scispace - formally typeset
Search or ask a question
Author

Wei-Hung Chen

Bio: Wei-Hung Chen is an academic researcher from Walter Reed Army Institute of Research. The author has contributed to research in topics: Neutralizing antibody & Medicine. The author has an hindex of 5, co-authored 11 publications receiving 89 citations. Previous affiliations of Wei-Hung Chen include Henry M. Jackson Foundation for the Advancement of Military Medicine.

Papers
More filters
Journal ArticleDOI
28 Oct 2021
TL;DR: In this paper, the authors evaluated whether a SARS-CoV-2 spike ferritin nanoparticle vaccine (SpFN), adjuvanted with the Army Liposomal Formulation QS21 (ALFQ), conferred protection against the Alpha (B.1.7), and Beta (B1.351) VOCs in Syrian golden hamsters.
Abstract: The emergence of SARS-CoV-2 variants of concern (VOC) requires adequate coverage of vaccine protection. We evaluated whether a SARS-CoV-2 spike ferritin nanoparticle vaccine (SpFN), adjuvanted with the Army Liposomal Formulation QS21 (ALFQ), conferred protection against the Alpha (B.1.1.7), and Beta (B.1.351) VOCs in Syrian golden hamsters. SpFN-ALFQ was administered as either single or double-vaccination (0 and 4 week) regimens, using a high (10 μg) or low (0.2 μg) dose. Animals were intranasally challenged at week 11. Binding antibody responses were comparable between high- and low-dose groups. Neutralizing antibody titers were equivalent against WA1, B.1.1.7, and B.1.351 variants following two high dose vaccinations. Dose-dependent SpFN-ALFQ vaccination protected against SARS-CoV-2-induced disease and viral replication following intranasal B.1.1.7 or B.1.351 challenge, as evidenced by reduced weight loss, lung pathology, and lung and nasal turbinate viral burden. These data support the development of SpFN-ALFQ as a broadly protective, next-generation SARS-CoV-2 vaccine.

42 citations

Posted ContentDOI
17 Mar 2020-bioRxiv
TL;DR: It is reported that this first-ever resolution of a human antibody in complex with SARS-CoV-2 and the broad reactivity of this set of antibodies to a conserved betacoronavirus epitope will allow antigenic assessment of vaccine candidates, and provide a framework for accelerated vaccine, immunotherapeutic and diagnostic strategies against Sars-Cov-2.
Abstract: SARS-CoV-2 is a zoonotic virus that has caused a pandemic of severe respiratory disease-COVID-19-within several months of its initial identification. Comparable to the first SARS-CoV, this novel coronavirus's surface Spike (S) glycoprotein mediates cell entry via the human ACE-2 receptor, and, thus, is the principal target for the development of vaccines and immunotherapeutics. Molecular information on the SARS-CoV-2 S glycoprotein remains limited. Here we report the crystal structure of the SARS-CoV-2 S receptor-binding-domain (RBD) at a the highest resolution to date, of 1.95 A. We identified a set of SARS-reactive monoclonal antibodies with cross-reactivity to SARS-CoV-2 RBD and other betacoronavirus S glycoproteins. One of these antibodies, CR3022, was previously shown to synergize with antibodies that target the ACE-2 binding site on the SARS-CoV RBD and reduce viral escape capacity. We determined the structure of CR3022, in complex with the SARS-CoV-2 RBD, and defined a broadly reactive epitope that is highly conserved across betacoronaviruses. This epitope is inaccessible in the "closed" prefusion S structure, but is accessible in "open" conformations. This first-ever resolution of a human antibody in complex with SARS-CoV-2 and the broad reactivity of this set of antibodies to a conserved betacoronavirus epitope will allow antigenic assessment of vaccine candidates, and provide a framework for accelerated vaccine, immunotherapeutic and diagnostic strategies against SARS-CoV-2 and related betacoronaviruses.

