1
|
Elliott IG, Fisher H, Chan HTC, Inzhelevskaya T, Mockridge CI, Penfold CA, Duriez PJ, Orr CM, Herniman J, Müller KTJ, Essex JW, Cragg MS, Tews I. Structure-guided disulfide engineering restricts antibody conformation to elicit TNFR agonism. Nat Commun 2025; 16:3495. [PMID: 40221417 PMCID: PMC11993666 DOI: 10.1038/s41467-025-58773-8] [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: 07/23/2024] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
A promising strategy in cancer immunotherapy is activation of immune signalling pathways through antibodies that target co-stimulatory receptors. hIgG2, one of four human antibody isotypes, is known to deliver strong agonistic activity, and modification of hIgG2 hinge disulfides can influence immune-stimulating activity. This was shown for antibodies directed against the hCD40 receptor, where cysteine-to-serine exchange mutations caused changes in antibody conformational flexibility. Here we demonstrate that the principles of increasing agonism by restricting antibody conformation through disulfide modification can be translated to the co-stimulatory receptor h4-1BB, another member of the tumour necrosis factor receptor superfamily. Furthermore, we explore structure-guided design of the anti-hCD40 antibody ChiLob7/4 and show that engineering additional disulfides between opposing F(ab') arms can elicit conformational restriction, concomitant with enhanced agonism. These results support a mode where subtle increases in rigidity can deliver significant improvements in immunostimulatory activity, thus providing a strategy for the rational design of more powerful antibody therapeutics.
Collapse
Affiliation(s)
- Isabel G Elliott
- School of Chemistry and Chemical Engineering, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, SO16 6YD, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Hayden Fisher
- School of Chemistry and Chemical Engineering, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, SO16 6YD, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- European Synchrotron Radiation Facility, Grenoble, Cedex 9, 38043, France
| | - H T Claude Chan
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, SO16 6YD, UK
| | - Tatyana Inzhelevskaya
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, SO16 6YD, UK
| | - C Ian Mockridge
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, SO16 6YD, UK
| | - Christine A Penfold
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, SO16 6YD, UK
| | - Patrick J Duriez
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, SO16 6YD, UK
| | | | - Julie Herniman
- School of Chemistry and Chemical Engineering, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Kri T J Müller
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, SO16 6YD, UK
| | - Jonathan W Essex
- School of Chemistry and Chemical Engineering, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Mark S Cragg
- Antibody & Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, SO16 6YD, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Ivo Tews
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| |
Collapse
|
2
|
Imamura H, Honda S. IgG4 and IgG1 undergo common acid-induced compaction into an alternatively folded state. FEBS Lett 2025. [PMID: 40095324 DOI: 10.1002/1873-3468.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/09/2025] [Accepted: 03/01/2025] [Indexed: 03/19/2025]
Abstract
Immunoglobulin G1 (IgG1) antibodies undergo denaturation in acidic conditions, resulting in an alternatively folded state (AFS). The AFS structure is more compact than the native state. However, the prevalence of AFS in other subclasses remains largely unexplored. This study provides evidence that humanized IgG4 can also adopt the AFS structure, as demonstrated through size-exclusion chromatography coupled with small-angle X-ray scattering (SEC-SAXS) analysis. These findings suggest that the anomalous compaction of immunoglobulins G (IgGs) is resilient to variations in sequence and structure among subclasses.
Collapse
Affiliation(s)
- Hiroshi Imamura
- Department of Biological Data Science, Nagahama Institute of Bio-Science and Technology, Japan
| | - Shinya Honda
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Messaoudi S, Wai K, Marple A, Baniahmad SF, Wylie RG, Pelletier M, Craig M, Durocher Y, Greschner AA, Gauthier MA. Rapid Systematic Screening of Bispecific Antibody Surrogate Geometries for T-Cell Engagement Using DNA Nanotechnology. J Am Chem Soc 2024; 146:29824-29835. [PMID: 39412838 PMCID: PMC11529601 DOI: 10.1021/jacs.4c11648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 11/01/2024]
Abstract
Bispecific antibodies (bsAbs) are emerging immune-therapeutics, and many formats exist that differ considerably in structure. However, little systematic data exist about how the spatial organization of their components influences activity, requiring innovative approaches combining empirical and quantitative frameworks. This study presents a modular DNA nanotechnology platform to generate numerous bsAbs with surrogate geometries that span the structural features of the BiTE, IgG-like, and IgG-conjugate platforms to screen for T-cell engagement. Results highlight interesting structure-activity relationships regarding bsAb potency and selectivity and raise questions regarding the molecular phenomena underlying activity. To elucidate some effects, the platform was paired with a simple mathematical model. This work is thus one of the first to systematically investigate and reveal the importance of the spatial organization of bsAb components on activity and equally provides an accessible and convenient tool for rapidly mapping out such trends for other combinations of target epitopes.
Collapse
Affiliation(s)
- Sabrine Messaoudi
- Institut
National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Quebec J3X 1P7, Canada
| | - Kevin Wai
- Department
of Chemistry and Chemical Biology, McMaster
University, Hamilton, Ontario L8S 4M1, Canada
| | - April Marple
- Department
of Chemistry and Chemical Biology, McMaster
University, Hamilton, Ontario L8S 4M1, Canada
| | - Seyed Farzad Baniahmad
- Human
Health Therapeutics Research Centre, National
Research Council Canada, Montreal, Quebec H4P 2R2, Canada
| | - Ryan G. Wylie
- Department
of Chemistry and Chemical Biology, McMaster
University, Hamilton, Ontario L8S 4M1, Canada
| | - Marianne Pelletier
- Sainte-Justine
University Hospital Azrieli Research Centre, Montreal, Quebec H3T 1C5, Canada
- Département
de Mathématiques et de Statistique, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Morgan Craig
- Sainte-Justine
University Hospital Azrieli Research Centre, Montreal, Quebec H3T 1C5, Canada
- Département
de Mathématiques et de Statistique, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Yves Durocher
- Human
Health Therapeutics Research Centre, National
Research Council Canada, Montreal, Quebec H4P 2R2, Canada
| | - Andrea A. Greschner
- Institut
National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Quebec J3X 1P7, Canada
| | - Marc A. Gauthier
- Institut
National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Quebec J3X 1P7, Canada
| |
Collapse
|
6
|
Bourel L, Bray F, Vivier S, Flament S, Guilbert L, Chepy A, Rolando C, Launay D, Dubucquoi S, Sobanski V. Comparative Analysis of Laboratory-Scale Immunoglobulin G Purification Methods from Human Serum. J Proteome Res 2024; 23:3933-3943. [PMID: 39140748 PMCID: PMC11468067 DOI: 10.1021/acs.jproteome.4c00268] [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: 04/02/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Immunoglobulin G (IgG) purification is a critical process for evaluating its role in autoimmune diseases, which are defined by the occurrence of autoantibodies. Affinity chromatography with protein G is widely considered to be the optimal technique for laboratory-scale purification. However, this technique has some limitations, including the exposure of IgG to low pH, which can compromise the quality of the purified IgG. Here, we show that alternative methods for IgG purification are possible while maintaining the quality of IgG. Different techniques for IgG purification from serum were evaluated and compared with protein G-based approaches: Melon Gel, caprylic acid-ammonium sulfate (CAAS) precipitation, anion-exchange chromatography with diethylamino ethyl (DEAE) following ammonium sulfate (AS) precipitation, and AS precipitation alone. The results demonstrated that the purification yield of these techniques surpassed that of protein G. However, differences in the purity of IgG were observed using GeLC-MS/MS. The avidity of purified IgG against selected targets (SARS-CoV-2 and topoisomerase-I) was similar between purified IgG obtained using all techniques and unpurified sera. Our work provides valuable insights for future studies of IgG function by recommending alternative purification methods that offer advantages in terms of yield, time efficiency, cost-effectiveness, and milder pH conditions than protein G.
Collapse
Affiliation(s)
- Louisa Bourel
- Univ.
Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational
Research in Inflammation, 59000 Lille, France
| | - Fabrice Bray
- Univ.
Lille, CNRS, UAR 3290 − MSAP − Miniaturisation pour
la Synthèse, l’Analyse et la Protéomique, F-59000 Lille, France
| | - Solange Vivier
- Univ.
Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational
Research in Inflammation, 59000 Lille, France
| | - Stéphanie Flament
- Univ.
Lille, CNRS, UAR 3290 − MSAP − Miniaturisation pour
la Synthèse, l’Analyse et la Protéomique, F-59000 Lille, France
| | - Lucile Guilbert
- Univ.
Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational
Research in Inflammation, 59000 Lille, France
- CHU
Lille, Institut d’Immunologie, Centre de Biologie Pathologie, 59000 Lille, France
| | - Aurélien Chepy
- Univ.
Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational
Research in Inflammation, 59000 Lille, France
- CHU
Lille, Département de Médecine Interne Et Immunologie
Clinique, Centre de référence des Maladies Auto-Immunes
et Auto-inflammatoires Systémiques rares de l’Adulte
du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), 59000 Lille, France
| | - Christian Rolando
- Univ.
Lille, CNRS, UAR 3290 − MSAP − Miniaturisation pour
la Synthèse, l’Analyse et la Protéomique, F-59000 Lille, France
- Shrieking
Sixties, 1-3 Allée
Lavoisier, F-59650 Villeneuve-d’Ascq, France
| | - David Launay
- Univ.
Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational
Research in Inflammation, 59000 Lille, France
- CHU
Lille, Département de Médecine Interne Et Immunologie
Clinique, Centre de référence des Maladies Auto-Immunes
et Auto-inflammatoires Systémiques rares de l’Adulte
du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), 59000 Lille, France
| | - Sylvain Dubucquoi
- Univ.
Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational
Research in Inflammation, 59000 Lille, France
- CHU
Lille, Institut d’Immunologie, Centre de Biologie Pathologie, 59000 Lille, France
| | - Vincent Sobanski
- Univ.
Lille, Inserm, CHU Lille, U1286-INFINITE—Institute for Translational
Research in Inflammation, 59000 Lille, France
- CHU
Lille, Département de Médecine Interne Et Immunologie
Clinique, Centre de référence des Maladies Auto-Immunes
et Auto-inflammatoires Systémiques rares de l’Adulte
du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), 59000 Lille, France
- Institut
Universitaire de France (IUF), 75005 Paris, France
| |
Collapse
|
7
|
Saurabh S, Zhang Q, Li Z, Seddon JM, Kalonia C, Lu JR, Bresme F. Mechanistic Insights into the Adsorption of Monoclonal Antibodies at the Water/Vapor Interface. Mol Pharm 2024; 21:704-717. [PMID: 38194618 PMCID: PMC10848294 DOI: 10.1021/acs.molpharmaceut.3c00821] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 01/11/2024]
Abstract
Monoclonal antibodies (mAbs) are active components of therapeutic formulations that interact with the water-vapor interface during manufacturing, storage, and administration. Surface adsorption has been demonstrated to mediate antibody aggregation, which leads to a loss of therapeutic efficacy. Controlling mAb adsorption at interfaces requires a deep understanding of the microscopic processes that lead to adsorption and identification of the protein regions that drive mAb surface activity. Here, we report all-atom molecular dynamics (MD) simulations of the adsorption behavior of a full IgG1-type antibody at the water/vapor interface. We demonstrate that small local changes in the protein structure play a crucial role in promoting adsorption. Also, interfacial adsorption triggers structural changes in the antibody, potentially contributing to the further enhancement of surface activity. Moreover, we identify key amino acid sequences that determine the adsorption of antibodies at the water-air interface and outline strategies to control the surface activity of these important therapeutic proteins.
