Pharmacological Improvement of Oncolytic Virotherapy

Total Page:16

File Type:pdf, Size:1020Kb

Pharmacological Improvement of Oncolytic Virotherapy PHARMACOLOGICAL IMPROVEMENT OF ONCOLYTIC VIROTHERAPY Mohammed Selman Thesis submitted to the Faculty of Graduate and Postdoctoral Studies in partial fulfillment of the requirements for the Doctorate of Philosophy in Biochemistry Department of Biochemistry, Microbiology & Immunology Faculty of Medicine University of Ottawa © Mohammed Selman, Ottawa, Canada, 2018 Abstract Oncolytic viruses (OV) are an emerging class of anticancer bio-therapeutics that induce antitumor immunity through selective replication in cancer cells. However, the efficacy of OVs as single agents remains limited. We postulate that resistance to oncolytic virotherapy results in part from the failure of tumor cells to be sufficiently infected. In this study, we provide evidence that in the context of sarcoma, a highly heterogeneous malignancy, the infection of tumors by different oncolytic viruses varies greatly. Similarly, for a given oncolytic virus, productive infection of tumors across patient samples varies by many orders of magnitude. To overcome this issue, we hypothesize that the infection of resistant tumors can be achieved through the use of selected small molecules. Here, we have identified two novel drug classes with the ability to improve the efficacy of OV therapy: fumaric and maleic acid esters (FMAEs) and vanadium compounds. FMAEs are enhancing infection of cancer cells by several oncolytic viruses in cancer cell lines and human tumor biopsies. The ability of FMAEs to enhance viral spread is due to their ability to inhibit type I IFN production and response, which is associated with their ability to block nuclear translocation of transcription factor NF-κB. Vanadium-based phosphatase inhibitors enhance OV infection of RNA viruses in vitro and ex vivo, in resistant cancer cell lines. Mechanistically, this involves subverting the antiviral type I IFN response towards a death-inducing and proinflammatory type II IFN response, leading to improved OV spread, increased bystander killing of cancer cells, and enhanced anti-tumor immune-stimulation. Both FMAEs and vanadium compounds improve therapeutic outcomes of OV treatment in syngeneic tumor models, leading to durable responses, even in models otherwise refractory to OV and drug alone. Overall, we showcased novel avenues for the development of improved immunotherapy strategies. ii Acknowledgements This thesis would not have been possible without the aid and support of several people. First, I would like to thank my supervisor Dr. Jean-Simon Diallo for his guidance, motivation and continuous support during the duration of my PhD. Furthermore, I would like to thank to my thesis committee members, Dr. Mary-Ellen Harper, Dr. Tommy Alain and Dr. Douglas Gray for their helpful advice throughout the course of the program. I would also like to thank the many collaborators for their work and valuable input: Dr. Debbie C. Crans, Dr. Brian A. Keller, Dr. Larissa Pikor, Dr. Fabrice Le Boeuf, Dr. John C. Bell and Dr. Carolina Ilkow. A special thanks to Dr. Fanny Tzelepis for teaching me new skills and all her effort and help with the many immunological experiments, and Andrew Chen for all his help managing the numerous in vivo experiments. Sincere thanks for all the hard work from honors students that I had the privilege to supervise and work with: Chris Rousso, Paula Ou and Anabel Bergeron. Finally, thanks to family, friends and all current and former lab members of the Diallo lab for their ongoing support. iii Table of Contents Abstract ii Acknowledgements iii List of Abbreviations vi List of Figures ix Chapter 1 - General Introduction 1 Cancer 1 Oncolytic virotherapy 1 Mechanism of action of oncolytic viruses 2 Oncolytic virus platforms 4 Oncolytic