A Newcastle Disease Virus (NDV) Expressing a Membrane-Anchored Spike as a Cost-Effective Inactivated SARS-CoV-2 Vaccine

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

Abstract

A successful severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine must not only be safe and protective, but must also meet the demand on a global scale at a low cost. Using the current influenza virus vaccine production capacity to manufacture an egg-based inactivated Newcastle disease virus (NDV)/SARS-CoV-2 vaccine would meet that challenge. Here, we report pre-clinical evaluations of an inactivated NDV chimera stably expressing the membrane-anchored form of the spike (NDV-S) as a potent coronavirus disease 2019 (COVID-19) vaccine in mice and hamsters. The inactivated NDV-S vaccine was immunogenic, inducing strong binding and/or neutralizing antibodies in both animal models. More importantly, the inactivated NDV-S vaccine protected animals from SARS-CoV-2 infections. In the presence of an adjuvant, antigen-sparing could be achieved, which would further reduce the cost while maintaining the protective efficacy of the vaccine.

Keywords: COVID-19; adjuvant; antigen-sparing; egg-based vaccine; hamster model; mouse-adapted SARS-CoV-2.

Conflict of interest statement

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”.

Figures

Figure 1
Figure 1
Design and concept of an inactivated Newcastle disease virus (NDV)-based severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine. (A) Design of the NDV-S vaccine. The sequence of the spike-fusion (S-F) chimera (green: ectodomain of S, and black: the transmembrane domain and cytoplasmic tail of the NDV F protein) was inserted between the P and M gene of the NDV LaSota (NDV_LS) strain L289A mutant (NDV_LS/L289A). NDV-S: NDV_LS/L289A_S-F. The polybasic cleavage site of the S was removed (682RRAR685 to A). (B) The concept overview of an inactivated NDV-based SARS-CoV-2 vaccine. The NDV-S vaccine could be produced using the current global influenza virus vaccine production capacity. Such an NDV-S vaccine displays abundant S proteins on the surface of the virions. The NDV-S vaccine could be inactivated by beta-propiolactone (BPL). The NDV-S vaccine could be administered intramuscularly (i.m.) to elicit protective antibody responses in humans.
Figure 2
Figure 2
The S-F chimera is stable. (A) Stability of the S-F chimera at 4 °C. Allantoic fluid containing the NDV-S virus was aliquoted into equal amounts (15 mL) and stored at 4 °C. Virus from each aliquot was concentrated through a 20% sucrose cushion, re-suspended in an equal amount of phosphate buffered saline (PBS), and then stored at −80 °C for several weeks (wk 0, wk 1, wk 2, and wk 3). One microgram of each concentrated virus was resolved onto 4–20% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE). Protein degradation was evaluated by Western blot using the S-specific mouse monoclonal antibody 2B3E5. The hemagglutinin-neuraminidase (HN) protein of NDV was used as an NDV protein control. (B) Inactivation of the virus by beta-propiolactone (BPL). Viruses in the allantoic fluid were inactivated by 0.05% BPL, as described previously. Clarified allantoic fluids with live and inactivated viruses were diluted in PBS (at 1000-fold dilution) and inoculated into 10-day-old embryonated chicken eggs. The eggs were incubated at 37 °C for 3 days. The loss of infectivity of the inactivated virus was confirmed by the lack of growth of the virus determined by a hemagglutination (HA) assay. (C) Stability of the S-F before and after BPL inactivation. Live or inactivated (using 0.05% BPL) NDV-S virus was concentrated through a 20% sucrose cushion, as described previously. Two micrograms of live or BPL-inactivated virus were loaded onto 4–20% SDS-PAGE. Stability loss of the S-F was evaluated by Western blot, as described in A.