Collapse
Affiliation(s)
- Suman Saurabh
- Department
of Chemistry, Molecular Sciences Research
Hub Imperial College, London W12 0BZ, U.K.
| | - Qinkun Zhang
- Department
of Chemistry, Molecular Sciences Research
Hub Imperial College, London W12 0BZ, U.K.
| | - Zongyi Li
- Biological
Physics Group, School of Physics and Astronomy, Faculty of Science
and Engineering, the University of Manchester, Manchester M13 9PL, U.K.
| | - John M. Seddon
- Department
of Chemistry, Molecular Sciences Research
Hub Imperial College, London W12 0BZ, U.K.
| | - Cavan Kalonia
- Dosage
Form Design and Development, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Jian R. Lu
- Biological
Physics Group, School of Physics and Astronomy, Faculty of Science
and Engineering, the University of Manchester, Manchester M13 9PL, U.K.
| | - Fernando Bresme
- Department
of Chemistry, Molecular Sciences Research
Hub Imperial College, London W12 0BZ, U.K.
| |
Collapse
|
8
|
Vanamee ÉS, Faustman DL. The benefits of clustering in TNF receptor superfamily signaling. Front Immunol 2023; 14:1225704. [PMID: 37662920 PMCID: PMC10469783 DOI: 10.3389/fimmu.2023.1225704] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
The tumor necrosis factor (TNF) receptor superfamily is a structurally and functionally related group of cell surface receptors that play crucial roles in various cellular processes, including apoptosis, cell survival, and immune regulation. This review paper synthesizes key findings from recent studies, highlighting the importance of clustering in TNF receptor superfamily signaling. We discuss the underlying molecular mechanisms of signaling, the functional consequences of receptor clustering, and potential therapeutic implications of targeting surface structures of receptor complexes.
Collapse
Affiliation(s)
- Éva S. Vanamee
- Immunobiology Department, Massachusetts General Hospital, Boston, MA, United States
| | - Denise L. Faustman
- Immunobiology Department, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| |
Collapse
|
9
|
Imamura H, Honda S. Cue to Acid-Induced Long-Range Conformational Changes in an Antibody Preceding Aggregation: The Structural Origins of the Subpeaks in Kratky Plots of Small-Angle X-ray Scattering. Int J Mol Sci 2023; 24:12042. [PMID: 37569415 PMCID: PMC10418478 DOI: 10.3390/ijms241512042] [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: 05/31/2023] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Antibody aggregation, followed by acid denaturation and neutralization of pH, is one of the reasons why the production of therapeutic monoclonal antibodies (mAbs) is expensive. Determining the structural details of acid-denatured antibodies is important for understanding their aggregation mechanism and for antibody engineering. Recent research has shown that monoclonal antibodies of human/humanized immunoglobulin G1 (IgG1) become smaller globules at pH 2 compared to their native structure at pH 7. This acid-denatured species is unstable at pH 7 and prone to aggregation by neutralization of pH. Small-angle X-ray scattering (SAXS) data have revealed an acid-induced reduction in the subpeaks in Kratky plot, indicating conformational changes that can lead to aggregation. The subpeaks are well resolved at pH > 3 but less pronounced at pH ≤ 2. One of the weakened subpeaks indicates loosely organized inter-region (Fab-Fab and Fab-Fc) correlations due to acid denaturation. However, the structural origin of the other subpeak (called q3 peak in this study) has not been established because its q region could represent the various inter-region, inter-domain, and intra-domain correlations in IgG1. In this study, we aimed to untangle the effects of domain-domain correlations on Kratky's q3 peak based on the computed SAXS of the crystal structure of IgG1. The q3 peak appeared in the static structure and was more prominent in the Fc region than in the Fab or isolated domains. Further brute-force analysis indicated that longer domain-domain correlations, including the inter-region, also positively contribute to Kratky's q3 peak. Thus, the distortion of the Fc region and a longer inter-region correlation initiate acid denaturation and aggregation.
Collapse
Affiliation(s)
- Hiroshi Imamura
- Department of Bio-Science, Nagahama Institute of Bio-Science and Technology, 1266 Tamura, Nagahama 526-0829, Japan
| | - Shinya Honda
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba 305-8566, Japan
| |
Collapse
|
10
|
Chowdhury AA, Manohar N, Witek MA, Woldeyes MA, Majumdar R, Qian KK, Kimball WD, Xu S, Lanzaro A, Truskett TM, Johnston KP. Subclass Effects on Self-Association and Viscosity of Monoclonal Antibodies at High Concentrations. Mol Pharm 2023; 20:2991-3008. [PMID: 37191356 DOI: 10.1021/acs.molpharmaceut.3c00023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The effects of a subclass of monoclonal antibodies (mAbs) on protein-protein interactions, formation of reversible oligomers (clusters), and viscosity (η) are not well understood at high concentrations. Herein, we quantify a short-range anisotropic attraction between the complementarity-determining region (CDR) and CH3 domains (KCDR-CH3) for vedolizumab IgG1, IgG2, or IgG4 subclasses by fitting small-angle X-ray scattering (SAXS) structure factor Seff(q) data with an extensive library of 12-bead coarse-grained (CG) molecular dynamics simulations. The KCDR-CH3 bead attraction strength was isolated from the strength of long-range electrostatic repulsion for the full mAb, which was determined from the theoretical net charge and a scaling parameter ψ to account for solvent accessibility and ion pairing. At low ionic strength (IS), the strongest short-range attraction (KCDR-CH3) and consequently the largest clusters and highest η were observed with IgG1, the subclass with the most positively charged CH3 domain. Furthermore, the trend in KCDR-CH3 with the subclass followed the electrostatic interaction energy between the CDR and CH3 regions calculated with the BioLuminate software using the 3D mAb structure and molecular interaction potentials. Whereas the equilibrium cluster size distributions and fractal dimensions were determined from fits of SAXS with the MD simulations, the degree of cluster rigidity under flow was estimated from the experimental η with a phenomenological model. For the systems with the largest clusters, especially IgG1, the inefficient packing of mAbs in the clusters played the largest role in increasing η, whereas for other systems, the relative contribution from stress produced by the clusters was more significant. The ability to relate η to short-range attraction from SAXS measurements at high concentrations and to theoretical characterization of electrostatic patches on the 3D surface is not only of fundamental interest but also of practical value for mAb discovery, processing, formulation, and subcutaneous delivery.
Collapse
Affiliation(s)
- Amjad A Chowdhury
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Neha Manohar
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Marta A Witek
- Eli Lilly and Company, Indianapolis, Indiana 46225, United States
| | | | - Ranajoy Majumdar
- Eli Lilly and Company, Indianapolis, Indiana 46225, United States
| | - Ken K Qian
- Eli Lilly and Company, Indianapolis, Indiana 46225, United States
| | - William D Kimball
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Shifeng Xu
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Alfredo Lanzaro
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Thomas M Truskett
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
- Department of Physics, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Keith P Johnston
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
11
|
Katagiri T, Tsukamoto S, Kuratani M, Tsuji S, Nakamura K, Ohte S, Kawaguchi Y, Takaishi K. A blocking monoclonal antibody reveals dimerization of intracellular domains of ALK2 associated with genetic disorders. Nat Commun 2023; 14:2960. [PMID: 37231012 PMCID: PMC10212922 DOI: 10.1038/s41467-023-38746-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/15/2023] [Indexed: 05/27/2023] Open
Abstract
Mutations in activin receptor-like kinase 2 (ALK2) can cause the pathological osteogenic signaling seen in some patients with fibrodysplasia ossificans progressiva and other conditions such as diffuse intrinsic pontine glioma. Here, we report that intracellular domain of wild-type ALK2 readily dimerizes in response to BMP7 binding to drive osteogenic signaling. This osteogenic signaling is pathologically triggered by heterotetramers of type II receptor kinases and ALK2 mutant forms, which form intracellular domain dimers in response to activin A binding. We develop a blocking monoclonal antibody, Rm0443, that can suppress ALK2 signaling. We solve the crystal structure of the ALK2 extracellular domain complex with a Fab fragment of Rm0443 and show that Rm0443 induces dimerization of ALK2 extracellular domains in a back-to-back orientation on the cell membrane by binding the residues H64 and F63 on opposite faces of the ligand-binding site. Rm0443 could prevent heterotopic ossification in a mouse model of fibrodysplasia ossificans progressiva that carries the human R206H pathogenic mutant.
Collapse
Affiliation(s)
- Takenobu Katagiri
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan.
- Project of Clinical and Basic Research for FOP, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan.
| | - Sho Tsukamoto
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
- Project of Clinical and Basic Research for FOP, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
| | - Mai Kuratani
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
| | - Shinnosuke Tsuji
- Specialty Medicine Research Laboratories I, R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Kensuke Nakamura
- Modality Research Laboratories, Biologics Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Satoshi Ohte
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
- Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Yoshiro Kawaguchi
- Modality Research Laboratories, Biologics Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Kiyosumi Takaishi
- Specialty Medicine Research Laboratories I, R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| |
Collapse
|
12
|
Imamura H, Ooishi A, Honda S. Getting Smaller by Denaturation: Acid-Induced Compaction of Antibodies. J Phys Chem Lett 2023; 14:3898-3906. [PMID: 37093025 PMCID: PMC10150727 DOI: 10.1021/acs.jpclett.3c00258] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/07/2023] [Indexed: 05/03/2023]
Abstract
Protein denaturation is a ubiquitous process that occurs both in vitro and in vivo. While our molecular understanding of the denatured structures of proteins is limited, it is commonly accepted that the loss of unique intramolecular contacts makes proteins larger. Herein, we report compaction of the immunoglobulin G1 (IgG1) protein upon acid denaturation. Small-angle X-ray scattering coupled with size exclusion chromatography revealed that IgG1 radii of gyration at pH 2 were ∼75% of those at a neutral pH. Scattering profiles showed a compact globular shape, supported by analytical ultracentrifugation. The acid denaturation of proteins with a decrease in size is energetically costly, and acid-induced compaction requires an attractive force for domain reorientation. Such intramolecular aggregation may be widespread in immunoglobulin proteins as noncanonical structures. Herein, we discuss the potential biological significance of these noncanonical structures of antibodies.
Collapse
Affiliation(s)
- Hiroshi Imamura
- Biomedical
Research Institute, National Institute of
Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
- Department
of Applied Chemistry, College of Life Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga 525-8577, Japan
- Department
of Bio-Science, Nagahama Institute of Bio-Science
and Technology, 1266 Tamura, Nagahama, Shiga 526-0829, Japan
| | - Ayako Ooishi
- Biomedical
Research Institute, National Institute of
Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Shinya Honda
- Biomedical
Research Institute, National Institute of
Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| |
Collapse
|
13
|
Rofo F, Meier SR, Metzendorf NG, Morrison JI, Petrovic A, Syvänen S, Sehlin D, Hultqvist G. A Brain-Targeting Bispecific-Multivalent Antibody Clears Soluble Amyloid-Beta Aggregates in Alzheimer's Disease Mice. Neurotherapeutics 2022; 19:1588-1602. [PMID: 35939261 PMCID: PMC9606191 DOI: 10.1007/s13311-022-01283-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2022] [Indexed: 12/04/2022] Open
Abstract
Amyloid-β (Aβ) oligomers and protofibrils are suggested to be the most neurotoxic Aβ species in Alzheimer's disease (AD). Hence, antibodies with strong and selective binding to these soluble Aβ aggregates are of therapeutic potential. We have recently introduced HexaRmAb158, a multivalent antibody with additional Aβ-binding sites in the form of single-chain fragment variables (scFv) on the N-terminal ends of Aβ protofibril selective antibody (RmAb158). Due to the additional binding sites and the short distance between them, HexaRmAb158 displayed a slow dissociation from protofibrils and strong binding to oligomers in vitro. In the current study, we aimed at investigating the therapeutic potential of this antibody format in vivo using mouse models of AD. To enhance BBB delivery, the transferrin receptor (TfR) binding moiety (scFv8D3) was added, forming the bispecific-multivalent antibody (HexaRmAb158-scFv8D3). The new antibody displayed a weaker TfR binding compared to the previously developed RmAb158-scFv8D3 and was less efficiently transcytosed in a cell-based BBB model. HexaRmAb158 detected soluble Aβ aggregates derived from brains of tg-ArcSwe and AppNL-G-F mice more efficiently compared to RmAb158. When intravenously injected, HexaRmAb158-scFv8D3 was actively transported over the BBB into the brain in vivo. Brain uptake was marginally lower than that of RmAb158-scFv8D3, but significantly higher than observed for conventional IgG antibodies. Both antibody formats displayed similar brain retention (72 h post injection) and equal capacity in clearing soluble Aβ aggregates in tg-ArcSwe mice. In conclusion, we demonstrate a bispecific-multivalent antibody format capable of passing the BBB and targeting a wide-range of sizes of soluble Aβ aggregates.