herpesvirus 5 Oncolytic poxviruses 6 Oncolytic reovirus 7 Oncolytic rhabdoviruses 9 Type I and type II interferon pathways 11 The NF-κB pathway 13 Defects in type I IFN signaling in cancer 14 Resistance to OV 16 Breaking resistance to OV: Viral sensitizers 17 Fumaric acid esters 21 DMF as cancer treatment 23 Vanadium 25 Vanadium compounds are promising anticancer agents 27 Sarcoma: a rare cancer in need of new therapies 28 Rationale and aim I 29 Rationale and hypothesis II 30 Chapter 2 - Oncolytic Maraba virus MG1 as a treatment for Sarcoma 32 Abstract 33 Introduction 34 Materials and Methods 36 Results 40 iv Discussion 52 Chapter 3 - Dimethyl Fumarate Potentiates Oncolytic Virotherapy through NF-kB inhibition 54 Abstract 55 Introduction 56 Results 58 Discussion 79 Chapter 4 - Multi-Modal Potentiation of Oncolytic Virotherapy by Vanadium Compounds 91 Abstract 92 Introduction 93 Results 95 Discussion 119 Materials and Methods 123 Chapter 5 - General discussion 132 Crosstalk between type I and II interferon pathways 132 The antiapoptotic and proapoptotic effects of vanadate 135 Expanding OV tropism 137 Improving Oncolytic rhabdovirus therapy in the treatment of Sarcoma 137 Cancer therapy targeting the NF-κB pathway 138 Treatment delivery 139 Beyond oncolytic immunotherapy 140 Concluding thoughts 141 Appendices I – Chapter 2 Supplemental information 143 Appendices II – Chapter 3 Supplemental information 147 Appendices III – Chapter 3 Supplemental information 157 References 178 v List of Abbreviations 7-AAD 7-Aminoactinomycin D ANOVA Analysis of variance Bax BCL2 Associated X BMOV Bismaltolato oxovanadium BSO Buthionine sulfoximine CDK Cyclin-dependent kinase CTCL Cutaneous T-cell lymphoma CTLA-4 Cytotoxic T-lymphocyte-associated protein-4 DEF Diethyl fumarate DEM Diethyl maleate DMEM Dulbecco’s Modified Eagle’s Medium DMF Dimethyl fumarate DMM Dimethyl maleate DNA Deoxyribonucleic acid ECM Extracellular matrix eIF2a eukaryotic initiation factor 2 alpha FA Fumaric acid FAE Fumaric acid esters FDA U S Food and Drug Administration FMAE Fumaric and maleic acid esters G Glycoprotein GAF IFN-γ-regulated DNA-binding factor GAS IFN-γ activation sequence GCL Glutamate Cysteine Ligase GM-CSF Granulocyte-macrophage colony-stimulating factor GO Gene Ontology GSH Glutathione HDAC Histone deacetylase inhibitor HEPS 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid HR Higher responders HSV-1 Herpes simplex virus 1 IAP Inhibitors of apoptosis proteins IFIT1 Interferon-induced protein with tetratricopeptide repeats 1 IFITM1 Interferon-induced transmembrane protein 1 IFN Interferon IFNGR1 IFN-γ receptor 1 IKK IκB kinase IRF IFN-regulatory factor vi IRSE Interferon-stimulated response element ISGF3 Interferon-stimulated gene factor 3 IT Intratumoral IV Intravenous IVIS in vivo imaging system JAK Janus kinase JC John Cunningham JX-594 Pexastimogene devacirepvec KEAP1 Kelch-like ECH-associated protein 1 LPS Lipopolysaccharides LR Lower responders M Matrix MG1 Marabex MMF Monomethyl fumarate MOI Multiplicity of infection MS Multiple sclerosis mTOR mammalian target of rapamycin MV Measles MxA Myxoma resistance protein A N Nucleoprotein NF-κB Nuclear factor kappa-light-chain-enhancer of activated B cells NK Natural killer NRF2 Nuclear factor (erythroid-derived 2)-like 2 OHSN-REB Ottawa Health Science Network Research Ethics Board OV Oncolytic Virus P Phosphoprotein PBS Phosphate-buffered saline PD-L1 Programmed death-ligand 1 PD1 Programmed cell death protein 1 PDA Pancreatic ductal adenocarcinomas PFU Plaque forming units PI3K Phosphoinositide 3-kinase PKR Protein kinase R PML Progressive multifocal leukoencephalopathy PRR Pattern recognition receptors PTP Protein tyrosine phosphatases PTP1B Protein-tyrosine phosphatase 1B qPCR Quantitative polymerase chain reaction RECIST Response Evaluation Criteria In Solid Tumors RIG-1 Retinoic Acid