Your browser doesn't support javascript.
An intranasal influenza virus-vectored vaccine prevents SARS-CoV-2 replication in respiratory tissues of mice and hamsters.
Deng, Shaofeng; Liu, Ying; Tam, Rachel Chun-Yee; Chen, Pin; Zhang, Anna Jinxia; Mok, Bobo Wing-Yee; Long, Teng; Kukic, Anja; Zhou, Runhong; Xu, Haoran; Song, Wenjun; Chan, Jasper Fuk-Woo; To, Kelvin Kai-Wang; Chen, Zhiwei; Yuen, Kwok-Yung; Wang, Pui; Chen, Honglin.
  • Deng S; Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • Liu Y; State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • Tam RC; Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • Chen P; State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • Zhang AJ; Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • Mok BW; State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • Long T; Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • Kukic A; State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • Zhou R; Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • Xu H; State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • Song W; Centre for Virology, Vaccinology and Therapeutics Limited, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • Chan JF; Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • To KK; State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • Chen Z; Centre for Virology, Vaccinology and Therapeutics Limited, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • Yuen KY; Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • Wang P; State Key Laboratory for Emerging Infectious Diseases, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
  • Chen H; Centre for Virology, Vaccinology and Therapeutics Limited, the University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
Nat Commun ; 14(1): 2081, 2023 04 12.
Article in English | MEDLINE | ID: covidwho-2294153
ABSTRACT
Current available vaccines for COVID-19 are effective in reducing severe diseases and deaths caused by SARS-CoV-2 infection but less optimal in preventing infection. Next-generation vaccines which are able to induce mucosal immunity in the upper respiratory to prevent or reduce infections caused by highly transmissible variants of SARS-CoV-2 are urgently needed. We have developed an intranasal vaccine candidate based on a live attenuated influenza virus (LAIV) with a deleted NS1 gene that encodes cell surface expression of the receptor-binding-domain (RBD) of the SARS-CoV-2 spike protein, designated DelNS1-RBD4N-DAF. Immune responses and protection against virus challenge following intranasal administration of DelNS1-RBD4N-DAF vaccines were analyzed in mice and compared with intramuscular injection of the BioNTech BNT162b2 mRNA vaccine in hamsters. DelNS1-RBD4N-DAF LAIVs induced high levels of neutralizing antibodies against various SARS-CoV-2 variants in mice and hamsters and stimulated robust T cell responses in mice. Notably, vaccination with DelNS1-RBD4N-DAF LAIVs, but not BNT162b2 mRNA, prevented replication of SARS-CoV-2 variants, including Delta and Omicron BA.2, in the respiratory tissues of animals. The DelNS1-RBD4N-DAF LAIV system warrants further evaluation in humans for the control of SARS-CoV-2 transmission and, more significantly, for creating dual function vaccines against both influenza and COVID-19 for use in annual vaccination strategies.
Subject(s)

Full text: Available Collection: International databases Database: MEDLINE Main subject: Orthomyxoviridae / Influenza Vaccines / COVID-19 Type of study: Experimental Studies Topics: Vaccines / Variants Limits: Animals / Humans Language: English Journal: Nat Commun Journal subject: Biology / Science Year: 2023 Document Type: Article

Similar

MEDLINE

...
LILACS

LIS


Full text: Available Collection: International databases Database: MEDLINE Main subject: Orthomyxoviridae / Influenza Vaccines / COVID-19 Type of study: Experimental Studies Topics: Vaccines / Variants Limits: Animals / Humans Language: English Journal: Nat Commun Journal subject: Biology / Science Year: 2023 Document Type: Article