Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cancer Res ; 67(13): 6383-91, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17616698

RESUMO

Clinical studies with bortezomib have validated the proteasome as a therapeutic target for the treatment of multiple myeloma and non-Hodgkin's lymphoma. However, significant toxicities have restricted the intensity of bortezomib dosing. Here we describe the antitumor activity of PR-171, a novel epoxyketone-based irreversible proteasome inhibitor that is currently in clinical development. In comparison to bortezomib, PR-171 exhibits equal potency but greater selectivity for the chymotrypsin-like activity of the proteasome. In cell culture, PR-171 is more cytotoxic than bortezomib following brief treatments that mimic the in vivo pharmacokinetics of both molecules. Hematologic tumor cells exhibit the greatest sensitivity to brief exposure, whereas solid tumor cells and nontransformed cell types are less sensitive to such treatments. Cellular consequences of PR-171 treatment include the accumulation of proteasome substrates and induction of cell cycle arrest and/or apoptosis. Administration of PR-171 to animals results in the dose-dependent inhibition of the chymotrypsin-like proteasome activity in all tissues examined with the exception of the brain. PR-171 is well tolerated when administered for either 2 or 5 consecutive days at doses resulting in >80% proteasome inhibition in blood and most tissues. In human tumor xenograft models, PR-171 mediates an antitumor response that is both dose and schedule dependent. The antitumor efficacy of PR-171 delivered on 2 consecutive days is stronger than that of bortezomib administered on its clinical dosing schedule. These studies show the tolerability, efficacy, and dosing flexibility of PR-171 and provide validation for the clinical testing of PR-171 in the treatment of hematologic malignancies using dose-intensive schedules.


Assuntos
Antineoplásicos/farmacologia , Oligopeptídeos/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Apoptose , Ácidos Borônicos/farmacologia , Bortezomib , Quimotripsina/metabolismo , Quimotripsina/farmacologia , Relação Dose-Resposta a Droga , Humanos , Concentração Inibidora 50 , Masculino , Camundongos , Transplante de Neoplasias , Pirazinas/farmacologia , Ratos , Ratos Sprague-Dawley
2.
Mol Cancer Ther ; 13(4): 890-901, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24523301

RESUMO

Glutamine serves as an important source of energy and building blocks for many tumor cells. The first step in glutamine utilization is its conversion to glutamate by the mitochondrial enzyme glutaminase. CB-839 is a potent, selective, and orally bioavailable inhibitor of both splice variants of glutaminase (KGA and GAC). CB-839 had antiproliferative activity in a triple-negative breast cancer (TNBC) cell line, HCC-1806, that was associated with a marked decrease in glutamine consumption, glutamate production, oxygen consumption, and the steady-state levels of glutathione and several tricarboxylic acid cycle intermediates. In contrast, no antiproliferative activity was observed in an estrogen receptor-positive cell line, T47D, and only modest effects on glutamine consumption and downstream metabolites were observed. Across a panel of breast cancer cell lines, GAC protein expression and glutaminase activity were elevated in the majority of TNBC cell lines relative to receptor positive cells. Furthermore, the TNBC subtype displayed the greatest sensitivity to CB-839 treatment and this sensitivity was correlated with (i) dependence on extracellular glutamine for growth, (ii) intracellular glutamate and glutamine levels, and (iii) GAC (but not KGA) expression, a potential biomarker for sensitivity. CB-839 displayed significant antitumor activity in two xenograft models: as a single agent in a patient-derived TNBC model and in a basal like HER2(+) cell line model, JIMT-1, both as a single agent and in combination with paclitaxel. Together, these data provide a strong rationale for the clinical investigation of CB-839 as a targeted therapeutic in patients with TNBC and other glutamine-dependent tumors.


Assuntos
Antineoplásicos/administração & dosagem , Benzenoacetamidas/farmacologia , Inibidores Enzimáticos/administração & dosagem , Glutaminase/antagonistas & inibidores , Neoplasia de Células Basais/tratamento farmacológico , Tiadiazóis/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Administração Oral , Animais , Antineoplásicos/uso terapêutico , Benzenoacetamidas/uso terapêutico , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/uso terapêutico , Feminino , Humanos , Neoplasias Mamárias Experimentais , Camundongos , Camundongos SCID , Pessoa de Meia-Idade , Neoplasia de Células Basais/patologia , Sulfetos/administração & dosagem , Sulfetos/uso terapêutico , Tiadiazóis/administração & dosagem , Tiadiazóis/uso terapêutico , Neoplasias de Mama Triplo Negativas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
3.
J Med Chem ; 52(9): 3028-38, 2009 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-19348473

RESUMO

Proteasome inhibition has been validated as a therapeutic modality in the treatment of multiple myeloma and non-Hodgkin's lymphoma. Carfilzomib, an epoxyketone currently undergoing clinical trials in malignant diseases, is a highly selective inhibitor of the chymotrypsin-like (CT-L) activity of the proteasome. A chemistry effort was initiated to discover orally bioavailable analogues of carfilzomib, which would have potential for improved dosing flexibility and patient convenience over intravenously administered agents. The lead compound, 2-Me-5-thiazole-Ser(OMe)-Ser(OMe)-Phe-ketoepoxide (58) (PR-047), selectively inhibited CT-L activity of both the constitutive proteasome (beta5) and immunoproteasome (LMP7) and demonstrated an absolute bioavailability of up to 39% in rodents and dogs. It was well tolerated with repeated oral administration at doses resulting in >80% proteasome inhibition in most tissues and elicited an antitumor response equivalent to intravenously administered carfilzomib in multiple human tumor xenograft and mouse syngeneic models. The favorable pharmacologic profile supports its further development for the treatment of malignant diseases.


Assuntos
Dipeptídeos/síntese química , Dipeptídeos/farmacologia , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Oligopeptídeos/síntese química , Oligopeptídeos/farmacologia , Inibidores de Proteassoma , Tiazóis/síntese química , Tiazóis/farmacologia , Administração Oral , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Disponibilidade Biológica , Linhagem Celular , Dipeptídeos/química , Dipeptídeos/farmacocinética , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacocinética , Humanos , Cinética , Camundongos , Oligopeptídeos/química , Oligopeptídeos/farmacocinética , Relação Estrutura-Atividade , Especificidade por Substrato , Tiazóis/química , Tiazóis/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA