key: cord-0006468-31srtult authors: Lan, Qiaoshuai; Xia, Shuai; Wang, Qian; Xu, Wei; Huang, Haiyan; Jiang, Shibo; Lu, Lu title: Development of oncolytic virotherapy: from genetic modification to combination therapy date: 2020-03-07 journal: Front Med DOI: 10.1007/s11684-020-0750-4 sha: 76d24b67f8dfb470083da0352c247b6d218686d5 doc_id: 6468 cord_uid: 31srtult Oncolytic virotherapy (OVT) is a novel form of immunotherapy using natural or genetically modified viruses to selectively replicate in and kill malignant cells. Many genetically modified oncolytic viruses (OVs) with enhanced tumor targeting, antitumor efficacy, and safety have been generated, and some of which have been assessed in clinical trials. Combining OVT with other immunotherapies can remarkably enhance the antitumor efficacy. In this work, we review the use of wild-type viruses in OVT and the strategies for OV genetic modification. We also review and discuss the combinations of OVT with other immunotherapies. Immunotherapy is an important antitumor therapy that involves stimulating or enhancing host antitumor immune response to kill and clear tumor cells [1, 2] . Oncolytic virotherapy (OVT) is a new antitumor immunotherapy that uses natural or genetically modified viruses to selectively infect and kill tumor cells [3] . OVT has various advantages over current antitumor therapies. Oncolytic viruses (OVs) can selectively replicate in tumor cells with high safety [4] and carry foreign genes, such as therapeutic and immunostimulatory genes [5] for specific expression at tumor sites [6] . Some OVs can cross the blood-brain barrier (BBB) to kill brain tumor cells, such as reovirus [7] and parvovirus H-1 (H-1PV) [8] , and OVs can turn "cold" tumors into "hot" tumors, thereby increasing the cellular sensitivity to other immunotherapies [9, 10] . To date, many OVs, including DNA viruses, such as adenovirus (AdV) [11, 12] , vaccinia virus (VACV) [13, 14] , herpesvirus [15, 16] , and parvovirus [17] and RNA viruses, such as reovirus [18, 19] , Newcastle disease virus (NDV) [20] , and measles virus (MV) [21] (Table 1 ) have been evaluated for cancer treatment (Fig. 1) . In the last century, scientists found that some viruses, such as NDV [32] , MV [33] , and parvovirus [34] , can naturally kill tumor cells. However, during this period, OVs were derived from wild-type or naturally attenuated virus strains, leaving considerable room for improvement in their safety and antitumor effects [35, 36] . In the 1990s, the emergence of recombinant DNA technology accelerated the development of OVT. Genetically modified OVs showed enhanced tumor targeting and killing efficiency against tumor cells with negligible damage to normal cells. The first genetically modified OV was herpes simplex virus (HSV)-1 developed by Martuza et al. in 1991 [37,38] . Oncorine (H101), a genetically modified oncolytic adenovirus (OAd), was approved by the State Food and Drug Administration (SFDA, now the National Medical Products Administration (NMPA)) in 2005 for the treatment of nasopharyngeal carcinoma in China [39, 40] . Talimogene laherparepvec (T-VEC; brand name Imlygic) was approved by the US Food and Drug Administration (FDA) and European Medicines Agency (EMA) for the treatment of advanced melanoma in 2015 [41, 42] . Researchers have recently attempted to combine OVT with other antitumor therapies, including chemotherapy [43] [44] [45] , radiotherapy [46, 47] , small molecules [48, 49] , and other immunotherapies [50, 51] , to achieve increased therapeutic effects against tumors. Among these treatments, the combination of OVT with other immunotherapies, especially regimens involving immune checkpoint inhibitors (ICIs) and chimeric antigen receptor-engineered T cell (CAR-T cell) immunotherapies, has shown promise as a cancer treatment [51, 52] . OVs can "heat up" tumors, thus enhancing their sensitivity to ICIs [53] . OVs can also piggyback CAR-T cells in a manner that enables them to overcome the immunosuppressive tumor microenvironment (TME) and increase their antitumor activity against solid tumors [54] . Recently, natural OVs have been continuously identified and widely applied in OVT. The renewed development of genetically modified OVs with enhanced tumor-targeting ability, antitumor efficacy, and safety has further promoted the development of OVT. In this work, we review the applications of wild-type viruses and the modifications for improving the tumor-targeting ability, antitumor efficacy, and safety of OVs. We also review and discuss the important research directions for OVT combination with other immunotherapies, including immune checkpoint blockade (ICB) and CAR-T cell therapies. Some wild-type OVs have a natural tropism for tumor cells based on their capacity to recognize highly expressed receptors on tumor cell surfaces or based on abnormal pathways or products in tumor cells, such as a defect in the interferon (IFN) signaling pathway and the activated Ras pathway. Among them, several representative OVs, such as coxsackievirus [55] , parvovirus [56] , and reovirus [57] , have been widely reported. Coxsackievirus A21 (CVA21) is a promising OV that depends on highly expressed receptor molecules on the surface of tumor cells, intercellular adhesion molecule-1 (ICAM-1, also named CD54), and decay-accelerating factor to preferentially enter, replicate in, and kill tumor cells. CVA21 exhibited antitumor efficacy in multiple cancers, including melanoma, prostate cancer, and multiple myeloma [30] . A CVA21-based OV (CAVATAK ® ) has been assessed in clinical trials for the treatment of malignant melanoma, bladder cancer [58] , and uveal melanoma with liver metastases [59] . The results revealed that CAVATAK ® has a good safety profile and can stimulate systemic antitumor immunity for the treatment of patients with melanoma [60] . CAVATAK ® can also induce immune cells to infiltrate the TME, indicating that its use in combination with other antitumor immunotherapies can increase antitumor efficacy. Wild-type H-1PV exhibits tumor selectivity, mainly depending on tumor-specific abnormal replication and transcription factors and the defective type I IFN-mediated antiviral pathway in tumor cells [17] . H-1PV nonstructural protein 1 (NS1) is important for promoting tumor cell death. Preclinical studies suggested that H-1PV can effectively kill various tumor cells [56] , including pancreatic ductal adenocarcinoma cells and glioma cells [61] . In particular, ParvOryx (wild-type H-1PV) showed an excellent safety profile in phase I/IIa clinical trials [8] . Wild-type reovirus can also selectively replicate in tumor cells in association with the abnormally activated Ras pathway in tumor cells [62] . Reovirus can cross the BBB to kill brain tumor cells through intravenous administration [7] . Reolysin ® (pelareorep) is a formulation of the reovirus serotype 3 Dearing strain and is being assessed in clinical trials for the treatment of multiple myeloma [63] , pancreatic adenocarcinoma [64] , and melanoma [65] . Reolysin ® has received an orphan drug designation from the US FDA and EMA for the treatment of gastric and pancreatic cancers [31] . In the treatment of multiple myeloma, the single use of Reolysin® did not show obvious antitumor activity [66] . However, when Reoly-sin® was combined with chemotherapy, such as carboplatin and paclitaxel, it exhibited improved therapeutic efficacy with an objective response rate (ORR) of up to 21% in the treatment of malignant melanoma [19] . Many alphaviruses, such as Semliki Forest virus (SFV) [67, 68] , Sindbis virus (SINV) [69] , and M1, exhibit antitumor activity. M1 is a recently identified Getah-like alphavirus from culicine mosquitoes [70, 71] . M1 has tumor selectivity related to zinc-finger antiviral protein (ZAP) deficiency in tumor cells and therefore can selectively replicate in tumor cells and induce apoptosis of the infected tumor cells by inducing irreversible endoplasmic reticulum (ER) stress [72] . M1 showed high tumor cell selectivity and tumor-killing activity during in vitro and in vivo experiments [73] . Moreover, its antitumor activity can be considerably increased by targeted inhibitors, such as valosin-containing protein (VCP) inhibitor and DNA-dependent kinase (DNA-PK) inhibitor [74, 75] , suggesting that combining M1 with these inhibitors is an important strategy. Genetic modification has accelerated the development of OVT [76] . In 1991, the first study reported on a genetically engineered OV, that is, an HSV with thymidine kinase (TK) deletion [38] . Many genetically modified OVs have been evaluated in clinical trials, as shown in Table 2 ; such OVs include AdV, VACV, and HSV. OVs mainly genetically modified through the deletion of virulence genes to improve the safety and insertion of foreign genes and improve antitumor efficacy or tumor targeting ability of OVs [77] . The genetic modifications of OVs can be (Fig. 2) . The tumor tropism of OVs is an important feature to ensure that they selectively replicate in and specifically kill tumor cells [79] . Wild-type OVs generally depend on tumorspecific cellular receptors or abnormal intracellular pathways and products to enter and selectively replicate in tumor cells and therefore can be optimized through genetic modifications to enhance their tumor targeting. Many genetically engineered OVs with enhanced tumor cell targeting ability have been developed on the basis of the tumor-specific high expression of certain receptors and the abnormal intracellular signaling pathways and metabolic status [80] . The nonspecificity, low affinity, or low expression of natural viral receptors on tumor cells can limit the application of wild-type OVs. Thus, OVs should be genetically modified to increase their binding affinity to specifically expressed receptors on tumor cells and increase their efficiency in entering and targeting tumor cells. Inserting targeting peptides can improve the entry efficiency of OVs [81] . For example, adenovirus type 5 (Ad5) is a widely used OV vector, but its receptor, coxsackie adenovirus receptor (CAR), is not highly expressed in many tumor cells [22] ; this phenomenon leads to the low entry efficiency of OAd. Through genetic engineering, an arginine-glycine-aspartic acid (RGD) motif was inserted into the fiber knob domain of Ad5 to generate a newly modified virus, which no longer depends on CAR to enter tumor cells and instead relies on integrins that are highly expressed on tumor cells, to enter tumor cells [82] . Other studies inserted the fiber knob domain derived from adenovirus type 3 (Ad3) into the backbone of Ad5 (also named serotype switching) to allow its entrance to the tumor cells by utilizing the highly expressed Ad3 receptor (desmoglein 2 as the primary receptor) on tumor cells to enter the tumor cells [79] . A similar modification involves inserting the fiber knob domain of adenovirus type 35 (Ad35) into the backbone of Ad5, allowing the OVs to utilize the Ad35 receptor (CD46 as the primary receptor) to enter tumor cells [81] . Some envelope glycoproteins (G) from other virus families can be inserted to improve tumor cell targeting. Betancourt et al. [83] reported replacing the G protein of vesicular stomatitis virus (VSV) with human immunodeficiency virus type 1 (HIV-1) gp160 to generate a new OV (VSV-gp160G); this method abandons the natural tissue tropism of VSV and specifically targets the new receptor, CD4. VSV-gp160G does not damage normal CD4 T cells; however, it exhibits potent killing activity against CD4expressing tumor cells, such as adult T cell leukemia/ lymphoma cell lines, in association with defective antiviral immune pathways in these tumor cells. A gene encoding a single-chain antibody (scAb) targeting human epidermal growth factor receptor 2 (HER2) was incorporated into HSV-1, making it fully targeted to tumor cells expressing HER2, and HER2-negative cells were left unharmed [84] [85] [86] . This modification also enhanced safety. Insertions of scAb targeting tumor-surface antigens, such as epithelial cell adhesion molecule (EpCAM) [87] and human carcinoembryonic antigen (CEA) [88] , have been reported to improve the tumor targeting of OVs. Shibata et al. [87] reported the insertion of the scAb targeting EpCAM (scEpCAM) into the HSV-1 genome. The engineered virus is highly specific to the EpCAM expressed on the surface of tumor cells to enter tumor cells; it can efficiently and specifically kill tumor cells expressing EpCAM. This strategy can improve the tumor targeting of OVs. However, these OVs have not yet entered clinical trials, and their clinical efficacy needs to be further confirmed. The various signaling pathways and the metabolic status of tumor cells are altered compared with those of normal cells [89] . Many features of tumor cells can be utilized by OVs to generate tumor selectivity [90] ; such features include defective antiviral pathways, such as a defective type I IFN signaling pathway [91] ; the loss of expression of tumor suppressor genes, such as loss of the retinoblastoma gene [92] ; and elevated signaling pathways, such as a transformed Ras pathway [93] . Therefore, targeting these abnormal pathways or products in tumor cells, deleted virulence factors and inserted tumor-specific promoters or microRNA-targeting sequences (miRNA-TSs) of OVs can regulate OVs specifically replicating in tumor cells [94, 95] . Deletion of virulence factors can improve the safety of OVs, which is discussed in the next section on the safety of genetically modified OVs. This section introduces the strategies for inserting tumor-specific promoters or miRNA-TSs to improve the tumor targeting of OVs. Insertion of tumor-specific promoters, such as the human telomerase reverse transcriptase (hTERT) promoter [96] and the prostate-specific antigen (PSA) promoter [97] , can improve the tumor selectivity of OVs [3] , which will, in turn, drive the expression of viral genes in tumor cells but not in normal cells. As an example, hTERT is highly expressed in most tumor cells but absent in normal cells [98] ; hence, the inserted hTERT promoter in OVs can initiate viral gene expression and replication in tumor cells with high hTERT expression [99] . OBP-301, an OAd with an inserted hTERT promoter, can replicate in tumor cells specifically, such as renal cell carcinoma cells and prostate cancer cells [100] . Different from the mechanisms of action of inserted tumor-specific promoters, inserted miRNA-TSs can lead to the viral RNA degradation through miRNAs in normal tissues; therefore, engineered OVs can specifically replicate in tumor cells with low miRNA expression [101, 102] . For example, targeting downregulated miRNA-7 in glioblastoma multiforme (GBM), researchers constructed a miRNA-7-sensitive oncolytic MV that specifically infects glioma cells without damaging normal cells [103] . The safety of OVs is important for their clinical use. Some natural viruses have the potential to kill tumor cells but can also damage normal cells. Numerous studies have suggested that the deletion of some genes necessary for viral replication in normal cells or virulence genes can improve OV safety in normal cells and maintain their oncolytic capacities; such genes include TK in oncolytic VACV [104, 105] and ICP34.5 in oncolytic HSV-1 [106, 107] . The viral products encoded by such genes are required for the replication of some OVs in normal cells, and they are abundant in tumor cells. Therefore, OVs can utilize tumor cell products to selectively replicate, but they cannot replicate in normal cells because relevant products are lacking. For example, TK, an important enzyme for nucleotide synthesis, is highly expressed in tumor cells [108] but only expressed at a low level in normal cells. TK encoded by VACV is essential for viral replication in normal cells. The deletion of the TK gene is an important genetic modification of oncolytic VACV [109, 110] to improve its safety. Oncolytic VACV with TK deletion can replicate in tumor cells but not in normal cells. Therefore, TK deletion can increase the safety of oncolytic VACV. Some oncolytic VACVs, such as GL-ONC1 and Pexa-Vec (JX-594), currently being assessed in clinical trials have TK deletions. GL-ONC1 has been assessed in clinical trials for the treatment of patients with peritoneal carcinomatosis (phase I/II) or cancer of the head and neck (phase I). Phase I clinical trial of GL-ONC1 for the treatment of peritoneal carcinomatosis [13, 111] showed its improved safety but also the emergence of minor adverse events. Phase II clinical trial results for Pexa-Vec showed that it could improve the survival of patients with liver cancer [14] ; thus, this OV has now entered phase III clinical trials for the treatment of hepatocellular carcinoma. Wild-type HSV-1 can cause latent infections in normal neurons, but the deletion of its neurovirulence factor (ICP34.5) can enhance its safety [112] . ICP34.5 antagonizes protein kinase R (PKR)-mediated immune response and is essential for infecting neurons. Oncolytic HSV with ICP34.5 deletion cannot replicate in neurons [24] . Given that PKR is usually downregulated in tumor cells, the virus can replicate normally in these tumor cells. HSV-1716, G207, and T-VEC are oncolytic HSVs with ICP34.5 deletions. T-VEC has been approved by the US FDA for the treatment of melanoma [113] . In a recent study, the ORR among patients with in-transit melanoma metastasis treated with T-VEC was 40.7% [114] . Zika virus (ZIKV) was recently found to have oncolytic properties [115] , but ZIKV infections in children can cause serious consequences [116, 117] . Wild-type ZIKV could infect glioblastoma stem cells (GSCs; an important target for glioblastoma treatment) and slow down the growth of tumors, but wild-type ZIKV also attacks normal nerve cells [115] ; hence, its safety should be further improved. A liveattenuated ZIKV vaccine (ZIKV-LAV) candidate [118] with 10 nucleotide deletions in the 3′ untranslated region in the ZIKV genome can maintain its oncolytic activity with increased safety and gene stability [119, 120] . In preclinical research, ZIKV-LAV effectively inhibited GSC growth and improved the survival of experimental animals compared with the mock control group. The median survival time of mice transplanted with 387 GSC cells was extended from 30 days to 48 days, while the median survival time of mice transplanted with 4121 GSC cells increased from 31 days to 53 days [119] . OVs can kill tumor cells through the following mechanisms: selectively replicating in tumor cells and causing the oncolysis of tumor cells [121] ; inducing the immunogenic cell death of tumor cells, thereby releasing damageassociated molecular pattern molecules, tumor-associated antigens, and pathogen-associated molecule patterns that stimulate antitumor immunity to kill other tumor cells [91] ; and destroying tumor vasculature to indirectly kill tumor cells [122, 123] . Insertion of genes coding for immunostimulatory molecules/cytokines, suicide genes, ECMdegrading enzymes, and anti-vasculature molecules can improve the antitumor efficacy of OVs [76, 124, 125] . OVs induce systemic antitumor immunity, and many cytokines, such as granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin (IL)-12 can increase the antitumor immunity. Thus, many OVs have been armed with immunostimulatory cytokines to improve their therapeutic efficacy [126] . For example, GM-CSF can recruit antigen-presenting cells (APCs) and promote the presentation of tumor antigens [78] . Various OVs armed with GM-CSF, including T-VEC (HSV-1), Pexa-Vec (VACV), and CG0070 (AdV), have been evaluated in clinical trials. IL-12 can stimulate IFN-g production, enhance the effect of T cells, and promote antitumor activity [127] . M032 is an oncolytic HSV expressing IL-12 [128] and is being assessed in the clinical trial for the treatment of GBM (the results of which have not yet been reported). Other cytokines, such as IL-2 [129] and IL-15 [130] , can also be inserted to improve antitumor efficiency. However, some secreted cytokines have systemic toxicity [131] . Liu et al. [132] have developed a new strategy to solve the side effects of the systemic administration of cytokines. They used membrane-bound IL-2, instead of secretory IL-2, to construct an oncolytic VACV that expresses membrane-bound IL-2 (named vvDD-IL-2-RG). Compared with the oncolytic VACV-expressing secretory IL-2, vvDD-IL-2-RG did not compromise antitumor activity and its toxicity was remarkably reduced. Inserting other immunostimulatory molecules, such as the costimulatory molecules CD40 ligand (CD40L) and 4-1BB ligand (4-1BBL), has also enhanced the antitumor effects of OVs [133, 134] . CD40 and 4-1BB belong to the tumor necrosis factor receptor (TNFR) superfamily. CD40 and 4-1BBL expressed on the surface of APCs and other cells can recognize and bind CD40L and 4-1BB on the surface of T cells, respectively, enhancing the ability of APCs to present antigens and promote T cell activation [135] . The recombinant adenovirus LOAd703 simultaneously expresses the immunostimulatory molecules CD40L and 4-1BBL [136] , resulting in the effective activation of T cells through the CD40 and 4-1BB signaling pathways, respectively; as such, they can enhance antitumor immunity. LOAd703 has entered clinical trials for the treatment of pancreatic cancer [137] but the results have not yet been reported. Suicide gene therapy includes two strategies: expressing pro-apoptotic proteins or toxin proteins to kill tumor cells directly and using prodrug-activating systems to kill tumor cells indirectly [138] . Engineered OVs can express proapoptotic proteins or toxin proteins to directly kill tumor cells, such as tumor necrosis factor-related apoptosisinducing ligand (TRAIL) [139, 140] and apoptin [141] . TRAIL can induce the apoptosis of tumor cells but does not damage normal cells [142] . Zhu et al. constructed a TRAIL-expressing OAd, which can efficiently kill triplenegative breast cancer cell lines in vitro without damaging normal cells and efficiently inhibit the growth of tumors in animal experiments [139] . Similarly, apoptin can induce the apoptosis of tumor cells but not that of normal cells, and apoptin-expressing OAd can effectively inhibit the growth of gastric carcinoma cells [143] . Conventional chemotherapies lack tumor selectivity and can cause damage to normal cells [144] . By contrast, virusdirected enzyme prodrug therapy is an ideal choice as it involves prodrug-activating enzymes expressed by recombinant OVs to turn nontoxic prodrugs into toxic drugs to specifically kill tumor cells [145] . TK and cytosine deaminase (CD) are widely used prodrug convertases. Chalikonda et al. [146] reported on the oncolytic VACV with an inserted CD gene; CD converts the nontoxic prodrug 5-fluorocytosine (5-FC) into 5-fluorouracil (5-FU) in the tumor sites, substantially enhancing the therapeutic effect owing to the presence of active drugs at the tumor sites. However, some tumor cells are resistant to 5-FU, reducing its therapeutic efficiency [147] . TG6002 is an oncolytic VACV expressing the suicide gene FCU1 [148] . FCU1 is a bifunctional gene comprising FCY1 and FUR1. FCY1 can convert 5-FC into 5-FU, and FUR1 can convert 5-FU into the more potent 5-fluorouracil-monophosphate [149] , exerting a strong antitumor effect. The results of clinical trials on TG6002 have yet to be reported. ECM in tumor tissues can inhibit the intratumoral spread of OVs and limit their antitumor efficacy [150] . Many studies have explored the effects of ECM-degrading enzymes, such as relaxin [151] , matrix metalloproteinase (MMP)-9 [152] , chondroitinase ABC [153] , and PH20 hyaluronidase [154] ; those ECM-degrading enzymes can increase the intratumoral spread of OVs and improve the antitumor activity of OVs through degrading ECM components. Therefore, expressing ECM-degrading enzymes is a feasible strategy to increase the antitumor activity of modified OVs. For example, GLV-1h255, an oncolytic VACV, can express MMP-9 to promote collagen IV degradation [152] and increase the intratumoral spread and antitumor efficacy of OV. VCN-01 is an OAd expressing PH20 hyaluronidase [155] , PH20 hyaluronidase can promote the spread of OVs between tumor cells by degrading the ECM. VCN-01 has now entered clinical trials for the treatment of pancreatic adenocarcinoma and retinoblastoma [156] . Tumor vasculature can transport nutrients and oxygen to tumor cells and is crucial to the growth of tumor cells and an ideal target for antitumor therapy [157] . Previous studies have found that anti-angiogenesis factors expressed by OVs can effectively inhibit tumor angiogenesis and Alexa et al. constructed an oncolytic VACV expressing scAb-targeting VEGF, and its antitumor efficiency was significantly improved [159] . In addition to targeting VEGF, OVs can express other anti-angiogenic factors, such as endostatin [160, 161] and thrombospondin-1 (TSP-1), to destroy tumor blood vessels [162] . Endostatin can inhibit tumor angiogenesis by preventing endothelial cell migration; oncolytic HSV expressing endostatin (HSV-Endo) effectively inhibited tumor angiogenesis and growth in the lung cancer model [160] . TSP-1 can inhibit tumor angiogenesis by inducing apoptosis and can improve the antitumor efficiency of OV; T-TSP-1 is an oncolytic HSV expressing TSP-1 that significantly inhibited the growth of gastric cancer in vivo [162] . Understanding the distribution and replication status of OVs in vivo helps in assessing the efficacy and safety of OVs [163] . OV replication can be monitored by incorporating them with reporter genes, such GFP and Rluc (usually used for optical imaging), NIS, and human norepinephrine transporter (NET) (usually used for deep tissue imaging) [164] . GFP insertion is widely used in OVT, and multiple OVs expressing GFP have been developed, such as MV-GFP (MV) [165] , JX-GFP (VACV) [104] , and rFMW/GFP (NDV) [166] . With the expression of GFP, fluorescence imaging can be used to directly observe the localization of OVs. However, monitoring OVs or tumors in deep tissues via GFP and Rluc is difficult, whereas NIS or NET can be used to monitor OV replication in deep tissues [164] . NIS is a membrane ion channel that mediates iodine transport. After NIS expressed by OVs takes up the iodide isotope, OV localization can be monitored using single-photon emission computed tomography or positron emission tomography [26, 167] . MV-NIS is an oncolytic MV expressing NIS, and it has entered clinical trials for the treatment of multiple myeloma [21] . In addition to the insertion of reporter genes, the replication status of OV in vivo can be reflected by the expression of soluble markers [168] , such as CEA. MV-CEA is an oncolytic MV expressing CEA; the serum CEA level can reflect the replication status of MV in vivo [169] , but it cannot be used for localization. When OVs are used as monotherapy, their antitumor efficacy can be limited by many factors [186] . Pre-existing antiviral immunity in human bodies could clear OVs rapidly [187] ; the heterogeneity of tumor cells might result in the low efficient replication and spread of OVs [187] ; the expression of immune checkpoint molecules might limit the antitumor activity of immune cells, which were attracted into the TME by OVs [51] . Researchers have begun to combine OVT with other antitumor therapies, particularly immunotherapies, including ICB (programmed cell death protein 1 (PD-1)/programmed death ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) signaling pathway blockade), and CAR-T cell therapies, to further improve the antitumor efficacy of OVs. ICB involves using ICIs to bind to immune checkpoint receptors or ligands and block immune inhibitory signals to treat cancer [188, 189] . The PD-1/PD-L1 and CTLA-4 pathways are two important immune inhibitory pathways. Several monoclonal antibodies blocking these two pathways have been approved by the US FDA. The treatment of some tumor types by using ICB is successful [190, 191] , but many factors that induce resistance of tumor cells to ICB, such as CD8 T cell deficiency in tumor tissues and low expression of immune signal molecules (such as PD-L1), have restricted the antitumor efficacy of ICB [192, 193] ; many patients also did not benefit from immune checkpoint therapy. Therefore, combinations of OVs with ICB have been widely explored in antitumor research to achieve better outcomes, and some combination therapies have been evaluated in clinical trials ( Table 3 ). The combination of OVs and ICB can exert a synergistic effect. OVs can attract CD8 T cells and natural killer (NK) cells into the TME. This phenomenon addresses the lack of immune cells in the TME and consequent inability of antibodies targeting the PD-1/PD-L1 and CTLA-4 pathways. Anti-PD-1/PD-L1 antibodies can block the PD-L1 induced by OVs (Fig. 3A) , and anti-CTLA-4 antibodies can block the corresponding inhibitory signaling pathway and enhance the antitumor activity of immune cells attracted by OVs. To date, six monoclonal antibodies targeting the PD-1/PD-L1 pathway have been approved by the US FDA for clinical use; among such antibodies, pembrolizumab, nivolumab, and cemiplimab are anti-PD-1 antibodies [209, 210] , whereas durvalumab, atezolizumab, and avelumab are anti-PD-L1 antibodies [211] . Tumeh et al. [212] analyzed samples from patients with melanoma in the phase I clinical trials of pembrolizumab and found that the CD8 T cell density at the tumor site was significantly lower in patients who did not respond to pembrolizumab compared with that of patients who responded to pembrolizumab. CD8 T cell deficiency in the TME is an important factor underlying the low efficacy of anti-PD-1 treatment. Studies have reported that OVs can increase the number of CD8 T cells in the TME and peripheral blood [28, 213, 214] , which is helpful in improving the therapeutic effect of PD-1/PD-L1 blockade [189] . OV infections can upregulate PD-L1 expression on the surface of tumor cells [7, 194, 196, 200] , while PD-1/PD-L1 blocking antibodies can block the PD-1/PD-L1 signaling pathway. Therefore, combining OVs with PD-1/PD-L1 blockade has synergistic roles in increasing antitumor effects. Chen et al. reported that combining oncolytic HSV-1716 with PD-1 blockade has a pronounced antitumor effect in murine rhabdomyosarcoma models compared with oncolytic HSV-1716 or anti-PD-1 antibody monotherapy; this improvement is associated with an increase in local antitumor T cell immune response [195] . Liu et al. combined oncolytic VACV with PD-L1 blocking antibody, which effectively inhibited tumor growth and improved the survival of experimental mice ( > 40% experimental mice were cured) [194] in colon cancer and ovarian cancer models. Fend et al. reported that combining the WR strain of VACV with ICB effectively improved the survival of experimental animals with MCA205 sarcoma [199] . Samson et al. combined oncolytic reovirus with ICB to treat brain tumors and found that sequential treatment with oncolytic reovirus and PD-1/PD-L1 blocking antibody effectively improved the survival of experimental mice with glioma [7] . Marie-Claude et al. also reported that oncolytic Maraba virus can induce long-term antitumor immunity and improve the immunosuppressive status of the TME, enhancing the sensitivity of TNBC to ICB [10] . In the treatment of non-solid tumors, such as acute myeloid leukemia (AML), the combination of OV and PD-1/PD-L1 blockade also produced promising outcomes. Shen et al. reported that oncolytic VSV (VSV-IFNβ-NIS) combined with anti-PD-L1 antibodies effectively killed murine AML cells and significantly improved the survival of AMLbearing mice [197] . Combinations of OVs and anti-PD-1/PD-L1 antibodies have been assessed in clinical trials. Ribas et al. reported the results of a phase Ib clinical trial of intratumoral T-VEC injection combined with pembrolizumab for the treatment of patients with advanced melanoma [200] . T-VEC enhanced the infiltration of CD4 and CD8 T cells and promoted the PD-L1 expression in tumor tissues, thereby improving the efficacy of pembrolizumab that led to an overall response rate of 62% and a complete response rate of 33%. This combination therapy did not increase toxicity compared with pembrolizumab monotherapy. Sun et al. reported the results of a clinical trial of T-VEC combined with anti-PD-1 antibody for unresectable stage III-IV melanoma treatment, leading to an overall response rate Combination therapy: more than 40% cures in models of colon and ovarian cancers [194] HSV1716 (HSV-1) Anti-PD-1 Combination therapy significantly prolongs survival of mice bearing M3-9-M tumors [195] MV-EGFR, MV-NIS (MV) Anti-PD-1 GL261 glioma cell G47-mIL12 (HSV-1) Anti-CTLA-4 and anti-PD-1 Median survival: 66.5 days (triple combination therapy) versus 20 days (mock-treated group) [198] WR (VACV) Anti-PD1 or anti-CTLA4 Combination therapy increased survival in MCA205 sarcoma model [199] Combinations with ICB in clinical trials (on-target lesions) of 90% (9/10) [201] . Zamarin et al. studied the combination of NDV and CTLA-4 blockade in the syngeneic mouse model [9] . The combination generated a synergistic effect, inhibiting the growth of local tumors. In addition, lymphocyte infiltration induced by NDV increased the sensitivity of distal tumor cells (where no OV was injected) to CTLA-4 blockade. Among the CTLA-4 blocking antibodies, ipilimumab has been approved by the US FDA for the treatment of melanoma [215] . In 2018, the results of the phase II clinical trial of T-VEC combined with ipilimumab for melanoma treatment were reported [203] , and phase Ib trial results were reported previously [202] . The combination of the two therapies increased antitumor efficacy compared with ipilimumab alone [203] (ORR: 39% versus 18%) with no reduction in safety. Comprehensive combination regimens have been used for some tumors with a highly immunosuppressed TME. GBM is one of the most lethal tumors, and its TME is highly immunosuppressed. A standalone ICI may not overcome the immunosuppressive state. Saha et al. combined anti-PD-1 antibody, anti-CTLA-4 antibody, and oncolytic HSV expressing IL-12 (G47-mIL-12) for GBM treatment [198] . Compared with dual therapy using anti-PD-1 and anti-CTLA-4 antibodies, triple therapy, including anti-PD-1 antibody, anti-CTLA-4 antibody, and G47-mIL-12, cured most experimental mice with 005 GSC GBM and significantly improved the survival of the CT-2A implanted mice. The median survival of CT-2A implanted mice was 66.5, 20, and 19 days for the triple therapy, mock treatment, and dual antibody therapy, respectively. Triple therapy can effectively trigger immune memory and prevent tumor recurrence compared with dual therapy. CAR-T cell therapy show considerable promise in cancer treatment. This therapy involves CAR-T cells derived from patients' T cells with an additional insertion of the chimeric antigen receptor (CAR) gene to enhance tumor cell recognition [216] . CAR-T cells targeting CD19 is the first CAR-T cell therapy approved by the US FDA for the treatment of B cell lymphoma [217] . CAR consists of an extracellular antigen recognition domain, a transmembrane region, and an intracellular signal domain [218] . The antigen recognition domain is an antibody fragment that recognizes targeted antigens. The intracellular signaling domain stimulates T cell activation and proliferation to kill target cells [219] . CAR-T cell therapies are successful in the treatment of several hematopoietic malignancies [220, 221] but not in the treatment of solid tumors [222] ; the immunosuppressive TME is a major obstacle to the role of CAR-T cells [223] . Given the many types of immunesuppressive cells, such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) in the TME [224] , and the lack of chemokines to attract tumor-specific T cells, the therapeutic efficacy of CAR-T cell therapy in solid tumors is limited [222] . Combining CAR-T cell therapy with genetically modified OVs can significantly increase CAR-T cell infiltration into the TME and enhance the therapeutic effect of CAR-T cells in solid tumors, as shown in Fig. 3B . Those modified OVs can attract CAR-T cells to the TME via cytokine expression. Nishio et al. inserted the genes encoding a chemokine (RANTES) and a cytokine (IL-15) into OAd to create Ad5D24.RANTES.IL-15. The intratumoral injection of Ad5D24.RANTES.IL-15 increased the infiltration of CAR-T cells targeting the tumor antigen GD2 (GD2. CAR-T cells) into the TME, thereby enhancing their tumor-killing ability [206] . Watanabe et al. constructed an AdV expressing TNF-α and IL-2 (OAd-TNFa-IL2) and proved that it could enhance the function of CAR-T cells [205] . Bispecific T cell engager (BiTE) is a fusion protein composed of two scAb fragments; one targets specific molecules on the surface of tumor cells, and the other binds to CD3e on the surface of T cells [225] . BiTE can bridge T cells and tumor cells, thereby promoting T cell activation to kill tumor cells [226] . Blinatumomab (BiTE targeting CD19) has been approved by the US FDA for the treatment of B cell acute lymphoblastic leukemia [227] . The combination of OV expressing BiTE with CAR-T cells can also produce potent antitumor activity [228] . Folate receptor α (FR-α), highly expressed in ovarian, breast, and lung tumor cells, is an important target for antitumor therapy [229] . However, small partial tumor cells remain with low or even no expression of FR-α in those tumor lesions that can lead to immune escape. Wing et al. [204] constructed an OAd armed with BiTE targeting epidermal growth factor receptor (OAd-BiTE). In the combination of OAd-BiTE and anti-FR-α CAR-T cells, EGFR-targeting BiTE expressed by OAd bridged the EGFR-overexpressing tumor cells and CAR-T cells to promote CAR-T cell activation and proliferation, and their ability to kill tumor cells was enhanced; as such, the survival of experimental mice was prolonged. All mice treated with CAR-T cells plus OAd-BiTE survived up to the experimental endpoint of 41 days, whereas the median survival of OAd-BiTEonly mice was 20 days and that of CAR-T cells-only mice was 38 days. Immune inhibitory molecules on the surface of cancer cells also affect the efficiency of CAR-T cell therapy. The transduction of immune checkpoint signals may decrease the therapeutic efficiency of CAR-T cells [230, 231] , while combinations of ICIs and CAR-T cells can increase the antitumor effects of CAR-T cells [232, 233] . Tanoue et al. used an OAd expressing anti-PD-L1 mini-antibody (CAd-VECPDL1) to block the transduction of the PD-1/PD-L1 inhibitory signaling pathway and improve the antitumor effect of CAR-T cells; the median survival of mice treated with CAd-VECPDL1 and HER2.CAR-T cells was 110 days, which is twofold longer than that of mice treated with single agent of OAd or CAR-T cells [208] . Compared with the systemic injection of anti-PD-L1 antibody, the anti-PD-L1 mini-antibody (mini-body) produced by OVs mainly localized in the tumor tissue with fewer adverse events [234] . Shaw et al. constructed an OAd expressing anti-PD-L1 mini-body and cytokine IL-12p70 (CAd12_PDL1) [207] . IL-12p70 expression could further promote CAR expression and enhance the tumor-killing effect. In the head and neck squamous cell carcinoma (HNSCC) xenograft model, combining CAd12_PDL1 and CAR-T cell therapy improved the median survival time of experimental animals by > 100 days in the CAd12_PDL1 + CAR-T group versus 21 or 24 days in the untreated group. At present, combinations of OVs and CAR-T cells for cancer treatment are still at the preclinical stage and have not yet entered clinical trials (Table 3) . With its development for more than 100 years, OVT has become highly efficient for cancer treatment [3] . OVs can specifically infect and kill tumor cells, remodel the immunosuppressive TME, and stimulate the systemic antitumor response. New viruses with oncolytic potential, such as M1 [75] and ZIKV [115, 119] , have been continuously discovered, thus further enriching OVT. Genetic modification can improve the safety and antitumor efficacy of OVs, which can be designed to carry different foreign genes to kill different kinds of tumor cells. T-VEC has been approved by the US FDA and EMA for the treatment of melanoma [41] . Some OVs, such as Pexa-Vec and Reolysin®, are being assessed in phase III clinical trials. The combination of OVT with ICB or CAR-T cell therapies can have synergistic effects, which might play an important role in clinical cancer treatment in the future. OVs can be also combined with other immunotherapies, such as tumor-infiltrating lymphocyte (TIL) therapies [235] . In TIL therapies, patients usually need to be injected with IL-2 to promote TIL proliferation, which might cause many adverse events [236] . The toxicity caused by systematic injections of high-dose IL-2 can be avoided by the intratumoral injection of OAd expressing IL-2 [237] . OVs combined with tumor-targeted monoclonal antibodies, such as trastuzumab [238] and bevacizumab [239] and some small-molecule antitumor compounds, such as trametinib [240] and NU7441 [75] , can generate synergistic effects and thus have broad applications in cancer treatment. However, some shortcomings or problems still confound the full use of OVT. For example, the small genomic capacities of some OVs, such as coxsackievirus and reovirus, limit their ability to accommodate large foreign genes that can optimize their antitumor efficacy [79] . Genetic modification by deleting virulence genes to improve the safety of OVs also reduces the antitumor efficacy of OVs [241] . Although encouraging results and broad applications have been achieved with the combination of OVT and other antitumor immunotherapies, further studies on the combination therapy schedules, including dosages, injection routes, and times, should be conducted to obtain the best antitumor efficiency. Qiaoshuai Lan, Shuai Xia, Qian Wang, Wei Xu, Haiyan Huang, Shibo Jiang, and Lu Lu declare no conflict of interest. This manuscript is a review article and does not involve a research protocol requiring approval by relevant institutional review board or ethics committee. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit https://creativecommons.org/ licenses/by/4.0/. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy Cancer immunotherapy: harnessing the immune system to battle cancer Oncolytic viruses: a new class of immunotherapy drugs Oncolytic viruses and their application to cancer immunotherapy Tcell engager-armed oncolytic vaccinia virus significantly enhances antitumor therapy Re-designing interleukin-12 to enhance its safety and potential as an anti-tumor immunotherapeutic agent Intravenous delivery of oncolytic reovirus to brain tumor patients immunologically primes for subsequent checkpoint blockade Oncolytic H-1 parvovirus shows safety and signs of immunogenic activity in a first phase I/IIa glioblastoma trial Localized oncolytic virotherapy overcomes systemic tumor resistance to immune checkpoint blockade immunotherapy Neoadjuvant oncolytic virotherapy before surgery sensitizes triple-negative breast cancer to immune checkpoint therapy Phase I study of DNX-2401 (Delta-24-RGD) oncolytic adenovirus: replication and immunotherapeutic effects in recurrent malignant glioma An open label, single-arm, phase II multicenter study of the safety and efficacy of CG0070 oncolytic vector regimen in patients with BCG-unresponsive non-muscle-invasive bladder cancer: interim results Phase I trial of intravenous oncolytic vaccinia virus (GL-ONC1) with cisplatin and radiotherapy in patients with locoregionally advanced head and neck carcinoma Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer OPTIM trial: a phase III trial of an oncolytic herpes virus encoding GM-CSF for unresectable stage III or IV melanoma Phase I dose-escalation clinical trial of HF10 oncolytic herpes virus in 17 Japanese patients with advanced cancer Molecular pathways: rodent parvoviruses-mechanisms of oncolysis and prospects for clinical cancer treatment Randomized phase 2 trial of the oncolytic virus Pelareorep (Reolysin) in upfront treatment of metastatic pancreatic adenocarcinoma A phase II study of REOLYSIN ® (pelareorep) in combination with carboplatin and paclitaxel for patients with advanced malignant melanoma Therapeutic potential of oncolytic Newcastle disease virus: a critical review Phase I trial of systemic administration of Edmonston strain of measles virus genetically engineered to express the sodium iodide symporter in patients with recurrent or refractory multiple myeloma Oncolytic viruses: adenoviruses Poxvirus oncolytic virotherapy Oncolytic virotherapy by HSV Immunotherapeutic potential of oncolytic H-1 parvovirus: hints of glioblastoma microenvironment conversion towards immunogenicity Clinical trials with oncolytic measles virus: current status and future prospects Fifty years of clinical application of Newcastle disease virus: time to celebrate Oncolytic VSV primes differential responses to immuno-oncology therapy Oncolytic polio virotherapy of cancer Errington-Mais F. Applications of coxsackievirus A21 in oncology Going (Reo)Viral: factors promoting successful reoviral oncolytic infection Observations on the repeated administration of viruses to a patient with acute leukemia. A preliminary report Hodgkin's disease: remissions after measles H-1 virus viremia in the human Oncolytic virusesinteraction of virus and tumor cells in the battle to eliminate cancer Oncolytic viral therapies-the clinical experience The current status and future prospects of oncolytic viruses in clinical trials against melanoma, glioma, pancreatic, and breast cancers Experimental therapy of human glioma by means of a genetically engineered virus mutant Oncorine, the world first oncolytic virus medicine and its update in China Fighting cancer with viruses: oncolytic virus therapy in China Talimogene laherparepvec (T-VEC) as cancer immunotherapy Talimogene laherparepvec: first in class oncolytic virotherapy Combination of paclitaxel and MG1 oncolytic virus as a successful strategy for breast cancer treatment Antitumor effect of oncolytic virus and paclitaxel encapsulated in extracellular vesicles for lung cancer treatment Chemovirotherapy of pancreatic adenocarcinoma by combining oncolytic vaccinia virus GLV-1h68 with nab-paclitaxel plus gemcitabine Oncolytic vaccinia virus combined with radiotherapy induces apoptotic cell death in sarcoma cells by down-regulating the inhibitors of apoptosis Combining oncolytic adenovirus with radiation-a paradigm for the future of radiosensitization In vitro screen of a small molecule inhibitor drug library identifies multiple compounds that synergize with oncolytic myxoma virus against human brain tumor-initiating cells First-in-class small molecule potentiators of cancer virotherapy Prospects for combined use of oncolytic viruses and CAR T-cells Oncolytic virus and PD-1/PD-L1 blockade combination therapy Oncolytic viruses: priming time for cancer immunotherapy Oncolytic reovirus sensitizes multiple myeloma cells to anti-PD-L1 therapy Lighting a fire in the tumor microenvironment using oncolytic immunotherapy Oncolytic coxsackievirus A21 as a novel therapy for multiple myeloma Preclinical testing of an oncolytic parvovirus: standard protoparvovirus H-1PV efficiently induces osteosarcoma cell lysis in vitro A phase I study of intravenous oncolytic reovirus type 3 Dearing in patients with advanced cancer Phase I trial of an ICAM-1-targeted immunotherapeutic-coxsackievirus A21 (CVA21) as an oncolytic agent against non muscle-invasive bladder cancer Viral targeting of non-muscle-invasive bladder cancer and priming of antitumor immunity following intravesical coxsackievirus A21 Final data from CALM: a phase II study of coxsackievirus A21 (CVA21) oncolytic virus immunotherapy in patients with advanced melanoma The oncolytic virotherapy era in cancer management: prospects of applying H-1 parvovirus to treat blood and solid cancers Oncolytic reovirus induces intracellular redistribution of Ras to promote apoptosis and progeny virus release A phase I trial of single-agent reolysin in patients with relapsed multiple myeloma A phase II study of Pelareorep (REOLYSIN ® ) in combination with gemcitabine for patients with advanced pancreatic adenocarcinoma Phase II trial of intravenous administration of Reolysin(®) (Reovirus Serotype-3-dearing Strain) in patients with metastatic melanoma Histone deacetylase inhibitors enhance the therapeutic potential of reovirus in multiple myeloma Safe and effective treatment of experimental neuroblastoma and glioblastoma using systemically delivered triple microRNA-detargeted oncolytic Semliki Forest Virus Virotherapy with a Semliki Forest virus-based vector encoding IL12 synergizes with PD-1/PD-L1 blockade Oncolytic Sindbis virus targets tumors defective in the interferon response and induces significant bystander antitumor immunity in vivo Identification and characterization of alphavirus M1 as a selective oncolytic virus targeting ZAP-defective human cancers Intravenous injections of the oncolytic virus M1 as a novel therapy for muscleinvasive bladder cancer Inhibition of the mevalonate pathway enhances cancer cell oncolysis mediated by M1 virus Naturally existing oncolytic virus M1 is nonpathogenic for the nonhuman primates after multiple rounds of repeated intravenous injections Targeting VCP enhances anticancer activity of oncolytic virus M1 in hepatocellular carcinoma DNA-PK inhibition synergizes with oncolytic virus M1 by inhibiting antiviral response and potentiating DNA damage From benchtop to bedside: a review of oncolytic virotherapy Designing and building oncolytic viruses Oncolytic virusesnatural and genetically engineered cancer immunotherapies Integrating oncolytic viruses in combination cancer immunotherapy New viruses for cancer therapy: meeting clinical needs Tropism and transduction of oncolytic adenovirus 5 vectors in cancer therapy: focus on fiber chimerism and mosaicism, hexon and pIX Oncolytic virotherapy for the treatment of malignant glioma Retargeting oncolytic vesicular stomatitis virus to human T-cell lymphotropic virus type 1-associated adult T-cell leukemia A fully-virulent retargeted oncolytic HSV armed with IL-12 elicits local immunity and vaccine therapy towards distant tumors Construction of a fully retargeted herpes simplex virus 1 recombinant capable of entering cells solely via human epidermal growth factor receptor 2 Eradication of glioblastoma by immuno-virotherapy with a retargeted oncolytic HSV in a preclinical model Development of an oncolytic HSV vector fully retargeted specifically to cellular EpCAM for virus entry and cell-to-cell spread Effective treatment of an orthotopic xenograft model of human glioblastoma using an EGFR-retargeted oncolytic herpes simplex virus Hallmarks of cancer: the next generation Oncolytic viruses: exploiting cancer's deal with the devil Molecular pathways: mechanism of action for Talimogene Laherparepvec, a new oncolytic virus immunotherapy The oncolytic adenovirus VCN-01 as therapeutic approach against pediatric osteosarcoma Oncolytic reovirus induces intracellular redistribution of Ras to promote apoptosis and progeny virus release Telomerase-specific oncolytic adenoviral therapy for orthotopic hepatocellular carcinoma in HBx transgenic mice MicroRNA-145 regulates oncolytic herpes simplex virus-1 for selective killing of human non-small cell lung cancer cells Telomerase-specific oncolytic virotherapy for human gastrointestinal cancer Oncolytic viruses driven by tumor-specific promoters A novel oHSV-1 targeting telomerase reverse transcriptase-positive cancer cells via tumorspecific promoters regulating the expression of ICP4 Enhanced oncolysis by a tropism-modified telomerase-specific replication-selective adenoviral agent OBP-405 ('Telomelysin-RGD') Potent antitumor effects of combined therapy with a telomerase-specific, replication-competent adenovirus (OBP-301) and IL-2 in a mouse model of renal cell carcinoma Targeting human breast cancer cells by an oncolytic adenovirus using microRNA-targeting strategy Enhanced control of oncolytic measles virus using microRNA target sites MicroRNA-sensitive oncolytic measles viruses for cancer-specific vector tropism Systemic cancer therapy with a tumorselective vaccinia virus mutant lacking thymidine kinase and vaccinia growth factor genes Viruses as nanomedicine for cancer Effect of g34.5 deletions on oncolytic herpes simplex virus activity in brain tumors Selective in vitro replication of herpes simplex virus type 1 (HSV-1) ICP34.5 null mutants in primary human CNS tumours-evaluation of a potentially effective clinical therapy Targeted and armed oncolytic poxviruses: a novel multi-mechanistic therapeutic class for cancer Oncolytic viral therapy for mesothelioma The mechanisms of genetically modified vaccinia viruses for the treatment of cancer Phase I study of oncolytic vaccinia virus GL-ONC1 in patients with peritoneal carcinomatosis Talimogene laherparepvec (T-VEC) for the treatment of advanced melanoma Talimogene laherparepvec (T-Vec) for the treatment of melanoma and other cancers Efficacy of Talimogene Laherparepvec (T-VEC) therapy in patients with in-transit melanoma metastasis decreases with increasing lesion size Zika virus has oncolytic activity against glioblastoma stem cells Zika virus: history of a newly emerging arbovirus Zika virus: an emerging flavivirus A live-attenuated Zika virus vaccine candidate induces sterilizing immunity in mouse models Treatment of human glioblastoma with a live attenuated Zika virus vaccine candidate Zika virus vaccine: progress and challenges First-in-man study of western reserve strain oncolytic vaccinia virus: safety, systemic spread, and antitumor activity Targeting tumor vasculature with an oncolytic virus Oncolytic vaccinia virus disrupts tumor-associated vasculature in humans Oncolytic immunotherapy: unlocking the potential of viruses to help target cancer Promising oncolytic agents for metastatic breast cancer treatment Oncolytic virus-based cytokine expression to improve immune activity in brain and solid tumors Evaluation of the safety and biodistribution of M032, an attenuated herpes simplex virus type 1 expressing hIL-12, after intracerebral administration to aotus nonhuman primates Design of a phase I clinical trial to evaluate M032, a genetically engineered HSV-1 expressing IL-12, in patients with recurrent/progressive glioblastoma multiforme, anaplastic astrocytoma, or gliosarcoma Recombinant Newcastle disease virus (NDV/Anh-IL-2) expressing human IL-2 as a potential candidate for suppresses growth of hepatoma therapy Oncolytic influenza A virus expressing interleukin-15 decreases tumor growth in vivo Development of an attenuated interleukin-2 fusion protein that can be activated by tumour-expressed proteases Modifying the cancer-immune set point using vaccinia virus expressing re-designed interleukin-2 Safety and biodistribution of a double-deleted oncolytic vaccinia virus encoding CD40 ligand in laboratory Beagles Therapeutic and tumor-specific immunity induced by combination of dendritic cells and oncolytic adenovirus expressing IL-12 and 4-1BBL The TNFRs OX40, 4-1BB, and CD40 as targets for cancer immunotherapy Shaping the tumor stroma and sparking immune activation by CD40 and 4-1BB signaling induced by an armed oncolytic virus Recent advances in oncolytic adenovirus therapies for cancer Cancer suicide gene therapy: a patent review Oncolytic adenovirus encoding tumor necrosis factor-related apoptosis inducing ligand (TRAIL) inhibits the growth and metastasis of triple-negative breast cancer Trail armed oncolytic poxvirus suppresses lung cancer cell by inducing apoptosis Recombinant adenoviruses expressing apoptin suppress the growth of MCF7 breast cancer cells and affect cell autophagy Telomerase-specific oncolytic adenovirus expressing TRAIL suppresses peritoneal dissemination of gastric cancer Therapeutic efficacy of an hTERT promoter-driven oncolytic adenovirus that expresses apoptin in gastric carcinoma Viral vectors for gene-directed enzyme prodrug therapy Gene-directed enzyme prodrug therapy Oncolytic virotherapy for ovarian carcinomatosis using a replication-selective vaccinia virus armed with a yeast cytosine deaminase gene Targeted chemotherapy for head and neck cancer with a chimeric oncolytic adenovirus coding for bifunctional suicide protein FCU1 The enhanced tumor specificity of TG6002, an armed oncolytic vaccinia virus deleted in two genes involved in nucleotide metabolism In vivo cancer gene therapy by adenovirus-mediated transfer of a bifunctional yeast cytosine deaminase/uracil phosphoribosyltransferase fusion gene Strategies to enhance viral penetration of solid tumors Relaxin expression from tumor-targeting adenoviruses and its intratumoral spread, apoptosis induction, and efficacy Vaccinia virus-mediated intra-tumoral expression of matrix metalloproteinase 9 enhances oncolysis of PC-3 xenograft tumors Chondroitinase ABC I-mediated enhancement of oncolytic virus spread and antitumor efficacy Hyaluronidase expression by an oncolytic adenovirus enhances its intratumoral spread and suppresses tumor growth Safety and efficacy of VCN-01, an oncolytic adenovirus combining fiber HSG-binding domain replacement with RGD and hyaluronidase expression Therapeutic targeting of the RB1 pathway in