6,342
Views
64
CrossRef citations to date
0
Altmetric
Review

Trial Watch: Oncolytic viro-immunotherapy of hematologic and solid tumors

, , , , , , ORCID Icon, , , , , , , ORCID Icon & show all
Article: e1503032 | Received 08 Jul 2018, Accepted 15 Jul 2018, Published online: 27 Aug 2018

ABSTRACT

Oncolytic viruses selectively target and kill cancer cells in an immunogenic fashion, thus supporting the establishment of therapeutically relevant tumor-specific immune responses. In 2015, the US Food and Drug Administration (FDA) approved the oncolytic herpes simplex virus T-VEC for use in advanced melanoma patients. Since then, a plethora of trials has been initiated to assess the safety and efficacy of multiple oncolytic viruses in patients affected with various malignancies. Here, we summarize recent preclinical and clinical progress in the field of oncolytic virotherapy.

Introduction

Since immune checkpoint inhibitors (ICIs) have revolutionized the care of melanoma patients, the field of immunotherapy has diversified and extended to multiple malignant indications. Cancer immunotherapies currently approved by the US Food and Drug Administration (FDA) for clinical use include: a) six ICIs that bind either to programmed cell death 1 (PDCD1; best known as PD-1) (i.e., nivolumab, pembrolizumab), either to CD274 (best known as PD-L1) (i.e., avelumab, atezolizumab and durvalumab), or to cytotoxic T-lymphocyte associated protein 4 (CTLA4) (i.e., ipilimumab);Citation1Citation15 b) multiple monoclonal antibodies (mAb) targeting surface tumor-associated antigens (TAAs) (e.g., the anti-CD20 mAb rituximab, the anti-ERBB2 mAb trastuzumab);Citation15Citation17 c) two immunostimulatory cytokines (i.e., interferon [IFN]-α and interleukin [IL]-2);Citation18 d) several immunogenic cell death inducers (e.g., anthracyclines, cyclophosphamide [CPA], oxaliplatin),Citation19Citation28 e) adoptive chimeric antigen receptor [CAR] T-cell therapy (i.e., tisagenlecleucel and axicabtagene ciloleucel),Citation29Citation33 f) bacillus Calmette-Guérin (BCG) immunotherapy,Citation34,Citation35 g) adoptive dendritic cell [DC]-based cancer vaccine (i.e., sipuleuceul-T)Citation31,Citation36Citation39 and h) oncolytic viro-immunotherapy (i.e., talimogene laherparepvec [T-VEC]).Citation40Citation46

Oncolytic viruses (OVs) are non-pathogenic replication-competent viruses that preferentially infect, replicate in and kill cancer cells while sparing their normal counterparts.Citation40,Citation47Citation49 The list of viruses that demonstrate oncolytic activity keeps expanding and includes agents of animal (e.g., vaccinia virus [VV]) and human (e.g., serotype 3 reovirus, type 1 herpes simplex virus [HSV-1]) origin.Citation50Citation64 Tumor selectivity, referred to as ‘oncotropism’, requires that cancer cells i) express the virus entry receptor and ii) demonstrate permissiveness to virus life cycle completion.Citation65Citation69 Alongside, OVs should undergo abortive infection in most normal tissues to prevent side effects and toxicity, which is of particular concern when treating immunosuppressed patients.Citation70,Citation71 Oncotropism can be natural or, more frequently, genetically acquired/restricted. Genetic engineering procedures implemented so far to OVs consisted of 1) modifying the OV attachment protein to redirect or strengthen its binding to a tumor-associated receptor,Citation72Citation81 2) inserting cis-regulatory elements in the OV genome to guarantee a transcriptional and/or post-transcriptional control of its expression/replication in target rather than off-target tissues,Citation61,Citation82Citation96 and/or 3) abolishing the activity of virulence factors in charge of hacking proliferation, survival and/or antiviral machineries of the host cell.Citation41,Citation92,Citation96Citation105 The latter strategy has been extensively applied in the development of most OVs currently evaluated into the clinic. Indeed, it generates viral strains with strongly attenuated infectivity in normal cells but with preserved replicative efficacy in cancer cells as the features of the malignant phenotype (e.g., defective/attenuated IFN response, sustained nucleotide and protein synthesis, resistance to cell death)Citation66,Citation106Citation111 complement the activity of the mutated viral factors. For instance, the FDA-approved OV T-VEC (trade name Imlygic, formerly called OncoVexGM-CSF, Amgen, Thousand Oaks, CA, USA) consists of an HSV-1 (JS1 strain) that underwent deletion of two neurovirulence factors, ICP34.5 and ICP47, responsible for counteracting innate and adaptive immunity, respectively.Citation41,Citation112Citation114 OV-induced cancer cell killing, referred to as ‘oncolysis’, is consecutive to multimodal mechanisms of action. First, tumor cell elimination can result from direct oncolysis due to completion of a lytic viral cycle and/or induction of programmed cancer cell death upon virus replication.Citation19,Citation20,Citation47Citation49,Citation112,Citation115Citation118 Accordingly, some OVs have been armed with pro-apoptotic factors or combined with cytotoxic agents to enhance direct oncolysis.Citation77,Citation87,Citation88,Citation92,Citation98,Citation118Citation139 Nevertheless, most of the efficacy of oncolytic virotherapy is attributed to indirect means. Indeed, OV replication within the tumor microenvironment (TME) remains limited in extent and transient due to the antiviral immune pressure, rendering direct oncolysis marginally efficient.Citation64,Citation68,Citation140Citation143 As an aside, multiple strategies have been introduced to favor OV targeting to, persistence and/or spread within the tumor by circumventing elimination from the host immune machinery. Genetic engineering, coating of the OV attachment protein,Citation59,Citation144Citation146 cell carrier,Citation78,Citation147Citation152 or biochemical/pharmacological immunosuppressionCitation153-Citation159 have proven efficient at limiting virus neutralization/clearing mediated by antibodies, the complement system or innate immune cells such as phagocytes. This is particularly important when OVs are administered systemically (rather than intralesionally) for the treatment of hardly accessible primary neoplasms or disseminated malignant tissues.Citation160Citation163 To facilitate the spread of OV progenies from one infected cell to uninfected neighbors, transgenes or agents affecting the cell junctions or the extracellular matrix have also been employed.Citation138,Citation156,Citation164Citation166 Of note, anti-angiogenic properties have also been ascribed to some OV strains for their ability to acutely disrupt the tumor neovasculature, thus inducing vast necrotic areas within infected neoplasms.Citation167Citation171 In line with this notion, several groups have equipped or associated OVs with genetic inserts or drugs impairing blood vessel development.Citation132,Citation172,Citation173 On the other hand, OVs demonstrate immunotherapeutic activity as viral infection profoundly reshapes the immunosuppressive TME in a way that supports the reinstatement of cancer immunosurveillance.Citation47,Citation49,Citation64,Citation140,Citation143,Citation174,Citation175 First, virus-induced cell stress and death produce pathogen/microbe-associated molecular patterns (PAMPs/MAMPs), release damage-associated molecular patterns (DAMPs) and favor tumor antigen spreading.Citation23,Citation112,Citation128,Citation175Citation186 The immediate consequence of such pro-inflammatory environment consists of the recruitment of innate and adaptive actors of the intrinsically linked antiviral and antitumor immunities. Citation112,Citation128,Citation139,Citation167,Citation174,Citation177,Citation187Citation195 Such antitumor response plays a critical role in the overall efficacy of oncolytic virotherapy by eliminating spare malignant cells.Citation128,Citation189,Citation191,Citation196 Additionally, the establishment of a tumor-specific immune memory contributes to prevent cancer recurrence.Citation191,Citation196,Citation197 Several strategies have been designed to support cancer immunosurveillance upon OV treatment. Pharmacological immunomodulators or transgenes expressing either immunomodulatory proteins (e.g., chemokines, cytokines, co-stimulatory molecules, antibodies), or shRNAs silencing mRNAs of immunoinhibitory factors, have been recurrently combined with/inserted into OVs.Citation112,Citation119,Citation126,Citation132,Citation192,Citation194,Citation197Citation220 For instance, T-VEC overexpresses colony stimulating factor 2 (CSF2, best known as GM-CSF), which drives chemotaxis and activation of antigen-presenting cells.Citation41,Citation112 Alternatively, tumor-associated antigens or epitopes can be either coated on viral particulesCitation221 or overexpressed from OVs to exacerbate their cancer vaccine potential. Citation222Citation231 Like prophylactic vaccines, the later strategy (referred as an ‘oncolytic vaccine’) authorizes prime-boost immunization protocols to further expand the adaptive arm of the antitumor response.Citation222Citation224,Citation226,Citation230,Citation231 Finally, combinations of OV therapy with complementary immunotherapeutic approaches (e.g., ICIs or CAR T, DC and natural killer [NK] cell therapies), have demonstrated enhanced efficacy in preclinical models, and more recently into the clinic, and are actively investigated against multiple indications.Citation112,Citation128,Citation133,Citation147,Citation187,Citation188,Citation191,Citation197Citation199,Citation201,Citation203,Citation204,Citation206,Citation207,Citation232Citation244 Not surprisingly, combinatorial regimens involving ICIs are predominant and have already confirmed synergistic effect with several strains of OVs.Citation112,Citation128,Citation147,Citation187,Citation188,Citation191,Citation197Citation199,Citation239Citation245 Interestingly, OV therapy input can revert resistance to ICI treatment; a property that can be attributed, at least in part, to their ability to broaden neoantigenome-directed T-cell responses and to stimulate the expression of inhibitory checkpoints on both immune and tumor cells.Citation139,Citation188,Citation197,Citation239,Citation242,Citation246,Citation247

Here, we provide an update on the preclinical and clinical progress achieved in the field of oncolytic viro-immunotherapy since the publication of the latest Trial Watch dealing with this topic (December 2015).Citation40

Preclinical and translational advances

A plethora of preclinical and translational studies dealing with oncolytic viruses have been published during the past 2.5 years (source http://www.ncbi.nlm.nih.gov/pubmed). The following paragraphs provide a non-exhaustive compilation of recent original findings in the field.

Adenoviruses. (1) Eriksson et al. (Lokon Pharma AB, Uppsala, Sweden) introduced LOAd703, an oncolytic adenovirus (oAd) of chimeric serotype 5/35 armed with co-immunostimulatory CD40L and 4-1BBL. LOAd703 demonstrated oncolytic activity against pancreatic cell lines in vitro. In mice, LOAd703 given intratumorally (i.t.) was able to control the growth of a syngeneic pancreatic tumor model, particularly when combined with gemcitabine (an antimetabolite used as standard of care for pancreatic cancer patients).Citation248Citation251 LOAd703 infection was associated with DC maturation/activation which in turn increased the activation/expansion of NK and tumor-specific T cells. Following on these encouraging preclinical data, the safety and oncolytic viro-immunotherapeutic efficacy of LOAd703 will be investigated clinically against pancreatic malignancy (for details, see “Recently initiated clinical trials” below).Citation200,Citation252 (2) Delta-24-RGD, also referred to as DNX-2401 into the clinic (DNAtrix, Houston, TX, USA), is an oAd engineered to preferentially infect, replicate in and lyze tumor cells. Precisely, a RGD peptide motif has been introduced in the Ad fiber to favor interaction with tumor integrins and 24 base pairs have been deleted in the E1A gene to restrict replication to tumor cells with abnormality of the p16/RB/E2F pathway. DNX-2401 is currently evaluated in a Phase II clinical trial against various brain malignancies (see following sections on clinical trials).Citation97 In a preclinical study, i.t. Delta-24-RGD also demonstrated efficacy against human pancreatic tumor xenografts in mice. OV-mediated cytotoxicity was inducing phosphatidylserine exposure on the outer layer of the plasma membrane. Interestingly, the therapeutic efficacy was further enhanced when combining Delta-24-RGD to 1N11, a fully human phosphatidylserine antibody.Citation253 Following infection, several immune subsets involved in anticancer innate immunity (i.e., macrophages and NK cells) were infiltrating the tumor bed, particularly when the OV was accompanied with the 1N11-based immunotherapy. Among the mechanisms involved were cited the antibody-dependent cell-mediated cytotoxicity (ADCC) and an enhanced anticancer immune response, notably through an increased differentiation of pro-tumor M2 macrophages into anti-tumor M1 macrophages.Citation97 In this line, Delta-24-RGD was armed with the co-immunostimulator OX40L,Citation254,Citation255 thus forming Delta-24-RGDOX. In an orthotopic model of glioma in immunocompetent syngeneic mice, Delta-24-RGDOX appeared to be more efficient than its precursor at recruiting CD4+ and CD8+ T lymphocytes in the tumor bed and exhibited superior ability to activate tumor-specific CD8+ T cells. As a consequence, i.t. injection of Delta-24-RGDOX demonstrated a more potent immune-mediated antiglioma activity than Delta-24-RGD. Cured animals were also protected against tumor rechallenge thus validating the establishment of a protective immune memory. To further overcome the immune suppression mediated by PD-L1 expression at the surface of cancer cells, virotherapy was accompanied with a PD-L1 antibody. The combinatorial treatment showed synergistic inhibition of gliomas and significantly increased mouse survival. Altogether, these studies demonstrated that Delta-24-RGD and its armed derivatives could be promising agents for brain and pancreatic cancer therapy, particularly when combined with immunotherapies.Citation197 (3) VCN-01 (VCN Biosciences, Barcelona, Spain) is an engineered Ad5 with enhanced infectivity through RGD modification of the viral fiber, conditional-replication in retinoblastoma protein (Rb)-deficient cells through 24-bp deletion in the E1A gene and improved tissue distribution via transgene expression of the soluble hyaluronidase PH-20. It is currently undergoing Phase I clinical investigations against pancreatic cancer and retinoblastoma (see section “Recently initiated clinical trials”). Martinez-Velez and colleagues evaluated the efficacy of VCN-01 against relevant xenograft models of osteosarcoma in mice. The OV demonstrated remarkable anti-tumor activity against both orthotopic and lung metastatic osteosarcoma following i.t. or systemic delivery, respectively. These results motivated the authors to propel a Phase I/II study with VCN-01 in pediatric osteosarcoma.Citation146 (4) Two groups have armed oAds with bispecific T cell-engagers (BiTEs), some artificial proteins generated by the fusion of the single-chain variable fragments (scFv) of two different monoclonal antibodies, one binding to the T lymphocyte marker CD3 and the second one targeting a surface TAA.Citation256Citation259 By co-engaging T cell effectors and cancer cells, BiTEs can mediate immune-mediated tumor cell lysis. Fajardo and collaborators (Bellvitge Biomedical Research Institute/IDIBELL, L’Hospitalet de Llobregat, Spain) expressed a human epidermal growth factor receptor (EGFR)-targeting BiTE antibody from ICOVIR-15K, an oAd with RGD-modified fiber and truncated E1A protein.Citation260 The BiTE was potently secreted from ICOVIR-15K-cBiTE-infected cells and specifically bound to CD3+ and EGFR+ cells. Cocultures with human peripheral blood mononuclear cells (PBMCs) promoted the activation, proliferation and antitumor cytotoxicity of T cells. In immunodeficient mice bearing EGFR+ tumor xenografts and transplanted with human PBMCs, i.t. treatment with ICOVIR-15K-cBiTE increased the accumulation and persistence of tumor-infiltrating lymphocytes (TILs) and enhanced antitumor efficacy in comparison to the parental strain ICOVIR-15K.Citation260 OV-mediated expression of BiTEs also benefited to CAR T cell therapy. Interestingly, the combinatorial strategy was unlocking activation and proliferation of tumor-infiltrating CAR T cells in the absence of the targeted antigen on cancer cells and improved outcome in mice bearing tumor xenografts. By bypassing limitations of CAR T cell monotherapy, clinical evaluation of such combination is encouraged.Citation261 Similarly, Freedman and colleagues introduced an EpCAM-targeting BiTE into EnAdenotucirev (EnAd), a chimeric Ad3/11p group B adenovirus (PsiOxus Therapeutics, Abingdon, UK).Citation208 The efficacy of EnAd-CMV-EpCAMBiTE was assessed on pleural effusions and peritoneal malignant ascites from patients with multiple indications of advanced carcinoma. Despite significant levels of immunosuppressive IL-10, EnAd-CMV-EpCAMBiTE adjunction appeared much more potent than its unarmed counterpart at activating endogenous T cells and eliminating endogenous cancer cells.Citation208 Translation into the clinic of this novel therapeutic is under consideration. (5) Ad5 is currently the benchmark oAd. However, high seroprevalence in the human population and known hepatotoxicity limits its delivery to localized and injectable malignant lesions.Citation262,Citation263 In order to circumvent these downsides, Nguyen et al. (Translational Immunovirology and Biodefense Program, Rochester, MN, USA) introduced the lower seroprevalence Ad6. Following systemic injection, Ad6 appeared perfectly tolerated, without any signs of toxicity, and demonstrated efficacy against syngeneic and xenografted tumors in rodents.Citation59 (6) Villanueva and colleagues (IDIBAPS, Barcelona, Spain) developed a novel strategy to improve OV tumor-selectivity based on the recent discovery that mRNA translational control by cytoplasmic polyadenylation element (CPE)-binding proteins is reactivated in cancer. They inserted CPE regulatory sequences in the 3ʹ-untranslated region of the adenoviral gene E1A. Following systemic delivery in mice, the resulting AdCPE conserved full potency against tumor xenografts with strongly attenuated replication in normal tissues such as liver. This approach could be extended to other OV platforms.Citation83 (7) Capasso et al. (University of Helsinki, Helsinki, Finland) developed a versatile and rapid system to adsorb major histocompatibility complex class I (MHC-I)-restricted tumor peptides onto the virus surface to stimulate oAd treatment immunogenicity.Citation221 The peptide-coated oAd (PeptiCRAd) retained intact oncolytic activity and allowed cross-presentation of the exogenous epitope on MHC-I. When administered to mice bearing syngeneic melanoma, PeptiCRAd coated with melanoma-associated epitopes showed enhanced antitumor activity towards treated and distant untreated tumors.Citation221 This platform efficiently redirected the immunodominance of viral antigens towards tumor antigens, making PeptiCRAd a promising approach for clinical evaluation.Citation221 (8) Osteosarcoma is generally refractory to chemotherapy, hence of poor prognosis. Osaki and collaborators showed that OBP-301, a telomerase-specific oAd (also named Telomelysin, Oncolys BioPharma, Tokyo, Japan),Citation264 synergistically suppressed the viability of osteosarcoma cell line and xenograft tumor models in combination with chemotherapeutic agents (cisplatin and doxorubicin).Citation137 Mechanistically, OBP-301 infection was promoting the siRNA-mediated knockdown of anti-apoptotic myeloid cell leukemia 1 (MCL1), thus sensitizing cancer cells to chemotoxicity. OBP-301 would appear as a promising strategy to attenuate chemoresistance in osteosarcoma patients.Citation137 (9) Dey et al. (The Brain Tumor Center, The University of Chicago, Chicago, IL, USA) emphasized the ability of neural stem cells (NSCs) to cross the blood-brain barrier and reach orthotopic glioma tumors following non-invasive intranasal delivery. Once engineered to overexpress C-X-C Motif Chemokine Receptor 4 (CXCR4), then loaded with an oncolytic Ad, infected NSCs extended survival of experimental animals in the context of radiotherapy.Citation233 This approach deserves optimization for future applications against disorders of the central nervous system (CNS).

Flaviviruses. Two groups reported the oncolytic property of Zika virus (ZIKV), a neurotropic flavivirus. Zhu and colleagues (University of California School of Medicine, San Diego, CA, USA) showed that ZIKV infects and kills human glioblastoma stem-like cells (GSCs) at higher rates than differentiated glioma or normal neuronal cells. In vivo, intracranial injection of a mouse-adapted ZIKV substantially extended survival of immunocompetent mice bearing syngeneic orthotopic glioblastoma (GB).Citation50 Lately, Kaid et al. (University of São Paulo, São Paulo, SP, Brazil) validated the potent ZIKV oncolytic activity against orthotopic xenografts of human embryonic tumors of the CNS. Mechanistically, the sensitivity of CNS tumor cells to ZIKV was relying on the basal activation of the Wnt signaling, as the pharmacological inhibition or stimulation of this pathway was respectively reducing or enhancing viral replication and oncolysis.Citation265

Herpesviruses. (1) In the clinic, the efficacy of poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi) is limited to DNA repair-deficient cancers and still faces resistance.Citation266 Rabkin’s team (Harvard Medical School, Boston, MA, USA) showed that two strains of oncolytic HSV-1 (oHSV-1), namely G47Δ (γ34.5Δ, ICP47-Us11 promoter Δ, ICP6, LacZ+) and MG18L (Us3Δ, ICP6, LacZ+), sensitized patient-derived GSCs to PARPi, irrespectively of their resistance status.Citation267 The synergistic effect resulted from the oHSV-mediated proteasomal degradation of the recombinase Rad51, a key protein of the DNA damage response.Citation268 It translated into an increased DNA damage and cell death in vitro and in vivo. The interaction between the approved PARPi olaparib and oHSVs significantly extended median survival of mice bearing PARPi-sensitive or -resistant GSC-derived brain tumors compared to either agent alone.Citation267 (2) The same group showed in a clinically relevant murine model of GB that combined application of G47Δ-mIL12, a G47Δ oHSV-1 expressing murine IL-12, with checkpoint inhibitors (anti-CTLA-4, -PD-1, -PD-L1) overcomes the highly immunosuppressive TME and eradicates tumors.Citation269 Mechanistically, the combination treatment promoted macrophage influx and M1-like macrophage polarization together with an increase of the effector/regulatory T cell ratio.Citation269 (3) Han & colleagues (The Ohio State University, Columbus, OH, USA) demonstrated that the immunosuppressive cytokine TGF-β could compromise NK cell-mediated cytotoxicity against oHSV-infected GB multiforme. Consequently, administration of TGF-β prior to oHSV treatment improved therapeutic efficacy against both xenograft and syngeneic GB multiforme mouse models.Citation158 In contrast, in GB multiforme refractory to temozolomide (TMZ, a standard-of-care alkylating agent) and typically enriched in cancer stem-like cells, Esaki and colleagues (Department of Neurosurgery, Harvard Medical School, Boston, MA, USA) showed that inhibition of the TGF-β signaling pathway benefited to oHSV therapy. Systemic administration of inhibitors of the TGF-β receptor together with i.t. injection of oHSV cured 60% of mice bearing orthotopic grafts of patient-derived recurrent GB multiforme. Mechanistically, the combination caused blockade of the mitogen-activated protein kinase (MAPK) pathways, thus affecting malignant cell clonogenicity, together with a boost of OV replication.Citation235 Similarly, Hutzen and co-workers (The Ohio State University, Columbus, OH, USA) also reported a benefit of TGF-β inhibition to oHSV therapy in murine models of rhabdomyosarcoma.Citation210 (4) Chen et al. (College of Medicine, Colombus, OH, USA) engineered NK cells to express last generation EGFR-CAR and demonstrated their cytotoxic activity against breast cancer cell lines expressing EGFR. Both in vitro and in vivo in a relevant model of breast cancer brain metastases, i.t. inoculation of both EGFR-CAR NK cells and oHSV1 demonstrated superior antitumor effectiveness than either therapy alone.Citation237 (5) In human breast cancer cell lines, using a genome-wide shRNA library, Workenhe et al. (McMaster Immunology Research Centre, Hamilton, Canada) observed that depleting serine/arginine-rich splicing factor 2 (SRSF2), or controlling its phosphorylation thanks to DNA topoisomerase I inhibitors, could enhance the oncolytic activity of the HSV-1 KM-100 (KOS strain, ICP0nCitation212 VP16inCitation1814).Citation238,Citation270 These results were confirmed in vivo as the combination of the approved topoisomerase inhibitor irinotecanCitation271 with KM-100 led to a synergistic antitumor activity against syngeneic murine breast cancer.Citation270 (6) A study by Oldfield and co-workers (The Johns Hopkins University, Baltimore, MD, USA) evaluated the potential of synthetic genome assembly methods for genome-wide engineering of HSV-1.Citation272 The genome of wild-type KOS strain was cloned using yeast transformation- associated recombination. By using the overlapping sequences between the adjacent pieces, 11 fragments of the genome were assembled into a complete HSV-1 genome in yeast. The assembled genome was transferred into an Escherichia coli host for transfection into mammalian cells. This method of HSV-1 engineering allowed deletion of up to 5 combinatorial deletions of genes that encode virion structural proteins. This technology is a suitable platform to modify oncolytic HSV-1.Citation272 (7) To circumvent the obstacle of systemic delivery and enable access of the virus to metastases, Leoni and colleagues (University of Bologna, Bologna, Italy) infected mesenchymal stromal cells (MSCs) with HER2-retargeted oHSV. Following infusion of immunocompromised mice with oHSV-infected MSCs, the burden of xenografted models of ovarian cancer lung metastases and breast cancer brain metastases was significantly reduced.Citation78 Confirming the reliability of the approach, Du et al. (Harvard Medical School, Boston, MA, USA) efficiently treated syngeneic murine melanoma brain metastases with oHSV-armed MSCs. Moreover, combination with PD-L1 blockade increased tumor-infiltrating cytotoxic CD8+ T lymphocytes (CTLs) and profoundly extended median survival of treated animals.Citation147

Orthomyxoviruses. (1) Using Raf-BxB transgenic mice that spontaneously develop non-small cell lung cancer (NSCLC), Masemann and colleagues demonstrated that one single intranasal inoculation of 500 particles of influenza A virus (IAV) rapidly eliminated 70% of the initial tumor mass.Citation273 Interestingly, IAV infection caused a functional reversion of immunosuppressive tumor-associated lung macrophages into a M1-like pro-inflammatory phenotype that supports virus-induced oncolysis. This study demonstrated, for the first time in an immunocompetent in vivo model, that infection with oncolytic IAV reshapes the TME and instates an anti-NSCLC immunity.Citation273 (2) Recent works from Palese’s team (Icahn School of Medicine, Mount Sinai, NY, USA) showed the feasibility to engineer an IAV that expresses an anti-CTLA-4 antibody. This recombinant virus significantly slowed tumor growth and extended overall survival of mice bearing aggressive melanoma. Importantly, the OV-mediated production of this immune checkpoint antibody successfully delayed the growth of distant, untreated, tumors thus supporting the induction of an abscopal effect.Citation198

Paramyxoviruses. (1) Kazimirsky et al. (Mina & Everad Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, Israel) demonstrated that glioma cells and glioma stem cells were more sensitive to the oncolytic effect of Newcastle disease virus (NDV) when the virus was delivered in MSCs. The therapeutic benefit was relying on the release of TNF-related apoptosis-inducing ligand (TRAIL) from infected MSCs.Citation152 (2) Ammayappan et al. (Mayo Clinic, Rochester, MN, USA) developed a reverse genetics system from a major isolate of the Urabe strain mumps virus (MuV) stock that allowed the construction and production of several recombinant strains. These recombinant MuVs demonstrated oncolytic activity against tumor cell lines in vitro and some efficacy against a syngeneic mouse model of colon carcinoma.Citation57 (3) Chen and colleagues (Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China) demonstrated the therapeutic advantage of a local administration of a measle virus vaccine strain Edmonston (MV-Edm) in improving the adoptive transfer of CD8+ NKG2D+ cells in a hepatocellular carcinoma mouse model. Moreover, they showed that this combination could be further improved by the administration of fludarabine, an inhibitor of the therapy-induced immune suppressor enzyme IDO1.Citation232

Picornaviruses. (1) PVSRIPO (Duke University, Durham, NC, USA), an engineered poliovirus Sabin type 1, is being evaluated clinically against recurrent grade IV glioma (see sections on clinical trials below). Recent preclinical studies investigated its efficacy against xenografted and syngeneic subcutaneous models of melanoma, breast and prostate cancers in mice. Following i.t. injection, PVSRIPO infection was stimulating the expression of numerous pro-inflammatory cytokines and chemokines within the tumor bed, primed the infiltration of innate immune cells (i.e., neutrophils, DCs) and the subsequent activation and recruitment of T lymphocytes including anticancer effectors. Ultimately, PVSRIPO viro-immunotherapy significantly slowed tumor growth and prolonged mouse survival.Citation142,Citation274 (2) Bell and Pavenko (Mayo Clinic, Rochester, MN, USA) developed new hybrid strains of the picornavirus Theiler’s murine encephalomyelitis virus (TMEV). In vitro, they identified one virus named GD7-KS1 as a potent oncolyic agent against melanoma cell lines. In melanoma-bearing immunocompetent mice, repeated i.t. injections of GD7-KS1 stimulated a strong infiltration of tumor-specific CTLs, delayed tumor outgrowth and promoted survival extension.Citation58

