3
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Conversion of monoclonal IgG to dimeric and secretory IgA restores neutralizing ability and prevents infection of Omicron lineages

      research-article
      a , b , c , a , d , e , d , a , e , f , e , g , a , e , h , h , a , a , d , i , j , k , l , m , n , o , o , p , p , q , r , d , s , d , h , t , o , f , u , v , w , x , e , y , b , n , w , x , a , g , d , b , c , 3 , , a , 3 ,
      Proceedings of the National Academy of Sciences of the United States of America
      National Academy of Sciences
      IgA, SARS-CoV-2, Omicron, antibody engineering, antibody therapy

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Significance

          Considering the high risk of breakthrough infections in individuals with an insufficient mucosal immunoglobulin A (IgA) response, we have engineered various forms of monoclonal IgA antibodies for direct administration to the mucosal surface. The dimerization of IgA, potentially through increased avidity, significantly enhances the potency of broadly neutralizing antibodies tested. Importantly, converting IgG to dimeric and secretory forms of IgA restores neutralizing ability against Omicron variants. When administered intranasally, the dimeric IgA antibody DXP-604 provided both prophylactic and therapeutic protection against Omicron BA.5 in transgenic mice expressing human ACE2. Thus, the nasal spray delivery of dimeric or secretory IgA antibodies holds the potential to effectively block viral infection and enhance mucosal immunity against severe acute respiratory syndrome coronavirus 2.

          Abstract

          The emergence of Omicron lineages and descendent subvariants continues to present a severe threat to the effectiveness of vaccines and therapeutic antibodies. We have previously suggested that an insufficient mucosal immunoglobulin A (IgA) response induced by the mRNA vaccines is associated with a surge in breakthrough infections. Here, we further show that the intramuscular mRNA and/or inactivated vaccines cannot sufficiently boost the mucosal secretory IgA response in uninfected individuals, particularly against the Omicron variant. We thus engineered and characterized recombinant monomeric, dimeric, and secretory IgA1 antibodies derived from four neutralizing IgG monoclonal antibodies (mAbs 01A05, rmAb23, DXP-604, and XG014) targeting the receptor-binding domain of the spike protein. Compared to their parental IgG antibodies, dimeric and secretory IgA1 antibodies showed a higher neutralizing activity against different variants of concern (VOCs), in part due to an increased avidity. Importantly, the dimeric or secretory IgA1 form of the DXP-604 antibody significantly outperformed its parental IgG antibody, and neutralized the Omicron lineages BA.1, BA.2, and BA.4/5 with a 25- to 75-fold increase in potency. In human angiotensin converting enzyme 2 (ACE2) transgenic mice, a single intranasal dose of the dimeric IgA DXP-604 conferred prophylactic and therapeutic protection against Omicron BA.5. Thus, dimeric or secretory IgA delivered by nasal administration may potentially be exploited for the treatment and prevention of Omicron infection, thereby providing an alternative tool for combating immune evasion by the current circulating subvariants and, potentially, future VOCs.

          Related collections

          Most cited references78

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Omicron escapes the majority of existing SARS-CoV-2 neutralizing antibodies

          The SARS-CoV-2 B.1.1.529 (Omicron) variant contains 15 mutations of the receptor-binding domain (RBD). How Omicron evades RBD-targeted neutralizing antibodies requires immediate investigation. Here we use high-throughput yeast display screening 1,2 to determine the profiles of RBD escaping mutations for 247 human anti-RBD neutralizing antibodies and show that the neutralizing antibodies can be classified by unsupervised clustering into six epitope groups (A–F)—a grouping that is highly concordant with knowledge-based structural classifications 3–5 . Various single mutations of Omicron can impair neutralizing antibodies of different epitope groups. Specifically, neutralizing antibodies in groups A–D, the epitopes of which overlap with the ACE2-binding motif, are largely escaped by K417N, G446S, E484A and Q493R. Antibodies in group E (for example, S309) 6 and group F (for example, CR3022) 7 , which often exhibit broad sarbecovirus neutralizing activity, are less affected by Omicron, but a subset of neutralizing antibodies are still escaped by G339D, N440K and S371L. Furthermore, Omicron pseudovirus neutralization showed that neutralizing antibodies that sustained single mutations could also be escaped, owing to multiple synergetic mutations on their epitopes. In total, over 85% of the tested neutralizing antibodies were escaped by Omicron. With regard to neutralizing-antibody-based drugs, the neutralization potency of LY-CoV016, LY-CoV555, REGN10933, REGN10987, AZD1061, AZD8895 and BRII-196 was greatly undermined by Omicron, whereas VIR-7831 and DXP-604 still functioned at a reduced efficacy. Together, our data suggest that infection with Omicron would result in considerable humoral immune evasion, and that neutralizing antibodies targeting the sarbecovirus conserved region will remain most effective. Our results inform the development of antibody-based drugs and vaccines against Omicron and future variants.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            SARS-CoV-2 neutralizing antibody structures inform therapeutic strategies

