RT Journal Article SR Electronic T1 A live attenuated influenza virus-vectored intranasal COVID-19 vaccine provides rapid, prolonged, and broad protection against SARS-CoV-2 infection JF bioRxiv FD Cold Spring Harbor Laboratory SP 2021.11.13.468472 DO 10.1101/2021.11.13.468472 A1 Junyu Chen A1 Pui Wang A1 Lunzhi Yuan A1 Liang Zhang A1 Limin Zhang A1 Hui Zhao A1 Congjie Chen A1 Yaode Chen A1 Jinle Han A1 Jizong Jia A1 Zhen Lu A1 Junping Hong A1 Liqiang Chen A1 Changfa Fan A1 Zicen Lu A1 Qian Wang A1 Rirong Chen A1 Minping Cai A1 Ruoyao Qi A1 Xijing Wang A1 Jian Ma A1 Min Zhou A1 Huan Yu A1 Chunlan Zhuang A1 Xiaohui Liu A1 Qiangyuan Han A1 Guosong Wang A1 Yingying Su A1 Quan Yuan A1 Tong Cheng A1 Ting Wu A1 Xiangzhong Ye A1 Changgui Li A1 Tianying Zhang A1 Jun Zhang A1 Huachen Zhu A1 Yixin Chen A1 Honglin Chen A1 Ningshao Xia YR 2021 UL http://biorxiv.org/content/early/2021/11/15/2021.11.13.468472.abstract AB Remarkable progress has been made in developing intramuscular vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); however, they are limited with respect to eliciting local immunity in the respiratory tract, which is the primary infection site for SARS-CoV-2. To overcome the limitations of intramuscular vaccines, we constructed a nasal vaccine candidate based on an influenza vector by inserting a gene encoding the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2, named CA4-dNS1-nCoV-RBD (dNS1-RBD). A preclinical study showed that in hamsters challenged 1 day and 7 days after single-dose vaccination or 6 months after booster vaccination, dNS1-RBD largely mitigated lung pathology, with no loss of body weight, caused by either the prototype-like strain or beta variant of SARS-CoV-2. Lasted data showed that the animals could be well protected against beta variant challenge 9 months after vaccination. Notably, the weight loss and lung pathological changes of hamsters could still be significantly reduced when the hamster was vaccinated 24 h after challenge. Moreover, such cellular immunity is relatively unimpaired for the most concerning SARS-CoV-2 variants. The protective immune mechanism of dNS1-RBD could be attributed to the innate immune response in the nasal epithelium, local RBD-specific T cell response in the lung, and RBD-specific IgA and IgG response. Thus, this study demonstrates that the intranasally delivered dNS1-RBD vaccine candidate may offer an important addition to fight against the ongoing COVID-19 pandemic, compensating limitations of current intramuscular vaccines, particularly at the start of an outbreak.Competing Interest StatementThe authors have declared no competing interest.