Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Oncology
Join Editorial Board Propose a Special Issue
Print ISSN: 1019-6439 Online ISSN: 1791-2423
Journal Cover
February-2026 Volume 68 Issue 2

Full Size Image

Cover Legend PDF

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
February-2026 Volume 68 Issue 2

Full Size Image

Cover Legend PDF

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Advancements on the synergistic application of oncolytic viruses and molecularly targeted therapies for the treatment of solid tumors (Review)

  • Authors:
    • Shiwen Bi
    • Tiyan Shan
    • Yong Tang
    • Qi Wang
  • View Affiliations / Copyright

    Affiliations: Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, P.R. China, University Engineering Research Center of Oncolytic & Nanosystem Development, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
    Copyright: © Bi et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 25
    |
    Published online on: December 16, 2025
       https://doi.org/10.3892/ijo.2025.5838
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Oncolytic virotherapy has emerged as a significant advancement in cancer treatment. However, the efficacy of monotherapies is limited by tumor heterogeneity, highlighting the need for combination strategies to overcome therapeutic limitations. This study provides a review of the molecular mechanisms, preclinical advancements and clinical outcomes associated with oncolytic virus (OV)‑targeted drug combinations over the past two decades, elaborating on the interaction mechanisms through which molecular targeted drugs and oncolytic viruses enhance antitumor effects. Additionally, the progress in translating OV‑based combination therapies for solid tumors into clinical practice is outlined and innovative strategies are proposed for developing novel therapeutic frameworks.

Introduction

Oncolytic viruses (OVs) represent an innovative approach in cancer therapy, characterized by their ability to selectively lyse tumor cells while sparing adjacent normal tissues. They also potentiate the anti-tumor immune response through the release of tumor-associated antigens and the activation of inflammatory pathways within the tumor microenvironment (TME) (1). Preclinical toxicology studies have demonstrated that OVs exhibit low toxicity and high tolerability (2). To date, five OVs have successfully undergone clinical translation (3), including the oncolytic adenovirus H101, talimogene laherparepvec (T-VEC), Rigvir, G47Δ and nadofaragene firadenovec-vncg. They show significant potential in clinical applications (4-8). However, in solid tumors, factors such as abnormal lymphatic structures, high vascular permeability, a dense extracellular matrix and the requirement for OVs to traverse the endothelial layer to reach target cells collectively reduce the penetration range of OVs. Additionally, interactions between OVs and antigen-presenting cells can trigger innate immune responses and antiviral immunity, increasing the likelihood of clearance by the host's immune system (9). Another key obstacle is the heterogeneity of tumors, which leads to incomplete responses to specific monotherapies (10). These limitations hinder the widespread clinical translation of OV therapy.

Targeted therapy functions by selectively interacting with specific sites to inhibit enzymes and growth factor receptors essential for proliferation, inducing apoptosis in cancer cells and modulating gene expression, thereby altering protein functions in normal cells to disrupt pathways associated with carcinogenesis and tumor progression (11). However, due to genetic alterations, adaptive responses or bypass mechanisms that allow cancer cells to endure the selective pressure exerted by the treatment, resistance may be acquired, which limits the long-term efficacy and developmental potential of targeted therapies (12).

Previously research indicates that the integration of OVs with targeted therapeutics can inhibit tumor initiation and progression through various mechanisms, effectively overcoming the limitations associated with OVs and targeted drug monotherapies, thereby enhancing the overall efficacy of cancer treatment. As a result, combination therapy is gradually becoming one of the leading approaches in oncology. Earlier reviews mainly explained the basic principles and potential strategies of combining OVs with key cellular signaling pathway modulators in cancer, without categorizing the specific mechanisms (13). Certain reviews have summarized the progress of OVs in combination with various therapies, such as chemotherapy, radiotherapy and mainly novel immunotherapies like immune checkpoint inhibitors (10,14,15). This article primarily elaborates on the mechanisms and progress of OVs combined with targeted drugs, categorizing them according to different mechanisms, while further exploring how to select more rational combination approaches based on the characteristics of different tumors, such as OVs targeting tumor surface nonspecific and specific receptors and monitoring of mutated targets. By discussing the differences between human and mouse immune systems, potential combination toxicities and their solutions, as well as future strategies to address drug resistance, the article highlights key points for translating preclinical research into clinical applications. Based on the above discussions, it connects preclinical evidence with trial design elements such as patient selection criteria, recommended endpoints and overlapping toxicity safety monitoring, providing a valuable reference for the future clinical translation of OVs combined with targeted drugs.

Mechanisms of action of OVs combined with molecularly targeted therapies

The mechanism of action of OVs combined with molecularly targeted drugs is mainly related to signaling pathways, immune responses, DNA damage, apoptosis and autophagic cell death. In this chapter, a detailed explanation of the above will be provided.

Inhibition of phosphorylation at specific sites modulates downstream signaling and interferon (IFN) pathways

Inhibiting key targets in signaling pathways related to cell proliferation can enhance the replication of OVs and their oncolytic effects, thereby exerting antitumor activity. The mitogen-activated protein kinase (MAPK), Janus kinase (JAK)/signal transducer and activator of transcription (STAT) and phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathways are jointly involved in regulating cell proliferation, differentiation, survival and apoptosis, with complex crosstalk between them. MAPK kinase (MEK) is one of the key targets in the MAPK signaling pathway (16). Lee et al (17) demonstrated that inhibiting MEK phosphorylation can activate STAT3, thereby promoting oncolytic vaccinia virus (OVV) replication in doxorubicin-resistant ovarian cancer and enhancing the anti-tumor effects. STAT3 activation also promotes the replication of oncolytic herpes simplex virus (oHSV) in glioma cells (18). MEK inhibitors also suppress the antiviral response facilitated by IFN signaling through STAT1/STAT2-dependent pathways. IFNs orchestrate innate immune responses, serving as a formidable first line of defense against pathogen invasion (19). This initiates a signaling cascade through the JAK-STAT pathway (20). STAT1 and STAT2 play pivotal roles in type I and type II IFN signaling, significantly contributing to the cellular antiviral response and adaptive immunity. The primary pathways through which IFNs exert their antiviral effects include the oligoadenylate synthetase-ribonuclease L system and the RNA-dependent protein kinase (PKR) pathway, which degrade viral RNA and inhibit viral protein synthesis, respectively. Zhou et al (21) found that trametinib boosts viral replication in BRAF V600E and BRAF wild-type (wt)/KRAS mutated tumor cells by reducing STAT1 and PKR phosphorylation. Additionally, the JAK-1/2 inhibitor ruxolitinib increases vesicular stomatitis virus (VSV) replication by inhibiting the JAK/STAT signaling pathway, enhancing its sensitivity in melanoma and lung cancer (22,23). Targeting the Cysteine-rich 61 (CCN1)-AKT pathway can enhance bevacizumab's effectiveness by reducing glioma infiltration. Elevated CCN1 expression is associated with increased AKT phosphorylation in tumor cells (24). Studies have shown that CCN1 stimulates the expression of PI3K/AKT in various types of cancer cells, including glioma, breast cancer, gastric cancer and renal cancer cells (25-27). Rapid antiangiogenesis mediated by oncolytic virus (RAMBO) is an engineered oHSV designed to express angiogenesis inhibitors and is capable of suppressing the expression of CCN1 (28). A study found that the angiostatin produced by RAMBO can inhibit bevacizumab-induced CCN1 expression, reducing bevacizumab-induced glioma infiltration by blocking the CCN1-AKT signaling pathway, thereby enhancing the efficacy of bevacizumab against malignant gliomas (28).

Tyrosine kinase inhibitor (TKI) synergistically augments the antitumor efficacy of OVs by inhibiting the phosphorylation of AKT, which are activated by OVs. G47Δ-mIL12, an advanced version based on the third-generation oHSV-1 G47Δ, enhancing tumor-specific replication and safety by deleting the α47 gene and US11 promoter to prevents class I major histocompatibility complex (MHC) downregulation and inhibiting angiogenesis (4,29,30). Research conducted by indicates that the selective TKI axitinib enhances the antitumor effect of G47Δ-mIL12 in glioblastoma (GBM) by blocking the platelet-derived growth factor receptor (PDGFR) pathway, inhibiting phosphorylated ERK1/2 and reducing G47Δ-mIL12-induced AKT phosphorylation. Furthermore, this combination also promotes apoptosis and affects stem-like cell characteristics (31). TKI can also promote OV replication by inhibiting the antiviral response mediated by IFN signaling. Research by Jha et al (32) demonstrated that multi-target TKI sunitinib weakens the antiviral response by inhibiting VSV-induced activation of the IFN pathway, thereby enhancing VSV replication.

Boosting natural or adaptive anti-tumor immunity

IFN is also a key pathway in antiviral immunity, and the transcription of IFN-regulated genes induced by the JAK/STAT pathway inhibits OVs replication through multiple mechanisms (19). By inhibiting the type I IFN pathway to weaken the antiviral response of specific tumor cells, thereby enhancing the spread and oncolytic activity of OVs in tumor cells. Trastuzumab-DM1 (T-DM1), an antibody-drug conjugate, consists of the targeted therapeutic agent trastuzumab linked to the microtubule-disrupting agent and mertansine derivative DM1. This conjugate primarily exerts its microtubule-inhibiting effect in human EGFR2 (HER2)-overexpressing tumor cells through the specific targeting of trastuzumab (33). Arulanandam et al (34) found that T-DM1 targets HER2-overexpressing tumor cells to release DM1, which inhibits the type I IFN pathway and enhances the cytotoxic effect of TNF-α on neighboring cells, thereby increasing the spread and oncolytic effect of the VSVΔ51 virus in VSV-resistant cancer cells. This study did not emphasize the antitumor effect of trastuzumab itself (34). In addition, IκB kinase inhibitors BMS-345541 and TPCA-1 enhance VSV replication by inhibiting type I IFN-mediated antiviral responses, thereby improving the efficacy against glioma (35).

Targeted drugs combined with OVs enhance cytotoxic effects on tumor cells in the TME by activating immune cells, promoting cytokine release and reducing immunosuppression. Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells that expand in inflammation and tumors and are effective inhibitors of T cell-mediated immune responses (36). Lawson et al (37) found that sunitinib can alleviate immunosuppression induced by coxsackievirus, such as the accumulation of MDSCs, and stimulate the number of immune-stimulating cells, enhancing the anti-tumor immune response in mouse renal cell carcinoma and lung squamous cell carcinoma. Similarly, Moehler et al (38) found that sunitinib with H-1 parvovirus significantly boosts immune stimulation in melanoma. S100A8/9 are low-molecular-weight calcium-binding proteins, which can regulate the accumulation of MDSCs (39). Chai et al (40) confirmed that neural stem cell-delivered OV combined with S100A8/9 inhibitor paquinimod enhanced anti-GBM efficacy by increasing T-cell infiltration, shifting macrophages to a pro-inflammatory phenotype and reducing MDSCs. A recent study reported that KRAS inhibitors themselves promote innate and adaptive immunity, and when combined with OVV, they reduce immunosuppressive cells such as MDSCs and T-regulatory cells (Tregs) in the TME, synergistically enhancing antitumor effects (41).

TANK-binding kinase 1 (TBK1) inhibitors promote OVs replication and their sensitivity to resistant cells by enhancing immune cell responses. Intercellular adhesion molecule 1 (ICAM-1) is a cell surface receptor used by immune cells for adhesion and migration, and it is often upregulated during viral infections (42). TBK1 is a serine/threonine kinase, and its increased expression or abnormal activity can promote the survival and proliferation of cancer cells (43). Guo et al (44) reported that TBK1 inhibitors can promote VSVΔ51 replication by enhancing ICAM-1-mediated natural killer (NK) cell immunity and increase the sensitivity of VSVΔ51 to chemoresistant colorectal cancer cells.

MEK inhibitors enhance viral replication by decreasing TNF-α secretion and blocking its apoptotic effects. Due to early cell death from TNF-α-induced apoptosis, the replication and antitumor impact of oHSV-1 are limited (45). Yoo et al (46) discovered that the MEK inhibitor trametinib inhibits apoptosis in GBM cells following oHSV infection by reducing TNF-α levels, thereby promoting viral replication. Furthermore, oHSV aids trametinib in crossing the blood-brain barrier and prevents MAPK pathway reactivation, enhancing tumor sensitivity to MEK inhibition.

Proteasome inhibition promotes OVs replication by increasing the expression of heat shock proteins (HSPs). HSP90 has previously been shown to be important for the nuclear localization of HSV polymerase (47). In line with this, Yoo et al (48) found that in head and neck squamous cell carcinoma, ovarian cancer, malignant peripheral nerve sheath tumor and glioma, the proteasome inhibitor bortezomib increases the expression of HSP90 by inducing unfolded protein response accumulation, thereby promoting oHSV-1 replication and enhancing antitumor efficacy.

Using OVs to infect tumor cells allows targeted drugs to be released and connects their targets with immune cell receptors to enhance cell-killing effects. Tian et al (49) created OV-Cmab-CCL5, an oHSV encoding a bispecific fusion protein that combines cetuximab's IgG1 form with C-C motif chemokine ligand 5 (CCL5). This targets EGFR and CCL5 receptors to increase CCL5 levels in tumors and boost immune cell infiltration. It can also enhance the killing effect on GBM cells by promoting antibody-dependent cellular phagocytosis and antibody-dependent cellular cytotoxicity through linking Fcgamma receptors on macrophages and NK cells with EGFR on tumor cells.

Increasing DNA damage by inhibiting DNA damage repair and increasing ROS production to promote tumor cell death

Poly ADP-ribose polymerase (PARP) inhibitors (PARPi) enhance antitumor effects by blocking DNA repair induced by OVs, causing persistent DNA damage and promoting cancer cell death. The dl922-947, a second-generation oncolytic adenovirus with a 24-base pair deletion in the E1A-conserved region 2, enhances viral genome production in host cells (50). Passaro et al (51) found that DNA damage induced by dl922-947 activates the PARP repair mechanism in the DNA damage response signaling pathway, while the PARPi olaparib can restore PARP activation induced by it and exacerbate DNA damage, thereby promoting apoptosis and enhancing the anti-tumor efficacy against thyroid cancer. The same antitumor mechanism was observed in the treatment of melanoma with reovirus type 3 Dearing strain (ReoT3D) combined with talazoparib (52). Recent research reports that PARP1 has been identified as a replication restriction factor for HSV-1, and olaparib promotes viral replication by inhibiting PARP1, thereby enhancing antitumor efficacy in GBM and triple-negative breast cancer (53).

Furthermore, the combination of OVs and PARPi can induce excessive reactive oxygen species (ROS) production, leading to increased DNA damage and S-phase arrest, thereby enhancing the inhibition of tumor cell proliferation. While ROS are common in cells, excessive ROS can cause cell death (54) and activate oncogenic pathways, leading to DNA damage and genetic instability (55). PARPi can have antitumor effects by increasing ROS and ROS-induced DNA damage (56,57). The measles virus (MV) is a promising alternative virus (58), which has been proven to exert oncolytic effects on ovarian cancer through ROS-induced apoptosis (59). Zhang et al (60) discovered that the recombinant Chinese Hu191 MV (rMV-Hu191) modified by a viral reverse genetic system, works with olaparib to enhance oxidative DNA damage in pancreatic ductal adenocarcinoma cells by increasing ROS levels. This combination also significantly halts the cell cycle in the S phase and inhibits cell proliferation by blocking DNA division (60).

Inducing tumor cell apoptosis by disrupting specific cell cycle stages

In mammalian cells, p53 and cyclin-dependent kinases jointly manage the G1/S transition, linking apoptosis to cell cycle progression (61). Irinotecan is a topoisomerase I inhibitor that can induce the expression and phosphorylation of the tumor antigen p53, promoting the expression of apoptosis-regulating factors (62). Napabucasin (BBI608) is a stem cell inhibitor that can directly inhibit the transcription of STAT3-derived genes and can also indirectly inhibit other related genes and pathways (63). Babaei et al (64) found that combining ReoT3D, Irinotecan (CPT-11) and BBI608 significantly reduced the S phase of CT26 cells, arrested them in G0/G1 and G2/M phases, induced apoptosis by causing a sub-G1 fraction to form and downregulated KRAS and STAT3 mRNA.

Inducing autophagic cell death to stop tumor cells from using autophagy to survive and grow in nutrient-poor conditions

Autophagy is a fundamental biological process that is crucial for preventing cellular damage, maintaining cell survival during nutrient scarcity and responding to cytotoxic stimuli. The regulation of autophagy by the PI3K/AKT/mTOR signaling pathway allows tumors to exploit this process for survival benefits through the dysregulation of the pathway (65-67). Everolimus (RAD001) is a widely used mTOR inhibitor that targets mTOR complex 1 and inhibits autophagy (68). Alonso et al (69) demonstrated that the combination of RAD001 with Delta-24-RGD, an oncolytic adenovirus with a 24-base pair deletion in the E1A gene (70), induces autophagic cell death and enhances anti-glioma efficacy, although the precise underlying mechanism remains to be elucidated. The anti-angiogenic and immunosuppressive properties of RAD001 may also contribute to these outcomes. Furthermore, they revealed that Delta-24-RGD increases glioma cell sensitivity to RAD001 by circumventing G1 phase arrest, without impacting the replication of Delta-24-RGD, indicating an additional potential anti-tumor mechanism.

