Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Jul 15, 2024; 16(7): 2867-2876
Published online Jul 15, 2024. doi: 10.4251/wjgo.v16.i7.2867
Oncolytic virotherapy for hepatocellular carcinoma: A potent immunotherapeutic landscape
Rong Xiao, Hao Jin, Biao Huang, Yi-Gang Wang, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
Fang Huang, Department of Pathology, Laboratory Medicine Center, Zhejiang Provincial Peoples’ Hospital, Peoples’ Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China
Hui Wang, Department of Oncology, Zhejiang Xiaoshan Hospital, Hangzhou 310018, Zhejiang Province, China
ORCID number: Fang Huang (0000-0002-3115-8703); Biao Huang (0000-0003-1883-0670); Yi-Gang Wang (0000-0003-4546-8179).
Co-corresponding authors: Hui Wang and Yi-Gang Wang.
Author contributions: Xiao R, Jin H, and Huang F performed the literature, and drafted the manuscript; Huang B, Wang H, and Wang YG revised the manuscript; and all the authors have read and approved the final version to be published.
Supported by the Public Welfare Technology Project of Zhejiang Province, No. LGF21H160033; Zhejiang Medical Technology Plan Project, No. 2021KY047; and Hangzhou Medical Health Science and Technology Project, No. B20220173.
Conflict-of-interest statement: The authors declare that there is no conflict of interest related to this study.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Yi-Gang Wang, PhD, Professor, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, No. 2 Street of Xiasha District, Hangzhou 310018, Zhejiang Province, China. wangyigang43@163.com
Received: December 28, 2023
Revised: April 24, 2024
Accepted: May 13, 2024
Published online: July 15, 2024
Processing time: 197 Days and 7.3 Hours

Abstract

Hepatocellular carcinoma (HCC) is a systemic disease with augmented malignant degree, high mortality and poor prognosis. Since the establishment of the immune mechanism of tumor therapy, people have realized that immunotherapy is an effective means for improvement of HCC patient prognosis. Oncolytic virus is a novel immunotherapy drug, which kills tumor cells and exempts normal cells by directly lysing tumor and inducing anti-tumor immune response, and it has been extensively examined as an HCC therapy. This editorial discusses oncolytic viruses for the treatment of HCC, emphasizing viral immunotherapy strategies and clinical applications related to HCC.

Key Words: Oncolytic virus; Immunotherapeutic; Hepatocellular carcinoma; Virotherapy; Editorial

Core Tip: Since the discovery of oncolytic virus, various preclinical studies and clinical trials have been carried out on the application of virus therapy in hepatocellular carcinoma (HCC), and people have constantly tried to optimize the efficacy of this new drug. Based on this background, this editorial introduces oncolytic viruses for the treatment of HCC, including immunotherapy strategies, and looks forward to more good clinical results.



INTRODUCTION

Hepatocellular carcinoma (HCC) is a frequently occurring malignancy and a major contributor to the global cancer-associated deaths. Environmental and genetic hazard agents strongly modulate HCC development. Among the most prominent and preventable hazard agents are oncogenic viral infection with hepatitis B virus or hepatitis C virus (HCV), alcohol abuse, obesity and diabetes[1]. With rising HCC incidences year by year, treatment options have also been improved, common therapies are surgical resection, liver transplantation, thermal ablation, intra-arterial therapy, radiotherapy, etc. Systemic therapies such as Sorafenib and Lenvatinib should be considered when the HCC is at an advanced stage and cannot be treated locally[2]. Despite the variety of treatment options for HCC, current treatments still do not extend overall survival for advanced HCC.

In recent ten years, with the deepening of cancer immunotherapy research, immunotherapy has become a hot spot for treating HCC. Immunotherapies are typically checkpoint inhibitors, adoptive cell therapy, and viral therapy[3]. Among them, viral therapy encompasses non-replicating viral delivery as delivery vectors and replicating oncolytic viruses. Oncolytic viruses belong to a large virus family that can directly kill tumor cells, and have been widely concerned owing to their strong oncolytic activity, good targeting and reduced drug resistance[4]. Nowadays, several oncolytic viruses are employed in cancer research, including adeno-, vaccinia-, reo-, vesicular stomatitis-, herpes simplex virus (HSV), and so on. The oncolytic mechanism of oncolytic virus mainly includes: (1) Direct oncolytic effect. The imbalance of specific proteins such as PTEN, TP53, RAS, and RB1 in tumor cells makes it preferentially infected by oncolytic virus[5]. The virus replicates itself with the help of host intracellular resources. When the amount of intracellular virus reaches a certain level, the virus lyses cells and releases excess progeny viruses to continue further infection of other tumor cells, therefore achieving oncolytic effect; (2) stimulate anti-tumor immune response, which is the most important part of anti-tumor effect[6]. Due to its own variability, tumor cells do not respond to the immune system, that is, forming cold tumors. However, restoring the tumor cells-induced immune response, and conversion of cold to hot tumors, is also a big challenge. As viral lysis of tumor cells (Figure 1), secretion of soluble tumor-related antigens, cell-derived damage associated molecular patterns, and viral pathogen associated molecular patterns are identified and sequestered by antigen-presenting cells, and both innate and adaptive immunity are activated, thus playing a proximal or distal anti-tumor role; (3) through genetic engineering, the virus can express some foreign genes, such as some tumor suppressor proteins or functional RNA. The insertion of foreign genes is accompanied by the deletion of some non-essential genes of the virus, which can give the virus higher safety and oncolytic effect. As the first oncolytic virus (OV), talimogene laherparepvec (T-Vec) received approval from the United States Food and Drug Administration, loss of ICP47, which encodes antigen-presenting inhibitors, facilitating entry of tumor-associated antigens and viral antigens into the major histocompatibility complex (MHC) Class I complex and promotes an immune response against tumor cells. Simultaneously, the granulocyte-macrophage colony-stimulating factor (GM-CSF) content accelerated dendritic cell maturation and enhanced their tumor cells-induced immune response[7]; and (4) OV can also generate neutrophil aggregations within blood vessels via its action on stromal cells in the tumor environment. These neutrophils promote fibrin accumulation and secrete clotting agents, thus ultimately impairing the tumor vasculature. Generally, the anti-tumor mechanisms of OV are diverse, and more and more preclinical studies have shown that oncolytic viruses have significant anti-tumor effects, and the clinical treatment results of OV are also satisfactory and encouraging.