retinoblastoma with the oncolytic adenovirus VCN-01 Tumor angiogenesis and vascular normalization: alternative therapeutic targets Vascular endothelial growth factor (VEGF) signaling in tumour vascularization: potential and challenges Anti-VEGF single-chain antibody GLAF-1 encoded by oncolytic vaccinia virus significantly enhances antitumor therapy Angiogenesis inhibition using an oncolytic herpes simplex virus expressing endostatin in a murine lung cancer model Treatment of medulloblastoma with oncolytic measles viruses expressing the angiogenesis inhibitors endostatin and angiostatin An armed oncolytic herpes simplex virus expressing thrombospondin-1 has an enhanced in vivo antitumor effect against human gastric cancer The use of the NIS reporter gene for optimizing oncolytic virotherapy Genetically engineered vaccinia viruses as agents for cancer treatment, imaging, and transgene delivery Measles Edmonston vaccine strain derivatives have potent oncolytic activity against osteosarcoma Recombinant oncolytic Newcastle disease virus displays antitumor activities in anaplastic thyroid cancer cells Measles to the rescue: a review of oncolytic measles virus Noninvasive in vivo monitoring of trackable viruses expressing soluble marker peptides Potential and clinical translation of oncolytic measles viruses Talimogene laherparepvec (T-VEC) for the treatment of advanced melanoma Viruses for tumor therapy Cutaneous head and neck melanoma in OPTiM, a randomized phase 3 trial of talimogene laherparepvec versus granulocyte-macrophage colony-stimulating factor for the treatment of unresected stage IIIB/IIIC/IV melanoma Genomic signature of the natural oncolytic herpes simplex virus HF10 and its therapeutic role in preclinical and clinical trials The oncolytic virus Delta-24-RGD elicits an antitumor effect in pediatric glioma and DIPG mouse models Use of telomelysin (OBP-301) in mouse xenografts of human head and neck cancer Pexa-Vec double agent engineered vaccinia: oncolytic and active immunotherapeutic Development of PROSTVAC immunotherapy in prostate cancer Recent advances in vesicular stomatitis virus-based oncolytic virotherapy: a 5-year update Cytotoxic and immunogenic mechanisms of recombinant oncolytic poliovirus Recurrent glioblastoma treated with recombinant poliovirus Preclinical development of peptide vaccination combined with oncolytic MG1-E6E7 for HPV-associated cancer Clinical development of reovirus for cancer therapy: an oncolytic virus with immunemediated antitumor activity Double-faceted mechanism of parvoviral oncosuppression A non-controlled, single arm, open label, phase II study of intravenous and intratumoral administration of ParvOryx in patients with metastatic, inoperable pancreatic cancer: ParvOryx02 protocol Phase 1 clinical experience using intravenous administration of PV701, an oncolytic Newcastle disease virus Armed therapeutic viruses -a disruptive therapy on the horizon of cancer immunotherapy Chemotherapy and oncolytic virotherapy: advanced tactics in the war against cancer Immune checkpoint blockade: a common denominator approach to cancer therapy The blockade of immune checkpoints in cancer immunotherapy Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial PD-1 and PD-L1 checkpoint signaling inhibition for cancer immunotherapy: mechanism, combinations, and clinical outcome Primary and acquired resistance to PD-1/PD-L1 blockade in cancer treatment Tumour-intrinsic resistance to immune checkpoint blockade Rational combination of oncolytic vaccinia virus and PD-L1 blockade works synergistically to enhance therapeutic efficacy Cooperation of oncolytic herpes virotherapy and PD-1 blockade in murine rhabdomyosarcoma models Immunovirotherapy with measles virus strains in combination with anti-PD-1 antibody blockade enhances antitumor activity in glioblastoma treatment Immunovirotherapy with vesicular stomatitis virus and PD-L1 blockade enhances therapeutic outcome in murine acute myeloid leukemia Macrophage polarization contributes to glioblastoma eradication by combination immunovirotherapy and immune checkpoint blockade Immune checkpoint blockade, immunogenic chemotherapy or IFN-α blockade boost the local and abscopal effects of oncolytic virotherapy Oncolytic virotherapy promotes intratumoral T cell infiltration and improves anti-PD-1 immunotherapy Talimogene Laherparepvec combined with anti-PD-1 based immunotherapy for unresectable stage III-IV melanoma: a case series Talimogene Laherparepvec in combination with ipilimumab in previously untreated, unresectable stage IIIB-IV melanoma Randomized, open-label phase II study evaluating the efficacy and safety of Talimogene Laherparepvec in combination with ipilimumab versus ipilimumab alone in patients with advanced, unresectable melanoma Improving CART-cell therapy of solid tumors with oncolytic virus-driven production of a bispecific T-cell engager Pancreatic cancer therapy with combined mesothelin-redirected chimeric antigen receptor T cells and cytokine-armed oncolytic adenoviruses Armed oncolytic virus enhances immune functions of chimeric antigen receptor-modified T cells in solid tumors Adenovirotherapy delivering cytokine and checkpoint inhibitor augments CAR T cells against metastatic head and neck cancer Armed oncolytic adenovirusexpressing PD-L1 mini-body enhances antitumor effects of chimeric antigen receptor T cells in solid tumors Monoclonal antibodies for the treatment of cancer Cemiplimab approved for treatment of CSCC Durvalumab: first global approval PD-1 blockade induces responses by inhibiting adaptive immune resistance T-cell subsets in peripheral blood and tumors of patients treated with oncolytic adenoviruses Oncolytic immunotherapy of advanced solid tumors with a CD40L-expressing replicating adenovirus: assessment of safety and immunologic responses in patients Ipilimumab: from preclinical development to future clinical perspectives in melanoma Chimeric antigen receptor (CAR) T-cell therapy A review of chimeric antigen receptor Tcells in lymphoma Chimeric antigen receptor T-cell therapies for multiple myeloma Driving CAR T-cells forward CAR T cell therapy of non-hematopoietic malignancies: detours on the road to clinical success CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia Chimeric antigen receptor T cell (CAR-T) immunotherapy for solid tumors: lessons learned and strategies for moving forward Cancer immunotherapy strategies based on overcoming barriers within the tumor microenvironment Innate immune landscape in early lung adenocarcinoma by paired single-cell analyses A BiTE from cancer's intracellular menu Bispecific T-cell engagers for cancer immunotherapy Utilizing the BiTE (bispecific T-cell engager) platform for immunotherapy of cancer Solid tumor immunotherapy with T cell engager-armed oncolytic viruses Targeting folate receptor alpha for cancer treatment Programmed cell death protein 1 activation preferentially inhibits CD28.CAR-T cells Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition Combination cancer immunotherapy and new immunomodulatory targets Enhancement of PSMAdirected CAR adoptive immunotherapy by PD-1/PD-L1 blockade Immune-related adverse events associated with immune checkpoint blockade Achievements and challenges of adoptive T cell therapy with tumor-infiltrating or blood-derived lymphocytes for metastatic melanoma: what is needed to achieve standard of care? Adoptive transfer of tumor-infiltrating lymphocytes in patients with metastatic melanoma: intent-to-treat analysis and efficacy after failure to prior immunotherapies Adenoviral production of interleukin-2 at the tumor site removes the need for systemic postconditioning in adoptive cell therapy Oncolytic reovirus combined with trastuzumab enhances antitumor efficacy through TRAIL signaling in human HER2-positive gastric cancer cells Combination therapy of oncolytic herpes simplex virus HF10 and bevacizumab against experimental model of human breast carcinoma xenograft MEK inhibition enhances oncolytic virus immunotherapy through increased tumor cell killing and T cell activation A novel chimeric oncolytic virus vector for improved safety and efficacy as a platform for the treatment of hepatocellular carcinoma