Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 9825, 2023 06 17.
Article in English | MEDLINE | ID: mdl-37330528

ABSTRACT

Interleukin (IL)-33 is a broad-acting alarmin cytokine that can drive inflammatory responses following tissue damage or infection and is a promising target for treatment of inflammatory disease. Here, we describe the identification of tozorakimab (MEDI3506), a potent, human anti-IL-33 monoclonal antibody, which can inhibit reduced IL-33 (IL-33red) and oxidized IL-33 (IL-33ox) activities through distinct serum-stimulated 2 (ST2) and receptor for advanced glycation end products/epidermal growth factor receptor (RAGE/EGFR complex) signalling pathways. We hypothesized that a therapeutic antibody would require an affinity higher than that of ST2 for IL-33, with an association rate greater than 107 M-1 s-1, to effectively neutralize IL-33 following rapid release from damaged tissue. An innovative antibody generation campaign identified tozorakimab, an antibody with a femtomolar affinity for IL-33red and a fast association rate (8.5 × 107 M-1 s-1), which was comparable to soluble ST2. Tozorakimab potently inhibited ST2-dependent inflammatory responses driven by IL-33 in primary human cells and in a murine model of lung epithelial injury. Additionally, tozorakimab prevented the oxidation of IL-33 and its activity via the RAGE/EGFR signalling pathway, thus increasing in vitro epithelial cell migration and repair. Tozorakimab is a novel therapeutic agent with a dual mechanism of action that blocks IL-33red and IL-33ox signalling, offering potential to reduce inflammation and epithelial dysfunction in human disease.


Subject(s)
Inflammation , Interleukin-1 Receptor-Like 1 Protein , Mice , Humans , Animals , Interleukin-1 Receptor-Like 1 Protein/metabolism , Inflammation/metabolism , Interleukin-33/metabolism , Cytokines/metabolism , ErbB Receptors/metabolism , Signal Transduction
2.
Sci Transl Med ; 13(595)2021 05 26.
Article in English | MEDLINE | ID: mdl-34039741

ABSTRACT

Plasmacytoid dendritic cells (pDCs) not only are specialized in their capacity to secrete large amounts of type I interferon (IFN) but also serve to enable both innate and adaptive immune responses through expression of additional proinflammatory cytokines, chemokines, and costimulatory molecules. Persistent activation of pDCs has been demonstrated in a number of autoimmune diseases. To evaluate the potential benefit of depleting pDCs in autoimmunity, a monoclonal antibody targeting the pDC-specific marker immunoglobulin-like transcript 7 was generated. This antibody, known as VIB7734, which was engineered for enhanced effector function, mediated rapid and potent depletion of pDCs through antibody-dependent cellular cytotoxicity. In cynomolgus monkeys, treatment with VIB7734 reduced pDCs in blood below the lower limit of normal by day 1 after the first dose. In two phase 1 studies in patients with autoimmune diseases, VIB7734 demonstrated an acceptable safety profile, comparable to that of placebo. In individuals with cutaneous lupus, VIB7734 profoundly reduced both circulating and tissue-resident pDCs, with a 97.6% median reduction in skin pDCs at study day 85 in VIB7734-treated participants. Reductions in pDCs in the skin correlated with a decrease in local type I IFN activity as well as improvements in clinical disease activity. Biomarker analysis suggests that responsiveness to pDC depletion therapy may be greater among individuals with high baseline type I IFN activity, supporting a central role for pDCs in type I IFN production in autoimmunity and further development of VIB7734 in IFN-associated diseases.


Subject(s)
Interferon Type I , Lupus Erythematosus, Cutaneous , Autoimmunity , Chemokines , Dendritic Cells , Humans
3.
Sci Rep ; 9(1): 1605, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30733557

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1) is a serine protease inhibitor (serpin) that regulates fibrinolysis, cell adhesion and cell motility via its interactions with plasminogen activators and vitronectin. PAI-1 has been shown to play a role in a number of diverse pathologies including cardiovascular diseases, obesity and cancer and is therefore an attractive therapeutic target. However the multiple patho-physiological roles of PAI-1, and understanding the relative contributions of these in any one disease setting, make the development of therapeutically relevant molecules challenging. Here we describe the identification and characterisation of fully human antibody MEDI-579, which binds with high affinity and specificity to the active form of human PAI-1. MEDI-579 specifically inhibits serine protease interactions with PAI-1 while conserving vitronectin binding. Crystallographic analysis reveals that this specificity is achieved through direct binding of MEDI-579 Fab to the reactive centre loop (RCL) of PAI-1 and at the same exosite used by both tissue and urokinase plasminogen activators (tPA and uPA). We propose that MEDI-579 acts by directly competing with proteases for RCL binding and as such is able to modulate the interaction of PAI-1 with tPA and uPA in a way not previously described for a human PAI-1 inhibitor.


Subject(s)
Antibodies, Neutralizing/immunology , Plasminogen Activator Inhibitor 1/immunology , Amino Acid Sequence , Animals , Antibodies, Neutralizing/chemistry , Antibody Specificity , Humans , Mice , Models, Molecular , Plasminogen Activator Inhibitor 1/chemistry , Protein Conformation , Rats
4.
Ann Rheum Dis ; 78(2): 228-237, 2019 02.
Article in English | MEDLINE | ID: mdl-30459279

ABSTRACT

OBJECTIVE: Immune complexes (ICs) play a critical role in the pathology of autoimmune diseases. The aim of this study was to generate and characterise a first-in-class anti-FcγRIIA antibody (Ab) VIB9600 (previously known as MEDI9600) that blocks IgG immune complex-mediated cellular activation for clinical development. METHODS: VIB9600 was humanised and optimised from the IV.3 Ab. Binding affinity and specificity were determined by Biacore and ELISA. Confocal microscopy, Flow Cytometry-based assays and binding competition assays were used to assess the mode of action of the antibody. In vitro cell-based assays were used to demonstrate suppression of IC-mediated inflammatory responses. In vivo target suppression and efficacy was demonstrated in FcγRIIA-transgenic mice. Single-dose pharmacokinetic (PK)/pharmacodynamic study multiple dose Good Laboratory Practice (GLP) toxicity studies were conducted in non-human primates. RESULTS: We generated a humanised effector-deficient anti-FcγRIIA antibody (VIB9600) that potently blocks autoantibody and IC-mediated proinflammatory responses. VIB9600 suppresses FcγRIIA activation by blocking ligand engagement and by internalising FcγRIIA from the cell surface. VIB9600 inhibits IC-induced type I interferons from plasmacytoid dendritic cells (involved in SLE), antineutrophil cytoplasmic antibody (ANCA)-induced production of reactive oxygen species by neutrophils (involved in ANCA-associated vasculitis) and IC-induced tumour necrosis factor α and interleukin-6 production (involved in rheumatoid arthritis). In FcγRIIA transgenic mice, VIB9600 suppressed antiplatelet antibody-induced thrombocytopaenia, acute anti-GBM Ab-induced nephritis and anticollagen Ab-induced arthritis. VIB9600 also exhibited favourable PK and safety profiles in cynomolgus monkey studies. CONCLUSIONS: VIB9600 is a specific humanised antibody antagonist of FcγRIIA with null effector function that warrants further clinical development for the treatment of IC-mediated diseases.


Subject(s)
Antibodies, Anti-Idiotypic/pharmacology , Antigen-Antibody Complex/drug effects , Autoimmune Diseases/drug therapy , Immunologic Factors/pharmacology , Receptors, IgG/immunology , Animals , Antibodies, Antineutrophil Cytoplasmic/immunology , Antigen-Antibody Complex/immunology , Autoimmune Diseases/immunology , Dendritic Cells/immunology , Humans , Immunoglobulin G/immunology , Interleukin-6/immunology , Macaca fascicularis , Mice , Mice, Transgenic , Neutrophils/immunology , Reactive Oxygen Species/immunology , Tumor Necrosis Factor-alpha/immunology
5.
Front Immunol ; 8: 1200, 2017.
Article in English | MEDLINE | ID: mdl-28993780

