Abstract
The emergence of rapidly spreading variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a major challenge to the ability of vaccines and therapeutic antibodies to provide immunity. These variants contain mutations at specific amino acids that might impede vaccine efficacy. BriLife® (rVSV-ΔG-spike) is a newly developed SARS-CoV-2 vaccine candidate currently in Phase II clinical trials. It is based on a replication competent vesicular stomatitis virus (VSV) platform. rVSV-ΔG-spike contains several spontaneously-acquired spike mutations that correspond to SARS-CoV-2 variants’ mutations. We show that human sera from BriLife® vaccinees preserve comparable neutralization titers towards alpha, gamma and delta variants, and show less than 3-fold reduction in neutralization capacity of beta and omicron compared to the original virus. Taken together, we show that human sera from BriLife® vaccinees overall maintain neutralizing antibody response against all tested variants. We suggest that BriLife® acquired mutations may prove advantageous against future SARS-CoV-2 VOCs.
Introduction
BriLife® is a recombinant replication-competent Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) vaccine candidate, based on Vesicular Stomatitis Virus (VSV) platform, in which VSV glycoprotein (G) was replaced by the SARS-CoV-2 spike protein (S), creating rVSV-ΔG-spike [1].
Since the emergence of the original SARS-CoV-2, several variants have been described, including alpha (B.1.1.7), beta (B.1.351), gamma (P.1), delta (B.1.617.2), and most recently omicron variant (B.1.1.529) (https://www.cdc.gov/coronavirus/2019-ncov/variants/variant-info.html(accessed on January 24th, 2022; Figure 1a)). The emergence of SARS-CoV-2 variants presents a challenge to the ability of vaccines to provide proper immune response.
We report here that during its development, BriLife® spontaneously acquired several mutations at specific sites, including but not exclusive to the receptor binding domain (RBD), some of which are identical, or correspond to some of the key mutations or areas of SARS-CoV-2 VOCs, namely N501, E484, Q493 and G685. Here, we examined the neutralization capacity of serum samples from participants in BriLife® phase II clinical trial against the original SARS-CoV-2 virus as well as the alpha, beta, gamma, delta and omicron variants. We show that BriLife®-elicited sera maintain neutralization against SARS-CoV-2 variants. This preservation of neutralization capacity among variant may be attributed to the unique genetic features of BriLife® vaccine.
Materials and methods
Samples
Serum samples of BriLife® vaccinees were obtained from participants in a randomized, multi-center, placebo-controlled, dose-escalation phase II of an ongoing clinical trial, aimed to evaluate the safety, immunogenicity and potential efficacy of BriLife®, an rVSV-SARS-CoV-2-S vaccine (IIBR-100) in adults (ClinicalTrials.gov -NCT04608305). All participants were vaccinated intramuscularly in a prime-boost regimen (28 days apart). Samples were obtained approximately two weeks following boost vaccination. This study was performed in compliance with International Council for Harmonization (ICH) Good Clinical Practices (GCP), including the archiving of essential documents as well as the ethical principles of the Declaration of Helsinki.
Serum samples of convalescent COVID-19 patients diagnosed at the first wave of the pandemic were collected by the National Blood Services of “Magen David Adom” in Israel within a protocol for plasma donation. All convalescent volunteers gave their informed consent to the National Blood services of Magen David Adom. The study was approved by the ethics committee of the Israeli Ministry of Health (0083-20-WOMC) [2].
Cells
Vero E6 cells (ATCC CRL-1586(tm)) were grown in DMEM containing 10% fetal bovine serum (FBS), MEM nonessential amino acids (NEAA), 2 mM L-glutamine, 100 Units/ml penicillin, 0.1 mg/ml streptomycin, 12.5 Units/ml nystatin (P/S/N). Calu3 cells (ATCC HTB-55) were grown in RPMI supplemented with 10% FBS, NEAA, 2 mM L-glutamine, P/S/N, and 1% Na-pyruvate, all reagents are from Biological Industries, Beit-Haemek, Israel. Cells were cultured at 37 °C, 5% CO2 with 95% humidity.
Viruses
Virus stocks of SARS-CoV-2 original virus (GISAID accession EPI_ISL_406862) were propagated (four passages) in Vero E6 cells. SARS-CoV-2 variants were provided by the Central Virology Lab of the Israel Ministry of health [3]. Alpha (B.1.1.7, GISAID accession EPI_ISL_4169857) was passaged once in Vero E6, followed by two passages in Calu3 cells. Beta (B.1.351, GISAID accession EPI_ISL_4169885), gamma (P.1, GISAID accession EPI_ISL_4169886) and delta (B.1.617.2, GISAID accession EPI_ISL_4169986) variants were propagated in Vero E6 cells. Omicron (B.1.1.529) was propagated in Calu3 cells. Variants were verified by next-generation sequencing (NGS). Recombinant VSV Indiana serotype (rVSV-WT) was propagated in Vero cells [1]. All virus stocks were tittered on Vero E6 cells as previously described [1]. Handling and working with SARS-CoV-2 virus were conducted in a BSL3 facility in accordance with the biosafety guidelines of the Israel Institute for Biological Research (IIBR).
Plaque reduction neutralization test (PRNT50)
PRNT50 was previously described [1]. Briefly, serial serum samples were incubated with 300 pfu/ml of SARS-CoV-2 original virus, its variants, or with rVSV-WT. Vero E6 cells were infected with the virus-serum mixtures and following incubation (24 h for rVSV-WT, 72 h for SARS-CoV-2 original virus, beta, gamma or delta variant, 96 h for alpha variant, or 120 h for omicron variant) plaques were counted and NT50 was calculated using GraphPad Prism 6 software (GraphPad Software Inc.).
Statistical analysis
Data were analyzed with GraphPad Prism 6 software. Exact p values are provided for each analysis. Statistical significance for SARS-CoV-2 variants neutralization were determined by unpaired t-test. Statistical significance for VSV vector immunity was determined by one-way ANOVA non-parametric test by Kruskal–Wallis test.
Results
BriLife® development was accompanied by spontaneous acquisition of several mutations, some of which are at the spike protein at sites of major importance to antibody-mediated immunity, such as E484, Q493, and N501 located in the receptor binding domain (RBD) (Figure 1a) [1]. An additional mutation is R685G at the RRAR multibasic furin cleavage site, located at the junction of receptor-binding (S1) and fusion (S2) domains of the spike protein [1].
We aimed at assessing the neutralization capacity induced by BriLife®. Therefore, we first tested blinded serum samples from two cohorts of participants in a BriLife® phase II clinical trial two weeks following intramuscular prime-boost vaccination for their ability to neutralize SARS-CoV-2 variants alpha, beta, gamma, delta or omicron. Inclusion criteria were negative SARS-CoV-2 nucleocapsid binding (data not shown) and positive SARS-CoV-2 neutralization response against the original virus.
Nine tested sera (“cohort 1”, Table S1) efficiently neutralized the original virus, as well as gamma variant, with mean titers of 246 and 357, respectively (0.7-fold change, Figure 1d, Table S1). Nearly all tested sera neutralized alpha (8/9 samples) and beta (7/9 samples) variants, with mean titers of 163 and 103, respectively (1.5-fold and 2.4-fold reduction relative to original virus, Figure 1b-c, Table S1).
Several months ago, delta variant of concern (VOC) became dominant worldwide exhibiting increased transmissibility, immune escape [4], and breakthrough infections in vaccinated individuals [5]. We therefore tested another set of 13 vaccinees’ sera (“cohort 2”, Table S2) for neutralization of original virus and delta VOC. Of 13 tested sera which neutralized the original virus, eleven also neutralized the delta variant, with no significant difference in neutralization titers (mean titers of 152 and 131, respectively, Figure 1e, Table S2).
Most recently, at the end of November 2021, omicron variant emerged leading to massive waves of infections worldwide, with increasing evidence of breakthrough infections of vaccinated individuals or previously infected individuals [6]. Thus, we also tested the ability of BriLife® vaccinees’ sera (“cohort 2”) to neutralize live omicron VOC. We show that most tested sera (10/13 samples) maintained neutralization capacity against omicron VOC, showing a ∼2.9-fold reduction in neutralization titers (Figure 1f, Table S2), with only 3 vaccinees’ sera below limit of detection (LOD).
Immunity to surface antigens of viral vectored-based vaccines (e.g. adenovirus-based platforms) might hamper their efficacy upon repeated usage. Replacement of VSV-G surface antigen with the antigen of interest reduces the risk of vector immunity [7]. We show that vector immunity was not significantly induced following BriLife® vaccination (Figure 1g, Table S3), minimizing the risk of reduced vaccine efficacy upon repeated vaccinations.
