Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Bioorg Med Chem Lett ; 29(17): 2503-2510, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31327531

RESUMO

Cancer cells reprogram their metabolism to support growth and to mitigate cellular stressors. The serine synthesis pathway has been identified as a metabolic pathway frequently altered in cancers and there has been considerable interest in developing pharmacological agents to target this pathway. Here, we report a series of indole amides that inhibit human 3-phosphoglycerate dehydrogenase (PHGDH), the enzyme that catalyzes the first committed step of the serine synthesis pathway. Using X-ray crystallography, we show that the indole amides bind the NAD+ pocket of PHGDH. Through structure-based optimization we were able to develop compounds with low nanomolar affinities for PHGDH in an enzymatic IC50 assay. In cellular assays, the most potent compounds inhibited de novo serine synthesis with low micromolar to sub-micromolar activities and these compounds successfully abrogated the proliferation of cancer cells in serine free media. The indole amide series reported here represent an important improvement over previously published PHGDH inhibitors as they are markedly more potent and their mechanism of action is better defined.


Assuntos
Amidas/química , Inibidores Enzimáticos/química , Indóis/química , Fosfoglicerato Desidrogenase/antagonistas & inibidores , Serina/biossíntese , Amidas/metabolismo , Amidas/farmacologia , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cristalografia por Raios X , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Simulação de Dinâmica Molecular , Fosfoglicerato Desidrogenase/metabolismo , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
2.
J Biol Chem ; 288(31): 22758-67, 2013 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-23792959

RESUMO

Phosphorylation of inhibitor of nuclear transcription factor κB (IκB) by IκB kinase (IKK) triggers the degradation of IκB and migration of cytoplasmic κB to the nucleus where it promotes the transcription of its target genes. Activation of IKK is achieved by phosphorylation of its main subunit, IKKß, at the activation loop sites. Here, we report the 2.8 Å resolution crystal structure of human IKKß (hIKKß), which is partially phosphorylated and bound to the staurosporine analog K252a. The hIKKß protomer adopts a trimodular structure that closely resembles that from Xenopus laevis (xIKKß): an N-terminal kinase domain (KD), a central ubiquitin-like domain (ULD), and a C-terminal scaffold/dimerization domain (SDD). Although hIKKß and xIKKß utilize a similar dimerization mode, their overall geometries are distinct. In contrast to the structure resembling closed shears reported previously for xIKKß, hIKKß exists as an open asymmetric dimer in which the two KDs are further apart, with one in an active and the other in an inactive conformation. Dimer interactions are limited to the C-terminal six-helix bundle that acts as a hinge between the two subunits. The observed domain movements in the structures of IKKß may represent trans-phosphorylation steps that accompany IKKß activation.


Assuntos
Quinase I-kappa B/química , Cristalização , Cristalografia por Raios X , Dimerização , Humanos , Quinase I-kappa B/antagonistas & inibidores , Quinase I-kappa B/metabolismo , Ligantes , Modelos Moleculares , Fosforilação
3.
J Biol Chem ; 288(24): 17408-19, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23632026

RESUMO

The immunoglobulin new antigen receptors (IgNARs) are a class of Ig-like molecules of the shark immune system that exist as heavy chain-only homodimers and bind antigens by their single domain variable regions (V-NARs). Following shark immunization and/or in vitro selection, V-NARs can be generated as soluble, stable, and specific high affinity monomeric binding proteins of ∼12 kDa. We have previously isolated a V-NAR from an immunized spiny dogfish shark, named E06, that binds specifically and with high affinity to human, mouse, and rat serum albumins. Humanization of E06 was carried out by converting over 60% of non-complementarity-determining region residues to those of a human germ line Vκ1 sequence, DPK9. The resulting huE06 molecules have largely retained the specificity and affinity of antigen binding of the parental V-NAR. Crystal structures of the shark E06 and its humanized variant (huE06 v1.1) in complex with human serum albumin (HSA) were determined at 3- and 2.3-Å resolution, respectively. The huE06 v1.1 molecule retained all but one amino acid residues involved in the binding site for HSA. Structural analysis of these V-NARs has revealed an unusual variable domain-antigen interaction. E06 interacts with HSA in an atypical mode that utilizes extensive framework contacts in addition to complementarity-determining regions that has not been seen previously in V-NARs. On the basis of the structure, the roles of various elements of the molecule are described with respect to antigen binding and V-NAR stability. This information broadens the general understanding of antigen recognition and provides a framework for further design and humanization of shark IgNARs.