Inducible Gene 1 vii RIPK3 Receptor Interacting Protein Kinase 3 RNA Ribonucleic acid ROS Reactive oxygen species RPMI Roswell Park Memorial Institute SC Sub-cutaneous Smac Second mitochondrial activator of caspase SMC Smac mimetic compounds STAT1 Signal transducer and activator of transcription 1 STAT2 Signal transducer and activator of transcription 2 T-Vec Talimogene laherparepvec TBK1 TANK-binding kinase 1 TFNα Tumor necrosis factor alpha TGF-β Transforming growth factor-β TK Thymidine kinase TLR Toll-like receptors VOx Vvanadium(V) oxytriethoxyde VS Vanadium(IV) oxide sulphate VSV Vesicular stomatitis virus VVdd double-deleted vaccinia virus wt wild-type viii List of Figures Figure 2.1. Cytopathic effect of oncolytic viruses in sarcoma cells lines in vitro. Figure 2.2. Productive infection of human sarcoma explants by MG1 and VV. Figure 2.3. Maraba virus MG1 infects and replicates in murine sarcoma S180 tumors, leading to persistent anti-tumor effects Figure 3.1. Dimethyl fumarate promotes viral infection Figure 3.2. DMF enhances infection ex vivo and in human clinical samples. Figure 3.3. Fumaric and maleic acid esters promote infection by VSVΔ51. Figure 3.4. Dimethyl fumarate enhances VSV∆51 therapeutic efficacy in syngeneic and xenograft tumor models. Figure 3.5. FMAEs inhibit antiviral cytokine production and response to type I interferon. Figure 3.6. DMF inhibits NF-κB translocation upon infection. Figure 4.1. Vanadate enhances VSV∆51 infection in cancer cells but not normal cells. Figure 4.2. Viral enhancement is dependent on Vanadium. Figure 4.3. Vanadate facilitates virus-induced type I interferon and ROS mediated cell death Figure 4.4. Vanadate increases VSV∆51 efficacy in resistant syngeneic tumor models. Figure 4.5. Vanadate /VSV∆51 co-treatment triggers T-cell infiltration and anti-tumor immunity. Figure 4.6. Vanadate re-wires antiviral type I IFN signaling towards pro-inflammatory type II IFN response ix Chapter 1 - General Introduction
Recommended publications
  • Promoting the Accumulation of Tumor-Specific T Cells in Tumor
    PROTOCOL Promoting the accumulation of tumor-specific T cells in tumor tissues by dendritic cell vaccines and chemokine-modulating agents Nataša Obermajer1 , Julie Urban2, Eva Wieckowski1,2,6, Ravikumar Muthuswamy1, Roshni Ravindranathan1, David L Bartlett1 & Pawel Kalinski1–5 1Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. 2Immunotransplantation Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. 3University of Pittsburgh Cancer Institute, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. 4Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. 5Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. 6Deceased. Correspondence should be addressed to P.K. ([email protected]). Published online 18 January 2018; doi:10.1038/nprot.2017.130 This protocol describes how to induce large numbers of tumor-specific cytotoxic T cells (CTLs) in the spleens and lymph nodes of mice receiving dendritic cell (DC) vaccines and how to modulate tumor microenvironments (TMEs) to ensure effective homing of the vaccination-induced CTLs to tumor tissues. We also describe how to evaluate the numbers of tumor-specific CTLs within tumors. The protocol contains detailed information describing how to generate a specialized DC vaccine with augmented ability to induce tumor-specific CTLs. We also describe methods to modulate the production of chemokines in the TME and show how to quantify tumor-specific CTLs in the lymphoid organs and tumor tissues of mice receiving different treatments. The combined experimental procedure, including tumor implantation, DC vaccine generation, chemokine-modulating (CKM) approaches, and the analyses of tumor-specific systemic and intratumoral immunity is performed over 30–40 d. The presented ELISpot-based ex vivo CTL assay takes 6 h to set up and 5 h to develop.
