Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ChemMedChem ; 17(13): e202200163, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35475323

RESUMO

Ligand-based 19 F NMR screening is a highly effective and well-established hit-finding approach. The high sensitivity to protein binding makes it particularly suitable for fragment screening. Different criteria can be considered for generating fluorinated fragment libraries. One common strategy is to assemble a large, diverse, well-designed and characterized fragment library which is screened in mixtures, generated based on experimental 19 F NMR chemical shifts. Here, we introduce a complementary knowledge-based 19 F NMR screening approach, named 19 Focused screening, enabling the efficient screening of putative active molecules selected by computational hit finding methodologies, in mixtures assembled and on-the-fly deconvoluted based on predicted 19 F NMR chemical shifts. In this study, we developed a novel approach, named LEFshift, for 19 F NMR chemical shift prediction using rooted topological fluorine torsion fingerprints in combination with a random forest machine learning method. A demonstration of this approach to a real test case is reported.


Assuntos
Flúor , Imageamento por Ressonância Magnética , Flúor/química , Ligantes , Espectroscopia de Ressonância Magnética/métodos , Ligação Proteica
2.
J Med Chem ; 63(15): 8088-8113, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32551603

RESUMO

The serine protease factor XI (FXI) is a prominent drug target as it holds promise to deliver efficacious anticoagulation without an enhanced risk of major bleeds. Several efforts have been described targeting the active form of the enzyme, FXIa. Herein, we disclose our efforts to identify potent, selective, and orally bioavailable inhibitors of FXIa. Compound 1, identified from a diverse library of internal serine protease inhibitors, was originally designed as a complement factor D inhibitor and exhibited submicromolar FXIa activity and an encouraging absorption, distribution, metabolism, and excretion (ADME) profile while being devoid of a peptidomimetic architecture. Optimization of interactions in the S1, S1ß, and S1' pockets of FXIa through a combination of structure-based drug design and traditional medicinal chemistry led to the discovery of compound 23 with subnanomolar potency on FXIa, enhanced selectivity over other coagulation proteases, and a preclinical pharmacokinetics (PK) profile consistent with bid dosing in patients.


Assuntos
Fator XIa/antagonistas & inibidores , Fator XIa/genética , Inibidores do Fator Xa/administração & dosagem , Inibidores do Fator Xa/química , Administração Oral , Sequência de Aminoácidos , Animais , Disponibilidade Biológica , Cães , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade
3.
ACS Med Chem Lett ; 11(2): 188-194, 2020 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-32071687

RESUMO

Inhibition of neprilysin (NEP) is widely studied as a therapeutic target for the treatment of hypertension, heart failure, and kidney disease. Sacubitril/valsartan (LCZ696) is a drug approved to reduce the risk of cardiovascular death in heart failure patients with reduced ejection fraction. LBQ657 is the active metabolite of sacubitril and an inhibitor of NEP. Previously, we have reported the crystal structure of NEP bound with LBQ657, whereby we noted the presence of a subsite in S1' that has not been explored before. We were also intrigued by the zinc coordination made by one of the carboxylic acids of LBQ657, leading us to explore alternative linkers to efficiently engage zinc for NEP inhibition. Structure-guided design culminated in the synthesis of selective, orally bioavailable, and subnanomolar inhibitors of NEP. A 17-fold boost in biochemical potency was observed upon addition of a chlorine atom that occupied the newly found subsite in S1'. We report herein the discovery and preclinical profiling of compound 13, which paved the path to our clinical candidate.

4.
Blood ; 133(13): 1507-1516, 2019 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-30692123

RESUMO

A large unmet medical need exists for safer antithrombotic drugs because all currently approved anticoagulant agents interfere with hemostasis, leading to an increased risk of bleeding. Genetic and pharmacologic evidence in humans and animals suggests that reducing factor XI (FXI) levels has the potential to effectively prevent and treat thrombosis with a minimal risk of bleeding. We generated a fully human antibody (MAA868) that binds the catalytic domain of both FXI (zymogen) and activated FXI. Our structural studies show that MAA868 traps FXI and activated FXI in an inactive, zymogen-like conformation, explaining its equally high binding affinity for both forms of the enzyme. This binding mode allows the enzyme to be neutralized before entering the coagulation process, revealing a particularly attractive anticoagulant profile of the antibody. MAA868 exhibited favorable anticoagulant activity in mice with a dose-dependent protection from carotid occlusion in a ferric chloride-induced thrombosis model. MAA868 also caused robust and sustained anticoagulant activity in cynomolgus monkeys as assessed by activated partial thromboplastin time without any evidence of bleeding. Based on these preclinical findings, we conducted a first-in-human study in healthy subjects and showed that single subcutaneous doses of MAA868 were safe and well tolerated. MAA868 resulted in dose- and time-dependent robust and sustained prolongation of activated partial thromboplastin time and FXI suppression for up to 4 weeks or longer, supporting further clinical investigation as a potential once-monthly subcutaneous anticoagulant therapy.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Anticoagulantes/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Fator XI/antagonistas & inibidores , Trombose/tratamento farmacológico , Adolescente , Adulto , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Anticoagulantes/farmacologia , Feminino , Humanos , Imunoglobulina G/farmacologia , Imunoglobulina G/uso terapêutico , Macaca fascicularis , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Simulação de Acoplamento Molecular , Trombose/sangue , Adulto Jovem
5.
Bioorg Med Chem Lett ; 26(17): 4340-4, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27476144

RESUMO

Proline-based trypsin inhibitors occupying the S1-S2-S1' region were identified by an HTS screening campaign. It was discovered that truncation of the P1' moiety and appropriate extension into the S4 region led to highly potent trypsin inhibitors with excellent selectivity against related serine proteases and a favorable hERG profile.


Assuntos
Pancreatite/tratamento farmacológico , Inibidores da Tripsina/síntese química , Inibidores da Tripsina/uso terapêutico , Cristalografia por Raios X , Ativação Enzimática/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Estrutura Molecular , Relação Estrutura-Atividade , Inibidores da Tripsina/química , Inibidores da Tripsina/farmacologia
6.
Sci Rep ; 6: 27909, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27302413

RESUMO

Sacubitril is an ethyl ester prodrug of LBQ657, the active neprilysin (NEP) inhibitor, and a component of LCZ696 (sacubitril/valsartan). We report herein the three-dimensional structure of LBQ657 in complex with human NEP at 2 Å resolution. The crystal structure unravels the binding mode of the compound occupying the S1, S1' and S2' sub-pockets of the active site, consistent with a competitive inhibition mode. An induced fit conformational change upon binding of the P1'-biphenyl moiety of the inhibitor suggests an explanation for its selectivity against structurally homologous zinc metallopeptidases.


Assuntos
Aminobutiratos/química , Compostos de Bifenilo/química , Neprilisina/química , Neprilisina/metabolismo , Aminobutiratos/metabolismo , Compostos de Bifenilo/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Combinação de Medicamentos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Humanos , Ligação de Hidrogênio , Modelos Moleculares , Neprilisina/antagonistas & inibidores , Domínios Proteicos , Tetrazóis/metabolismo , Valsartana
7.
ACS Med Chem Lett ; 5(7): 787-92, 2014 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-25050166

RESUMO

A cis-configured 3,5-disubstituted piperidine direct renin inhibitor, (syn,rac)-1, was discovered as a high-throughput screening hit from a target-family tailored library. Optimization of both the prime and the nonprime site residues flanking the central piperidine transition-state surrogate resulted in analogues with improved potency and pharmacokinetic (PK) properties, culminating in the identification of the 4-hydroxy-3,5-substituted piperidine 31. This compound showed high in vitro potency toward human renin with excellent off-target selectivity, 60% oral bioavailability in rat, and dose-dependent blood pressure lowering effects in the double-transgenic rat model.

8.
J Biomol Screen ; 19(6): 870-7, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24487225

RESUMO

Fluorescence lifetime (FLT)-based assays have developed to become highly attractive tools in drug discovery. All recently published examples of FLT-based assays essentially describe their use for monitoring enzyme-mediated peptide modifications, such as proteolytic cleavage or phosphorylation/dephosphorylation. Here we report the development of competitive binding assays as novel, inhibitor-centric assays, principally employing the FLT of the acridone dye Puretime 14 (PT14) as the readout parameter. Exemplified with two case studies on human serine proteases, the details of the rationale for both the design and synthesis of probes (i.e., active site-directed low-molecular-weight inhibitors conjugated to PT14) are provided. Data obtained from testing inhibitors with the novel assay format match those obtained with alternative formats such as FLT-based protease activity and time-resolved fluorescence resonance energy transfer-based competitive binding assays.


