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

Weina Sun, Sarah R Leist, Stephen McCroskery, Yonghong Liu, Stefan Slamanig, Justine Oliva, Fatima Amanat, Alexandra Schäfer, Kenneth H Dinnon 3rd, Adolfo García-Sastre, Florian Krammer, Ralph S Baric, Peter Palese, Weina Sun, Sarah R Leist, Stephen McCroskery, Yonghong Liu, Stefan Slamanig, Justine Oliva, Fatima Amanat, Alexandra Schäfer, Kenneth H Dinnon 3rd, Adolfo García-Sastre, Florian Krammer, Ralph S Baric, Peter Palese

Abstract

Background: 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.

Methods: Here, we describe Newcastle disease virus (NDV) vector vaccines expressing the spike protein of SARS-CoV-2 in its wild type format or a membrane-anchored format lacking the polybasic cleavage site. All described NDV vector vaccines grow to high titers in embryonated chicken eggs. In a proof of principle mouse study, the immunogenicity and protective efficacy of these NDV-based vaccines were investigated.

Findings: We report that the NDV vector vaccines elicit high levels of antibodies that are neutralizing when the vaccine is given intramuscularly in mice. 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.

Interpretation: The results suggested that the NDV vector expressing either the wild type S or membrane-anchored S without the polybasic cleavage site could be used as live vector vaccine against SARS-CoV-2.

Funding: This work is supported by an NIAID funded Center of Excellence for Influenza Research and Surveillance (CEIRS) contract, the Collaborative Influenza Vaccine Innovation Centers (CIVIC) contract, philanthropic donations and NIH grants.

Keywords: Coronavirus vaccine; Intramuscular administration; Live COVID-19 vaccine; Mouse-adapted SARS-CoV-2; Neutralizing antibodies; Viral vector vaccine.

Conflict of interest statement

Declaration of Competing Interests The Icahn School of Medicine at Mount Sinai has filed patent applications entitled “RECOMBINANT NEWCASTLE DISEASE VIRUS EXPRESSING SARS-COV-2 SPIKE PROTEIN AND USES THEREOF” (63/057,267), in which W.S., F.K., A.G.S and P.P. were listed as inventors. A.G.S and P.P. also declared COI as consultants for AviMex and patents entitled “USE OF RECOMBINANT NDV EXPRESSING CHIMERIC ANTIGENS FOR VETERINARIAN VACCINE” (9387,242) and “USE OF RECOMBINANT NDV FOR ONCOLYTIC THERAPIES” (10,251,922). S.R.L, S.M., Y.L, S.S., J.O, F.A., A.S, K.H.D. and R.S.B. have nothing to declare

Copyright © 2020 The Authors. Published by Elsevier B.V. All rights reserved.

