#Bamlanivimab does not neutralize two #SARS-CoV-2 #variants carrying #E484K in vitro (MedRxIV, abstract)

[Source: MedRxIV, full page: (LINK). Abstract, edited.]

Bamlanivimab does not neutralize two SARS-CoV-2 variants carrying E484K in vitro

Marek Widera, Alexander Wilhelm, Sebastian Hoehl, Christiane Pallas, Niko Kohmer, Timo Wolf, Holger F Rabenau, Victor M Corman, Christian Drosten, Maria JGT Vehreschild, Udo Goetsch, Rene Gottschalk, Sandra Ciesek

doi: https://doi.org/10.1101/2021.02.24.21252372 | This article is a preprint and has not been certified by peer review. It reports new medical research that has yet to be evaluated and so should not be used to guide clinical practice.

Abstract

The IgG1 monoclonal antibody (mAb) bamlanivimab (LY-CoV555) prevents viral attachment and entry into human cells by blocking attachment to the ACE2 receptor. However, whether bamlanivimab is equally effective against SARS-CoV-2 emerging variants of concern (VOC) is not fully known. Hence, the aim of this study was to determine whether bamlanivimab is equally effective against SARS-CoV-2 emerging VOC. The ability of bamlanivimab to neutralize five SARS-CoV-2 variants including B.1.1.7 (mutations include N501Y and del69/70), B.1.351 (mutations include E484K and N501Y) and P.2 (mutations include E484K in the absence of a N501Y mutation) was analyzed in infectious cell culture using CaCo2 cells. Additionally, we analyzed vaccine-elicited sera after immunization with BNT162b2, and convalescent sera for its ability to neutralize SARS-CoV-2 variants. We found that the variant B.1.1.7, as well as two isolates from early 2020 (FFM1 and FFM7) could be efficiently neutralized by bamlanivimab (titer 1/1280, respectively), however, no neutralization effect could be detected against either B.1.135 or P.2, both harboring the E484K substitution. Vaccine-elicited sera showed slightly decreased neutralizing activity against B1.1.7, B.1.135 and P.2 Our in vitro findings indicate that, in contrast to vaccine-elicited sera, bamlanivimab may not provide efficacy against SARS-CoV-2 variants harboring the E484K substitution. Confirmation of the SARS-CoV-2 variant, including screening for E484K, may be needed before initiating mAb treatment with bamlanivimab to ensure both efficacious and efficient use of the antibody product. Hence, variant-specific mAb agents may be required to treat emerging VOC.

Competing Interest Statement: The authors have declared no competing interest.

Funding Statement: No external funding was received

Author Declarations

I confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.

Yes

The details of the IRB/oversight body that provided approval or exemption for the research described are given below:

Ethik-Kommission des Fachbereiches Medizin der Goethe Universitaet Frankfurt (250719)

All necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived.

Yes

I understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).

Yes

I have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.

Yes

Paper in collection COVID-19 SARS-CoV-2 preprints from medRxiv and bioRxiv

Copyright – The copyright holder for this preprint is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission.

Keywords: SARS-CoV-2; COVID-19; Monoclonal antibodies; E484K; Immune escape.

——

Therapeutic activity of an inhaled potent #SARS-CoV-2 neutralizing #human monoclonal #antibody in hamsters (Cell Rep Med., abstract)

[Source: Cell Reports Medicine, full page: (LINK). Abstract, edited.]

Therapeutic activity of an inhaled potent SARS-CoV-2 neutralizing human monoclonal antibody in hamsters

Michael S. Piepenbrink, Jun-Gyu Park, Fatai S. Oladunni, Ashlesha Deshpande, Madhubanti Basu, Sanghita Sarkar, Andreas Loos, Jennifer Woo, Phillip Lovalenti, Derek Sloan, Chengjin Ye, Kevin Chiem, Christopher W. Bates, Reuben E. Burch, Nathaniel B. Erdmann, Paul A. Goepfert, Vu L. Truong, Mark R. Walter, Luis Martinez-Sobrido, James J. Kobie

Open Access | Published: February 24, 2021 | DOI: https://doi.org/10.1016/j.xcrm.2021.100218

Highlights

  • The 1212C2 human monoclonal antibody potently neutralizes SARS-CoV-2
  • 1212C2 mAb was isolated from an IgM memory B cell of a recovered COVID-19 patient
  • Inhaled 1212C2 mAb is rapidly distributed in the lungs
  • Inhaled 1212C2 mAb treatment reduces viral burden and lung pathology in hamsters

Summary

SARS-CoV-2 infection results in viral burden in the respiratory tract, enabling transmission and leading to substantial lung pathology. The 1212C2 fully human monoclonal antibody was derived from an IgM memory B cell of a COVID-19 patient, has high affinity for the Spike protein Receptor Binding Domain, neutralizes SARS-CoV-2 and exhibits in vivo prophylactic and therapeutic activity in hamsters when delivered intraperitoneally, reducing upper and lower respiratory viral burden and lung pathology. Inhalation of nebulized 1212C2 at levels as low as 0.6mg/kg, corresponding to 0.03mg/kg of lung deposited dose, reduced viral burden below the detection limit, and mitigated lung pathology. The therapeutic efficacy of an exceedingly low-dose of inhaled 1212C2 supports the rationale for local lung delivery for dose-sparing benefits as compared to the conventional parenteral route of administration. These results suggest clinical development of 1212C2 formulated and delivered via inhalation for the treatment of SARS-CoV-2 infection should be considered.

Publication History Accepted: February 17, 2021 – Received in revised form: December 23, 2020 – Received: November 18, 2020

Publication stage In Press Accepted Manuscript

Identification DOI: https://doi.org/10.1016/j.xcrm.2021.100218

Copyright © 2021 The Author(s).

Keywords: SARS-CoV-2; COVID-19; Monoclonal antibodies; Animal models.

——

Robust #SARS-CoV-2 #Infection in #Nasal #Turbinates after #Treatment with Systemic Neutralizing #Antibodies (Cell Host Microbe, abstract)

[Source: Cell Host and Microbe, full page: (LINK). Abstract, edited.]

Robust SARS-CoV-2 Infection in Nasal Turbinates after Treatment with Systemic Neutralizing Antibodies

Dongyan Zhou, Jasper Fuk-Woo Chan, Biao Zhou, Runhong Zhou, Shuang Li, Sisi Shan, Li Liu, Anna Jinxia Zhang, Serena J. Chen, Chris Chung-Sing Chan, Haoran Xu, Vincent Kwok-Man Poon, Shuofeng Yuan, Cun Li, Kenn Ka-Heng Chik, Chris Chun-Yiu Chan, Jianli Cao, Chun-Yin Chan, Ka-Yi Kwan, Zhenglong Du, Thomas Tsz-Kan Lau, Qi Zhang, Jie Zhou, Kelvin Kai-Wang To, Linqi Zhang, David D. Ho, Kwok-Yung Yuen, Zhiwei Chen

Published: February 24, 2021 | DOI: https://doi.org/10.1016/j.chom.2021.02.019

Highlights

  • Systemic HuNAb or vaccine fails full prevention of SARS-CoV-2 infection in nasal turbinate
  • Post-exposure HuNAb suppresses SARS-CoV-2 in lungs but poorly in nasal turbinate
  • Live SARS-CoV-2 persists in nasal turbinate for several days despite systemic HuNAb
  • Robust SARS-CoV-2 infection in nasal turbinate is a mode to evade systemic HuNAb

SUMMARY

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is characterized by a burst in the upper respiratory portal for high transmissibility. To determine human neutralizing antibodies (HuNAbs) for entry protection, we tested three potent HuNAbs (IC50 range, 0.0007-0.35 μg/ml) against live SARS-CoV-2 infection in the golden Syrian hamster model. These HuNAbs inhibit SARS-CoV-2 infection by competing with human angiotensin converting enzyme-2 for binding to the viral receptor binding domain (RBD). Prophylactic intraperitoneal or intranasal injection of individual HuNAb or DNA vaccination significantly reduces infection in the lungs but not in the nasal turbinates of hamsters intranasally challenged with SARS-CoV-2. Although postchallenge HuNAb therapy suppresses viral loads and lung damage, robust infection is observed in nasal turbinates treated within 1-3 days. Our findings demonstrate that systemic HuNAb suppresses SARS-CoV-2 replication and injury in lungs; however, robust viral infection in nasal turbinate may outcompete the antibody with significant implications to subprotection, reinfection and vaccine.

