cells play a crucial role as they can recognize both the viral infection and tumor antigens, thereby generating a significantly stronger antitumor immune response (30). Macrophages, in particular, when polarized to an M1 phenotype, are known to support the antitumor immune response (162), whereas T cells, especially CD8 cytotoxic T-lymphocytes, directly kill cancer cells (168). Recent developments in OVT have focused on the modification of viral specificity and tropism and the modification of OVs to provide immune-stimulatory molecules to increase viral immunogenicity (169). For instance, viruses can be equipped with cytokines such as GM-CSF, which enhance the antitumor function of lymphocytes, or chemokines, which attract more immune cells into the TME (170). Additionally, incorporating ICIs into the virus can counteract the immunosuppressive state of the TME and enhance the immune targeting of CRC (32).
Biomarker-driven combination therapies in precision OVT for personalized CRC management
Chemotherapy and radiation synergy with OVT in CRC
Chemotherapy and radiation therapy are known treatments for the therapy of CRC with the overall goal of therapeutic mitigation of tumors and the associated symptoms (21). These drugs act by causing permanent defects in the DNA of actively dividing cancer cells, inhibiting growth. Nevertheless, these treatments also target healthy cells and, therefore, cause side effects and may not help all patients, particularly those diagnosed with MSS CRC, since this disease typically does not respond well to immunotherapy (171-173). In this context, the combination of OVT with chemotherapy or radiotherapy has emerged as a promising strategy in CRC treatment. Such combinations aim to enhance therapeutic efficacy by leveraging complementary mechanisms, including the induction of ICD and the modulation of the TME (21). The current research states that integrating these therapies exploits similar pathways, including ICD. OVT, chemotherapy and radiotherapy all release danger associated molecular patterns, which enhance innate and adaptive immunity (174). This improves the overall tumouricidal activity and seems beneficial in permitting lower doses of cytotoxic agents to be deployed in combination with enzymes, thereby diminishing toxicity. For instance, genetically modified oncolytic viruses can be programmed to express enzymes such as cytosine deaminase to transform prodrugs, such as 5-fluorocytosine, into toxic agents wherever the virus is replicating but minimize the side effect on other organs of the body (175).
Targeted therapies and OVT: Promising drug combinations
Targeted therapy for CRC is aimed at proteins or molecules that directly impact cancer rather than harming all dividing cells, such as in chemotherapy. These therapies target critical signaling pathways misregulated in CRC, including SHH/Gli, Wnt/β-cat, TGF-β/SMA, EGFR and Notch. For example, vismodegib, a hedgehog (Hh) inhibitor, suppresses the Hh pathway by binding to the Smoothened receptor and thus induces apoptosis in colon cancer cells (176). Similarly, Albring et al (177) have demonstrated the ability of berberine and its derivatives to modulate the Wnt/β-catenin signaling cascade.
The combination of targeted agents can improve the outcomes of OVT in managing CRC. When combined with targeted drugs that modulate molecular pathways, OVT generates enhanced toxicity for cancer cells while preserving standard tissue tolerance. In this regard, the use of monoclonal antibodies to EGFR, such as cetuximab and panitumumab, improves the response of patients with CRC to OVT by increasing the sensitivity of cancer cells to the OV (178). The same drugs can positively affect tumor vessels; for instance, bevacizumab can prevent the formation of short and irregular vessels typical for tumor-related vasculature, increasing the effectiveness of OV (179). This implies that there is an opportunity to improve the effectiveness of OVT using targeted therapies that make cancer cells sensitive to the treatment while at the same time lowering the general toxicity. When used together, there is the potential for drug candidates such as anti-angiogenic agents for CRC and EGFR inhibitors to enhance the effectiveness of OVT by making cancer cells more sensitive to treatment while reducing overall toxicity. Certain anti-angiogenic agents, such as regorafenib (an oral multikinase inhibitor), combined with immunotherapies such as nivolumab, have been shown to be beneficial in patients with CRC, especially those with MSS tumors, which are less sensitive to immunotherapies (180). This combination demonstrated an overall response rate of ~44% in the REGONIVO study (181).
Bevacizumab is an anti-angiogenic agent recently licensed in CRC and has demonstrated increased overall survival and progression-free survival (PFS) in metastatic CRC (182). Furthermore, research has shown that combining chemotherapy with bevacizumab in patients with metastatic CRC is influenced by PD-L1 expression, suggesting that PD-L1 status plays a crucial role in the effectiveness of this therapeutic approach (183,184). Cetuximab target the EGFR signaling cascade, which is frequently mutated in CRC, and have shown efficacy in KRAS wild-type metastatic CRC (185). These potent agents have been evaluated in several clinical trials for the first-line, second-line or third-line treatment of metastatic CRCs.
OVT combined with targeted therapies in CRC also poses certain issues, such as resistance, toxicity and dosage. Crossover and bypass can result in innate and acquired resistance due to existing interactions between pathways (186). One major issue with combining these therapies is that, as the number of treatment sessions increases, the rate of complications also rises (187). The dosage needs to be carefully fine-tuned to allow for effective targeted therapy while avoiding toxicity due to the druggability of targets in the TME, which have not been entirely elucidated (188). Some factors that should be weighed include prophylactic considerations, cost/benefit considerations, patient selection and follow-up, given the inter-compliant variation results (189,190).
Table II summarizes various therapeutic agents, including targeted therapies, ICIs and chemotherapy drugs, and their mechanisms of action in enhancing OVT efficacy. These combinations leverage synergistic effects to improve viral replication, immune activation and tumor-specific cytotoxicity.
Novel biomarker-based strategies for personalized OVT
Biomarkers are critical in precision medicine in CRC due to their guiding role in identifying various aspects of the tumor molecular profile that define its management. The American Society of Clinical Oncology, European Society for Medical Oncology and National Comprehensive Cancer Network recognize mutations within these genes as essential pharmacogenomic biomarkers (191-193). Notably, BRAF mutation is considered to be adverse for survival, highlighting the role of biomarkers as a guide for the selection of treatments based on tumor characteristics (194). In particular, PD-L1, TMB and MSI are established as possible indicators of ICIs. PD-L1 is currently used to predict response to anti-PD-1/anti-PD-L1 therapy, while TMB and MSI have some predictive roles in numerous cancer types, including colon cancer (195,196). Another molecular biomarker associated with improved clinical outcomes in ICI therapy for patients with CRC is MSI status, particularly in tumors that are MSI-H or dMMR (197). However, the predictive value of PD-L1 has been criticized since some patients with low or undetectable PD-L1 expression respond to this targeted therapy (198). This indicates that there is a requirement for improved predictive biomarkers for ICI therapy.
Abnormalities in the RAS, BRAF and EGFR genes are considered valid prognostic indicators in CRC (199). More specifically, anti-EGFR receptor therapies fail to show efficiency when KRAS mutations exist in the malignant cells. Detecting individuals with these genetic changes is recommended as these treatments can be unhelpful and expensive (200). In locally advanced rectal cancer, patients harboring wild-type KRAS have improved pathological complete response rates with cetuximab, a targeted agent (201). These biomarkers may be utilized in individualized treatment planning, which will benefit patients by reducing toxicities and misguided treatment costs.
Overcoming challenges in precision OVT for CRC
Addressing virus delivery and tumor targeting limitations
Delivering OVs into CRC tumors is challenging since CRC tumors are developed and heterogeneous. Although the concept of systemic delivery of OVs is thought-provoking due to its capability to target disseminated metastases, the approach is also faced with challenges such as the short biological half-life OVs in the circulatory system and the poor ability to target as well as sediment at the tumor site, thereby reducing the possibility of delivering OVs to the metastatic deposits. Intratumoral delivery, while less versatile as it is restricted to accessible tumors, can garner higher viral shed-load and produce vigorous local antitumor effects (42). Furthermore, other factors, such as the secretion of stromal cell-derived factor-1, can also worsen the mobilization nadir as well as greatly enhance the number of circulating progenitor cells, but the responses are typically relatively weak at untreated distal sites (202).
To enhance the efficiency of systemic delivery, a new method involving nanoparticle encapsulation and engineered vesicles has been proposed. Nanoparticles improve the resistance, solubility, crossover ability and circulation time of OVs; in other words, they make viral vectors safer and more effective in clinical applications (203). A recent innovation in nanoparticle construction has incorporated more hierarchical structures, bio-feedback guidance components and conjugates, which make OVs reach tumor-specific locale more efficiently (204,205). For example, biomimetic nanoparticles coated by cell membranes can use immune evasion, prolonged circulation and disease-specific targeting to enhance the oncolytic effect of OVs at the tumor site (206). The systemic and intratumoral delivery methods are compared in Fig. 4.
In summary, different delivery systems of OVs possess certain limitations, namely, immune clearance and insufficient tumor accumulation, which can be resolved by nanoparticle encapsulation and engineered vesicles. Modifications to nanoparticles, such as surface functionalization with targeting ligands or the incorporation of stimuli-responsive elements, have been shown to significantly enhance their ability to deliver OVs specifically to tumor lesions while minimizing off-target effects (207). These strategies will likely enhance the clinical use of OVT in CRC.
Resistance mechanisms and OVT
Understanding the mechanisms of cancer drug resistance can guide the optimization of existing targeted therapies, identify therapeutic targets valuable to discovering new and improved agents and form the basis of therapeutic advances in cancer treatment (208). As with other cancer treatments, CRC cells may interfere with or become resistant to OVT via several processes. This mechanism involves the activation of the PI3K-γ/AKT pathway in tumor-associated myeloid cells (TAMCs), which fosters an immunosuppressive environment by downregulating cytotoxic CD8+ T lymphocytes and inhibiting their activity (209-212). Additionally, CRC cells may have upregulated expression of immune checkpoint molecules, such as PD-L1, and thereby inhibit T cell function. The TME is also involved and the mechanical property of the tumor tissue and matrix stiffness hampers immune cell infiltration, which is essential for the effectiveness of OVT (213,214).
Understanding mechanisms of resistance is needed to outline how to counter this type of adaptive resistance. However, when the current OVTs are combined with players such as HDIs, the viral replication, oncolytic activity and recognition of NK cell activating ligands and TAAs are all boosted (215-217). Inhibition of PI3K-γ can overcome immuno-suppression in TAMCs and improve the therapeutic effect of OVT (218). The aberrant activation of the PI3K/AKT/mTOR signaling pathway in CRC promotes therapeutic resistance by enhancing cancer cell survival, metabolic reprogramming and immune evasion through mechanisms such as PTEN loss or PIK3CA mutations (219). Targeting this pathway with PI3K inhibitors (such as alpelisib and copanlisib) or mTOR inhibitors (such as everolimus) may counteract resistance by suppressing downstream oncogenic signaling and restoring apoptotic sensitivity (220). Combining these inhibitors with immune checkpoint blockers (such as pembrolizumab and nivolumab) could further enhance efficacy by reversing PI3K/AKT-mediated immunosuppression and reinvigorating antitumor immunity (221). Additionally, epigenetic therapies targeting DNA methyltransferases or histone deacetylases may reactivate tumor suppressor genes silenced by PI3K/AKT-driven hypermethylation, while miRNA-based strategies (such as with miR-34a) could downregulate pathway components (222). Thus, it can be stated that using selective targeting of specific resistance mechanisms and combining OVT with other treatment approaches, it is possible to effectively overcome the adaptive resistance of CRC cells to OVT.
Managing safety and toxicity in precision OVT
Potential issues connected with OVT for colon cancer are related to uncontrolled proliferation of the virus, the reaction of the immune defense mechanisms, other types of infections as well as unpredictable side effects. Furthermore, combined interventions of OVT with other treatments may increase such possibilities, posing threats to the patient's well-being and thus mandating close supervision (175). Further studies are being conducted to determine the correct dosage of OVs. Specific indicators, such as biomarkers or patient characteristics, are used to select the most suitable candidates for OVT and to anticipate possible side effects linked to this innovative treatment protocol (175).
Measures that help to avoid contact with the OV, including strict measures for transportation and administration of the OV, are necessary to minimize the potential exposure. Moreover, the training of clinicians and information for patients on how to take care of the area where injections are administered is also required. Adverse effects surveillance in patients administered OVT faces challenges such as under-reporting and bias in data collection, which can obscure the true incidence of adverse events. Additionally, distinguishing between genuine product-related effects and coincidental events complicates accurate surveillance and assessment (57). As effective as OVT may be for precision therapy in CRC, it must also be regulated for safety due to possible unintended consequences and viral transmission. Genetic engineering for specificity, containing the virus or making the treatment highly supervised, and monitoring the treatment closely and strictly can guarantee the use of OVs in cancer therapy.
