1
|
Goldberg BS, Ackerman ME. Underappreciated layers of antibody-mediated immune synapse architecture and dynamics. mBio 2025; 16:e0190024. [PMID: 39660921 PMCID: PMC11708040 DOI: 10.1128/mbio.01900-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Abstract
The biologic activities of antibody drugs are dictated by structure-function relationships-emerging from the kind, composition, and degree of interactions with a target antigen and with soluble and cellular antibody receptors of the innate immune system. These activities are canonically understood to be both modular: antigen recognition is driven by the heterodimeric antigen-binding fragment, and innate immune recruitment by the homodimeric constant/crystallizable fragment. The model that treats these domains with a high degree of independence has served the field well but is not without limitations. Here, we consider how new insights, particularly from structural studies, complicate the model of neat biophysical separation between these domains and shape our understanding of antibody effector functions. The emerging model endeavors to explain the phenotypic impact of both antibody intrinsic characteristics and extrinsic features-fitting them within a spatiotemporal paradigm that better accounts for observed antibody activities. In this review, we will use insights from recent models of classical complement complexes and T cell immune synapse formation to explore how structural differences in antibody-mediated immune synapses may relate to their functional diversity.
Collapse
Affiliation(s)
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
2
|
Yu X, Zhang H, Zhou T, Pan K, Raza SHA, Shen X, Lei H. A non-classical view of antibody properties: Allosteric effect between variable and constant regions. Biotechnol Adv 2025; 78:108482. [PMID: 39579911 DOI: 10.1016/j.biotechadv.2024.108482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/10/2024] [Accepted: 11/16/2024] [Indexed: 11/25/2024]
Abstract
Historically, antibodies have been divided into two functionally independent domains, the variable (V) region for antigen binding and the constant (C) region for mediating effector functions. However, this classical view of antibody function has been severely challenged by a large and growing number of studies, which reveal long-range conformational interactions and allosteric links between the V and C regions. This review comprehensively summarizes the existing studies on antibody allostery, including allosteric conformational changes induced by covalent modifications or noncovalent ligand binding. In addition, we discuss how intramolecular allosteric signals are transmitted from the V to C regions and vice versa. This review argues that there is sufficient evidence to revisit the structure-function relationship of antibodies. These advances in antibody allostery will provide a blueprint for regulating antibody functions in a simple and highly predictable manner. More focus on antibody allostery will definitely benefit antibody engineering and vaccine design in the field of biotechnology.
Collapse
Affiliation(s)
- Xiaoting Yu
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Huiling Zhang
- College of Mathematics and Informatics, South China Agricultural University, Guangzhou 510642, China
| | - Tao Zhou
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Kangliang Pan
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Sayed Haidar Abbas Raza
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Xing Shen
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Hongtao Lei
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
3
|
Ribeiro R, Vítor JMB, Voronovska A, Moreira JN, Goncalves J. Novel Strategy of Antibody Affinity Maturation and Enhancement of Nucleolin-Mediated Antibody-Dependent Cellular Cytotoxicity Against Triple-Negative Breast Cancer. Biotechnol J 2025; 20:e202400380. [PMID: 39868978 DOI: 10.1002/biot.202400380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 01/28/2025]
Abstract
Triple-negative breast cancer (TNBC) is a clinically aggressive subtype of breast cancer that remains an unmet medical need. Because TNBC cells do not express the most common markers of breast cancers, there is an active search for novel molecular targets in triple-negative tumors. Additionally, this subtype of breast cancer presents strong immunogenic characteristics which have been encouraging the development of immunotherapeutic approaches against the disease. In this context, nucleolin arises as a promising target for immunotherapy against TNBC. Our group has previously developed an anti-nucleolin VHH-Fc antibody capable of eliciting antibody-dependent cellular cytotoxicity (ADCC). Moreover, we constructed and characterized an antibody library, that was screened against nucleolin-overexpressing cells, originating an enriched anti-nucleolin antibody pool. In this work, a strategy to select individual clones from the pool was designed, combining NGS data with 3D modeling. Two antibodies demonstrated a significant 4.4- and 6.1-fold increase in binding to nucleolin-overexpressing and TNBC cells, and an improvement in affinity to the sub-micromolar range (0.19 µM and 83.69 nM). Additionally, an increment in 4.6- and 3.1-fold in ADCC activity against respective cell lines was observed for the M2 antibody clone. Herein, the affinity maturation strategy developed was validated and corroborated a positive, but not proportional, correlation between antibody binding, affinity, and ADCC.
Collapse
Affiliation(s)
- Rita Ribeiro
- CNC-UC-Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), Faculty of Medicine (Polo 1), University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, iMed.ULisboa - Research Institute for Medicines, University of Lisbon, Lisbon, Portugal
- Univ Coimbra - University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, University of Coimbra, Coimbra, Portugal
| | - Jorge M B Vítor
- Pathogen Genome Bioinformatics and Computational Biology, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
| | - Anastasiya Voronovska
- CNC-UC-Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), Faculty of Medicine (Polo 1), University of Coimbra, Coimbra, Portugal
| | - João N Moreira
- CNC-UC-Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), Faculty of Medicine (Polo 1), University of Coimbra, Coimbra, Portugal
- Univ Coimbra - University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, University of Coimbra, Coimbra, Portugal
| | - João Goncalves
- Faculty of Pharmacy, iMed.ULisboa - Research Institute for Medicines, University of Lisbon, Lisbon, Portugal
| |
Collapse
|
4
|
Fournier L, Guarnera E, Kolmar H, Becker S. Allosteric antibodies: a novel paradigm in drug discovery. Trends Pharmacol Sci 2024:S0165-6147(24)00218-9. [PMID: 39562213 DOI: 10.1016/j.tips.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/30/2024] [Accepted: 10/18/2024] [Indexed: 11/21/2024]
Abstract
Allostery represents a fundamental mechanism in protein regulation, enabling modulation of protein function from sites distal to the active site. While traditionally explored in the context of small molecules, allosteric modulation is gaining traction as a main mode of action in the realm of antibodies, which offer enhanced specificity and reduced toxicity. This review delves into the rapidly growing field of allosteric antibodies, highlighting recent therapeutic advancements and novel druggability avenues. We also explore the potential of these antibodies as innovative tools in drug discovery and discuss contemporary strategies for designing novel allosteric antibodies, leveraging state-of-the-art computational approaches.
Collapse
Affiliation(s)
- Léxane Fournier
- Early Protein Supply and Characterization, Merck Healthcare KGaA, Darmstadt, Germany; Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Enrico Guarnera
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany.
