International Journal of Nanomedicine (Jul 2024)

An Immunoreceptor-Targeting Strategy with Minimalistic C3b Peptide Fusion Enhances SARS-CoV-2 RBD mRNA Vaccine Immunogenicity

  • Chiu CT,
  • Tsai HH,
  • Chen JY,
  • Hu CMJ,
  • Chen HW

Journal volume & issue
Vol. Volume 19
pp. 7201 – 7214

Abstract

Read online

Chun-Ta Chiu,1 Hsiao-Han Tsai,2 Jing-Yuan Chen,1,3 Che-Ming Jack Hu,2 Hui-Wen Chen1,3 1Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan; 2Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan; 3Animal Resource Center, National Taiwan University, Taipei, 10617, TaiwanCorrespondence: Hui-Wen Chen, Department of Veterinary Medicine, National Taiwan University, 1 Sec 4 Roosevelt Road, Taipei, 10617, Taiwan, Tel +886 2 33669450, Email [email protected]: The clinical success of mRNA vaccine during the COVID-19 pandemic has inspired emerging approaches to elevate mRNA vaccine immunogenicity. Among them, antigen fusion protein designs for improved immune cell targeting have been shown to augment humoral immunity against small antigen targets.Methods: This research demonstrates that SARS-CoV-2 receptor binding domain (RBD) fusion with a minimalistic peptide segment of complement component 3b (C3b, residues 727– 767) ligand can improve mRNA vaccine immunogenicity through antigen targeting to complement receptor 1 (CR1). We affirm vaccines’ antigenicity and targeting ability towards specific receptors through Western blot and immunofluorescence assay. Furthermore, mice immunization studies help the investigation of the antibody responses.Results: Using SARS-CoV-2 Omicron RBD antigen, we compare mRNA vaccine formulations expressing RBD fusion protein with mouse C3b peptide (RBD-mC3), RBD fusion protein with mouse Fc (RBD-Fc), and wild-type RBD. Our results confirm the proper antigenicity and normal functionality of RBD-mC3. Upon validating comparable antigen expression by the different vaccine formulations, receptor-targeting capability of the fusion antigens is further confirmed. In mouse immunization studies, we show that while both RBD-mC3 and RBD-Fc elevate vaccine immunogenicity, RBD-mC3 leads to more sustained RBD-specific titers over the RBD-Fc design, presumably due to reduced antigenic diversion by the minimalistic targeting ligand.Conclusion: The study demonstrates a novel C3b-based antigen design strategy for immune cell targeting and mRNA vaccine enhancement. Keywords: vaccinology, nanotechnology, complement component 3b, follicular dendritic cells, complement receptor 1, immune cell targeting

Keywords