1
|
Stephens AD, Wilkinson T. Discovery of Therapeutic Antibodies Targeting Complex Multi-Spanning Membrane Proteins. BioDrugs 2024; 38:769-794. [PMID: 39453540 PMCID: PMC11530565 DOI: 10.1007/s40259-024-00682-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/26/2024]
Abstract
Complex integral membrane proteins, which are embedded in the cell surface lipid bilayer by multiple transmembrane spanning polypeptides, encompass families of proteins that are important target classes for drug discovery. These protein families include G protein-coupled receptors, ion channels, transporters, enzymes, and adhesion molecules. The high specificity of monoclonal antibodies and the ability to engineer their properties offers a significant opportunity to selectively bind these target proteins, allowing direct modulation of pharmacology or enabling other mechanisms of action such as cell killing. Isolation of antibodies that bind these types of membrane proteins and exhibit the desired pharmacological function has, however, remained challenging due to technical issues in preparing membrane protein antigens suitable for enabling and driving antibody drug discovery strategies. In this article, we review progress and emerging themes in defining discovery strategies for a generation of antibodies that target these complex membrane protein antigens. We also comment on how this field may develop with the emerging implementation of computational techniques, artificial intelligence, and machine learning.
Collapse
Affiliation(s)
- Amberley D Stephens
- Department of Biologics Engineering, Oncology R&D, The Discovery Centre, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0AA, UK
| | - Trevor Wilkinson
- Department of Biologics Engineering, Oncology R&D, The Discovery Centre, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0AA, UK.
| |
Collapse
|
2
|
Fontaine T, Busch A, Laeremans T, De Cesco S, Liang YL, Jaakola VP, Sands Z, Triest S, Masiulis S, Dekeyzer L, Samyn N, Loeys N, Perneel L, Debaere M, Martini M, Vantieghem C, Virmani R, Skieterska K, Staelens S, Barroco R, Van Roy M, Menet C. Structure elucidation of a human melanocortin-4 receptor specific orthosteric nanobody agonist. Nat Commun 2024; 15:7029. [PMID: 39353917 PMCID: PMC11445563 DOI: 10.1038/s41467-024-50827-7] [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: 09/21/2023] [Accepted: 07/23/2024] [Indexed: 10/03/2024] Open
Abstract
The melanocortin receptor 4 (MC4R) belongs to the melanocortin receptor family of G-protein coupled receptors and is a key switch in the leptin-melanocortin molecular axis that controls hunger and satiety. Brain-produced hormones such as α-melanocyte-stimulating hormone (agonist) and agouti-related peptide (inverse agonist) regulate the molecular communication of the MC4R axis but are promiscuous for melanocortin receptor subtypes and induce a wide array of biological effects. Here, we use a chimeric construct of conformation-selective, nanobody-based binding domain (a ConfoBody Cb80) and active state-stabilized MC4R-β2AR hybrid for efficient de novo discovery of a sequence diverse panel of MC4R-specific, potent and full agonistic nanobodies. We solve the active state MC4R structure in complex with the full agonistic nanobody pN162 at 3.4 Å resolution. The structure shows a distinct interaction with pN162 binding deeply in the orthosteric pocket. MC4R peptide agonists, such as the marketed setmelanotide, lack receptor selectivity and show off-target effects. In contrast, the agonistic nanobody is highly specific and hence can be a more suitable agent for anti-obesity therapeutic intervention via MC4R.
Collapse
MESH Headings
- Receptor, Melanocortin, Type 4/agonists
- Receptor, Melanocortin, Type 4/metabolism
- Receptor, Melanocortin, Type 4/chemistry
- Receptor, Melanocortin, Type 4/genetics
- Humans
- Single-Domain Antibodies/chemistry
- Single-Domain Antibodies/pharmacology
- Single-Domain Antibodies/metabolism
- alpha-MSH/chemistry
- alpha-MSH/pharmacology
- alpha-MSH/metabolism
- HEK293 Cells
- Protein Binding
- Binding Sites
- Crystallography, X-Ray
- Models, Molecular
- Animals
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Simonas Masiulis
- Materials and Structural Analysis, Thermo Fisher Scientific, Eindhoven, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Hutchings CJ, Sato AK. Phage display technology and its impact in the discovery of novel protein-based drugs. Expert Opin Drug Discov 2024; 19:887-915. [PMID: 39074492 DOI: 10.1080/17460441.2024.2367023] [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: 11/28/2023] [Accepted: 06/07/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Phage display technology is a well-established versatile in vitro display technology that has been used for over 35 years to identify peptides and antibodies for use as reagents and therapeutics, as well as exploring the diversity of alternative scaffolds as another option to conventional therapeutic antibody discovery. Such successes have been responsible for spawning a range of biotechnology companies, as well as many complementary technologies devised to expedite the drug discovery process and resolve bottlenecks in the discovery workflow. AREAS COVERED In this perspective, the authors summarize the application of phage display for drug discovery and provide examples of protein-based drugs that have either been approved or are being developed in the clinic. The amenability of phage display to generate functional protein molecules to challenging targets and recent developments of strategies and techniques designed to harness the power of sampling diverse repertoires are highlighted. EXPERT OPINION Phage display is now routinely combined with cutting-edge technologies to deep-mine antibody-based repertoires, peptide, or alternative scaffold libraries generating a wealth of data that can be leveraged, e.g. via artificial intelligence, to enable the potential for clinical success in the discovery and development of protein-based therapeutics.
Collapse
|
4
|
Macrophage polarization is involved in liver fibrosis induced by β 1-adrenoceptor autoantibody. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1100-1112. [PMID: 35983976 PMCID: PMC9828683 DOI: 10.3724/abbs.2022102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Accumulating evidence suggests that liver injury can be induced by the over-expression of β 1-adrenergic receptors (β 1-ARs). High titers of autoantibodies specific to β 1-adrenergic receptors (β 1-AA) are detected in the sera of heart failure patients, potentially playing agonist-like roles. However, the role of β 1-AA in liver function has not been characterized. In this study, we collect the sera of primary biliary cholangitis (PBC) patients, a condition which easily develops into liver fibrosis, and analyze the relationship between PBC and β 1-AA. A passive immunization model is established to assess the effect of β 1-AA on the liver. Subsequently, the effect of β 1-AA on macrophages is investigated in vitro. Results show that PBC patients have a high titer and ratio of β 1-AA, compared to controls. Liver injury and fibrosis are induced by β 1-AA. In vitro experiments with ROS probe demonstrate that β 1-AA induces macrophages to produce ROS and secrete TNFα. These effects can be partially reversed by metoprolol, a blocker for β 1-AR. Results from the transwell and phagocytosis assays show that β 1-AA promotes macrophage migration and phagocytosis. FCM tests suggest that β 1-AA induces the alteration of M1 rather than M2 markers in macrophages. Finally, the Annexin V/PI assay indicates that macrophage culture supernatants stimulated by β 1-AA cause hepatocyte apoptosis. Overall, these results suggest that β 1-AA is involved in PBC. The β 1-AA-induced activation, phagocytosis and phenotypic modification of macrophages may play an important role in the development of hepatic fibrosis and injury.