Figure 1
Figure 1 Oncolytic virus promotes both regional and systemic anti-tumor activity. Following viral infection of tumor cells, the virus replicates proficiently within the cell, and after lysis of tumor cells, the soluble tumor-associated antigens, viral pathogen-associated molecular models, and cell-derived damage-associated molecular models release recruits and activates antigen-presenting cells (APCs). APC then migrates to the lymph nodes and initiates adaptive T cell immunity to the tumor. These cytotoxic T cells identify and destroy proximal or distal tumor cells. CTL: Cytotoxic T cell; APC: Antigen-presenting cell; DAMP: Derived damage-associated molecular model; PAMP: Pathogen-associated molecular models.
ONCOLYTIC VIRUS THERAPY FOR HCC

Oncolytic viral treatment is highly efficacious in eradicating tumors. Oncolytic viruses used for treatment can be mainly divided into ssRNA and dsDNA viruses. dsDNA viruses are typically adenovirus, vaccinia virus (VV), herpes virus, and so on. ssRNA viruses are either just viruses (Coxsackie, Seneca Valley, or poliovirus) or negative viruses [measles, Newcastle disease, or vesicular stomatitis virus (VSV)[8]. DNA viruses possess extensive genome which facilitate genetic engineering operations, their life cycle and molecular biology are well understood, and most clinical studies use DNA viruses because they are relatively stable. RNA viruses also have the unique advantages of strong oncolytic effect and weak pathogenicity. Tumor selection mechanisms of oncolytic viruses vary, the oncolytic viral HCC treatment is also varied depending on the viral characteristics.

Adenovirus is an icosahedral, unenveloped double-stranded DNA virus with about 34-36 KB genome size[9]. Adenovirus type 5 proliferation ability, within the host genome copy number is high, the virus particles have good stability, can produce high drop degree of virus particles in order to meet the experimental requirements, they can be used for gene therapy. The genome structure and life cycle of adenoviruses have long been well understood, and molecular modification of adenoviruses is relatively easy. Adenovirus replication-related genes are mainly E1-E4 genes. Certain gene deletions can enhance safety and oncolytic performance of adenovirus. Such as delete defeat of E1A-CR2 in tumor cell specificity for Ad replication[10]. In addition, replacing the E1A promoter of the virus with the liver cancer tissue-specific promoter alpha-fetoprotein may also enhance the safety of the virus. ZD55 is a novel oncolytic adenovirus vector with the E1B-55K deletion constructed by our group, which can express more foreign inserted genes than Ad. We constructed a recombinant ZD55-IL-24 adenovirus and successful IL-24 expression enhanced the oncolytic effect of the virus (about 100 times higher than that of ONYX-015), in addition, ZD55-IL-24 did not have significant cytotoxic and apoptotic effects on normal cells. Meanwhile, the ZD55-IL24 and cisplatin co-therapy increased tumor cell death[11]. We also employed this vector to deploy one fused gene (ZD55-TRAIL-IETD-Smac) into HCC cells. The ZD55-TRAIL-IETD-Smac combination therapy facilitated quick and effective apoptotic tumor cell activation and total elimination of tumor xenografts in all exposed animal models, its effect of completely eliminating xenograft liver cancer was matched with ZD55-TRAIL + ZD55-Smac which could be an effective treatment strategy for HCC[12]. Tumor suppressor lung cancer 1 (TSLC1) is frequently down-regulated in multiple cancer forms, such as, HCC, and we found that TSLC1 inhibits tumor growth by affecting the Wnt signaling pathway[13]. Further, we designed a dual-regulated oncolytic adenovirus Ad-wnt-E1A(△24bp)-TSLC1, which modulates the Wnt and Rb axes and harbors the tumor suppressor gene TSLC1. The Ad-wnt-E1A(△24bp)-TSLC1 can induce tumor cell autophagy death. In vivo investigations revealed that this recombinant adenovirus effectively induces hepatic cancer stem-like cell apoptosis, inhibits tumor metastasis and growth, and prolongs mice survivability. Hence, this new oncolytic adenovirus has a promising prospect in the treatment of liver cancer[14].

VV is a member of the poxviridae family and is a linear double-stranded DNA virus with 190kb genome size encoding over 200 genes[15]. VV has its own unique biological characteristics: The replication time is shorter, and the first generation of the virus is released 8 h after VV infects cells, and 48-72 h after the infected cells are lysed[16]. Genetically safe and stable, unlike other DNA viruses, VV always exists in the cytoplasm without entry into the nucleus from the beginning of its entry into the cell. Therefore, the viral DNA does not insert itself into the host genome and does not induce host genetic changes. The unique infection mode of VV determines that it can infect almost all cell lines, unlike adenovirus, and it does not need to enter the target cells through specific cell surface receptors, but infects the target cells through multiple membrane fusion pathways. In addition, compared with somatic cells in the non-dividing/resting phase, VV is more likely to infect cancer cells with active cell cycles. Even without the recognition of specific receptors on tumor cell surfaces, VV still has natural targeting to cancer cells. VV virus particles are stable, which is convenient for storage, transportation and clinical experiments[17]. However, the large genome of VV also brings risks to treatment, and a large number of unclear genes bring certain risks to treatment[18]. At present, VV is mainly modified around targeting and efficacy. For example, by missing thymidine kinase (TK), VV is highly targeted to tumor cells. Because VV replication requires a high concentration of nucleotides, but normal cell proliferation requires TK, so the nucleotide concentration is usually low. However, tumor cells do not rely on TK for cell proliferation, so they have a higher concentration of nucleotide pools, which is conducive to VV replication. Therefore, oncolytic VV lacking TK tends to replicate in tumor cells[19,20]. The specificity of TK-deficient VV in tumor cells was reported in animal models of colon cancer, melanoma, sarcoma, and liver cancer[21]. Zhou et al[22] have evaluated the effectiveness of VV carrying Marine lectins. This study was to assess the cytotoxic effects of oncoVV harboring tachypleus tridentatus lectin (oncoVV-TTL), aphrocallistes vastus lectin (oncoVV-AVL), white-spotted charr lectin (oncoVV-WCL), and asterina pectinifera lectin (oncoVV-APL) on HCC, the results show that oncoVV-AVL has the strongest anti-tumor effect, which may promote self-replication through various pathways including MAPK, Hippo, PI3K, lipid metabolism, and androgen pathway.

HSV is a coated double-stranded linear DNA virus carrying a sizable (152 kd) genome. HSV contains two serotypes, HSV-1 and HSV-2, among which type 1 is widely examined[23]. HSV as an oncolytic virus has the following advantages: The genome is large; it can introduce large fragments of genes or multiple foreign genes; the virus replication ability is strong; the host gene mutation is not easy to occur; and the host range is wide. However, the strong HSV-1 virulence and immunogenicity itself has a “double-edged sword” consequence, and its prompt immune-regulated viral clearance and possible cytotoxicity can cause delivery and safety issues, which increase the difficulty of administration, and the accumulation of muscular injection in the liver. Therefore, most therapeutic administration regimens are intratumoral injection[24]. Nakatake et al[25] used third-generation oncolytic HSV type 1 (HSV-1) T-01 to examine antitumor activities of HCCs. The results showed that T-01 was toxic to 13 of 14 cell lines (in vitro), the in vivo efficacy of T-01 was studied using human cells HuH-7, KYN-2, PLC/PRF/5, HepG2, and mouse cells Hepa1-6. T-01 inhibited human hepatoma- and hepatoblastoma cell line-mediated tumor growth. At present, several HSV mutants have been successfully created to adapt to different therapeutic strategies. For example, G207, reportedly the first HSV examined in clinical trials (CTs), replicates specifically in cancer, but not normal cells and neurons[26]. In preclinical studies, tumor model G207 induced systemic antitumor immune response[27] in a G207 treatment children with high grade glioma[28]. Herein, twelve high-grade glioma patients aged 7-18 years were treated. Median overall survival was 12.2 months (95% confidence interval: 8.0-16.4). As of June 5, 2020, 4 patients survived 18 months following G207 treatment. Evidence showed that during G207 treatment, “cold” to “hot” tumor transformation significantly increased the tumor-infiltrating lymphocyte quantity, (ClinicalTrials.gov number: NCT02457845) it is hoped that future research will be carried out in HCC therapy.

VSV is a coated negative strand RNA rhabdovirus[29]. Types of infection, almost can infect all types of cells, including normal, healthy cells, but owing to the form of interferon antiviral responses in healthy cells, the types of even after the infection of normal cells cannot copy within the cell[30]. However, interferon (IFN) signaling within tumor cells is often defective, so VSV is able to infect and selectively destroy tumor cells[31]. The G protein of VSV initiates the viral infection process by binding to cell surface receptors, and the M protein causes tumor cell apoptosis by inhibiting the host antiviral response[32]. Shinozaki et al[33] found that the recombinant VSV vector harboring B-galactosidase (VSV-b-gal) was injected through arteries into rats with orthotopic transplantation of multifocal HCC, resulting in extensive tumor lysis in hepatic multifocal HCC. This suggests that oncolytic VSV is a potential efficacious and safe therapy for multifocal HCC patients in the near future. Through hepatic artery drug types, within the scope of the maximum tolerated dose (MTD), selective replication can enter HCC lesions, leading to a large number of tumor necrosis, but to give the medicine more than MTD, experimental animals cause nerve toxicity and acute liver toxicity. When the MTD of the host to VSV is increased, the dosage of VSV can be increased to obtain better curative effect. Shinozaki et al[34] verified this hypothesis by using exogenous IFN in preventive treatment of rats, which improved the VSV MTD in rats, and the IFN presence had no effect on the replication ability of VSV. No damage to the curative effect of HCC, which in order to solve the limitations of clinical dosage provides a new train of thought.

Reovirus (REO) is a double-stranded RNA virus and benign human pathogen that discreetly replicates and lysis cells in cells with abnormal Ras pathways, which are associated with most cancers[35]. Wild type REO to tumor cells have natural targeting[36], and HCV is a critical contributor to viral liver cancer, and evidences revealed that REO-induced cytokine response effectively inhibits HCV replication in vitro and in vivo during HCV-HCC therapy, while showing good efficacy in HCC preclinical models by activating innate degranulated immune cells[37]. Therefore, REO is a potential alternative to complement and support the available HCV-HCC treatment.

Influenza virus (IV) is an enveloped negative strand RNA virus, which is known to cause influenza. It consists of four genera, A, B, C, and D, with type A being the most widely examined[38]. Because of its lack of reverse transcriptase and genome integration activity, it is an ideal carrier for oncolytic therapy[39]. However the health problems caused by the IV need to be special attention, therefore, which the IV is used as a soluble tumor agent needs to enhance its anti-cancer properties, and avoid the infectious pathogenic, and lack of nonstructural protein1 (delNS1) is a more attractive strategy. García-Sastre et al[40] demonstrated that influenza A virus delNS1 could not replicate in mice with normal IFN, nonetheless, it was lethal in mice with abnormal IFN signaling pathways. Due to many cancer IFN signaling pathway is not active, the lack of NS1 protein makes influenza A virus to malignant tissue targeting property[41]. A study successfully constructed an influenza A virus (delNS1-GM-CSF) expressing GM-CSF with partial deletion of NS1, and in vitro experiments found that the virus had selective cytotoxicity in multiple HCC cell lines, whereas no alteration was evident in normal liver cell line LO2. In a xenotransplantation model of human Hep-G2 liver cancer cell line, delNS1-GM-CSF significantly suppressed tumor development in a dose-reliant fashion after intratumoral injection, and the tumor inhibitory effect of delNS1-GM-CSF was observed in HCC clinical samples. These findings suggest that delNS1-GM-CSF is a highly efficacious oncolytic agent against HCC.

Newcastle disease virus (NDV) is a coated negative strand RNA virus that has a natural host in birds and they gain entry to cells via association with sialic acid residues that reside on human and rodent cancer cells[43]. The natural sensitivity of NDV to IFN, combined with defects in tumor cells’ own IFN signaling pathway, makes NDV an ideal candidate for OV therapy[44]. However, compared to other Ovs, NDV virulence and pathogenicity were in the human body is lower, thus NDV in soluble tumor need to significantly improve performance. NDV enters cells by membrane fusion, a process in which fusion protein F is required[45]. Altomonte et al[46] demonstrated a new NDV vector that contains a mutation in the F gene leucine at amino acid 289 (L289A). Relative to the rNDV/F3aa control virus, recombinant vector rNDV/F3aa (L289A) enhances HCC fusion and cytotoxicity in vitro.

OVS COMBINED WITH IMMUNOTHERAPY

HCC is considered highly immune tolerance of the tumor, abnormal T cell activation, immune checkpoints to rise, immunosuppressive factors produced, immunosuppressive cells clustering cause of HCC was established highly immunosuppressive tumor microenvironment, the antitumor immune response paralysis[47]. Although researchers have made some advances in immunotherapy in recent years, such as the use of drugs such as the immune checkpoint inhibitor Immune checkpoint inhibitors, tumors have evolved immunoediting and thus have a greater ability to escape immune, so the treatment of HCC patients with these drugs remains limited. Emerging evidences suggest a strong role of the immune system in oncolytic virus therapy, and the use of oncolytic virus can reactivate the immune system and establish immune control against tumors.

A more attractive strategy is to add different immunomodulatory transgenes to the virus. For example, cytokines are proteins that regulate the immune response, and the VV performance is improved by carrying certain immune-stimulating molecules through VV. Arming VVS with cytokines namely GM-CSF, IL-2, IL-6, IL-12, IL-15, IL-23, and IL-24 exhibited superior anti-cancer activities and significant safety profiles in clinical and preclinical examinations[48,49]. For example, IL-24 suppresses tumor development, invasion, angiogenesis and metastasis, Promote cell apoptosis[50]. Deng et al[51] generated a VV Light 9 (VG9-IL24) containing IL-24 gene, and the results showed that VG9-IL24 had a strong ability to infect HCC cell lines in vitro, and could not infect normal cells. After VG9-IL24 infected cells, it significantly upregulated IL-24 content and promoted cell apoptosis. In animal models, tumor development was drastically inhibited with prolonged survival duration among mice treated with VG9-IL24. By combining oncolytic adenovirus with TIM-3 immune checkpoint molecular specific antibody (α-TIM-3), we also successfully constructed a novel adenovirus Ad-GD55-α-Tim-3, which can induce α-TIM-3 expression in hepatoma cells after infection, and augment anti-tumor efficiency and local immune response of oncolytic virus. Ad-GD55-α-TIM-3 is involved in a remarkably potent regional immune response in a simulated tumor immune microenvironment. Ad-GD55-α-TIM-3 oncolytic adenovirus is an HCC therapeutic candidate[52]. Zhang et al[53] successfully constructed an adenovirus Ad5-PC harboring bi-specific protein, one end of which expresses the PD-1 extracellular domain and the other end contains the CD137L extracellular domain (PD1/CD137L). Ad5-PC stimulates high level of interferon gamma production, continuously activates CD8+ T cells, and promotes immune cell infiltration. Induce anti-tumor immune response. Ad5PC significantly improved anti-tumor function in both ascites and subcutaneous HCC tumor models, with long-term cure rates of 70% and 60%, respectively. In addition, human IFN-b gene is specifically present within tumor cells and inhibit the growth of tumor cells, which is also a new tumor therapeutic target[54]. And, the double specificity and three specific antibodies in immune therapy also showed great prospect, a frontier study show that, the researchers used BITE forces soluble tumor adenovirus, This BITE interacts with the fibroblast activating protein on cancer-associated fibroblasts (CAF) and CD3e on T cells, effectively activating T cells while also inducing fibroblast death. Armed OV resulted in CAF-related immunosuppressive factor depletion, pro-inflammatory cytokine elevation, and upregulation of the antigen presentation, t cell function, and transport marker expressions, successfully killing tumor cells and fibroblasts. Clinical studies are expected[55]. The relatively new chimeric antigen receptor T (CAR-T) immunotherapy is reported to have excellent efficiency in hematological tumors, however, it does not efficaciously treat solid tumors[56]. Several investigations revealed that OV can be used as a complementary agent of CAR-T in solid tumors, helping CAR-T survival and accumulation, significantly improving the efficacy. This may be the future direction of the OV treatment for HCC[57].

Excitingly, immunomodulatory drugs could also act as perfect companions for oncolytic viruses. It is difficult for the immune system stimulated by oncolytic virus to obtain sustained anti-tumor response, and the existence of antiviral immune response is the main reason[58]. Once the OV in the host is cleared by the immune response, the tumor microenvironment with immunosuppressive response will be restored, and the tumor is easy to relapse. The up-regulated immune checkpoint in the tumor is an important reason for the generation of immunosuppressive tumor microenvironment. ICIs can inhibit the immunosuppressive signaling pathway, and the combination therapy with OV will be a promising anti-tumor strategy. OVH-aMPD-1, for example, is a kind of HSV targeting the PD-1/PD-L1 axis. Under interaction with anti-TIGIT antibodies, the percentage of immune-specific cells in tumor cells is significantly increased. Combined therapy blocks immune checkpoints and enhances tumor-specific immune response of subcutaneous implanted tumors, highlighting the benefits of collaborative therapy[59]. Pexastimogene devacirevec (Pexa-Vec, otherwise called JX-594) is a VV, and several CTs have examined its role in treating liver cancer. A phase I/II CT a JX-594 joint worth of sheet resistance as a first-line treatment for advanced HCC patients (ClinicalTrials.org: NCT03071094) was performed. Subjects received injections of JX-594 three times every two weeks and OPDIVO intravenously every two weeks (starting at week 2). The overall response rate for 12 cancer patients was 33.3% (9.9% to 65.1%). However, the severe adverse events risk in stage I and Stage II a was 85.71% and 80.00%, respectively, so the prospect of synergistic therapy should also consider how to overcome cytotoxicity problems.

CTS OF OVS FOR HCC

So far, we have searched for OV CTs on HCC at clinicaltrials.org (Table 1). Some experimental results have been encouraging. A Phase I CT (NCT00844623) evaluated the feasibility and safety of intratumoral first-generation HSV-TK gene-encoding adenovirus vector (Ad.TK) administration, with subsequent systemic ganciclovir therapy among advanced HCC patients. The results showed no hepatotoxicity, even in patients with cirrhosis. Fever, influenza-like syndrome, administration location pain, and pancytopenia were the most prevalent complications. There were no partial responses, and 60% participants exhibited stable tumors with injectable lesions. Importantly, the 2 patients given the highest dose exhibited some necrosis within the tumor on imaging. Among them, 1 attained sustained stability and was alive for 26 months[60]. CTs confirmed the good efficiency and safety profile of therapy.