            The coronavirus disease 2019 (COVID-19) pandemic presents an urgent health crisis. Human neutralizing antibodies that target the host ACE2 receptor-binding domain (RBD) of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) spike protein1-5 show promise therapeutically and are being evaluated clinically6-8. Here, to identify the structural correlates of SARS-CoV-2 neutralization, we solved eight new structures of distinct COVID-19 human neutralizing antibodies5 in complex with the SARS-CoV-2 spike trimer or RBD. Structural comparisons allowed us to classify the antibodies into categories: (1) neutralizing antibodies encoded by the VH3-53 gene segment with short CDRH3 loops that block ACE2 and bind only to 'up' RBDs; (2) ACE2-blocking neutralizing antibodies that bind both up and 'down' RBDs and can contact adjacent RBDs; (3) neutralizing antibodies that bind outside the ACE2 site and recognize both up and down RBDs; and (4) previously described antibodies that do not block ACE2 and bind only to up RBDs9. Class 2 contained four neutralizing antibodies with epitopes that bridged RBDs, including a VH3-53 antibody that used a long CDRH3 with a hydrophobic tip to bridge between adjacent down RBDs, thereby locking the spike into a closed conformation. Epitope and paratope mapping revealed few interactions with host-derived N-glycans and minor contributions of antibody somatic hypermutations to epitope contacts. Affinity measurements and mapping of naturally occurring and in vitro-selected spike mutants in 3D provided insight into the potential for SARS-CoV-2 to escape from antibodies elicited during infection or delivered therapeutically. These classifications and structural analyses provide rules for assigning current and future human RBD-targeting antibodies into classes, evaluating avidity effects and suggesting combinations for clinical use, and provide insight into immune responses against SARS-CoV-2.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Rapid epidemic expansion of the SARS-CoV-2 Omicron variant in southern Africa

              The SARS-CoV-2 epidemic in southern Africa has been characterized by three distinct waves. The first was associated with a mix of SARS-CoV-2 lineages, while the second and third waves were driven by the Beta (B.1.351) and Delta (B.1.617.2) variants, respectively 1–3 . In November 2021, genomic surveillance teams in South Africa and Botswana detected a new SARS-CoV-2 variant associated with a rapid resurgence of infections in Gauteng province, South Africa. Within three days of the first genome being uploaded, it was designated a variant of concern (Omicron, B.1.1.529) by the World Health Organization and, within three weeks, had been identified in 87 countries. The Omicron variant is exceptional for carrying over 30 mutations in the spike glycoprotein, which are predicted to influence antibody neutralization and spike function 4 . Here we describe the genomic profile and early transmission dynamics of Omicron, highlighting the rapid spread in regions with high levels of population immunity.
                Bookmark

                Author and article information

                Contributors
                Journal
                Proc Natl Acad Sci U S A
                Proc Natl Acad Sci U S A
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                9 January 2024
                16 January 2024
                9 January 2024
                : 121
                : 3
                : e2315354120
                Affiliations
                [1] aDivision of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , Stockholm 17165, Sweden
                [2] bChangping Laboratory , Beijing 102206, People’s Republic of China
                [3] cSchool of Life Sciences, Biomedical Pioneering Innovation Center, Peking University , Beijing 100871, People’s Republic of China
                [4] dInstitute for Research in Biomedicine, Università della Svizzera italiana , Bellinzona 6500, Switzerland
                [5] eMicrobiology and Virology Department, Fondazione Istituto di ricovero e cura a carattere scientifico (IRCCS) Policlinico San Matteo , Pavia 27100, Italy
                [6] fDivision of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University , Linköping 58185, Sweden
                [7] gYunnan Key Laboratory of Biodiversity Information, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming 650023, People’s Republic of China
                [8] hDepartment of Medical Biotechnology, Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig , Braunschweig 38106, Germany
                [9] iDepartment of Laboratory Medicine, Hospital St. Georg , Leipzig 04129, Germany
                [10] jImmunoDeficiencyCenter Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg , Leipzig 04129, Germany
                [11] kResearch Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences , Tehran 14194, Iran
                [12] lDivision of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet , Stockholm 14186, Sweden
                [13] mDepartment of Immunology, Peking University Health Science Center , Beijing 100191, People’s Republic of China
                [14] nState Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University , Beijing 100871, People’s Republic of China
                [15] oState Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, People’s Republic of China
                [16] pDivision of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital , Stockholm 17176, Sweden
                [17] qRheumatology Unit, Karolinska University Hospital , Stockholm 17176, Sweden
                [18] rEuropean Commission, Joint Research Centre , Ispra 21027, Italy
                [19] sKey Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University , 200032 Shanghai 200032, People’s Republic of China
                [20] tState Key laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences , Wuhan, Hubei 430071, People’s Republic of China
                [21] uDivision of Infectious Diseases, Department of Medicine, Karolinska Institute , Stockholm 17177, Sweden
                [22] vGuangzhou Laboratory , Guangzhou 510005, People’s Republic of China
                [23] wBeijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, National Health Commission Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College , Beijing 100021, People’s Republic of China
                [24] xNational Center of Technology Innovation for Animal Model , Beijing 102206, People’s Republic of China
                [25] yDepartment of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia , Pavia 27100, Italy
                Author notes
                3To whom correspondence may be addressed. Email: sunneyxie@ 123456biopic.pku.edu.cn or qiang.pan-hammarstrom@ 123456ki.se .

                Contributed by Xiaoliang Sunney Xie; received November 7, 2023; accepted November 10, 2023; reviewed by Michel Nussenzweig and Fusheng Wang

                1H.M., Y.C., F.Z., L. Simonelli, contributed equally to this work.

                2J.X., C.Q., L.H., X.Y., L.V., X.S.X., and Q.P.-H. contributed equally to this work.

                Author information
                https://orcid.org/0000-0002-7168-349X
                https://orcid.org/0000-0001-5918-1078
                https://orcid.org/0000-0002-9770-4623
                https://orcid.org/0000-0002-4838-0407
                https://orcid.org/0000-0001-6011-0744
                https://orcid.org/0000-0001-9306-8458
                https://orcid.org/0000-0003-1822-1701
                Article
                202315354
                10.1073/pnas.2315354120
                10801922
                38194459
                e85c5462-1eb8-46bf-b286-2fb32bb51f0b
                Copyright © 2024 the Author(s). Published by PNAS.

                This open access article is distributed under Creative Commons Attribution License 4.0 (CC BY).

                History
                : 07 November 2023
                : 10 November 2023
                Page count
                Pages: 11, Words: 7272
                Funding
                Funded by: The European Union's Horizon 2020 Research and Innovation Program;
                Award ID: ATAC 101003650
                Award Recipient : Harold Marcotte Award Recipient : Luigi Calzolai Award Recipient : Andrea Cavalli Award Recipient : Davide Robbiani Award Recipient : Michael Hust Award Recipient : Liqiang Feng Award Recipient : Fausto Baldanti Award Recipient : Lennart Hammarström Award Recipient : Xinglou Yang Award Recipient : Luca Varani Award Recipient : Qiang Pan-Hammarström
                Funded by: Swiss National Science Foundation;
                Award ID: BRIDGE 40B2-0 _ 203488
                Award Recipient : Harold Marcotte Award Recipient : Luigi Calzolai Award Recipient : Andrea Cavalli Award Recipient : Davide Robbiani Award Recipient : Michael Hust Award Recipient : Liqiang Feng Award Recipient : Fausto Baldanti Award Recipient : Lennart Hammarström Award Recipient : Xinglou Yang Award Recipient : Luca Varani Award Recipient : Qiang Pan-Hammarström
                Funded by: National Natural Science Foundation of China;
                Award ID: No. 82061138006
                Award Recipient : Harold Marcotte Award Recipient : Luigi Calzolai Award Recipient : Andrea Cavalli Award Recipient : Davide Robbiani Award Recipient : Michael Hust Award Recipient : Liqiang Feng Award Recipient : Fausto Baldanti Award Recipient : Lennart Hammarström Award Recipient : Xinglou Yang Award Recipient : Luca Varani Award Recipient : Qiang Pan-Hammarström
                Categories
                covid-19, Coronavirus (COVID-19)
                dataset, Dataset
                research-article, Research Article
                app-bio, Applied Biological Sciences
                403
                530
                Biological Sciences
                Applied Biological Sciences
                Custom metadata
                free

                iga,sars-cov-2,omicron,antibody engineering,antibody therapy

                Comments

                Comment on this article