Collapse
|
5
|
Mitgau J, Franke J, Schinner C, Stephan G, Berndt S, Placantonakis DG, Kalwa H, Spindler V, Wilde C, Liebscher I. The N Terminus of Adhesion G Protein–Coupled Receptor GPR126/ADGRG6 as Allosteric Force Integrator. Front Cell Dev Biol 2022; 10:873278. [PMID: 35813217 PMCID: PMC9259995 DOI: 10.3389/fcell.2022.873278] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/12/2022] [Indexed: 12/15/2022] Open
Abstract
The adhesion G protein–coupled receptor (aGPCR) GPR126/ADGRG6 plays an important role in several physiological functions, such as myelination or peripheral nerve repair. This renders the receptor an attractive pharmacological target. GPR126 is a mechano-sensor that translates the binding of extracellular matrix (ECM) molecules to its N terminus into a metabotropic intracellular signal. To date, the structural requirements and the character of the forces needed for this ECM-mediated receptor activation are largely unknown. In this study, we provide this information by combining classic second-messenger detection with single-cell atomic force microscopy. We established a monoclonal antibody targeting the N terminus to stimulate GPR126 and compared it to the activation through its known ECM ligands, collagen IV and laminin 211. As each ligand uses a distinct mode of action, the N terminus can be regarded as an allosteric module that can fine-tune receptor activation in a context-specific manner.
Collapse
Affiliation(s)
- Jakob Mitgau
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Julius Franke
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Camilla Schinner
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Gabriele Stephan
- Department of Neurosurgery, Kimmel Center for Stem Cell Biology, Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, United States
| | - Sandra Berndt
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Dimitris G. Placantonakis
- Department of Neurosurgery, Kimmel Center for Stem Cell Biology, Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, United States
| | - Hermann Kalwa
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany
| | - Volker Spindler
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Caroline Wilde
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
- *Correspondence: Ines Liebscher,
| |
Collapse
|
6
|
To'a Salazar G, Huang Z, Zhang N, Zhang XG, An Z. Antibody Therapies Targeting Complex Membrane Proteins. ENGINEERING 2021; 7:1541-1551. [DOI: 10.1016/j.eng.2020.11.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
7
|
Scott MJ, Jowett A, Orecchia M, Ertl P, Ouro-Gnao L, Ticehurst J, Gower D, Yates J, Poulton K, Harris C, Mullin MJ, Smith KJ, Lewis AP, Barton N, Washburn ML, de Wildt R. Rapid identification of highly potent human anti-GPCR antagonist monoclonal antibodies. MAbs 2021; 12:1755069. [PMID: 32343620 PMCID: PMC7188403 DOI: 10.1080/19420862.2020.1755069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Complex cellular targets such as G protein-coupled receptors (GPCRs), ion channels, and other multi-transmembrane proteins represent a significant challenge for therapeutic antibody discovery, primarily because of poor stability of the target protein upon extraction from cell membranes. To assess whether a limited set of membrane-bound antigen formats could be exploited to identify functional antibodies directed against such targets, we selected a GPCR of therapeutic relevance (CCR1) and identified target binders using an in vitro yeast-based antibody discovery platform (AdimabTM) to expedite hit identification. Initially, we compared two different biotinylated antigen formats overexpressing human CCR1 in a ‘scouting’ approach using a subset of the antibody library. Binders were isolated using streptavidin-coated beads, expressed as yeast supernatants, and screened using a high-throughput binding assay and flow cytometry on appropriate cell lines. The most suitable antigen was then selected to isolate target binders using the full library diversity. This approach identified a combined total of 183 mAbs with diverse heavy chain sequences. A subset of clones exhibited high potencies in primary cell chemotaxis assays, with IC50 values in the low nM/high pM range. To assess the feasibility of any further affinity enhancement, full-length hCCR1 protein was purified, complementary-determining region diversified libraries were constructed from a high and lower affinity mAb, and improved binders were isolated by fluorescence-activated cell sorting selections. A significant affinity enhancement was observed for the lower affinity parental mAb, but not the high affinity mAb. These data exemplify a methodology to generate potent human mAbs for challenging targets rapidly using whole cells as antigen and define a route to the identification of affinity-matured variants if required.
Collapse
Affiliation(s)
- Martin J Scott
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Amanda Jowett
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Martin Orecchia
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Peter Ertl
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Larissa Ouro-Gnao
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Julia Ticehurst
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - David Gower
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - John Yates
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Katie Poulton
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Carol Harris
- Department of Protein & Cellular Sciences, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Michael J Mullin
- Department of Protein & Cellular Sciences, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Kathrine J Smith
- Department of Protein & Cellular Sciences, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Alan P Lewis
- Department of Data & Computational Sciences, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Nick Barton
- Department of Data & Computational Sciences, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Michael L Washburn
- Experimental Medicine Unit, Glaxo Smith Kline Research & Development, Collegeville, PA, USA
| | - Ruud de Wildt
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| |
Collapse
|
8
|
Chatterjee T, Zhang S, Posey TA, Jacob J, Wu L, Yu W, Francisco LE, Liu QJ, Carmon KS. Anti-GPR56 monoclonal antibody potentiates GPR56-mediated Src-Fak signaling to modulate cell adhesion. J Biol Chem 2021; 296:100261. [PMID: 33837725 PMCID: PMC7948743 DOI: 10.1016/j.jbc.2021.100261] [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: 11/19/2020] [Revised: 12/28/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022] Open
Abstract
GPR56 is a member of the adhesion G-protein-coupled receptor family shown to play important roles in cell adhesion, brain development, immune function, and tumorigenesis. GPR56 is highly upregulated in colorectal cancer and correlates with poor prognosis. Several studies have shown GPR56 couples to the Gα12/13 class of heterotrimeric G-proteins to promote RhoA activation. However, due to its structural complexity and lack of a high-affinity receptor-specific ligand, the complete GPR56 signaling mechanism remains largely unknown. To delineate the activation mechanism and intracellular signaling functions of GPR56, we generated a monoclonal antibody (mAb) that binds with high affinity and specificity to the extracellular domain (ECD). Using deletion mutants, we mapped the mAb binding site to the GAIN domain, which mediates membrane-proximal autoproteolytic cleavage of the ECD. We showed that GPR56 overexpression in 293T cells leads to increased phosphorylation of Src, Fak, and paxillin adhesion proteins and activation of the Gα12/13-RhoA-mediated serum response factor (SRF) pathway. Treatment with the mAb potentiated Src-Fak phosphorylation, RhoA–SRF signaling, and cell adhesion. Consistently, GPR56 knockdown in colorectal cancer cells decreased Src–Fak pathway phosphorylation and cell adhesion. Interestingly, GPR56-mediated activation of Src–Fak phosphorylation occurred independent of RhoA, yet mAb-induced potentiation of RhoA–SRF signaling was Src-dependent. Furthermore, we show that the C-terminal portion of the Serine–Threonine–Proline-rich (STP) region, adjacent to the GAIN domain, was required for Src–Fak activation. However, autoproteolytic cleavage of the ECD was dispensable. These data support a new ECD-dependent mechanism by which GPR56 functions to regulate adhesion through activation of Src–Fak signaling.