Poxviruses. (1) Bartee and collaborators (Medical University of South Carolina, Charleston, SC, USA) developed an oncolytic myxomavirus (MYXV) that expresses a soluble PD-1 molecule allowing its secretion from infected cells. Interestingly, i.t. injection of the virus, referred to as vPD1, induced prolonged tumor-specific CD8+ T lymphocyte response and demonstrated superior anticancer efficacy than the combination of the unmodified MYXV with PD-1 antibodies administered systemically.Citation202 (2) Liu and colleagues (University of Pittsburgh School of Medicine, Pittsburgh, PA, USA) demonstrated in preclinical murine colon and ovarian cancer models that oncolytic vvDD (a Western Reserve [WR] VV with a double-deletion of the genes encoding the thymidine kinase [TK] and the vaccinia growth factor [VGF]) attracts effector T cells and induces PD-L1 expression on both cancer and immune cells in the tumor.Citation242 Interestingly, the combination of poxvirus plus ICI reduced the count of PD-L1+ cells and increased CD8+ and CD4+ T cell tumor infiltration as well as the expression of the activation/effector markers IFN-γ, Inducible T-cell COStimulator (ICOS), granzyme B and perforin. Overall, the treatment favored a sustained activation of effector T cells leading to extended survival.Citation242 In another study, a synergistic interaction between the ICIs anti-CTLA-4 or anti-PD-1 and the oncolytic VV-FCU1 has also been reported. VV-FCU1 (referred to as TG6002 into the clinic; Transgene, Illkirch-Graffenstaden, France) consists of a WR strain deleted of the TK and I4L/ribonucleotide reductase genes and armed with the fusion suicide gene FCU1 derived from the yeast cytosine deaminase (yCD) and uracil phosphoribosyltransferase genes. Noteworthy, the combination with ICIs was also accompanied with regressions of distant untreated tumors (abscopal effect). The VV-FCU1-based treatment was particularly beneficial against cancer cells deficient for the interferon-α/β receptor (IFNAR) signaling pathway.Citation128,Citation129 In the same way, Thorne’s group (University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA) had observed synergistic associations between the oncolytic vvDD and WR B18r−/- TK−/- and various immune checkpoint blockades (PD-1, PD-L1 or CTLA-4) against several preclinical murine tumor models. However, a particular concern was raised over the careful selection of the VV strain, antibody and timing of administration as some combinations actually produced antagonistic effects and loss of therapeutic activity.Citation275 Considering the variety of VV strains currently under clinical investigation (i.e., recombinant variants of WR, Lister, Copenhagen or Wyeth strains; for instance, see ) and the anticipated introduction of ICIs, critical attention will have to be paid on identifying the correct settings of OV/ICI association to achieve best efficacy. (3) Thorne’s group also identified myeloid-derived suppressor cells (MDSCs) as key mediators of resistance to immunotherapies, including OV therapy. Cumulative reports have identified cyclooxygenase (COX)-2-mediated production of prostaglandin E2 (PGE2) as a determinant of MDSC migration and maintenance of their immunosuppressive phenotype.Citation276Citation280 Therefore, their team aimed at targeting PGE2 by engineering the VV WR.TK strain to express the prostaglandin-inactivating enzyme 15-hydroxyprostaglandin dehydrogenase [NAD+] (HPGD). Treatment with the resultant VV WR.TK.HPGD+ was reducing MDSC tumor infiltration, enhanced the immune response within and against the tumor and re-sensitized resistant neoplasms not only to virotherapy but also to other immunotherapies such as adoptive T cell-based cancer treatment.Citation281 (4) To selectively target cancer cells, Potts et al. (Faculty of Medicine and Dentistry, University of Alberta, Edmontion, Canada) constructed a VV strain that is deleted for the F4L gene which encodes a ribonucleotide reductase; an enzyme essential for virus replication and complemented in malignant cells.Citation282 The F4L-deleted VV demonstrated its safety and efficiency against syngeneic rat and xenografted human orthotopic bladder tumor models. Importantly, this treatment led to the establishment of a protective immune memory since tumor rechallenge of cured rats failed to reinstate a neoplasm. Thus, F4L-deleted VV could constitute a promising alternative against bladder cancers, particularly in patients that fail responding to standard intravesical BCG immunotherapy.Citation34,Citation282 (5) Hirvinen and colleagues (Laboratory of ImmunoViroTherapy, Faculty of Phamacy, University of Helsinki, Finland) experienced an original approach that consisted in overexpressing an intracellular pattern recognition receptor, the DNA-dependent activator of IFN-regulatory factors (DAI), from vvDD with the aim of enhancing the production of type I IFNs and the stimulation of both innate and adaptive arms of antitumor immunity. Compared to VV alone, local administration of vvDD-DAI resulted in an increase of tumor-infiltrating CD8+ T-cells, which translated into better efficacy against melanoma in both syngeneic and humanized mouse models.Citation220 (6) In a model of syngeneic orthotopic glioma established from intracranial injection of brain tumor-initiating cells in mice, Pisklakova and colleagues (Moffitt Cancer Center, Tampa, FL, USA) demonstrated that removing the gene M011L, which encodes an anti-apoptotic Bcl2 homolog, from wild-type MYXV sensitizes brain tumor cells to virus-induced cell death and prolongs survival of tumor-bearing mice. Interestingly, the efficacy of vMyx-M011L-KO treatment was significantly enhanced in combination with the chemotherapeutic standard-of-care TMZ.Citation131 (7) Francis et al. (University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA) reported on a novel combination regimen to improve OV immunotherapeutic activity. In a syngeneic peritoneal carcinomatosis model, oncolysis was first induced by a vvDD expressing the chemokine CXCL11 administered through i.p. injection. Subsequently, a chemokine modulating drug cocktail consisting of IFNα, plus the TLR3 agonist polyI:C, plus the COX-2 inhibitor celecoxib, was administered repeatedly. The dual treatment promoted the trafficking and supported the activity of OV-induced tumor-targeting CTLs and NK cells within the TME. Upon combination therapy, median and long-term survivals dramatically extended thus encouraging further evaluations of the approach.Citation194 (8) In a model of spontaneous pancreatic neuroendocrine tumors, Kim et al. (University of California, San Francisco, USA) showed that systemic mouse-prototype VV JX-594 (best referred to as pexastimogene devacirepvec/Pexa-Vec/TG6006 into the clinic – see sections below on clinical trials) targets and disrupts tumor blood vessels, sequentially spreads to malignant cells, and produces widespread CD8+ T cell-dependent cancer cell killing in primary and metastatic lesions. Furthermore, these effects were amplified by concurrent or sequential administration of the multi-targeted receptor tyrosine kinase inhibitor (RTKi) Sunitinib.Citation167,Citation283

Table 1. Clinical trials recently initiated to investigate the safety and efficacy of oncolytic viruses in cancer patients.

Reoviruses. (1) In a window-of-opportunity clinical trial, Melcher’s group (University of Leeds, Leeds, UK) demonstrated that neoadjuvant reovirus successfully infected cancer cells within high-grade glioma and brain metastases following i.v. infusion in 9 patients.Citation188 OV administration enhanced infiltration of CTLs in the tumor bed and promoted the intra-tumoral expression of PD-L1 in a type I and II IFN-dependent manner. In mice affected with syngeneic brain tumors, sequential treatment using systemic reovirus followed by the blockade of the PD-1/PD-L1 axis improved survival. Altogether, these data support clinical investigation of viro-immunotherapeutic combinations against primary and disseminated neoplasms.Citation188 Additionally, the same team demonstrated that reovirus-induced proinflammatory antiviral immune responses can target hepatitis C virus (HCV)-associated hepatocellular carcinoma (HCC) and also hamper HCV replication.Citation284 (2) Rajani and collaborators (Mayo Clinic, Rochester, Minnesota, USA) demonstrated that the timely combination of reovirus with anti-PD-1 antibody also enhanced the therapeutic efficacy against subcutaneous B16 melanoma in mice. This study showed that checkpoint inhibition potentiated both NK cell-mediated tumor cell killing and the CD8+ T cell anti-tumor immune response while reducing regulatory T cell (Treg) activity.Citation285 (3) Ilett et al. (Mayo Clinic, Rochester, Minnesota, USA) utilized reovirus in a prime-boost vaccination approach in combination with anti-PD-1 therapy. Reovirus was applied as the immune primer and vesicular stomatitis viruses [VSV] expressing a cDNA library of melanoma antigens (VSV-ASMEL) were used as boosters. The regimen formulated an effective systemic anti-tumor immuno-virotherapy that significantly enhanced survival of B16 melanoma-bearing mice, with long-term cures.Citation223 (4) Recent discoveries highlighted the ability of histone deacetylase (HDAC) inhibition to upregulate the reovirus entry receptor and junctional adhesion molecule 1 (JAM-1), thus facilitating reovirus infection and tumor cell killing both in vitro and in myeloma-bearing nude mice.Citation157 Jaime-Ramirez et al. investigated the anti-tumor efficacy of the pan-HDAC inhibitor suberoylanilide hydroxamic acid (SAHA, also known as Vorinostat) in conjunction with REOLYSIN®, a proprietary reovirus serotype 3-Dearing strain (also referred to as Pelareorep; Oncolytics Biotech Inc., Calgary, AB, Canada), in head and neck carcinoma (HNC). This combination therapy stimulated tumor infiltration by multiple immune cells, including macrophages, NK cells and CD8+ T lymphocytes, and demonstrated improved anti-tumor efficacy, in comparison to either treatment alone, both against human tumor xenografts and syngeneic squamous tumors in mice.Citation156 (5) Finally, Sakurai et al. (Osaka University, Osaka, Japan) complexed reovirus with a cationic liposome transfection reagent and achieved enhanced tumor cell-killing in reovirus-resistant tumor cells by promoting endo-/lyso-somal escape of the virus.Citation286

Rhabdoviruses. (1) VSV-IFNβ-NIS (Mayo Clinic, Rochester, Minnesota, USA) is an oncolytic VSV that expresses IFN-β and the sodium/iodide symporter (NIS). IFN-β enhances VSV tumor-specificity by activating antiviral innate immunity in normal cells. Additionally, due to the enhanced iodine uptake by infected cells, administration of radioactive iodine Citation123I allows non-invasive imaging of OV spread across tumor lesions and can utilize radiotherapy to enhance cancer killing.Citation119 Currently, VSV-IFNβ-NIS is being evaluated clinically against multiple solid malignancies following intralesional administration (see sections on clinical trials below). To support the clinical testing of intravenous (i.v.) VSV-IFNβ-NIS against hematologic malignancies, preclinical studies have been engaged in mouse models. Systemic VSV-IFNβ-NIS was well tolerated up to high dose of 5 × 10Citation10 TCID50. As expected, sequestration of the OV by macrophages was observed in the spleen and liver. Dose-limiting toxicities (DLT) included hepatic toxicity, thrombocytopenia and lymphopenia. Infectious viral particles were only detected in the tumor.Citation287 VSV-IFNβ-NIS delivered i.v. demonstrated noticeable therapeutic efficacy, prolonging survival of mice bearing subcutaneous syngeneic plasmacytomas or human myeloma xenografts.Citation287 Antitumor activity was also demonstrated against acute myeloid leukemia (AML), ex vivo on mononuclear cells collected from affected patients and in vivo against a syngeneic murine AML. In the latter model, therapeutic efficacy was improved when VSV-IFNβ-NIS was combined to PD-L1 blockade.Citation288 Lately, Naik et al. also demonstrated the tolerability and safety of i.v. VSV-IFNβ-NIS for the treatment of spontaneous advanced or metastatic cancers in dogs.Citation119 Altogether, the safety profile in mice and canine patients motivated the translation of systemic VSV-IFNβ-NIS therapy into humans. (2) Kim and collaborators demonstrated that the second mitochondrial-derived activator of caspases (Smac)-mimetic compound LCL161 synergizes with the oncolytic VSVΔM51 allowing to cure mice bearing syngeneic breast tumors. Tumor regression was depending on CTLs. LCL161 contribution consisted in creating an immunosupportive TME through the polarization of tumor-associated macrophages toward M1-like phenotype and in rescuing CD8+ T-cell exhaustion within the TME. Moreover, the VSVΔM51 + SMC combination treatment was also synergizing with anti-PD-1 immunotherapy.Citation126 (3) Peritoneal carcinomatosis remains one of the most common causes of death from abdominal cancers. The Auer’s group reported that intraperitoneal (i.p.) injection of an infected cell vaccine (ICV), consisting of autologous tumor cells infected ex vivo with an oncolytic Maraba MG1 virus expressing IL-12, promotes the migration of activated natural killer (NK) cells to the peritoneal cavity. As a consequence, MG1-IL12-ICV reduced tumor burden and remarkably improved survival in a murine colon cancer model of peritoneal carcinomatosis.Citation289 (4) Le Boeuf and colleagues evaluated the efficacy of MG1 Maraba virus against sarcoma, a poor prognosis cancer with limited therapeutic options. In comparison to four other clinically relevant OVs, namely the mutant VSVΔM51, HSV-1 N212, vvDD and type-3 reovirus, MG1 demonstrated the strongest cytotoxicity in vitro against human and canine cell lines. Ex vivo, MG1 was able to infect and replicate in human explants of sarcoma originating from diverse histotypes. In vivo, treatment of murine syngeneic sarcoma significantly prolonged survival and allowed complete regression. Efficacy was relying on immune effector activity and generated memory that protected against cancer rechallenge. Altogether, these results highlight the therapeutic potential of Maraba MG1 for the care of sarcoma, as a standalone intervention or in a combinatorial regimen.Citation290 (5) Bourgeois-Daigneault et al. investigated the efficacy of Maraba MG1 in a neoadjuvant setting against triple-negative breast cancer (TNBC). In patient-derived tumor xenografts, MG1 demonstrated direct oncolysis of infected malignant cells and minimized the metastatic burden. In immunocompetent animals, neoadjuvant MG1 induced a potent antitumor immunity that actively participated in controlling/protecting against outgrowth of rechallenged breast tumors. By microarray, the authors characterized the pro-inflammatory signature of Maraba-infected TNBC human cell lines and patient-derived xenografts. The intracellular virus sensors MyD88 and RIG-I appeared determinant in stimulating the production of IFNs and of the chemokines responsible for driving Th1/Tc1 effector cell migration (e.g., CXCL9 to 11, CCL2,5). MG1-induced inflammation was accompanied by an overexpression of PD-L1 in tumor cells and an increase of tumor-infiltrating Tregs. Based on this observation, immune checkpoint therapy with anti-PD-1 plus anti-CTLA-4 was introduced post-surgery and translated into a significant extension of long-term survival. This work describes MG1 as a promising neoadjuvant treatment against TNBC for its dual ability to induce potent antitumor immunity and to sensitize the TME to immune checkpoint blockade.Citation187 (6) Atherton and colleagues developed a heterologous prime-boost vaccine strategy to treat neoplasm positive for the human papillomavirus (HPV) E6/E7 antigens. A CD8+ T cell antitumor response was primed through intramuscular injection of an Ad vaccine expressing E6/E7 then boosted through systemic delivery of a Maraba MG1-based oncolytic vaccine expressing the same tumor antigens. This approach generated a weak response against the HPV-E6 antigen but an exceptionally robust reactivity against the E7 protein. Such strong anticancer immunity translated into curing 75% of mice bearing HPV E6/E7-positive syngeneic tumors.Citation222 Alternatively, the team reached similar efficacy by priming the anti-E6/E7 adaptive immunity with E6/E7-derived synthetic long peptides rather than the Ad vaccine.Citation291,Citation292 Finally, the team extended this oncolytic immunotherapy to murine syngeneic prostate tumors using Ad and MG1 vaccines expressing the human six-transmembrane antigen of the prostate (STEAP) protein.Citation293 Therapeutic efficacy was remarkable. CTL responses against multiple STEAP epitopes were mounted and revealed a functional breach of tolerance. Activated TILs and MHC-I up-regulation were also detected.Citation293 Altogether, these preclinical studies provide rationale for the clinical evaluation of the Ad prime – MG1 boost oncolytic vaccination in patients affected with HPV-positive malignancies or against advanced prostate cancer.

Togaviruses. (1) Ramachandran et al. (Department of Immunology, Genetics and Pathology, Uppsala University, Sweden) successfully attenuated the neurovirulence of the Semliki Forest Virus-4 (SFV4). For this purpose, they inserted target sequences for miR124, miR125 and miR134 within the viral genome which prevented its replication within healthy cells of the central nervous system. The systemic delivery of the resulting SFV4miRT demonstrated a safety profile and potent oncolytic efficacy against experimental GB multiforme and neuroblastoma models. Thus, SFV4miRT constitutes a promising oncolytic candidate for the treatment of brain cancers.Citation82 (2) The alphavirus M1 is a Getah-like virus isolated from China whose natural oncolytic property has been reported in 2014. M1 preferentially kills cancer cells that are deficient in zinc-finger antiviral protein (ZAP).Citation62 Several compounds have since been described as potentiators of M1 oncolysis against human xenograft models in mice (e.g., CRC, HCC), including activators of the cyclic adenosine monophosphate (cAMP) signaling, an inhibitor of the valosin-containing protein (VCP) or else Smac mimetics, which each potentiates virus replication, induces prolonged endoplasmic reticulum stress and ultimately stimulates M1-induced apoptosis.Citation294Citation296 In non-human primates, Zhang and colleagues (Sun Yat-sen University, Guangzhou, China) validated the safety of repeated i.v. administrations of M1 opening the way to its clinical evaluation against malignant indications.Citation297

Completed and advanced clinical studies

Since the submission of our latest Trial Watch dealing with OVs (December 2015),Citation40 preliminary or definitive results from about 50 clinical trials testing this immunotherapeutic approach in cancer patients have been published in the peer-reviewed scientific literature (source http://www.ncbi.nlm.nih.gov/pubmed) or presented at the 2016–2018 meetings of the American Society of Clinical Oncology (ASCO) or the American Association for Cancer Research (AACR) (sources https://meetinglibrary.asco.org/ and http://aacrjournals.org/site/Meetings/, respectively).

Adenoviruses. (1) Alonso and colleagues evaluated the safety and efficacy of DNX-2401 into the clinic (DNAtrix, Houston, TX, USA), in 31 patients with glioma (NCT01956734).Citation298 All subjects were recruited upon first recurrence after ongoing surgery and standard care with radiotherapy and TMZ. DNX-2401 was injected into the brain parenchyma followed by several cycles of TMZ. The therapeutic regimen was well tolerated with no severe adverse events (AEs) associated with the virus observed. At the time of the report, objective responses (OR) were witnessed in 3 patients alive 30, 27 and 19 months post-combination therapy. Interestingly, the expression level of fibroblast growth factor 2 (FGF-2) in glioma biopsies appeared negatively correlating with IFN-γ production and positively correlating with overall survival (OS).Citation298 Of note, a complex interplay between FGF-2 and type I & III IFNs was uncovered the past years, with apparent antagonistic functions on cell proliferation, differentiation and antiviral response.Citation299Citation302 Along this line, FGF-2, which is mainly produced by cancer-associated fibroblasts in the TME, has been shown to enhance the susceptibility of malignant cells to infection and replication by multiple RNA and DNA oncolytic viruses.Citation299 Further investigations are encouraged to validate FGF-2 expression level as a prognostic biomarker for oncolytic virotherapy. (2) Ranki T et al. assessed the safety profile of ONCOS-102 (Targovax, Oslo, Norway), previously called CGTG-102 or Ad5/3-D24-GMCSF, in 12 patients affected with various solid tumors that failed standard therapeutic attempts (NCT01598129).Citation303 ONCOS-102 is an oncolytic Ad5/3 with a 24-bp deletion in the E1A gene and expressing GM-CSF. ONCOS-102 was injected topically following a 3 + 3 dose escalation design together with daily low-dose CPA to deplete immunosuppressive CD4+ Tregs. The study did not reveal DLT or maximum tolerated dose (MTD). Forty percent of evaluable patients demonstrated stable disease (SD) at 3 months and median OS was 9.3 months. An acute increase in systemic pro-inflammatory cytokines/chemokines together with a marked increase of TILs was observed post-treatment in 11 out of the 12 patients. In 2 patients affected with pleural mesothelioma or ovarian cancer, tumor-infiltration of CD8+ T cells was concomitant with the detection of circulating tumor-specific CD8+ T cells targeting cancer-testis (CT) antigens such as melanoma antigen family A3 (MAGE-A3). Finally, ONCOS-102 was stimulating the expression of PD-L1, thus encouraging further combination with ICBs.Citation303 Along this line, one Stage IV treatment-refractory ovarian cancer patient was introduced to a FDA-approved compassionate program involving ONCOS-102. The protocol incorporates ONCOS-102 in a multi-therapeutic regimen with systemic administration of low-dose CPA together with dual ICB treatment with ipilimumab and nivolumab. In this patient, the ONCOS-102-containing therapeutic regimen appeared well tolerated with no virus-related AEs observed.Citation304 A similar combinatorial approach is being evaluated clinically against melanoma (see “Recently initiated clinical trials” below). (3) McNeish and colleagues are evaluating EnAd (PsiOxus Therapeutics, Abingdon, UK) in platinum-resistant epithelial ovarian cancers (NCT02028117). EnAd is injected i.p. either as a monotherapy or in association with paclitaxel. Preliminary analyses indicate that the virus is reaching and replicating in the tumor with limited systemic exposure. Safety profile was acceptable at the dose tested. A dose escalation protocol will determine the MTD to be evaluated in Phase II.Citation305 (4) Hemminki’s group (University of Helsinki, Helsinki, Finland) performed a retrospective analysis which incorporated clinical data from 290 patients into a Cox regression model to estimate prognostic factors significant in the context of oncolytic Ad therapy.Citation306 Subjects had solid tumors refractory to standard treatment prior enrollment in the Advanced Therapy Access Program (ATAP, a personalized therapy program ongoing 2007–2012)Citation307 where they received one oncolytic Ad, among Ad5-d24-GMCSF, Ad5/3-d24-GMCSF, Ad5-RGD-d24-GMCSF, Ad5/3-E2F-d24-GMCSF, Ad3-hTERT-E1, Ad5-d24-RGD, ICOVIR-7, Ad5/3-Cox2L-d24, Ad5/3-hTERT-CD40L, Ad5/3-d24-hNIS. Results highlighted that 1) absence of liver metastases correlated with an improved rate of disease control, 2) patients treated with viruses coding for GM-CSF tended to have better prognosis, as well as 3) women, regardless of the cancer type, 4) participants administered via i.p. injection were more likely to achieve disease control and 5) subjects with low neutrophil/lymphocyte ratio prior oAd therapy had significantly longer OS.Citation306 To uncover potential prognostic biomarkers, an additional study monitored mRNA and/or protein levels in tumor biopsies or fluid samples from cancer patients treated with oAd.Citation130 Significant modulations in several signaling pathways and genes associated with innate immunity appeared to be associated with poor prognosis. Conversely, lack of chronic innate inflammation at baseline may predict improved oAd therapy outcome.Citation130 Finally, the team provided preclinical evidences that low-dose CPA increases the efficacy of Ad5/3-D24-GMCSF against a TNBC model.Citation215 In the ATAP program, Ad5/3-D24-GMCSF treatment was administered to 16 patients with advanced breast cancer refractory to previous therapies; 13 of which received concomitant CPA including 3 TNBC. The combination treatment appeared safe and well-tolerated. Thirteen patients were evaluable according to modified Response Evaluation Criteria in Solid Tumours (RECIST) 1.1 criteria: 1 patient had a minor response, 2 had SD and 10 had progressive disease (PD). One patient was still alive at the end of the follow-up, 1771 days post-treatment. These preliminary data encourage further development of oAd against breast cancer, including TNBC.Citation215 (5) Ramirez et al. (Hospital Infantil Universitario Niño Jesús, Madrid, Spain) reported on the results of the first-in-man trial using Celyvir, a therapy consisting of autologous MSCs infected with the oAd ICOVIR5 (NCT01844661). Celyvir was well tolerated in the 9 patients affected with relapsed/refractory pediatric solid tumors that completed the treatment. Ad replication and antibodies were detected in 7 and 8 subjects, respectively. Rise in circulating effector CD8+ T cells was observed. Two patients with neuroblastoma showed SD per RECIST, one of them continued on treatment for up to 6 additional weeks. Further evaluation in Phase II is warranted.Citation308