    [Show full text]
  • (T-VEC): an Intralesional Cancer Immunotherapy for Advanced Melanoma
    cancers Review Talimogene Laherparepvec (T-VEC): An Intralesional Cancer Immunotherapy for Advanced Melanoma Pier Francesco Ferrucci 1,* , Laura Pala 2, Fabio Conforti 2 and Emilia Cocorocchio 3 1 Tumor Biotherapy Unit, Department of Experimental Oncology, European Institute of Oncology, IRCCS, 20141 Milan, Italy 2 Division of Melanoma, Sarcoma and Rare Tumors, European Institute of Oncology, IRCCS, 20141 Milan, Italy; [email protected] (L.P.); [email protected] (F.C.) 3 Hemato-Oncology Division, European Institute of Oncology, IRCCS, 20141 Milan, Italy; [email protected] * Correspondence: [email protected]; Tel.: +39-0294371094 Simple Summary: Talimogene laherparepvec (T-VEC; IMLYGIC®, Amgen Inc.) is the first oncolytic vi- ral immunotherapy to be approved for the local treatment of unresectable metastatic stage IIIB/C–IVM1a melanoma. Its direct intratumoral injection aim to trigger local and systemic immunologic responses leading to tumor cell lysis, followed by release of tumor-derived antigens and subsequent activation of tumor-specific effector T-cells. Its approval has fueled the interest to study its possible sinergy with other immunotherapeutics in preclinical models as well as in clinical contextes. In fact, it has been shown that intratumoral administration of this immunostimulatory agent successfully synergizes with immune checkpoint inhibitors. The objectives of this review are to resume the current state of the art of T-VEC treatment when used in monotherapy or in combination with immune checkpoint inhibitors, describing the strong rationale of its development, the Citation: Ferrucci, P.F.; Pala, L.; adverse events of interest and the clinical outcome in selected patient’s populations. Conforti, F.; Cocorocchio, E.
    [Show full text]
  • Intratumoral Injection of HSV1716, an Oncolytic Herpes Virus, Is Safe and Shows Evidence of Immune Response and Viral Replication in Young Cancer Patients Keri A
    Published OnlineFirst May 11, 2017; DOI: 10.1158/1078-0432.CCR-16-2900 Cancer Therapy: Clinical Clinical Cancer Research Intratumoral Injection of HSV1716, an Oncolytic Herpes Virus, Is Safe and Shows Evidence of Immune Response and Viral Replication in Young Cancer Patients Keri A. Streby1,2, James I. Geller3, Mark A. Currier2, Patrick S. Warren4, John M. Racadio5, Alexander J. Towbin5, Michele R. Vaughan1, Melinda Triplet1, Kristy Ott-Napier1, Devon J. Dishman1, Lori R. Backus3, Beth Stockman3, Marianne Brunner6, Kathleen Simpson7, Robert Spavin7, Joe Conner7, and Timothy P. Cripe1,2 Abstract Purpose: HSV1716 is an oncolytic herpes simplex virus-1 included low-grade fever, chills, and mild cytopenias. Six of (HSV-1) studied in adults via injection into the brain and super- eight HSV-1 seronegative patients at baseline showed serocon- ficial tumors. To determine the safety of administering HSV1716 version on day 28. Six of nine patients had detectable HSV-1 to pediatric patients with cancer, we conducted a phase I trial of genomes by PCR in peripheral blood appearing on day þ4 image-guided injection in young patients with relapsed or refrac- consistent with de novo virus replication. Two patients had tory extracranial cancers. transient focal increases in metabolic activity on 18fluorine- Experimental Design: We delivered a single dose of 105 to 107 deoxyglucose PET, consistent with inflammatory reactions. In infectious units of HSV1716 via computed tomography–guided onecase,thesamegeographicregionthatflared later appeared intratumoral injection and measured tumor responses by imag- necrotic on imaging. No patient had an objective response to ing. Patients were eligible for up to three more doses if they HSV1716.