Collapse
Affiliation(s)
- Treena Chatterjee
- The Brown Foundation Institute of Molecular Medicine and Center for Translational Cancer Research, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Sheng Zhang
- The Brown Foundation Institute of Molecular Medicine and Center for Translational Cancer Research, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Tressie A Posey
- The Brown Foundation Institute of Molecular Medicine and Center for Translational Cancer Research, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Joan Jacob
- The Brown Foundation Institute of Molecular Medicine and Center for Translational Cancer Research, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ling Wu
- The Brown Foundation Institute of Molecular Medicine and Center for Translational Cancer Research, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Wangsheng Yu
- The Brown Foundation Institute of Molecular Medicine and Center for Translational Cancer Research, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Liezl E Francisco
- The Brown Foundation Institute of Molecular Medicine and Center for Translational Cancer Research, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Qingyun J Liu
- The Brown Foundation Institute of Molecular Medicine and Center for Translational Cancer Research, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Kendra S Carmon
- The Brown Foundation Institute of Molecular Medicine and Center for Translational Cancer Research, University of Texas Health Science Center at Houston, Houston, Texas, USA.
| |
Collapse
|
9
|
Skiba MA, Kruse AC. Autoantibodies as Endogenous Modulators of GPCR Signaling. Trends Pharmacol Sci 2020; 42:135-150. [PMID: 33358695 DOI: 10.1016/j.tips.2020.11.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/24/2020] [Accepted: 11/28/2020] [Indexed: 02/06/2023]
Abstract
Endogenous self-reactive autoantibodies (AAs) recognize a range of G-protein-coupled receptors (GPCRs). They are frequently associated with cardiovascular, neurological, and autoimmune disorders, and in some cases directly impact disease progression. Many GPCR AAs modulate receptor signaling, but molecular details of their modulatory activity are not well understood. Technological advances have provided insight into GPCR biology, which now facilitates deeper understanding of GPCR AA function at the molecular level. Most GPCR AAs are allosteric modulators and exhibit a broad range of pharmacological properties, altering both receptor signaling and trafficking. Understanding GPCR AAs is not only important for defining how these unusual GPCR modulators function in disease, but also provides insight into the potential use and limitations of using therapeutic antibodies to modulate GPCR signaling.
Collapse
Affiliation(s)
- Meredith A Skiba
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew C Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Wölfel A, Sättele M, Zechmeister C, Nikolaev VO, Lohse MJ, Boege F, Jahns R, Boivin-Jahns V. Unmasking features of the auto-epitope essential for β 1 -adrenoceptor activation by autoantibodies in chronic heart failure. ESC Heart Fail 2020; 7:1830-1841. [PMID: 32436653 PMCID: PMC7373925 DOI: 10.1002/ehf2.12747] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/31/2020] [Accepted: 04/21/2020] [Indexed: 12/15/2022] Open
Abstract
Aims Chronic heart failure (CHF) can be caused by autoantibodies stimulating the heart via binding to first and/or second extracellular loops of cardiac β1‐adrenoceptors. Allosteric receptor activation depends on conformational features of the autoantibody binding site. Elucidating these features will pave the way for the development of specific diagnostics and therapeutics. Our aim was (i) to fine‐map the conformational epitope within the second extracellular loop of the human β1‐adrenoceptor (β1ECII) that is targeted by stimulating β1‐receptor (auto)antibodies and (ii) to generate competitive cyclopeptide inhibitors of allosteric receptor activation, which faithfully conserve the conformational auto‐epitope. Methods and results Non‐conserved amino acids within the β1ECII loop (compared with the amino acids constituting the ECII loop of the β2‐adrenoceptor) were one by one replaced with alanine; potential intra‐loop disulfide bridges were probed by cysteine–serine exchanges. Effects on antibody binding and allosteric receptor activation were assessed (i) by (auto)antibody neutralization using cyclopeptides mimicking β1ECII ± the above replacements, and (ii) by (auto)antibody stimulation of human β1‐adrenoceptors bearing corresponding point mutations. With the use of stimulating β1‐receptor (auto)antibodies raised in mice, rats, or rabbits and isolated from exemplary dilated cardiomyopathy patients, our series of experiments unmasked two features of the β1ECII loop essential for (auto)antibody binding and allosteric receptor activation: (i) the NDPK211–214 motif and (ii) the intra‐loop disulfide bond C209↔C215. Of note, aberrant intra‐loop disulfide bond C209↔C216 almost fully disrupted the functional auto‐epitope in cyclopeptides. Conclusions The conformational auto‐epitope targeted by cardio‐pathogenic β1‐receptor autoantibodies is faithfully conserved in cyclopeptide homologues of the β1ECII loop bearing the NDPK211–214 motif and the C209↔C215 bridge while lacking cysteine C216. Such molecules provide promising tools for novel diagnostic and therapeutic approaches in β1‐autoantibody‐positive CHF.