ABSTRACT

Autoantibodies directed against citrullinated epitopes of proteins are highly diagnostic of rheumatoid arthritis (RA), and elevated levels of protein citrullination can be found in the joints of patients with RA. Calcium-dependent peptidyl-arginine deiminases (PAD) are the enzymes responsible for citrullination. PAD2 and PAD4 are enriched in neutrophils and likely drive citrullination under inflammatory conditions. PADs may be released during NETosis or cell death, but the mechanisms responsible for PAD activity under physiological conditions have not been fully elucidated. To understand how PADs citrullinate extracellular proteins, we investigated the cellular localization and activity of PAD2 and PAD4, and we report that viable neutrophils from healthy donors have active PAD4 exposed on their surface and spontaneously secrete PAD2. Neutrophil activation by some stimulatory agents increased the levels of immunoreactive PAD4 on the cell surface, and some stimuli reduced PAD2 secretion. Our data indicate that live neutrophils have the inherent capacity to express active extracellular PADs. These novel pathways are distinguished from intracellular PAD activation during NETosis and calcium influx-mediated hypercitrullination. Our study implies that extracellular PADs may have a physiological role under non-pathogenic conditions as well as a pathological role in RA.

6.
Nat Commun ; 6: 8327, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26365875

ABSTRACT

In response to infections and irritants, the respiratory epithelium releases the alarmin interleukin (IL)-33 to elicit a rapid immune response. However, little is known about the regulation of IL-33 following its release. Here we report that the biological activity of IL-33 at its receptor ST2 is rapidly terminated in the extracellular environment by the formation of two disulphide bridges, resulting in an extensive conformational change that disrupts the ST2 binding site. Both reduced (active) and disulphide bonded (inactive) forms of IL-33 can be detected in lung lavage samples from mice challenged with Alternaria extract and in sputum from patients with moderate-severe asthma. We propose that this mechanism for the rapid inactivation of secreted IL-33 constitutes a 'molecular clock' that limits the range and duration of ST2-dependent immunological responses to airway stimuli. Other IL-1 family members are also susceptible to cysteine oxidation changes that could regulate their activity and systemic exposure through a similar mechanism.


Subject(s)
Asthma/immunology , Interleukin-33/metabolism , Receptors, Cell Surface/immunology , Receptors, Interleukin/immunology , Animals , Asthma/genetics , Asthma/metabolism , Humans , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33/genetics , Interleukin-33/immunology , Male , Mice , Mice, Inbred BALB C , Oxidation-Reduction , Receptors, Cell Surface/genetics , Receptors, Interleukin/genetics
7.
PLoS Pathog ; 9(8): e1003520, 2013.
Article in English | MEDLINE | ID: mdl-23935498

ABSTRACT

Human rhinoviruses (HRV) cause the majority of common colds and acute exacerbations of asthma and chronic obstructive pulmonary disease (COPD). Effective therapies are urgently needed, but no licensed treatments or vaccines currently exist. Of the 100 identified serotypes, ∼90% bind domain 1 of human intercellular adhesion molecule-1 (ICAM-1) as their cellular receptor, making this an attractive target for development of therapies; however, ICAM-1 domain 1 is also required for host defence and regulation of cell trafficking, principally via its major ligand LFA-1. Using a mouse anti-human ICAM-1 antibody (14C11) that specifically binds domain 1 of human ICAM-1, we show that 14C11 administered topically or systemically prevented entry of two major groups of rhinoviruses, HRV16 and HRV14, and reduced cellular inflammation, pro-inflammatory cytokine induction and virus load in vivo. 14C11 also reduced cellular inflammation and Th2 cytokine/chemokine production in a model of major group HRV-induced asthma exacerbation. Interestingly, 14C11 did not prevent cell adhesion via human ICAM-1/LFA-1 interactions in vitro, suggesting the epitope targeted by 14C11 was specific for viral entry. Thus a human ICAM-1 domain-1-specific antibody can prevent major group HRV entry and induction of airway inflammation in vivo.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Immunoglobulin G/pharmacology , Intercellular Adhesion Molecule-1/immunology , Picornaviridae Infections/immunology , Pneumonia, Viral/immunology , Rhinovirus/immunology , Virus Internalization/drug effects , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Chemokines/genetics , Chemokines/immunology , HeLa Cells , Humans , Immunoglobulin G/immunology , Intercellular Adhesion Molecule-1/genetics , Jurkat Cells , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/immunology , Mice , Mice, Transgenic , Picornaviridae Infections/drug therapy , Picornaviridae Infections/genetics , Picornaviridae Infections/pathology , Pneumonia, Viral/diet therapy , Pneumonia, Viral/genetics , Pneumonia, Viral/pathology , Th2 Cells/immunology
8.
Inflamm Allergy Drug Targets ; 12(2): 99-108, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23517646