Summary of molecular mechanisms of different inhibitors in promoting OV replication and enhancing antitumor effects

The above content regarding composite molecular machineries involves various targeted drugs that exhibit significant differences in targets, mechanisms of action and tumor sensitivity. MAPK inhibitors primarily act on kinases within the MAPK pathway, such as BRAF, MEK and ERK. MEK inhibitors promote OVV replication by activating STAT3 and can also increase viral replication by reducing antiviral responses through the inhibition of STAT activation. TKIs synergistically enhance antitumor effects by reducing AKT phosphorylation activated by OVs, while OVs engineered to specifically express endostatin can increase tumor sensitivity to bevacizumab by inhibiting AKT. Pathway inhibitors can also suppress targets related to the IFN antiviral pathway, promoting OV replication. The presence of immunosuppressive cells in the TME is also a key reason for poor efficacy. Targeted drugs combined with OVs synergistically enhance antitumor effects by boosting innate and adaptive immunity and reducing immunosuppression. PARPi primarily target PARP enzymes responsible for repairing double-strand DNA damage, exerting antitumor effects by inducing sustained DNA damage through increased ROS and inhibition of OV-induced DNA repair. Furthermore, OVs can synergize with inhibitors related to apoptosis and autophagy pathways for antitumor effects (Fig. 1). The different characteristics of these inhibitors provide clearer guidance for selecting targeted drugs in combination therapy. Choosing the appropriate drugs based on tumors with different target mutations can markedly enhance the precision and antitumor efficacy of combination therapy. Due to the numerous molecular target mutations in tumors, a wide variety of targeted drugs have been developed and approved for use. However, the exploration of combination studies remains limited, with the combined mechanisms focusing only on major pathways such as MAPK, JAK/STAT and PI3K/AKT. The Wnt, Notch and Hippo signaling pathways, which also play important roles in multiple cancers (71-73), have not yet been reported, to the best of our knowledge. The potential interactions between OVs and the toxic side effects of the targeted drugs themselves could reduce the therapeutic window and introduce unpredictable pharmacokinetics/pharmacodynamics. Furthermore, this strategy also has some inherent limitations. Tumor heterogeneity may lead to certain tumors being insensitive to both treatments, and since OVs are mainly administered via local injection, their efficacy against distant metastatic lesions is limited. The use of certain potent apoptosis-inducing targeted drugs may prematurely eliminate OVs, which in turn could restrict viral replication.

Schematic depicting the main
mechanism. The left panel mainly focuses on the mechanisms by which
tumor cells interact with signaling pathways and DNA damage.
Trametinib combined with oHSV inhibits STAT1, thereby affecting the
type I interferon-mediated antiviral response pathway; Ruxolitinib
promotes VSV replication by inhibiting JAK1/2; MEK inhibitors
promote OVV replication by inhibiting STAT3 phosphorylation;
Axitinib combined with G47Δ-mIL12 inhibits p-AKT and p-ERK to exert
anti-tumor effects; RAMBO increases tumor sensitivity to
bevacizumab by inhibiting CCN1 expression. Olaparib combined with
rMV-Hu191 enhances DNA damage by increasing ROS levels; Olaparib
combined with dl922-947 exacerbates DNA damage to exert anti-tumor
effects. Bortezomib promotes oHSV-1 replication by increasing UPR
accumulation and enhancing HSP90 expression. The right panel mainly
describes the mechanism of action of combinations in the tumor
immune microenvironment. Trametinib mitigates the reduction of oHSV
induced by the exogenous phagocytosis pathway through the
inhibition of TNF-α secretion by macrophages; OV-Cmab-CCL5 enhances
the engagement of Fcγ receptors on the surfaces of macrophages and
NK cells by EGFR tumor cells, thereby facilitating ADCP and ADCC;
the combination of reovirus with sunitinib has been shown to
decrease IFN-γ secretion and reduce the presence of Treg and MDSC
within immunosuppressive environments. The co-administration of
H-1PV with sunitinib significantly augments immune stimulation by
promoting DC maturation, IL-6 release, and the activation of
cytotoxic T lymphocytes. NSCs-OV combined with paquinimod enhances
anti-tumor efficacy by increasing T cell infiltration and reducing
MDSCs. IFN-γ, interferon γ; RAMBO, Rapid antiangiogenesis mediated
by oncolytic virus; CCN1, Cysteine-rich 61; IFN-α, interferon α;
IFN-β, interferon β; JAK, janus kinase; STAT, signal transducer and
activator of transcription; TYK, tyrosine kinase; IRF9, interferon
regulatory factor 9; VSV, vesicular stomatitis virus; MEK,
mitogen-activated protein; OVV, oncolytic vaccinia virus; PI3K,
phosphatidylinositol 3-kinase; PDGFR, platelet-derived growth
factor receptor; ERK, extracellular regulated protein kinases; UPR,
unfolded protein response; HSP90, heat-shock protein 90; HSV,
herpes simplex virus; ROS, reactive oxygen species; IL-6,
interleukin 6; MDSC, myeloid-derived suppressor cell; Treg,
Regulatory T cell; ADCC, Antibody dependent cell-mediated
cytotoxicity; ADCP, Antibody-dependent cell-mediated phagocytosis;
H-1PV, H-1 parvovirus; NSC, neural stem cell; Cmab, cetuximab;
CCL5, C-C motif chemokine ligand 5; TNF, tumor-necrosis factor; NK,
natural killer; DC, Dendritic Cell.

Figure 1

Schematic depicting the main mechanism. The left panel mainly focuses on the mechanisms by which tumor cells interact with signaling pathways and DNA damage. Trametinib combined with oHSV inhibits STAT1, thereby affecting the type I interferon-mediated antiviral response pathway; Ruxolitinib promotes VSV replication by inhibiting JAK1/2; MEK inhibitors promote OVV replication by inhibiting STAT3 phosphorylation; Axitinib combined with G47Δ-mIL12 inhibits p-AKT and p-ERK to exert anti-tumor effects; RAMBO increases tumor sensitivity to bevacizumab by inhibiting CCN1 expression. Olaparib combined with rMV-Hu191 enhances DNA damage by increasing ROS levels; Olaparib combined with dl922-947 exacerbates DNA damage to exert anti-tumor effects. Bortezomib promotes oHSV-1 replication by increasing UPR accumulation and enhancing HSP90 expression. The right panel mainly describes the mechanism of action of combinations in the tumor immune microenvironment. Trametinib mitigates the reduction of oHSV induced by the exogenous phagocytosis pathway through the inhibition of TNF-α secretion by macrophages; OV-Cmab-CCL5 enhances the engagement of Fcγ receptors on the surfaces of macrophages and NK cells by EGFR tumor cells, thereby facilitating ADCP and ADCC; the combination of reovirus with sunitinib has been shown to decrease IFN-γ secretion and reduce the presence of Treg and MDSC within immunosuppressive environments. The co-administration of H-1PV with sunitinib significantly augments immune stimulation by promoting DC maturation, IL-6 release, and the activation of cytotoxic T lymphocytes. NSCs-OV combined with paquinimod enhances anti-tumor efficacy by increasing T cell infiltration and reducing MDSCs. IFN-γ, interferon γ; RAMBO, Rapid antiangiogenesis mediated by oncolytic virus; CCN1, Cysteine-rich 61; IFN-α, interferon α; IFN-β, interferon β; JAK, janus kinase; STAT, signal transducer and activator of transcription; TYK, tyrosine kinase; IRF9, interferon regulatory factor 9; VSV, vesicular stomatitis virus; MEK, mitogen-activated protein; OVV, oncolytic vaccinia virus; PI3K, phosphatidylinositol 3-kinase; PDGFR, platelet-derived growth factor receptor; ERK, extracellular regulated protein kinases; UPR, unfolded protein response; HSP90, heat-shock protein 90; HSV, herpes simplex virus; ROS, reactive oxygen species; IL-6, interleukin 6; MDSC, myeloid-derived suppressor cell; Treg, Regulatory T cell; ADCC, Antibody dependent cell-mediated cytotoxicity; ADCP, Antibody-dependent cell-mediated phagocytosis; H-1PV, H-1 parvovirus; NSC, neural stem cell; Cmab, cetuximab; CCL5, C-C motif chemokine ligand 5; TNF, tumor-necrosis factor; NK, natural killer; DC, Dendritic Cell.

Animal model studies on combining OVs with targeted drugs

Research indicates that combining OVs with targeted drugs is safer and more effective for treating solid tumors than using them separately. Some animal studies suggest that small molecule targeted drugs can hinder tumor growth by reducing angiogenesis, interstitial fluid pressure (IFP) and barriers to OVs reaching target cells. HF10 is a spontaneously occurring HSV-1 (74). Yamamura et al (75) found that erlotinib and HF10 work synergistically in a pancreatic cancer model by reducing angiogenesis and IFP, leading to a higher intratumoral virus distribution and significant tumor growth inhibition. Tan et al (76) found that combining the OV HF10 with bevacizumab improved tumor suppression compared to either treatment alone in breast cancer models. This synergy was linked to increased vascular permeability, VEGF-induced viral replication in endothelial cells, immune attacks on infected vessels, enhanced tumor hypoxia boosting viral protein synthesis and increased apoptosis. Another study reported that bevacizumab in combination with dl922-947 significantly inhibited tumor growth in tumor xenografts in thyroid anaplastic carcinoma tumors and increased the distribution of the virus in tumors (77). However, Mahller et al (78) found that erlotinib in combination with oHSV did not show enhanced antitumor effects in a xenograft mouse model of human malignant peripheral nerve sheath tumor. The distribution of OVs is strongly influenced by the TME, among which IFP is a great challenge to efficient virus distribution, and tumor angiogenesis is one of the main causes of high IFP.

The aforementioned preclinical studies have demonstrated that the combination of OVs with small molecule targeted therapies can partially overcome the barrier of the endothelial layer, thereby facilitating the distribution and replication of the virus within the tumor and enhancing its anti-tumor efficacy. Animal models have further indicated that this combination exhibits significant anti-tumor effects. However, the precise molecular mechanisms underlying these effects remain unidentified, and it is elusive whether they align with the mechanisms discussed in this article or involve alternative pathways. Given the discrepancies between the TMEs in vivo and in vitro, in vitro findings alone are insufficient to substantiate the translation of these therapies into clinical practice. Consequently, extensive preclinical research, including exploratory studies on the mechanisms involved, is necessary to advance the development of this combination therapy.

Although the immune organs of humans and mice are highly conserved in their macroscopic anatomical structure, there are significant differences in the composition, distribution and expression of key immune molecules in their immune cells (Table I) (79). Due to significant immune differences between the TME of mouse tumor models and human spontaneous tumors, their clinical application value is limited. The discovery and application of immunodeficient mice provide a crucial breakthrough for reconstructing the human immune system in vivo, thereby accurately simulating the human TME. Their development has undergone continuous iterative upgrades, evolving from simple T-cell deficiencies (such as nude mice) to severe combined immunodeficiency models with multiple functional deficiencies in immune cells such as T, B and NK cells (79). Most of the research data in this study are derived from this type of model. However, immunodeficient mice completely lose the ability to initiate adaptive immune responses, displaying severe defects in the innate immune system. They are not suitable for studying the human immune system itself or the diseases it participates in, but this is the primary requirement for constructing humanized mouse models from immunodeficient mice (80). Humanized mice refer to immunodeficient mice co-implanted with human tumors and immune components, which can realistically assess the mechanisms of immune system activation and efficacy in preclinical studies of OVs in combination with targeted drugs, providing more optimized combination treatment strategies for clinical trial design.

Table I

Comparison of the differences between the human and mouse immune systems.

Table I

Comparison of the differences between the human and mouse immune systems.

ItemHuman immune systemMouse immune system
Neutrophil proportion, %40-654-6
Lymphocyte proportion, %15-4060-80
IgG classificationIgG1, IgG2a, IgG2b, IgG3IgG1, IgG2, IgG3, IgG4
Splenic T cellsScattered in GCsClustered distribution
Epidermal immune cellsαβTγδT
NK cell inhibitory receptorKIRLy49
NKG2D binding ligandsMHC-I, UL16H-60, Rae1β
Anti-parasitic infectionTh2 cells activate eosinophils and B cells to secrete IgETh1 cells produce IFN-γ

[i] Th1, type 1 T-helper; Ig, immunoglobulin; NK, natural killer; NKG2D, NK group 2 member D; GC, germinal center; KIR, killer-cell immunoglobulin-like receptors; MHC, major histocompatibility complex; Th2, type 2 T-helper; IFN, interferon.

Clinical studies of OVs with targeted drugs

Certain OVs have the ability to preferentially infect certain types of tumor cells before any modifications. This natural tropism primarily depends on the specific binding between viral surface proteins and specific receptors on the host cell surface. Many tumor cells overexpress certain receptors required for viral invasion. For example, the coxsackievirus and adenovirus receptor (CAR) is expressed at low levels in normal tissues but is often overexpressed in various malignancies, allowing coxsackievirus and adenovirus to preferentially target and infect these tumors such as laryngeal cancer, thyroid cancer, lung cancer and female reproductive system tumors (81,82). CD46 is mainly present on the surface of prostate cancer and bladder cancer and can be recognized by adenoviruses and measles viruses (83-86). The expression of integrins in tumor cells is usually abnormal, allowing various viruses such as adenovirus, coxsackievirus and reovirus to enter cells through integrin-associated ligands (87-91). Table II shows the natural affinity of many viruses for nonspecific receptors on the surface of certain tumor cells (Table II). It can serve directly as a therapeutic option or as a framework for viral modification. In addition to common receptors, the surface receptors of different types of tumors also have their specific characteristics. OVs Ankara-5T4, constructed to target the specific receptors 5T4 for colorectal cancer, have shown higher specificity and efficacy (92,93). In addition, colorectal cancer is associated with gut microbiota dysbiosis, but there are no reports on the genetic modification of OVs in relation to this connection (94,95). R-405, designed to target the prostate cancer-specific receptor prostate-specific membrane antigen, also demonstrates strong antitumor effects (96,97). The development and research of OVs targeting different tumor-specific receptors are shown in the table (Table III). Therefore, we can enable OVs to acquire the ability to bind new receptors and further enhance tumor targeting by inserting antibody fragments or peptides that specifically bind to tumor-associated antigens.

Table II

Currently common types of OVs, their nonspecific receptors and the expression of these receptors in major solid tumors.

Table II

Currently common types of OVs, their nonspecific receptors and the expression of these receptors in major solid tumors.

OV typeNonspecific receptorTumor expression(Refs.)
AdenovirusCARElevated expression in laryngeal cancer, thyroid cancer, lung cancer and female reproductive system tumors; decreased expression in kidney cancer and prostate cancer(81,82)
CD46Prostate cancer and bladder cancer(83-85)
IntegrinThe expression of integrins in tumor cells is usually abnormal(87-89)
Desmoglein 2Upregulated in melanoma, malignant skin cancer, lung cancer, colon adenocarcinoma, primary liver cancer and cervical cancer; downregulated in gastric cancer, malignant ovarian tumors and pancreatic cancer(169-178)
Sialic acidWidely overexpressed in cancer(179,180)
Herpes virusHerpesvirus entry mediatorLung cancer, gastric cancer, breast cancer, esophageal squamous cell carcinoma(181-186)
Nectin-1Bladder urothelial carcinoma, colorectal cancer(187-189)
Vaccinia virusHeparan sulfate proteoglycanBladder cancer, breast cancer, gallbladder cancer(190-194)
ReovirusIntegrinSame as adenovirus(90)
Junctional adhesion molecule AEsophageal squamous cell carcinoma, glioma, stomach cancer, breast cancer(195-199)
Measles virusCD46Same as adenovirus(86)
Signaling lymphocytic activation moleculeHepatocellular carcinoma, head and neck squamous cell carcinoma(200,201)
PoliovirusCD155Pancreatic cancer, breast cancer, osteosarcoma, colorectal cancer(202-206)
Vesicular stomatitis virusLow-density lipoprotein receptorBreast cancer, renal cell carcinoma, ovarian cancer(207-210)
Coxsackie virusCARSame as adenovirus(81,82)
IntegrinSame as adenovirus(91)
ICAM-1/CD54Liver cancer, pancreatic cancer, ovarian cancer, prostate cancer(211-215)
DAF/CD55Colorectal cancer, breast cancer, malignant glioma, medullary thyroid carcinoma(216-220)
Scavenger receptor class B member 2Breast cancer, hepatocellular carcinoma, glioma(221-223)

[i] OV, oncolytic virus; ICAM-1, intercellular adhesion molecule 1, CAR, coxsackievirus and adenovirus receptor; DAF, decay-accelerating factor.

Table III

Tumor specificity and corresponding OVs of the world's 10 most ommon cancers.

Table III

Tumor specificity and corresponding OVs of the world's 10 most ommon cancers.