40 citations

Journal ArticleDOI
Hannah A.D. King1, Hannah A.D. King2, M. Gordon Joyce2, M. Gordon Joyce1, Ines Lakhal-Naouar2, Ines Lakhal-Naouar1, Aslaa Ahmed1, Camila Macedo Cincotta2, Camila Macedo Cincotta1, Caroline Subra2, Caroline Subra1, Kristina K. Peachman1, Holly R. Hack2, Holly R. Hack1, Rita E. Chen3, Paul V. Thomas1, Paul V. Thomas2, Wei-Hung Chen1, Wei-Hung Chen2, Rajeshwer S. Sankhala2, Rajeshwer S. Sankhala1, Agnes Hajduczki2, Agnes Hajduczki1, Elizabeth J. Martinez1, Elizabeth J. Martinez2, Caroline E. Peterson2, Caroline E. Peterson1, William C. Chang1, William C. Chang2, Misook Choe2, Misook Choe1, Clayton A. Smith, Jarrett A. Headley1, Jarrett A. Headley2, Hanne A. Elyard, Anthony L. Cook, Alexander R. A. Anderson1, Alexander R. A. Anderson2, Kathryn McGuckin Wuertz1, Ming Dong1, Ming Dong2, Isabella Swafford1, Isabella Swafford2, James Brett Case4, Jeffrey R. Currier1, Kerri G. Lal2, Kerri G. Lal1, Mihret F. Amare1, Mihret F. Amare2, Vincent Dussupt1, Vincent Dussupt2, Sebastian Molnar2, Sebastian Molnar1, Sharon P. Daye1, Xiankun Zeng5, Erica K. Barkei1, Kendra J. Alfson6, Hilary M. Staples6, Ricardo Carrion6, Shelly J. Krebs1, Shelly J. Krebs2, Dominic Paquin-Proulx2, Dominic Paquin-Proulx1, Nicos Karasavvas1, Nicos Karasavvas2, Victoria R. Polonis1, Linda L. Jagodzinski1, Sandhya Vasan1, Sandhya Vasan2, Paul T. Scott1, Yaoxing Huang7, Manoj S. Nair7, David D. Ho7, Natalia de Val, Michael S. Diamond3, Mark G. Lewis, Mangala Rao1, Gary R. Matyas1, Gregory D. Gromowski1, Sheila A. Peel1, Nelson L. Michael1, Kayvon Modjarrad1, Diane L. Bolton1, Diane L. Bolton2 
TL;DR: In this article, a ferritin nanoparticle vaccine displaying the receptor-binding domain of the SARS-CoV-2 spike protein (RFN) adjuvanted with Army Liposomal Formulation QS-21 (ALFQ) was evaluated.
Abstract: Emergence of novel variants of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) underscores the need for next-generation vaccines able to elicit broad and durable immunity. Here we report the evaluation of a ferritin nanoparticle vaccine displaying the receptor-binding domain of the SARS-CoV-2 spike protein (RFN) adjuvanted with Army Liposomal Formulation QS-21 (ALFQ). RFN vaccination of macaques using a two-dose regimen resulted in robust, predominantly Th1 CD4+ T cell responses and reciprocal peak mean serum neutralizing antibody titers of 14,000 to 21,000. Rapid control of viral replication was achieved in the upper and lower airways of animals after high-dose SARS-CoV-2 respiratory challenge, with undetectable replication within 4 d in seven of eight animals receiving 50 µg of RFN. Cross-neutralization activity against SARS-CoV-2 variant B.1.351 decreased only approximately twofold relative to WA1/2020. In addition, neutralizing, effector antibody and cellular responses targeted the heterotypic SARS-CoV-1, highlighting the broad immunogenicity of RFN-ALFQ for SARS-CoV-like Sarbecovirus vaccine development.

36 citations

Journal ArticleDOI
TL;DR: Krebs et al. as mentioned in this paper identified several potent neutralizing antibodies directed against either the N-terminal domain (NTD) or the receptor-binding domain (RBD) of the spike protein.
Abstract: Prevention of viral escape and increased coverage against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern require therapeutic monoclonal antibodies (mAbs) targeting multiple sites of vulnerability on the coronavirus spike glycoprotein. Here we identify several potent neutralizing antibodies directed against either the N-terminal domain (NTD) or the receptor-binding domain (RBD) of the spike protein. Administered in combinations, these mAbs provided low-dose protection against SARS-CoV-2 infection in the K18-human angiotensin-converting enzyme 2 mouse model, using both neutralization and Fc effector antibody functions. The RBD mAb WRAIR-2125, which targets residue F486 through a unique heavy-chain and light-chain pairing, demonstrated potent neutralizing activity against all major SARS-CoV-2 variants of concern. In combination with NTD and other RBD mAbs, WRAIR-2125 also prevented viral escape. These data demonstrate that NTD/RBD mAb combinations confer potent protection, likely leveraging complementary mechanisms of viral inactivation and clearance. Krebs and colleagues identify multiple mAbs that recognize either the RBD or the NTD of SARS-CoV-2 spike protein that have potent cross-neutralizing activities against variants of concern. Combinatorial mAb cocktails have complementary effects on viral neutralization and Fc effector functions and can protect against SARS-CoV-2 escape mutants.

