SORA

Advancing, promoting and sharing knowledge of health through excellence in teaching, clinical practice and research into the prevention and treatment of illness

A Dual-Approach Strategy to Optimize the Safety and Efficacy of Anti-Zika Virus Monoclonal Antibody Therapeutics.

Sun, H; Yang, M; Lai, H; Neupane, B; Teh, AY-H; Jugler, C; Ma, JK-C; Steinkellner, H; Bai, F; Chen, Q (2023) A Dual-Approach Strategy to Optimize the Safety and Efficacy of Anti-Zika Virus Monoclonal Antibody Therapeutics. Viruses, 15 (5). p. 1156. ISSN 1999-4915 https://doi.org/10.3390/v15051156
SGUL Authors: Teh, Yi Hui

[img]
Preview
PDF Published Version
Available under License Creative Commons Attribution.

Download (3MB) | Preview
[img] Archive (ZIP) (Supplementary material) Published Version
Available under License Creative Commons Attribution.

Download (170kB)

Abstract

Antibody-dependent enhancement of infection (ADE) is clinically relevant to Dengue virus (DENV) infection and poses a major risk to the application of monoclonal antibody (mAb)-based therapeutics against related flaviviruses such as the Zika virus (ZIKV). Here, we tested a two-tier approach for selecting non-cross-reactive mAbs combined with modulating Fc glycosylation as a strategy to doubly secure the elimination of ADE while preserving Fc effector functions. To this end, we selected a ZIKV-specific mAb (ZV54) and generated three ZV54 variants using Chinese hamster ovary cells and wild-type (WT) and glycoengineered ΔXF Nicotiana benthamiana plants as production hosts (ZV54CHO, ZV54WT, and ZV54ΔXF). The three ZV54 variants shared an identical polypeptide backbone, but each exhibited a distinct Fc N-glycosylation profile. All three ZV54 variants showed similar neutralization potency against ZIKV but no ADE activity for DENV infection, validating the importance of selecting the virus/serotype-specific mAbs for avoiding ADE by related flaviviruses. For ZIKV infection, however, ZV54CHO and ZV54ΔXF showed significant ADE activity while ZV54WT completely forwent ADE, suggesting that Fc glycan modulation may yield mAb glycoforms that abrogate ADE even for homologous viruses. In contrast to the current strategies for Fc mutations that abrogate all effector functions along with ADE, our approach allowed the preservation of effector functions as all ZV54 glycovariants retained antibody-dependent cellular cytotoxicity (ADCC) against the ZIKV-infected cells. Furthermore, the ADE-free ZV54WT demonstrated in vivo efficacy in a ZIKV-infection mouse model. Collectively, our study provides further support for the hypothesis that antibody-viral surface antigen and Fc-mediated host cell interactions are both prerequisites for ADE, and that a dual-approach strategy, as shown herein, contributes to the development of highly safe and efficacious anti-ZIKV mAb therapeutics. Our findings may be impactful to other ADE-prone viruses, including SARS-CoV-2.

Item Type: Article
Additional Information: © 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Keywords: Fc effector function, Zika virus, antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent enhancement of infection (ADE), glycosylation, monoclonal antibody (mAb), plant-made antibody, plant-made pharmaceutical, Animals, Mice, Cricetinae, Zika Virus, Zika Virus Infection, CHO Cells, Dengue Virus, Dengue, COVID-19, Cricetulus, SARS-CoV-2, Antibodies, Viral, Flavivirus, Antibodies, Monoclonal, Cross Reactions, Antibodies, Neutralizing, CHO Cells, Animals, Cricetulus, Mice, Flavivirus, Dengue Virus, Dengue, Antibodies, Monoclonal, Antibodies, Viral, Cross Reactions, Cricetinae, Antibodies, Neutralizing, Zika Virus, Zika Virus Infection, COVID-19, SARS-CoV-2, Zika virus, monoclonal antibody (mAb), plant-made antibody, antibody-dependent enhancement of infection (ADE), antibody-dependent cellular cytotoxicity (ADCC), glycosylation, Fc effector function, plant-made pharmaceutical, 0605 Microbiology
SGUL Research Institute / Research Centre: Academic Structure > Infection and Immunity Research Institute (INII)
Journal or Publication Title: Viruses
ISSN: 1999-4915
Language: eng
Dates:
DateEvent
11 May 2023Published
8 May 2023Accepted
Publisher License: Creative Commons: Attribution 4.0
Projects:
Project IDFunderFunder ID
R33AI101329National Institute of Allergy and Infectious Diseaseshttp://dx.doi.org/10.13039/100000060
R15AI135893National Institute of Allergy and Infectious Diseaseshttp://dx.doi.org/10.13039/100000060
I 4328-BAustrian Science Fundhttp://dx.doi.org/10.13039/501100002428
I 3721-B30Austrian Science Fundhttp://dx.doi.org/10.13039/501100002428
PubMed ID: 37243242
Web of Science ID: WOS:000996744500001
Go to PubMed abstract
URI: https://openaccess.sgul.ac.uk/id/eprint/115529
Publisher's version: https://doi.org/10.3390/v15051156

Actions (login required)

Edit Item Edit Item