Newcastle disease virus (NDV) expressing the spike protein of SARS-CoV-2 as vaccine candidate

Due to the lack of protective immunity of humans towards the newly emerged SARS-CoV-2, this virus has caused a massive pandemic across the world resulting in hundreds of thousands of deaths. Thus, a vaccine is urgently needed to contain the spread of the virus. Here, we describe Newcastle disease virus (NDV) vector vaccines expressing the spike protein of SARS-CoV-2 in its wild type or a pre-fusion membrane anchored format. All described NDV vector vaccines grow to high titers in embryonated chicken eggs. In a proof of principle mouse study, we report that the NDV vector vaccines elicit high levels of antibodies that are neutralizing when the vaccine is given intramuscularly. Importantly, these COVID-19 vaccine candidates protect mice from a mouse-adapted SARS-CoV-2 challenge with no detectable viral titer and viral antigen in the lungs. Research in context Evidence before this study The spike (S) protein of the SARS-CoV-2 is the major antigen that notably induces neutralizing antibodies to block viral entry. Many COVID-19 vaccines are under development, among them viral vectors expressing the S protein of SARS-CoV-2 exhibit many benefits. Viral vector vaccines have the potential of being used as both live or inactivated vaccines and they can induce Th1 and Th2-based immune responses following different immunization regimens. Additionally, viral vector vaccines can be handled under BSL-2 conditions and they grow to high titers in cell cultures or other species restricted-hosts. For a SARS-CoV-2 vaccine, several viral vectors are being tested, such as adenovirus, measles virus and Modified vaccinia Ankara. Added value of this study The NDV vector vaccine against SARS-CoV-2 described in this study has advantages similar to those of other viral vector vaccines. But the NDV vector can be amplified in embryonated chicken eggs, which allows for high yields and low costs per dose. Also, the NDV vector is not a human pathogen, therefore the delivery of the foreign antigen would not be compromised by any pre-existing immunity in humans. Finally, NDV has a very good safety record in humans, as it has been used in many oncolytic virus trials. This study provides an important option for a cost-effective SARS-CoV-2 vaccine. Implications of all the available evidence This study informs of the value of a viral vector vaccine against SARS-CoV-2. Specifically, for this NDV based SARS-CoV-2 vaccine, the existing egg-based influenza virus vaccine manufactures in the U.S. and worldwide would have the capacity to rapidly produce hundreds of millions of doses to mitigate the consequences of the ongoing COVID-19 pandemic.

[1]  J. Mascola,et al.  An mRNA Vaccine against SARS-CoV-2 — Preliminary Report , 2020, The New England journal of medicine.

[2]  R. Albrecht,et al.  An In Vitro Microneutralization Assay for SARS‐CoV‐2 Serology and Drug Screening , 2020, Current protocols in microbiology.

[3]  J. Mascola,et al.  SARS-CoV-2 mRNA Vaccine Development Enabled by Prototype Pathogen Preparedness , 2020, bioRxiv.

[4]  L. Ren,et al.  Effect of Convalescent Plasma Therapy on Time to Clinical Improvement in Patients With Severe and Life-threatening COVID-19: A Randomized Clinical Trial. , 2020, JAMA.

[5]  A. Basu,et al.  Faculty Opinions recommendation of Safety, tolerability, and immunogenicity of a recombinant adenovirus type-5 vectored COVID-19 vaccine: a dose-escalation, open-label, non-randomised, first-in-human trial. , 2020, Faculty Opinions – Post-Publication Peer Review of the Biomedical Literature.

[6]  C. Poh,et al.  Two linear epitopes on the SARS-CoV-2 spike protein that elicit neutralising antibodies in COVID-19 patients , 2020, Nature Communications.

[7]  Y. Hu,et al.  Safety, tolerability, and immunogenicity of a recombinant adenovirus type-5 vectored COVID-19 vaccine: a dose-escalation, open-label, non-randomised, first-in-human trial , 2020, The Lancet.

[8]  Amalio Telenti,et al.  Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody , 2020, Nature.

[9]  C. Cunningham-Rundles,et al.  A serological assay to detect SARS-CoV-2 seroconversion in humans , 2020, Nature Medicine.