ABSTRACT

Chronic respiratory diseases such as asthma and chronic obstructive pulmonary disease (COPD) represent a significant health burden worldwide and are a major unmet medical need. Asthma affects over 300 million people and leads to 250,000 deaths per year, with an increasing prevalence particularly in developing countries. Although a large proportion of asthmatics are maintained on beta agonists and corticosteroids, there still remains a group of patients where these medicines fail to modulate symptoms and who may therefore benefit from monoclonal antibody based drugs that are aimed at controlling the disease. COPD is a cigarette smoke-driven chronic inflammatory airway disease with an increasing global prevalence. Given that current therapies fail to prevent disease progression or mortality, this patient population is also a focus for the development of monoclonal antibody therapies. At present anti-IgE (omalizumab, Xolair®) is the only monoclonal antibody based drug approved in the respiratory space for the treatment of asthma. However, an increasing number of antibodies targeting key mediators/pathways of disease are in clinical development for both asthma and COPD, including targeting the Th2 pathway for asthma (anti-IL-4/5/13) and the pro-inflammatory cytokine IL-1 for COPD. This review will examine the antibody engineering approaches used to develop the next generation of antibodies, with a focus on respiratory disease.


Subject(s)
Antibodies, Anti-Idiotypic/chemistry , Antibodies, Anti-Idiotypic/therapeutic use , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/therapeutic use , Asthma/drug therapy , Pulmonary Disease, Chronic Obstructive/drug therapy , Animals , Humans
9.
J Biomol Screen ; 18(3): 237-46, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23207740

ABSTRACT

Infection with human rhinovirus (HRV) is thought to result in acute respiratory exacerbations of chronic obstructive pulmonary disorder (COPD). Consequently, prevention of HRV infection may provide therapeutic benefit to these patients. As all major group HRV serotypes infect cells via an interaction between viral coat proteins and intercellular adhesion molecule-1 (ICAM-1), it is likely that inhibitors of this interaction would prevent or reduce infections. Our objective was to use phage display technology in conjunction with naive human antibody libraries to identify anti-ICAM-1 antibodies capable of functional blockade of HRV infection. Key to success was the development of a robust, functionally relevant high-throughput screen (HTS) compatible with the specific challenges of antibody screening. In this article, we describe the development of a novel homogeneous time-resolved fluorescence (HTRF) assay based on the inhibition of soluble ICAM-1 binding to live HRV16. We describe the implementation of the method in an antibody screening campaign and demonstrate the biological relevance of the assay by confirming the activity of resultant antibodies in a cell-based in vitro HRV infection assay.


Subject(s)
High-Throughput Screening Assays/methods , Picornaviridae Infections/immunology , Rhinovirus/immunology , Antibodies/immunology , Antibodies/metabolism , Cell Line, Tumor , Fluorescence , HeLa Cells , Humans , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Picornaviridae Infections/metabolism , Rhinovirus/metabolism
10.
MAbs ; 4(6): 664-72, 2012.
Article in English | MEDLINE | ID: mdl-22926024

ABSTRACT

Antibodies are a unique class of proteins with the ability to adapt their binding sites for high affinity and high specificity to a multitude of antigens. Many analyses have been performed on antibody sequences and structures to elucidate which amino acids have a predominant role in antibody interactions with antigens. These studies have generally not distinguished between amino acids selected for broad antigen specificity in the primary immune response and those selected for high affinity in the secondary immune response. By studying a large data set of affinity matured antibodies derived from in vitro directed evolution experiments, we were able to specifically highlight a subset of amino acids associated with affinity improvements. In a comparison of affinity maturations using either tailored or full amino acid diversification, the tailored approach was found to be at least as effective at improving affinity while requiring fewer mutagenesis libraries than the traditional method. The resulting sequence data also highlight the potential for further reducing amino acid diversity for high affinity binding interactions.


