Development of Group B Coxsackievirus As an Oncolytic Virus: Opportunities and Challenges

Total Page:16

File Type:pdf, Size:1020Kb

Development of Group B Coxsackievirus As an Oncolytic Virus: Opportunities and Challenges viruses Review Development of Group B Coxsackievirus as an Oncolytic Virus: Opportunities and Challenges Huitao Liu 1,2 and Honglin Luo 1,3,* 1 Centre for Heart Lung Innovation, St. Paul’s Hospital—University of British Columbia, Vancouver, BC V6Z 1Y6, Canada; [email protected] 2 Department of Experimental Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada 3 Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada * Correspondence: [email protected] Abstract: Oncolytic viruses have emerged as a promising strategy for cancer therapy due to their dual ability to selectively infect and lyse tumor cells and to induce systemic anti-tumor immunity. Among various candidate viruses, coxsackievirus group B (CVBs) have attracted increasing attention in recent years. CVBs are a group of small, non-enveloped, single-stranded, positive-sense RNA viruses, belonging to species human Enterovirus B in the genus Enterovirus of the family Picornaviridae. Preclinical studies have demonstrated potent anti-tumor activities for CVBs, particularly type 3, against multiple cancer types, including lung, breast, and colorectal cancer. Various approaches have been proposed or applied to enhance the safety and specificity of CVBs towards tumor cells and to further increase their anti-tumor efficacy. This review summarizes current knowledge and strategies for developing CVBs as oncolytic viruses for cancer virotherapy. The challenges arising from these studies and future prospects are also discussed in this review. Citation: Liu, H.; Luo, H. Keywords: coxsackievirus B; oncolytic virus; cancer; virotherapy; cancer; toxicity; anti-tumor immunity Development of Group B Coxsackievirus as an Oncolytic Virus: Opportunities and Challenges. Viruses 2021, 13, 1082. https://doi.org/ 1. Introduction 10.3390/v13061082 Oncolytic virotherapy represents a new and promising option for cancer treatment. This approach uses engineered or naturally occurring viruses, termed oncolytic viruses, to Academic Editor: Caroline Tapparel target and destroy tumor cells while sparing normal cells [1,2]. Current evidence supports that both direct lysis of cancer cells and anti-tumor immunity triggered by viral infection Received: 4 May 2021 contribute to the anti-tumor efficacy of virotherapy [1,2]. Oncolytic viruses selectively Accepted: 3 June 2021 infect and replicate in tumor cells, inducing cell destruction and the consequent release Published: 5 June 2021 and spread of progeny virions to infect adjacent cells. Oncolytic viruses can also stimulate immunogenic cell death through promoting the innate and adaptive immune response. Publisher’s Note: MDPI stays neutral The advantage of virotherapy over other classical cancer therapies (e.g., chemo- and with regard to jurisdictional claims in published maps and institutional affil- radiotherapy) lies in the fact that candidate viruses can be genetically manipulated to iations. increase their potency against specific cancer types. As a milestone in virotherapy, in 2015, the US FDA approved the first oncolytic agent Imlygic (Talimogene laherparepvec, T-Vec) for the treatment of melanoma [3,4]. T-Vec is a modified herpes simplex virus-1 (HSV-1) genetically engineered to encode the im- munostimulatory cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF). Copyright: © 2021 by the authors. Although the survival benefits remain controversial, the approval of T-Vec has promoted Licensee MDPI, Basel, Switzerland. significant interest in the development of oncolytic viruses as either a monotherapy or in This article is an open access article combination with other anti-tumor therapies [3,4]. According to clinicaltrials.gov, there are distributed under the terms and conditions of the Creative Commons currently 105 registered oncolytic viral trials in the USA, 21 in Canada, 18 in Spain, and 15 Attribution (CC BY) license (https:// in France among other top countries (Figure1A). In recent years, the number of initiated creativecommons.org/licenses/by/ clinical studies on virotherapy has increased significantly (Figure1B). 4.0/). Viruses 2021, 13, 1082. https://doi.org/10.3390/v13061082 https://www.mdpi.com/journal/viruses Viruses 2021, 13, 1082 2 of 16 Viruses 2021, 13, 1082 2 of 15 Spain, and 15 in France among other top countries (Figure 1A). In recent years, the number of initiated clinical studies on virotherapy has increased significantly (Figure 1B). A Rank Countries Clinical trials recorded* B 1 United States 105 25 2 Canada 21 y d s l u a t 20 3 Spain 18 i s r t s l l a 4 France 15 a i c i r 15 t n i l l 5 United Kingdom 14 a c c f i o n i 10 r 6 China (mainland) 11 l e c b f 7 Germany 8 o m . 5 u O N 7 Australia 8 N 9 Republic of Korea 7 0 00 1 1 2 2 33 44 5 5 66 77 88 99 00 11 22 33 44 55 6 6 77 88 99 00 11 22 0 0 0 0 0 0 0 0 0 0 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 2 2 2 2 2 2 10 Japan 5 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 2 2 2 2 2 2 2 2 2 2 2 2 Year2 2 2 2 2 2 2 2 2 2 2 * Same clinical trials may be taken in multiple countries. Years Figure 1.FigureNumber 1. Number of registered of registered clinical clinical trials trials on on oncolytic oncolytic viralviral studies studies from from clinicaltrials.gov clinicaltrials.gov in top in10 topcountries 10 countries (A) from (A) from 2001 to 20212001 (toB ).2021 Note (B). thatNote thethat datathe data of 2021of 2021 only only cover cover clinicalclinical studies studies registered registered until until 31 May 31 [ May2021. Search 2021. keywords Search keywords ‘oncolytic’ and ‘tumor’ found 182 records in total with five excluded for not involving oncolytic virus. ‘oncolytic’ and ‘tumor’ found 182 records in total with five excluded for not involving oncolytic virus. Research in the field of oncolytic RNA viruses has grown rapidly over the past dec- ade.Research Several in RNA the viruses field of that oncolytic exhibit oncolytic RNA viruses activity has hav growne been rapidlytested in overearly theclinical past decade. Severaltrials, RNA which viruses include thatNewcastle exhibit disease oncolytic virus [5], activity measles have virus been [6], vesicular tested in stomatitis early clinical tri- als,virus which (VSV include) [7], Seneca Newcastle valley virus disease [8], reovirus virus [[9],5], respiratory measles virus syncytial [6], virus vesicular [10], po- stomatitis virusliovirus (VSV) [11], [7], and Seneca coxsackievirus valley virus [12]. A [8mong], reovirus enteroviruses, [9], respiratory the PVSRIPO, syncytial a modified virus po- [10], po- liovirusliovirus [11 ],carrying and coxsackievirus an internal ribosomal [12]. Amongentry site enteroviruses,(IRES) of human the rhinovirus PVSRIPO, type a 2 modified poliovirus[11,13,14] carrying, and the CVA21 an internal-based CAVATAK ribosomal [15 entry–19] are site the (IRES) two best of-characterized human rhinovirus onco- type lytic viruses. A number of clinical trials have been conducted on them and shown prom- 2 [11ising,13,14 efficacy.], and theIn 2018, CVA21-based the CAVATAK CAVATAK start-up company, [15–19] are Viralytics, the two was best-characterized acquired by on- colyticMerck viruses. for $394 A million. number This of remarkable clinical trialsnews has have been been very conducted encouraging, on especially them and to shown promisingthose interested efficacy. inIn developing 2018, the enteroviruses CAVATAK for start-up cancer company,therapy. While Viralytics, PVSRIPO was and acquired by MerckCAVATAK for $394 have million.demonstrated This effectiveness remarkable again newsst certain has been tumors, very they encouraging, have less effi- especially to thosecacy against interested other in tumors developing and can sometimes enteroviruses cause foradverse cancer side therapy. effects. Thus, While investiga- PVSRIPO and CAVATAKtion into have additional demonstrated enteroviruses effectiveness is needed. against certain tumors, they have less efficacy Coxsackieviruses are a group of non-enveloped, positive-sense, single-stranded against other tumors and can sometimes cause adverse side effects. Thus, investigation RNA viruses belonging to the species human Enterovirus in the family of Picornaviridae into[20] additional. Coxsackieviruses enteroviruses were isorig needed.inally classified into two groups—A (CVA) and B (CoxsackievirusesCVB)—on the basis of are early a group observation of non-enveloped, of the differences positive-sense, in tissue damage single-stranded induced in RNA virusesnewborn belonging mice [21]. to The the first species report humanon oncolyticEnterovirus CVB was publishedin the family in 1997, of whichPicornaviridae inves- [20]. Coxsackievirusestigated the anti- weretumor originallyproperties of classified CVB1 against into human two groups—A colon cancer (CVA) [22]. Since and then, B (CVB)—on the basisseveral of efforts early by observation different laboratories of the differenceshave been made in tissueto assess damage the oncolytic induced potential in newborn miceand [21 safety]. The of first CVBs report targeting on oncolyticvarious forms CVB of tumor was published in pre-clinical in 1997,studies which [23–30]. investigated In this the review, we summarize the recent progress in developing CVBs, with a focus on CVB3, as anti-tumor properties of CVB1 against human colon cancer [22]. Since then, several efforts an oncolytic virus against different types of cancer. We also discuss the potential barriers by differentand future laboratories perspectives for have the beenclinical made use of toCVBs assess in cancer the oncolytictherapy. potential and safety of CVBs targeting various forms of tumor in pre-clinical studies [23–30]. In this review, we summarize2. Viral Structure the recent and progress Life-Cycle in developing CVBs, with a focus on CVB3, as an oncolytic virus againstThe CVBs different contain types six viral of cancer.types, named We also CVB1 discuss-CVB6, thewhich potential are common barriers human and future perspectivespathogens forassociated the clinical with a usewide of range CVBs of diseases in cancer from therapy.
Recommended publications
  • 1 Systemic Combination Virotherapy for Melanoma with Tumor Antigen-Expressing Vesicular Stomatitis Virus and Adoptive T Cell
    Author Manuscript Published OnlineFirst on July 26, 2012; DOI: 10.1158/0008-5472.CAN-12-0600 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Systemic Combination Virotherapy For Melanoma With Tumor Antigen-Expressing Vesicular Stomatitis Virus And Adoptive T Cell Transfer Diana M. Rommelfanger1,2, Phonphimon Wongthida1, Rosa M. Diaz1,3, Karen M. Kaluza1,3, Jill M. Thompson1, Timothy J. Kottke1, and Richard G. Vile1,3 1Department of Molecular Medicine, 2Department of Molecular Pharmacology and Experimental Therapeutics, 3Department of Immunology, Mayo Clinic, Rochester, MN 55905 Running title: Antitumor effects of systemic combination viro-immunotherapy Keywords: vesicular stomatitis virus, adoptive cell transfer, melanoma, virotherapy, tumor- associated antigen This work was supported by the Richard M. Schulze Family Foundation, the Mayo Foundation, and by NIH grants CA107082, CA130878, and CA132734. Correspondence: Richard Vile, Ph.D. / Mayo Clinic / 200 First St SW / Rochester, MN 55905 Phone: 507-284-9941 / Fax: 507-266-2122 / Email: [email protected] The authors declare no conflict of interest. 1 Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 26, 2012; DOI: 10.1158/0008-5472.CAN-12-0600 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Abstract Oncolytic virotherapy offers the potential to treat tumors both as a single agent and in combination with traditional modalities such as chemotherapy and radiotherapy. Here we describe an effective, fully systemic treatment regimen, which combines virotherapy, acting essentially as an adjuvant immunotherapy, with adoptive cell transfer (ACT).
    [Show full text]
  • 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]
  • 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.
    [Show full text]
  • Adjuvant Oncolytic Virotherapy for Personalized Anti-Cancer Vaccination
    ARTICLE https://doi.org/10.1038/s41467-021-22929-z OPEN Adjuvant oncolytic virotherapy for personalized anti-cancer vaccination D. G. Roy 1,2, K. Geoffroy 3,4,5, M. Marguerie1,2, S. T. Khan1,2, N. T. Martin1, J. Kmiecik6,7, D. Bobbala6, A. S. Aitken1,2, C. T. de Souza1, K. B. Stephenson6, B. D. Lichty6,8, R. C. Auer1,2, D. F. Stojdl2,6,7, J. C. Bell 1,2 & ✉ M.-C. Bourgeois-Daigneault 3,4,5 By conferring systemic protection and durable benefits, cancer immunotherapies are emer- 1234567890():,; ging as long-term solutions for cancer treatment. One such approach that is currently undergoing clinical testing is a therapeutic anti-cancer vaccine that uses two different viruses expressing the same tumor antigen to prime and boost anti-tumor immunity. By providing the additional advantage of directly killing cancer cells, oncolytic viruses (OVs) constitute ideal platforms for such treatment strategy. However, given that the targeted tumor antigen is encoded into the viral genomes, its production requires robust infection and therefore, the vaccination efficiency partially depends on the unpredictable and highly variable intrinsic sensitivity of each tumor to OV infection. In this study, we demonstrate that anti-cancer vaccination using OVs (Adenovirus (Ad), Maraba virus (MRB), Vesicular stomatitis virus (VSV) and Vaccinia virus (VV)) co-administered with antigenic peptides is as efficient as antigen-engineered OVs and does not depend on viral replication. Our strategy is particularly attractive for personalized anti-cancer vaccines targeting patient-specific mutations. We suggest that the use of OVs as adjuvant platforms for therapeutic anti-cancer vaccination warrants testing for cancer treatment.
    [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]
  • A Bibliometric Review of Oncolytic Virus Research As a Novel Approach For
    Mozafari Nejad et al. Virol J (2021) 18:98 https://doi.org/10.1186/s12985-021-01571-7 REVIEW Open Access A bibliometric review of oncolytic virus research as a novel approach for cancer therapy Amir Sasan Mozafari Nejad1 , Tehjeeb Noor2, Ziaul Haque Munim3, Mohammad Yousef Alikhani4* and Amir Ghaemi5* Abstract Background: In recent years, oncolytic viruses (OVs) have drawn attention as a novel therapy to various types of can- cers, both in clinical and preclinical cancer studies all around the world. Consequently, researchers have been actively working on enhancing cancer therapy since the early twentieth century. This study presents a systematic review of the literature on OVs, discusses underlying research clusters and, presents future directions of OVs research. Methods: A total of 1626 published articles related to OVs as cancer therapy were obtained from the Web of Science (WoS) database published between January 2000 and March 2020. Various aspects of OVs research, including the countries/territories, institutions, journals, authors, citations, research areas, and content analysis to fnd trending and emerging topics, were analysed using the bibliometrix package in the R-software. Results: In terms of the number of publications, the USA based researchers were the most productive (n 611) followed by Chinese (n 197), and Canadian (n 153) researchers. The Molecular Therapy journal ranked frst= both in terms of the number= of publications (n 133)= and local citations (n 1384). The most prominent institution was Mayo Clinic from the USA (n 117) followed= by the University of Ottawa= from Canada (n 72), and the University of Helsinki from Finland (n 63).= The most impactful author was Bell J.C with the highest number= of articles (n 67) and total local citations (n =885).
    [Show full text]
  • CRISPR-Cas9: a New and Promising Player in Gene Therapy
    Downloaded from http://jmg.bmj.com/ on October 22, 2015 - Published by group.bmj.com Methods CRISPR-Cas9: a new and promising player in gene therapy 1 2 3 4,1 5 Editor’s choice Lu Xiao-Jie, Xue Hui-Ying, Ke Zun-Ping, Chen Jin-Lian, Ji Li-Juan Scan to access more free content 1Department of ABSTRACT as insertional mutations and non-physical expres- Gastroenterology, Shanghai First introduced into mammalian organisms in 2013, the sion of proteins, the programmable nucleases use a East Hospital, Tongji University ‘ ’ School of Medicine, Shanghai, RNA-guided genome editing tool CRISPR-Cas9 (clustered cut-and-paste strategy, that is, remove the defect China regularly interspaced short palindromic repeats/CRISPR- and install the correct, thus representing an prefer- 2The Reproductive Center, associated nuclease 9) offers several advantages over able tool for gene therapy. Recently, a RNA-guided Jiangsu Huai’an Maternity and conventional ones, such as simple-to-design, easy-to-use genome editing tool termed CRISPR-Cas9 (clus- ’ Children Hospital, Huai an, and multiplexing (capable of editing multiple genes tered regularly interspaced short palindromic China 3Department of Cardiology, simultaneously). Consequently, it has become a cost- repeats/CRISPR-associated nuclease 9) added to the The Fifth People’s Hospital effective and convenient tool for various genome editing list of programmable nucleases, offers several of Shanghai, Fudan University, purposes including gene therapy studies. In cell lines or advantages over its counterparts and shows thera- Shanghai, China animal models, CRISPR-Cas9 can be applied for peutic potentials. Herein, we introduce the basic 4Department of Gastroenterology, Shanghai therapeutic purposes in several ways.
    [Show full text]