scispace - formally typeset
Search or ask a question
Journal ArticleDOI

Epitope diversity of SARS-CoV-2 hyperimmune intravenous human immunoglobulins and neutralization of variants of concern.

TL;DR: In this article, the authors explored the antibody epitope repertoire, and virus neutralizing capacity of six hCoV-2IG batches as well as nine convalescent plasma (CP) against SARS-CoV2 and emerging variants of concern (VOC).
About: This article is published in iScience.The article was published on 2021-08-20 and is currently open access. It has received 21 citations till now. The article focuses on the topics: Neutralization & Epitope.
Citations
More filters
Journal ArticleDOI
TL;DR: In this paper, SARS-CoV-2 proteome-wide immunoprofiling of children with mild/moderate or severe coronavirus disease 2019 (COVID-19) versus multisystem inflammatory syndrome in children versus hospitalized control patients revealed differential cytokine responses, IgM/IgG-IgA diversity, antibody binding and avidity.
Abstract: There is limited understanding of the viral antibody fingerprint following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in children. Herein, SARS-CoV-2 proteome-wide immunoprofiling of children with mild/moderate or severe coronavirus disease 2019 (COVID-19) versus multisystem inflammatory syndrome in children versus hospitalized control patients revealed differential cytokine responses, IgM/IgG/IgA epitope diversity, antibody binding and avidity. Apart from spike and nucleocapsid, IgG/IgA recognized epitopes in nonstructural protein (NSP) 2, NSP3, NSP12-NSP14 and open reading frame (ORF) 3a-ORF9. Peptides representing epitopes in NSP12, ORF3a and ORF8 demonstrated SARS-CoV-2 serodiagnosis. Antibody-binding kinetics with 24 SARS-CoV-2 proteins revealed antibody parameters that distinguish children with mild/moderate versus severe COVID-19 or multisystem inflammatory syndrome in children. Antibody avidity to prefusion spike correlated with decreased illness severity and served as a clinical disease indicator. The fusion peptide and heptad repeat 2 region induced SARS-CoV-2-neutralizing antibodies in rabbits. Thus, we identified SARS-CoV-2 antibody signatures in children associated with disease severity and delineate promising serodiagnostic and virus neutralization targets. These findings might guide the design of serodiagnostic assays, prognostic algorithms, therapeutics and vaccines in this important but understudied population.

32 citations

Journal ArticleDOI
TL;DR: In this paper , the authors evaluated the neutralization capacity of antibodies against Omicron after a prior SARS-CoV-2 infection in children and adolescents and found that less than 10% of the vaccinated patients showed neutralizing antibody titers against omicron.
Abstract: Abstract Neutralization capacity of antibodies against Omicron after a prior SARS-CoV-2 infection in children and adolescents is not well studied. Therefore, we evaluated virus-neutralizing capacity against SARS-CoV-2 Alpha, Beta, Gamma, Delta and Omicron variants by age-stratified analyses (<5, 5–11, 12–21 years) in 177 pediatric patients hospitalized with severe acute COVID-19, acute MIS-C, and in convalescent samples of outpatients with mild COVID-19 during 2020 and early 2021. Across all patients, less than 10% show neutralizing antibody titers against Omicron. Children <5 years of age hospitalized with severe acute COVID-19 have lower neutralizing antibodies to SARS-CoV-2 variants compared with patients >5 years of age. As expected, convalescent pediatric COVID-19 and MIS-C cohorts demonstrate higher neutralization titers than hospitalized acute COVID-19 patients. Overall, children and adolescents show some loss of cross-neutralization against all variants, with the most pronounced loss against Omicron. In contrast to SARS-CoV-2 infection, children vaccinated twice demonstrated higher titers against Alpha, Beta, Gamma, Delta and Omicron. These findings can influence transmission, re-infection and the clinical disease outcome from emerging SARS-CoV-2 variants and supports the need for vaccination in children.

32 citations

Journal ArticleDOI
TL;DR: This review article provides insights into the basic aspects of monoclonal antibodies for the therapy of COVID-19, as well as its advancement in terms of clinical trial and current approval status.
Abstract: ABSTRACT Background The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has affected more than 529 million people, and today the world is facing different mutant strains of the virus, leading to increased morbidity rates, fatality rates, and surfacing re-infections. Various therapies, such as prophylactic treatments, repurposed drug treatments, convalescent plasma, and polyclonal antibody therapy have been developed to help combat the coronavirus disease 2019 (COVID-19). Area covered This review article provides insights into the basic aspects of monoclonal antibodies (mAbs) for the therapy of COVID-19, as well as its advancement in terms of clinical trial and current approval status. Expert opinion Monoclonal antibodies represents the most effective and viable therapy and/or prophylaxis option against COVID-19, and have shown a reduction of the viral load, as well as lowering hospitalizations and death rates. In different countries, various mAbs are undergoing different phases of clinical trials, with a few of them having entered phases III and IV. Due to the soaring number of cases worldwide, the FDA has given emergency approval for the mAb combinations bamlanivimab with etesevimab and casirivimab with imdevimab.

30 citations

Journal ArticleDOI
TL;DR: In this article , the authors measured the durability of neutralization capacity and antibody affinity maturation generated following two versus three doses of SARS-CoV-2 mRNA vaccines in naïve versus convalescent individuals (hybrid immunity) against the highly transmissible Omicron BA.1, BA.2 and BA.3 subvariants.
Abstract: Abstract There is limited knowledge on durability of neutralization capacity and antibody affinity maturation generated following two versus three doses of SARS-CoV-2 mRNA vaccines in naïve versus convalescent individuals (hybrid immunity) against the highly transmissible Omicron BA.1, BA.2 and BA.3 subvariants. Virus neutralization titers against the vaccine-homologous strain (WA1) and Omicron sublineages are measured in a pseudovirus neutralization assay (PsVNA). In addition, antibody binding and antibody affinity against spike proteins from WA1, BA.1, and BA.2 is determined using surface plasmon resonance (SPR). The convalescent individuals who after SARS-CoV-2 infection got vaccinated develop hybrid immunity that shows broader neutralization activity and cross-reactive antibody affinity maturation against the Omicron BA.1 and BA.2 after either second or third vaccination compared with naïve individuals. Neutralization activity correlates with antibody affinity against Omicron subvariants BA.1 and BA.2 spikes. Importantly, at four months post-third vaccination the neutralization activity and antibody affinity against the Omicron subvariants is maintained and trended higher for the individuals with hybrid immunity compared with naïve adults. These findings about hybrid immunity resulting in superior immune kinetics, breadth, and durable high affinity antibodies support the need for booster vaccinations to provide effective protection from emerging SARS-CoV-2 variants like the rapidly spreading Omicron subvariants.