Herpesviruses. (1) T-VEC (Amgen, Thousand Oaks, CA, USA) has been approved by the FDA for the treatment of melanoma.Citation41,Citation44Citation46 A randomized, open-label, Phase II study evaluated neoadjuvant T-VEC + surgery (Arm 1, n = 76) versus surgery alone (Arm 2, n = 74) for resectable stage IIIB-IVM1a melanoma (NCT02211131).Citation309 At the interim analysis, the pathological complete response [CR] rate was 21% in patients of the Arm 1 that underwent scheduled surgery (15.8% when including the 19 patients that finally did not benefit from resection). Negative margin resection (R0) rates were 56.1% for Arm 1 and 40.6% for Arm 2. No unexpected toxicities were noticed. After 12 weeks of neoadjuvant T-VEC, the pathological CR rate was higher than observed by ORs (14.7%) and may account for the higher R0 margin in Arm 1. The primary analysis of relapse-free survival is ongoing.Citation309 Additional clinical trials investigated the efficacy of T-VEC along with the ICIs ipilimumab or pembrolizumab.Citation239,Citation310Citation313 An initial multi-center open label Phase I study evaluated the combined application of T-VEC and ipilimumab in 19 patients harbouring a Stage IIIB-IVM1c melanoma that is not suitable for surgical resection but injectable (NCT01740297). The OV was injected directly into cutaneous, subcutaneous or nodal lesions that have not received systemic therapy, except a prior adjuvant therapy.Citation313 Dose escalation studies in the trial showed that the two monotherapies (either T-VEC or ipilimumab) can be administered at their full therapeutic doses without DLTs. Reported severe AEs consisted of Grade 3 nausea and a Grade 4 elevation of the lipase and amylase levels attributed to ipilimumab. Overall, the study concluded that T-VEC + ipilimumab combination was tolerable without unexpected AEs.Citation313 Subsequently, the efficacy of intralesional administration of T-VEC + ipilimumab versus ipilimumab alone (n ~ 100 patients per arm) has been evaluated in a Phase II trial in patients with advanced unresectable melanoma of stages IIIB to IV, with no more than one prior therapy if BRAF wild type, no more than two prior therapies if BRAF mutant, and that present measurable/injectable lesions and no symptomatic autoimmunity or clinically significant immunosuppression.Citation311 The primary end point was objective response rate (ORR) evaluated by investigators per immune-related (ir) response criteria (irRC). ORR was significantly higher for T-VEC + ipilimumab versus ipilimumab alone with 39% (38 patients) in the combination arm and 18% (18 patients) in the ipilimumab arm.Citation311 Responses were not limited to injected sites as decreases of visceral lesions were observed in 52% of patients in the combination arm and 23% of patients in the ipilimumab arm.Citation311 Finally, a Phase Ib clinical trial evaluated the ability of T-VEC to allow infiltration of CTLs within the tumor and its potential for synergy with pembrolizumab (NCT02263508).Citation239 Twenty-one patients harbouring advanced melanoma have been injected with T-VEC i.t. followed by systemic administration of pembrolizumab with baseline and on-therapy biopsies.Citation239 Seven of the 21 patients (33%) had received prior anticancer therapy (including adjuvant therapy) and four (19%) had received prior radiotherapy. Patient had a median follow-up time of 18.6 months. Overall, there was no DLT and serious AEs were solely related to pembrolizumab and included autoimmune hepatitis, meningitis and pneumonitis. Confirmed OR and CR rates were 62% and 33%, respectively.Citation239 Overall, the responses occurred across all substages of melanoma. Median progression-free survival [PFS] and OS were not reached at the time of the last follow-up. A >50% reduction of the lesions has been observed with the combination treatment in: 82% of the injected non-visceral lesions, 43% of non-injected non-visceral ones, and 33% of non-injected visceral ones.Citation239 Patients who responded to combination therapy had increased CD8+ T cells, elevated PD-L1 protein expression and IFN-γ gene expression on several tumor-infiltrating cell subsets following T-VEC administration. Response to combination therapy did not seem associated with baseline CD8+ T cell infiltration or baseline IFN-γ signature.Citation239 In summary, this study showed that i.t. injection of T-VEC modifies the TME by attracting T cells and induces a systemic response in distant metastases after subsequent blockade of PD-1 with pembrolizumab (see also: Citation314Citation316). On top of melanoma, intralesional T-VEC is being evaluated in a multicenter Phase Ib study against recurrent or metastatic HNC in combination with pembrolizumab (NCT02626000/MASTERKEY-232). Preliminary results have been disclosed at the 2018 ASCO meeting.Citation317 Thirty-six patients were treated. Overall, 66.7% participants had serious treatment-emergent AEs, among which pyrexia, dyspnea and fatigue were the most common. One DLT (fatal arterial hemorrhage) was reported. Seven deaths were recorded during the study, 1 of which was related to T-VEC (the DLT) and none to pembrolizumab. The ORR was 16.7% and the disease control rate (OR/SD) was 38.9%.Citation317 Preliminary results have also been revealed regarding the Phase II evaluation of T-VEC combined to pembrolizumab against metastatic and/or locally advanced sarcoma (NCT03069378).Citation318 Twenty patients were enrolled. Grade 3 treatment-related AEs occurred in 2 participants (10%): fever from T-VEC and one case of pneumonitis from pembrolizumab that led to treatment abortion. Among 19 evaluable patients, 4 PR (21%), 9 SD (47%) and 6 progressions (32%) were observed by RECIST 1.1.Citation318 (2) A Phase Ib study is evaluating the safety and efficacy of OrienX010, a recombinant CL1 strain of HSV-1 (ICP34.5−/- ICP47 ICP6) expressing human GM-CSF, in unresected Stage IIIC to IV acral melanoma, a main subtype of melanoma in Chinese patients.Citation319 Twelve patients received on average 10 intra-lesional injections. Overall, the OV was tolerated with no DLT. Grade 1/2 AEs included pyrexia, injection site pain, leucopenia, rash and nausea. ORR was 16.7% (2 partial responses [PR]) and SD rate was 41.7%.Citation319 Time to response was 6–12 weeks, median PFS of 12 weeks and duration of response was 24 weeks. OS was not reached. A related Phase Ic trial is testing the safety and efficacy of intralesional administration of OrienX010 in liver metastases of Stage IV melanoma patients that have undergone standard therapies (NCT03048253, ).Citation320 The OV was tolerated with Grade 1/2 AEs that included pyrexia, fatigue, injection site pain, nausea/vomiting, hepatotoxicity and leucopenia. ORR was 8.3% (1 PR), disease control rate was 41.7% (1 PR, 4 SD).Citation320 OS was not reached. (3) Two Phase I clinical trials have been registered by the University of Alabama at Birmingham (Birmingham, AL, USA) in the past years to assess the safety and tolerability of two genetically-engineered HSV-1 strain F against brain tumors: G207 (ICP34.5−/- ICP6 LacZ+) in children (NCT02457845) and M032 (ICP34.5−/- human IL-12+) in adults (NCT02062827).Citation321,Citation322 If no results have been disclosed yet, related reports have been published. In Phase I trials (including NCT00157703 and NCT00028158), Waters and colleagues validated the safety of G207 in adults suffering from recurrent/progressive high-grade gliomas.Citation321 No DLTs were seen in the patients and no MTD was reached. Approximately half of the 35 treated adults had radiographic or neuropathologic evidence of response at a minimum of one time point. These clinical observations support the current trial involving i.t. inoculation of G207 in pediatric recurrent or progressive supratentorial malignant tumors (NCT02457845). Additionally, Patel at al. validated the safety of intracerebral M032 in 30 HSV-hypersensitive non-human primates at a per-kilogram dose which far exceeds that intended to be injected in patients with recurrent and progressive GB multiforme, anaplastic astrocytoma, or gliosarcoma.Citation322 Finally, Ren and collaborators (Duke University Medical Center, Durham, NC, USA) evaluated the combination of local delivery of OrienX010 with adoptive immunotherapy based on the infusion of autologous dendritic cells and cytokine-induced killer cells (DC-CIK).Citation206 Nine patients with advanced malignancy were treated with OrienX010, of whom seven experienced SD. Five of the OV-treated patients underwent DC-CIK adoptive transfer, and two had SD, whereas three progressed. Overall, this treatment was well tolerated and long-term event-free and overall survivals were observed. Moreover, the combination allowed further systemic expansion of multiple T cell clone populations than with OV therapy alone.Citation206 (4) Steby et al. (The Ohio State University, Columbus, OH, USA) validated the safety and tolerability of i.t. HSV1716 in 9 pediatric patients with relapsed or refractory extracranial cancers.Citation323 Detection of HSV-1 genome in the blood of 6 patients and increases in metabolic activity on 18fluorine-deoxyglucose PET in 2 patients were consistent with de novo virus replication and inflammation, respectively. Such observations are encouraging further clinical investigations.Citation323 (5) Finally, two interesting clinical investigations have exploited oHSV-1 as a cancer diagnostic tool based on their oncotropism.Citation324,Citation325 One clinical study involved NV1066, a replication-competent HSV-1 F strain with single-copy deletions of ICP0, ICP4 and ICP34.5 and which expresses the enhanced green fluorescent protein (GFP). The study evaluated NV1066 for the rapid detection of peritoneal dissemination of pancreatic cancer from peritoneal washings of 96 patients undergoing diagnostic laparoscopy for presumed or biopsy-proven pancreatic cancer. Cytology based on peritoneal fluid is the gold standard test owing to its clinical relevance and high specificity. In this trial, cytology assay detected any metastasis and peritoneal metastasis at rates of 44% and 89%, respectively. In contrast, HSV-GFP-based diagnosis of any metastasis or peritoneal metastasis demonstrated superior sensitivities with 94% and 100% detection rate, respectively.Citation324 The second clinical study involved a telomerase-specific replication-selective oHSV-1 that expresses GFP for the detection of circulating tumor cells (CTCs).Citation325 CTCs were detected in 59–100% of 326 blood samples from subjects affected with 6 different solid cancers (lung, colon, liver, gastric or pancreatic cancer and glioma) and lymphomas. Interestingly, CTC-positive rates increased remarkably with tumor progression in each of 5 tested cancers and decreased in response to the treatment. In hematological malignancies, the monitoring of CTC values provided an efficient treatment response indicator. Compared to CellSearch (a commercialized test for detecting CTCs of epithelial origin), the oHSV-1-based method demonstrated higher sensitivity in 40 NSCLCs, regardless of the stage, and was less affected by chemotherapy.Citation325 Further investigations are encouraged to validate OV-based diagnostic assays as a new gold standard for the detection of advanced cancers. (6) In a single-arm Phase I dose-escalation trial (UMIN-CTR UMIN000010150), 10 patients affected with unresectable locally advanced pancreatic cancer received i.t. the HSV-1 TBI-1401, best referred to as HF10 (Takara Bio, Kusatsu, Japan), together with the RTKi erlotinib and gemcitabine.Citation326 The combination treatment was safe. Out of 9 subjects who completed the therapy, tumor responses were 3 PR, 4 SD and 2 PD. Two subjects ultimately achieved surgical CR. The median PFS and OS were of 6.3 and 15.5 months, respectively. The association of oncolytic HF10 with chemotherapy will deserve further prospective evaluations.Citation326 In a Phase II trial, HF10 has been evaluated in combination with ipilimumab in unresectable/unresected Stage IIIB-IV melanoma patients (NCT02272855).Citation327 HF10 AE profile appeared similar in combination with ipilimumab as in monotherapy. Twenty-eight percent of the patients had treatment-related ≥ Grade 3 AEs; the majority due to ipilimumab. Of the 44 evaluable participants, irRC best ORR at 24 weeks was 41% (18% irCR and 23% irPR); SD rate was 68% (27% irSD). As of February 2018, median PFS and OS were 19 and 26 months, respectively. Responding tumors exhibited an activation of the adaptive immune response with an increase of total TILs and CD8+ T-cells together with a decrease of CD4+ T-cells.Citation327

Paramyxoviruses. MV-NIS (Vyriad, Rochester, MN, USA) is an attenuated oncolytic Edmonston strain of measles virus that expresses the sodium/iodide symporter (NIS). In a Phase I trial (NCT00450814), Dispenzieri et al. (Department of Hematology, Mayo Clinic, Rochester, MN, USA) demonstrated that systemic administration of MV-NIS, either alone (cohort 1) or following CPA treatment (cohort 2), was reasonably well tolerated in 29 patients with relapsed or refractory multiple myeloma.Citation328 Some severe AEs possibly related to therapy were reported in both cohorts at all dose levels and included leukocyte count decreased (n = 5), neutropenia (n = 9), thrombocytopenia (n = 2) or CD4+ T lymphocytes decreased, anemia and lymphopenia (n = 1 each). Citation123I SPECT/CT scan of malignant lesions and MV genome detection by RT-qPCR from gargle specimens, blood and urine body fluids up to one month post-infusion validated the ability of the virus to reach and amplify in the disseminated tumors. One patient achieved a CR and transient drops in serum free light chains (markers of disease progression) were seen in other patients. No DLT were observed and Phase II dose was determined.Citation328

Parvoviruses. Systemic or local treatment with ParvOryx (ORYX GmbH & Co. KG, Baldham, Germany), a wild-type rat H-1 parvovirus (H-1PV), has been investigated in 18 patients with recurrent GB in a first Phase I/IIa clinical trial (NCT01301430/ParvOryx01). ParvOryx was administered before and after surgical resection. ParvOryx treatment appeared safe and well tolerated with no MTD reached. The OV succeeded to cross the blood-brain/tumor barrier and spread widely through the tumor. Markers of microglia/macrophage activation and CTL infiltration were detected in infected tumors, suggesting that ParvOryx may trigger an immunogenic stimulus with immunotherapeutic potential. Median survival was extended in comparison with recent meta-analyses. Based on these encouraging data, further clinical development of ParvOryx will follow (see section on “Recently initiated clinical tirals” below).Citation329

Picornaviruses. (1) CAVATAK (Viralytics, Sydney, Australia), a proprietary formulation of the common cold coxsackievirus type A21 (CVA21), is being investigated against advanced malignancies in several clinical studies.Citation330Citation334 The completed Phase II CALM trial (NCT01227551) demonstrated the tolerability and efficacy of intralesional CVA21 monotherapy in 57 patients affected by late-stage melanoma.Citation334,Citation335 The ORR assessed by irRECIST was 28.1% with a ≥ 6 months durable response rate (DRR) of 21.1%. Tumor responses were observed in both injected and non-injected, local and distant, lesions suggesting the generation of systemic antitumor immunity.Citation334,Citation335 A CALM extension study was conducted in 13 patients and investigated CVA21-induced changes in immune cell infiltrates within the TME. In responders, increases in CD8+ TILs and in the expression of PD-L1 were observed in injected lesions. Reconstitution of immune cell infiltrates was reported in some CVA21-treated lesions from patients that failed prior therapy with immune checkpoint blockers.Citation334 Along this line, the Phase Ib STORM trial (NCT02043665) was designed to evaluate the safety and efficacy of CVA21 administered systemically either as a standalone therapy (part A, n = 16) or in combination with the anti-PD-1 pembrolizumab (part B, still recruiting) against late-stage solid tumors.Citation330Citation332 Each arm includes 3 cohorts in a dose escalation design. In part A, no severe AEs were observed. Tumor targeting was confirmed in biopsies of melanoma, non-small cell lung cancer (NSCLC) and bladder cancer subjects. Thirteen patients were eligible for response assessment and demonstrated 8 SD, 1 PR and 4 PD. Out of the 6 patients enrolled in part B so far (cohorts 1 and 2 with the lowest doses), one Grade 3 virus-related hyponatremia with no DLT for the combination treatment was reported. Generally, systemic delivery of CVA21 appeared well tolerated and able to reach metastatic sites.Citation330Citation332 The latter observation is encouraging for a synergistic effect with pembrolizumab considering the ability of CVA21 tumor infection to restore immune cell infiltration and upregulate the expression of immune checkpoint molecules.Citation334 Finally, the Phase I/II CANON trial (NCT02316171) is investigating the safety and efficacy of escalating doses of neoadjuvant CVA21, delivered i.v. in 16 first-line patients with bladder cancer, either alone or in combination with low-dose mitomycin C.Citation333 Observations indicate general tolerance of i.v. CVA21. Evidences of complete tumor response, viral replication and signs of virus-induced tumor inflammation demonstrated clinical activity of CVA21, likely associated with the induction of local and systemic anti-tumor immunity.Citation333 (2) Two case reports suggested that oncolytic virotherapy with Rigvir, a wild-type enterovirus ECHO-7 (Riga Eastern Clinical University Hospital, Riga, Latvia), could be applied for long-term treatment of patients with melanoma stage IV M1c (n = 2), small cell lung cancer stage IIIA (n = 1), and histiocytic sarcoma stage IV (n = 1). Prospective clinical studies are encouraged.Citation336,Citation337

Poxviruses. (1) Genelux (San Diego, CA, USA) developed a portfolio based on replicating VV Lister backbone flexible to express imaging genes, anti-tumor therapy genes and immune-modulator genes. Their lead candidate consists of GL-ONC1 (formerly named GLV-1h68), a genetically-engineered oncolytic VV encoding a Renilla luciferase-Aequorea GFP fusion protein as well as the β-galactosidase and β-glucuronidase. A Phase I clinical trial involving GL-ONC1 has been completed in 2015 and the results published recently (NCT01584284).Citation338 Primary outcome of the study was to determine the safety of GL-ONC1 when delivered i.v. with chemoradiotherapy to patients with HNC. An escalating dose of GL-ONC1 was given to 19 subjects with locoregionally advanced unresected, non-metastatic HNC. The MTD was not reached. Grade 3 hypotension, mucositis, nausea and vomiting were observed. Overall, systemic delivery of GL-ONC1 was considered safe and feasible in patients with locoregionally advanced HNC undergoing standard chemoradiotherapy. Interestingly, in several patients, viral presence in tumor was confirmed. With a median follow-up of 30 months, 1-year (2-year) PFS and OS were 74.4% (64.1%) and 84.6% (69.2%), respectively.Citation338 In a Phase Ib trial, 11 women with platinum-resistant/refractory recurrent ovarian cancer received repeated i.p. infusions of GL-ONC1 monotherapy (NCT02759588). Promising safety data, anti-tumor activity and immune activation mechanisms were documented. Disease control (PR+ CR≥ 15 weeks) was observed in 55% of patients. Extended PFS of 23, 35, 59 & 71 weeks were observed in 4 participants.Citation339 (2) Downs-Canner and collaborators (University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA) published the results of a Phase I trial assessing the safety of systemic administration of vvDD in cancer patients (NCT00574977). The OV was infused in 11 patients with standard treatment-refractory advanced colorectal cancer (CRC) or other solid cancers. No DLT and severe AEs were observed. Interestingly, in two patients, prolonged virus replication was detected in malignant tissues but not in normal ones, with the exception of a healed injury site and an oral thrush. One patient showed a mixed response on PET-CT with resolution of some liver metastases and another patient with cutaneous melanoma demonstrated clinical regression of some lesions.Citation340

Reoviruses. REOLYSIN® (Oncolytics Biotech Inc., Calgary, AB, Canada) has been investigated following i.v. delivery in combination with paclitaxel and/or carboplatinCitation341 in several Phase II trials: a) in 36 evaluable patients with metastatic pancreatic adenocarcinoma (NCT01280058),Citation342,Citation343 b) in 14 subjects with advanced malignant melanoma (NCT00984464),Citation344 c) in 37 patients with NSCLC (NCT00861627),Citation345 d) in 52 women with recurrent ovarian, tubal, or peritoneal cancer (NCT01199263),Citation346 and e) in 36 subjects with metastatic breast cancer (NCT01656538).Citation347 Furthermore, REOLYSIN has been administered in combination with pemetrexed (a folate antimetabolite)Citation348 or docetaxel (an antimitotic drug that inhibits microtubule depolymerization)Citation349 in 152 randomized advanced stage refractory platinum doublet NSCLC participants (NCT01708993, Phase II)Citation350 and in 85 randomized metastatic castration resistant prostate cancer patients (NCT01619813/IND.209, Phase II). Citation351 Also, REOLYSIN has been combined with gemcitabine in 34 patients with advanced pancreatic ductal adenocarcinoma (NCT00998322, Phase II).Citation352 Lastly, it has been examined in combination with monoclonal antibodies: i) pembrolizumab plus either 5-fluorouracilCitation353 (5-FU; n = 3), gemcitabine (n = 6), or irinotecan (n = 2) in 11 patients with relapsed metastatic adenocarcinoma of the pancreas (NCT02620423, Phase I)Citation354 and ii) FOLFOX6/bevacizumab for 51 patients with metastatic CRC (NCT01622543, Phase II).Citation355 Of the abovementioned completedCitation344,Citation345,Citation350Citation352 or activeCitation342,Citation343,Citation346,Citation347,Citation354,Citation355 clinical trials, REOLYSIN was well tolerated with no major signs of toxicity/AEs for combinations with paclitaxel and/or carboplatin, pemetrexed, docetaxel, or with pembrolizumab and 5-FU, gemcitabine, and irinotecan combinations. Nonetheless, AEs can arise with REOLYSIN® combination therapy, as was observed in its combination regimen with FOLFOX6/bevacizumab, showing increased toxicity and decreased quality of life in metastatic CRC patients.Citation355 Regarding efficacy, REOLYSIN did not prove superiority in comparison to chemotherapy alone when combined with carboplatin/paclitaxel neither in metastatic pancreatic cancer (NCT01280058),Citation343 nor in recurrent ovarian, tubal, or peritoneal cancer (NCT01199263).Citation346 Detrimental consequences of the therapeutic association were even reported on OS or PFS with docetaxel in advanced castration resistant prostate cancer (NCT01619813/IND.209)Citation351 and with FOLFOX6/bevacizumab in metastatic CRC (NCT01622543/IND.210).Citation355 Mitigated results were reported in patients with advanced NSCLC. Indeed, no significant benefit of salvage REOLYSIN plus docetaxel/pemetrexed arms over standard salvage chemotherapy arms was observed. Nevertheless, PFS tended to improve in the female gender and in the subgroups with epidermal growth factor receptor (EGFR)-mutated and p53-mutated status (NCT01708993/IND.211).Citation350 In this line, in patients with advanced EGFR-mutated/amplified or KRAS-mutated NSCLC, an ORR of 31% (11 PR, 20 SD) suggested a benefit of the reovirus in combination with paclitaxel and carboplatin. The median PFS, median OS and 12-month OS rate were 4 months, 13.1 months and 57%, respectively. Seven patients were alive after a median follow-up of 34.2 months.Citation345 As interesting, the combination of REOLYSIN with paclitaxel in treatment-refractory breast cancer did not impact PFS or ORR but significantly extended OS (IND.213).Citation347 In patients with advanced melanoma, REOLYSIN combined with carboplatin and paclitaxel was potentially efficacious as the study met its efficacy goal for the first stage with an ORR of 21% (3 PR). The median PFS, median OS and 1-year OS were 5.2 months, 10.9 months, and 43%, respectively, with a disease control rate of 85%.Citation344 In the single-arm study combining i.v. REOLYSIN with gemcitabine against pancreatic adenocarcinoma, 1 PR, 23 SD and 5 PD were witnessed with a median OS of 10.2 months, and a 1- and 2-year survival rate of 45% and 24%, respectively. When considering the overall poor prognosis of pancreatic cancer (1 and 5-year survival rates of ~ 18% and 7%),Citation356 REOLYSIN appeared to complement single agent gemcitabine against the disease. Moreover, upregulation of PD-L1 in the biopsy of a tumor treated with the combination therapy encourages future supplementation with PD-L1/PD-1 checkpoint inhibitors.Citation352

Rhabdoviruses. The oncolytic rhabdovirus Maraba MG1 expressing the CT antigen MAGE-A3 (MG1MA3; Turnstone Biologics, Ottawa, ON, Canada) is currently being investigated in a Phase I/II trial against incurable advanced/metastatic MAGE-A3-positive solid tumors (NCT02285816/CCTG IND.214).Citation102,Citation357 First results from Phase I have been disclosed at the 2017 ASCO meeting.Citation357 The oncolytic vaccine MG1MA3 was infused systemically either alone (arm A, n = 9 patients) or as a booster in a heterologous prime-boost vaccination strategy (arm C, n = 25). In the latter setting, a replication-deficient Ad vaccine expressing MAGE-A3 (AdMA3) was injected intramuscularly 14 days prior to MG1MA3 in order to prime MAGE-A3 immunity.Citation222,Citation224,Citation357,Citation358 A third cohort received only AdMA3 (arm B, n = 6). A 3 + 3 escalating dose design in the arms A and C allowed to establish the recommended Phase II dose of MG1MA3. Acute AEs related to the Maraba virus in the arm C commonly included fatigue, flu-like symptoms, diarrhea, hypophosphatemia and hypotension. Interestingly, a pro-inflammatory gene signature was characterized in tumor biopsies, with markers of infiltration/activation of myeloid and lymphoid cell populations detected. Moreover, humoral and cellular immune responses specific to MAGE-A3 were observed in the blood of 3 out of 6 evaluable patients of the arm C. Remarkably, over 1% of the circulating CD8+ T cells were reacting against MAGE-A3 in one patient. Overall, AdMA3 prime followed by the oncolytic MG1MA3 boost is applicable and able to induce potent antitumor immunity in cancer patients.

Recently initiated clinical trials

Since the submission of our latest Trial Watch on oncolytic virotherapy,Citation40 no less than 63 clinical studies have been initiated to evaluate this immunotherapeutic paradigm in cancer patients (; source: https://www.clinicaltrials.gov/).

Adenoviruses. (1) A Phase I dose escalation study will determine the MTD of oncolytic Ad5-yCD/mutTKSR39rep-ADP, also known as Theragene® (Henry Ford Health System, Detroit, MI, USA in partnership with Newgenpharm, Seoul, South Korea), in medically inoperable stage I/IIA NSCLC. The OV is a replication-competent Ad5 armed with a fusion gene expressing yCD and the mutant sr39 HSV TK (mutTKsr39) together with the 11.6 kDa adenoviral death protein (ADP) gene.Citation359Citation361 Following i.t. delivery of Ad5-yCD/mutTKSR39rep-ADP, patients will be given 5-fluorocytosine (5-FC) and valganciclovir prodrugs, respectively metabolized by yCD and mutTKsr39, along with stereotactic body radiotherapy (NCT03029871). Moreover, Ad5-yCD/mutTKSR39rep-ADP and its IL-12-expressing derivative Ad5-yCD/mutTKSR39rep-hIL12 are being evaluated in association to 5-FC and standard chemotherapy in patients suffering from metastatic pancreatic cancer (NCT02894944, NCT03281382). (2) AdVince (Uppsala University, Uppsala, Sweden), formerly named Ad5[CgA-E1A-miR122]PTD, is an oncolytic Ad that has been designed to treat liver metastases of neuroendocrine tumors (NETs).Citation362 A Phase I/IIa clinical trial is recruiting patients to determine the MTD and evaluate the safety of repeated infusions of AdVince into the hepatic artery of patients with metastatic NETs (NCT02749331/RADNET). (3) A Phase I study is assessing the tolerability and safety of neoadjuvant neural stem cell (NSC)-based virotherapy for patients with newly diagnosed malignant glioma. The strategy relies on the administration of NSCs loaded with oncolytic CRAd-Survivin-pk7; a conditionally replicative glioma-tropic Ad.Citation363Citation365 Patients undergo intracranial injection of NSC-CRAd-Survivin-pk7 together with standard chemoradiotherapy, after surgery if resection is feasible (NCT03072134; Northwestern Memorial Hospital, Chicago, IL, USA). (4) The efficacy, safety and immunological response of i.t. OBP-301 (Telomelysin, Oncolys BioPharma, Tokyo, Japan) are being assessed in an open-label, multi-center Phase IIa trial in patients with unresectable metastatic melanoma (NCT03190824). Two Phase I trials are recruiting participants to determine the safety and tolerability of OBP-301 in combination with pembrolizumab against advanced solid tumors (NCT03172819)Citation366 or with radiotherapy against esophageal cancer (NCT03213054). (5) A Phase I study investigates the safety and antitumor activity of the recently introduced VCN-01 (VCN Biosciences, Barcelona, Spain)Citation146,Citation367 administered through intravitreal injection in 13 patients with refractory retinoblastoma (NCT03284268). (6) Up to 36 patients are currently recruited in a Phase II study to evaluate the efficacy of DNX-2401 (DNAtrix, Houston, TX, USA) against recurrent glioblastoma or gliosarcoma. The virus is delivered directly into the tumor followed by repeated i.v. administrations of the ICI pembrolizumab (NCT02798406/CAPTIVE). Additionally, the safety, tolerability and toxicity (hematologic and neurologic) of DNX-2401 as neoadjuvant therapy for diffuse intrinsic pontine glioma is being assessed in an unicentric non-randomized Phase I study. Administration of the OV is performed through injection in the cerebellar peduncle of newly diagnosed pediatric patients (NCT03178032).Citation368 (7) A Phase II trial will recruit a total of 30 patients with pleural mesothelioma to determine the safety, tolerability and efficacy of i.t. ONCOS-102 (Targovax, Oslo, Norway), preceded by CPA and followed by pemetrexed/cisplatin (NCT02879669). A Phase I/II study is investigating the safety, biologic and anti-tumor activities of i.p. ONCOS-102 in association with durvalumab (anti-PD-L1) in subjects with advanced CRC or platinum-resistant ovarian cancers (NCT02963831). A Phase I pilot study evaluates the safety of i.t. ONCOS-102, preceded by CPA and followed by pembrolizumab, in two cohorts of patients with advanced or unresectable melanoma progressing after ICB therapy. Cohort 1 regroups subjects that failed prior anti-PD-1 monotherapy while cohort 2 failed prior anti-PD-1 plus ipilimumab (NCT03003676). (8) Two Phase I/II trials have been registered to evaluate the safety and efficacy of LOAd703Citation200,Citation252 (Lokon Pharma AB, Uppsala, Sweden) in patients with pancreatic, biliary, colorectal or ovarian cancers. LOAd703 will be co-administered i.t. together with the standard-of-care chemotherapy (NCT03225989; NCT02705196). (9) Harb WA and colleagues (PsiOxus Therapeutics, Abingdon, UK) have initiated a Phase I study (named SPICE; NCT02636036) to evaluate the safety profile of i.v. EnAd combined to pembrolizumab in patients with metastatic or advanced HNC, CRC, urothelial and salivary gland carcinomas.Citation369

Herpesviruses. (1) The safety and efficacy of T-VEC (Amgen, Thousand Oaks, CA, USA) is being evaluated in various types of advanced and metastatic cancers. Intralesional T-VEC monotherapy is undergoing testing in breast (NCT02658812), advanced and metastatic pancreatic cancer refractory to chemotherapy (NCT03086642), unresectable advanced Stage IIIB-IV malignant melanoma (NCT03064763) and advanced non-CNS pediatric tumors (NCT02756845). Moreover, T-VEC is being tested in combination with ICIs: i) pembrolizumab in patients with advanced sarcoma (NCT03069378),Citation318 melanoma (NCT02965716)Citation370 and recurrent metastatic HNC (NCT02626000/MASTERKEY232), ii) nivolumab in refractory lymphomas and metastatic non-melanoma skin cancers (NCT02978625) and iii) atezolizumab in patients harboring TNBC and metastatic CRC (NCT03256344). Additionally, T-VEC is being evaluated for efficacy in combination with a) standard chemotherapy against TNBC (NCT02779855) and CRC (NCT03300544), b) targeted therapies such as the BRAF and MEK inhibitors dabrafenib and trametinib, respectively, against BRAF-mutated advanced melanoma (NCT03088176) and c) radiotherapy in soft tissue sarcoma (NCT02923778), various forms of CRC (NCT03300544), and melanoma, Merkel cell carcinoma and other solid tumors (NCT02819843). One Phase I trial is recruiting participants to evaluate the mechanisms of action of T-VEC in patients with non-melanoma skin cancer through determination of local immune effects after repeated T-VEC injections (NCT03458117). Assays will include quantifying some local immune activation markers in skin biopsies of injected lesions (such as IFN, 2-prime, 5-prime oligoadenylate synthetase 1 [OAS1], Interferon-induced GTP-binding protein MxA [MXA] and C-X-C motif chemokine 11 [CXCL11]) as well as immune response markers in sera and peripheral blood mononuclear cells. Lastly, a prospective study is evaluating the risk of herpetic infection in melanoma patients that received T-VEC (NCT02910557). (2) Two single-arm open label Phase II studies, initiated by Takara Bio Inc, aim at determining the efficacy and saftey of i.t. injections of oncolytic HSV-1 HF10Citation371 in combination with ipilimumab in Japanese patients with Stage IIIb/C or IV unresectable or metastatic melanoma (NCT03153085) or in a neoadjuvant setting together with nivolumab against resectable Stage IIIB, IIIC, IVM1a melanoma (NCT03259425). Additionally, a Phase I dose trial is determining the recommended dose of HF10 in combination with chemotherapy (gemcitabine + Nab-paclitaxel) in Japanese patients with Stage III or IV unresectable pancreatic cancer (NCT03252808).

