Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
MAbs ; 11(6): 996-1011, 2019.
Article in English | MEDLINE | ID: mdl-31156033

ABSTRACT

Agonism of members of the tumor necrosis factor receptor superfamily (TNFRSF) with monoclonal antibodies is of high therapeutic interest due to their role in immune regulation and cell proliferation. A major hurdle for pharmacologic activation of this receptor class is the requirement for high-order clustering, a mechanism that imposes a reliance in vivo on Fc receptor-mediated crosslinking. This extrinsic dependence represents a potential limitation of virtually the entire pipeline of agonist TNFRSF antibody drugs, of which none have thus far been approved or reached late-stage clinical trials. We show that tetravalent biepitopic targeting enables robust intrinsic antibody agonism for two members of this family, OX40 and DR5, that is superior to extrinsically crosslinked native parental antibodies. Tetravalent biepitopic anti-OX40 engagement co-stimulated OX40low cells, obviated the requirement for CD28 co-signal for T cell activation, and enabled superior pharmacodynamic activity relative to native IgG in a murine vaccination model. This work establishes a proof of concept for an engineering approach that addresses a major gap for the therapeutic activation of this important receptor class.


Subject(s)
Antibodies, Monoclonal/immunology , Immunologic Capping , OX40 Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , CD28 Antigens/immunology , CHO Cells , Cricetulus , Humans , Jurkat Cells , Mice , Mice, SCID , Mice, Transgenic , OX40 Ligand/immunology , Receptors, Fc/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , T-Lymphocytes/cytology
2.
Nat Chem Biol ; 7(1): 41-50, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21113169

ABSTRACT

Bruton's tyrosine kinase (Btk) is a therapeutic target for rheumatoid arthritis, but the cellular and molecular mechanisms by which Btk mediates inflammation are poorly understood. Here we describe the discovery of CGI1746, a small-molecule Btk inhibitor chemotype with a new binding mode that stabilizes an inactive nonphosphorylated enzyme conformation. CGI1746 has exquisite selectivity for Btk and inhibits both auto- and transphosphorylation steps necessary for enzyme activation. Using CGI1746, we demonstrate that Btk regulates inflammatory arthritis by two distinct mechanisms. CGI1746 blocks B cell receptor-dependent B cell proliferation and in prophylactic regimens reduces autoantibody levels in collagen-induced arthritis. In macrophages, Btk inhibition abolishes FcγRIII-induced TNFα, IL-1ß and IL-6 production. Accordingly, in myeloid- and FcγR-dependent autoantibody-induced arthritis, CGI1746 decreases cytokine levels within joints and ameliorates disease. These results provide new understanding of the function of Btk in both B cell- or myeloid cell-driven disease processes and provide a compelling rationale for targeting Btk in rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , B-Lymphocytes/drug effects , Benzamides/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Myeloid Cells/drug effects , Protein Kinase Inhibitors/therapeutic use , Agammaglobulinaemia Tyrosine Kinase , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Autoantibodies/immunology , Autoantibodies/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Benzamides/chemistry , Benzamides/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Mice , Myeloid Cells/immunology , Myeloid Cells/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/pharmacology , Protein-Tyrosine Kinases/therapeutic use , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
3.
J Clin Invest ; 120(6): 2218-29, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20458139

ABSTRACT

IgE-mediated hypersensitivity is central to the pathogenesis of asthma and other allergic diseases. Although neutralization of serum IgE with IgE-specific antibodies is in general an efficacious treatment for allergic asthma, one limitation of this approach is its lack of effect on IgE production. Here, we have developed a strategy to disrupt IgE production by generating monoclonal antibodies that target a segment of membrane IgE on human IgE-switched B cells that is not present in serum IgE. This segment is known as the M1' domain, and using genetically modified mice that contain the human M1' domain inserted into the mouse IgE locus, we demonstrated that M1'-specific antibodies reduced serum IgE and IgE-producing plasma cells in vivo, without affecting other immunoglobulin isotypes. M1'-specific antibodies were effective when delivered prophylactically and therapeutically in mouse models of immunization, allergic asthma, and Nippostrongylus brasiliensis infection, likely by inducing apoptosis of IgE-producing B cells. In addition, we generated a humanized M1'-specific antibody that was active on primary human cells in vivo, as determined by its reduction of serum IgE levels and IgE plasma cell numbers in a human PBMC-SCID mouse model. Thus, targeting of human IgE-producing B cells with apoptosis-inducing M1'-specific antibodies may be a novel treatment for asthma and allergy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies/immunology , B-Lymphocytes/drug effects , Hypersensitivity, Immediate/immunology , Mice, SCID/immunology , Animals , Antibodies, Monoclonal/immunology , Asthma/immunology , B-Lymphocytes/immunology , Humans , Hypersensitivity/immunology , Immunization , Mice , Mice, Transgenic , Nippostrongylus/drug effects , Nippostrongylus/immunology
4.
Proc Natl Acad Sci U S A ; 107(19): 8712-7, 2010 May 11.
Article in English | MEDLINE | ID: mdl-20421466