    [Show full text]
  • Oncolytic Virotherapy with an HSV Amplicon Vector Expressing
    Cancer Gene Therapy (2007) 14, 918–926 r 2007 Nature Publishing Group All rights reserved 0929-1903/07 $30.00 www.nature.com/cgt ORIGINAL ARTICLE Oncolytic virotherapy with an HSV amplicon vector expressing granulocyte–macrophage colony-stimulating factor using the replication-competent HSV type 1 mutant HF10 as a helper virus S-i Kohno1,2, C Luo1,2, A Nawa3, Y Fujimoto4, D Watanabe5, F Goshima1, T Tsurumi6 and Y Nishiyama1 1Department of Virology, Graduate School of Medicine, Nagoya University, Nagoya, Japan; 2Research Division, M’s Science Corporation, Kobe, Japan; 3Department of Obstetrics and Genecology, Graduate School of Medicine, Nagoya University, Nagoya, Japan; 4Department of Otorhinolaryngology, Graduate School of Medicine, Nagoya University, Nagoya, Japan; 5Department of Dermatology, Aichi Medical University, Aichi, Japan and 6Division of Virology, Aichi Cancer Center Research Institute, Nagoya, Japan Direct viral infection of solid tumors can cause tumor cell death, but these techniques offer the opportunity to express exogenous factors to enhance the antitumor response. We investigated the antitumor effects of a herpes simplex virus (HSV) amplicon expressing mouse granulocyte–macrophage colony-stimulating factor (mGM-CSF) using the replication-competent HSV type 1 mutant HF10 as a helper virus. HF10-packaged mGM-CSF-expressing amplicon (mGM-CSF amplicon) was used to infect subcutaneously inoculated murine colorectal tumor cells (CT26 cells) and the antitumor effects were compared to tumors treated with only HF10. The mGM-CSF amplicon efficiently replicated in CT26 cells with similar oncolytic activity to HF10 in vitro. However, when mice subcutaneously inoculated with CT26 cells were intratumorally injected with HF10 or mGM-CSF amplicon, greater tumor regression was seen in mGM-CSF amplicon-treated animals.
    [Show full text]
  • Oncolytic Herpes Simplex Virus-Based Therapies for Cancer
    cells Review Oncolytic Herpes Simplex Virus-Based Therapies for Cancer Norah Aldrak 1 , Sarah Alsaab 1,2, Aliyah Algethami 1, Deepak Bhere 3,4,5 , Hiroaki Wakimoto 3,4,5,6 , Khalid Shah 3,4,5,7, Mohammad N. Alomary 1,2,* and Nada Zaidan 1,2,* 1 Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology, P.O. Box 6086, Riyadh 11451, Saudi Arabia; [email protected] (N.A.); [email protected] (S.A.); [email protected] (A.A.) 2 National Center for Biotechnology, Life Science and Environmental Research Institute, King Abdulaziz City for Science and Technology, P.O. Box 6086, Riyadh 11451, Saudi Arabia 3 Center for Stem Cell Therapeutics and Imaging (CSTI), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; [email protected] (D.B.); [email protected] (H.W.); [email protected] (K.S.) 4 Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA 5 BWH Center of Excellence for Biomedicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA 6 Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA 7 Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA * Correspondence: [email protected] (M.N.A.); [email protected] (N.Z.) Abstract: With the increased worldwide burden of cancer, including aggressive and resistant cancers, oncolytic virotherapy has emerged as a viable therapeutic option. Oncolytic herpes simplex virus (oHSV) can be genetically engineered to target cancer cells while sparing normal cells.
    [Show full text]
  • IL-6 Signaling Blockade During CD40-Mediated Immune Activation Favors Antitumor Factors by Reducing TGF-Β, Collagen Type I
    IL-6 Signaling Blockade during CD40-Mediated Immune Activation Favors Antitumor Factors by Reducing TGF-β, Collagen Type I, and PD-L1/PD-1 This information is current as of September 26, 2021. Emma Eriksson, Ioanna Milenova, Jessica Wenthe, Rafael Moreno, Ramon Alemany and Angelica Loskog J Immunol 2019; 202:787-798; Prepublished online 7 January 2019; doi: 10.4049/jimmunol.1800717 Downloaded from http://www.jimmunol.org/content/202/3/787 References This article cites 50 articles, 12 of which you can access for free at: http://www.jimmunol.org/ http://www.jimmunol.org/content/202/3/787.full#ref-list-1 Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists by guest on September 26, 2021 • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2019 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology IL-6 Signaling Blockade during CD40-Mediated Immune Activation Favors Antitumor Factors by Reducing TGF-b, Collagen Type I, and PD-L1/PD-1 Emma Eriksson,* Ioanna Milenova,* Jessica Wenthe,* Rafael Moreno,† Ramon Alemany,† and Angelica Loskog*,‡ IL-6 plays a role in cancer pathogenesis via its connection to proteins involved in the formation of desmoplastic stroma and to immunosuppression by driving differentiation of myeloid suppressor cells together with TGF-b.
    [Show full text]
  • Development of a New Hyaluronic Acid Based Redox-Responsive Nanohydrogel for the Encapsulation of Oncolytic Viruses for Cancer Immunotherapy
    nanomaterials Article Development of a New Hyaluronic Acid Based Redox-Responsive Nanohydrogel for the Encapsulation of Oncolytic Viruses for Cancer Immunotherapy Siyuan Deng 1, Alessandra Iscaro 2, Giorgia Zambito 3,4, Yimin Mijiti 5 , Marco Minicucci 5 , Magnus Essand 6, Clemens Lowik 3,4 , Munitta Muthana 2, Roberta Censi 1, Laura Mezzanotte 3,4 and Piera Di Martino 1,* 1 School of Pharmacy, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy; [email protected] (S.D.); [email protected] (R.C.) 2 Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; [email protected] (A.I.); m.muthana@sheffield.ac.uk (M.M.) 3 Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; [email protected] (G.Z.); [email protected] (C.L.); [email protected] (L.M.) 4 Department of Molecular Genetics, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands 5 Physics Division, School of Science and Technology, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy; [email protected] (Y.M.); [email protected] (M.M.) 6 Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden; [email protected] * Correspondence: [email protected]; Tel.: +39-0737-40-2215 Abstract: Oncolytic viruses (OVs) are emerging as promising and potential anti-cancer thera- peutic agents, not only able to kill cancer cells directly by selective intracellular viral replication, Citation: Deng, S.; Iscaro, A.; but also to promote an immune response against tumor. Unfortunately, the bioavailability Zambito, G.; Mijiti, Y.; Minicucci, M.; under systemic administration of OVs is limited because of undesired inactivation caused by Essand, M.; Lowik, C.; Muthana, M.; host immune system and neutralizing antibodies in the bloodstream.
    [Show full text]
  • Treating Cancerous Cells with Viruses: Insights from a Minimal Model for Oncolytic Virotherapy
    LETTERS IN BIOMATHEMATICS 2018, VOL. 5, NO. S1, S117–S136 https://doi.org/10.1080/23737867.2018.1440977 RESEARCH ARTICLE OPEN ACCESS Treating cancerous cells with viruses: insights from a minimal model for oncolytic virotherapy Adrianne L. Jennera, Adelle C. F. Costerb, Peter S. Kima and Federico Frascolic aSchool of Mathematics and Statistics, University of Sydney, Sydney, Australia; bSchool of Mathematics and Statistics, University of New South Wales, Sydney, Australia; cFaculty of Science, Engineering and Technology, Department of Mathematics, Swinburne University of Technology, Melbourne, Australia ABSTRACT ARTICLE HISTORY In recent years, interest in the capability of virus particles as a Received 13 December 2017 treatment for cancer has increased. In this work, we present a Accepted 30 January 2018 mathematical model embodying the interaction between tumour KEYWORDS cells and virus particles engineered to infect and destroy cancerous Oncolytic virotherapy; tissue. To quantify the effectiveness of oncolytic virotherapy, we bifurcation analysis; conduct a local stability analysis and bifurcation analysis of our model. oscillations In the absence of tumour growth or viral decay, the model predicts that oncolytic virotherapy will successfully eliminate the tumour cell population for a large proportion of initial conditions. In comparison, for growing tumours and decaying viral particles there are no stable equilibria in the model; however, oscillations emerge for certain regions in our parameter space. We investigate how the period and amplitude of oscillations depend on tumour growth and viral decay. We find that higher tumour replication rates result in longer periods between oscillations and lower amplitudes for uninfected tumour cells. From our analysis, we conclude that oncolytic viruses can reduce growing tumours into a stable oscillatory state, but are insufficient to completely eradicate them.
    [Show full text]
  • Combination Therapy of Novel Oncolytic Adenovirus with Anti-PD1 Resulted in Enhanced Anti-Cancer Effect in Syngeneic Immunocompetent Melanoma Mouse Model
    pharmaceutics Article Combination Therapy of Novel Oncolytic Adenovirus with Anti-PD1 Resulted in Enhanced Anti-Cancer Effect in Syngeneic Immunocompetent Melanoma Mouse Model Mariangela Garofalo 1,* , Laura Bertinato 1, Monika Staniszewska 2, Magdalena Wieczorek 3 , Stefano Salmaso 1 , Silke Schrom 4, Beate Rinner 4, Katarzyna Wanda Pancer 3 and Lukasz Kuryk 3,5,* 1 Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; [email protected] (L.B.); [email protected] (S.S.) 2 Centre for Advanced Materials and Technologies, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland; [email protected] 3 Department of Virology, National Institute of Public Health—National Institute of Hygiene, Chocimska 24, 00-791 Warsaw, Poland; [email protected] (M.W.); [email protected] (K.W.P.) 4 Division of Biomedical Research, Medical University of Graz, Roseggerweg 48, 8036 Graz, Austria; [email protected] (S.S.); [email protected] (B.R.) 5 Clinical Science, Targovax Oy, Lars Sonckin kaari 14, 02600 Espoo, Finland * Correspondence: [email protected] (M.G.); [email protected] (L.K.) Abstract: Malignant melanoma, an aggressive form of skin cancer, has a low five-year survival rate Citation: Garofalo, M.; Bertinato, L.; in patients with advanced disease. Immunotherapy represents a promising approach to improve sur- Staniszewska, M.; Wieczorek, M.; vival rates among patients at advanced stage. Herein, the aim of the study was to design and produce, Salmaso, S.; Schrom, S.; Rinner, B.; by using engineering tools, a novel oncolytic adenovirus AdV-D24- inducible co-stimulator ligand Pancer, K.W.; Kuryk, L.
    [Show full text]
  • Review the Oncolytic Virotherapy Treatment Platform for Cancer
    Cancer Gene Therapy (2002) 9, 1062 – 1067 D 2002 Nature Publishing Group All rights reserved 0929-1903/02 $25.00 www.nature.com/cgt Review The oncolytic virotherapy treatment platform for cancer: Unique biological and biosafety points to consider Richard Vile,1 Dale Ando,2 and David Kirn3,4 1Molecular Medicine Program, Mayo Clinic, Rochester, Minnesota, USA; 2Cell Genesys Corp., Foster City, California, USA; 3Department of Pharmacology, Oxford University Medical School, Oxford, UK; and 4Kirn Oncology Consulting, San Francisco, California, USA. The field of replication-selective oncolytic viruses (virotherapy) has exploded over the last 10 years. As with many novel therapeutic approaches, initial overexuberance has been tempered by clinical trial results with first-generation agents. Although a number of significant hurdles to this approach have now been identified, novel solutions have been proposed and improvements are being made at a furious rate. This article seeks to initiate a discussion of these hurdles, approaches to overcome them, and unique safety and regulatory issues to consider. Cancer Gene Therapy (2002) 9, 1062 – 1067 doi:10.1038/sj.cgt.7700548 Keywords: oncolytic; virotherapy; experimental therapeutics; cancer ew cancer treatments are needed. These agents must limitation of levels of delivery to cancer cells in a solid tumor Nhave novel mechanisms of action and thereby lack mass. Over 10 different virotherapy agents have entered, or cross-resistance with currently available treatments. Viruses will soon be entering, clinical trials; one such adenovirus have evolved to infect, replicate in, and kill human cells (dl1520) has entered a Phase III clinical trial in recurrent through diverse mechanisms. Clinicians treated hundreds of head and neck carcinoma.
    [Show full text]
  • Oncolytic Virotherapy Blockade by Microglia and Macrophages Requires STAT1/3 Zahid M
    Published OnlineFirst November 8, 2017; DOI: 10.1158/0008-5472.CAN-17-0599 Cancer Translational Science Research Oncolytic Virotherapy Blockade by Microglia and Macrophages Requires STAT1/3 Zahid M. Delwar1,2,3, Yvonne Kuo1, Yan H. Wen1,4, Paul S. Rennie3, and William Jia1,2 Abstract The first oncolytic virotherapy employing HSV-1 (oHSV-1) STAT1/3 was determined to be responsible for suppressing was approved recently by the FDA to treat cancer, but further oHSV-1 replication in macrophages/microglia. Treatment improvements in efficacy are needed to eradicate challenging with the oxindole/imidazole derivative C16 rescued oHSV-1 refractory tumors, such as glioblastomas (GBM). Microglia/ replication in microglia/macrophages by inhibiting STAT1/3 macrophages comprising approximately 40% of a GBM tumor activity. In the U87 xenograft model of GBM, C16 treatment may limit virotherapeutic efficacy. Here, we show these cells overcame the microglia/macrophage barrier, thereby facilitat- suppress oHSV-1 growth in gliomas by internalizing the virus ing tumor regression without causing a spread of the virus to through phagocytosis. Internalized virus remained capable of normal organs. Collectively, our results suggest a strategy to expressing reporter genes while viral replication was blocked. relieve a STAT1/3-dependent therapeutic barrier and enhance Macrophage/microglia formed a nonpermissive OV barrier, oHSV-1 oncolytic activity in GBM. preventing dissemination of oHSV-1 in the glioma mass. The Significance: These findings suggest a strategy to enhance the deficiency in viral replication in microglial cells was associated therapeutic efficacy of oncolytic virotherapy in glioblastoma. with silencing of particular viral genes. Phosphorylation of Cancer Res; 78(3); 718–30.
    [Show full text]
  • Forced Degradation Studies to Identify Critical Process Parameters for the Purification of Infectious Measles Virus
    viruses Article Forced Degradation Studies to Identify Critical Process Parameters for the Purification of Infectious Measles Virus Daniel Loewe 1,2 , Julian Häussler 1, Tanja A. Grein 1 , Hauke Dieken 1, Tobias Weidner 1, Denise Salzig 1 and Peter Czermak 1,2,3,* 1 Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstraße 14, 35390 Giessen, Germany 2 Faculty of Biology and Chemistry, University of Giessen, Heinrich-Buff-Ring 17, 35392 Giessen, Germany 3 Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Project group Bioresources, Winchesterstr. 3, 35394 Giessen, Germany * Correspondence: [email protected]; Tel.: +49-641-309-2551 Received: 25 June 2019; Accepted: 3 August 2019; Published: 7 August 2019 Abstract: Oncolytic measles virus (MV) is a promising treatment for cancer but titers of up to 1011 infectious particles per dose are needed for therapeutic efficacy, which requires an efficient, robust, and scalable production process. MV is highly sensitive to process conditions, and a substantial fraction of the virus is lost during current purification processes. We therefore conducted forced degradation studies under thermal, pH, chemical, and mechanical stress to determine critical process parameters. We found that MV remained stable following up to five freeze–thaw cycles, but was inactivated during short-term incubation (< 2 h) at temperatures exceeding 35 ◦C. The infectivity of MV declined at pH < 7, but was not influenced by different buffer systems or the ionic strength/osmolality, except high concentrations of CaCl2 and MgSO4. We observed low shear sensitivity (dependent on the flow rate) caused by the use of a peristaltic pump.
    [Show full text]