Open Access

Resistance to oncolytic virotherapy: Multidimensional mechanisms and therapeutic breakthroughs (Review)

  • Authors:
    • Jinzhou Xu
    • Zhiyu Xia
    • Shaogang Wang
    • Qidong Xia
  • View Affiliations

  • Published online on: August 21, 2025     https://doi.org/10.3892/ijmm.2025.5612
  • Article Number: 171
  • Copyright: © Xu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Oncolytic viruses (OV) are an emerging form of immunotherapy that utilize naturally occurring or engineered viruses to specifically infect and lyse tumor cells. They achieve tumor treatment through direct tumor cell killing or by inducing immunogenic cell death to enhance immune responses. However, the efficacy of OV has been suboptimal in clinical trials. This review comprehensively examines mechanisms of resistance to OV through three interconnected dimensions: The characteristics of tumors and tumor cells, factors related to stromal cells and the extracellular matrix (ECM) and the host immune status. Potential solutions targeting these mechanisms are also proposed. For instance, OV typically achieve tumor selectivity through tumor‑specific receptors or specific promoters. However, due to inter‑ and intratumoral heterogeneity, the lack of such specific receptors or promoters in tumor cells can lead to off‑target effects of OV, resulting in treatment resistance. The ECM in the tumor microenvironment, such as hyaluronic acid, may also impede viral transport. Additionally, the clearance of OV by immune cells can contribute to suboptimal therapeutic outcomes of OV treatment. Consequently, investigating predictive biomarkers of OV efficacy, utilizing ECM‑degrading enzymes and combining with immune checkpoint inhibitors represents a promising strategy to augment the therapeutic effects of OV. Synthesizing current evidence, it is anticipated that future investigations will optimize the therapeutic effects of OV treatment and bring better immunotherapeutic outcomes for cancer patients.

Introduction

Cancer remains one of the leading causes of mortality worldwide, driving the relentless pursuit of novel therapeutic strategies (1). Oncolytic virus (OV) therapy is a novel treatment that utilizes viruses to specifically infect and lyse tumor cells. Unlike traditional radiotherapy and chemotherapy, OVs work through the following mechanisms: Selectively infecting tumor cells, inducing immunogenic cell death and enhancing immune responses. OVs have been explored across a broad spectrum of malignancies, encompassing various solid tumors and some hematological cancers. However, despite decades of research and promising preclinical data, the clinical efficacy of OV therapy, as measured by objective response rates and durable remissions, has often been suboptimal in larger trials. While some patients experience significant and sometimes long-lasting benefits, a substantial proportion derive limited or no clinical benefit (2). This discrepancy between promising mechanisms and variable clinical outcomes highlights a critical challenge: Resistance to OV therapy.

The development of resistance is a complex and multifaceted phenomenon, likely arising from dynamic interactions within the intricate ecosystem of the tumor and its microenvironment (TME). Factors contributing to this resistance span multiple interconnected levels: The heterogeneity of patients and tumors, changes in the components of tumor stromal cells and extracellular matrix (ECM), and the complexity of the tumor immune microenvironment. This review aims to provide an in-depth understanding of the reasons for OV resistance and offer a basis for future research directions and optimization of treatment plans (Fig. 1).

Tumor cells

The foundation of OV application lies in its ability to specifically infect tumor cells, replicate within them and not harm normal cells. This is typically achieved by exploiting the hallmarks of cancer cells, such as differences in surface protein expression, metabolic characteristics and genetic features. However, due to the significant heterogeneity of tumors, when the strategies targeting the tumor are not applicable to the respective patient, resistance to OV therapy can occur (3).

Virus entry

OVs typically require the recognition of specific membrane receptors to enter tumor cells. Taking advantage of the receptor recognition process is an important approach for achieving tumor selectivity for OVs. Certain OVs are designed to target tumor cells via natural receptors, which have differing expression levels between tumor and normal cells (4). For instance, Nectin-like protein 5 (CD155) is the natural receptor for poliovirus and is typically weakly expressed in normal human tissues but widely overexpressed in malignant tumors, and is associated with poor prognosis and tumor progression (5,6). Adenovirus type 5 (Ad5) infects cells via the Coxsackie and Ad receptor (CAR), while Ad3 has a different tropism and enters cells through CD46 (7). Expression of CAR and CD46 was reported to influence therapeutic effectiveness of oncolytic virotherapy, while neither CAR nor CD46 is highly expressed in all tumors (8-10). Although not all viral sensitivities are associated with the expression levels of viral receptors, the presence of Galectin-1, which is involved in the entry of H-1PV, a kind of protoparvovirus, into cells, was found to have a positive correlation with the cell-killing effect induced by H-1PV (11,12). Cell lines of pancreatic ductal adenocarcinoma are known to exhibit reduced expression of the low-density lipoprotein receptor, which is also the receptor for vesicular stomatitis virus (VSV) and pancreatic ductal adenocarcinoma cells thereby had an OV-resistant phenotype (13). Besides natural receptors, another strategy is to genetically modify the virus to target tumor-specific membrane antigens, such as CD20 (14), epidermal growth factor receptor (EGFR) (15) and human EGFR2 (16). Notably, tumor heterogeneity poses a significant clinical challenge. The absence of these receptors on tumor cells compromises OV specificity and infectivity, thereby driving therapeutic resistance (17). The macrophage receptor with collagenous structure (MARCO) exhibits context-dependent duality in viral pathogenesis. As a viral entry facilitator, MARCO directly binds herpes simplex virus type 1 (HSV-1) glycoprotein C to mediate keratinocyte adsorption (18), enhances vaccinia virus infection in skin epithelia (19) and serves as an Ad entry receptor via hexon protein interactions in macrophages (20). Consistently, Ghosh et al (21) found that MARCO-deficient mice exhibited significantly reduced morbidity and mortality due to influenza pneumonia compared to wild-type controls. Conversely, in systemic defense roles, MARCO+ lymphatic endothelial cells sequester arthritogenic alphaviruses in draining lymph nodes to limit viremia, while MARCO+ Kupffer cells clear circulating virions in the liver (22). In respiratory immunity, High et al (23) identified a loss-of-function single nucleotide polymorphism in the human MARCO promoter that ablates gene expression, amplifies in vitro inflammatory responses to respiratory syncytial virus (RSV) infection, and confers an increased risk of severe disease in two independent pediatric cohorts with RSV. Examples of OV achieving tumor selectivity through natural receptors and tumor-specific antigens are listed in Table I (24-33). Additionally, mutations in the PI3K/AKT pathway within tumor cells have been found to be associated with the ability of CF33, a novel chimeric orthopoxvirus, to enter tumors (34). However, AKT mutations occur at low frequencies (~3-5%) and in tumors without mutations in the AKT pathway, this OV may exhibit therapeutic resistance (35). The successful and selective entry of OVs into tumor cells is a crucial step in their therapeutic effectiveness. However, due to the heterogeneity of tumor cells, some may resist the entry of OVs, which directly affects the therapeutic outcome. If this resistance mechanism cannot be overcome, OV therapy will struggle to spread effectively and exert its function, potentially leading to treatment failure. Therefore, understanding and addressing the impact of tumor heterogeneity on OVs is vital for improving the success rate of OV therapy (36).

Table I

Examples of OV achieving tumor selectivity through natural receptors and tumor-specific antigens.

Table I

Examples of OV achieving tumor selectivity through natural receptors and tumor-specific antigens.

A, OV targets tumor cells through natural receptors
NameViral typeTargetTarget characteristics and expression statusRelated tumor types(Refs.)
Ad5/F35Ad5CAR and CD46Widely expressed in the majority of tumor cellsMultiple tumors(7)
MeV-SCDMVCD46The expression level varies in tumors, which affects the efficacy of OV therapyNeuroendocrine neoplasms(9)
Ad5/F11pAd5CD46The expression level varies in tumors, which affects the efficacy of oncolytic virus therapyBladder cancer(10)
H-1PV ProtoparvovirusGalectin-1Positively correlated with the cytotoxic effect of H-1PVPDAC(12)
-VSVLDLRLow expression in pancreatic ductal adenocarcinoma cells, leading to resistance to VSV.PDAC(13)
-PoliovirusCD155Low expression in normal tissues, but widely overexpressed in malignant tumors and associated with poor prognosisMultiple solid tumors (such as gliomas)(30)
-NDVSialic acid receptorsAffects the efficacy of OV therapy-(26)
-Alphaviru s M1MXRA8Directly binds to the virus and its expression is positively correlated with the induced tumor lysisMelanoma(25)

B, Targeting tumor overexpressed surface targets with engineered OV

NameViral typeTargetTarget characteristics and expression statusRelated tumor types(Refs.)

MVgreenH blindantiCD 20MVCD20A marker of B-cell non-Hodgkin lymphoma and also the target molecule for rituximabB-cell non-Hodgkin's lymphomas(14)
gD:scEGFR Δ2-24HSVEGFRLowly expressed in normal tissues but widely overexpressed in various malignant tumorsGlioblastoma, head and neck cancer, lung cancer, breast cancer(15)
R-LM113/R-LM249HSVHER-2Overexpressed in a number of cancersMalignant epithelial tumor(16)
OV-Cmab-CCL5HSVEGFRAs aboveGlioblastoma(33)
AdCMVLac Z 425-S11AdEGFRAs above-(27)
KGNEpHSVEpCAMA recognized carcinoma-associated antigen; is associated with poor prognosisMultiple tumors(32)
Ad5-ffCXCL12Ad5CXCR4/CXCR7Expression is associated with the occurrence and progression of breast cancerBreast cancer(31)
KNTc-gD:GDNFHSVGFRα1Low expression in normal tissues, but highly expressed in breast cancerBreast cancer(28)
R-593HSVPSMASpecifically overexpressed in prostate cancer; has been widely used in clinical applicationsProstate cancer(29)
VSVΔG-αFRVSVFRUsually highly expressed in tumor tissues, while its expression is low or absent in normal tissuesOvarian cancer and myeloid leukemia(24)

[i] Ad, adenovirus; MV, measles virus; CAR, Coxsackie and adenovirus receptor; PDAC, pancreatic ductal adenocarcinoma; NDV, Newcastle disease virus; VSV, vesicular stomatitis virus; LDLR, low-density lipoprotein receptor; MXRA8, matrix remodeling associated 8; HSV, herpes simplex virus; HER-2, human epidermal growth factor receptor 2; EGFR, epidermal growth factor receptor; EpCAM, epithelial cell adhesion molecule; CXCR4, C-X-C chemokine receptor type 4; GFRα1, GDNF family receptor alpha 1; PSMA, prostate-specific membrane antigen; FR, folate receptor; OV, oncolytic virus.

Virus replication

Another feature that makes OVs selectively target tumor cells is their ability to selectively replicate and proliferate within tumor cells. This selectivity relies on the characteristics of high metabolism and rapid division of tumor cells, as well as mutations in tumor genetic features (37). The world's first approved OV, H101, has the E1B 55kD gene deleted (38). Cells infected with the virus lacking the E1B 55kD gene are more susceptible to p53-induced apoptosis, which is typically mutated in tumor cells, leading to selective replication of OV in tumor cells (39). Modification of E1A protein, whose interaction with retinoblastoma protein (Rb) is the initiation for Ad infection, is a common approach to achieve selective replication in tumor cells (40). Mutations in the E1A gene prevent it from binding to the Rb, which diminishes the virus's replication capability in normal cells. However, due to the typical dysregulation of the Rb gene and over-activated state of E2F in tumor cells, the virus is able to replicate within them (41,42). However, the E2F family is not activated in all tumors. Among the target genes of E2F, some have been identified as negative regulators of growth, which can suppress tumor growth (43-45). High activity of telomerase is a crucial characteristic that allows tumor cells to maintain telomere length and achieve immortality, which is different from the low activity of telomerase in normal cells (46,47). Based on this, controlling the replication of OVs selectively in telomerase-expressing cells can make the OVs tumor-specific (48,49). Using the carcinoembryonic antigen (CEA) promoter, which is highly expressed in numerous tumors, is another approach for targeting tumor cells (50-52). However, telomerase is also expressed in many normal cell types, such as stem cells, germline cells and lymphocytes, which can diminish the targeting specificity of OVs (53). CEA is not universally expressed in all cancers and it has also been detected in mature normal cells, such as goblet cells of the colon and epithelial cells of the prostate (54). In addition, certain OVs can be engineered to possess tissue specificity. For instance, OVs controlled by the cyclooxygenase-2 (COX-2) promoter can specifically target breast cancer, while those with prostate-specific antigen (PSA) selectivity can target prostate cancer (55,56). However, it has been reported that only ~36% of patients with breast cancer express COX-2 and PSA is also expressed in normal prostate tissue and can be elevated in prostatitis (57,58). Notably, the incorporation of a transgene that replaces the E3 region of an HAdV-5 vector enables replication-dependent expression of the transgene in human A549 cells, but not in the murine 4T1 cells (59). The metabolic and genetic alterations in tumors are specific, and when the characteristics of tumor cells change, understanding their overall condition is crucial to minimize resistance to OV therapy.

Cell death

After OVs enter tumor cells, sufficient survival time is critical for viral replication and gene expression. Striking a balance is essential: Premature cell death may abort viral replication, while delayed death allows progeny accumulation. This balance varies across cell death modalities impacting therapeutic efficacy (60). Cell death induced by OVs is a form of immunogenic cell death. The oncolytic effect is an important way for OVs to combat tumors (61). OVs can induce tumor cell death through various mechanisms, including apoptosis, necroptosis and pyroptosis.

Apoptosis modulated by viral infection has been found to be mediated by a death-inducing signaling complex formed by death receptors, such as Fas, TNF-related apoptosis-inducing ligand-receptor and TNF-R. A study has reported that the introduction of the second mitochondria-derived activator of caspase/diablo IAP-binding mitochondrial protein gene can enhance tumor apoptosis and improve OV efficacy, thereby overcoming resistance to OV therapy caused by insufficient oncolytic effects (62). A study by Dobson et al (63) also found that the combination of VSV with an antagonist of apoptosis inhibitors can enhance the oncolytic effect, thereby improving therapeutic efficacy. Using Ad armed with secretable trimeric TNF-related apoptosis-inducing ligand promotes apoptosis and enhances the killing and therapeutic efficacy against glioblastoma (49). However, on the contrary, Zhou et al (64) discovered that animal cells infected with diarrhea virus promote apoptosis by upregulating the phosphorylation of p53, thereby inhibiting viral replication. Mansour et al (65) discovered that the resistance of tumor cells to apoptosis is the basis for the oncolytic selectivity of OVs. They used a non-small cell lung cancer (NSCLC) cell line overexpressing the anti-apoptotic protein Bcl-xL and found that it was resistant to early apoptotic stimuli from Newcastle disease virus (NDV), yet paradoxically exhibited increased apoptosis later on (65). This paradox arises as apoptotic resistance extends viral replication windows, facilitating syncytia formation to form syncytia. Stanziale et al (66) also found that cell death induced by HSV-1 OV infection in human gastric cancer cell lines may hinder the generation of viral progeny. The use of the apoptosis inhibitor N-acetylcysteine was shown to enhance the efficacy of the virus (66). These studies highlight the paradoxical and complex role of apoptosis in OV therapy, where both the inhibition and promotion of apoptosis can potentially enhance the efficacy of OVs. Apoptosis may facilitate tumor killing effects and could also prematurely terminate viral replication, reducing virus spread. The net effect may depend on gene mutation status, viral status and tumor type. Therefore, the role of apoptosis in the treatment resistance of specific OVs and particular cancer types requires further in-depth investigation.

Necroptosis, a form of programmed cell death, typically involves the activation of receptor-interacting protein kinase 1 (RIPK1) and RIPK3. RIPK1 and RIPK3 transmit signals by forming a complex known as the 'necrosome', leading to necroptosis (67). Oncolytic HSV intervention can induce early necroptosis after formation of multinucleated giant cells (68). Research by Okamura et al (69) found Coxsackievirus A11 could induce various modes of cell death in malignant pleural mesothelioma, including necroptosis. OV M1 can induce necroptosis in triple-negative breast cancer and its combination with doxorubicin further enhances the level of necroptosis (70). In a previous study, a necroptosis-inducing factor, MLKL, was inserted into Vaccinia viruses to further promote necroptosis in tumor cells and the therapeutic efficacy was found to be enhanced (71). In addition to focusing on the role of necroptosis in promoting tumor cell death, attention should also be paid to the potential impact of necroptosis on viral infection and replication. Wen et al (72) found that RIPK3-dependent necroptosis is induced and restricts viral replication in human astrocytes infected with Zika virus. Similar conclusions have also been made with pseudorabies virus (73). However, Hu et al (74) found that RIPK3-driven necroptosis induced by Coxsackievirus A10 actually enhances viral replication. Furthermore, Cotia poxviruses possess proteins that closely resemble the pseudoactive domain sequence of MLKL. By utilizing their kinase domain to sequester RIPK3, these proteins can impede the engagement and phosphorylation of MLKL, subsequently suppressing the process of necroptosis (75). The M45 protein from cytomegaloviruses (CMV) effectively suppresses necroptosis and the activation of the nuclear factor-κB pathway. This capability to inhibit necroptosis is vital for CMV's infection in certain cell types that are sensitive to this form of cell death, as well as for the virus's spread throughout the host organism (76). These results demonstrate that, from an evolutionary and genetic perspective, necroptosis is a way for host cells to clear viral infections. It is noteworthy that van den Wollenberg et al (77) found that reoviruses can induce necroptosis in mouse cells, but this is not common in human cell lines. This indicates differences between animal experimental models and clinical applications and may also account for the resistance to OVs observed in clinical trials.

Pyroptosis, orchestrated by the Gasdermin (GSDM) family, is another type of programmed cell death. It involves distinct morphological alterations such as cellular swelling, break-down of the plasma membrane and chromatin condensation. This process culminates in the discharge of pro-inflammatory factors from the cell, subsequently instigating an inflammatory response (78). Lin et al (79) discovered that oncolytic Parapoxvirus can promote pyroptosis in tumor cells by reducing the ubiquitination of GSDME, particularly in cells with low expression of GSDME. The recombinant measles virus vaccine, rMV-Hu191, also demonstrates oncolytic activity against esophageal squamous cell carcinoma through the induction of caspase-3/GSDME-dependent pyroptosis (80). Genetically modified OVs can more effectively induce pyroptosis to provide anticancer effects (81,82). Certain novel nanoparticle drugs are also used in combination with OVs to promote pyroptosis in tumor cells and enhance the effectiveness of cancer treatment (83). Furthermore, mouse CMV M84 protein engages with the pyrin domain found in absent in melanoma 2 (AIM2) and Apoptosis associated speck like protein containing a CARD, thereby inhibiting the formation of the inflammasome complex. This interaction can prevent caspase-1-dependent pyroptosis (84). Another protein from human CMV UL83 was also proven to interact with AIM2 and downregulate interleukin (IL)-1β, which is a vital molecule in pyroptosis (85). However, these results do not seem to focus on the impact of pyroptosis on the replication and spread of OVs. In fact, certain studies have found that pyroptosis can promote viral infection, including Coxsackievirus A16, A10, B3 and enterovirus 71 (86,87). By contrast, other studies have found that pyroptosis can enhance antiviral immunity and inhibit viral replication and spread (88,89). Furthermore, the chronic inflammatory tumor microenvironment (TME) induced by pyroptosis may promote tumor growth (90). Therefore, the therapeutic role of pyroptosis-inducing OV therapy within tumors requires further comprehensive exploration (91).

Autophagy involves the encapsulation and transport of cellular waste, including dysfunctional organelles and proteins, by autophagosomes to the lysosomes for destruction and subsequent recycling. It plays a vital role in preserving cellular stability, eliminating damaged elements and coping with stress (92). Lei et al (93) found that enhancing cellular autophagy via arming with an autophagic gene, Beclin-1, can boost the antitumor effects of vaccinia virus in leukemia and myeloma. However, autophagy may also impede the production of viral progeny. For instance, Li et al (94) discovered that the virus M1 induces autophagy in glioma cells, and inhibiting the inositol-requiring enzyme 1α-mediated autophagy pathway can enhance the oncolytic effect. Xu et al (95) also found that modifying and inhibiting cellular autophagy can enhance the effectiveness of OVs in pancreatic cancer.

Collectively, these findings suggest that different forms of cell death seem to play contradictory roles in viral infections. Based on this, it may be hypothesized that the death of tumor cells and the replication of OVs are in a delicate balance (Fig. 2). During the infection of tumor cells by OVs, if an overly strong cell death response is triggered, it can reduce the survival of tumor cells, but it may also inhibit viral replication and spread, thereby diminishing the therapeutic effect of the virus within the tumor. Therefore, in the treatment of different types of cancer, exploring the appropriate administration methods and dosages of OVs is crucial to achieve the best induction of cell death, enhance the therapeutic effect of the virus, and suppress tumor growth and metastasis to the greatest extent. Such targeted strategies can help increase the success rate of OV therapy and play a greater role in personalized cancer treatment to avoid OV resistance. The forms of tumor cell death caused by OVs and the impact of cell death on the efficacy of OVs are summarized in Table II (82,96-111).

Table II

Potential impact of different forms of cell death on resistance to OV.

Table II

Potential impact of different forms of cell death on resistance to OV.

A, Apoptosis
NameViral typeImpact on the OV efficacyRelated findingsRelated tumor types(Refs.)
H5CmTERT-Ad/TRAILAdImproveAd armed with secretable trimeric TRAIL promotes apoptosis and enhances efficacyGlioblastoma(49)
VSV-SVSVImproveIntroducing the Smac gene to promote apoptosis and overcome OV resistanceBreast cancer(62)
VSVΔ51-GFPVSVImproveCombining apoptosis-inducing agents can enhance the efficacy of OV Rhabdomyosarcoma(63)
-NDVDecreaseThe activity and replication of OV are significantly enhanced in tumor cells overexpressing anti-apoptotic proteinsNon-small-cell lung cancer(65)
NV1066HSVDecreaseCombination with an inhibitor of apoptosis increased tumor killingGastric cancer(66)
CV-B5/FCoxsackievirusImprovementOncolytic activity through apoptosis induced by endoplasmic reticulum stressNon-small-cell lung cancer(99)

B, Necroptosis

NameViral typeImpact on the OV efficacyRelated findingsRelated tumor types(Refs.)

