Immunogenicity of influenza virus vaccine is increased by anti-gal-mediated targeting to antigen-presenting cells

UM Abdel-Motal, HM Guay, K Wigglesworth… - Journal of …, 2007 - Am Soc Microbiol
UM Abdel-Motal, HM Guay, K Wigglesworth, RM Welsh, U Galili
Journal of virology, 2007Am Soc Microbiol
This study describes a method for increasing the immunogenicity of influenza virus vaccines
by exploiting the natural anti-Gal antibody to effectively target vaccines to antigen-presenting
cells (APC). This method is based on enzymatic engineering of carbohydrate chains on virus
envelope hemagglutinin to carry the α-Gal epitope (Galα1-3Galβ1-4GlcNAc-R). This epitope
interacts with anti-Gal, the most abundant antibody in humans (1% of immunoglobulins).
Influenza virus vaccine expressing α-Gal epitopes is opsonized in situ by anti-Gal …
Abstract
This study describes a method for increasing the immunogenicity of influenza virus vaccines by exploiting the natural anti-Gal antibody to effectively target vaccines to antigen-presenting cells (APC). This method is based on enzymatic engineering of carbohydrate chains on virus envelope hemagglutinin to carry the α-Gal epitope (Galα1-3Galβ1-4GlcNAc-R). This epitope interacts with anti-Gal, the most abundant antibody in humans (1% of immunoglobulins). Influenza virus vaccine expressing α-Gal epitopes is opsonized in situ by anti-Gal immunoglobulin G. The Fc portion of opsonizing anti-Gal interacts with Fcγ receptors on APC and induces effective uptake of the vaccine virus by APC. APC internalizes the opsonized virus to transport it to draining lymph nodes for stimulation of influenza virus-specific T cells, thereby eliciting a protective immune response. The efficacy of such an influenza vaccine was demonstrated in α1,3galactosyltransferase (α1,3GT) knockout mice, which produce anti-Gal, using the influenza virus strain A/Puerto Rico/8/34-H1N1 (PR8). Synthesis of α-Gal epitopes on carbohydrate chains of PR8 virus (PR8αgal) was catalyzed by recombinant α1,3GT, the glycosylation enzyme that synthesizes α-Gal epitopes in cells of nonprimate mammals. Mice immunized with PR8αgal displayed much higher numbers of PR8-specific CD8+ and CD4+ T cells (determined by intracellular cytokine staining and enzyme-linked immunospot assay) and produced anti-PR8 antibodies with much higher titers than mice immunized with PR8 lacking α-Gal epitopes. Mice immunized with PR8αgal also displayed a much higher level of protection than PR8 immunized mice after being challenged with lethal doses of live PR8 virus. We suggest that a similar method for increasing immunogenicity may be applicable to avian influenza vaccines.
American Society for Microbiology