28 citations

Journal ArticleDOI
27 Apr 2022-BioDrugs
TL;DR: More than three dozen antibodies or antibody combinations have been forwarded into clinical trials, including Eli Lilly's bamlanivimab and etesevimab, Regeneron's mixture of imdevimab with casirivimabs, Vir's sotrovimab (SOTV), Celltrion's regdanvimab, and Lilly's bebtelovimab as mentioned in this paper .
Abstract: The COVID-19 pandemic is now approaching 2 years old, with more than 440 million people infected and nearly six million dead worldwide, making it the most significant pandemic since the 1918 influenza pandemic. The severity and significance of SARS-CoV-2 was recognized immediately upon discovery, leading to innumerable companies and institutes designing and generating vaccines and therapeutic antibodies literally as soon as recombinant SARS-CoV-2 spike protein sequence was available. Within months of the pandemic start, several antibodies had been generated, tested, and moved into clinical trials, including Eli Lilly’s bamlanivimab and etesevimab, Regeneron’s mixture of imdevimab and casirivimab, Vir’s sotrovimab, Celltrion’s regdanvimab, and Lilly’s bebtelovimab. These antibodies all have now received at least Emergency Use Authorizations (EUAs) and some have received full approval in select countries. To date, more than three dozen antibodies or antibody combinations have been forwarded into clinical trials. These antibodies to SARS-CoV-2 all target the receptor-binding domain (RBD), with some blocking the ability of the RBD to bind human ACE2, while others bind core regions of the RBD to modulate spike stability or ability to fuse to host cell membranes. While these antibodies were being discovered and developed, new variants of SARS-CoV-2 have cropped up in real time, altering the antibody landscape on a moving basis. Over the past year, the search has widened to find antibodies capable of neutralizing the wide array of variants that have arisen, including Alpha, Beta, Gamma, Delta, and Omicron. The recent rise and dominance of the Omicron family of variants, including the rather disparate BA.1 and BA.2 variants, demonstrate the need to continue to find new approaches to neutralize the rapidly evolving SARS-CoV-2 virus. This review highlights both convalescent plasma- and polyclonal antibody-based approaches as well as the top approximately 50 antibodies to SARS-CoV-2, their epitopes, their ability to bind to SARS-CoV-2 variants, and how they are delivered. New approaches to antibody constructs, including single domain antibodies, bispecific antibodies, IgA- and IgM-based antibodies, and modified ACE2-Fc fusion proteins, are also described. Finally, antibodies being developed for palliative care of COVID-19 disease, including the ramifications of cytokine release syndrome (CRS) and acute respiratory distress syndrome (ARDS), are described.

16 citations

References
More filters
Journal ArticleDOI
Paul Bastard1, Paul Bastard2, Paul Bastard3, Lindsey B. Rosen4, Qian Zhang3, Eleftherios Michailidis3, Hans-Heinrich Hoffmann3, Yu Zhang4, Karim Dorgham2, Quentin Philippot1, Quentin Philippot2, Jérémie Rosain2, Jérémie Rosain1, Vivien Béziat2, Vivien Béziat3, Vivien Béziat1, Jeremy Manry2, Jeremy Manry1, Elana Shaw4, Liis Haljasmägi5, Pärt Peterson5, Lazaro Lorenzo1, Lazaro Lorenzo2, Lucy Bizien1, Lucy Bizien2, Sophie Trouillet-Assant6, Kerry Dobbs4, Adriana Almeida de Jesus4, Alexandre Belot6, Anne Kallaste7, Emilie Catherinot, Yacine Tandjaoui-Lambiotte1, Jérémie Le Pen3, Gaspard Kerner2, Gaspard Kerner1, Benedetta Bigio3, Yoann Seeleuthner1, Yoann Seeleuthner2, Rui Yang3, Alexandre Bolze, András N Spaan3, András N Spaan8, Ottavia M. Delmonte4, Michael S. Abers4, Alessandro Aiuti9, Giorgio Casari9, Vito Lampasona9, Lorenzo Piemonti9, Fabio Ciceri9, Kaya Bilguvar10, Richard P. Lifton3, Richard P. Lifton10, Marc Vasse, David M. Smadja2, Mélanie Migaud1, Mélanie Migaud2, Jérôme Hadjadj2, Benjamin Terrier2, Darragh Duffy11, Lluis Quintana-Murci12, Lluis Quintana-Murci11, Diederik van de Beek13, Lucie Roussel14, Donald C. Vinh14, Stuart G. Tangye15, Stuart G. Tangye16, Filomeen Haerynck17, David Dalmau18, Javier Martinez-Picado19, Javier Martinez-Picado20, Petter Brodin21, Petter Brodin22, Michel C. Nussenzweig3, Michel C. Nussenzweig23, Stéphanie Boisson-Dupuis3, Stéphanie Boisson-Dupuis1, Stéphanie Boisson-Dupuis2, Carlos Rodríguez-Gallego, Guillaume Vogt2, Trine H. Mogensen24, Trine H. Mogensen25, Andrew J. Oler4, Jingwen Gu4, Peter D. Burbelo4, Jeffrey I. Cohen4, Andrea Biondi26, Laura Rachele Bettini26, Mariella D'Angiò26, Paolo Bonfanti26, Patrick Rossignol27, Julien Mayaux2, Frédéric Rieux-Laucat2, Eystein S. Husebye28, Eystein S. Husebye29, Eystein S. Husebye30, Francesca Fusco, Matilde Valeria Ursini, Luisa Imberti31, Alessandra Sottini31, Simone Paghera31, Eugenia Quiros-Roldan32, Camillo Rossi, Riccardo Castagnoli33, Daniela Montagna33, Amelia Licari33, Gian Luigi Marseglia33, Xavier Duval, Jade Ghosn2, Hgid Lab4, Covid Clinicians5, Covid-Storm Clinicians§4, CoV-Contact Cohort§2, Amsterdam Umc Covid Biobank3, Amsterdam Umc Covid Biobank1, Amsterdam Umc Covid Biobank2, Covid Human Genetic Effort3, John S. Tsang4, Raphaela Goldbach-Mansky4, Kai Kisand5, Michail S. Lionakis4, Anne Puel1, Anne Puel3, Anne Puel2, Shen-Ying Zhang1, Shen-Ying Zhang2, Shen-Ying Zhang3, Steven M. Holland4, Guy Gorochov2, Emmanuelle Jouanguy3, Emmanuelle Jouanguy2, Emmanuelle Jouanguy1, Charles M. Rice3, Aurélie Cobat2, Aurélie Cobat3, Aurélie Cobat1, Luigi D. Notarangelo4, Laurent Abel1, Laurent Abel2, Laurent Abel3, Helen C. Su4, Jean-Laurent Casanova 
23 Oct 2020-Science
TL;DR: A means by which individuals at highest risk of life-threatening COVID-19 can be identified is identified, and the hypothesis that neutralizing auto-Abs against type I IFNs may underlie critical CO VID-19 is tested.
Abstract: Interindividual clinical variability in the course of SARS-CoV-2 infection is immense. We report that at least 101 of 987 patients with life-threatening COVID-19 pneumonia had neutralizing IgG auto-Abs against IFN-ω (13 patients), the 13 types of IFN-α (36), or both (52), at the onset of critical disease; a few also had auto-Abs against the other three type I IFNs. The auto-Abs neutralize the ability of the corresponding type I IFNs to block SARS-CoV-2 infection in vitro. These auto-Abs were not found in 663 individuals with asymptomatic or mild SARS-CoV-2 infection and were present in only 4 of 1,227 healthy individuals. Patients with auto-Abs were aged 25 to 87 years and 95 were men. A B cell auto-immune phenocopy of inborn errors of type I IFN immunity underlies life-threatening COVID-19 pneumonia in at least 2.6% of women and 12.5% of men.

1,913 citations

Journal ArticleDOI
08 Mar 2021-Nature
TL;DR: In this paper, the authors show that B.1.7 is refractory to neutralization by most monoclonal antibodies against the N-terminal domain of the spike protein and is relatively resistant to a few monoclanal antibody against the receptor-binding domain.
Abstract: The COVID-19 pandemic has had widespread effects across the globe, and its causative agent, SARS-CoV-2, continues to spread. Effective interventions need to be developed to end this pandemic. Single and combination therapies with monoclonal antibodies have received emergency use authorization1-3, and more treatments are under development4-7. Furthermore, multiple vaccine constructs have shown promise8, including two that have an approximately 95% protective efficacy against COVID-199,10. However, these interventions were directed against the initial SARS-CoV-2 virus that emerged in 2019. The recent detection of SARS-CoV-2 variants B.1.1.7 in the UK11 and B.1.351 in South Africa12 is of concern because of their purported ease of transmission and extensive mutations in the spike protein. Here we show that B.1.1.7 is refractory to neutralization by most monoclonal antibodies against the N-terminal domain of the spike protein and is relatively resistant to a few monoclonal antibodies against the receptor-binding domain. It is not more resistant to plasma from individuals who have recovered from COVID-19 or sera from individuals who have been vaccinated against SARS-CoV-2. The B.1.351 variant is not only refractory to neutralization by most monoclonal antibodies against the N-terminal domain but also by multiple individual monoclonal antibodies against the receptor-binding motif of the receptor-binding domain, which is mostly due to a mutation causing an E484K substitution. Moreover, compared to wild-type SARS-CoV-2, B.1.351 is markedly more resistant to neutralization by convalescent plasma (9.4-fold) and sera from individuals who have been vaccinated (10.3-12.4-fold). B.1.351 and emergent variants13,14 with similar mutations in the spike protein present new challenges for monoclonal antibody therapies and threaten the protective efficacy of current vaccines.

1,641 citations

Journal ArticleDOI
TL;DR: In this article, the authors show that SARS-CoV-2 501Y.1.V2 spike protein completely escapes three classes of therapeutically relevant antibodies and exhibits substantial to complete escape from neutralization, but not binding, by convalescent plasma.
Abstract: SARS-CoV-2 501Y.V2 (B.1.351), a novel lineage of coronavirus causing COVID-19, contains substitutions in two immunodominant domains of the spike protein. Here, we show that pseudovirus expressing 501Y.V2 spike protein completely escapes three classes of therapeutically relevant antibodies. This pseudovirus also exhibits substantial to complete escape from neutralization, but not binding, by convalescent plasma. These data highlight the prospect of reinfection with antigenically distinct variants and foreshadows reduced efficacy of spike-based vaccines. Substitutions in SARS-CoV-2 spike protein present in the B.1.351 variant first detected in South Africa, when expressed in pseudoviruses, mediate escape from neutralization by monoclonal antibodies under clinical development and by plasma from individuals previously infected with SARS-CoV-2, but do not prevent binding of convalescent plasma to recombinant spike protein containing B.1.351 lineage substitutions.

982 citations

Journal ArticleDOI
25 Feb 2021-Nature
TL;DR: It is demonstrated that relatively low antibody titers are sufficient for protection against SARS-CoV-2 in rhesus macaques, and that cellular immune responses may also contribute to protection if antibody responses are suboptimal.
Abstract: Recent studies have reported the protective efficacy of both natural1 and vaccine-induced2–7 immunity against challenge with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in rhesus macaques. However, the importance of humoral and cellular immunity for protection against infection with SARS-CoV-2 remains to be determined. Here we show that the adoptive transfer of purified IgG from convalescent rhesus macaques (Macaca mulatta) protects naive recipient macaques against challenge with SARS-CoV-2 in a dose-dependent fashion. Depletion of CD8+ T cells in convalescent macaques partially abrogated the protective efficacy of natural immunity against rechallenge with SARS-CoV-2, which suggests a role for cellular immunity in the context of waning or subprotective antibody titres. These data demonstrate that relatively low antibody titres are sufficient for protection against SARS-CoV-2 in rhesus macaques, and that cellular immune responses may contribute to protection if antibody responses are suboptimal. We also show that higher antibody titres are required for treatment of SARS-CoV-2 infection in macaques. These findings have implications for the development of SARS-CoV-2 vaccines and immune-based therapeutic agents. Adoptive transfer of purified IgG from convalescent macaques protects naive macaques against SARS-CoV-2 infection, and cellular immune responses contribute to protection against rechallenge with SARS-CoV-2.

881 citations

Journal ArticleDOI
29 Apr 2021-Cell
TL;DR: In this paper, a structure-function analysis of B.1.351 was performed using a large cohort of convalescent and vaccinee serum samples, and it was shown that 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.

831 citations

Related Papers (5)