Subject(s)
Antibody Affinity , Models, Molecular , Single-Chain Antibodies/metabolism , Amino Acids/genetics , Antibody Affinity/genetics , Antibody Diversity/genetics , Binding Sites, Antibody/genetics , Computational Biology , Drug Design , Humans , Immunoglobulin Variable Region/genetics , Immunologic Memory , Picornaviridae Infections/immunology , Protein Conformation , Protein Engineering , Rhinovirus/immunology , Single-Chain Antibodies/genetics
11.
Bioorg Med Chem Lett ; 14(15): 3975-8, 2004 Aug 02.
Article in English | MEDLINE | ID: mdl-15225710

ABSTRACT

The first inhibitors of fungal protein: mannosyl transferase 1 (PMT1) are described. They are based upon rhodanine-3-acetic acid and several compounds have been identified, for example, 5-[[3-(1-phenylethoxy)-4-(2-phenylethoxy)phenyl]methylene]-4-oxo-2-thioxo-3-thiazolidineacetic acid (5a), which inhibit Candida albicans PMT1 with IC(50)s in the range 0.2-0.5 microM. Members of the series are effective in inducing changes in morphology of C. albicans in vitro that have previously been associated with loss of the transferase activity. These compounds could serve as useful tools for studying the effects of protein O-mannosylation and its relevance in the search for novel antifungal agents.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Mannosyltransferases/antagonists & inhibitors , Rhodanine/analogs & derivatives , Rhodanine/pharmacology , Candida albicans/drug effects , Candida albicans/enzymology , Enzyme Inhibitors/pharmacology , Fungi/drug effects , Fungi/enzymology , Microbial Sensitivity Tests , Rhodanine/chemical synthesis , Structure-Activity Relationship
12.
Genetics ; 166(2): 707-19, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15020461

ABSTRACT

Saccharomyces cerevisiae whi2Delta cells are unable to halt cell division in response to nutrient limitation and are sensitive to a wide variety of stresses. A synthetic lethal screen resulted in the isolation of siw mutants that had a phenotype similar to that of whi2Delta. Among these were mutations affecting SIW14, FEN2, SLT2, and THR4. Fluid-phase endocytosis is severely reduced or abolished in whi2Delta, siw14Delta, fen2Delta, and thr4Delta mutants. Furthermore, whi2Delta and siw14Delta mutants produce large actin clumps in stationary phase similar to those seen in prk1Delta ark1Delta mutants defective in protein kinases that regulate the actin cytoskeleton. Overexpression of SIW14 in a prk1Delta strain resulted in a loss of cortical actin patches and cables and was lethal. Overexpression of SIW14 also rescued the caffeine sensitivity of the slt2 mutant isolated in the screen, but this was not due to alteration of the phosphorylation state of Slt2. These observations suggest that endocytosis and the organization of the actin cytoskeleton are required for the proper response to nutrient limitation. This hypothesis is supported by the observation that rvs161Delta, sla1Delta, sla2Delta, vrp1Delta, ypt51Delta, ypt52Delta, and end3Delta mutations, which disrupt the organization of the actin cytoskeleton and/or reduce endocytosis, have a phenotype similar to that of whi2Delta mutants.


Subject(s)
Actins/genetics , Endocytosis/genetics , Genes, Lethal , Saccharomyces cerevisiae/genetics , Actins/metabolism , Cell Cycle/genetics , Cell Cycle/physiology , Endocytosis/physiology , Fungal Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phenotype , Protein Kinase C , Protein Serine-Threonine Kinases/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Deletion , Symporters/genetics , Symporters/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...