Collapse
Affiliation(s)
- Angela Wölfel
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, D-97078, Wuerzburg, Germany.,Rudolf-Virchow-Centre, Josef-Schneider-Str. 2, 97080, Würzburg, Germany.,Rudolf-Virchow-Centre, Pierre Fabre Dermo-Kosmetik GmbH, Jechtinger Straße 13, 79111, Freiburg, Germany
| | - Mathias Sättele
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, D-97078, Wuerzburg, Germany
| | - Christina Zechmeister
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, D-97078, Wuerzburg, Germany.,Interdisciplinary Bank of Biomaterials and Data (ibdw), University Hospital of Würzburg, Straubmühlweg 2A, D-97078, Würzburg, Germany.,Comprehensive Heart Failure Centre (CFHC), Am Schwarzenberg 11, 978078, Würzburg, Germany
| | - Viacheslav O Nikolaev
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, D-97078, Wuerzburg, Germany.,Institute for Molecular Cardiology, Department of Cardiology and Pneumology, University Hospital Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Martin J Lohse
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, D-97078, Wuerzburg, Germany.,Rudolf-Virchow-Centre, Josef-Schneider-Str. 2, 97080, Würzburg, Germany.,Institute Max Delbrück Center for Molecular Research, Berlin-Buch, Robert-Koch-Str. 40, 1000, Berlin, Germany
| | - Fritz Boege
- Rudolf-Virchow-Centre, Institute of Clinical Chemistry and Laboratory Diagnostics, University Hospital, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Roland Jahns
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, D-97078, Wuerzburg, Germany.,Interdisciplinary Bank of Biomaterials and Data (ibdw), University Hospital of Würzburg, Straubmühlweg 2A, D-97078, Würzburg, Germany.,Comprehensive Heart Failure Centre (CFHC), Am Schwarzenberg 11, 978078, Würzburg, Germany
| | - Valérie Boivin-Jahns
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, D-97078, Wuerzburg, Germany.,Comprehensive Heart Failure Centre (CFHC), Am Schwarzenberg 11, 978078, Würzburg, Germany
| |
Collapse
|
11
|
Generating therapeutic monoclonal antibodies to complex multi-spanning membrane targets: Overcoming the antigen challenge and enabling discovery strategies. Methods 2020; 180:111-126. [PMID: 32422249 DOI: 10.1016/j.ymeth.2020.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/21/2020] [Accepted: 05/13/2020] [Indexed: 12/17/2022] Open
Abstract
Complex integral membrane proteins, which are embedded in the cell surface lipid bilayer by multiple transmembrane spanning helices, encompass families of proteins which are important target classes for drug discovery. These protein families include G protein-coupled receptors, ion channels and transporters. Although these proteins have typically been targeted by small molecule drugs and peptides, the high specificity of monoclonal antibodies offers a significant opportunity to selectively modulate these target proteins. However, it remains the case that isolation of antibodies with desired pharmacological function(s) has proven difficult due to technical challenges in preparing membrane protein antigens suitable to support antibody drug discovery. In this review recent progress in defining strategies for generation of membrane protein antigens is outlined. We also highlight antibody isolation strategies which have generated antibodies which bind the membrane protein and modulate the protein function.
Collapse
|
12
|
Hutchings CJ. A review of antibody-based therapeutics targeting G protein-coupled receptors: an update. Expert Opin Biol Ther 2020; 20:925-935. [PMID: 32264722 DOI: 10.1080/14712598.2020.1745770] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION G protein-coupled receptors (GPCRs) play key roles in many biological functions and are linked to many diseases across all therapeutic areas. As such, GPCRs represent a significant opportunity for antibody-based therapeutics. AREAS COVERED The structure of the major GPCR families is summarized in the context of choice of antigen source employed in the drug discovery process and receptor biology considerations which may impact on targeting strategies. An overview of the therapeutic GPCR-antibody target landscape and the diversity of current therapeutic programs is provided along with summary case studies for marketed antibody drugs or those in advanced clinical studies. Antibodies in early clinical studies and the emergence of next-generation modalities are also highlighted. EXPERT OPINION The GPCR-antibody pipeline has progressed significantly with a number of technical developments enabling the successful resolution of some of the challenges previously encountered and this has contributed to the growing interest in antibody-based therapeutics addressing this target class.
Collapse
|
13
|
Therapeutic Monoclonal Antibodies to Complex Membrane Protein Targets: Antigen Generation and Antibody Discovery Strategies. BioDrugs 2019; 32:339-355. [PMID: 29934752 DOI: 10.1007/s40259-018-0289-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cell surface membrane proteins comprise a wide array of structurally and functionally diverse proteins involved in a variety of important physiological and homeostatic processes. Complex integral membrane proteins, which are embedded in the lipid bilayer by multiple transmembrane-spanning helices, are represented by families of proteins that are important target classes for drug discovery. Such protein families include G-protein-coupled receptors, ion channels and transporters. Although these targets have typically been the domain of small-molecule drugs, the exquisite specificity of monoclonal antibodies offers a significant opportunity to selectively modulate these target proteins. Nevertheless, the isolation of antibodies with desired pharmacological functions has proved difficult because of technical challenges in preparing membrane protein antigens for antibody drug discovery. In this review, we describe recent progress in defining strategies for the generation of membrane protein antigens. We also describe antibody-isolation strategies that identify antibodies that bind the membrane protein and modulate protein function.
Collapse
|
14
|
Vij R, Lin Z, Chiang N, Vernes JM, Storek KM, Park S, Chan J, Meng YG, Comps-Agrar L, Luan P, Lee S, Schneider K, Bevers J, Zilberleyb I, Tam C, Koth CM, Xu M, Gill A, Auerbach MR, Smith PA, Rutherford ST, Nakamura G, Seshasayee D, Payandeh J, Koerber JT. A targeted boost-and-sort immunization strategy using Escherichia coli BamA identifies rare growth inhibitory antibodies. Sci Rep 2018; 8:7136. [PMID: 29740124 PMCID: PMC5940829 DOI: 10.1038/s41598-018-25609-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/25/2018] [Indexed: 12/19/2022] Open
Abstract
Outer membrane proteins (OMPs) in Gram-negative bacteria are essential for a number of cellular functions including nutrient transport and drug efflux. Escherichia coli BamA is an essential component of the OMP β-barrel assembly machinery and a potential novel antibacterial target that has been proposed to undergo large (~15 Å) conformational changes. Here, we explored methods to isolate anti-BamA monoclonal antibodies (mAbs) that might alter the function of this OMP and ultimately lead to bacterial growth inhibition. We first optimized traditional immunization approaches but failed to identify mAbs that altered cell growth after screening >3000 hybridomas. We then developed a “targeted boost-and-sort” strategy that combines bacterial cell immunizations, purified BamA protein boosts, and single hybridoma cell sorting using amphipol-reconstituted BamA antigen. This unique workflow improves the discovery efficiency of FACS + mAbs by >600-fold and enabled the identification of rare anti-BamA mAbs with bacterial growth inhibitory activity in the presence of a truncated lipopolysaccharide layer. These mAbs represent novel tools for dissecting the BamA-mediated mechanism of β-barrel folding and our workflow establishes a new template for the efficient discovery of novel mAbs against other highly dynamic membrane proteins.