Collapse
Affiliation(s)
- Fadi Rofo
- Department of Pharmacy, Uppsala University, 75124, Uppsala, Sweden
| | - Silvio R Meier
- Department of Public Health and Caring Sciences, Uppsala University, 75185, Uppsala, Sweden
| | | | - Jamie I Morrison
- Department of Pharmacy, Uppsala University, 75124, Uppsala, Sweden
| | - Alex Petrovic
- Department of Pharmacy, Uppsala University, 75124, Uppsala, Sweden
| | - Stina Syvänen
- Department of Public Health and Caring Sciences, Uppsala University, 75185, Uppsala, Sweden
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Uppsala University, 75185, Uppsala, Sweden
| | - Greta Hultqvist
- Department of Pharmacy, Uppsala University, 75124, Uppsala, Sweden.
| |
Collapse
|
14
|
Communication pathways bridge local and global conformations in an IgG4 antibody. Sci Rep 2021; 11:23197. [PMID: 34853348 PMCID: PMC8636491 DOI: 10.1038/s41598-021-02323-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/15/2021] [Indexed: 12/15/2022] Open
Abstract
The affinity of an antibody for its antigen is primarily determined by the specific sequence and structural arrangement of the complementarity-determining regions (CDRs). Recent evidence, however, points toward a nontrivial relation between the CDR and distal sites: variations in the binding strengths have been observed upon mutating residues separated from the paratope by several nanometers, thus suggesting the existence of a communication network within antibodies, whose extension and relevance might be deeper than insofar expected. In this work, we test this hypothesis by means of molecular dynamics (MD) simulations of the IgG4 monoclonal antibody pembrolizumab, an approved drug that targets the programmed cell death protein 1 (PD-1). The molecule is simulated in both the apo and holo states, totalling 4 μs of MD trajectory. The analysis of these simulations shows that the bound antibody explores a restricted range of conformations with respect to the apo one, and that the global conformation of the molecule correlates with that of the CDR. These results support the hypothesis that pembrolizumab featues a multi-scale hierarchy of intertwined global and local conformational changes. The analysis pipeline developed in this work is general, and it can help shed further light on the mechanistic aspects of antibody function.
Collapse
|
15
|
Tang Y, Cain P, Anguiano V, Shih JJ, Chai Q, Feng Y. Impact of IgG subclass on molecular properties of monoclonal antibodies. MAbs 2021; 13:1993768. [PMID: 34763607 PMCID: PMC8726687 DOI: 10.1080/19420862.2021.1993768] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Immunoglobulin G-based monoclonal antibodies (mAbs) have become a dominant class of biotherapeutics in recent decades. Approved antibodies are mainly of the subclasses IgG1, IgG2, and IgG4, as well as their derivatives. Over the decades, the selection of IgG subclass has frequently been based on the needs of Fc gamma receptor engagement and effector functions for the desired mechanism of action, while the effect on drug product developability has been less thoroughly characterized. One of the major reasons is the lack of systematic understanding of the impact of IgG subclass on the molecular properties. Several efforts have been made recently to compare molecular property differences among these IgG subclasses, but the conclusions from these studies are sometimes obscured by the interference from variable regions. To further establish mechanistic understandings, we conducted a systematic study by grafting three independent variable regions onto human IgG1, an IgG1 variant, IgG2, and an IgG4 variant constant domains and evaluating the impact of subclass and variable regions on their molecular properties. Structural and computational analysis revealed specific molecular features that potentially account for the differential behavior of the IgG subclasses observed experimentally. Our data indicate that IgG subclass plays a significant role on molecular properties, either through direct effects or via the interplay with the variable region, the IgG1 mAbs tend to have higher solubility than either IgG2 or IgG4 mAbs in a common pH 6 buffer matrix, and solution behavior relies heavily on the charge status of the antibody at the desirable pH.
Collapse
Affiliation(s)
- Yu Tang
- Pharmaceutical Development, Syndax Pharmaceuticals, Waltham, Massachusetts, USA
| | - Paul Cain
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, Indiana, USA
| | - Victor Anguiano
- Bioproduct Research & Development, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, Indiana, USA
| | - James J Shih
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, California, USA
| | - Qing Chai
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, California, USA
| | - Yiqing Feng
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, Indiana, USA
| |
Collapse
|
16
|
Martí D, Martín-Martínez E, Torras J, Bertran O, Turon P, Alemán C. In silico antibody engineering for SARS-CoV-2 detection. Comput Struct Biotechnol J 2021; 19:5525-5534. [PMID: 34642596 PMCID: PMC8496930 DOI: 10.1016/j.csbj.2021.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/06/2021] [Accepted: 10/06/2021] [Indexed: 12/11/2022] Open
Abstract
Engineered immunoglobulin-G molecules (IgGs) are of wide interest for the development of detection elements in protein-based biosensors with clinical applications. The strategy usually employed for the de novo design of such engineered IgGs consists on merging fragments of the three-dimensional structure of a native IgG, which is immobilized on the biosensor surface, and of an antibody with an exquisite target specificity and affinity. In this work conventional and accelerated classical molecular dynamics (cMD and aMD, respectively) simulations have been used to propose two IgG-like antibodies for COVID-19 detection. More specifically, the crystal structure of the IgG1 B12 antibody, which inactivates the human immunodeficiency virus-1, has been merged with the structure of the antibody CR3022 Fab tightly bounded to SARS-CoV-2 receptor-binding domain (RBD) and the structure of the S309 antibody Fab fragment complexed with SARS-CoV-2 RBD. The two constructed antibodies, named IgG1-CR3022 and IgG1-S309, respectively, have been immobilized on a stable gold surface through a linker. Analyses of the influence of both the merging strategy and the substrate on the stability of the two constructs indicate that the IgG1-S309 antibody better preserves the neutralizing structure than the IgG1-CR3022 one. Overall, results indicate that the IgG1-S309 is appropriated for the generation of antibody based sensors for COVID-19 diagnosis.
Collapse
Affiliation(s)
- Didac Martí
- Departament d'Enginyeria Química, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany, 10-14, Ed. I2, 08019 Barcelona, Spain
| | - Eduard Martín-Martínez
- Departament d'Enginyeria Química, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany, 10-14, Ed. I2, 08019 Barcelona, Spain
| | - Juan Torras
- Departament d'Enginyeria Química, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany, 10-14, Ed. I2, 08019 Barcelona, Spain.,Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, C/ Eduard Maristany, 10-14, 08019 Barcelona, Spain
| | - Oscar Bertran
- Departament de Física EETAC, Universitat Politècnica de Catalunya, c/ Esteve Terrades, 7, 08860 Castelldefels, Spain
| | - Pau Turon
- B. Braun Surgical, S.A.U. Carretera de Terrassa 121, 08191 Rubí (Barcelona), Spain
| | - Carlos Alemán
- Departament d'Enginyeria Química, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany, 10-14, Ed. I2, 08019 Barcelona, Spain.,Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, C/ Eduard Maristany, 10-14, 08019 Barcelona, Spain.,Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| |
Collapse
|
17
|
Rofo F, Buijs J, Falk R, Honek K, Lannfelt L, Lilja AM, Metzendorf NG, Gustavsson T, Sehlin D, Söderberg L, Hultqvist G. Novel multivalent design of a monoclonal antibody improves binding strength to soluble aggregates of amyloid beta. Transl Neurodegener 2021; 10:38. [PMID: 34579778 PMCID: PMC8477473 DOI: 10.1186/s40035-021-00258-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/14/2021] [Indexed: 11/30/2022] Open
Abstract
Background Amyloid-β (Aβ) immunotherapy is a promising therapeutic strategy in the fight against Alzheimer’s disease (AD). A number of monoclonal antibodies have entered clinical trials for AD. Some of them have failed due to the lack of efficacy or side-effects, two antibodies are currently in phase 3, and one has been approved by FDA. The soluble intermediate aggregated species of Aβ, termed oligomers and protofibrils, are believed to be key pathogenic forms, responsible for synaptic and neuronal degeneration in AD. Therefore, antibodies that can strongly and selectively bind to these soluble intermediate aggregates are of great diagnostic and therapeutic interest. Methods We designed and recombinantly produced a hexavalent antibody based on mAb158, an Aβ protofibril-selective antibody. The humanized version of mAb158, lecanemab (BAN2401), is currently in phase 3 clinical trials for the treatment of AD. The new designs involved recombinantly fusing single-chain fragment variables to the N-terminal ends of mAb158 antibody. Real-time interaction analysis with LigandTracer and surface plasmon resonance were used to evaluate the kinetic binding properties of the generated antibodies to Aβ protofibrils. Different ELISA setups were applied to demonstrate the binding strength of the hexavalent antibody to Aβ aggregates of different sizes. Finally, the ability of the antibodies to protect cells from Aβ-induced effects was evaluated by MTT assay. Results Using real-time interaction analysis with LigandTracer, the hexavalent design promoted a 40-times enhanced binding with avidity to protofibrils, and most of the added binding strength was attributed to the reduced rate of dissociation. Furthermore, ELISA experiments demonstrated that the hexavalent design also had strong binding to small oligomers, while retaining weak and intermediate binding to monomers and insoluble fibrils. The hexavalent antibody also reduced cell death induced by a mixture of soluble Aβ aggregates. Conclusion We provide a new antibody design with increased valency to promote binding avidity to an enhanced range of sizes of Aβ aggregates. This approach should be general and work for any aggregated protein or repetitive target. Supplementary Information The online version contains supplementary material available at 10.1186/s40035-021-00258-x.
