Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 26(23): 6196-6203, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32887725

RESUMO

PURPOSE: The safety and preliminary efficacy of MEDI1873, an agonistic IgG1 fusion protein targeting glucocorticoid-induced TNF receptor-related protein (GITR), were evaluated in an open-label, first-in-human, phase I, dose escalation study in previously treated patients with advanced solid tumors. PATIENTS AND METHODS: Two single-patient cohorts at 1.5 and 3 mg i.v. were followed by 3+3 dose escalation in six cohorts at 7.5, 25, 75, 250, 500, and 750 mg, all every 2 weeks, for up to 52 weeks. Primary endpoints were safety and tolerability, dose-limiting toxicities (DLT), and MTD. Secondary endpoints included antitumor activity, pharmacokinetics, immunogenicity, and pharmacodynamics. RESULTS: Forty patients received MEDI1873. Three experienced DLTs: grade 3 worsening tumor pain (250 mg); grade 3 nausea, vomiting, and headache (500 mg); and grade 3 non-ST segment elevation myocardial infarction (750 mg). An MTD was not reached and treatment was well tolerated up to 500 mg. Most common treatment-related adverse events were headache (25%), infusion-related reaction (17.5%), and decreased appetite (17.5%). MEDI1873 exposure was dose proportional. Antidrug-antibody incidence was low. MEDI1873 increased peripheral CD4+ effector memory T-cell proliferation as well as cytokines associated with effector T-cell activation at dose levels ≥75 mg. The best response was stable disease (SD) in 17 patients (42.5%), including 1 unconfirmed partial response. Eight patients (20.0%) had SD ≥24 weeks. CONCLUSIONS: MEDI1873 showed acceptable safety up to 500 mg i.v. every 2 weeks with pharmacodynamics activity, and prolonged SD in some patients. However, further development is not planned because of lack of demonstrated tumor response.


Assuntos
Antineoplásicos/uso terapêutico , Proteína Relacionada a TNFR Induzida por Glucocorticoide/agonistas , Imunoglobulina G/química , Neoplasias/tratamento farmacológico , Adulto , Idoso , Estudos de Casos e Controles , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/patologia , Prognóstico , Estudos Retrospectivos
2.
Sci Rep ; 9(1): 1605, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30733557

RESUMO

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.


Assuntos
Anticorpos Neutralizantes/imunologia , Inibidor 1 de Ativador de Plasminogênio/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/química , Especificidade de Anticorpos , Humanos , Camundongos , Modelos Moleculares , Inibidor 1 de Ativador de Plasminogênio/química , Conformação Proteica , Ratos
3.
Am J Respir Crit Care Med ; 185(1): 34-43, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21997333

RESUMO

RATIONALE: Inflammation and oxidative stress are linked to the deleterious effects of cigarette smoke in producing chronic obstructive pulmonary disease (COPD). Myeloperoxidase (MPO), a neutrophil and macrophage product, is important in bacterial killing, but also drives inflammatory reactions and tissue oxidation. OBJECTIVES: To determine the role of MPO in COPD. METHODS: We treated guinea pigs with a 2-thioxanthine MPO inhibitor, AZ1, in a 6-month cigarette smoke exposure model, with one group receiving compound from Smoking Day 1 and another group treated after 3 months of smoke exposure. RESULTS: At 6 months both treatments abolished smoke-induced increases in lavage inflammatory cells, largely ameliorated physiological changes, and prevented or stopped progression of morphologic emphysema and small airway remodeling. Cigarette smoke caused a marked increase in immunohistochemical staining for the myeloperoxidase-generated protein oxidation marker dityrosine, and this effect was considerably decreased with both treatment arms. Serum 8-isoprostane, another marker of oxidative stress, showed similar trends. Both treatments also prevented muscularization of the small intrapulmonary arteries, but only partially ameliorated smoke-induced pulmonary hypertension. Acutely, AZ1 prevented smoke-induced increases in expression of cytokine mediators and nuclear factor-κB binding. CONCLUSIONS: We conclude that an MPO inhibitor is able to stop progression of emphysema and small airway remodeling and to partially protect against pulmonary hypertension, even when treatment starts relatively late in the course of long-term smoke exposure, suggesting that inhibition of MPO may be a novel and useful therapeutic treatment for COPD. Protection appears to relate to inhibition of oxidative damage and down-regulation of the smoke-induced inflammatory response.


Assuntos
Inibidores Enzimáticos/farmacologia , Peroxidase/antagonistas & inibidores , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/metabolismo , Purinas/uso terapêutico , Fumar/efeitos adversos , Tionas/uso terapêutico , Remodelação das Vias Aéreas/efeitos dos fármacos , Animais , Dinoprosta/análogos & derivados , Dinoprosta/sangue , Modelos Animais de Doenças , Progressão da Doença , Feminino , Cobaias , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/prevenção & controle , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/prevenção & controle , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Peroxidase/metabolismo , Doença Pulmonar Obstrutiva Crônica/etiologia , Tioxantenos/antagonistas & inibidores , Tioxantenos/metabolismo , Tirosina/análogos & derivados , Tirosina/efeitos dos fármacos
4.
J Mol Biol ; 411(4): 791-807, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21723291

RESUMO

The differentiation of therapeutic monoclonal antibodies in an increasingly competitive landscape requires optimization of clinical efficacy combined with increased patient convenience. We describe here the generation of MEDI5117, a human anti-interleukin (IL)-6 antibody generated by variable domain engineering, to achieve subpicomolar affinity for IL-6, combined with Fc (fragment crystallizable) engineering to enhance pharmacokinetic half-life. MEDI5117 was shown to be highly potent in disease-relevant cellular assays. The pharmacokinetics of MEDI5117 were evaluated and compared to those of its progenitor, CAT6001, in a single-dose study in cynomolgus monkeys. The antibodies were administered, either subcutaneously or intravenously, as a single dose of 5 mg/kg. The half-life of MEDI5117 was extended by approximately 3-fold, and clearance was reduced by approximately 4-fold when compared to CAT6001. MEDI5117 therefore represents a potential 'next-generation' antibody; future studies are planned to determine the potential for affinity-driven efficacy and/or less frequent administration.


Assuntos
Anticorpos Anti-Idiotípicos/farmacologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/farmacocinética , Interleucina-6/antagonistas & inibidores , Interleucina-6/imunologia , Macaca fascicularis/imunologia , Animais , Anticorpos Anti-Idiotípicos/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Células Cultivadas , Meia-Vida , Humanos , Interleucina-6/genética , Rim/citologia , Rim/metabolismo , Modelos Químicos , Mutagênese , Engenharia de Proteínas , Receptores de Interleucina/imunologia , Ressonância de Plasmônio de Superfície , Linfócitos T/metabolismo , Distribuição Tecidual , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
J Mol Biol ; 406(1): 160-75, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21167836

RESUMO

Interleukin (IL) 15 is an inflammatory cytokine that plays an essential role in the activation, proliferation, and maintenance of specific natural killer cell and T-cell populations, and has been implicated as a mediator of inflammatory diseases. An anti-IL-15 antibody that blocked IL-15-dependent cellular responses was isolated by phage display and optimised via mutagenesis of the third complementarity-determining regions (CDRs) of variable heavy (VH) and variable light chains. Entire repertoires of improved variants were recombined with each other to explore the maximum potential sequence space. DISC0280, the most potent antibody isolated using this comprehensive strategy, exhibits a 228-fold increase in affinity and a striking 40,000-fold increase in cellular potency compared to its parent. Such a wholesale recombination strategy therefore represents a useful method for exploiting synergistic potency gains as part of future antibody engineering efforts. The crystal structure of DISC0280 Fab (fragment antigen binding), in complex with human IL-15, was determined in order to map the structural epitope and paratope. The most remarkable feature revealed lies within the paratope and is a novel six-amino-acid α-helix that sits within the VH CDR3 loop at the center of the antigen binding site. This is the first report to describe an α-helix as a principal component of a naturally derived VH CDR3 following affinity maturation.