Figure 3
Figure 3
Inactivated NDV-S vaccine elicits high antibody responses in mice. (A) Immunization regimen and groups. BALB/c mice were given two immunizations via the intramuscular administration route with a 2-week interval. Mice were bled pre-boost and 11 days after the boost for in vitro serological assays. Mice were challenged with a mouse-adapted SARS-CoV-2 strain 19 days after the boost. Ten groups described in the table were included in this study. Group 1, 2, and 3 were immunized with 5, 10, and 20 μg of vaccine, respectively; group 4, 5, and 6 were immunized with 0.2, 1, and 5 μg of vaccine formulated with the R-enantiomer of the cationic lipid DOTAP (R-DOTAP), respectively; group 7, 8, and 9 were immunized with 0.2, 1, and 5 μg of vaccine combined with AddaVax, respectively; and group 10 was immunized with 20 μg of WT NDV virus as the vector-only control. (B) Spike-specific serum IgG titers. Serum IgG titers from animals after prime (pattern bars) and boost (solid bars) toward the recombinant trimeric spike protein were measured by an enzyme linked immunosorbent assay (ELISA). Endpoint titers were shown as the readout for ELISA. (C) Neutralization titers of serum antibodies. Microneutralization assays were performed to determine the neutralizing activities of serum antibodies from animals after the boost (D26) using the USA-WA1/2020 SARS-CoV-2 strain. The 50% of inhibitory dilution (ID50) of serum samples showing no neutralizing activity (WT NDV) was set as 10 (LoD: limit of detection).
Figure 4
Figure 4
Inactivated NDV-S vaccine protects mice from SARS-CoV-2 infection. (A) Weight loss of mice infected with SARS-CoV-2. Weight loss of mice challenged with a mouse-adapted SARS-CoV-2 strain was monitored for 4 days. (B) Viral titers in the lung. Lungs of mice were harvested at day 4 post-infection. Viral titers of the lung homogenates were determined by a plaque assay. Geometric mean titer (PFU/lobe) is shown (LoD: limit of detection). Statistical analysis was performed using the Kruskal–Wallis test with Dunn’s correction for multiple comparisons. P-values between groups were shown.
Figure 5
Figure 5
Inactivated NDV-S vaccine attenuates SARS-CoV-2 infection in hamsters. (A) Immunization regimen and groups. Golden Syrian hamsters were vaccinated with inactivated NDV-S following a prime-boost regimen with a 2-week interval. Hamsters were challenged 24 days after the boost with the USA-WA1/2020 SARS-CoV-2 strain. Four groups of hamsters (n = 8) were included in this study. Group 1 received 10 μg of inactivated NDV-S vaccine without any adjuvant. Group 2 received 5 μg of inactivated NDV-S vaccine adjuvanted with AddaVax. Group 3 receiving the 10 μg of inactivated WT NDV was included as the vector-only (negative) control. Group 4 animals receiving no vaccine were mock challenged with PBS as healthy controls. (B) Spike-specific serum IgG titers. Hamsters were bled pre-boost and a subset of hamsters were terminally bled at 2 days post-infection (dpi). Vaccine-induced serum IgG titers towards the trimeric spike protein were determined by ELISA. Endpoint titers are shown as the readout for ELISA. (C) Weight loss of hamsters challenged with SARS-CoV-2. Weight loss of SARS-CoV-2-infected hamsters was monitored for 5 days. (D) Viral titers in the lungs. Viral titers in the upper right (UR) and lower right (LR) lung lobes of the animals at 2 and 5 dpi were measured by a plaque assay (LoD: limit of detection). Statistical analysis was performed using the Kruskal–Wallis test with Dunn’s correction for multiple comparisons. P-values between groups were shown.

References

    1. Jackson L.A., Anderson E.J., Rouphael N.G., Roberts P.C., Makhene M., Coler R.N., McCullough M.P., Chappell J.D., Denison M.R., Stevens L.J., et al. An mRNA Vaccine against SARS-CoV-2—Preliminary Report. N. Engl. J. Med. 2020;383:1920–1931. doi: 10.1056/NEJMoa2022483.
    1. Corbett K.S., Edwards D.K., Leist S.R., Abiona O.M., Boyoglu-Barnum S., Gillespie R.A., Himansu S., Schafer A., Ziwawo C.T., DiPiazza A.T., et al. SARS-CoV-2 mRNA vaccine design enabled by prototype pathogen preparedness. Nature. 2020;586:567–571. doi: 10.1038/s41586-020-2622-0.
    1. Gao Q., Bao L., Mao H., Wang L., Xu K., Yang M., Li Y., Zhu L., Wang N., Lv Z., et al. Rapid development of an inactivated vaccine candidate for SARS-CoV-2. Science. 2020;369:77–81. doi: 10.1126/science.abc1932.
    1. Zhu F.C., Li Y.H., Guan X.H., Hou L.H., Wang W.J., Li J.X., Wu S.P., Wang B.S., Wang Z., Wang L., 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. Lancet. 2020;395:1845–1854. doi: 10.1016/S0140-6736(20)31208-3.
    1. DiNapoli J.M., Kotelkin A., Yang L., Elankumaran S., Murphy B.R., Samal S.K., Collins P.L., Bukreyev A. Newcastle disease virus, a host range-restricted virus, as a vaccine vector for intranasal immunization against emerging pathogens. Proc. Natl. Acad. Sci. USA. 2007;104:9788–9793. doi: 10.1073/pnas.0703584104.
    1. Liu R.Q., Ge J.Y., Wang J.L., Shao Y., Zhang H.L., Wang J.L., Wen Z.Y., Bu Z.G. Newcastle disease virus-based MERS-CoV candidate vaccine elicits high-level and lasting neutralizing antibodies in Bactrian camels. J. Integr. Agric. 2017;16:2264–2273. doi: 10.1016/S2095-3119(17)61660-5.
    1. Li K., Li Z., Wohlford-Lenane C., Meyerholz D.K., Channappanavar R., An D., Perlman S., McCray P.B., Jr., He B. Single-Dose, Intranasal Immunization with Recombinant Parainfluenza Virus 5 Expressing Middle East Respiratory Syndrome Coronavirus (MERS-CoV) Spike Protein Protects Mice from Fatal MERS-CoV Infection. mBio. 2020;11:e00554-20. doi: 10.1128/mBio.00554-20.
    1. Liniger M., Zuniga A., Tamin A., Azzouz-Morin T.N., Knuchel M., Marty R.R., Wiegand M., Weibel S., Kelvin D., Rota P.A., et al. Induction of neutralising antibodies and cellular immune responses against SARS coronavirus by recombinant measles viruses. Vaccine. 2008;26:2164–2174. doi: 10.1016/j.vaccine.2008.01.057.
    1. Koch T., Dahlke C., Fathi A., Kupke A., Krahling V., Okba N.M.A., Halwe S., Rohde C., Eickmann M., Volz A., et al. Safety and immunogenicity of a modified vaccinia virus Ankara vector vaccine candidate for Middle East respiratory syndrome: An open-label, phase 1 trial. Lancet Infect. Dis. 2020;20:827–838. doi: 10.1016/S1473-3099(20)30248-6.
    1. Sun W., Leist S.R., McCroskery S., Liu Y., Slamanig S., Oliva J., Amanat F., Schaefer A., Dinnon K., Garcia-Sastre A., et al. Newcastle disease virus (NDV) expressing the spike protein of SARS-CoV-2 as vaccine candidate. EBioMedicine (Accepted) 2020 doi: 10.1016/j.ebiom.2020.103132.
    1. Vasievich E.A., Ramishetti S., Zhang Y., Huang L. Trp2 peptide vaccine adjuvanted with (R)-DOTAP inhibits tumor growth in an advanced melanoma model. Mol. Pharm. 2012;9:261–268. doi: 10.1021/mp200350n.
    1. Smalley Rumfield C., Pellom S.T., Morillon Ii Y.M., Schlom J., Jochems C. Immunomodulation to enhance the efficacy of an HPV therapeutic vaccine. J. Immunother. Cancer. 2020;8:e000612. doi: 10.1136/jitc-2020-000612.
    1. Riehl M., Harms M., Hanefeld A., Baleeiro R.B., Walden P., Mader K. Combining R-DOTAP and a particulate antigen delivery platform to trigger dendritic cell activation: Formulation development and in-vitro interaction studies. Int. J. Pharm. 2017;532:37–46. doi: 10.1016/j.ijpharm.2017.08.119.