M1AlphavirusImprovementCombining with doxorubicin can significantly trigger necroptosis and inhibit tumor growthTriple-negative breast cancer(70)
MVA/MLKLVaccinia virusImprovementInsertion of MLKL gene promotes necroptosis and enhances therapeutic efficacyMultiple tumors(71)
-Reoviruses-Reoviruses have difficulty inducing necroptosis in human cellsColorectal adenocarcinoma(77)
WR-GSVaccinia virusImprovementCombining with radiotherapy can enhance necroptosis and antitumor efficacyLung cancer(98)
-NDVImprovementBlocking necroptosis has blocked the immunogenic death of tumor cellsGlioma(102)

C, Pyroptosis

NameViral typeImpact on the OV efficacyRelated findingsRelated tumor types(Refs.)

rMV-Hu191MVImprovementShows antitumor effect through caspase-3/GSDME-mediated pyroptosisEsophageal cancer(80)
VSV-SVSVImproveIntroducing the Smac gene to promote PANoptosis to produce a more potent antitumor effectHead and neck squamous cell carcinoma(97)
rM1-mGSDME_FLAlphavirusImprovementArming gasdermin E can promote pyroptosis and improve the efficacyBreast cancer(82)
HSV-1Δ34.5/Δ47HSVImprovementCombining with novel nanoparticle drugs can promote pyroptosis and improve the effectiveness of treatmentBreast cancer and oral cancer(106)
H101AdImprovePromotes anti-tumor immunity via inducing pyroptosisMelanoma(109)
CVB3Coxsackie virusImprovePromotes gasdermin E-mediated pyroptosis to induce oncolytic activityColon cancer(111)

D, Autophagy

NameViral typeImpact on the OV efficacyRelated findingsRelated tumor types(Refs.)

M1AlphavirusDecreaseBlocking the UPR-autophagy axis significantly enhances the antitumor effectGlioma(94)
OAd.R.shPKM2AdDecreaseInhibiting autophagy can exhibit a potent cytotoxic effectPancreatic cancer(95)
dl922-947AdDecreaseInhibition of autophagy enhances the effectsGlioma(96)
saOAsAdImprovePromotes anti-tumor effects through sonosensitizer-induced autophagyMelanoma(100)
-NDVImprovementInhibiting autophagy can inhibit tumor cell death and fail to enhance the efficacy of OVBreast cancer(105)
OBP-301/OBP-702AdImprovementAd armed with p53 induces autophagy and promote antitumor effectColorectal cancer(108)
rMV-Hu191MVDecreaseInhibiting autophagy with inhibitors can enhance the efficacy of OVColorectal cancer(110)

E, Ferroptosis

NameViral typeImpact on the OV efficacyRelated findingsRelated tumor types(Refs.)
-NDVImprovementNDV kills tumor cells through ferroptosisGlioma(101)
KD01AdImprovementFerroptosis inducer increased the cytotoxicity of oncolytic virotherapyMultiple tumors(103)
vvDD-IL15RαVaccinia virusImprovementFerroptosis inducer promotes OV efficacyMultiple tumors(104)
-NDVImprovementNDV kills tumor cells through ferroptosisMultiple tumors(107)

[i] OV, oncolytic virus; Ad, adenovirus; VSV, vesicular stomatitis virus; HSV, herpes simplex virus; NDV, Newcastle disease virus; GSDME, gasdermin E; UPR, unfolded protein response; TRAIL, tumor necrosis factor-related apoptosis-inducing ligand; PANoptosis, pyroptosis, apoptosis and necroptosis combined cell death.

Anti-virus response

The interferon (IFN) pathway is a primary route of cellular antiviral innate immunity (112). Pattern recognition receptors identify viral infections to activate signaling pathways, leading to the production of IFN. IFN induces the expression of IFN-stimulated genes (ISGs), and the antiviral function is carried out by numerous approaches, including degrading viral genetic material, blocking the synthesis of viral mRNA, preventing the transport of viral nucleoproteins and affecting the release of viruses (113). For instance, a recent study identified an ISG, tripartite motif containing 21, which can affect the ubiquitination of the channel transient receptor potential cation channel subfamily V member 2 through autocrine and paracrine mechanisms, leading to its degradation and thereby modulating the susceptibility of itself and neighboring cells to viral infections (114,115). Within the TME, OVs exhibit significant sensitivity to the antiviral state that is triggered by IFNs. This sensitivity is crucial because the IFN-induced antiviral response can substantially impact the efficacy of OVs (116). In addition, this antiviral effect is thought to be regulated by an intracellular amplifier circuit that depends on the unphosphorylated and phosphorylated forms of IFN-stimulated gene factor 3 (ISGF3) and gamma activation factor complexes, as well as IFN regulatory factor 1 (IRF1) to enhance or fine-tune the antiviral response by modulating the expression of other ISGs or IFNs (117). At the same time, viruses have developed various strategies to resist host elimination and to avoid IFN-dependent innate immunity, such as hiding their genetic material, disrupting interactions with host factors that trigger IFN responses, altering phosphorylation and ubiquitination pathways within host cells, degrading proteins essential for IFN stimulation through cleavage or tagging, controlling the transcription and translation of host genes, manipulating RNA processing and transport, and employing protein-based deception tactics (118). For instance, cells infected with Dengue virus can activate the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) pathway by releasing mitochondrial DNA, which in turn generates an antiviral immune response through the production of cyclic GMP-AMP (119). Additionally, it has been found that the cofactor of the NS2B protease from Dengue virus targets cGAS for degradation in lysosomes, thereby avoiding the detection of mitochondrial DNA during the infection. This process ultimately inhibits the production of type I IFNs in the infected cells (120). This is a game of cellular resistance to viral infection and viral evasion (121).

Cancerous cells that lack a functional IFN induction or response mechanism can gain benefits in terms of survival and proliferation. Consequently, the accumulation of IFN signaling pathway mutations is closely linked to the process of cancer development (122,123). The lack of IFN response in tumor cells is also one of the foundations of OV therapy for cancer. However, research has found that tumor cells still exhibit IFN responses and activation ISGs. Delaunay et al (124) found that in pleural mesothelioma, the homozygous deletion of IFN-I genes is very frequent and increases the sensitivity to oncolytic measles virus. However, they also discovered that even IFN-I−/− cell lines can still produce some IFN-I-independent IFN-I responses and pro-inflammatory responses when exposed to the virus (124). Lipatova et al (125) discovered that in patient-derived glioblastoma multiforme, the expression levels of ISGs are similar to those in normal cells, and the silencing of certain IFN pathway-related genes such as IFN-induced protein with tetratricopeptide repeats 3 and phospholipid scramblase 1 can actually reduce the virus's ability to enter cells. If there is activation of the IFN pathway in tumors, it may potentially develop resistance to OV therapy by interfering with viral replication and degrading viral RNA. In addition, the pathways involved in antiviral actions are characterized by both cell and virus specificity (113).

Furthermore, it is important to recognize the differences in viral immunity between animal experimental models and the human body. In patients deficient in ISG15, there is no increased sensitivity to the virus, a finding that stands in marked contrast to the findings with ISG15-deficient mice (126). These differences may also be one of the reasons for the resistance to OV treatment observed in clinical trials.

Possible solution

To avoid OV therapy resistance, comprehensive gene testing of the tumor may provide a personalized treatment plan for the patient. Screening for receptors of OVs can help select the type of OV to be used to help OV entry. For instance, the expression level of nectin-1 has also been described as being associated with the sensitivity to OV therapy derived from HSV (127). Matrix remodeling associated 8 has been identified as an entry receptor for the OV M1, and together with the intracellular factor zinc finger antiviral protein, it can serve as a predictive biomarker for OV therapy (128). Furthermore, certain gene variations were reported to be correlated with OV replication and OV sensitivity. For instance, Höti et al (129) found that the integrity of p21 in normal cells affects the normal life cycle of OVs and mutations in p21 can enhance Ad replication. If genetic testing can detect mutations in genes related to the IFN pathway, it may, to a certain extent, predict the sensitivity to OV therapy (130). The expression of CD24 can predict weaker antiviral capabilities and increased sensitivity to Zika virus in neuroblastoma cells (131). Mutations in IDH1 also lead to impaired type I IFN antiviral response in glioma cells (132). Other biomarkers can also be anticipated to be further discovered (133,134). For instance, tumor mutational burden (TMB) refers to the total number of mutations within a tumor, indicating the level of genomic instability in the cancer cells and microsatellite instability (MSI) describes the variation in microsatellite regions within tumor cells due to errors during DNA replication, reflecting a heightened mutation rate in these areas (135-137). They are commonly used as biomarkers to predict the effectiveness of immune checkpoint therapies. However, the relationship between TMB or MSI and the sensitivity and efficacy of OV therapy has remained to be elucidated. However, in addition to the heterogeneity between tumors leading to the failure of OV therapy, the heterogeneity within a tumor can also result in the following scenario: Even if the presence of a certain biomarker is detected, the OV can only have a limited effect on tumor restriction (138). Therefore, in this case, to better cover the mutated characteristics of the tumor, the combination of multiple different types of OVs can broaden the therapeutic spectrum of OV therapy, thereby enhancing the efficacy of OVs.

Rationally combining already clinically used antitumor drugs with OVs can potentially enhance the therapeutic efficacy of cancer treatment. Certain drugs can facilitate the entry of OVs. For instance, the use of histone deacetylase inhibitors can upregulate the expression of virus receptor CAR on the surface of tumor cells, thereby facilitating OV entry (139-141). Jaime-Ramirez et al (142) also found that histone deacetylase inhibition could increase the levels of reovirus receptor junctional adhesion molecule 1 in head and neck squamous cell carcinomas and the combination could facilitate both virus infection and tumor lysis. Certain drugs may enhance antitumor efficacy by promoting tumor cell death. Phosphoinositol-3 kinase (PI3K) inhibitors can enhance the antitumor effects of OVs by promoting the expression of death receptor 5 on multiple myeloma cells, thereby facilitating apoptosis (143). PI3K inhibitors have also been shown to enhance the activity against prostate cancer stem cells when used in combination with HSV, while no significant effects were observed when applying a combination of OV with radiotherapy, docetaxel or inhibitors of the Wnt or NOTCH pathways (144). It is worth mentioning the contradictory role of EGFR/RAS signaling in viral replication and tumor growth. The activation of the EGFR/RAS signaling pathway is quite common in tumors and plays a crucial role in their initiation and progression (145). Oncolytic HSV in combination with EGFR inhibitors such as erlotinib or monoclonal antibodies like cetuximab has shown superior therapeutic effects compared to monotherapy in the treatment of malignant peripheral nerve sheath tumors, pancreatic cancer and colorectal cancer (146-148). Jha et al (149) also found that the combination of sunitinib can inhibit innate immunity to enhance VSV oncolytic virotherapy in prostate, breast and kidney malignant tumors. Notably, the activation of the RAS signal is also an important factor for the selective infection and replication of certain OVs in tumors. For instance, the infection and the propagation of the virus following the death of cancer cells of reovirus requires the activation of the RAS signaling pathway (150,151). In addition, the presence of RAS mutations boosts the replicative capacity and oncolytic activity of M1 virus within cancer cells and dampening the RAS signaling pathway can hinder M1 infection and the subsequent cytopathic effects (152). Based on current evidence, it may be proposed that whether this combination yields synergistic or antagonistic effects may primarily depend on the specific stage of the viral life cycle involved. Thus, while EGFR inhibitors can suppress tumor growth and exert synergistic effects when combined with OVs, they may also impair the replication of RAS-dependent OVs (153). Therefore, it is conducive that such combinations should not be employed in a random or trial-and-error manner. Instead, they should be used logically, based on an understanding of the underlying mechanisms. Otherwise, it could more readily lead to therapeutic failure.

Stromal cells and ECM

Stromal cells such as cancer-associated fibroblasts (CAFs), endothelial cells and pericytes are integral components of the TME and exert significant regulatory effects (154). The intricate interplay between cancer cells and stromal cells, in addition to the interactions between immune cells and stromal cells, plays a critical role in the development, advancement, metastasis and resistance to therapy of solid tumors. Dalin et al (155) discovered that pancreatic stellate cells can induce drug resistance to gemcitabine treatment by the secretion of deoxycytidine through equilibrative nucleoside transporters. Besides promoting the formation of new blood vessels, factors originating from tumors can also modify the genetic transcription in endothelial cells of the tumor, resulting in resistance to treatment (156,157). The ECM, which encompasses a variety of structural molecules secreted by cells and encircling them within tissues, is primarily made up of collagen, fibronectin, elastin, laminin and hyaluronic acid (HA) (158). The stiffness of the matrix triggered lipolysis in hepatic stellate cells through the focal adhesion kinase/Yes-associated protein signaling pathway, which contributed to the resistance against anti-angiogenic monoclonal antibody therapy observed in liver metastases from colorectal cancer (159). Cescon et al (160) also discovered that collagen VI maintains the stem-like properties and chemotherapy resistance in glioblastoma cells. It is evident that stromal cells and components of the ECM contribute to therapy resistance in tumors. Moving forward, this review will focus on their role in resistance to OV treatment (Fig. 3).

Tumor vasculature

Currently, the administration of OVs is mostly through intratumoral injection, which, however, is ineffective for small lesions, multiple metastatic foci and deeply located tumors that are difficult to access anatomically (161). In such cases, intravenous administration could be an alternative. Yet, when administering intravenously, the successful delivery of the virus becomes a critical factor in determining the efficacy of OV therapy. Tumor vasculature differs significantly from healthy blood vessels, being immature and chaotic, often manifesting as spherical and twisted vessels that impede efficient blood flow (162). The discontinuity of basement membranes, incomplete endothelial coverage and poor pericyte wrapping contribute to the irregularity and dysfunction of tumor blood vessels, which are also prone to leakage. While the leaky nature of these vessels permits the retention in the perivascular area and the seepage of viruses into areas beneath the endothelium, the elevated interstitial pressure from leakage also hinders the convective transport of OVs from the tumor's periphery towards its core (163). Miller et al (164) identified perfusion pressure as an essential factor influencing both the delivery and effectiveness of OVs within the tumor environment. Their research indicated that increased blood pressure, induced by physical activity, improves the ability of OVs to target tumors effectively (164). Consistent with this, Hong et al (165) found that under conditions of cell death or stress, the active or passive release of high-mobility group box 1 (HMGB1) into the extracellular environment leads to a significant increase in endothelial permeability, which results in further leakage and edema in the tumor vasculature, contributing to higher tumor interstitial pressure and blocking HMGB1 can lead to improved outcomes with OV therapy. Nevertheless, it has been posited that OV vectors are large particles to face challenges in extravasating from the tumor vasculature and enhancing tumor vascular permeability through increasing leakage can substantially boost the impact of OVs (166). OV therapy has also been observed to increase vascular permeability and immune cell infiltration in rat glioma models, yet the relationship between this enhanced tumor vascular permeability and the efficacy of the OV has not been discussed in the study (167).

In addition to affecting OV extravasation from blood vessels, endothelial cells are also involved in the process of angiogenesis. Angiogenesis is considered to be closely associated with the occurrence, progression, metastasis and therapy resistance of tumors and inhibiting angiogenesis is considered an important target in cancer therapy (168). However, the relationship between current OV therapy and tumor angiogenesis has yielded some contradictory conclusions. On the one hand, OVs have been shown in certain studies to disrupt the tumor vasculature and reduce tumor perfusion by leveraging platelet pathways, neutrophil infiltration or downregulating the vascular endothelial growth factor (VEGF) signaling pathway (169-173). Various OVs have been reported to target tumor endothelial cells and it has been found that OV intervention significantly reduces tumor vascular density (174). While these phenomena may appear linked, it is not yet clear whether the infection of endothelial cells is related to the collapse of tumor blood vessels or whether the infection of endothelial cells by OVs leads to some form of cell death (174). In in vivo experiments, tumor endothelial cells were able to effectively clear OVs without cell lysis (175). It is indeed thought-provoking to consider whether the reduction in tumor perfusion due to the destruction of tumor vasculature may impede the penetration of OVs into the tumor, particularly for OVs administered intravenously (176). On the other hand, studies have found that OVs exhibit characteristics that promote angiogenesis, but this phenomenon appears to have been only observed in HSVs thus far (176,177). Angiogenesis is a common phenomenon in viral infections and its mechanisms may be associated with tissue damage, immune cell infiltration and cytokine storms (178,179). The evidence presented above shows conflicting angiogenesis roles of OVs. These differing results may be due to variations in viral strains, tumor types and experimental conditions. At the same time, Arulanandam et al (180) found that VEGF signaling can increase the sensitivity to OVs and promote OV infection by inhibiting type I IFN. Further research is needed to determine whether OV-induced angiogenesis may further promote the metastasis and drug resistance of tumor cells.

CAFs

The influence of CAFs on tumors is complex and involves multiple factors. CAFs can promote tumor growth by regulating inflammatory responses, modifying the ECM to prevent T-cell infiltration and enhancing cell survival. However, certain subtypes of CAFs have been shown to have tumor-inhibiting effects (181). CAFs are also associated with resistance to cancer therapies. For instance, CAFs can suppress ferroptosis and induce gemcitabine resistance in pancreatic cancer cells and can promote castration resistance in prostate cancer after antiandrogen treatment (182,183). Similarly, CAFs can also affect the efficacy of OVs, ultimately leading to treatment resistance. Vähä-Koskela et al (184) found that after virus-infected cells undergo death, CAFs can rapidly form connective tissue to protect tumor cells. Arwert et al (185) discovered that direct interaction between cancer cells and CAFs stimulates fibroblasts to produce various inflammation-modulating factors. This process activates STING and induces the expression of IFN-β1 and other cytokines through IRF3, which significantly impairs the effectiveness of OVs in both in vitro and in vivo settings (185). Yasui et al (186) found that hepatocyte growth factor secreted by fibroblasts under in vitro culture conditions can inhibit Ad infection of pancreatic ductal adenocarcinoma cells by downregulating the expression of virus receptors. In addition, fibroblast growth factors (FGFs), such as FGF9 and FGF16, have been shown to inhibit the replication of viruses such as influenza A, vesicular stomatitis virus and Coxsackievirus (187,188). These factors secreted by fibroblasts are thought to be upregulated during early infection, inducing local IFN responses and inflammatory effects, thereby exerting antiviral activity. However, certain FGFs, including FGF7 and FGF10, have been found to promote the replication of viruses such as HSV I, lymphocytic choriomeningitis virus and Zika virus (189). It is important to note that part of these studies about FGFs were not conducted in tumor models and the TME may be more complex.

Given the crucial role of CAFs in remodeling the TME and contributing to therapy resistance, OVs to directly target CAFs have been designed, achieving success in preclinical research. For instance, i-leader truncation of Ads can target CAFs to enhance antitumor effects (190). Additionally, targeting specific markers of CAFs can be effective, with commonly used markers including fibroblast activation protein (FAP), the urokinase-type plasminogen activator receptor (uPAR) and junctional adhesion molecule-A with Ad and reovirus (191-194). These studies have found that targeting CAFs can effectively disrupt the tumor stromal barrier and reverse the tumor-suppressive immune microenvironment, while also reducing antiviral immune responses, promoting the spread and replication of OVs. Notably, clinical trial outcomes have demonstrated limited efficacy (195). On the one hand, this can be attributed to the limitations of preclinical models, and on the other hand, it may be strongly related to the heterogeneity of CAFs (196). The subtypes of CAFs have been extensively elaborated on in previous reviews (197). Certain subtypes promote tumor growth, suppress immune activation and enhance treatment resistance, while others, such as myofibroblastic CAFs, inhibit tumor growth. It is important to note that the targets of OV modifications aimed at CAFs do not actually distinguish between CAF subtypes and are not exclusively expressed on CAFs. For instance, the FAP protein is not only expressed in CAFs that inhibit tumor growth but also in macrophages (198). Similarly, another target, uPAR, is expressed on macrophages and other bone marrow-derived cells (199). This situation can easily lead to off-target effects of OVs, resulting in suboptimal therapeutic efficacy.

Furthermore, CAFs can regulate the composition of the ECM by secreting matrix metalloproteinase (MMP) proteins, synthesizing ECM components and mechanically reshaping the microenvironment to form physical barriers for virus spread. The relationship between the ECM and OVs will be discussed in the next chapter.

ECM

After the lysis of tumor cells by OVs, they are released into the ECM. However, most targeted therapeutics encounter substantial delivery obstacles within the tumor, and OVs, being larger in size than small molecule drugs, face even greater challenges (200). Solid tumors efficiently restrict macromolecule transport via physical barriers, with high interstitial fluid pressure and the composition of the ECM being the main physical barriers to the effective delivery of OVs (201). Asiatic acid was reported to improve the drug delivery and antitumor efficacy of pegylated liposomal doxorubicin by degrading collagen within the tumor stroma (202). Haseley et al (203) found that ECM protein cellular communication network factor 1 (CCN1) restricts the effectiveness of OV in glioma. Therefore, manipulating the ECM may also potentially enhance the spread of OV.

For instance, early studies have shown that pre-treatment with elastase can increase the efficiency of Ad gene transduction, and proteases such as collagenase/dispase and trypsin can also enhance the effectiveness of Ad gene therapy for cancer treatment by facilitating transduction (204-206). The co-injection of bacterial collagenase to modify the ECM results in a marked expansion of the initial viral distribution area within the tumor, increasing it to three times the original size in melanoma xenografts (207). HA, another key component of the ECM primarily composed of glycosaminoglycans, plays a significant role in regulating the tumor immune microenvironment, tumor migration and invasion, and resistance to cancer therapies (208). Ganesh et al (209) discovered that combination of hyaluronidase enzyme and Ads can enhance the antitumor activity of the OV and improve the prognosis of mice in animal models. Furthermore, relaxin, a peptide hormone, can effectively prompt the restructuring of collagen by enhancing the expression and activity of specific MMPs, including MMP-1, MMP-2 and MMP-14 (210,211). The intratumoral dissemination, apoptosis induction and therapeutic effectiveness of OV can be enhanced when expressing relaxin (212). Chondroitinase ABC, an enzyme derived from bacteria, facilitates the dissemination of OVs within tumors by degrading chondroitin sulfate glycosaminoglycans, a major component of the ECM (213). Additionally, MMP-1, MMP-8 and MMP-9 improve the distribution and effectiveness of OVs by influencing sulfated glycosaminoglycans in tumors (214,215). The use of epithelial junction opener, derived from Escherichia coli, in combination with oncolytic Ads can also enhance therapeutic efficacy (216). It should be noted that hyaluronidase can facilitate the growth of glioblastoma in mouse models via astrocytes, and elevated levels of relaxin may contribute to the metastasis of breast cancer (217,218). Hence, a more cautious consideration is required when disrupting the ECM. Bacterial chondroitinase ABC has been shown to enhance the spread and antitumor efficacy of OVs without significantly promoting the migration and invasion of glioma cells (219). Additionally, neutralizing antibodies present in the stromal components can significantly impact the infection and spread of OVs. Compared to the relatively simple environment in animal models, the complex environment of clinical trial patients may lead to a greater presence of anti-viral antibodies, thereby reducing the efficacy of OVs (220). Evidence indicates that in tumors with high stromal reaction, nanoparticles administered directly into the tumor mass do not disperse widely from the injection point (221). The aforementioned investigations into ECM degradation and reconfiguration, which have been shown to boost the distribution and effectiveness of OVs, further demonstrate the ECM's role as a barrier to viral spread. The failure of viral spread is a significant barrier to tumor resistance against OV therapy.

Possible solution

To address the suboptimal therapeutic effects of OVs due to stromal cells and ECM factors, three approaches can be summarized to enhance antitumor efficacy: Optimizing the OV delivery system, genetically modifying the OV and combining it with other treatments.

Using novel delivery systems of OVs can provide advantages such as improvement of the in vivo distribution of the virus, offering better biocompatibility, enhanced ability to penetrate high-pressure ECMs and protection against neutralizing antibodies (222). The main approach for 'cloaking' OVs involves encapsulation using a variety of agents, including synthetic and biological materials such as immune cells, copolymers, nanoparticles and biodegradable substances (223). For example, using polyethylene glycol as a carrier to encapsulate OVs has been found to prevent neutralization by antibodies (224). Wang et al (225) utilized a dendrimer platform and injectable hydrogel encapsulation system to deliver OVs, which reduced the generation of reactive antibodies. In fact, the exploration and application of using polymers to encapsulate OVs have been widely discussed (226,227). Nanotechnology also represents an emerging avenue for OV carriers. The use of nanoparticles to encapsulate OVs can effectively increase the circulation lifespan and overcome the insufficient penetration caused by high interstitial pressure in tumors and obstruction of dense ECM (228). The application of magnetic nanoparticles can even enable non-invasive therapeutic monitoring (229). However, it is important to note that certain nanoparticles may have inhibitory and cytotoxic effects on OVs (230). Another promising delivery system is extracellular vesicles (EVs). The use of EVs to encapsulate OVs has been explored by certain researchers. It has been found that EVs can bypass the issue of low viral receptor expression and protect the virus from neutralization by antibodies (231). Additionally, EVs offer advantages such as high biocompatibility and ease of modification. Hong et al (232) have explored the use of PH20 hyaluronidase to modify EVs for the degradation of the ECM, which holds great promise for overcoming ECM-induced resistance. Furthermore, tumor-derived EVs have a natural tropism for tumors (233). The use of effective delivery systems not only prevents OVs from being neutralized by antibodies but also, to a certain extent, avoids phagocytosis by circulating immune cells, a point that will be discussed later in the text.

Modifying the genome of OVs can enable them to express specific proteins upon infection of tumor cells. To address the physical barrier posed by the ECM, genetic modification of OVs to express ECM-degrading proteins, such as relaxin, MMPs and hyaluronidase, is a common strategy. For instance, Jung et al (234) used a relaxin-expressing oncolytic Ad that effectively degraded ECM components and increased the permeability of other drugs. OVs expressing hyaluronidase PH20 or Hyal1 have been shown to effectively induce tumor regression in both immunocompetent and immunodeficient animal models, improving OV infection and spread (201,235,236). A genetically modified vaccinia virus expressing MMP9 has been shown to promote the degradation of collagen IV, facilitating OV spread and demonstrating stronger antitumor effects (237). Regarding endothelial cells, Swanner et al (238) found that oncolytic HSV-1 armed with a decoy receptor for advanced glycation end products can block the signaling of high-mobility group box 1 (HMGB1), thereby preventing the activation of endothelial cells in response to OV infection, thus promoting the efficacy of OVs. Jeong and Yoo (239) constructed an oncolytic vaccinia virus carrying the angiopoietin 1 gene to promote tumor vascular normalization, which further facilitated the activation of antitumor immunity and enhanced antitumor effects. Armed with internalizing Arginine-Glycine-Aspartic tumor-penetrating peptide, by binding to αv-integrins on the tumor vasculature or tumor cells, and through proteolysis exposing a C-terminal motif that binds to neuropilin-1 and facilitates cell internalization, the entry of OVs into tumor cells is enhanced (240,241). OV modifications targeting CAFs have also been explored in preclinical studies. However, due to the limitations of specificity for CAFs and CAF subtypes, more research is needed on OV targeting CAFs (242).

Combining OVs with other treatments can also enhance their therapeutic efficacy. Similar to having OVs express small-molecule proteins, the use of ECM-degrading molecules in combination can effectively improve the replication and spread of OVs, thereby increasing their anti-tumor efficacy. In addition to the aforementioned MMPs, hyaluronidase and relaxin, Lavilla-Alonso et al (243) demonstrated that macrophage metalloelastase can enhance the antitumor effects of OVs in subcutaneous colorectal cancer xenografts. Furthermore, epithelial cell junctions within tumors also form an important mechanism of therapeutic resistance. Therefore, OVs combined with epithelial junction openers have been found to have better intratumoral spread and therapeutic effects (216). Targeting tumor vasculature, β-adrenergic receptor inhibitors can enhance the effect of oncolytic herpesvirus in colon cancer through anti-angiogenic actions (244). Interestingly, anti-angiogenesis has also been observed to degrade ECM components, thereby enhancing the efficacy of OVs (245). Other approaches, such as using neutralizing antibodies against HMGB1, can reduce vascular leakage and edema, thereby decreasing interstitial pressure and improving the therapeutic effect on tumors (165). To prevent the neutralization of intravenously injected OVs, certain studies have used the immunosuppressant cyclophosphamide to inhibit both innate and induced neutralizing antibody responses against HSV, thereby promoting the spread of oncolytic HSV in vivo (246,247). In addition, certain physical methods can be employed. For instance, Carlisle et al (248) and Bazan-Peregrino et al (249) found that the use of ultrasound to induce cavitation can also enhance the penetration and infection of OVs. Sun et al (250) found that reversible electroporation can induce the PI3K/Akt pathway in pancreatic cancer cells, thereby promoting M1 infection and enhancing its oncolytic effect.

Immune cells

OV therapy is considered a new generation of immunotherapy, promoting antigen-presenting cells to activate the immune system by inducing immunogenic cell death. However, the activation of the immune system is partly due to tumor antigens released from dying tumor cells and partly due to a series of antiviral responses triggered by viral infection. There is a balance between antitumor and antiviral responses during OV therapy. Moderate antiviral immunity can enhance antitumor immune effects, while excessive antiviral immunity can lead to viral clearance and resistance to OV therapy (251). In addition, Donat et al (252) found that the oncolytic vaccinia virus GLV-1H68 has a lymphangiolytic effect when treating human PC-3 prostate cancer xenografts. The authors suggest that this could prevent lymphatic metastasis of the tumor, but it remains to be determined whether this effect would also impact the process of tumor antigen presentation and tumor infiltration. Therefore, the relationship between the tumor, the immune system and the virus becomes even more complex after the introduction of the virus. Below, the potential mechanisms by which immune cells may cause failure of OV therapy are discussed (Fig. 4).

Immune checkpoint expression

Immune checkpoints refer to a series of regulatory mechanisms within the immune system that normally maintain the balance of immune responses, preventing excessive reactions that could lead to autoimmune diseases. The upregulation of immune checkpoints typically suppresses antitumor immunity, making immune checkpoint inhibitors a promising approach for cancer treatment (253). Originally characterized as regulators of T-cell function, further research has revealed that various immune cells, including macrophages, natural killer (NK) cells, dendritic cells, regulatory T cells and B cells, are also affected by the expression of immune checkpoints (254). Consequently, the upregulation of immune checkpoints in the tumor immune microenvironment can create a suppressive immune environment, leading to resistance and failure of immunotherapy. In preclinical models, numerous studies have observed evidence of OVs upregulating immune checkpoints. Kwan et al (255) also found that the introduction of HSV1716, a modified HSV, can lead to increased expression of programmed death-1 (PD-1) on T cells in a breast cancer model. In melanoma, the use of Talimogene laherparepvec has also been observed to lead to an upregulation of programmed death-ligand 1 (PD-L1) (256). Mostafa et al (257) found that oncolytic reovirus treatment in breast cancer also leads to an upregulation of PD-L1 expression in tumor cells. MEDI5395, a recombinant NDV, has similarly been found to upregulate PD-L1 after infecting myeloid cells (258). In treatments with oncolytic vaccinia virus, upregulation of PD-1, PD-L1, cytotoxic T-lymphocyte associated protein 4 (CTLA-4) and lymphocyte activation gene 3 (LAG-3) in the tumor immune microenvironment has also been observed (259). In clinical trials, similar observations of immune checkpoint upregulation following OV intervention have also been made (260). Bernstock et al (261) found that treatment with oncolytic HSV-1 G207 in a Phase 1 trial of pediatric patients with glioblastoma induced significant upregulation of immune checkpoint proteins PD-1, PD-L1, and CTLA-4. In bladder cancer, a Phase I clinical trial found an increase in PD-L1 and LAG-3 following treatment with Coxsackievirus A21 (262). In a study on oncolytic HSV-2, an increase in PD-L1 was also observed following intratumoral injection in solid tumors (263). In fact, the upregulation of immune checkpoints following viral introduction is rational. On the one hand, for immune cells, an activated immune system requires the upregulation of immune checkpoints to achieve negative feedback regulation and prevent excessive immune effects. On the other hand, for tumor cells, the upregulation of PD-L1 is a self-protective biological response to cope with the stress from viruses and the immune system.

Based on this, combining immune checkpoint inhibitors or genetically modifying OVs are the most feasible solutions. For instance, Saha et al (264) used a triple combination of anti-CTLA-4, anti-PD-1 and oncolytic HSV to treat glioblastoma, which can modulate the proportion of CD8-positive T cells in the immune microenvironment and the polarization of macrophages. An Ad has been designed to express a soluble fusion protein of PD-1/CD137L, which can simultaneously block PD-1 signaling and maintain T-cell activation signals (265). Certain studies have also used small molecule inhibitors to intervene in the upregulation of immune checkpoints, such as reducing PD-L1 upregulation in NSCLC following OV treatment through JAK/STAT inhibition (266). It is worth noting that, as previously mentioned, the IFN response of tumor cells is a factor limiting the efficacy of OVs, while mutations in the IFN pathway can lead to resistance to immune checkpoint inhibitor therapy and enhance sensitivity to OV therapy (267). This further provides a rationale for the combined use of OVs and immune checkpoint inhibitors. Therefore, several clinical trials are currently exploring the feasibility and effectiveness of combining immune checkpoint inhibitors with OVs. Current clinical trials of OV in combination with immune checkpoint inhibitors are summarized in Table III.

Table III

Clinical trials of oncolytic viruses in combination with immune checkpoint therapy.

Table III

Clinical trials of oncolytic viruses in combination with immune checkpoint therapy.

Viral nameViral typeCheckpoint inhibitorCheckpointIndicationClinical phaseIdentifier
RT-01AdNivolumabPD-1Advanced solid tumorsPhase INCT05228119
H101AdCamrelizumabPD-1Pancreatic cancerPhase IINCT06196671
RT-01AdNivolumabPD-1Advanced solid tumorsPhase INCT05122572
RevottackVesicular stomatitisPD-1 InhibitorPD-1Advanced solid tumorsPhase INCT05644509
-AdAnti-PD1PD-1Pancreatic cancerPhase INCT06346808
MG1-E6E7Maraba virusAtezolizumabPD-L1HPV-associated cancersPhase I/1bNCT03618953
Pexa-VecVaccinia virusIpilimumabCTLA-4Metastatic/advanced solid tumorsPhase INCT02977156
ADV/HSV-tkHSVPembrolizumabPD-1 MetastaticTNBC/NSCLCPhase IINCT03004183
DNX-2401AdPembrolizumabPD-1 Glioblastoma/gliosarcomaPhase IINCT02798406
Pexa-VecVaccinia virusDurvalumabPD-L1Colorectal cancerPhase I/IINCT03206073
PelareorepReovirusNivolumabPD-1Multiple myelomaPhase INCT03605719
RP1HSVNivolumabPD-1Cutaneous squamous cell carcinomaPhase IINCT04050436
MEM-288AdNivolumabPD-1Solid tumors/NSCLCPhase INCT05076760
CF33-hNISVaccinia virusPembrolizumabPD-1Advanced solid tumorsPhase INCT05346484
TILT-123AdAvelumabPD-L1Ovarian cancerPhase INCT05271318
VSV-IFNβ-NISVesicular stomatitisPembrolizumabPD-1Solid tumors/NSCLCPhase I/IINCT03647163
Olvi-VecVaccinia virusPembrolizumabPD-1NSCLCPhase IINCT06463665
H101AdCamrelizumabPD-1Malignant pleural mesotheliomaPhase INCT06031636
rHSV2hGM-CSFHSVHX-008PD-1Melanoma stage IVPhase INCT05070221
rHSV2hG M-CSFHSVHX-008PD-1Melanoma stage IVPhase INCT05068453
BioTTT001AdToripalimabPD-1Gastric cancerPhase IINCT06283121
M1-c6v1M1SHR-1210PD-1Hepatocellular carcinomaPhase INCT04665362
TILT-123AdAvelumabPD-L1Solid tumorsPhase INCT05222932
T3011HSVToripalimabPD-1Colorectal cancerPhase INCT06283303
OH2HSVHX-008PD-1MelanomaPhase I/IINCT04616443
BioTTT001AdToripalimabPD-1Colorectal cancerPhase INCT06283134
TBio-6517Vesicular stomatitisPembrolizumabPD-1Solid tumorsPhase I/IINCT04301011
LOAd703AdAtezolizumabPD-L1Malignant melanomaPhase I/IINCT04123470
ONCR-177HSVPembrolizumabPD-1Solid tumorsPhase INCT04348916
RP3HSVNivolumabPD-1Head and neck squamous cell carcinomaPhase IINCT05743270
HF10HSVIpilimumabCTLA-4MelanomaPhase IINCT02272855
VG161HSVNivolumabPD-1Gastric cancerPhase I/IINCT06008925
CVA21CoxsackievirusIpilimumabCTLA-4Uveal melanomaPhase INCT03408587
OBP-301AdPembrolizumabPD-1Head and neck squamous cell carcinomaPhase IINCT04685499
VSV-IFNβ-NISVesicular stomatitisPembrolizumabPD-1Solid tumorsPhase I/IINCT03647163
H101AdCamrelizumabPD-1Cervical cancerPhase IINCT05234905
H101AdTislelizumabPD-1Pancreatic cancerPhase INCT05303090
OncorineAdTislelizumabPD-1Hepatocellular carcinomaPhase INCT05675462

[i] Ad, adenovirus; VSV, vesicular stomatitis virus; HSV, herpes simplex virus; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1; CTLA-4, cytotoxic T-lymphocyte associated protein 4; NSCLC, non-small cell lung cancer; TNBC, triple-negative breast cancer; HPV, human papillomavirus.

Macrophages

Macrophages are pivotal in the antiviral response, capable of identifying a range of virus-specific antigens such as double-stranded RNA, single-stranded RNA and DNA. Recognition of these antigens leads to membrane reorganization and the creation of new vacuoles that engulf and transport the virus into the cell's cytoplasm. These immune cells can also engulf infected cells, processing and identifying viral elements within phagocytic vesicles (268). Activated macrophages secrete reactive oxygen species and nitric oxide, directly inhibiting viral replication through ribosomal reductase impairment (269). It has been found after extensive research that simply classifying macrophages into M1 and M2 types is somewhat outdated. In order to avoid using outdated terminology, the descriptions of 'pro-inflammatory' (historically M1) and 'immunosuppressive' (historically M2) phenotypes are now used for clarity (270). Here, the pro-inflammatory type represents the classically activated antitumor phenotype, while the immunosuppressive type is generally considered the protumor phenotype. Current research typically finds that after OV intervention, the proportion of pro-inflammatory macrophages in the immune microenvironment increases. The intervention can promote the polarization of pro-inflammatory macrophages and can even revert the immunosuppressive phenotype back to the pro-inflammatory phenotype (271,272). The pro-inflammatory macrophage phenotype, on one hand, promotes the generation of antitumor immunity, and on the other hand, also generates anti-OV immunity. Applying microglia with the pro-inflammatory phenotype to organotypic slice cultures implanted with glioma results in reduced infection of tumor cells (273). Liu et al (274) found that during OV therapy for breast cancer, low concentrations of IFN-β derived from macrophages mediated a robust antiviral immune state. Additionally, TNF-α is also considered a key effector molecule in the antiviral immune response of macrophages (275). In addition to activated inflammatory macrophages exerting antiviral effects locally within tumors, macrophages also play a role in clearing OVs when administered intravenously. This necessitates a larger dose of OV to achieve an appropriate and effective titer of OV in the TME (276). It is worth noting that OVs can also infect macrophages, turning them into carriers for dissemination and establishing chronic infections to achieve long-term therapeutic effects. Although these infections are found non-productive, whether they could lead to off-target effects and a decrease in viral therapeutic efficacy requires further consideration and discussion (277).

Based on the above evidence, strategies targeting macrophages focus on two aspects: Preventing the phagocytosis of OVs and inhibiting the function of immunosuppressive-type macrophages. Administering the PI3Kδ selective inhibitor IC87114 or the clinically approved drug Idelalisib (CAL-101) to immunocompetent mice before intravenous injection of tumor-tropic vaccinia virus can prevent viral attachment to macrophages and achieve better anti-tumor effects (278). Meisen et al (271) discovered that the therapeutic efficacy of oncolytic HSVs in treating glioblastoma was significantly improved through the use of TNF-α blocking antibodies or macrophages derived from TNF-α knockout mice. Blocking integrin β1 can also inhibited macrophage migration and enhanced viral replication and oncolytic effects (279). Thorne et al (280) found that the activation of CCN1 promotes macrophage activity and increases viral clearance. Therefore, neutralizing integrin αMβ2 can reverse this activation and the macrophage-mediated viral clearance (280). A novel oncolytic Ad engineered to express IL-13 has been shown to decrease the proportion of immunosuppressive macrophages within the TME, resulting in improved antitumor activity in oral cancer (281).

Neutrophils

Neutrophils are the most abundant subgroup of white blood cells in the peripheral blood. In recent years, the role of neutrophils in tumor occurrence, development and treatment has gradually gained attention (282). Typically, the infiltration of neutrophils in tumors is associated with poor clinical outcomes (283). Neutrophils can promote angiogenesis, tumor spread and metastasis in tumors. However, there is also a type of neutrophil, known as N1, which has been identified in humans and mice and is capable of exerting antitumor effects (284). Neutrophils were previously thought to play a crucial role primarily in combating bacterial and fungal infections, but have recently been found to also have a key role in antiviral responses. Neutrophils can act similarly to macrophages by phagocytosing viruses, thereby activating antiviral responses (285). Another powerful function of neutrophils is the release of neutrophil extracellular traps (NETs) to capture and inactivate viruses. The production of NETs is also commonly associated with poor clinical outcomes such as low immune infiltration in tumors, treatment resistance and metastasis (286,287). The above discussions have explored the relationships between neutrophils and tumors, as well as between neutrophils and viral infections. However, current research directly investigating the role of neutrophils in OV treatment resistance is not yet sufficient (288). Dai et al (289) noted that the use of oncolytic HSV in treating gliomas triggers insulin-like growth factor (IGF) 2 mRNA binding protein 3-induced NETosis. This is a specific form of inflammatory cell death in neutrophils, marked by the creation of NETs, which contributes to resistance against therapy. Price et al (290) found that vaccinia virus Ankara can rapidly recruit the infiltration of white blood cells, particularly neutrophils. The direct phagocytosis of vaccinia virus by neutrophils has also been observed in radiophagocytosis assays (291). Breitbach et al (292) also found that vesicular stomatitis virus intervention increased neutrophil infiltration in tumors, and pre-clearing neutrophils would allow the virus to replicate and spread more widely within the tumor. Taipale et al (293) found in clinical trials that patients with a lower neutrophil-to-lymphocyte ratio (NLR) prior to Ad oncolytic immunotherapy had significantly longer overall survival, and He and Lin (294) also found that in the treatment of hepatocellular carcinoma with Ad5 H101, the NLR was associated with a higher hazard ratio, indirectly suggesting that the level of neutrophils may negatively regulate the efficacy of OV therapy. Furthermore, inhibiting neutrophil function can significantly enhance the antitumor effects of intravenously administered oncolytic vaccinia viruses (295). Of note, certain studies have also found that neutrophils play a key role in tumor regression during OV therapy (296,297). Given the heterogeneity of neutrophils, further precise molecular subtyping of neutrophils may be necessary to more comprehensively explore the dual roles of neutrophils in different cancer types and with different OVs.

There are certain strategies that can inhibit the function of neutrophils to enhance the therapeutic effect of OV. For instance, pharmacological targeting of PI3Kγ can mitigate immune suppression mediated by tumor-associated myeloid cells, thereby improving the therapeutic efficacy of OV M1 in ovarian cancer (298). IFNs secreted by myeloid cells can enhance the resistance of malignant neural progenitor cells to Zika virus (299). However, inhibiting the JAK1/2 signaling pathway can restore their susceptibility. It should be noted that the term myeloid cells used here is a general reference and does not specifically pinpoint a particular subpopulation. Noh et al (300) found that in patients with recurrent glioblastoma after treatment with oncolytic HSV-1, IGF2 was upregulated. By expressing a decoy receptor for IGF2 via OV, the infiltration of neutrophils was reduced, thereby decreasing the production of immunosuppressive molecules and enhancing the efficacy of OV in animal models (300). This target is consistent with the findings of Dai et al (289) reported earlier.

NK cells

NK cells are an important component of the innate immune system, playing a crucial role in both antitumor and antiviral immunity. NK cells can directly kill tumor cells or virus-infected cells by releasing cytotoxic granules or through antibody-dependent cellular cytotoxicity. They can also enhance antitumor immunity and viral clearance by activating adaptive immunity (301). Alvarez-Breckenridge et al (302) observed that during the treatment of glioblastoma with oncolytic HSV, NK cells were recruited to the infected area within a few hours after infection and preferentially lysed infected cells through recognition by NK cell natural cytotoxicity receptors. Meanwhile, Reale et al (303) found that after infecting glioblastoma with HSV expressing green fluorescent protein, it took a period of time measured in days to observe significant viral spread and replication. It is required to consider whether the rapid response of NK cells to viral infection is too quick for the virus to survive and spread within the tumor. Of note, depleting endogenous NK cells two days before OV treatment and replenishing exogenous NK cells three days after OV intervention to control the number of NK cells can effectively extend survival (304). It is worth mentioning that OV treatment potentiates exogenous NK-cell cytotoxicity (305). Specifically, Wang et al (306) found that oncolytic HSV can upregulate checkpoint molecule NK group 2 member A on NK cells, indicating a more complex interaction between oncolytic virotherapy and tumor immune microenvironment. Expressing IL-15 via OVs can stimulate the activation of NK cells and more effectively promote tumor regression (307). Of course, this cytokine-mediated antitumor effect may not be generated through NK-cell activation, as no strict causal relationship has been identified.

Overall, most studies have found that NK cells do not seem to be a helpful ally in OV therapy. Therefore, most strategies targeting NK cells are aimed at suppressing their numbers and functions. In an orthotopic glioblastoma mouse model, pretreatment with valproic acid can reduce the recruitment of NK cells and macrophages, and valproic acid can also inhibit the function of NK cells (308). The recombinant virus rVSV-UL141 has been proposed, which expresses a protein that downregulates the expression of the NK-cell activation ligand CD155, reducing NK-cell recruitment and activation and enhancing viral replication, leading to increased tumor necrosis and extended survival (309). Administering a single dose of TGF-β before OV treatment for glioblastoma can temporarily inhibit innate immune cells, primarily macrophages and NK cells, that restrict therapeutic efficacy, thus improving the effectiveness of OV therapy (310).

Future perspective

The potential mechanisms of OV treatment resistance identified in the above studies are mostly based on biological foundations. Therefore, addressing OV treatment resistance will likely require further in-depth exploration of the effects of different virus types on different tumors and their underlying biological mechanisms in order to better intervene in this process. In addition to the exploration of biomarkers, pathways and immune cells mentioned earlier, metabolic changes within tumors may represent a new avenue for investigation. For instance, a chemical derivative of itaconate, a Krebs cycle-derived metabolite, has been found to enhance the efficacy of OV in various tumor models (311). The homeostasis of calcium ions in the TME also has a significant impact on the antitumor effects of OV (312).

Additionally, emerging biological research techniques could be highly significant for OV studies. For instance, considering the spatial dynamics within tumors, 3D cell culture techniques may more accurately reflect real tumor treatment scenarios. These techniques can systematically investigate how viral characteristics (such as viral spread rate), tumor features (such as the emergence rate of drug-resistant cells and the cost of resistance), the resistance of healthy stromal cells to the virus and the timing of treatment collectively influence therapeutic outcomes (313,314). Brachtlova et al (315) proposed a method that can detect virus-associated RNA through liquid biopsy in preclinical in vivo models, which can provide a minimally invasive detection method for in vivo replication of OV. Going further, to address animal model limitations in tumor immune microenvironment studies, humanized mouse models offer a translational platform for assessing the impact of OVs on the tumor immune microenvironment (316).

In addition to biological techniques, the intersection of biology with other disciplines is more conducive to the study and development of OV. For instance, according to mathematical models, monotherapy alone is insufficient to completely eradicate tumors and needs to be supplemented with other therapies (317). By employing a finite volume method coupled with a fractional step method and utilizing the nonlinear solver nksol for algebraic systems, Kim et al (304) determined the optimal number of endogenous immune cells to deplete and exogenous immune cells to reinfuse in order to best enhance the effects of OV therapy. Assuming the support of more powerful computing capabilities and more refined models, which could provide more accurate predictions of real-world scenarios would significantly advance the study of OV resistance. For instance, agent-based models simulating spatial OV spread in 3D tumor architectures and ordinary differential equation models optimizing immune cell depletion kinetics could predict resistance evolution (318,319).

Literature search strategy

The literature review for this article was conducted using major databases such as PubMed (https://pubmed.ncbi.nlm.nih.gov/), Web of Science (https://www.webofscience.com/) and Scopus (https://www.elsevier.com/en-in/products/scopus/search). Key words included 'oncolytic virus therapy', 'drug resistance', 'tumor microenvironment' and 'immune response'. Studies were selected based on their relevance to the topic and quality of evidence, with a focus on recent advancements and key findings in the field.

Conclusion

In this review, the challenges of drug resistance in OV therapy for cancer treatment were thoroughly explored. OVs demonstrate significant antitumor potential by selectively infecting and lysing tumor cells and inducing immunogenic cell death. However, the clinical application of OV therapy is still limited due to factors such as the high heterogeneity of tumor cells, the complexity of the TME and the diversity of host antiviral immune responses. The drug resistance mechanisms of tumor cells were analyzed in detail, including the obstruction of viral entry, the imbalance of cell death mechanisms, the activation of antiviral responses, the physical barrier effect of tumor stromal cells and the ECM, and the factors related to immune cells. Meanwhile, this paper proposes several possible solutions, such as personalized genetic testing (e.g. to test mutations in E1A or E2F, or the expression status of OV receptors to prevent treatment resistance caused by improper virus selection), combination therapy strategies (e.g. to combine with immune checkpoint inhibitors to address treatment resistance caused by immunosuppressive microenvironments), optimization of viral delivery systems (e.g. to address treatment resistance caused by antibody neutralization and insufficient tissue penetration) and genetic modification of viruses (e.g. to express ECM-degrading enzymes to overcome resistance due to physical barriers for viral spread), in order to improve the clinical efficacy of OV therapy. Future studies should prioritize investigating the interaction mechanisms between OVs and tumor cells and immune cells, develop more effective biomarkers and therapeutic targets to achieve the goal of precision medicine and facilitate broader clinical translation of OV therapy in cancer treatment.

Availability of data and materials

Not applicable.

Authors' contributions

JX performed the literature search and manuscript writing. ZX contributed to the literature search. SW was involved in manuscript review & editing. QX was responsible for study design and supervision. All authors have 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.

Abbreviations:

Ad

adenovirus

CAFs

cancer-associated fibroblasts

CAR

Coxsackie and Ad receptor

CPA

cyclophosphamide

CTLA-4

cytotoxic T-lymphocyte associated protein 4

CXCR4

C-X-C chemokine receptor type 4

ECM

extracellular matrix

EGFR

epidermal growth factor receptor

EpCAM

epithelial cell adhesion molecule

EVs

extracellular vesicles

FAP

fibroblast activation protein

FR

folate receptor

GFRα1

GDNF family receptor α1

GSDME

gasdermin E

HA

hyaluronic acid

HER-2

human EGFR2

HMGB1

high-mobility group box 1

HPV

human papillomavirus

HSV

herpes simplex virus

ICB

immune checkpoint blockade

ICD

immunogenic cell death

IFN

interferon

ISGs

IFN-stimulated genes

JAK/STAT

Janus kinase/signal transducer and activator of transcription

JAM-A

junctional adhesion molecule-A

LDLR

low-density lipoprotein receptor

MMPs

matrix metalloproteinases

MSI

microsatellite instability

MV

measles virus

MXRA8

matrix remodeling associated 8

NDV

Newcastle disease virus

NETs

neutrophil extracellular traps

NK cells

natural killer cells

NLR

neutrophil-to-lymphocyte ratio

NSCLC

non-small cell lung cancer

OV

oncolytic virus

PANoptosis

pyroptosis, apoptosis and necroptosis combined cell death

PD-1

programmed cell death protein 1

PD-L1

programmed death-ligand 1

PDAC

pancreatic ductal adenocarcinoma

PI3K

phosphoinositide 3-kinase

PRRs

pattern recognition receptors

PSMA

prostate-specific membrane antigen

RAS

rat sarcoma

T-VEC

Talimogene laherparepvec

TGF-β

transforming growth factor β

TILs

tumor-infiltrating lymphocytes

TMB

tumor mutational burden

TME

tumor microenvironment

TNF-α

tumor necrosis factor α

TNBC

triple-negative breast cancer

TRAIL

TNF-related apoptosis-inducing ligand

UPAR

urokinase-type plasminogen activator receptor

UPR

unfolded protein response

VEGF

vascular endothelial growth factor

VSV

vesicular stomatitis virus

Acknowledgements

Not applicable.

Funding

No funding was received.

References

1 

Siegel RL, Kratzer TB, Giaquinto AN, Sung H and Jemal A: Cancer statistics, 2025. CA Cancer J Clin. 75:10–45. 2025.PubMed/NCBI

2 

Pol J, Buqué A, Aranda F, Bloy N, Cremer I, Eggermont A, Erbs P, Fucikova J, Galon J, Limacher JM, et al: Trial watch-oncolytic viruses and cancer therapy. Oncoimmunology. 5:e11177402016. View Article : Google Scholar : PubMed/NCBI

3 

Dagogo-Jack I and Shaw AT: Tumour heterogeneity and resistance to cancer therapies. Nat Rev Clin Oncol. 15:81–94. 2018. View Article : Google Scholar

4 

Ma R, Li Z, Chiocca EA, Caligiuri MA and Yu J: The emerging field of oncolytic virus-based cancer immunotherapy. Trends Cancer. 9:122–139. 2023. View Article : Google Scholar :

5 

Nandi SS, Gohil T, Sawant SA, Lambe UP, Ghosh S and Jana S: CD155: A key receptor playing diversified roles. Curr Mol Med. 22:594–607. 2022. View Article : Google Scholar

6 

Kučan Brlić P, Lenac Roviš T, Cinamon G, Tsukerman P, Mandelboim O and Jonjić S: Targeting PVR (CD155) and its receptors in anti-tumor therapy. Cell Mol Immunol. 16:40–52. 2019. View Article : Google Scholar

7 

Yang M, Yang CS, Guo W, Tang J, Huang Q, Feng S, Jiang A, Xu X, Jiang G and Liu YQ: A novel fiber chimeric conditionally replicative adenovirus-Ad5/F35 for tumor therapy. Cancer Biol Ther. 18:833–840. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Phung AT, Shah JR, Dong T, Reid T, Larson C, Sanchez AB, Oronsky B, Trogler WC, Kummel AC, Aisagbonhi O and Blair SL: CAR expression in invasive breast carcinoma and its effect on adenovirus transduction efficiency. Breast Cancer Res. 26:1312024. View Article : Google Scholar : PubMed/NCBI

9 

Scheicher NV, Berchtold S, Beil J, Smirnow I, Schenk A and Lauer UM: In vitro sensitivity of neuroendocrine neoplasms to an armed oncolytic measles vaccine virus. Cancers (Basel). 16:4882024. View Article : Google Scholar : PubMed/NCBI

10 

Cao W, Tian J, Li C, Gao Y, Liu X, Lu J, Wang Y, Wang Z, Svatek RS and Rodriguez R: A novel bladder cancer-specific oncolytic adenovirus by CD46 and its effect combined with cisplatin against cancer cells of CAR negative expression. Virol J. 14:1492017. View Article : Google Scholar

11 

Lipatova AV, Le TH, Sosnovtseva AO, Babaeva FE, Kochetkov DV and Chumakov PM: Relationship between cell receptors and tumor cell sensitivity to oncolytic enteroviruses. Bull Exp Biol Med. 166:58–62. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Schäfer TE, Knol LI, Haas FV, Hartley A, Pernickel SCS, Jády A, Finkbeiner MSC, Achberger J, Arelaki S, Modic Ž, et al: Biomarker screen for efficacy of oncolytic virotherapy in patient-derived pancreatic cancer cultures. EBioMedicine. 105:1052192024. View Article : Google Scholar : PubMed/NCBI

13 

Felt SA, Droby GN and Grdzelishvili VZ: Ruxolitinib and polycation combination treatment overcomes multiple mechanisms of resistance of pancreatic cancer cells to oncolytic vesicular stomatitis virus. J Virol. 91:e00461–17. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Yaiw KC, Miest TS, Frenzke M, Timm M, Johnston PB and Cattaneo R: CD20-targeted measles virus shows high oncolytic specificity in clinical samples from lymphoma patients independent of prior rituximab therapy. Gene Ther. 18:313–317. 2011. View Article : Google Scholar

15 

Ingusci S, Hall BL, Cohen JB and Glorioso JC: Oncolytic herpes simplex viruses designed for targeted treatment of EGFR-bearing tumors. Mol Ther Oncol. 32:2007612024. View Article : Google Scholar : PubMed/NCBI

16 

Leoni V, Vannini A, Gatta V, Rambaldi J, Sanapo M, Barboni C, Zaghini A, Nanni P, Lollini PL, Casiraghi C and Campadelli-Fiume G: A fully-virulent retargeted oncolytic HSV armed with IL-12 elicits local immunity and vaccine therapy towards distant tumors. PLoS Pathog. 14:e10072092018. View Article : Google Scholar : PubMed/NCBI

17 

Shiao SL, Gouin KH III, Ing N, Ho A, Basho R, Shah A, Mebane RH, Zitser D, Martinez A, Mevises NY, et al: Single-cell and spatial profiling identify three response trajectories to pembrolizumab and radiation therapy in triple negative breast cancer. Cancer Cell. 42:70–84.e8. 2024. View Article : Google Scholar : PubMed/NCBI

18 

MacLeod DT, Nakatsuji T, Yamasaki K, Kobzik L and Gallo RL: HSV-1 exploits the innate immune scavenger receptor MARCO to enhance epithelial adsorption and infection. Nat Commun. 4:19632013. View Article : Google Scholar : PubMed/NCBI

19 

MacLeod DT, Nakatsuji T, Wang Z, di Nardo A and Gallo RL: Vaccinia virus binds to the scavenger receptor MARCO on the surface of keratinocytes. J Invest Dermatol. 135:142–150. 2015. View Article : Google Scholar

20 

Stichling N, Suomalainen M, Flatt JW, Schmid M, Pacesa M, Hemmi S, Jungraithmayr W, Maler MD, Freudenberg MA, Plückthun A, et al: Lung macrophage scavenger receptor SR-A6 (MARCO) is an adenovirus type-specific virus entry receptor. PLoS Pathog. 14:e10069142018. View Article : Google Scholar : PubMed/NCBI

21 

Ghosh S, Gregory D, Smith A and Kobzik L: MARCO regulates early inflammatory responses against influenza: A useful macrophage function with adverse outcome. Am J Respir Cell Mol Biol. 45:1036–1044. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Carpentier KS, Sheridan RM, Lucas CJ, Davenport BJ, Li FS, Lucas ED, McCarthy MK, Reynoso GV, May NA, Tamburini BAJ, et al: MARCO(+) lymphatic endothelial cells sequester arthritogenic alphaviruses to limit viremia and viral dissemination. EMBO J. 40:e1089662021. View Article : Google Scholar : PubMed/NCBI

23 

High M, Cho HY, Marzec J, Wiltshire T, Verhein KC, Caballero MT, Caballero MT, Acosta PL, Ciencewicki J, McCaw ZR, et al: Determinants of host susceptibility to murine respiratory syncytial virus (RSV) disease identify a role for the innate immunity scavenger receptor MARCO gene in human infants. EBioMedicine. 11:73–84. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Ayala-Breton C, Barber GN, Russell SJ and Peng KW: Retargeting vesicular stomatitis virus using measles virus envelope glycoproteins. Hum Gene Ther. 23:484–491. 2012. View Article : Google Scholar :

25 

Dan J, Cai J, Zhong Y, Wang C, Huang S, Zeng Y, Fan Z, Xu C, Hu L, Zhang J, et al: Oncolytic virus M1 functions as a bifunctional checkpoint inhibitor to enhance the antitumor activity of DC vaccine. Cell Rep Med. 4:1012292023. View Article : Google Scholar : PubMed/NCBI

26 

Dong B, Tang N, Guan Y, Qu G, Miao L, Han W and Shen Z: Type and abundance of sialic acid receptors on host cell membrane affect infectivity and viral titer of different strains of Newcastle disease virus. J Virol Methods. 302:1144882022. View Article : Google Scholar : PubMed/NCBI

27 

Haisma HJ, Grill J, Curiel DT, Hoogeland S, van Beusechem VW, Pinedo HM and Gerritsen WR: Targeting of adenoviral vectors through a bispecific single-chain antibody. Cancer Gene Ther. 7:901–904. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Hall BL, Leronni D, Miyagawa Y, Goins WF, Glorioso JC and Cohen JB: Generation of an oncolytic herpes simplex viral vector completely retargeted to the GDNF receptor GFRα1 for specific infection of breast cancer cells. Int J Mol Sci. 21:88152020. View Article : Google Scholar

29 

Menotti L, Avitabile E, Gatta V, Malatesta P, Petrovic B and Campadelli-Fiume G: HSV as a platform for the generation of retargeted, armed, and reporter-expressing oncolytic viruses. Viruses. 10:3522018. View Article : Google Scholar : PubMed/NCBI

30 

Merrill MK, Bernhardt G, Sampson JH, Wikstrand CJ, Bigner DD and Gromeier M: Poliovirus receptor CD155-targeted oncolysis of glioma. Neuro Oncol. 6:208–217. 2004. View Article : Google Scholar : PubMed/NCBI

31 

O'Bryan SM and Mathis JM: CXCL12 retargeting of an oncolytic adenovirus vector to the chemokine CXCR4 and CXCR7 receptors in breast cancer. J Cancer Ther. 12:311–336. 2021. View Article : Google Scholar : PubMed/NCBI

32 

Shibata T, Uchida H, Shiroyama T, Okubo Y, Suzuki T, Ikeda H, Yamaguchi M, Miyagawa Y, Fukuhara T, Cohen JB, et al: Development of an oncolytic HSV vector fully retargeted specifically to cellular EpCAM for virus entry and cell-to-cell spread. Gene Ther. 23:479–488. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Tian L, Xu B, Chen Y, Li Z, Wang J, Zhang J, Ma R, Cao S, Hu W, Chiocca EA, et al: Specific targeting of glioblastoma with an oncolytic virus expressing a cetuximab-CCL5 fusion protein via innate and adaptive immunity. Nat Cancer. 3:1318–1335. 2022. View Article : Google Scholar : PubMed/NCBI

34 

Choi AH, O'Leary MP, Lu J, Kim SI, Fong Y and Chen NG: Endogenous akt activity promotes virus entry and predicts efficacy of novel chimeric orthopoxvirus in triple-negative breast cancer. Mol Ther Oncolytics. 9:22–29. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Yi KH and Lauring J: Recurrent AKT mutations in human cancers: Functional consequences and effects on drug sensitivity. Oncotarget. 7:4241–4251. 2016. View Article : Google Scholar :

36 

Pelin A, Boulton S, Tamming LA, Bell JC and Singaravelu R: Engineering vaccinia virus as an immunotherapeutic battleship to overcome tumor heterogeneity. Expert Opin Biol Ther. 20:1083–1097. 2020. View Article : Google Scholar : PubMed/NCBI

37 

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

38 

Garber K: China approves world's first oncolytic virus therapy for cancer treatment. J Natl Cancer Inst. 98:298–300. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Cheng PH, Wechman SL, McMasters KM and Zhou HS: Oncolytic replication of E1b-deleted adenoviruses. Viruses. 7:5767–5779. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Goradel NH, Mohajel N, Malekshahi ZV, Jahangiri S, Najafi M, Farhood B, Mortezaee K, Negahdari B and Arashkia A: Oncolytic adenovirus: A tool for cancer therapy in combination with other therapeutic approaches. J Cell Physiol. 234:8636–8646. 2019. View Article : Google Scholar

41 

Abudoureyimu M, Lai Y, Tian C, Wang T, Wang R and Chu X: Oncolytic Adenovirus-A nova for gene-targeted oncolytic viral therapy in HCC. Front Oncol. 9:11822019. View Article : Google Scholar : PubMed/NCBI

42 

Jakubczak JL, Ryan P, Gorziglia M, Clarke L, Hawkins LK, Hay C, Huang Y, Kaloss M, Marinov A, Phipps S, et al: An oncolytic adenovirus selective for retinoblastoma tumor suppressor protein pathway-defective tumors: dependence on E1A, the E2F-1 promoter, and viral replication for selectivity and efficacy. Cancer Res. 63:1490–1499. 2003.PubMed/NCBI

43 

Ortega S, Malumbres M and Barbacid M: Cyclin D-dependent kinases, INK4 inhibitors and cancer. Biochim Biophys Acta. 1602:73–87. 2002.PubMed/NCBI

44 

Bracken AP, Ciro M, Cocito A and Helin K: E2F target genes: Unraveling the biology. Trends Biochem Sci. 29:409–417. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Kent LN and G: The broken cycle: E2F dysfunction in cancer. Nat Rev Cancer. 19:326–338. 2019. View Article : Google Scholar : PubMed/NCBI

46 

Shay JW: Role of telomeres and telomerase in aging and cancer. Cancer Discov. 6:584–593. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Boccardi V and Marano L: Aging, cancer, and inflammation: The telomerase connection. Int J Mol Sci. 25:85422024. View Article : Google Scholar : PubMed/NCBI

48 

Alfano A, Cafferata EGA, Gangemi M, Nicola Candia A, Malnero CM, Bermudez I, Lopez MV, Ríos GD, Rotondaro C, Cuneo N, et al: In vitro and in vivo efficacy of a stroma-targeted, tumor microenviron ment responsive oncolytic adenovirus in different preclinical models of cancer. Int J Mol Sci. 24:99922023. View Article : Google Scholar

49 

Oh E, Hong J, Kwon OJ and Yun CO: A hypoxia- and telomerase-responsive oncolytic adenovirus expressing secretable trimeric TRAIL triggers tumour-specific apoptosis and promotes viral dispersion in TRAIL-resistant glioblastoma. Sci Rep. 8:14202018. View Article : Google Scholar : PubMed/NCBI

50 

Xu C, Sun Y, Wang Y, Yan Y, Shi Z, Chen L, Lin H, Lü S, Zhu M, Su C and Li Z: CEA promoter-regulated oncolytic adenovirus-mediated Hsp70 expression in immune gene therapy for pancreatic cancer. Cancer Lett. 319:154–163. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Chouljenko DV, Murad YM, Lee IF, Delwar Z, Ding J, Liu G, Liu X, Bu X, Sun Y, Samudio I and Jia WW: Targeting carcinoembryonic antigen-expressing tumors using a novel transcriptional and translational dual-regulated oncolytic herpes simplex virus type 1. Mol Ther Oncolytics. 28:334–348. 2023. View Article : Google Scholar : PubMed/NCBI

52 

Cho WK, Choi DH, Park HC, Park W, Yu JI, Park YS, Park JO, Lim HY, Kang WK, Kim HC, et al: Elevated CEA is associated with worse survival in recurrent rectal cancer. Oncotarget. 8:105936–105941. 2017. View Article : Google Scholar : PubMed/NCBI

53 

Saretzki G: The telomerase connection of the brain and its implications for neurod egenerative diseases. Stem Cells. 41:233–241. 2023. View Article : Google Scholar

54 

Aquino A, Formica V, Prete SP, Correale PP, Massara MC, Turriziani M, De Vecchis L and Bonmassar E: Drug-induced increase of carcinoembryonic antigen expression in cancer cells. Pharmacol Res. 49:383–396. 2004. View Article : Google Scholar : PubMed/NCBI

55 

DeWeese TL, van der Poel H, Li S, Mikhak B, Drew R, Goemann M, Hamper U, DeJong R, Detorie N, Rodriguez R, et al: A phase I trial of CV706, a replication-competent, PSA selective oncolytic adenovirus, for the treatment of locally recurrent prostate cancer following radiation therapy. Cancer Res. 61:7464–7472. 2001.PubMed/NCBI

56 

Robert S, Roman Ortiz NI, LaRocca CJ, Ostrander JH and Davydova J: Oncolytic adenovirus for the targeting of paclitaxel-resistant breast cancer stem cells. Viruses. 16:5672024. View Article : Google Scholar : PubMed/NCBI

57 

Denkert C, Winzer KJ, Müller BM, Weichert W, Pest S, Köbel M, Kristiansen G, Reles A, Siegert A, Guski H and Hauptmann S: Elevated expression of cyclooxygenase-2 is a negative prognostic factor for disease free survival and overall survival in patients with breast carcinoma. Cancer. 97:2978–2987. 2003. View Article : Google Scholar : PubMed/NCBI

58 

Hugosson J, Godtman RA, Wallstrom J, Axcrona U, Bergh A, Egevad L, Geterud K, Khatami A, Socratous A, Spyratou V, et al: Results after four years of screening for prostate cancer with PSA and MRI. N Engl J Med. 391:1083–1095. 2024. View Article : Google Scholar : PubMed/NCBI

59 

Del Papa J, Petryk J, Bell JC and Parks RJ: An oncolytic adenovirus vector expressing p14 FAST protein induces widespread syncytium formation and reduces tumor growth rate in vivo. Mol Ther Oncolytics. 14:107–120. 2019. View Article : Google Scholar : PubMed/NCBI

60 

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

61 

Weibel S, Raab V, Yu YA, Worschech A, Wang E, Marincola FM and Szalay AA: Viral-mediated oncolysis is the most critical factor in the late-phase of the tumor regression process upon vaccinia virus infection. BMC Cancer. 11:682011. View Article : Google Scholar : PubMed/NCBI

62 

Li W, Turaga RC, Li X, Sharma M, Enadi Z, Dunham Tompkins SN, Hardy KC, Mishra F, Tsao J, Liu ZR, et al: Overexpression of Smac by an armed vesicular stomatitis virus overcomes tumor resistance. Mol Ther Oncolytics. 14:188–195. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Dobson CC, Naing T, Beug ST, Faye MD, Chabot J, St-Jean M, Walker DE, LaCasse EC, Stojdl DF, Korneluk RG and Holcik M: Oncolytic virus synergizes with Smac mimetic compounds to induce rhabdomyosarcoma cell death in a syngeneic murine model. Oncotarget. 8:3495–3508. 2017. View Article : Google Scholar :

64 

Zhou H, Zhang Y, Wang J, Yan Y, Liu Y, Shi X, Zhang Q and Xu X: The CREB and AP-1-dependent cell communication network factor 1 regulates porcine epidemic diarrhea virus-induced cell apoptosis inhibiting virus replication through the p53 pathway. Front Microbiol. 13:8318522022. View Article : Google Scholar : PubMed/NCBI

65 

Mansour M, Palese P and Zamarin D: Oncolytic specificity of Newcastle disease virus is mediated by selectivity for apoptosis-resistant cells. J Virol. 85:6015–6023. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Stanziale SF, Petrowsky H, Adusumilli PS, Ben-Porat L, Gonen M and Fong Y: Infection with oncolytic herpes simplex virus-1 induces apoptosis in neighboring human cancer cells: A potential target to increase anticancer activity. Clin Cancer Res. 10:3225–3232. 2004. View Article : Google Scholar : PubMed/NCBI

67 

Grootjans S, Vanden Berghe T and Vandenabeele P: Initiation and execution mechanisms of necroptosis: An overview. Cell Death Differ. 24:1184–1195. 2017. View Article : Google Scholar : PubMed/NCBI

68 

Suzuki T and Uchida H: Induction of necroptosis in multinucleated giant cells induced by conditionally replicating syncytial oHSV in co-cultures of cancer cells and non-cancerous cells. Mol Ther Oncol. 32:2008032024. View Article : Google Scholar : PubMed/NCBI

69 

Okamura K, Inoue H, Tanaka K, Ikematsu Y, Furukawa R, Ota K, Yoneshima Y, Iwama E and Okamoto I: Immunostimulatory oncolytic activity of coxsackievirus A11 in human malignant pleural mesothelioma. Cancer Sci. 114:1095–1107. 2023. View Article : Google Scholar :

70 

Zhang J, Liu Y, Tan J, Zhang Y, Wong CW, Lin Z, Liu X, Sander M, Yang X, Liang L, et al: Necroptotic virotherapy of oncolytic alphavirus M1 cooperated with Doxorubicin displays promising therapeutic efficacy in TNBC. Oncogene. 40:4783–4795. 2021. View Article : Google Scholar : PubMed/NCBI

71 

Van Hoecke L, Riederer S, Saelens X, Sutter G and Rojas JJ: Recombinant viruses delivering the necroptosis mediator MLKL induce a potent antitumor immunity in mice. Oncoimmunology. 9:18029682020. View Article : Google Scholar : PubMed/NCBI

72 

Wen C, Yu Y, Gao C, Qi X, Cardona CJ and Xing Z: RIPK3-dependent necroptosis Is induced and restricts viral replication in human astrocytes infected with zika virus. Front Cell Infect Microbiol. 11:6377102021. View Article : Google Scholar : PubMed/NCBI

73 

Gou H, Bian Z, Cai R, Chu P, Song S, Li Y, Jiang Z, Zhang K, Yang D and Li C: RIPK3-dependent necroptosis limits PRV replication in PK-15 cells. Front Microbiol. 12:6643532021. View Article : Google Scholar : PubMed/NCBI

74 

Hu Y, Deng X, Lv Y, Liu C, Chen J, Song J and Zhang Y: Coxsackievirus-A10 induced RIPK3-driven necroptosis to promote the formation of inflammatory response and enhance virus production via being recognized by TLR3. Mol Immunol. 178:107–116. 2025. View Article : Google Scholar : PubMed/NCBI

75 

Petrie EJ, Sandow JJ, Lehmann WIL, Liang LY, Coursier D, Young SN, Kersten WJA, Fitzgibbon C, Samson AL, Jacobsen AV, et al: Viral MLKL homologs subvert necroptotic cell death by sequestering cellular RIPK3. Cell Rep. 28:3309–3319.e5. 2019. View Article : Google Scholar : PubMed/NCBI

76 

Deng Y, Águeda-Pinto A and Brune W: No time to die: How cytomegaloviruses suppress apoptosis, necroptosis, and pyroptosis. Viruses. 16:12722024. View Article : Google Scholar : PubMed/NCBI

77 

van den Wollenberg DJM, Kemp V, Rabelink MJWE and Hoeben RC: Reovirus type 3 dearing variants do not induce necroptosis in RIPK3-expressing human tumor cell lines. Int J Mol Sci. 24:23202023. View Article : Google Scholar : PubMed/NCBI

78 

Panganiban RA, Nadeau KC and Lu Q: Pyroptosis, gasdermins and allergic diseases. Allergy. 79:2380–2395. 2024. View Article : Google Scholar : PubMed/NCBI

79 

Lin J, Sun S, Zhao K, Gao F, Wang R, Li Q, Zhou Y, Zhang J, Li Y, Wang X, et al: Oncolytic parapoxvirus induces gasdermin E-mediated pyroptosis and activates antitumor immunity. Nat Commun. 14:2242023. View Article : Google Scholar : PubMed/NCBI

80 

Wu A, Li Z, Wang Y, Chen Y, Peng J, Zhu M, Li Y, Song H, Zhou D, Zhang C, et al: Recombinant measles virus vaccine rMV-Hu191 exerts an oncolytic effect on esophageal squamous cell carcinoma via caspase-3/GSDME-mediated pyroptosis. Cell Death Discov. 9:1712023. View Article : Google Scholar : PubMed/NCBI

81 

Liu Z, Li Y, Zhu Y, Li N, Li W, Shang C, Song G, Li S, Cong J, Li T, et al: Apoptin induces pyroptosis of colorectal cancer cells via the GSDME-dependent pathway. Int J Biol Sci. 18:717–730. 2022. View Article : Google Scholar : PubMed/NCBI

82 

Chen XY, Liu Y, Zhu WB, Li SH, Wei S, Cai J, Lin Y, Liang JK, Yan GM, Guo L and Hu C: Arming oncolytic M1 virus with gasdermin E enhances antitumor efficacy in breast cancer. iScience. 27:1111482024. View Article : Google Scholar : PubMed/NCBI

83 

Wang YY, Wang J, Wang S, Yang QC, Song A, Zhang MJ, Wang WD, Liu YT, Zhang J, Wang WM, et al: Dual-responsive epigenetic inhibitor nanoprodrug combined with oncolytic virus synergistically boost cancer immunotherapy by igniting gasdermin E-mediated pyroptosis. ACS Nano. Jul 22–2024.Epub ahead of print.

84 

Deng Y, Ostermann E and Brune W: A cytomegalovirus inflammasome inhibitor reduces proinflammatory cytokine release and pyroptosis. Nat Commun. 15:7862024. View Article : Google Scholar : PubMed/NCBI

85 

Huang Y, Ma D, Huang H, Lu Y, Liao Y, Liu L, Liu X and Fang F: Interaction between HCMV pUL83 and human AIM2 disrupts the activation of the AIM2 inflammasome. Virol J. 14:342017. View Article : Google Scholar : PubMed/NCBI

86 

Wang Y, Qin Y, Wang T, Chen Y, Lang X, Zheng J, Gao S, Chen S, Zhong X, Mu Y, et al: Pyroptosis induced by enterovirus 71 and coxsackievirus B3 infection affects viral replication and host response. Sci Rep. 8:28872018. View Article : Google Scholar : PubMed/NCBI

87 

Hu Y, Zhao W, Lv Y, Li H, Li J, Zhong M, Pu D, Jian F, Song J and Zhang Y: NLRP3-dependent pyroptosis exacerbates coxsackievirus A16 and coxsackievirus A10-induced inflammatory response and viral replication in SH-SY5Y cells. Virus Res. 345:1993862024. View Article : Google Scholar : PubMed/NCBI

88 

Jiang R, Chen D, Zhang Y, Zhou L, Ge X, Han J, Guo X and Yang H: PRRSV infection inhibits CSFV C-strain replication via GSDMD-mediated pyroptosis. Vet Microbiol. 298:1102432024. View Article : Google Scholar : PubMed/NCBI

89 

Jiang J, Shen W, He Y, Liu J, Ouyang J, Zhang C and Hu K: Overexpression of NLRP12 enhances antiviral immunity and alleviates herpes simplex keratitis via pyroptosis/IL-18/IFN-γ signaling. Int Immunopharmacol. 137:1124282024. View Article : Google Scholar

90 

Zhang Z, Zhang Y and Lieberman J: Lighting a fire: Can we harness pyroptosis to ignite antitumor immunity? Cancer Immunol Res. 9:2–7. 2021. View Article : Google Scholar : PubMed/NCBI

91 

Guo ZS, Liu Z and Bartlett DL: Oncolytic immunotherapy: Dying the right way is a key to eliciting potent antitumor immunity. Front Oncol. 4:742014. View Article : Google Scholar : PubMed/NCBI

92 

Debnath J, Gammoh N and Ryan KM: Autophagy and autophagy-related pathways in cancer. Nat Rev Mol Cell Biol. 24:560–575. 2023. View Article : Google Scholar : PubMed/NCBI

93 

Lei W, Wang S, Xu N, Chen Y, Wu G, Zhang A, Chen X, Tong Y and Qian W: Enhancing therapeutic efficacy of oncolytic vaccinia virus armed with Beclin-1, an autophagic Gene in leukemia and myeloma. Biomed Pharmacother. 125:1100302020. View Article : Google Scholar : PubMed/NCBI

94 

Li K, Hu C, Xing F, Gao M, Liang J, Xiao X, Cai J, Tan Y, Hu J, Zhu W, et al: Deficiency of the IRE1α-autophagy axis enhances the antitumor effects of the oncolytic virus M1. J Virol. 92:e01331–17. 2018. View Article : Google Scholar :

95 

Xu Y, Chu L, Yuan S, Yang Y, Yang Y, Xu B, Zhang K, Liu XY, Wang R, Fang L, et al: RGD-modified oncolytic adenovirus-harboring shPKM2 exhibits a potent cytotoxic effect in pancreatic cancer via autophagy inhibition and apoptosis promotion. Cell Death Dis. 8:e28352017. View Article : Google Scholar : PubMed/NCBI

96 

Botta G, Passaro C, Libertini S, Abagnale A, Barbato S, Maione AS, Hallden G, Beguinot F, Formisano P and Portella G: Inhibition of autophagy enhances the effects of E1A-defective oncolytic adenovirus dl922-947 against glioma cells in vitro and in vivo. Hum Gene Ther. 23:623–634. 2012. View Article : Google Scholar : PubMed/NCBI

97 

Chen F, Lang L, Yang J, Yang F, Tang S, Fu Z, Saba NF, Luo M and Teng Y: SMAC-armed oncolytic virotherapy enhances the anticancer activity of PD1 blockade by modulating PANoptosis. Biomark Res. 13:82025. View Article : Google Scholar : PubMed/NCBI

98 

Chen WY, Chen YL, Lin HW, Chang CF, Huang BS, Sun WZ and Cheng WF: Stereotactic body radiation combined with oncolytic vaccinia virus induces potent anti-tumor effect by triggering tumor cell necroptosis and DAMPs. Cancer Lett. 523:149–161. 2021. View Article : Google Scholar : PubMed/NCBI

99 

Cui B, Song L, Wang Q, Li K, He Q, Wu X, Gao F, Liu M, An C, Gao Q, et al: Non-small cell lung cancers (NSCLCs) oncolysis using coxsackievirus B5 and synergistic DNA-damage response inhibitors. Signal Transduct Target Ther. 8:3662023. View Article : Google Scholar : PubMed/NCBI

100 

Ding J, Su R, Yang R, Xu J, Liu X, Yao T, Li S, Wang C, Zhang H, Yue Q, et al: Enhancing the antitumor efficacy of oncolytic adenovirus through sonodynamic therapy-augmented virus replication. ACS Nano. 18:18282–18298. 2024. View Article : Google Scholar : PubMed/NCBI

101 

Kan X, Yin Y, Song C, Tan L, Qiu X, Liao Y, Liu W, Meng S, Sun Y and Ding C: Newcastle-disease-virus-induced ferroptosis through nutrient deprivation and ferritinophagy in tumor cells. iScience. 24:1028372021. View Article : Google Scholar : PubMed/NCBI

102 

Koks CA, Garg AD, Ehrhardt M, Riva M, Vandenberk L, Boon L, De Vleeschouwer S, Agostinis P, Graf N and Van Gool SW: Newcastle disease virotherapy induces long-term survival and tumor-specific immune memory in orthotopic glioma through the induction of immunogenic cell death. Int J Cancer. 136:E313–E325. 2015. View Article : Google Scholar

103 

Li W, Ji T, Ye J, Xiong S, Si Y, Sun X, Li F and Dai Z: Ferroptosis enhances the therapeutic potential of oncolytic adenoviruses KD01 against cancer. Cancer Gene Ther. 32:403–417. 2025. View Article : Google Scholar : PubMed/NCBI

104 

Liu W, Chen H, Zhu Z, Liu Z, Ma C, Lee YJ, Bartlett DL and Guo ZS: Ferroptosis inducer improves the efficacy of oncolytic virus-mediated cancer immunotherapy. Biomedicines. 10:14252022. View Article : Google Scholar : PubMed/NCBI

105 

Rozilah MI, Yusoff K, Chia SL and Ismail S: Autophagy inhibition suppresses Newcastle disease virus-induced cell death by inhibiting viral replication in human breast cancer cells. Virology. 590:1099572024. View Article : Google Scholar

106 

Su W, Qiu W, Li SJ, Wang S, Xie J, Yang QC, Xu J, Zhang J, Xu Z and Sun ZJ: A dual-responsive STAT3 inhibitor nanoprodrug combined with oncolytic virus elicits synergistic antitumor immune responses by igniting pyroptosis. Adv Mater. 35:e22093792023. View Article : Google Scholar

107 

Sun Y, Tang L, Kan X, Tan L, Song C, Qiu X, Liao Y, Nair V, Ding C, Liu X and Sun Y: Oncolytic Newcastle disease virus induced degradation of YAP through E3 ubiquitin ligase PRKN to exacerbate ferroptosis in tumor cells. J Virol. 98:e01897232024. View Article : Google Scholar : PubMed/NCBI

108 

Tamura S, Tazawa H, Hori N, Li Y, Yamada M, Kikuchi S, Kuroda S, Urata Y, Kagawa S and Fujiwara T: p53-armed oncolytic adenovirus induces autophagy and apoptosis in KRAS and BRAF-mutant colorectal cancer cells. PLoS One. 18:e02944912023. View Article : Google Scholar : PubMed/NCBI

109 

Wang ZM, Li MK, Yang QL, Duan SX, Lou XY, Yang XY, Liu Y, Zhong YW, Qiao Y, Wang ZS, et al: Recombinant human adenovirus type 5 promotes anti-tumor immunity via inducing pyroptosis in tumor endothelial cells. Acta Pharmacol Sin. 45:2646–2656. 2024. View Article : Google Scholar : PubMed/NCBI

110 

Zhang CD, Wang YL, Zhou DM, Zhu MY, Lv Y, Hao XQ, Qu CF, Chen Y, Gu WZ, Wu BQ, et al: A recombinant Chinese measles virus vaccine strain rMV-Hu191 inhibits human colorectal cancer growth through inducing autophagy and apoptosis regulating by PI3K/AKT pathway. Transl Oncol. 14:1010912021. View Article : Google Scholar : PubMed/NCBI

111 

Zhang Y, Xu T, Tian H, Wu J, Yu X, Zeng L, Liu F, Liu Q and Huang X: Coxsackievirus group B3 has oncolytic activity against colon cancer through gasdermin E-mediated pyroptosis. Cancers (Basel). 14:62062022. View Article : Google Scholar : PubMed/NCBI

112 

Crow YJ and Casanova JL: Human life within a narrow range: The lethal ups and downs of type I interferons. Sci Immunol. 9:eadm81852024. View Article : Google Scholar : PubMed/NCBI

113 

Mesev EV, LeDesma RA and Ploss A: Decoding type I and III interferon signalling during viral infection. Nat Microbiol. 4:914–924. 2019. View Article : Google Scholar : PubMed/NCBI

114 

Guo YY, Gao Y, Zhao YL, Xie C, Gan H, Cheng X, Yang LP, Hu J, Shu HB, Zhong B, et al: Viral infection and spread are inhibited by the polyubiquitination and downregulation of TRPV2 channel by the interferon-stimulated gene TRIM21. Cell Rep. 43:1140952024. View Article : Google Scholar : PubMed/NCBI

115 

Guo YY, Gao Y, Hu YR, Zhao Y, Jiang D, Wang Y, Zhang Y, Gan H, Xie C, Liu Z, et al: The transient receptor potential vanilloid 2 (TRPV2) channel facilitates virus infection through the Ca(2+) -LRMDA axis in myeloid cells. Adv Sci (Weinh). 9:e22028572022. View Article : Google Scholar : PubMed/NCBI

116 

Ebrahimi S, Ghorbani E, Khazaei M, Avan A, Ryzhikov M, Azadmanesh K and Hassanian SM: Interferon-mediated tumor resistance to oncolytic virotherapy. J Cell Biochem. 118:1994–1999. 2017. View Article : Google Scholar : PubMed/NCBI

117 

Michalska A, Blaszczyk K, Wesoly J and Bluyssen HAR: A positive feedback amplifier circuit that regulates interferon (IFN)-stimulated gene expression and controls type I and II IFN responses. Front Immunol. 9:11352018. View Article : Google Scholar

118 

Sobol PT, Hummel JL, Rodrigues RM and Mossman KL: PML has a predictive role in tumor cell permissiveness to interferon-sensitive oncolytic viruses. Gene Ther. 16:1077–1087. 2009. View Article : Google Scholar : PubMed/NCBI

119 

Sun B, Sundström KB, Chew JJ, Bist P, Gan ES, Tan HC, Goh KC, Chawla T, Tang CK and Ooi EE: Dengue virus activates cGAS through the release of mitochondrial DNA. Sci Rep. 7:35942017. View Article : Google Scholar : PubMed/NCBI

120 

Aguirre S, Luthra P, Sanchez-Aparicio MT, Maestre AM, Patel J, Lamothe F, Fredericks AC, Tripathi S, Zhu T, Pintado-Silva J, et al: Dengue virus NS2B protein targets cGAS for degradation and prevents mitochondrial DNA sensing during infection. Nat Microbiol. 2:170372017. View Article : Google Scholar : PubMed/NCBI

121 

Liikanen I, Monsurrò V, Ahtiainen L, Raki M, Hakkarainen T, Diaconu I, Escutenaire S, Hemminki O, Dias JD, Cerullo V, et al: Induction of interferon pathways mediates in vivo resistance to oncolytic adenovirus. Mol Ther. 19:1858–1866. 2011. View Article : Google Scholar : PubMed/NCBI

122 

Li S, Zhu M, Pan R, Fang T, Cao YY, Chen S, Zhao X, Lei CQ, Guo L, Chen Y, et al: The tumor suppressor PTEN has a critical role in antiviral innate immunity. Nat Immunol. 17:241–249. 2016. View Article : Google Scholar

123 

Pikor LA, Bell JC and Diallo JS: Oncolytic viruses: Exploiting cancer's deal with the devil. Trends Cancer. 1:266–277. 2015. View Article : Google Scholar : PubMed/NCBI

124 

Delaunay T, Achard C, Boisgerault N, Grard M, Petithomme T, Chatelain C, Dutoit S, Blanquart C, Royer PJ, Minvielle S, et al: Frequent homozygous deletions of type I interferon genes in pleural mesothelioma confer sensitivity to oncolytic measles virus. J Thorac Oncol. 15:827–842. 2020. View Article : Google Scholar : PubMed/NCBI

125 

Lipatova AV, Soboleva AV, Gorshkov VA, Bubis JA, Solovyeva EM, Krasnov GS, Kochetkov DV, Vorobyev PO, Ilina IY, Moshkovskii SA, et al: Multi-omics analysis of glioblastoma cells' sensitivity to oncolytic viruses. Cancers (Basel). 13:52682021. View Article : Google Scholar : PubMed/NCBI

126 

Speer SD, Li Z, Buta S, Payelle-Brogard B, Qian L, Vigant F, Rubino E, Gardner TJ, Wedeking T, Hermann M, et al: ISG15 deficiency and increased viral resistance in humans but not mice. Nat Commun. 7:114962016. View Article : Google Scholar : PubMed/NCBI

127 

Friedman GK, Bernstock JD, Chen D, Nan L, Moore BP, Kelly VM, Youngblood SL, Langford CP, Han X, Ring EK, et al: Enhanced sensitivity of patient-derived pediatric high-grade brain tumor xenografts to oncolytic HSV-1 virotherapy correlates with nectin-1 expression. Sci Rep. 8:139302018. View Article : Google Scholar : PubMed/NCBI

128 

Song D, Jia X, Liu X, Hu L, Lin K, Xiao T, Qiao Y, Zhang J, Dan J, Wong C, et al: Identification of the receptor of oncolytic virus M1 as a therapeutic predictor for multiple solid tumors. Signal Transduct Target Ther. 7:1002022. View Article : Google Scholar : PubMed/NCBI

129 

Höti N, Johnson TJ, Chowdhury WH and Rodriguez R: Loss of cyclin-dependent kinase inhibitor alters oncolytic adenovirus replication and promotes more efficient virus production. Cancers (Basel). 10:2022018. View Article : Google Scholar : PubMed/NCBI

130 

Matveeva OV and Chumakov PM: Defects in interferon pathways as potential biomarkers of sensitivity to oncolytic viruses. Rev Med Virol. 28:e20082018. View Article : Google Scholar : PubMed/NCBI

131 

Kedarinath K, Fox CR, Crowgey E, Mazar J, Phelan P, Westmoreland TJ, Alexander KA and Parks GD: CD24 Expression dampens the basal antiviral state in human neuroblastoma cells and enhances permissivity to zika virus infection. Viruses. 14:17352022. View Article : Google Scholar : PubMed/NCBI

132 

Chen X, Liu J, Li Y, Zeng Y, Wang F, Cheng Z, Duan H, Pan G, Yang S, Chen Y, et al: IDH1 mutation impairs antiviral response and potentiates oncolytic virotherapy in glioma. Nat Commun. 14:67812023. View Article : Google Scholar : PubMed/NCBI

133 

Stavrakaki E, Dirven CMF and Lamfers MLM: Personalizing oncolytic virotherapy for glioblastoma: In search of biomarkers for response. Cancers (Basel). 13:6142021. View Article : Google Scholar : PubMed/NCBI

134 

Vasilevska J, De Souza GA, Stensland M, Skrastina D, Zhulenvovs D, Paplausks R, Kurena B, Kozlovska T and Zajakina A: Comparative protein profiling of B16 mouse melanoma cells susceptible and non-susceptible to alphavirus infection: Effect of the tumor microenvironment. Cancer Biol Ther. 17:1035–1050. 2016. View Article : Google Scholar : PubMed/NCBI

135 

Qian ZY, Pan YQ, Li XX, Chen YX, Wu HX, Liu ZX, Kosar M, Bartek J, Wang ZX and Xu RH: Modulator of TMB-associated immune infiltration (MOTIF) predicts immunotherapy response and guides combination therapy. Sci Bull (Beijing). 69:803–822. 2024. View Article : Google Scholar : PubMed/NCBI

136 

Cheng Y, Bu D, Zhang Q, Sun R, Lyle S, Zhao G, Dong L, Li H, Zhao Y, Yu J and Hao X: Genomic and transcriptomic profiling indicates the prognosis significance of mutational signature for TMB-high subtype in Chinese patients with gastric cancer. J Adv Res. 51:121–134. 2023. View Article : Google Scholar :

137 

Wilbur HC, Le DT and Agarwal P: Immunotherapy of MSI cancer: Facts and hopes. Clin Cancer Res. 30:1438–1447. 2024. View Article : Google Scholar

138 

Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM, Wakimoto H, Cahill DP, Nahed BV, Curry WT, Martuza RL, et al: Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science. 344:1396–1401. 2014. View Article : Google Scholar : PubMed/NCBI

139 

Bieler A, Mantwill K, Dravits T, Bernshausen A, Glockzin G, Köhler-Vargas N, Lage H, Gansbacher B and Holm PS: Novel three-pronged strategy to enhance cancer cell killing in glioblastoma cell lines: Histone deacetylase inhibitor, chemotherapy, and oncolytic adenovirus dl520. Hum Gene Ther. 17:55–70. 2009. View Article : Google Scholar

140 

Kitazono M, Goldsmith ME, Aikou T, Bates S and Fojo T: Enhanced adenovirus transgene expression in malignant cells treated with the histone deacetylase inhibitor FR901228. Cancer Res. 61:6328–6330. 2001.PubMed/NCBI

141 

Chang HG, Choi YH, Hong J, Choi JW, Yoon AR and Yun CO: GM101 in combination with histone deacetylase inhibitor enhances anti-tumor effects in desmoplastic microenvironment. Cells. 10:28112021. View Article : Google Scholar : PubMed/NCBI

142 

Jaime-Ramirez AC, Yu JG, Caserta E, Yoo JY, Zhang J, Lee TJ, Hofmeister C, Lee JH, Kumar B, Pan Q, et al: Reolysin and histone deacetylase inhibition in the treatment of head and neck squamous cell carcinoma. Mol Ther Oncolytics. 5:87–96. 2017. View Article : Google Scholar : PubMed/NCBI

143 

Tong Y, Zhu W, Huang X, You L, Han X, Yang C and Qian W: PI3K inhibitor LY294002 inhibits activation of the Akt/mTOR pathway induced by an oncolytic adenovirus expressing TRAIL and sensitizes multiple myeloma cells to the oncolytic virus. Oncol Rep. 31:1581–1588. 2014. View Article : Google Scholar : PubMed/NCBI

144 

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

145 

Hsu WH, LaBella KA, Lin Y, Xu P, Lee R, Hsieh CE, Yang L, Zhou A, Blecher JM, Wu CJ, et al: Oncogenic KRAS drives lipofibrogenesis to promote angiogenesis and colon cancer progression. Cancer Discov. 13:2652–2673. 2023. View Article : Google Scholar : PubMed/NCBI

146 

Mahller YY, Vaikunth SS, Currier MA, Miller SJ, Ripberger MC, Hsu YH, Mehrian-Shai R, Collins MH, Crombleholme TM, Ratner N and Cripe TP: Oncolytic HSV and erlotinib inhibit tumor growth and angiogenesis in a novel malignant peripheral nerve sheath tumor xenograft model. Mol Ther. 15:279–286. 2007. View Article : Google Scholar : PubMed/NCBI

147 

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

148 

Yamamura K, Kasuya H, Sahin TT, Tan G, Hotta Y, Tsurumaru N, Fukuda S, Kanda M, Kobayashi D, Tanaka C, et al: Combination treatment of human pancreatic cancer xenograft models with the epidermal growth factor receptor tyrosine kinase inhibitor erlotinib and oncolytic herpes simplex virus HF10. Ann Surg Oncol. 21:691–698. 2014. View Article : Google Scholar

149 

Jha BK, Dong B, Nguyen CT, Polyakova I and Silverman RH: Suppression of antiviral innate immunity by sunitinib enhances oncolytic virotherapy. Mol Ther. 21:1749–1757. 2013. View Article : Google Scholar : PubMed/NCBI

150 

Coffey MC, Strong JE, Forsyth PA and Lee PW: Reovirus therapy of tumors with activated Ras pathway. Science. 282:1332–1334. 1998. View Article : Google Scholar : PubMed/NCBI

151 

Gong J and Mita MM: Activated ras signaling pathways and reovirus oncolysis: An update on the mechanism of preferential reovirus replication in cancer cells. Front Oncol. 4:1672014. View Article : Google Scholar : PubMed/NCBI

152 

Cai J, Lin K, Cai W, Lin Y, Liu X, Guo L, Zhang J, Xu W, Lin Z, Wong CW, et al: Tumors driven by RAS signaling harbor a natural vulnerability to oncolytic virus M1. Mol Oncol. 14:3153–3168. 2020. View Article : Google Scholar : PubMed/NCBI

153 

Zhu Z, McGray AJR, Jiang W, Lu B, Kalinski P and Guo ZS: Improving cancer immunotherapy by rationally combining oncolytic virus with modulators targeting key signaling pathways. Mol Cancer. 21:1962022. View Article : Google Scholar : PubMed/NCBI

154 

Hingorani SR: Epithelial and stromal co-evolution and complicity in pancreatic cancer. Nat Rev Cancer. 23:57–77. 2023. View Article : Google Scholar :

155 

Dalin S, Sullivan MR, Lau AN, Grauman-Boss B, Mueller HS, Kreidl E, Fenoglio S, Luengo A, Lees JA, Vander Heiden MG, et al: Deoxycytidine release from pancreatic stellate cells promotes gemcitabine resistance. Cancer Res. 79:5723–5733. 2019. View Article : Google Scholar : PubMed/NCBI

156 

Fischer A and Alsina-Sanchis E: Disturbed endothelial cell signaling in tumor progression and therapy resistance. Curr Opin Cell Biol. 86:1022872024. View Article : Google Scholar

157 

Sun Y: Tumor microenvironment and cancer therapy resistance. Cancer Lett. 380:205–215. 2016. View Article : Google Scholar

158 

Chen L, Xu YX, Wang YS, Ren YY, Dong XM, Wu P, Xie T, Zhang Q and Zhou JL: Prostate cancer microenvironment: Multidimensional regulation of immune cells, vascular system, stromal cells, and microbiota. Mol Cancer. 23:2292024. View Article : Google Scholar : PubMed/NCBI

159 

Zheng Y, Zhou R, Cai J, Yang N, Wen Z, Zhang Z, Sun H, Huang G, Guan Y, Huang N, et al: Matrix stiffness triggers lipid metabolic cross-talk between tumor and stromal cells to mediate bevacizumab resistance in colorectal cancer liver metastases. Cancer Res. 83:3577–3592. 2023. View Article : Google Scholar : PubMed/NCBI

160 

Cescon M, Rampazzo E, Bresolin S, Da Ros F, Manfreda L, Cani A, Della Puppa A, Braghetta P, Bonaldo P and Persano L: Collagen VI sustains cell stemness and chemotherapy resistance in glioblastoma. Cell Mol Life Sci. 80:2332023. View Article : Google Scholar : PubMed/NCBI

161 

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

162 

Guelfi S, Hodivala-Dilke K and Bergers G: Targeting the tumour vasculature: From vessel destruction to promotion. Nat Rev Cancer. 24:655–675. 2024. View Article : Google Scholar : PubMed/NCBI

163 

Wan PK, Ryan AJ and Seymour LW: Beyond cancer cells: Targeting the tumor microenvironment with gene therapy and armed oncolytic virus. Mol Ther. 29:1668–1682. 2021. View Article : Google Scholar : PubMed/NCBI

164 

Miller A, Nace R, Ayala-Breton CC, Steele M, Bailey K, Peng KW and Russell SJ: Perfusion pressure is a critical determinant of the intratumoral extravasation of oncolytic viruses. Mol Ther. 24:306–317. 2016. View Article : Google Scholar :

165 

Hong B, Muili K, Bolyard C, Russell L, Lee TJ, Banasavadi-Siddegowda Y, Yoo JY, Yan Y, Ballester LY, Bockhorst KH and Kaur B: Suppression of HMGB1 released in the glioblastoma tumor microenvironment reduces tumoral edema. Mol Ther Oncolytics. 12:93–102. 2018. View Article : Google Scholar

166 

Tseng JC, Granot T, DiGiacomo V, Levin B and Meruelo D: Enhanced specific delivery and targeting of oncolytic Sindbis viral vectors by modulating vascular leakiness in tumor. Cancer Gene Ther. 17:244–255. 2010. View Article : Google Scholar :

167 

Kurozumi K, Hardcastle J, Thakur R, Yang M, Christoforidis G, Fulci G, Hochberg FH, Weissleder R, Carson W, Chiocca EA and Kaur B: Effect of tumor microenvironment modulation on the efficacy of oncolytic virus therapy. J Natl Cancer Inst. 99:1768–1781. 2007. View Article : Google Scholar : PubMed/NCBI

168 

Liu ZL, Chen HH, Zheng LL, Sun LP and Shi L: Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct Target Ther. 8:1982023. View Article : Google Scholar : PubMed/NCBI

169 

Yousaf I, Kaeppler J, Frost S, Seymour LW and Jacobus EJ: Attenuation of the Hypoxia inducible factor pathway after oncolytic adenovirus infection coincides with decreased vessel perfusion. Cancers (Basel). 12:8512020. View Article : Google Scholar : PubMed/NCBI

170 

Breitbach CJ, De Silva NS, Falls TJ, Aladl U, Evgin L, Paterson J, Sun YY, Roy DG, Rintoul JL, Daneshmand M, et al: Targeting tumor vasculature with an oncolytic virus. Mol Ther. 19:886–894. 2011. View Article : Google Scholar : PubMed/NCBI

171 

Kim M, Nitschké M, Sennino B, Murer P, Schriver BJ, Bell A, Subramanian A, McDonald CE, Wang J, Cha H, et al: Amplification of oncolytic vaccinia virus widespread tumor cell killing by sunitinib through multiple mechanisms. Cancer Res. 78:922–937. 2018. View Article : Google Scholar

172 

Hou W, Chen H, Rojas J, Sampath P and Thorne SH: Oncolytic vaccinia virus demonstrates antiangiogenic effects mediated by targeting of VEGF. Int J Cancer. 135:1238–1246. 2014. View Article : Google Scholar : PubMed/NCBI

173 

Rizzo R, D'Accolti M, Bortolotti D, Caccuri F, Caruso A, Di Luca D and Caselli E: Human Herpesvirus 6A and 6B inhibit in vitro angiogenesis by induction of Human Leukocyte Antigen G. Sci Rep. 8:176832018. View Article : Google Scholar : PubMed/NCBI

174 

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

175 

Nair M, Khosla M, Otani Y, Yeh M, Park F, Shimizu T, Kang JM, Bolyard C, Yu JG, Kumar Banasavadi-Siddegowda Y, et al: Enhancing antitumor efficacy of heavily vascularized tumors by RAMBO virus through decreased tumor endothelial cell activation. Cancers (Basel). 12:10402020. View Article : Google Scholar : PubMed/NCBI

176 

Aghi M, Rabkin SD and Martuza RL: Angiogenic response caused by oncolytic herpes simplex virus-induced reduced thrombospondin expression can be prevented by specific viral mutations or by administering a thrombospondin-derived peptide. Cancer Res. 67:440–444. 2007. View Article : Google Scholar : PubMed/NCBI

177 

Kurozumi K, Hardcastle J, Thakur R, Shroll J, Nowicki M, Otsuki A, Chiocca EA and Kaur B: Oncolytic HSV-1 infection of tumors induces angiogenesis and upregulates CYR61. Mol Ther. 16:1382–1391. 2008. View Article : Google Scholar : PubMed/NCBI

178 

Hassan M, Selimovic D, El-Khattouti A, Soell M, Ghozlan H, Haikel Y, Abdelkader O and Megahed M: Hepatitis C virus-mediated angiogenesis: Molecular mechanisms and therapeutic strategies. World J Gastroenterol. 20:15467–15475. 2014. View Article : Google Scholar : PubMed/NCBI

179 

Wigner-Jeziorska P, Janik-Karpińska E, Niwald M, Saluk J and Miller E: Effect of SARS-CoV-2 infection and BNT162b2 vaccination on the mRNA expression of genes associated with angiogenesis. Int J Mol Sci. 24:160942023. View Article : Google Scholar : PubMed/NCBI

180 

Arulanandam R, Batenchuk C, Angarita FA, Ottolino-Perry K, Cousineau S, Mottashed A, Burgess E, Falls TJ, De Silva N, Tsang J, et al: VEGF-mediated induction of PRD1-BF1/Blimp1 expression sensitizes tumor vasculature to oncolytic virus infection. Cancer Cell. 28:210–224. 2015. View Article : Google Scholar : PubMed/NCBI

181 

Cords L, Engler S, Haberecker M, Rüschoff JH, Moch H, de Souza N and Bodenmiller B: Cancer-associated fibroblast phenotypes are associated with patient outcome in non-small cell lung cancer. Cancer Cell. 42:396–412.e5. 2024. View Article : Google Scholar : PubMed/NCBI

182 

Wang H, Li N, Liu Q, Guo J, Pan Q, Cheng B, Xu J, Dong B, Yang G, Yang B, et al: Antiandrogen treatment induces stromal cell reprogramming to promote castration resistance in prostate cancer. Cancer Cell. 41:1345–1362.e9. 2023. View Article : Google Scholar : PubMed/NCBI

183 

Qi R, Bai Y, Li K, Liu N, Xu Y, Dal E, Wang Y, Lin R, Wang H, Liu Z, et al: Cancer-associated fibroblasts suppress ferroptosis and induce gemcitabine resistance in pancreatic cancer cells by secreting exosome-derived ACSL4-targeting miRNAs. Drug Resist Updat. 68:1009602023. View Article : Google Scholar : PubMed/NCBI

184 

Vähä-Koskela MJ, Kallio JP, Jansson LC, Heikkilä JE, Zakhartchenko VA, Kallajoki MA, Kähäri VM and Hinkkanen AE: Oncolytic capacity of attenuated replicative semliki forest virus in human melanoma xenografts in severe combined immunodeficient mice. Cancer Res. 66:7185–7194. 2066. View Article : Google Scholar

185 

Arwert EN, Milford EL, Rullan A, Derzsi S, Hooper S, Kato T, Mansfield D, Melcher A, Harrington KJ and Sahai E: STING and IRF3 in stromal fibroblasts enable sensing of genomic stress in cancer cells to undermine oncolytic viral therapy. Nat Cell Biol. 22:758–766. 2020. View Article : Google Scholar : PubMed/NCBI

186 

Yasui T, Ohuchida K, Zhao M, Onimaru M, Egami T, Fujita H, Ohtsuka T, Mizumoto K and Tanaka M: Tumor-stroma interactions reduce the efficacy of adenoviral therapy through the HGF-MET pathway. Cancer Sci. 102:484–491. 2011. View Article : Google Scholar

187 

van Asten SD, Raaben M, Nota B and Spaapen RM: Secretome screening reveals fibroblast growth factors as novel inhibitors of viral replication. J Virol. 92:e00260–18. 2018. View Article : Google Scholar : PubMed/NCBI

188 

Hiller BE, Yin Y, Perng YC, de Araujo Castro Í, Fox LE, Locke MC, Monte KJ, López CB, Ornitz DM and Lenschow DJ: Fibroblast growth factor-9 expression in airway epithelial cells amplifies the type I interferon response and alters influenza A virus pathogenesis. PLoS Pathog. 18:e10102282022. View Article : Google Scholar : PubMed/NCBI

189 

Maddaluno L, Urwyler C, Rauschendorfer T, Meyer M, Stefanova D, Spörri R, Wietecha M, Ferrarese L, Stoycheva D, Bender D, et al: Antagonism of interferon signaling by fibroblast growth factors promotes viral replication. EMBO Mol Med. 12:e117932020. View Article : Google Scholar : PubMed/NCBI

190 

Puig-Saus C, Gros A, Alemany R and Cascalló M: Adenovirus i-leader truncation bioselected against cancer-associated fibroblasts to overcome tumor stromal barriers. Mol Ther. 20:54–62. 2012. View Article : Google Scholar :

191 

Kurisu N, Kaminade T, Eguchi M, Ishigami I, Mizuguchi H and Sakurai F: Oncolytic reovirus-mediated killing of mouse cancer-associated fibroblasts. Int J Pharm. 610:1212692021. View Article : Google Scholar : PubMed/NCBI

192 

de Sostoa J, Fajardo CA, Moreno R, Ramos MD, Farrera-Sal M and Alemany R: Targeting the tumor stroma with an oncolytic adenovirus secreting a fibroblast activation protein-targeted bispecific T-cell engager. J Immunother Cancer. 7:192019. View Article : Google Scholar : PubMed/NCBI

193 

Harryvan TJ, Golo M, Dam N, Schoonderwoerd MJA, Farshadi EA, Hornsveld M, Hoeben RC, Hawinkels LJAC and Kemp V: Gastrointestinal cancer-associated fibroblasts expressing Junctional Adhesion Molecule-A are amenable to infection by oncolytic reovirus. Cancer Gene Ther. 29:1918–1929. 2022. View Article : Google Scholar : PubMed/NCBI

194 

Jing Y, Chavez V, Ban Y, Acquavella N, El-Ashry D, Pronin A, Chen X and Merchan JR: Molecular effects of stromal-selective targeting by uPAR-retargeted oncolytic virus in breast cancer. Mol Cancer Res. 15:1410–1420. 2017. View Article : Google Scholar : PubMed/NCBI

195 

Shahvali S, Rahiman N, Jaafari MR and Arabi L: Targeting fibroblast activation protein (FAP): Advances in CAR-T cell, antibody, and vaccine in cancer immunotherapy. Drug Deliv Transl Res. 13:2041–2056. 2023. View Article : Google Scholar : PubMed/NCBI

196 

Al-Obaidi I, Sandhu C, Qureshi B and Seymour LW: The implications of oncolytic viruses targeting fibroblasts in enhancing the antitumoural immune response. Heliyon. 10:e392042024. View Article : Google Scholar : PubMed/NCBI

197 

Chen X and Song E: Turning foes to friends: Targeting cancer-associated fibroblasts. Nat Rev Drug Discov. 18:99–115. 2019. View Article : Google Scholar

198 

Long F, Zhong W, Zhao F, Xu Y, Hu X, Jia G, Huang L, Yi K, Wang N, Si H, et al: DAB2 (+) macrophages support FAP (+) fibroblasts in shaping tumor barrier and inducing poor clinical outcomes in liver cancer. Theranostics. 14:4822–4843. 2024. View Article : Google Scholar : PubMed/NCBI

199 

Yashaswini CN, Qin T, Bhattacharya D, Amor C, Lowe S, Lujambio A, Wang S and Friedman SL: Phenotypes and ontogeny of senescent hepatic stellate cells in metabolic dysfunction-associated steatohepatitis. J Hepatol. 81:207–217. 2024. View Article : Google Scholar : PubMed/NCBI

200 

Yang T, Zhai J, Hu D, Yang R, Wang G, Li Y and Liang G: 'Targeting Design' of nanoparticles in tumor therapy. Pharmaceutics. 14:19192022. View Article : Google Scholar

201 

Guedan S, Rojas JJ, Gros A, Mercade E, Cascallo M and Alemany R: Hyaluronidase expression by an oncolytic adenovirus enhances its intratumoral spread and suppresses tumor growth. Mol Ther. 18:1275–1283. 2010. View Article : Google Scholar : PubMed/NCBI

202 

Fang L, Kong SS, Zhong LK, Wang CM, Liu YJ, Ding HY, Sun J, Zhang YW, Li FZ and Huang P: Asiatic acid enhances intratumor delivery and the antitumor effect of pegylated liposomal doxorubicin by reducing tumor-stroma collagen. Acta Pharmacol Sin. 40:539–545. 2019. View Article : Google Scholar :

203 

Haseley A, Boone S, Wojton J, Yu L, Yoo JY, Yu J, Kurozumi K, Glorioso JC, Caligiuri MA and Kaur B: Extracellular matrix protein CCN1 limits oncolytic efficacy in glioma. Cancer Res. 72:1353–1362. 2012. View Article : Google Scholar : PubMed/NCBI

204 

Maillard L, Ziol M, Tahlil O, Le Feuvre C, Feldman LJ, Branellec D, Bruneval P and Steg P: Pre-treatment with elastase improves the efficiency of percutaneous adenovirus-mediated gene transfer to the arterial media. Gene Ther. 5:1023–1030. 1998. View Article : Google Scholar

205 

Kuriyama N, Kuriyama H, Julin CM, Lamborn KR and Israel MA: Protease pretreatment increases the efficacy of adenovirus-mediated gene therapy for the treatment of an experimental glioblastoma model. Cancer Res. 61:1805–1809. 2001.PubMed/NCBI

206 

Kuriyama N, Kuriyama H, Julin CM, Lamborn K and Israel MA: Pretreatment with protease is a useful experimental strategy for enhancing adenovirus-mediated cancer gene therapy. Hum Gene Ther. 11:2219–2230. 2000. View Article : Google Scholar : PubMed/NCBI

207 

McKee TD, Grandi P, Mok W, Alexandrakis G, Insin N, Zimmer JP, Bawendi MG, Boucher Y, Breakefield XO and Jain RK: Degradation of fibrillar collagen in a human melanoma xenograft improves the efficacy of an oncolytic herpes simplex virus vector. Cancer Res. 66:2509–2513. 2006. View Article : Google Scholar : PubMed/NCBI

208 

Bhattacharyya M, Jariyal H and Srivastava A: Hyaluronic acid: More than a carrier, having an overpowering extracellular and intracellular impact on cancer. Carbohydr Polym. 317:1210812023. View Article : Google Scholar : PubMed/NCBI

209 

Ganesh S, Gonzalez-Edick M, Gibbons D, Van Roey M and Jooss K: Intratumoral coadministration of hyaluronidase enzyme and oncolytic adenoviruses enhances virus potency in metastatic tumor models. Clin Cancer Res. 14:3933–3941. 2008. View Article : Google Scholar : PubMed/NCBI

210 

Samuel CS, Hewitson TD, Unemori EN and Tang ML: Drugs of the future: the hormone relaxin. Cell Mol Life Sci. 64:1539–1557. 2007. View Article : Google Scholar : PubMed/NCBI

211 

Perentes JY, McKee TD, Ley CD, Mathiew H, Dawson M, Padera TP, Munn LL, Jain RK and Boucher Y: In vivo imaging of extracellular matrix remodeling by tumor-associated fibroblasts. Nat Methods. 6:143–145. 2009. View Article : Google Scholar : PubMed/NCBI

212 

Kim JH, Lee YS, Kim H, Huang JH, Yoon AR and Yun CO: Relaxin expression from tumor-targeting adenoviruses and its intratumoral spread, apoptosis induction, and efficacy. J Natl Cancer Inst. 98:1482–1493. 2006. View Article : Google Scholar : PubMed/NCBI

213 

Kim Y, Lee HG, Dmitrieva N, Kim J, Kaur B and Friedman A: Choindroitinase ABC I-mediated enhancement of oncolytic virus spread and anti tumor efficacy: A mathematical model. PLoS One. 9:e1024992014. View Article : Google Scholar : PubMed/NCBI

214 

Mok W, Boucher Y and Jain RK: Matrix metalloproteinases-1 and -8 improve the distribution and efficacy of an oncolytic virus. Cancer Res. 67:10664–10668. 2007. View Article : Google Scholar : PubMed/NCBI

215 

Hong CS, Fellows W, Niranjan A, Alber S, Watkins S, Cohen JB, Glorioso JC and Grandi P: Ectopic matrix metalloproteinase-9 expression in human brain tumor cells enhances oncolytic HSV vector infection. Gene Ther. 17:1200–1205. 2010. View Article : Google Scholar : PubMed/NCBI

216 

Yumul R, Richter M, Lu ZZ, Saydaminova K, Wang H, Wang CH, Carter D and Lieber A: Epithelial junction opener improves oncolytic adenovirus therapy in mouse tumor models. Hum Gene Ther. 27:325–337. 2016. View Article : Google Scholar : PubMed/NCBI

217 

Binder C, Simon A, Binder L, Hagemann T, Schulz M, Emons G, Trümper L and Einspanier A: Elevated concentrations of serum relaxin are associated with metastatic disease in breast cancer patients. Breast Cancer Res Treat. 87:157–166. 2004. View Article : Google Scholar : PubMed/NCBI

218 

Daginakatte GC and Gutmann DH: Neurofibromatosis-1 (Nf1) heterozygous brain microglia elaborate paracrine factors that promote Nf1-deficient astrocyte and glioma growth. Hum Mol Genet. 16:1098–1112. 2007. View Article : Google Scholar : PubMed/NCBI

219 

Dmitrieva N, Yu L, Viapiano M, Cripe TP, Chiocca EA, Glorioso JC and Kaur B: Chondroitinase ABC I-mediated enhancement of oncolytic virus spread and antitumor efficacy. Clin Cancer Res. 17:1362–1372. 2011. View Article : Google Scholar

220 

Kanzaki A, Kasuya H, Yamamura K, Sahin TT, Nomura N, Shikano T, Shirota T, Tan G, Fukuda S, Misawa M, et al: Antitumor efficacy of oncolytic herpes simplex virus adsorbed onto antigen-specific lymphocytes. Cancer Gene Ther. 19:292–298. 2012. View Article : Google Scholar : PubMed/NCBI

221 

Netti PA, Berk DA, Swartz MA, Grodzinsky AJ and Jain RK: Role of extracellular matrix assembly in interstitial transport in solid tumors. Cancer Res. 60:2497–2503. 2000.PubMed/NCBI

222 

Yokoda R, Nagalo BM, Vernon B, Oklu R, Albadawi H, DeLeon TT, Zhou Y, Egan JB, Duda DG and Borad MJ: Oncolytic virus delivery: From nano-pharmacodynamics to enhanced oncolytic effect. Oncolytic Virother. 6:39–49. 2017. View Article : Google Scholar : PubMed/NCBI

223 

Kwan A, Winder N and Muthana M: Oncolytic virotherapy treatment of breast cancer: Barriers and recent advances. Viruses. 13:11282021. View Article : Google Scholar : PubMed/NCBI

224 

Fisher KD, Stallwood Y, Green NK, Ulbrich K, Mautner V and Seymour LW: Polymer-coated adenovirus permits efficient retargeting and evades neutralising antibodies. Gene Ther. 8:341–348. 2001. View Article : Google Scholar : PubMed/NCBI

225 

Wang J, Guo C, Wang XY and Yang H: 'Double-punch' strategy for delivery of viral immunotherapy with prolonged tumor retention and enhanced transfection efficacy. J Control Release. 329:328–336. 2021. View Article : Google Scholar

226 

Thambi T, Hong J, Yoon AR and Yun CO: Challenges and progress toward tumor-targeted therapy by systemic delivery of polymer-complexed oncolytic adenoviruses. Cancer Gene Ther. 29:1321–1331. 2022. View Article : Google Scholar : PubMed/NCBI

227 

Kim J, Kim PH, Kim SW and Yun CO: Enhancing the therapeutic efficacy of adenovirus in combination with biomaterials. Biomaterials. 33:1838–1850. 2012. View Article : Google Scholar

228 

Howard F and Muthana M: Designer nanocarriers for navigating the systemic delivery of oncolytic viruses. Nanomedicine (Lond). 15:93–110. 2020. View Article : Google Scholar

229 

Almstätter I, Mykhaylyk O, Settles M, Altomonte J, Aichler M, Walch A, Rummeny EJ, Ebert O, Plank C and Braren R: Characterization of magnetic viral complexes for targeted delivery in oncology. Theranostics. 5:667–685. 2015. View Article : Google Scholar : PubMed/NCBI

230 

Lara HH, Ayala-Nuñez NV, Ixtepan-Turrent L and Rodriguez-Padilla C: Mode of antiviral action of silver nanoparticles against HIV-1. J Nanobiotechnology. 8:12010. View Article : Google Scholar : PubMed/NCBI

231 

Ran L, Tan X, Li Y, Zhang H, Ma R, Ji T, Dong W, Tong T, Liu Y, Chen D, et al: Delivery of oncolytic adenovirus into the nucleus of tumorigenic cells by tumor microparticles for virotherapy. Biomaterials. 89:56–66. 2016. View Article : Google Scholar : PubMed/NCBI

232 

Hong Y, Kim YK, Kim GB, Nam GH, Kim SA, Park Y, Yang Y and Kim IS: Degradation of tumour stromal hyaluronan by small extracellular vesicle-PH20 stimulates CD103(+) dendritic cells and in combination with PD-L1 blockade boosts anti-tumour immunity. J Extracell Vesicles. 8:16708932019. View Article : Google Scholar : PubMed/NCBI

233 

Liu L, Liu W, Han Z, Shan Y, Xie Y, Wang J, Qi H and Xu Q: Extracellular Vesicles-in-Hydrogel (EViH) targeting pathophysiology for tissue repair. Bioact Mater. 44:283–318. 2024.PubMed/NCBI

234 

Jung BK, Ko HY, Kang H, Hong J, Ahn HM, Na Y, Kim H, Kim JS and Yun CO: Relaxin-expressing oncolytic adenovirus induces remodeling of physical and immunological aspects of cold tumor to potentiate PD-1 blockade. J Immunother Cancer. 8:e0007632020. View Article : Google Scholar : PubMed/NCBI

235 

Wang S, Li Y, Xu C, Dong J and Wei J: An oncolytic vaccinia virus encoding hyaluronidase reshapes the extracellular matrix to enhance cancer chemotherapy and immunotherapy. J Immunother Cancer. 12:e0084312024. View Article : Google Scholar : PubMed/NCBI

236 

Farrera-Sal M, Moreno R, Mato-Berciano A, Maliandi MV, Bazan-Peregrino M and Alemany R: Hyaluronidase expression within tumors increases virotherapy efficacy and T cell accumulation. Mol Ther Oncolytics. 22:27–35. 2021. View Article : Google Scholar : PubMed/NCBI

237 

Schäfer S, Weibel S, Donat U, Zhang Q, Aguilar RJ, Chen NG and Szalay AA: Vaccinia virus-mediated intra-tumoral expression of matrix metalloproteinase 9 enhances oncolysis of PC-3 xenograft tumors. BMC Cancer. 12:3662012. View Article : Google Scholar : PubMed/NCBI

238 

Swanner J, Shim JS, Rivera-Caraballo KA, Vázquez-Arreguín K, Hong B, Bueso-Perez AJ, Lee TJ, Banasavadi-Siddegowda YK, Kaur B and Yoo JY: esRAGE-expressing oHSV enhances anti-tumor efficacy by inhibition of endothelial cell activation. Mol Ther Oncolytics. 28:171–181. 2023. View Article : Google Scholar : PubMed/NCBI

239 

Jeong SN and Yoo SY: Novel oncolytic virus armed with cancer suicide gene and normal vasculogenic gene for improved anti-tumor activity. Cancers (Basel). 12:10702020. View Article : Google Scholar : PubMed/NCBI

240 

Al-Zaher AA, Moreno R, Fajardo CA, Arias-Badia M, Farrera M, de Sostoa J, Rojas LA and Alemany R: Evidence of anti-tumoral efficacy in an immune competent setting with an iRGD-Modified hyaluronidase-armed oncolytic adenovirus. Mol Ther Oncolytics. 8:62–70. 2018. View Article : Google Scholar : PubMed/NCBI

241 

Puig-Saus C, Rojas LA, Laborda E, Figueras A, Alba R, Fillat C and Alemany R: iRGD tumor-penetrating peptide-modified oncolytic adenovirus shows enhanced tumor transduction, intratumoral dissemination and antitumor efficacy. Gene Ther. 21:767–774. 2014. View Article : Google Scholar : PubMed/NCBI

242 

Everts A, Bergeman M, McFadden G and Kemp V: Simultaneous tumor and stroma targeting by oncolytic viruses. Biomedicines. 8:4742020. View Article : Google Scholar : PubMed/NCBI

243 

Lavilla-Alonso S, Bauer MM, Abo-Ramadan U, Ristimäki A, Halavaara J, Desmond RA, Wang D, Escutenaire S, Ahtiainen L, Saksela K, et al: Macrophage metalloelastase (MME) as adjuvant for intra-tumoral injection of oncolytic adenovirus and its influence on metastases development. Cancer Gene Ther. 19:126–134. 2012. View Article : Google Scholar

244 

Hu J, Chen C, Lu R, Zhang Y, Wang Y, Hu Q, Li W, Wang S, Jing O, Yi H, et al: β-adrenergic receptor inhibitor and oncolytic herpesvirus combination therapy shows enhanced antitumoral and antiangiogenic effects on colorectal cancer. Front Pharmacol. 12:7352782021. View Article : Google Scholar

245 

Thaci B, Ulasov IV, Ahmed AU, Ferguson SD, Han Y and Lesniak MS: Anti-angiogenic therapy increases intratumoral adenovirus distribution by inducing collagen degradation. Gene Ther. 20:318–327. 2013. View Article : Google Scholar

246 

Ikeda K, Ichikawa T, Wakimoto H, Silver JS, Deisboeck TS, Finkelstein D, Harsh GR IV, Louis DN, Bartus RT, Hochberg FH and Chiocca EA: Oncolytic virus therapy of multiple tumors in the brain requires suppression of innate and elicited antiviral responses. Nat Med. 5:881–887. 1999. View Article : Google Scholar : PubMed/NCBI

247 

Ikeda K, Wakimoto H, Ichikawa T, Jhung S, Hochberg FH, Louis DN and Chiocca EA: Complement depletion facilitates the infection of multiple brain tumors by an intravascular, replication-conditional herpes simplex virus mutant. J Virol. 74:4765–4775. 2000. View Article : Google Scholar : PubMed/NCBI

248 

Carlisle R, Choi J, Bazan-Peregrino M, Laga R, Subr V, Kostka L, Ulbrich K, Coussios CC and Seymour LW: Enhanced tumor uptake and penetration of virotherapy using polymer stealthing and focused ultrasound. J Natl Cancer Inst. 105:1701–1710. 2013. View Article : Google Scholar : PubMed/NCBI

249 

Bazan-Peregrino M, Arvanitis CD, Rifai B, Seymour LW and Coussios CC: Ultrasound-induced cavitation enhances the delivery and therapeutic efficacy of an oncolytic virus in an in vitro model. J Control Release. 157:235–242. 2012. View Article : Google Scholar

250 

Sun S, Liu Y, He C, Hu W, Liu W, Huang X, Wu J, Xie F, Chen C, Wang J, et al: Combining NanoKnife with M1 oncolytic virus enhances anticancer activity in pancreatic cancer. Cancer Lett. 502:9–24. 2021. View Article : Google Scholar : PubMed/NCBI

251 

Otani Y, Yoo JY, Shimizu T, Kurozumi K, Date I and Kaur B: Implications of immune cells in oncolytic herpes simplex virotherapy for glioma. Brain Tumor Pathol. 39:57–64. 2022. View Article : Google Scholar : PubMed/NCBI

252 

Donat U, Weibel S, Hess M, Stritzker J, Härtl B, Sturm JB, Chen NG, Gentschev I and Szalay AA: Preferential colonization of metastases by oncolytic vaccinia virus strain GLV-1h68 in a human PC-3 prostate cancer model in nude mice. PLoS One. 7:e459422012. View Article : Google Scholar : PubMed/NCBI

253 

Lynch C, Pitroda SP and Weichselbaum RR: Radiotherapy, immunity, and immune checkpoint inhibitors. Lancet Oncol. 25:e352–e362. 2024. View Article : Google Scholar : PubMed/NCBI

254 

Munir AZ, Gutierrez A, Qin J, Lichtman AH and Moslehi JJ: Immune-checkpoint inhibitor-mediated myocarditis: CTLA4, PD1 and LAG3 in the heart. Nat Rev Cancer. 24:540–553. 2024. View Article : Google Scholar : PubMed/NCBI

255 

Kwan A, Winder N, Atkinson E, Al-Janabi H, Allen RJ, Hughes R, Moamin M, Louie R, Evans D, Hutchinson M, et al: Macrophages Mediate the Antitumor Effects of the Oncolytic Virus HSV1716 in Mammary Tumors. Mol Cancer Ther. 20:589–601. 2021. View Article : Google Scholar

256 

Bommareddy PK, Aspromonte S, Zloza A, Rabkin SD and Kaufman HL: MEK inhibition enhances oncolytic virus immunotherapy through increased tumor cell killing and T cell activation. Sci Transl Med. 10:eaau04172018. View Article : Google Scholar : PubMed/NCBI

257 

Mostafa AA, Meyers DE, Thirukkumaran CM, Liu PJ, Gratton K, Spurrell J, Shi Q, Thakur S and Morris DG: Oncolytic reovirus and immune checkpoint inhibition as a novel immunotherapeutic strategy for breast cancer. Cancers (Basel). 10:2052018. View Article : Google Scholar : PubMed/NCBI

258 

Burke S, Shergold A, Elder MJ, Whitworth J, Cheng X, Jin H, Wilkinson RW, Harper J and Carroll DK: Oncolytic Newcastle disease virus activation of the innate immune response and priming of antitumor adaptive responses in vitro. Cancer Immunol Immunother. 69:1015–1027. 2020. View Article : Google Scholar : PubMed/NCBI

259 

Chon HJ, Lee WS, Yang H, Kong SJ, Lee NK, Moon ES, Choi J, Han EC, Kim JH, Ahn JB, et al: Tumor microenvironment remodeling by intratumoral oncolytic vaccinia virus enhances the efficacy of immune-checkpoint blockade. Clin Cancer Res. 25:1612–1623. 2019. View Article : Google Scholar

260 

Lovatt C and Parker AL: Oncolytic viruses and immune checkpoint inhibitors: The 'Hot' new power couple. Cancers (Basel). 15:41782023. View Article : Google Scholar

261 

Bernstock JD, Vicario N, Rong L, Valdes PA, Choi BD, Chen JA, DiToro D, Osorio DS, Kachurak K, Gessler F, et al: A novel in situ multiplex immunofluorescence panel for the assessment of tumor immunopathology and response to virotherapy in pediatric glioblastoma reveals a role for checkpoint protein inhibition. Oncoimmunology. 8:e16789212019. View Article : Google Scholar : PubMed/NCBI

262 

Annels NE, Mansfield D, Arif M, Ballesteros-Merino C, Simpson GR, Denyer M, Sandhu SS, Melcher AA, Harrington KJ, Davies B, et al: Phase I Trial of an ICAM-1-targeted immunotherapeutic-coxsackievirus A21 (CVA21) as an oncolytic agent against non muscle-invasive bladder cancer. Clin Cancer Res. 25:5818–5831. 2019. View Article : Google Scholar : PubMed/NCBI

263 

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

264 

Saha D, Martuza RL and Rabkin SD: Macrophage polarization contributes to glioblastoma eradication by combination immunovirotherapy and immune checkpoint blockade. Cancer Cell. 32:253–267.e5. 2017. View Article : Google Scholar : PubMed/NCBI

265 

Liang Y, Wang B, Chen Q, Fu X, Jiang C, Lin Z, Zhuang Q, Zeng Y, Liu X and Zhang D: Systemic delivery of glycosylated-PEG-masked oncolytic virus enhances targeting of antitumor immuno-virotherapy and modulates T and NK cell infiltration. Theranostics. 13:5452–5468. 2023. View Article : Google Scholar : PubMed/NCBI

266 

Patel MR, Dash A, Jacobson BA, Ji Y, Baumann D, Ismail K and Kratzke RA: JAK/STAT inhibition with ruxolitinib enhances oncolytic virotherapy in non-small cell lung cancer models. Cancer Gene Ther. 26:411–418. 2019. View Article : Google Scholar : PubMed/NCBI

267 

Nguyen TT, Ramsay L, Ahanfeshar-Adams M, Lajoie M, Schadendorf D, Alain T and Watson IR: Mutations in the IFNγ-JAK-STAT pathway causing resistance to immune checkpoint inhibitors in melanoma increase sensitivity to oncolytic virus treatment. Clin Cancer Res. 27:3432–3442. 2021. View Article : Google Scholar : PubMed/NCBI

268 

Lecoultre M, Walker PR and El Helali A: Oncolytic virus and tumor-associated macrophage interactions in cancer immunotherapy. Clin Exp Med. 24:2022024. View Article : Google Scholar : PubMed/NCBI

269 

Fujikura Y, Kudlackova P, Vokurka M, Krijt J and Melkova Z: The effect of nitric oxide on vaccinia virus-encoded ribonucleotide reductase. Nitric Oxide. 20:114–121. 2009. View Article : Google Scholar

270 

Pittet MJ, Michielin O and Migliorini D: Clinical relevance of tumour-associated macrophages. Nat Rev Clin Oncol. 19:402–421. 2022. View Article : Google Scholar : PubMed/NCBI

271 

Meisen WH, Wohleb ES, Jaime-Ramirez AC, Bolyard C, Yoo JY, Russell L, Hardcastle J, Dubin S, Muili K, Yu J, et al: The impact of macrophage- and microglia-secreted TNFα on Oncolytic HSV-1 therapy in the glioblastoma tumor microenvironment. Clin Cancer Res. 21:3274–3285. 2015. View Article : Google Scholar : PubMed/NCBI

272 

van den Bossche WBL, Kleijn A, Teunissen CE, Voerman JSA, Teodosio C, Noske DP, van Dongen JJM, Dirven CMF and Lamfers MLM: Oncolytic virotherapy in glioblastoma patients induces a tumor macrophage phenotypic shift leading to an altered glioblastoma microenvironment. Neuro Oncol. 20:1494–1504. 2018. View Article : Google Scholar : PubMed/NCBI

273 

Kober C, Rohn S, Weibel S, Geissinger U, Chen NG and Szalay AA: Microglia and astrocytes attenuate the replication of the oncolytic vaccinia virus LIVP 1.1.1 in murine GL261 gliomas by acting as vaccinia virus traps. J Transl Med. 13:2162015. View Article : Google Scholar : PubMed/NCBI

274 

Liu YP, Suksanpaisan L, Steele MB, Russell SJ and Peng KW: Induction of antiviral genes by the tumor microenvironment confers resistance to virotherapy. Sci Rep. 3:23752013. View Article : Google Scholar : PubMed/NCBI

275 

Zhang Q and Liu F: Advances and potential pitfalls of oncolytic viruses expressing immunomodulatory transgene therapy for malignant gliomas. Cell Death Dis. 11:4852020. View Article : Google Scholar : PubMed/NCBI

276 

Shen Z, Liu X, Fan G, Na J, Liu Q, Lin F, Zhang Z and Zhong L: Improving the therapeutic efficacy of oncolytic viruses for cancer: Targeting macrophages. J Transl Med. 21:8422023. View Article : Google Scholar : PubMed/NCBI

277 

Blitz SE, Kappel AD, Gessler FA, Klinger NV, Arnaout O, Lu Y, Peruzzi PP, Smith TR, Chiocca EA, Friedman GK and Bernstock JD: Tumor-associated macrophages/microglia in glioblastoma oncolytic virotherapy: A double-edged sword. Int J Mol Sci. 23:18082022. View Article : Google Scholar : PubMed/NCBI

278 

Ferguson MS, Chard Dunmall LS, Gangeswaran R, Marelli G, Tysome JR, Burns E, Whitehead MA, Aksoy E, Alusi G, Hiley C, et al: Transient inhibition of PI3Kδ enhances the therapeutic effect of intravenous delivery of oncolytic vaccinia virus. Mol Ther. 28:1263–1275. 2020. View Article : Google Scholar : PubMed/NCBI

279 

Lee TJ, Nair M, Banasavadi-Siddegowda Y, Liu J, Nallanagulagari T, Jaime-Ramirez AC, Guo JY, Quadri H, Zhang J, Bockhorst KH, et al: Enhancing therapeutic efficacy of oncolytic herpes simplex virus-1 with integrin β1 blocking antibody OS2966. Mol Cancer Ther. 18:1127–1136. 2019. View Article : Google Scholar : PubMed/NCBI

280 

Thorne AH, Meisen WH, Russell L, Yoo JY, Bolyard CM, Lathia JD, Rich J, Puduvalli VK, Mao H, Yu J, et al: Role of cysteine-rich 61 protein (CCN1) in macrophage-mediated oncolytic herpes simplex virus clearance. Mol Ther. 22:1678–1687. 2014. View Article : Google Scholar : PubMed/NCBI

281 

Zhang KL, Li RP, Zhang BP, Gao ST, Li B, Huang CJ, Cao R, Cheng JY, Xie XD, Yu ZH and Feng XY: Efficacy of a new oncolytic adenovirus armed with IL-13 against oral carcinoma models. Onco Targets Ther. 12:6515–6523. 2019. View Article : Google Scholar : PubMed/NCBI

282 

Hedrick CC and Malanchi I: Neutrophils in cancer: Heterogeneous and multifaceted. Nat Rev Immunol. 22:173–187. 2022. View Article : Google Scholar

283 

Ng MSF, Kwok I, Tan L, Shi C, Cerezo-Wallis D, Tan Y, Leong K, Calvo GF, Yang K, Zhang Y, et al: Deterministic reprogramming of neutrophils within tumors. Science. 383:eadf64932024. View Article : Google Scholar : PubMed/NCBI

284 

Andzinski L, Kasnitz N, Stahnke S, Wu CF, Gereke M, von Köckritz-Blickwede M, Schilling B, Brandau S, Weiss S and Jablonska J: Type I IFNs induce anti-tumor polarization of tumor associated neutrophils in mice and human. Int J Cancer. 138:1982–1993. 2016. View Article : Google Scholar

285 

Naumenko V, Turk M, Jenne CN and Kim SJ: Neutrophils in viral infection. Cell Tissue Res. 371:505–516. 2018. View Article : Google Scholar : PubMed/NCBI

286 

Taifour T, Attalla SS, Zuo D, Gu Y, Sanguin-Gendreau V, Proud H, Solymoss E, Bui T, Kuasne H, Papavasiliou V, et al: The tumor-derived cytokine Chi3l1 induces neutrophil extracellular traps that promote T cell exclusion in triple-negative breast cancer. Immunity. 56:2755–2772.e8. 2023. View Article : Google Scholar : PubMed/NCBI

287 

He XY, Gao Y, Ng D, Michalopoulou E, George S, Adrover JM, Sun L, Albrengues J, Daßler-Plenker J, Han X, et al: Chronic stress increases metastasis via neutrophil-mediated changes to the microenvironment. Cancer Cell. 42:474–486.e12. 2024. View Article : Google Scholar : PubMed/NCBI

288 

Zhou D, Zhang C, Sun J and Yuan M: Neutrophils in oncolytic virus immunotherapy. Front Immunol. 15:14904142024. View Article : Google Scholar : PubMed/NCBI

289 

Dai W, Tian R, Yu L, Bian S, Chen Y, Yin B, Luan Y, Chen S, Fan Z, Yan R, et al: Overcoming therapeutic resistance in oncolytic herpes virotherapy by targeting IGF2BP3-induced NETosis in malignant glioma. Nat Commun. 15:1312024. View Article : Google Scholar : PubMed/NCBI

290 

Price PJ, Luckow B, Torres-Domínguez LE, Brandmüller C, Zorn J, Kirschning CJ, Sutter G and Lehmann MH: Chemokine (C-C Motif) receptor 1 is required for efficient recruitment of neutrophils during respiratory infection with modified vaccinia virus Ankara. J Virol. 88:10840–10850. 2014. View Article : Google Scholar : PubMed/NCBI

291 

West BC, Escheté ML, Cox ME and King JW: Neutrophil uptake of vaccinia virus in vitro. J Infect Dis. 156:597–606. 1987. View Article : Google Scholar : PubMed/NCBI

292 

Breitbach CJ, Paterson JM, Lemay CG, Falls TJ, McGuire A, Parato KA, Stojdl DF, Daneshmand M, Speth K, Kirn D, et al: Targeted inflammation during oncolytic virus therapy severely compromises tumor blood flow. Mol Ther. 15:1686–1693. 2007. View Article : Google Scholar : PubMed/NCBI

293 

Taipale K, Liikanen I, Koski A, Heiskanen R, Kanerva A, Hemminki O, Oksanen M, Grönberg-Vähä-Koskela S, Hemminki K, Joensuu T and Hemminki A: Predictive and prognostic clinical variables in cancer patients treated with adenoviral oncolytic immunotherapy. Mol Ther. 24:1323–1332. 2016. View Article : Google Scholar : PubMed/NCBI

294 

He CB and Lin XJ: Inflammation scores predict the survival of patients with hepatocellular carcinoma who were treated with transarterial chemoembolization and recombinant human type-5 adenovirus H101. PLoS One. 12:e01747692017. View Article : Google Scholar : PubMed/NCBI

295 

Zhou D, Xu W, Ding X, Guo H, Wang J, Zhao G, Zhang C, Zhang Z, Wang Z, Wang P, et al: Transient inhibition of neutrophil functions enhances the antitumor effect of intravenously delivered oncolytic vaccinia virus. Cancer Sci. 115:1129–1140. 2024. View Article : Google Scholar : PubMed/NCBI

296 

Fu X, Tao L, Rivera A, Xu H and Zhang X: Virotherapy induces massive infiltration of neutrophils in a subset of tumors defined by a strong endogenous interferon response activity. Cancer Gene Ther. 18:785–794. 2011. View Article : Google Scholar : PubMed/NCBI

297 

Minott JA, van Vloten JP, Chan L, Mehrani Y, Bridle BW and Karimi K: The role of neutrophils in oncolytic orf virus-mediated cancer immunotherapy. Cells. 11:28582018. View Article : Google Scholar

298 

Liu Y, Xu C, Xiao X, Chen Y, Wang X, Liu W, Tan Y, Zhu W, Hu J, Liang J, et al: Overcoming resistance to oncolytic virus M1 by targeting PI3K-γ in tumor-associated myeloid cells. Mol Ther. 30:3677–3693. 2022. View Article : Google Scholar : PubMed/NCBI

299 

Bulstrode H, Girdler GC, Gracia T, Aivazidis A, Moutsopoulos I, Young AMH, Hancock J, He X, Ridley K, Xu Z, et al: Myeloid cell interferon secretion restricts Zika flavivirus infection of developing and malignant human neural progenitor cells. Neuron. 110:3936–3951.e10. 2022. View Article : Google Scholar : PubMed/NCBI

300 

Noh MH, Kang JM, Miller AA, Nguyen G, Huang M, Shim JS, Bueso-Perez AJ, Murphy SA, Rivera-Caraballo KA, Otani Y, et al: Targeting IGF2 to reprogram the tumor microenvironment for enhanced viro-immunotherapy. Neuro Oncol. 26:1602–1616. 2024. View Article : Google Scholar : PubMed/NCBI

301 

Bald T, Krummel MF, Smyth MJ and Barry KC: The NK cell-cancer cycle: Advances and new challenges in NK cell-based immunotherapies. Nat Immunol. 21:835–847. 2020. View Article : Google Scholar : PubMed/NCBI

302 

Alvarez-Breckenridge CA, Yu J, Price R, Wojton J, Pradarelli J, Mao H, Wei M, Wang Y, He S, Hardcastle J, et al: NK cells impede glioblastoma virotherapy through NKp30 and NKp46 natural cytotoxicity receptors. Nat Med. 18:1827–1834. 2012. View Article : Google Scholar : PubMed/NCBI

303 

Reale A, Gatta A, Shaik AKB, Shallak M, Chiaravalli AM, Cerati M, Zaccaria M, La Rosa S, Calistri A, Accolla RS and Forlani G: An oncolytic HSV-1 vector induces a therapeutic adaptive immune response against glioblastoma. J Transl Med. 22:8622024. View Article : Google Scholar : PubMed/NCBI

304 

Kim Y, Yoo JY, Lee TJ, Liu J, Yu J, Caligiuri MA, Kaur B and Friedman A: Complex role of NK cells in regulation of oncolytic virus-bortezomib therapy. Proc Natl Acad Sci USA. 115:4927–4932. 2018. View Article : Google Scholar : PubMed/NCBI

305 

Varudkar N, Oyer JL, Copik A and Parks GD: Oncolytic parainfluenza virus combines with NK cells to mediate killing of infected and non-infected lung cancer cells within 3D spheroids: role of type I and type III interferon signaling. J Immunother Cancer. 9:e0023732021. View Article : Google Scholar : PubMed/NCBI

306 

Wang Y, Jin J, Li Y, Zhou Q, Yao R, Wu Z, Hu H, Fang Z, Dong S, Cai Q, et al: NK cell tumor therapy modulated by UV-inactivated oncolytic herpes simplex virus type 2 and checkpoint inhibitors. Transl Res. 240:64–86. 2022. View Article : Google Scholar

307 

Yan Y, Li S, Jia T, Du X, Xu Y, Zhao Y, Li L, Liang K, Liang W, Sun H and Li R: Combined therapy with CTL cells and oncolytic adenovirus expressing IL-15-induced enhanced antitumor activity. Tumour Biol. 36:4535–4543. 2015. View Article : Google Scholar : PubMed/NCBI

308 

Alvarez-Breckenridge CA, Yu J, Price R, Wei M, Wang Y, Nowicki MO, Ha YP, Bergin S, Hwang C, Fernandez SA, et al: The histone deacetylase inhibitor valproic acid lessens NK cell action against oncolytic virus-infected glioblastoma cells by inhibition of STAT5/T-BET signaling and generation of gamma interferon. J Virol. 86:4566–4577. 2012. View Article : Google Scholar : PubMed/NCBI

309 

Altomonte J, Wu L, Meseck M, Chen L, Ebert O, Garcia-Sastre A, Fallon J, Mandeli J and Woo SL: Enhanced oncolytic potency of vesicular stomatitis virus through vector-mediated inhibition of NK and NKT cells. Cancer Gene Ther. 16:266–278. 2009. View Article : Google Scholar

310 

Han J, Chen X, Chu J, Xu B, Meisen WH, Chen L, Zhang L, Zhang J, He X, Wang QE, et al: TGFβ treatment enhances glioblastoma virotherapy by inhibiting the innate immune response. Cancer Res. 75:5273–5282. 2015. View Article : Google Scholar : PubMed/NCBI

311 

Kurmasheva N, Said A, Wong B, Kinderman P, Han X, Rahimic AHF, Kress A, Carter-Timofte ME, Holm E, van der Horst D, et al: Octyl itaconate enhances VSVdelta51 oncolytic virotherapy by multitarget inhibition of antiviral and inflammatory pathways. Nat Commun. 15:40962024. View Article : Google Scholar

312 

Taverner WK, Jacobus EJ, Christianson J, Champion B, Paton AW, Paton JC, Su W, Cawood R, Seymour LW and Lei-Rossmann J: Calcium influx caused by ER stress inducers enhances oncolytic adenovirus enadenotucirev replication and killing through PKCα activation. Mol Ther Oncolytics. 15:117–130. 2019. View Article : Google Scholar :

313 

Bhatt DK, Janzen T, Daemen T and Weissing FJ: Modelling the spatial dynamics of oncolytic virotherapy in the presence of virus-resistant tumour cells. PLoS Comput Biol. 18:e10100762022. View Article : Google Scholar : PubMed/NCBI

314 

Kloker LD, Yurttas C and Lauer UM: Three-dimensional tumor cell cultures employed in virotherapy research. Oncolytic Virother. 7:79–93. 2018. View Article : Google Scholar : PubMed/NCBI

315 

Brachtlova T, Li J, van der Meulen-Muileman IH, Sluiter F, von Meijenfeldt W, Witte I, Massaar S, van den Oever R, de Vrij J and van Beusechem VW: Quantitative virus-associated RNA detection to monitor oncolytic adenovirus replication. Int J Mol Sci. 25:65512024. View Article : Google Scholar : PubMed/NCBI

316 

Karnik I, Her Z, Neo SH, Liu WN and Chen Q: Emerging preclinical applications of humanized mouse models in the discovery and validation of novel immunotherapeutics and their mechanisms of action for improved cancer treatment. Pharmaceutics. 15:16002023. View Article : Google Scholar : PubMed/NCBI

317 

Ambegoda P, Wei HC and Jang SR: The role of immune cells in resistance to oncolytic viral therapy. Math Biosci Eng. 21:5900–5946. 2024. View Article : Google Scholar : PubMed/NCBI

318 

West J, Robertson-Tessi M and Anderson ARA: Agent-based methods facilitate integrative science in cancer. Trends Cell Biol. 33:300–311. 2023. View Article : Google Scholar

319 

Sachak-Patwa R, Lafferty EI, Schmit CJ, Thompson RN and Byrne HM: A target-cell limited model can reproduce influenza infection dynamics in hosts with differing immune responses. J Theor Biol. 567:1114912023. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2025
Volume 56 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu J, Xia Z, Wang S and Xia Q: Resistance to oncolytic virotherapy: Multidimensional mechanisms and therapeutic breakthroughs (Review). Int J Mol Med 56: 171, 2025.
APA
Xu, J., Xia, Z., Wang, S., & Xia, Q. (2025). Resistance to oncolytic virotherapy: Multidimensional mechanisms and therapeutic breakthroughs (Review). International Journal of Molecular Medicine, 56, 171. https://doi.org/10.3892/ijmm.2025.5612
MLA
Xu, J., Xia, Z., Wang, S., Xia, Q."Resistance to oncolytic virotherapy: Multidimensional mechanisms and therapeutic breakthroughs (Review)". International Journal of Molecular Medicine 56.5 (2025): 171.
Chicago
Xu, J., Xia, Z., Wang, S., Xia, Q."Resistance to oncolytic virotherapy: Multidimensional mechanisms and therapeutic breakthroughs (Review)". International Journal of Molecular Medicine 56, no. 5 (2025): 171. https://doi.org/10.3892/ijmm.2025.5612