Assuntos
Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/isolamento & purificação , Regiões Determinantes de Complementaridade/química , Interleucina-15/antagonistas & inibidores , Interleucina-15/imunologia , Engenharia de Proteínas , Sequência de Aminoácidos , Anticorpos Neutralizantes/genética , Sítios de Ligação de Anticorpos/genética , Regiões Determinantes de Complementaridade/genética , Epitopos/química , Epitopos/genética , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/imunologia , Cadeias Pesadas de Imunoglobulinas/química , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/imunologia , Cadeias Leves de Imunoglobulina/química , Cadeias Leves de Imunoglobulina/genética , Cadeias Leves de Imunoglobulina/imunologia , Dados de Sequência Molecular , Mutação
6.
Protein Expr Purif ; 68(1): 42-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19435602

RESUMO

The physiological activities of Interleukin-15 (IL-15) suggest that it could be useful as an immunomodulator to activate the innate immune system, however, the expression and purification yields of recombinant mature IL-15 have typically been low. In this report, a method was optimised to generate milligram quantities of this cytokine. Human IL-15 with an N-terminal (His)(6)-tag was expressed in Escherichia coli as an insoluble protein. The IL-15 material was purified from other cellular proteins by dissolution in 6M guanidine HCl, followed by Ni-NTA chromatography in a buffer containing 8M urea. Use of a multi-component screen identified the optimal conditions for folding (His)(6)-tagged human IL-15 and the method was scaled up to produce milligram quantities of folded material in its native conformation, with two intra-molecular disulphides as determined by electrospray mass spectrometry. Mature IL-15 was generated by cleavage with recombinant enterokinase, which was subsequently removed by Ni-NTA chromatography. Identical methods were used to produce mature cynomolgus monkey (Macaca fascicularis) IL-15 in similar quantities. Human and cynomolgus IL-15 were both active in two IL-15 dependent assays; mouse CTLL2 cell proliferation and human and cynomolgus CD69 upregulation on CD3(-) CD8+ lymphocytes in whole blood. Despite being 96% identical at the amino acid level the human IL-15 was 10-fold more potent than the cynomolgus IL-15 in both assays. The methods described here are useful for producing both mature IL-15 proteins in sufficient quantity for in vivo and in vitro studies, including X-ray crystallography.


Assuntos
Escherichia coli/metabolismo , Interleucina-15/isolamento & purificação , Interleucina-15/metabolismo , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Linhagem Celular , Proliferação de Células , Cromatografia em Gel , Diálise , Escherichia coli/genética , Humanos , Interleucina-15/química , Interleucina-15/genética , Lectinas Tipo C , Macaca fascicularis/genética , Peso Molecular , Dobramento de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Regulação para Cima
7.
J Biomol Screen ; 14(3): 263-72, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19211775

RESUMO

Inducible nitric oxide synthase (iNOS) is active as a homodimer. A cell-based assay suitable for high-throughput screening (HTS) was generated to identify inhibitors of iNOS dimerization using the InteraX enzyme complementation technology of Applied Biosystems. The cells contain 2 chimeric proteins of complementing deletion mutants of beta-galactosidase, each fused to the oxygenase domain of human iNOS. The assay was characterized using known iNOS dimerization inhibitors, which gave a decrease in beta-galactosidase activity. Surprisingly, the assay was also able to identify compounds that have the same profile as known inhibitors of fully formed dimeric iNOS by causing an increase in beta-galactosidase activity. The iNOS InteraX assay was used to screen approximately 800,000 compounds in a 384-well format. After hit confirmation, 3359 compounds were taken forward for full IC50 determination in InteraX and cytotoxicity assays. Of these compounds 40.5% were confirmed as greater than 10-fold more active in InteraX compared to a cytotoxicity assay and were classified as potential iNOS dimerization inhibitors as they did not inhibit beta-galactosidase alone. In the same primary screen, 901 compounds gave a significant increase in beta-galactosidase activity. Many of these were known inhibitors of iNOS. After IC50 determination in InteraX and cytotoxicity assays, 182 novel compounds remained as potential arginine-competitive inhibitors of dimeric iNOS.