[10]  Lisa E. Gralinski,et al.  A mouse-adapted SARS-CoV-2 model for the evaluation of COVID-19 medical countermeasures , 2020, bioRxiv.

[11]  Xiangxi Wang,et al.  Development of an inactivated vaccine candidate for SARS-CoV-2 , 2020, Science.

[12]  Velizar Shivarov,et al.  Potential SARS-CoV-2 Preimmune IgM Epitopes , 2020, Frontiers in Immunology.

[13]  Yi Wang,et al.  Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial , 2020, The Lancet.

[14]  I. Wilson,et al.  A highly conserved cryptic epitope in the receptor binding domains of SARS-CoV-2 and SARS-CoV , 2020, Science.

[15]  F. Krammer,et al.  SARS-CoV-2 Vaccines: Status Report , 2020, Immunity.

[16]  Nicholas C. Wu,et al.  A highly conserved cryptic epitope in the receptor binding domains of SARS-CoV-2 and SARS-CoV , 2020, Science.

[17]  Arturo Casadevall,et al.  The convalescent sera option for containing COVID-19. , 2020, The Journal of clinical investigation.

[18]  R. Scheuermann,et al.  A Sequence Homology and Bioinformatic Approach Can Predict Candidate Targets for Immune Responses to SARS-CoV-2 , 2020, Cell Host & Microbe.

[19]  Matthew R. McKay,et al.  Preliminary Identification of Potential Vaccine Targets for the COVID-19 Coronavirus (SARS-CoV-2) Based on SARS-CoV Immunological Studies , 2020, bioRxiv.

[20]  D. Zamarin,et al.  Design and Production of Newcastle Disease Virus for Intratumoral Immunomodulation. , 2019, Methods in molecular biology.

[21]  P. Palese,et al.  Oncolytic Newcastle disease virus expressing a checkpoint inhibitor as a radioenhancing agent for murine melanoma , 2019, EBioMedicine.

[22]  Z. Bu,et al.  Newcastle disease virus-based MERS-CoV candidate vaccine elicits high-level and lasting neutralizing antibodies in Bactrian camels , 2017, Journal of Integrative Agriculture.

[23]  V. Schirrmacher Fifty Years of Clinical Application of Newcastle Disease Virus: Time to Celebrate! , 2016, Biomedicines.

[24]  P. Palese,et al.  Oncolytic Newcastle disease virus for cancer therapy: old challenges and new directions. , 2012, Future microbiology.

[25]  A. García-Sastre,et al.  Recombinant Newcastle Disease Virus as a Vaccine Vector for Cancer Therapy , 2008, Molecular Therapy.

[26]  B. Murphy,et al.  Newcastle disease virus, a host range-restricted virus, as a vaccine vector for intranasal immunization against emerging pathogens , 2007, Proceedings of the National Academy of Sciences.

[27]  Jaap Goudsmit,et al.  Human Monoclonal Antibody Combination against SARS Coronavirus: Synergy and Coverage of Escape Mutants , 2006, PLoS medicine.

[28]  A. García-Sastre,et al.  Engineered viral vaccine constructs with dual specificity: avian influenza and Newcastle disease. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[29]  J. Gomori,et al.  Phase I/II trial of intravenous NDV-HUJ oncolytic virus in recurrent glioblastoma multiforme. , 2006, Molecular therapy : the journal of the American Society of Gene Therapy.

[30]  Jianrong Li,et al.  Decreased Dependence on Receptor Recognition for the Fusion Promotion Activity of L289A-Mutated Newcastle Disease Virus Fusion Protein Correlates with a Monoclonal Antibody-Detected Conformational Change , 2005, Journal of Virology.

[31]  J. Marshall,et al.  Phase I trial of intravenous administration of PV701, an oncolytic virus, in patients with advanced solid cancers. , 2002, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[32]  E. Mundt,et al.  Generation of recombinant lentogenic Newcastle disease virus from cDNA. , 1999, The Journal of general virology.

[33]  B. Peeters,et al.  Rescue of Newcastle Disease Virus from Cloned cDNA: Evidence that Cleavability of the Fusion Protein Is a Major Determinant for Virulence , 1999, Journal of Virology.