1
|
Medernach JG, Li RC, Zhao XY, Yin B, Noonan EA, Etter EF, Raghavan SS, Borish LC, Wilson JM, Barnes BH, Platts-Mills TAE, Ewald SE, Sauer BG, McGowan EC. Immunoglobulin G4 in eosinophilic esophagitis: Immune complex formation and correlation with disease activity. Allergy 2023; 78:3193-3203. [PMID: 37497566 DOI: 10.1111/all.15826] [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: 11/11/2022] [Revised: 05/15/2023] [Accepted: 06/02/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND Recent studies have shown deposition of immunoglobulin G4 (IgG4) and food proteins in the esophageal mucosa of eosinophilic esophagitis (EoE) patients. Our aims were to assess whether co-localization of IgG4 and major cow's milk proteins (CMPs) was associated with EoE disease activity and to investigate the proteins enriched in proximity to IgG4 deposits. METHODS This study included adult subjects with EoE (n = 13) and non-EoE controls (n = 5). Esophageal biopsies were immunofluorescence stained for IgG4 and CMPs. Co-localization in paired samples from active disease and remission was assessed and compared to controls. The proteome surrounding IgG4 deposits was evaluated by the novel technique, AutoSTOMP. IgG4-food protein interactions were confirmed with co-immunoprecipitation and mass spectrometry. RESULTS IgG4-CMP co-localization was higher in the active EoE group compared to paired remission samples (Bos d 4, p = .02; Bos d 5, p = .002; Bos d 8, p = .002). Co-localization was also significantly higher in the active EoE group compared to non-EoE controls (Bos d 4, p = .0013; Bos d 5, p = .0007; Bos d 8, p = .0013). AutoSTOMP identified eosinophil-derived proteins (PRG 2 and 3, EPX, RNASE3) and calpain-14 in IgG4-enriched areas. Co-immunoprecipitation and mass spectrometry confirmed IgG4 binding to multiple food allergens. CONCLUSION These findings further contribute to the understanding of the interaction of IgG4 with food antigens as it relates to EoE disease activity. These data strongly suggest the immune complex formation of IgG4 and major cow's milk proteins. These immune complexes may have a potential role in the pathophysiology of EoE by contributing to eosinophil activation and disease progression.
Collapse
Affiliation(s)
- Jonathan G Medernach
- Division of Pediatric Gastroenterology and Hepatology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Rung-Chi Li
- Division of Allergy and Immunology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Xiao-Yu Zhao
- Department of Microbiology, Immunology and Cancer Biology and The Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Bocheng Yin
- Department of Microbiology, Immunology and Cancer Biology and The Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Emily A Noonan
- Division of Allergy and Immunology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Elaine F Etter
- Division of Allergy and Immunology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Shyam S Raghavan
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Larry C Borish
- Division of Allergy and Immunology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Jeffrey M Wilson
- Division of Allergy and Immunology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Barrett H Barnes
- Division of Pediatric Gastroenterology and Hepatology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Thomas A E Platts-Mills
- Division of Allergy and Immunology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Sarah E Ewald
- Department of Microbiology, Immunology and Cancer Biology and The Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Bryan G Sauer
- Division of Gastroenterology and Hepatology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Emily C McGowan
- Division of Allergy and Immunology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
2
|
In vivo pharmacokinetic enhancement of monomeric Fc and monovalent bispecific designs through structural guidance. Commun Biol 2021; 4:1048. [PMID: 34497355 PMCID: PMC8426389 DOI: 10.1038/s42003-021-02565-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/18/2021] [Indexed: 11/08/2022] Open
Abstract
In a biologic therapeutic landscape that requires versatility in targeting specificity, valency and half-life modulation, the monomeric Fc fusion platform holds exciting potential for the creation of a class of monovalent protein therapeutics that includes fusion proteins and bispecific targeting molecules. Here we report a structure-guided approach to engineer monomeric Fc molecules to adapt multiple versions of half-life extension modifications. Co-crystal structures of these monomeric Fc variants with Fc neonatal receptor (FcRn) shed light into the binding interactions that could serve as a guide for engineering the half-life of antibody Fc fragments. These engineered monomeric Fc molecules also enabled the generation of a novel monovalent bispecific molecular design, which translated the FcRn binding enhancement to improvement of in vivo serum half-life. Lu Shan et al. present a structure-guided approach to engineer a monovalent form of the fragment crystallizable (Fc) region of an IgG4 antibody to adapt multiple versions of half-life extension modifications and bispecific targeting. Additionally, they report co-crystal structures of the variants bound to the Fc neonatal receptor that allow insights into the binding interactions.