32 citations

Posted ContentDOI
Michael Gordon Joyce1, Michael Gordon Joyce2, Hannah A.D. King1, Hannah A.D. King2, Ines Elakhal Naouar1, Aslaa Ahmed, Kristina K. Peachman1, Camila Macedo Cincotta1, Caroline Subra1, Rita E. Chen3, Paul V. Thomas2, Paul V. Thomas1, Wei-Hung Chen2, Wei-Hung Chen1, Rajeshwer S. Sankhala1, Rajeshwer S. Sankhala2, Agnes Hajduczki2, Agnes Hajduczki1, Elizabeth J. Martinez2, Elizabeth J. Martinez1, Caroline E. Peterson2, Caroline E. Peterson1, William C. Chang1, William C. Chang2, Misook Choe1, Misook Choe2, Clayton Smith4, Parker J. Lee2, Parker J. Lee1, Jarrett A. Headley2, Jarrett A. Headley1, Mekdi G. Taddese1, Mekdi G. Taddese2, Hanne A. Elyard, Anthony L. Cook, Alexander R. A. Anderson1, Alexander R. A. Anderson2, Kathryn McGuckin-Wuertz, Ming Dong1, Ming Dong2, Isabella Swafford1, Isabella Swafford2, James B. Case3, Jeffrey R. Currier, Kerri G. Lal1, Kerri G. Lal2, Sebastian Molnar2, Sebastian Molnar1, Manoj S. Nair5, Vincent Dussupt2, Vincent Dussupt1, Sharon P. Daye, Xiankun Zeng6, Erica K. Barkei, Hilary M. Staples7, Kendra J. Alfson7, Ricardo Carrion7, Shelly J. Krebs2, Shelly J. Krebs1, Dominic Paquin-Proulx2, Dominic Paquin-Proulx1, Nicos Karasavva1, Victoria R. Polonis, Linda L. Jagodzinski, Mihret F. Amare2, Mihret F. Amare1, Sandhya Vasan2, Sandhya Vasan1, Paul T. Scott2, Yaoxing Huang5, David D. Ho5, Natalia de Val4, Michael S. Diamond3, Mark G. Lewis, Mangala Rao, Gary R. Matyas, Gregory D. Gromowski, Sheila A. Peel, Nelson L. Michael, Diane L. Bolton2, Diane L. Bolton1, Kayvon Modjarrad2 
25 Mar 2021-bioRxiv
TL;DR: In this paper, a Spike protein ferritin nanoparticle (SpFN) vaccine was developed and evaluated for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants in nonhuman primates.
Abstract: The emergence of novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants stresses the continued need for next-generation vaccines that confer broad protection against coronavirus disease 2019 (COVID-19). We developed and evaluated an adjuvanted SARS-CoV-2 Spike Ferritin Nanoparticle (SpFN) vaccine in nonhuman primates (NHPs). High-dose (50 {micro}g) SpFN vaccine, given twice within a 28 day interval, induced a Th1-biased CD4 T cell helper response and a peak neutralizing antibody geometric mean titer of 52,773 against wild-type virus, with activity against SARS-CoV-1 and minimal decrement against variants of concern. Vaccinated animals mounted an anamnestic response upon high-dose SARS-CoV-2 respiratory challenge that translated into rapid elimination of replicating virus in their upper and lower airways and lung parenchyma. SpFNs potent and broad immunogenicity profile and resulting efficacy in NHPs supports its utility as a candidate platform for SARS-like betacoronaviruses. One-Sentence SummaryA SARS-CoV-2 Spike protein ferritin nanoparticle vaccine, co-formulated with a liposomal adjuvant, elicits broad neutralizing antibody responses that exceed those observed for other major vaccines and rapidly protects against respiratory infection and disease in the upper and lower airways and lung tissue of nonhuman primates.

28 citations


Cited by
More filters
Journal ArticleDOI
18 May 2020-Nature
TL;DR: Several monoclonal antibodies that target the S glycoprotein of SARS-CoV-2, which was identified from memory B cells of an individual who was infected with severe acute respiratory syndrome coronavirus (SARS- coV) in 2003, and one antibody (named S309) potently neutralization, which may limit the emergence of neutralization-escape mutants.
Abstract: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a newly emerged coronavirus that is responsible for the current pandemic of coronavirus disease 2019 (COVID-19), which has resulted in more than 3.7 million infections and 260,000 deaths as of 6 May 20201,2. Vaccine and therapeutic discovery efforts are paramount to curb the pandemic spread of this zoonotic virus. The SARS-CoV-2 spike (S) glycoprotein promotes entry into host cells and is the main target of neutralizing antibodies. Here we describe several monoclonal antibodies that target the S glycoprotein of SARS-CoV-2, which we identified from memory B cells of an individual who was infected with severe acute respiratory syndrome coronavirus (SARS-CoV) in 2003. One antibody (named S309) potently neutralizes SARS-CoV-2 and SARS-CoV pseudoviruses as well as authentic SARS-CoV-2, by engaging the receptor-binding domain of the S glycoprotein. Using cryo-electron microscopy and binding assays, we show that S309 recognizes an epitope containing a glycan that is conserved within the Sarbecovirus subgenus, without competing with receptor attachment. Antibody cocktails that include S309 in combination with other antibodies that we identified further enhanced SARS-CoV-2 neutralization, and may limit the emergence of neutralization-escape mutants. These results pave the way for using S309 and antibody cocktails containing S309 for prophylaxis in individuals at a high risk of exposure or as a post-exposure therapy to limit or treat severe disease.