Collapse
Affiliation(s)
- Fadi Rofo
- Protein Drug Design, Faculty of Pharmacy, Uppsala University, 75124, Uppsala, Sweden
| | - Jos Buijs
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185, Uppsala, Sweden.,Ridgeview Instruments, 75237, Uppsala, Sweden
| | | | - Ken Honek
- BioArctic AB, 11251, Stockholm, Sweden
| | - Lars Lannfelt
- BioArctic AB, 11251, Stockholm, Sweden.,Department of Public Health and Caring Sciences, Uppsala University, 75185, Uppsala, Sweden
| | | | - Nicole G Metzendorf
- Protein Drug Design, Faculty of Pharmacy, Uppsala University, 75124, Uppsala, Sweden
| | - Tobias Gustavsson
- Department of Public Health and Caring Sciences, Uppsala University, 75185, Uppsala, Sweden
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Uppsala University, 75185, Uppsala, Sweden
| | | | - Greta Hultqvist
- Protein Drug Design, Faculty of Pharmacy, Uppsala University, 75124, Uppsala, Sweden.
| |
Collapse
|
18
|
Chen Z, Qian Y, Song Y, Xu X, Tao L, Mussa N, Ghose S, Li ZJ. Design of next-generation therapeutic IgG4 with improved manufacturability and bioanalytical characteristics. MAbs 2021; 12:1829338. [PMID: 33044887 PMCID: PMC7577236 DOI: 10.1080/19420862.2020.1829338] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Manufacturability of immunoglobulin G4 (IgG4) antibodies from the Chemistry, Manufacture, and Controls (CMC) perspective has received little attention during early drug discovery. Despite the success of protein engineering in improving antibody biophysical properties, a clear gap still exists between rational design of IgG4 candidates and their manufacturing suitability. Here, we illustrate that undesirable two-peak elution profiles in cation-exchange chromatography are attributed to the S228P mutation (in IgG4 core-hinge region) intentionally designed to prevent Fab-arm exchange. A new scaffolding platform for engineering IgG4 antibodies amenable to bioprocessing and bioanalysis is proposed by introducing an “IgG1-like” single-point mutation in the hinge or CH1 region of IgG4S228P. This work offers insight into the design, discovery, and development of innovative therapeutic antibodies that are well suited for robust biomanufacturing and quality control.
Collapse
Affiliation(s)
- Zhiqiang Chen
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb Company , Devens, MA, USA
| | - Yueming Qian
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb Company , Devens, MA, USA
| | - Yuanli Song
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb Company , Devens, MA, USA
| | - Xuankuo Xu
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb Company , Devens, MA, USA
| | - Li Tao
- Biophysical Characterization, Global Product Development and Supply, Bristol Myers Squibb Company , New Brunswick, NJ, USA
| | - Nesredin Mussa
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb Company , Devens, MA, USA
| | - Sanchayita Ghose
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb Company , Devens, MA, USA
| | - Zheng Jian Li
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb Company , Devens, MA, USA
| |
Collapse
|
19
|
Jendroszek A, Kjaergaard M. Nanoscale spatial dependence of avidity in an IgG1 antibody. Sci Rep 2021; 11:12663. [PMID: 34135438 PMCID: PMC8209022 DOI: 10.1038/s41598-021-92280-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 06/04/2021] [Indexed: 11/16/2022] Open
Abstract
Antibodies are secreted proteins that are crucial to recognition of pathogens by the immune system and are also efficient pharmaceuticals. The affinity and specificity of target recognition can increase remarkably through avidity effects, when the antibody can bind a multivalent antigen through more than one epitope simultaneously. A key goal of antibody engineering is thus to optimize avidity, but little is known about the nanoscale spatial dependence of avidity in antibodies. Here, we develop a set of anti-parallel coiled-coils spanning from 7 to 20 nm and validate their structure using biophysical techniques. We use the coiled-coils to control the spacing between two epitopes, and measure how antigen spacing affects the stability of the bivalent antibody:antigen complex. We find a maximal avidity enhancement at a spacing of 13 nm. In contrast to recent studies, we find the avidity to be relatively insensitive to epitope spacing near the avidity maximum as long as it is within the spatial tolerance of the antibody. We thus only see a ~ twofold variation of avidity in the range from 7 to 20 nm. The coiled-coil systems developed here may prove a useful protein nanocaliper for profiling the spatial tolerance and avidity profile of bispecific antibodies.
Collapse
Affiliation(s)
- Agnieszka Jendroszek
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus, Denmark
| | - Magnus Kjaergaard
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark. .,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus, Denmark. .,Aarhus Institute of Advanced Studies (AIAS), Aarhus, Denmark. .,The Center for Proteins in Memory (PROMEMO), Aarhus, Denmark.
| |
Collapse
|
20
|
Hodge CD, Rosenberg DJ, Grob P, Wilamowski M, Joachimiak A, Hura GL, Hammel M. Rigid monoclonal antibodies improve detection of SARS-CoV-2 nucleocapsid protein. MAbs 2021; 13:1905978. [PMID: 33843452 PMCID: PMC8043170 DOI: 10.1080/19420862.2021.1905978] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/10/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
Monoclonal antibodies (mAbs) are the basis of treatments and diagnostics for pathogens and other biological phenomena. We conducted a structural characterization of mAbs against the N-terminal domain of nucleocapsid protein (NPNTD) from SARS-CoV-2 using small-angle X-ray scattering and transmission electron microscopy. Our solution-based results distinguished the mAbs' flexibility and how this flexibility affects the assembly of multiple mAbs on an antigen. By pairing two mAbs that bind different epitopes on the NPNTD, we show that flexible mAbs form a closed sandwich-like complex. With rigid mAbs, a juxtaposition of the antigen-binding fragments is prevented, enforcing a linear arrangement of the mAb pair, which facilitates further mAb polymerization. In a modified sandwich enzyme-linked immunosorbent assay, we show that rigid mAb-pairings with linear polymerization led to increased NPNTD detection sensitivity. These enhancements can expedite the development of more sensitive and selective antigen-detecting point-of-care lateral flow devices, which are critical for early diagnosis and epidemiological studies of SARS-CoV-2 and other pathogens.
Collapse
Affiliation(s)
- Curtis D. Hodge
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Daniel. J. Rosenberg
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Graduate Group in Biophysics, University of California, Berkeley, CA, USA
| | - Patricia Grob
- Howard Hughes Medical Institute, UC Berkeley, Berkeley, CA, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA, USA
| | - Mateusz Wilamowski
- Center for Structural Genomics of Infectious Diseases, Consortium for Advanced Science and Engineering, University of Chicago, Chicago, IL, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Andrzej Joachimiak
- Center for Structural Genomics of Infectious Diseases, Consortium for Advanced Science and Engineering, University of Chicago, Chicago, IL, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
- Structural Biology Center, X-ray Science Division, Argonne National Laboratory, Argonne, IL, USA
| | - Greg L. Hura
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Chemistry and Biochemistry Department, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Michal Hammel
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| |
Collapse
|
21
|
Bivalent antibody pliers inhibit β-tryptase by an allosteric mechanism dependent on the IgG hinge. Nat Commun 2020; 11:6435. [PMID: 33353951 PMCID: PMC7755903 DOI: 10.1038/s41467-020-20143-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
Human β-tryptase, a tetrameric trypsin-like serine protease, is an important mediator of allergic inflammatory responses in asthma. Antibodies generally inhibit proteases by blocking substrate access by binding to active sites or exosites or by allosteric modulation. The bivalency of IgG antibodies can increase potency via avidity, but has never been described as essential for activity. Here we report an inhibitory anti-tryptase IgG antibody with a bivalency-driven mechanism of action. Using biochemical and structural data, we determine that four Fabs simultaneously occupy four exosites on the β-tryptase tetramer, inducing allosteric changes at the small interface. In the presence of heparin, the monovalent Fab shows essentially no inhibition, whereas the bivalent IgG fully inhibits β-tryptase activity in a hinge-dependent manner. Our results suggest a model where the bivalent IgG acts akin to molecular pliers, pulling the tetramer apart into inactive β-tryptase monomers, and may provide an alternative strategy for antibody engineering. β-tryptases are responsible for most of the proteolytic activity during mast cell activation. Here, the authors develop β-tryptase-inhibiting antibodies and provide structural and biochemical evidence that the bivalency of the antibodies is a prerequisite for their inhibitory activity.
Collapse
|
22
|
Abdo AIK, Ngoh YY, Lew MH, Dass SA, Rahumatullah A, Noordin R, Tye GJ. Generation of human scFv-IgG1Fc antibodies for detection of lymphatic filarial recombinant antigens, BmR1 and BmSXP. Biotechnol Appl Biochem 2020; 69:70-76. [PMID: 33258152 DOI: 10.1002/bab.2082] [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: 06/15/2020] [Accepted: 11/22/2020] [Indexed: 11/08/2022]
Abstract
Lymphatic filariasis is a neglected parasitic disease that affects millions in tropical and subtropical countries and is caused by Wuchereria and Brugia species. Specific and sensitive detection methods are essential in mapping infected areas where rapid tests are needed to cover underdeveloped and remote regions, which facilitates eliminating the disease as a public health problem. A few commercialized rapid tests based on antigen or antibody detection are available, but the former only detects infection by Wuchereria species and cross-reacts with nonlymphatic filaria, whereas antibody detection might provide positive results of previous infection. Here, we report the production of three different recombinant immunoglobulin gamma (IgG)1 antibodies based on scFvs previously generated via human antibody phage display technology, that is, anti-BmR1 clone 4, anti-BmXSP clone 5B, and anti-BmXSP clone 2H2. The scFv sequences were cloned into a pCMV-IgG1 vector, then transfected into a HEK293F cell line. The generated antibodies were found to be able to bind to their respective targets even at relatively low concentration. Conjugation of Fc to scFv induces binder stability and provides multiple labeling sites for probes and signaling molecules that can be used in rapid tests.
Collapse
Affiliation(s)
- Ahmad Ismail Khaled Abdo
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Ying Yuan Ngoh
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Min Han Lew
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Sylvia Annabel Dass
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Anizah Rahumatullah
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Rahmah Noordin
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| |
Collapse
|
23
|
Stachowski TR, Snell ME, Snell EH. SAXS studies of X-ray induced disulfide bond damage: Engineering high-resolution insight from a low-resolution technique. PLoS One 2020; 15:e0239702. [PMID: 33201877 PMCID: PMC7671560 DOI: 10.1371/journal.pone.0239702] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/12/2020] [Indexed: 12/17/2022] Open
Abstract
A significant problem in biological X-ray crystallography is the radiation chemistry caused by the incident X-ray beam. This produces both global and site-specific damage. Site specific damage can misdirect the biological interpretation of the structural models produced. Cryo-cooling crystals has been successful in mitigating damage but not eliminating it altogether; however, cryo-cooling can be difficult in some cases and has also been shown to limit functionally relevant protein conformations. The doses used for X-ray crystallography are typically in the kilo-gray to mega-gray range. While disulfide bonds are among the most significantly affected species in proteins in the crystalline state at both cryogenic and higher temperatures, there is limited information on their response to low X-ray doses in solution, the details of which might inform biomedical applications of X-rays. In this work we engineered a protein that dimerizes through a susceptible disulfide bond to relate the radiation damage processes seen in cryo-cooled crystals to those closer to physiologic conditions. This approach enables a low-resolution technique, small angle X-ray scattering (SAXS), to detect and monitor a residue specific process. A dose dependent fragmentation of the engineered protein was seen that can be explained by a dimer to monomer transition through disulfide bond cleavage. This supports the crystallographically derived mechanism and demonstrates that results obtained crystallographically can be usefully extrapolated to physiologic conditions. Fragmentation was influenced by pH and the conformation of the dimer, providing information on mechanism and pointing to future routes for investigation and potential mitigation. The novel engineered protein approach to generate a large-scale change through a site-specific interaction represents a promising tool for advancing radiation damage studies under solution conditions.