Collapse
|
3
|
A structural perspective on the design of decoy immune modulators. Pharmacol Res 2021; 170:105735. [PMID: 34146695 DOI: 10.1016/j.phrs.2021.105735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/23/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
Therapeutic mAbs have dominated the class of immunotherapeutics in general and immune checkpoint inhibitors in particular. The high specificity of mAbs to the target molecule as well as their extended half-life and (or) the effector functions raised by the Fc part are some of the important aspects that contribute to the success of this class of therapeutics. Equally potential candidates are decoys and their fusions that can address some of the inherent limitations of mAbs, like immunogenicity, resistance development, low bio-availability and so on, besides maintaining the advantages of mAbs. The decoys are molecules that trap the ligands and prevent them from interacting with the signaling receptors. Although a few FDA-approved decoy immune modulators are very successful, the potential of this class of drugs is yet to be fully realized. Here, we review various strategies employed in fusion protein therapeutics with a focus on the design of decoy immunomodulators from the structural perspective and discuss how the information on protein structure and function can strategically guide the development of next-generation immune modulators.
Collapse
|
4
|
Arslan FB, Ozturk Atar K, Calis S. Antibody-mediated drug delivery. Int J Pharm 2021; 596:120268. [PMID: 33486037 DOI: 10.1016/j.ijpharm.2021.120268] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 01/10/2023]
Abstract
Passive and active targeted nanoparticulate delivery systems show promise to compensate for lacking properties of conventional therapy such as side effects, insufficient efficiency and accumulation of the drug at target site, poor pharmacokinetic properties etc. For active targeting, physically or covalently conjugated ligands, including monoclonal antibodies and their fragments, are consistently used and researched for targeting delivery systems or drugs to their target site. Currently, there are several FDA approved actively targeted antibody-drug conjugates, whereas no active targeted delivery system is in clinical use at present. However, efforts to successfully formulate actively targeted delivery systems continue. The scope of this review will be the use of monoclonal antibodies and their fragments as targeting ligands. General information about targeted delivery and antibodies will be given at the first half of the review. As for the second half, fragmentation of antibodies and conjugation approaches will be explained. Monoclonal antibodies and their fragments as targeting ligands and approaches for conjugating these ligands to nanoparticulate delivery systems and drugs will be the main focus of this review, polyclonal antibodies will not be included.