Assuntos
Anticorpos Monoclonais Humanizados/química , Tubarões/imunologia , Anticorpos de Cadeia Única/química , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Cristalografia por Raios X , Proteínas de Peixes , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Engenharia de Proteínas , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Ratos , Homologia de Sequência de Aminoácidos , Albumina Sérica/química
4.
J Med Chem ; 67(18): 16773-16795, 2024 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-39231272

RESUMO

Inhibition of Hepatitis B Virus (HBV) replication by small molecules that modulate capsid assembly and the encapsidation of pgRNA and viral polymerase by HBV core protein is a clinically validated approach toward the development of new antivirals. Through definition of a minimal pharmacophore, a series of isoquinolinone-based capsid assembly modulators (CAMs) was identified. Structural biology analysis revealed that lead molecules possess a unique binding mode, exploiting electrostatic interactions with accessible phenylalanine and tyrosine residues. Key analogs demonstrated excellent primary potency, absorption, distribution, metabolism, and excretion (ADME) and pharmacokinetic properties, and efficacy in a mouse model of HBV. The optimized lead also displayed potent inhibition of capsid uncoating in HBV-infected HepG2 cells expressing the sodium-taurocholate cotransporting polypeptide (NTCP) receptor, affecting the generation of HBsAg and cccDNA establishment. Based on these results, isoquinolinone derivative AB-836 was advanced into clinical development. In Phase 1b trials, AB-836 demonstrated >3 log10 reduction in serum HBV DNA, however, further development was discontinued due to the observation of incidental alanine aminotransferase (ALT) elevations.


Assuntos
Antivirais , Desenho de Fármacos , Vírus da Hepatite B , Humanos , Relação Estrutura-Atividade , Vírus da Hepatite B/efeitos dos fármacos , Animais , Antivirais/farmacologia , Antivirais/síntese química , Antivirais/química , Antivirais/farmacocinética , Camundongos , Células Hep G2 , Capsídeo/efeitos dos fármacos , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/antagonistas & inibidores , Isoquinolinas/farmacologia , Isoquinolinas/química , Isoquinolinas/síntese química , Quinolonas/farmacologia , Quinolonas/síntese química , Quinolonas/química , Montagem de Vírus/efeitos dos fármacos
5.
RSC Med Chem ; 13(3): 343-349, 2022 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-35434625

RESUMO

Disruption of the HBV viral life cycle with small molecules that prevent the encapsidation of pregenomic RNA and viral polymerase through binding to HBV core protein is a clinically validated approach to inhibiting HBV viral replication. Herein we report the further optimisation of clinical candidate AB-506 through core modification with a focus on increasing oral exposure and oral half-life. Maintenance of high levels of anti-HBV cellular potency in conjunction with improvements in pharmacokinetic properties led to multi-log10 reductions in serum HBV DNA following low, once-daily oral dosing for key analogues in a preclinical animal model of HBV replication.

6.
Bioorg Med Chem Lett ; 21(15): 4602-7, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21715165

RESUMO

An extension of our previously reported series of macrocyclic ortho-aminobenzamide Hsp90 inhibitors is reported. Addition of a second methyl group to the tether provided analogs that show increased potency in binding as well as cell-proliferation assays and, more importantly, are stable toward microsomes. We wish to disclose the discovery of a macrocycle which showed impressive biomarker activity 24-h post dosing and which demonstrated prolonged exposure in tumors. When studied in a lung cancer xenograft model, the compound demonstrated significant tumor size reduction.


Assuntos
Antineoplásicos/química , Benzamidas/química , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Neoplasias Pulmonares/tratamento farmacológico , Compostos Macrocíclicos/química , Administração Oral , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Benzamidas/farmacocinética , Benzamidas/uso terapêutico , Sítios de Ligação , Biomarcadores/metabolismo , Avaliação Pré-Clínica de Medicamentos , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Camundongos , Camundongos Nus , Microssomos Hepáticos/metabolismo , Estrutura Terciária de Proteína , Ratos , Transplante Heterólogo
7.
Bioengineered ; 12(2): 11076-11086, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34895054