Clinical trial insights and challenges
Challenges
The gaps in the patient selection criteria for current OVT clinical trials for CRC are multifaceted. One significant gap is the historical underrepresentation of specific populations in clinical trials, limiting the generalizability of results and affecting the diversity of eligible participants. Specifically, the strict eligibility criteria often exclude a large proportion of patients, particularly those who are older, female or non-Latinx Black, and those with a lower socioeconomic status, thus limiting the pool of eligible candidates. The lack of broadened eligibility criteria may potentially exclude patients who could benefit from the treatment (223). Furthermore, the challenges in validating combination treatment strategies using patient-derived organoids highlight the need for more refined and representative models to guide patient selection in clinical trials (153). Addressing these gaps is crucial for optimizing patient selection, ensuring more inclusive trials and improving the clinical applicability of OVT in CRC.
In OVT clinical trials for CRC, selecting predictive endpoints is crucial for determining success. According to the latest research, endpoints such as PFS and immune reactivity are considered highly predictive of success (224). PFS is a critical secondary endpoint that measures the time from trial randomization to the occurrence of disease progression or death, and it is a standard measure of the effectiveness of a treatment in delaying disease progression. However, immune reactivity is assessed through the objective response rate, which measures the proportion of patients achieving a complete or partial response, and the disease control rate, which includes patients with stable disease for a defined period (225). These endpoints are integral to evaluating the immunotherapeutic effects of OVT.
Clinical trials
Recent progress in OVT for CRC has occurred through efforts to enhance the viral agents' specificity and carcinoid efficacy by gene engineering and distinct delivery system methods in the last decade. New strategies include elements of viral genomes that promote antitumor immune responses and the integration of OVT with ICIs to overcome TME (226). To illustrate this progress, Table III summarizes ongoing and completed clinical trials investigating OVT in CRC, highlighting the use of engineered viruses, immune-modulating therapies and innovative delivery systems.
Regulatory, ethical and practical aspects of precision OVT
Precision oncology, mainly with the use of virtual trials and OVT, opens new possibilities in clinical practice. However, their implementation should also consider compliance with regulatory requirements concerning the safety and efficacy of applied approaches, data correctness and ethical standards. Additionally, patient privacy, data security measures and consent are essential for successfully approving and developing precision OVT (227). The application of precision to OVT for patients has notable challenges, such as restraints in safety and efficacy, Good Manufacturing Practice standards and patient consent. Long-term monitoring is necessary for continuous post-market surveillance (226). The germline genetic profile, comorbidities and age-specific mutational processes should inform the creation of these regimens. The modified form of OVT can increase the efficacy of treatments due to the targeted receptor contributing to immune evasion, thus leading to improved patient results (228). Researchers hope that adjusting the therapy according to the properties of the tumor will enhance the antitumor immune response and overall survival. Additionally, the synergistic effects of OVT with other immunotherapies may be apparent, improving the overall therapeutic advantage (55).
The use of genetically modified OVs introduces unique ethical challenges, particularly concerning patient safety and potential long-term risks. While these viruses are engineered to selectively target cancer cells and minimize off-target effects, unintended consequences such as viral shedding, transmission to non-target tissues or unforeseen immune reactions could pose significant risks (229). Ensuring rigorous preclinical testing, transparent informed consent processes and long-term post-market surveillance is critical to mitigating these concerns. Additionally, equitable access to precision OVT must be considered, as high costs and complex logistical requirements may limit availability in low-resource settings. Addressing these ethical dimensions will not only safeguard patients but also foster public trust in this transformative therapeutic approach.
Conclusions
OVT represents a new paradigm in the field of CRC treatment, as it has the potential to circumvent the drawbacks associated with conventional therapies while utilizing targeting and immune-stimulating properties. During the last decade, genetic engineering and biomarkers have opened the door for targeted treatment options, a style that remains novel in CRC due to intersquamous tumor heterogeneity. Additionally, combining ICIs with other conventional methods such as chemotherapy and targeted therapy produces synergistic effects on treatment outcomes.
Nevertheless, the OV technique has certain limitations, which have posed the following challenges: How best to deliver viruses, what strategies to counter viral resistance and the question of safety when the viruses will be used clinically. These include integrating new, efficient delivery systems (nanoparticle encapsulation) and creating new biomarkers. Although OVT holds significant promise, its implementation in low- and middle-income countries is hindered by notable challenges, such as inadequate healthcare infrastructure, prohibitive treatment costs and limited access to advanced therapeutic technologies. To bridge these gaps, it is crucial to develop innovative strategies, including cost-effective viral production techniques, simplified and scalable delivery systems as well as fostering international collaborations aimed at promoting equitable access to OVT-based treatments. Such efforts could pave the way for broader global adoption of this transformative cancer therapy.
Future directions for enhancing translation involve incorporating artificial intelligence into these findings and utilizing well-established clinical trials for patient selection. By addressing these issues through a behavioral/precision OVT approach, CRC treatment may undergo a radical transformation, establishing a new standard in the oncology process. With ongoing research, this innovative treatment modality could eventually transition from experimental to routine practice, offering millions of patients a renewed chance at life worldwide.
Availability of data and materials
Not applicable.
Authors' contributions
Conceptualization was conducted by MHS and YW; MHS, XJ and YW collected and organized the relevant literature; YX and QZ conducted a systematic review of the literature to identify key themes and trends; MHS, QZ, YX, XJ and YW critically evaluated the sources and synthesized the findings; project administration was conducted by YW; the original draft was written by MHS; reviewing and editing the manuscript was performed by MHS, YW and XJ. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Acknowledgments
Not applicable.
Funding
This work was supported by the Natural Science Foundation of Zhejiang Province (grant no. LGF22C010005) and the Science Foundation of Zhejiang Sci-Tech University (grant no. 18042291-Y).
References
Fadlallah H, El Masri J, Fakhereddine H, Youssef J, Chemaly C, Doughan S and Abou-Kheir W: Colorectal cancer: Recent advances in management and treatment. World J Clin Oncol. 15:1136–1156. 2024. View Article : Google Scholar : PubMed/NCBI | |
Morgan E, Arnold M, Gini A, Lorenzoni V, Cabasag CJ, Laversanne M, Vignat J, Ferlay J, Murphy N and Bray F: Global burden of colorectal cancer in 2020 and 2040: Incidence and mortality estimates from GLOBOCAN. Gut. 72:338–344. 2023. View Article : Google Scholar : PubMed/NCBI | |
Pan H, Zhao Z, Deng Y, Zheng Z, Huang Y, Huang S and Chi P: The global, regional, and national early-onset colorectal cancer burden and trends from 1990 to 2019: Results from the global burden of disease study 2019. BMC Public Health. 22:18962022. View Article : Google Scholar : PubMed/NCBI | |
Maida M, Dahiya DS, Shah YR, Tiwari A, Gopakumar H, Vohra I, Khan A, Jaber F, Ramai D and Facciorusso A: Screening and surveillance of colorectal cancer: A review of the literature. Cancers (Basel). 16:27462024. View Article : Google Scholar : PubMed/NCBI | |
Nordlinger B, Sorbye H, Glimelius B, Poston GJ, Schlag PM, Rougier P, Bechstein WO, Primrose JN, Walpole ET, Finch-Jones M, et al: Perioperative FOLFOX4 chemotherapy and surgery versus surgery alone for resectable liver metastases from colorectal cancer (EORTC 40983): Long-term results of a randomised, controlled, phase 3 trial. Lancet Oncol. 14:1208–1215. 2013. View Article : Google Scholar : PubMed/NCBI | |
Zabaleta J, Iida T, Falcoz PE, Salah S, Jarabo JR, Correa AM, Zampino MG, Matsui T, Cho S, Ardissone F, et al: Individual data meta-analysis for the study of survival after pulmonary metastasectomy in colorectal cancer patients: A history of resected liver metastases worsens the prognosis. Eur J Surg Oncol. 44:1006–1012. 2018. View Article : Google Scholar : PubMed/NCBI | |
Verwaal VJ, van Ruth S, de Bree E, van Slooten GW, van Tinteren H, Boot H and Zoetmulder FAN: Randomized trial of cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy and palliative surgery in patients with peritoneal carcinomatosis of colorectal cancer. J Clin Oncol. 21:3737–3743. 2003. View Article : Google Scholar : PubMed/NCBI | |
Cercek A, Lumish M, Sinopoli J, Weiss J, Shia J, Lamendola-Essel M, El Dika IH, Segal N, Shcherba M, Sugarman R, et al: PD-1 blockade in mismatch repair-deficient, locally advanced rectal cancer. N Engl J Med. 386:2363–2376. 2022. View Article : Google Scholar : PubMed/NCBI | |
Chalabi M, Verschoor YL, van den Berg J, Sikorska K, Beets G, Lent AV, Grootscholten MC, Aalbers A, Buller N, Marsman H, et al: LBA7 Neoadjuvant immune checkpoint inhibition in locally advanced MMR-deficient colon cancer: The NICHE-2 study. Ann Oncol. 33(Suppl 7): S13892022. View Article : Google Scholar | |
Yang J, Nie J, Ma X, Wei Y, Peng Y and Wei X: Targeting PI3K in cancer: Mechanisms and advances in clinical trials. Mol Cancer. 18:262019. View Article : Google Scholar : PubMed/NCBI | |
Javid H, Hashemian P, Yazdani S, Sharbaf Mashhad A and Karimi-Shahri M: The role of heat shock proteins in metastatic colorectal cancer: A review. J Cell Biochem. 123:1704–1735. 2022. View Article : Google Scholar : PubMed/NCBI | |