ABSTRACT

Tim-4 is a phosphatidylserine (PS) receptor that is expressed on various macrophage subsets. It mediates phagocytosis of apoptotic cells by peritoneal macrophages. The in vivo functions of Tim-4 in phagocytosis and immune responses, however, are still unclear. In this study, we show that Tim-4 quickly forms punctate caps on contact with apoptotic cells, in contrast to its normal diffused expression on the surface of phagocytes. Despite its expression in marginal zone and tingible body macrophages, Tim-4 deficiency only minimally affects outcomes of several acute immune challenges, including the trapping of apoptotic cells in the marginal zone, the clearance apoptotic cells by tingible body macrophages, and the formation of germinal centers and elicitation of antibody responses against sheep red blood cells (SRBCs). In addition, Tim-4(-/-) resident peritoneal macrophages (rPMs) phagocytose necrotic cells and other opsonized targets normally. However, their ability to bind and engulf apoptotic cells is significantly compromised both in vitro and in vivo. Most importantly, Tim-4 deficiency results in increased cellularity in the peritoneum. Resting rPMs produce higher TNF-alpha in culture. Their response to LPS, on the contrary, is dampened. Our data support an indispensible role of Tim-4 in maintaining the homeostasis of rPMs.


Subject(s)
Homeostasis/immunology , Macrophages, Peritoneal/immunology , Membrane Proteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Antibodies/immunology , Antibody Formation/immunology , Apoptosis/immunology , Cell Adhesion , Cell Count , Cell Line , Erythrocytes/immunology , Humans , Hypersensitivity, Delayed/immunology , Macrophages, Peritoneal/cytology , Membrane Proteins/deficiency , Mice , Phagocytosis/immunology , Protein Transport , Receptors, Complement/immunology , Sheep , Spleen/cytology , Spleen/immunology , Tumor Necrosis Factor-alpha/biosynthesis
5.
Blood ; 110(12): 3959-67, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17687108

ABSTRACT

Removal of pathogenic B lymphocytes by depletion of monoclonal antibodies (mAbs) or deprivation of B-cell survival factors has demonstrated clinical benefit in both oncologic and immunologic diseases. Partial clinical responses and emerging data demonstrating incomplete B-cell depletion after immunotherapy fuels the need for improved therapeutic modalities. Lessons from the first generation of therapeutics directed against B-cell-specific antigens (CD20, CD22) are being applied to develop novel antibodies with additional functional attributes. We describe the generation of a novel class of B-cell-directed therapy (anti-BR3 mAbs) that combines the depleting capacity of a therapeutic mAb and blockade of B-cell-activating factor (BAFF)-BR3 B-cell survival. In mice, treatment with antagonistic anti-BR3 antibodies results in quantitatively greater reduction in some B-cell subsets and qualitatively different effects on bone marrow plasma cells compared with BR3-Fc BAFF blockade or with anti-CD20 treatment. Comparative analysis of BR3-Fc and anti-BR3 mAb reveals a lower B-cell dependence for BAFF-mediated survival in nonhuman primates than in mice. This novel class of B-cell-targeted therapies shows species characteristics in mice and primates that will guide translation to treatment of human disease.


Subject(s)
Antibodies, Monoclonal/pharmacology , B-Cell Activation Factor Receptor/antagonists & inhibitors , Immune System Diseases/drug therapy , Immunotherapy , Lymphocyte Depletion , Neoplasms/drug therapy , Plasma Cells/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , B-Cell Activating Factor/antagonists & inhibitors , B-Cell Activating Factor/immunology , B-Cell Activation Factor Receptor/immunology , Bone Marrow Cells/immunology , Cell Survival/drug effects , Cell Survival/immunology , Immune System Diseases/immunology , Macaca fascicularis , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Species Specificity
6.
J Biol Chem ; 281(31): 22299-22311, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16737968

ABSTRACT

We present a detailed comparative analysis of the PDZ domains of the human LAP proteins Erbin, Densin-180, and Scribble and the MAGUK ZO-1. Phage-displayed peptide libraries and in vitro affinity assays were used to define ligand binding profiles for each domain. The analysis reveals the importance of interactions with all four C-terminal residues of the ligand, which constitute a core recognition motif, and also the role of interactions with more upstream ligand residues that support and modulate the core binding interaction. In particular, the results highlight the importance of site(-1), which interacts with the penultimate residue of ligand C termini. Site(-1) was found to be monospecific in the Erbin PDZ domain (accepts tryptophan only), bispecific in the first PDZ domain of ZO-1 (accepts tryptophan or tyrosine), and promiscuous in the Scribble PDZ domains. Furthermore, it appears that the level of promiscuity within site(-1) greatly influences the range of potential biological partners and functions that can be associated with each protein. These findings show that subtle changes in binding specificity can significantly alter the range of biological partners for PDZ domains, and the insights enhance our understanding of this diverse family of peptide-binding modules.