Collapse
Affiliation(s)
- Rajesh Vij
- Department of Antibody Engineering, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Zhonghua Lin
- Department of Antibody Engineering, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Nancy Chiang
- Department of Antibody Engineering, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Jean-Michel Vernes
- Department of Biochemical and Cellular Pharmacology, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Kelly M Storek
- Department of Infectious Diseases, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Summer Park
- Department of Translational Immunology, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Joyce Chan
- Department of Biochemical and Cellular Pharmacology, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Y Gloria Meng
- Department of Biochemical and Cellular Pharmacology, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Laetitia Comps-Agrar
- Department of Biochemical and Cellular Pharmacology, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Peng Luan
- Department of Antibody Engineering, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Sophia Lee
- Department of Antibody Engineering, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Kellen Schneider
- Department of Antibody Engineering, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Jack Bevers
- Department of Antibody Engineering, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Inna Zilberleyb
- Department of BioMolecular Resources, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Christine Tam
- Department of BioMolecular Resources, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Christopher M Koth
- Department of Structural Biology, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Min Xu
- Department of Translational Immunology, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Avinash Gill
- Department of Antibody Engineering, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Marcy R Auerbach
- Department of Infectious Diseases, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Peter A Smith
- Department of Infectious Diseases, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Steven T Rutherford
- Department of Infectious Diseases, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Gerald Nakamura
- Department of Antibody Engineering, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Dhaya Seshasayee
- Department of Antibody Engineering, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Jian Payandeh
- Department of Structural Biology, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA.
| | - James T Koerber
- Department of Antibody Engineering, Genentech, 1 DNA Way, South San Francisco, California, 94080, USA.
| |
Collapse
|
15
|
The G protein-coupled receptors deorphanization landscape. Biochem Pharmacol 2018; 153:62-74. [PMID: 29454621 DOI: 10.1016/j.bcp.2018.02.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 02/13/2018] [Indexed: 12/14/2022]
Abstract
G protein-coupled receptors (GPCRs) are usually highlighted as being both the largest family of membrane proteins and the most productive source of drug targets. However, most of the GPCRs are understudied and hence cannot be used immediately for innovative therapeutic strategies. Besides, there are still around 100 orphan receptors, with no described endogenous ligand and no clearly defined function. The race to discover new ligands for these elusive receptors seems to be less intense than before. Here, we present an update of the various strategies employed to assign a function to these receptors and to discover new ligands. We focus on the recent advances in the identification of endogenous ligands with a detailed description of newly deorphanized receptors. Replication being a key parameter in these endeavors, we also discuss the latest controversies about problematic ligand-receptor pairings. In this context, we propose several recommendations in order to strengthen the reporting of new ligand-receptor pairs.
Collapse
|
16
|
Soave M, Cseke G, Hutchings CJ, Brown AJH, Woolard J, Hill SJ. A monoclonal antibody raised against a thermo-stabilised β 1-adrenoceptor interacts with extracellular loop 2 and acts as a negative allosteric modulator of a sub-set of β 1-adrenoceptors expressed in stable cell lines. Biochem Pharmacol 2017; 147:38-54. [PMID: 29102678 PMCID: PMC5770334 DOI: 10.1016/j.bcp.2017.10.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 10/31/2017] [Indexed: 11/30/2022]
Abstract
Recent interest has focused on antibodies that can discriminate between different receptor conformations. Here we have characterised the effect of a monoclonal antibody (mAb3), raised against a purified thermo-stabilised turkey β1-adrenoceptor (β1AR-m23 StaR), on β1-ARs expressed in CHO-K1 or HEK 293 cells. Immunohistochemical and radioligand-binding studies demonstrated that mAb3 was able to bind to ECL2 of the tβ1-AR, but not its human homologue. Specific binding of mAb3 to tβ1-AR was inhibited by a peptide based on the turkey, but not the human, ECL2 sequence. Studies with [3H]-CGP 12177 demonstrated that mAb3 prevented the binding of orthosteric ligands to a subset (circa 40%) of turkey β1-receptors expressed in both CHO K1 and HEK 293 cells. MAb3 significantly reduced the maximum specific binding capacity of [3H]-CGP-12177 without influencing its binding affinity. Substitution of ECL2 of tβ1-AR with its human equivalent, or mutation of residues D186S, P187D, Q188E prevented the inhibition of [3H]-CGP 12177 binding by mAb3. MAb3 also elicited a negative allosteric effect on agonist-stimulated cAMP responses. The identity of the subset of turkey β1-adrenoceptors influenced by mAb3 remains to be established but mAb3 should become an important tool to investigate the nature of β1-AR conformational states and oligomeric complexes.
Collapse
Affiliation(s)
- Mark Soave
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Gabriella Cseke
- Heptares Therapeutics Ltd., Bio Park, Welwyn Garden City AL7 3AX, UK
| | | | | | - Jeanette Woolard
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK.
| | - Stephen J Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK.
| |
Collapse
|
17
|
Opportunities for therapeutic antibodies directed at G-protein-coupled receptors. Nat Rev Drug Discov 2017; 16:787-810. [PMID: 28706220 DOI: 10.1038/nrd.2017.91] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
G-protein-coupled receptors (GPCRs) are activated by a diverse range of ligands, from large proteins and proteases to small peptides, metabolites, neurotransmitters and ions. They are expressed on all cells in the body and have key roles in physiology and homeostasis. As such, GPCRs are one of the most important target classes for therapeutic drug discovery. The development of drugs targeting GPCRs has therapeutic value across a wide range of diseases, including cancer, immune and inflammatory disorders as well as neurological and metabolic diseases. The progress made by targeting GPCRs with antibody-based therapeutics, as well as technical hurdles to overcome, are presented and discussed in this Review. Antibody therapeutics targeting C-C chemokine receptor type 4 (CCR4), CCR5 and calcitonin gene-related peptide (CGRP) are used as illustrative clinical case studies.
Collapse
|
18
|
Discovery of functional monoclonal antibodies targeting G-protein-coupled receptors and ion channels. Biochem Soc Trans 2017; 44:831-7. [PMID: 27284048 DOI: 10.1042/bst20160028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Indexed: 11/17/2022]
Abstract
The development of recombinant antibody therapeutics is a significant area of growth in the pharmaceutical industry with almost 50 approved monoclonal antibodies on the market in the US and Europe. Despite this growth, however, certain classes of important molecular targets have remained intractable to therapeutic antibodies due to complexity of the target molecules. These complex target molecules include G-protein-coupled receptors and ion channels which represent a large potential target class for therapeutic intervention with monoclonal antibodies. Although these targets have typically been addressed by small molecule approaches, the exquisite specificity of antibodies provides a significant opportunity to provide selective modulation of these target proteins. Given this opportunity, substantial effort has been applied to address the technical challenges of targeting these complex membrane proteins with monoclonal antibodies. In this review recent progress made in the strategies for discovery of functional monoclonal antibodies for these challenging membrane protein targets is addressed.