Golestaneh M, Firoozrai M, Javid H and Hashemy SI: The substance P/neurokinin-1 receptor signaling pathway mediates metastasis in human colorectal SW480 cancer cells. Mol Biol Rep. 49:4893–4900. 2022. View Article : Google Scholar : PubMed/NCBI | |
Aapro M, Jelkmann W, Constantinescu SN and Leyland-Jones B: Effects of erythropoietin receptors and erythropoiesis-stimulating agents on disease progression in cancer. Br J Cancer. 106:1249–1258. 2012. View Article : Google Scholar : PubMed/NCBI | |
González-Perera I, Gutiérrez-Nicolás F, Nazco-Casariego GJ, Ramos-Díaz R, Hernández-San Gil R, Pérez-Pérez JA, González García J and González De La Fuente GA: 5-fluorouracil toxicity in the treatment of colon cancer associated with the genetic polymorphism 2846 A>G (rs67376798). J Oncol Pharm Pract. 23:396–398. 2017. View Article : Google Scholar | |
Javid H, Karimi-Shahri M, Khorramdel M, Mashhad AS, Tabrizi AT, Sathyapalan T, Afshari AR and Sahebkar A: Probiotics as an adjuvant for management of gastrointestinal cancers through their anti-inflammatory effects: A mechanistic review. Curr Med Chem. 30:390–406. 2023. View Article : Google Scholar | |
Ogura A, Konishi T, Cunningham C, Garcia-Aguilar J, Iversen H, Toda S, Lee IK, Lee HX, Uehara K, Lee P, et al: Neoadjuvant (Chemo)radiotherapy with total mesorectal excision only is not sufficient to prevent lateral local recurrence in enlarged nodes: Results of the multicenter lateral node study of patients with low cT3/4 rectal cancer. J Clin Oncol. 37:33–43. 2019. View Article : Google Scholar : | |
Ye LY, Li YS, Ge T, Liu LC, Si JX, Yang X, Fan WJ, Liu XZ, Zhang YN, Wang JW, et al: Engineered luminescent oncolytic vaccinia virus activation of photodynamic-immune combination therapy for colorectal cancer. Adv Healthc Mater. 13:e23041362024. View Article : Google Scholar : PubMed/NCBI | |
Ruan Z, Chi J, Kong Y, Li C, Ruan X, Zhou X, Chen Y, Li Y and Luo Z: Natural oncolysis of enterovirus 71 in antitumor therapy of colorectal cancer. Adv Biol (Weinh). 7:e22003362023. View Article : Google Scholar : PubMed/NCBI | |
Komant S, Wang J, Favis N, Alex C, Evans DH, Noyce RS and Baldwin TA: Oncolytic vaccinia virus as a precision cancer vaccine platform. bioRxiv. 2024.08.16.608170. 2024. | |
Nasar RT, Uche IK and Kousoulas KG: Targeting cancers with oHSV-based oncolytic viral immunotherapy. Curr Issues Mol Biol. 46:5582–5594. 2024. View Article : Google Scholar : PubMed/NCBI | |
Ren Y, Miao JM, Wang YY, Fan Z, Kong XB, Yang L and Cheng G: Oncolytic viruses combined with immune checkpoint therapy for colorectal cancer is a promising treatment option. Front Immunol. 13:9617962022. View Article : Google Scholar : PubMed/NCBI | |
Wang XL, Xu HW and Liu NN: Oral microbiota: A new insight into cancer progression, diagnosis and treatment. Phenomics. 3:535–547. 2023. View Article : Google Scholar : PubMed/NCBI | |
Mai Z, Lin Y, Lin P, Zhao X and Cui L: Modulating extracellular matrix stiffness: A strategic approach to boost cancer immunotherapy. Cell Death Dis. 15:3072024. View Article : Google Scholar : PubMed/NCBI | |
Kalli M, Poskus MD, Stylianopoulos T and Zervantonakis IK: Beyond matrix stiffness: Targeting force-induced cancer drug resistance. Trends Cancer. 9:937–954. 2023. View Article : Google Scholar : PubMed/NCBI | |
Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, Zhang Z, Yang S and Xiao M: Extracellular matrix remodeling in tumor progression and immune escape: From mechanisms to treatments. Mol Cancer. 22:482023. View Article : Google Scholar : PubMed/NCBI | |
Ahmed H, Mahmud AR, Siddiquee MF, Shahriar A, Biswas P, Shimul MEK, Ahmed SZ, Ema TI, Rahman N, Khan MA, et al: Role of T cells in cancer immunotherapy: Opportunities and challenges. Cancer Pathog Ther. 1:116–126. 2022. View Article : Google Scholar : PubMed/NCBI | |
Noro J, Vilaça-Faria H, Reis RL and Pirraco RP: Extracellular matrix-derived materials for tissue engineering and regenerative medicine: A journey from isolation to characterization and application. Bioact Mater. 34:494–519. 2024.PubMed/NCBI | |
Sorokin M, Zolotovskaia M, Nikitin D, Suntsova M, Poddubskaya E, Glusker A, Garazha A, Moisseev A, Li X, Sekacheva M, et al: Personalized targeted therapy prescription in colorectal cancer using algorithmic analysis of RNA sequencing data. BMC Cancer. 22:11132022. View Article : Google Scholar : PubMed/NCBI | |
Hu H, Zhang S, Cai L, Duan H, Li Y, Yang J, Wang Y and Liu B, Dong S, Fang Z and Liu B: A novel cocktail therapy based on quintuplet combination of oncolytic herpes simplex virus-2 vectors armed with interleukin-12, interleukin-15, GM-CSF, PD1v, and IL-7 x CCL19 results in enhanced antitumor efficacy. Virol J. 19:742022. View Article : Google Scholar | |
Ribas A, Dummer R, Puzanov I, VanderWalde A, Andtbacka RHI, Michielin O, Olszanski AJ, Malvehy J, Cebon J, Fernandez E, et al: Oncolytic virotherapy promotes intratumoral T cell infiltration and improves Anti-PD-1 immunotherapy. Cell. 170:1109–1119.e10. 2017. View Article : Google Scholar : PubMed/NCBI | |
Garcia-Carbonero R, Salazar R, Duran I, Osman-Garcia I, Paz-Ares L, Bozada JM, Boni V, Blanc C, Seymour L, Beadle J, et al: Phase 1 study of intravenous administration of the chimeric adenovirus enadenotucirev in patients undergoing primary tumor resection. J Immunother Cancer. 5:712017. View Article : Google Scholar : PubMed/NCBI | |
Zhang H, Ren Y, Wang F, Tu X, Tong Z, Liu L, Zheng Y, Zhao P, Cheng J, Li J, et al: The long-term effectiveness and mechanism of oncolytic virotherapy combined with anti-PD-L1 antibody in colorectal cancer patient. Cancer Gene Ther. 31:1412–1426. 2024. View Article : Google Scholar : PubMed/NCBI | |
Berkey SE, Thorne SH and Bartlett DL: Oncolytic virotherapy and the tumor microenvironment. Tumor Immune Microenvironment in Cancer Progression and Cancer Therapy. Kalinski P: Springer International Publishing; Cham: pp. 157–172. 2017 | |
Cai L, Chen A and Tang D: A new strategy for immunotherapy of microsatellite-stable (MSS)-type advanced colorectal cancer: Multi-pathway combination therapy with PD-1/PD-L1 inhibitors. Immunology. 173:209–226. 2024. View Article : Google Scholar : PubMed/NCBI | |
Hajeri PB, Sharma NS and Yamamoto M: Oncolytic adenoviruses: Strategies for improved targeting and specificity. Cancers (Basel). 12:15042020. View Article : Google Scholar : PubMed/NCBI | |
Samson A, Smolenschi C, Cassier P, Patel JV, Hammond C, Kurzawa M, Sainte-Croix S, West E, Sadoun A and Bendjama K: Abstract CT190: Oncolytic virus TG6002 safety and activity after intrahepatic artery administration in patients with liver-dominant metastatic colorectal cancer. Cancer Res. 83(8 Suppl): CT1902023. View Article : Google Scholar | |
Lavilla-Alonso S, Bauerschmitz G, Abo-Ramadan U, Halavaara J, Escutenaire S, Diaconu I, Tatlisumak T, Kanerva A, Hemminki A and Pesonen S: Adenoviruses with an αvβ integrin targeting moiety in the fiber shaft or the HI-loop increase tumor specificity without compromising antitumor efficacy in magnetic resonance imaging of colorectal cancer metastases. J Transl Med. 8:802010. View Article : Google Scholar | |
Naumenko VA, Stepanenko AA, Lipatova AV, Vishnevskiy DA and Chekhonin VP: Infection of non-cancer cells: A barrier or support for oncolytic virotherapy? Mol Ther Oncolytics. 24:663–682. 2022. View Article : Google Scholar : PubMed/NCBI | |
Chen G, Wu K, Li H, Xia D and He T: Role of hypoxia in the tumor microenvironment and targeted therapy. Front Oncol. 12:9616372022. View Article : Google Scholar : PubMed/NCBI | |
Kingsak M, Meethong T, Jongkhumkrong J, Cai L and Wang Q: Therapeutic potential of oncolytic viruses in the era of precision oncology. Biomater Transl. 4:67–84. 2023. | |
Peng Z, Kalim M and Lu Y: Improving systemic delivery of oncolytic virus by cellular carriers. Cancer Biol Med. 21:1104–1119. 2025. View Article : Google Scholar : PubMed/NCBI | |
Chen L, Zuo M, Zhou Q and Wang Y: Oncolytic virotherapy in cancer treatment: Challenges and optimization prospects. Front Immunol. 14:13088902023. View Article : Google Scholar | |
Crupi MJF, Taha Z, Janssen TJA, Petryk J, Boulton S, Alluqmani N, Jirovec A, Kassas O, Khan ST, Vallati S, et al: Oncolytic virus driven T-cell-based combination immunotherapy platform for colorectal cancer. Front Immunol. 13:10292692022. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Wang G, Qin L, Hu B and Li J: Intestinal microbiota modulates the antitumor effect of oncolytic virus vaccines in colorectal cancer. Dig Dis Sci. 69:1228–1241. 2024. View Article : Google Scholar : PubMed/NCBI | |
Kontermann RE, Ungerechts G and Nettelbeck DM: Viro-antibody therapy: Engineering oncolytic viruses for genetic delivery of diverse antibody-based biotherapeutics. MAbs. 13:19824472021. View Article : Google Scholar : PubMed/NCBI | |
Zheng M, Huang J, Tong A and Yang H: Oncolytic viruses for cancer therapy: Barriers and recent advances. Mol Ther Oncolytics. 15:234–247. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhu J, Ma J, Huang M, Deng H and Shi G: Emerging delivery strategy for oncolytic virotherapy. Mol Ther Oncol. 32:2008092024. View Article : Google Scholar : PubMed/NCBI | |
Song Z, Tao Y, Liu Y and Li J: Advances in delivery systems for CRISPR/Cas-mediated cancer treatment: A focus on viral vectors and extracellular vesicles. Front Immunol. 15:14444372024. View Article : Google Scholar : PubMed/NCBI | |
Mainenti PP, Stanzione A, Guarino S, Romeo V, Ugga L, Romano F, Storto G, Maurea S and Brunetti A: Colorectal cancer: Parametric evaluation of morphological, functional and molecular tomographic imaging. World J Gastroenterol. 25:5233–5256. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhang T, Jiang S, Zhang L, Liu Y, Zheng H, Zhao H, Du S, Xu Y and Lu X: A bibliometric analysis of oncolytic virotherapy combined with immunotherapy. Hum Vaccin Immunother. 20:24066212024. View Article : Google Scholar : PubMed/NCBI | |
Chai C, Zhang J, Zhou Y, Yin H, Zhang F, Diao Y, Zan X, Ma Y, Wang Y, Wu Y and Wang W: The effects of oncolytic pseudorabies virus vaccine strain inhibited the growth of colorectal cancer HCT-8 cells in vitro and in vivo. Animals (Basel). 12:24162022. View Article : Google Scholar : PubMed/NCBI | |
Girod M, Geisler A, Hinze L, Elsner L, Dieringer B, Beling A, Kurreck J and Fechner H: Combination of FOLFOXIRI drugs with oncolytic coxsackie B3 virus PD-H synergistically induces oncolysis in the refractory colorectal cancer cell line Colo320. Int J Mol Sci. 25:56182024. View Article : Google Scholar : PubMed/NCBI | |
Enow JA, Sheikh HI and Rahman MM: Tumor tropism of DNA viruses for oncolytic virotherapy. Viruses. 15:22622023. View Article : Google Scholar : PubMed/NCBI | |
Tian Y, Xie D and Yang L: Engineering strategies to enhance oncolytic viruses in cancer immunotherapy. Signal Transduct Target Ther. 7:1172022. View Article : Google Scholar : PubMed/NCBI | |