Keywords: SARS-CoV-2 – COVID-19 – Human neutralizing antibody – Receptor binding domain – Nasal turbinate – Lung injury – Phage display – Upper respiratory tract

Publication History Accepted: February 19, 2021 – Received in revised form: February 1, 2021 – Received: December 21, 2020

Publication stage In Press Accepted Manuscript

Identification DOI: https://doi.org/10.1016/j.chom.2021.02.019

Copyright © 2021 Elsevier Inc.

Keywords: SARS-CoV-2; COVID-19; Monoclonal antibodies; Immune escape.

——

No higher #infectivity but #immune #escape of #SARS-CoV-2 #501YV2 #variants (Cell, abstract)

[Source: Cell, full page: (LINK). Abstract, edited.]

No higher infectivity but immune escape of SARS-CoV-2 501Y.V2 variants

Qianqian Li, Jianhui Nie, Jiajing Wu, Li Zhang, Ruxia Ding, Haixin Wang, Yue Zhang, Tao Li, Shuo Liu, Mengyi Zhang, Chenyan Zhao, Huan Liu, Lingling Nie, Haiyang Qin, Meng Wang, Qiong Lu, Xiaoyu Li, Junkai Liu, Haoyu Liang, Yi Shi, Yuelei Shen, Liangzhi Xie, Linqi Zhang, Xiaowang Qu, Wenbo Xu, Weijin Huang, Youchun Wang

Published: February 23, 2021 | DOI: https://doi.org/10.1016/j.cell.2021.02.042

Highlights

  • 501Y.V2 showed no higher infectivity in cells with hACE2 comparing to 614G variant.
  • 501Y.V2 showed increased infectivity in cells with mACE2 compared to 614G variant.
  • 501Y.V2 escaped neutralization by most of neutralizing monoclonal antibodies.
  • 501Y.V2 significantly compromised the inhibitory effects of polyclonal antibodies.

Summary

The 501Y.V2 variants of SARS-CoV-2 containing multiple mutations in Spike are now dominant in South Africa and are rapidly spreading to other countries. Here, experiments with 18 pseudotyped viruses showed that the 501Y.V2 variants do not confer increased infectivity in multiple cell types except for murine ACE2-overexpressing cells, where a substantial increase in infectivity was observed. Notably, the susceptibility of the 501Y.V2 variants to 12 of 17 neutralizing monoclonal antibodies was substantially diminished, and the neutralization ability of the sera from convalescent patients and immunized mice was also reduced for these variants. The neutralization resistance was mainly caused by E484K and N501Y mutations in the receptor-binding domain of Spike. The enhanced infectivity in murine ACE2-overexpressing cells suggests the possibility of spillover of the 501Y.V2 variants to mice. Moreover, the neutralization resistance we detected for the 501Y.V2 variants suggests the potential for compromised efficacy of monoclonal antibodies and vaccines.

Publication History Accepted: February 18, 2021 – Received in revised form: February 11, 2021 – Received: January 28, 2021

Publication stage In Press Accepted Manuscript

Identification DOI: https://doi.org/10.1016/j.cell.2021.02.042

Copyright © 2021 Elsevier Inc.

Keywords: SARS-CoV-2; COVID-19; B1351; E484K; Immune escape; Vaccines; Monoclonal antibodies.

——

Extremely potent #human #monoclonal #antibodies from #COVID19 #convalescent patients (Cell, abstract)

[Source: Cell, full page: (LINK). Abstract, edited.]

Extremely potent human monoclonal antibodies from COVID-19 convalescent patients

Emanuele Andreano, Emanuele Nicastri, Ida Paciello, Piero Pileri, Noemi Manganaro, Giulia Piccini, Alessandro Manenti, Elisa Pantano, Anna Kabanova, Marco Troisi, Fabiola Vacca, Dario Cardamone, Concetta De Santi, Jonathan L. Torres, Gabriel Ozorowski, Linda Benincasa, Hyesun Jang, Cecilia Di Genova, Lorenzo Depau, Jlenia Brunetti, Chiara Agrati, Maria Rosaria Capobianchi, Concetta Castilletti, Arianna Emiliozzi, Massimiliano Fabbiani, Francesca Montagnani, Luisa Bracci, Giuseppe Sautto, Ted M. Ross, Emanuele Montomoli, Nigel Temperton, Andrew B. Ward, Claudia Sala, Giuseppe Ippolito, Rino Rappuoli

Published: February 23, 2021 | DOI: https://doi.org/10.1016/j.cell.2021.02.035

Highlights

  • Human memory B cells encoding extremely potent neutralizing antibodies are rare.
  • Most potent antibodies recognize the tip of the spike receptor binding domain.
  • Selected neutralizing antibody neutralizes SARS-CoV-2 emerging variants.
  • Potent antibody prevents and treats hamster infection without Fc-functions

Summary

Human monoclonal antibodies are safe, preventive and therapeutic tools, that can be rapidly developed to help restore the massive health and economic disruption caused by the coronavirus disease 2019 (COVID-19) pandemic. By single cell sorting 4,277 SARS-CoV-2 spike protein specific memory B cells from 14 COVID-19 survivors, 453 neutralizing antibodies were identified. The most potent neutralizing antibodies recognized the spike protein receptor binding domain, followed in potency by antibodies that recognize the S1 domain, the spike protein trimer and the S2 subunit. Only 1.4% of them neutralized the authentic virus with a potency of 1-10 ng/mL. The most potent monoclonal antibody, engineered to reduce the risk of antibody dependent enhancement and prolong half-life, neutralized the authentic wild type virus and emerging variants containing D614G, E484K and N501Y substitutions. Prophylactic and therapeutic efficacy in the hamster model was observed at 0.25 and 4 mg/kg respectively in absence of Fc-functions.

Publication History Accepted: February 16, 2021 – Received in revised form: January 25, 2021 – Received: November 24, 2020

Publication stage In Press Accepted Manuscript

Identification DOI: https://doi.org/10.1016/j.cell.2021.02.035

Copyright © 2021 Elsevier Inc.

Keywords: SARS-CoV-2; COVID-19; Monoclonal antibodies; Animal models.

——

#Evidence of #escape of #SARS-CoV-2 #variant #B1351 from natural and #vaccine induced #sera (Cell, abstract)

[Source: Cell, full page: (LINK). Abstract, edited.]

Evidence of escape of SARS-CoV-2 variant B.1.351 from natural and vaccine induced sera

Daming Zhou, Wanwisa Dejnirattisai, Piyada Supasa, Chang Liu, Alexander J. Mentzer, Helen M. Ginn, Yuguang Zhao, Helen M.E. Duyvesteyn, Aekkachai Tuekprakhon, Rungtiwa Nutalai, Beibei Wang, Guido C. Paesen, Cesar Lopez-Camacho, Jose Slon-Campos, Bassam Hallis, Naomi Coombes, Kevin Bewley, Sue Charlton, Thomas S. Walter, Donal Skelly, Sheila F. Lumley, Christina Dold, Robert Levin, Tao Dong, Andrew J. Pollard, Julian C. Knight, Derrick Crook, Teresa Lambe, Elizabeth Clutterbuck, Sagida Bibi, Amy Flaxman, Mustapha Bittaye, Sandra Belij-Rammerstorfer, Sarah Gilbert, William James, Miles W. Carroll, Paul Klenerman, Eleanor Barnes, Susanna J. Dunachie, Elizabeth E. Fry, Juthathip Mongkolspaya, Jingshan Ren, David I. Stuart, Gavin R. Screaton

Open Access | Published: February 23, 2021 | DOI: https://doi.org/10.1016/j.cell.2021.02.037

Highlights

  • Reduced B.1.351 neutralisation by mAbs and sera induced by early SARS-C0V-2 isolates
  • Neutralisation titre for B.1.351 reduced 8 to 9-fold for Pfizer and AstraZeneca vaccinees
  • E484K, K417N and N501Y cause widespread escape from monoclonal antibodies
  • NTD deletion in B.1.351 abrogates neutralization by a potent neutralizing human mAb

Summary

The race to produce vaccines against SARS-CoV-2 began when the first sequence was published, and this forms the basis for vaccines currently deployed globally. Independent lineages of SARS-CoV-2 have recently been reported: UK–B.1.1.7, South Africa–B.1.351 and Brazil–P.1. These variants have multiple changes in the immunodominant spike protein which facilitates viral cell entry via the Angiotensin converting enzyme-2 (ACE2) receptor. Mutations in the receptor recognition site on the spike are of great concern for their potential for immune escape. Here we describe a structure-function analysis of B.1.351 using a large cohort of convalescent and vaccinee serum samples. The receptor binding domain mutations provide tighter ACE2 binding and widespread escape from monoclonal antibody neutralization largely driven by E484K although K417N and N501Y act together against some important antibody classes. In a number of cases it would appear that convalescent and some vaccine serum offers limited protection against this variant.

Publication History Accepted: February 17, 2021 – Received in revised form: February 16, 2021 – Received: February 8, 2021

Publication stage In Press Accepted Manuscript

Identification DOI: https://doi.org/10.1016/j.cell.2021.02.037

Copyright © 2021 The Author(s). Published by Elsevier Inc.

Keywords: SARS-CoV-2; COVID-19; Immune escape; B1351; Neutralizing antibodies; Monoclonal antibodies; Vaccines.

——

Complete #map of #SARS-CoV-2 #RBD #mutations that #escape the #monoclonal #antibody #LYCoV555 and its #cocktail with LY-CoV016 (BioRxIV, abstract)

[Source: BioRxIV, full page: (LINK). Abstract, edited.]

Complete map of SARS-CoV-2 RBD mutations that escape the monoclonal antibody LY-CoV555 and its cocktail with LY-CoV016

Tyler N Starr,  Allison J Greaney,  Adam S Dingens,  Jesse D Bloom

doi: https://doi.org/10.1101/2021.02.17.431683 | This article is a preprint and has not been certified by peer review.

Abstract

Monoclonal antibodies and antibody cocktails are a promising therapeutic and prophylaxis for COVID-19. However, ongoing evolution of SARS-CoV-2 can render monoclonal antibodies ineffective. Here we completely map all mutations to the SARS-CoV-2 spike receptor binding domain (RBD) that escape binding by a leading monoclonal antibody, LY-CoV555, and its cocktail combination with LY-CoV016. Individual mutations that escape binding by each antibody are combined in the circulating B.1.351 and P.1 SARS-CoV-2 lineages (E484K escapes LY-CoV555, K417N/T escape LY-CoV016). Additionally, the L452R mutation in the B.1.429 lineage escapes LY-CoV555. Furthermore, we identify single amino acid changes that escape the combined LY-CoV555+LY-CoV016 cocktail. We suggest that future efforts should diversify the epitopes targeted by antibodies and antibody cocktails to make them more resilient to antigenic evolution of SARS-CoV-2.

Competing Interest Statement: The authors have declared no competing interest.

Paper in collection COVID-19 SARS-CoV-2 preprints from medRxiv and bioRxiv

Copyright – The copyright holder for this preprint is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under a CC-BY 4.0 International license.

Keywords: SARS-CoV-2; COVID-19; B1351; P1; B1429; E484K; Immune escape; Monoclonal antibodies.