Collapse
|
19
|
Ayoub MA, Crépieux P, Koglin M, Parmentier M, Pin JP, Poupon A, Reiter E, Smit M, Steyaert J, Watier H, Wilkinson T. Antibodies targeting G protein-coupled receptors: Recent advances and therapeutic challenges. MAbs 2017; 9:735-741. [PMID: 28475474 DOI: 10.1080/19420862.2017.1325052] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Le STUDIUM conference was held November 24-25, 2016 in Tours, France as a satellite workshop of the 5th meeting of the French GDR 3545 on "G Protein-Coupled Receptors (GPCRs) -From Physiology to Drugs," which was held in Tours during November 22-24, 2016. The conference gathered speakers from academia and industry considered to be world leaders in the molecular pharmacology and signaling of GPCRs, with a particular interest in the development of therapeutic GPCR antibodies (Abs). The main topics were new advances and challenges in the development of antibodies targeting GPCRs and their potential applications to the study of the structure and function of GPCRs, as well as their implication in physiology and pathophysiology. The conference included 2 sessions, with the first dedicated to the recent advances in methodological strategies used for GPCR immunization using thermo-stabilized and purified GPCRs, and the development of various formats of Abs such as monoclonal IgG, single-chain variable fragments and nanobodies (Nbs) by in vitro and in silico approaches. The second session focused on GPCR Nbs as a "hot" field of research on GPCRs. This session started with discussion of the pioneering Nbs developed against GPCRs and their application to structural studies, then transitioned to talks on original ex vivo and in vivo studies on GPCR-selective Nbs showing promising therapeutic applications of Nbs in important physiologic systems, such as the central nervous and the immune systems, as well as in cancer. The conference ended with the consensus that Abs and especially Nbs are opening a new era of research on GPCR structure, pharmacology and pathophysiology.
Collapse
Affiliation(s)
- Mohammed Akli Ayoub
- a PRC, INRA, CNRS, Université François-Rabelais de Tours , Nouzilly , France.,b LE STUDIUM® Loire Valley Institute for Advanced Studies , Orléans , France.,c Biology Department , College of Science, United Arab Emirates University , Al Ain , United Arab Emirates
| | - Pascale Crépieux
- a PRC, INRA, CNRS, Université François-Rabelais de Tours , Nouzilly , France
| | - Markus Koglin
- d Heptares Therapeutics Ltd , BioPark, Welwyn Garden City, Hertfordshire , UK
| | - Marc Parmentier
- e Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles , Brussels , Belgium.,f Welbio, Université Libre de Bruxelles , Brussels , Belgium
| | - Jean-Philippe Pin
- g Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS UMR5203 , Montpellier , France.,h INSERM U1091 , Montpellier , France
| | - Anne Poupon
- a PRC, INRA, CNRS, Université François-Rabelais de Tours , Nouzilly , France
| | - Eric Reiter
- a PRC, INRA, CNRS, Université François-Rabelais de Tours , Nouzilly , France
| | - Martine Smit
- i Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit , Amsterdam , The Netherlands
| | - Jan Steyaert
- j Structural Biology Brussels, Vrije Universiteit Brussels , Brussels , Belgium.,k Structural Biology Research Center, Vlaams Instituut voor Biotechnologie , Brussels , Belgium
| | - Hervé Watier
- l Université François-Rabelais de Tours, CNRS, UMR 7292 , Tours , France.,m Laboratoire d'Immunologie, CHRU de Tours , Tours , France
| | - Trevor Wilkinson
- n Antibody Discovery and Protein Engineering, MedImmune , Cambridge , UK
| |
Collapse
|
20
|
van der Woning B, De Boeck G, Blanchetot C, Bobkov V, Klarenbeek A, Saunders M, Waelbroeck M, Laeremans T, Steyaert J, Hultberg A, De Haard H. DNA immunization combined with scFv phage display identifies antagonistic GCGR specific antibodies and reveals new epitopes on the small extracellular loops. MAbs 2016; 8:1126-35. [PMID: 27211075 PMCID: PMC4968103 DOI: 10.1080/19420862.2016.1189050] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/02/2016] [Accepted: 05/09/2016] [Indexed: 10/21/2022] Open
Abstract
The identification of functional monoclonal antibodies directed against G-protein coupled receptors (GPCRs) is challenging because of the membrane-embedded topology of these molecules. Here, we report the successful combination of llama DNA immunization with scFv-phage display and selections using virus-like particles (VLP) and the recombinant extracellular domain of the GPCR glucagon receptor (GCGR), resulting in glucagon receptor-specific antagonistic antibodies. By immunizing outbred llamas with plasmid DNA containing the human GCGR gene, we sought to provoke their immune system, which generated a high IgG1 response. Phage selections on VLPs allowed the identification of mAbs against the extracellular loop regions (ECL) of GCGR, in addition to multiple VH families interacting with the extracellular domain (ECD) of GCGR. Identifying mAbs binding to the ECL regions of GCGR is challenging because the large ECD covers the small ECLs in the energetically most favorable 'closed conformation' of GCGR. Comparison of Fab with scFv-phage display demonstrated that the multivalent nature of scFv display is essential for the identification of GCGR specific clones by selections on VLPs because of avid interaction. Ten different VH families that bound 5 different epitopes on the ECD of GCGR were derived from only 2 DNA-immunized llamas. Seven VH families demonstrated interference with glucagon-mediated cAMP increase. This combination of technologies proved applicable in identifying multiple functional binders in the class B GPCR context, suggesting it is a robust approach for tackling difficult membrane proteins.
Collapse
Affiliation(s)
| | | | | | - Vladimir Bobkov
- Argenx BVBA, Zwijnaarde, Belgium
- AIMMS, Division Medicinal Chemistry, VU University Amsterdam, The Netherlands
| | - Alex Klarenbeek
- Dept. of Cell Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | | | | | | | - Jan Steyaert
- Confotherapeutics, Brussels, Belgium
- VIB Structural Biology Research Center, Brussels, Belgium
| | | | | |
Collapse
|
21
|
Development of therapeutic antibodies to G protein-coupled receptors and ion channels: Opportunities, challenges and their therapeutic potential in respiratory diseases. Pharmacol Ther 2016; 169:113-123. [PMID: 27153991 DOI: 10.1016/j.pharmthera.2016.04.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The development of recombinant antibody therapeutics continues to be a significant area of growth in the pharmaceutical industry with almost 50 approved monoclonal antibodies on the market in the US and Europe. Therapeutic drug targets such as soluble cytokines, growth factors and single transmembrane spanning receptors have been successfully targeted by recombinant monoclonal antibodies and the development of new product candidates continues. Despite this growth, however, certain classes of important disease targets have remained intractable to therapeutic antibodies due to the complexity of the target molecules. These complex target molecules include G protein-coupled receptors and ion channels which represent a large target class for therapeutic intervention with monoclonal antibodies. Although these targets have typically been addressed by small molecule approaches, the exquisite specificity of antibodies provides a significant opportunity to provide selective modulation of these important regulators of cell function. Given this opportunity, a significant effort has been applied to address the challenges of targeting these complex molecules and a number of targets are linked to the pathophysiology of respiratory diseases. In this review, we provide a summary of the importance of GPCRs and ion channels involved in respiratory disease and discuss advantages offered by antibodies as therapeutics at these targets. We highlight some recent GPCRs and ion channels linked to respiratory disease mechanisms and describe in detail recent progress made in the strategies for discovery of functional antibodies against challenging membrane protein targets such as GPCRs and ion channels.