Lin D, Shen Y and Liang T: Oncolytic virotherapy: Basic principles, recent advances and future directions. Signal Transduct Target Ther. 8:1562023. View Article : Google Scholar : PubMed/NCBI | |
Robilotti E, Zeitouni NC and Orloff M: Biosafety and biohazard considerations of HSV-1-based oncolytic viral immunotherapy. Front Mol Biosci. 10:11783822023. View Article : Google Scholar : PubMed/NCBI | |
Shalhout SZ, Miller DM, Emerick KS and Kaufman HL: Therapy with oncolytic viruses: progress and challenges. Nat Rev Clin Oncol. 20:160–177. 2023. View Article : Google Scholar : PubMed/NCBI | |
Alkayyal AA, Darwish M, Ajina R, Alabbas SY, Alotaibi MA, Alsofyani A, Bokhamseen M, Hakami M, Albaradie OA, Moglan AM, et al: Repurposing the oncolytic virus VSV∆51M as a COVID-19 vaccine. Front Bioeng Biotechnol. 11:11508922023. View Article : Google Scholar | |
Ranki T, Särkioja M, Hakkarainen T, von Smitten K, Kanerva A and Hemminki A: Systemic efficacy of oncolytic adenoviruses in imagable orthotopic models of hormone refractory metastatic breast cancer. Int J Cancer. 121:165–174. 2007. View Article : Google Scholar : PubMed/NCBI | |
Niemann J and Kühnel F: Oncolytic viruses: Adenoviruses. Virus Genes. 53:700–706. 2017. View Article : Google Scholar : PubMed/NCBI | |
Bolyard C, Yoo JY, Wang PY, Saini U, Rath KS, Cripe TP, Zhang J, Selvendiran K and Kaur B: Doxorubicin synergizes with 34.5ENVE to enhance antitumor efficacy against metastatic ovarian cancer. Clin Cancer Res. 20:6479–6494. 2014. View Article : Google Scholar : PubMed/NCBI | |
Meisen WH, Dubin S, Sizemore ST, Mathsyaraja H, Thies K, Lehman NL, Boyer P, Jaime-Ramirez AC, Elder JB, Powell K, et al: Changes in BAI1 and nestin expression are prognostic indicators for survival and metastases in breast cancer and provide opportunities for dual targeted therapies. Mol Cancer Ther. 14:307–314. 2015. View Article : Google Scholar : | |
Medrano RFV, Hunger A, Mendonça SA, Barbuto JAM and Strauss BE: Immunomodulatory and antitumor effects of type I interferons and their application in cancer therapy. Oncotarget. 8:71249–71284. 2017. View Article : Google Scholar : PubMed/NCBI | |
Hirvinen M, Rajecki M, Kapanen M, Parviainen S, Rouvinen-Lagerström N, Diaconu I, Nokisalmi P, Tenhunen M, Hemminki A and Cerullo V: Immunological effects of a tumor necrosis factor alpha-armed oncolytic adenovirus. Hum Gene Ther. 26:134–144. 2015. View Article : Google Scholar : PubMed/NCBI | |
Zhang B, Wang X and Cheng P: Remodeling of tumor immune microenvironment by oncolytic viruses. Front Oncol. 10:5613722021. View Article : Google Scholar : PubMed/NCBI | |
Hazafa A, Mumtaz M, Farooq MF, Bilal S, Chaudhry SN, Firdous M, Naeem H, Ullah MO, Yameen M, Mukhtiar MS and Zafar F: CRISPR/Cas9: A powerful genome editing technique for the treatment of cancer cells with present challenges and future directions. Life Sci. 263:1185252020. View Article : Google Scholar : PubMed/NCBI | |
Zhang N, Li J, Yu J, Wan Y, Zhang C, Zhang H and Cao Y: Construction of an IL12 and CXCL11 armed oncolytic herpes simplex virus using the CRISPR/Cas9 system for colon cancer treatment. Virus Res. 323:1989792023. View Article : Google Scholar : | |
Wang L, Chen Y, Liu X, Li Z and Dai X: The application of CRISPR/Cas9 technology for cancer immunotherapy: Current status and problems. Front Oncol. 11:7049992022. View Article : Google Scholar : PubMed/NCBI | |
Ilkow CS and Bell JC: Optimizing oncolytic virus design: A 'Swiss army knife' approach to create a systemically delivered therapeutic. Signal Transduct Target Ther. 9:822024. View Article : Google Scholar | |
Xiao B, Ying C, Chen Y, Huang F, Wang B, Fang H, Guo W, Liu T, Zhou X, Huang B, et al: Doxorubicin hydrochloride enhanced antitumour effect of CEA-regulated oncolytic virotherapy in live cancer cells and a mouse model. J Cell Mol Med. 24:13431–13439. 2020. View Article : Google Scholar : PubMed/NCBI | |
Xiao B, Zhang L, Liu H, Fang H, Wang C, Huang B, Liu X, Zhou X and Wang Y: Oncolytic adenovirus cd55-smad4 suppresses cell proliferation, metastasis, and tumor stemness in colorectal cancer by regulating wnt/β-catenin signaling pathway. Biomedicines. 8:5932020. View Article : Google Scholar | |
Zhang Y, Ye M, Huang F, Wang S, Wang H, Mou X and Wang Y: Oncolytic adenovirus expressing ST13 increases antitumor effect of tumor necrosis factor-related apoptosis-inducing ligand against pancreatic ductal adenocarcinoma. Hum Gene Ther. 31:891–903. 2020. View Article : Google Scholar : PubMed/NCBI | |
Luo Q, Song H, Deng X, Li J, Jian W, Zhao J, Zheng X, Basnet S, Ge H, Daniel T, et al: A triple-regulated oncolytic adenovirus carrying MicroRNA-143 exhibits potent antitumor efficacy in colorectal cancer. Mol Ther Oncolytics. 16:219–229. 2020. View Article : Google Scholar : PubMed/NCBI | |
Toropko M, Chuvpilo S and Karabelsky A: miRNA-mediated mechanisms in the generation of effective and safe oncolytic viruses. Pharmaceutics. 16:9862024. View Article : Google Scholar : PubMed/NCBI | |
Yu B, Kang J, Lei H, Li Z, Yang H and Zhang M: Immunotherapy for colorectal cancer. Front Immunol. 15:14333152024. View Article : Google Scholar : PubMed/NCBI | |
Zhao JL, Lin BL, Luo C, Yi YL, Huang P, Chen Y, Zhao S, Huang ZJ, Ma XY and Huang L: Challenges and strategies toward oncolytic virotherapy for leptomeningeal metastasis. J Transl Med. 22:10002024. View Article : Google Scholar : PubMed/NCBI | |
Oronsky B, Gastman B, Conley AP, Reid C, Caroen S and Reid T: Oncolytic adenoviruses: The cold war against cancer finally turns hot. Cancers (Basel). 14:47012022. View Article : Google Scholar : PubMed/NCBI | |
Yin L, Zhao C, Han J, Li Z, Zhen Y, Xiao R, Xu Z and Sun Y: Antitumor effects of oncolytic herpes simplex virus type 2 against colorectal cancer in vitro and in vivo. Ther Clin Risk Manag. 13:117–130. 2017. View Article : Google Scholar : PubMed/NCBI | |
Gong J, Sachdev E, Mita AC and Mita MM: Clinical development of reovirus for cancer therapy: An oncolytic virus with immune-mediated antitumor activity. World J Methodol. 6:25–42. 2016. View Article : Google Scholar : PubMed/NCBI | |
Li M, Zhang M, Ye Q, Liu Y and Qian W: Preclinical and clinical trials of oncolytic vaccinia virus in cancer immunotherapy: A comprehensive review. Cancer Biol Med. 20:646–661. 2023.PubMed/NCBI | |
Davis D and Lahiri SS: Application of oncolytic viruses for cure of colorectal cancer. Cancer Res J. 3:76–93. 2015. View Article : Google Scholar | |
Kana SI and Essani K: Immuno-oncolytic viruses: Emerging options in the treatment of colorectal cancer. Mol Diagn Ther. 25:301–313. 2021. View Article : Google Scholar : PubMed/NCBI | |
Larocca CJ, Wilber S, Jensen E, Steer C and Davydova J: 428 Development of an Oncolytic Adenovirus to Treat Metastatic Colorectal Cancer. J Clin Transl Sci. 7(Suppl 1): 1282023. View Article : Google Scholar : | |
Huang L, Zhao H, Shan M, Chen H, Xu B, He Y, Zhao Y, Liu Z, Chen J and Xu Q: Oncolytic adenovirus H101 ameliorate the efficacy of anti-PD-1 monotherapy in colorectal cancer. Cancer Med. 11:4575–4587. 2022. View Article : Google Scholar : PubMed/NCBI | |
Li YS, Ye LY, Luo YX, Zheng WJ, Si JX, Yang X, Zhang YN, Wang SB, Zou H, Jin KT, et al: Tumor-targeted delivery of copper-manganese biomineralized oncolytic adenovirus for colorectal cancer immunotherapy. Acta Biomater. 179:243–255. 2024. View Article : Google Scholar : PubMed/NCBI | |
Scanlan H, Coffman Z, Bettencourt J, Shipley T and Bramblett DE: Herpes simplex virus 1 as an oncolytic viral therapy for refractory cancers. Front Oncol. 12:9400192022. View Article : Google Scholar : PubMed/NCBI | |
Ferrucci PF, Pala L, Conforti F and Cocorocchio E: Talimogene laherparepvec (T-VEC): An intralesional cancer immunotherapy for advanced melanoma. Cancers (Basel). 13:13832021. View Article : Google Scholar : PubMed/NCBI | |
Park SH, Breitbach CJ, Lee J, Park JO, Lim HY, Kang WK, Moon A, Mun JH, Sommermann EM, Maruri Avidal L, et al: Phase 1b trial of biweekly intravenous pexa-Vec (JX-594), an oncolytic and immunotherapeutic vaccinia virus in colorectal cancer. Mol Ther. 23:1532–1540. 2015. View Article : Google Scholar : PubMed/NCBI | |
Harrop R, Drury N, Shingler W, Chikoti P, Redchenko I, Carroll MW, Kingsman SM, Naylor S, Melcher A, Nicholls J, et al: Vaccination of colorectal cancer patients with modified vaccinia ankara encoding the tumor antigen 5T4 (TroVax) given alongside chemotherapy induces potent immune responses. Clin Cancer Res. 13:4487–4494. 2007. View Article : Google Scholar : PubMed/NCBI | |
Koch MS, Lawler SE and Chiocca EA: HSV-1 oncolytic viruses from bench to bedside: An overview of current clinical trials. Cancers (Basel). 12:35142020. View Article : Google Scholar : PubMed/NCBI | |
Snook AE, Baybutt TR, Xiang B, Abraham TS, Flickinger JC Jr, Hyslop T, Zhan T, Kraft WK, Sato T and Waldman SA: Split tolerance permits safe Ad5-GUCY2C-PADRE vaccine-induced T-cell responses in colon cancer patients. J Immunother Cancer. 7:1042019. View Article : Google Scholar : PubMed/NCBI | |
Balint JP, Gabitzsch ES, Rice A, Latchman Y, Xu Y, Messerschmidt GL, Chaudhry A, Morse MA and Jones FR: Extended evaluation of a phase 1/2 trial on dosing, safety, immunogenicity, and overall survival after immunizations with an advanced-generation Ad5 [E1-, E2b-]-CEA(6D) vaccine in late-stage colorectal cancer. Cancer Immunol Immunother. 64:977–987. 2015. View Article : Google Scholar : PubMed/NCBI | |
Gögenur M, Balsevicius L, Bulut M, Colak N, Justesen TF, Fiehn AK, Jensen MB, Høst-Rasmussen K, Cappelen B, Gaggar S, et al: Neoadjuvant intratumoral influenza vaccine treatment in patients with proficient mismatch repair colorectal cancer leads to increased tumor infiltration of CD8+ T cells and upregulation of PD-L1: A phase 1/2 clinical trial. J Immunother Cancer. 11:e0067742023. View Article : Google Scholar : PubMed/NCBI | |
Jooss K, Ertl HC and Wilson JM: Cytotoxic T-lymphocyte target proteins and their major histocompatibility complex class I restriction in response to adenovirus vectors delivered to mouse liver. J Virol. 72:2945–2954. 1998. View Article : Google Scholar : PubMed/NCBI | |
Xue Y, Ruan Y, Wang Y, Xiao P and Xu J: Signaling pathways in liver cancer: Pathogenesis and targeted therapy. Mol Biomed. 5:202024. View Article : Google Scholar : PubMed/NCBI | |
Calistri D, Rengucci C, Seymour I, Lattuneddu A, Polifemo AM, Monti F, Saragoni L and Amadori D: Mutation analysis of p53, K-ras, and BRAF genes in colorectal cancer progression. J Cell Physiol. 204:484–488. 2005. View Article : Google Scholar : PubMed/NCBI | |
Fodde R: The APC gene in colorectal cancer. Eur J Cancer. 38:867–871. 2002. View Article : Google Scholar : PubMed/NCBI | |