Collapse
Affiliation(s)
- Timothy R. Stachowski
- Hauptman-Woodward Medical Research Institute, Buffalo, New York, United States of America
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States of America
| | - Mary E. Snell
- Hauptman-Woodward Medical Research Institute, Buffalo, New York, United States of America
| | - Edward H. Snell
- Hauptman-Woodward Medical Research Institute, Buffalo, New York, United States of America
- Department of Materials Design and Innovation, State University at New York at Buffalo, Buffalo, New York, United States of America
| |
Collapse
|
24
|
Taking the Hinge off: An Approach to Effector-Less Monoclonal Antibodies. Antibodies (Basel) 2020; 9:antib9040050. [PMID: 32977708 PMCID: PMC7709103 DOI: 10.3390/antib9040050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/05/2020] [Accepted: 08/03/2020] [Indexed: 12/25/2022] Open
Abstract
A variety of Fc domain engineering approaches for abrogating the effector functions of mAbs exists. To address some of the limitations of the current Fc domain silencing approaches, we are exploring a less commonly considered option which relies on the deletion of the hinge. Removal of the hinge domain in humanized IgG1 and IgG4 mAbs obliterates their ability to bind to activating human Fc gamma receptors I and IIIA, while leaving their ability to engage their target antigen intact. Deletion of the hinge also reduces binding to the Fc neonatal receptor, although Fc engineering allows partial recovery of affinity. Engineering of the CH3 domain, stabilizes hinge deleted IgG4s and prevents Fab arm exchange. The faster clearing properties together with the pacified Fc make modality of the hinge deleted mAb an appealing solution for therapeutic and diagnostic applications.
Collapse
|
25
|
Koneczny I. Update on IgG4-mediated autoimmune diseases: New insights and new family members. Autoimmun Rev 2020; 19:102646. [PMID: 32801046 DOI: 10.1016/j.autrev.2020.102646] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 03/08/2020] [Indexed: 12/23/2022]
Abstract
Antibodies of IgG4 subclass are exceptional players of the immune system, as they are considered to be immunologically inert and functionally monovalent, and as such may be part of classical tolerance mechanisms. IgG4 antibodies are found in a range of different diseases, including IgG4-related diseases, allergy, cancer, rheumatoid arthritis, helminth infection and IgG4 autoimmune diseases, where they may be pathogenic or protective. IgG4 autoimmune diseases are an emerging new group of diseases that are characterized by pathogenic, antigen-specific autoantibodies of IgG4 subclass, such as MuSK myasthenia gravis, pemphigus vulgaris and thrombotic thrombocytopenic purpura. The list of IgG4 autoantigens is rapidly growing and to date contains 29 candidate antigens. Interestingly, IgG4 autoimmune diseases are restricted to four distinct organs: 1) the central and peripheral nervous system, 2) the kidney, 3) the skin and mucous membranes and 4) the vascular system and soluble antigens in the blood circulation. The pathogenicity of IgG4 can be validated using our classification system, and is usually excerted by functional blocking of protein-protein interaction.
Collapse
Affiliation(s)
- Inga Koneczny
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Währingergürtel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
26
|
Jensen RK, Jabs F, Miehe M, Mølgaard B, Pfützner W, Möbs C, Spillner E, Andersen GR. Structure of intact IgE and the mechanism of ligelizumab revealed by electron microscopy. Allergy 2020; 75:1956-1965. [PMID: 32037590 DOI: 10.1111/all.14222] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/10/2020] [Accepted: 01/22/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND IgE is the central antibody isotype in TH2-biased immunity and allergic diseases. The structure of intact IgE and the impact of IgE-targeting molecules on IgE however remain elusive. In order to obtain insights into IgE biology and the clinical impact, we aimed for structure determination of IgE and the complex of IgE with the anti-IgE antibody ligelizumab. METHODS Structures of two distinct intact IgE with specificity for cross-reactive carbohydrate determinants and Der p 2 as well as complexes of ligelizumab-Fab with IgE and IgE Fc were assessed by negative stain electron microscopy and solution scattering. Inhibition of IgE binding and displacement of receptor-bound IgE were assessed using cellular assays, basophil activation testing and ELIFAB assays. RESULTS Our data reveal that the investigated IgE molecules share an overall rigid conformation. In contrast to the IgE Fc fragment, the IgE Fc in intact IgE is significantly less asymmetrically bent. The proximal and the distal Fabs are rigidly tethered to the Fc. Binding of ligelizumab to IgE in a 2:1 stoichiometry induces an extended and twofold symmetrical conformation of IgE, which retains a rigid Fab-Fc architecture. Analyses of effector cell activation revealed that ligelizumab inhibits IgE binding without displacing receptor-bound IgE. Together with an interference of CD23 binding, the data underline a functional activity similar to omalizumab. CONCLUSIONS Our data reveal the first structures of intact IgE suggesting that the IgE Fab is fixed relative to the Fc. Furthermore, we provide a structural rationale for the inhibitory mechanism of ligelizumab.
Collapse
Affiliation(s)
- Rasmus K. Jensen
- Department of Molecular Biology and Genetics Aarhus University Aarhus Denmark
| | - Frederic Jabs
- Immunological Biotechnology Department of Engineering Aarhus University Aarhus Denmark
| | - Michaela Miehe
- Immunological Biotechnology Department of Engineering Aarhus University Aarhus Denmark
| | - Brian Mølgaard
- Immunological Biotechnology Department of Engineering Aarhus University Aarhus Denmark
| | - Wolfgang Pfützner
- Clinical and Experimental Allergology, Department of Dermatology and Allergology Philipps-Universität Marburg Marburg Germany
| | - Christian Möbs
- Clinical and Experimental Allergology, Department of Dermatology and Allergology Philipps-Universität Marburg Marburg Germany
| | - Edzard Spillner
- Immunological Biotechnology Department of Engineering Aarhus University Aarhus Denmark
| | - Gregers R. Andersen
- Department of Molecular Biology and Genetics Aarhus University Aarhus Denmark
| |
Collapse
|
27
|
Disentangling the role of solvent polarity and protein solvation in folding and self-assembly of α-lactalbumin. J Colloid Interface Sci 2020; 561:749-761. [DOI: 10.1016/j.jcis.2019.11.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/29/2019] [Accepted: 11/14/2019] [Indexed: 12/31/2022]
|
28
|
Reese HR, Shanahan CC, Proulx C, Menegatti S. Peptide science: A "rule model" for new generations of peptidomimetics. Acta Biomater 2020; 102:35-74. [PMID: 31698048 DOI: 10.1016/j.actbio.2019.10.045] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/17/2019] [Accepted: 10/30/2019] [Indexed: 02/07/2023]
Abstract
Peptides have been heavily investigated for their biocompatible and bioactive properties. Though a wide array of functionalities can be introduced by varying the amino acid sequence or by structural constraints, properties such as proteolytic stability, catalytic activity, and phase behavior in solution are difficult or impossible to impart upon naturally occurring α-L-peptides. To this end, sequence-controlled peptidomimetics exhibit new folds, morphologies, and chemical modifications that create new structures and functions. The study of these new classes of polymers, especially α-peptoids, has been highly influenced by the analysis, computational, and design techniques developed for peptides. This review examines techniques to determine primary, secondary, and tertiary structure of peptides, and how they have been adapted to investigate peptoid structure. Computational models developed for peptides have been modified to predict the morphologies of peptoids and have increased in accuracy in recent years. The combination of in vitro and in silico techniques have led to secondary and tertiary structure design principles that mirror those for peptides. We then examine several important developments in peptoid applications inspired by peptides such as pharmaceuticals, catalysis, and protein-binding. A brief survey of alternative backbone structures and research investigating these peptidomimetics shows how the advancement of peptide and peptoid science has influenced the growth of numerous fields of study. As peptide, peptoid, and other peptidomimetic studies continue to advance, we will expect to see higher throughput structural analyses, greater computational accuracy and functionality, and wider application space that can improve human health, solve environmental challenges, and meet industrial needs. STATEMENT OF SIGNIFICANCE: Many historical, chemical, and functional relations draw a thread connecting peptides to their recent cognates, the "peptidomimetics". This review presents a comprehensive survey of this field by highlighting the width and relevance of these familial connections. In the first section, we examine the experimental and computational techniques originally developed for peptides and their morphing into a broader analytical and predictive toolbox. The second section presents an excursus of the structures and properties of prominent peptidomimetics, and how the expansion of the chemical and structural diversity has returned new exciting properties. The third section presents an overview of technological applications and new families of peptidomimetics. As the field grows, new compounds emerge with clear potential in medicine and advanced manufacturing.
Collapse
|
29
|
Abstract
In vertebrates, immunoglobulins (Igs), commonly known as antibodies, play an integral role in the armamentarium of immune defense against various pathogens. After an antigenic challenge, antibodies are secreted by differentiated B cells called plasma cells. Antibodies have two predominant roles that involve specific binding to antigens to launch an immune response, along with activation of other components of the immune system to fight pathogens. The ability of immunoglobulins to fight against innumerable and diverse pathogens lies in their intrinsic ability to discriminate between different antigens. Due to this specificity and high affinity for their antigens, antibodies have been a valuable and indispensable tool in research, diagnostics and therapy. Although seemingly a simple maneuver, the association between an antibody and its antigen, to make an antigen-antibody complex, is comprised of myriads of non-covalent interactions. Amino acid residues on the antigen binding site, the epitope, and on the antibody binding site, the paratope, intimately contribute to the energetics needed for the antigen-antibody complex stability. Structural biology methods to study antigen-antibody complexes are extremely valuable tools to visualize antigen-antibody interactions in detail; this helps to elucidate the basis of molecular recognition between an antibody and its specific antigen. The main scope of this chapter is to discuss the structure and function of different classes of antibodies and the various aspects of antigen-antibody interactions including antigen-antibody interfaces-with a special focus on paratopes, complementarity determining regions (CDRs) and other non-CDR residues important for antigen binding and recognition. Herein, we also discuss methods used to study antigen-antibody complexes, antigen recognition by antibodies, types of antigens in complexes, and how antigen-antibody complexes play a role in modern day medicine and human health. Understanding the molecular basis of antigen binding and recognition by antibodies helps to facilitate the production of better and more potent antibodies for immunotherapy, vaccines and various other applications.
Collapse
Affiliation(s)
- A Brenda Kapingidza
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, USA
| | - Krzysztof Kowal
- Department of Allergology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
- Department of Experimental Allergology and Immunology, Medical University of Bialystok, Bialystok, Poland
| | - Maksymilian Chruszcz
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, USA.
| |
Collapse
|
30
|
Law-Hine D, Rudiuk S, Bonestebe A, Ienco R, Huille S, Tribet C. Distinctive Low-Resolution Structural Features of Dimers of Antibody-Drug Conjugates and Parent Antibody Determined by Small-Angle X-ray Scattering. Mol Pharm 2019; 16:4902-4912. [PMID: 31618040 DOI: 10.1021/acs.molpharmaceut.9b00792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Structural features of lysine-conjugated antibody-drug conjugate (ADC) from humanized IgG1 were studied by small-angle X-ray scattering (SAXS). As the physicochemical properties of the cytotoxic drug (payload) and linker may impact the conformational and colloidal stabilities of the conjugated monoclonal antibody (mAb), it is essential to characterize how the conjugation may affect the overall higher order structure and therefore the physical stability and integrity of the ADCs upon storage conditions. Here, the ADC monomer and aggregates generated upon thermal stress were analyzed by high performance liquid chromatography coupled to SAXS with a particular focus on the fraction of dimers (3-10% depending on the storage conditions at 25 and 40 °C). In addition to average parameters such as radius of gyration, molecular weight, and maximal end-to-end distance, the structural information obtained from SAXS patterns were visualized as a low-resolution average envelope of both monomers and dimers (implementation of two methods: ab initio reconstruction and modeling Fab and Fc as rigid bodies with a flexible hinge). We showed that the monomer envelope of the ADC was similar to the corresponding (nonconjugated) parent monoclonal antibody (mAb). ADC dimers appeared more compact and less polydisperse than the dimers of mAb, which was also confirmed by atomic force microscopy. The generated envelopes of the mAb dimers suggest elongated structures with one or few inter-mAb contacts at the outermost region of Fab or Fc domains. The structural features of ADC dimers are independent of the tested pH buffering system (pH 5.0/acetate and pH 6.0/histidine with or without NaCl) and characterized by multiple, tighter contacts between the Fab and Fc domains and distortion of the monomer native shape. Results from the SAXS structural study show in the present case that conjugation has favored innermost inter-ADC contacts in the dimer, which differ from the inter-mAb ones. In general, it is likely that many parameters affect inter-ADC association, including the chemical nature of linkers and drugs, degree of conjugation, conjugation sites, etc. Making a qualitative difference between mAb and ADC dimers as a function of these parameters can help point to the presence of tight associations that must be abolished in protein drug formulations.