Collapse
|
22
|
Generation of Recombinant Monoclonal Antibodies from Immunised Mice and Rabbits via Flow Cytometry and Sorting of Antigen-Specific IgG+ Memory B Cells. PLoS One 2016; 11:e0152282. [PMID: 27022949 PMCID: PMC4811437 DOI: 10.1371/journal.pone.0152282] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/12/2016] [Indexed: 11/29/2022] Open
Abstract
Single B cell screening strategies, which avoid both hybridoma fusion and combinatorial display, have emerged as important technologies for efficiently sampling the natural antibody repertoire of immunized animals and humans. Having access to a range of methods to interrogate different B cell subsets provides an attractive option to ensure large and diverse panels of high quality antibody are produced. The generation of multiple antibodies and having the ability to find rare B cell clones producing IgG with unique and desirable characteristics facilitates the identification of fit-for-purpose molecules that can be developed into therapeutic agents or research reagents. Here, we describe a multi-parameter flow cytometry single-cell sorting technique for the generation of antigen-specific recombinant monoclonal antibodies from single IgG+ memory B cells. Both mouse splenocytes and rabbit PBMC from immunised animals were used as a source of B cells. Reagents staining both B cells and other unwanted cell types enabled efficient identification of class-switched IgG+ memory B cells. Concurrent staining with antigen labelled separately with two spectrally-distinct fluorophores enabled antigen-specific B cells to be identified, i.e. those which bind to both antigen conjugates (double-positive). These cells were then typically sorted at one cell per well using FACS directly into a 96-well plate containing reverse transcriptase reaction mix. Following production of cDNA, PCR was performed to amplify cognate heavy and light chain variable region genes and generate transcriptionally-active PCR (TAP) fragments. These linear expression cassettes were then used directly in a mammalian cell transfection to generate recombinant antibody for further testing. We were able to successfully generate antigen-specific recombinant antibodies from both the rabbit and mouse IgG+ memory B cell subset within one week. This included the generation of an anti-TNFR2 blocking antibody from mice with an affinity of 90 pM.
Collapse
|
23
|
Pauthner M, Yeung J, Ullman C, Bakker J, Wurch T, Reichert JM, Lund-Johansen F, Bradbury AR, Carter PJ, Melis JP. Antibody Engineering & Therapeutics, the annual meeting of The Antibody Society December 7-10, 2015, San Diego, CA, USA. MAbs 2016; 8:617-52. [PMID: 26909869 PMCID: PMC4966842 DOI: 10.1080/19420862.2016.1153211] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 02/08/2016] [Indexed: 10/31/2022] Open
Abstract
The 26th Antibody Engineering & Therapeutics meeting, the annual meeting of The Antibody Society united over 800 participants from all over the world in San Diego from 6-10 December 2015. The latest innovations and advances in antibody research and development were discussed, covering a myriad of antibody-related topics by more than 100 speakers, who were carefully selected by The Antibody Society. As a prelude, attendees could join the pre-conference training course focusing, among others, on the engineering and enhancement of antibodies and antibody-like scaffolds, bispecific antibody engineering and adaptation to generate chimeric antigen receptor constructs. The main event covered 4 d of scientific sessions that included antibody effector functions, reproducibility of research and diagnostic antibodies, new developments in antibody-drug conjugates (ADCs), preclinical and clinical ADC data, new technologies and applications for bispecific antibodies, antibody therapeutics for non-cancer and orphan indications, antibodies to harness the cellular immune system, building comprehensive IgVH-gene repertoires through discovering, confirming and cataloging new germline IgVH genes, and overcoming resistance to clinical immunotherapy. The Antibody Society's special session focused on "Antibodies to watch" in 2016. Another special session put the spotlight on the limitations of the new definitions for the assignment of antibody international nonproprietary names introduced by the World Health Organization. The convention concluded with workshops on computational antibody design and on the promise and challenges of using next-generation sequencing for antibody discovery and engineering from synthetic and in vivo libraries.
Collapse
Affiliation(s)
- Matthias Pauthner
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, USA
| | | | | | | | | | | | | | | | - Paul J. Carter
- Antibody Engineering Department, Genentech, South San Francisco, USA
| | | |
Collapse
|
24
|
Douthwaite JA, Sridharan S, Huntington C, Hammersley J, Marwood R, Hakulinen JK, Ek M, Sjögren T, Rider D, Privezentzev C, Seaman JC, Cariuk P, Knights V, Young J, Wilkinson T, Sleeman M, Finch DK, Lowe DC, Vaughan TJ. Affinity maturation of a novel antagonistic human monoclonal antibody with a long VH CDR3 targeting the Class A GPCR formyl-peptide receptor 1. MAbs 2015; 7:152-66. [PMID: 25484051 PMCID: PMC4622665 DOI: 10.4161/19420862.2014.985158] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Therapeutic monoclonal antibodies targeting G-protein-coupled receptors (GPCRs) are desirable for intervention in a wide range of disease processes. The discovery of such antibodies is challenging due to a lack of stability of many GPCRs as purified proteins. We describe here the generation of Fpro0165, a human anti-formyl peptide receptor 1 (FPR1) antibody generated by variable domain engineering of an antibody derived by immunization of transgenic mice expressing human variable region genes. Antibody isolation and subsequent engineering of affinity, potency and species cross-reactivity using phage display were achieved using FPR1 expressed on HEK cells for immunization and selection, along with calcium release cellular assays for antibody screening. Fpro0165 shows full neutralization of formyl peptide-mediated activation of primary human neutrophils. A crystal structure of the Fpro0165 Fab shows a long, protruding VH CDR3 of 24 amino acids and in silico docking with a homology model of FPR1 suggests that this long VH CDR3 is critical to the predicted binding mode of the antibody. Antibody mutation studies identify the apex of the long VH CDR3 as key to mediating the species cross-reactivity profile of the antibody. This study illustrates an approach for antibody discovery and affinity engineering to typically intractable membrane proteins such as GPCRs.
Collapse
Key Words
- CDR, complementarity determining region
- FLIPR, Fluorescent Imaging Plate Reader
- FMAT, fluorometric microvolume assay technology
- FPR, formyl peptide receptor
- Fv, variable domain
- G-protein coupled receptor
- GPCR, G-protein coupled receptor
- IgG, immunoglobulin G
- MPL, magnetic proteoliposome
- RIMMS, repetitive immunization at multiple sites
- VH, variable heavy
- VL, variable light
- affinity maturation
- antibody crystal structure
- antibody engineering
- formyl peptide receptor-1
- homology modeling
- long CDR
- phage display
- scFv, single chain Fv fragment
Collapse
|
25
|
Vazquez-Lombardi R, Phan TG, Zimmermann C, Lowe D, Jermutus L, Christ D. Challenges and opportunities for non-antibody scaffold drugs. Drug Discov Today 2015; 20:1271-83. [PMID: 26360055 DOI: 10.1016/j.drudis.2015.09.004] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 08/06/2015] [Accepted: 09/01/2015] [Indexed: 12/22/2022]
Abstract
The first candidates from the promising class of small non-antibody protein scaffolds are now moving into clinical development and practice. Challenges remain, and scaffolds will need to be further tailored toward applications where they provide real advantages over established therapeutics to succeed in a rapidly evolving drug development landscape.