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany; Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany
| | - Stefan Becker
- Early Protein Supply and Characterization, Merck Healthcare KGaA, Darmstadt, Germany.
| |
Collapse
|
5
|
Whitehead CA, Wines BD, Davies AM, McDonnell JM, Trist HM, Esparon SE, Hogarth PM. Stellabody: A novel hexamer-promoting mutation for improved IgG potency. Immunol Rev 2024; 328:438-455. [PMID: 39364646 PMCID: PMC11659935 DOI: 10.1111/imr.13400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
Advances in antibody engineering are being directed at the development of next generation immunotherapeutics with improved potency. Hexamerisation of IgG is a normal physiological aspect of IgG biology and recently described mutations that facilitate this process have a substantial impact upon monoclonal antibody behavior resulting in the elicitation of dramatically enhanced complement-dependent cytotoxicity, Fc receptor function, and enhanced antigen binding effects, such as targeted receptor agonism or microbe neutralization. Whereas the discovery of IgG hexamerisation enhancing mutations has largely focused on residues with exposure at the surface of the Fc-Fc and CH2-CH3 interfaces, our unique approach is the engineering of the mostly buried residue H429 in the CH3 domain. Selective substitution at position 429 forms the basis of Stellabody technology, where the choice of amino acid results in distinct hexamerisation outcomes. H429F results in monomeric IgG that hexamerises after target binding, so called "on-target" hexamerisation, while the H429Y mutant forms pH-sensitive hexamers in-solution prior to antigen binding. Moreover, Stellabody technologies are broadly applicable across the family of antibody-based biologic therapeutics, including conventional mAbs, bispecific mAbs, and Ig-like biologics such as Fc-fusions, with applications in diverse diseases.
Collapse
Affiliation(s)
- Clarissa A. Whitehead
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Bruce D. Wines
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Anna M. Davies
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's HouseLondonUK
| | - James M. McDonnell
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's HouseLondonUK
| | - Halina M. Trist
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
| | | | - P. Mark Hogarth
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
- Department of Clinical PathologyThe University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
6
|
Edgar JE, Bournazos S. Fc-FcγR interactions during infections: From neutralizing antibodies to antibody-dependent enhancement. Immunol Rev 2024; 328:221-242. [PMID: 39268652 PMCID: PMC11659939 DOI: 10.1111/imr.13393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
Advances in antibody technologies have resulted in the development of potent antibody-based therapeutics with proven clinical efficacy against infectious diseases. Several monoclonal antibodies (mAbs), mainly against viruses such as SARS-CoV-2, HIV-1, Ebola virus, influenza virus, and hepatitis B virus, are currently undergoing clinical testing or are already in use. Although these mAbs exhibit potent neutralizing activity that effectively blocks host cell infection, their antiviral activity results not only from Fab-mediated virus neutralization, but also from the protective effector functions mediated through the interaction of their Fc domains with Fcγ receptors (FcγRs) on effector leukocytes. Fc-FcγR interactions confer pleiotropic protective activities, including the clearance of opsonized virions and infected cells, as well as the induction of antiviral T-cell responses. However, excessive or inappropriate activation of specific FcγR pathways can lead to disease enhancement and exacerbated pathology, as seen in the context of dengue virus infections. A comprehensive understanding of the diversity of Fc effector functions during infection has guided the development of engineered antiviral antibodies optimized for maximal effector activity, as well as the design of targeted therapeutic approaches to prevent antibody-dependent enhancement of disease.
Collapse
Affiliation(s)
- Julia E. Edgar
- The London School of Hygiene and Tropical MedicineLondonUK
| | - Stylianos Bournazos
- The Laboratory of Molecular Genetics and ImmunologyThe Rockefeller UniversityNew YorkNew YorkUSA
| |
Collapse
|
7
|
Lagassé HD, Ou J, Sauna ZE, Golding B. Factor VIII moiety of recombinant Factor VIII Fc fusion protein impacts Fc effector function and CD16 + NK cell activation. Front Immunol 2024; 15:1341013. [PMID: 38655263 PMCID: PMC11035769 DOI: 10.3389/fimmu.2024.1341013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/28/2024] [Indexed: 04/26/2024] Open
Abstract
Recombinant Factor VIII-Fc fusion protein (rFVIIIFc) is an enhanced half-life therapeutic protein product used for the management of hemophilia A. Recent studies have demonstrated that rFVIIIFc interacts with Fc gamma receptors (FcγR) resulting in the activation or inhibition of various FcγR-expressing immune cells. We previously demonstrated that rFVIIIFc, unlike recombinant Factor IX-Fc (rFIXFc), activates natural killer (NK) cells via Fc-mediated interactions with FcγRIIIA (CD16). Additionally, we showed that rFVIIIFc activated CD16+ NK cells to lyse a FVIII-specific B cell clone. Here, we used human NK cell lines and primary NK cells enriched from peripheral blood leukocytes to study the role of the FVIII moiety in rFVIIIFc-mediated NK cell activation. Following overnight incubation of NK cells with rFVIIIFc, cellular activation was assessed by measuring secretion of the inflammatory cytokine IFNγ by ELISA or by cellular degranulation. We show that anti-FVIII, anti-Fc, and anti-CD16 all inhibited indicating that these molecules were involved in rFVIIIFc-mediated NK cell activation. To define which domains of FVIII were involved, we used antibodies that are FVIII domain-specific and demonstrated that blocking FVIII C1 or C2 domain-mediated membrane binding potently inhibited rFVIIIFc-mediated CD16+ NK cell activation, while targeting the FVIII heavy chain domains did not. We also show that rFVIIIFc binds CD16 with about five-fold higher affinity than rFIXFc. Based on our results we propose that FVIII light chain-mediated membrane binding results in tethering of the fusion protein to the cell surface, and this, together with increased binding affinity for CD16, allows for Fc-CD16 interactions to proceed, resulting in NK cellular activation. Our working model may explain our previous results where we observed that rFVIIIFc activated NK cells via CD16, whereas rFIXFc did not despite having identical IgG1 Fc domains.
Collapse
Affiliation(s)
- H.A. Daniel Lagassé
- Division of Hemostasis, Office of Plasma Protein Therapeutics CMC, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Jiayi Ou
- Division of Hemostasis, Office of Plasma Protein Therapeutics CMC, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Zuben E. Sauna
- Division of Hemostasis, Office of Plasma Protein Therapeutics CMC, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Basil Golding
- Office of Plasma Protein Therapeutics CMC, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
8
|
Guo D, De Sciscio ML, Chi-Fung Ng J, Fraternali F. Modelling the assembly and flexibility of antibody structures. Curr Opin Struct Biol 2024; 84:102757. [PMID: 38118364 DOI: 10.1016/j.sbi.2023.102757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/22/2023]
Abstract
Antibodies are large protein assemblies capable of both specifically recognising antigens and engaging with other proteins and receptors to coordinate immune action. Traditionally, structural studies have been dedicated to antibody variable regions, but efforts to determine and model full-length antibody structures are emerging. Here we review the current knowledge on modelling the structures of antibody assemblies, focusing on their conformational flexibility and the challenge this poses to obtaining and evaluating structural models. Integrative modelling approaches, combining experiments (cryo-electron microscopy, mass spectrometry, etc.) and computational methods (molecular dynamics simulations, deep-learning based approaches, etc.), hold the promise to map the complex conformational landscape of full-length antibody structures.
Collapse
Affiliation(s)
- Dongjun Guo
- Institute of Structural and Molecular Biology, University College London, Darwin Building, Gower Street, London, WC1E 6BT, United Kingdom; Randall Centre for Cell & Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, United Kingdom
| | - Maria Laura De Sciscio
- Institute of Structural and Molecular Biology, University College London, Darwin Building, Gower Street, London, WC1E 6BT, United Kingdom; Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, Rome, 00185, Italy
| | - Joseph Chi-Fung Ng
- Institute of Structural and Molecular Biology, University College London, Darwin Building, Gower Street, London, WC1E 6BT, United Kingdom
| | - Franca Fraternali
- Institute of Structural and Molecular Biology, University College London, Darwin Building, Gower Street, London, WC1E 6BT, United Kingdom.
| |
Collapse
|
9
|
Damelang T, de Taeye SW, Rentenaar R, Roya-Kouchaki K, de Boer E, Derksen NIL, van Kessel K, Lissenberg-Thunnissen S, Rooijakkers SHM, Jongerius I, Mebius MM, Schuurman J, Labrijn AF, Vidarsson G, Rispens T. The Influence of Human IgG Subclass and Allotype on Complement Activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1725-1735. [PMID: 37843500 PMCID: PMC10656437 DOI: 10.4049/jimmunol.2300307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 09/20/2023] [Indexed: 10/17/2023]
Abstract
Complement activation via the classical pathway is initiated when oligomeric Igs on target surfaces are recognized by C1 of the complement cascade. The strength of this interaction and activation of the complement system are influenced by structural variation of the Ab, including Ab isotype, subclass, and glycosylation profile. Polymorphic variants of IgG have also been described to influence Fc-dependent effector functions. Therefore, we assessed complement binding, deposition, and complement-dependent cytotoxicity (CDC) of 27 known IgG allotypes with anti-trinitrophenyl specificity. Differences between allotypes within subclasses were minor for IgG1, IgG3, and IgG4 allotypes, and more substantial for IgG2. Allelic variant IGHG2*06, containing a unique serine at position 378 in the CH3 domain, showed less efficient complement activation and CDC compared with other IgG2 polymorphisms. We also observed variable cell lysis between IgG1 and IgG3, with IgG3 being superior in lysis of human RBCs and Ramos cells, and IgG1 being superior in lysis of Raji and Wien133 cells, demonstrating that a long-standing conundrum in the literature depends on cellular context. Furthermore, we compared IgG1 and IgG3 under different circumstances, showing that Ag density and Ab hinge length, but not complement regulators, define the context dependency of Ab-mediated CDC activity. Our results point toward a variation in the capacity of IgG subclasses to activate complement due to single amino acid changes and hinge length differences of allotypes to activate complement, which might give new insights on susceptibility to infectious, alloimmune, or autoimmune diseases and aid the design of Ab-based therapeutics.
Collapse
Affiliation(s)
- Timon Damelang
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Steven W. de Taeye
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands
| | - Rosa Rentenaar
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Kasra Roya-Kouchaki
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands
| | - Esther de Boer
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Amsterdam, the Netherlands
| | - Ninotska I. L. Derksen
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Kok van Kessel
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | - Suzan H. M. Rooijakkers
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ilse Jongerius
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Amsterdam, the Netherlands
| | | | | | | | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
10
|
Burn Aschner C, Muthuraman K, Kucharska I, Cui H, Prieto K, Nair MS, Wang M, Huang Y, Christie-Holmes N, Poon B, Lam J, Sultana A, Kozak R, Mubareka S, Rubinstein JL, Rujas E, Treanor B, Ho DD, Jetha A, Julien JP. A multi-specific, multi-affinity antibody platform neutralizes sarbecoviruses and confers protection against SARS-CoV-2 in vivo. Sci Transl Med 2023; 15:eadf4549. [PMID: 37224226 DOI: 10.1126/scitranslmed.adf4549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 04/26/2023] [Indexed: 05/26/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has been responsible for a global pandemic. Monoclonal antibodies (mAbs) have been used as antiviral therapeutics; however, these therapeutics have been limited in efficacy by viral sequence variability in emerging variants of concern (VOCs) and in deployment by the need for high doses. In this study, we leveraged the multi-specific, multi-affinity antibody (Multabody, MB) platform, derived from the human apoferritin protomer, to enable the multimerization of antibody fragments. MBs were shown to be highly potent, neutralizing SARS-CoV-2 at lower concentrations than their corresponding mAb counterparts. In mice infected with SARS-CoV-2, a tri-specific MB targeting three regions within the SARS-CoV-2 receptor binding domain was protective at a 30-fold lower dose than a cocktail of the corresponding mAbs. Furthermore, we showed in vitro that mono-specific MBs potently neutralize SARS-CoV-2 VOCs by leveraging augmented avidity, even when corresponding mAbs lose their ability to neutralize potently, and that tri-specific MBs expanded the neutralization breadth beyond SARS-CoV-2 to other sarbecoviruses. Our work demonstrates how avidity and multi-specificity combined can be leveraged to confer protection and resilience against viral diversity that exceeds that of traditional monoclonal antibody therapies.
Collapse
Affiliation(s)
- Clare Burn Aschner
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Krithika Muthuraman
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Iga Kucharska
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Hong Cui
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Katherine Prieto
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Manoj S Nair
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Maple Wang
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Yaoxing Huang
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | | | - Betty Poon
- Combined Containment Level 3 Unit, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jessica Lam
- Combined Containment Level 3 Unit, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Azmiri Sultana
- Combined Containment Level 3 Unit, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Robert Kozak
- Department of Laboratory Medicine and Molecular Diagnostics, Division of Microbiology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Samira Mubareka
- Department of Laboratory Medicine and Molecular Diagnostics, Division of Microbiology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Division of Infectious Diseases, Sunnybrook Health Sciences Centre and Department of Medicine, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - John L Rubinstein
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Edurne Rujas
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
- Pharmacokinetic, Nanotechnology and Gene Therapy Group, Faculty of Pharmacy, University of the Basque Country UPV/EHU, 01006 Vitoria, Spain
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, 01006 Vitoria, Spain
| | - Bebhinn Treanor
- Department of Immunology, University of Toronto, ON M5S 1A8, Canada
- Department of Cell and Systems Biology, University of Toronto, ON M5S 3G5, Canada
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON M1C 1A4, Canada
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Arif Jetha
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Jean-Philippe Julien
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Immunology, University of Toronto, ON M5S 1A8, Canada
| |
Collapse
|
11
|
Rosenberg YJ, Ordonez T, Khanwalkar US, Barnette P, Pandey S, Backes IM, Otero CE, Goldberg BS, Crowley AR, Leib DA, Shapiro MB, Jiang X, Urban LA, Lees J, Hessell AJ, Permar S, Haigwood NL, Ackerman ME. Evidence for the Role of a Second Fc-Binding Receptor in Placental IgG Transfer in Nonhuman Primates. mBio 2023; 14:e0034123. [PMID: 36946726 PMCID: PMC10127586 DOI: 10.1128/mbio.00341-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 02/21/2023] [Indexed: 03/23/2023] Open
Abstract
Transplacental transfer of maternal antibodies provides the fetus and newborn with passive protection against infectious diseases. While the role of the highly conserved neonatal Fc receptor (FcRn) in transfer of IgG in mammals is undisputed, recent reports have suggested that a second receptor may contribute to transport in humans. We report poor transfer efficiency of plant-expressed recombinant HIV-specific antibodies, including engineered variants with high FcRn affinity, following subcutaneous infusion into rhesus macaques close to parturition. Unexpectedly, unlike those derived from mammalian tissue culture, plant-derived antibodies were essentially unable to cross macaque placentas. This defect was associated with poor Fcγ receptor binding and altered Fc glycans and was not recapitulated in mice. These results suggest that maternal-fetal transfer of IgG across the three-layer primate placenta may require a second receptor and suggest a means of providing maternal antibody treatments during pregnancy while avoiding fetal harm. IMPORTANCE This study compared the ability of several human HIV envelope-directed monoclonal antibodies produced in plants with the same antibodies produced in mammalian cells for their ability to cross monkey and mouse placentas. We found that the two types of antibodies have comparable transfer efficiencies in mice, but they are differentially transferred across macaque placentas, consistent with a two-receptor IgG transport model in primates. Importantly, plant-produced monoclonal antibodies have excellent binding characteristics for human FcRn receptors, permitting desirable pharmacokinetics in humans. The lack of efficient transfer across the primate placenta suggests that therapeutic plant-based antibody treatments against autoimmune diseases and cancer could be provided to the mother while avoiding transfer and preventing harm to the fetus.