Paramyxoviruses. A Phase I trial, soon to be open for participant recruitment, will evaluate the safety and toxicity of MV-NIS (Vyriad, Rochester, MN, USA) in patients with urothelial carcinoma undergoing cystectomy. Antitumor efficacy of intravesical MV-NIS neoadjuvant therapy will be assessed as secondary outcome measures (NCT03171493). Safety of MV-NIS is being assessed in a Phase I study in children and young adults with recurrent medulloblastoma or atypical teratoid rhabdoid tumor. The OV is injected in the tumor bed for local recurrence (during surgical resection) or via lumbar puncture for disseminated recurrence (NCT02962167). I.t. MV-NIS monotherapy is also investigated against malignant unresectable or recurrent peripheral nerve sheath tumor (NCT02700230). A fourth Phase I trial aims at defining the MTD of i.t. MV-NIS in combination with atezolizumab, an anti-PD-L1, in patients with metastatic NSCLC (NCT02919449).

Parvoviruses. Safety and tolerability of ParvOryx (ORYX GmbH & Co. KG, Baldham, Germany) will be evaluated in a Phase I/II trial in subjects suffering from metastatic, inoperable pancreatic cancer. Parvovirus H-1 will be administered at three increasing dose levels with 40% of the total dose given i.v. and the remaining 60% given i.t. into a hepatic metastasis of the pancreatic cancer (NCT02653313/ParvOryx02)

Picornaviruses. (1) A Phase I study will evaluate the safety and efficacy of i.v. CAVATAK/CVA21 (Viralytics, Sydney, Australia) in combination with pembrolizumab in patients with advanced NSCLC (NCT02824965). (2) In a Phase Ib trial, the safety and toxicity of a single dose of i.t. PVSRIPO (Duke University, Durham, NC, USA) is evaluated in children affected with diverse brain cancers, including advanced glioma. A secondary objective will consist of estimating OS in this population (NCT03043391). A Phase II trial aims at evaluating the impact of PVSRIPO on the survival of adult patients with recurrent severe glioma, relative to the survival observed in a historical control group. PVSRIPO will be delivered i.t. either alone or with a single dose of lomustine, an alkylating nitrosourea compound used as standard-of-care chemotherapy. The safety of the latter combination treatment will also be assessed (NCT02986178).

Poxviruses. (1) Two Phase I/II trials have been initiated by Genelux (San Diego, CA, USA) involving their lead VV candidate, GL-ONC1. A Phase Ib trial (NCT02714374) enrolled 36 patients with solid organ cancers. The study will evaluate the tolerability of single or multiple doses of GL-ONC1 delivered i.v., with or without eculizumab (a humanized monoclonal antibody functioning as a terminal complement inhibitor), prior to surgery. A Phase Ib/II trial aims at recruiting 52 patients to investigate the safety and tolerability of repeated i.p. infusions of GL-ONC1 in women diagnosed with recurrent ovarian cancer, peritoneal carcinomatosis or fallopian tube cancer (NCT02759588/VIRO-15). PFS will also be assessed and tumor response to treatment evaluated by RECIST 1.1 and irRECIST 1.1. Results of the Phase Ib have been disclosed at the 2018 ASCO meeting and reported in the previous section on “advanced clinical trials”.Citation339 (2) Pexa-Vec, also known as JX-594 or TG6006, is a TK GM-CSF+ LacZ+ variant of the VV Wyeth strain (SillaJen, San Francisco, CA, USA in partnership with Transgene, Illkirch-Graffenstaden, France; Green Cross, Kyoto, Japan; Lee’s Pharmaceuticals, Hong Kong, China and Rex Medical, Conshohocken, PA, USA). Pexa-Vec is one of the most extensively investigated VV-based oncolytic biotherapeutic with no less than 19 clinical trials registered on the clinicaltrial.gov website. A multi-center, randomized, open-label, Phase 3 study has been initiated in 2015 and will enroll 600 patients to compare Pexa-Vec followed by sorafenib (a multikinase inhibitor) versus sorafenib alone in patients with advanced HCC without prior systemic therapy (NCT02562755/PHOCUS). During the past 2 years, 5 clinical trials involving Pexa-Vec have been initiated. A two-part Phase I clinical trial aims at assessing the DLT of escalating doses of ipilimumab (up to 5 injections given i.t.) in combination with a fixed dose of Pexa-Vec (up to 6 injections given i.t.) in 60 patients with advanced/metastatic solid tumors (NCT02977156/ISI-JX). The second part of the study will determine the ORR according to irRC. A second two-part Phase I trial will enroll up to 89 patients with renal cell carcinoma (NCT03294083). The first part will determine the MTD and maximum feasible dose (MFD) of Pexa-Vec administered by i.v. infusion in combination with REGN2810, an anti-PD-1 monoclonal antibody. The second part of the study will assess the safety of Pexa-Vec administered by i.v. infusions or i.t. injections in combination with i.v. REGN2810 and evaluate anti-tumor activity and efficacy with respect to ORR per RECIST 1.1. Secondary outcome evaluations will consist in measuring PFS and OS. A Phase I/IIa trial is recruiting 30 participants to evaluate the safety and efficacy of the combination of i.t. Pexa-Vec with Nivolumab as first-line therapy of advanced HCC (NCT03071094). Another clinical trial is determining the safety, tolerability and feasibility of Pexa-Vec in combination with immune checkpoint inhibitors in 35 patients with refractory metastatic CRC (NCT03206073). Precisely, two different doses of Pexa-Vec will be injected i.v. for 4 times together with repeated infusions of anti-PD-L1 durvalumab (cohort A) or durvalumab + anti-CTLA-4 tremelimumab (cohort B). Finally, a Phase Ib/II clinical trial is expecting to recruit 118 patients with advanced breast cancer, soft-tissue sarcoma or other solid tumors to assess the tolerability and antitumor efficacy of i.v. Pexa-Vec in combination with metronomic CPA (NCT02630368/METROmaJX). After determining the MTD in 14 subjects (Phase Ib), the antitumor activity of the combination therapy will be determined based on 6-month non-progression in soft-tissue sarcoma patients (48 receiving the dual therapy versus 24 ingesting CPA alone) or based on OR to treatment in the breast cancer cohort (single-arm, n = 32), following RECIST 1.1 criteria. (3) TG6002 (Transgene, Illkirch-Graffenstaden, France) is an engineered oncolytic vaccinia virus deleted from two genes expressing the viral TK and ribonucleotide reductase and outfitted with the suicide gene FCU1. FCU1 gene encodes a bifunctional chimeric protein that catalyzes the conversion of the non-toxic 5-FC into the toxic 5-FU. A Phase I/II aims at evaluating the safety and efficacy of i.v. TG6002 plus oral 5-FC in recurrent GB patients (NCT03294486/ONCOVIRAC).Citation372

Reoviruses. The safety and therapeutic efficacy of REOLYSIN® is being assessed in pancreatic adenocarcinoma patients, who are being treated with i.v. REOLYSIN® in combination with chemotherapy (gemcitabine, irinotecan, leucovorin, or 5-FU) and pembrolizumab (NCT02620423); in multiple myeloma patients, receiving REOLYSIN® with either lenalidomide or pomalidomide (NCT03015922/MUKeleven); as well as in individuals with skin melanoma in combination with GM-CSF (NCT03282188).

Retroviruses. Toca 511, also referred to as vocimagene amiretrorepvec (Tocagen, San Diego, CA, USA), is a replication-competent tumor-selective retrovirus that expresses an optimized yCD.Citation362 When combine to Toca FC, an extended-release formulation of the prodrug 5-FC, it locally generates toxic 5-FU. A Phase II study will establish the recommended dose of Toca FC to be used in combination with Toca 511, together with standard-of-care chemoradiation and TMZ, in subjects with newly diagnosed high grade glioma (HGG). Toca 511 will be administered by intracranial parenchymal injection into the cavity resulting from surgical resection (NCT02598011/Toca7).

Rhabdoviruses. (1) Three Phase I trials aim at determining the tolerability and safety of VSV-IFNβ-NIS (Vyriad, Rochester, MN, USA) administered either i.t. against refractory advanced/metastatic solid tumors (NCT02923466) or given i.v. against relapsed or refractory multiple myeloma, acute myeloid leukemia (AML) or T-cell lymphoma (NCT03017820) or against stage IV or recurrent endometrial cancers (NCT03120624). (2) A Phase I/II trial will determine the MTD and efficacy of the oncolytic vaccine Maraba MG1MA3 (Turnstone Biologics, Ottawa, ON, Canada) in patients with NSCLC who have completed a first standard therapy with a platinum-based chemotherapy. MG1MA3 will be given i.v. following an intramuscular delivery of the non-replicating AdMA3 vaccine and in association with pembrolizumab (NCT02879760).

Status changes

The following studies discussed in our previous Trial Watches related to oncolytic virotherapy Citation40,Citation373,Citation374 have changed status during the past 2.5 years: NCT02293850, NCT02457845, NCT02509507, NCT02068794, NCT02562755 and NCT02414165 which are currently “recruiting”; NCT02263508, NCT02365818, NCT02197169, NCT02045589, NCT02272855, NCT02366195, NCT02211131, NCT01491893, NCT01766739, NCT01274624, NCT01740297, which are now « active, not recruiting »; NCT00390299 which has been suspended; NCT02031965, NCT01438112 and NCT01017601, which have been terminated; NCT02028442, NCT01656538, NCT01844661, NCT01985256, NCT01156584, NCT02014441, NCT01864759, NCT01956734, NCT02053220, NCT02428036, NCT01721018, NCT01935453, NCT02316171, NCT01636882, NCT00794131, NCT01394939, NCT01469611, NCT01636284, NCT01171651, NCT01380600, NCT00602277, NCT01619813, NCT01708993 and NCT00861627 which are now completed.

Although their status is not completed, preliminary results are available for the trials NCT02263508 Citation228 (a Phase Ib/III study testing the combination of T-VEC with pembrolizumab in patients with unresected melanoma), NCT01740297 Citation297 (a Phase Ib/II trial evaluating the safety and efficacy of T-VEC in combination with ipilimumab in subjects with unresected melanoma) and NCT01656538 Citation325 (a Phase II trial investigating the benefit of REOLYSIN when given with Paclitaxel in subjects with advanced/metastatic breast cancer).

The results from the “completed” studies NCT02316171 Citation314 (a Phase I study assessing the safety and efficacy of CVA21 with or without sequencial combination with mitomycin C in individuals with bladder cancer), NCT01619813 Citation327 (a Phase II trial testing the efficacy of combining REOLYSIN with docetaxel and predisone in metastatic castration resistant prostate cancer patients), NCT01708993 Citation326 (a Phase II study evaluating the combination of REOLYSIN with docetaxel or pemetrexed in subjects with advanced or metastatic NSCLC), and NCT00861627 Citation323 (a Phase II trial combining REOLYSIN with paclitaxel and carboplatin in NSCLC patients) have been considered in the “completed and advanced clinical studies” section above.

Finally, based on our investigations, the results from NCT02028442 (a Phase I/II trial testing EnAd in patients suffering from epithelial solid tumors or metastatic colorectal/bladder cancer), NCT01656538 (a Phase II study evaluating the safety of REOLYSIN in combination with the standard treatment Paclitaxel in metastatic or advanced breast cancer patients), NCT01985256 (a Phase I trial assessing the tolerability of the retroviral vector Toca 511 in patients undergoing brain tumor surgery), NCT01156584 (a Phase I study investigating the safety of Toca 511 in individuals that have undergone surgery of recurrent glioma), NCT02014441 (a Phase II study assessing the biodistribution of T-VEC after intralesional injection in subjects suffering from melanoma), NCT01864759 (a Phase I trial assessing the safety of ICOVIR-5 in subjects with locally advanced or metastatic melanoma), NCT01956734 (a Phase I study combining DNX-2401 with TMZ in glioblastoma patients), NCT02053220 (a Phase I trial determining the viral delivery and viral expression of ColoAd1 in tumor tissues of patients eligible for primary tumor resection), NCT02428036 (a Phase I trial testing the i.t. administration of HF10 in individuals with solid tumors comprising superficial lesions), NCT01721018 (a Phase I/IIa study assessing the safety and tolerability of HSV1716 inoculation into pleural cavity of subjects with inoperable malignant pleural mesothelioma), NCT01935453 (a Phase I trial investigating the safety of OrienX010 in patients with solid tumors), NCT01636882 (a Phase II extented use study aiming at continue the treatment with CAVATAK in individuals with stage IIIc and stage IV malignant melanoma), NCT00794131 (a Phase I trial evaluating the safety of GL-ONC1 i.v. administration in subjects with advanced solid tumors), NCT01394939 (a Phase I/IIa study of i.v. administration of Pexa-Vec with or without Irinotecan in patients with metastatic, refractory colorectal carcinoma), NCT01469611 (a Phase Ib trial designed to determine the MTD/MFD of Pexa-Vec and the safety of its biweekly i.v. infusion in individuals with advanced CRC), NCT01636284 (a Phase IIa study evaluating the efficacy of Pexa-Vec in HCC patients who had not formerly received sorafenib), NCT01171651 (a Phase II trial assessing the safety of Pexa-Vec i.v. and i.t. administration before standard therapy with sorafenib in subjects with unresectable primary HCC), NCT01380600 (a Phase Ib trial investigating the safety of Pexa-Vec biweekly i.v. administration in CRC individuals) and NCT00602277 (a Phase I study evaluating the REOLYSIN viral therapy in patients with ovarian, primary peritoneal and fallopian tube cancer who did not respond to platinum therapy), have not been disclosed yet even though their status is currently reported as “completed”.

Concluding remarks

Based on the sizeable number of clinical trials reported, the attractiveness of oncolytic virotherapy has grown since the approval of T-VEC in 2015. The broad repertoire of OV strains allows their application for virtually any type of malignancy, whether hematological or solid, localized or disseminated. With that said, considerable efforts are being made on evaluating OVs against highly morbid cancers that keep resisting to the arsenal of approved drugs, such as brain and pancreatic tumors. OV therapeutic efficacy mostly relies on its ability to prime and adjuvantize both innate (NK cells, DCs, M1-like macrophages) and adaptive (CTLs) arms of antitumor immunity. For this reason, cumulative evidences evoke the use of oncolytic virotherapy in support of current standards of care. If, for instance, T-VEC can intervene as a first-line monotherapy, the OV platform seems adequately fitted for combinatory strategies and has already demonstrated promising benefits in association with surgery (in both neoadjuvant and adjuvant settings), chemotherapy, radiotherapy and immunotherapy. Regarding the latter, synergistic interaction between OVs and ICIs has been quasi systematically witnessed, regardless of the virus strain. Interestingly, antitumor activity was detected not only in the primary lesion but also at distant sites that were not necessarily targeted by the virus (abscopal effect). However, caution to the experimental design has been raised to prevent antagonistic interactions. If much of the focus has been dragged by anti-PD-1/CTLA-4, oncolytic virotherapy also appeared beneficial to other immunotherapeutic approaches, including CAR T cells and BiTE antibodies. The characterization of markers responsible for resistance to the various OV strains, as well as markers of good prognosis, will contribute to define and optimize the associations that best suit the therapeutic needs. Finally, OV application to the field of oncology could expand beyond treatment as their tumor selectivity can be exploited as a sensitive diagnostic tool, or for imaging tumor lesions, or for clearing residual malignant entities from cell preparations destined to autologous transfer.

Abbreviations

5-FC=

5-fluorocytosine

5-FU=

5-fluorouracil

Ad=

adenovirus

AE=

adverse event

BiTE=

bispecific T cell-engager

CAR=

chimeric antigen receptor

CNS=

central nervous system

CPA=

cyclophosphamide

CRC=

colorectal cancer

CTL=

cytotoxic T lymphocyte

DC=

dendritic cell

DLT=

dose limiting toxicity

GB=

glioblastoma

GSC=

glioblastoma stem-like cell

HCC=

hepatocellular carcinoma

HNC=

head and neck cancer

HSV=

herpes simplex virus

IAV=

influenza A virus

ICI=

immune checkpoint inhibitor

IFN=

interferon

IL=

interleukin

irRC=

immune-related Response Criteria

MDSC=

myeloid-derived suppressor cell

MSC=

mesenchymal stem cell

MTD=

maximum tolerated dose

MV=

measle virus

MYXV=

myxomavirus

NDV=

Newcastle disease virus

NSCLC=

non-small cell lung carcinoma

OR=

objective response

ORR=

overall response rate

OS=

overall survival

OV=

oncolytic virus

PFS=

progression-free survival

SD=

stable disease

TAA=

tumor-associated antigen

TIL=

tumor-infiltrating lymphocyte

TME=

tumor microenvironment

TMZ=

temozolomide

TNBC=

triple-negative breast cancer

Treg=

regulatory T cell

VV=

vaccinia virus

yCD=

yeast cytosine deaminase.

Disclosures

SL, STW, FLB, SG, DRC, KLM, GK and LG have no conflicts of interest to declare. JGP and JCB share ownership of a patent for cancer vaccine composition involving oncolytic Maraba MG1 (publication number: EP2958994A1). JCB is a co-founder, equity holder and on the Board of Directors of Turnstone Biologics. JEF and LF are, respectively, funded by and employee of Transgene. LZ is on the Board of Directors of Transgene.

Additional information

Funding

LG is supported by an intramural startup from the Department of Radiation Oncology of Weill Cornell Medical College (New York, US), and by donations from Sotio a.s. (Prague, Czech Republic) and Phosplatin (New York, US). GK is supported by the Ligue contre le Cancer (Equipe labelisée); Agence Nationale de la Recherche (ANR)—Projets blancs; ANR under the frame of E-Rare-2, the ERA-Net for Research on Rare Diseases; Association pour la recherche sur le cancer (ARC); Cancéropôle Ile-de-France; Institut National du Cancer (INCa); Institut Universitaire de France; Fondation pour la Recherche Médicale (FDM20140630126 and FDM 40739); the European Commission (ArtForce); the European Research Council (ERC); the Leducq Foundation; the Seerave Foundation; the LabEx Immuno-Oncology; the RHU Torino Lumière; the SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE); the SIRIC Cancer Research and Personalized Medicine (CARPEM); and the Paris Alliance of Cancer Research Institutes (PACRI).

References

  • Yue C, Jiang Y, Li P, Wang Y, Xue J, Li N, Li D, Wang R, Dang Y, Hu Z, et al. Dynamic change of PD-L1 expression on circulating tumor cells in advanced solid tumor patients undergoing PD-1 blockade therapy. Oncoimmunology. 2018;7:e1438111. doi:10.1080/2162402X.2018.1438111.
  • Wu X, Giobbie-Hurder A, Connolly EM, Li J, Liao X, Severgnini M, Zhou J, Rodig S, Hodi FS. Anti-CTLA-4 based therapy elicits humoral immunity to galectin-3 in patients with metastatic melanoma. Oncoimmunology. 2018;7:e1440930. doi:10.1080/2162402X.2018.1440930.
  • Johnson CB, Win SY. Combination therapy with PD-1/PD-L1 blockade: an overview of ongoing clinical trials. Oncoimmunology. 2017;7. doi:10.1080/2162402X.2017.1408744.
  • Kaderbhai CG, Richard C, Fumet JD, Aarnink A, Ortiz-Cuaran S, Perol M, Foucher P, Coudert B, Favier L, Lagrange A, et al. Response to first line chemotherapy regimen is associated with efficacy of nivolumab in non-small-cell lung cancer. Oncoimmunology. 2017;6:e1339856. doi:10.1080/2162402X.2017.1339856.
  • Garg AD, Vandenberk L, Van Woensel M, Belmans J, Schaaf M, Boon L, De Vleeschouwer S, Agostinis P. Preclinical efficacy of immune-checkpoint monotherapy does not recapitulate corresponding biomarkers-based clinical predictions in glioblastoma. Oncoimmunology. 2017;6:e1295903. doi:10.1080/2162402X.2017.1295903.
  • Galluzzi L, Eggermont A, Kroemer G. Doubling the blockade for melanoma immunotherapy. Oncoimmunology. 2016;5:e1106127. doi:10.1080/2162402X.2015.1106127.
  • Jelinek T, Hajek R. PD-1/PD-L1 inhibitors in multiple myeloma: the present and the future. Oncoimmunology. 2016;5:e1254856. doi:10.1080/2162402X.2016.1254856.
  • Rekoske BT, Olson BM, McNeel DG. Antitumor vaccination of prostate cancer patients elicits PD-1/PD-L1 regulated antigen-specific immune responses. Oncoimmunology. 2016;5:e1165377. doi:10.1080/2162402X.2016.1165377.
  • Houot R, Gaulard P, Schreiber R, Mellman I, Lambotte O, Coulie PG, Fest T, Korman A, Levy R, Shipp M, et al. Immunomodulatory antibodies for the treatment of lymphoma: report on the CALYM Workshop. Oncoimmunology. 2016;5:e1186323. doi:10.1080/2162402X.2016.1186323.
  • Karydis I, Chan PY, Wheater M, Arriola E, Szlosarek PW, Ottensmeier CH. Clinical activity and safety of Pembrolizumab in Ipilimumab pre-treated patients with uveal melanoma. Oncoimmunology. 2016;5:e1143997. doi:10.1080/2162402X.2016.1143997.
  • Buque A, Bloy N, Aranda F, Castoldi F, Eggermont A, Cremer I, Fridman WH, Fucikova J, Galon J, Marabelle A, et al. Trial Watch: immunomodulatory monoclonal antibodies for oncological indications. Oncoimmunology. 2015;4:e1008814. doi:10.1080/2162402X.2015.1008371.
  • Vanella V, Festino L, Strudel M, Simeone E, Grimaldi AM, Ascierto PA. PD-L1 inhibitors in the pipeline: promise and progress. Oncoimmunology. 2017;7:e1365209. doi:10.1080/2162402X.2017.1365209.
  • Vetizou M, Pitt JM, Daillere R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CP, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350:1079–1084. doi:10.1126/science.aad1329.
  • Aranda F, Vacchelli E, Eggermont A, Galon J, Fridman WH, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: immunostimulatory monoclonal antibodies in cancer therapy. Oncoimmunology. 2014;3:e27297. doi:10.4161/onci.27297.
  • Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautes-Fridman C, Tartour E, Zucman-Rossi J, Zitvogel L, Kroemer G. Trial Watch: monoclonal antibodies in cancer therapy. Oncoimmunology. 2012;1:28–37. doi:10.4161/onci.1.1.17938.
  • Vacchelli E, Pol J, Bloy N, Eggermont A, Cremer I, Fridman WH, Galon J, Marabelle A, Kohrt H, Zitvogel L, et al. Trial watch: tumor-targeting monoclonal antibodies for oncological indications. Oncoimmunology. 2015;4:e985940. doi:10.1080/2162402X.2015.1008371.
  • Chaganty BK, Lu Y, Qiu S, Somanchi SS, Lee DA, Fan Z. Trastuzumab upregulates expression of HLA-ABC and T cell costimulatory molecules through engagement of natural killer cells and stimulation of IFNgamma secretion. Oncoimmunology. 2016;5:e1100790. doi:10.1080/2162402X.2015.1100790.
  • Vacchelli E, Aranda F, Bloy N, Buque A, Cremer I, Eggermont A, Fridman WH, Fucikova J, Galon J, Spisek R, et al. Trial Watch-Immunostimulation with cytokines in cancer therapy. Oncoimmunology. 2016;5:e1115942. doi:10.1080/2162402X.2015.1115942.
  • Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, et al. Molecular mechanisms of cell death: recommendations of the nomenclature committee on cell death 2018. Cell Death Differ. 2018;25:486–541. doi:10.1038/s41418-017-0012-4.
  • Galluzzi L, Buque A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol. 2017;17:97–111. doi:10.1038/nri.2016.107.
  • Garg AD, More S, Rufo N, Mece O, Sassano ML, Agostinis P, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: immunogenic cell death induction by anticancer chemotherapeutics. Oncoimmunology. 2017;6:e1386829. doi:10.1080/2162402X.2017.1386829.
  • Hanoteau A, Henin C, Svec D, Bisilliat Donnet C, Denanglaire S, Colau D, Romero P, Leo O, Van den Eynde B, Moser M. Cyclophosphamide treatment regulates the balance of functional/exhausted tumor-specific CD8(+) T cells. Oncoimmunology. 2017;6:e1318234. doi:10.1080/2162402X.2017.1318234.
  • Di Blasio S, Wortel IM, van Bladel DA, de Vries LE, Duiveman-de Boer T, Worah K, de Haas N, Buschow SI, de Vries IJ, Figdor CG, et al. Human CD1c(+) DCs are critical cellular mediators of immune responses induced by immunogenic cell death. Oncoimmunology. 2016;5:e1192739. doi:10.1080/2162402X.2016.1192739.
  • van Dodewaard-de Jong JM, Santegoets SJ, van de Ven PM, Versluis J, Verheul HM, de Gruijl TD, Gerritsen WR, van den Eertwegh AJ. Improved efficacy of mitoxantrone in patients with castration-resistant prostate cancer after vaccination with GM-CSF-transduced allogeneic prostate cancer cells. Oncoimmunology. 2016;5:e1105431. doi:10.1080/2162402X.2015.1105431.
  • Galluzzi L, Buque A, Kepp O, Zitvogel L, Kroemer G. Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell. 2015;28:690–714. doi:10.1016/j.ccell.2015.10.012.
  • Pol J, Vacchelli E, Aranda F, Castoldi F, Eggermont A, Cremer I, Sautes-Fridman C, Fucikova J, Galon J, Spisek R, et al. Trial Watch: immunogenic cell death inducers for anticancer chemotherapy. Oncoimmunology. 2015;4:e1008866. doi:10.1080/2162402X.2015.1008371.
  • Vacchelli E, Aranda F, Eggermont A, Galon J, Sautes-Fridman C, Cremer I, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: chemotherapy with immunogenic cell death inducers. Oncoimmunology. 2014;3:e27878. doi:10.4161/onci.27878.
  • Martins I, Galluzzi L, Kroemer G. Hormesis, cell death and aging. Aging (Albany NY). 2011;3:821–828. doi:10.18632/aging.100380.
  • Zheng PP, Kros JM, Li J. Approved CAR T cell therapies: ice bucket challenges on glaring safety risks and long-term impacts. Drug Discov Today. 2018; 23:1175–1182. doi:10.1016/j.drudis.2018.02.012.
  • Galluzzi L, Martin P. CARs on a highway with roadblocks. Oncoimmunology. 2017;6:e1388486. doi:10.1080/2162402X.2017.1388486.
  • Berraondo P, Labiano S, Minute L, Etxeberria I, Vasquez M, Sanchez-Arraez A, Teijeira A, Melero I. Cellular immunotherapies for cancer. Oncoimmunology. 2017;6:e1306619. doi:10.1080/2162402X.2017.1306619.
  • Watanabe N, Bajgain P, Sukumaran S, Ansari S, Heslop HE, Rooney CM, Brenner MK, Leen AM, Vera JF. Fine-tuning the CAR spacer improves T-cell potency. Oncoimmunology. 2016;5:e1253656. doi:10.1080/2162402X.2016.1253656.
  • Zhang Q, Zhang Z, Peng M, Fu S, Xue Z, Zhang R. CAR-T cell therapy in gastrointestinal tumors and hepatic carcinoma: from bench to bedside. Oncoimmunology. 2016;5:e1251539. doi:10.1080/2162402X.2016.1251539.
  • Garcia-Cuesta EM, Esteso G, Ashiru O, Lopez-Cobo S, Alvarez-Maestro M, Linares A, Ho MM, Martinez-Pineiro L, H TR, Vales-Gomez M. Characterization of a human anti-tumoral NK cell population expanded after BCG treatment of leukocytes. Oncoimmunology. 2017;6:e1293212. doi:10.1080/2162402X.2017.1293212.
  • Qu K, Gu J, Ye Y, Williams SB, Dinney CP, Wu X, Kamat A. High baseline levels of interleukin-8 in leukocytes and urine predict tumor recurrence in non-muscle invasive bladder cancer patients receiving bacillus calmette-guerin therapy: A long-term survival analysis. Oncoimmunology. 2017;6:e1265719. doi:10.1080/2162402X.2016.1265719.
  • Gulley JL, Mulders P, Albers P, Banchereau J, Bolla M, Pantel K, Powles T. Perspectives on sipuleucel-T: its role in the prostate cancer treatment paradigm. Oncoimmunology. 2016;5:e1107698. doi:10.1080/2162402X.2015.1107698.
  • Wei XX, Fong L, Small EJ. Prostate Cancer Immunotherapy with Sipuleucel-T: current standards and future directions. Expert Rev Vaccines. 2015;14:1529–1541. doi:10.1586/14760584.2015.1099437.
  • Vacchelli E, Vitale I, Eggermont A, Fridman WH, Fucikova J, Cremer I, Galon J, Tartour E, Zitvogel L, Kroemer G, et al. Trial watch: dendritic cell-based interventions for cancer therapy. Oncoimmunology. 2013;2:e25771. doi:10.4161/onci.25771.
  • Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–422. doi:10.1056/NEJMoa1001294.
  • Pol J, Buque A, Aranda F, Bloy N, Cremer I, Eggermont A, Erbs P, Fucikova J, Galon J, Limacher JM, et al. Trial Watch-Oncolytic viruses and cancer therapy. Oncoimmunology. 2016;5:e1117740. doi:10.1080/2162402X.2015.1117740.
  • Pol J, Kroemer G, Galluzzi L. First oncolytic virus approved for melanoma immunotherapy. Oncoimmunology. 2016;5:e1115641. doi:10.1080/2162402X.2015.1115641.
  • Schmidt C. Oncolytic Virus Approved To Treat Melanoma. J Natl Cancer Inst. 2016;108.
  • Ott PA, Hodi FS. Talimogene Laherparepvec for the Treatment of Advanced Melanoma. Clin Cancer Res. 2016;22:3127–3131. doi:10.1158/1078-0432.CCR-15-2709.
  • Andtbacka RH, Agarwala SS, Ollila DW, Hallmeyer S, Milhem M, Amatruda T, Nemunaitis JJ, Harrington KJ, Chen L, Shilkrut M, et al. 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. Head Neck. 2016;38:1752–1758. doi:10.1002/hed.24522.
  • Andtbacka RH, Kaufman HL, Collichio F, Amatruda T, Senzer N, Chesney J, Delman KA, Spitler LE, Puzanov I, Agarwala SS, et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol. 2015;33:2780–2788. doi:10.1200/JCO.2014.58.3377.
  • Harrington KJ, Andtbacka RH, Collichio F, Downey G, Chen L, Szabo Z, Kaufman HL. Efficacy and safety of talimogene laherparepvec versus granulocyte-macrophage colony-stimulating factor in patients with stage IIIB/C and IVM1a melanoma: subanalysis of the Phase III OPTiM trial. Onco Targets Ther. 2016;9:7081–7093. doi:10.2147/OTT.S115245.
  • Breitbach CJ, Lichty BD, Bell JC. Oncolytic viruses: therapeutics with an identity crisis. EBioMedicine. 2016;9:31–36. doi:10.1016/j.ebiom.2016.06.046.
  • Kaufman HL, Kohlhapp FJ, Zloza A. Oncolytic viruses: a new class of immunotherapy drugs. Nat Rev Drug Discov. 2015;14:642–662. doi:10.1038/nrd4663.
  • Lichty BD, Breitbach CJ, Stojdl DF, Bell JC. Going viral with cancer immunotherapy. Nat Rev Cancer. 2014;14:559–567. doi:10.1038/nrc3770.
  • Zhu Z, Gorman MJ, McKenzie LD, Chai JN, Hubert CG, Prager BC, et al. Zika virus has oncolytic activity against glioblastoma stem cells. J Exp Med. 2017;214:2843–2857. doi:10.1084/jem.20171093.
  • Cassady KA, Bauer DF, Roth J, Chambers MR, Shoeb T, Coleman J, Prichard M, Gillespie GY, Markert JM. Pre-clinical assessment of c134, a chimeric oncolytic herpes simplex virus, in mice and non-human primates. Mol Ther Oncolytics. 2017;5:1–10. doi:10.1016/j.omto.2017.02.001.
  • Kuhn I, Bauzon M, Green N, Seymour L, Fisher K, Hermiston T. OvAd1, a novel, potent, and selective chimeric oncolytic virus developed for ovarian cancer by 3D-directed evolution. Mol Ther Oncolytics. 2017;4:55–66. doi:10.1016/j.omto.2016.12.001.
  • Deng L, Fan J, Ding Y, Zhang J, Zhou B, Zhang Y, Huang B. Oncolytic efficacy of thymidine kinase-deleted vaccinia virus strain Guang9. Oncotarget. 2017;8:40533–40543. doi:10.18632/oncotarget.17125.
  • Zhang T, Essani K. Tanapoxvirus lacking the 15L gene inhibits melanoma cell growth in vitro by inducing interferon-lambda1 release. Virus Genes. 2017;53:477–482. doi:10.1007/s11262-017-1434-2.
  • Zhang T, Suryawanshi YR, Kordish DH, Woyczesczyk HM, Jeng D, Essani K. Tanapoxvirus lacking a neuregulin-like gene regresses human melanoma tumors in nude mice. Virus Genes. 2017;53:52–62. doi:10.1007/s11262-016-1402-2.
  • Garcia JA, Ferreira HL, Vieira FV, Gameiro R, Andrade AL, Eugenio FR, Flores EF, Cardoso TC. Tumour necrosis factor-alpha-induced protein 8 (TNFAIP8) expression associated with cell survival and death in cancer cell lines infected with canine distemper virus. Vet Comp Oncol. 2017;15:336–344. doi:10.1111/vco.12168.
  • Ammayappan A, Russell SJ, Federspiel MJ. Recombinant mumps virus as a cancer therapeutic agent. Mol Ther Oncolytics. 2016;3:16019. doi:10.1038/mto.2016.19.
  • Bell MP, Pavelko KD. Enhancing the tumor selectivity of a picornavirus virotherapy promotes tumor regression and the accumulation of infiltrating CD8+ T cells. Mol Cancer Ther. 2016;15:523–530. doi:10.1158/1535-7163.MCT-15-0459.
  • Nguyen TV, Heller GJ, Barry ME, Crosby CM, Turner MA, Barry MA. Evaluation of polymer shielding for adenovirus serotype 6 (Ad6) for systemic virotherapy against human prostate cancers. Mol Ther Oncolytics. 2016;3. doi:10.1038/mto.2015.21.
  • Guerrero CA, Guerrero RA, Silva E, Acosta O, Barreto E. Experimental adaptation of rotaviruses to tumor cell lines. PLoS One. 2016;11:e0147666. doi:10.1371/journal.pone.0147666.
  • Ruiz AJ, Hadac EM, Nace RA, Russell SJ. MicroRNA-detargeted mengovirus for oncolytic virotherapy. J Virol. 2016;90:4078–4092. doi:10.1128/JVI.02810-15.
  • Lin Y, Zhang H, Liang J, Li K, Zhu W, Fu L, Wang F, Zheng X, Shi H, Wu S, et al. Identification and characterization of alphavirus M1 as a selective oncolytic virus targeting ZAP-defective human cancers. Proc Natl Acad Sci U S A. 2014;111:E4504–12. doi:10.1073/pnas.1408759111.
  • Kasloff SB, Pizzuto MS, Silic-Benussi M, Pavone S, Ciminale V, Capua I. Oncolytic activity of avian influenza virus in human pancreatic ductal adenocarcinoma cell lines. J Virol. 2014;88:9321–9334. doi:10.1128/JVI.00929-14.
  • Pol JG, Rességuier J, Lichty B. Oncolytic viruses: a step into cancer immunotherapy. Virus Adaptation and Treatment. 2011;2012:1–21.
  • Howells A, Marelli G, Lemoine NR, Wang Y. Oncolytic viruses-interaction of virus and tumor cells in the battle to eliminate cancer. Front Oncol. 2017;7:195. doi:10.3389/fonc.2017.00195.
  • Ilkow CS, Swift SL, Bell JC, Diallo JS. From scourge to cure: tumour-selective viral pathogenesis as a new strategy against cancer. PLoS Pathog. 2014;10:e1003836. doi:10.1371/journal.ppat.1003836.
  • Angelova AL, Geletneky K, Nuesch JP, Rommelaere J. Tumor selectivity of oncolytic parvoviruses: from in vitro and animal models to cancer patients. Front Bioeng Biotechnol. 2015;3:55. doi:10.3389/fbioe.2015.00055.
  • Goldufsky J, Sivendran S, Harcharik S, Pan M, Bernardo S, Stern RH, Friedlander P, Ruby CE, Saenger Y, Kaufman HL. Oncolytic virus therapy for cancer. Oncolytic Virother. 2013;2:31–46. doi:10.2147/OV.S38901.
  • Geekiyanage H, Galanis E. MiR-31 and miR-128 regulates poliovirus receptor-related 4 mediated measles virus infectivity in tumors. Mol Oncol. 2016;10:1387–1403. doi:10.1016/j.molonc.2016.07.007.
  • Burke J, Nieva J, Borad MJ, Breitbach CJ. Oncolytic viruses: perspectives on clinical development. Curr Opin Virol. 2015;13:55–60. doi:10.1016/j.coviro.2015.03.020.
  • Jhawar SR, Thandoni A, Bommareddy PK, Hassan S, Kohlhapp FJ, Goyal S, Schenkel JM, Silk AW, Zloza A. Oncolytic viruses-natural and genetically engineered cancer immunotherapies. Front Oncol. 2017;7:202. doi:10.1128/JVI.02154-16.
  • van den Pol AN, Mao G, Chattopadhyay A, Rose JK, Davis JN. Chikungunya, Influenza, Nipah, and semliki forest chimeric viruses with vesicular stomatitis virus: actions in the Brain. J Virol. 2017;91. doi:10.3389/fonc.2017.00202
  • Kuznetsova I, Arnold T, Aschacher T, Schwager C, Hegedus B, Garay T, Stukova M, Pisareva M, Pleschka S, Bergmann M, et al. Targeting an oncolytic influenza a virus to tumor tissue by elastase. Mol Ther Oncolytics. 2017;7:37–44. doi:10.1016/j.omto.2017.09.002.
  • Petrovic B, Gianni T, Gatta V, Campadelli-Fiume G. Insertion of a ligand to HER2 in gB retargets HSV tropism and obviates the need for activation of the other entry glycoproteins. PLoS Pathog. 2017;13:e1006352. doi:10.1371/journal.ppat.1006352.
  • Campadelli-Fiume G, Petrovic B, Leoni V, Gianni T, Avitabile E, Casiraghi C, Gatta V. Retargeting strategies for oncolytic herpes simplex viruses. Viruses. 2016;8:63. doi:10.3390/v8030063.
  • Shibata T, Uchida H, Shiroyama T, Okubo Y, Suzuki T, Ikeda H, Yamaguchi M, Miyagawa Y, Fukuhara T, Cohen JB, et al. Development of an oncolytic HSV vector fully retargeted specifically to cellular EpCAM for virus entry and cell-to-cell spread. Gene Ther. 2016;23:479–488. doi:10.1038/gt.2016.17.
  • Wang Z, Yu B, Wang B, Yan J, Feng X, Wang L, Zhang H, Wu H, Wu J, Kong W, et al. A novel capsid-modified oncolytic recombinant adenovirus type 5 for tumor-targeting gene therapy by intravenous route. Oncotarget. 2016;7:47287–47301. doi:10.18632/oncotarget.10075.
  • Leoni V, Gatta V, Palladini A, Nicoletti G, Ranieri D, Dall'Ora M, Grosso V, Rossi M, Alviano F, Bonsi L, et al. Systemic delivery of HER2-retargeted oncolytic-HSV by mesenchymal stromal cells protects from lung and brain metastases. Oncotarget. 2015;6:34774–34787. doi:10.18632/oncotarget.5793.
  • Betancourt D, Ramos JC, Barber GN. Retargeting oncolytic vesicular stomatitis virus to human T-cell lymphotropic virus type 1-associated adult T-cell leukemia. J Virol. 2015;89:11786–11800. doi:10.1128/JVI.01356-15.
  • Gatta V, Petrovic B, Campadelli-Fiume G. The engineering of a novel ligand in gH confers to HSV an expanded tropism independent of gD activation by its receptors. PLoS Pathog. 2015;11:e1004907. doi:10.1371/journal.ppat.1004907.
  • Sakr HI, Coleman DT, Cardelli JA, Mathis JM. Characterization of an oncolytic adenovirus vector constructed to target the cMet receptor. Oncolytic Virother. 2015;4:119–132. doi:10.2147/OV.S87369.
  • Ramachandran M, Yu D, Dyczynski M, Baskaran S, Zhang L, Lulla A, Lulla V, Saul S, Nelander S, Dimberg A, et al. Safe and effective treatment of experimental neuroblastoma and glioblastoma using systemically delivered triple microRNA-detargeted oncolytic semliki forest virus. Clin Cancer Res. 2017;23:1519–1530. doi:10.1158/1078-0432.CCR-16-0925.
  • Villanueva E, Navarro P, Rovira-Rigau M, Sibilio A, Mendez R, Fillat C. Translational reprogramming in tumour cells can generate oncoselectivity in viral therapies. Nat Commun. 2017;8:14833. doi:10.1038/ncomms14833.
  • Machitani M, Sakurai F, Wakabayashi K, Tachibana M, Fujiwara T, Mizuguchi H. Enhanced oncolytic activities of the telomerase-specific replication-competent adenovirus expressing short-hairpin RNA against dicer. Mol Cancer Ther. 2017;16:251–259. doi:10.1158/1535-7163.MCT-16-0383.
  • Futami M, Sato K, Miyazaki K, Suzuki K, Nakamura T, Tojo A. Efficacy and safety of doubly-regulated vaccinia virus in a mouse xenograft model of multiple myeloma. Mol Ther Oncolytics. 2017;6:57–68. doi:10.1016/j.omto.2017.07.001.
  • Li X, Su Y, Sun B, Ji W, Peng Z, Xu Y, Wu M, Su C. An artificially designed interfering lncrna expressed by oncolytic adenovirus competitively consumes oncomirs to exert antitumor efficacy in hepatocellular carcinoma. Mol Cancer Ther. 2016;15:1436–1451. doi:10.1158/1535-7163.MCT-16-0096.
  • Takehara K, Tazawa H, Okada N, Hashimoto Y, Kikuchi S, Kuroda S, Kishimoto H, Shirakawa Y, Narii N, Mizuguchi H, et al. Targeted photodynamic virotherapy armed with a genetically encoded photosensitizer. Mol Cancer Ther. 2016;15:199–208. doi:10.1158/1535-7163.MCT-15-0344.
  • Lei W, Wang S, Yang C, Huang X, Chen Z, He W, Shen J, Liu X, Qian W. Combined expression of miR-34a and Smac mediated by oncolytic vaccinia virus synergistically promote anti-tumor effects in multiple myeloma. Sci Rep. 2016;6:32174. doi:10.1038/srep32174.
  • Zhang KX, Matsui Y, Lee C, Osamu O, Skinner L, Wang J, So A, Rennie PS, Jia WW. Intravesical treatment of advanced urothelial bladder cancers with oncolytic HSV-1 co-regulated by differentially expressed microRNAs. Gene Ther. 2016;23:460–468. doi:10.1038/gt.2016.18.
  • Delwar ZM, Liu G, Kuo Y, Lee C, Bu L, Rennie PS, Jia WW. Tumour-specific triple-regulated oncolytic herpes virus to target glioma. Oncotarget. 2016;7:28658–28669. doi:10.18632/oncotarget.8637.
  • Ruiz AJ, Russell SJ. MicroRNAs and oncolytic viruses. Curr Opin Virol. 2015;13:40–48. doi:10.1016/j.coviro.2015.03.007.
  • Su BH, Shieh GS, Tseng YL, Shiau AL, Wu CL. Etoposide enhances antitumor efficacy of MDR1-driven oncolytic adenovirus through autoupregulation of the MDR1 promoter activity. Oncotarget. 2015;6:38308–38326. doi:10.18632/oncotarget.5702.
  • Baertsch MA, Leber MF, Bossow S, Singh M, Engeland CE, Albert J, Grossardt C, Jager D, von Kalle C, Ungerechts G. MicroRNA-mediated multi-tissue detargeting of oncolytic measles virus. Cancer Gene Ther. 2014;21:373–380. doi:10.1038/cgt.2014.40.
  • Takagi-Kimura M, Yamano T, Tagawa M, Kubo S. Oncolytic virotherapy for osteosarcoma using midkine promoter-regulated adenoviruses. Cancer Gene Ther. 2014;21:126–132. doi:10.1038/cgt.2014.7.
  • Ylosmaki E, Martikainen M, Hinkkanen A, Saksela K. Attenuation of semliki forest virus neurovirulence by microRNA-mediated detargeting. J Virol. 2013;87:335–344. doi:10.1128/JVI.01940-12.
  • Zhou X, Xie G, Wang S, Wang Y, Zhang K, Zheng S, Chu L, Xiao L, Yu Y, Zhang Y, et al. Potent and specific antitumor effect for colorectal cancer by CEA and Rb double regulated oncolytic adenovirus harboring ST13 gene. PLoS One. 2012;7:e47566. doi:10.1371/journal.pone.0047566.
  • Dai B, Roife D, Kang Y, Gumin J, Rios Perez MV, Li X, Pratt M, Brekken RA, Fueyo-Margareto J, Lang FF, et al. Preclinical evaluation of sequential combination of oncolytic adenovirus delta-24-RGD and phosphatidylserine-targeting antibody in pancreatic ductal adenocarcinoma. Mol Cancer Ther. 2017;16:662–670. doi:10.1158/1535-7163.MCT-16-0526.
  • Pantelidou C, Cherubini G, Lemoine NR, Hallden G. The E1B19K-deleted oncolytic adenovirus mutant AdDelta19K sensitizes pancreatic cancer cells to drug-induced DNA-damage by down-regulating Claspin and Mre11. Oncotarget. 2016;7:15703–15724. doi:10.18632/oncotarget.7310.
  • LaRocca CJ, Han J, Salzwedel AO, Davydova J, Herzberg MC, Gopalakrishnan R, Yamamoto M. Oncolytic adenoviruses targeted to human papilloma virus-positive head and neck squamous cell carcinomas. Oral Oncol. 2016;56:25–31. doi:10.1016/j.oraloncology.2016.02.014.
  • Mansour M, Palese P, Zamarin D. Oncolytic specificity of newcastle disease virus is mediated by selectivity for apoptosis-resistant cells. J Virol. 2011;85:6015–6023. doi:10.1128/JVI.01537-10.
  • Breitbach CJ, Burke J, Jonker D, Stephenson J, Haas AR, Chow LQ, Nieva J, Hwang TH, Moon A, Patt R, et al. Intravenous delivery of a multi-mechanistic cancer-targeted oncolytic poxvirus in humans. Nature. 2011;477:99–102. doi:10.1038/nature10358.
  • Brun J, McManus D, Lefebvre C, Hu K, Falls T, Atkins H, Bell JC, McCart JA, Mahoney D, Stojdl DF. Identification of genetically modified Maraba virus as an oncolytic rhabdovirus. Mol Ther. 2010;18:1440–1449. doi:10.1038/mt.2010.103.
  • Stojdl DF, Lichty BD, tenOever BR, Paterson JM, Power AT, Knowles S, Marius R, Reynard J, Poliquin L, Atkins H, et al. VSV strains with defects in their ability to shutdown innate immunity are potent systemic anti-cancer agents. Cancer Cell. 2003;4:263–275.
  • Varghese S, Rabkin SD. Oncolytic herpes simplex virus vectors for cancer virotherapy. Cancer Gene Ther. 2002;9:967–978. doi:10.1038/sj.cgt.7700537.
  • Mineta T, Rabkin SD, Yazaki T, Hunter WD, Martuza RL. Attenuated multi-mutated herpes simplex virus-1 for the treatment of malignant gliomas. Nat Med. 1995;1:938–943.
  • Galluzzi L, Vanpouille-Box C, Bakhoum SF, Demaria S. SnapShot: CGAS-STING Signaling. Cell. 2018;173:276- e1. doi:10.1016/j.cell.2018.03.015.
  • Ying L, Cheng H, Xiong XW, Yuan L, Peng ZH, Wen ZW, Ka LJ, Xiao X, Jing C, Qian TY, et al. Interferon alpha antagonizes the anti-hepatoma activity of the oncolytic virus M1 by stimulating anti-viral immunity. Oncotarget. 2017;8:24694–24705. doi:10.18632/oncotarget.15788.
  • Xia T, Konno H, Barber GN. Recurrent loss of STING signaling in melanoma correlates with susceptibility to viral oncolysis. Cancer Res. 2016;76:6747–6759. doi:10.1158/0008-5472.CAN-16-1404.
  • Xia T, Konno H, Ahn J, Barber GN. Deregulation of STING signaling in colorectal carcinoma constrains DNA damage responses and correlates with tumorigenesis. Cell Rep. 2016;14:282–297. doi:10.1016/j.celrep.2015.12.029.
  • Danziger O, Shai B, Sabo Y, Bacharach E, Ehrlich M. Combined genetic and epigenetic interferences with interferon signaling expose prostate cancer cells to viral infection. Oncotarget. 2016;7:52115–52134. doi:10.18632/oncotarget.10313.
  • Naik S, Russell SJ. Engineering oncolytic viruses to exploit tumor specific defects in innate immune signaling pathways. Expert Opin Biol Ther. 2009;9:1163–1176. doi:10.1517/14712590903170653.
  • Kohlhapp FJ, Kaufman HL. Molecular pathways: mechanism of action for talimogene laherparepvec, a new oncolytic virus immunotherapy. Clin Cancer Res. 2016;22:1048–1054. doi:10.1158/1078-0432.CCR-15-2667.
  • Li Y, Zhang C, Chen X, Yu J, Wang Y, Yang Y, Du M, Jin H, Ma Y, He B, et al. ICP34.5 protein of herpes simplex virus facilitates the initiation of protein translation by bridging eukaryotic initiation factor 2alpha (eIF2alpha) and protein phosphatase 1. J Biol Chem. 2011;286:24785–24792. doi:10.1074/jbc.M111.232439.
  • Goldsmith K, Chen W, Johnson DC, Hendricks RL. Infected cell protein (ICP)47 enhances herpes simplex virus neurovirulence by blocking the CD8+ T cell response. J Exp Med. 1998;187:341–348.
  • Jacobson BA, Sadiq AA, Tang S, Jay-Dixon J, Patel MR, Drees J, Sorenson BS, Russell SJ, Kratzke RA. Cap-dependent translational control of oncolytic measles virus infection in malignant mesothelioma. Oncotarget. 2017;8:63096–63109. doi:10.18632/oncotarget.18656.
  • Liao Y, Wang HX, Mao X, Fang H, Wang H, Li Y, Sun Y, Meng C, Tan L, Song C, et al. RIP1 is a central signaling protein in regulation of TNF-alpha/TRAIL mediated apoptosis and necroptosis during newcastle disease virus infection. Oncotarget. 2017;8:43201–43217. doi:10.18632/oncotarget.17970.
  • Garant KA, Shmulevitz M, Pan L, Daigle RM, Ahn DG, Gujar SA, Lee PW. Oncolytic reovirus induces intracellular redistribution of Ras to promote apoptosis and progeny virus release. Oncogene. 2016;35:771–782. doi:10.1038/onc.2015.136.
  • McEntee G, Kyula JN, Mansfield D, Smith H, Wilkinson M, Gregory C, Roulstone V, Coffey M, Harrington KJ. Enhanced cytotoxicity of reovirus and radiotherapy in melanoma cells is mediated through increased viral replication and mitochondrial apoptotic signalling. Oncotarget. 2016;7:48517–48532. doi:10.18632/oncotarget.10365.
  • Naik S, Galyon GD, Jenks NJ, Steele MB, Miller AC, Allstadt SD, Suksanpaisan L, Peng KW, Federspiel MJ, Russell SJ, et al. Comparative oncology evaluation of intravenous recombinant oncolytic vesicular stomatitis virus therapy in spontaneous canine cancer. Mol Cancer Ther. 2017;17:316–326.
  • Dobson CC, Naing T, Beug ST, Faye MD, Chabot J, St-Jean M, Walker DE, LaCasse EC, Stojdl DF, Korneluk RG, et al. Oncolytic virus synergizes with Smac mimetic compounds to induce rhabdomyosarcoma cell death in a syngeneic murine model. Oncotarget. 2017;8:3495–3508. doi:10.18632/oncotarget.13849.
  • Jahan N, Lee JM, Shah K, Wakimoto H. Therapeutic targeting of chemoresistant and recurrent glioblastoma stem cells with a proapoptotic variant of oncolytic herpes simplex virus. Int J Cancer. 2017;141:1671–1681. doi:10.1002/ijc.30811.
  • Tan Y, Lin Y, Li K, Xiao X, Liang J, Cai J, Guo L, Li C, Zhu W, Xing F, et al. Selective Antagonism of Bcl-xL potentiates M1 oncolysis by enhancing mitochondrial apoptosis. Hum Gene Ther. 2017. doi:10.1089/hum.2017.055.
  • Binz E, Berchtold S, Beil J, Schell M, Geisler C, Smirnow I, Lauer UM. Chemovirotherapy of pancreatic adenocarcinoma by combining oncolytic vaccinia virus GLV-1h68 with nab-paclitaxel plus gemcitabine. Mol Ther Oncolytics. 2017;6:10–21. doi:10.1016/j.omto.2017.04.001.
  • Bressy C, Hastie E, Grdzelishvili VZ. Combining oncolytic virotherapy with p53 tumor suppressor gene therapy. Mol Ther Oncolytics. 2017;5:20–40. doi:10.1016/j.omto.2017.03.002.
  • Le Boeuf F, Gebremeskel S, McMullen N, He H, Greenshields AL, Hoskin DW, Bell JC, Johnston B, Pan C, Duncan R. Reovirus FAST protein enhances vesicular stomatitis virus oncolytic virotherapy in primary and metastatic tumor models. Mol Ther Oncolytics. 2017;6:80–89. doi:10.1016/j.omto.2017.08.001.
  • Kim DS, Dastidar H, Zhang C, Zemp FJ, Lau K, Ernst M, Rakic A, Sikdar S, Rajwani J, Naumenko V, et al. Smac mimetics and oncolytic viruses synergize in driving anticancer T-cell responses through complementary mechanisms. Nat Commun. 2017;8:344. doi:10.1038/s41467-017-00324-x.
  • Currier MA, Sprague L, Rizvi TA, Nartker B, Chen CY, Wang PY, Hutzen BJ, Franczek MR, Patel AV, Chaney KE, et al. Aurora A kinase inhibition enhances oncolytic herpes virotherapy through cytotoxic synergy and innate cellular immune modulation. Oncotarget. 2017;8:17412–17427. doi:10.18632/oncotarget.14885.
  • Fend L, Yamazaki T, Remy C, Fahrner C, Gantzer M, Nourtier V, Preville X, Quemeneur E, Kepp O, Adam J, et al. Immune checkpoint blockade, immunogenic chemotherapy or ifn-alpha blockade boost the local and abscopal effects of oncolytic virotherapy. Cancer Res. 2017;77:4146–4157. doi:10.1158/0008-5472.CAN-16-2165.
  • Fend L, Remy-Ziller C, Foloppe J, Kempf J, Cochin S, Barraud L, Accart N, Erbs P, Fournel S, Preville X. Oncolytic virotherapy with an armed vaccinia virus in an orthotopic model of renal carcinoma is associated with modification of the tumor microenvironment. Oncoimmunology. 2016;5:e1080414. doi:10.1080/2162402X.2015.1080414.
  • Wilkinson MJ, Smith HG, McEntee G, Kyula-Currie J, Pencavel TD, Mansfield DC, Khan AA, Roulstone V, Hayes AJ, Harrington KJ. Oncolytic vaccinia virus combined with radiotherapy induces apoptotic cell death in sarcoma cells by down-regulating the inhibitors of apoptosis. Oncotarget. 2016;7:81208–81222. doi:10.18632/oncotarget.12820.
  • Pisklakova A, McKenzie B, Zemp F, Lun X, Kenchappa RS, Etame AB, Rahman MM, Reilly K, Pilon-Thomas S, McFadden G, et al. M011L-deficient oncolytic myxoma virus induces apoptosis in brain tumor-initiating cells and enhances survival in a novel immunocompetent mouse model of glioblastoma. Neuro Oncol. 2016; doi:10.1093/neuonc/now006.
  • Ashshi AM, El-Shemi AG, Dmitriev IP, Kashentseva EA, Curiel DT. Combinatorial strategies based on CRAd-IL24 and CRAd-ING4 virotherapy with anti-angiogenesis treatment for ovarian cancer. J Ovarian Res. 2016;9:38. doi:10.1186/s13048-016-0248-5.
  • Yoo JY, Jaime-Ramirez AC, Bolyard C, Dai H, Nallanagulagari T, Wojton J, Hurwitz BS, Relation T, Lee TJ, Lotze MT, et al. Bortezomib treatment sensitizes oncolytic HSV-1-treated tumors to NK cell immunotherapy. Clin Cancer Res. 2016;22:5265–5276. doi:10.1158/1078-0432.CCR-16-1003.
  • Mansfield DC, Kyula JN, Rosenfelder N, Chao-Chu J, Kramer-Marek G, Khan AA, Roulstone V, McLaughlin M, Melcher AA, Vile RG, et al. Oncolytic vaccinia virus as a vector for therapeutic sodium iodide symporter gene therapy in prostate cancer. Gene Ther. 2016;23:357–368. doi:10.1038/gt.2016.5.
  • Garofalo M, Iovine B, Kuryk L, Capasso C, Hirvinen M, Vitale A, Yliperttula M, Bevilacqua MA, Cerullo V. Oncolytic adenovirus loaded with l-carnosine as novel strategy to enhance the antitumor activity. Mol Cancer Ther. 2016;15:651–660. doi:10.1158/1535-7163.MCT-15-0559.
  • Al-Shammari AM, Rameez H, Al-Taee MF. Newcastle disease virus, rituximab, and doxorubicin combination as anti-hematological malignancy therapy. Oncolytic Virother. 2016;5:27–34. doi:10.2147/OV.S95250.
  • Osaki S, Tazawa H, Hasei J, Yamakawa Y, Omori T, Sugiu K, Komatsubara T, Fujiwara T, Sasaki T, Kunisada T, et al. Ablation of MCL1 expression by virally induced microRNA-29 reverses chemoresistance in human osteosarcomas. Sci Rep. 2016;6:28953. doi:10.1038/srep28953.
  • Yang Y, Xu W, Neill T, Hu Z, Wang CH, Xiao X, Stock SR, Guise T, Yun CO, Brendler CB, et al. Systemic delivery of an oncolytic adenovirus expressing decorin for the treatment of breast cancer bone metastases. Hum Gene Ther. 2015;26:813–825. doi:10.1089/hum.2015.098.
  • Patel MR, Jacobson BA, Ji Y, Drees J, Tang S, Xiong K, Wang H, Prigge JE, Dash AS, Kratzke AK, et al. Vesicular stomatitis virus expressing interferon-beta is oncolytic and promotes antitumor immune responses in a syngeneic murine model of non-small cell lung cancer. Oncotarget. 2015;6:33165–33177. doi:10.18632/oncotarget.5320.
  • Fukuhara H, Ino Y, Todo T. Oncolytic virus therapy: A new era of cancer treatment at dawn. Cancer Sci. 2016;107:1373–1379. doi:10.1111/cas.13027.
  • Taipale K, Liikanen I, Juhila J, Turkki R, Tahtinen S, Kankainen M, Vassilev L, Ristimaki A, Koski A, Kanerva A, et al. Chronic activation of innate immunity correlates with poor prognosis in cancer patients treated with oncolytic adenovirus. Mol Ther. 2016;24:175–183. doi:10.1038/mt.2015.143.
  • Holl EK, Brown MC, Boczkowski D, McNamara MA, George DJ, Bigner DD, Gromeier M, Nair SK. Recombinant oncolytic poliovirus, PVSRIPO, has potent cytotoxic and innate inflammatory effects, mediating therapy in human breast and prostate cancer xenograft models. Oncotarget. 2016;7:79828–79841. doi:10.18632/oncotarget.12975.
  • Prestwich RJ, Harrington KJ, Pandha HS, Vile RG, Melcher AA, Errington F. Oncolytic viruses: a novel form of immunotherapy. Expert Rev Anticancer Ther. 2008;8:1581–1588. doi:10.1586/14737140.8.10.1581.
  • Rojas JJ, Sampath P, Bonilla B, Ashley A, Hou W, Byrd D, Thorne SH. Manipulating TLR signaling increases the anti-tumor T cell response induced by viral cancer therapies. Cell Rep. 2016;15:264–273. doi:10.1016/j.celrep.2016.03.017.
  • Nosaki K, Hamada K, Takashima Y, Sagara M, Matsumura Y, Miyamoto S, Hijikata Y, Okazaki T, Nakanishi Y, Tani K. A novel, polymer-coated oncolytic measles virus overcomes immune suppression and induces robust antitumor activity. Mol Ther Oncolytics. 2016;3:16022. doi:10.1038/mto.2016.22.
  • Martinez-Velez N, Xipell E, Vera B, Acanda de la Rocha A, Zalacain M, Marrodan L, Gonzalez-Huarriz M, Toledo G, Cascallo M, Alemany R, et al. The oncolytic adenovirus VCN-01 as therapeutic approach against pediatric osteosarcoma. Clin Cancer Res. 2016;22:2217–2225. doi:10.1158/1078-0432.CCR-15-1899.
  • Du W, Seah I, Bougazzoul O, Choi G, Meeth K, Bosenberg MW, Wakimoto H, Fisher D, Shah K. Stem cell-released oncolytic herpes simplex virus has therapeutic efficacy in brain metastatic melanomas. Proc Natl Acad Sci U S A. 2017;114:E6157–E65. doi:10.1073/pnas.1700363114.
  • Kaczorowski A, Hammer K, Liu L, Villhauer S, Nwaeburu C, Fan P, Zhao Z, Gladkich J, Gross W, Nettelbeck DM, et al. Delivery of improved oncolytic adenoviruses by mesenchymal stromal cells for elimination of tumorigenic pancreatic cancer cells. Oncotarget. 2016;7:9046–9059. doi:10.18632/oncotarget.7031.
  • Melen GJ, Franco-Luzon L, Ruano D, Gonzalez-Murillo A, Alfranca A, Casco F, Lassaletta A, Alonso M, Madero L, Alemany R, et al. Influence of carrier cells on the clinical outcome of children with neuroblastoma treated with high dose of oncolytic adenovirus delivered in mesenchymal stem cells. Cancer Lett. 2016;371:161–170. doi:10.1016/j.canlet.2015.11.036.
  • Yuan X, Zhang Q, Li Z, Zhang X, Bao S, Fan D, Ru Y, Dong S, Zhang Y, Ye Z, et al. Mesenchymal stem cells deliver and release conditionally replicative adenovirus depending on hepatic differentiation to eliminate hepatocellular carcinoma cells specifically. Cancer Lett. 2016;381:85–95. doi:10.1016/j.canlet.2016.07.019.
  • Bauer DF, Pereboeva L, Gillespie GY, Cloud GA, Elzafarany O, Langford C, Markert JM, Lamb LS, Jr. Effect of HSV-IL12 loaded tumor cell-based vaccination in a mouse model of high-grade neuroblastoma. J Immunol Res. 2016;2016:2568125. doi:10.1155/2016/2568125.
  • Kazimirsky G, Jiang W, Slavin S, Ziv-Av A, Brodie C. Mesenchymal stem cells enhance the oncolytic effect of newcastle disease virus in glioma cells and glioma stem cells via the secretion of TRAIL. Stem Cell Res Ther. 2016;7:149. doi:10.1186/s13287-016-0414-0.
  • Dold C, Rodriguez Urbiola C, Wollmann G, Egerer L, Muik A, Bellmann L, Fiegl H, Marth C, Kimpel J, von Laer D. Application of interferon modulators to overcome partial resistance of human ovarian cancers to VSV-GP oncolytic viral therapy. Mol Ther Oncolytics. 2016;3:16021. doi:10.1038/mto.2016.21.
  • Li K, Liang J, Lin Y, Zhang H, Xiao X, Tan Y, Cai J, Zhu W, Xing F, Hu J, et al. A classical PKA inhibitor increases the oncolytic effect of M1 virus via activation of exchange protein directly activated by cAMP 1. Oncotarget. 2016;7:48443–48455. doi:10.18632/oncotarget.10305.
  • Evgin L, Ilkow CS, Bourgeois-Daigneault MC, de Souza CT, Stubbert L, Huh MS, Jennings VA, Marguerie M, Acuna SA, Keller BA, et al. Complement inhibition enables tumor delivery of LCMV glycoprotein pseudotyped viruses in the presence of antiviral antibodies. Mol Ther Oncolytics. 2016;3:16027. doi:10.1038/mto.2016.27.
  • Jaime-Ramirez AC, Yu JG, Caserta E, Yoo JY, Zhang J, Lee TJ, Hofmeister C, Lee JH, Kumar B, Pan Q, et al. Reolysin and histone deacetylase inhibition in the treatment of head and neck squamous cell carcinoma. Mol Ther Oncolytics. 2017;5:87–96. doi:10.1016/j.omto.2017.05.002.
  • Stiff A, Caserta E, Sborov DW, Nuovo GJ, Mo X, Schlotter SY, Canella A, Smith E, Badway J, Old M, et al. Histone deacetylase inhibitors enhance the therapeutic potential of reovirus in multiple myeloma. Mol Cancer Ther. 2016;15:830–841. doi:10.1158/1535-7163.MCT-15-0240-T.
  • Han J, Chen X, Chu J, Xu B, Meisen WH, Chen L, Zhang L, Zhang J, He X, Wang QE, et al. TGFbeta treatment enhances glioblastoma virotherapy by inhibiting the innate immune response. Cancer Res. 2015;75:5273–5282. doi:10.1158/0008-5472.CAN-15-0894.
  • Chard LS, Maniati E, Wang P, Zhang Z, Gao D, Wang J, Cao F, Ahmed J, El Khouri M, Hughes J, et al. A vaccinia virus armed with interleukin-10 is a promising therapeutic agent for treatment of murine pancreatic cancer. Clin Cancer Res. 2015;21:405–416. doi:10.1158/1078-0432.CCR-14-0464.
  • Pease DF, Kratzke RA. Oncolytic viral therapy for mesothelioma. Front Oncol. 2017;7:179. doi:10.3389/fonc.2017.00179.
  • Rehman H, Silk AW, Kane MP, Kaufman HL. Into the clinic: talimogene laherparepvec (T-VEC), a first-in-class intratumoral oncolytic viral therapy. J Immunother Cancer. 2016;4:53. doi:10.1186/s40425-016-0158-5.
  • Sze DY, Reid TR, Rose SC. Oncolytic virotherapy. J Vasc Interv Radiol. 2013;24:1115–1122. doi:10.1016/j.jvir.2013.05.040.
  • Ferguson MS, Lemoine NR, Wang Y. Systemic delivery of oncolytic viruses: hopes and hurdles. Adv Virol. 2012;2012:805629. doi:10.1155/2012/805629.
  • Jung KH, Choi IK, Lee HS, Yan HH, Son MK, Ahn HM, Hong J, Yun CO, Hong SS. Oncolytic adenovirus expressing relaxin (YDC002) enhances therapeutic efficacy of gemcitabine against pancreatic cancer. Cancer Lett. 2017;396:155–166. doi:10.1016/j.canlet.2017.03.009.
  • Yumul R, Richter M, Lu ZZ, Saydaminova K, Wang H, Wang CH, Carter D, Lieber A. Epithelial junction opener improves oncolytic adenovirus therapy in mouse tumor models. Hum Gene Ther. 2016;27:325–337. doi:10.1089/hum.2016.022.
  • Tedcastle A, Illingworth S, Brown A, Seymour LW, Fisher KD. Actin-resistant DNAse I expression from oncolytic adenovirus enadenotucirev enhances its intratumoral spread and reduces tumor growth. Mol Ther. 2016;24:796–804. doi:10.1038/mt.2015.233.
  • Kim M, Nitschke M, Sennino B, Murer P, Schriver BJ, Bell AM, Subramanian A, McDonald CE, Wang J, Cha H, et al. Amplification of oncolytic vaccinia virus widespread tumor cell killing by sunitinib through multiple mechanisms. Cancer Res. 2017;78:922–937.
  • Arulanandam R, Batenchuk C, Angarita FA, Ottolino-Perry K, Cousineau S, Mottashed A, et al. VEGF-Mediated Induction of PRD1-BF1/Blimp1 expression sensitizes tumor vasculature to oncolytic virus infection. Cancer Cell. 2015;28:210–224. doi:10.1016/j.ccell.2015.06.009.
  • Hou W, Chen H, Rojas J, Sampath P, Thorne SH. Oncolytic vaccinia virus demonstrates antiangiogenic effects mediated by targeting of VEGF. Int J Cancer. 2014;135:1238–1246. doi:10.1002/ijc.28747.
  • Breitbach CJ, Arulanandam R, De Silva N, Thorne SH, Patt R, Daneshmand M, Moon A, Ilkow C, Burke J, Hwang TH, et al. Oncolytic vaccinia virus disrupts tumor-associated vasculature in humans. Cancer Res. 2013;73:1265–1275. doi:10.1158/0008-5472.CAN-12-2687.
  • Breitbach CJ, De Silva NS, Falls TJ, Aladl U, Evgin L, Paterson J, Sun YY, Roy DG, Rintoul JL, Daneshmand M, et al. Targeting tumor vasculature with an oncolytic virus. Mol Ther. 2011;19:886–894. doi:10.1038/mt.2011.26.
  • Bolyard C, Meisen WH, Banasavadi-Siddegowda Y, Hardcastle J, Yoo JY, Wohleb ES, Wojton J, Yu JG, Dubin S, Khosla M, et al. BAI1 orchestrates macrophage inflammatory response to HSV infection-implications for oncolytic viral therapy. Clin Cancer Res. 2017;23:1809–1819. doi:10.1158/1078-0432.CCR-16-1818.
  • Toro Bejarano M, Merchan JR. Targeting tumor vasculature through oncolytic virotherapy: recent advances. Oncolytic Virother. 2015;4:169–181. doi:10.2147/OV.S66045.
  • Gujar S, Pol JG, Kim Y, Lee PW, Kroemer G. Antitumor benefits of antiviral immunity: an underappreciated aspect of oncolytic virotherapies. Trends Immunol. 2017.
  • Aurelian L. Oncolytic viruses as immunotherapy: progress and remaining challenges. Onco Targets Ther. 2016;9:2627–2637. doi:10.2147/OTT.S63049.
  • Delaunay T, Violland M, Boisgerault N, Dutoit S, Vignard V, Munz C, Gannage M, Dreno B, Vaivode K, Pjanova D, et al. Oncolytic viruses sensitize human tumor cells for NY-ESO-1 tumor antigen recognition by CD4+ effector T cells. Oncoimmunology. 2018;7:e1407897. doi:10.1080/2162402X.2017.1407897.
  • van Vloten JP, Workenhe ST, Wootton SK, Mossman KL, Bridle BW. Critical interactions between immunogenic cancer cell death, oncolytic viruses, and the immune system define the rational design of combination immunotherapies. J Immunol. 2018;200:450–458. doi:10.4049/jimmunol.1701021.
  • De Munck J, Binks A, McNeish IA, Aerts JL. Oncolytic virus-induced cell death and immunity: a match made in heaven? J Leukoc Biol. 2017;102:631–643. doi:10.1189/jlb.5RU0117-040R.
  • Dyer A, Di Y, Calderon H, Illingworth S, Kueberuwa G, Tedcastle A, Jakeman P, Chia SL, Brown A, Silva MA, et al. Oncolytic group B adenovirus enadenotucirev mediates non-apoptotic cell death with membrane disruption and release of inflammatory mediators. Mol Ther Oncolytics. 2017;4:18–30. doi:10.1016/j.omto.2016.11.003.
  • Delaunay T, Violland M, Boisgerault N, Dutoit S, Vignard V, Münz C, et al. Oncolytic viruses sensitize human tumor cells for NY-ESO-1 tumor antigen recognition by CD4+ effector T cells. Oncoimmunology. 2017.
  • Workenhe ST, Mossman KL. Oncolytic virotherapy and immunogenic cancer cell death: sharpening the sword for improved cancer treatment strategies. Mol Ther. 2014;22:251–256. doi:10.1038/mt.2013.220.
  • Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, Agostinis P, Apetoh L, Aranda F, Barnaba V, Bloy N, et al. Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology. 2014;3:e955691. doi:10.4161/21624011.2014.955691.
  • Cauwels A, Van Lint S, Garcin G, Bultinck J, Paul F, Gerlo S, Van der Heyden J, Bordat Y, Catteeuw D, De Cauwer L, et al. A safe and highly efficient tumor-targeted type I interferon immunotherapy depends on the tumor microenvironment. Oncoimmunology. 2017;7:e1398876.
  • Riganti C, Lingua MF, Salaroglio IC, Falcomatà C, Righi L, Morena D, Picca F, Oddo D, Kopecka J, Pradotto M, et al. Bromodomain inhibition exerts its therapeutic potential in malignant pleural mesothelioma by promoting immunogenic cell death and changing the tumor immune-environment. Oncoimmunology. 2017;e1398874.
  • Showalter A, Limaye A, Oyer JL, Igarashi R, Kittipatarin C, Copik AJ, Khaled AR. Cytokines in immunogenic cell death: applications for cancer immunotherapy. Cytokine. 2017;97:123–132. doi:10.1016/j.cyto.2017.05.024.
  • Galluzzi L, Kroemer G. Calreticulin and type I interferon: an unsuspected connection. Oncoimmunology. 2017;6:e1288334. doi:10.1080/2162402X.2017.1288334.
  • Bourgeois-Daigneault MC, Roy DG, Aitken AS, El Sayes N, Martin NT, Varette O, Falls T, St-Germain LE, Pelin A, Lichty BD, et al. Neoadjuvant oncolytic virotherapy before surgery sensitizes triple-negative breast cancer to immune checkpoint therapy. Sci Transl Med. 2018;10.
  • Samson A, Scott KJ, Taggart D, West EJ, Wilson E, Nuovo GJ, Thomson S, Corns R, Mathew RK, Fuller MJ, et al. Intravenous delivery of oncolytic reovirus to brain tumor patients immunologically primes for subsequent checkpoint blockade. Sci Transl Med. 2018;10. doi:10.1126/scitranslmed.aao1641.
  • Achard C, Guillerme JB, Bruni D, Boisgerault N, Combredet C, Tangy F, Jouvenet N, Gregoire M, Fonteneau JF. Oncolytic measles virus induces tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated cytotoxicity by human myeloid and plasmacytoid dendritic cells. Oncoimmunology. 2017;6:e1261240. doi:10.1080/2162402X.2016.1261240.
  • Li X, Wang P, Li H, Du X, Liu M, Huang Q, Wang Y, Wang S. The efficacy of oncolytic adenovirus is mediated by T-cell responses against virus and tumor in syrian hamster model. Clin Cancer Res. 2017;23:239–249. doi:10.1158/1078-0432.CCR-16-0477.
  • Moesta AK, Cooke K, Piasecki J, Mitchell P, Rottman JB, Fitzgerald K, Zhan J, Yang B, Le T, Belmontes B, et al. Local delivery of oncoVEXmGM-CSF generates systemic antitumor immune responses enhanced by cytotoxic T-lymphocyte-associated protein blockade. Clin Cancer Res. 2017;23:6190–6202. doi:10.1158/1078-0432.CCR-17-0681.
  • Hock K, Laengle J, Kuznetsova I, Egorov A, Hegedus B, Dome B, Wekerle T, Sachet M, Bergmann M. Oncolytic influenza A virus expressing interleukin-15 decreases tumor growth in vivo. Surgery. 2017;161:735–746. doi:10.1016/j.surg.2016.08.045.
  • Aurelian L, Bollino D, Colunga A. The oncolytic virus DeltaPK has multimodal anti-tumor activity. Pathog Dis. 2016;74.doi:10.1093/femspd/ftw050.
  • Francis L, Guo ZS, Liu Z, Ravindranathan R, Urban JA, Sathaiah M, Magge D, Kalinski P, Bartlett DL. Modulation of chemokines in the tumor microenvironment enhances oncolytic virotherapy for colorectal cancer. Oncotarget. 2016;7:22174–22185. doi:10.18632/oncotarget.7907.
  • Passaro C, Borriello F, Vastolo V, Di Somma S, Scamardella E, Gigantino V, Franco R, Marone G, Portella G. The oncolytic virus dl922-947 reduces IL-8/CXCL8 and MCP-1/CCL2 expression and impairs angiogenesis and macrophage infiltration in anaplastic thyroid carcinoma. Oncotarget. 2016;7:1500–1515. doi:10.18632/oncotarget.6430.
  • Schwaiger T, Knittler MR, Grund C, Roemer-Oberdoerfer A, Kapp JF, Lerch MM, Mettenleiter TC, Mayerle J, Blohm U. Newcastle disease virus mediates pancreatic tumor rejection via NK cell activation and prevents cancer relapse by prompting adaptive immunity. Int J Cancer. 2017;141:2505–2516. doi:10.1002/ijc.31026.
  • Jiang H, Rivera-Molina Y, Gomez-Manzano C, Clise-Dwyer K, Bover L, Vence LM, Yuan Y, Lang FF, Toniatti C, Hossain MB, et al. Oncolytic adenovirus and tumor-targeting immune modulatory therapy improve autologous cancer vaccination. Cancer Res. 2017;77:3894–3907. doi:10.1158/0008-5472.CAN-17-0468.
  • Hamilton JR, Vijayakumar G, Palese P. A recombinant antibody-expressing influenza virus delays tumor growth in a mouse model. Cell Rep. 2018;22:1–7. doi:10.1016/j.celrep.2017.12.025.
  • Zamarin D, Holmgaard RB, Ricca J, Plitt T, Palese P, Sharma P, Merghoub T, Wolchok JD, Allison JP. Intratumoral modulation of the inducible co-stimulator ICOS by recombinant oncolytic virus promotes systemic anti-tumour immunity. Nat Commun. 2017;8:14340. doi:10.1038/ncomms14340.
  • Eriksson E, Milenova I, Wenthe J, Stahle M, Leja-Jarblad J, Ullenhag G, Dimberg A, Moreno R, Alemany R, Loskog A. Shaping the tumor stroma and sparking immune activation by CD40 and 4-1BB signaling induced by an armed oncolytic virus. Clin Cancer Res. 2017;23:5846–5857. doi:10.1158/1078-0432.CCR-17-0285.
  • Kim SY, Kang D, Choi HJ, Joo Y, Kim JH, Song JJ. Prime-boost immunization by both DNA vaccine and oncolytic adenovirus expressing GM-CSF and shRNA of TGF-beta2 induces anti-tumor immune activation. Oncotarget. 2017;8:15858–15877. doi:10.18632/oncotarget.15008.
  • Bartee MY, Dunlap KM, Bartee E. Tumor-Localized secretion of soluble PD1 enhances oncolytic virotherapy. Cancer Res. 2017;77:2952–2963. doi:10.1158/0008-5472.CAN-16-1638.
