Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
SLAS Discov ; 23(6): 554-560, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29420107

RESUMO

Kynurenine 3-monooxygenase (KMO) is a well-validated therapeutic target for the treatment of neurodegenerative diseases, including Alzheimer's disease (AD) and Huntington's disease (HD). This work reports a facile fluorescence-based KMO assay optimized for high-throughput screening (HTS) that achieves a throughput approximately 20-fold higher than the fastest KMO assay currently reported. The screen was run with excellent performance (average Z' value of 0.80) from 110,000 compounds across 341 plates and exceeded all statistical parameters used to describe a robust HTS assay. A subset of molecules was selected for validation by ultra-high-performance liquid chromatography, resulting in the confirmation of a novel hit with an IC50 comparable to that of the well-described KMO inhibitor Ro-61-8048. A medicinal chemistry program is currently underway to further develop our novel KMO inhibitor scaffolds.


Assuntos
Inibidores Enzimáticos/química , Ensaios de Triagem em Larga Escala/métodos , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Cromatografia Líquida de Alta Pressão/métodos , Fluorescência
2.
Br J Pharmacol ; 168(6): 1316-28, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23126308

RESUMO

BACKGROUND AND PURPOSE: Growing evidence implicates iron in the aetiology of gastrointestinal cancer. Furthermore, studies demonstrate that iron chelators possess potent anti-tumour activity, although whether iron chelators show activity against oesophageal cancer is not known. EXPERIMENTAL APPROACH: The effect of the iron chelators, deferoxamine (DFO) and deferasirox, on cellular iron metabolism, viability and proliferation was assessed in two oesophageal adenocarcinoma cell lines, OE33 and OE19, and the squamous oesophageal cell line, OE21. A murine xenograft model was employed to assess the effect of deferasirox on oesophageal tumour burden. The ability of chelators to overcome chemoresistance and to enhance the efficacy of standard chemotherapeutic agents (cisplatin, fluorouracil and epirubicin) was also assessed. KEY RESULTS: Deferasirox and DFO effectively inhibited cellular iron acquisition and promoted intracellular iron mobilization. The resulting reduction in cellular iron levels was reflected by increased transferrin receptor 1 expression and reduced cellular viability and proliferation. Treating oesophageal tumour cell lines with an iron chelator in addition to a standard chemotherapeutic agent resulted in a reduction in cellular viability and proliferation compared with the chemotherapeutic agent alone. Both DFO and deferasirox were able to overcome cisplatin resistance. Furthermore, in human xenograft models, deferasirox was able to significantly suppress tumour growth, which was associated with decreased tumour iron levels. CONCLUSIONS AND IMPLICATIONS: The clinically established iron chelators, DFO and deferasirox, effectively deplete iron from oesophageal tumour cells, resulting in growth suppression. These data provide a platform for assessing the utility of these chelators in the treatment of oesophageal cancer patients.


Assuntos
Antineoplásicos/uso terapêutico , Benzoatos/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Neoplasias Esofágicas/tratamento farmacológico , Esôfago/efeitos dos fármacos , Quelantes de Ferro/uso terapêutico , Triazóis/uso terapêutico , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Benzoatos/administração & dosagem , Benzoatos/farmacologia , Linhagem Celular Tumoral , Cisplatino/administração & dosagem , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Deferasirox , Desferroxamina/administração & dosagem , Desferroxamina/farmacologia , Desferroxamina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Esofágicas/sangue , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Esôfago/metabolismo , Esôfago/patologia , Feminino , Humanos , Ferro/sangue , Ferro/metabolismo , Quelantes de Ferro/administração & dosagem , Quelantes de Ferro/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Triazóis/administração & dosagem , Triazóis/farmacologia , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Curr Drug Deliv ; 7(3): 194-207, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20507267

RESUMO

Targeting essential nutrients (eg., those required for DNA synthesis) to inhibit cancer cell growth is a well established therapeutic strategy. A good example is the highly successful folate antagonist, methotrexate. However, up until recently, strategies to target iron which is also crucial for DNA synthesis have not been systematically explored to develop agents for the treatment of cancer. Over the last 15 years, our laboratory has embarked upon structure-activity studies designed to develop novel Fe chelators with anti-cancer efficacy. These studies have led to the development of the dipyridyl thiosemicarbazone chelators that show potent and selective anti-cancer activity and which overcome resistance to other cytotoxic agents. This class of compounds include the chelator, di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (Dp44mT), which at optimal doses markedly inhibits tumour growth and is well tolerated. Moreover, this ligand does not induce overt Fe-depletion in vivo, probably because very low doses (0.4 mg/kg) are effective at inhibiting tumour growth. Importantly, our compounds are far more active and less toxic than the chelator, Triapine®, that is being assessed in a wide variety of international clinical trials. A vital part of the mechanism of action of these compounds is their ability to form a redox-active Fe complex that generates radicals to inhibit tumour growth. Due to their relatively high lipophilicity and low molecular weight of this class of compounds, oral activity may be expected in addition to their well known efficacy via the intravenous route.


Assuntos
Antineoplásicos/uso terapêutico , Desenho de Fármacos , Quelantes de Ferro/uso terapêutico , Ferro/metabolismo , Neoplasias/tratamento farmacológico , Tiossemicarbazonas/uso terapêutico , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/química , Antineoplásicos/metabolismo , Transporte Biológico , Humanos , Quelantes de Ferro/efeitos adversos , Quelantes de Ferro/química , Quelantes de Ferro/metabolismo , Estrutura Molecular , Neoplasias/metabolismo , Neoplasias/patologia , Piridinas/uso terapêutico , Relação Estrutura-Atividade , Tiossemicarbazonas/efeitos adversos , Tiossemicarbazonas/química , Tiossemicarbazonas/metabolismo , Resultado do Tratamento
4.
Br J Cancer ; 100(1): 96-105, 2009 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-19127267

RESUMO

Increased retinoic acid receptor beta (RARbeta(2)) gene expression is a hallmark of cancer cell responsiveness to retinoid anticancer effects. Moreover, low basal or induced RARbeta(2) expression is a common feature of many human cancers, suggesting that RARbeta(2) may act as a tumour suppressor gene in the absence of supplemented retinoid. We have previously shown that low RARbeta(2) expression is a feature of advanced neuroblastoma. Here, we demonstrate that the ABC domain of the RARbeta(2) protein alone was sufficient for the growth inhibitory effects of RARbeta(2) on neuroblastoma cells. ATP7A, the copper efflux pump, is a retinoid-responsive gene, was upregulated by ectopic overexpression of RARbeta(2). The ectopic overexpression of the RARbeta(2) ABC domain was sufficient to induce ATP7A expression, whereas, RARbeta(2) siRNA blocked the induction of ATP7A expression in retinoid-treated neuroblastoma cells. Forced downregulation of ATP7A reduced copper efflux and increased viability of retinoid-treated neuroblastoma cells. Copper supplementation enhanced cell growth and reduced retinoid-responsiveness, whereas copper chelation reduced the viability and proliferative capacity. Taken together, our data demonstrates ATP7A expression is regulated by retinoic acid receptor beta and it has effects on intracellular copper levels, revealing a link between the anticancer action of retinoids and copper metabolism.


Assuntos
Adenosina Trifosfatases/fisiologia , Proteínas de Transporte de Cátions/fisiologia , Neuroblastoma/tratamento farmacológico , Receptores do Ácido Retinoico/fisiologia , Adenosina Trifosfatases/genética , Proteínas de Transporte de Cátions/genética , Linhagem Celular Tumoral , Proliferação de Células , Cobre/metabolismo , ATPases Transportadoras de Cobre , Regulação Neoplásica da Expressão Gênica , Humanos , Neuroblastoma/patologia , Retinoides/farmacologia , Retinoides/uso terapêutico
5.
Biochim Biophys Acta ; 1790(7): 702-17, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18485918

RESUMO

Cancer contributes to 50% of deaths worldwide and new anti-tumour therapeutics with novel mechanisms of actions are essential to develop. Metabolic inhibitors represent an important class of anti-tumour agents and for many years, agents targeting the nutrient folate were developed for the treatment of cancer. This is because of the critical need of this factor for DNA synthesis. Similarly to folate, Fe is an essential cellular nutrient that is critical for DNA synthesis. However, in contrast to folate, there has been limited effort applied to specifically design and develop Fe chelators for the treatment of cancer. Recently, investigations have led to the generation of novel di-2-pyridylketone thiosemicarbazone (DpT) and 2-benzoylpyridine thiosemicarbazone (BpT) group of ligands that demonstrate marked and selective anti-tumour activity in vitro and also in vivo against a wide spectrum of tumours. Indeed, administration of these compounds to mice did not induce whole body Fe-depletion or disturbances in haematological or biochemical indices due to the very low doses required. The mechanism of action of these ligands includes alterations in expression of molecules involved in cell cycle control and metastasis suppression, as well as the generation of redox-active Fe complexes. This review examines the alterations in Fe metabolism in tumour cells and the systematic development of novel aroylhydrazone and thiosemicarbazone Fe chelators for cancer treatment.


Assuntos
Antineoplásicos/uso terapêutico , Quelantes de Ferro/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos de Neoplasias/fisiologia , Peptídeos Catiônicos Antimicrobianos/fisiologia , Proteínas de Transporte de Cátions , Ciclo Celular/efeitos dos fármacos , FMN Redutase/metabolismo , Hepcidinas , Humanos , Absorção Intestinal , Proteínas Reguladoras de Ferro/fisiologia , Antígenos Específicos de Melanoma , Metástase Neoplásica/fisiopatologia , Proteínas de Neoplasias/fisiologia , Neovascularização Patológica/fisiopatologia , Receptores da Transferrina/metabolismo
6.
Curr Med Chem ; 10(12): 1035-49, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12678675

RESUMO

The chelator currently used to treat iron (Fe) overload disease, desferrioxamine (DFO), has shown anti-proliferative activity against leukemia and neuroblastoma cells in vitro, in vivo and in clinical trials. Collectively, these studies suggest that Fe-deprivation may be a useful anti-cancer strategy. However, the efficacy of DFO is severely limited due to its poor ability to permeate cell membranes and bind intracellular Fe pools. These limitations have encouraged the development of other Fe chelators that are far more effective than DFO. One group of ligands that have been extensively investigated are those of the pyridoxal isonicotinoyl hydrazone (PIH) class. In this review the marked anti-proliferative effects of the PIH analogs are discussed with reference to their mechanisms of action and structure-activity relationships. In particular, we discuss the activity of a novel group of ligands that are "hybrid" chelators derived from our most effective PIH analogs and thiosemicarbazones. The anti-tumor activity of the PIH analogs and other chelators such as tachpyridine, O-trensox and the desferrithiocin analogs have been well characterized in vitro. However, further studies in animals are critical to evaluate their selective anti-tumor activity and potential as therapeutic agents.


Assuntos
Antineoplásicos/farmacologia , Hidrazonas/farmacologia , Quelantes de Ferro/farmacologia , Neoplasias/tratamento farmacológico , Animais , Desferroxamina/farmacologia , Humanos , Ferro/metabolismo , Ferro/fisiologia , Neoplasias/metabolismo , Relação Estrutura-Atividade
7.
Expert Opin Investig Drugs ; 9(6): 1257-70, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11060741

RESUMO

The metal complexes of a variety of ligands show diverse pharmacological properties. The potential of these compounds as antineoplastic agents is underlined by the success of the clinically used platinum complex cisplatin (cis-[(NH(3))(2)PtCl(2)]). In the current review, specific examples of gallium, copper, ruthenium and titanium complexes are discussed with special relevance to their use in the treatment of cancer. Some of these complexes have demonstrated marked activity in a number of animal models and for some compounds, clinical trials are anticipated or have already begun. Collectively, the results in the literature indicate that the study of metal complexes as antineoplastic agents deserves continued intensive investigation.


Assuntos
Antineoplásicos/uso terapêutico , Gálio/uso terapêutico , Neoplasias/tratamento farmacológico , Compostos Organometálicos/uso terapêutico , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...