Table 1 Clinical trials of oncolytic virus for hepatocellular carcinoma.
Trial number
Virus
Disease
Status
Interventions
Trail phase
Study start
NCT00844623TK99UNHCCCompletedAloneI2002
NCT00003147Ad5CMV-p53 geneLiver cancerCompletedAloneI1998
NCT02509169rAd-p53HCCUnknown statusAloneII2014
NCT00300521ADV-TKAdvanced HCCCompletedAloneII2000
NCT02418988rAd-p53Advanced HCCUnknown statusTACEII2014
NCT01869088H101HCCUnknown statusTACEIII2013
NCT03790059H101HCCUnknown statusRFANot applicable2016
NCT03313596ADV-TkAdvanced HCCUnknown statusLTIII2013
NCT05113290H101HCCUnknown statusSorafenibIV2021
NCT00629759JX-594Primary hepatocellular carcinomaCompletedAloneI2006
NCT00554372JX-594Primary hepatocellular carcinomaCompletedAloneII2008
NCT02562755JX-594HCCCompletedSorafenibIII2015
NCT01387555JX-594HCC failed sorafenib treatmentCompletedAloneII2008
NCT02509507T-VecHCC et alActive, not recruitingPembrolizumabI/II2016
NCT04313868GEN2Metastatic HCCRecruitingAloneI2014
NCT05223816VG161HCC et alRecruitingNivolumabII2023
NCT01628640VSV (IFN-β)HCCActive, not recruitingAloneI2012
NCT04665362M1 virusAdvanced/metastatic hepatocellular carcinomaUnknown statusAnti PD-1 antibody and apatinibI2021

JX-594 has good antitumor and immunotherapeutic effects, and is well tolerated, and is widely used in liver cancer. In a Phase I CT, 14 patients with refractory primary or metastatic HCC underwent JX-594 intratumoral shot in 3 cases of partial remission and 6 of stable disease[16]. Phase II CT to study the drug dose for the effects of the treatment of cancer of the liver (ClinicalTrials.org: NCT00554372) in 30 patients with advanced liver cancer, Survival (median survival 14.1 months) was substantially elevated among patients receiving the higher vs lower dose (6.7 months)[17]. It was primarily JX-594 that induced anti-tumor immunity in patients, and in addition, its anti-angiogenesis also resulted in improved survival.

OV from natural virus to recombinant virus, from single therapy to combination therapy, this is the clinical development trend, when patients tolerate drug therapy, the addition of OV can reverse this adverse situation, in the future, there could be more CTs of OV, which can enhance knowledge of OVT, far-reaching significance for subsequent treatment. A variety of treatment modalities will also benefit HCC patients.

CONCLUSION

Viruses are commonly used in cancer therapy for over a century, and with the understanding of the mechanism of virus action, coupled with the establishment of genetic engineering technology, OVTs have gradually become a mature means of cancer treatment from natural strains to recombinant viruses, the representative OVs discussed for the treatment of hepatocellular carcinoma are shown in Table 2. More and more studies have shown that OVs is also a powerful weapon of immunotherapy, OVs strongly dissolves tumor cells, and utilizes intricate modulatory networks namely immune system activation. However, the efficacy of single therapy may be limited, and the clinical research of OV therapy for HCC has gradually shifted from single therapy to OV combination therapy.

Table 2 Representative oncolytic virus for hepatocellular carcinoma.
Virus name
Mode of actions/target
Virus Modification
Potential implications
Ref.
ZD55-IL-24Combine cisplatin in vitro/Bel-7404, SMMC-7721IL-24 gene, E1B 55 kDa deletionEnhance selective cytotoxicity[11]
ZD55-TRAIL-IETD-SmacIntratumoral injection/Bel-7404 nude mice modelTRAIL and Smac gene, E1B 55 kDa deletionEnhance synergistic anti-tumor effect[12]
Ad.wnt-E1A (Δ24bp)-TSLC1Intratumoral injection/MHCC-97H nude mice modelTSLC1 gene, 24 bp deletion of E1A CR2Enhance tumor selectivity antitumor activity[14]
OncoVV-AVLIn vitro/Hep-3B, PLC/PRF/5, Huh7Aphrocallistes vastus lectinEnhance the ability of replication[22]
T-01Left lobe of the liver injection/HuH-7, KYN-2, PLC/PRF/5, HepG2 athymic mice model-Enhance oncolytic safety[25]
VSV-β-galAdministered via the hepatic artery/McA-RH7777 ‘‘multifocal’’ HCC in the rat liverβ-galactosidase Enhance tumor-selective viral replication, and extensive oncolysis[33]
Wild type REOOral gavage/Huh7 SCID mice model-Inhibit the replication of HCV in HCV-HCC[37]
delNS1Inoculation in vivo/STAT12/2 mice, STAT11/1 mice, C57BL/6 micePartial deletion of NSInhibit the host antiviral response[40]
delNS1-GM-CSFIntratumoral injection/HepG2 nude mice modelPartial deletion of NS, GM-CSF insertedEnhance oncolytic efficacy and safety[42]
rNDV/F3aa (L289A)Administered via the hepatic artery/McA-RH7777 ‘‘multifocal’’ HCC in the rat liverA single amino acid substitution from alanine to L289A in the F proteinEnhance tumor specific syncytial formation and necrosis[46]
VG9-IL-24Intratumoral injection/SMMC-7721 nude mice modelIL-24Induce apoptosis of tumor cells[51]
Ad-GD55-α-TIM-3Intratumoral injection/PBMC mixed Bel-7404 BALB/c nude mice modelGP73 promoter and α-TIM-3Enhance the antitumor efficacy and the local immune response[52]
OVH-aMPD-1Combination with TIGIT blockade In vivo/hepa1-6 nude mice modelExpress a scFv against PD-1 (aMPD-1 scFv)Remodel immunosuppressive tumor microenvironment[59]

But there are still some problems to be solved. First of all, safety should be guaranteed first, in addition, the clinical drug OVs preparation is challenging and complex, and further research is needed to extract pure non-toxic single virus. Second, the system’s OV management route still has shortcomings. In the systematic administration of OVs, intratumoral injection has limitations and cannot completely eradicate the tumor, and intravenous injection overcomes the obstacle of intratumoral administration, but OV suffers from the destruction of host immunity during systemic transport, and these fatal limitations remain to be solved[61]. In addition, the targeting specificity and oncolytic efficiency of OVs are important, and additional CTs are required to confirm the effectiveness of various strategies for arming and modifying viruses[62].