  • Rosewell Shaw A, Porter CE, Watanabe N, Tanoue K, Sikora A, Gottschalk S, Brenner MK, Suzuki M. Adenovirotherapy delivering cytokine and checkpoint inhibitor augments CAR T cells against metastatic head and neck cancer. Mol Ther. 2017; doi:10.1016/j.ymthe.2017.09.010.
  • Moon E, Wang L-CS, Bekdache K, Lynn R, Lo A, Thorne S, Albelda S. Intra-tumoral delivery of CXCL11 via a vaccinia virus, but not by modified T cells, enhances the efficacy of adoptive T cell therapy and vaccines. Oncoimmunology. 2017.
  • Veinalde R, Grossardt C, Hartmann L, Bourgeois-Daigneault MC, Bell JC, Jager D, von Kalle C, Ungerechts G, Engeland CE. Oncolytic measles virus encoding interleukin-12 mediates potent antitumor effects through T cell activation. Oncoimmunology. 2017;6:e1285992. doi:10.1080/2162402X.2017.1285992.
  • Ren J, Gwin WR, Zhou X, Wang X, Huang H, Jiang N, Zhou L, Agarwal P, Hobeika A, Crosby E, et al. Adaptive T cell responses induced by oncolytic herpes simplex virus-granulocyte macrophage-colony-stimulating factor therapy expanded by dendritic cell and cytokine-induced killer cell adoptive therapy. Oncoimmunology. 2017;6:e1264563. doi:10.1080/2162402X.2016.1264563.
  • Zafar S, Parviainen S, Siurala M, Hemminki O, Havunen R, Tahtinen S, Bramante S, Vassilev L, Wang H, Lieber A, et al. Intravenously usable fully serotype 3 oncolytic adenovirus coding for CD40L as an enabler of dendritic cell therapy. Oncoimmunology. 2017;6:e1265717. doi:10.1080/2162402X.2016.1265717.
  • Freedman JD, Freedman JD, Hagel J, Scott EM, Psallidas I, Gupta A, Spiers L, Miller P, Kanellakis N, Ashfield R, Fisher KD, et al. Oncolytic adenovirus expressing bispecific antibody targets T-cell cytotoxicity in cancer biopsies. EMBO Mol Med. 2017;9:1067–1087. doi:10.15252/emmm.201707567.
  • Dinsart C, Pervolaraki K, Stroh-Dege A, Lavie M, Ronsse I, Rommelaere J, Van Damme J, Van Raemdonck K, Struyf S. Recombinant parvoviruses armed to deliver CXCL4L1 and CXCL10 are impaired in their antiangiogenic and antitumoral effects in a kaposi sarcoma tumor model due to the chemokines’ interference with the virus cycle. Hum Gene Ther. 2017;28:295–306. doi:10.1089/hum.2016.108.
  • Hutzen B, Chen CY, Wang PY, Sprague L, Swain HM, Love J, Conner J, Boon L, Cripe TP. TGF-beta inhibition improves oncolytic herpes viroimmunotherapy in murine models of rhabdomyosarcoma. Mol Ther Oncolytics. 2017;7:17–26. doi:10.1016/j.omto.2017.09.001.
  • Ren G, Tian G, Liu Y, He J, Gao X, Yu Y, Liu X, Zhang X, Sun T, Liu S, et al. Recombinant newcastle disease virus encoding IL-12 and/or IL-2 as potential candidate for hepatoma carcinoma therapy. Technol Cancer Res Treat. 2016;15:NP83–94. doi:10.1177/1533034615601521.
  • Komorowski MP, McGray AR, Kolakowska A, Eng K, Gil M, Opyrchal M, Litwinska B, Nemeth MJ, Odunsi KO, Kozbor D. Reprogramming antitumor immunity against chemoresistant ovarian cancer by a CXCR4 antagonist-armed viral oncotherapy. Mol Ther Oncolytics. 2016;3:16034. doi:10.1038/mto.2016.34.
  • Liikanen I, Tahtinen S, Guse K, Gutmann T, Savola P, Oksanen M, Kanerva A, Hemminki A. Oncolytic adenovirus expressing monoclonal antibody trastuzumab for treatment of HER2-positive cancer. Mol Cancer Ther. 2016;15:2259–2269. doi:10.1158/1535-7163.MCT-15-0819.
  • Kochneva G, Sivolobova G, Tkacheva A, Grazhdantseva A, Troitskaya O, Nushtaeva A, Tkachenko A, Kuligina E, Richter V, Koval O. Engineering of double recombinant vaccinia virus with enhanced oncolytic potential for solid tumor virotherapy. Oncotarget. 2016;7:74171–74188. doi:10.18632/oncotarget.12367.
  • Bramante S, Koski A, Liikanen I, Vassilev L, Oksanen M, Siurala M, Heiskanen R, Hakonen T, Joensuu T, Kanerva A, et al. Oncolytic virotherapy for treatment of breast cancer, including triple-negative breast cancer. Oncoimmunology. 2016;5:e1078057. doi:10.1080/2162402X.2015.1078057.
  • Kuryk L, Haavisto E, Garofalo M, Capasso C, Hirvinen M, Pesonen S, Ranki T, Vassilev L, Cerullo V. Synergistic anti-tumor efficacy of immunogenic adenovirus ONCOS-102 (Ad5/3-D24-GM-CSF) and standard of care chemotherapy in preclinical mesothelioma model. Int J Cancer. 2016;139:1883–1893. doi:10.1002/ijc.30228.
  • Kleinpeter P, Fend L, Thioudellet C, Geist M, Sfrontato N, Koerper V, Fahrner C, Schmitt D, Gantzer M, Remy-Ziller C, et al. Vectorization in an oncolytic vaccinia virus of an antibody, a fab and a scFv against programmed cell death −1 (PD-1) allows their intratumoral delivery and an improved tumor-growth inhibition. Oncoimmunology. 2016;5:e1220467. doi:10.1080/2162402X.2016.1220467.
  • Liu Z, Ravindranathan R, Li J, Kalinski P, Guo ZS, Bartlett DL. CXCL11-Armed oncolytic poxvirus elicits potent antitumor immunity and shows enhanced therapeutic efficacy. Oncoimmunology. 2016;5:e1091554. doi:10.1080/2162402X.2015.1091554.
  • Bourgeois-Daigneault MC, Roy DG, Falls T, Twumasi-Boateng K, St-Germain LE, Marguerie M, Garcia V, Selman M, Jennings VA, Pettigrew J, et al. Oncolytic vesicular stomatitis virus expressing interferon-gamma has enhanced therapeutic activity. Mol Ther Oncolytics. 2016;3:16001. doi:10.1038/mto.2016.1.
  • Hirvinen M, Capasso C, Guse K, Garofalo M, Vitale A, Ahonen M, Kuryk L, Vaha-Koskela M, Hemminki A, Fortino V, et al. Expression of DAI by an oncolytic vaccinia virus boosts the immunogenicity of the virus and enhances antitumor immunity. Mol Ther Oncolytics. 2016;3:16002. doi:10.1038/mto.2016.2.
  • Capasso C, Hirvinen M, Garofalo M, Romaniuk D, Kuryk L, Sarvela T, Vitale A, Antopolsky M, Magarkar A, Viitala T, et al. Oncolytic adenoviruses coated with MHC-I tumor epitopes increase the antitumor immunity and efficacy against melanoma. Oncoimmunology. 2016;5:e1105429. doi:10.1080/2162402X.2015.1105429.
  • Atherton MJ, Stephenson KB, Pol J, Wang F, Lefebvre C, Stojdl DF, Nikota JK, Dvorkin-Gheva A, Nguyen A, Chen L, et al. Customized Viral Immunotherapy for HPV-Associated Cancer. Cancer Immunol Res. 2017;5:847–859. doi:10.1158/2326-6066.CIR-17-0102.
  • Ilett E, Kottke T, Thompson J, Rajani K, Zaidi S, Evgin L, Coffey M, Ralph C, Diaz R, Pandha H, et al. Prime-boost using separate oncolytic viruses in combination with checkpoint blockade improves anti-tumour therapy. Gene Ther. 2017;24:21–30. doi:10.1038/gt.2016.70.
  • Pol JG, Zhang L, Bridle BW, Stephenson KB, Resseguier J, Hanson S, Chen L, Kazdhan N, Bramson JL, Stojdl DF, et al. Maraba virus as a potent oncolytic vaccine vector. Mol Ther. 2014;22:420–429. doi:10.1038/mt.2013.249.
  • Boisgerault N, Kottke T, Pulido J, Thompson J, Diaz RM, Rommelfanger-Konkol D, Embry A, Saenz D, Poeschla E, Pandha H, et al. Functional cloning of recurrence-specific antigens identifies molecular targets to treat tumor relapse. Mol Ther. 2013;21:1507–1516. doi:10.1038/mt.2013.116.
  • Bridle BW, Clouthier D, Zhang L, Pol J, Chen L, Lichty BD, Bramson JL, Wan Y. Oncolytic vesicular stomatitis virus quantitatively and qualitatively improves primary CD8 T-cell responses to anticancer vaccines. Oncoimmunology. 2013;2:e26013. doi:10.4161/onci.26013.
  • Pulido J, Kottke T, Thompson J, Galivo F, Wongthida P, Diaz RM, Rommelfanger D, Ilett E, Pease L, Pandha H, et al. Using virally expressed melanoma cDNA libraries to identify tumor-associated antigens that cure melanoma. Nat Biotechnol. 2012;30:337–343. doi:10.1038/nbt.2157.
  • Rommelfanger DM, Wongthida P, Diaz RM, Kaluza KM, Thompson JM, Kottke TJ, Vile RG. Systemic combination virotherapy for melanoma with tumor antigen-expressing vesicular stomatitis virus and adoptive T-cell transfer. Cancer Res. 2012;72:4753–4764. doi:10.1158/0008-5472.CAN-12-0600.
  • Kottke T, Errington F, Pulido J, Galivo F, Thompson J, Wongthida P, Diaz RM, Chong H, Ilett E, Chester J, et al. Broad antigenic coverage induced by vaccination with virus-based cDNA libraries cures established tumors. Nat Med. 2011;17:854–859. doi:10.1038/nm.2390.
  • Bridle BW, Stephenson KB, Boudreau JE, Koshy S, Kazdhan N, Pullenayegum E, Brunelliere J, Bramson JL, Lichty BD, Wan Y.Potentiating cancer immunotherapy using an oncolytic virus. Mol Ther. 2010;18:1430–1439. doi:10.1038/mt.2010.98.
  • Naslund TI, Uyttenhove C, Nordstrom EK, Colau D, Warnier G, Jondal M, Van den Eynde BJ, Liljestrom P. Comparative prime-boost vaccinations using Semliki Forest virus, adenovirus, and ALVAC vectors demonstrate differences in the generation of a protective central memory CTL response against the P815 tumor. J Immunol. 2007;178:6761–6769.
  • Chen A, Zhang Y, Meng G, Jiang D, Zhang H, Zheng M, Xia M, Jiang A, Wu J, Beltinger C, et al. Oncolytic measles virus enhances antitumour responses of adoptive CD8+NKG2D+ cells in hepatocellular carcinoma treatment. Sci Rep. 2017;7:5170. doi:10.1038/s41598-017-05500-z.
  • Dey M, Yu D, Kanojia D, Li G, Sukhanova M, Spencer DA, Pituch KC, Zhang L, Han Y, Ahmed AU, et al. Intranasal oncolytic virotherapy with CXCR4-enhanced stem cells extends survival in mouse model of glioma. Stem Rep. 2016;7:471–482. doi:10.1016/j.stemcr.2016.07.024.
  • Siurala M, Vaha-Koskela M, Havunen R, Tahtinen S, Bramante S, Parviainen S, Mathis JM, Kanerva A, Hemminki A. Syngeneic syrian hamster tumors feature tumor-infiltrating lymphocytes allowing adoptive cell therapy enhanced by oncolytic adenovirus in a replication permissive setting. Oncoimmunology. 2016;5:e1136046. doi:10.1080/2162402X.2015.1136046.
  • Esaki S, Nigim F, Moon E, Luk S, Kiyokawa J, Curry W, Jr., Cahill DP, Chi AS, Iafrate AJ, Martuza RL, et al. Blockade of transforming growth factor-beta signaling enhances oncolytic herpes simplex virus efficacy in patient-derived recurrent glioblastoma models. Int J Cancer. 2017;141:2348–2358. doi:10.1002/ijc.30929.
  • Zhao X, Ouyang W, Chester C, Long S, Wang N, He Z. Cytokine-induced killer cell delivery enhances the antitumor activity of oncolytic reovirus. PLoS One. 2017;12:e0184816. doi:10.1371/journal.pone.0184816.
  • Chen X, Han J, Chu J, Zhang L, Zhang J, Chen C, Chen L, Wang Y, Wang H, Yi L, et al. A combinational therapy of EGFR-CAR NK cells and oncolytic herpes simplex virus 1 for breast cancer brain metastases. Oncotarget. 2016;7:27764–27777. doi:10.18632/oncotarget.8526.
  • Workenhe ST, Pol JG, Lichty BD, Cummings DT, Mossman KL. Combining oncolytic HSV-1 with immunogenic cell death-inducing drug mitoxantrone breaks cancer immune tolerance and improves therapeutic efficacy. Cancer Immunol Res. 2013;1:309–319. doi:10.1158/2326-6066.CIR-13-0059-T.
  • Ribas A, Dummer R, Puzanov I, VanderWalde A, Andtbacka RHI, Michielin O, Olszanski AJ, Malvehy J, Cebon J, Fernandez E, et al. Oncolytic Virotherapy Promotes Intratumoral T Cell Infiltration and Improves Anti-PD-1 Immunotherapy. Cell. 2017;170:1109–19 e10. doi:10.1016/j.cell.2017.08.027.
  • Jonas BA. Combination of an oncolytic virus with PD-L1 blockade keeps cancer in check. Sci Transl Med. 2017;9. doi:10.1126/scitranslmed.aan2781.
  • Hardcastle J, Mills L, Malo CS, Jin F, Kurokawa C, Geekiyanage H, Schroeder M, Sarkaria J, Johnson AJ, Galanis E. Immunovirotherapy with measles virus strains in combination with anti-PD-1 antibody blockade enhances antitumor activity in glioblastoma treatment. Neuro Oncol. 2017;19:493–502. doi:10.1093/neuonc/now179.
  • Liu Z, Ravindranathan R, Kalinski P, Guo ZS, Bartlett DL. Rational combination of oncolytic vaccinia virus and PD-L1 blockade works synergistically to enhance therapeutic efficacy. Nat Commun. 2017;8:14754. doi:10.1038/ncomms14754.
  • Chen CY, Wang PY, Hutzen B, Sprague L, Swain HM, Love JK, et al. Cooperation of oncolytic herpes virotherapy and PD-1 blockade in murine rhabdomyosarcoma models. Sci Rep. 2017;7:2396. doi:10.1038/s41598-017-02503-8.
  • Cockle JV, Rajani K, Zaidi S, Kottke T, Thompson J, Diaz RM, Shim K, Peterson T, Parney IF, Short S, et al. Combination viroimmunotherapy with checkpoint inhibition to treat glioma, based on location-specific tumor profiling. Neuro Oncol. 2016;18:518–527. doi:10.1093/neuonc/nov173.
  • Cervera-Carrascon V, Siurala M, Santos JM, Havunen R, Tahtinen S, Karell P, et al. TNFa and IL-2 armed adenoviruses enable complete responses by anti-PD-1 checkpoint blockade. Oncoimmunology. 2018;7:e1412902. doi:10.1080/2162402X.2017.1412902.
  • Cervera-Carrascon V, Siurala M, Santos JM, Havunen R, Tahtinen S, Karell P, Sorsa S, Kanerva A, Hemminki A. Overcoming resistance to anti-PD immunotherapy in a syngeneic mouse lung cancer model using locoregional virotherapy. Oncoimmunology. 2018;7:e1376156. doi:10.1080/2162402X.2017.1376156.
  • Woller N, Gurlevik E, Fleischmann-Mundt B, Schumacher A, Knocke S, Kloos AM, Saborowski M, Geffers R, Manns MP, Wirth TC, et al. Viral infection of tumors overcomes resistance to PD-1-immunotherapy by broadening neoantigenome-directed T-cell responses. Mol Ther. 2015;23:1630–1640. doi:10.1038/mt.2015.115.
  • Gravett AM, Trautwein N, Stevanović S, Dalgleish AG, Copier J. Gemcitabine alters the proteasome composition and immunopeptidome of tumour cells. Oncoimmunology. 2018;7:e1438107. doi:10.1080/2162402X.2018.1438107.
  • Chen A, Zhang Y, Meng G, Jiang D, Zhang H, Zheng M, Xia M, Jiang A, Wu J, Beltinger C, et al. Gemcitabine enhances antitumor efficacy of recombinant lipoimmunogen-based immunotherapy. Oncoimmunology. 2016;5:e1095433. doi:10.1080/2162402X.2015.1095433.
  • McDonnell AM, Joost Lesterhuis W, Khong A, Nowak AK, Lake RA, Currie AJ, Robinson BW. Restoration of defective cross-presentation in tumors by gemcitabine. Oncoimmunology. 2015;4:e1005501. doi:10.1080/2162402X.2015.1008371.
  • Gujar SA, Clements D, Lee PW. Two is better than one: complementing oncolytic virotherapy with gemcitabine to potentiate antitumor immune responses. Oncoimmunology. 2014;3:e27622. doi:10.4161/onci.27622.
  • Wenthe J, Eriksson E, Milenova I, Moreno R, Alemany R, Loskog A. Meeting abstract | A novel oncolytic adenovirus expressing tumor microenvironment stimulators to evoke and facilitate anti-tumor immune responses. Mol Ther. 2016;24:S206. doi:10.1016/S1525-0016(16)33325-1.
  • Gong J, Gray M, Hutchins J, Freimark B. Anti-tumor responses by ibrutinib and anti-PD-1 blockade is enhanced by phosphatidylserine-targeting antibody therapy. Blood. 2016;128:5379. doi:10.1182/blood-2016-06-724161.
  • Buchan SL, Rogel A, Al-Shamkhani A. The immunobiology of CD27 and OX40 and their potential as targets for cancer immunotherapy. Blood. 2017;doi:10.1182/blood-2017-07-741025.
  • Sapski S, Beha N, Kontermann R, Muller D. Tumor-targeted costimulation with antibody-fusion proteins improves bispecific antibody-mediated immune response in presence of immunosuppressive factors. Oncoimmunology. 2017;6:e1361594. doi:10.1080/2162402X.2017.1361594.
  • Scott EM, Duffy MR, Freedman JD, Fisher KD, Seymour LW. Solid tumor immunotherapy with T cell engager-armed oncolytic viruses. Macromol Biosci. 2017;18. doi:10.1002/mabi.201700187.
  • Gohil SH, Paredes-Moscosso SR, Harrasser M, Vezzalini M, Scarpa A, Morris E, Davidoff AM, Sorio C, Nathwani AC, Della Peruta M. An ROR1 bi-specific T-cell engager provides effective targeting and cytotoxicity against a range of solid tumors. Oncoimmunology. 2017;6:e1326437. doi:10.1080/2162402X.2017.1326437.
  • Kebenko M, Goebeler M-E, Wolf M, Hasenburg A, Seggewiss-Bernhardt R, Ritter B, Rautenberg B, Atanackovic D, Kratzer A, Rottman JB, et al. A multicenter phase 1 study of solitomab (MT110, AMG 110), a bispecific EpCAM/CD3 T-cell engager (BiTE®) antibody construct, in patients with refractory solid tumors. Oncoimmunology. 2018;e1450710. doi:10.1080/2162402X.2018.1450710.
  • Koopmans I, Hendriks D, Samplonius DF, van Ginkel RJ, Heskamp S, Wierstra PJ, Bremer E, Helfrich W. A novel bispecific antibody for EGFR-directed blockade of the PD-1/PD-L1 immune checkpoint. Oncoimmunology. 2018;e1466016. doi:10.1080/2162402X.2018.1466016.
  • Fajardo CA, Guedan S, Rojas LA, Moreno R, Arias-Badia M, de Sostoa J, June CH, Alemany R. Oncolytic adenoviral delivery of an EGFR-targeting T-cell engager improves antitumor efficacy. Cancer Res. 2017;77:2052–2063. doi:10.1158/0008-5472.CAN-16-1708.
  • Wing A, Fajardo CA, Posey AD, Jr., Shaw C, Da T, Young RM, Alemany R, June CH, Guedan S. Improving CART-cell therapy of solid tumors with oncolytic virus-driven production of a bispecific T-cell engager. Cancer Immunol Res. 2018; doi:10.1158/2326-6066.CIR-17-0314.
  • Shayakhmetov DM, Gaggar A, Ni S, Li ZY, Lieber A. Adenovirus binding to blood factors results in liver cell infection and hepatotoxicity. J Virol. 2005;79:7478–7491. doi:10.1128/JVI.79.12.7478-7491.2005.
  • Yu B, Zhou Y, Wu H, Wang Z, Zhan Y, Feng X, Geng R, Wu Y, Kong W, Yu X. Seroprevalence of neutralizing antibodies to human adenovirus type 5 in healthy adults in China. J Med Virol. 2012;84:1408–1414. doi:10.1002/jmv.23325.
  • Hashimoto Y, Watanabe Y, Shirakiya Y, Uno F, Kagawa S, Kawamura H, Nagai K, Tanaka N, Kumon H, Urata Y, et al. Establishment of biological and pharmacokinetic assays of telomerase-specific replication-selective adenovirus. Cancer Sci. 2008;99:385–390. doi:10.1111/j.1349-7006.2007.00665.x.
  • Kaid C, Goulart E, Caires-Junior LC, Araujo BHS, Soares-Schanoski A, Siqueira Bueno HM, Silva KAT, Astray RM, Assoni AF, Ribeiro Junior AF, et al. Zika virus selectively kills aggressive human embryonal CNS tumor cells in vitro and in vivo. Cancer Res. 2018; doi:10.1158/0008-5472.CAN-17-3201.
  • Sistigu A, Manic G, Obrist F, Vitale I. Trial watch - inhibiting PARP enzymes for anticancer therapy. Mol Cell Oncol. 2016;3:e1053594. doi:10.1080/23723556.2015.1053594.
  • Ning J, Wakimoto H, Peters C, Martuza RL, Rabkin SD. Rad51 degradation: role in Oncolytic Virus-Poly(ADP-Ribose) polymerase inhibitor combination therapy in glioblastoma. J Natl Cancer Inst. 2017;109:1–13. doi:10.1093/jnci/djw229.
  • Cruz C, Castroviejo-Bermejo M, Gutierrez-Enriquez S, Llop-Guevara A, Ibrahim YH, Gris-Oliver A, Bonache S, Morancho B, Bruna A, Rueda OM, et al. RAD51 foci as a functional biomarker of homologous recombination repair and PARP inhibitor resistance in germline BRCA mutated breast cancer. Ann Oncol. 2018; doi:10.1093/annonc/mdy099.
  • Saha D, Martuza RL, Rabkin SD. Macrophage polarization contributes to glioblastoma eradication by combination immunovirotherapy and immune checkpoint blockade. Cancer Cell. 2017;32:253–67 e5. doi:10.1016/j.ccell.2017.07.006.
  • Workenhe ST, Ketela T, Moffat J, Cuddington BP, Mossman KL. Genome-wide lentiviral shRNA screen identifies serine/arginine-rich splicing factor 2 as a determinant of oncolytic virus activity in breast cancer cells. Oncogene. 2016;35:2465–2474. doi:10.1038/onc.2015.303.
  • Roselli M, Formica V, Cereda V, Jochems C, Richards J, Grenga I, Orlandi A, Ferroni P, Guadagni F, Schlom J. The association of clinical outcome and peripheral T-cell subsets in metastatic colorectal cancer patients receiving first-line FOLFIRI plus bevacizumab therapy. Oncoimmunology. 2016;5:e1188243. doi:10.1080/2162402X.2016.1188243.
  • Oldfield LM, Grzesik P, Voorhies AA, Alperovich N, MacMath D, Najera CD, Chandra DS, Prasad S, Noskov VN, Montague MG, et al. Genome-wide engineering of an infectious clone of herpes simplex virus type 1 using synthetic genomics assembly methods. Proc Natl Acad Sci U S A. 2017;114:E8885–E94. doi:10.1073/pnas.1700534114.
  • Masemann D, Kother K, Kuhlencord M, Varga G, Roth J, Lichty BD, Rapp UR, Wixler V, Ludwig S. Oncolytic influenza virus infection restores immunocompetence of lung tumor-associated alveolar macrophages. Oncoimmunology. 2018;7:e1423171. doi:10.1080/2162402X.2017.1423171.
  • Brown MC, Holl EK, Boczkowski D, Dobrikova E, Mosaheb M, Chandramohan V, Bigner DD, Gromeier M, Nair SK. Cancer immunotherapy with recombinant poliovirus induces IFN-dominant activation of dendritic cells and tumor antigen-specific CTLs. Sci Transl Med. 2017;9.
  • Rojas JJ, Sampath P, Hou W, Thorne SH. Defining effective combinations of immune checkpoint blockade and oncolytic virotherapy. Clin Cancer Res. 2015;21:5543–5551. doi:10.1158/1078-0432.CCR-14-2009.
  • Kalinski P. Regulation of immune responses by prostaglandin E2. J Immunol. 2012;188:21–28. doi:10.4049/jimmunol.1101029.
  • Obermajer N, Muthuswamy R, Lesnock J, Edwards RP, Kalinski P. Positive feedback between PGE2 and COX2 redirects the differentiation of human dendritic cells toward stable myeloid-derived suppressor cells. Blood. 2011;118:5498–5505. doi:10.1182/blood-2011-07-365825.
  • Fujita M, Kohanbash G, Fellows-Mayle W, Hamilton RL, Komohara Y, Decker SA, Ohlfest JR, Okada H. COX-2 blockade suppresses gliomagenesis by inhibiting myeloid-derived suppressor cells. Cancer Res. 2011;71:2664–2674. doi:10.1158/0008-5472.CAN-10-3055.
  • Donkor MK, Lahue E, Hoke TA, Shafer LR, Coskun U, Solheim JC, Gulen D, Bishay J, Talmadge JE. Mammary tumor heterogeneity in the expansion of myeloid-derived suppressor cells. Int Immunopharmacol. 2009;9:937–948. doi:10.1016/j.intimp.2009.03.021.
  • Rodriguez PC, Hernandez CP, Quiceno D, Dubinett SM, Zabaleta J, Ochoa JB, Gilbert J, Ochoa AC. Arginase I in myeloid suppressor cells is induced by COX-2 in lung carcinoma. J Exp Med. 2005;202:931–939. doi:10.1084/jem.20050715.
  • Hou W, Sampath P, Rojas JJ, Thorne SH. Oncolytic virus-mediated targeting of PGE2 in the tumor alters the immune status and sensitizes established and resistant tumors to immunotherapy. Cancer Cell. 2016;30:108–119. doi:10.1016/j.ccell.2016.05.012.
  • Potts KG, Irwin CR, Favis NA, Pink DB, Vincent KM, Lewis JD, Moore RB, Hitt MM, Evans DH. Deletion of F4L (ribonucleotide reductase) in vaccinia virus produces a selective oncolytic virus and promotes anti-tumor immunity with superior safety in bladder cancer models. EMBO Mol Med. 2017;9:638–654. doi:10.15252/emmm.201607296.
  • Liu D, Li G, Avella DM, Kimchi ET, Kaifi JT, Rubinstein MP, Camp ER, Rockey DC, Schell TD, Staveley-O'Carroll KF. Sunitinib represses regulatory T cells to overcome immunotolerance in a murine model of hepatocellular cancer. Oncoimmunology. 2017;7:e1372079. doi:10.1080/2162402X.2017.1372079.
  • Samson A, Bentham MJ, Scott K, Nuovo G, Bloy A, Appleton E, Adair RA, Dave R, Peckham-Cooper A, Toogood G, et al. Oncolytic reovirus as a combined antiviral and anti-tumour agent for the treatment of liver cancer. Gut. 2016.
  • Rajani K, Parrish C, Kottke T, Thompson J, Zaidi S, Ilett L, et al. Combination therapy with reovirus and anti-PD-1 blockade controls tumor growth through innate and adaptive immune responses. Mol Ther. 2016;24:166–174. doi:10.1038/mt.2015.156.
  • Samson A, Bentham MJ, Scott K, Nuovo G, Bloy A, Appleton E, Adair RA, Dave R, Peckham-Cooper A, Toogood G Cationic liposome-mediated delivery of reovirus enhances the tumor cell-killing efficiencies of reovirus in reovirus-resistant tumor cells. Int J Pharm. 2017;524:238–247. doi:10.1016/j.ijpharm.2017.04.006.
  • Zhang L, Steele MB, Jenks N, Grell J, Suksanpaisan L, Naik S, Federspiel MJ, Lacy MQ, Russell SJ, Peng KW. Safety studies in tumor and non-tumor-bearing mice in support of clinical trials using oncolytic VSV-IFNbeta-NIS. Hum Gene Ther Clin Dev. 2016;27:111–122. doi:10.1089/humc.2016.061.
  • Shen W, Patnaik MM, Ruiz A, Russell SJ, Peng KW. Immunovirotherapy with vesicular stomatitis virus and PD-L1 blockade enhances therapeutic outcome in murine acute myeloid leukemia. Blood. 2016;127:1449–1458. doi:10.1182/blood-2015-06-652503.
  • Alkayyal AA, Tai LH, Kennedy MA, de Souza CT, Zhang J, Lefebvre C, Sahi S, Ananth AA, Mahmoud AB, Makrigiannis AP, et al. NK-cell recruitment is necessary for eradication of peritoneal carcinomatosis with an IL12-expressing maraba virus cellular vaccine. Cancer Immunol Res. 2017;5:211–221. doi:10.1158/2326-6066.CIR-16-0162.
  • Le Boeuf F, Selman M, Son HH, Bergeron A, Chen A, Tsang J, Butterwick D, Arulanandam R, Forbes NE, Tzelepis F, et al. Oncolytic maraba virus MG1 as a treatment for sarcoma. Int J Cancer. 2017;141:1257–1264. doi:10.1002/ijc.30813.
  • Atherton MJ, Stephenson KB, Nikota JK, Hu QN, Nguyen A, Wan Y, Lichty BD. Preclinical development of peptide vaccination combined with oncolytic MG1-E6E7 for HPV-associated cancer. Vaccine. 2018;36:2181–2192. doi:10.1016/j.vaccine.2018.02.070.
  • Aranda F, Vacchelli E, Eggermont A, Galon J, Sautes-Fridman C, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: peptide vaccines in cancer therapy. Oncoimmunology. 2013;2:e26621. doi:10.4161/onci.26621.
  • Atherton MJ, Stephenson KB, Tzelepis F, Bakhshinyan D, Nikota JK, Son HH, et al. Transforming the prostatic tumor microenvironment with oncolytic virotherapy. Oncoimmunology. 2018;7:e1445459. doi:10.1080/2162402X.2018.1445459.
  • Zhang H, Li K, Lin Y, Xing F, Xiao X, Cai J, Zhu W, Liang J, Tan Y, Fu L, et al. Targeting VCP enhances anticancer activity of oncolytic virus M1 in hepatocellular carcinoma. Sci Transl Med. 2017;9. doi:10.1126/scitranslmed.aam7996.
  • Cai J, Lin Y, Zhang H, Liang J, Tan Y, Cavenee WK, Yan G. Selective replication of oncolytic virus M1 results in a bystander killing effect that is potentiated by smac mimetics. Proc Natl Acad Sci U S A. 2017;114:6812–6817. doi:10.1073/pnas.1701002114.
  • Li K, Zhang H, Qiu J, Lin Y, Liang J, Xiao X, Fu L, Wang F, Cai J, Tan Y, et al. Activation of cyclic adenosine monophosphate pathway increases the sensitivity of cancer cells to the oncolytic virus M1. Mol Ther. 2016;24:156–165. doi:10.1038/mt.2015.172.
  • Zhang H, Lin Y, Li K, Liang J, Xiao X, Cai J, Tan Y, Xing F, Mai J, Li Y, et al. Naturally existing oncolytic virus M1 is nonpathogenic for the nonhuman primates after multiple rounds of repeated intravenous injections. Hum Gene Ther. 2016;27:700–711. doi:10.1089/hum.2016.038.
  • Alonso MM, García-Moure M, Gonzalez-Huarriz M, Marigil M, Hernandez-Alcoceba R, Buñales M, Hervás S, Gallego J, Gomez-Manzano C, Fueyo, et al. Meeting library | Abstract CT027: oncolytic virus DNX-2401 with a short course of temozolomide for glioblastoma at first recurrence: clinical data and prognostic biomarkers. Cancer Research. 2017;77:CT027–CT. doi:10.1158/1538-7445.AM2017-CT027.
  • Ilkow CS, Marguerie M, Batenchuk C, Mayer J, Ben Neriah D, Cousineau S, et al. Reciprocal cellular cross-talk within the tumor microenvironment promotes oncolytic virus activity. Nat Med. 2015;21:530–536. doi:10.1038/nm.3848.
  • Kleiderman S, Gutbier S, Ugur Tufekci K, Ortega F, Sa JV, Teixeira AP, Brito C, Glaab E, Berninger B, Alves PM, et al. Conversion of nonproliferating astrocytes into neurogenic neural stem cells: control by FGF2 and interferon-gamma. Stem Cells. 2016;34:2861–2874. doi:10.1002/stem.2483.
  • Lee BJ, Moon HG, Shin TS, Jeon SG, Lee EY, Gho YS, Lee CG, Zhu Z, Elias JA, Kim YK. Protective effects of basic fibroblast growth factor in the development of emphysema induced by interferon-gamma. Exp Mol Med. 2011;43:169–178. doi:10.3858/emm.2011.43.4.018.
  • Scheil S, Ludecke G, Unsicker K. Interleukin-1 beta and interferon gamma interact with fibroblast growth factor-2 in the control of neuroblastoma cell proliferation and differentiation. Int J Dev Neurosci. 1994;12:405–410.
  • Ranki T, Pesonen S, Hemminki A, Partanen K, Kairemo K, Alanko T, Lundin J, Linder N, Turkki R, Ristimaki A, et al. Phase I study with ONCOS-102 for the treatment of solid tumors - an evaluation of clinical response and exploratory analyses of immune markers. J Immunother Cancer. 2016;4:17. doi:10.1186/s40425-016-0121-5.
  • Feister K, Collins S, Kasahara N, Slomovitz B. Meeting Library | Abstract B36: compassionate use of an oncolytic adenovirus in a Stage IV treatment-refractory ovarian cancer patient. Clin Cancer Res. 2017;23:B36–B.
  • McNeish IA, Michael A, Jayson GC, Glasspool RM, Brown R, Fisher K, Alvis S, Barrett J, Blanc CW, McElwaine-Johnn H. A phase I/II study of enadenotucirev, a chimeric Ad11/Ad3 oncolytic group B adenovirus, administered intraperitoneally (IP) in platinum-resistant epithelial ovarian cancer: pharmacokinetic (PK) and tolerability data from phase I. J Clin Oncol. 2016;34:5543.
  • Taipale K, Liikanen I, Koski A, Heiskanen R, Kanerva A, Hemminki O, Oksanen M, Gronberg-Vaha-Koskela S, Hemminki K, Joensuu T, et al. Predictive and prognostic clinical variables in cancer patients treated with adenoviral oncolytic immunotherapy. Mol Ther. 2016;24:1323–1332. doi:10.1038/mt.2016.67.
  • Hemminki A, Oksanen M, Merisalo-Soikkeli M. Oncolytic virotherapy trials–letter. Clin Cancer Res. 2013;19:4541–4542. doi:10.1158/1078-0432.CCR-13-1471.
  • Ramirez M, Ruano D, Moreno L, Lassaletta A, Bautista-Sirvent F, Andión M, Hernández C, González-Murillo A, Melen G, Alemany R, et al. Meeting library | first-in-child trial of celyvir (autologous mesenchymal stem cells carrying the oncolytic virus ICOVIR-5) in patients with relapsed and refractory pediatric solid tumors. J Clin Oncol. 2018;36. (suppl; abstr 10543) http://abstracts.asco.org/214/AbstView_214_220281.html.
  • Andtbacka RH, Dummer R, Gyorki D, Berger A, Conry R, Demidov L, Chan E, Treichel S, Faries M, Ross M. Meeting library | interim analysis of a randomized, open-label phase 2 study of talimogene laherparepvec (T-VEC) neoadjuvant treatment (neotx) plus surgery (surgx) vs surgx for resectable stage IIIB-IVM1a melanoma (MEL). J Clin Oncol. 2018;36. (suppl; abstr 9508) http://abstracts.asco.org/214/AbstView_214_213921.html.
  • Chesney J, Collichio F, Andtbacka RHI, Puzanov I, Glaspy J, Milhem M, Hamid O, Cranmer L, Saenger Y, Ross M, et al. Interim safety and efficacy of a randomized (1:1), open-label phase 2 study of talimogene laherparepvec (T) and ipilimumab (I) vs I alone in unresected, stage IIIB-IV melanoma. Ann Oncol. 2016;27:1108PD–PD. doi:10.1093/annonc/mdw379.04.
  • Chesney J, Puzanov I, Collichio F, Singh P, Milhem MM, Glaspy J, Hamid O, Ross M, Friedlander P, Garbe C, et al. 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. J Clin Oncol. 2017;JCO2017737379.
  • Chesney JA, Puzanov I, Ross MI, Collichio FA, Milhem MM, Chen L, Kim JJ, Garbe C, Hauschild A, Andtbacka RHI. Primary results from a randomized (1:1), open-label phase II study of talimogene laherparepvec (T) and ipilimumab (I) vs I alone in unresected stage IIIB- IV melanoma. J Clin Oncol. 2017;35:9509.
  • Puzanov I, Milhem MM, Minor D, Hamid O, Li A, Chen L, Chastain M, Gorski KS, Anderson A, Chou J, et al. Talimogene laherparepvec in combination with ipilimumab in previously untreated, unresectable stage IIIB-IV melanoma. J Clin Oncol. 2016;34:2619–2626. doi:10.1200/JCO.2016.67.1529.
  • Kaufman HL, Amatruda T, Reid T, Gonzalez R, Glaspy J, Whitman E, Harrington K, Nemunaitis J, Zloza A, Wolf M, et al. Systemic versus local responses in melanoma patients treated with talimogene laherparepvec from a multi-institutional phase II study. J Immunother Cancer. 2016;4:12. doi:10.1186/s40425-016-0116-2.
  • Kaufman HL, Andtbacka RHI, Collichio FA, Wolf M, Zhao Z, Shilkrut M, Puzanov I, Ross M. Durable response rate as an endpoint in cancer immunotherapy: insights from oncolytic virus clinical trials. J Immunother Cancer. 2017;5:72. doi:10.1186/s40425-017-0276-8.
  • Schvartsman G, Perez K, Flynn JE, Myers JN, Tawbi H. Safe and effective administration of T-VEC in a patient with heart transplantation and recurrent locally advanced melanoma. J Immunother Cancer. 2017;5:45. doi:10.1186/s40425-017-0250-5.
  • Harrington K, Kong A, Mach N, Rordorf T, Jaime J, Espeli V, Treichel S, Gumuscu B, Kim J, Chesney JA. Meeting library | Safety and preliminary efficacy of talimogene laherparepvec (T-VEC) in combination (combo) with pembrobrolizumab (Pembro) in patients (pts) with recurrent or metastatic squamous cell carcinoma of the head and neck (R/M HNSCC): A multicenter, phase 1b study (MASTERKEY-232). J Clin Oncol. 2018;36. (suppl; abstr 6036) http://abstracts.asco.org/214/AbstView_214_224119.html.
  • Kelly C, Bowler T, Munhoz R, Chi P, Dickson M, Gounder M, Keohan M, Dholakia R, Condy M, Singer S, et al. Meeting library | A phase II study of talimogene laherparepvec (T-VEC) and pembrolizumab in patients with metastatic sarcoma. J Clin Oncol. 2018;36 (suppl; abstr 11516). http://abstracts.asco.org/214/AbstView_214_222395.html.
  • Cui C, Chi Z, Si L, Sheng X, Li D, Wang X, Lian B, Tang B, Mao L, Yan X, et al. Meeting library | OrienX010 oncolytic viral therapy in phase Ib trial of intralesional injection in unresected stage IIIC to IV (M1a/M1b) acral melanoma patients in China. J Clin Oncol. 2016;34. (suppl; abstr e21001) http://ascopubs.org/doi/abs/10.1200/JCO.2016.34.15_suppl.e21001.
  • Cui C, Cui C, Lian B, Chi Z, Si L, Sheng X, Li D, Li S, Tang B, Mao L, Wang X, et al. Meeting library | OrienX010 oncolytic viral therapy in phase Ic trial of intralesional injection in liver metastases among patients with stage IV melanoma after standard treatment. J Clin Oncol. 2017;35. (suppl; abstr e21013) http://ascopubs.org/doi/abs/10.1200/JCO.2017.35.15_suppl.e21013.
  • Waters AM, Johnston JM, Reddy AT, Fiveash J, Madan-Swain A, Kachurak K, Bag AK, Gillespie GY, Markert JM, Friedman GK. Rationale and design of a phase 1 clinical trial to evaluate HSV G207 alone or with a single radiation dose in children with progressive or recurrent malignant supratentorial brain tumors. Hum Gene Ther Clin Dev. 2017;28:7–16. doi:10.1089/humc.2017.002.
  • Patel DM, Foreman PM, Nabors LB, Riley KO, Gillespie GY, Markert JM. 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. Hum Gene Ther Clin Dev. 2016;27:69–78. doi:10.1089/humc.2016.031.
  • Streby KA, Geller JI, Currier MA, Warren PS, Racadio JM, Towbin AJ, Vaughan MR, Triplet M, Ott-Napier K, Dishman DJ, et al. Intratumoral injection of hsv1716, an oncolytic herpes virus, is safe and shows evidence of immune response and viral replication in young cancer patients. Clin Cancer Res. 2017;23:3566–3574. doi:10.1158/1078-0432.CCR-16-2900.
  • Kelly KJ, Wong J, Gonen M, Allen P, Brennan M, Coit D, Fong Y. Human trial of a genetically modified herpes simplex virus for rapid detection of positive peritoneal cytology in the staging of pancreatic cancer. EBioMedicine. 2016;7:94–99. doi:10.1016/j.ebiom.2016.03.043.
  • Zhang W, Bao L, Yang S, Qian Z, Dong M, Yin L, Zhao Q, Ge K, Deng Z, Zhang J, et al. Tumor-selective replication herpes simplex virus-based technology significantly improves clinical detection and prognostication of viable circulating tumor cells. Oncotarget. 2016;7:39768–39783. doi:10.18632/oncotarget.9465.
  • Hirooka Y, Kasuya H, Ishikawa T, Kawashima H, Ohno E, Villalobos IB, Naoe Y, Ichinose T, Koyama N, Tanaka M, et al. A phase I clinical trial of EUS-guided intratumoral injection of the oncolytic virus, HF10 for unresectable locally advanced pancreatic cancer. BMC Cancer. 2018;18:596. doi:10.1186/s12885-018-4242-8.
  • Andtbacka R, Ross M, Agarwala S, Taylor M, Vetto J, Neves R, Daud A, Khong H, Ungerleider R, Tanaka M. Meeting library | Efficacy and genetic analysis for a phase II multicenter trial of HF10, a replication-competent HSV-1 oncolytic immunotherapy, and ipilimumab combination treatment in patients with stage IIIb-IV unresectable or metastatic melanoma. J Clin Oncol. 2018;36(suppl; abstr 9541). http://abstracts.asco.org/214/AbstView_214_229247.html.
  • Dispenzieri A, Tong C, LaPlant B, Lacy MQ, Laumann K, Dingli D, Zhou Y, Federspiel MJ, Gertz MA, Hayman S, et al. 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. Leukemia. 2017;31:2791–2798. doi:10.1038/leu.2017.120.
  • Geletneky K, Hajda J, Angelova AL, Leuchs B, Capper D, Bartsch AJ, Neumann JO, Schoning T, Husing J, Beelte B, et al. Oncolytic H-1 parvovirus shows safety and signs of immunogenic activity in a first phase I/IIa glioblastoma trial. Mol Ther. 2017;25:2620–2634. doi:10.1016/j.ymthe.2017.08.016.
  • Pandha H, Harrington K, Ralph C, Melcher A, Gupta S, Akerley W, Sandborn RE, Rudin C, Rosenberg J, Kaufman D, et al. Meeting library | Abstract CT115: phase 1b KEYNOTE 200 (STORM study): A study of an intravenously delivered oncolytic virus, coxsackievirus A21 in combination with pembrolizumab in advanced cancer patients.. Cancer Research. 2017;77:CT115–CT. doi:10.1158/1538-7445.AM2017-CT115.
  • Pandha HS, Ralph C, Harrington K, Curti BD, Sanborn RE, Akerley WL, Gupta S, Rudin CM, Rosenberg JE, Kaufman DR, et al. Meeting library | Keynote-200 phase 1b: A novel combination study of intravenously delivered coxsackievirus A21 and pembrolizumab in advanced cancer patients. J Clin Oncol. 2017;35:TPS3108–TPS.
  • Pandha HS, Harrington KJ, Ralph C, Melcher A, Mansfield D, Grose M, Davies B, Karpathy R, Shafren D. Meeting library | Phase I STORM study (KEYNOTE 200): intravenous delivery of a novel oncolytic immunotherapy agent, Coxsackievirus A21 in combination with pembrolizumab in advanced cancer patients. J Clin Oncol. 2016;34:TPS3108–TPS.
  • Pandha HS, Annels NE, Simpson G, Mostafid H, Harrington KJ, Melcher A, Grose M, Davies B, Au GG, Karpathy R, et al. Meeting library | Phase I/II CANON study: oncolytic immunotherapy for the treatment of non-muscle invasive bladder (NMIBC) cancer using intravesical coxsackievirus A21. J Clin Oncol. 2016;34:e16016–e.
  • Andtbacka R, Curti B, Kaufman H, Nemunaitis J, Daniels GA, Hallmeyer S, Whitman E, Lutzky J, Schultz S, Spitler LE, et al. Meeting library | dynamics of tumor response in advanced melanoma patients treated with Coxsackievirus A21. J Clin Oncol. 2016;34.
  • Andtbacka R, Curti B, Kaufman H, Daniels G, Nemunaitis J, Spitler L, Hallmeyer S, Lutzky J, Schultz S, Whitman E, et al. Final data from CALM: A phase II study of Coxsackievirus A21 (CVA21) oncolytic virus immunotherapy in patients with advanced melanoma. J Clin Oncol. 2015;33:9030.
  • Alberts P, Olmane E, Brokane L, Krastina Z, Romanovska M, Kupcs K, Isajevs S, Proboka G, Erdmanis R, Nazarovs J, et al. Long-term treatment with the oncolytic ECHO-7 virus Rigvir of a melanoma stage IV M1c patient, a small cell lung cancer stage IIIA patient, and a histiocytic sarcoma stage IV patient-three case reports. APMIS. 2016;124:896–904. doi:10.1111/apm.12576.
  • Proboka G, Tilgase A, Isajevs S, Rasa A, Alberts P. Melanoma unknown primary brain metastasis treatment with ECHO-7 oncolytic virus rigvir: a case report. Front Oncol. 2018;8:43. doi:10.3389/fonc.2018.00043.
  • Mell LK, Brumund KT, Daniels GA, Advani SJ, Zakeri K, Wright ME, Onyeama SJ, Weisman RA, Sanghvi PR, Martin PJ, et al. Phase I trial of intravenous oncolytic vaccinia virus (GL-ONC1) with cisplatin and radiotherapy in patients with locoregionally advanced head and neck carcinoma. Clin Cancer Res. 2017;23:5696–5702. doi:10.1158/1078-0432.CCR-16-3232.
  • Holloway R, Kendrick J, Stephens A, Kennard J, Burt J, LeBlanc J, Sellers K, Smith J, Coakley S. Meeting library | Phase 1b study of oncolytic vaccinia virus GL-ONC1 in recurrent ovarian cancer (ROC). J Clin Oncol. 2018;36(suppl; abstr 5577). http://abstracts.asco.org/214/AbstView_214_227961.html.
  • Downs-Canner S, Guo ZS, Ravindranathan R, Breitbach CJ, O’Malley ME, Jones HL, et al. Phase 1 study of intravenous oncolytic poxvirus (vvDD) in patients with advanced solid cancers. Mol Ther. 2016;24:1492–1501. doi:10.1038/mt.2016.101.
  • Formenti SC, Golden EB, Goldberg JD, Li X, Taff J, Fenton-Kerimian MB, Chandrasekhar S, Demaria S, Novik Y. Results of a phase I-II study of adjuvant concurrent carboplatin and accelerated radiotherapy for triple negative breast cancer. Oncoimmunology. 2017;6:e1274479. doi:10.1080/2162402X.2016.1274479.
  • Farren MR, Mace TA, Geyer S, Mikhail S, Wu C, Ciombor K, Tahiri S, Ahn D, Noonan AM, Villalona-Calero M, et al. Systemic immune activity predicts overall survival in treatment-naive patients with metastatic pancreatic cancer. Clin Cancer Res. 2016;22:2565–2574. doi:10.1158/1078-0432.CCR-15-1732.
  • Noonan AM, Farren MR, Geyer SM, Huang Y, Tahiri S, Ahn D, Mikhail S, Ciombor KK, Pant S, Aparo S, et al. Randomized phase 2 trial of the oncolytic virus pelareorep (Reolysin) in upfront treatment of metastatic pancreatic adenocarcinoma. Mol Ther. 2016;24:1150–1158. doi:10.1038/mt.2016.66.
  • Mahalingam D, Fountzilas C, Moseley J, Noronha N, Tran H, Chakrabarty R, Selvaggi G, Coffey M, Thompson B, Sarantopoulos J. A phase II study of REOLYSIN(R) (pelareorep) in combination with carboplatin and paclitaxel for patients with advanced malignant melanoma. Cancer Chemother Pharmacol. 2017;79:697–703. doi:10.1007/s00280-017-3260-6.
  • Villalona-Calero MA, Lam E, Otterson GA, Zhao W, Timmons M, Subramaniam D, et al. Oncolytic reovirus in combination with chemotherapy in metastatic or recurrent non-small cell lung cancer patients with KRAS-activated tumors. Cancer. 2016;122:875–883. doi:10.1002/cncr.29856.
  • Cohn DE, Sill MW, Walker JL, O'Malley D, Nagel CI, Rutledge TL, Bradley W, Richardson DL, Moxley KM, Aghajanian C. Randomized phase IIB evaluation of weekly paclitaxel versus weekly paclitaxel with oncolytic reovirus (Reolysin(R)) in recurrent ovarian, tubal, or peritoneal cancer: an NRG oncology/gynecologic oncology group study. Gynecol Oncol. 2017;146:477–483. doi:10.1016/j.ygyno.2017.07.135.
  • Bernstein V, Ellard SL, Dent SF, Tu D, Mates M, Dhesy-Thind SK, et al. A randomized phase II study of weekly paclitaxel with or without pelareorep in patients with metastatic breast cancer: final analysis of Canadian cancer trials group IND.213. Breast Cancer Res Treat. 2017.
  • Lester JF, Casbard AC, Al-Taei S, Harrop R, Katona L, Attanoos RL, et al. A single centre phase II trial to assess the immunological activity of TroVax® plus pemetrexed/cisplatin in patients with malignant pleural mesothelioma – the SKOPOS trial. Oncoimmunology. 2018;00. doi:10.1080/2162402X.2018.1457597.
  • Mikyskova R, Indrova M, Stepanek I, Kanchev I, Bieblova J, Vosahlikova S, et al. Dendritic cells pulsed with tumor cells killed by high hydrostatic pressure inhibit prostate tumor growth in TRAMP mice. Oncoimmunology. 2017;6:e1362528. doi:10.1080/2162402X.2017.1362528.
  • Morris D, Tu D, Tehfe M, Nicholas G, Goffin J, Gregg R, et al. Meeting library | A randomized Phase II study of reolysin in patients with previously treated advanced or metatstatic non small cell lung cancer (NSCLC) receiving standard salvage chemotherapy – Canadian cancer trials group IND 211. J Clin Oncol. 2016;34:e20512.
  • Eigl BJ, Winquist E, Tu D, Hotte SJ, Canil CM, Gregg RW, et al. Meeting Library | A randomized phase II study of pelareorep (REO) plus docetaxel vs. docetaxel alone in patients with metastatic castration resistant prostate cancer (mCRPC): Canadian cancer trials group study IND 209. J Clin Oncol. 2017;35:5021.
  • Mahalingam D, Goel S, Aparo S, Patel Arora S, Noronha N, Tran H, et al. A phase ii study of pelareorep (REOLYSIN) in combination with gemcitabine for patients with advanced pancreatic adenocarcinoma. Cancers (Basel). 2018;10. doi:10.3390/cancers10060160.
  • Malesci A, Bianchi P, Celesti G, Basso G, Marchesi F, Grizzi F, et al. Tumor-associated macrophages and response to 5-fluorouracil adjuvant therapy in stage III colorectal cancer. Oncoimmunology. 2017;6:e1342918. doi:10.1080/2162402X.2017.1342918.
  • Mahalingam D, Fountzilas C, Moseley JL, Noronha N, Cheetham K, Dzugalo A, et al. Meeting Library | A study of REOLYSIN in combination with pembrolizumab and chemotherapy in patients (pts) with relapsed metastatic adenocarcinoma of the pancreas (MAP). J Clin Oncol. 2017;35:e15753–e.
  • Tang P, Jonker D, Kennecke H, Welch S, Cripps M, Asmis T, Chalchal H, Tomiak A, Ko Y, Chen E, et al. Meeting library | IND.210: A randomized phase 2 trial of FOLFOX6/bevacizumab with or without reovirus (Reo) in patients (pts) with metastatic colorectal cancer (mCRC). J Clin Oncol. 2016;34.
  • Adamska A, Domenichini A, Falasca M. Pancreatic ductal adenocarcinoma: current and evolving therapies. Int J Mol Sci. 2017;18.
  • Jonker D, Hotte S, Razak A, Renouf D, Lichty B, Bell J, Powers J, Breitbach CJ, Stojdl D, Stephenson K, et al. Meeting library | phase I study of oncolytic virus (OV) MG1 maraba/MAGE-A3 (MG1MA3), with and without transgenic MAGE-A3 adenovirus vaccine (AdMA3) in incurable advanced/metastatic MAGE-A3-expressing solid tumours: CCTGIND.214. J Clin Oncol. 2017;35:e14637–e14637.
  • Hummel J, Bienzle D, Morrison A, Cieplak M, Stephenson K, DeLay J, Woods JP, Lichty BD, Bridle BW. Maraba virus-vectored cancer vaccines represent a safe and novel therapeutic option for cats. Sci Rep. 2017;7:15738. doi:10.1038/s41598-017-15992-4:3546–3546
  • Freytag SO, Stricker H, Lu M, Elshaikh M, Aref I, Pradhan D, Levin K, Kim JH, Peabody J, Siddiqui F, et al. Prospective randomized phase 2 trial of intensity modulated radiation therapy with or without oncolytic adenovirus-mediated cytotoxic gene therapy in intermediate-risk prostate cancer. Int J Radiat Oncol Biol Phys. 2014;89:268–276. doi:10.1016/j.ijrobp.2014.02.034.
  • Freytag SO, Barton KN, Brown SL, Narra V, Zhang Y, Tyson D, Nall C, Lu M, Ajlouni M, Movsas B, et al. Replication-competent adenovirus-mediated suicide gene therapy with radiation in a preclinical model of pancreatic cancer. Mol Ther. 2007;15:1600–1606. doi:10.1038/sj.mt.6300212.
  • Barton KN, Paielli D, Zhang Y, Koul S, Brown SL, Lu M, Seely J, Kim JH, Freytag SO. Second-generation replication-competent oncolytic adenovirus armed with improved suicide genes and ADP gene demonstrates greater efficacy without increased toxicity. Mol Ther. 2006;13:347–356. doi:10.1016/j.ymthe.2005.10.005.
  • Yu D, Leja-Jarblad J, Loskog A, Hellman P, Giandomenico V, Oberg K, Essand M. Preclinical evaluation of advince, an oncolytic adenovirus adapted for treatment of liver metastases from neuroendocrine cancer. Neuroendocrinology. 2017;105:54–66. doi:10.1159/000448430.
  • Kim JW, Auffinger B, Spencer DA, Miska J, Chang AL, Kane JR, Young JS, Kanojia D, Qiao J, Mann JF, et al. Single dose GLP toxicity and biodistribution study of a conditionally replicative adenovirus vector, CRAd-S-pk7, administered by intracerebral injection to Syrian hamsters. J Transl Med. 2016;14:134. doi:10.1186/s12967-016-0867-z.
  • Thaci B, Ahmed AU, Ulasov IV, Tobias AL, Han Y, Aboody KS, Lesniak MS. Pharmacokinetic study of neural stem cell-based cell carrier for oncolytic virotherapy: targeted delivery of the therapeutic payload in an orthotopic brain tumor model. Cancer Gene Ther. 2012;19:431–442. doi:10.1038/cgt.2012.21.
  • Ulasov IV, Zhu ZB, Tyler MA, Han Y, Rivera AA, Khramtsov A, Curiel DT, Lesniak MS. Survivin-driven and fiber-modified oncolytic adenovirus exhibits potent antitumor activity in established intracranial glioma. Hum Gene Ther. 2007;18:589–602. doi:10.1089/hum.2007.002.
  • Kojima T, Fujiwara T, Shirakawa Y, Ono H, Nakamoto M, Hasegawa H, Hirano N, Wakabayashi M, Nomura S, Togashi Y, et al. Meeting Library | an open label phase I study to evaluate the safety and efficacy of OBP-301 with pembrolizumab in patients with advanced solid tumors. J Clin Oncol. 2018;36(suppl; abstr TPS3117). http://abstracts.asco.org/214/AbstView_214_216803.html.
  • Rodriguez-Garcia A, Gimenez-Alejandre M, Rojas JJ, Moreno R, Bazan-Peregrino M, Cascallo M, Alemany R. Safety and efficacy of VCN-01, an oncolytic adenovirus combining fiber HSG-binding domain replacement with RGD and hyaluronidase expression. Clin Cancer Res. 2015;21:1406–1418. doi:10.1158/1078-0432.CCR-14-2213.
  • Tejada S, Alonso M, Patino A, Fueyo J, Gomez-Manzano C, Diez-Valle R. Phase I trial of DNX-2401 for diffuse intrinsic pontine glioma newly diagnosed in pediatric patients. Neurosurgery. 2017;doi:10.1093/neuros/nyx507.
  • Harb WA, Cerec V, McElwaine-Johnn H, Champion B, Alvis S, Jain N, Ellis C, Fisher K, Beadle JW. Meeting library | A phase I study of pembrolizumab in combination with enadenotucirev (EnAd) (SPICE) in subjects with metastatic or advanced carcinoma. J Clin Oncol. 2016;34:TPS3112–TPS.
  • Hu-Lieskovan S, Moon J, Campos D, Grossmann K, Sosman J, Ryan C, Wu M, Ribas A. Meeting library | reversing resistance to PD-1 blockade by combination of talimogene laherparepvec (T-VEC) with pembrolizumab (pembro) in advanced melanoma patients following progression on a prior PD-1 inhibitor: SWOG S1607 (NCT02965716). J Clin Oncol. 2018;36 (suppl; abstr TPS9603). http://abstracts.asco.org/214/AbstView_214_221999.html.
  • Hotta Y, Kasuya H, Bustos I, Naoe Y, Ichinose T, Tanaka M, Kodera Y. Curative effect of HF10 on liver and peritoneal metastasis mediated by host antitumor immunity. Oncolytic Virother. 2017;6:31–38. doi:10.2147/OV.S127179.
  • Heinrich B, Klein J, Delic M, Goepfert K, Engel V, Geberzahn L, Lusky M, Erbs P, Preville X, Moehler M. Immunogenicity of oncolytic vaccinia viruses JX-GFP and TG6002 in a human melanoma in vitro model: studying immunogenic cell death, dendritic cell maturation and interaction with cytotoxic T lymphocytes. Onco Targets Ther. 2017;10:2389–2401. doi:10.2147/OTT.S126320.
  • Vacchelli E, Eggermont A, Sautes-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch:: oncolytic viruses for cancer therapy. Oncoimmunology. 2014;3:e28694. doi:10.4161/onci.28694.
  • Vacchelli E, Eggermont A, Sautes-Fridman C, Galon J, Zitvogel L, Kroemer G, et al. Trial watch: oncolytic viruses for cancer therapy. Oncoimmunology. 2013;2:e24612. doi:10.4161/onci.24612.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.