Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
MAbs ; 8(1): 65-73, 2016.
Article in English | MEDLINE | ID: mdl-26496237

ABSTRACT

The pH-dependent antigen binding antibody, termed a recycling antibody, has recently been reported as an attractive type of second-generation engineered therapeutic antibody. A recycling antibody can dissociate antigen in the acidic endosome, and thus bind to its antigen multiple times. As a consequence, a recycling antibody can neutralize large amounts of antigen in plasma. Because this approach relies on histidine residues to achieve pH-dependent antigen binding, which could limit the epitopes that can be targeted and affect the rate of antigen dissociation in the endosome, we explored an alternative approach for generating recycling antibodies. Since calcium ion concentration is known to be lower in endosome than in plasma, we hypothesized that an antibody with antigen-binding properties that are calcium-dependent could be used as recycling antibody. Here, we report a novel anti-interleukin-6 receptor (IL-6R) antibody, identified from a phage library that binds to IL-6R only in the presence of a calcium ion. Thermal dynamics and a crystal structure study revealed that the calcium ion binds to the heavy chain CDR3 region (HCDR3), which changes and possibly stabilizes the structure of HCDR3 to make it bind to antigen calcium dependently (PDB 5AZE). In vitro and in vivo studies confirmed that this calcium-dependent antigen-binding antibody can dissociate its antigen in the endosome and accelerate antigen clearance from plasma, making it a novel approach for generating recycling antibody.


Subject(s)
Antigens , Calcium , Endosomes , Receptors, Interleukin-6 , Single-Chain Antibodies , Antigens/chemistry , Antigens/metabolism , Calcium/chemistry , Calcium/metabolism , Cell Line , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/metabolism , Endosomes/chemistry , Endosomes/metabolism , Humans , Hydrogen-Ion Concentration , Receptors, Interleukin-6/chemistry , Receptors, Interleukin-6/metabolism , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/metabolism
2.
Biochim Biophys Acta ; 1844(11): 1943-1950, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25125373

ABSTRACT

Monoclonal antibodies have become a general modality in therapeutic development. However, even with infinite binding affinity to an antigen, a conventional antibody is limited in that it can bind to the antigen only once, and this results in antigen-mediated antibody clearance when the a membrane-bound antigen is targeted, or in antibody-mediated antigen accumulation when a soluble antigen is targeted. Recently, a pH-dependent antigen-binding antibody that binds to an antigen in plasma at neutral pH and dissociates from the antigen in endosome at acidic pH has been reported to overcome this limitation and to reduce antigen-mediated antibody clearance and antibody-mediated antigen accumulation. A pH-dependent binding antibody against a soluble antigen can be further improved by Fc engineering to enhance the Fc receptor binding. Various approaches, including histidine-based engineering, direct cloning from immunized animals, and synthetic and combinatorial libraries, have been successfully applied to generate pH-dependent binding antibodies against various antigens. This review discusses the features, approaches, advantages, and challenges of developing a pH-dependent binding antibody as a novel therapeutic modality. This article is part of a Special Issue entitled: Recent advances in molecular engineering of antibody.

3.
Mol Immunol ; 58(1): 132-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24334029

ABSTRACT

Enhancing the effector function by optimizing the interaction between Fc and Fcγ receptor (FcγR) is a promising approach to enhance the potency of anticancer monoclonal antibodies (mAbs). To date, a variety of Fc engineering approaches to modulate the interaction have been reported, such as afucosylation in the heavy chain Fc region or symmetrically introducing amino acid substitutions into the region, and there is still room to improve FcγR binding and thermal stability of the CH2 domain with these approaches. Recently, we have reported that asymmetric Fc engineering, which introduces different substitutions into each Fc region of heavy chain, can further improve the FcγR binding while maintaining the thermal stability of the CH2 domain by fine-tuning the asymmetric interface between the Fc domain and FcγR. However, the structural mechanism by which the asymmetrically engineered Fc improved FcγR binding remained unclear. In order to elucidate the mechanism, we solved the crystal structure of a novel asymmetrically engineered Fc, asym-mAb23, in complex with FcγRIIIa. Asym-mAb23 has enhanced binding affinity for both FcγRIIIa and FcγRIIa at the highest level of previously reported Fc variants. The structural analysis reveals the features of the asymmetrically engineered Fc in comparison with symmetric Fc and how each asymmetrically introduced substitution contributes to the improved interaction between asym-mAb23 and FcγRIIIa. This crystal structure could be utilized to enable us to design a more potent asymmetric Fc.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Affinity , Immunoglobulin Fc Fragments/ultrastructure , Receptors, IgG/immunology , Amino Acid Substitution/genetics , Antibodies, Monoclonal/genetics , Antibody-Dependent Cell Cytotoxicity/immunology , Crystallography, X-Ray , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/immunology , Protein Binding/genetics , Protein Binding/immunology , Protein Engineering , Recombinant Proteins/genetics , Recombinant Proteins/immunology
4.
Protein Eng Des Sel ; 26(10): 589-98, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23744091