Discussion
SARS-CoV-2 emerging variants pose a challenge to the battle against COVID-19. Amino acid substitutions in SARS-CoV-2 variants such as delta and omicron were shown to compromise the protection afforded by vaccines, therapeutic antibodies or antibodies derived from previous SARS-CoV-2 infection [8-10]. As described, BriLife® development was accompanied by spontaneous acquisition of mutations, such as E484, Q493, N501 at the RBD and R685G at the furin cleavage site (Figure 1a) [1]. These mutations are similar or identical to some of the key naturally-occurring mutations in SARS-CoV-2 variants. N501Y substitution is found in alpha, beta, gamma and omicron variants (Figure 1a) [11]. E484 is mutated in BriLife® to aspartic acid to form E484D. In beta and gamma, E484 is mutated to Lysine (E484K). In omicron, E484 is mutated to Alanine (E484A). E484K, E484D, E484A and E484G substitutions were shown to escape neutralization by convalescent human sera [11], further consolidating the role of E484 in antibody binding. N501Y as a single mutation does not significantly impair sensitivity to vaccine-induced antibodies [12]. However, combination of E484K, N501Y and K417N or K417T found in beta, omicron, or gamma variants, was shown to significantly affect neutralization capacity in sera of convalescent or vaccinated individuals [13]. In omicron, Q493 is mutated to Lysine (Q493K); mutation was previously shown to enable evasion from antibody-mediated immunity [14]. We suggest that the above-mentioned BriLife® mutations might provide an advantage in combating SARS-CoV-2 variants.
P681 located just before the RRAR multibasic motif in furin cleavage site is replaced by Histidine at alpha and omicron variants (P681H) and by Arginine at delta variant (P681R). This region is of major importance to transmissibility and spreading of SARS-CoV-2 [11]. Whether the spontaneously-acquired R685G mutation of BriLife® have a beneficial effect on vaccine efficacy, specifically against alpha, delta, and omicron, awaits further investigation.
As currently approved vaccines correspond to the original virus sequence [4, 15], there is major concern regarding their ability to protect against SARS-CoV-2 variants. Neutralization titers against variants following two-dose vaccination with BNT162b2, mRNA-1273 or ChAdOx1 nCoV-19 were reported, showing for most only a minor reduction in alpha neutralization, but a strong reduction in beta and gamma neutralization [8], while others report no significant reduction in vaccine efficacy against gamma variant [4, 10, 13]. An overall reduction in neutralization potential has been reported against beta variant ranging between ∼2-30-fold, depending on the report [10, 11, 13, 16]. For delta variant, some report a modest reduction in the effectiveness of two-dose vaccines compared to the alpha variant or to the original virus. An overall reduction ranges between ∼2-11 fold [3, 4, 10, 16].
As for omicron, there are increasing reports on its escape from therapeutic monoclonal antibodies, convalescent patients’ antibodies, or vaccine-induced antibodies. The reduction in omicron neutralization by sera post-immunization was reported more so following two-dose vaccination, but also following three vaccine doses, to a lesser extent [9, 17-19]. Several studies addressing neutralization capacity of VOCs in sera obtained at early time points, ranging between 10 days to 3 months following two dose administration of mRNA or adenoviral vaccines show a significant and profound neutralization reduction compared to the original virus. Carreño et al show a 23.3-fold reduction for BNT162b2 at 14-21 days post 2nd vaccination, and a 42.6-fold reduction for mRNA-1273 at 14-36 days post 2nd vaccination [20]; Cele report a ∼22-fold reduction for BNT162b2 at 10-33 days post 2nd vaccination [21]. An additional study shows a ∼108-fold decrease and 18.9-fold decrease between omicron and the original virus at 28 days following 2nd vaccination with BNT162b2, or ADZ1222, respectively [17]. The time points addressed by these reports span the 14 days-time point tested in the current study.
Taken together, most of the above-mentioned studies show a significant reduction following 2nd dose of vaccination against beta, delta, and omicron, already at early time points, which span the 14 days-time point tested in the current study. Our data show only a mild reduction, of 2.4-and 2.9-fold for beta and omicron, respectively. Thus, we suggest that the maintenance of neutralization capacity against variants at these early time point indicates the potential of BriLife® as an efficacious vaccine against variants. It would be interesting to asses neutralization capacity of BriLife® vaccinees’ sera obtained at later time points in future work.
Significant reduction in neutralization potential of convalescent sera from early pandemic stages against variants was also reported [8, 10, 22]. We also compared neutralization capacity of 9 convalescent sera from early pandemic infections (Figure S1) against the original virus and alpha, beta, gamma or delta variants. We show no significant difference in neutralization titers of the original virus and that of alpha, beta or gamma variants (Supplementary Figure S1a-d, Table S4). However, we do see a significant 3.8-fold reduction in neutralization titers of convalescent sera against delta VOC compared to the original virus (Supplementary Figure S1d, Table S4).
Overall, it appears that beta is the most resistant variant to neutralization either by convalescent sera, currently available vaccines, or BriLife® vaccination [8]. Recent cross-neutralization data from convalescent sera from different pandemic waves shed light on the possible contribution of specific spike mutations to protection from variants [8] and gives rise to a need for updated vaccines. Convalescent sera also show lower, or no neutralization of omicron VOC, depending on the variant of infection and on time post infection [9, 19].
Taken together, our data indicates that BriLife®-induced antibodies maintain neutralizing potential against all tested variants, and most importantly against delta, and the recently emerged omicron VOCs. We suggest that spontaneously-acquired mutations that occurred during BriLife® development and correspond to naturally-occurring mutations of SARS-CoV-2 variants, may increase the potential of BriLife® to maintain effectiveness against current SARS-CoV-2 variants, and potentially against future VOC.
Data Availability
All data produced in the present work are contained in the manuscript
SUPPLEMENTARY MATERIAL
Author Contributions
Conceptualization, Y.Y.R., N.E., T.I., N.P. ; methodology, Y.Y.R., N.E., T.I., N.P. ; formal analysis, Y.Y.R., N.E. ; investigation, Y.Y.R., N.E., M.F., H.T., B.P., H.A., S.M., I.G., A.M., S.W., I.C.G., O.I., A.B.D., A.Z. ; resources, M.M., M.I., D.C., E.S., H.M., N.M.B., Y.C., R.Z. ; writing—original draft preparation, Y.Y.R., T.I., N.P. ; writing—review and editing, Y.Y.R, T.I., N.P. ; supervision, T.I., N.P. ; All authors have read and agreed to the published version of the manuscript.
Funding
This research received no external funding.
Institutional Review Board Statement
BriLife® study - was performed in compliance with International Council for Harmonization (ICH) Good Clinical Practices (GCP), including the archiving of essential documents as well as the ethical principles of the Declaration of Helsinki. Convalescent COVID-19 sera - The study was approved by the ethics committee of the Israeli Ministry of Health (0083-20-WOMC)2.
Informed Consent Statement
Informed consent was obtained from all BriLife® participants involved in the study. All convalescent volunteers gave their informed consent to the National Blood services of Magen David Adom.
Data Availability Statement
Data available on request due to restrictions, e.g., privacy or ethical.
Conflicts of Interest
The authors declare no conflict of interest.
Acknowledgments
We thank the participants in BriLife® clinical trial for their altruism and their dedication to this trial. We thank Prof. Yoseph Caraco from Hadassah Medical Center (Jerusalem), Dr. Eytan Ben-Ami from Sheba Medical Center (Ramat Gan), Dr. Avivit Peer from Rambam Health Care Campus (Haifa), Prof. David Zeltser from Tel Aviv Sourasky Medical Center (Tel Aviv), Prof. Shlomo Maayan from Barzilai Medical Center (Ashkelon), Dr. Victor Vishlitzky from Meir Medical Center (Kefar Sava), Dr. Tal Brosh from Assuta (Ashdod), Dr. Noa Eliakim from Rabin Medical Center (Petah Tiqwa), and the respective Clinical Center teams in Israel participating in this study. We also acknowledge the contributions of the IIBR20-001 Clinical Trial Group at IIBR, Gsap (Haifa, Israel) and IQVIA (Durham NC, USA).
SARS-CoV-2 original virus was kindly provided by Bundeswehr Institute of Microbiology, Munich, Germany. SARS-CoV-2 variants alpha, beta, gamma and delta were kindly provided by Prof. Ella Mendelson, Dr. Ital Nemet and Limor Kliker from the Central Virology Lab of the Israel Ministry of Health. rVSV-WT was kindly provided by Prof. Eran Bacharach from Tel Aviv University.
Footnotes
yfatyr{at}iibr.gov.il (Y.Y.-R.); noame{at}iibr.gov.il (N.E.); morlyf{at}iibr.gov.il (M.F.) hadast{at}iibr.gov.il (H.T.); boazp{at}iibr.gov.il (B.P.); hagita{at}iibr.gov.il (H.A.); sharonm{at}iibr.gov.il (S.M.); itaig{at}iibr.gov.il (I.G.); shayw{at}iibr.gov.il (S.W.); advam{at}iibr.gov.il (A.M.), tomeri{at}iibr.gov.il (T.I.), nirp{at}iibr.gov.il (N.P.).
inbarg{at}iibr.gov.il (I.C.G.), ofiri{at}iibr.gov.il (O.I.) adib{at}iibr.gov.il (A.B.D.), anatz{at}iibr.gov.il (A.Z.)
marinai{at}mdais.co.il (M.I.), eilats{at}mdais.co.il (E.S.)
michal.mandelboim{at}sheba.health.gov.il (M.M.)
CARACO{at}hadassah.org.il (Y.C)
noamb{at}iibr.gov.il
ranz{at}iibr.gov.il (R.Z.); hadarm{at}iibr.gov.il (H.M.)
dancohen{at}tauex.tau.ac.il (D.C.)
Omicron VOC data was added