Collapse
Affiliation(s)
- Fatma Betul Arslan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Kivilcim Ozturk Atar
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Sema Calis
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
5
|
Miyashita K, Lutz J, Hudgins LC, Toib D, Ashraf AP, Song W, Murakami M, Nakajima K, Ploug M, Fong LG, Young SG, Beigneux AP. Chylomicronemia from GPIHBP1 autoantibodies. J Lipid Res 2020; 61:1365-1376. [PMID: 32948662 PMCID: PMC7604722 DOI: 10.1194/jlr.r120001116] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Some cases of chylomicronemia are caused by autoantibodies against glycosylphosphatidylinositol-anchored HDL binding protein 1 (GPIHBP1), an endothelial cell protein that shuttles LPL to the capillary lumen. GPIHBP1 autoantibodies prevent binding and transport of LPL by GPIHBP1, thereby disrupting the lipolytic processing of triglyceride-rich lipoproteins. Here, we review the "GPIHBP1 autoantibody syndrome" and summarize clinical and laboratory findings in 22 patients. All patients had GPIHBP1 autoantibodies and chylomicronemia, but we did not find a correlation between triglyceride levels and autoantibody levels. Many of the patients had a history of pancreatitis, and most had clinical and/or serological evidence of autoimmune disease. IgA autoantibodies were present in all patients, and IgG4 autoantibodies were present in 19 of 22 patients. Patients with GPIHBP1 autoantibodies had low plasma LPL levels, consistent with impaired delivery of LPL into capillaries. Plasma levels of GPIHBP1, measured with a monoclonal antibody-based ELISA, were very low in 17 patients, reflecting the inability of the ELISA to detect GPIHBP1 in the presence of autoantibodies (immunoassay interference). However, GPIHBP1 levels were very high in five patients, indicating little capacity of their autoantibodies to interfere with the ELISA. Recently, several GPIHBP1 autoantibody syndrome patients were treated successfully with rituximab, resulting in the disappearance of GPIHBP1 autoantibodies and normalization of both plasma triglyceride and LPL levels. The GPIHBP1 autoantibody syndrome should be considered in any patient with newly acquired and unexplained chylomicronemia.
Collapse
Affiliation(s)
- Kazuya Miyashita
- Department of Clinical Laboratory Medicine, Gunma University, Graduate School of Medicine, Maebashi, Japan
- Immuno-Biological Laboratories (IBL), Fujioka, Gunma, Japan
| | - Jens Lutz
- Medical Clinic, Nephrology-Infectious Diseases, Central Rhine Hospital Group, Koblenz, Germany
| | - Lisa C Hudgins
- Rogosin Institute, Weill Cornell Medical College, New York, NY, USA
| | - Dana Toib
- Department of Pediatrics, Drexel University, Philadelphia, PA, USA
- Section of Pediatric Rheumatology, St. Christopher's Hospital for Children, Philadelphia, PA, USA
| | - Ambika P Ashraf
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wenxin Song
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Masami Murakami
- Department of Clinical Laboratory Medicine, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Katsuyuki Nakajima
- Department of Clinical Laboratory Medicine, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Michael Ploug
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
- Biotechnology Research Innovation Center, Copenhagen University, Copenhagen, Denmark
| | - Loren G Fong
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Stephen G Young
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Anne P Beigneux
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
6
|
Botzanowski T, Hernandez-Alba O, Malissard M, Wagner-Rousset E, Deslignière E, Colas O, Haeuw JF, Beck A, Cianférani S. Middle Level IM–MS and CIU Experiments for Improved Therapeutic Immunoglobulin Subclass Fingerprinting. Anal Chem 2020; 92:8827-8835. [DOI: 10.1021/acs.analchem.0c00293] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Thomas Botzanowski
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Oscar Hernandez-Alba
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Martine Malissard
- IRPF—Centre d’Immunologie Pierre-Fabre (CIPF), 74160 Saint-Julien-en-Genevois, France
| | - Elsa Wagner-Rousset
- IRPF—Centre d’Immunologie Pierre-Fabre (CIPF), 74160 Saint-Julien-en-Genevois, France
| | - Evolène Deslignière
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Olivier Colas
- IRPF—Centre d’Immunologie Pierre-Fabre (CIPF), 74160 Saint-Julien-en-Genevois, France
| | - Jean-François Haeuw
- IRPF—Centre d’Immunologie Pierre-Fabre (CIPF), 74160 Saint-Julien-en-Genevois, France
| | - Alain Beck