Assuntos
Descoberta de Drogas/métodos , Inibidores de Proteases/química , Espectrometria de Fluorescência/métodos , Acridonas/química , Ligação Competitiva , Soluções Tampão , Domínio Catalítico , Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes/química , Humanos , Concentração de Íons de Hidrogênio , Concentração Inibidora 50 , Cinética , Pulmão/enzimologia , Conformação Molecular , Peso Molecular , Peptídeos/química , Ligação Proteica , Proteínas Recombinantes/química , Serina Proteases/química , Triptases/química
9.
Proc Natl Acad Sci U S A ; 108(52): 21052-6, 2011 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-22160684

RESUMO

Hepatitis C virus (HCV) infection is a global health burden with over 170 million people infected worldwide. In a significant portion of patients chronic hepatitis C infection leads to serious liver diseases, including fibrosis, cirrhosis, and hepatocellular carcinoma. The HCV NS3 protein is essential for viral polyprotein processing and RNA replication and hence viral replication. It is composed of an N-terminal serine protease domain and a C-terminal helicase/NTPase domain. For full activity, the protease requires the NS4A protein as a cofactor. HCV NS3/4A protease is a prime target for developing direct-acting antiviral agents. First-generation NS3/4A protease inhibitors have recently been introduced into clinical practice, markedly changing HCV treatment options. To date, crystal structures of HCV NS3/4A protease inhibitors have only been reported in complex with the protease domain alone. Here, we present a unique structure of an inhibitor bound to the full-length, bifunctional protease-helicase NS3/4A and show that parts of the P4 capping and P2 moieties of the inhibitor interact with both protease and helicase residues. The structure sheds light on inhibitor binding to the more physiologically relevant form of the enzyme and supports exploring inhibitor-helicase interactions in the design of the next generation of HCV NS3/4A protease inhibitors. In addition, small angle X-ray scattering confirmed the observed protease-helicase domain assembly in solution.


Assuntos
Proteínas de Transporte/química , Hepatite C/tratamento farmacológico , Modelos Moleculares , Inibidores de Proteases/química , Conformação Proteica , Proteínas não Estruturais Virais/química , Proteínas de Transporte/metabolismo , Cromatografia em Gel , Cristalização , Cristalografia por Raios X , Escherichia coli , Concentração Inibidora 50 , Peptídeos e Proteínas de Sinalização Intracelular , Inibidores de Proteases/metabolismo , Inibidores de Proteases/uso terapêutico , Espalhamento a Baixo Ângulo , Proteínas não Estruturais Virais/metabolismo
10.
Proteins ; 78(16): 3281-91, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20886466

RESUMO

(19)F NMR screening of fluorinated fragments with different Local Environment of Fluorine, a.k.a. LEF library, is an experimental methodology which, beyond providing useful starting fragments for fragment-based drug discovery projects, offers, in combination with crystal and computational analysis, an approach for the identification of fluorophilic hot-spots in the proteins of interest. The application of this approach in the identification of fluorinated fragments binding to the serine protease trypsin, and the X-ray structures of the complexes are presented. The specific nature of the observed fluorine-protein interactions is discussed and compared with the interactions detected for other fluorinated ligands reported in the protein data bank. The presence of similar 3D arrangements of protein atoms at the fluorine sub-sites is identified with a newly developed tool. In this approach, protein sub-sites are extracted around each fluorine contained in the protein data bank and compared with the query of interest by using a pharmacophoric description.


Assuntos
Biologia Computacional/métodos , Compostos de Flúor/química , Tripsina/química , Animais , Bovinos , Cristalografia por Raios X , Bases de Dados de Proteínas , Flúor/química , Halogenação , Espectroscopia de Ressonância Magnética
11.
J Biomol Screen ; 15(9): 1029-41, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20855559

RESUMO

Fragment-based screening (FBS) has gained acceptance in the pharmaceutical industry as an attractive approach for the identification of new chemical starting points for drug discovery programs in addition to classical strategies such as high-throughput screening. There is the concern that screening of fragments at high µM concentrations in biochemical assays results in increased false-positive and false-negative rates. Here the authors systematically compare the data quality of FBS obtained by enzyme activity-based fluorescence intensity, fluorescence lifetime, and mobility shift assays with the data quality from surface plasmon resonance (SPR) and nuclear magnetic resonance (NMR) methods. The serine protease trypsin and the matrix metalloprotease MMP12 were selected as model systems. For both studies, 352 fragments were selected each. From the data generated, all 3 biochemical protease assay methods can be used for screening of fragments with low false-negative and low false-positive rates, comparable to those achieved with the SPR-based assays. It can also be concluded that only fragments with a solubility higher than the screening concentration determined by means of NMR should be used for FBS purposes. Extrapolated to 10,000 fragments, the biochemical assays speed up the primary FBS process by approximately a factor of 10 and reduce the protease consumption by approximately 10,000-fold compared to NMR protein observation experiments.