——

Structural Basis for Accommodation of Emerging #B1351 and #B117 #Variants by Two Potent #SARS-CoV-2 Neutralizing #Antibodies (BioRxIV, abstract)

[Source: BioRxIV, full page: (LINK). Abstract, edited.]

Structural Basis for Accommodation of Emerging B.1.351 and B.1.1.7 Variants by Two Potent SARS-CoV-2 Neutralizing Antibodies

Gabriele Cerutti, Micah Rapp, Yicheng Guo, Fabiana Bahna, Jude Bimela, Eswar R Reddem, Jian Yu, Pengfei Wang, Lihong Liu, Yaoxing Huang, David D Ho, Peter D Kwong, Zizhang Sheng, Lawrence Shapiro

doi: https://doi.org/10.1101/2021.02.21.432168 | This article is a preprint and has not been certified by peer review.

Abstract

Emerging SARS-CoV-2 strains, B.1.1.7 and B.1.351, from the UK and South Africa, respectively show decreased neutralization by monoclonal antibodies and convalescent or vaccinee sera raised against the original wild-type virus, and are thus of clinical concern. However, the neutralization potency of two antibodies, 1-57 and 2-7, which target the receptor-binding domain (RBD) of spike, was unaffected by these emerging strains. Here, we report cryo-EM structures of 1-57 and 2-7 in complex with spike, revealing each of these antibodies to utilize a distinct mechanism to bypass or accommodate RBD mutations. Notably, each antibody represented a response with recognition distinct from those of frequent antibody classes. Moreover, many epitope residues recognized by 1-57 and 2-7 were outside hotspots of evolutionary pressure for both ACE2 binding and neutralizing antibody escape. We suggest the therapeutic use of antibodies like 1-57 and 2-7, which target less prevalent epitopes, could ameliorate issues of monoclonal antibody escape.

Competing Interest Statement: DDH, YH, JY, LL and PW are inventors of a patent describing some of the antibodies reported on here.

Paper in collection COVID-19 SARS-CoV-2 preprints from medRxiv and bioRxiv

Copyright – The copyright holder for this preprint is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission.

Keywords: SARS-CoV-2; COVID-19; B117; B1351; Monoclonal antibodies; Immune escape.

——-

Decreased #neutralization of #SARS-CoV-2 #global #variants by therapeutic anti-spike protein #monoclonal #antibodies (BioRxIV, abstract)

[Source: BioRxIV, full page: (LINK). Abstract, edited.]

Decreased neutralization of SARS-CoV-2 global variants by therapeutic anti-spike protein monoclonal antibodies

Takuya Tada,  Belinda M Dcosta, Hao Zhou, Ada Vaill, Wes Kazmierski, Nathaniel R Landau

doi: https://doi.org/10.1101/2021.02.18.431897 | This article is a preprint and has not been certified by peer review.

Abstract

Monoclonal antibodies against the SARS-CoV-2 spike protein, notably, those developed by Regeneron Pharmaceuticals and Eli Lilly and Company have proven to provide protection against severe COVID-19. The emergence of SARS-CoV-2 variants with heavily mutated spike proteins raises the concern that the therapy could become less effective if any of the mutations disrupt epitopes engaged by the antibodies. In this study, we tested monoclonal antibodies REGN10933 and REGN10987 that are used in combination, for their ability to neutralize SARS-CoV-2 variants B.1.1.7, B.1.351, mink cluster 5 and COH.20G/677H. We report that REGN10987 maintains most of its neutralization activity against viruses with B.1.1.7, B.1.351 and mink cluster 5 spike proteins but that REGN10933 has lost activity against B.1.351 and mink cluster 5. The failure of REGN10933 to neutralize B.1.351 is caused by the K417N and E484K mutations in the receptor binding domain; the failure to neutralize the mink cluster 5 spike protein is caused by the Y453F mutation. The REGN10933 and REGN10987 combination was 9.1-fold less potent on B.1.351 and 16.2-fold less potent on mink cluster 5, raising concerns of reduced efficacy in the treatment of patients infected with variant viruses. The results suggest that there is a need to develop additional monoclonal antibodies that are not affected by the current spike protein mutations.

Competing Interest Statement: The authors have declared no competing interest.

Paper in collection COVID-19 SARS-CoV-2 preprints from medRxiv and bioRxiv

Copyright  – The copyright holder for this preprint is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission.

Keywords: SARS-CoV-2; COVID-19; Monoclonal antibodies; B117; B1351; Escape variants.

—–

Broad and potent #activity against #SARS-like viruses by an engineered human #monoclonal #antibody (Science, abstract)

[Source: Science, full page: (LINK). Abstract, edited.]

Broad and potent activity against SARS-like viruses by an engineered human monoclonal antibody

C. Garrett Rappazzo1,*, Longping V. Tse2,*, Chengzi I. Kaku1, Daniel Wrapp3, Mrunal Sakharkar1, Deli Huang4, Laura M. Deveau1, Thomas J. Yockachonis5, Andrew S. Herbert6,7, Michael B. Battles1, Cecilia M. O’Brien6,7, Michael E. Brown1, James C. Geoghegan1, Jonathan Belk1, Linghang Peng4, Linlin Yang4, Yixuan Hou2, Trevor D. Scobey2, Dennis R. Burton4,8,9,10, David Nemazee4, John M. Dye6, James E. Voss4, Bronwyn M. Gunn5, Jason S. McLellan3, Ralph S. Baric2,11,†, Lisa E. Gralinski2,†, Laura M. Walker1,12,†

1 Adimab, LLC, Lebanon, NH 03766, USA. 2 Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA. 3 Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA. 4 Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA. 5 Paul G. Allen School of Global Animal Health, Washington State University, Pullman, WA 99164, USA. 6 U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA. 7 The Geneva Foundation, Tacoma, WA 98402, USA. 8 IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA. 9 Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA. 10 Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA 02139, USA. 11 Departments of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA. 12 Adagio Therapeutics, Inc., Waltham, MA 02451, USA.

†Corresponding author. Email: rbaric@email.unc.edu (R.S.B.); lgralins@email.unc.edu (L.E.G.); laura.walker@adimab.com (L.M.W.)

* These authors contributed equally to this work.

Science  19 Feb 2021: Vol. 371, Issue 6531, pp. 823-829 | DOI: 10.1126/science.abf4830

Targeting sarbecoviruses

As we continue to battle the COVID-19 pandemic, we must confront the possibility of new pathogenic coronaviruses emerging in humans in the future. With this in mind, Rappazzo et al.isolated antibodies from a survivor of the 2003 severe acute respiratory syndrome coronavirus (SARS-CoV), used yeast display libraries to introduce diversity into these antibodies, and then screened for binding to SARS-CoV-2. One of the affinity-matured progeny strongly neutralized SARS-CoV-2, SARS-CoV, and two SARS-related viruses from bats. In addition, this antibody bound to the receptor-binding domains from a panel of sarbecoviruses, suggesting broader activity, and provided protection against SARS-CoV and SARS-CoV-2 in mouse models.

Science, this issue p. 823

Abstract

The recurrent zoonotic spillover of coronaviruses (CoVs) into the human population underscores the need for broadly active countermeasures. We employed a directed evolution approach to engineer three severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibodies for enhanced neutralization breadth and potency. One of the affinity-matured variants, ADG-2, displays strong binding activity to a large panel of sarbecovirus receptor binding domains and neutralizes representative epidemic sarbecoviruses with high potency. Structural and biochemical studies demonstrate that ADG-2 employs a distinct angle of approach to recognize a highly conserved epitope that overlaps the receptor binding site. In immunocompetent mouse models of SARS and COVID-19, prophylactic administration of ADG-2 provided complete protection against respiratory burden, viral replication in the lungs, and lung pathology. Altogether, ADG-2 represents a promising broad-spectrum therapeutic candidate against clade 1 sarbecoviruses.

Keywords: SARS-CoV-2; COVID-19; Monoclonal antibodies; Animal models.

——