Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Cancer Ther ; 10(10): 1929-38, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21831962

RESUMO

CBP501 is an anticancer drug currently in randomized phase II clinical trials for patients with non-small cell lung cancer and malignant pleural mesothelioma. CBP501 was originally described as a unique G(2) checkpoint-directed agent that binds to 14-3-3, inhibiting the actions of Chk1, Chk2, mitogen-activated protein kinase-activated protein kinase 2, and C-Tak1. However, unlike a G(2) checkpoint inhibitor, CBP501 clearly enhances the accumulation of tumor cells at G(2)-M phase that is induced by cisplatin or bleomycin at low doses and short exposure. By contrast, CBP501 does not similarly affect the accumulation of tumor cells at G(2)-M that is induced by radiation, doxorubicin, or 5-fluorouracil treatment. Our recent findings point to an additional mechanism of action for CBP501. The enhanced accumulation of tumor cells at G(2)-M upon combined treatment with cisplatin and CBP501 results from an increase in intracellular platinum concentrations, which leads to increased binding of platinum to DNA. The observed CBP501-enhanced platinum accumulation is negated in the presence of excess Ca(2+). Some calmodulin inhibitors behave similarly to, although less potently than, CBP501. Furthermore, analysis by surface plasmon resonance reveals a direct, high-affinity molecular interaction between CBP501 and CaM (K(d) = 4.62 × 10(-8) mol/L) that is reversed by Ca(2+), whereas the K(d) for the complex between CBP501 and 14-3-3 is approximately 10-fold weaker and is Ca(2+) independent. We conclude that CaM inhibition contributes to CBP501's activity in sensitizing cancer cells to cisplatin or bleomycin. This article presents an additional mechanism of action which might explain the clinical activity of the CBP501-cisplatin combination.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Bleomicina/farmacologia , Calmodulina/metabolismo , Cisplatino/farmacologia , Neoplasias/tratamento farmacológico , Fragmentos de Peptídeos/farmacologia , Fosfatases cdc25/farmacologia , Bleomicina/administração & dosagem , Cloreto de Cálcio/farmacologia , Calmodulina/antagonistas & inibidores , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/administração & dosagem , Cisplatino/farmacocinética , Adutos de DNA/biossíntese , Sinergismo Farmacológico , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Fragmentos de Peptídeos/farmacocinética , Fosfatases cdc25/farmacocinética
2.
Mol Cancer Ther ; 6(1): 147-53, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17237275

RESUMO

Cell cycle G(2) checkpoint abrogation is an attractive strategy for sensitizing cancer cells to DNA-damaging anticancer agent without increasing adverse effects on normal cells. However, there is no single proven molecular target for this therapeutic approach. High-throughput screening for molecules inhibiting CHK1, a kinase that is essential for the G(2) checkpoint, has not yet yielded therapeutic G(2) checkpoint inhibitors, and the tumor suppressor phenotypes of ATM and CHK2 suggest they may not be ideal targets. Here, we optimized two G(2) checkpoint-abrogating peptides, TAT-S216 and TAT-S216A, based on their ability to reduce G(2) phase accumulation of DNA-damaged cells without affecting M phase accumulation of cells treated with a microtubule-disrupting compound. This approach yielded a peptide CBP501, which has a unique, focused activity against molecules that phosphorylate Ser(216) of CDC25C, including MAPKAP-K2, C-Tak1, and CHK1. CBP501 is >100-fold more potent than TAT-S216A and retains its selectivity for cancer cells. CBP501 is unusually stable, enters cells rapidly, and increases the cytotoxicity of DNA-damaging anticancer drugs against cancer cells without increasing adverse effects. These findings highlight the potency of CBP501 as a G(2)-abrogating drug candidate. This report also shows the usefulness of the cell cycle phenotype-based protocol for identifying G(2) checkpoint-abrogating compounds as well as the potential of peptide-based compounds as focused multitarget inhibitors.


Assuntos
Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Fase G2/efeitos dos fármacos , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Peptídeos/farmacologia , Fosfatases cdc25/metabolismo , Fosfatases cdc25/farmacologia , Sequência de Aminoácidos , Animais , Antineoplásicos/química , Bleomicina/efeitos adversos , Bleomicina/farmacologia , Proteínas de Ciclo Celular/metabolismo , Morte Celular/efeitos dos fármacos , Cisplatino/efeitos adversos , Cisplatino/farmacologia , Células HCT116 , Humanos , Células Jurkat , Masculino , Camundongos , Camundongos SCID , Modelos Moleculares , Dados de Sequência Molecular , Neoplasias/patologia , Fragmentos de Peptídeos/química , Peptídeos/química , Fenótipo , Fosforilação/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Fosfatases cdc25/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...