Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Pharmacol Exp Ther ; 338(1): 134-42, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21505059

RESUMO

N-[4-(3-amino-1H-indazol-4-yl)phenyl]-N1-(2-fluoro-5-methyl phenyl)-urea (ABT-869) is a novel multitargeted receptor tyrosine kinase inhibitor that demonstrates single-agent activity in preclinical studies and has undergone phase I and II clinical trials. We characterized the mechanism of action of ABT-869 by examining vascular changes after treatment (25 mg/kg per day) in HT1080 fibrosarcoma and SW620 colon carcinoma cells, using immunohistochemistry, dynamic contrast enhanced-magnetic resonance imaging (DCE-MRI), and hypoxic protein detection. We observed the inhibition of vascular endothelial growth factor receptor 2 and platelet-derived growth factor receptor ß phosphorylation in both tumors and changes in tumor vasculature. Reductions in microvessel density and diameter were observed. Vascular-wall integrity was assessed by colocalization of pericytes and basement membrane. Although both microvessel density and total number of pericytes decreased with treatment, the percentage of pericyte coverage on remaining vessels significantly increased. These data suggest the selective ablation of microvessels lacking pericyte coverage. Functional vascular measures DCE-MRI and hypoxia formation were also tested. After 2 days of treatment on the HT1080 model, vascular permeability, K(trans), was reduced by >60% and hypoxic tumor fraction was significantly decreased, which was also seen in the SW620 tumors after 4 days of treatment. Taken together, decreases in vascular permeability and changes in vascular integrity observed in these studies define the mode of action of ABT-869 and may aid in optimizing the timing of therapeutic window for combination therapies.


Assuntos
Permeabilidade Capilar/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Indazóis/farmacologia , Indazóis/uso terapêutico , Neovascularização Patológica/tratamento farmacológico , Compostos de Fenilureia/farmacologia , Compostos de Fenilureia/uso terapêutico , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Permeabilidade Capilar/fisiologia , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos/métodos , Endotélio Vascular/metabolismo , Humanos , Camundongos , Neovascularização Patológica/enzimologia , Neovascularização Patológica/patologia , Receptores Proteína Tirosina Quinases/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
2.
J Immunol ; 181(2): 1282-7, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18606682

RESUMO

Preclinical animal studies have shown that Ab12.6, an agonistic human Ab targeting the erythropoietin receptor (EPOR), exhibits several potential dosing and safety features that make it an attractive clinical candidate for the treatment of anemia. Ab12.6 was derived by yeast display affinity maturation of parental Ab12, a strategy initially intended to improve off-rate and affinity for EPOR, thereby enhancing erythropoietic activity. Analysis of full-length IgGs derived from yeast clones identified sequences within Ab12 CDRH2 that independently influenced both affinity and potency. The Ab12.6 derivative displayed improved in vitro potency and in vivo efficacy, although its binding affinity to the EPOR was lower than that of the parent Ab12. Additional Ab12 derivatives also exhibited an inverse correlation between affinity and potency. These results suggest that for this class of agonistic Abs, faster off-rates may permit continuous receptor stimulation and more efficient erythropoiesis.


Assuntos
Anticorpos/imunologia , Afinidade de Anticorpos , Eritropoese , Eritropoetina/metabolismo , Receptores da Eritropoetina/agonistas , Anticorpos/metabolismo , Eritropoetina/imunologia , Humanos , Mimetismo Molecular , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo
3.
Blood ; 110(7): 2408-13, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17620453

RESUMO

Recombinant human erythropoietin (rHu-EPO) is used to treat anemia by activating the erythropoietin receptor (EPOR) in erythroid progenitor cells, leading to proliferation and differentiation into mature red blood cells. To allow less frequent dosing, a hyperglycosylated version of EPO has been developed with a longer half-life. In principle, an agonistic antibody targeting EPOR would offer an even longer half-life, support robust monthly dosing, and, unlike EPO products, reduce the risk of pure red cell aplasia. The efficiency of signaling and corresponding potency of previously reported antibody mimics are generally suboptimal compared with EPO and not suitable for clinical use. Here we describe a potent, fully human, agonistic antibody (ABT007) targeting EPOR that supports potent, more sustained, and less pulsatile elevation of hematocrit in a human EPOR-expressing transgenic mouse model compared with standard doses of rHu-EPO while requiring less frequent dosing. Resolution of the crystal structure of the EPOR extracellular domain (ECD) complexed to the ABT007 Fab fragment, determined at 0.32 nm, identifies a binding site that is consistent with a novel mechanism of receptor activation based on a unique antibody-imposed conformational change. These results demonstrate that a symmetric molecule can serve as a potent activator of the EPOR.