Tumor typeTumor-specific receptorGenetically engineered OV targeting tumor-specific receptor(Refs.)
Colorectal cancer5T4Ankara-5T4(92,93)
Abnormal gut microbiota-(94,95)
CEArV(224,225)
Prostate cancerPSMAR-405(96,97)
PSCA-
TMPRSS2-
STEAP1-
Stomach cancerCLDN18.2OV-BiTE(226,227)
Liver cancerAFPHa2bm-d19(228)
ASGPR-(229)
GPC3-(230)
Thyroid cancerTSHR-(231)
RET
Bladder cancerPD-L1-(232)
FGFR-(233)
MelanomaBRAF V600E/K-(234)
GD2-(235)
CSPG4/MCSPCoxsackievirus B3(236)

[i] OV, oncolytic virus; CEA, carcinoembryonic antigen; PSMA, prostate-specific membrane antigen; PSCA, prostate stem cell antigen; TMPRSS2, transmembrane serine protease 2; STEAP1, six-transmembrane epithelial antigen of prostate 1; CLDN18.2, claudin18.2; AFP, alpha-fetoprotein; ASGPR, asialoglycoprotein receptor; GPC3, glypican-3; TSHR, thyrotropin receptor; PD-L1, programmed cell death ligand 1; FGFR, fibroblast growth factor receptor; CSPG4/MCSP, chondroitin sulfate proteoglycan 4.

Over the past two decades, clinical research has progressively investigated the integration of OVs with targeted therapies, yielding promising outcomes in certain instances. A meta-analysis encompassing 12 studies evaluated the objective response rate, survival rate and incidence of adverse reactions in cancer patients receiving a combination of OVs and conventional therapy compared to conventional therapy alone. The findings revealed that the combination therapy was significantly associated with an increased objective response rate (ORR) (P=0.04) and a higher incidence of grade ≥3 adverse reactions (P=0.02) and grade ≥3 neutropenia (P=0.01), although no significant difference in survival was observed (98). Nonetheless, further investigation is warranted. In a phase I trial conducted by Hirooka et al (99), patients with histologically confirmed locally advanced pancreatic cancer without distant metastasis and unresectable tumors received one cycle of erlotinib and gemcitabine, followed by intratumoral injection of HF10 under endoscopic ultrasound guidance. The primary endpoint was safety assessment and the secondary endpoint was efficacy evaluation. The study results showed that among the 10 subjects, 2 experienced serious adverse events unrelated to HF10, indicating that the doses used in this trial were safe and effective. Among the 9 subjects who completed all four HF10 injections, the overall response included 3 partial responses, 4 patients with stable disease and 2 cases with progressive disease, with a median progression-free survival (PFS) of 6.3 months and a median overall survival (OS) of 15.5 months, and 2 subjects were able to undergo surgical resection. The results suggest the safety and efficacy of this therapeutic approach (100). However, the study only compared the efficacy between different HF10 titers and did not set up a control group or erlotinib monotherapy group, gemcitabine monotherapy group or erlotinib plus gemcitabine to further demonstrate the superiority of HF10 injection after treatment with erlotinib and gemcitabine.

Sorafenib in combination with JX-594 has been studied in hepatocellular carcinoma (HCC). JX-594 (Pexa-Vec) was constructed by inserting human granulocyte-monocyte colony-stimulating factor and lacZ into the thymidine kinase gene region of the Wyeth strain vaccinia virus (101). After infecting tumor cells, it can stimulate toll like receptors, activate dendritic cells (DCs), increase NK, cytotoxic T-lymphocyte and white blood cell infiltration, and promote the secretion of a variety of cytokines, such as IFN, IL-1, IL-6 and IL-12 (101). Heo et al (102) discovered that administering JX-594 before sorafenib significantly inhibited tumor growth compared to control and other treatment sequences in mouse models. Encouraged by these findings, they tested this sequence in patients with advanced HCC and found it to be well-tolerated, leading to rapid and significant tumor necrosis, with a 50-100% increase in tumor necrosis for patients who did not achieve durable objective tumor remission with JX-594 alone completed JX-594 treatment, and JX-594 was injected directly into the tumors under imaging guidance (102). However, only 3 patients were evaluated in this study and are not representative of the entire HCC population, and did not set evaluation endpoints or provide statistical analysis results, resulting in poor generalizability of the results and reduced reliability, potentially leading to misleading conclusions. Rare adverse events could also not be detected. In a randomized, multicenter phase IIb trial, intravenous injection of Pexa-Vec in patients with advanced HCC who have progressed after sorafenib treatment or are intolerant to sorafenib, OS is primarily being evaluated, with secondary endpoints including time to progression (TTP), ORR, disease control rate (DCR), time to symptomatic progression, safety, tolerability and quality of life. The study results showed that Pexa-Vec was generally well-tolerated, but there were no significant differences in the evaluated endpoints, which may be due to a high dropout rate preventing accurate assessment of the outcomes (103). In a phase Ⅲ, randomized, open-label study, for patients with advanced HCC who have not received systematic treatment, Pexa-Vec was first administered via intratumoral injection, followed by sorafenib, to evaluate OS compared to sorafenib alone. Secondary objectives included TTP, PFS, ORR and DCR, as well as assessing and comparing the safety of the two treatment groups. The study results showed that compared to sorafenib alone, treatment with sorafenib following Pexa-Vec did not show a statistically significant benefit and was less effective, with a higher incidence of serious adverse events (104). The study indicates that the dosing sequence of JX-594 and sorafenib may impact their effectiveness. Investigating the mechanisms behind their interaction could advance the use of OVs with targeted drugs in clinical research. Table IV summarizes the study subjects, treatment methods, endpoints, results and limitations of the aforementioned clinical studies on OVs combined with targeted drugs and/or other chemotherapeutic agents, as well as those have not reported results. There are currently relatively few clinical trials of OVs combined with targeted drugs, suggesting that there may be obstacles to clinical translation and highlighting the importance of preclinical research in clinical trials.

Table IV

Completed or ongoing clinical trials of oncolytic viruses with targeted and other drugs.

Table IV

Completed or ongoing clinical trials of oncolytic viruses with targeted and other drugs.

Type of virusObjectGroupsPrimary endpointKey secondary endpointsResultsLimitationsClinical trial phaseClinical trial registration no.
Oncolytic herpes simplex virusUnresectable locally advanced pancreatic cancerHF10 + Erlotinib + gemcitabineSafety (n=10)Efficacy (n=9)No AEs related to HF10; PR (n=3), SD (n=4), PD (n=2)-ⅠUMIN000010150
Late-stage microsatellite stable and mismatch repair proficient colorectal cancerRP2/RP3 + Bevacizumab + AtezolizumabORROS, PFS, DOR, DoCB--II (incomplete)NCT05733611
Locally advanced unresectable or metastatic HCCRP2 + Bevacizumab + AtezolizumabORRSafety, DOR--II (incomplete)NCT05733598
Oncolytic vaccinia virusAdvanced HCC not previously treated with sorafenibJX-594 + Sorafenib--Significant tumor necrosis (tumor density quantification)The small number of subjects cannot represent the entire HCC population, no control groupIINCT00554372
Sorafenib-failed HCCPexa-vec + BSC BSCOSQoL, TTP, safetyNo statistical difference at the endpointsHigh rates of drop-out and inability to assessIIbNCT01387555
Advanced HCC not previously treated with systemic therapyPexa-vec + Sorafenib SorafenibOSTTP, PFS, ORR, DCRNo significant difference at the endpointsPatient samples not collected after treatment to assess whether there is benefit from Pexa-vecIIINCT02562755
Platinum-resistant/refractory ovarian cancerOlvi-Vec + Bevacizumab + PlatinumPFSDOR, ORR, OS--III (incomplete)NCT05281471
Natural alphavirus M1Advanced HCCM1-c6v1 + Camrelizumab + ApatinibSafety, toleranceORR, OS, PFSORR was 70%, OS was 15.4 months and PFS was 8.9 months-INCT04665362
Oncolytic adenovirusAdvanced HCCH101 + SorafenibORRChanges in AFP serum levels, DCR, PFS--IVNCT05113290
Colorectal cancer liver metastasisBioTTT001 + Regorafenib, ToripalimabSafety, toleranceOS, PFS, ORR, plasma adenovirus copies--INCT06283134
ReovirusKRAS-mutant metastatic colorectal cancerREOLYSIN + Bevacizumab + FOLFIRIMTDORR, PFS, OS--INCT01274624

[i] HCC, hepatocellular carcinoma; PR, partial response; SD, stable disease; PD, progressive disease; ORR, objective response rate; OS, overall survival; DCR, disease control rate; PFS, progression-free survival; DOR, duration of response; DoCB, duration of clinical benefit; BSC, best supportive care; RR, relative risk; TTP, time to progression; DCR, disease control rate; QoL, quality of life; MTD, maximum tolerated dose.

In some rare case studies, OVs combined with molecularly targeted drugs can alleviate the patient's disease and improve the prognosis. CF33-hNIS-anti-PD-L1 (CHECKvacc) is a novel chimeric orthopoxvirus encoding a single-stranded variable fragment of human sodium iodide symporter and anti-programmed cell death ligand 1, with potent anticancer activity in triple-negative breast cancer xenografts (105). A pathology report reported that a patient with metastatic triple-negative breast cancer progressed after multiple lines of therapy with extensive skin metastases, received intratumoral injection of CHECKvacc followed by sequential treatment with trastuzumab deruxtecan, with a clinical complete response lasting 7 months and a DFS of 10 months, The tumor marker CA15-3 was significantly reduced (106). The patient's decision to cease therapy after multiple treatments makes it unclear whether remission is solely due to the combination therapy, requiring more research. A pathology report highlighted a patient with advanced poorly differentiated rectal adenocarcinoma and liver metastases who achieved complete remission and stable disease for 7 years following FOLFOX-4 chemotherapy, bevacizumab and Rigvir (107). However, since most pathological reports are retrospective, issues of data completeness and consistency cannot be verified, and they also have disadvantages such as high selectivity, poor population representativeness and a high risk of bias, and the results do not represent the safety and efficacy of this combination in the overall population, but can be linked to the existing literature as a way to share lessons learned to help identify new trends and potential uses.

Table V summarizes the classification, genetic modification, immunostimulatory properties and current clinical status of OVs involved in combination therapies discussed in this article. oHSV is the most genetically modified OV, and most applications of modified OVs remain at the preclinical research stage and have not yet progressed to clinical trials.

Table V

Comparison of the characteristics of OVs.

Table V

Comparison of the characteristics of OVs.

Type of virusGenetic modificationImmune microenvironmentClinical trial phaseClinical trial registration no.
Oncolytic vaccinia virusJX-594Insertion mutations (hGM-CSF)TLRs stimulation, DCs activation, NK, CTL, WBC expansion, IFN, IL-1, IL-6 and IL-12 secretionⅢNCT02562755
CF33-Hnis-anti-PD-L1Insertion mutations (hNIS and PD-L1)IFN-γ, IL-1β, IL-6, IL-10, TNF-α and TGF-β1 secretionINCT05081492
Oncolytic HSVG207γ34.5 deletion, ICP insertion mutationSpecific CTL responsesIINCT00028158
G47Δγ34.5 deletion, ICP insertion mutation, α47 deletionRecovery of MHC expression in HSV-1-infected human cells, antiangiogenicIIUMIN000015995
G47Δ-mIL12mIL-12Recovery of MHC expression in HSV-1-infected human cells, antiangiogenic, IFN-γ secretion, Th1 differentiation, immunosuppression improvement--
RAMBOAngiotinDownregulation of CCN1 expression--
OV-Cmab-CCL5IgG1 form of cetuximab and CCL5Enhances migration and activation of NK cells, macrophages and T cells--
HF10Gene deletions, insertional mutations, frameshift mutationsCD8 T-cell infiltration, IL-2, IFN-γ and TNF-α secretionINCT01017185
ReovirusReoT3D-Promotes DC maturation, IFN-α, IFN-γ and TNF-α secretion, NK cell activation--
ParvovirusH-1PV-Increases phagocytosis, maturation and cross-presentation of DCsIINCT02653313
Oncolytic adenovirusDelta-24-RGD24bp E1A deletionStimulates Th1 type immunity, high expression of OX40L on DCs and macrophages, increase in NK and T cells, IFN-γ secretionIINCT01582516
dl922-94724bp E1A deletionDecreased secretion of IL-8 and CCL5, vascular damage and reduced macrophage infiltration--
VSVVSVΔ51M protein mutations---
MVrMV-Hu191Defective MTase region of MV L proteinPyroptosis, apoptosis--

[i] OV, oncolytic virus; TLRs, Toll-like receptors; CTL, cytotoxic T lymphocyte; Th1, type 1 T-helper cell; CCL5, C-C motif chemokine ligand 5; NK, natural killer; DC, dendritic cell; IFN, interferon; MV, measles virus; VSV, vesicular stomatitis virus; CCN1, Cysteine-rich 61; HSV, herpes simplex virus; hGM-CSF, human granulocyte-monocyte colony-stimulating factor; WBC, white blood cell; IL, interleukin; TNF, tumor-necrosis factor; TGF, transforming growth factor; hNIS, human sodium-iodide symporter; PD-L1, programmed cell death ligand 1; MHC, major histocompatibility complex; RAMBO, rapid antiangiogenesis mediated by oncolytic virus; Ig, immunoglobulin; ReoT3D, reovirus type 3 Dearing strain; H-1PV, H-1 parvovirus; OX40L, OX40 ligand; Cmab, cetuximab.

Discussion

Malignant tumors constitute the primary cause of mortality worldwide, with their incidence continuously rising (108). Therapeutic strategies, encompassing surgery, chemotherapy, radiotherapy, immunotherapy and targeted therapy, have progressed from monotherapy to combination therapy to address tumor drug resistance (109,110). OVs have emerged as an innovative approach to cancer treatment, distinguished by their tumor-targeting capabilities, which minimize harm to normal cells and reduce the occurrence of side effects. Clinical trials have not reported any fatalities or severe adverse events attributable to OV therapy, highlighting its favorable safety profile. However, the inherent limitations of OVs may affect their therapeutic efficacy. Targeted drugs can activate the phosphorylation of relevant targets to promote viral replication, and inhibit pathway activation and antiviral responses caused by OVs, thereby increasing OV replication, distribution and oncolytic effects. This enhancement may extend to other targets involved in tumorigenesis or immune microenvironment regulation. The relationship between MEK and STAT3 leads to the inhibition of MEK promoting STAT3 activation, thereby enhancing viral replication. Existing studies have reported a negative correlation between ERK and STAT3 (111-113). This negative correlation has been confirmed as a potential mechanism underlying the development of drug resistance, suggesting that a similar mechanism may also exist. Targeted drugs can also synergize with OVs to enhance immunity, exacerbate DNA damage and promote tumor cell apoptosis, exerting a strong antitumor effect. The diversity in the types and mechanisms of action of targeted drugs, coupled with the variety of OVs and the potential for their genetic modification, provides numerous opportunities. These opportunities include the elimination of factors that adversely affect their anti-tumor effects, as well as the potential for synergistic and cumulative effects. Such interactions underscore the promising prospects for the combined use of OVs and targeted drugs. In addition to the simultaneous administration of these therapeutic agents, researchers have also developed genetically modified OVs that incorporate molecularly targeted drugs. This innovative strategy seeks to leverage the anti-tumor properties of both the targeted drugs and the OVs. However, extensive genetic modifications limit the replication and efficacy of the virus. Therefore, revisiting the fundamental principles of virology and basic immunology is crucial for rationally designing more effective oncolytic virus constructs (114). Multiple preclinical studies have confirmed that OVs combined with targeted drugs can enhance anti-tumor immune responses through multiple mechanisms, and initial results have been achieved in clinical trial progress. To this end, in the present study, preclinical evidence was connected with trial design elements, including patient selection, recommended endpoints, and overlapping toxicity safety monitoring, to construct a clinical translation pathway (Fig. 2). Based on mechanisms of action, potential combination strategies and priority tumor types can be suggested, which in turn provides criteria for patient selection, while the combined efficacy observed in animal models offers guidance on drug dosage and administration sequence. Continuous safety monitoring during these explorations may help prevent serious adverse events caused by potential toxicities. How to further optimize OVs and targeted drugs based on their own characteristics and limitations to advance clinical trials more effectively is a key focus for the future.

Clinical translation roadmap. In
preclinical studies, in vitro experiments provide guidance
on tumor type choice for patient selection in clinical trial
design, and further help set appropriate recommended endpoints
according to different stages of the trial. Mechanistic exploration
suggests possible combination strategies, and the combined efficacy
in animal models provides references for drug dosage and
administration sequence. Potential toxicities in these three areas
indicate the need for safety monitoring to avoid adverse
events.

Figure 2

Clinical translation roadmap. In preclinical studies, in vitro experiments provide guidance on tumor type choice for patient selection in clinical trial design, and further help set appropriate recommended endpoints according to different stages of the trial. Mechanistic exploration suggests possible combination strategies, and the combined efficacy in animal models provides references for drug dosage and administration sequence. Potential toxicities in these three areas indicate the need for safety monitoring to avoid adverse events.

Future development direction: OVs and molecularly targeted drugs combined with precision strategy and OV delivery optimization

Overactivation of signaling pathways is a cause of cancer pathogenesis, being involved in various tumors and playing a critical role in guiding clinical treatment choices and classification (115). Different gene mutation statuses, such as EGFR and KRAS mutations in lung adenocarcinoma, show significant differences in biological behavior and clinicopathological features, leading to completely different treatment options (116). In breast cancer, classification is based on the expression of estrogen receptor, progesterone receptor and HER2 (Luminal A, Luminal B, HER2-positive, triple-negative), which directly determines subsequent treatment strategies (117). Once the key signaling pathways driving tumor growth are identified, targeted drugs can be developed against critical nodes in these pathways. These pathways can also serve as predictive markers for evaluating treatment efficacy and prognosis. For instance, phosphatase and tensin homolog protein loss leading to overactivation of the PI3K-AKT pathway in prostate cancer generally indicates poor prognosis (118). Single-cell RNA sequencing suggests that the C2 insulin-like growth factor 2 tumor subtype is associated with poor prognosis in high-grade serous ovarian cancer, providing a promising target for future therapies (119). Additionally, signaling pathway targets can be used as monitoring indicators for studying resistance mechanisms and adjusting treatment plans; analyzing pathway changes through re-biopsy can reveal resistance mechanisms and guide subsequent treatment. Approximately half of the resistance to first-generation EGFR inhibitors is due to the emergence of the secondary T790M mutation. To address this, the third-generation EGFR inhibitor Osimertinib was developed, which effectively overcomes T790M resistance and has become the standard treatment option after resistance develops (120). Through time-of-flight cytometry, it was found that AXL inhibition can induce JAK1-STAT3 signaling to compensate for the loss of AXL, enhancing the potential for distant metastasis in lung cancer (121). AXL is a member of the TAM receptor family, typically overexpressed in cancers, and has become a potential target for malignant tumors (122). The use of sequencing technology to predict targeted drug targets has formed a set of multi-omics integration strategies. By sequencing the genome, transcriptome and by other omics, key genes related to diseases can be systematically identified, and their potential as drug targets can be evaluated. Advances in targeted drug development have led to new drugs addressing various mutations, enhancing the effectiveness and overcoming the limitations of older treatments. Consequently, precise combination strategies based on molecular typing can significantly enhance anti-tumor effects and reduce drug resistance.

As the first line of defense against the invasion of external pathogenic microorganisms, the antiviral response pathway, in addition to the IFN pathway, The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) and retinoic-acid-inducible gene-I (RIG-I)/mitochondrial antiviral signaling protein (MAVS), also play key roles in antiviral innate immunity, belonging to the cellular stress response pathway. The release of viral DNA and tumor cell nuclear DNA can activate the cGAS-STING pathway, subsequently triggering a series of signaling cascades, thereby inducing the production of type I IFNs and initiating adaptive immune responses (123). However, in various cancers, interruption or loss of the cGAS-STING pathway has been observed, which prevents OVs from effectively activating this pathway, resulting in a failure to initiate immune responses (124-126). The development of various STING agonists has shown enhanced stability and efficacy in anti-tumor applications (127-129). Sibal et al (130) found that the STING activator 2'3'-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) combined with the attenuated HSV-1 strain C-REV could induce a more sustained anti-tumor immune response, but they did not conduct an in-depth analysis of how 2'3'-cGAMP affects the intracellular process dynamics related to viral replication mechanisms (130). The RIG-I/MAVS pathway primarily recognizes viral RNA in the cytoplasm. RIG-I has high affinity for double-stranded (ds)RNA with three phosphates on the 5' end. After binding, it also activates the transcription and expression of type I IFNs and various pro-inflammatory cytokines (131). It was reported that HSV-1 infection induces mitochondrial damage and release of mitochondrial DNA, which can trigger cGAS/STING/interferon regulatory factor 3 and RIG-I-MAVS signaling pathways, suggesting that the RIG-I-MAVS pathway can recognize more than just viral RNA (132). Current research mainly focuses on OVs transforming 'cold' tumors into 'hot' tumors and promoting immune cell infiltration and tumor killing by activating the cGAS-STING and RIG-I/MAVS pathway, generating a strong antitumor immune response, and tumors lacking cGAS or STING expression are more easily infected, thereby enhancing oncolytic activity (133-137). However, if this pathway, which is central to antiviral immunity, is excessively or prematurely activated, it may lead to viral clearance, thereby weakening the direct oncolytic effect of the virus. Precisely regulating the balance between viral replication and immune activation is a major challenge for studying the combined use of OVs and related pathway activators. Furthermore, there are no reports in the literature on whether specific inhibitors exist that can temporarily modulate innate antiviral pathways to allow OV spread without causing systemic immunosuppression, which may become a direction for future research.

When OVs enter the body, their ability to induce a strong anti-tumor immune response is also influenced by another key factor. Previous research has indicated that OVs are swiftly neutralized by antibodies upon entering the body, thereby significantly diminishing their therapeutic efficacy (138,139). To overcome this challenge, researchers have proposed employing vector delivery systems to shield OVs from complement and neutralizing antibodies, thereby enhancing tumor targeting. Polymer coatings (140) and cell-derived nanovesicles (141) has been identified as potential vectors for delivering OVs. Another promising approach involves cell-based drug delivery systems, which provide advantages such as extended drug circulation, improved efficacy, controlled drug release and reduced immunogenicity and cytotoxicity (142). Furthermore, chimeric antigen receptor T-cells have been reported as effective carriers for OVs, offering significant advantages in OV therapy against distant tumors (143). However, the immunosuppressive mechanisms present within the TME may restrict the effectiveness of T cells as OV vectors to tumors that are not classified as immune deserts (144). By contrast, mesenchymal stem cells have been extensively investigated as vectors for various OVs, including oncolytic adenovirus, oncolytic HSV, oncolytic MV and oncolytic reovirus (145). This is attributed to their advantageous properties, such as tumor homing, intrinsic anti-cancer capabilities, protection of OVs from neutralizing antibodies and the ability to deliver viruses to tumor sites via trojan horse strategies (146-148). The development and refinement of targeted delivery systems for OVs substantially mitigate the risk of clearance by the host immune system, thereby addressing some of the challenges associated with combining OVs with molecular targeted therapies and enhancing their anti-tumor efficacy. However, in the development of targeted delivery systems, we must consider not only the immunogenicity of the OVs themselves, but also evaluate whether the delivery system could synergize with the OVs to trigger potential risks such as cytokine storms. Therefore, it is necessary to make a rational selection and modification of the carrier. In addition, efficiently and stably assembling OVs with the delivery system while maintaining the activity of both, and achieving large-scale production at a relatively low cost, remains a significant challenge.

Core challenges in clinical translation

The integration of OVs with molecularly targeted therapies constitutes a promising domain for further exploration. Current research predominantly focuses on preclinical trials, with relatively few studies advancing to clinical trials. This limited progression is likely due to the fact that many viruses do not successfully advance beyond phase I trials (149), which are primarily designed to assess safety and determine appropriate dosing parameters. To date, only 13 viruses have exhibited adequate safety profiles and preliminary clinical success to warrant progression to phase II trials, which are intended to assess clinical efficacy (149). Current methodologies for drug combination exhibit inherent limitations, such as the issue of cumulative toxicity, necessitating further clinical trials to ascertain their safety and efficacy. It is crucial to evaluate the rationale behind these combination strategies and to optimize the use of both the shared and unique properties of the drugs, especially in cases where combinations are ineffective (114). Given that tumor cells frequently develop resistance to monotherapies, employing a combination of diverse therapeutic approaches may partially alleviate this resistance (150-152). However, the potential development of new drug resistance following combination therapy, and the subsequent challenges it may pose to tumor eradication, warrant further rigorous investigation and validation.

The dosing sequence guides the transition from preclinical studies to clinical trials

The efficacy of JX-594 in combination with sorafenib in HCC mentioned in the aforementioned clinical study above is affected by the order of administration, and priority administration of JX-594 may be superior to sorafenib. The underlying mechanisms may be related to the following factors. Immune 'cold tumors' may be more dependent on OV to preferentially initiate immunity (133-135). As a result, preferential injection of OVs can induce immunogenic death of tumor cells, release tumor-associated antigens, promote DC maturation, T-cell infiltration and cytokine secretion, transform 'cold tumors' into 'hot tumors' and provide a more favorable immune environment for subsequent targeted drugs to exert anti-tumor effects (136,153). Some targeted drugs may inhibit OV replication, so prioritizing OV treatment ensures sufficient viral spread and prevents targeted drugs from weakening its oncolytic effect. When OVs are combined with certain antiangiogenic drugs, which induced reduction in blood supply to tumors may limit delivery of OV (154,155). Conversely, targeted drug pretreatment can promote OV replication by downregulating the IFN signaling pathway, a tumor cell defense mechanism (21,34). The use of immunomodulatory drugs to remove immunosuppressive cells such as Tregs and relieve immunosuppression may improve the efficacy of immunotherapy for OV (37,40,41). IFP is one of the key factors hindering the intratumoral distribution of OVs, and the prior use of chemotherapy drugs such as cyclophosphamide can reduce tumor IFP, further improve the intratumoral distribution of OV, and enhance its oncolytic effect (156). Considering the above factors, based on the dynamic changes of tumor characteristics, viral characteristics, drug mechanism and TME, adjusting the order and dose of drugs can enhance the anti-tumor effect.

Potential toxicity and safety management of combinations

OVs and targeted drugs each have their own toxic effects, and when used in combination, careful consideration should be given to whether potential additive toxicity could cause serious adverse events. To date, the combination of therapies mentioned in this study has not yet undergone clinical translation, and the unique toxicities that may arise warrant attention. Protein kinase inhibitors (PKIs) inhibit downstream signaling by targeting specific oncogenic kinases and are one of the research focuses in cancer therapy. However, most analyzed PKI product characteristic summaries and European public assessment reports indicated severe hepatotoxicity (157). Furthermore, the hepatotoxicity caused by OVs themselves also needs to be taken seriously when combined with targeted therapy. Therefore, before treatment, patients at high risk of hepatotoxicity should be identified through relevant biomarkers. In addition, the nonspecific receptor CAR targeted by OVs is also present in normal liver cells (158), and the virus can be genetically engineered to avoid being taken up by the liver or infecting liver cells.

Sunitinib, which is the most involved targeted drug in mechanistic studies, shows a higher prevalence of reduced left ventricular ejection fraction and bone marrow suppression (159). OVs therapy is generally not considered to have significant bone marrow suppression characteristics, and the incidence and severity of hematologic toxicity are usually low. However, certain wild-type OVs, such as oHSV-1, when administered locally to lesions, do not cause viremia, but in rare cases, intravenous administration can lead to a strong systemic immune response due to large-scale viral replication, which may indirectly affect bone marrow function and result in transient cytopenia (160). In addition, bone marrow suppression is also the most common side effect of PARPi (161). Therefore, when considering the combination of sunitinib or PARPi with OVs, the hematological toxicity caused by both should be of concern. The complete blood count should be monitored regularly, and when grade ≥3 toxicity occurs, it is usually necessary to suspend treatment. Once recovery to grade ≤1 is achieved, the dose needs to be reduced upon resuming to avoid irreversible consequences.

Delta-24-RGD is commonly used for intratumoral injection in the treatment of recurrent gliomas, with neurological symptoms being the most common adverse events, such as headaches, speech disorders and cerebral edema (70). ONC201 has shown significant efficacy against certain high-grade gliomas in clinical trials, but it can also lead to adverse events related to the nervous system (162). The therapeutic effect of their combined treatment on tumors is undeniable, but the potential overlapping side effects on the central nervous system should not be overlooked either. Furthermore, distinguishing whether these neurological symptoms are caused by treatment-related inflammation, which may indicate the effectiveness of the treatment, or by the progression of the tumor itself, is crucial in clinical practice and is also a challenge in management. Certain wild-type viruses naturally have neurotropism, and by genetically modifying these viruses, their ability to infect and damage normal nerve cells can be significantly weakened (163-165).

At the same time, the size and location of the tumor, previous treatment history such as radiotherapy that can damage the blood-brain barrier, and pre-existing neurological deficits may all amplify the neurotoxicity of combined therapy (166). Bevacizumab combined with RAMBO can enhance the efficacy against malignant glioma through the CCN1-AKT pathway, but its dual anti-angiogenic effect could significantly increase its impact on the vascular system, excessively suppressing angiogenesis in both the tumor and surrounding normal tissues, leading to tissue ischemia and necrosis (167,168). Therefore, when conducting further clinical research, the optimal order, dosage and interval for administering the two drugs should be determined to balance efficacy and toxicity. In addition, the new genes carried by the genetically engineered recombinant oncolytic viruses may lead to additional side effects. Based on studies of relevant OVs and the toxicity of targeted drugs, future clinical development requires cautious dose exploration and safety management.

In conclusion, the present study collected literature on the mechanisms and clinical research of OVs combined with targeted drugs over the past two decades. There may be potential selection bias, while literature on this research area is relatively sparse and outdated. Additionally, the weighting between preclinical and clinical evidence is uneven and quantitative synthesis of efficacy across studies is limited.

Availability of data and materials

Not applicable.

Authors' contributions

SB and TS conceived the study and wrote the manuscript, and conducted the literature search/selection and data extraction. YT and QW revised and edited the manuscript. All authors have read and approved the final 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.

Acknowledgements

Not applicable.

Funding

This research was funded by the Guangxi Science and Technology Program (grant nos. 2025GXNSFDA069034 and AD25069077), the National Natural Science Foundation of China (grant nos. 82260484, 81860459, 82360587 and 82560548), the Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumors at Guangxi Medical University, Ministry of Education (grant nos. GKE-ZZ2023021 and GKE-ZZ202407) and the Key Projects of Guangxi Natural Science Foundation (grant no. 2024GXNSFDA010022). Additionally, support was provided by the '139' Plan for Cultivating High-Level and Key Talents in Guangxi Medicine, China (grant no. 201903036).

References

1 

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 :

2 

Rasa A and Alberts P: Oncolytic virus preclinical toxicology studies. J Appl Toxicol. 43:620–648. 2023. View Article : Google Scholar

3 

Shalhout SZ, Miller DM, Emerick KS and Kaufman HL: Therapy with oncolytic viruses: Progress and challenges. Nat Rev Clin Oncol. 20:160–177. 2023. View Article : Google Scholar : PubMed/NCBI

4 

Todo T, Ito H, Ino Y, Ohtsu H, Ota Y, Shibahara J and Tanaka M: Intratumoral oncolytic herpes virus G47 for residual or recurrent glioblastoma: A phase 2 trial. Nat Med. 28:1630–1639. 2022. View Article : Google Scholar : PubMed/NCBI

5 

Gazal S, Gazal S, Kaur P, Bhan A and Olagnier D: Breaking barriers: Animal viruses as oncolytic and immunotherapeutic agents for human cancers. Virology. 600:1102382024. View Article : Google Scholar : PubMed/NCBI

6 

Kaufman HL, Shalhout SZ and Iodice G: Talimogene laherparepvec: Moving from first-in-class to best-in-class. Front Mol Biosci. 9:8348412022. View Article : Google Scholar : PubMed/NCBI

7 

Xi P, Zeng D, Chen M, Jiang L, Zhang Y, Qin D, Yao Z and He C: Enhancing pancreatic cancer treatment: The role of H101 oncolytic virus in irreversible electroporation. Front Immunol. 16:15462422025. View Article : Google Scholar : PubMed/NCBI

8 

Lee T, Gianchandani A, Boorjian SA, Shore ND, Narayan VM, Dinney CPN and Kamat AM: Intravesical interferon-α2b gene therapy with nadofaragene firadenovec-vncg: A contemporary review. Future Oncol. 21:2429–2438. 2025. View Article : Google Scholar : PubMed/NCBI

9 

Chen L, Zuo M, Zhou Q and Wang Y: Oncolytic virotherapy in cancer treatment: Challenges and optimization prospects. Front Immunol. 14:13088902023. View Article : Google Scholar

10 

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

11 

Lee YT, Tan YJ and Oon CE: Molecular targeted therapy: Treating cancer with specificity. Eur J Pharmacol. 834:188–196. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Aldea M, Andre F, Marabelle A, Dogan S, Barlesi F and Soria JC: Overcoming resistance to tumor-targeted and immune-targeted therapies. Cancer Discov. 11:874–899. 2021. View Article : Google Scholar : PubMed/NCBI

13 

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

14 

Zhou X, Hu S and Wang X: Recent advances in oncolytic virus combined immunotherapy in tumor treatment. Genes Dis. 12:1015992025. View Article : Google Scholar : PubMed/NCBI

15 

Chen C, Cillis J, Deshpande S, Park AK, Valencia H, Kim SI, Lu J, Vashi Y, Yang A, Zhang Z, et al: Oncolytic virotherapy in solid tumors: A current review. BioDrugs. 39:857–876. 2025. View Article : Google Scholar : PubMed/NCBI

16 

Ullah R, Yin Q, Snell AH and Wan L: RAF-MEK-ERK pathway in cancer evolution and treatment. Semin Cancer Biol. 85:123–154. 2022. View Article : Google Scholar

17 

Lee S, Yang W, Kim DK, Kim H, Shin M, Choi KU, Suh DS, Kim YH, Hwang TH and Kim JH: Inhibition of MEK-ERK pathway enhances oncolytic vaccinia virus replication in doxorubicin-resistant ovarian cancer. Mol Ther Oncolytics. 25:211–224. 2022. View Article : Google Scholar : PubMed/NCBI

18 

Okemoto K, Wagner B, Meisen H, Haseley A, Kaur B and Chiocca EA: STAT3 activation promotes oncolytic HSV1 replication in glioma cells. PLoS One. 8:e719322013. View Article : Google Scholar : PubMed/NCBI

19 

Zhao Q, Zhang R, Qiao C, Miao Y, Yuan Y and Zheng H: Ubiquitination network in the type I IFN-induced antiviral signaling pathway. Eur J Immunol. 53:e23503842023. View Article : Google Scholar : PubMed/NCBI

20 

Schneider W M, Chevillotte M D and Rice CM: Interferon-stimulated genes: A complex web of host defenses. Annu Rev Immunol. 32:513–545. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Zhou X, Zhao J, Zhang JV, Wu Y, Wang L, Chen X, Ji D and Zhou GG: Enhancing therapeutic efficacy of oncolytic herpes simplex virus with MEK inhibitor trametinib in some BRAF or KRAS-Mutated colorectal or lung carcinoma models. Viruses. 13:17582021. View Article : Google Scholar : PubMed/NCBI

22 

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

23 

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

24 

Otani Y, Ishida J, Kurozumi K, Oka T, Shimizu T, Tomita Y, Hattori Y, Uneda A, Matsumoto Y, Michiue H, et al: PIK3R1Met326Ile germline mutation correlates with cysteine-rich protein 61 expression and poor prognosis in glioblastoma. Sci Rep. 7:73912017. View Article : Google Scholar : PubMed/NCBI

25 

Long QZ, Zhou M, Liu XG, Du YF, Fan JH, Li X and He DL: Interaction of CCN1 with αvβ3 integrin induces P-glycoprotein and confers vinblastine resistance in renal cell carcinoma cells. Anticancer Drugs. 24:810–817. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Lin BR, Chang CC, Chen LR, Wu MH, Wang MY, Kuo IH, Chu CY, Chang KJ, Lee PH, Chen WJ, et al: Cysteine-rich 61 (CCN1) enhances chemotactic migration, transendothelial cell migration, and intravasation by concomitantly up-regulating chemokine receptor 1 and 2. Mol Cancer Res. 5:1111–1123. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Di Y, Zhang Y, Nie Q and Chen X: CCN1/Cyr61-PI3K/AKT signaling promotes retinal neovascularization in oxygen-induced retinopathy. Int J Mol Med. 36:1507–1518. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Tomita Y, Kurozumi K, Yoo JY, Fujii K, Ichikawa T, Matsumoto Y, Uneda A, Hattori Y, Shimizu T, Otani Y, et al: Oncolytic herpes virus armed with vasculostatin in combination with bevacizumab abrogates glioma invasion via the CCN1 and AKT signaling pathways. Mol Cancer Ther. 18:1418–1429. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Sugawara K, Iwai M, Ito H, Tanaka M, Seto Y and Todo T: Oncolytic herpes virus G47Δ works synergistically with CTLA-4 inhibition via dynamic intratumoral immune modulation. Mol Ther Oncolytics. 22:129–142. 2021. View Article : Google Scholar : PubMed/NCBI

30 

Ma W, He H and Wang H: Oncolytic herpes simplex virus and immunotherapy. BMC Immunol. 19:402018. View Article : Google Scholar : PubMed/NCBI

31 

Saha D, Wakimoto H, Peters CW, Antoszczyk SJ, Rabkin SD and Martuza RL: Combinatorial effects of VEGFR kinase inhibitor axitinib and oncolytic virotherapy in mouse and human glioblastoma stem-like cell models. Clin Cancer Res. 24:3409–3422. 2018. View Article : Google Scholar : PubMed/NCBI

32 

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

33 

Khoury R, Saleh K, Khalife N, Saleh M, Chahine C, Ibrahim R and Lecesne A: Mechanisms of resistance to antibody-drug conjugates. Int J Mol Sci. 24:96742023. View Article : Google Scholar : PubMed/NCBI

34 

Arulanandam R, Taha Z, Garcia V, Selman M, Chen A, Varette O, Jirovec A, Sutherland K, Macdonald E, Tzelepis F, et al: The strategic combination of trastuzumab emtansine with oncolytic rhabdoviruses leads to therapeutic synergy. Commun Biol. 3:2542020. View Article : Google Scholar : PubMed/NCBI

35 

Du Z, Whitt MA, Baumann J, Garner JM, Morton CL, Davidoff AM and Pfeffer LM: Inhibition of type I interferon-mediated antiviral action in human glioma cells by the IKK inhibitors BMS-345541 and TPCA-1. J Interferon Cytokine Res. 32:368–377. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Dolcetti L, Marigo I, Mantelli B, Peranzoni E, Zanovello P and Bronte V: Myeloid-derived suppressor cell role in tumor-related inflammation. Cancer Lett. 267:216–225. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Lawson KA, Mostafa AA, Shi ZQ, Spurrell J, Chen W, Kawakami J, Gratton K, Thakur S and Morris DG: Repurposing sunitinib with oncolytic reovirus as a novel immunotherapeutic strategy for renal cell carcinoma. Clin Cancer Res. 22:5839–5850. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Moehler M, Sieben M, Roth S, Springsguth F, Leuchs B, Zeidler M, Dinsart C, Rommelaere J and Galle PR: Activation of the human immune system by chemotherapeutic or targeted agents combined with the oncolytic parvovirus H-1. BMC Cancer. 11:4642011. View Article : Google Scholar : PubMed/NCBI

39 

Ichikawa M, Williams R, Wang L, Vogl T and Srikrishna G: S100A8/A9 activate key genes and pathways in colon tumor progression. Mol Cancer Res. 9:133–148. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Chai H, Xu H, Jiang S, Zhang T, Chen J, Zhu R, Wang Y, Sun M, Liu B, Wang X, et al: Neural stem cell-delivered oncolytic virus via intracerebroventricular administration enhances glioblastoma therapy and immune modulation. J Immunother Cancer. 13:e0129342025. View Article : Google Scholar : PubMed/NCBI

41 

Zhu Z, Chen H, Feng C, Chen L, Ma C, Liu Z, Qu Z, Bartlett DL, Lu B, Li K and Guo ZS: Specific inhibitor to KRASG12C induces tumor-specific immunity and synergizes with oncolytic virus for enhanced cancer immunotherapy. J Immunother Cancer. 13:e0105142025. View Article : Google Scholar

42 

Herman ML, Geanes ES, McLennan R, Greening GJ, Mwitanti H and Bradley T: ICAM-1 autoantibodies detected in healthy individuals and cross-react with functional epitopes. Immunohorizons. 9:vlaf0252025. View Article : Google Scholar : PubMed/NCBI

43 

Shin J, Lim J, Han D, Lee S, Sung NS, Kim JS, Kim DK, Lee HY, Lee SK, Shin J, et al: TBK1 inhibitor amlexanox exerts anti-cancer effects against endometrial cancer by regulating AKT/NF-κB signaling. Int J Biol Sci. 21:143–159. 2025. View Article : Google Scholar :

44 

Guo X, Feng H, Xi Z, Zhou J, Huang Z, Guo J, Zheng J, Lyu Z, Liu Y, Zhou J, et al: Targeting TBK1 potentiates oncolytic virotherapy via amplifying ICAM1-mediated NK cell immunity in chemo-resistant colorectal cancer. J Immunother Cancer. 13:e0114552025. View Article : Google Scholar : PubMed/NCBI

45 

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

46 

Yoo JY, Swanner J, Otani Y, Nair M, Park F, Banasavadi-Siddegowda Y, Liu J, Jaime-Ramirez AC, Hong B, Geng F, et al: Oncolytic HSV therapy increases trametinib access to brain tumors and sensitizes them in vivo. Neuro Oncol. 21:1131–1140. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Burch AD and Weller SK: Herpes simplex virus type 1 DNA polymerase requires the mammalian chaperone hsp90 for proper localization to the nucleus. J Virol. 79:10740–10749. 2005. View Article : Google Scholar : PubMed/NCBI

48 

Yoo JY, Hurwitz BS, Bolyard C, Yu JG, Zhang J, Selvendiran K, Rath KS, He S, Bailey Z, Eaves D, et al: Bortezomib-induced unfolded protein response increases oncolytic HSV-1 replication resulting in synergistic antitumor effects. Clin Cancer Res. 20:3787–3798. 2014. View Article : Google Scholar : PubMed/NCBI

49 

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

50 

Napolitano F, Di Somma S, Castellano G, Amato J, Pagano B, Randazzo A, Portella G and Malfitano AM: Combination of dl922-947 oncolytic adenovirus and G-quadruplex binders uncovers improved antitumor activity in breast cancer. Cells. 11:24822022. View Article : Google Scholar : PubMed/NCBI

51 

Passaro C, Volpe M, Botta G, Scamardella E, Perruolo G, Gillespie D, Libertini S and Portella G: PARP inhibitor olaparib increases the oncolytic activity of dl922-947 in in vitro and in vivo model of anaplastic thyroid carcinoma. Mol Oncol. 9:78–92. 2015. View Article : Google Scholar

52 

Kyula-Currie J, Roulstone V, Wright J, Butera F, Legrand A, Elliott R, McLaughlin M, Bozhanova G, Krastev D, Pettitt S, et al: The PARP inhibitor talazoparib synergizes with reovirus to induce cancer killing and tumour control in vivo in mouse models. Nat Commun. 16:62992025. View Article : Google Scholar : PubMed/NCBI

53 

Zhong Y, Le H, Zhang X, Dai Y, Guo F, Ran X, Hu G, Xie Q, Wang D and Cai Y: Identification of restrictive molecules involved in oncolytic virotherapy using genome-wide CRISPR screening. J Hematol Oncol. 17:362024. View Article : Google Scholar : PubMed/NCBI

54 

Tapeinos C and Pandit A: Physical, chemical, and biological structures based on ROS-Sensitive moieties that are able to respond to oxidative microenvironments. Adv Mater. 28:5553–5585. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Schieber M and Chandel NS: ROS function in redox signaling and oxidative stress. Curr Biol. 24:R453–R462. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Wang L, Wang D, Sonzogni O, Ke S, Wang Q, Thavamani A, Batalini F, Stopka SA, Regan MS, Vandal S, et al: PARP-inhibition reprograms macrophages toward an anti-tumor phenotype. Cell Rep. 41:1114622022. View Article : Google Scholar : PubMed/NCBI

57 

Tubbs A and Nussenzweig A: Endogenous DNA damage as a source of genomic instability in cancer. Cell. 168:644–656. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Packiriswamy N, Upreti D, Zhou Y, Khan R, Miller A, Diaz RM, Rooney CM, Dispenzieri A, Peng KW and Russell SJ: Oncolytic measles virus therapy enhances tumor antigen-specific T-cell responses in patients with multiple myeloma. Leukemia. 34:3310–3322. 2020. View Article : Google Scholar : PubMed/NCBI

59 

Zhou S, Li Y, Huang F, Zhang B, Yi T, Li Z, Luo H, He X, Zhong Q, Bian C, et al: Live-attenuated measles virus vaccine confers cell contact loss and apoptosis of ovarian cancer cells via ROS-induced silencing of E-cadherin by methylation. Cancer Lett. 318:14–25. 2012. View Article : Google Scholar : PubMed/NCBI

60 

Zhang CD, Jiang LH, Zhou X, He YP, Liu Y, Zhou DM, Lv Y, Wu BQ and Zhao ZY: Synergistic antitumor efficacy of rMV-Hu191 and Olaparib in pancreatic cancer by generating oxidative DNA damage and ROS-dependent apoptosis. Transl Oncol. 39:1018122024. View Article : Google Scholar

61 

Meikrantz W and Schlegel R: Apoptosis and the cell cycle. J Cell Biochem. 58:160–174. 1995. View Article : Google Scholar : PubMed/NCBI

62 

Lee B, Min JA, Nashed A, Lee SO, Yoo JC, Chi SW and Yi GS: A novel mechanism of irinotecan targeting MDM2 and Bcl-xL. Biochem Biophys Res Commun. 514:518–523. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Hubbard JM and Grothey A: Napabucasin: An update on the first-in-class cancer stemness inhibitor. Drugs. 77:1091–1103. 2017. View Article : Google Scholar : PubMed/NCBI

64 

Babaei A, Soleimanjahi H, Soleimani M and Arefian E: The synergistic anticancer effects of ReoT3D, CPT-11, and BBI608 on murine colorectal cancer cells. Daru. 28:555–565. 2020. View Article : Google Scholar : PubMed/NCBI

65 

Chen Y and Zhou X: Research progress of mTOR inhibitors. Eur J Med Chem. 208:1128202020. View Article : Google Scholar : PubMed/NCBI

66 

Wang Y and Zhang H: Regulation of autophagy by mTOR signaling pathway. Adv Exp Med Biol. 1206:67–83. 2019. View Article : Google Scholar : PubMed/NCBI

67 

Yoshida GJ: Therapeutic strategies of drug repositioning targeting autophagy to induce cancer cell death: From pathophysiology to treatment. J Hematol Oncol. 10:672017. View Article : Google Scholar : PubMed/NCBI

68 

Saran U, Foti M and Dufour JF: Cellular and molecular effects of the mTOR inhibitor everolimus. Clin Sci (Lond). 129:895–914. 2015. View Article : Google Scholar : PubMed/NCBI

69 

Alonso MM, Jiang H, Yokoyama T, Xu J, Bekele NB, Lang FF, Kondo S, Gomez-Manzano C and Fueyo J: Delta-24-RGD in combination with RAD001 induces enhanced anti-glioma effect via autophagic cell death. Mol Ther. 16:487–493. 2008. View Article : Google Scholar : PubMed/NCBI

70 

Lang FF, Conrad C, Gomez-Manzano C, Yung WKA, Sawaya R, Weinberg JS, Prabhu SS, Rao G, Fuller GN, Aldape KD, et al: Phase I study of DNX-2401 (Delta-24-RGD) oncolytic adenovirus: Replication and immunotherapeutic effects in recurrent malignant glioma. J Clin Oncol. 36:1419–1427. 2018. View Article : Google Scholar : PubMed/NCBI

71 

Xue C, Chu Q, Shi Q, Zeng Y, Lu J and Li L: Wnt signaling pathways in biology and disease: Mechanisms and therapeutic advances. Signal Transduct Target Ther. 10:1062025. View Article : Google Scholar : PubMed/NCBI

72 

Zhang H, Hang W, Jing Z, Liu B, Wang X, Li Y, Luo H, Lv H, Tao X, Timashev P, et al: The role of notch signaling pathway in cancer: Mechanistic insights, therapeutic potential, and clinical progress. Front Immunol. 16:15675242025. View Article : Google Scholar : PubMed/NCBI

73 

Yang H, Yang J, Zheng X, Chen T, Zhang R, Chen R, Cao T, Zeng F and Liu Q: The hippo pathway in breast cancer: The extracellular matrix and hypoxia. Int J Mol Sci. 25:128682024. View Article : Google Scholar : PubMed/NCBI

74 

Ushijima Y, Luo C, Goshima F, Yamauchi Y, Kimura H and Nishiyama Y: Determination and analysis of the DNA sequence of highly attenuated herpes simplex virus type 1 mutant HF10, a potential oncolytic virus. Microbes Infect. 9:142–149. 2007. View Article : Google Scholar : PubMed/NCBI

75 

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

76 

Tan G, Kasuya H, Sahin TT, Yamamura K, Wu Z, Koide Y, Hotta Y, Shikano T, Yamada S, Kanzaki A, et al: Combination therapy of oncolytic herpes simplex virus HF10 and bevacizumab against experimental model of human breast carcinoma xenograft. Int J Cancer. 136:1718–1730. 2015. View Article : Google Scholar

77 

Libertini S, Iacuzzo I, Perruolo G, Scala S, Ieranò C, Franco R, Hallden G and Portella G: Bevacizumab increases viral distribution in human anaplastic thyroid carcinoma xenografts and enhances the effects of E1A-defective adenovirus dl922-947. Clin Cancer Res. 14:6505–6514. 2008. View Article : Google Scholar : PubMed/NCBI

78 

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

79 

Bin Y, Ren J and Zhang H, Zhang T, Liu P, Xin Z, Yang H, Feng Z, Chen Z and Zhang H: Against all odds: The road to success in the development of human immune reconstitution mice. Animal Model Exp Med. 7:460–470. 2024. View Article : Google Scholar : PubMed/NCBI

80 

Chen J, Liao S, Xiao Z and Pan Q, Wang X, Shen K, Wang S, Yang L, Guo F, Liu HF and Pan Q: The development and improvement of immunodeficient mice and humanized immune system mouse models. Front Immunol. 13:10075792022. View Article : Google Scholar : PubMed/NCBI

81 

Reeh M, Bockhorn M, Görgens D, Vieth M, Hoffmann T, Simon R, Izbicki JR, Sauter G, Schumacher U and Anders M: Presence of the coxsackievirus and adenovirus receptor (CAR) in human neoplasms: A multitumour array analysis. Br J Cancer. 109:1848–1858. 2013. View Article : Google Scholar : PubMed/NCBI

82 

Cohen CJ, Shieh JT, Pickles RJ, Okegawa T, Hsieh JT and Bergelson JM: The coxsackievirus and adenovirus receptor is a transmembrane component of the tight junction. Proc Natl Acad Sci USA. 98:15191–15196. 2001. View Article : Google Scholar : PubMed/NCBI

83 

Su Y, Liu Y, Behrens CR, Bidlingmaier S, Lee NK, Aggarwal R, Sherbenou DW, Burlingame AL, Hann BC, Simko JP, et al: Targeting CD46 for both adenocarcinoma and neuroendocrine prostate cancer. JCI Insight. 3:e1214972018. View Article : Google Scholar : PubMed/NCBI

84 

Do MH, To PK, Cho YS, Kwon SY, Hwang EC, Choi C, Cho SH, Lee SJ, Hemmi S and Jung C: Targeting CD46 enhances anti-tumoral activity of adenovirus type 5 for bladder cancer. Int J Mol Sci. 19:26942018. View Article : Google Scholar : PubMed/NCBI

85 

Trinh HV, Lesage G, Chennamparampil V, Vollenweider B, Burckhardt CJ, Schauer S, Havenga M, Greber UF and Hemmi S: Avidity binding of human adenovirus serotypes 3 and 7 to the membrane cofactor CD46 triggers infection. J Virol. 86:1623–1637. 2012. View Article : Google Scholar :

86 

Dhiman N, Jacobson RM and Poland GA: Measles virus receptors: SLAM and CD46. Rev Med Virol. 14:217–229. 2004. View Article : Google Scholar : PubMed/NCBI

87 

Nestić D, Uil TG, Ma J, Roy S, Vellinga J, Baker AH, Custers J and Majhen D: αvβ3 integrin is required for efficient infection of epithelial cells with human adenovirus type 26. J Virol. 93:e01474–18. 2018.

