RT Journal Article SR Electronic T1 An ultra-high affinity synthetic nanobody blocks SARS-CoV-2 infection by locking Spike into an inactive conformation JF bioRxiv FD Cold Spring Harbor Laboratory SP 2020.08.08.238469 DO 10.1101/2020.08.08.238469 A1 Michael Schoof A1 Bryan Faust A1 Reuben A. Saunders A1 Smriti Sangwan A1 Veronica Rezelj A1 Nick Hoppe A1 Morgane Boone A1 Christian Bache Billesbølle A1 Marcell Zimanyi A1 Ishan Deshpande A1 Jiahao Liang A1 Aditya A. Anand A1 Niv Dobzinski A1 Beth Shoshana Zha A1 Benjamin Barsi-Rhyne A1 Vladislav Belyy A1 Andrew W. Barile-Hill A1 Sayan Gupta A1 Camille R. Simoneau A1 Kristoffer Leon A1 Kris M. White A1 Silke Nock A1 Yuwei Liu A1 Nevan J. Krogan A1 Corie Y. Ralston A1 Danielle L. Swaney A1 Adolfo García-Sastre A1 Melanie Ott A1 Marco Vignuzzi A1 QCRG Structural Biology Consortium A1 Peter Walter A1 Aashish Manglik YR 2020 UL http://biorxiv.org/content/early/2020/08/10/2020.08.08.238469.abstract AB Without an effective prophylactic solution, infections from SARS-CoV-2 continue to rise worldwide with devastating health and economic costs. SARS-CoV-2 gains entry into host cells via an interaction between its Spike protein and the host cell receptor angiotensin converting enzyme 2 (ACE2). Disruption of this interaction confers potent neutralization of viral entry, providing an avenue for vaccine design and for therapeutic antibodies. Here, we develop single-domain antibodies (nanobodies) that potently disrupt the interaction between the SARS-CoV-2 Spike and ACE2. By screening a yeast surface-displayed library of synthetic nanobody sequences, we identified a panel of nanobodies that bind to multiple epitopes on Spike and block ACE2 interaction via two distinct mechanisms. Cryogenic electron microscopy (cryo-EM) revealed that one exceptionally stable nanobody, Nb6, binds Spike in a fully inactive conformation with its receptor binding domains (RBDs) locked into their inaccessible down-state, incapable of binding ACE2. Affinity maturation and structure-guided design of multivalency yielded a trivalent nanobody, mNb6-tri, with femtomolar affinity for SARS-CoV-2 Spike and picomolar neutralization of SARS-CoV-2 infection. mNb6-tri retains stability and function after aerosolization, lyophilization, and heat treatment. These properties may enable aerosol-mediated delivery of this potent neutralizer directly to the airway epithelia, promising to yield a widely deployable, patient-friendly prophylactic and/or early infection therapeutic agent to stem the worst pandemic in a century.Competing Interest StatementM.Schoof, B.Faust, R.Saunders, N.Hoppe, P.Walter, and A.Manglik are inventors on a provisional patent describing anti-Spike nanobodies described in this manuscript.