Collapse
Affiliation(s)
| | - Tracy Ordonez
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | - Philip Barnette
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Shilpi Pandey
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Iara M. Backes
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Claire E. Otero
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | | | - Andrew R. Crowley
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - David A. Leib
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Mariya B. Shapiro
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | | | | | - Ann J. Hessell
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Sallie Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Nancy L. Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
12
|
A subset of antibodies targeting citrullinated proteins confers protection from rheumatoid arthritis. Nat Commun 2023; 14:691. [PMID: 36754962 PMCID: PMC9908943 DOI: 10.1038/s41467-023-36257-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/20/2023] [Indexed: 02/10/2023] Open
Abstract
Although elevated levels of anti-citrullinated protein antibodies (ACPAs) are a hallmark of rheumatoid arthritis (RA), the in vivo functions of these antibodies remain unclear. Here, we have expressed monoclonal ACPAs derived from patients with RA, and analyzed their functions in mice, as well as their specificities. None of the ACPAs showed arthritogenicity nor induced pain-associated behavior in mice. However, one of the antibodies, clone E4, protected mice from antibody-induced arthritis. E4 showed a binding pattern restricted to skin, macrophages and dendritic cells in lymphoid tissue, and cartilage derived from mouse and human arthritic joints. Proteomic analysis confirmed that E4 strongly binds to macrophages and certain RA synovial fluid proteins such as α-enolase. The protective effect of E4 was epitope-specific and dependent on the interaction between E4-citrullinated α-enolase immune complexes with FCGR2B on macrophages, resulting in increased IL-10 secretion and reduced osteoclastogenesis. These findings suggest that a subset of ACPAs have therapeutic potential in RA.
Collapse
|
13
|
Crowley AR, Richardson SI, Tuyishime M, Jennewein M, Bailey MJ, Lee J, Alter G, Ferrari G, Morris L, Ackerman ME. Functional consequences of allotypic polymorphisms in human immunoglobulin G subclasses. Immunogenetics 2023; 75:1-16. [PMID: 35904629 PMCID: PMC9845132 DOI: 10.1007/s00251-022-01272-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/07/2022] [Indexed: 01/21/2023]
Abstract
Heritable polymorphisms within the human IgG locus, collectively termed allotypes, have often been linked by statistical associations, but rarely mechanistically, to a wide range of disease states. One potential explanation for these associations is that IgG allotype alters host cell receptors' affinity for IgG, dampening or enhancing an immune response depending on the nature of the change and the receptors. In this work, a panel of allotypic antibody variants were evaluated using multiplexed, label-free biophysical methods and cell-based functional assays to determine what effect, if any, human IgG polymorphisms have on antibody function. While we observed several differences in FcγR affinity among allotypes, there was little evidence of dramatically altered FcγR-based effector function or antigen recognition activity associated with this aspect of genetic variability.
Collapse
Affiliation(s)
- Andrew R Crowley
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | - Simone I Richardson
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, 2131, Gauteng, South Africa
- MRC Antibody Immunity Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Marina Tuyishime
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | | | - Meredith J Bailey
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA
| | - Jiwon Lee
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Lynn Morris
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, 2131, Gauteng, South Africa
- MRC Antibody Immunity Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Margaret E Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA.
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA.
| |
Collapse
|
14
|
Iwasaki YW, Tharakaraman K, Subramanian V, Khongmanee A, Hatas A, Fleischer E, Rurak TT, Ngok-ngam P, Tit-oon P, Ruchirawat M, Satayavivad J, Fuangthong M, Sasisekharan R. Generation of bispecific antibodies by structure-guided redesign of IgG constant regions. Front Immunol 2023; 13:1063002. [PMID: 36703993 PMCID: PMC9871890 DOI: 10.3389/fimmu.2022.1063002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023] Open
Abstract
Bispecific antibodies (BsAbs) form an exciting class of bio-therapeutics owing to their multispecificity. Although numerous formats have been developed, generation of hetero-tetrameric IgG1-like BsAbs having acceptable safety and pharmacokinetics profiles from a single cell culture system remains challenging due to the heterogeneous pairing between the four chains. Herein, we employed a structure-guided approach to engineer mutations in the constant domain interfaces (CH1-CL and CH3-CH3) of heavy and κ light chains to prevent heavy-light mispairing in the antigen binding fragment (Fab) region and heavy-heavy homodimerization in the Fc region. Transient co-transfection of mammalian cells with heavy and light chains of pre-existing antibodies carrying the engineered constant domains generates BsAbs with percentage purity ranging from 78% to 85%. The engineered BsAbs demonstrate simultaneous binding of both antigens, while retaining the thermal stability, Fc-mediated effector properties and FcRn binding properties of the parental antibodies. Importantly, since the variable domains were not modified, the mutations may enable BsAb formation from antibodies belonging to different germline origins and isotypes. The rationally designed mutations reported in this work could serve as a starting point for generating optimized solutions required for large scale production.
Collapse
Affiliation(s)
- Yordkhwan W. Iwasaki
- Program in Environmental Toxicology, Chulabhorn Graduate Institute, Bangkok, Thailand
| | - Kannan Tharakaraman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Vidya Subramanian
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Amnart Khongmanee
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand
| | - Andrew Hatas
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Eduardo Fleischer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Troy T. Rurak
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Patchara Ngok-ngam
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand
| | - Phanthakarn Tit-oon
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand
| | - Mathuros Ruchirawat
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand,Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand
| | - Jutamaad Satayavivad
- Program in Environmental Toxicology, Chulabhorn Graduate Institute, Bangkok, Thailand,Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand,Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
| | - Mayuree Fuangthong
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand,Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand,Program in Applied Biological Sciences, Chulabhorn Graduate Institute, Bangkok, Thailand,*Correspondence: Mayuree Fuangthong, ; Ram Sasisekharan,
| | - Ram Sasisekharan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States,*Correspondence: Mayuree Fuangthong, ; Ram Sasisekharan,
| |
Collapse
|
15
|
Crowley AR, Mehlenbacher MR, Sajadi MM, DeVico AL, Lewis GK, Ackerman ME. Evidence of variable human Fcγ receptor-Fc affinities across differentially-complexed IgG. MAbs 2023; 15:2231128. [PMID: 37405954 PMCID: PMC10324447 DOI: 10.1080/19420862.2023.2231128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/07/2023] Open
Abstract
Antibody-mediated effector functions are widely considered to unfold according to an associative model of IgG-Fcγ receptor (FcγR) interactions. The associative model presupposes that Fc receptors cannot discriminate antigen-bound IgG from free IgG in solution and have equivalent affinities for each. Therefore, the clustering of Fcγ receptors (FcγR) in the cell membrane, cross-activation of intracellular signaling domains, and the formation of the immune synapse are all the result of avid interactions between the Fc region of IgG and FcγRs that collectively overcome the individually weak, transient interactions between binding partners. Antibody allostery, specifically conformational allostery, is a competing model in which antigen-bound antibody molecules undergo a physical rearrangement that causes them to stand out from the background of free IgG by virtue of greater FcγR affinity. Various evidence exists in support of this model of antibody allostery, but it remains controversial. We report observations from multiplexed, label-free kinetic experiments in which the affinity values of FcγR were characterized for covalently immobilized, captured, and antigen-bound IgG. Across the strategies tested, receptors had greater affinity for the antigen-bound mode of IgG presentation. This phenomenon was observed across multiple FcγRs and generalized to multiple antigens, antibody specificities, and subclasses. Furthermore, the thermodynamic signatures of FcγR binding to free or immune-complexed IgG in solution differed when measured by an orthogonal label-free method, but the failure to recapitulate the trend in overall affinity leaves open questions as to what additional factors may be at play.
Collapse
Affiliation(s)
- Andrew R. Crowley
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | | | - Mohammad M. Sajadi
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, USA
- Baltimore VA Medical Center, VA Maryland Health Care System, Baltimore, USA
| | - Anthony L. DeVico
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, USA
| | - George K. Lewis
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, USA
| | - Margaret E. Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
- Department of Chemistry, Dartmouth College, Hanover, NH, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| |
Collapse
|
16
|
Wines BD, Kurtovic L, Trist HM, Esparon S, Lopez E, Chappin K, Chan LJ, Mordant FL, Lee WS, Gherardin NA, Patel SK, Hartley GE, Pymm P, Cooney JP, Beeson JG, Godfrey DI, Burrell LM, van Zelm MC, Wheatley AK, Chung AW, Tham WH, Subbarao K, Kent SJ, Hogarth PM. Fc engineered ACE2-Fc is a potent multifunctional agent targeting SARS-CoV2. Front Immunol 2022; 13:889372. [PMID: 35967361 PMCID: PMC9369017 DOI: 10.3389/fimmu.2022.889372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/27/2022] [Indexed: 01/26/2023] Open
Abstract
Joining a function-enhanced Fc-portion of human IgG to the SARS-CoV-2 entry receptor ACE2 produces an antiviral decoy with strain transcending virus neutralizing activity. SARS-CoV-2 neutralization and Fc-effector functions of ACE2-Fc decoy proteins, formatted with or without the ACE2 collectrin domain, were optimized by Fc-modification. The different Fc-modifications resulted in distinct effects on neutralization and effector functions. H429Y, a point mutation outside the binding sites for FcγRs or complement caused non-covalent oligomerization of the ACE2-Fc decoy proteins, abrogated FcγR interaction and enhanced SARS-CoV-2 neutralization. Another Fc mutation, H429F did not improve virus neutralization but resulted in increased C5b-C9 fixation and transformed ACE2-Fc to a potent mediator of complement-dependent cytotoxicity (CDC) against SARS-CoV-2 spike (S) expressing cells. Furthermore, modification of the Fc-glycan enhanced cell activation via FcγRIIIa. These different immune profiles demonstrate the capacity of Fc-based agents to be engineered to optimize different mechanisms of protection for SARS-CoV-2 and potentially other viral pathogens.
Collapse
Affiliation(s)
- Bruce D. Wines
- Immune therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - Liriye Kurtovic
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Halina M. Trist
- Immune therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia
| | - Sandra Esparon
- Immune therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia
| | - Ester Lopez
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Klasina Chappin
- Immune therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia
| | - Li-Jin Chan
- Infectious Diseases and Immune Defence Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Francesca L. Mordant
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Wen Shi Lee
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Nicholas A. Gherardin
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Sheila K. Patel
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Gemma E. Hartley
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Phillip Pymm
- Infectious Diseases and Immune Defence Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - James P. Cooney
- Infectious Diseases and Immune Defence Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - James G. Beeson
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Microbiology, Monash University, Clayton VIC, Australia
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Louise M. Burrell
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Menno C. van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Alfred Hospital, Melbourne, VIC, Australia
| | - Adam K. Wheatley
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
- Australian Research Council Centre for Excellence in Convergent Bio-Nano Science and Technology, The University of Melbourne, Melbourne, VIC, Australia
| | - Amy W. Chung
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Wai-Hong Tham
- Infectious Diseases and Immune Defence Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
- World Health Organization (WHO) Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J. Kent
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
- Australian Research Council Centre for Excellence in Convergent Bio-Nano Science and Technology, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - P. Mark Hogarth
- Immune therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
17
|
Non-covalent Fc-Fab interactions significantly alter internal dynamics of an IgG1 antibody. Sci Rep 2022; 12:9321. [PMID: 35661134 PMCID: PMC9167292 DOI: 10.1038/s41598-022-13370-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 05/24/2022] [Indexed: 11/17/2022] Open
Abstract
The fragment-antigen-binding arms (Fab1 and Fab2) in a canonical immunoglobulin G (IgG) molecule have identical sequences and hence are always expected to exhibit symmetric conformations and dynamics. Using long all atom molecular simulations of a human IgG1 crystal structure 1HZH, we demonstrate that the translational and rotational dynamics of Fab1 and Fab2 also strongly depend on their interactions with each other and with the fragment-crystallizable (Fc) region. We show that the Fab2 arm in the 1HZH structure is non-covalently bound to the Fc region via long-lived hydrogen bonds, involving its light chain and both heavy chains of the Fc region. These highly stable interactions stabilize non-trivial conformer states with constrained fluctuations. We observe subtle modifications in Fab1 dynamics in response to Fab2-Fc interactions that points to novel allosteric interactions between the Fab arms. These results yield novel insights into the inter- and intra-fragment motions of immunoglobulins which could help us better understand the relation between their structure and function.