RESUMO

To further elucidate the mechanism of action and binding properties of eptinezumab to calcitonin gene-related peptide (CGRP), X-ray crystallography, computational alanine scanning, and molecular dynamics were used. X-ray diffraction data were collected to determine the three-dimensional structures of the unbound eptinezumab antigen-binding fragment (Fab) and the Fab:CGRP complex. Molecular dynamics simulations were performed to analyze the transition between uncomplexed and complex states. The amidated C-terminus of CGRP was shown to bind in a pocket formed by the Fab heavy and light chains. There was extensive contact between all six complementarity-determining regions (CDRs; composed of light-chain [L1, L2, and L3] and heavy-chain [H1, H2, H3]) of eptinezumab and CGRP. The complex demonstrated a high ligand-binding surface area dominated by aromatic residues. CDR L3 contains a disulfide bond that stabilizes the loop, contributes surface area to the binding pocket, and provides van der Waals contacts. Comparison of the uncomplexed and complex structures revealed motion near the binding cleft. The CDR loops H2 and H3 were displaced ~1.4-2.0 Å and residue H-Tyr33 changed conformation, creating a 'latch-and-lock' mechanism for binding CGRP and preventing dissociation. Computational alanine scanning of CGRP identified energetic 'hot spots' that contribute to binding energy; mutating these positions to residues in homologous neuropeptides resulted in unfavorable binding energies. The attributes of the Fab region and the conformational changes that occur in eptinezumab during binding to CGRP contribute to the specificity, durability, and strength of the interaction, and likely underlie the rapid and sustained migraine preventive effect observed in clinical studies.


Assuntos
Anticorpos Monoclonais Humanizados/química , Peptídeo Relacionado com Gene de Calcitonina/química , Epitopos/química , Humanos , Ligação de Hidrogênio , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Estabilidade Proteica , Água/química , Difração de Raios X
9.
Bioorg Med Chem Lett ; 20(22): 6597-605, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20880704

RESUMO

The identification of small molecule aminohydantoins as potent and selective human ß-secretase inhibitors is reported. These analogs exhibit good brain permeability (40-70%), low nanomolar potency for BACE1, and demonstrate >100-fold selectivity for the structurally related aspartyl proteases cathepsin D, renin and pepsin. Alkyl and alkoxy groups at the meta-position of the P1 phenyl, which extend toward the S3 region of the enzyme, have contributed to the ligand's reduced affinity for the efflux transporter protein P-gp, and decreased topological polar surface area, thus resulting in enhanced brain permeability. A fluorine substitution at the para-position of the P1 phenyl has contributed to 100-fold decrease of CYP3A4 inhibition and enhancement of compound metabolic stability. The plasma and brain protein binding properties of these new analogs are affected by substitutions at the P1 phenyl moiety. Higher compound protein binding was observed in the brain than in the plasma. Two structurally diverse potent BACE1 inhibitors (84 and 89) reduced 30% plasma Aß40 in the Tg2576 mice in vivo model at 30 mg/kg p.o..


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Encéfalo/metabolismo , Inibidores Enzimáticos/síntese química , Hidantoínas/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Hidantoínas/química , Hidantoínas/farmacologia , Permeabilidade
10.
Bioorg Med Chem Lett ; 20(7): 2068-73, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20223661

RESUMO

The proteolytic enzyme beta-secretase (BACE1) plays a central role in the synthesis of the pathogenic beta-amyloid in Alzheimer's disease. Recently, we reported small molecule acylguanidines as potent BACE1 inhibitors. However, many of these acylguanidines have a high polar surface area (e.g. as measured by the topological polar surface area or TPSA), which is unfavorable for crossing the blood-brain barrier. Herein, we describe the identification of the 2-aminopyridine moiety as a bioisosteric replacement of the acylguanidine moiety, which resulted in inhibitors with lower TPSA values and superior brain penetration. X-ray crystallographic studies indicated that the 2-aminopyridine moiety interacts directly with the catalytic aspartic acids Asp32 and Asp228 via a hydrogen-bonding network.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Aminopiridinas/química , Aminopiridinas/farmacocinética , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/metabolismo , Encéfalo/metabolismo , Doença de Alzheimer/enzimologia , Aminopiridinas/farmacologia , Secretases da Proteína Precursora do Amiloide/química , Ácido Aspártico Endopeptidases/química , Cristalografia por Raios X , Humanos , Modelos Moleculares , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa