key: cord-316589-f1hq0xl5 authors: Nagalo, Bolni Marius; Breton, Camilo Ayala; Zhou, Yumei; Arora, Mansi; Bogenberger, James M.; Barro, Oumar; Steele, Michael B.; Jenks, Nathan J.; Baker, Alexander T.; Duda, Dan G.; Roberts, Lewis Rowland; Russell, Stephen J.; Peng, Kah Whye; Borad, Mitesh J. title: Oncolytic Virus With Attributes of Vesicular Stomatitis Virus and Measles Virus in Hepatobiliary and Pancreatic Cancers date: 2020-08-19 journal: Mol Ther Oncolytics DOI: 10.1016/j.omto.2020.08.007 sha: doc_id: 316589 cord_uid: f1hq0xl5 Abstract Recombinant vesicular stomatitis virus (VSV)-fusion and hemagglutinin (FH) was developed by substituting the promiscuous VSV-G glycoprotein (G) gene in the backbone of VSV with genes encoding for the measles virus envelope proteins F and H. Hybrid VSV-FH exhibited a multifaceted mechanism of cancer-cell killing, and improved neurotolerability over parental VSV in preclinical studies. In this study, we evaluated VSV-FH in vitro and in vivo in models of hepatobiliary and pancreatic cancers. Our results indicate that high intrahepatic doses of VSV-FH did not result in any significant toxicity and were well tolerated by transgenic mice expressing the measles virus receptor CD46. Furthermore, single intratumoral treatments with VSV-FH yielded improved survival and complete tumor regressions in a proportion of mice in the Hep3B hepatocellular carcinoma model, but not in mice xenografted with BxPC3 pancreatic cancer cells. Our preliminary findings indicate that VSV-FH can induce potent oncolysis in hepatocellular and pancreatic cancer cell lines with concordant results in vivo in hepatocellular cancer and discordant in pancreatic cancer, without the VSV-mediated toxic effects previously observed in laboratory animals. Further study of VSV-FH as an oncolytic virotherapy is warranted in hepatocellular carcinoma and pancreatic cancer, to understand broader applicability and mechanisms of sensitivity and resistance. vehicles. [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] However, successful clinical translation of VSV has been hindered by a number of factors, including neurovirulence, liver toxicity, and variable sensitivity of malignant tumors to VSV oncotherapy. 3, 4, [13] [14] [15] [16] [17] [18] Several studies have attempted to remedy these limitations, with varying degrees of success. [19] [20] [21] However, additional investigations and new approaches are needed before VSV-based virotherapy can be used clinically to treat human cancers. able to cross the human blood-brain barrier and propagate in the brain, treatment with VSV-FH will allow for mitigation of neurotoxicity because most normal neuronal cells lack surface expression of CD46, [27] [28] [29] [30] which VSV-FH uses for cellular entry by virtue of its oncolytic MV attribute. 22, 25, 26 VSV-FH has a robust and rapid lytic cycle attributable to the VSV platform and a preferential tropism for CD46-overexpressed cells from the MV platform. CD46 is ubiquitously expressed in all nucleated cells; however, its overexpression has been reported in human tumor cells and tissues, including HPBCs. [31] [32] [33] [34] [35] [36] [37] We have previously shown that hybrid VSV-FH mediates a higher degree of human tumor selectivity, which is strongly associated with the number of CD46 molecules expressed on target cells. 26 We also demonstrated that the multifaceted mechanisms of cancer-cell killing by VSV-FH are enhanced because of its inherent fusogenic activity. 25, 26 Furthermore, we showed that treatment with VSV-FH has the added benefit of being well-tolerated and less neurotoxic in laboratory rodents than parental wild-type (wt)-VSV, which causes severe encephalitis in laboratory animals. 25, 26 HPBCs have extremely high rates of mortality and are increasing in incidence. 38 HBPCs. As previously described, the oncolytic virus VSV-FH-enhanced green fluorescent protein (eGFP) was generated by substitution of the VSV- To further investigate the difference in the infectivity and CPE of To determine whether VSV-FH is causally associated with liver damage and liver toxicity in animal models. Analyses of multiple toxicology parameters, including complete blood counts (CBC), serum chemistry, and liver function tests, were performed at 2, 21, and 45 days after treatment. As shown in Figure although body weight consistently increased from 4 days after injection until the end of the study ( Figure 4B ). As shown in Figure Collectively, these data indicate that intrahepatic injection of VSV-FH did not elicit significant liver toxicity in the treated animals (100% survival) or result in impairment (Kaplan-Meier method, P<.05). Hep3B ( Hep3B cohort resulted in a significant difference in overall survival (P=.01 [ Figure 5C ]), indicating effectiveness of treatment with VSV-FH. In this study, we evaluated whether treatment with oncolytic VSV-FH could trigger a potent cytotoxicity effect in HBPC cell lines in vitro and in vivo using animal models. We conducted a causality assessment in the context of potential VSV-FH-induced liver damage and toxicity in a human CD46 transgenic mouse model, expressing murine interferon alpha receptor (CD46 IFNαR wt/wt). This particular mouse strain has been shown to be susceptible to infections by viruses that use human CD46 as entry receptor, including measles virus, VSV-FH and some adenoviruses. 30,45 46 Additionally, we compared cytotoxic effects of VSV-FH with that of another recombinant, neuro-attenuated VSV that is currently being evaluated in a clinical trial (VSV-hIFNβ-NIS). 