Collapse
Affiliation(s)
- Rodrigo Vazquez-Lombardi
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; The University of New South Wales, Faculty of Medicine, St Vincent's Clinical School, Darlinghurst, Sydney, NSW 2010, Australia
| | - Tri Giang Phan
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; The University of New South Wales, Faculty of Medicine, St Vincent's Clinical School, Darlinghurst, Sydney, NSW 2010, Australia
| | - Carsten Zimmermann
- University of San Diego, School of Business Administration, 5998 Alcala Park, San Diego, CA 92110, USA
| | - David Lowe
- MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK
| | - Lutz Jermutus
- MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK; Trinity Hall, University of Cambridge, Trinity Lane CB2 1TJ, UK.
| | - Daniel Christ
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; The University of New South Wales, Faculty of Medicine, St Vincent's Clinical School, Darlinghurst, Sydney, NSW 2010, Australia.
| |
Collapse
|
26
|
Abstract
G protein-coupled receptors (GPCRs) are of particular importance for drug discovery, being the targets of many existing drugs, and being linked to many diseases where new therapies are required. However, as integral membrane proteins, they are generally unstable when removed from their membrane environment, precluding them from the wide range of structural and biophysical techniques which can be applied to soluble proteins such as kinases. Through the use of protein engineering methods, mutations can be identified which both increase the thermostability of GPCRs when purified in detergent, as well as biasing the receptor toward a specific physiologically relevant conformational state. The resultant stabilized receptor (known as a StaR) can be purified in multiple-milligram quantities, whilst retaining correct folding, thus enabling the generation of reagents suitable for a broad range of structural and biophysical studies. Example protocols for the purification of StaR proteins for analysis, ligand screening with the thiol-specific fluorochrome N-[4-(7-diethylamino-4-methyl-3-coumarinyl)phenyl]maleimide (CPM), surface plasmon resonance (SPR), and crystallization for structural studies are presented.
Collapse
|
27
|
Abstract
Antibody drugs have become an increasingly significant component of the therapeutic landscape. Their success has been driven by some of their unique properties, in particular their very high specificity and selectivity, in contrast to the off-target liabilities of small molecules (SMs). Antibodies can bring additional functionality to the table with their ability to interact with the immune system, and this can be further manipulated with advances in antibody engineering. This review summarizes what antibody therapeutics have achieved to date and what opportunities and challenges lie ahead. The target landscape for large molecules (LMs) versus SMs and some of the challenges for antibody drug development are discussed. Effective penetration of membrane barriers and intracellular targeting is one challenge, particularly across the highly resistant blood-brain barrier. The expanding pipeline of antibody-drug conjugates offers the potential to combine SM and LM modalities in a variety of creative ways, and antibodies also offer exciting potential to build bi- and multispecific molecules. The ability to pursue more challenging targets can also be further exploited but highlights the need for earlier screening in functional cell-based assays. I discuss how this might be addressed given the practical constraints imposed by high-throughput screening sample type and process differences in antibody primary screening.
Collapse
Affiliation(s)
- Alison J. Smith
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd, Cambridge, UK
| |
Collapse
|
28
|
Wilkinson TCI, Gardener MJ, Williams WA. Discovery of Functional Antibodies Targeting Ion Channels. ACTA ACUST UNITED AC 2014; 20:454-67. [DOI: 10.1177/1087057114560698] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Ion channels play critical roles in physiology and disease by modulation of cellular functions such as electrical excitability, secretion, cell migration, and gene transcription. Ion channels represent an important target class for drug discovery that has been largely addressed, to date, using small-molecule approaches. A significant opportunity exists to target these channels with antibodies and alternative formats of biologics. Antibodies display high specificity and affinity for their target antigen, and they have the potential to target ion channels very selectively. Nevertheless, isolating antibodies to this target class is challenging due to the difficulties in expression and purification of ion channels in a format suitable for antibody drug discovery in addition to the complexity of screening for function. In this article, we will review the current state of ion channel biologics discovery and the progress that has been made. We will also highlight the challenges in isolating functional antibodies to these targets and how these challenges may be addressed. Finally, we also illustrate successful approaches to isolating functional monoclonal antibodies targeting ion channels by way of a number of case studies drawn from recent publications.
Collapse
Affiliation(s)
| | | | - Wendy A. Williams
- Antibody Discovery and Protein Engineering, MedImmune, Cambridge, UK
| |
Collapse
|
29
|
Bock A, Kostenis E, Tränkle C, Lohse MJ, Mohr K. Pilot the pulse: controlling the multiplicity of receptor dynamics. Trends Pharmacol Sci 2014; 35:630-8. [PMID: 25455830 DOI: 10.1016/j.tips.2014.10.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/07/2014] [Accepted: 10/08/2014] [Indexed: 12/30/2022]
Abstract
G protein-coupled receptors (GPCRs) are involved in almost every (patho)physiological process, which explains their importance as drug targets. GPCRs have long been regarded as on/off-switches, which is reflected by direct activation or blockade of these receptors through the majority of marketed GPCR drugs. In recent years, however, our view of GPCRs has changed dramatically. GPCRs are now appreciated as integrative and highly dynamic signaling machines which can adopt numerous distinct conformations enabling them to initiate a highly ramified signaling network. We argue here that it may be possible to chemically encode distinct signaling profiles into ligands by rational ligand design. We exemplify our hypothesis by fine-tuning partial and biased agonism, thereby exploiting two new principles of GPCR modulation - dynamic and dualsteric ligand binding. We propose that the emerging understanding of the multiplicity of receptor dynamics will eventually lead to rationally designed new drugs which pilot the pulse; in other words, that stabilize distinct receptor states to fine-tune GPCR signaling.
Collapse
Affiliation(s)
- Andreas Bock
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Strasse 9, 97078 Würzburg, Germany.
| | - Evi Kostenis
- Molecular-, Cellular-, and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Christian Tränkle
- Pharmacology and Toxicology Section, Institute of Pharmacy, University of Bonn, Gerhard-Domagk-Strasse 3, 53121 Bonn, Germany
| | - Martin J Lohse
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Strasse 9, 97078 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, Versbacher Strasse 9, 97078 Würzburg, Germany
| | - Klaus Mohr
- Pharmacology and Toxicology Section, Institute of Pharmacy, University of Bonn, Gerhard-Domagk-Strasse 3, 53121 Bonn, Germany.
| |
Collapse
|