Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Oncogene ; 31(42): 4567-76, 2012 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-22249249

RESUMO

Multiple DNA methylation changes in the cancer methylome are associated with the acquisition of drug resistance; however it remains uncertain how many represent critical DNA methylation drivers of chemoresistance. Using isogenic, cisplatin-sensitive/resistant ovarian cancer cell lines and inducing resensitizaton with demethylating agents, we aimed to identify consistent methylation and expression changes associated with chemoresistance. Using genome-wide DNA methylation profiling across 27 578 CpG sites, we identified loci at 4092 genes becoming hypermethylated in chemoresistant A2780/cp70 compared with the parental-sensitive A2780 cell line. Hypermethylation at gene promoter regions is often associated with transcriptional silencing; however, expression of only 245 of these hypermethylated genes becomes downregulated in A2780/cp70 as measured by microarray expression profiling. Treatment of A2780/cp70 with the demethylating agent 2-deoxy-5'-azacytidine induces resensitization to cisplatin and re-expression of 41 of the downregulated genes. A total of 13/41 genes were consistently hypermethylated in further independent cisplatin-resistant A2780 cell derivatives. CpG sites at 9 of the 13 genes (ARHGDIB, ARMCX2, COL1A, FLNA, FLNC, MEST, MLH1, NTS and PSMB9) acquired methylation in ovarian tumours at relapse following chemotherapy or chemoresistant cell lines derived at the time of patient relapse. Furthermore, 5/13 genes (ARMCX2, COL1A1, MDK, MEST and MLH1) acquired methylation in drug-resistant ovarian cancer-sustaining (side population) cells. MLH1 has a direct role in conferring cisplatin sensitivity when reintroduced into cells in vitro. This combined genomics approach has identified further potential key drivers of chemoresistance whose expression is silenced by DNA methylation that should be further evaluated as clinical biomarkers of drug resistance.


Assuntos
Cisplatino/farmacologia , Metilação de DNA , Resistencia a Medicamentos Antineoplásicos/genética , Epigenômica/métodos , Perfilação da Expressão Gênica/métodos , Neoplasias Ovarianas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Linhagem Celular Tumoral , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Ilhas de CpG/genética , Decitabina , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Ácidos Hidroxâmicos/farmacologia , Midkina , Proteína 1 Homóloga a MutL , Fatores de Crescimento Neural/genética , Proteínas Nucleares/genética , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Proteínas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sulfonamidas/farmacologia
2.
Cancer Chemother Pharmacol ; 67(6): 1273-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20706839

RESUMO

PURPOSE: The primary objective of this sub-study, undertaken as an extension to the previously reported phase-I study, was to explore the feasibility, tolerability and pharmacokinetics (PK) of belinostat when administered by the oral route. Preliminary pharmacodynamic (PD) studies were also performed to enable comparison of the biological effects of the oral and intravenous formulations. PATIENTS AND METHODS: Oral belinostat was administered in a range of doses and schedules (once, twice or thrice daily), on either day 1 or days 1-5, of the second or a subsequent treatment cycle in 15 patients who were included in the phase-I trial of intravenous belinostat. Serial blood samples were collected for PK and PD (histone acetylation) analyses, and the results compared with corresponding analyses following intravenous administration. RESULTS: A total mean daily AUC of 2,767 ± 1,453 ng h/ml (8.7 ± 4.6 µM h) resulted from a dose of 1,000 mg/m(2) once daily (qd). There was no clear evidence of drug accumulation on twice daily dosing (bid); however, a trend towards accumulation was apparent when belinostat was given three times daily (tid). Mean half-life (T½) of a single dose of 1,000 mg/m(2) was 1.5 h (± 0.3 h) and peak levels were reached in an average of 1.9 h (± 0.3 h). The half-life was found to be independent of dose, but a trend towards increasing half-life following multiple dosing was observed. Histone H4 hyperacetylation in PBMCs estimated after oral dosing was comparable to that achieved after intravenous administration. CONCLUSIONS: High doses of oral belinostat, up to 1,000 mg/m(2) bid for 5 consecutive days, have been tolerated in this small study. An oral formulation could lead to enhanced drug exposure and, more importantly, prolonged effects on the intended drug target. Future trials are required to establish the optimal dose and schedule of oral administration of belinostat.


Assuntos
Antineoplásicos/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Acetilação , Administração Oral , Adulto , Idoso , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Estudos de Viabilidade , Inibidores de Histona Desacetilases/administração & dosagem , Inibidores de Histona Desacetilases/farmacocinética , Histonas/sangue , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Ácidos Hidroxâmicos/farmacocinética , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Pessoa de Meia-Idade , Sulfonamidas
3.
Cancer Nanotechnol ; 1(1-6): 29-34, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-26069477

RESUMO

The present study was designed to explore the ability of polymeric nanoparticles to restore drug sensitivity to P-glycoprotein over-expressing cancer cells. A multidrug-resistant cell line 2780 AD and its sensitive parent cell line A2780 were studied in cell culture and as a xenografted tumour model. Paclitaxel was incorporated in poly(lactide-co-glycolide) nanoparticles of average diameter 125 nm stabilised by a positively charged surfactant. The nanoparticulate formulation was shown to be about sevenfold more potent than free paclitaxel against cell line A2780 and the poly(lactic-co-glycolic acid) (PLGA) nanoparticles alone were nontoxic to the cells at the concentrations required to deliver the drug. Whilst the oral formulation of paclitaxel was not as potent as the free drug in the A2780 xenografts, it showed significant activity against 2780 AD tumours, which are resistant to the maximum tolerated intravenous dose of paclitaxel. The efficacy of orally delivered paclitaxel in this drug-resistant model supports the concept of exploring nanoparticles for improved drug delivery.

4.
Br J Cancer ; 100(5): 758-63, 2009 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-19259094

RESUMO

Histone deacetylation and DNA methylation have a central role in the control of gene expression in tumours, including transcriptional repression of tumour suppressor genes and genes involved in sensitivity to chemotherapy. Treatment of cisplatin-resistant cell lines with an inhibitor of DNA methyltransferases, 2-deoxy-5'azacytidine (decitabine), results in partial reversal of DNA methylation, re-expression of epigenetically silenced genes including hMLH1 and sensitisation to cisplatin both in vitro and in vivo. We have investigated whether the combination of decitabine and a clinically relevant inhibitor of histone deacetylase activity (belinostat, PXD101) can further increase the re-expression of genes epigenetically silenced by DNA methylation and enhance chemo-sensitisation in vivo at well-tolerated doses. The cisplatin-resistant human ovarian cell line A2780/cp70 has the hMLH1 gene methylated and is resistant to cisplatin both in vitro and when grown as a xenograft in mice. Treatment of A2780/cp70 with decitabine and belinostat results in a marked increase in expression of epigenetically silenced MLH1 and MAGE-A1 both in vitro and in vivo when compared with decitabine alone. The combination greatly enhanced the effects of decitabine alone on the cisplatin sensitivity of xenografts. As the dose of decitabine that can be given to patients and hence the maximum pharmacodynamic effect as a demethylating agent is limited by toxicity and eventual re-methylation of genes, we suggest that the combination of decitabine and belinostat could have a role in the efficacy of chemotherapy in tumours that have acquired drug resistance due to DNA methylation and gene silencing.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Inibidores Enzimáticos/administração & dosagem , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histona Acetiltransferases/antagonistas & inibidores , Neoplasias Ovarianas/tratamento farmacológico , Acetilação/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Azacitidina/administração & dosagem , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Metilação de DNA/efeitos dos fármacos , Decitabina , Resistencia a Medicamentos Antineoplásicos/genética , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Antígenos Específicos de Melanoma , Camundongos , Camundongos Nus , Proteína 1 Homóloga a MutL , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Sulfonamidas , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Biochem Soc Trans ; 32(Pt 6): 916-7, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15506923

RESUMO

Aberrant methylation of CpG islands (CpG-rich regions of DNA associated with the promoters of many genes) is associated with transcriptional inactivation of genes involved in tumour development. Genes involved in key DNA damage response pathways, such as cell-cycle control, apoptosis signalling and DNA repair can frequently become epigenetically silenced and methylated in tumours. This may lead to differences in intrinsic sensitivity of tumours to chemotherapy, depending on the specific function of the gene inactivated. Furthermore, chemotherapy itself may exert a selective pressure on epigenetically silenced drug sensitivity genes present in subpopulations of cells, leading to acquired chemoresistance. Clinical trials of epigenetic therapies are now in progress, and epigenetic profiling using DNA methylation will provide guidance on optimization of the use of these therapies with conventional chemotherapy, as well as helping to identify patient populations who may particularly benefit from such approaches.


Assuntos
Fosfatos de Dinucleosídeos/genética , Neoplasias/tratamento farmacológico , Animais , Resistencia a Medicamentos Antineoplásicos , Epigênese Genética , Inativação Gênica , Humanos , Neoplasias/genética
6.
Biochem Soc Trans ; 32(Pt 6): 1095-7, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15506976

RESUMO

Histone deacetylation and DNA methylation have a central role in the control of gene expression, including transcriptional repression of tumour suppressor genes. Loss of DNA mismatch repair due to methylation of the hMLH1 gene promoter results in resistance to cisplatin in vitro and in vivo. The cisplatin-resistant cell line A2780/cp70 is 8-fold more resistant to cisplatin than the non-resistant cell line, and has the hMLH1 gene methylated. Treatment with an inhibitor of DNA methyltransferase, DAC (2-deoxy-5'-azacytidine), results in a partial reversal of DNA methylation, re-expression of MLH1 (mutL homologue 1) and sensitization to cisplatin both in vitro and in vivo. PXD101 is a novel hydroxamate type histone deacetylase inhibitor that shows antitumour activity in vivo and is currently in phase I clinical evaluation. Treatment of A2780/cp70 tumour-bearing mice with DAC followed by PXD101 results in a marked increase in the number of cells that re-express MLH1. Since the clinical use of DAC may be limited by toxicity and eventual re-methylation of genes, we suggest that the combination of DAC and PXD101 could have a role in increasing the efficacy of chemotherapy in patients with tumours that lack MLH1 expression due to hMLH1 gene promoter methylation.


Assuntos
Terapia Genética/métodos , Antineoplásicos/uso terapêutico , Carcinoma de Células Pequenas/genética , Linhagem Celular Tumoral , Cisplatino/uso terapêutico , Metilação de DNA , Humanos , Neoplasias Pulmonares/genética
7.
Oncogene ; 20(53): 7797-803, 2001 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-11753658

RESUMO

Telomerase activation is considered to be a critical step in cancer progression due to its role in cellular immortalization. The prevalence of telomerase expression in human cancers makes it an attractive candidate for new mechanism-based targets for cancer therapy. The selective killing of cancer cells can be achieved by gene-directed enzyme pro-drug therapy (GDEPT). In this study we have tested the feasibility of using the transcriptional regulatory sequences from the hTERT and hTR genes to regulate expression of the bacterial nitroreductase enzyme in combination with the pro-drug CB1954 in a suicide gene therapy strategy. hTERT and hTR promoter activity was compared in a panel of 10 cell lines and showed a wide distribution in activity; low activity was observed in normal cells and telomerase-negative immortal ALT cell lines, with up to 300-fold higher activity observed in telomerase positive cancer lines. Placing the nitroreductase gene under the control of the telomerase gene promoters sensitized cancer cells in tissue culture to the pro-drug CB1954 and promoter activity was predictive of sensitization to the pro-drug (2-20-fold sensitization), with cell death restricted to lines exhibiting high levels of promoter activity. The in vivo relevance of these data was tested using two xenograft models (C33a and GLC4 cells). Significant tumour reduction was seen with both telomerase promoters and the promoter-specific patterns of sensitization observed in tissue culture were retained in xenograft models. Thus, telomerase-specific suicide gene therapy vectors expressing bacterial nitroreductase sensitize human cancer cells to the pro-drug CB1954.


Assuntos
Aziridinas/farmacologia , Terapia Genética/métodos , Neoplasias/genética , Neoplasias/terapia , Nitrorredutases/genética , Nitrorredutases/uso terapêutico , Pró-Fármacos/farmacologia , Telomerase/genética , Animais , Aziridinas/uso terapêutico , Bactérias/enzimologia , Carcinoma de Células Pequenas/genética , Carcinoma de Células Pequenas/metabolismo , Carcinoma de Células Pequenas/patologia , Carcinoma de Células Pequenas/terapia , Divisão Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Regulação Enzimológica da Expressão Gênica , Vetores Genéticos/genética , Humanos , Camundongos , Transplante de Neoplasias , Neoplasias/metabolismo , Neoplasias/patologia , Nitrorredutases/metabolismo , Pró-Fármacos/uso terapêutico , Regiões Promotoras Genéticas/genética , Telomerase/metabolismo , Transgenes/genética , Transplante Heterólogo , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/terapia
8.
Cancer Res ; 61(2): 749-58, 2001 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11212278

RESUMO

The overexpression of P-glycoprotein (P-gp) on the surface of tumor cells causes multidrug resistance (MDR). This protein acts as an energy-dependent drug efflux pump reducing the intracellular concentration of structurally unrelated drugs. Modulators of P-gp function can restore the sensitivity of MDR cells to such drugs. XR9576 is a novel anthranilic acid derivative developed as a potent and specific inhibitor of P-gp, and in this study we evaluate the in vitro and in vivo modulatory activity of this compound. The in vitro activity of XR9576 was evaluated using a panel of human (H69/LX4, 2780AD) and murine (EMT6 AR1.0, MC26) MDR cell lines. XR9576 potentiated the cytotoxicity of several drugs including doxorubicin, paclitaxel, etoposide, and vincristine; complete reversal of resistance was achieved in the presence of 25-80 nM XR9576. Direct comparative studies with other modulators indicated that XR9576 was one of the most potent modulators described to date. Accumulation and efflux studies with the P-gp substrates, [3H]daunorubicin and rhodamine 123, demonstrated that XR9576 inhibited P-gp-mediated drug efflux. The inhibition of P-gp function was reversible, but the effects persisted for >22 h after removal of the modulator from the incubation medium. This is in contrast to P-gp substrates such as cyclosporin A and verapamil, which lose their activity within 60 min, suggesting that XR9576 is not transported by P-gp. Also, XR9576 was a potent inhibitor of photoaffinity labeling of P-gp by [3H]azidopine implying a direct interaction with the protein. In mice bearing the intrinsically resistant MC26 colon tumors, coadministration of XR9576 potentiated the antitumor activity of doxorubicin without a significant increase in toxicity; maximum potentiation was observed at 2.5-4.0 mg/kg dosed either i.v. or p.o. In addition, coadministration of XR9576 (6-12 mg/kg p.o.) fully restored the antitumor activity of paclitaxel, etoposide, and vincristine against two highly resistant MDR human tumor xenografts (2780AD, H69/LX4) in nude mice. Importantly all of the efficacious combination schedules appeared to be well tolerated. Furthermore, i.v. coadministration of XR9576 did not alter the plasma pharmacokinetics of paclitaxel. These results demonstrate that XR9576 is an extremely potent, selective, and effective modulator with a long duration of action. It exhibits potent i.v. and p.o. activity without apparently enhancing the plasma pharmacokinetics of paclitaxel or the toxicity of coadministered drugs. Hence, XR9576 holds great promise for the treatment of P-gp-mediated MDR cancers.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/fisiologia , Resistência a Múltiplos Medicamentos , Quinolinas/farmacologia , Tetra-Hidroisoquinolinas , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Acridinas/farmacologia , Animais , Antibióticos Antineoplásicos/farmacologia , Antineoplásicos/farmacologia , Azidas/metabolismo , Ligação Competitiva/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Ciclosporinas/farmacologia , Daunorrubicina/metabolismo , Daunorrubicina/farmacologia , Di-Hidropiridinas/metabolismo , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Feminino , Humanos , Isoquinolinas/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/patologia , Neoplasias Experimentais/prevenção & controle , Paclitaxel/farmacocinética , Fatores de Tempo , Resultado do Tratamento , Trítio , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Cancer Res ; 60(21): 6039-44, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11085525

RESUMO

Loss of DNA mismatch repair because of hypermethylation of the hMLH1 gene promoter occurs at a high frequency in a number of human tumors. A role for loss of mismatch repair (MMR) in resistance to a number of clinically important anticancer drugs has been shown. We have investigated whether the demethylating agent 2'-deoxy-5-azacytidine (DAC) can be used in vivo to sensitize MMR-deficient, drug-resistant ovarian (A2780/cp70) and colon (SW48) tumor xenografts that are MLH1 negative because of gene promoter hypermethylation. Treatment of tumor-bearing mice with the demethylating agent DAC at a nontoxic dose induces MLH1 expression. Re-expression of MLH1 is associated with a decrease in hMLH1 gene promoter methylation. DAC treatment alone has no effect on the growth rate of the tumors. However, DAC treatment sensitizes the xenografts to cisplatin, carboplatin, temozolomide, and epirubicin. Sensitization is comparable with that obtained by reintroduction of the hMLH1 gene by chromosome 3 transfer. Consistent with loss of MMR having no effect on sensitivity in vitro to Taxol, DAC treatment has no effect on the Taxol sensitivity of the xenografts. DAC treatment does not sensitize xenografts of HCT116, which lacks MMR because of hMLH1 mutation. Because there is emerging data on the role of loss of MMR in clinical drug resistance, DAC could have a role in increasing the efficacy of chemotherapy for patients whose tumors lack MLH1 expression because of hMLH1 promoter methylation.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Metilação de DNA/efeitos dos fármacos , Dacarbazina/análogos & derivados , Proteínas de Neoplasias/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antineoplásicos/farmacologia , Pareamento Incorreto de Bases , Carboplatina/farmacologia , Proteínas de Transporte , Cisplatino/farmacologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Reparo do DNA , Dacarbazina/farmacologia , Decitabina , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Nus , Proteína 1 Homóloga a MutL , Proteínas de Neoplasias/biossíntese , Proteínas Nucleares , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Temozolomida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Dis Aquat Organ ; 39(2): 143-6, 2000 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-10715819

RESUMO

Juvenile largemouth bass Micropterus salmoides were intraperitoneally injected with largemouth bass virus (LMBV), a member of the genus Ranavirus, family Iridoviridae. Moribund fish which had been injected with 10(6.2) tissue culture infectious doses, 50% endpoint (TCID50), were sampled 4 d after injection; other largemouth bass injected with this dose died between 3 and 5 d after injection. Fish injected with 10(2.8) TCID50 of LMBV were also examined after 4 d and had lesions similar to those of fish injected with the high dose. Clinical signs included darker pigmentation, inflammation and necrosis at the site of injection, distended abdomen, corkscrew swimming, and lateral recumbency. Internally, fish had focally pale livers, bright red spleens and reddened intestinal ceca. Histologically acute fibrinous peritonitis affected the surface of all organs in the peritoneal cavity, but deeper portions of organs appeared normal. There was also necrosis of the gastrointestinal mucosa. Except for the injection site, lesions were confined to the peritoneal cavity.


Assuntos
Bass , Infecções por Vírus de DNA/veterinária , Doenças dos Peixes/patologia , Ranavirus , Animais , Infecções por Vírus de DNA/patologia , Injeções Intraperitoneais , Intestinos/patologia , Fígado/patologia
11.
Exp Physiol ; 84(2): 285-97, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10226171

RESUMO

Experiments were undertaken using an ovarian adenocarcinoma cell line (A2780) and a drug-resistant strain (A2780.ad) derived from this line. P-glycoprotein could not be detected in A2780 cells but was essentially ubiquitous in A2780.ad cells, although removing the selective pressure for drug resistance led to reduced expression. However, the amount of P-glycoprotein present was used to predict the capacity of these cells to extrude rhodamine-123 (R-123) and their resistance to adriamycin, a cytotoxic drug. This accords with the role of P-glycoprotein as a drug pump. Although hypotonic solutions increased anion efflux from A2780 and A2780.ad cells, larger responses occurred in the parental line. Moreover, R-123 extrusion and anion efflux appeared to be mutually independent processes and so these data do not support the view that P-glycoprotein is involved in the control of volume-sensitive anion channels. Hypotonic solutions increased intracellular free calcium ([Ca2+]i) in drug-resistant cells but not in the parental line, and so establishing a drug-resistant strain may affect the control of [Ca2+]i during osmotic swelling. This could account for effects that were previously attributed to P-glycoprotein.


Assuntos
Cálcio/metabolismo , Células Epiteliais/metabolismo , Ovário/metabolismo , Preparações Farmacêuticas/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Antibióticos Antineoplásicos/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular , Sobrevivência Celular , Doxorrubicina/farmacologia , Resistência a Medicamentos , Feminino , Humanos , Imuno-Histoquímica , Radioisótopos do Iodo , Transporte de Íons , Ovário/citologia , Espectrometria de Fluorescência , Células Tumorais Cultivadas , Verapamil/farmacologia
12.
Methods Mol Med ; 28: 17-23, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-21374023

RESUMO

The use of cell culture systems to assess the toxicity of anticancer agents began over 50 years ago following the observation of the antineoplastic effects of nitrogen mustard (1). There are a wide variety of assays designed to evaluate cellular drug sensitivity described in the literature. These assays essentially fall into two groups; those that measure cell survival and those that measure cytotoxicity. Cytotoxicity assays include methods such as trypan blue dye exclusion, (51)Cr release and (3)H-thymidine incorporation (2-4) and these assays assess the structural integrity and metabolic function of the cells following drug exposure. In contrast, cell survival assays measure the end result of these effects on the cell which can be either cell death or recovery. A cell survival assay thus requires a measure of the ability of cells to proliferate and this is usually an estimate of the ability of individual cells to form colonies. However, cytotoxicity assays can also measure the ability of cells to proliferate if the cells are allowed a period of growth following drug exposure. This recovery time is comparable to the time taken for formation of colonies in a clonogenic assay.

13.
Methods Mol Med ; 28: 25-30, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-21374024

RESUMO

The clonogenic assay described in Chapter 2 is not suitable for all cell lines. Many adherent cell lines do not form colonies and clearly it is not applicable to non-adherent cell lines. Furthermore, it is slow and time consuming. This chapter describes an alternative cytotoxicity assay that has a number of advantages when compared with a clonogenic assay. It is quick and easy and allows a large number of assays to be carried out in one batch. This is an important consideration when making comparisons between cell lines, between cytotoxic agents, or when evaluating combinations of drugs. No one cytotoxicity assay is ideal and it is always advisable to support results with those from alternative assays where possible.

14.
Vet Immunol Immunopathol ; 58(2): 181-90, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9336886

RESUMO

The role of peritoneal macrophages in immunity to enteric septicemia of catfish (ESC) after infection with live Edwardsiella ictaluri was investigated. Channel catfish macrophage-mediated bacteriocidal activity was dependent on the macrophage:bacteria ratio. Ratios of 1:1 to 1:12 exhibited significant differences (P < or = 0.05) in killing between macrophages from immune fish when compared to killing by macrophages from susceptible fish at 2.5 h. At 5 h, macrophages from immune fish were capable of effective killing (83.3%) at a 1:24 effector:target ratio, whereas macrophages from susceptible fish killed significantly (P < or = 0.05) less (56.9%). Macrophage bacteriocidal activity was significantly greater (P < or = 0.05) in macrophages from individual immune fish (93.4%) compared to macrophages from individual susceptible fish (85.4%). The kinetics of macrophage killing showed a linear increase in bacteriocidal activity from 1 to 3 h. Opsonization with immune serum enabled macrophages from immune fish to kill bacteria more effectively (93.8 vs. 75.9%) at 2.5 h. Opsonization of E. ictaluri with immune serum significantly suppressed the killing ability of macrophages from susceptible fish (46.2%) at 2.5 h. The results suggest that macrophages from fish immune to ESC had a greater capacity to kill E. ictaluri than macrophages from susceptible fish especially when E. ictaluri were opsonized with anti-E. ictaluri antibody.


Assuntos
Bacteriemia/veterinária , Doenças dos Peixes/imunologia , Bactérias Gram-Negativas/imunologia , Infecções por Bactérias Gram-Negativas/imunologia , Ictaluridae/imunologia , Macrófagos Peritoneais/imunologia , Animais , Bacteriemia/imunologia , Técnicas In Vitro , Cinética , Proteínas Opsonizantes/metabolismo , Fagocitose
15.
Biochim Biophys Acta ; 1362(1): 29-38, 1997 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-9434097

RESUMO

Growth factors and certain oncogenes activate a range of phospholipid-mediated signal transduction pathways resulting in cell proliferation. Demethoxyviridin (DMV), a structural analogue of wortmannin and recently reported as a potent inhibitor of phosphoinositide-3-kinase, inhibited bombesin plus insulin-stimulated increase in cell number in Swiss 3T3 cells, a model of cell proliferation. The drug produced cytostatic effects at concentrations below 1 microM and cytotoxic effects at 10 microM. In intact Swiss 3T3 cells DMV inhibited insulin-stimulated PI 3- and 4-kinases and bombesin-stimulated phospholipases C, D and A2 in the nanomolar range. DMV also inhibited bombesin-stimulated tyrosine phosphorylation of a range of proteins at nM concentrations. This study shows that DMV inhibited multiple stimulated signalling pathways which lead to increased Swiss 3T3 cell proliferation. A stable analogue of DMV may have chemotherapeutic potential.


Assuntos
Androstenos/farmacologia , Divisão Celular/efeitos dos fármacos , Inibidores de Fosfoinositídeo-3 Quinase , Fosfolipídeos/fisiologia , Transdução de Sinais/efeitos dos fármacos , Células 3T3 , Androstadienos , Androstenos/toxicidade , Animais , Bombesina/farmacologia , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/toxicidade , Insulina/farmacologia , Camundongos , Fosfatidilinositóis/metabolismo , Fosfolipases/antagonistas & inibidores , Fosfolipases/metabolismo , Fosforilação , Receptores Proteína Tirosina Quinases/metabolismo , Tirosina/metabolismo , Wortmanina
16.
Anticancer Drug Des ; 11(6): 421-38, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8836108

RESUMO

Bioreducible anti-tumour agents are prodrugs which are intended to be inactive in normal cells, but are able to undergo metabolic reduction in cancer cells to produce toxic species that can damage biomolecules. A series of N-oxides of heterocyclic aliphatic amines were designed and prepared as mentioned below as bioreducible drugs based on the reported anti-cancer activity of 2,6-bis(halomethyl)piperidines. In order to study structure-activity relationships in these conformationally restricted nitrogen mustards, samples of cis- and trans-2,6-dihydroxymethyl-N-methylpiperidine were prepared and converted into a number of carbamate or halogen derivatives. The free bases were designed to be bifunctional alkylating agents via aziridinium ion formation. The corresponding N-oxides were also prepared for comparison in cytotoxicity tests. In total, 21 new compounds were synthesized plus cis-N-methyl-2,6-bis(chloromethyl)-piperidine (prepared previously but lacking spectroscopic data) and tested against two human colon carcinoma cell lines, HT 29 (high DT-diaphorase) and BE (no DT-diaphorase), under oxic and hypoxic conditions. The majority of the free bases were equally toxic against both cell lines. The most toxic compounds were cis- and trans-N-methyl-2,6-bis(bromomethyl)piperidine with oxic IC50 values between 6 and 11 microM against both cell lines. The N-oxides were relatively non-toxic under both oxic and hypoxic conditions apart from the N-oxide of trans-N-methyl-2,6-bis(bromomethyl)-piperidine. Their low toxicity suggested that the N-oxides are not reduced under hypoxic conditions. We conclude that: (i) 2,6-disubstituted N-methylpiperidine derivatives are chemically versatile cytotoxic entities that are suitable for prodrugging to enhance their therapeutic selectivity; and (ii) N-oxide prodrugs of these compounds are deactivated chemically and display reduced cytotoxicity compared to the parent amines but are apparently not reduced under hypoxic conditions. At least in the colorectal cell lines tested the latter issue would need to be addressed by modifying the redox properties in future work to progress this approach.


Assuntos
Antineoplásicos Alquilantes/síntese química , Carcinoma/tratamento farmacológico , Neoplasias Colorretais/tratamento farmacológico , Piperidinas/síntese química , Pró-Fármacos/síntese química , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/toxicidade , Biotransformação , Carcinoma/enzimologia , Carcinoma/patologia , Hipóxia Celular , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/patologia , Di-Hidrolipoamida Desidrogenase/metabolismo , Humanos , Proteínas de Neoplasias/metabolismo , Nitrogênio/química , Oxirredução , Óxidos/síntese química , Óxidos/farmacologia , Óxidos/toxicidade , Piperidinas/farmacologia , Piperidinas/toxicidade , Pró-Fármacos/farmacologia , Pró-Fármacos/toxicidade , Relação Estrutura-Atividade , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/enzimologia
17.
Free Radic Res ; 25(1): 75-86, 1996 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8814445

RESUMO

Fruits and vegetables contain several classes of compounds that can potentially contribute to antioxidant activity, including vitamins, simple and complex phenolics, sulphur-containing compounds and glucosinolates. The glucosinolates are found in high concentration in many cruciferous vegetables, and it is well established that their breakdown products induce endogenous antioxidant defences such as quinone reductase and glutathione S-transferase in cells and in vivo. Despite the anticarcinogenic effect of these compounds in animal models, the direct antioxidant properties of this class of compounds have not been systematically studied. We therefore examined the free radical-scavenging properties of representative extracts and of purified glucosinolates from cruciferous vegetables, by measuring their effect on ascorbate- or NADPH/iron-induced peroxidation of human liver microsomes, ascorbate/iron-induced peroxidation on phospholipid liposomes, iron chelation and hydroxyl radical scavenging using the deoxyribose assay, total antioxidant potential using ABTS (2,2'-azinobis(3-ethyl-benzothiazoline-6-sulphonate)) and the bleomycin assay. Most of the extracts from cruciferous vegetables exhibited some antioxidant properties, although extracts from cooked Brussels sprouts increased the rate of microsomal lipid peroxidation. The effects in these assays were dependent upon processing and species of crucifer, and the glucosinolate content appeared to play a minor role in these effects, since purified glucosinolates exhibited only weak antioxidant properties. The total antioxidant activities of extracts from cooked and autolysed Brussels sprouts were identical within experimental error. This is probably due to the content of phenolics which is unaltered by autolysis, despite the differences between these samples in other assays especially NADPH-iron-induced lipid peroxidation of human liver microsomes. The results demonstrate that glucosinolates are unlikely to account for the direct antioxidant effects of extracts from cruciferous vegetables.


Assuntos
Antioxidantes/farmacologia , Glucosinolatos/farmacologia , Extratos Vegetais/farmacologia , Verduras/química , Ácido Ascórbico/química , Ácido Ascórbico/metabolismo , Ácido Ascórbico/farmacologia , Benzotiazóis , Bleomicina/farmacologia , Brassica/química , Dano ao DNA/efeitos dos fármacos , Desoxirribose/química , Desoxirribose/metabolismo , Sequestradores de Radicais Livres , Glucosinolatos/isolamento & purificação , Glucosinolatos/metabolismo , Humanos , Radical Hidroxila , Ferro/química , Ferro/metabolismo , Ferro/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Lipossomos/metabolismo , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , NADP/química , NADP/metabolismo , Oxirredução , Fosfolipídeos/metabolismo , Extratos Vegetais/química , Especificidade da Espécie , Ácidos Sulfônicos/metabolismo , Ácidos Sulfônicos/farmacologia
18.
Br J Urol ; 77(3): 363-6, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8814839

RESUMO

OBJECTIVES: To determine the effect of pH on the cytotoxicity of mitomycin C on a wide range of established cell lines. MATERIALS AND METHODS: Cultures of three human tumour cell lines, including two bladder carcinomas and one renal adenocarcinoma, and two mouse cell lines were treated with mitomycin C (concentration range 320 pM-50 microM) at a range of pH (5.8-7.8) for 2 h. The cytotoxicity of mitomycin C over this range was determined using a tetrazolium-dye-based microtitration assay. RESULTS: The cytotoxicity of mitomycin C was significantly greater (confidence level > or = 95%) in acidic conditions (pH 5.8-6.0) than in neutral to alkaline pH (7.2-7.8) for three of the five cell lines. The same trend, although less marked, also occurred in the other two cell lines. CONCLUSION: The cytotoxicity of mitomycin C was greater under acidic conditions for all these cell lines confirming that the activity of mitomycin C depends on pH and suggesting that the pH of the vehicle used for intravesical chemotherapy may be an important factor in the successful treatment of bladder carcinomas. The modification of pH by urine may explain some of the variation in success rates among patients treated intravesically with mitomycin C.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Mitomicina/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , Adenocarcinoma/tratamento farmacológico , Animais , Relação Dose-Resposta a Droga , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Neoplasias da Bexiga Urinária/tratamento farmacológico
19.
Carcinogenesis ; 16(5): 1191-4, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7767984

RESUMO

We have tested the ability of a representative range of dietary glucosinolates and their breakdown products, found in high concentrations in cruciferous vegetables, to act as blocking agents against carcinogenesis by inducing the activity of the anticarcinogenic phase II marker enzyme quinone reductase in murine hepa1c1c7 cells. Breakdown of glucosinolates was catalysed by the endogenous plant enzyme thioglucoside glucohydrolase at neutral and acid pH. Only two unmodified glucosinolates, p-hydroxybenzyl and 2-hydroxybut-3-enyl, significantly induced quinone reductase activity. However, after enzymic hydrolysis at near-neutral pH, some of the glucosinolates yielded breakdown products that significantly induced quinone reductase in the order: 3-methylsulphinylpropyl-->prop-2-enyl-->pent-4-enyl approximately 2-phenylethyl approximately benzyl-->all others tested. Incubation with myrosinase at acidic pH resulted in induction of quinone reductase activity by the hydrolysis products of only three of the tested glucosinolates:3-methylsulphinyl-propyl approximately 2-phenylethyl-->benzyl-->all others, activity due to the two alkenyl compounds being lost. The results show that the potential cancer-blocking action of both intact and thioglucoside glucohydrolase-treated glucosinolates, as assessed by induction of phase II enzyme activity, is dependent on the nature of the side chain of the parent glucosinolate.


Assuntos
Anticarcinógenos/farmacologia , Glucosinolatos/farmacologia , Neoplasias Hepáticas Experimentais/enzimologia , NAD(P)H Desidrogenase (Quinona)/biossíntese , Animais , Anticarcinógenos/metabolismo , Biotransformação , Sobrevivência Celular/efeitos dos fármacos , Dieta , Indução Enzimática , Glucosinolatos/administração & dosagem , Glucosinolatos/metabolismo , Glicosídeo Hidrolases/metabolismo , Cinética , Camundongos , Mostardeira/enzimologia , Plantas Medicinais , Células Tumorais Cultivadas
20.
Br J Cancer ; 70(6): 1136-43, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7526885

RESUMO

Aerobic sensitivity to indoloquinone EO9 has been shown to correlate with cellular levels of the two-electron reducing enzyme DT-diaphorase. However, little is known about the relative roles of one- and two-electron reducing enzymes in the hypoxic cytotoxicity of EO9. We have characterised a panel of 23 human tumour cell lines for both bioreductive enzyme activities and aerobic sensitivity to EO9. Eight cell lines were then selected for a comparison of aerobic and hypoxic sensitivities. Activities of DT-diaphorase showed a wide range (> 10,000-fold), while activities of the one-electron reducing cytochrome b5 and cytochrome P450 reductases were generally lower and showed only a 15- and 25-fold range respectively. The aerobic cytotoxicity of EO9 was clearly related to the cellular levels of DT-diaphorase (r = 0.87), with higher levels giving increased sensitivity, but not to the levels of one-electron reducing enzymes. In contrast, there was no relationship between sensitivity to BCNU, cisplatin or the bioreductive agent SR 4233 (tirapazamine) and activities of any of these reducing enzymes. Under hypoxic conditions sensitivity to EO9 was markedly increased in cell lines with low levels of DT-diaphorase activity, while cell lines with high levels show only a small increase in sensitivity. This is reflected by a clear correlation (r = 0.98) between cellular DT-diaphorase activity and the ratio of aerobic to hypoxic sensitivity to EO9. However, we have now for the first time demonstrated an inverse correlation (r = 0.93) between the cellular activity of DT-diaphorase and hypoxic sensitivity to EO9, that is sensitivity decreases with increasing DT-diaphorase activity. Moreover, this correlation was lost when cells were exposed to drug in the presence of dicoumarol, supporting an involvement of DT-diaphorase in this relationship. These observations question the previously straightforward role for DT-diaphorase in the metabolic activation of EO9. Whereas DT-diaphorase is associated with increased toxicity in air, it appears to reduce the cytotoxicity of EO9 in hypoxic conditions. This suggests either that the one-electron reduction product of EO9 metabolism, the semiquinone, is more toxic than the two-electron reduction product, the hydroquinone, or that the hydroquinone is not cytotoxic and aerobic toxicity is due to the transient appearance of the semiquinone upon back oxidation of the hydroquinone.


Assuntos
Aziridinas/toxicidade , Indolquinonas , Indóis/toxicidade , Fígado/efeitos dos fármacos , NAD(P)H Desidrogenase (Quinona)/metabolismo , Aerobiose , Aziridinas/metabolismo , Biotransformação , Redutases do Citocromo/metabolismo , Citocromo-B(5) Redutase , Dicumarol/farmacologia , Humanos , Técnicas In Vitro , Indóis/metabolismo , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Oxirredução , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...