Mehrvarz Sarshekeh A, Advani S, Overman MJ, Manyam G, Kee BK, Fogelman DR, Dasari A, Raghav K, Vilar E, Manuel S, et al: Association of SMAD4 mutation with patient demographics, tumor characteristics, and clinical outcomes in colorectal cancer. PLoS One. 12:e01733452017. View Article : Google Scholar : PubMed/NCBI | |
Chung Y, Wi YC, Kim Y, Bang SS, Yang JH, Jang K, Min KW and Paik SS: The Smad4/PTEN expression pattern predicts clinical outcomes in colorectal adenocarcinoma. J Pathol Transl Med. 52:37–44. 2018. View Article : Google Scholar : | |
Vaughn CP, ZoBell SD, Furtado LV, Baker CL and Samowitz WS: Frequency of KRAS, BRAF, and NRAS mutations in colorectal cancer. Genes Chromosomes Cancer. 50:307–312. 2011. View Article : Google Scholar : PubMed/NCBI | |
Überall I, Kolář Z, Trojanec R, Berkovcová J and Hajdúch M: The status and role of ErbB receptors in human cancer. Exp Mol Pathol. 84:79–89. 2008. View Article : Google Scholar : PubMed/NCBI | |
Kerk SA, Papagiannakopoulos T, Shah YM and Lyssiotis CA: Metabolic networks in mutant KRAS-driven tumours: Tissue specificities and the microenvironment. Nat Rev Cancer. 21:510–525. 2021. View Article : Google Scholar : PubMed/NCBI | |
Schmitz KJ, Ademi C, Bertram S, Schmid KW and Baba HA: Prognostic relevance of autophagy-related markers LC3, p62/sequestosome 1, Beclin-1 and ULK1 in colorectal cancer patients with respect to KRAS mutational status. World J Surg Oncol. 14:1892016. View Article : Google Scholar : PubMed/NCBI | |
Jiffry J, Thavornwatanayong T, Rao D, Fogel EJ, Saytoo D, Nahata R, Guzik H, Chaudhary I, Augustine T, Goel S and Maitra R: Oncolytic reovirus (pelareorep) induces autophagy in KRAS-mutated colorectal cancer. Clin Cancer Res. 27:865–876. 2021. View Article : Google Scholar : | |
Goel S, Ocean AJ, Parakrama RY, Ghalib MH, Chaudhary I, Shah U, Viswanathan S, Kharkwal H, Coffey M and Maitra R: Elucidation of pelareorep pharmacodynamics in A phase I trial in patients with KRAS-mutated colorectal cancer. Mol Cancer Ther. 19:1148–1156. 2020. View Article : Google Scholar : PubMed/NCBI | |
Su Z, El Hage M and Linnebacher M: Mutation patterns in colorectal cancer and their relationship with prognosis. Heliyon. 10:e365502024. View Article : Google Scholar : PubMed/NCBI | |
Sommariva S, Caviglia G, Ravera S, Frassoni F, Benvenuto F, Tortolina L, Castagnino N, Parodi S and Piana M: Computational quantification of global effects induced by mutations and drugs in signaling networks of colorectal cancer cells. Sci Rep. 11:196022021. View Article : Google Scholar : PubMed/NCBI | |
Zeng M, Zhang W, Li Y and Yu L: Harnessing adenovirus in cancer immunotherapy: Evoking cellular immunity and targeting delivery in cell-specific manner. Biomark Res. 12:362024. View Article : Google Scholar : PubMed/NCBI | |
Liu CC, Liu JH, Wu SC, Yen CC, Chen WS and Tsai YC: A novel E1B-55kD-deleted oncolytic adenovirus carrying mutant KRAS-regulated hdm2 transgene exerts specific antitumor efficacy on colorectal cancer cells. Mol Cancer Ther. 9:450–460. 2010. View Article : Google Scholar : PubMed/NCBI | |
Planchard D, Besse B, Groen HJM, Souquet PJ, Quoix E, Baik CS, Barlesi F, Kim TM, Mazieres J, Novello S, et al: Dabrafenib plus trametinib in patients with previously treated BRAF(V600E)-mutant metastatic non-small cell lung cancer: An open-label, multicentre phase 2 trial. Lancet Oncol. 17:984–993. 2016. View Article : Google Scholar : PubMed/NCBI | |
Kopetz S, Grothey A, Yaeger R, Van Cutsem E, Desai J, Yoshino T, Wasan H, Ciardiello F, Loupakis F, Hong YS, et al: Encorafenib, binimetinib, and cetuximab in BRAF V600E-mutated colorectal cancer. N Engl J Med. 381:1632–1643. 2019. View Article : Google Scholar : PubMed/NCBI | |
Kopetz S, Murphy DA, Pu J, Ciardiello F, Desai J, Van Cutsem E, Wasan HS, Yoshino T, Saffari H, Zhang X, et al: Molecular profiling of BRAF-V600E-mutant metastatic colorectal cancer in the phase 3 BEACON CRC trial. Nat Med. 30:3261–3271. 2024. View Article : Google Scholar : PubMed/NCBI | |
Ivanova M, Venetis K, Guerini-Rocco E, Bottiglieri L, Mastropasqua MG, Garrone O, Fusco N and Ghidini M: HER2 in metastatic colorectal cancer: Pathology, somatic alterations, and perspectives for novel therapeutic schemes. Life (Basel). 12:14032022.PubMed/NCBI | |
Yonesaka K, Zejnullahu K, Okamoto I, Satoh T, Cappuzzo F, Souglakos J, Ercan D, Rogers A, Roncalli M, Takeda M, et al: Activation of ERBB2 signaling causes resistance to the EGFR-directed therapeutic antibody cetuximab. Sci Transl Med. 3:99ra86. 2011. View Article : Google Scholar : PubMed/NCBI | |
Takegawa N and Yonesaka K: HER2 as an emerging oncotarget for colorectal cancer treatment after failure of anti-epidermal growth factor receptor therapy. Clin Colorectal Cancer. 16:247–251. 2017. View Article : Google Scholar : PubMed/NCBI | |
Liu H, Zhou D, Liu D, Xu X, Zhang K, Hu R, Xiong P, Wang C, Zeng X, Wang L and Zhang S: Synergistic antitumor activity between HER2 antibody-drug conjugate and chemotherapy for treating advanced colorectal cancer. Cell Death Dis. 15:1872024. View Article : Google Scholar : PubMed/NCBI | |
Li K, Luo H, Huang L, Luo H and Zhu X: Microsatellite instability: A review of what the oncologist should know. Cancer Cell Int. 20:162020. View Article : Google Scholar : PubMed/NCBI | |
Schumacher TN and Schreiber RD: Neoantigens in cancer immunotherapy. Science. 348:69–74. 2015. View Article : Google Scholar : PubMed/NCBI | |
Xiao B, Qin Y, Ying C, Ma B, Wang B, Long F, Wang R, Fang L and Wang Y: Combination of oncolytic adenovirus and luteolin exerts synergistic antitumor effects in colorectal cancer cells and a mouse model. Mol Med Rep. 16:9375–9382. 2017. View Article : Google Scholar : PubMed/NCBI | |
Calu V, Ionescu A, Stanca L, Geicu OI, Iordache F, Pisoschi AM, Serban AI and Bilteanu L: Key biomarkers within the colorectal cancer related inflammatory microenvironment. Sci Rep. 11:79402021. View Article : Google Scholar : PubMed/NCBI | |
Mao C, Yang ZY, Hu XF, Chen Q and Tang JL: PIK3CA exon 20 mutations as a potential biomarker for resistance to anti-EGFR monoclonal antibodies in KRAS wild-type metastatic colorectal cancer: A systematic review and meta-analysis. Ann Oncol. 23:1518–1525. 2012. View Article : Google Scholar | |
Wu X, Yan H, Qiu M, Qu X, Wang J, Xu S, Zheng Y, Ge M, Yan L and Liang L: Comprehensive characterization of tumor microenvironment in colorectal cancer via molecular analysis. Elife. 12:e860322023. View Article : Google Scholar : PubMed/NCBI | |
Hirata E and Sahai E: Tumor microenvironment and differential responses to therapy. Cold Spring Harb Perspect Med. 7:a0267812017. View Article : Google Scholar : PubMed/NCBI | |
Jia Q, Wang A, Yuan Y, Zhu B and Long H: Heterogeneity of the tumor immune microenvironment and its clinical relevance. Exp Hematol Oncol. 11:242022. View Article : Google Scholar : PubMed/NCBI | |
Zhang L, Yu X, Zheng L, Zhang Y, Li Y, Fang Q, Gao R, Kang B, Zhang Q, Huang JY, et al: Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature. 564:268–272. 2018. View Article : Google Scholar : PubMed/NCBI | |
Karlsson S and Nyström H: The extracellular matrix in colorectal cancer and its metastatic settling-Alterations and biological implications. Crit Rev Oncol Hematol. 175:1037122022. View Article : Google Scholar | |
Becht E, de Reyniès A, Giraldo NA, Pilati C, Buttard B, Lacroix L, Selves J, Sautès-Fridman C, Laurent-Puig P and Fridman WH: Immune and stromal classification of colorectal cancer is associated with molecular subtypes and relevant for precision immunotherapy. Clin Cancer Res. 22:4057–4066. 2016. View Article : Google Scholar : PubMed/NCBI | |
Chen DS and Mellman I: Elements of cancer immunity and the cancer-immune set point. Nature. 541:321–330. 2017. View Article : Google Scholar : PubMed/NCBI | |
Jin MZ and Jin WL: The updated landscape of tumor microenvironment and drug repurposing. Signal Transduct Target Ther. 5:1662020. View Article : Google Scholar : PubMed/NCBI | |
Ohta A, Kini R, Ohta A, Subramanian M, Madasu M and Sitkovsky M: The development and immunosuppressive functions of CD4(+) CD25(+) FoxP3(+) regulatory T cells are under influence of the adenosine-A2A adenosine receptor pathway. Front Immunol. 3:1902012. View Article : Google Scholar : PubMed/NCBI | |
Schmidt A, Oberle N and Krammer PH: Molecular mechanisms of treg-mediated T cell suppression. Front Immunol. 3:512012. View Article : Google Scholar : PubMed/NCBI | |
Ghiringhelli F, Puig PE, Roux S, Roux S, Parcellier A, Schmitt E, Solary E, Kroemer G, Martin F, Chauffert B and Zitvogel L: Tumor cells convert immature myeloid dendritic cells into TGF-beta-secreting cells inducing CD4+CD25+ regulatory T cell proliferation. J Exp Med. 202:919–929. 2005. View Article : Google Scholar : PubMed/NCBI | |
Morello S, Pinto A, Blandizzi C and Antonioli L: Myeloid cells in the tumor microenvironment: Role of adenosine. Oncoimmunology. 5:e11085152016. View Article : Google Scholar : PubMed/NCBI | |
Aristin Revilla S, Kranenburg O and Coffer PJ: Colorectal cancer-infiltrating regulatory T cells: Functional heterogeneity, metabolic adaptation, and therapeutic targeting. Front Immunol. 13:9035642022. View Article : Google Scholar : PubMed/NCBI | |
Hong J, Chen X, Chen L, Wang Y, Huang B and Fang H: Clinical value of combined detection of serum sTim-3 and CEA or CA19-9 for postoperative recurrence of colorectal cancer diagnosis. Cancer Manag Res. 15:563–572. 2023. View Article : Google Scholar : PubMed/NCBI | |
He X and Xu C: Immune checkpoint signaling and cancer immunotherapy. Cell Res. 30:660–669. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH and Diaz LA Jr: Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat Rev Gastroenterol Hepatol. 16:361–375. 2019. View Article : Google Scholar : PubMed/NCBI | |
Dong H, Li M, Yang C, Wei W, He X, Cheng G and Wang S: Combination therapy with oncolytic viruses and immune checkpoint inhibitors in head and neck squamous cell carcinomas: An approach of complementary advantages. Cancer Cell Int. 23:12023. View Article : Google Scholar : PubMed/NCBI | |
Song D, Hou S, Ma N, Yan B and Gao J: Efficacy and safety of PD-1/PD-L1 and CTLA-4 immune checkpoint inhibitors in the treatment of advanced colorectal cancer: A systematic review and meta-analysis. Front Immunol. 15:14853032024. View Article : Google Scholar : PubMed/NCBI | |
Chen EX, Jonker DJ, Loree JM, Kennecke HF, Berry SR, Couture F, Ahmad CE, Goffin JR, Kavan P, Harb M, et al: Effect of combined immune checkpoint inhibition vs best supportive care alone in patients with advanced colorectal cancer: The Canadian cancer trials group CO.26 study. JAMA Oncol. 6:831–838. 2020. View Article : Google Scholar : PubMed/NCBI | |
Overman MJ, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, Morse MA, Van Cutsem E, McDermott R, Hill A, et al: Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J Clin Oncol. 36:773–779. 2018. View Article : Google Scholar : PubMed/NCBI | |
Thibaudin M, Fumet JD, Chibaudel B, Bennouna J, Borg C, Martin-Babau J, Cohen R, Fonck M, Taieb J, Limagne E, et al: First-line durvalumab and tremelimumab with chemotherapy in RAS-mutated metastatic colorectal cancer: a phase 1b/2 trial. Nat Med. 29:2087–2098. 2023. View Article : Google Scholar : PubMed/NCBI | |
Shi T, Song X, Wang Y, Liu F and Wei J: Combining oncolytic viruses with cancer immunotherapy: Establishing a new generation of cancer treatment. Front Immunol. 11:6832020. View Article : Google Scholar : PubMed/NCBI | |
Xu H, Russell SN, Steiner K, O'Neill E and Jones KI: Targeting PI3K-gamma in myeloid driven tumour immune suppression: A systematic review and meta-analysis of the preclinical literature. Cancer Immunol Immunother. 73:2042024. View Article : Google Scholar : PubMed/NCBI | |
Arai H, Battaglin F, Wang J, Lo JH, Soni S, Zhang W and Lenz HJ: Molecular insight of regorafenib treatment for colorectal cancer. Cancer Treat Rev. 81:1019122019. View Article : Google Scholar : PubMed/NCBI | |
Zhou J, Ji Q and Li Q: Resistance to anti-EGFR therapies in metastatic colorectal cancer: Underlying mechanisms and reversal strategies. J Exp Clin Cancer Res. 40:3282021. View Article : Google Scholar : PubMed/NCBI | |
Leiphrakpam PD and Are C: PI3K/Akt/mTOR signaling pathway as a target for colorectal cancer treatment. Int J Mol Sci. 25:31782024. View Article : Google Scholar : PubMed/NCBI | |
Stefani C, Miricescu D, Stanescu-Spinu II, Nica RI, Greabu M, Totan AR and Jinga M: Growth factors, PI3K/AKT/mTOR and MAPK signaling pathways in colorectal cancer pathogenesis: Where are we now? Int J Mol Sci. 22:102602021. View Article : Google Scholar : PubMed/NCBI | |
Diaz LA Jr, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt C, Smith D, Garcia-Carbonero R, Benavides M, Gibbs P, et al: Pembrolizumab versus chemotherapy for microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer (KEYNOTE-177): Final analysis of a randomised, open-label, phase 3 study. Lancet Oncol. 23:659–670. 2022. View Article : Google Scholar : PubMed/NCBI | |
Kalyan A, Kircher S, Shah H, Mulcahy M and Benson A: Updates on immunotherapy for colorectal cancer. J Gastrointest Oncol. 9:160–169. 2018. View Article : Google Scholar : PubMed/NCBI | |
Huo G, Liu W, Zhang S and Chen P: Efficacy of PD-1/PD-L1 plus CTLA-4 inhibitors in solid tumors based on clinical characteristics: a meta-analysis. Immunotherapy. 15:189–207. 2023. View Article : Google Scholar : PubMed/NCBI | |
Shi Y, Wang X, Meng Y, Ma J, Zhang Q, Shao G, Wang L, Cheng X, Hong X, Wang Y, et al: A novel mechanism of endoplasmic reticulum stress- and c-Myc-degradation-mediated therapeutic benefits of antineurokinin-1 receptor drugs in colorectal cancer. Adv Sci (Weinh). 8:e21019362021. View Article : Google Scholar : PubMed/NCBI | |
Benboubker V, Ramzy GM, Jacobs S and Nowak-Sliwinska P: Challenges in validation of combination treatment strategies for CRC using patient-derived organoids. J Exp Clin Cancer Res. 43:2592024. View Article : Google Scholar : PubMed/NCBI | |
Sillo TO, Beggs AD, Morton DG and Middleton G: Mechanisms of immunogenicity in colorectal cancer. Br J Surg. 106:1283–1297. 2019. View Article : Google Scholar : PubMed/NCBI | |
Hu Z, Li Y, Yang J, Liu J, Zhou H, Sun C, Tian C, Zhu C, Shao M, Wang S, et al: 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. 14:236712024. View Article : Google Scholar | |
Bommareddy PK, Shettigar M and Kaufman HL: Integrating oncolytic viruses in combination cancer immunotherapy. Nat Rev Immunol. 18:498–513. 2018. View Article : Google Scholar : PubMed/NCBI | |
Liu Z, Ravindranathan R, Kalinski P, Guo ZS and Bartlett DL: Rational combination of oncolytic vaccinia virus and PD-L1 blockade works synergistically to enhance therapeutic efficacy. Nat Commun. 8:147542017. View Article : Google Scholar : PubMed/NCBI | |
Xie X, Lv J, Zhu W, Tian C, Li J, Liu J, Zhou H, Sun C, Hu Z and Li X: The combination therapy of oncolytic HSV-1 armed with anti-PD-1 antibody and IL-12 enhances anti-tumor efficacy. Transl Oncol. 15:1012872022. View Article : Google Scholar | |
Jung I, Shin S, Baek MC and Yea K: Modification of immune cell-derived exosomes for enhanced cancer immunotherapy: Current advances and therapeutic applications. Exp Mol Med. 56:19–31. 2024. View Article : Google Scholar : PubMed/NCBI | |
Fan T, Zhang M, Yang J, Zhu Z, Cao W and Dong C: Therapeutic cancer vaccines: Advancements, challenges, and prospects. Signal Transduct Target Ther. 8:4502023. View Article : Google Scholar : PubMed/NCBI | |
Senzer NN, Kaufman H, Amatruda T, Nemunaitis M, Daniels GA, Glaspy J, Goldsweig H, Coffin RS and Nemunaitis J: Phase II clinical trial with a second generation, GM-CSF encoding, oncolytic herpesvirus in unresectable metastatic melanoma. J Clin Oncol. 27(15 Suppl): S90352009. View Article : Google Scholar | |
Ott PA and Hodi FS: Talimogene laherparepvec for the treatment of advanced melanoma. Clin Cancer Res. 22:3127–3131. 2016. View Article : Google Scholar : PubMed/NCBI | |
Ylösmäki E, Ylösmäki L, Fusciello M, Martins B, Ahokas P, Cojoc H, Uoti A, Feola S, Kreutzman A, Ranki T, et al: Characterization of a novel OX40 ligand and CD40 ligand-expressing oncolytic adenovirus used in the PeptiCRAd cancer vaccine platform. Mol Ther Oncolytics. 20:459–469. 2021. View Article : Google Scholar : PubMed/NCBI | |
Huang Q, Cai WQ, Han ZW, Wang MY, Zhou Y, Cheng JT, Zhang Y, Wang YY, Xin Q, Wang XW, et al: Bispecific T cell engagers and their synergistic tumor immunotherapy with oncolytic viruses. Am J Cancer Res. 11:2430–2455. 2021.PubMed/NCBI | |
Guo ZS, Lotze MT, Zhu Z, Storkus WJ and Song XT: Bi- and tri-specific T cell engager-armed oncolytic viruses: Next-generation cancer immunotherapy. Biomedicines. 8:2042020. View Article : Google Scholar : PubMed/NCBI | |
Jiang J, Li J, Zhou X, Zhao X, Huang B and Qin Y: Exosomes regulate the epithelial-mesenchymal transition in cancer. Front Oncol. 12:8649802022. View Article : Google Scholar : PubMed/NCBI | |
Xiang Z, Hua M, Hao Z, Biao H, Zhu C, Zhai G and Wu J: The roles of mesenchymal stem cells in gastrointestinal cancers. Front Immunol. 13:8440012022. View Article : Google Scholar : PubMed/NCBI | |
Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, Citrin DE, Restifo NP, Robbins PF, Wunderlich JR, et al: Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res. 17:4550–4557. 2011. View Article : Google Scholar : PubMed/NCBI | |
Nguyen HM, Bommareddy PK, Silk AW and Saha D: Optimal timing of PD-1 blockade in combination with oncolytic virus therapy. Semin Cancer Biol. 86:971–980. 2022. View Article : Google Scholar | |
Pol J, Kroemer G and Galluzzi L: First oncolytic virus approved for melanoma immunotherapy. Oncoimmunology. 5:e11156412015. View Article : Google Scholar | |
Overman MJ, Ernstoff MS and Morse MA: Where we stand with immunotherapy in colorectal cancer: deficient mismatch repair, proficient mismatch repair, and toxicity management. Am Soc Clin Oncol Educ Book. 38:239–247. 2018. View Article : Google Scholar : PubMed/NCBI | |
Creasy JM, Sadot E, Koerkamp BG, Chou JF, Gonen M, Kemeny NE, Balachandran VP, Kingham TP, DeMatteo RP, Allen PJ, et al: Actual 10-year survival after hepatic resection of colorectal liver metastases: What factors preclude cure? Surgery. 163:1238–1244. 2018. View Article : Google Scholar : PubMed/NCBI | |
Yu J, Green MD, Li S, Sun Y, Journey SN, Choi JE, Rizvi SM, Qin A, Waninger JJ, Lang X, et al: Liver metastasis restrains immunotherapy efficacy via macrophage-mediated T cell elimination. Nat Med. 27:152–164. 2021. View Article : Google Scholar : PubMed/NCBI | |
Garg P, Pareek S, Kulkarni P, Horne D, Salgia R and Singhal SS: Next-generation immunotherapy: Advancing clinical applications in cancer treatment. J Clin Med. 13:65372024. View Article : Google Scholar : PubMed/NCBI | |
Jin KT, Du WL, Liu YY, Lan HR, Si JX and Mou XZ: Oncolytic virotherapy in solid tumors: The challenges and achievements. Cancers (Basel). 13:5882021. View Article : Google Scholar : PubMed/NCBI | |
Chen Q, Zhang H, Wu M, Wang Q, Luo L, Ma H, Zhang X and He S: Discovery of a potent hedgehog pathway inhibitor capable of activating caspase8-dependent apoptosis. J Pharmacol Sci. 137:256–264. 2018. View Article : Google Scholar : PubMed/NCBI | |
Albring KF, Weidemüller J, Mittag S, Weiske J, Friedrich K, Geroni MC, Lombardi P and Huber O: Berberine acts as a natural inhibitor of Wnt/β-catenin signaling-identification of more active 13-arylalkyl derivatives. Biofactors. 39:652–662. 2013. View Article : Google Scholar : PubMed/NCBI | |
García-Foncillas J, Sunakawa Y, Aderka D, Wainberg Z, Ronga P, Witzler P and Stintzing S: Distinguishing features of cetuximab and panitumumab in colorectal cancer and other solid tumors. Front Oncol. 9:8492019. View Article : Google Scholar : PubMed/NCBI | |
Xie YH, Chen YX and Fang JY: Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct Target Ther. 5:222020. View Article : Google Scholar : PubMed/NCBI | |
Wang D, Zhang H, Xiang T and Wang G: Clinical application of adaptive immune therapy in MSS colorectal cancer patients. Front Immunol. 12:7623412021. View Article : Google Scholar : PubMed/NCBI | |
Fukuoka S, Hara H, Takahashi N, Kojima T, Kawazoe A, Asayama M, Yoshii T, Kotani D, Tamura H, Mikamoto Y, et al: Regorafenib plus nivolumab in patients with advanced gastric or colorectal cancer: An open-label, dose-escalation, and dose-expansion phase Ib trial (REGONIVO, EPOC1603). J Clin Oncol. 38:2053–2061. 2020. View Article : Google Scholar : PubMed/NCBI | |
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. 350:2335–2342. 2004. View Article : Google Scholar : PubMed/NCBI | |
Qin Y, Ma FY, Zhang Z, Zhao CH and Huang B: Vascular endothelial growth factor pathway's influence on bevacizumab efficacy in metastatic colorectal cancer treatment. World J Gastrointest Oncol. 16:4514–4517. 2024. View Article : Google Scholar : PubMed/NCBI | |
Tilgase A, Olmane E, Nazarovs J, Brokāne L, Erdmanis R, Rasa A and Alberts P: Multimodality treatment of a colorectal cancer stage IV patient with FOLFOX-4, bevacizumab, rigvir oncolytic virus, and surgery. Case Rep Gastroenterol. 12:457–465. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wu Z, Ichinose T, Naoe Y, Matsumura S, Villalobos IB, Eissa IR, Yamada S, Miyajima N, Morimoto D, Mukoyama N, et al: Combination of cetuximab and oncolytic virus canerpaturev synergistically inhibits human colorectal cancer growth. Mol Ther Oncolytics. 13:107–115. 2019. View Article : Google Scholar : PubMed/NCBI | |
Van Cutsem E, Tabernero J, Lakomy R, Prenen H, Prausová J, Macarulla T, Ruff P, van Hazel GA, Moiseyenko V, Ferry D, et al: Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J Clin Oncol. 30:3499–3506. 2012. View Article : Google Scholar : PubMed/NCBI | |