The application value of OV is extremely promising. In the future, more extensive and in-depth basic research and CTs will pave the way for the treatment of OV. The safety and anti-tumor activity of OVs will be the focus of future research, and combination therapy and the continuous optimization of drug administration strategy will be the future research direction.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Oncology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade B

Creativity or Innovation: Grade C

Scientific Significance: Grade B

P-Reviewer: Abid S, Pakistan S-Editor: Chen YL L-Editor: A P-Editor: Guo X

References
1.  Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A, Roberts LR. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol. 2019;16:589-604.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2184]  [Cited by in F6Publishing: 2532]  [Article Influence: 506.4]  [Reference Citation Analysis (16)]
2.  Vogel A, Meyer T, Sapisochin G, Salem R, Saborowski A. Hepatocellular carcinoma. Lancet. 2022;400:1345-1362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 845]  [Article Influence: 422.5]  [Reference Citation Analysis (40)]
3.  Kennedy LB, Salama AKS. A review of cancer immunotherapy toxicity. CA Cancer J Clin. 2020;70:86-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 472]  [Cited by in F6Publishing: 791]  [Article Influence: 197.8]  [Reference Citation Analysis (36)]
4.  Li X, Sun X, Wang B, Li Y, Tong J. Oncolytic virus-based hepatocellular carcinoma treatment: Current status, intravenous delivery strategies, and emerging combination therapeutic solutions. Asian J Pharm Sci. 2023;18:100771.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 13]  [Reference Citation Analysis (40)]
5.  Shmulevitz M, Marcato P, Lee PW. Unshackling the links between reovirus oncolysis, Ras signaling, translational control and cancer. Oncogene. 2005;24:7720-7728.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 70]  [Article Influence: 3.7]  [Reference Citation Analysis (36)]
6.  Bommareddy PK, Shettigar M, Kaufman HL. Integrating oncolytic viruses in combination cancer immunotherapy. Nat Rev Immunol. 2018;18:498-513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 288]  [Cited by in F6Publishing: 306]  [Article Influence: 51.0]  [Reference Citation Analysis (16)]
7.  Lan Q, Xia S, Wang Q, Xu W, Huang H, Jiang S, Lu L. Development of oncolytic virotherapy: from genetic modification to combination therapy. Front Med. 2020;14:160-184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 39]  [Article Influence: 9.8]  [Reference Citation Analysis (17)]
8.  Lin D, Shen Y, Liang T. Oncolytic virotherapy: basic principles, recent advances and future directions. Signal Transduct Target Ther. 2023;8:156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 85]  [Reference Citation Analysis (36)]
9.  Choi JW, Lee JS, Kim SW, Yun CO. Evolution of oncolytic adenovirus for cancer treatment. Adv Drug Deliv Rev. 2012;64:720-729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 98]  [Article Influence: 8.2]  [Reference Citation Analysis (39)]
10.  Baird SK, Aerts JL, Eddaoudi A, Lockley M, Lemoine NR, McNeish IA. Oncolytic adenoviral mutants induce a novel mode of programmed cell death in ovarian cancer. Oncogene. 2008;27:3081-3090.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 59]  [Article Influence: 3.5]  [Reference Citation Analysis (41)]
11.  Wu YM, Zhang KJ, Yue XT, Wang YQ, Yang Y, Li GC, Li N, Wang YG. Enhancement of tumor cell death by combining cisplatin with an oncolytic adenovirus carrying MDA-7/IL-24. Acta Pharmacol Sin. 2009;30:467-477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 31]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
12.  Wang SB, Tan Y, Lei W, Wang YG, Zhou XM, Jia XY, Zhang KJ, Chu L, Liu XY, Qian WB. Complete eradication of xenograft hepatoma by oncolytic adenovirus ZD55 harboring TRAIL-IETD-Smac gene with broad antitumor effect. Hum Gene Ther. 2012;23:992-1002.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
13.  Wang Y, Huang P, Hu Y, Guo K, Jia X, Huang B, Liu X, He X, Huang F. An oncolytic adenovirus delivering TSLC1 inhibits Wnt signaling pathway and tumor growth in SMMC-7721 xenograft mice model. Acta Biochim Biophys Sin (Shanghai). 2021;53:766-774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
14.  Zhang J, Lai W, Li Q, Yu Y, Jin J, Guo W, Zhou X, Liu X, Wang Y. A novel oncolytic adenovirus targeting Wnt signaling effectively inhibits cancer-stem like cell growth via metastasis, apoptosis and autophagy in HCC models. Biochem Biophys Res Commun. 2017;491:469-477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 47]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
15.  Haddad D. Genetically Engineered Vaccinia Viruses As Agents for Cancer Treatment, Imaging, and Transgene Delivery. Front Oncol. 2017;7:96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 52]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
16.  Park BH, Hwang T, Liu TC, Sze DY, Kim JS, Kwon HC, Oh SY, Han SY, Yoon JH, Hong SH, Moon A, Speth K, Park C, Ahn YJ, Daneshmand M, Rhee BG, Pinedo HM, Bell JC, Kirn DH. Use of a targeted oncolytic poxvirus, JX-594, in patients with refractory primary or metastatic liver cancer: a phase I trial. Lancet Oncol. 2008;9:533-542.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 354]  [Cited by in F6Publishing: 322]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
17.  Heo J, Reid T, Ruo L, Breitbach CJ, Rose S, Bloomston M, Cho M, Lim HY, Chung HC, Kim CW, Burke J, Lencioni R, Hickman T, Moon A, Lee YS, Kim MK, Daneshmand M, Dubois K, Longpre L, Ngo M, Rooney C, Bell JC, Rhee BG, Patt R, Hwang TH, Kirn DH. Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer. Nat Med. 2013;19:329-336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 490]  [Cited by in F6Publishing: 587]  [Article Influence: 53.4]  [Reference Citation Analysis (0)]
18.  Cripe TP, Ngo MC, Geller JI, Louis CU, Currier MA, Racadio JM, Towbin AJ, Rooney CM, Pelusio A, Moon A, Hwang TH, Burke JM, Bell JC, Kirn DH, Breitbach CJ. Phase 1 study of intratumoral Pexa-Vec (JX-594), an oncolytic and immunotherapeutic vaccinia virus, in pediatric cancer patients. Mol Ther. 2015;23:602-608.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 119]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
19.  Prichard MN, Keith KA, Johnson MP, Harden EA, McBrayer A, Luo M, Qiu S, Chattopadhyay D, Fan X, Torrence PF, Kern ER. Selective phosphorylation of antiviral drugs by vaccinia virus thymidine kinase. Antimicrob Agents Chemother. 2007;51:1795-1803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 22]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
20.  Shugar D. Viral and host-cell protein kinases: enticing antiviral targets and relevance of nucleoside, and viral thymidine, kinases. Pharmacol Ther. 1999;82:315-335.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
21.  Breitbach CJ, Parato K, Burke J, Hwang TH, Bell JC, Kirn DH. Pexa-Vec double agent engineered vaccinia: oncolytic and active immunotherapeutic. Curr Opin Virol. 2015;13:49-54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
22.  Zhou Y, Wang Q, Ying Q, Zhang X, Chen K, Ye T, Li G. Effects of Oncolytic Vaccinia Viruses Harboring Different Marine Lectins on Hepatocellular Carcinoma Cells. Int J Mol Sci. 2023;24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
23.  Pan C, Cai Q, Li X, Li L, Yang L, Chen Y, Liu J, Liu W, Gao M, Sui T, Wang X, Fan H, Ruan J, Shi Y, Chen S, Cheng LS, Liu J, Yang H, Cheng G. Enhancing the HSV-1-mediated antitumor immune response by suppressing Bach1. Cell Mol Immunol. 2022;19:516-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
24.  Liu S, Dai M, You L, Zhao Y. Advance in herpes simplex viruses for cancer therapy. Sci China Life Sci. 2013;56:298-305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 9]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
25.  Nakatake R, Kaibori M, Nakamura Y, Tanaka Y, Matushima H, Okumura T, Murakami T, Ino Y, Todo T, Kon M. Third-generation oncolytic herpes simplex virus inhibits the growth of liver tumors in mice. Cancer Sci. 2018;109:600-610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
26.  Andtbacka RH, Kaufman HL, Collichio F, Amatruda T, Senzer N, Chesney J, Delman KA, Spitler LE, Puzanov I, Agarwala SS, Milhem M, Cranmer L, Curti B, Lewis K, Ross M, Guthrie T, Linette GP, Daniels GA, Harrington K, Middleton MR, Miller WH Jr, Zager JS, Ye Y, Yao B, Li A, Doleman S, VanderWalde A, Gansert J, Coffin RS. Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma. J Clin Oncol. 2015;33:2780-2788.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1504]  [Cited by in F6Publishing: 1591]  [Article Influence: 176.8]  [Reference Citation Analysis (0)]
27.  Todo T, Rabkin SD, Sundaresan P, Wu A, Meehan KR, Herscowitz HB, Martuza RL. Systemic antitumor immunity in experimental brain tumor therapy using a multimutated, replication-competent herpes simplex virus. Hum Gene Ther. 1999;10:2741-2755.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 166]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
28.  Friedman GK, Johnston JM, Bag AK, Bernstock JD, Li R, Aban I, Kachurak K, Nan L, Kang KD, Totsch S, Schlappi C, Martin AM, Pastakia D, McNall-Knapp R, Farouk Sait S, Khakoo Y, Karajannis MA, Woodling K, Palmer JD, Osorio DS, Leonard J, Abdelbaki MS, Madan-Swain A, Atkinson TP, Whitley RJ, Fiveash JB, Markert JM, Gillespie GY. Oncolytic HSV-1 G207 Immunovirotherapy for Pediatric High-Grade Gliomas. N Engl J Med. 2021;384:1613-1622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 208]  [Article Influence: 69.3]  [Reference Citation Analysis (0)]
29.  Ebert O, Shinozaki K, Huang TG, Savontaus MJ, García-Sastre A, Woo SL. Oncolytic vesicular stomatitis virus for treatment of orthotopic hepatocellular carcinoma in immune-competent rats. Cancer Res. 2003;63:3605-3611.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Barber GN. VSV-tumor selective replication and protein translation. Oncogene. 2005;24:7710-7719.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 114]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
31.  Sasso E, D’Alise AM, Zambrano N, Scarselli E, Folgori A, Nicosia A. New viral vectors for infectious diseases and cancer. Semin Immunol. 2020;50:101430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 57]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
32.  Marozin S, Altomonte J, Stadler F, Thasler WE, Schmid RM, Ebert O. Inhibition of the IFN-beta response in hepatocellular carcinoma by alternative spliced isoform of IFN regulatory factor-3. Mol Ther. 2008;16:1789-1797.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 40]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
33.  Shinozaki K, Ebert O, Kournioti C, Tai YS, Woo SL. Oncolysis of multifocal hepatocellular carcinoma in the rat liver by hepatic artery infusion of vesicular stomatitis virus. Mol Ther. 2004;9:368-376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 59]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
34.  Shinozaki K, Ebert O, Suriawinata A, Thung SN, Woo SL. Prophylactic alpha interferon treatment increases the therapeutic index of oncolytic vesicular stomatitis virus virotherapy for advanced hepatocellular carcinoma in immune-competent rats. J Virol. 2005;79:13705-13713.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 31]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
35.  Comins C, Heinemann L, Harrington K, Melcher A, De Bono J, Pandha H. Reovirus: viral therapy for cancer ‘as nature intended’. Clin Oncol (R Coll Radiol). 2008;20:548-554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
36.  Kelly K, Nawrocki S, Mita A, Coffey M, Giles FJ, Mita M. Reovirus-based therapy for cancer. Expert Opin Biol Ther. 2009;9:817-830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
37.  Samson A, Bentham MJ, Scott K, Nuovo G, Bloy A, Appleton E, Adair RA, Dave R, Peckham-Cooper A, Toogood G, Nagamori S, Coffey M, Vile R, Harrington K, Selby P, Errington-Mais F, Melcher A, Griffin S. Oncolytic reovirus as a combined antiviral and anti-tumour agent for the treatment of liver cancer. Gut. 2018;67:562-573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 39]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
38.  Kabiljo J, Laengle J, Bergmann M. From threat to cure: understanding of virus-induced cell death leads to highly immunogenic oncolytic influenza viruses. Cell Death Discov. 2020;6:48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 19]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
39.  Zhu Z, Fodor E, Keown JR. A structural understanding of influenza virus genome replication. Trends Microbiol. 2023;31:308-319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 26]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
40.  García-Sastre A, Egorov A, Matassov D, Brandt S, Levy DE, Durbin JE, Palese P, Muster T. Influenza A virus lacking the NS1 gene replicates in interferon-deficient systems. Virology. 1998;252:324-330.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 804]  [Cited by in F6Publishing: 789]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
41.  Katlinski KV, Gui J, Katlinskaya YV, Ortiz A, Chakraborty R, Bhattacharya S, Carbone CJ, Beiting DP, Girondo MA, Peck AR, Puré E, Chatterji P, Rustgi AK, Diehl JA, Koumenis C, Rui H, Fuchs SY. Inactivation of Interferon Receptor Promotes the Establishment of Immune Privileged Tumor Microenvironment. Cancer Cell. 2017;31:194-207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 177]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