Figures

Fig. 1
Fig. 1
NDV vectors expressing the spike protein of SARS-CoV-2. (a) Two forms of spike proteins expressed by NDV. Spike (S) has the wild type amino acid sequence. The Spike-F chimera (S-F) consists of the ectodomain of S without the polybasic cleavage site and the transmembrane domain (TM) and cytoplasmic tail (CT) of the F protein from NDV. (b) Illustration of genome structures of wild type NDV LaSota (WT NDV_LS), NDV expressing the S or S-F in the wild type LaSota backbone (NDV_LS_S or NDV_LS_S-F) or NDV expressing the S-F in the L289A mutant backbone (NDV_LS/L289A_S-F). The L289A mutation supports the HN-independent fusion of the F protein. (c) Titers of NDV vectors grown in embryonated chicken eggs. The rescued viruses were grown in 10-day old embryonated chicken eggs for 2 or 3 days at 37 °C at limited dilutions. The peak titers of each virus were determined by immunofluorescence assay (IFA). The error bars represent standard deviation (SD).
Fig. 2
Fig. 2
Expression of spike protein in infected cells and NDV particles. (a) Expression of the S and S-F protein in infected cells. Vero E6 cells were infected with three NDV vectors encoding the S or S-F for 16 to 18 h. A WT NDV control was included. The next day, cells were fixed with methanol-free paraformaldehyde. Surface proteins were stained with anti-NDV rabbit serum or a spike receptor-binding domain (RBD)-specific monoclonal antibody CR3022. A scale bar of 200 µM were shown. (b) Incorporation of S and S-F into NDV particles. Three NDV vectors expressing the S or S-F including the NDV_LS_S (green), NDV_LS_S-F (red) and NDV_LS/L289A_S-F (blue) were concentrated through a 20% sucrose cushion. Two clones were shown for NDV_LS_S and NDV_LS_S-F. The concentrated WT NDV expressing no transgenes was used as a control. Two micrograms of each concentrated virus were resolved on a 4–20% SDS-PAGE, the spike protein and NDV HN protein were detected by western blot using an anti-spike 2B3E5 mouse monoclonal antibody and an anti-HN 8H2 mouse monoclonal antibody.
Fig. 3
Fig. 3
NDV vector vaccines elicit high titers of binding and neutralizing antibodies in mice. (a) Vaccination groups and regimen. A prime-boost vaccination regimen was used with a three-week interval. Mice (n = 5) were bled pre-boost and 8 days after the boost. Mice were challenged with a mouse-adapted SARS-CoV-2 MA strain 11 days after the boost. A total of ten groups of mice were used in a vaccination and challenge study. Group 1 (10 µg) and 2 (50 µg) received the WT NDV; Group 3 (10 µg) and 4 (50 µg) received the NDV_LS_S; Group 5 (10 µg) and 6 (50 µg) received NDV_LS_S-F; Group 7 (10 µg) and 8 (50 µg) received NDV_LS/L289A_S-F; Group 9 received PBS as negative controls. An age-matched healthy control group 10 was provided upon challenge. (b) Spike-specific serum IgG titers measured by ELISAs. Sera from animals at 3 weeks after-prime (patterned bars) and 8 days after-boost (solid bars) were isolated. Serum IgG was measured against a recombinant trimeric spike protein by ELISAs. The endpoint titers (mean with SD) were calculated as the readout for ELISAs. (c) Neutralization titers of serum antibodies. Sera from 3 weeks after-prime and 8 days after-boost were pooled within each group. Technical duplicates were performed to measure neutralization activities of serum antibodies using a USA-WA1/2020 SARS-CoV-2 strain. The ID50 value was calculated as the readout of the neutralization assay. For the samples (WT NDV and PBS groups) showing no neutralizing activity in the assay, an ID50 of 10 was given as the starting dilution of the sera is 1:20 (LoD: limit of detection). The error bars represent SD.
Fig. 4
Fig. 4
NDV vector vaccines protected mice from the SARS-CoV-2 challenge. (a) Viral titers in the lungs. All mice were infected intranasally with 104 PFU SARS-CoV-2 MA strain except the healthy control group, which was mock infected with PBS. At day 4 post-challenge, lungs (n = 3) were collected and homogenized in PBS. Viral titers in the lung homogenates were determined by plaque assay. Plaque-forming units (PFU) per lung lobe was calculated. Geometric mean titer was shown for all the groups (LoD: limit of detection). Statistical analysis was performed using one-way analysis of variance (ANOVA), and corrected for multiple comparisons using Dunnett's test (ns, not significant; **, p<0.01). The error bars represent SD. (b) Immunohistochemistry (IHC) staining of lungs. A SARS-CoV-2 NP specific antibody was used for IHC to detect viral antigens. Slides were counterstained with hematoxylin. A presentative image was shown for each group. The brown staining indicates the presence of NP protein of SARS-CoV-2. A scale bar of 100 µm is shown.

References

    1. Jackson L.A., Anderson E.J., Rouphael N.G., Roberts P.C., Makhene M., Coler R.N. An mRNA vaccine against SARS-CoV-2 - preliminary report. N Engl J Med. 2020
    1. Corbett K.S., Edwards D.K., Leist S.R., Abiona O.M., Boyoglu-Barnum S., Gillespie R.A. SARS-CoV-2 mRNA vaccine design enabled by prototype pathogen preparedness. Nature. 2020;586(7830):567–571.
    1. Gao Q., Bao L., Mao H., Wang L., Xu K., Yang M. Rapid development of an inactivated vaccine candidate for SARS-CoV-2. Science. 2020
    1. Zhu F.C., Li Y.H., Guan X.H., Hou L.H., Wang W.J., Li J.X. 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. Lancet. 2020;395(10240):1845–1854.
    1. Amanat F., Krammer F. SARS-CoV-2 Vaccines: status Report. Immunity. 2020;52(4):583–589.
    1. Poh C.M., Carissimo G., Wang B., Amrun S.N., Lee C.Y., Chee R.S. Two linear epitopes on the SARS-CoV-2 spike protein that elicit neutralising antibodies in COVID-19 patients. Nat Commun. 2020;11(1):2806.
    1. Grifoni A., Sidney J., Zhang Y., Scheuermann R.H., Peters B., Sette A. A sequence homology and bioinformatic approach can predict candidate targets for immune responses to SARS-CoV-2. Cell Host Microbe. 2020;27(4):671–680. e2.
    1. Ahmed S.F., Quadeer A.A., McKay M.R. Preliminary identification of potential vaccine targets for the COVID-19 coronavirus (SARS-CoV-2) based on SARS-CoV immunological studies. Viruses. 2020;12(3)
    1. Shivarov V., Petrov P.K., Pashov A.D. Potential SARS-CoV-2 pre-immune IgM epitopes. Front Immunol. 2020;11:932.
    1. Pinto D., Park Y.J., Beltramello M., Walls A.C., Tortorici M.A., Bianchi S. Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody. Nature. 2020
    1. Peeters B.P., de Leeuw O.S., Koch G., Gielkens A.L. Rescue of Newcastle disease virus from cloned cDNA: evidence that cleavability of the fusion protein is a major determinant for virulence. J Virol. 1999;73(6):5001–5009.
    1. Romer-Oberdorfer A., Mundt E., Mebatsion T., Buchholz U.J., Mettenleiter T.C. Generation of recombinant lentogenic Newcastle disease virus from cDNA. J Gen Virol. 1999;80(Pt 11):2987–2995.
    1. Zamarin D., Palese P. Oncolytic Newcastle disease virus for cancer therapy: old challenges and new directions. Fut Microbiol. 2012;7(3):347–367.
    1. Vigil A., Martinez O., Chua M.A., Garcia-Sastre A. Recombinant Newcastle disease virus as a vaccine vector for cancer therapy. Mol Ther. 2008;16(11):1883–1890.
    1. Vijayakumar G., Palese P., Goff P.H. Oncolytic Newcastle disease virus expressing a checkpoint inhibitor as a radioenhancing agent for murine melanoma. EBioMedicine. 2019;49:96–105.
    1. DiNapoli J.M., Kotelkin A., Yang L., Elankumaran S., Murphy B.R., Samal S.K. Newcastle disease virus, a host range-restricted virus, as a vaccine vector for intranasal immunization against emerging pathogens. Proc Natl Acad Sci U S A. 2007;104(23):9788–9793.
    1. Liu R.Q., Ge J.Y., Wang J.L., Shao Y., Zhang H.L., Wang J.L. Newcastle disease virus-based MERS-CoV candidate vaccine elicits high-level and lasting neutralizing antibodies in Bactrian camels. J Integr Agric. 2017;16(10):2264–2273.
    1. Freeman A.I., Zakay-Rones Z., Gomori J.M., Linetsky E., Rasooly L., Greenbaum E. Phase I/II trial of intravenous NDV-HUJ oncolytic virus in recurrent glioblastoma multiforme. Mol Ther. 2006;13(1):221–228.
    1. Pecora A.L., Rizvi N., Cohen G.I., Meropol N.J., Sterman D., Marshall J.L. Phase I trial of intravenous administration of PV701, an oncolytic virus, in patients with advanced solid cancers. J Clin Oncol. 2002;20(9):2251–2266.
    1. Schirrmacher V. Fifty years of clinical application of newcastle disease virus: time to celebrate! Biomedicines. 2016;4(3)
    1. Amanat F., Stadlbauer D., Strohmeier S., Nguyen T.H.O., Chromikova V., McMahon M. A serological assay to detect SARS-CoV-2 seroconversion in humans. Nat Med. 2020;26(7):1033–1036.
    1. Vijayakumar G., Zamarin D. Design and production of newcastle disease virus for intratumoral immunomodulation. Methods Mol Biol. 2020;2058:133–154.
    1. Li J., Melanson V.R., Mirza A.M., Iorio R.M. 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. J Virol. 2005;79(2):1180–1190.
    1. ter Meulen J., van den Brink E.N., Poon L.L., Marissen W.E., Leung C.S., Cox F. Human monoclonal antibody combination against SARS coronavirus: synergy and coverage of escape mutants. PLoS Med. 2006;3(7):e237.
    1. Yuan M., Wu N.C., Zhu X., Lee C.D., So R.T.Y., Lv H. A highly conserved cryptic epitope in the receptor binding domains of SARS-CoV-2 and SARS-CoV. Science. 2020;368(6491):630–633.
    1. Percie du Sert N., Hurst V., Ahluwalia A., Alam S., Avey M.T., Baker M. The ARRIVE guidelines 2.0: updated guidelines for reporting animal research. PLoS Biol. 2020;18(7)
    1. Dinnon K.H., 3rd, Leist S.R., Schafer A., Edwards C.E., Martinez D.R., Montgomery S.A. A mouse-adapted model of SARS-CoV-2 to test COVID-19 countermeasures. Nature. 2020;586(7830):560–566.
    1. Amanat F., White K.M., Miorin L., Strohmeier S., McMahon M., Meade P. An in vitro microneutralization assay for SARS-CoV-2 serology and drug screening. Curr Protoc Microbiol. 2020;58(1):e108.
    1. Park M.S., Steel J., Garcia-Sastre A., Swayne D., Palese P. Engineered viral vaccine constructs with dual specificity: avian influenza and Newcastle disease. Proc Natl Acad Sci U S A. 2006;103(21):8203–8208.
    1. Casadevall A., Pirofski L.A. The convalescent sera option for containing COVID-19. J Clin Invest. 2020;130(4):1545–1548.
    1. Li L., Zhang W., Hu Y., Tong X., Zheng S., Yang J. Effect of convalescent plasma therapy on time to clinical improvement in patients with severe and life-threatening COVID-19: a randomized clinical trial. JAMA. 2020
    1. Wang Y., Zhang D., Du G., Du R., Zhao J., Jin Y. Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial. Lancet. 2020;395(10236):1569–1578.
    1. Rohaim M.A., Munir M. A scalable topical vectored vaccine candidate against SARS-CoV-2. Vaccines (Basel) 2020;8(3)

Source: PubMed

3
Subscribe