88 

Weis SM and Cheresh DA: αV integrins in angiogenesis and cancer. Cold Spring Harb Perspect Med. 1:a0064782011. View Article : Google Scholar

89 

Lyle C and McCormick F: Integrin alphavbeta5 is a primary receptor for adenovirus in CAR-negative cells. Virol J. 7:1482010. View Article : Google Scholar : PubMed/NCBI

90 

Koehler M, Petitjean SJL, Yang J, Aravamudhan P, Somoulay X, Lo Giudice C, Poncin MA, Dumitru AC, Dermody TS and Alsteens D: Reovirus directly engages integrin to recruit clathrin for entry into host cells. Nat Commun. 12:21492021. View Article : Google Scholar : PubMed/NCBI

91 

Ioannou M and Stanway G: Tropism of Coxsackie virus A9 depends on the +1 position of the RGD (arginine-glycine-aspartic acid) motif found at the C' terminus of its VP1 capsid protein. Virus Res. 294:1982922021. View Article : Google Scholar

92 

Stern PL and Harrop R: 5T4 oncofoetal antigen: An attractive target for immune intervention in cancer. Cancer Immunol Immunother. 66:415–426. 2017. View Article : Google Scholar

93 

Scurr M, Pembroke T, Bloom A, Roberts D, Thomson A, Smart K, Bridgeman H, Adams R, Brewster A, Jones R, et al: Effect of modified vaccinia ankara-5T4 and low-dose cyclophosphamide on antitumor immunity in metastatic colorectal cancer: A Randomized clinical trial. JAMA Oncol. 3:e1725792017. View Article : Google Scholar : PubMed/NCBI

94 

Jian C, Jing Z, Yinhang W, Jinlong D, Yuefen P, Quan Q and Shuwen H: Colorectal cancer and gut viruses: A visualized analysis based on CiteSpace knowledge graph. Front Microbiol. 14:12398182023. View Article : Google Scholar : PubMed/NCBI

95 

Yi J, Quji S, Guo L, Chai Z, Kong X and Meng J: Exploring novel strategies of oncolytic viruses and gut microbiota to enhance CAR-T cell therapy for colorectal cancer. Cell Immunol. 417:1050262025. View Article : Google Scholar : PubMed/NCBI

96 

Boixareu C, Taha T, Venkadakrishnan VB, de Bono J and Beltran H: Targeting the tumour cell surface in advanced prostate cancer. Nat Rev Urol. 22:569–589. 2025. View Article : Google Scholar : PubMed/NCBI

97 

Vannini A, Parenti F, Bressanin D, Barboni C, Zaghini A, Campadelli-Fiume G and Gianni T: Towards a precision medicine approach and in situ vaccination against prostate cancer by PSMA-Retargeted oHSV. Viruses. 13:20852021. View Article : Google Scholar : PubMed/NCBI

98 

Li Y, Shen Y, Tang T, Tang Z, Song W, Yang Z, Zhang X, Wang M, Bai X and Liang T: Oncolytic virus combined with traditional treatment versus traditional treatment alone in patients with cancer: A meta-analysis. Int J Clin Oncol. 25:1901–1913. 2020. View Article : Google Scholar : PubMed/NCBI

99 

Hirooka Y, Kasuya H, Ishikawa T, Kawashima H, Ohno E, Villalobos IB, Naoe Y, Ichinose T, Koyama N, Tanaka M, et al: A Phase I clinical trial of EUS-guided intratumoral injection of the oncolytic virus, HF10 for unresectable locally advanced pancreatic cancer. BMC Cancer. 18:5962018. View Article : Google Scholar : PubMed/NCBI

100 

Kim JH, Oh JY, Park BH, Lee DE, Kim JS, Park HE, Roh MS, Je JE, Yoon JH, Thorne SH, et al: Systemic armed oncolytic and immunologic therapy for cancer with JX-594, a targeted poxvirus expressing GM-CSF. Mol Ther. 14:361–370. 2006. View Article : Google Scholar : PubMed/NCBI

101 

Breitbach CJ, Parato K, Burke J, Hwang TH, Bell JC and Kirn DH: Pexa-Vec double agent engineered vaccinia: Oncolytic and active immunotherapeutic. Curr Opin Virol. 13:49–54. 2015. View Article : Google Scholar : PubMed/NCBI

102 

Heo J, Breitbach CJ, Moon A, Kim CW, Patt R, Kim MK, Lee YK, Oh SY, Woo HY, Parato K, et al: Sequential therapy with JX-594, a targeted oncolytic poxvirus, followed by sorafenib in hepatocellular carcinoma: Preclinical and clinical demonstration of combination efficacy. Mol Ther. 19:1170–1179. 2011. View Article : Google Scholar : PubMed/NCBI

103 

Moehler M, Heo J, Lee HC, Tak WY, Chao Y, Paik SW, Yim HJ, Byun KS, Baron A, Ungerechts G, et al: Vaccinia-based oncolytic immunotherapy Pexastimogene Devacirepvec in patients with advanced hepatocellular carcinoma after sorafenib failure: A randomized multicenter Phase IIb trial (TRAVERSE). Oncoimmunology. 8:16158172019. View Article : Google Scholar : PubMed/NCBI

104 

Abou-Alfa GK, Galle PR, Chao Y, Erinjeri J, Heo J, Borad MJ, Luca A, Burke J, Pelusio A, Agathon D, et al: PHOCUS: A phase 3, Randomized, open-label study of sequential treatment with pexa-vec (JX-594) and sorafenib in patients with advanced hepatocellular carcinoma. Liver Cancer. 13:248–264. 2024. View Article : Google Scholar : PubMed/NCBI

105 

Chaurasiya S, Yang A, Zhang Z, Lu J, Valencia H, Kim SI, Woo Y, Warner SG, Olafsen T, Zhao Y, et al: A comprehensive preclinical study supporting clinical trial of oncolytic chimeric poxvirus CF33-hNIS-anti-PD-L1 to treat breast cancer. Mol Ther Methods Clin Dev. 24:102–116. 2021. View Article : Google Scholar

106 

Yuan Y, Egelston C, Colunga Flores O, Chaurasiya S, Lin D, Chang H, Chong LMO, Seiz A, Shah M, Meisen WH, et al: CF33-hNIS-anti-PD-L1 oncolytic virus followed by trastuzumab-deruxtecan in a patient with metastatic triple negative breast cancer: A case study. Ther Adv Med Oncol. 15:175883592312106752023. View Article : Google Scholar : PubMed/NCBI

107 

Tilgase A, Olmane E, Nazarovs J, Brokāne L, Erdmanis R, Rasa A and Alberts P: Multimodality treatment of a colorectal cancer stage IV patient with FOLFOX-4, bevacizumab, rigvir oncolytic virus, and surgery. Case Rep Gastroenterol. 12:457–465. 2018. View Article : Google Scholar : PubMed/NCBI

108 

Filho AM, Laversanne M, Ferlay J, Colombet M, Piñeros M, Znaor A, Parkin DM, Soerjomataram I and Bray F: The GLOBOCAN 2022 cancer estimates: Data sources, methods, and a snapshot of the cancer burden worldwide. Int J Cancer. 156:1336–1346. 2025. View Article : Google Scholar

109 

Liu S, Jiang W, Sheng J, Wang L and Cui M: Adoptive cell therapy for cancer: Combination strategies and biomarkers. Front Immunol. 16:16037922025. View Article : Google Scholar : PubMed/NCBI

110 

Goswami S, Pauken KE, Wang L and Sharma P: Next-generation combination approaches for immune checkpoint therapy. Nat Immunol. 25:2186–2199. 2024. View Article : Google Scholar : PubMed/NCBI

111 

Xie B, Zhang L, Hu W, Fan M, Jiang N, Duan Y, Jing D, Xiao W, Fragoso RC, Lam KS, et al: Dual blockage of STAT3 and ERK1/2 eliminates radioresistant GBM cells. Redox Biol. 24:1011892019. View Article : Google Scholar : PubMed/NCBI

112 

Nagathihalli NS, Castellanos JA, Lamichhane P, Messaggio F, Shi C, Dai X, Rai P, Chen X, VanSaun MN and Merchant NB: Inverse correlation of STAT3 and MEK signaling mediates resistance to RAS pathway inhibition in pancreatic cancer. Cancer Res. 78:6235–6246. 2018. View Article : Google Scholar : PubMed/NCBI

113 

Vultur A, Villanueva J, Krepler C, Rajan G, Chen Q, Xiao M, Li L, Gimotty PA, Wilson M, Hayden J, et al: MEK inhibition affects STAT3 signaling and invasion in human melanoma cell lines. Oncogene. 33:1850–1861. 2014. View Article : Google Scholar

114 

Appleton E, Chiocca EA, Ungerechts G, Melcher A and Vile R: Oncolytic viruses as anticancer agents: clinical progress and remaining challenges. Lancet. 406:1295–1312. 2025. View Article : Google Scholar : PubMed/NCBI

115 

Hossain MA: Targeting the RAS upstream and downstream signaling pathway for cancer treatment. Eur J Pharmacol. 979:1767272024. View Article : Google Scholar : PubMed/NCBI

116 

Zou J, Han W, Hu Y, Zeng C, Li J, Lei W, Cao J, Fei Q, Shao M, Yi J, et al: Gene mutation, clinical characteristics and pathology in resectable lung adenocarcinoma. World J Surg Oncol. 23:162025. View Article : Google Scholar : PubMed/NCBI

117 

Yang T, Li W, Huang T and Zhou J: Genetic testing enhances the precision diagnosis and treatment of breast cancer. Int J Mol Sci. 24:166072023. View Article : Google Scholar : PubMed/NCBI

118 

Chaudagar K, Hieromnimon HM, Khurana R, Labadie B, Hirz T, Mei S, Hasan R, Shafran J, Kelley A, Apostolov E, et al: Reversal of lactate and PD-1-mediated macrophage immunosuppression controls growth of PTEN/p53-deficient prostate cancer. Clin Cancer Res. 29:1952–1968. 2023. View Article : Google Scholar : PubMed/NCBI

119 

Zhao F, Jiang X, Li Y, Huang T, Xiahou Z, Nie W and Li Q: Characterizing tumor biology and immune microenvironment in high-grade serous ovarian cancer via single-cell RNA sequencing: Insights for targeted and personalized immunotherapy strategies. Front Immunol. 15:15001532025. View Article : Google Scholar : PubMed/NCBI

120 

Cooper AJ, Sequist LV and Lin JJ: Third-generation EGFR and ALK inhibitors: Mechanisms of resistance and management. Nat Rev Clin Oncol. 19:499–514. 2022. View Article : Google Scholar : PubMed/NCBI

121 

Taverna JA, Hung CN, DeArmond DT, Chen M, Lin CL, Osmulski PA, Gaczynska ME, Wang CM, Lucio ND, Chou CW, et al: Single-cell proteomic profiling identifies combined AXL and JAK1 inhibition as a novel therapeutic strategy for lung cancer. Cancer Res. 80:1551–1563. 2020. View Article : Google Scholar : PubMed/NCBI

122 

Yadav M, Sharma A, Patne K, Tabasum S, Suryavanshi J, Rawat L, Machaalani M, Eid M, Singh RP, Choueiri TK, et al: AXL signaling in cancer: From molecular insights to targeted therapies. Signal Transduct Target Ther. 10:372025. View Article : Google Scholar : PubMed/NCBI

123 

Shen M, Jiang X, Peng Q, Oyang L, Ren Z, Wang J, Peng M, Zhou Y, Deng X and Liao Q: The cGAS-STING pathway in cancer immunity: mechanisms, challenges, and therapeutic implications. J Hematol Oncol. 18:402025. View Article : Google Scholar

124 

Xia T, Konno H, Ahn J and Barber GN: Deregulation of STING signaling in colorectal carcinoma constrains DNA damage responses and correlates with tumorigenesis. Cell Rep. 14:282–297. 2016. View Article : Google Scholar : PubMed/NCBI

125 

Xia T, Konno H and Barber GN: Recurrent loss of STING signaling in melanoma correlates with susceptibility to viral oncolysis. Cancer Res. 76:6747–6759. 2016. View Article : Google Scholar : PubMed/NCBI

126 

de Queiroz N, Xia T, Konno H and Barber GN: Ovarian cancer cells commonly exhibit defective STING signaling which affects sensitivity to viral oncolysis. Mol Cancer Res. 17:974–986. 2019. View Article : Google Scholar :

127 

Meric-Bernstam F, Sweis RF, Hodi FS, Messersmith WA, Andtbacka RHI, Ingham M, Lewis N, Chen X, Pelletier M, Chen X, et al: Phase I dose-escalation trial of MIW815 (ADU-S100), an intratumoral STING agonist, in patients with advanced/metastatic solid tumors or lymphomas. Clin Cancer Res. 28:677–688. 2022. View Article : Google Scholar

128 

Chang W, Altman MD, Lesburg CA, Perera SA, Piesvaux JA, Schroeder GK, Wyss DF, Cemerski S, Chen Y, DiNunzio E, et al: Discovery of MK-1454: A potent cyclic dinucleotide stimulator of interferon genes agonist for the treatment of cancer. J Med Chem. 65:5675–5689. 2022. View Article : Google Scholar : PubMed/NCBI

129 

Song Z, Wang X, Zhang Y, Gu W, Shen A, Ding C, Li H, Xiao R, Geng M, Xie Z and Zhang A: Structure-activity relationship study of amidobenzimidazole analogues leading to potent and systemically administrable stimulator of interferon gene (STING) agonists. J Med Chem. 64:1649–1669. 2021. View Article : Google Scholar : PubMed/NCBI

130 

Sibal PA, Matsumura S, Ichinose T, Bustos-Villalobos I, Morimoto D, Eissa IR, Abdelmoneim M, Aboalela MAM, Mukoyama N, Tanaka M, et al: STING activator 2'3'-cGAMP enhanced HSV-1-based oncolytic viral therapy. Mol Oncol. 18:1259–1277. 2024. View Article : Google Scholar : PubMed/NCBI

131 

Thoresen D, Wang W, Galls D, Guo R, Xu L and Pyle AM: The molecular mechanism of RIG-I activation and signaling. Immunol Rev. 304:154–168. 2021. View Article : Google Scholar : PubMed/NCBI

132 

Berry N, Suspène R, Caval V, Khalfi P, Beauclair G, Rigaud S, Blanc H, Vignuzzi M, Wain-Hobson S and Vartanian JP: Herpes simplex virus type 1 infection disturbs the mitochondrial network, leading to type I interferon production through the RNA polymerase III/RIG-I pathway. mBio. 12:e02557212021. View Article : Google Scholar : PubMed/NCBI

133 

Farhangnia P, Khorramdelazad H, Nickho H and Delbandi AA: Current and future immunotherapeutic approaches in pancreatic cancer treatment. J Hematol Oncol. 17:402024. View Article : Google Scholar : PubMed/NCBI