42.  Penghui Y, Fang S, Ruilan W, Guanglin L, Hongjing G, Yueqiang D, Zhongpeng Z, Xiaolan Y, Zhaohai W, Shaogeng Z, Xiliang W. Oncolytic Activity of a Novel Influenza A Virus Carrying Granulocyte-Macrophage Colony-Stimulating Factor in Hepatocellular Carcinoma. Hum Gene Ther. 2019;30:330-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 10]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
43.  Ganar K, Das M, Sinha S, Kumar S. Newcastle disease virus: current status and our understanding. Virus Res. 2014;184:71-81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 237]  [Article Influence: 23.7]  [Reference Citation Analysis (1)]
44.  Schirrmacher V, van Gool S, Stuecker W. Breaking Therapy Resistance: An Update on Oncolytic Newcastle Disease Virus for Improvements of Cancer Therapy. Biomedicines. 2019;7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 53]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
45.  Bello MB, Yusoff K, Ideris A, Hair-Bejo M, Jibril AH, Peeters BPH, Omar AR. Exploring the Prospects of Engineered Newcastle Disease Virus in Modern Vaccinology. Viruses. 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
46.  Altomonte J, Marozin S, Schmid RM, Ebert O. Engineered newcastle disease virus as an improved oncolytic agent against hepatocellular carcinoma. Mol Ther. 2010;18:275-284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 69]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
47.  Zhu L, Lei Y, Huang J, An Y, Ren Y, Chen L, Zhao H, Zheng C. Recent advances in oncolytic virus therapy for hepatocellular carcinoma. Front Oncol. 2023;13:1172292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
48.  Wei M, Zuo S, Chen Z, Qian P, Zhang Y, Kong L, Gao H, Wei J, Dong J. Oncolytic vaccinia virus expressing a bispecific T-cell engager enhances immune responses in EpCAM positive solid tumors. Front Immunol. 2022;13:1017574.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 6]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
49.  Yu F, Wang X, Guo ZS, Bartlett DL, Gottschalk SM, Song XT. T-cell engager-armed oncolytic vaccinia virus significantly enhances antitumor therapy. Mol Ther. 2014;22:102-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 142]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
50.  Emdad L, Bhoopathi P, Talukdar S, Pradhan AK, Sarkar D, Wang XY, Das SK, Fisher PB. Recent insights into apoptosis and toxic autophagy: The roles of MDA-7/IL-24, a multidimensional anti-cancer therapeutic. Semin Cancer Biol. 2020;66:140-154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 51]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
51.  Deng L, Yang X, Ding Y, Fan J, Peng Y, Xu D, Huang B, Hu Z. Oncolytic therapy with vaccinia virus carrying IL-24 for hepatocellular carcinoma. Virol J. 2022;19:44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
52.  Qiang L, Huili Z, Leilei Z, Xiaoyan W, Hui W, Biao H, Yigang W, Fang H, Yiqiang W. Intratumoral delivery of a Tim-3 antibody-encoding oncolytic adenovirus engages an effective antitumor immune response in liver cancer. J Cancer Res Clin Oncol. 2023;149:18201-18213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
53.  Zhang Y, Zhang H, Wei M, Mou T, Shi T, Ma Y, Cai X, Li Y, Dong J, Wei J. Recombinant Adenovirus Expressing a Soluble Fusion Protein PD-1/CD137L Subverts the Suppression of CD8(+) T Cells in HCC. Mol Ther. 2019;27:1906-1918.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 34]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
54.  He LF, Wang YG, Xiao T, Zhang KJ, Li GC, Gu JF, Chu L, Tang WH, Tan WS, Liu XY. Suppression of cancer growth in mice by adeno-associated virus vector-mediated IFN-beta expression driven by hTERT promoter. Cancer Lett. 2009;286:196-205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
55.  Freedman JD, Duffy MR, Lei-Rossmann J, Muntzer A, Scott EM, Hagel J, Campo L, Bryant RJ, Verrill C, Lambert A, Miller P, Champion BR, Seymour LW, Fisher KD. An Oncolytic Virus Expressing a T-cell Engager Simultaneously Targets Cancer and Immunosuppressive Stromal Cells. Cancer Res. 2018;78:6852-6865.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 121]  [Article Influence: 20.2]  [Reference Citation Analysis (0)]
56.  Yang J, Chen Y, Jing Y, Green MR, Han L. Advancing CAR T cell therapy through the use of multidimensional omics data. Nat Rev Clin Oncol. 2023;20:211-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 44]  [Article Influence: 44.0]  [Reference Citation Analysis (0)]
57.  Watanabe K, Luo Y, Da T, Guedan S, Ruella M, Scholler J, Keith B, Young RM, Engels B, Sorsa S, Siurala M, Havunen R, Tähtinen S, Hemminki A, June CH. Pancreatic cancer therapy with combined mesothelin-redirected chimeric antigen receptor T cells and cytokine-armed oncolytic adenoviruses. JCI Insight. 2018;3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 183]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
58.  Ramelyte E, Tastanova A, Balázs Z, Ignatova D, Turko P, Menzel U, Guenova E, Beisel C, Krauthammer M, Levesque MP, Dummer R. Oncolytic virotherapy-mediated anti-tumor response: a single-cell perspective. Cancer Cell. 2021;39:394-406.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 53]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
59.  Lin C, Ren W, Luo Y, Li S, Chang Y, Li L, Xiong D, Huang X, Xu Z, Yu Z, Wang Y, Zhang J, Huang C, Xia N. Intratumoral Delivery of a PD-1-Blocking scFv Encoded in Oncolytic HSV-1 Promotes Antitumor Immunity and Synergizes with TIGIT Blockade. Cancer Immunol Res. 2020;8:632-647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 41]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
60.  Sangro B, Mazzolini G, Ruiz M, Ruiz J, Quiroga J, Herrero I, Qian C, Benito A, Larrache J, Olagüe C, Boan J, Peñuelas I, Sádaba B, Prieto J. A phase I clinical trial of thymidine kinase-based gene therapy in advanced hepatocellular carcinoma. Cancer Gene Ther. 2010;17:837-843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 88]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
61.  Zheng M, Huang J, Tong A, Yang H. Oncolytic Viruses for Cancer Therapy: Barriers and Recent Advances. Mol Ther Oncolytics. 2019;15:234-247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 179]  [Article Influence: 35.8]  [Reference Citation Analysis (0)]
62.  Zhu Z, McGray AJR, Jiang W, Lu B, Kalinski P, Guo ZS. Improving cancer immunotherapy by rationally combining oncolytic virus with modulators targeting key signaling pathways. Mol Cancer. 2022;21:196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 43]  [Reference Citation Analysis (0)]