ABSTRACT

Engaging inhibitory FcγRIIb by Fc region has been recently reported to be an attractive approach for improving the efficacy of antibody therapeutics. However, the previously reported S267E/L328F variant with enhanced binding affinity to FcγRIIb, also enhances binding affinity to FcγRIIa(R131) allotype to a similar degree because FcγRIIb and FcγRIIa(R131) are structurally similar. In this study, we applied comprehensive mutagenesis and structure-guided design based on the crystal structure of the Fc/FcγRIIb complex to identify a novel Fc variant with selectively enhanced FcγRIIb binding over both FcγRIIa(R131) and FcγRIIa(H131). This novel variant has more than 200-fold stronger binding affinity to FcγRIIb than wild-type IgG1, while binding affinity to FcγRIIa(R131) and FcγRIIa(H131) is comparable with or lower than wild-type IgG1. This selectivity was achieved by conformational change of the C(H)2 domain by mutating Pro to Asp at position 238. Fc variant with increased binding to both FcγRIIb and FcγRIIa induced platelet aggregation and activation in an immune complex form in vitro while our novel variant did not. When applied to agonistic anti-CD137 IgG1 antibody, our variant greatly enhanced the agonistic activity. Thus, the selective enhancement of FcγRIIb binding achieved by our Fc variant provides a novel tool for improving the efficacy of antibody therapeutics.


Subject(s)
Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Protein Engineering , Receptors, IgG/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Animals , Crystallography, X-Ray , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin G/chemistry , Mice , Models, Molecular , Mutagenesis , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
5.
Protein Eng Des Sel ; 23(5): 385-92, 2010 May.
Article in English | MEDLINE | ID: mdl-20159773

ABSTRACT

Fc engineering to increase the binding affinity of IgG antibodies to FcRn has been reported to reduce the elimination of IgG antibodies. Herein, we present a novel non-FcRn-dependent approach to reduce the elimination of IgG antibodies. Pharmacokinetic studies conducted in normal mice of various humanized IgG4 antibodies, which had identical constant regions but different variable region sequences, revealed that an antibody with a lower isoelectric point (pI) has a longer half-life. These antibodies exhibited comparable binding affinity to FcRn, and with the antibodies with lower pIs, a longer half-life was also observed in beta2-microglobulin knockout mice, suggesting that differences in the pharmacokinetics were due to a non-FcRn-dependent mechanism. On the basis of our findings, we attempted to engineer the pharmacokinetic properties of a humanized anti-IL6 receptor IgG1 antibody. Selected substitutions in the variable region, without substitution in the Fc region, lowered the pI but did not reduce the biological activity and showed a significant reduction in the clearance of the antibody in cynomolgus monkey. These results suggest that lowering the pI by engineering the variable region could reduce the elimination of IgG antibodies and could provide an alternative to Fc engineering of IgG antibodies.


Subject(s)
Genetic Engineering/methods , Immunoglobulin G/genetics , Immunoglobulin Variable Region/genetics , Protein Engineering/methods , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , CHO Cells , Cricetinae , Cricetulus , Half-Life , Histocompatibility Antigens Class I/metabolism , Humans , Immunoglobulin G/metabolism , Isoelectric Point , Mice , Receptors, Fc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...