1,546 citations

Journal ArticleDOI
12 Nov 2020-Cell
TL;DR: It is found that both the magnitude of Ab responses to SARS-CoV-2 spike (S) and nucleoprotein and nAb titers correlate with clinical scores, and the immunodominance of the receptor-binding motif will guide the design of COVID-19 vaccines and therapeutics.

1,042 citations

Journal ArticleDOI
11 Dec 2020-Science
TL;DR: Using diverse assays for antibodies recognizing SARS-CoV-2 proteins, preexisting humoral immunity was detected and antibodies targeting the S2 subunit were particularly prevalent in children and adolescents.
Abstract: Zoonotic introduction of novel coronaviruses may encounter preexisting immunity in humans. Using diverse assays for antibodies recognizing SARS-CoV-2 proteins, we detected preexisting humoral immunity. SARS-CoV-2 spike glycoprotein (S)–reactive antibodies were detectable using a flow cytometry–based method in SARS-CoV-2–uninfected individuals and were particularly prevalent in children and adolescents. They were predominantly of the immunoglobulin G (IgG) class and targeted the S2 subunit. By contrast, SARS-CoV-2 infection induced higher titers of SARS-CoV-2 S–reactive IgG antibodies targeting both the S1 and S2 subunits, and concomitant IgM and IgA antibodies, lasting throughout the observation period. SARS-CoV-2–uninfected donor sera exhibited specific neutralizing activity against SARS-CoV-2 and SARS-CoV-2 S pseudotypes. Distinguishing preexisting and de novo immunity will be critical for our understanding of susceptibility to and the natural course of SARS-CoV-2 infection.

721 citations

Journal ArticleDOI
06 May 2020-Viruses
TL;DR: It is demonstrated how these pseudotyped lentiviral particles could be used to measure the neutralizing activity of human sera or plasma against SARS-CoV-2 in convenient luciferase-based assays, thereby providing a valuable complement to ELISA-based methods that measure antibody binding rather than neutralization.
Abstract: SARS-CoV-2 enters cells using its Spike protein, which is also the main target of neutralizing antibodies. Therefore, assays to measure how antibodies and sera affect Spike-mediated viral infection are important for studying immunity. Because SARS-CoV-2 is a biosafety-level-3 virus, one way to simplify such assays is to pseudotype biosafety-level-2 viral particles with Spike. Such pseudotyping has now been described for single-cycle lentiviral, retroviral, and vesicular stomatitis virus (VSV) particles, but the reagents and protocols are not widely available. Here, we detailed how to effectively pseudotype lentiviral particles with SARS-CoV-2 Spike and infect 293T cells engineered to express the SARS-CoV-2 receptor, ACE2. We also made all the key experimental reagents available in the BEI Resources repository of ATCC and the NIH. Furthermore, we demonstrated how these pseudotyped lentiviral particles could be used to measure the neutralizing activity of human sera or plasma against SARS-CoV-2 in convenient luciferase-based assays, thereby providing a valuable complement to ELISA-based methods that measure antibody binding rather than neutralization.

609 citations

Posted ContentDOI
20 Apr 2020-bioRxiv
TL;DR: It is demonstrated how these pseudotyped lentiviral particles can be used to measure the neutralizing activity of human sera or plasma against SARS-CoV-2 in convenient luciferase-based assays, thereby providing a valuable complement to ELISA-based methods that measure antibody binding rather than neutralization.
Abstract: SARS-CoV-2 enters cells using its Spike protein, which is also the main target of neutralizing antibodies. Therefore, assays to measure how antibodies and sera affect Spike-mediated viral infection are important for studying immunity. Because SARS-CoV-2 is a biosafety-level-3 virus, one way to simplify such assays is to pseudotype biosafety-level-2 viral particles with Spike. Such pseudotyping has now been described for single-cycle lentiviral, retroviral and VSV particles, but the reagents and protocols are not widely available. Here we detail how to effectively pseudotype lentiviral particles with SARS-CoV-2 Spike and infect 293T cells engineered to express the SARS-CoV-2 receptor, ACE2. We also make all the key experimental reagents available in the BEI Resources repository of ATCC and the NIH. Furthermore, we demonstrate how these pseudotyped lentiviral particles can be used to measure the neutralizing activity of human sera or plasma against SARS-CoV-2 in convenient luciferase-based assays, thereby providing a valuable complement to ELISA-based methods that measure antibody binding rather than neutralization.

297 citations