Collapse
|
18
|
Kosuge H, Nagatoishi S, Kiyoshi M, Ishii-Watabe A, Terao Y, Ide T, Tsumoto K. Biophysical Characterization of the Contribution of the Fab Region to the IgG-FcγRIIIa Interaction. Biochemistry 2022; 62:262-269. [PMID: 35605982 PMCID: PMC9850916 DOI: 10.1021/acs.biochem.1c00832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The cell-surface receptor FcγRIIIa is crucial to the efficacy of therapeutic antibodies as well as the immune response. The interaction of the Fc region of IgG molecules with FcγRIIIa has been characterized, but until recently, it was thought that the Fab regions were not involved in the interaction. To evaluate the influence of the Fab regions in a biophysical context, we carried out surface plasmon resonance analyses using recombinant FcγRIIIa ligands. A van't Hoff analysis revealed that compared to the interaction of the papain-digested Fc fragment with FcγRIIIa, the interaction of commercially available, full-length rituximab with FcγRIIIa had a more favorable binding enthalpy, a less favorable binding entropy, and a slower off rate. Similar results were obtained from analyses of IgG1 molecules and an IgG1-Fc fragment produced by Expi293 cells. For further validation, we also prepared a maltose-binding protein-linked IgG1-Fc fragment (MBP-Fc). The binding enthalpy of MBP-Fc was nearly equal to that of the IgG1-Fc fragment for the interaction with FcγRIIIa, indicating that such alternatives to the Fab domains as MBP do not positively contribute to the IgG-FcγRIIIa interactions. Our investigation strongly suggests that the Fab region directly interacts with FcγRIIIa, resulting in an increase in the binding enthalpy and a decrease in the dissociation rate, at the expense of favorable binding entropy.
Collapse
Affiliation(s)
- Hirofumi Kosuge
- School
of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Satoru Nagatoishi
- The
Institute of Medical Science, The University
of Tokyo, 4-6-1, Shirokanedai,
Minato-ku, Tokyo 108-8639, Japan,Center
for Drug Design Research, National Institutes
of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki
City, Osaka 567-0085, Japan,
| | - Masato Kiyoshi
- Division
of Biological Chemistry and Biologicals, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| | - Akiko Ishii-Watabe
- Division
of Biological Chemistry and Biologicals, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| | - Yosuke Terao
- Tosoh
Corporation, 2743-1, Hayakawa, Ayase, Kanagawa 252-1123, Japan
| | - Teruhiko Ide
- Tosoh
Corporation, 2743-1, Hayakawa, Ayase, Kanagawa 252-1123, Japan
| | - Kouhei Tsumoto
- School
of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan,The
Institute of Medical Science, The University
of Tokyo, 4-6-1, Shirokanedai,
Minato-ku, Tokyo 108-8639, Japan,Center
for Drug Design Research, National Institutes
of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki
City, Osaka 567-0085, Japan,
| |
Collapse
|
19
|
Tee WV, Wah Tan Z, Guarnera E, Berezovsky IN. Conservation and diversity in allosteric fingerprints of proteins for evolutionary-inspired engineering and design. J Mol Biol 2022; 434:167577. [PMID: 35395233 DOI: 10.1016/j.jmb.2022.167577] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 11/26/2022]
Abstract
Hand-in-hand work of physics and evolution delivered protein universe with diversity of forms, sizes, and functions. Pervasiveness and advantageous traits of allostery made it an important component of the protein function regulation, calling for thorough investigation of its structural determinants and evolution. Learning directly from nature, we explored here allosteric communication in several major folds and repeat proteins, including α/β and β-barrels, β-propellers, Ig-like fold, ankyrin and α/β leucine-rich repeat proteins, which provide structural platforms for many different enzymatic and signalling functions. We obtained a picture of conserved allosteric communication characteristic in different fold types, modifications of the structure-driven signalling patterns via sequence-determined divergence to specific functions, as well as emergence and potential diversification of allosteric regulation in multi-domain proteins and oligomeric assemblies. Our observations will be instrumental in facilitating the engineering and de novo design of proteins with allosterically regulated functions, including development of therapeutic biologics. In particular, results described here may guide the identification of the optimal structural platforms (e.g. fold type, size, and oligomerization states) and the types of diversifications/perturbations, such as mutations, effector binding, and order-disorder transition. The tunable allosteric linkage across distant regions can be used as a pivotal component in the design/engineering of modular biological systems beyond the traditional scaffolding function.
Collapse
Affiliation(s)
- Wei-Ven Tee
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01, Matrix, Singapore 138671
| | - Zhen Wah Tan
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01, Matrix, Singapore 138671
| | - Enrico Guarnera
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01, Matrix, Singapore 138671
| | - Igor N Berezovsky
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01, Matrix, Singapore 138671; Department of Biological Sciences (DBS), National University of Singapore (NUS), 8 Medical Drive, Singapore 117597.
| |
Collapse
|
20
|
Delidakis G, Kim JE, George K, Georgiou G. Improving Antibody Therapeutics by Manipulating the Fc Domain: Immunological and Structural Considerations. Annu Rev Biomed Eng 2022; 24:249-274. [PMID: 35363537 PMCID: PMC9648538 DOI: 10.1146/annurev-bioeng-082721-024500] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Interactions between the crystallizable fragment (Fc) domain of antibodies and a plethora of cellular Fc receptors (FcRs) or soluble proteins form a critical link between humoral and innate immunity. In particular, the immunoglobulin G Fc domain is critical for the clearance of target cells by processes that include (a) cytotoxicity, phagocytosis, or complement lysis; (b) modulation of inflammation; (c) antigen presentation; (d) antibody-mediated receptor clustering; and (e) cytokine release. More than 30 Fc-engineered antibodies aimed primarily at tailoring these effects for optimal therapeutic outcomes are in clinical evaluation or have already been approved. Nonetheless, our understanding of how FcR engagement impacts various immune cell phenotypes is still largely incomplete. Recent insights into FcR biology coupled with advances in Fc:FcR structural analysis, Fc engineering, and mouse models that recapitulate human biology are helping to fill in existing knowledge gaps. These advances will provide a blueprint on how to fine-tune the Fc domain to achieve optimal therapeutic efficacy. Expected final online publication date for the Annual Review of Biomedical Engineering, Volume 24 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- George Delidakis
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA;
| | - Jin Eyun Kim
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Katia George
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - George Georgiou
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA; .,Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA.,Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
21
|
Wang W, Chen Q. Antigen improves binding of IgGs to FcγRs in SPR analysis. Anal Biochem 2022; 640:114411. [PMID: 34648807 DOI: 10.1016/j.ab.2021.114411] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/01/2021] [Accepted: 10/07/2021] [Indexed: 11/22/2022]
Abstract
FcγR binding characterization is one of the critical attributes during the development of therapeutic antibodies. Here, we report a novel assay format to characterize IgG-FcγR interaction in the presence of antigen using Surface plasmon resonance (SPR). The new assay format was developed by creating stable antigen/antibody immunocomplexes on a sensor chip surface before injection of FcγRs. In this assay format, binding activity of both huIgG1 (including IgG1 Fc fusion Protein) and huIgG2 increased significantly to most activating human FcγRs, especially to FcγRI, FcγRIIa-131H and FcγRIIIa-158F. To our knowledge, this study provides the first set of evidence using a biophysical method to demonstrate antigen binding facilitating IgG-FcγR interaction, especially for huIgG2 where previous studies did not indicate its binding to human FcγRI or FcγRIIIa-158F. Although further studies are needed to investigate the correlation of the binding data with effector function data in vivo, our results suggest that it may be useful to evaluate the IgG-FcγR interaction in the presence of antigen to help design safer and more effective biotherapeutics.