- IRPF—Centre d’Immunologie Pierre-Fabre (CIPF), 74160 Saint-Julien-en-Genevois, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| |
Collapse
|
7
|
Blech M, Hörer S, Kuhn AB, Kube S, Göddeke H, Kiefer H, Zang Y, Alber Y, Kast SM, Westermann M, Tully MD, Schäfer LV, Garidel P. Structure of a Therapeutic Full-Length Anti-NPRA IgG4 Antibody: Dissecting Conformational Diversity. Biophys J 2019; 116:1637-1649. [PMID: 31023536 DOI: 10.1016/j.bpj.2019.03.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 03/20/2019] [Accepted: 03/25/2019] [Indexed: 01/02/2023] Open
Abstract
We report the x-ray crystal structure of intact, full-length human immunoglobulin (IgG4) at 1.8 Å resolution. The data for IgG4 (S228P), an antibody targeting the natriuretic peptide receptor A, show a previously unrecognized type of Fab-Fc orientation with a distorted λ-shape in which one Fab-arm is oriented toward the Fc portion. Detailed structural analysis by x-ray crystallography and molecular simulations suggest that this is one of several conformations coexisting in a dynamic equilibrium state. These results were confirmed by small angle x-ray scattering in solution. Furthermore, electron microscopy supported these findings by preserving molecule classes of different conformations. This study fosters our understanding of IgG4 in particular and our appreciation of antibody flexibility in general. Moreover, we give insights into potential biological implications, specifically for the interaction of human anti-natriuretic peptide receptor A IgG4 with the neonatal Fc receptor, Fcγ receptors, and complement-activating C1q by considering conformational flexibility.
Collapse
Affiliation(s)
- Michaela Blech
- Innovation Unit, Pharmaceutical Development Biologics, Biberach (Riss), Germany.
| | - Stefan Hörer
- Department Lead Identification and Optimization Support, Structural Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | | | - Sebastian Kube
- Innovation Unit, Pharmaceutical Development Biologics, Biberach (Riss), Germany
| | - Hendrik Göddeke
- Theoretical Chemistry, Ruhr University Bochum, Bochum, Germany
| | - Hans Kiefer
- University of Applied Sciences Biberach, Biberach (Riss), Germany
| | - Yuguo Zang
- University of Applied Sciences Biberach, Biberach (Riss), Germany
| | - Yannic Alber
- Physikalische Chemie III, Technische Universität Dortmund, Dortmund, Germany
| | - Stefan M Kast
- Physikalische Chemie III, Technische Universität Dortmund, Dortmund, Germany
| | - Martin Westermann
- Elektronenmikroskopisches Zentrum, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Mark D Tully
- European Synchrotron Radiation Facility, Grenoble, France
| | - Lars V Schäfer
- Theoretical Chemistry, Ruhr University Bochum, Bochum, Germany
| | - Patrick Garidel
- Innovation Unit, Pharmaceutical Development Biologics, Biberach (Riss), Germany.
| |
Collapse
|
8
|
Hinge-deleted IgG4 blocker therapy for acetylcholine receptor myasthenia gravis in rhesus monkeys. Sci Rep 2017; 7:992. [PMID: 28428630 PMCID: PMC5430546 DOI: 10.1038/s41598-017-01019-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 03/28/2017] [Indexed: 11/08/2022] Open
Abstract
Autoantibodies against ion channels are the cause of numerous neurologic autoimmune disorders. Frequently, such pathogenic autoantibodies have a restricted epitope-specificity. In such cases, competing antibody formats devoid of pathogenic effector functions (blocker antibodies) have the potential to treat disease by displacing autoantibodies from their target. Here, we have used a model of the neuromuscular autoimmune disease myasthenia gravis in rhesus monkeys (Macaca mulatta) to test the therapeutic potential of a new blocker antibody: MG was induced by passive transfer of pathogenic acetylcholine receptor-specific monoclonal antibody IgG1-637. The effect of the blocker antibody (IgG4Δhinge-637, the hinge-deleted IgG4 version of IgG1-637) was assessed using decrement measurements and single-fiber electromyography. Three daily doses of 1.7 mg/kg IgG1-637 (cumulative dose 5 mg/kg) induced impairment of neuromuscular transmission, as demonstrated by significantly increased jitter, synaptic transmission failures (blockings) and a decrease in the amplitude of the compound muscle action potentials during repeated stimulations (decrement), without showing overt symptoms of muscle weakness. Treatment with three daily doses of 10 mg/kg IgG4Δhinge-637 significantly reduced the IgG1-637-induced increase in jitter, blockings and decrement. Together, these results represent proof-of principle data for therapy of acetylcholine receptor-myasthenia gravis with a monovalent antibody format that blocks binding of pathogenic autoantibodies.
Collapse
|
9
|
Generation and Characterization of an IgG4 Monomeric Fc Platform. PLoS One 2016; 11:e0160345. [PMID: 27479095 PMCID: PMC4968834 DOI: 10.1371/journal.pone.0160345] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/18/2016] [Indexed: 01/07/2023] Open
Abstract
The immunoglobulin Fc region is a homodimer consisted of two sets of CH2 and CH3 domains and has been exploited to generate two-arm protein fusions with high expression yields, simplified purification processes and extended serum half-life. However, attempts to generate one-arm fusion proteins with monomeric Fc, with one set of CH2 and CH3 domains, are often plagued with challenges such as weakened binding to FcRn or partial monomer formation. Here, we demonstrate the generation of a stable IgG4 Fc monomer with a unique combination of mutations at the CH3-CH3 interface using rational design combined with in vitro evolution methodologies. In addition to size-exclusion chromatography and analytical ultracentrifugation, we used multi-angle light scattering (MALS) to show that the engineered Fc monomer exhibits excellent monodispersity. Furthermore, crystal structure analysis (PDB ID: 5HVW) reveals monomeric properties supported by disrupted interactions at the CH3-CH3 interface. Monomeric Fc fusions with Fab or scFv achieved FcRn binding and serum half-life comparable to wildtype IgG. These results demonstrate that this monomeric IgG4 Fc is a promising therapeutic platform to extend the serum half-life of proteins in a monovalent format.
Collapse
|
10
|
|
11
|
Vazquez-Lombardi R, Phan TG, Zimmermann C, Lowe D, Jermutus L, Christ D. Challenges and opportunities for non-antibody scaffold drugs. Drug Discov Today 2015; 20:1271-83. [PMID: 26360055 DOI: 10.1016/j.drudis.2015.09.004] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 08/06/2015] [Accepted: 09/01/2015] [Indexed: 12/22/2022]
Abstract
The first candidates from the promising class of small non-antibody protein scaffolds are now moving into clinical development and practice. Challenges remain, and scaffolds will need to be further tailored toward applications where they provide real advantages over established therapeutics to succeed in a rapidly evolving drug development landscape.
Collapse
Affiliation(s)
- Rodrigo Vazquez-Lombardi
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; The University of New South Wales, Faculty of Medicine, St Vincent's Clinical School, Darlinghurst, Sydney, NSW 2010, Australia
| | - Tri Giang Phan
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; The University of New South Wales, Faculty of Medicine, St Vincent's Clinical School, Darlinghurst, Sydney, NSW 2010, Australia
| | - Carsten Zimmermann
- University of San Diego, School of Business Administration, 5998 Alcala Park, San Diego, CA 92110, USA
| | - David Lowe
- MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK
| | - Lutz Jermutus
- MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK; Trinity Hall, University of Cambridge, Trinity Lane CB2 1TJ, UK.
| | - Daniel Christ
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; The University of New South Wales, Faculty of Medicine, St Vincent's Clinical School, Darlinghurst, Sydney, NSW 2010, Australia.