Holch JW, Ricard I, Stintzing S, Modest DP and Heinemann V: The relevance of primary tumour location in patients with metastatic colorectal cancer: A meta-analysis of first-line clinical trials. Eur J Cancer. 70:87–98. 2017. View Article : Google Scholar | |
Schmoll HJ, Tabernero J, Maroun J, de Braud F, Price T, Van Cutsem E, Hill M, Hoersch S, Rittweger K and Haller DG: Capecitabine plus oxaliplatin compared with fluorouracil/folinic acid as adjuvant therapy for stage iii colon cancer: Final results of the NO16968 randomized controlled phase III trial. J Clin Oncol. 33:3733–3740. 2015. View Article : Google Scholar : PubMed/NCBI | |
Lenz HJ, Ou FS, Venook AP, Hochster HS, Niedzwiecki D, Goldberg RM, Mayer RJ, Bertagnolli MM, Blanke CD, Zemla T, et al: Impact of consensus molecular subtype on survival in patients with metastatic colorectal cancer: Results from CALGB/SWOG 80405 (alliance). J Clin Oncol. 37:1876–1885. 2019. View Article : Google Scholar : PubMed/NCBI | |
Dienstmann R, Vermeulen L, Guinney J, Kopetz S, Tejpar S and Tabernero J: Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer. Nat Rev Cancer. 17:79–92. 2017. View Article : Google Scholar : PubMed/NCBI | |
Westphalen CB, Martins-Branco D, Beal JR, Cardone C, Coleman N, Schram AM, Halabi S, Michiels S, Yap C, André F, et al: The ESMO tumour-agnostic classifier and screener (ETAC-S): A tool for assessing tumour-agnostic potential of molecularly guided therapies and for steering drug development. Ann Oncol. 35:936–953. 2024. View Article : Google Scholar : PubMed/NCBI | |
McKean WB, Moser JC, Rimm D and Hu-Lieskovan S: Biomarkers in precision cancer immunotherapy: Promise and challenges. Am Soc Clin Oncol Educ Book. 40:e275–e291. 2020. View Article : Google Scholar : PubMed/NCBI | |
Steuerwald NM, Morris S, Nguyen DG and Patel JN: Understanding the biology and testing techniques for pharmacogenomics in oncology: A practical guide for the clinician. JCO Oncol Pract. 20:1441–1451. 2024. View Article : Google Scholar : PubMed/NCBI | |
Lișcu HD, Verga N, Atasiei DI, Badiu DC, Dumitru AV, Ultimescu F, Pavel C, Stefan RE, Manole DC and Ionescu AI: Biomarkers in colorectal cancer: Actual and future perspectives. Int J Mol Sci. 25:115352024. View Article : Google Scholar : | |
Marcus L, Fashoyin-Aje LA, Donoghue M, Yuan M, Rodriguez L, Gallagher PS, Philip R, Ghosh S, Theoret MR, Beaver JA, et al: FDA approval summary: Pembrolizumab for the treatment of tumor mutational burden-high solid tumors. Clin Cancer Res. 27:4685–4689. 2021. View Article : Google Scholar : PubMed/NCBI | |
Yarchoan M, Hopkins A and Jaffee EM: Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med. 377:2500–2501. 2017. View Article : Google Scholar : PubMed/NCBI | |
Puccini A, Seeber A and Berger MD: Biomarkers in metastatic colorectal cancer: Status quo and future perspective. Cancers (Basel). 14:48282022. View Article : Google Scholar : PubMed/NCBI | |
Sunshine J and Taube JM: PD-1/PD-L1 inhibitors. Curr Opin Pharmacol. 23:32–38. 2015. View Article : Google Scholar : PubMed/NCBI | |
Matsuhashi N, Yamada T, Nagasaka T, Kataoka K, Sakamoto K, Koda K, Hiramatsu K, Matsuoka H, Kuramochi H, Ishida H, et al: Impact of RAS and BRAF heterogeneity on the efficacy of EGFR blockade in patients with metastatic colorectal cancer. J Clin Oncol. 43(4 Suppl): S2552025. View Article : Google Scholar | |
Ogunwobi OO, Mahmood F and Akingboye A: Biomarkers in colorectal cancer: Current research and future prospects. Int J Mol Sci. 21:53112020. View Article : Google Scholar : PubMed/NCBI | |
De Mattia E, Polesel J, Mezzalira S, Palazzari E, Pollesel S, Toffoli G and Cecchin E: Predictive and prognostic value of oncogene mutations and microsatellite instability in locally-advanced rectal cancer treated with neoadjuvant radiation-based therapy: A systematic review and meta-analysis. Cancers (Basel). 15:14692023. View Article : Google Scholar : PubMed/NCBI | |
Santollani L, Maiorino L, Zhang YJ, Palmeri JR, Stinson JA, Duhamel LR, Qureshi K, Suggs JR, Porth OT, Pinney W III, et al: Local delivery of cell surface-targeted immunocytokines programs systemic antitumor immunity. Nat Immunol. 25:1820–1829. 2024. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Shi X, Shen Y, Dong X, He R, Chen G, Zhang Y, Tan H and Zhang K: Nanoengineering-armed oncolytic viruses drive antitumor response: Progress and challenges. MedComm. 5:e7552024. View Article : Google Scholar : PubMed/NCBI | |
Hwang SR, Chakraborty K, An JM, Mondal J, Yoon HY and Lee Y: Pharmaceutical aspects of nanocarriers for smart anticancer therapy. Pharmaceutics. 13:18752021. View Article : Google Scholar : PubMed/NCBI | |
Choi JW, Nam JP, Nam K, Lee YS, Yun CO and Kim SW: Oncolytic adenovirus coated with multidegradable bioreducible core-cross-linked polyethylenimine for cancer gene therapy. Biomacromolecules. 16:2132–2143. 2015. View Article : Google Scholar : PubMed/NCBI | |
Yang Y, Liu Q, Wang M, Li L, Yu Y, Pan M, Hu D, Chu B, Qu Y and Qian Z: Genetically programmable cell membrane-camouflaged nanoparticles for targeted combination therapy of colorectal cancer. Signal Transduct Target Ther. 9:1582024. View Article : Google Scholar : PubMed/NCBI | |
Fang C, Xiao G, Wang T, Song L, Peng B, Xu B and Zhang K: Emerging nano-/biotechnology drives oncolytic virus-activated and combined cancer immunotherapy. Research (Wash D C). 6:01082023.PubMed/NCBI | |
Zhou F, Zhu H and Fu C: Editorial: Clinical therapeutic development against cancers resistant to targeted therapies. Front Pharmacol. 12:8168962022. View Article : Google Scholar : PubMed/NCBI | |
Kaufman HL, Kohlhapp FJ and Zloza A: Erratum: Oncolytic viruses: A new class of immunotherapy drugs. Nat Rev Drug Discov. 15:6602016. View Article : Google Scholar | |
Lichty BD, Breitbach CJ, Stojdl DF and Bell JC: Going viral with cancer immunotherapy. Nat Rev Cancer. 14:559–567. 2014. View Article : Google Scholar : PubMed/NCBI | |
Lawler SE, Speranza MC, Cho CF and Chiocca EA: Oncolytic viruses in cancer treatment: A review. JAMA Oncol. 3:841–849. 2017. View Article : Google Scholar | |
Li X and Cheng Z: Oncolytic viruses in cancer immunotherapy. Adv Ther. 7:23004452024. View Article : Google Scholar | |
Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, Kadel EE III, Koeppen H, Astarita JL, Cubas R, et al: TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 554:544–548. 2018. View Article : Google Scholar : PubMed/NCBI | |
Lv D, Fei Y, Chen H, Wang J, Han W, Cui B, Feng Y, Zhang P and Chen J: Crosstalk between T lymphocyte and extracellular matrix in tumor microenvironment. Front Immunol. 15:13407022024. View Article : Google Scholar : PubMed/NCBI | |
Fox CR and Parks GD: Histone deacetylase inhibitors enhance cell killing and block interferon-beta synthesis elicited by infection with an oncolytic parainfluenza virus. Viruses. 11:4312019. View Article : Google Scholar : PubMed/NCBI | |
Jennings VA, Scott GB, Rose AMS, Scott KJ, Migneco G, Keller B, Reilly K, Donnelly O, Peach H, Dewar D, et al: Potentiating oncolytic virus-induced immune-mediated tumor cell killing using histone deacetylase inhibition. Mol Ther. 27:1139–1152. 2019. View Article : Google Scholar : PubMed/NCBI | |
Roulstone V, Pedersen M, Kyula J, Mansfield D, Khan AA, McEntee G, Wilkinson M, Karapanagiotou E, Coffey M, Marais R, et al: BRAF- and MEK-targeted small molecule inhibitors exert enhanced antimelanoma effects in combination with oncolytic reovirus through ER stress. Mol Ther. 23:931–942. 2015. View Article : Google Scholar : PubMed/NCBI | |
De Henau O, Rausch M, Winkler D, Campesato LF, Liu C, Cymerman DH, Budhu S, Ghosh A, Pink M, Tchaicha J, et al: Overcoming resistance to checkpoint blockade therapy by targeting PI3Kγ in myeloid cells. Nature. 539:443–447. 2016. View Article : Google Scholar : PubMed/NCBI | |
Choi HJ, Chung TW, Kang SK, Lee YC, Ko JH, Kim JG and Kim CH: Ganglioside GM3 modulates tumor suppressor PTEN-mediated cell cycle progression-transcriptional induction of p21(WAF1) and p27(kip1) by inhibition of PI-3K/AKT pathway. Glycobiology. 16:573–583. 2006. View Article : Google Scholar : PubMed/NCBI | |
Kazanets A, Shorstova T, Hilmi K, Marques M and Witcher M: Epigenetic silencing of tumor suppressor genes: Paradigms, puzzles, and potential. Biochim Biophys Acta. 1865:275–288. 2016.PubMed/NCBI | |
Wallace BD, Wang H, Lane KT, Scott JE, Orans J, Koo JS, Venkatesh M, Jobin C, Yeh LA, Mani S and Redinbo MR: Alleviating cancer drug toxicity by inhibiting a bacterial enzyme. Science. 330:831–835. 2010. View Article : Google Scholar : PubMed/NCBI | |
Jung G, Hernández-Illán E, Moreira L, Balaguer F and Goel A: Epigenetics of colorectal cancer: Biomarker and therapeutic potential. Nat Rev Gastroenterol Hepatol. 17:111–130. 2020. View Article : Google Scholar : PubMed/NCBI | |
Kaur M, Frahm F, Lu Y, Ascha MS, Guadamuz JS, Dotan E, Gottesman AS, Leybovich BC, Sondhi A, Zhao Y, et al: Broadening eligibility criteria and diversity among patients for cancer clinical trials. NEJM Evid. 3:EVIDoa23002362024. View Article : Google Scholar : PubMed/NCBI | |
Veen T, Kanani A, Lea D and Søreide K: Clinical trials of neoadjuvant immune checkpoint inhibitors for early-stage operable colon and rectal cancer. Cancer Immunol Immunother. 72:3135–3147. 2023. View Article : Google Scholar : PubMed/NCBI | |
Li C, Ferro A, Mhatre SK, Lu D, Lawrance M, Li X, Li S, Allen S, Desai J, Fakih M, et al: Hybrid-control arm construction using historical trial data for an early-phase, randomized controlled trial in metastatic colorectal cancer. Commun Med (Lond). 2:902022. View Article : Google Scholar : PubMed/NCBI | |
Qi Z, Gu J, Qu L, Shi X, He Z, Sun J, Tan L and Sun M: Advancements of engineered live oncolytic biotherapeutics (microbe/virus/cells): Preclinical research and clinical progress. J Control Release. 375:209–235. 2024. View Article : Google Scholar : PubMed/NCBI | |
Xu Z, Li W, Dong X, Chen Y, Zhang D, Wang J, Zhou L and He G: Precision medicine in colorectal cancer: Leveraging multi-omics, spatial omics, and artificial intelligence. Clin Chim Acta. 559:1196862024. View Article : Google Scholar : PubMed/NCBI | |
Cornish AJ, Gruber AJ, Kinnersley B, Chubb D, Frangou A, Caravagna G, Noyvert B, Lakatos E, Wood HM, Thorn S, et al: The genomic landscape of 2,023 colorectal cancers. Nature. 633:127–136. 2024. View Article : Google Scholar : PubMed/NCBI | |
Gujar S, Pol JG, Kim Y, Lee PW and Kroemer G: Antitumor benefits of antiviral immunity: An underappreciated aspect of oncolytic virotherapies. Trends Immunol. 39:209–221. 2018. View Article : Google Scholar | |
Su Y, Su C and Qin L: Current landscape and perspective of oncolytic viruses and their combination therapies. Transl Oncol. 25:1015302022. View Article : Google Scholar : PubMed/NCBI | |