14 In this study, we showed that VSV-FH High intrahepatic doses of VSV-FH in healthy transgenic mice expressing human CD46 were found to be manageable, safe, and tolerable. There was a no statistically significant increase in liver enzyme levels (ALT, AST), granulocytes, and total bilirubin at the end of study, but not in alkaline phosphatase. In the absence of severe toxic effects, we deduced that these increases may not result in clinically significant sequelae and could have been confounded by blood hemolysis from the terminal cardiac puncture, which is known to interfere with the measurement of liver VSV-FH-eGFP was used to infect 5×10 5 To determine the production of VSV-FH infectious particles, For cell viability assays, 1. Under a Mayo Clinic Institutional Animal Care and Use Committee-approved protocol, we conducted the following in vivo evaluations. To assess potential toxicity, especially hepatotoxicity, 30 Recent advances in vesicular stomatitis virus-based oncolytic virotherapy: a 5-year update Oncolytic Recombinant Vesicular Stomatitis Virus (VSV) Is Nonpathogenic and Nontransmissible in Pigs, a Natural Host of VSV Safety Studies in Tumor and Non-Tumor-Bearing Mice in Support of Clinical Trials Using Oncolytic VSV-IFNbeta-NIS A Novel Chimeric Oncolytic Virus Vector for Improved Safety and Efficacy as a Platform for the Treatment of Hepatocellular Carcinoma Pre-clinical development of a vaccine against Lassa fever A VSV-based Zika virus vaccine protects mice from lethal challenge Long-term protection from SARS coronavirus infection conferred by a single immunization with an attenuated VSVbased vaccine The Oncolytic Virus VSV-GP Is Effective against Malignant Melanoma A recombinant VSV-vectored MERS-CoV vaccine induces neutralizing antibody and T cell responses in rhesus monkeys after single dose immunization Enhanced Safety and Efficacy of Oncolytic VSV Therapy by Combination with T Cell Receptor Transgenic T Cells as Carriers Comparative Oncology Evaluation of Intravenous Recombinant Oncolytic Vesicular Stomatitis Virus Therapy in Spontaneous Canine Cancer The vesicular stomatitis virus-based Ebola virus vaccine: From concept to clinical trials Re-engineering vesicular stomatitis virus to abrogate neurotoxicity, circumvent humoral immunity, and enhance oncolytic potency Potent systemic therapy of multiple myeloma utilizing oncolytic vesicular stomatitis virus coding for interferon-beta Neurovirulence properties of recombinant vesicular stomatitis virus vectors in non-human primates A human case of encephalitis associated with vesicular stomatitis virus (Indiana serotype) infection. Am Safety studies on intrahepatic or intratumoral injection of oncolytic vesicular stomatitis virus expressing interferon-beta in rodents and nonhuman primates Relative neurotropism of a recombinant rhabdovirus expressing a green fluorescent envelope glycoprotein Vesiculovirus neutralization by natural IgM and complement Characteristics of oncolytic vesicular stomatitis virus displaying tumor-targeting ligands rVSV(M Delta 51)-M3 is an effective and safe oncolytic virus for cancer therapy Retargeting vesicular stomatitis virus using measles virus envelope glycoproteins. Hum Lassa-vesicular stomatitis chimeric virus safely destroys brain tumors CD30-targeted oncolytic viruses as novel therapeutic approach against classical Hodgkin lymphoma Oncolytic potency of HER-2 retargeted VSV-FH hybrid viruses: the role of receptor ligand affinity Faster replication and higher expression levels of viral glycoproteins give the vesicular stomatitis virus/measles virus hybrid VSV-FH a growth advantage over measles virus Nectin 4 is the epithelial cell receptor for measles virus Tumor cell marker PVRL4 (nectin 4) is an epithelial cell receptor for measles virus Measles virus spread between neurons requires cell contact but not CD46 expression, syncytium formation, or extracellular virus production A matrix-less measles virus is infectious and elicits extensive cell fusion: consequences for propagation in the brain High CD46 receptor density determines preferential killing of tumor cells by oncolytic measles virus Systematic immunohistochemical analysis of the expression of CD46, CD55, and CD59 in colon cancer Antibody-drug conjugate targeting CD46 eliminates multiple myeloma cells RNA interference characterization of proteins discovered by proteomic analysis of pancreatic cancer reveals function in cell growth and survival p53 regulates CD46 expression and measles virus infection in myeloma cells Bioinformatic analysis of the membrane cofactor protein CD46 and microRNA expression in hepatocellular carcinoma Cell-surface density of complement restriction factors (CD46, CD55, and CD59): oral squamous cell carcinoma versus other solid tumors. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 103 Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States Pancreatic cancer: A review of clinical diagnosis, epidemiology, treatment and outcomes Systemic Treatment Options in Hepatocellular Carcinoma Amalgamating oncolytic viruses to enhance their safety, consolidate their killing mechanisms, and accelerate their spread Oncolytic measles viruses displaying a single-chain antibody against CD38, a myeloma cell marker Preclinical pharmacology and toxicology of intravenous MV-NIS, an oncolytic measles virus administered with or without cyclophosphamide Roles of macrophages in measles virus infection of genetically modified mice Vaccines within vaccines: the use of adenovirus types 4 and 7 as influenza vaccine vectors Vesicular stomatitis virus expressing interferon-beta is oncolytic and promotes antitumor immune responses in a syngeneic murine model of non-small cell lung cancer Vascularization, Oxygenation, and the Effect of Sunitinib Treatment in Pancreatic Ductal Adenocarcinoma Xenografts Role of the tumor microenvironment in pancreatic cancer Antitumor effect of angiotensin II type 1 receptor blocker losartan for orthotopic rat pancreatic adenocarcinoma Hyaluronidase expression by an oncolytic adenovirus enhances its intratumoral spread and suppresses tumor growth