[HTML][HTML] An HSV-2 nucleoside-modified mRNA genital herpes vaccine containing glycoproteins gC, gD, and gE protects mice against HSV-1 genital lesions and latent …

KP Egan, LM Hook, A Naughton, N Pardi… - PLoS …, 2020 - journals.plos.org
KP Egan, LM Hook, A Naughton, N Pardi, S Awasthi, GH Cohen, D Weissman, HM Friedman
PLoS Pathogens, 2020journals.plos.org
HSV-1 causes 50% of first-time genital herpes infections in resource-rich countries and
affects 190 million people worldwide. A prophylactic herpes vaccine is needed to protect
against genital infections by both HSV-1 and HSV-2. Previously our laboratory developed a
trivalent vaccine that targets glycoproteins C, D, and E present on the HSV-2 virion. We
reported that this vaccine protects animals from genital disease and recurrent virus shedding
following lethal HSV-2 challenge. Importantly the vaccine also generates cross-reactive …
HSV-1 causes 50% of first-time genital herpes infections in resource-rich countries and affects 190 million people worldwide. A prophylactic herpes vaccine is needed to protect against genital infections by both HSV-1 and HSV-2. Previously our laboratory developed a trivalent vaccine that targets glycoproteins C, D, and E present on the HSV-2 virion. We reported that this vaccine protects animals from genital disease and recurrent virus shedding following lethal HSV-2 challenge. Importantly the vaccine also generates cross-reactive antibodies that neutralize HSV-1, suggesting it may provide protection against HSV-1 infection. Here we compared the efficacy of this vaccine delivered as protein or nucleoside-modified mRNA immunogens against vaginal HSV-1 infection in mice. Both the protein and mRNA vaccines protected mice from HSV-1 disease; however, the mRNA vaccine provided better protection as measured by lower vaginal virus titers post-infection. In a second experiment, we compared protection provided by the mRNA vaccine against intravaginal challenge with HSV-1 or HSV-2. Vaccinated mice were totally protected against death, genital disease and infection of dorsal root ganglia caused by both viruses, but somewhat better protected against vaginal titers after HSV-2 infection. Overall, in the two experiments, the mRNA vaccine prevented death and genital disease in 54/54 (100%) mice infected with HSV-1 and 20/20 (100%) with HSV-2, and prevented HSV DNA from reaching the dorsal root ganglia, the site of virus latency, in 29/30 (97%) mice infected with HSV-1 and 10/10 (100%) with HSV-2. We consider the HSV-2 trivalent mRNA vaccine to be a promising candidate for clinical trials for prevention of both HSV-1 and HSV-2 genital herpes.
PLOS