    1. Gandhapudi S.K., Ward M., Bush J.P.C., Bedu-Addo F., Conn G., Woodward J.G. Antigen Priming with Enantiospecific Cationic Lipid Nanoparticles Induces Potent Antitumor CTL Responses through Novel Induction of a Type I IFN Response. J. Immunol. 2019;202:3524–3536. doi: 10.4049/jimmunol.1801634.
    1. Langley J.M., Wang L., Aggarwal N., Bueso A., Chandrasekaran V., Cousin L., Halperin S.A., Li P., Liu A., McNeil S., et al. Immunogenicity and Reactogenicity of an Inactivated Quadrivalent Influenza Vaccine Administered Intramuscularly to Children 6 to 35 Months of Age in 2012–2013: A Randomized, Double-Blind, Controlled, Multicenter, Multicountry, Clinical Trial. J. Pediatr. Infect. Dis Soc. 2015;4:242–251. doi: 10.1093/jpids/piu098.
    1. Madan A., Ferguson M., Rheault P., Seiden D., Toma A., Friel D., Soni J., Li P., Innis B.L., Schuind A. Immunogenicity and safety of an AS03-adjuvanted H7N1 vaccine in adults 65years of age and older: A phase II, observer-blind, randomized, controlled trial. Vaccine. 2017;35:1865–1872. doi: 10.1016/j.vaccine.2017.02.057.
    1. Wang L., Chandrasekaran V., Domachowske J.B., Li P., Innis B.L., Jain V.K. Immunogenicity and Safety of an Inactivated Quadrivalent Influenza Vaccine in US Children 6-35 Months of Age During 2013–2014: Results From A Phase II Randomized Trial. J. Pediatr. Infect. Dis. Soc. 2016;5:170–179. doi: 10.1093/jpids/piv041.
    1. Arifin M.A., Mel M., Abdul Karim M.I., Ideris A. Production of Newcastle disease virus by Vero cells grown on cytodex 1 microcarriers in a 2-litre stirred tank bioreactor. J. Biomed. Biotechnol. 2010;2010:586363. doi: 10.1155/2010/586363.
    1. Amanat F., Stadlbauer D., Strohmeier S., Nguyen T.H.O., Chromikova V., McMahon M., Jiang K., Arunkumar G.A., Jurczyszak D., Polanco J., et al. A serological assay to detect SARS-CoV-2 seroconversion in humans. Nat. Med. 2020;26:1033–1036. doi: 10.1038/s41591-020-0913-5.
    1. Vijayakumar G., Zamarin D. Design and Production of Newcastle Disease Virus for Intratumoral Immunomodulation. Methods Mol. Biol. 2020;2058:133–154.
    1. Dinnon K.H., 3rd, Leist S.R., Schafer A., Edwards C.E., Martinez D.R., Montgomery S.A., West A., Yount B.L., Jr., Hou Y.J., Adams L.E., et al. A mouse-adapted model of SARS-CoV-2 to test COVID-19 countermeasures. Nature. 2020;586:560–566. doi: 10.1038/s41586-020-2708-8.
    1. Amanat F., White K.M., Miorin L., Strohmeier S., McMahon M., Meade P., Liu W.C., Albrecht R.A., Simon V., Martinez-Sobrido L., et al. An In Vitro Microneutralization Assay for SARS-CoV-2 Serology and Drug Screening. Curr. Protoc. Microbiol. 2020;58:e108. doi: 10.1002/cpmc.108.
    1. Ania Wajnberg F.A., Firpo A., Altman D., Bailey M., Mansour M., McMahon M., Meade P., Mendu D.R., Muellers K., Stadlbauer D., et al. SARS-CoV-2 infection induces robust, neutralizing antibody responses that are stable for at least three months. medRxiv. 2020 doi: 10.1101/2020.07.14.20151126.
    1. Imai M., Iwatsuki-Horimoto K., Hatta M., Loeber S., Halfmann P.J., Nakajima N., Watanabe T., Ujie M., Takahashi K., Ito M., et al. Syrian hamsters as a small animal model for SARS-CoV-2 infection and countermeasure development. Proc. Natl. Acad. Sci. USA. 2020;117:16587–16595. doi: 10.1073/pnas.2009799117.
    1. Chan J.F., Zhang A.J., Yuan S., Poon V.K., Chan C.C., Lee A.C., Chan W.M., Fan Z., Tsoi H.W., Wen L., et al. Simulation of the clinical and pathological manifestations of Coronavirus Disease 2019 (COVID-19) in golden Syrian hamster model: Implications for disease pathogenesis and transmissibility. Clin. Infect. Dis. 2020 doi: 10.1093/cid/ciaa644.

Source: PubMed

3
Tilaa