134 

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

135 

Shen KY, Zhu Y, Xie SZ and Qin LX: Immunosuppressive tumor microenvironment and immunotherapy of hepatocellular carcinoma: Current status and prospectives. J Hematol Oncol. 17:252024. View Article : Google Scholar : PubMed/NCBI

136 

Wang JW, Feng Q, Liu JH and Xun JJ: Opportunities, challenges, and future perspectives of oncolytic virus therapy for malignant melanoma. Front Immunol. 16:16536832025. View Article : Google Scholar : PubMed/NCBI

137 

Wang JW, Liu JH, Liu YL, Xu WZ and Zhang ZB: Oncolytic virus therapy in the elderly: Immune frailty, challenges, and perspectives. Front Immunol. 16:16866592025. View Article : Google Scholar : PubMed/NCBI

138 

Lang SI, Giese NA, Rommelaere J, Dinsart C and Cornelis JJ: Humoral immune responses against minute virus of mice vectors. J Gene Med. 8:1141–1150. 2006. View Article : Google Scholar : PubMed/NCBI

139 

Hirasawa K, Nishikawa SG, Norman KL, Coffey MC, Thompson BG, Yoon CS, Waisman DM and Lee PW: Systemic reovirus therapy of metastatic cancer in immune-competent mice. Cancer Res. 63:348–353. 2003.PubMed/NCBI

140 

Chen G, Yuan Y, Li Y, He Q, Qin Z, Hu H, Gao C, Xu Z, Xu Q, Gao Q and Li F: Enhancing oncolytic virus efficiency with methionine and N-(3-aminoprolil)methacrylamide modified acrylamide cationic block polymer. J Mater Chem B. 12:3741–3750. 2024. View Article : Google Scholar : PubMed/NCBI

141 

Perera AS and Coppens MO: Re-designing materials for biomedical applications: From biomimicry to nature-inspired chemical engineering. Philos Trans A Math Phys Eng Sci. 377:201802682019.PubMed/NCBI

142 

Pang L, Zhang C, Qin J, Han L, Li R, Hong C, He H and Wang J: A novel strategy to achieve effective drug delivery: Exploit cells as carrier combined with nanoparticles. Drug Deliv. 24:83–91. 2017. View Article : Google Scholar : PubMed/NCBI

143 

Zhang Z, Yang N, Xu L, Lu H, Chen Y, Wang Z, Lu Q, Zhong K, Zhu Z, Wang G, et al: Systemic delivery of oncolytic herpes virus using CAR-T cells enhances targeting of antitumor immuno-virotherapy. Cancer Immunol Immunother. 73:1732024. View Article : Google Scholar : PubMed/NCBI

144 

Reale A, Calistri A and Altomonte J: Giving oncolytic viruses a free ride: Carrier cells for oncolytic virotherapy. Pharmaceutics. 13:21922021. View Article : Google Scholar : PubMed/NCBI

145 

Ghasemi Darestani N, Gilmanova AI, Al-Gazally ME, Zekiy AO, Ansari MJ, Zabibah RS, Jawad MA, Al-Shalah SAJ, Rizaev JA, Alnassar YS, et al: Mesenchymal stem cell-released oncolytic virus: An innovative strategy for cancer treatment. Cell Commun Signal. 21:432023. View Article : Google Scholar : PubMed/NCBI

146 

Collet G, Grillon C, Nadim M and Kieda C: Trojan horse at cellular level for tumor gene therapies. Gene. 525:208–216. 2013. View Article : Google Scholar : PubMed/NCBI

147 

Mader EK, Maeyama Y, Lin Y, Butler GW, Russell HM, Galanis E, Russell SJ, Dietz AB and Peng KW: Mesenchymal stem cell carriers protect oncolytic measles viruses from antibody neutralization in an orthotopic ovarian cancer therapy model. Clin Cancer Res. 15:7246–7255. 2009. View Article : Google Scholar : PubMed/NCBI

148 

Hakkarainen T, Särkioja M, Lehenkari P, Miettinen S, Ylikomi T, Suuronen R, Desmond RA, Kanerva A and Hemminki A: Human mesenchymal stem cells lack tumor tropism but enhance the antitumor activity of oncolytic adenoviruses in orthotopic lung and breast tumors. Hum Gene Ther. 18:627–641. 2007. View Article : Google Scholar : PubMed/NCBI

149 

Cook M and Chauhan A: Clinical application of oncolytic viruses: A systematic review. Int J Mol Sci. 21:75052020. View Article : Google Scholar : PubMed/NCBI

150 

Rathod LS, Sakle NS and Mokale SN: KRAS inhibitors in drug resistance and potential for combination therapy. Tumori. 111:20–40. 2025. View Article : Google Scholar

151 

Rahimi A, Baghernejadan Z, Hazrati A, Malekpour K, Samimi LN, Najafi A, Falak R and Khorramdelazad H: Combination therapy with immune checkpoint inhibitors in colorectal cancer: Challenges, resistance mechanisms, and the role of microbiota. Biomed Pharmacother. 186:1180142025. View Article : Google Scholar : PubMed/NCBI

152 

He X, Deng H, Liu W, Hu L and Tan X: Advances in understanding drug resistance mechanisms and innovative clinical treatments for melanoma. Curr Treat Options Oncol. 25:1615–1633. 2024. View Article : Google Scholar : PubMed/NCBI

153 

Xuan Y, Yan W, Wang R, Wang X, Guo Y, Dun H, Huan Z, Xu L, Han R, Sun X, et al: GM-CSF and IL-21-armed oncolytic vaccinia virus significantly enhances anti-tumor activity and synergizes with anti-PD1 immunotherapy in pancreatic cancer. Front Immunol. 15:15066322025. View Article : Google Scholar : PubMed/NCBI

154 

Ottolino-Perry K, Tang N, Head R, Ng C, Arulanandam R, Angarita FA, Acuna SA, Chen Y, Bell J, Dacosta RS and McCart JA: Tumor vascularization is critical for oncolytic vaccinia virus treatment of peritoneal carcinomatosis. Int J Cancer. 134:717–730. 2014. View Article : Google Scholar

155 

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

156 

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

157 

Maliepaard M, Faber YS and van Bussel MTJ: Reported hepatotoxicity and hepatotoxicity guidance in the product information of protein kinase inhibitors in oncology registered at the European medicines agency. Pharmacol Res Perspect. 11:e010672023. View Article : Google Scholar : PubMed/NCBI

158 

Godoy P, Hewitt NJ, Albrecht U, Andersen ME, Ansari N, Bhattacharya S, Bode JG, Bolleyn J, Borner C, Böttger J, et al: Recent advances in 2D and 3D in vitro systems using primary hepatocytes, alternative hepatocyte sources and non-parenchymal liver cells and their use in investigating mechanisms of hepatotoxicity, cell signaling and ADME. Arch Toxicol. 87:1315–1530. 2013. View Article : Google Scholar : PubMed/NCBI

159 

Rock EP, Goodman V, Jiang JX, Mahjoob K, Verbois SL, Morse D, Dagher R, Justice R and Pazdur R: Food and drug administration drug approval summary: Sunitinib malate for the treatment of gastrointestinal stromal tumor and advanced renal cell carcinoma. Oncologist. 12:107–113. 2007. View Article : Google Scholar : PubMed/NCBI

160 

Fukuhara H, Ino Y and Todo T: Oncolytic virus therapy: A new era of cancer treatment at dawn. Cancer Sci. 107:1373–1379. 2016. View Article : Google Scholar : PubMed/NCBI

161 

LaFargue CJ, Dal Molin GZ, Sood AK and Coleman RL: Exploring and comparing adverse events between PARP inhibitors. Lancet Oncol. 20:e15–e28. 2019. View Article : Google Scholar : PubMed/NCBI

162 

Arrillaga-Romany I, Gardner SL, Odia Y, Aguilera D, Allen JE, Batchelor T, Butowski N, Chen C, Cloughesy T, Cluster A, et al: ONC201 (Dordaviprone) in recurrent H3 K27M-Mutant diffuse midline glioma. J Clin Oncol. 42:1542–1552. 2024. View Article : Google Scholar : PubMed/NCBI

163 

Peters C, Paget M, Tshilenge KT, Saha D, Antoszczyk S, Baars A, Frost T, Martuza RL, Wakimoto H and Rabkin SD: Restriction of replication of oncolytic herpes simplex virus with a deletion of γ34.5 in glioblastoma stem-like cells. J Virol. 92:e00246–18. 2018. View Article : Google Scholar :

164 

Nakashima H, Nguyen T, Kasai K, Passaro C, Ito H, Goins WF, Shaikh I, Erdelyi R, Nishihara R, Nakano I, et al: Toxicity and efficacy of a novel GADD34-expressing oncolytic HSV-1 for the treatment of experimental glioblastoma. Clin Cancer Res. 24:2574–2584. 2018. View Article : Google Scholar : PubMed/NCBI

165 

Balathasan L, Tang VA, Yadollahi B, Brun J, Labelle M, Lefebvre C, Swift SL and Stojdl DF: Activating peripheral innate immunity enables safe and effective oncolytic virotherapy in the brain. Mol Ther Oncolytics. 7:45–56. 2017. View Article : Google Scholar : PubMed/NCBI

166 

Takata F, Nakagawa S, Matsumoto J and Dohgu S: Blood-Brain barrier dysfunction amplifies the development of neuroinflammation: Understanding of cellular events in brain microvascular endothelial cells for prevention and treatment of BBB Dysfunction. Front Cell Neurosci. 15:6618382021. View Article : Google Scholar : PubMed/NCBI

167 

Zhuang H, Shi S, Yuan Z and Chang JY: Bevacizumab treatment for radiation brain necrosis: mechanism, efficacy and issues. Mol Cancer. 18:212019. View Article : Google Scholar : PubMed/NCBI

168 

Seet RC, Rabinstein AA, Lindell PE, Uhm JH and Wijdicks EF: Cerebrovascular events after bevacizumab treatment: an early and severe complication. Neurocrit Care. 15:421–427. 2011. View Article : Google Scholar : PubMed/NCBI

169 

Zhou BX and Li Y: Significance of desmoglein-2 on cell malignant behaviors via mediating MAPK signaling in cervical cancer. Kaohsiung J Med Sci. 36:336–343. 2020. View Article : Google Scholar : PubMed/NCBI

170 

Sun R, Ma C, Wang W and Yang S: Upregulation of desmoglein 2 and its clinical value in lung adenocarcinoma: A comprehensive analysis by multiple bioinformatics methods. PeerJ. 8:e84202020. View Article : Google Scholar : PubMed/NCBI

171 

Han CP, Yu YH, Wang AG, Tian Y, Zhang HT, Zheng ZM and Liu YS: Desmoglein-2 overexpression predicts poor prognosis in hepatocellular carcinoma patients. Eur Rev Med Pharmacol Sci. 22:5481–5489. 2018.PubMed/NCBI

172 

Cai F, Zhu Q, Miao Y, Shen S, Su X and Shi Y: Desmoglein-2 is overexpressed in non-small cell lung cancer tissues and its knockdown suppresses NSCLC growth by regulation of p27 and CDK2. J Cancer Res Clin Oncol. 143:59–69. 2017. View Article : Google Scholar

173 

Tan LY, Mintoff C, Johan MZ, Ebert BW, Fedele C, Zhang YF, Szeto P, Sheppard KE, McArthur GA, Foster-Smith E, et al: Desmoglein 2 promotes vasculogenic mimicry in melanoma and is associated with poor clinical outcome. Oncotarget. 7:46492–46508. 2016. View Article : Google Scholar : PubMed/NCBI

174 

Kamekura R, Kolegraff KN, Nava P, Hilgarth RS, Feng M, Parkos CA and Nusrat A: Loss of the desmosomal cadherin desmoglein-2 suppresses colon cancer cell proliferation through EGFR signaling. Oncogene. 33:4531–4536. 2014. View Article : Google Scholar :

175 

Brennan D and Mahoney MG: Increased expression of Dsg2 in malignant skin carcinomas: A tissue-microarray based study. Cell Adh Migr. 3:148–154. 2009. View Article : Google Scholar : PubMed/NCBI

176 

Chen L, Liu X, Zhang J and Liu Y, Gao A, Xu Y, Lin Y, Du Q, Zhu Z, Hu Y and Liu Y: Characterization of desmoglein 2 expression in ovarian serous tumors and its prognostic significance in high-grade serous carcinoma. Int J Clin Exp Pathol. 11:4977–4986. 2018.PubMed/NCBI

177 

Ramani VC, Hennings L and Haun RS: Desmoglein 2 is a substrate of kallikrein 7 in pancreatic cancer. BMC Cancer. 8:3732008. View Article : Google Scholar : PubMed/NCBI

178 

Biedermann K, Vogelsang H, Becker I, Plaschke S, Siewert JR, Höfler H and Keller G: Desmoglein 2 is expressed abnormally rather than mutated in familial and sporadic gastric cancer. J Pathol. 207:199–206. 2005. View Article : Google Scholar : PubMed/NCBI

179 

Mundy RM, Baker AT, Bates EA, Cunliffe TG, Teijeira-Crespo A, Moses E, Rizkallah PJ and Parker AL: Broad sialic acid usage amongst species D human adenovirus. Npj Viruses. 1:12023. View Article : Google Scholar : PubMed/NCBI

180 

Rodrigues E and Macauley MS: Hypersialylation in cancer: Modulation of inflammation and therapeutic opportunities. Cancers (Basel). 10:2072018. View Article : Google Scholar : PubMed/NCBI

181 

Hu K, He S, Xiao J, Li M, Luo S, Zhang M and Hu Q: Interaction between herpesvirus entry mediator and HSV-2 glycoproteins mediates HIV-1 entry of HSV-2-infected epithelial cells. J Gen Virol. 98:2351–2361. 2017. View Article : Google Scholar : PubMed/NCBI

182 

Montgomery RI, Warner MS, Lum BJ and Spear PG: Herpes simplex virus-1 entry into cells mediated by a novel member of the TNF/NGF receptor family. Cell. 87:427–436. 1996. View Article : Google Scholar : PubMed/NCBI

183 

Ren S, Tian Q, Amar N, Yu H, Rivard CJ, Caldwell C, Ng TL, Tu M, Liu Y, Gao D, et al: The immune checkpoint, HVEM may contribute to immune escape in non-small cell lung cancer lacking PD-L1 expression. Lung Cancer. 125:115–120. 2018. View Article : Google Scholar : PubMed/NCBI

184 

Tsang JYS, Chan KW, Ni YB, Hlaing T, Hu J, Chan SK, Cheung SY and Tse GM: Expression and clinical significance of herpes virus entry mediator (HVEM) in breast cancer. Ann Surg Oncol. 24:4042–4050. 2017. View Article : Google Scholar : PubMed/NCBI

185 

Lan X, Li S, Gao H, Nanding A, Quan L, Yang C, Ding S and Xue Y: Increased BTLA and HVEM in gastric cancer are associated with progression and poor prognosis. Onco Targets Ther. 10:919–926. 2017. View Article : Google Scholar : PubMed/NCBI

186 

Migita K, Sho M, Shimada K, Yasuda S, Yamato I, Takayama T, Matsumoto S, Wakatsuki K, Hotta K, Tanaka T, et al: Significant involvement of herpesvirus entry mediator in human esophageal squamous cell carcinoma. Cancer. 120:808–817. 2014. View Article : Google Scholar

187 

Ahn AR, Noh SJ, Hussein UK, Park HS, Chung MJ, Lee H, Moon WS, Kang MJ, Kim HJ, Lee NR, et al: FAM83H and Nectin1 expression are related with survival and relapse of bladder urothelial carcinoma patients. BMC Urol. 21:1432021. View Article : Google Scholar : PubMed/NCBI

188 

Tampakis A, Tampaki EC, Nonni A, Droeser R, Posabella A, Tsourouflis G, Kontzoglou K, Patsouris E, von Flüe M and Kouraklis G: Nectin-1 expression in colorectal cancer: Is there a group of patients with high risk for early disease recurrence? Oncology. 96:318–325. 2019. View Article : Google Scholar : PubMed/NCBI

189 

Cocchi F, Menotti L, Dubreuil P, Lopez M and Campadelli-Fiume G: Cell-to-cell spread of wild-type herpes simplex virus type 1, but not of syncytial strains, is mediated by the immunoglobulin-like receptors that mediate virion entry, nectin1 (PRR1/HveC/HIgR) and nectin2 (PRR2/HveB). J Virol. 74:3909–3917. 2000. View Article : Google Scholar : PubMed/NCBI

190 

Girgis NM, Dehaven BC, Fan X, Viner KM, Shamim M and Isaacs SN: Cell surface expression of the vaccinia virus complement control protein is mediated by interaction with the viral A56 protein and protects infected cells from complement attack. J Virol. 82:4205–4214. 2008. View Article : Google Scholar : PubMed/NCBI

191 

Marques C, Reis CA, Vivès RR and Magalhães A: Heparan sulfate biosynthesis and sulfation profiles as modulators of cancer signalling and progression. Front Oncol. 11:7787522021. View Article : Google Scholar : PubMed/NCBI

192 

Li C, Luo P, Guo F, Jia X, Shen M, Zhang T, Wang S and Du T: Identification of HSPG2 as a bladder pro-tumor protein through NID1/AKT signaling. Cancer Cell Int. 24:3452024. View Article : Google Scholar : PubMed/NCBI

193 

Lambrecht V, Le Bourhis X, Toillon RA, Boilly B and Hondermarck H: Alterations in both heparan sulfate proteoglycans and mitogenic activity of fibroblast growth factor-2 are triggered by inhibitors of proliferation in normal and breast cancer epithelial cells. Exp Cell Res. 245:239–244. 1998. View Article : Google Scholar : PubMed/NCBI

194 

Yi B, Qiu Y, Ji W, Wei M, Liu C, Peng Z, Zhang Y, Quan Z, Tang Z and Su C: Desulfation of cell surface HSPG is an effective strategy for the treatment of gallbladder carcinoma. Cancer Lett. 381:349–358. 2016. View Article : Google Scholar : PubMed/NCBI

195 

Xiong R, Long Q, Zhang X, Xu J, Liu Y, Xiong L, Yang S, Feng G, Song G and Liu K: HOXD11 upregulates JAM-A and exerts oncogenic properties via NF-κB signaling pathway in esophageal squamous cell carcinoma. Hum Cell. 36:244–257. 2023. View Article : Google Scholar

196 

Aravamudhan P, Guzman-Cardozo C, Urbanek K, Welsh OL, Konopka-Anstadt JL, Sutherland DM and Dermody TS: The murine neuronal receptor NgR1 is dispensable for reovirus pathogenesis. J Virol. 96:e00055222022. View Article : Google Scholar : PubMed/NCBI

197 

Rosager AM, Sørensen MD, Dahlrot RH, Boldt HB, Hansen S, Lathia JD and Kristensen BW: Expression and prognostic value of JAM-A in gliomas. J Neurooncol. 135:107–117. 2017. View Article : Google Scholar : PubMed/NCBI

198 

Ikeo K, Oshima T, Shan J, Matsui H, Tomita T, Fukui H, Watari J and Miwa H: Junctional adhesion molecule-A promotes proliferation and inhibits apoptosis of gastric cancer. Hepatogastroenterology. 62:540–545. 2015.PubMed/NCBI

199 

McSherry EA, McGee SF, Jirstrom K, Doyle EM, Brennan DJ, Landberg G, Dervan PA, Hopkins AM and Gallagher WM: JAM-A expression positively correlates with poor prognosis in breast cancer patients. Int J Cancer. 125:1343–1351. 2009. View Article : Google Scholar : PubMed/NCBI

200 

Marcq I, Nyga R, Cartier F, Amrathlal RS, Ossart C, Ouled-Haddou H, Ghamlouch H, Galmiche A, Chatelain D, Lamotte L, et al: Identification of SLAMF3 (CD229) as an inhibitor of hepatocellular carcinoma cell proliferation and tumour progression. PLoS One. 8:e829182013. View Article : Google Scholar :

201 

Agresta L, Lehn M, Lampe K, Cantrell R, Hennies C, Szabo S, Wise-Draper T, Conforti L, Hoebe K and Janssen EM: CD244 represents a new therapeutic target in head and neck squamous cell carcinoma. J Immunother Cancer. 8:e0002452020. View Article : Google Scholar : PubMed/NCBI

202 

Zhang J, Zhu Y, Wang Q, Kong Y, Sheng H, Guo J, Xu J and Dai B: Poliovirus receptor CD155 is up-regulated in muscle-invasive bladder cancer and predicts poor prognosis. Urol Oncol. 38:41.e11–41.e18. 2020. View Article : Google Scholar

203 

Nishiwada S, Sho M, Yasuda S, Shimada K, Yamato I, Akahori T, Kinoshita S, Nagai M, Konishi N and Nakajima Y: Clinical significance of CD155 expression in human pancreatic cancer. Anticancer Res. 35:2287–2297. 2015.PubMed/NCBI

204 

Li YC, Zhou Q, Song QK, Wang RB, Lyu S, Guan X, Zhao YJ and Wu JP: Overexpression of an immune checkpoint (CD155) in breast cancer associated with prognostic significance and exhausted tumor-infiltrating lymphocytes: A cohort study. J Immunol Res. 2020:39489282020. View Article : Google Scholar : PubMed/NCBI

205 

Zhuo B, Li Y, Gu F, Li Z, Sun Q, Shi Y, Shen Y, Zhang F, Wang R and Wang X: Overexpression of CD155 relates to metastasis and invasion in osteosarcoma. Oncol Lett. 15:7312–7318. 2018.PubMed/NCBI

206 

Murakami D, Matsuda K, Iwamoto H, Mitani Y, Mizumoto Y, Nakamura Y, Matsuzaki I, Iwamoto R, Takahashi Y, Kojima F, et al: Prognostic value of CD155/TIGIT expression in patients with colorectal cancer. PLoS One. 17:e02659082022. View Article : Google Scholar : PubMed/NCBI

207 

Finkelshtein D, Werman A, Novick D, Barak S and Rubinstein M: LDL receptor and its family members serve as the cellular receptors for vesicular stomatitis virus. Proc Natl Acad Sci USA. 110:7306–7311. 2013. View Article : Google Scholar : PubMed/NCBI

208 

Behrouj H, Erfani M and Mokarram P: Examining the expression of low-density lipoprotein receptor (LDLR) and low-density lipoprotein receptor-related protein 6 (LRP6) genes in breast cancer cell lines. Mol Biol Res Commun. 13:85–88. 2024.PubMed/NCBI

209 

Zhang GM, Chen W, Yao Y, Luo L and Sun LJ: LDLR promotes growth and invasion in renal cell carcinoma and activates the EGFR pathway. Neoplasma. 69:113–122. 2022. View Article : Google Scholar

210 

Tang S, Chen K, Zheng F, Fu Z, Niu Y, Liu X, Ni H, Yuan X, Cui Z, Lu W, et al: High serum LDL promotes EMT and stemness through LDLR/FOXQ1/NF-κB1 pathway in epithelial ovarian cancer. Oncogene. 44:4587–4600. 2025. View Article : Google Scholar : PubMed/NCBI

211 

Örbom A, Evans-Axelsson S, Jansson B, Vilhelmsson Timmermand O, Tran TA, Bjartell A and Strand SE: Intratumoral distribution and pharmacokinetics of the radiolabeled ICAM-1 targeting monoclonal antibody R6.5 in a prostate cancer mouse model. Nuklearmedizin. 64:163–169. 2025. View Article : Google Scholar : PubMed/NCBI

212 

Gultekin O, Gonzalez-Molina J, Sarhan D, Groes-Kofoed N, Hassan MU, Lehti K and Salehi S: Systemic and tumor-specific inflammatory markers VCAM-1 and ICAM-1 as indicators of extent of surgery and oncologic outcome in advanced ovarian cancer. Transl Oncol. 59:1024622025. View Article : Google Scholar : PubMed/NCBI

213 

Chen YH, Chu CC, Liu JF, Lai HS and Chen YT: C-X-C motif ligand 1 induces cell migration by upregulating ICAM-1 expression by activating PI3K/Akt and NF-κB signaling pathway in liver cancer. Adv Biol (Weinh). 9:e24002952025. View Article : Google Scholar

214 

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

215 

Tempia-Caliera AA, Horvath LZ, Zimmermann A, Tihanyi TT, Korc M, Friess H and Büchler MW: Adhesion molecules in human pancreatic cancer. J Surg Oncol. 79:93–100. 2002. View Article : Google Scholar : PubMed/NCBI

216 

Wang Q, Yang Q, Liu C, Wang G, Song H, Shang G, Peng R, Qu X, Liu S, Cui Y, et al: Molecular basis of differential receptor usage for naturally occurring CD55-binding and -nonbinding coxsackievirus B3 strains. Proc Natl Acad Sci USA. 119:e21185901192022. View Article : Google Scholar : PubMed/NCBI

217 

Koretz K, Brüderlein S, Henne C and Möller P: Decay-accelerating factor (DAF, CD55) in normal colorectal mucosa, adenomas and carcinomas. Br J Cancer. 66:810–814. 1992. View Article : Google Scholar : PubMed/NCBI

218 

Madjd Z, Durrant LG, Bradley R, Spendlove I, Ellis IO and Pinder SE: Loss of CD55 is associated with aggressive breast tumors. Clin Cancer Res. 10:2797–2803. 2004. View Article : Google Scholar : PubMed/NCBI

219 

Mäenpää A, Junnikkala S, Hakulinen J, Timonen T and Meri S: Expression of complement membrane regulators membrane cofactor protein (CD46), decay accelerating factor (CD55), and protectin (CD59) in human malignant gliomas. Am J Pathol. 148:1139–1152. 1996.PubMed/NCBI

220 

Mustafa T, Klonisch T, Hombach-Klonisch S, Kehlen A, Schmutzler C, Koehrle J, Gimm O, Dralle H and Hoang-Vu C: Expression of CD97 and CD55 in human medullary thyroid carcinomas. Int J Oncol. 24:285–294. 2004.PubMed/NCBI

221 

Yamayoshi S, Iizuka S, Yamashita T, Minagawa H, Mizuta K, Okamoto M, Nishimura H, Sanjoh K, Katsushima N, Itagaki T, et al: Human SCARB2-dependent infection by coxsackievirus A7, A14, and A16 and enterovirus 71. J Virol. 86:5686–5696. 2012. View Article : Google Scholar : PubMed/NCBI

222 

Zhang D, Fang J, Shan J, Xu L, Wu Y, Lu B, Zhang X, Wang C, Sun P and Wang Q: SCARB2 associates with tumor-infiltrating neutrophils and predicts poor prognosis in breast cancer. Breast Cancer Res Treat. 207:15–24. 2024. View Article : Google Scholar : PubMed/NCBI

223 

Wang F, Gao Y, Xue S, Zhao L, Jiang H, Zhang T, Li Y, Zhao C, Wu F, Siqin T, et al: SCARB2 drives hepatocellular carcinoma tumor initiating cells via enhanced MYC transcriptional activity. Nat Commun. 14:59172023. View Article : Google Scholar : PubMed/NCBI

224 

Hance KW, Rogers CJ, Zaharoff DA, Canter D, Schlom J and Greiner JW: The antitumor and immunoadjuvant effects of IFN-alpha in combination with recombinant poxvirus vaccines. Clin Cancer Res. 15:2387–2396. 2009. View Article : Google Scholar : PubMed/NCBI

225 

Duggan MC, Jochems C, Donahue RN, Richards J, Karpa V, Foust E, Paul B, Brooks T, Tridandapani S, Olencki T, et al: A phase I study of recombinant (r) vaccinia-CEA(6D)-TRICOM and rFowlpox-CEA(6D)-TRICOM vaccines with GM-CSF and IFN-α-2b in patients with CEA-expressing carcinomas. Cancer Immunol Immunother. 65:1353–1364. 2016. View Article : Google Scholar : PubMed/NCBI

226 

Qi C, Liu C, Peng Z, Zhang Y, Wei J, Qiu W, Zhang X, Pan H, Niu Z, Qiu M, et al: Claudin-18 isoform 2-specific CAR T-cell therapy (satri-cel) versus treatment of physician's choice for previously treated advanced gastric or gastro-oesophageal junction cancer (CT041-ST-01): A randomised, open-label, phase 2 trial. Lancet. 405:2049–2060. 2025. View Article : Google Scholar : PubMed/NCBI

227 

Liu S, Li F, Deng L, Ma Q, Lu W, Zhao Z, Liu H, Zhou Y, Hu M, Wang H, et al: Claudin18.2 bispecific T cell engager armed oncolytic virus enhances antitumor effects against pancreatic cancer. Mol Ther Oncolytics. 30:275–285. 2023. View Article : Google Scholar : PubMed/NCBI

228 

Yoon AR, Hong J, Kim M and Yun CO: Hepatocellular carcinoma-targeting oncolytic adenovirus overcomes hypoxic tumor microenvironment and effectively disperses through both central and peripheral tumor regions. Sci Rep. 8:22332018. View Article : Google Scholar : PubMed/NCBI

229 

Cordeiro R, Oliveira D, Santo D, Coelho J and Faneca H: Mesoporous silica-glycopolymer hybrid nanoparticles for dual targeted chemotherapy and gene therapy to liver cancer cells. Int J Pharm. 675:1255532025. View Article : Google Scholar : PubMed/NCBI

230 

Deng Y, Yang B, Yang Z, Xiao H, Zou Y, Zou C, Yang S, Sun X, Wang Y, Bai J, et al: Engineered E. coli OMVs carrying the membrane-binding hGC33 fragment precisely target liver cancer and effectively treat tumor. Int J Nanomedicine. 20:6573–6590. 2025. View Article : Google Scholar : PubMed/NCBI

231 

West CE, Mirshahi UL, Ruth KS, Sharp LN, Arni AM, Turnbull C, Wright CF, Vaidya B, Owens MM, Carey DJ and Patel KA: Medullary thyroid cancer risk and mortality in carriers of incidentally identified MEN2A RET variants. JAMA Netw Open. 8:e25179372025. View Article : Google Scholar : PubMed/NCBI

232 

Cañizo CG, Guerrero-Ramos F, Perez Escavy M, Lodewijk I, Suárez-Cabrera C, Morales L, Nunes SP, Munera-Maravilla E, Rubio C, Sánchez R, et al: Characterisation of the tumour microenvironment and PD-L1 granularity reveals the prognostic value of cancer-associated myofibroblasts in non-invasive bladder cancer. Oncoimmunology. 14:24382912025. View Article : Google Scholar

233 

Okato A, Utsumi T, Ranieri M, Zheng X, Zhou M, Pereira LD, Chen T, Kita Y, Wu D, Hyun H, et al: FGFR inhibition augments anti-PD-1 efficacy in murine FGFR3-mutant bladder cancer by abrogating immunosuppression. J Clin Invest. 134:e1692412024. View Article : Google Scholar : PubMed/NCBI

234 

Cheng L, Lopez-Beltran A, Massari F, MacLennan GT and Montironi R: Molecular testing for BRAF mutations to inform melanoma treatment decisions: A move toward precision medicine. Mod Pathol. 31:24–38. 2018. View Article : Google Scholar :

235 

Rosenkrans ZT, Erbe AK, Clemons NB, Feils AS, Medina-Guevara Y, Jeffery JJ, Barnhart TE, Engle JW, Sondel PM and Hernandez R: ImmunoPET demonstrates that Co-Targeting GD2 and B7-H3 with bispecific antibodies enhances tumor selectivity in preclinical melanoma models. Bioconjug Chem. 36:1595–1603. 2025. View Article : Google Scholar : PubMed/NCBI

236 

Krug C, Birkholz K, Paulus A, Schwenkert M, Schmidt P, Hoffmann N, Hombach A, Fey G, Abken H, Schuler G, et al: Stability and activity of MCSP-specific chimeric antigen receptors (CARs) depend on the scFv antigen-binding domain and the protein backbone. Cancer Immunol Immunother. 64:1623–1635. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Bi S, Shan T, Tang Y and Wang Q: Advancements on the synergistic application of oncolytic viruses and molecularly targeted therapies for the treatment of solid tumors (Review). Int J Oncol 68: 25, 2026.
APA
Bi, S., Shan, T., Tang, Y., & Wang, Q. (2026). Advancements on the synergistic application of oncolytic viruses and molecularly targeted therapies for the treatment of solid tumors (Review). International Journal of Oncology, 68, 25. https://doi.org/10.3892/ijo.2025.5838
MLA
Bi, S., Shan, T., Tang, Y., Wang, Q."Advancements on the synergistic application of oncolytic viruses and molecularly targeted therapies for the treatment of solid tumors (Review)". International Journal of Oncology 68.2 (2026): 25.
Chicago
Bi, S., Shan, T., Tang, Y., Wang, Q."Advancements on the synergistic application of oncolytic viruses and molecularly targeted therapies for the treatment of solid tumors (Review)". International Journal of Oncology 68, no. 2 (2026): 25. https://doi.org/10.3892/ijo.2025.5838
Copy and paste a formatted citation
x
Spandidos Publications style
Bi S, Shan T, Tang Y and Wang Q: Advancements on the synergistic application of oncolytic viruses and molecularly targeted therapies for the treatment of solid tumors (Review). Int J Oncol 68: 25, 2026.
APA
Bi, S., Shan, T., Tang, Y., & Wang, Q. (2026). Advancements on the synergistic application of oncolytic viruses and molecularly targeted therapies for the treatment of solid tumors (Review). International Journal of Oncology, 68, 25. https://doi.org/10.3892/ijo.2025.5838
MLA
Bi, S., Shan, T., Tang, Y., Wang, Q."Advancements on the synergistic application of oncolytic viruses and molecularly targeted therapies for the treatment of solid tumors (Review)". International Journal of Oncology 68.2 (2026): 25.
Chicago
Bi, S., Shan, T., Tang, Y., Wang, Q."Advancements on the synergistic application of oncolytic viruses and molecularly targeted therapies for the treatment of solid tumors (Review)". International Journal of Oncology 68, no. 2 (2026): 25. https://doi.org/10.3892/ijo.2025.5838
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team