Collapse
Affiliation(s)
- Didier Law-Hine
- Département de Chimie, PASTEUR, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris 75005, France
| | - Sergii Rudiuk
- Département de Chimie, PASTEUR, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris 75005, France
| | - Audrey Bonestebe
- Biologics Formulation & Process Development, Biologics Development, SANOFI R&D, 13 quai Jules Guesde- BP 14, Vitry-sur-Seine 94403, France
| | - Romain Ienco
- Biologics Formulation & Process Development, Biologics Development, SANOFI R&D, 13 quai Jules Guesde- BP 14, Vitry-sur-Seine 94403, France
| | - Sylvain Huille
- Biologics Formulation & Process Development, Biologics Development, SANOFI R&D, 13 quai Jules Guesde- BP 14, Vitry-sur-Seine 94403, France
| | - Christophe Tribet
- Département de Chimie, PASTEUR, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris 75005, France
| |
Collapse
|
31
|
Wright DW, Elliston ELK, Hui GK, Perkins SJ. Atomistic Modeling of Scattering Curves for Human IgG1/4 Reveals New Structure-Function Insights. Biophys J 2019; 117:2101-2119. [PMID: 31708160 PMCID: PMC6895691 DOI: 10.1016/j.bpj.2019.10.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/03/2019] [Accepted: 10/18/2019] [Indexed: 11/11/2022] Open
Abstract
Small angle x-ray and neutron scattering are techniques that give solution structures for large macromolecules. The creation of physically realistic atomistic models from known high-resolution structures to determine joint x-ray and neutron scattering best-fit structures offers a, to our knowledge, new method that significantly enhances the utility of scattering. To validate this approach, we determined scattering curves for two human antibody subclasses, immunoglobulin G (IgG) 1 and IgG4, on five different x-ray and neutron instruments to show that these were reproducible, then we modeled these by Monte Carlo simulations. The two antibodies have different hinge lengths that connect their antigen-binding Fab and effector-binding Fc regions. Starting from 231,492 and 190,437 acceptable conformations for IgG1 and IgG4, respectively, joint x-ray and neutron scattering curve fits gave low goodness-of-fit R factors for 28 IgG1 and 2748 IgG4 structures that satisfied the disulphide connectivity in their hinges. These joint best-fit structures showed that the best-fit IgG1 models had a greater separation between the centers of their Fab regions than those for IgG4, in agreement with their hinge lengths of 15 and 12 residues, respectively. The resulting asymmetric IgG1 solution structures resembled its crystal structure. Both symmetric and asymmetric solution structures were determined for IgG4. Docking simulations with our best-fit IgG4 structures showed greater steric clashes with its receptor to explain its weaker FcγRI receptor binding compared to our best-fit IgG1 structures with fewer clashes and stronger receptor binding. Compared to earlier approaches for fitting molecular antibody structures by solution scattering, we conclude that this joint fit approach based on x-ray and neutron scattering data, combined with Monte Carlo simulations, significantly improved our understanding of antibody solution structures. The atomistic nature of the output extended our understanding of known functional differences in Fc receptor binding between IgG1 and IgG4.
Collapse
Affiliation(s)
- David W Wright
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, United Kingdom
| | - Emma L K Elliston
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, United Kingdom
| | - Gar Kay Hui
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, United Kingdom
| | - Stephen J Perkins
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, United Kingdom.
| |
Collapse
|
32
|
Jaccoulet E, Daniel T, Prognon P, Caudron E. Forced Degradation of Monoclonal Antibodies After Compounding: Impact on Routine Hospital Quality Control. J Pharm Sci 2019; 108:3252-3261. [DOI: 10.1016/j.xphs.2019.06.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 05/16/2019] [Accepted: 06/04/2019] [Indexed: 10/26/2022]
|
33
|
Liu X, Zhao Y, Shi H, Zhang Y, Yin X, Liu M, Zhang H, He Y, Lu B, Jin T, Li F. Human immunoglobulin G hinge regulates agonistic anti-CD40 immunostimulatory and antitumour activities through biophysical flexibility. Nat Commun 2019; 10:4206. [PMID: 31562320 PMCID: PMC6765011 DOI: 10.1038/s41467-019-12097-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 08/20/2019] [Indexed: 12/31/2022] Open
Abstract
Human immunoglobulin G (IgG) agonistic antibodies targeting costimulatory immunoreceptors represent promising cancer immunotherapies yet to be developed. Whether, and how, human IgG hinge and Fc impact on their agonistic functions have been disputed. Here, we show that different natural human IgGs confer divergent agonistic anti-CD40 immunostimulatory and antitumour activities in FcγR-humanized mice, including inactive IgG3 and superior IgG2. This divergence is primarily due to their CH1-hinges despite all human IgGs requiring Fc-FcγR binding for optimal agonistic activities. Unexpectedly, biophysical flexibility of these CH1-hinges inversely correlates with, and can modulate, their agonistic potency. Furthermore, IgG Fcs optimized for selective FcγR binding synergize with and still require IgG hinge, selected for rigidity, to confer improved anti-CD40 immunostimulatory and antitumour activities. These findings highlight the importance of both hinge rigidity and selective FcγR binding in antibody agonistic function, and the need for newer strategies to modulate antibody agonism for improved clinical application. Conserved regions of the antibody molecule impact its downstream biological effects. Here the authors show that a rigid hinge conformation increases the agonistic activity of CD40 and DR5 antibodies, distinctly from FcγR-binding, suggesting that the hinge and FcR binding regions can be separately modified to optimize therapies.
Collapse
Affiliation(s)
- Xiaobo Liu
- Shanghai Institute of Immunology, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yingjie Zhao
- Shanghai Institute of Immunology, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Huan Shi
- Shanghai Institute of Immunology, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yan Zhang
- Shanghai Institute of Immunology, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xueying Yin
- Laboratory of Structural Immunology, CAS Key Laboratory of Innate Immunity and Chronic Disease, CAS Center for Excellence in Molecular Cell Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | | | | | - Yongning He
- State Key Laboratory of Molecular Biology, National Center for Protein Science, Shanghai Science Research Center, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Boxun Lu
- State Key Laboratory of Genetic Engineering, Department of Biophysics, School of Life Sciences, Fudan University, Shanghai, China
| | - Tengchuan Jin
- Laboratory of Structural Immunology, CAS Key Laboratory of Innate Immunity and Chronic Disease, CAS Center for Excellence in Molecular Cell Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Fubin Li
- Shanghai Institute of Immunology, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Collaborative Innovation Center of Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
34
|
Hui GK, Gardener AD, Begum H, Eldrid C, Thalassinos K, Gor J, Perkins SJ. The solution structure of the human IgG2 subclass is distinct from those for human IgG1 and IgG4 providing an explanation for their discrete functions. J Biol Chem 2019; 294:10789-10806. [PMID: 31088911 PMCID: PMC6635440 DOI: 10.1074/jbc.ra118.007134] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 05/03/2019] [Indexed: 11/06/2022] Open
Abstract
Human IgG2 antibody displays distinct therapeutically-useful properties compared with the IgG1, IgG3, and IgG4 antibody subclasses. IgG2 is the second most abundant IgG subclass, being able to bind human FcγRII/FcγRIII but not to FcγRI or complement C1q. Structural information on IgG2 is limited by the absence of a full-length crystal structure for this. To this end, we determined the solution structure of human myeloma IgG2 by atomistic X-ray and neutron-scattering modeling. Analytical ultracentrifugation disclosed that IgG2 is monomeric with a sedimentation coefficient (s20, w0) of 7.2 S. IgG2 dimer formation was ≤5% and independent of the buffer conditions. Small-angle X-ray scattering in a range of NaCl concentrations and in light and heavy water revealed that the X-ray radius of gyration (Rg ) is 5.2-5.4 nm, after allowing for radiation damage at higher concentrations, and that the neutron Rg value of 5.0 nm remained unchanged in all conditions. The X-ray and neutron distance distribution curves (P(r)) revealed two peaks, M1 and M2, that were unchanged in different buffers. The creation of >123,000 physically-realistic atomistic models by Monte Carlo simulations for joint X-ray and neutron-scattering curve fits, constrained by the requirement of correct disulfide bridges in the hinge, resulted in the determination of symmetric Y-shaped IgG2 structures. These molecular structures were distinct from those for asymmetric IgG1 and asymmetric and symmetric IgG4 and were attributable to the four hinge disulfides. Our IgG2 structures rationalize the existence of the human IgG1, IgG2, and IgG4 subclasses and explain the receptor-binding functions of IgG2.
Collapse
Affiliation(s)
- Gar Kay Hui
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, United Kingdom
| | - Antoni D Gardener
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, United Kingdom
| | - Halima Begum
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, United Kingdom
| | - Charles Eldrid
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, United Kingdom
| | - Konstantinos Thalassinos
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, United Kingdom; Institute of Structural and Molecular Biology, Birkbeck College, University of London, London, WC1E 7HX, United Kingdom
| | - Jayesh Gor
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, United Kingdom
| | - Stephen J Perkins
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, United Kingdom.
| |
Collapse
|
35
|
Yanaka S, Yogo R, Inoue R, Sugiyama M, Itoh SG, Okumura H, Miyanoiri Y, Yagi H, Satoh T, Yamaguchi T, Kato K. Dynamic Views of the Fc Region of Immunoglobulin G Provided by Experimental and Computational Observations. Antibodies (Basel) 2019; 8:antib8030039. [PMID: 31544845 PMCID: PMC6784063 DOI: 10.3390/antib8030039] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/08/2019] [Accepted: 06/12/2019] [Indexed: 01/08/2023] Open
Abstract
The Fc portion of immunoglobulin G (IgG) is a horseshoe-shaped homodimer, which interacts with various effector proteins, including Fcγ receptors (FcγRs). These interactions are critically dependent on the pair of N-glycans packed between the two CH2 domains. Fucosylation of these N-glycans negatively affects human IgG1-FcγRIIIa interaction. The IgG1-Fc crystal structures mostly exhibit asymmetric quaternary conformations with divergent orientations of CH2 with respect to CH3. We aimed to provide dynamic views of IgG1-Fc by performing long-timescale molecular dynamics (MD) simulations, which were experimentally validated by small-angle X-ray scattering and nuclear magnetic resonance spectroscopy. Our simulation results indicated that the dynamic conformational ensembles of Fc encompass most of the previously reported crystal structures determined in both free and complex forms, although the major Fc conformers in solution exhibited almost symmetric, stouter quaternary structures, unlike the crystal structures. Furthermore, the MD simulations suggested that the N-glycans restrict the motional freedom of CH2 and endow quaternary-structure plasticity through multiple intramolecular interaction networks. Moreover, the fucosylation of these N-glycans restricts the conformational freedom of the proximal tyrosine residue of functional importance, thereby precluding its interaction with FcγRIIIa. The dynamic views of Fc will provide opportunities to control the IgG interactions for developing therapeutic antibodies.