Assuntos
Anticorpos/imunologia , Eritropoetina/metabolismo , Mimetismo Molecular , Animais , Sítios de Ligação , Linhagem Celular , Cristalografia por Raios X , Eritropoese , Hematócrito , Humanos , Camundongos , Camundongos Knockout , Modelos Moleculares , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Receptores da Eritropoetina/química , Receptores da Eritropoetina/deficiência , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Homologia Estrutural de Proteína
4.
Bioorg Med Chem Lett ; 17(10): 2817-22, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17350258

RESUMO

A series of aryl sulfonamides of 5,6-disubstituted anthranilic acids were identified as potent inhibitors of methionine aminopeptidase-2 (MetAP2). Small alkyl groups and 3-furyl were tolerated at the 5-position of anthranilic acid, while -OCH(3), CH(3), and Cl were found optimal for the 6-position. Placement of 2-aminoethoxy group at the 6-position enabled interaction with the second Mn(2+) but did not result in enhancement in potency. Introduction of a tertiary amino moiety at the ortho-position of the sulfonyl phenyl ring gave reduced protein binding and improved cellular activity, but led to lower oral bioavailability.


Assuntos
Aminopeptidases/antagonistas & inibidores , Chumbo/química , Metaloendopeptidases/antagonistas & inibidores , Sulfonamidas/química , ortoaminobenzoatos/farmacologia , Aminopeptidases/química , Metaloendopeptidases/química , Modelos Moleculares , Estrutura Molecular , Conformação Proteica , Relação Estrutura-Atividade , ortoaminobenzoatos/química
5.
J Med Chem ; 49(13): 3832-49, 2006 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-16789740

RESUMO

Methionine aminopeptidase-2 (MetAP2) is a novel target for cancer therapy. As part of an effort to discover orally active reversible inhibitors of MetAP2, a series of anthranilic acid sulfonamides with micromolar affinities for human MetAP2 were identified using affinity selection by mass spectrometry (ASMS) screening. These micromolar hits were rapidly improved to nanomolar leads on the basis of insights from protein crystallography; however, the compounds displayed extensive binding to human serum albumin and had limited activity in cellular assays. Modifications based on structural information on the binding of lead compounds to both MetAP2 and domain III of albumin allowed the identification of compounds with significant improvements in both parameters, which showed good cellular activity in both proliferation and methionine processing assays.


Assuntos
Aminopeptidases/química , Antineoplásicos/síntese química , Metaloendopeptidases/química , Albumina Sérica/química , Sulfonamidas/síntese química , ortoaminobenzoatos/síntese química , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Técnicas In Vitro , Espectrometria de Massas , Metionina/metabolismo , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Ratos , Relação Estrutura-Atividade , Sulfonamidas/química , Sulfonamidas/farmacologia , ortoaminobenzoatos/química , ortoaminobenzoatos/farmacologia
6.
Bioorg Med Chem Lett ; 16(13): 3574-7, 2006 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-16632353

RESUMO

We have screened molecules for inhibition of MetAP2 as a novel approach toward antiangiogenesis and anticancer therapy using affinity selection/mass spectrometry (ASMS) employing MetAP2 loaded with Mn(2+) as the active site metal. After a series of anthranilic acid sulfonamides with micromolar affinities was identified, chemistry efforts were initiated. The micromolar hits were quickly improved to potent nanomolar inhibitors by chemical modifications guided by insights from X-ray crystallography.


Assuntos
Aminopeptidases/antagonistas & inibidores , Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Glicoproteínas/antagonistas & inibidores , Sulfonamidas/farmacologia , Inibidores da Angiogênese/síntese química , Inibidores da Angiogênese/química , Antineoplásicos/síntese química , Antineoplásicos/química , Sítios de Ligação , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Cristalografia por Raios X , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Manganês/química , Espectrometria de Massas/métodos , Metionil Aminopeptidases , Modelos Moleculares , Estrutura Molecular , Sensibilidade e Especificidade , Estereoisomerismo , Relação Estrutura-Atividade , Sulfonamidas/síntese química , Sulfonamidas/química
7.
J Med Chem ; 48(8): 2838-46, 2005 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-15828822

RESUMO

The heptapeptide 1, NAc-Gly-Val-DIle-Thr-Arg-Ile-ArgNHEt, a structurally modified fragment derived from the second type-1 repeat of thrombospondin-1 (TSP-1), is known to possess antiangiogenic activity. However, therapeutic utility could not be demonstrated because this peptide has a very short half-life in rodents. To optimize the PD/PK profile of 1, we initiated a systematic SAR study. The initial structural modifications were performed at positions 5 and 7 of peptide 1 and at the N- and C-termini. Out of several hundred peptides synthesized, the nonapeptide 5 (ABT-526) emerged as a promising lead. ABT-526 inhibited VEGF-induced HMVEC cell migration and tube formation in the nanomolar range and increased apoptosis of HUAEC cells. ABT-526 showed acceptable PK in rodents, dog, and monkey. ABT-526, when incorporated in an angiogenic pellet implanted in the rat cornea at 10 microM, reduced neovascularization by 92%. Substitution of DalloIle in place of DIle in ABT-526 provided nonapeptide 6 (ABT-510), which was 30-fold less active than ABT-526 in the EC migration but 20-fold more active in the tube formation assay. In comparison to ABT-526, ABT-510 has increased water solubility and slower clearance in dog and monkey. Radiolabeled ABT-510 demonstrated saturable binding to HMVEC cells at 0.02-20 nM concentrations and was displaceable by TSP-1. ABT-510 and ABT-526 were shown to significantly increase apoptosis of HUAEC cells. ABT-510 was effective in blocking neovascularization in the mouse Matrigel plug model and inhibited tumor growth in the mouse Lewis lung carcinoma model. Previous studies had shown that ABT-510 was effective in inhibiting the outgrowth of murine melanoma metastases in syngeneic mice and in blocking the growth of human bladder carcinoma implanted in nude mice. It had been also shown that ABT-510 could regress tumor lesions in pet dogs or cause unexpected stabilization of the disease in advanced canine cancer. ABT-526 and ABT-510 are the first compounds in the class of potent inhibitors of angiogenesis that mimic the antiangiogenic function of TSP-1. ABT-510 is currently in phase II clinical studies.


Assuntos
Inibidores da Angiogênese/síntese química , Oligopeptídeos/síntese química , Trombospondina 1/química , Inibidores da Angiogênese/farmacocinética , Inibidores da Angiogênese/farmacologia , Animais , Apoptose , Capilares/citologia , Quimiotaxia/efeitos dos fármacos , Córnea/irrigação sanguínea , Córnea/efeitos dos fármacos , Cães , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Endotélio Vascular/citologia , Feminino , Haplorrinos , Humanos , Técnicas In Vitro , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos C57BL , Mimetismo Molecular , Neovascularização Fisiológica/efeitos dos fármacos , Oligopeptídeos/farmacocinética , Oligopeptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Cordão Umbilical/citologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Bioorg Med Chem Lett ; 14(4): 865-8, 2004 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-15012983

RESUMO

Substituted 3-amino-2-hydroxyamides and related hydroxyamides and acylhydrazines were identified as inhibitors of human methionine aminopeptidase-2 (MetAP2). Examination of substituents through parallel synthesis and iterative structure-based design allowed the identification of potent inhibitors with good selectivity against MetAP1. Diacylhydrazine 3t (A-357300) was identified as an analogue displaying inhibition of methionine processing and cellular proliferation in human microvascular endothelial cells (HMVEC).


Assuntos
Amidas/química , Amidas/farmacologia , Aminopeptidases/antagonistas & inibidores , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Metaloendopeptidases/antagonistas & inibidores , Sítios de Ligação/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Humanos , Metionina/efeitos dos fármacos , Modelos Biológicos , Modelos Moleculares , Estrutura Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA