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










Base de dados
Tipo de estudo
Intervalo de ano de publicação
1.
J Med Chem ; 59(7): 3439-51, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-26938120

RESUMO

Combretastatin A-4 (CA-4) in phosphate and serine pro-drug forms is under phase II clinical trials. With our interest of discovering CA-4 inspired new chemical entities, a novel series of 4,5-diaryl-2-aminoimidazole analogues of the compound was designed and synthesized by an efficient and diversity feasible route involving atom economical arene C-H bond arylation. Interestingly, four compounds showed potent cell-based antiproliferative activities in nanomolar concentrations. Among the compounds, compound 12 inhibited the proliferation of several types of cancer cells much more efficiently than CA-4. It depolymerized microtubules, induced spindle defects, and stalled mitosis in cells. Compound 12 bound to tubulin and inhibited the polymerization of tubulin in vitro. In addition, podophyllotoxin and CA-4 inhibited the binding of compound 12 to tubulin. The distinctive pharmacophoric features of the bridging motif as well as quinoline nucleus were explored. We noted also a valuable quality of compound 12 as a potential probe in characterizing new CA-4 analogues.


Assuntos
Bibenzilas/química , Neoplasias da Mama/tratamento farmacológico , Imidazóis/química , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacologia , Tubulina (Proteína)/química , Apoptose/efeitos dos fármacos , Western Blotting , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Microtúbulos/metabolismo , Mitose/efeitos dos fármacos , Estrutura Molecular , Pró-Fármacos/química , Relação Estrutura-Atividade , Tubulina (Proteína)/metabolismo , Células Tumorais Cultivadas
2.
Expert Opin Ther Targets ; 19(7): 957-72, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25787715

RESUMO

INTRODUCTION: The centrosome plays an essential role in the cell cycle. The centrosome and its associated proteins assist in nucleating and organizing microtubules. A structural or a functional aberration in the centrosome is known to cause abnormal cell proliferation leading to tumors. Therefore, the centrosome is considered as a promising anti-cancer target. AREAS COVERED: This review begins with a brief introduction to the centrosome and its role in the cell cycle. We elaborate on the centrosome-associated proteins that regulate microtubule dynamics. In addition, we discuss the centrosomal protein kinase targets such as cyclin-dependent, polo-like and aurora kinases. Inhibitors targeting these kinases are undergoing clinical trials for cancer chemotherapy. Further, we shed light on new approaches to target the centrosomal proteins for cancer therapy. EXPERT OPINION: Insights into the functioning of the centrosomal proteins will be extremely beneficial in validating the centrosome as a target in cancer therapy. New strategies either as a single entity or in combination with current chemotherapeutic agents should be researched or exploited to reveal the promises that the centrosome holds for future cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Centrossomo/metabolismo , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/administração & dosagem , Ciclo Celular/fisiologia , Proliferação de Células/fisiologia , Humanos , Terapia de Alvo Molecular , Neoplasias/patologia
3.
PLoS One ; 8(10): e75182, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24124473

RESUMO

Maytansinoid conjugates are currently under different phases of clinical trials and have been showing promising activity for various types of cancers. In this study, we have elucidated the mechanism of action of ansamitocin P3, a structural analogue of maytansine for its anticancer activity. Ansamitocin P3 potently inhibited the proliferation of MCF-7, HeLa, EMT-6/AR1 and MDA-MB-231 cells in culture with a half-maximal inhibitory concentration of 20±3, 50±0.5, 140±17, and 150±1.1 pM, respectively. Ansamitocin P3 strongly depolymerized both interphase and mitotic microtubules and perturbed chromosome segregation at its proliferation inhibitory concentration range. Treatment of ansamitocin P3 activated spindle checkpoint surveillance proteins, Mad2 and BubR1 and blocked the cells in mitotic phase of the cell cycle. Subsequently, cells underwent apoptosis via p53 mediated apoptotic pathway. Further, ansamitocin P3 was found to bind to purified tubulin in vitro with a dissociation constant (Kd) of 1.3±0.7 µM. The binding of ansamitocin P3 induced conformational changes in tubulin. A docking analysis suggested that ansamitocin P3 may bind partially to vinblastine binding site on tubulin in two different positions. The analysis indicated that the binding of ansamitocin P3 to tubulin is stabilized by hydrogen bonds. In addition, weak interactions such as halogen-oxygen interactions may also contribute to the binding of ansamitocin P3 to tubulin. The study provided a significant insight in understanding the antiproliferative mechanism of action of ansamitocin P3.


Assuntos
Apoptose/efeitos dos fármacos , Maitansina/análogos & derivados , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Vimblastina/metabolismo , Western Blotting , Ciclo Celular , Linhagem Celular , Células HeLa , Humanos , Maitansina/farmacologia , Microscopia de Fluorescência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...