Assuntos
Bioensaio/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Metaloproteinase 12 da Matriz/metabolismo , Fragmentos de Peptídeos/análise , Tripsina/metabolismo , Animais , Bovinos , Cromatografia Líquida , Reações Falso-Negativas , Reações Falso-Positivas , Estudos de Viabilidade , Fluorescência , Humanos , Cinética , Luz , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Fragmentos de Peptídeos/química , Espalhamento de Radiação , Solubilidade , Ressonância de Plasmônio de Superfície
13.
Cancer Lett ; 289(2): 228-36, 2010 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-19783361

RESUMO

Point mutations emerge as one of the rate-limiting steps in tumor response to small molecule inhibitors of protein kinases. Here we characterized the response of the MET mutated variants, V1110I, V1238I, V1206L and H1112L to the small molecule SU11274. Our results reveal a distinct inhibition pattern of the four mutations with IC(50) values for autophosphorylation inhibition ranging between 0.15 and 1.5muM. Differences were further seen on the ability of SU11274 to inhibit phosphorylation of downstream MET transducers such as AKT, ERK, PLCgamma and STAT3 and a variety of MET-dependent biological endpoints. In all the assays, H1112L was the most sensitive to SU11274, while V1206L was less affected under the used concentration range. The differences in responses to SU11274 are discussed based on a structural model of the MET kinase domain.


Assuntos
Indóis/farmacologia , Mutação/efeitos dos fármacos , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-met/química , Proteínas Proto-Oncogênicas c-met/genética , Sulfonamidas/farmacologia , Animais , Western Blotting , Adesão Celular/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mutação/genética , Células NIH 3T3 , Proteína Oncogênica v-akt/metabolismo , Fosfolipase C gama/metabolismo , Fosforilação/efeitos dos fármacos , Conformação Proteica , Proteínas Proto-Oncogênicas c-met/metabolismo , Fator de Transcrição STAT3/metabolismo , Cicatrização/efeitos dos fármacos
14.
Artigo em Inglês | MEDLINE | ID: mdl-16511290

RESUMO

Both dengue and West Nile virus infections are an increasing risk to humans, not only in tropical and subtropical areas, but also in North America and parts of Europe. These viral infections are generally transmitted by mosquitoes, but may also be tick-borne. Infection usually results in mild flu-like symptoms, but can also cause encephalitis and fatalities. Approximately 2799 severe West Nile virus cases were reported this year in the United States, resulting in 102 fatalities. With this alarming increase in the number of West Nile virus infections in western countries and the fact that dengue virus already affects millions of people per year in tropical and subtropical climates, there is a real need for effective medicines. A possible therapeutic target to combat these viruses is the protease, which is essential for virus replication. In order to provide structural information to help to guide a lead identification and optimization program, crystallizations of the NS2B-NS3 protease complexes from both dengue and West Nile viruses have been initiated. Crystals that diffract to high resolution, suitable for three-dimensional structure determinations, have been obtained.


Assuntos
Cisteína Endopeptidases/isolamento & purificação , Vírus da Dengue/enzimologia , Serina Endopeptidases/isolamento & purificação , Proteínas não Estruturais Virais/isolamento & purificação , Vírus do Nilo Ocidental/enzimologia , Cristalização , Cristalografia por Raios X , Cisteína Endopeptidases/química , Vírus da Dengue/isolamento & purificação , Humanos , Complexos Multiproteicos/química , Complexos Multiproteicos/isolamento & purificação , Serina Endopeptidases/química , Proteínas não Estruturais Virais/química , Vírus do Nilo Ocidental/isolamento & purificação
15.
Nat Struct Mol Biol ; 13(4): 372-3, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16532006

RESUMO

The replication of flaviviruses requires the correct processing of their polyprotein by the viral NS3 protease (NS3pro). Essential for the activation of NS3pro is a 47-residue region of NS2B. Here we report the crystal structures of a dengue NS2B-NS3pro complex and a West Nile virus NS2B-NS3pro complex with a substrate-based inhibitor. These structures identify key residues for NS3pro substrate recognition and clarify the mechanism of NS3pro activation.


Assuntos
Vírus da Dengue/enzimologia , Serina Endopeptidases/química , Serina Endopeptidases/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Vírus do Nilo Ocidental/enzimologia , Sítios de Ligação , Ativação Enzimática , Ligação de Hidrogênio , Substâncias Macromoleculares , Modelos Moleculares , Oligopeptídeos , Inibidores de Proteases/química , Inibidores de Proteases/metabolismo , RNA Helicases/química , RNA Helicases/metabolismo
16.
Biochemistry ; 44(43): 14110-9, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16245927

RESUMO

Met receptor tyrosine kinase plays a crucial role in the regulation of a large number of cellular processes and, when deregulated by overexpression or mutations, leads to tumor growth and invasion. The Y1235D mutation identified in metastases was shown to induce constitutive activation and a motile-invasive phenotype on transduced carcinoma cells. Wild-type Met activation requires phosphorylation of both Y1234 and Y1235 in the activation loop. We mapped the major phosphorylation sites in the kinase domain of a recombinant Met protein and identified the known residues Y1234 and Y1235 as well as a new phosphorylation site at Y1194 in the hinge region. Combining activating and silencing mutations at these sites, we characterized in depth the mechanism of activation of wild-type and mutant Met proteins. We found that the phosphotyrosine mimetic mutation Y1235D is sufficient to confer constitutive kinase activity, which is not influenced by phosphorylation at Y1234. However, the specific activity of this mutant was lower than that observed for fully activated wild-type Met and induced less phosphorylation of Y1349 in the signaling site, indicating that this mutation cannot entirely compensate for a phosphorylated tyrosine at this position. The Y1194F silencing mutation yielded an enzyme that could be activated to a similar extent as the wild type but with significantly slower activation kinetics, underlying the importance of this residue, which is conserved among different tyrosine kinase receptors. Finally, we observed different interactions of wild-type and mutant Met with the inhibitor K252a that may have therapeutic implications for the selective inhibition of this kinase.


Assuntos
Mutação , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Ácido Aspártico/genética , Sequência de Bases , Sítios de Ligação , Carbazóis/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Inativação Gênica , Humanos , Alcaloides Indólicos , Cinética , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Fenótipo , Fosforilação , Fosfotirosina/química , Fosfotirosina/metabolismo , Conformação Proteica , Proteínas Proto-Oncogênicas c-met/química , Proteínas Proto-Oncogênicas c-met/genética , Receptores Proteína Tirosina Quinases/química , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução Genética , Células Tumorais Cultivadas , Tirosina/genética
17.
Proc Natl Acad Sci U S A ; 100(22): 12654-9, 2003 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-14559966

RESUMO

The protooncogene c-met codes for the hepatocyte growth factor receptor tyrosine kinase. Binding of its ligand, hepatocyte growth factor/scatter factor, stimulates receptor autophosphorylation, which leads to pleiotropic downstream signaling events in epithelial cells, including cell growth, motility, and invasion. These events are mediated by interaction of cytoplasmic effectors, generally through Src homology 2 (SH2) domains, with two phosphotyrosine-containing sequence motifs in the unique C-terminal tail of c-Met (supersite). There is a strong link between aberrant c-Met activity and oncogenesis, which makes this kinase an important cancer drug target. The furanosylated indolocarbazole K-252a belongs to a family of microbial alkaloids that also includes staurosporine. It was recently shown to be a potent inhibitor of c-Met. Here we report the crystal structures of an unphosphorylated c-Met kinase domain harboring a human cancer mutation and its complex with K-252a at 1.8-A resolution. The structure follows the well established architecture of protein kinases. It adopts a unique, inhibitory conformation of the activation loop, a catalytically noncompetent orientation of helix alphaC, and reveals the complete C-terminal docking site. The first SH2-binding motif (1349YVHV) adopts an extended conformation, whereas the second motif (1356YVNV), a binding site for Grb2-SH2, folds as a type II Beta-turn. The intermediate portion of the supersite (1353NATY) assumes a type I Beta-turn conformation as in an Shc-phosphotyrosine binding domain peptide complex. K-252a is bound in the adenosine pocket with an analogous binding mode to those observed in previously reported structures of protein kinases in complex with staurosporine.


Assuntos
Carbazóis/química , Inibidores Enzimáticos/química , Proteínas Tirosina Quinases/química , Proteínas Proto-Oncogênicas c-met/química , Sequência de Aminoácidos , Sítios de Ligação , Clonagem Molecular , Cristalografia por Raios X/métodos , Alcaloides Indólicos , Metionina , Modelos Moleculares , Conformação Proteica , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Recombinantes/química , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...