Collapse
Affiliation(s)
- Wei Wang
- Department of Therapeutic Discovery, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA, 91320, USA.
| | - Qing Chen
- Department of Therapeutic Discovery, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA, 91320, USA.
| |
Collapse
|
22
|
Yamaguchi Y, Wakaizumi N, Irisa M, Maruno T, Shimada M, Shintani K, Nishiumi H, Yogo R, Yanaka S, Higo D, Torisu T, Kato K, Uchiyama S. The Fab portion of immunoglobulin G has sites in the CL domain that interact with Fc gamma receptor IIIa. MAbs 2022; 14:2038531. [PMID: 35291930 PMCID: PMC8932917 DOI: 10.1080/19420862.2022.2038531] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/16/2022] [Accepted: 02/02/2022] [Indexed: 02/02/2023] Open
Abstract
The interaction between IgG and Fc gamma receptor IIIa (FcγRIIIa) is essential for mediating immune responses. Recent studies have shown that the antigen binding fragment (Fab) and Fc are involved in IgG-FcγRIII interactions. Here, we conducted bio-layer interferometry (BLI) and isothermal titration calorimetry to measure the kinetic and thermodynamic parameters that define the role of Fab in forming the IgG-FcγRIII complex using several marketed therapeutic antibodies. Moreover, hydrogen/deuterium exchange mass spectrometry (HDX-MS) and crosslinking mass spectrometry (XL-MS) were used to clarify the interaction sites and structural changes upon formation of these IgG-FcγRIII complexes. The results showed that Fab in IgG facilitates the interaction via slower dissociation and a larger enthalpy gain. However, a larger entropy loss led to only a marginal change in the equilibrium dissociation constant. Combined HDX-MS and XL-MS analysis revealed that the CL domain of Fab in IgG was in close proximity to FcγRIIIa, indicating that this domain specifically interacts with the extracellular membrane-distal domain (D1) and membrane-proximal domain (D2) of FcγRIIIa. Together with previous studies, these results demonstrate that IgG-FcγRIII interactions are predominantly mediated by the binding of Fc to D2, and the Fab-FcγRIII interaction stabilizes complex formation. These interaction schemes were essentially fucosylation-independent, with Fc-D2 interactions enhanced by afucosylation and the contribution of Fab slightly reduced. Furthermore, the influence of antigen binding on IgG-FcγRIII interactions was also investigated. Combined BLI and HDX-MS results indicate that structural alterations in Fab caused by antigen binding facilitate stabilization of IgG-FcγRIII interactions. This report provides a comprehensive understanding of the interaction between IgG and FcγRIII.
Collapse
Affiliation(s)
- Yuki Yamaguchi
- Graduate School of Engineering, Osaka University, Osaka, Japan
| | | | - Mine Irisa
- Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Takahiro Maruno
- Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Mari Shimada
- Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Koya Shintani
- Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Haruka Nishiumi
- Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Rina Yogo
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi, Japan
- Institute for Molecular Science (IMS), National Institutes of Natural Sciences, Aichi, Japan
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Aichi, Japan
| | - Saeko Yanaka
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi, Japan
- Institute for Molecular Science (IMS), National Institutes of Natural Sciences, Aichi, Japan
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Aichi, Japan
| | | | - Tetsuo Torisu
- Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Koichi Kato
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi, Japan
- Institute for Molecular Science (IMS), National Institutes of Natural Sciences, Aichi, Japan
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Aichi, Japan
| | - Susumu Uchiyama
- Graduate School of Engineering, Osaka University, Osaka, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi, Japan
| |
Collapse
|
23
|
Grasso L, Kline JB, Nicolaides NC. Block-Removed Immunoglobulin Technology to enhance rituximab effector function by counteracting CA125-mediated immunosuppression. Oncol Lett 2021; 23:2. [PMID: 34820001 PMCID: PMC8607236 DOI: 10.3892/ol.2021.13120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 10/01/2021] [Indexed: 11/16/2022] Open
Abstract
Rituximab (RTX) is a CD20-targeting antibody that is the standard-of-care for patients with non-Hodgkin Lymphoma (NHL) cases. RTX's mechanism of action includes complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC). Recent clinical evidence suggests that high serum levels of the tumor-produced mucin 16 (MUC16) and cancer antigen 125 (CA125) have a negative impact on the effectiveness of RTX clinical activity in up to 40% of patients with follicular lymphoma. The present study sought to understand the possible mechanism underlying these results; therefore, cellular and molecular analyses of RTX and CA125 interaction were peformed, and a library of RTX variants was generated using a proprietary technology called Block-Removed Immunoglobulin Technology that combines randomized amino acid substitutions and high-throughput functional screenings to identify CA125-refractory RTX variants. The present study demonstrated that CA125 could bind to RTX and reduce its tumor cell killing activity. Furthermore, the study characterized an RTX variant, named NAV-006 (RTX-N109D), which was more refractory to the immunosuppressive effects mediated by CA125 as evidenced by its reduced CA125 interaction and increased activity of ADCC and CDC when compared with parent RTX. Taken together, these findings warranted further investigation on NAV-006 as a next generation anti-CD20 antibody that could improve the efficacy of parent RTX in NHL patients with high levels of CA125.
Collapse
|
24
|
James EI, Murphree TA, Vorauer C, Engen JR, Guttman M. Advances in Hydrogen/Deuterium Exchange Mass Spectrometry and the Pursuit of Challenging Biological Systems. Chem Rev 2021; 122:7562-7623. [PMID: 34493042 PMCID: PMC9053315 DOI: 10.1021/acs.chemrev.1c00279] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
![]()
Solution-phase hydrogen/deuterium
exchange (HDX) coupled to mass
spectrometry (MS) is a widespread tool for structural analysis across
academia and the biopharmaceutical industry. By monitoring the exchangeability
of backbone amide protons, HDX-MS can reveal information about higher-order
structure and dynamics throughout a protein, can track protein folding
pathways, map interaction sites, and assess conformational states
of protein samples. The combination of the versatility of the hydrogen/deuterium
exchange reaction with the sensitivity of mass spectrometry has enabled
the study of extremely challenging protein systems, some of which
cannot be suitably studied using other techniques. Improvements over
the past three decades have continually increased throughput, robustness,
and expanded the limits of what is feasible for HDX-MS investigations.
To provide an overview for researchers seeking to utilize and derive
the most from HDX-MS for protein structural analysis, we summarize
the fundamental principles, basic methodology, strengths and weaknesses,
and the established applications of HDX-MS while highlighting new
developments and applications.