Subject(s)
Membrane Proteins/chemistry , Protein Interaction Mapping/methods , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Binding Sites , Humans , Ligands , Membrane Proteins/metabolism , Peptide Library , Peptides/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Binding , Protein Structure, Tertiary , Sialoglycoproteins/chemistry , Sialoglycoproteins/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Zonula Occludens-1 Protein
7.
Assay Drug Dev Technol ; 2(2): 131-40, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15165509

ABSTRACT

In developing a screening assay for a serine/threonine kinase, we evaluated various formats of an in-plate enzyme-linked immunosorbent assay (ELISA), as well as solution-phase kinase assays using either ELISA or AlphaScreen detection. Substrate was available both as a biotinylated 15-residue peptide and as a 25-residue peptide containing the same sequence expressed as a glutathione S-transferase fusion protein. When increasing concentrations of either of these substrates were coated directly onto ELISA plates, the rates of the kinase reactions progressively increased. In contrast, when the biotin-peptide was captured onto NeutrAvidin-coated plates, the finite peptide binding capacity of the plates limited the amount of substrate that could be incorporated into the assay system and thereby limited the rate of the reaction at a given kinase concentration. Solution-phase kinase reactions can tolerate high substrate concentrations; however, analysis of kinase reaction samples containing biotin-peptide concentrations higher than the binding capacity of NeutrAvidin-coated plates resulted in an inability to detect differences between reactions run at different substrate concentrations. For AlphaScreen detection following solution-phase kinase reactions, limitations in the binding capacity of the donor and acceptor beads caused loss of signal for substrate concentrations above the maximum binding capacity. Overall, the solution-phase assays required significantly more kinase than the in-plate assays (1-4 microg/ml versus <100 ng/ml, respectively). These studies demonstrate that the amount of substrate that can be incorporated into an assay system substantially affects the rate of the kinase reaction and therefore the amount of kinase required for the assay.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Technology, Pharmaceutical/methods , Enzyme-Linked Immunosorbent Assay/methods , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/chemistry , Substrate Specificity
8.
J Biol Chem ; 278(9): 7645-54, 2003 Feb 28.
Article in English | MEDLINE | ID: mdl-12446668

ABSTRACT

The LAP (leucine-rich repeat and PDZ-containing) family of proteins play a role in maintaining epithelial and neuronal cell size, and mutation of these proteins can have oncogenic consequences. The LAP protein Erbin has been implicated previously in a number of cellular activities by virtue of its PDZ domain-dependent association with the C termini of both ERB-B2 and the p120-catenins. The present work describes the NMR structure of Erbin PDZ in complex with a high affinity peptide ligand and includes a comprehensive energetic analysis of both the ligand and PDZ domain side chains responsible for binding. C-terminal phage display has been used to identify preferred ligands, whereas binding affinity measurements provide precise details of the energetic importance of each ligand side chain to binding. Alanine and homolog scanning mutagenesis (in a combinatorial phage display format) identifies Erbin side chains that make energetically important contacts with the ligand. The structure of a phage-optimized peptide (Ac-TGW(-4)ETW(-1)V; IC(50) = approximately 0.15 microm) in complex with Erbin PDZ provides a structural context to understand the binding energetics. In particular, the very favorable interactions with Trp(-1) are not Erbin side chain-mediated (and therefore may be generally applicable to many PDZ domains), whereas the beta2-beta3 loop provides a binding site for the Trp(-4) side chain (specific to Erbin because it has an unusually long loop). These results contribute to a growing appreciation for the importance of at least five ligand C-terminal side chains in determining PDZ domain binding energy and highlight the mechanisms of ligand discrimination among the several hundred PDZ domains present in the human genome.


Subject(s)
Carrier Proteins/chemistry , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA/metabolism , Genome , Humans , Inhibitory Concentration 50 , Ligands , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligonucleotides/pharmacology , Peptide Library , Peptides/chemistry , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Sequence Homology, Amino Acid , Tryptophan/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...