Search for authorsSearch for similar articles
6
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      SARS-CoV-2 RBD protein enhances the oncolytic activity of the vesicular stomatitis virus

      brief-report

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Despite recent advances in the research on oncolytic viruses (OVs), a better understanding of how to enhance their replication is key to improving their therapeutic index. Understanding viral replication is important to improve treatment outcomes based on enhanced viral spreading within the tumor milieu. The VSV-Δ51 oncolytic virus has been widely used as an anticancer agent with a high selectivity profile. In this study, we examined the role of the SARS-CoV-2 spike protein receptor-binding domain (RBD) in enhancing VSV-Δ51 viral production and oncolytic activity. To test this hypothesis, we first generated a novel VSV-Δ51 mutant that encoded the SARS-COV-2 RBD and compared viral spreading and viral yield between VSV-Δ51-RBD and VSV-Δ51 in vitro. Using the viral plaque assay, we demonstrated that the presence of the SARS-CoV-2 RBD in the VSV-Δ51 genome is associated with a significantly larger viral plaque surface area and significantly higher virus titers. Subsequently, using an ATP release-based assay, we demonstrated that the SARS-CoV-2 RBD could enhance VSV-Δ51 oncolytic activity in vitro. This observation was further supported using the B16F10 tumor model. These findings highlighted a novel use of the SARS-CoV-2 RBD as an anticancer agent.

          Related collections

          Most cited references33

          • Record: found
          • Abstract: found
          • Article: not found

          ONCOLYTIC VIROTHERAPY

          Oncolytic virotherapy is an emerging treatment modality which uses replication competent viruses to destroy cancers. Advances in the past two years include preclinical proof of feasibility for a single-shot virotherapy cure, identification of drugs that accelerate intratumoral virus propagation, new strategies to maximize the immunotherapeutic potential of oncolytic virotherapy, and clinical confirmation of a critical viremic thereshold for vascular delivery and intratumoral virus replication. The primary clinical milestone was completion of accrual in a phase III trial of intratumoral herpes simplex virus therapy using talimogene laherparepvec for metastatic melanoma. Challenges for the field are to select ‘winners’ from a burgeoning number of oncolytic platforms and engineered derivatives, to transiently suppress but then unleash the power of the immune system to maximize both virus spread and anticancer immunity, to develop more meaningful preclinical virotherapy models and to manufacture viruses with orders of magnitude higher yields compared to established vaccine manufacturing processes.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Oncolytic viruses: a new class of immunotherapy drugs

            Key Points Oncolytic viruses mediate anti-tumour responses through a dual mechanism involving viral oncolysis of cancer cells and induction of host anti-tumour immunity. The molecular and cellular mechanisms of action are not fully elucidated but are likely to depend on viral replication within transformed cells, induction of primary cell death, interaction with tumour cell antiviral elements, release of danger signals and initiation of innate and adaptive anti-tumour immunity. A variety of native and genetically modified viruses have been utilized as oncolytic vectors in preclinical studies, which have demonstrated therapeutic activity against several types of cancer. Oncolytic viruses can be genetically modified to decrease pathogenicity, increase lytic potential and enhance immunogenicity, improving the risk–benefit ratio for clinical development. The approval of a modified adenovirus, H101, in China and the pending approval of a modified herpes simplex virus type 1 (HSV-1) encoding granulocyte–macrophage colony stimulating factor (GM-CSF), termed talimogene laherparepvec (T-VEC), by the US Food and Drug Administration (FDA) in the United States is likely to promote further drug development within this new class of cancer therapeutics. Oncolytic viruses face unique challenges in drug development, including the need for optimal clinical trial designs and response assessment that capture therapeutic responses, different regulatory and commercialization pathways, the need for live culture scale-up procedures, and novel biosafety concerns related to viral persistence in patients and transmission to household contacts and health-care providers. Supplementary information The online version of this article (doi:10.1038/nrd4663) contains supplementary material, which is available to authorized users.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              VSV strains with defects in their ability to shutdown innate immunity are potent systemic anti-cancer agents.

              Ideally, an oncolytic virus will replicate preferentially in malignant cells, have the ability to treat disseminated metastases, and ultimately be cleared by the patient. Here we present evidence that the attenuated vesicular stomatitis strains, AV1 and AV2, embody all of these traits. We uncover the mechanism by which these mutants are selectively attenuated in interferon-responsive cells while remaining highly lytic in 80% of human tumor cell lines tested. AV1 and AV2 were tested in a xenograft model of human ovarian cancer and in an immune competent mouse model of metastatic colon cancer. While highly attenuated for growth in normal mice, both AV1 and AV2 effected complete and durable cures in the majority of treated animals when delivered systemically.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                31 January 2023
                2023
                31 January 2023
                : 14
                : 1082191
                Affiliations
                [1] 1 Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk , Tabuk, Saudi Arabia
                [2] 2 Immunology Research Program, King Abdullah International Medical Research Center , Riyadh, Saudi Arabia
                [3] 3 Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences , Riyadh, Saudi Arabia
                [4] 4 Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Instituto Nacional de Tecnología Agropecuaria (INTA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), De los Reseros y N. Repetto s/n, Hurlingham , Buenos Aires, Argentina
                [5] 5 Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján , Buenos Aires, Argentina
                [6] 6 College of Medicine, Taibah University , Almadinah Almunwarah, Saudi Arabia
                [7] 7 King Abdullah International Medical Research Centre, King Saud University for Health Sciences, Ministry of National Guard Health Affairs , Jeddah, Saudi Arabia
                [8] 8 King Abdulaziz City for Science and Technology-Brigham and Women's Hospital (KACST-BWH) Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST) , Riyadh, Saudi Arabia
                [9] 9 Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women’s Hospital, Harvard Medical School , Boston, MA, United States
                [10] 10 Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, MA, United States
                [11] 11 Harvard Stem Cell Institute, Harvard University , Cambridge, MA, United States
                [12] 12 Department of Immunology, Ministry of the National Guard - Health Affairs , Riyadh, Saudi Arabia
                [13] 13 Faculty of Medicine, King Saud bin Abdulaziz University for Health Sciences , Riyadh, Saudi Arabia
                [14] 14 College of Applied Medical Sciences, Taibah University , Madinah, Saudi Arabia
                [15] 15 Strategic Research and Innovation Laboratories, Taibah University , Madinah, Saudi Arabia
                [16] 16 Immunology Research Program, King Abdullah International Medical Research Center , Jeddah, Saudi Arabia
                Author notes

                Edited by: Nitin Saksena, Victoria University, Australia

                Reviewed by: Gregor Ebert, Technical University of Munich, Germany; Hongzhao Li, National Centre for Foreign Animal Disease (NCFAD), Canada

                *Correspondence: Almohanad A. Alkayyal, aalkayyal@ 123456ut.edu.sa ; Ahmad Bakur Mahmoud, abamahmoud@ 123456taibahu.edu.sa

                This article was submitted to Vaccines and Molecular Therapeutics, a section of the journal Frontiers in Immunology

                Article
                10.3389/fimmu.2023.1082191
                9927213
                36798114
                549e9f6f-ce81-4004-9c90-be1cd4dfa48b
                Copyright © 2023 Alkayyal, Ajina, Cacciabue, Alkayyal, Saeedi, Hussain Alshehry, Kaboha, Alotaibi, Zaidan, Shah, Alroqi and Bakur Mahmoud

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 27 October 2022
                : 13 January 2023
                Page count
                Figures: 4, Tables: 0, Equations: 0, References: 29, Pages: 7, Words: 3187
                Funding
                The authors extend their appreciation to the Deputyship for Research & Innovation, Ministry of Education in Saudi Arabia for funding this research work through project number (0026-1442-S).
                Categories
                Immunology
                Brief Research Report

                Immunology
                sars-cov-2 rbd,oncolytic virotherapy,vsv-δ51 enhancement,vsv-δ51 production,b16f10 melanoma models

                Comments

                Comment on this article