Collapse
Affiliation(s)
- Ellie I James
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Taylor A Murphree
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Clint Vorauer
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - John R Engen
- Department of Chemistry & Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Miklos Guttman
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
25
|
Sun Y, Estevez A, Schlothauer T, Wecksler AT. Antigen physiochemical properties allosterically effect the IgG Fc-region and Fc neonatal receptor affinity. MAbs 2021; 12:1802135. [PMID: 32795110 PMCID: PMC7531492 DOI: 10.1080/19420862.2020.1802135] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The neonatal Fc receptor (FcRn) is a key membrane protein that plays an integral role in serum immunoglobulin (IgG) recycling, which extends the half-life of antibody. In addition, FcRn is known to traffic antigen-bound immunoglobulins (Ag-IgGs), and to interact with immune complexes to facilitate the antigen cross-presentation of peptides derived from the immune complexes in antigen-presenting cells (APCs). Studies on the IgG-FcRn molecular interactions have primarily focused on the Fc region, and only recently have shown the potential impact of the antigen-binding fragment physiochemical properties on FcRn binding. However, the effect of the antigen physiochemical properties on IgG structure as it relates to Ag-IgG-FcRn binding is not well understood. Here we used an IgG-peptide antigen complex as a model system to investigate the structural effects of the antigen's physiochemical properties on the IgG structure, and the subsequent effects of Ag-IgG-FcRn interactions. We used hydroxyl radical footprinting-mass spectrometry to investigate the structural impact on an IgG upon antigen binding, and observed that the physicochemical properties of the antigen differentially induce conformational changes in the IgG FcRn binding region. The extent of these structural changes directly correlates to the magnitude of the affinity differences between the Ag-IgG complexes and FcRn. Moreover, the antigen's physicochemical properties differentially induce structural differences within the Ag-IgG-FcRn ternary complex. We also provide electron microscopy data that shows corroborating Fab-FcRn interactions, and confirms the hypothesis of potential 2:1 FcRn:IgG binding stoichiometry. These data demonstrate antigen-induced Fc structural rearrangements affect both the affinity toward FcRn and the trimeric antigen-IgG-FcRn complex, providing novel molecular insights in the first steps toward understanding interactions of FcRn-containing large(r)-sized immune complex.
Collapse
Affiliation(s)
- Yue Sun
- Protein Analytical Chemistry, Genentech Inc ., South San Francisco, CA, USA
| | - Alberto Estevez
- Structural Biology, Genentech Inc ., South San Francisco, CA, USA
| | - Tilman Schlothauer
- Roche Pharma Research & Early Development, Roche Innovation Center Munich , Penzberg, Germany.,Biological Technologies, Genentech Inc ., South San Francisco, CA, USA
| | - Aaron T Wecksler
- Protein Analytical Chemistry, Genentech Inc ., South San Francisco, CA, USA
| |
Collapse
|
26
|
Rincon Pabon JP, Kochert BA, Liu YH, Richardson DD, Weis DD. Protein A does not induce allosteric structural changes in an IgG1 antibody during binding. J Pharm Sci 2021; 110:2355-2361. [PMID: 33640336 DOI: 10.1016/j.xphs.2021.02.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 10/22/2022]
Abstract
Affinity chromatography is widely used for antibody purification in biopharmaceutical production. Although there is evidence suggesting that affinity chromatography might induce structural changes in antibodies, allosteric changes in structure have not been well-explored. Here, we used hydrogen exchange-mass spectrometry (HX-MS) to reveal conformational changes in the NIST mAb upon binding with a protein A (ProA) matrix. HX-MS measurements of NIST mAb bound to in-solution and resin forms of ProA revealed regions of the CH2 and CH3 domains with increased protection from HX upon ProA binding, consistent with the known ProA binding region. In-solution ProA experiments revealed regions in the Fab with increased HX uptake when the ProA:mAb molar ratio was increased to 2:1, suggesting an allosterically induced increase in backbone flexibility. Such effects were not observed with lower ProA concentration (1:1 molar ratio) or when ProA resin was used, suggesting some kind of change in binding mode. Since all pharmaceutical processes use ProA bound to resin, our results rule out reversible allosteric effects on the NIST mAb during interaction with resin ProA. However, irreversible effects cannot be ruled out since the NIST mAb was previously exposed to ProA during its original purification.
Collapse
Affiliation(s)
- Juan P Rincon Pabon
- Department of Chemistry and the Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Brent A Kochert
- Analytical Research & Development Mass Spectrometry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Yan-Hui Liu
- Analytical Research & Development Mass Spectrometry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Douglas D Richardson
- Analytical Research & Development Mass Spectrometry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - David D Weis
- Department of Chemistry and the Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, KS, United States.
| |
Collapse
|
27
|
Doepker LE, Danon S, Harkins E, Ralph DK, Yaffe Z, Garrett ME, Dhar A, Wagner C, Stumpf MM, Arenz D, Williams JA, Jaoko W, Mandaliya K, Lee KK, Matsen FA, Overbaugh JM. Development of antibody-dependent cell cytotoxicity function in HIV-1 antibodies. eLife 2021; 10:e63444. [PMID: 33427196 PMCID: PMC7884072 DOI: 10.7554/elife.63444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/08/2021] [Indexed: 11/27/2022] Open
Abstract
A prerequisite for the design of an HIV vaccine that elicits protective antibodies is understanding the developmental pathways that result in desirable antibody features. The development of antibodies that mediate antibody-dependent cellular cytotoxicity (ADCC) is particularly relevant because such antibodies have been associated with HIV protection in humans. We reconstructed the developmental pathways of six human HIV-specific ADCC antibodies using longitudinal antibody sequencing data. Most of the inferred naive antibodies did not mediate detectable ADCC. Gain of antigen binding and ADCC function typically required mutations in complementarity determining regions of one or both chains. Enhancement of ADCC potency often required additional mutations in framework regions. Antigen binding affinity and ADCC activity were correlated, but affinity alone was not sufficient to predict ADCC potency. Thus, elicitation of broadly active ADCC antibodies may require mutations that enable high-affinity antigen recognition along with mutations that optimize factors contributing to functional ADCC activity.
Collapse
Affiliation(s)
- Laura E Doepker
- Human Biology Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Sonja Danon
- Human Biology Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Elias Harkins
- Public Health Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Duncan K Ralph
- Public Health Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Zak Yaffe
- Human Biology Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Medical Scientist Training Program, University of Washington School of MedicineSeattleUnited States
| | - Meghan E Garrett
- Human Biology Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Molecular and Cellular Biology Graduate Program, University of Washington and Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Amrit Dhar
- Public Health Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Department of Statistics, University of WashingtonSeattleUnited States
| | - Cassia Wagner
- Medical Scientist Training Program, University of Washington School of MedicineSeattleUnited States
| | - Megan M Stumpf
- Human Biology Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Dana Arenz
- Human Biology Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - James A Williams
- Department of Medicinal Chemistry, University of WashingtonSeattleUnited States
| | - Walter Jaoko
- Department of Medicinal Microbiology, University of NairobiNairobiKenya
| | - Kishor Mandaliya
- Coast Provincial General Hospital, Women’s Health ProjectMombasaKenya
| | - Kelly K Lee
- Department of Medicinal Chemistry, University of WashingtonSeattleUnited States
| | - Frederick A Matsen
- Public Health Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Julie M Overbaugh
- Human Biology Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Public Health Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| |
Collapse
|