Collapse
Affiliation(s)
- Saeko Yanaka
- Exploratory Research Center on Life and Living Systems (ExCELLS) and Institute for Molecular Science (IMS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
- Department of Functional Molecular Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan
| | - Rina Yogo
- Exploratory Research Center on Life and Living Systems (ExCELLS) and Institute for Molecular Science (IMS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Rintaro Inoue
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, 2-1010 Asashiro-Nishi, Kumatori, Osaka 590-0494, Japan
| | - Masaaki Sugiyama
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, 2-1010 Asashiro-Nishi, Kumatori, Osaka 590-0494, Japan
| | - Satoru G Itoh
- Exploratory Research Center on Life and Living Systems (ExCELLS) and Institute for Molecular Science (IMS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
- Department of Structural Molecular Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Hisashi Okumura
- Exploratory Research Center on Life and Living Systems (ExCELLS) and Institute for Molecular Science (IMS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
- Department of Structural Molecular Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Yohei Miyanoiri
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hirokazu Yagi
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Tadashi Satoh
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Takumi Yamaguchi
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
- School of Materials Science, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi 923-1292, Japan
| | - Koichi Kato
- Exploratory Research Center on Life and Living Systems (ExCELLS) and Institute for Molecular Science (IMS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan.
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan.
- Department of Functional Molecular Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan.
| |
Collapse
|
36
|
Kapelski S, Cleiren E, Attar RM, Philippar U, Häsler J, Chiu ML. Influence of the bispecific antibody IgG subclass on T cell redirection. MAbs 2019; 11:1012-1024. [PMID: 31242061 PMCID: PMC6748600 DOI: 10.1080/19420862.2019.1624464] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/03/2019] [Accepted: 05/23/2019] [Indexed: 01/07/2023] Open
Abstract
T cell redirection mediated by bispecific antibodies (BsAbs) is a promising cancer therapy. Dual antigen binding is necessary for potent T cell redirection and is influenced by the structural characteristics of a BsAb, which are dependent on its IgG subclass. In this study, model BsAbs targeting CD19xCD3 were generated in variants of IgG1, IgG2, and IgG4 carrying Fc mutations that reduce FcγR interaction, and two chimeric IgG subclasses termed IgG1:2 and IgG4:2, in which the IgG1- or IgG4-F(ab)2 are grafted on an IgG2 Fc. Molecules containing an IgG2 or IgG4-F(ab)2 domain were confirmed to be the most structurally compact molecules. All BsAbs were shown to bind both of their target proteins (and corresponding cells) equally well. However, CD19xCD3 IgG2 did not bind both antigens simultaneously as measured by the absence of cellular clustering of T cells with target cells. This translated to a reduced potency of IgG2 BsAbs in T-cell redirection assays. The activity of IgG2 BsAbs was fully restored in the chimeric subclasses IgG4:2 and IgG1:2. This confirmed the major contribution of the F(ab)2 region to the BsAb's functional activity and demonstrated that function of BsAbs can be modulated by engineering molecules combining different Fc and F(ab)2 domains. Abbreviations: ADCC: Antibody-dependent cellular cytotoxicity; AlphaScreenTM: Amplified Luminescent Proximity Homogeneous Assay Screening; ANOVA: Analysis of variance; BiTE: bispecific T-cell engager; BSA: bovine serum albumin; BsAb: bispecific antibody; cFAE: controlled Fab-arm exchange; CDC: complement-dependent cellular cytotoxicity; CIEX: cation-exchange; CIR: chimeric immune receptor; DPBS: Dulbecco's phosphate-buffered saline; EC50 value: effective concentration to reach half-maximum effect; EGFR: epidermal growth factor receptor; EI: expansion index (RAt=x/RAt=0); FACS: fluorescence-activated cell sorting; FVD: fixable viability dye; HI-HPLC: hydrophobic interaction HPLC; HI-FBS: heat-inactivated fetal bovine serum; HPLC: high-pressure liquid chromatography; IC50 value: effective concentration to reach half-maximum inhibition; IQ: Inhibition Quotient; IS: immunological synapse; MES: 2-(N-morpholino)ethanesulfonic acid; R-PE: recombinant phycoerythrin; RA: red area in μm2/well; RD: receptor density; RFP: red fluorescent protein; Rg: radius of gyration; RSV: respiratory syncytial virus; SAXS: small-angle x-ray scattering; scFv: single-chain variable fragment; SD: standard deviation; SPR: surface plasmon resonance; WT: wild-type.
Collapse
Affiliation(s)
- Stephanie Kapelski
- Biologics Discovery, Janssen BioTherapeutics, Janssen Research and Development, Beerse, Belgium
- Oncology Biology & Discovery, Janssen Research and Development, Beerse, Belgium
| | - Erna Cleiren
- Former Discovery Sciences, LD-Screening BE, Janssen Research and Development, Beerse, Belgium
- Charles River Laboratories, Beerse, Belgium
| | - Ricardo M. Attar
- Oncology Biology & Discovery, Janssen Research and Development, Spring House, PA,USA
| | - Ulrike Philippar
- Oncology Biology & Discovery, Janssen Research and Development, Beerse, Belgium
| | - Julien Häsler
- Biologics Discovery, Janssen BioTherapeutics, Janssen Research and Development, Beerse, Belgium
| | - Mark L. Chiu
- BioTherapeutics Analytical Development, Discovery, Product Development & Supply, Janssen Research and Development, Malvern, PA, USA
| |
Collapse
|
37
|
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: 18] [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
|
38
|
Shaw A, Hoffecker IT, Smyrlaki I, Rosa J, Grevys A, Bratlie D, Sandlie I, Michaelsen TE, Andersen JT, Högberg B. Binding to nanopatterned antigens is dominated by the spatial tolerance of antibodies. NATURE NANOTECHNOLOGY 2019; 14:184-190. [PMID: 30643273 PMCID: PMC6420075 DOI: 10.1038/s41565-018-0336-3] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 11/21/2018] [Indexed: 05/17/2023]
Abstract
Although repetitive patterns of antigens are crucial for certain immune responses, an understanding of how antibodies bind and dynamically interact with various spatial arrangements of molecules is lacking. Hence, we introduced a new method in which molecularly precise nanoscale patterns of antigens are displayed using DNA origami and immobilized in a surface plasmon resonance set-up. Using antibodies with identical antigen-binding domains, we found that all the subclasses and isotypes studied bind bivalently to two antigens separated at distances that range from 3 to 17 nm. The binding affinities of these antibodies change with the antigen distances, with a distinct preference for antigens separated by approximately 16 nm, and considerable differences in spatial tolerance exist between IgM and IgG and between low- and high-affinity antibodies.
Collapse
Affiliation(s)
- Alan Shaw
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ian T Hoffecker
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ioanna Smyrlaki
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Joao Rosa
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Algirdas Grevys
- Centre for Immune Regulation (CIR), Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- CIR, Department of Biosciences, University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Diane Bratlie
- Department of Infectious Disease Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Inger Sandlie
- Centre for Immune Regulation (CIR), Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- CIR, Department of Biosciences, University of Oslo, Oslo, Norway
| | - Terje Einar Michaelsen
- Department of Infectious Disease Immunology, Norwegian Institute of Public Health, Oslo, Norway
- School of Pharmacy, University of Oslo, Oslo, Norway
| | - Jan Terje Andersen
- Centre for Immune Regulation (CIR), Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Björn Högberg
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
39
|
Hansen K, Lau AM, Giles K, McDonnell JM, Struwe WB, Sutton BJ, Politis A. A Mass-Spectrometry-Based Modelling Workflow for Accurate Prediction of IgG Antibody Conformations in the Gas Phase. Angew Chem Int Ed Engl 2018; 57:17194-17199. [PMID: 30408305 PMCID: PMC6392142 DOI: 10.1002/anie.201812018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Indexed: 11/09/2022]
Abstract
Immunoglobulins are biomolecules involved in defence against foreign substances. Flexibility is key to their functional properties in relation to antigen binding and receptor interactions. We have developed an integrative strategy combining ion mobility mass spectrometry (IM-MS) with molecular modelling to study the conformational dynamics of human IgG antibodies. Predictive models of all four human IgG subclasses were assembled and their dynamics sampled in the transition from extended to collapsed state during IM-MS. Our data imply that this collapse of IgG antibodies is related to their intrinsic structural features, including Fab arm flexibility, collapse towards the Fc region, and the length of their hinge regions. The workflow presented here provides an accurate structural representation in good agreement with the observed collision cross section for these flexible IgG molecules. These results have implications for studying other nonglobular flexible proteins.
Collapse
Affiliation(s)
- Kjetil Hansen
- Department of ChemistryKing's College London7 Trinity StreetLondonSE1 1DBUK
| | - Andy M. Lau
- Department of ChemistryKing's College London7 Trinity StreetLondonSE1 1DBUK
| | | | | | | | - Brian J. Sutton
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonUK
| | - Argyris Politis
- Department of ChemistryKing's College London7 Trinity StreetLondonSE1 1DBUK
| |
Collapse
|
40
|
Hansen K, Lau AM, Giles K, McDonnell JM, Struwe WB, Sutton BJ, Politis A. A Mass‐Spectrometry‐Based Modelling Workflow for Accurate Prediction of IgG Antibody Conformations in the Gas Phase. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201812018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Kjetil Hansen
- Department of Chemistry King's College London 7 Trinity Street London SE1 1DB UK
| | - Andy M. Lau
- Department of Chemistry King's College London 7 Trinity Street London SE1 1DB UK
| | - Kevin Giles
- Waters Corp. Stamford Road Wilmslow SK9 4AX UK
| | - James M. McDonnell
- Randall Centre for Cell and Molecular Biophysics King's College London UK
| | | | - Brian J. Sutton
- Randall Centre for Cell and Molecular Biophysics King's College London UK
| | - Argyris Politis
- Department of Chemistry King's College London 7 Trinity Street London SE1 1DB UK
| |
Collapse
|
41
|
Datta-Mannan A, Choi H, Stokell D, Tang J, Murphy A, Wrobleski A, Feng Y. The Properties of Cysteine-Conjugated Antibody-Drug Conjugates Are Impacted by the IgG Subclass. AAPS JOURNAL 2018; 20:103. [DOI: 10.1208/s12248-018-0263-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 09/06/2018] [Indexed: 01/11/2023]
|
42
|
Sutton EJ, Bradshaw RT, Orr CM, Frendéus B, Larsson G, Teige I, Cragg MS, Tews I, Essex JW. Evaluating Anti-CD32b F(ab) Conformation Using Molecular Dynamics and Small-Angle X-Ray Scattering. Biophys J 2018; 115:289-299. [PMID: 30021105 PMCID: PMC6050753 DOI: 10.1016/j.bpj.2018.03.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 02/16/2018] [Accepted: 03/27/2018] [Indexed: 11/04/2022] Open
Abstract
Complementary strategies of small-angle x-ray scattering (SAXS) and crystallographic analysis are often used to determine atomistic three-dimensional models of macromolecules and their variability in solution. This combination of techniques is particularly valuable when applied to macromolecular complexes to detect changes within the individual binding partners. Here, we determine the x-ray crystallographic structure of a F(ab) fragment in complex with CD32b, the only inhibitory Fc-γ receptor in humans, and compare the structure of the F(ab) from the crystal complex to SAXS data for the F(ab) alone in solution. We investigate changes in F(ab) structure by predicting theoretical scattering profiles for atomistic structures extracted from molecular dynamics (MD) simulations of the F(ab) and assessing the agreement of these structures to our experimental SAXS data. Through principal component analysis, we are able to extract principal motions observed during the MD trajectory and evaluate the influence of these motions on the agreement of structures to the F(ab) SAXS data. Changes in the F(ab) elbow angle were found to be important to reach agreement with the experimental data; however, further discrepancies were apparent between our F(ab) structure from the crystal complex and SAXS data. By analyzing multiple MD structures observed in similar regions of the principal component analysis, we were able to pinpoint these discrepancies to a specific loop region in the F(ab) heavy chain. This method, therefore, not only allows determination of global changes but also allows identification of localized motions important for determining the agreement between atomistic structures and SAXS data. In this particular case, the findings allowed us to discount the hypothesis that structural changes were induced upon complex formation, a significant find informing the drug development process. The methodology described here is generally applicable to deconvolute global and local changes of macromolecular structures and is well suited to other systems.