Assuntos
Bioensaio/métodos , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Animais , Sítios de Ligação , Carcinoma/metabolismo , Carcinoma/patologia , Linhagem Celular , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Dimerização , Inibidores Enzimáticos/química , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Indicadores e Reagentes/metabolismo , Concentração Inibidora 50 , Rim/citologia , Camundongos , Modelos Biológicos , Estrutura Molecular , Oxazinas/metabolismo , Ligação Proteica , Proteínas/metabolismo , Reprodutibilidade dos Testes , Retroviridae/genética , Transdução Genética , Xantenos/metabolismo , beta-Galactosidase/análise , beta-Galactosidase/metabolismo
8.
Nat Chem Biol ; 4(11): 700-7, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18849972

RESUMO

Nitric oxide synthase (NOS) enzymes synthesize nitric oxide, a signal for vasodilatation and neurotransmission at low concentrations and a defensive cytotoxin at higher concentrations. The high active site conservation among all three NOS isozymes hinders the design of selective NOS inhibitors to treat inflammation, arthritis, stroke, septic shock and cancer. Our crystal structures and mutagenesis results identified an isozyme-specific induced-fit binding mode linking a cascade of conformational changes to a new specificity pocket. Plasticity of an isozyme-specific triad of distant second- and third-shell residues modulates conformational changes of invariant first-shell residues to determine inhibitor selectivity. To design potent and selective NOS inhibitors, we developed the anchored plasticity approach: anchor an inhibitor core in a conserved binding pocket, then extend rigid bulky substituents toward remote specificity pockets, which become accessible upon conformational changes of flexible residues. This approach exemplifies general principles for the design of selective enzyme inhibitors that overcome strong active site conservation.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Óxido Nítrico Sintase/antagonistas & inibidores , Sequência de Aminoácidos , Aminopiridinas/química , Aminopiridinas/farmacologia , Animais , Bovinos , Cristalografia por Raios X , Modelos Animais de Doenças , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Expressão Gênica , Humanos , Isoenzimas/antagonistas & inibidores , Masculino , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Mutação , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Quinazolinas/química , Quinazolinas/farmacologia , Ratos
9.
Mol Pharmacol ; 74(5): 1193-202, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18676678

RESUMO

The chemokine receptors CXCR1 and CXCR2 are G-protein-coupled receptors (GPCRs) implicated in mediating cellular functions associated with the inflammatory response. Potent CXCR2 receptor antagonists have been discovered, some of which have recently entered clinical development. The aim of this study was to identify key amino acid residue differences between CXCR1 and CXCR2 that influence the relative antagonism by two compounds that have markedly different chemical structures. By investigating the effects of domain switching and point mutations, we found that the second extracellular loop, which contained significant amino acid sequence diversity, was not important for compound antagonism. We were surprised to find that switching the intracellular C-terminal 60 amino acid domains of CXCR1 and CXCR2 caused an apparent reversal of antagonism at these two receptors. Further investigation showed that a single amino acid residue, lysine 320 in CXCR2 and asparagine 311 in CXCR1, plays a predominant role in describing the relative antagonism of the two compounds. Homology modeling studies based on the structure of bovine rhodopsin indicated a potential intracellular antagonist binding pocket involving lysine 320. We conclude that residue 320 in CXCR2 forms part of a potential allosteric binding pocket on the intracellular side of the receptor, a site that is distal to the orthosteric site commonly assumed to be the location of antagonist binding to GPCRs. The existence of a common intracellular allosteric binding site at GPCRs related to CXCR2 may be of value in the design of novel antagonists for therapeutic intervention.


Assuntos
Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/metabolismo , Sítio Alostérico , Sequência de Aminoácidos , Linhagem Celular , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ensaio Radioligante , Receptores de Interleucina-8A/química , Receptores de Interleucina-8A/efeitos dos fármacos , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8B/química , Receptores de Interleucina-8B/efeitos dos fármacos , Receptores de Interleucina-8B/genética , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...