Bi Y, Sun L, Gao D, Ding C, Li Z, Li Y, Cun W and Li Q: High-efficiency targeted editing of large viral genomes by RNA-guided nucleases. PLoS Pathog. 10:e10040902014. View Article : Google Scholar : PubMed/NCBI | |
Hsu PD, Lander ES and Zhang F: Development and applications of CRISPR-Cas9 for genome engineering. Cell. 157:1262–1278. 2014. View Article : Google Scholar : PubMed/NCBI | |
Cornall AM, Phillips S, Cummins E, Garland SM and Tabrizi SN: In vitro assessment of the effect of vaccine-targeted human papillomavirus (HPV) depletion on detection of non-vaccine HPV types: Implications for post-vaccine surveillance studies. J Virol Methods. 214:10–14. 2015. View Article : Google Scholar | |
Monge C, Xie C, Myojin Y, Coffman K, Hrones DM, Wang S, Hernandez JM, Wood BJ, Levy EB, Juburi I, et al: Phase I/II study of PexaVec in combination with immune checkpoint inhibition in refractory metastatic colorectal cancer. J Immunother Cancer. 11:e0056402023. View Article : Google Scholar : PubMed/NCBI | |
Zhang B, Huang J, Tang J, Hu S, Luo S, Luo Z, Zhou F, Tan S, Ying J, Chang Q, et al: Intratumoral OH2, an oncolytic herpes simplex virus 2, in patients with advanced solid tumors: A multicenter, phase I/II clinical trial. J Immunother Cancer. 9:e0022242021. View Article : Google Scholar : PubMed/NCBI | |
Zhang L, Pakmehr SA, Shahhosseini R, Hariri M, Fakhrioliaei A, Karkon Shayan F, Xiang W and Karkon Shayan S: Oncolytic viruses improve cancer immunotherapy by reprogramming solid tumor microenvironment. Med Oncol. 41:82023. View Article : Google Scholar : PubMed/NCBI | |
Fakih M, Raghav KPS, Chang DZ, Larson T, Cohn AL, Huyck TK, Cosgrove D, Fiorillo JA, Tam R, D'Adamo D, et al: Regorafenib plus nivolumab in patients with mismatch repair-proficient/microsatellite stable metastatic colorectal cancer: A single-arm, open-label, multicentre phase 2 study. EClinicalMedicine. 58:1019172023. View Article : Google Scholar : PubMed/NCBI | |
Wang N, Wang J, Zhang Z, Cao H, Yan W, Chu Y, Chard Dunmall LS and Wang Y: A novel vaccinia virus enhances anti-tumor efficacy and promotes a long-term anti-tumor response in a murine model of colorectal cancer. Mol Ther Oncolytics. 20:71–81. 2020. View Article : Google Scholar | |
Chen L, Chen H, Ye J, Ge Y, Wang H, Dai E, Ren J, Liu W, Ma C, Ju S, et al: Intratumoral expression of interleukin 23 variants using oncolytic vaccinia virus elicit potent antitumor effects on multiple tumor models via tumor microenvironment modulation. Theranostics. 11:6668–6681. 2021. View Article : Google Scholar : PubMed/NCBI | |
Deng L, Yang X, Fan J, Ding Y, Peng Y, Xu D, Huang B and Hu Z: IL-24-Armed oncolytic vaccinia virus exerts potent antitumor effects via multiple pathways in colorectal cancer. Oncol Res. 28:579–590. 2021. View Article : Google Scholar : | |
Li J, O'Malley M, Sampath P, Kalinski P, Bartlett DL and Thorne SH: Expression of CCL19 from oncolytic vaccinia enhances immunotherapeutic potential while maintaining oncolytic activity. Neoplasia. 14:1115–1121. 2012. View Article : Google Scholar | |
Flanagan K, Glover RT, Hörig H, Yang W and Kaufman HL: Local delivery of recombinant vaccinia virus expressing secondary lymphoid chemokine (SLC) results in a CD4 T-cell dependent antitumor response. Vaccine. 22:2894–2903. 2004. View Article : Google Scholar : PubMed/NCBI | |
Bereta M, Bereta J, Park J, Medina F, Kwak H and Kaufman HL: Immune properties of recombinant vaccinia virus encoding CD154 (CD40L) are determined by expression of virally encoded CD40L and the presence of CD40L protein in viral particles. Cancer Gene Ther. 11:808–818. 2004. View Article : Google Scholar : PubMed/NCBI | |
Warner SG, Kim SI, Chaurasiya S, O'Leary MP, Lu J, Sivanandam V, Woo Y, Chen NG and Fong Y: A novel chimeric poxvirus encoding hNIS Is tumor-tropic, imageable, and synergistic with radioiodine to sustain colon cancer regression. Mol Ther Oncolytics. 13:82–92. 2019. View Article : Google Scholar : PubMed/NCBI | |
Xing M, Wang X, Chi Y and Zhou D: Gene therapy for colorectal cancer using adenovirus-mediated full-length antibody, cetuximab. Oncotarget. 7:28262–28272. 2016. View Article : Google Scholar : PubMed/NCBI | |
Rong Y, Ning Y, Zhu J, Feng P, Zhu W, Zhao X, Xiong Z, Ruan C, Jin J, Wang H, et al: Oncolytic adenovirus encoding decorin and CD40 ligand inhibits tumor growth and liver metastasis via immune activation in murine colorectal tumor model. Mol Biomed. 5:392024. View Article : Google Scholar : PubMed/NCBI | |
Nie ZL, Pan YQ, He BS, Gu L, Chen LP, Li R, Xu YQ, Gao TY, Song GQ, Hoffman AR, et al: Gene therapy for colorectal cancer by an oncolytic adenovirus that targets loss of the insulin-like growth factor 2 imprinting system. Mol Cancer. 11:862012. View Article : Google Scholar : PubMed/NCBI | |
Hecht JR, Raman SS, Chan A, Kalinsky K, Baurain JF, Jimenez MM, Garcia MM, Berger MD, Lauer UM, Khattak A, et al: Phase Ib study of talimogene laherparepvec in combination with atezolizumab in patients with triple negative breast cancer and colorectal cancer with liver metastases. ESMO Open. 8:1008842023. View Article : Google Scholar : PubMed/NCBI | |
Tian L, Liu T, Jiang S, Cao Y, Kang K, Su H, Ren G, Wang Z, Xiao W and Li D: Oncolytic Newcastle disease virus expressing the co-stimulator OX40L as immunopotentiator for colorectal cancer therapy. Gene Ther. 30:64–74. 2023. View Article : Google Scholar | |
Vigil A, Park MS, Martinez O, Chua MA, Xiao S, Cros JF, Martínez-Sobrido L, Woo SL and García-Sastre A: Use of reverse genetics to enhance the oncolytic properties of Newcastle disease virus. Cancer Res. 67:8285–8292. 2007. View Article : Google Scholar : PubMed/NCBI | |
Li X, Wang Z, Zhang S, Yao Q, Chen W and Liu F: Ruxolitinib induces apoptosis of human colorectal cancer cells by downregulating the JAK1/2-STAT1-Mcl-1 axis. Oncol Lett. 21:3522021. View Article : Google Scholar : PubMed/NCBI | |
Ghonime MG and Cassady KA: Combination therapy using ruxolitinib and oncolytic HSV renders resistant MPNSTs susceptible to virotherapy. Cancer Immunol Res. 6:1499–1510. 2018. View Article : Google Scholar : PubMed/NCBI | |
Hong YS, Hong SW, Kim SM, Jin DH, Shin JS, Yoon DH, Kim KP, Lee JL, Heo DS, Lee JS and Kim TW: Bortezomib induces G2-M arrest in human colon cancer cells through ROS-inducible phosphorylation of ATM-CHK1. Int J Oncol. 41:76–82. 2012.PubMed/NCBI | |
Kim Y, Lee J, Lee D and Othmer HG: Synergistic effects of bortezomib-OV therapy and anti-invasive strategies in glioblastoma: A mathematical model. Cancers (Basel). 11:2152019. View Article : Google Scholar : PubMed/NCBI | |
Li X, Hu W, Shen J, Li M and Gong W: Targeting proteasome enhances anticancer activity of oncolytic HSV-1 in colorectal cancer. Virology. 578:13–21. 2023. View Article : Google Scholar | |
Dinu IM, Mihăilă M, Diculescu MM, Croitoru VM, Turcu-Stiolica A, Bogdan D, Miron MI, Lungulescu CV, Alexandrescu ST, Dumitrașcu T, et al: Bevacizumab treatment for metastatic colorectal cancer in real-world clinical practice. Medicina (Kaunas). 59:3502023. View Article : Google Scholar : PubMed/NCBI | |
Saoudi González N, Ros J, Baraibar I, Salvà F, Rodríguez-Castells M, Alcaraz A, García A, Tabernero J and Élez E: Cetuximab as a key partner in personalized targeted therapy for metastatic colorectal cancer. Cancers (Basel). 16:4122024. View Article : Google Scholar : PubMed/NCBI | |
Yang S, Li X, Guan W, Qian M, Yao Z, Yin X and Zhao H: NVP-BKM120 inhibits colon cancer growth via FoxO3a-dependent PUMA induction. Oncotarget. 8:83052–83062. 2017. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Ning J, Wakimoto H, Wu S, Wu CL, Humphrey MR, Rabkin SD and Martuza RL: Oncolytic herpes simplex virus and PI3K inhibitor BKM120 synergize to promote killing of prostate cancer stem-like cells. Mol Ther Oncolytics. 13:58–66. 2019. View Article : Google Scholar : PubMed/NCBI | |
Soltantoyeh T, Akbari B, Shahosseini Z, Mirzaei HR and Hadjati J: Simultaneous targeting of Tim3 and A2a receptors modulates MSLN-CAR T cell antitumor function in a human cervical tumor xenograft model. Front Immunol. 15:13629042024. View Article : Google Scholar : PubMed/NCBI | |
Chin K, Chand VK and Nuyten DSA: Avelumab: Clinical trial innovation and collaboration to advance anti-PD-L1 immunotherapy. Ann Oncol. 28:1658–1666. 2017. View Article : Google Scholar : PubMed/NCBI | |
Redman JM, O'Sullivan Coyne G, Reed CT, Madan RA, Strauss J, Steinberg SJ, Marté J, Cordes L, Heery C and Gulley JL: Avelumab in patients with metastatic colorectal cancer. Oncologist. 28:823–e804. 2023. View Article : Google Scholar : PubMed/NCBI | |
Español-Rego M, Fernández-Martos C, Elez E, Foguet C, Pedrosa L, Rodríguez N, Ruiz-Casado A, Pineda E, Cid J, Cabezón R, et al: A phase I-II multicenter trial with Avelumab plus autologous dendritic cell vaccine in pre-treated mismatch repair-proficient (MSS) metastatic colorectal cancer patients; GEMCAD 1602 study. Cancer Immunol Immunother. 72:827–840. 2023. View Article : Google Scholar : | |
Zhu WM and Middleton MR: Combination therapies for the optimisation of bispecific T-cell engagers in cancer treatment. Immunother Adv. 3:ltad0132023. View Article : Google Scholar : PubMed/NCBI | |
Heidbuechel JPW and Engeland CE: Oncolytic viruses encoding bispecific T cell engagers: A blueprint for emerging immunovirotherapies. J Hematol Oncol. 14:632021. View Article : Google Scholar : PubMed/NCBI | |
Puca E, Schmitt-Koopmann C, Furter M, Murer P, Probst P, Dihr M, Bajic D and Neri D: The targeted delivery of interleukin-12 to the carcinoembryonic antigen increases the intratumoral density of NK and CD8+ T cell in an immunocompetent mouse model of colorectal cancer. J Gastrointest Oncol. 11:803–811. 2020. View Article : Google Scholar : PubMed/NCBI | |
Li Q, Oduro PK, Guo R, Li R, Leng L, Kong X, Wang Q and Yang L: Oncolytic viruses: Immunotherapy drugs for gastrointestinal malignant tumors. Front Cell Infect Microbiol. 12:9215342022. View Article : Google Scholar : PubMed/NCBI | |
Sumransub N, Vantanasiri K, Prakash A and Lou E: Advances and new frontiers for immunotherapy in colorectal cancer: Setting the stage for neoadjuvant success? Mol Ther Oncolytics. 22:1–12. 2021. View Article : Google Scholar : PubMed/NCBI | |
Yoo SY, Bang SY, Jeong SN, Kang DH and Heo J: A cancer-favoring oncolytic vaccinia virus shows enhanced suppression of stem-cell like colon cancer. Oncotarget. 7:16479–16489. 2016. View Article : Google Scholar : PubMed/NCBI | |
Shebbo S, Binothman N, Darwaish M, Niaz HA, Abdulal RH, Borjac J, Hashem AM and Mahmoud AB: Redefining the battle against colorectal cancer: A comprehensive review of emerging immunotherapies and their clinical efficacy. Front Immunol. 15:13502082024. View Article : Google Scholar : PubMed/NCBI |