Collapse
Affiliation(s)
- Emma J Sutton
- Antibody & Vaccine Group, Cancer Sciences Unit, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom; Department of Chemistry, University of Southampton, Highfield Campus, Southampton, United Kingdom; Department of Biological Sciences, Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton, United Kingdom
| | - Richard T Bradshaw
- Department of Chemistry, University of Southampton, Highfield Campus, Southampton, United Kingdom
| | - Christian M Orr
- Antibody & Vaccine Group, Cancer Sciences Unit, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom; Department of Biological Sciences, Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton, United Kingdom
| | | | | | | | - Mark S Cragg
- Antibody & Vaccine Group, Cancer Sciences Unit, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Ivo Tews
- Department of Biological Sciences, Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton, United Kingdom
| | - Jonathan W Essex
- Department of Chemistry, University of Southampton, Highfield Campus, Southampton, United Kingdom.
| |
Collapse
|
43
|
Castellanos MM, Howell SC, Gallagher DT, Curtis JE. Characterization of the NISTmAb Reference Material using small-angle scattering and molecular simulation. Anal Bioanal Chem 2018; 410:2141-2159. [DOI: 10.1007/s00216-018-0868-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/11/2017] [Accepted: 01/10/2018] [Indexed: 12/12/2022]
|
44
|
Castellanos MM, Snyder JA, Lee M, Chakravarthy S, Clark NJ, McAuley A, Curtis JE. Characterization of Monoclonal Antibody-Protein Antigen Complexes Using Small-Angle Scattering and Molecular Modeling. Antibodies (Basel) 2017; 6:25. [PMID: 30364605 PMCID: PMC6197476 DOI: 10.3390/antib6040025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2017] [Indexed: 01/01/2023] Open
Abstract
The determination of monoclonal antibody interactions with protein antigens in solution can lead to important insights guiding physical characterization and molecular engineering of therapeutic targets. We used small-angle scattering (SAS) combined with size-exclusion multi-angle light scattering high-performance liquid chromatography to obtain monodisperse samples with defined stoichiometry to study an anti-streptavidin monoclonal antibody interacting with tetrameric streptavidin. Ensembles of structures with both monodentate and bidentate antibody-antigen complexes were generated using molecular docking protocols and molecular simulations. By comparing theoretical SAS profiles to the experimental data it was determined that the primary component(s) were compact monodentate and/or bidentate complexes. SAS profiles of extended monodentate complexes were not consistent with the experimental data. These results highlight the capability for determining the shape of monoclonal antibody-antigen complexes in solution using SAS data and physics-based molecular modeling.
Collapse
Affiliation(s)
- Maria Monica Castellanos
- NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Mail Stop 6102, Gaithersburg, MD 20899, USA; (M.M.C.); (J.A.S.); (M.L.)
- Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Drive, Rockville, MD 20850, USA
| | - James A. Snyder
- NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Mail Stop 6102, Gaithersburg, MD 20899, USA; (M.M.C.); (J.A.S.); (M.L.)
| | - Melody Lee
- NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Mail Stop 6102, Gaithersburg, MD 20899, USA; (M.M.C.); (J.A.S.); (M.L.)
| | - Srinivas Chakravarthy
- Biophysics Collaborative Access Team-Sector 18ID, Illinois Institute of Technology, Advanced Photon Source, Argonne National Laboratory, Lemont, IL 60439, USA;
| | - Nicholas J. Clark
- Department of Drug Product Development, Amgen Incorporated, One Amgen Center Drive, Thousand Oaks, CA 91230, USA; (N.J.C.); (A.M.)
| | - Arnold McAuley
- Department of Drug Product Development, Amgen Incorporated, One Amgen Center Drive, Thousand Oaks, CA 91230, USA; (N.J.C.); (A.M.)
| | - Joseph E. Curtis
- NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Mail Stop 6102, Gaithersburg, MD 20899, USA; (M.M.C.); (J.A.S.); (M.L.)
| |
Collapse
|
45
|
Human myeloma IgG4 reveals relatively rigid asymmetric Y-like structure with different conformational stability of C H 2 domains. Mol Immunol 2017; 92:199-210. [DOI: 10.1016/j.molimm.2017.10.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 10/10/2017] [Accepted: 10/17/2017] [Indexed: 11/20/2022]
|
46
|
Piche-Nicholas NM, Avery LB, King AC, Kavosi M, Wang M, O'Hara DM, Tchistiakova L, Katragadda M. Changes in complementarity-determining regions significantly alter IgG binding to the neonatal Fc receptor (FcRn) and pharmacokinetics. MAbs 2017; 10:81-94. [PMID: 28991504 PMCID: PMC5800364 DOI: 10.1080/19420862.2017.1389355] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
A large body of data exists demonstrating that neonatal Fc receptor (FcRn) binding of an IgG via its Fc CH2-CH3 interface trends with the pharmacokinetics (PK) of IgG. We have observed that PK of IgG molecules vary widely, even when they share identical Fc domains. This led us to hypothesize that domains distal from the Fc could contribute to FcRn binding and affect PK. In this study, we explored the role of these IgG domains in altering the affinity between IgG and FcRn. Using a surface plasmon resonance-based assay developed to examine the steady-state binding affinity (KD) of IgG molecules to FcRn, we dissected the contributions of IgG domains in modulating the affinity between FcRn and IgG. Through analysis of a broad collection of therapeutic antibodies containing more than 50 unique IgG molecules, we demonstrated that variable domains, and in particular complementarity-determining regions (CDRs), significantly alter binding affinity to FcRn in vitro. Furthermore, a panel of IgG molecules differing only by 1–5 mutations in CDRs altered binding affinity to FcRn in vitro, by up to 79-fold, and the affinity values correlated with calculated isoelectric point values of both variable domains and CDR-L3. In addition, tighter affinity values trend with faster in vivo clearance of a set of IgG molecules differing only by 1–3 mutations in human FcRn transgenic mice. Understanding the role of CDRs in modulation of IgG affinity to FcRn in vitro and their effect on PK of IgG may have far-reaching implications in the optimization of IgG therapeutics.
Collapse
Affiliation(s)
| | | | - Amy C King
- a BioMedicine Design, Pfizer Inc. , Cambridge , MA , USA
| | - Mania Kavosi
- b BioMedicine Design, Pfizer Inc. , Andover , MA , USA
| | - Mengmeng Wang
- b BioMedicine Design, Pfizer Inc. , Andover , MA , USA
| | | | | | | |
Collapse
|
47
|
König N, Paulus M, Julius K, Schulze J, Voetz M, Tolan M. Antibodies under pressure: A Small-Angle X-ray Scattering study of Immunoglobulin G under high hydrostatic pressure. Biophys Chem 2017. [PMID: 28622937 DOI: 10.1016/j.bpc.2017.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In the present work two subclasses of the human antibody Immunoglobulin G (IgG) have been investigated by Small-Angle X-ray Scattering under high hydrostatic pressures up to 5kbar. It is shown that IgG adopts a symmetric T-shape in solution which differs significantly from available crystal structures. Moreover, high-pressure experiments verify the high stability of the IgG molecule. It is not unfolded by hydrostatic pressures of up to 5kbar but a slight increase of the radius of gyration was observed at elevated pressures.
Collapse
Affiliation(s)
- Nico König
- Fakultät Physik/DELTA, TU Dortmund, Dortmund 44221, Germany; Bayer AG, Leverkusen 51368, Germany.
| | - Michael Paulus
- Fakultät Physik/DELTA, TU Dortmund, Dortmund 44221, Germany
| | - Karin Julius
- Fakultät Physik/DELTA, TU Dortmund, Dortmund 44221, Germany
| | - Julian Schulze
- Fakultät Physik/DELTA, TU Dortmund, Dortmund 44221, Germany
| | | | - Metin Tolan
- Fakultät Physik/DELTA, TU Dortmund, Dortmund 44221, Germany
| |
Collapse
|
48
|
Utility of Solution X-Ray Scattering for the Development of Antibody Biopharmaceuticals. J Pharm Sci 2016; 105:3278-3289. [DOI: 10.1016/j.xphs.2016.07.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 07/14/2016] [Accepted: 07/26/2016] [Indexed: 12/31/2022]
|
49
|
Valenzuela NM, Hickey MJ, Reed EF. Antibody Subclass Repertoire and Graft Outcome Following Solid Organ Transplantation. Front Immunol 2016; 7:433. [PMID: 27822209 PMCID: PMC5075576 DOI: 10.3389/fimmu.2016.00433] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 10/03/2016] [Indexed: 12/20/2022] Open
Abstract
Long-term outcomes in solid organ transplantation are constrained by the development of donor-specific alloantibodies (DSA) against human leukocyte antigen (HLA) and other targets, which elicit antibody-mediated rejection (ABMR). However, antibody-mediated graft injury represents a broad continuum, from extensive complement activation and tissue damage compromising the function of the transplanted organ, to histological manifestations of endothelial cell injury and mononuclear cell infiltration but without concurrent allograft dysfunction. In addition, while transplant recipients with DSA as a whole fare worse than those without, a substantial minority of patients with DSA do not experience poorer graft outcome. Taken together, these observations suggest that not all DSA are equally pathogenic. Antibody effector functions are controlled by a number of factors, including antibody concentration, antigen availability, and antibody isotype/subclass. Antibody isotype is specified by many integrated signals, including the antigen itself as well as from antigen-presenting cells or helper T cells. To date, a number of studies have described the repertoire of IgG subclasses directed against HLA in pretransplant patients and evaluated the clinical impact of different DSA IgG subclasses on allograft outcome. This review will summarize what is known about the repertoire of antibodies to HLA and non-HLA targets in transplantation, focusing on the distribution of IgG subclasses, as well as the general biology, etiology, and mechanisms of injury of different humoral factors.
Collapse
Affiliation(s)
- Nicole M Valenzuela
- UCLA Immunogenetics Center, University of California Los Angeles, Los Angeles, CA, USA; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Michelle J Hickey
- UCLA Immunogenetics Center, University of California Los Angeles, Los Angeles, CA, USA; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Elaine F Reed
- UCLA Immunogenetics Center, University of California Los Angeles, Los Angeles, CA, USA; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
50
|
Abstract
IgG4, the least represented human IgG subclass in serum, is an intriguing antibody with unique biological properties, such as the ability to undergo Fab-arm exchange and limit immune complex formation. The lack of effector functions, such as antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity, is desirable for therapeutic purposes. IgG4 plays a protective role in allergy by acting as a blocking antibody, and inhibiting mast cell degranulation, but a deleterious role in malignant melanoma, by impeding IgG1-mediated anti-tumor immunity. These findings highlight the importance of understanding the interaction between IgG4 and Fcγ receptors. Despite a wealth of structural information for the IgG1 subclass, including complexes with Fcγ receptors, and structures for intact antibodies, high-resolution crystal structures were not reported for IgG4-Fc until recently. Here, we highlight some of the biological properties of human IgG4, and review the recent crystal structures of IgG4-Fc. We discuss the unexpected conformations adopted by functionally important Cγ2 domain loops, and speculate about potential implications for the interaction between IgG4 and FcγRs.
Collapse
Affiliation(s)
- Anna M Davies
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK.,Medical Research Council & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Brian J Sutton
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK.,Medical Research Council & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| |
Collapse
|