Your browser doesn't support javascript.
loading
A single-administration therapeutic interfering particle reduces SARS-CoV-2 viral shedding and pathogenesis in hamsters
Sonali Chaturvedi; Nathan Beutler; Michael Pablo; Gustavo Vasen; Xinyue Chen; Giuliana Calia; Lauren Buie; Robert Rodick; Davey Smith; Thomas Rogers; Leor Weinberger.
Afiliação
  • Sonali Chaturvedi; The Gladstone Institutes, San Francisco, CA 94158, USA
  • Nathan Beutler; The Scripps Research Institute, La Jolla, CA 92037, USA
  • Michael Pablo; The Gladstone Institutes, San Francisco, CA 94158, USA
  • Gustavo Vasen; The Gladstone Institutes, San Francisco, CA 94158, USA
  • Xinyue Chen; The Gladstone Institutes, San Francisco, CA 94158, USA
  • Giuliana Calia; The Gladstone Institutes, San Francisco, CA 94158, USA
  • Lauren Buie; VxBiosciences Inc., Berkeley, CA 94707, USA
  • Robert Rodick; VxBiosciences Inc., Berkeley, CA 94707, USA
  • Davey Smith; Division of Infectious Diseases, Department of Medicine, University of California, San Diego, CA 92121, USA
  • Thomas Rogers; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
  • Leor Weinberger; The Gladstone Institutes, San Francisco, CA 94158, USA
Preprint em Inglês | bioRxiv | ID: ppbiorxiv-503534
ABSTRACT
The high transmissibility of SARS-CoV-2 is a primary driver of the COVID-19 pandemic. While existing interventions prevent severe disease, they exhibit mixed efficacy in preventing transmission, presumably due to their limited antiviral effects in the respiratory mucosa, whereas interventions targeting the sites of viral replication might more effectively limit respiratory virus transmission. Recently, intranasally administered RNA-based therapeutic interfering particles (TIPs) were reported to suppress SARS-CoV-2 replication, exhibit a high barrier to resistance, and prevent serious disease in hamsters. Since TIPs intrinsically target the tissues with the highest viral replication burden (i.e., respiratory tissues for SARS-CoV-2), we tested the potential of TIP intervention to reduce SARS-CoV-2 shedding. Here, we report that a single, post-exposure TIP dose lowers SARS-CoV-2 nasal shedding and at 5 days post-infection infectious virus shed is below detection limits in 4 out of 5 infected animals. Furthermore, TIPs reduce shedding of Delta variant or WA-1 from infected to uninfected hamsters. Co-housed contact animals exposed to infected, TIP-treated, animals exhibited significantly lower viral loads, reduced inflammatory cytokines, no severe lung pathology, and shortened shedding duration compared to animals co-housed with untreated infected animals. TIPs may represent an effective countermeasure to limit SARS-CoV-2 transmission. SignificanceCOVID-19 vaccines are exceptionally effective in preventing severe disease and death, but they have mixed efficacy in preventing virus transmission, consistent with established literature that parenteral vaccines for other viruses fail to prevent mucosal virus shedding or transmission. Likewise, small-molecule antivirals, while effective in reducing viral-disease pathogenesis, also appear to have inconsistent efficacy in preventing respiratory virus transmission including for SARS-CoV-2. Recently, we reported the discovery of a single-administration antiviral Therapeutic Interfering Particle (TIP) against SARS-CoV-2 that prevents severe disease in hamsters and exhibits a high genetic barrier to the evolution of resistance. Here, we report that TIP intervention also reduces SARS-CoV-2 transmission between hamsters.
Licença
cc_no
Texto completo: Disponível Coleções: Preprints Base de dados: bioRxiv Idioma: Inglês Ano de publicação: 2022 Tipo de documento: Preprint
Texto completo: Disponível Coleções: Preprints Base de dados: bioRxiv Idioma: Inglês Ano de publicação: 2022 Tipo de documento: Preprint
...