Abstract

The outbreak of the SARS-CoV-2 virus and its rapid spread into a global pandemic made the urgent development of scalable vaccines to prevent coronavirus disease (COVID-19) a global health and economic imperative. Here, we characterized and compared the immunogenicity of two alphavirus-based DNA-launched self-replicating (DREP) vaccine candidates encoding either SARS-CoV-2 spike glycoprotein (DREP-S) or a spike ectodomain trimer stabilized in prefusion conformation (DREP-Secto). We observed that the two DREP constructs were immunogenic in mice inducing both binding and neutralizing antibodies as well as T cell responses. Interestingly, the DREP coding for the unmodified spike turned out to be more potent vaccine candidate, eliciting high titers of SARS-CoV-2 specific IgG antibodies that were able to efficiently neutralize pseudotyped virus after a single immunization. In addition, both DREP constructs were able to efficiently prime responses that could be boosted with a heterologous spike protein immunization. These data provide important novel insights into SARS-CoV-2 vaccine design using a rapid response DNA vaccine platform. Moreover, they encourage the use of mixed vaccine modalities as a strategy to combat SARS-CoV-2.

Highlights

  • The outbreak of the SARS-CoV-2 virus and its rapid spread into a global pandemic made the urgent development of scalable vaccines to prevent coronavirus disease (COVID-19) a global health and economic imperative

  • The full-length proteins, similar in size to recombinant purified spike, were detected in cell lysates for both constructs and in culture supernatant for the DNA-launched self-replicating (DREP)-Secto construct encoding the spike ectodomain, while no protein product of this size was detected in cells transfected with the control DREP expressing chikungunya virus g­ lycoproteins[23,28] (Fig. 1B)

  • In order to determine how the DREP vaccine candidates affected T helper 1 cell (Th1) and T helper 2 cell (Th2) induction, we evaluated the ratio of IgG2c and IgG1 antibody titers against SARS-CoV-2 spike antigen (Fig. 3)

Read more

Summary

Introduction

The outbreak of the SARS-CoV-2 virus and its rapid spread into a global pandemic made the urgent development of scalable vaccines to prevent coronavirus disease (COVID-19) a global health and economic imperative. The DREP coding for the unmodified spike turned out to be more potent vaccine candidate, eliciting high titers of SARS-CoV-2 specific IgG antibodies that were able to efficiently neutralize pseudotyped virus after a single immunization Both DREP constructs were able to efficiently prime responses that could be boosted with a heterologous spike protein immunization. Replicon vectors have inherent adjuvant properties due to their self-amplifying activity which generates single-stranded and double-stranded RNA intermediates that stimulate different pattern recognition receptors (PRR) including the endosomal TLR3, TLR7, and ­TLR88,9 or the cytoplasmic MDA-510, RIG-I10,11 and P­ KR12,13 This results in induction of a type I interferon (IFN) response, translational shutoff of a host cell and induction of apoptosis thereby promoting antigen presentation on MHC class I molecules and cross-priming[11,14,15,16]. DREP platform combines the safety profile of conventional plasmid DNA vaccines that proved safe in multiple clinical

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.