| |
Collapse
|
12
|
Silva JP, Vetterlein O, Jose J, Peters S, Kirby H. The S228P mutation prevents in vivo and in vitro IgG4 Fab-arm exchange as demonstrated using a combination of novel quantitative immunoassays and physiological matrix preparation. J Biol Chem 2015; 290:5462-9. [PMID: 25568323 DOI: 10.1074/jbc.m114.600973] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Human immunoglobulin G isotype 4 (IgG4) antibodies (Abs) are potential candidates for immunotherapy when reduced effector functions are desirable. IgG4 Abs are dynamic molecules able to undergo a process known as Fab arm exchange (FAE). This results in functionally monovalent, bispecific antibodies (bsAbs) with unknown specificity and hence, potentially, reduced therapeutic efficacy. IgG4 FAE is suggested to be an important biological mechanism that provides the basis for the anti-inflammatory activity attributed to IgG4 Abs. To date, the mechanism of FAE is not entirely understood and studies measuring FAE in ex vivo matrices have been hampered by the presence and abundance of endogenous IgG4 wild-type (WT) Abs. Using representative humanized WT IgG4 monoclonal Abs, namely, anti-IL-6 and anti-TNF, and a core-hinge stabilized serine 228 to proline (S228P) anti-IL-6 IgG4 mutant, it is demonstrated for the first time how anti-IgG4 affinity chromatography can be used to prepare physiologically relevant matrices for assessing and quantifying FAE. A novel method for quantifying FAE using a single MSD immunoassay is also reported and confirms previous findings that, dependent on the redox conditions, the S228P mutation can prevent IgG4 FAE to undetectable levels both in vitro and in vivo. Together, the findings and novel methodologies will allow researchers to monitor and quantify FAE of their own IgG4 molecules in physiologically relevant matrices.
Collapse
Affiliation(s)
- John-Paul Silva
- From the Department of Bioanalytical Sciences, Non-Clinical Development and
| | - Olivia Vetterlein
- From the Department of Bioanalytical Sciences, Non-Clinical Development and
| | - Joby Jose
- From the Department of Bioanalytical Sciences, Non-Clinical Development and
| | - Shirley Peters
- the Department of Antibody Technology and Biology, UCB Pharma, Slough, SL1 3WE United Kingdom
| | - Hishani Kirby
- From the Department of Bioanalytical Sciences, Non-Clinical Development and
| |
Collapse
|
13
|
Rispens T, Davies AM, Ooijevaar-de Heer P, Absalah S, Bende O, Sutton BJ, Vidarsson G, Aalberse RC. Dynamics of inter-heavy chain interactions in human immunoglobulin G (IgG) subclasses studied by kinetic Fab arm exchange. J Biol Chem 2014; 289:6098-109. [PMID: 24425871 PMCID: PMC3937676 DOI: 10.1074/jbc.m113.541813] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interdomain interactions between the CH3 domains of antibody heavy chains are the first step in antibody assembly and are of prime importance for maintaining the native structure of IgG. For human IgG4 it was shown that CH3-CH3 interactions are weak, resulting in the potential for half-molecule exchange (“Fab arm exchange”). Here we systematically investigated non-covalent interchain interactions for CH3 domains in the other human subclasses, including polymorphisms (allotypes), using real-time monitoring of Fab arm exchange with a FRET-based kinetic assay. We identified structural variation between human IgG subclasses and allotypes at three amino acid positions (Lys/Asn-392, Val/Met-397, Lys/Arg-409) to alter the strength of inter-domain interactions by >6 orders of magnitude. Each substitution affected the interactions independent from the other substitutions in terms of affinity, but the enthalpic and entropic contributions were non-additive, suggesting a complex interplay. Allotypic variation in IgG3 resulted in widely different CH3 interaction strengths that were even weaker for IgG3 than for IgG4 in the case of allotype G3m(c3c5*/6,24*), whereas G3m(s*/15*) was equally stable to IgG1. These interactions are sufficiently strong to maintain the structural integrity of IgG1 during its normal life span; for IgG2 and IgG3 the inter-heavy chain disulfide bonds are essential to prevent half-molecule dissociation, whereas the labile hinge disulfide bonds favor half-molecule exchange in vivo for IgG4.
Collapse
Affiliation(s)
- Theo Rispens
- From Sanquin Research, 1066 CX Amsterdam, The Netherlands, and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1105 AZ, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|