Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Invest New Drugs ; 30(5): 2035-45, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21912889

RESUMO

PURPOSE: The human tumour suppressor protein p53 is mutated in nearly half of human tumours and most mutant proteins have single amino acid changes. Several drugs including the quinazoline derivative 1 (CP-31398) have been reported to restore p53 activity in mutant cells. The side chain of 1 contains a styryl linkage that compromises its stability and we wished to explore the activity of analogues containing more stable side chains. METHODS: Reactivation of p53 function was measured by flow cytometry as the ability to potentiate radiation-induced G(1)-phase cell cycle arrest and by western blotting to determine expression of p21(WAF1). DNA binding was measured by competition with ethidium and preliminary pharmacological and xenograft studies were carried out. RESULTS: Screening of analogues for potentiation of radiation-induced G(1)-phase cell cycle arrest using NZOV11, an ovarian tumour cell line containing a p53(R248Q) mutation, demonstrated that the (2-benzofuranyl)-quinazoline derivative 5 was among the most active of the analogues. Compound 5 showed similar effects in several other p53 mutant human tumour cell lines but not in a p53 null cell line. 5 also potentiated p21(WAF1) expression induced by radiation. DNA binding affinity was measured and found to correlate with p53 reactivation activity. Plasma concentrations of 5 in mice were sufficient to suggest in vivo activity and a small induced tumour growth delay (7 days) of NZM4 melanoma xenografts was observed. CONCLUSION: Compound 5 restores p53-like function to a human tumour cells lines expressing a variety of mutant p53 proteins, thus providing a basis for the design of further new drugs.


Assuntos
Mutação/efeitos dos fármacos , Quinazolinas/farmacologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , DNA Topoisomerases Tipo II/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Camundongos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo
2.
Mutat Res ; 716(1-2): 59-65, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21939672

RESUMO

Wheat bran protects against mutations and cancer, but contains different plant cell types that are likely to have different protective effects. We previously described the production and chemical characterisation of an aleurone-rich fraction (ARF) and a pericarp-rich fraction (PRF) from wheat grain. We compared these with whole bran (WB), fed to rats as 10% of a high fat AIN-76 diet. All bran-supplemented diets increased faecal bulk, in the order PRF>WB>ARF. PRF increased the activity of NAD(P)H:quinone acceptor oxidoreductase only in the forestomach, whereas ARF and WB enhanced levels of glutathione S-transferase in the duodenum. ARF but not PRF was digested and fermented, and also encouraged bacterial growth. Rats were gavaged with the radioactive mutagen (14)C-labelled IQ (2-amino-3-methylimidazo[4,5-f]quinoline), and effects of the brans on plasma radioactivity measured. Compared with the control diet, all bran-supplemented diets reduced the concentration of radioactivity in plasma, in the order ARF>PRF>WB. All brans increased faecal elimination of radioactivity, but only ARF and PRF enhanced urinary radioactivity. These data suggest that wheat bran may reduce mutation and cancers through direct adsorption and enhanced elimination of a dietary mutagen and/or its metabolites, and that wheat bran enriched in pericarp or aleurone cell walls may exert protective effects through different mechanisms.


Assuntos
Antimutagênicos/farmacologia , Fibras na Dieta/farmacologia , Quinolinas/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Carcinógenos/metabolismo , Fezes/química , Feminino , Absorção Intestinal/efeitos dos fármacos , Quinolinas/sangue , Quinolinas/toxicidade , Quinolinas/urina , Ratos , Ratos Wistar
3.
Invest New Drugs ; 27(3): 280-4, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18696010

RESUMO

AIM: 5,6-Dimethylxanthenone-4-acetic acid (DMXAA) (ASA404), a low molecular weight antivascular drug currently in clinical trial, acts both directly on the tumour vascular endothelium and indirectly through the induction of inflammatory cytokines and other vasoactive molecules from macrophages and other host cells. We wished to determine whether co-administration of non-steroidal anti-inflammatory drugs (NSAIDs) could modulate the antivascular effects of DMXAA in mice. METHODS: The effects of diclofenac, salicylate, ibuprofen, celecoxib and rofecoxib on the antitumour response to DMXAA were compared using growth delay assays of Colon 38 adenocarcinomas in C57Bl mice. Concentrations of DMXAA in mice were measured by high performance liquid chromatography. RESULTS: Administration of DMXAA alone (25 mg/kg i.p.) or of NSAIDs alone induced small tumour growth delays from 2 to 7 days. Co-administration of each of the NSAIDs augmented DMXAA effects with tumour growth delays from 4.5 to >20 days. The possibility of a pharmacokinetic interaction was investigated using diclofenac and it was found that diclofenac did not affect DMXAA pharmacokinetics. CONCLUSIONS: NSAIDs increase the antitumour activity of DMXAA in a murine tumour model. The effects are consistent with hypothesis that NSAIDs antagonises some of the protective effects of prostaglandins released in response to vascular injury. Co-administration of NSAIDs with DMXAA might be considered as a possible strategy for use in combination cancer therapy.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Anti-Inflamatórios não Esteroides/uso terapêutico , Xantonas/uso terapêutico , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Neoplasias do Colo/tratamento farmacológico , Inibidores de Ciclo-Oxigenase/farmacologia , Inibidores de Ciclo-Oxigenase/uso terapêutico , Diclofenaco/uso terapêutico , Ensaios de Seleção de Medicamentos Antitumorais , Quimioterapia Combinada , Camundongos , Camundongos Endogâmicos C57BL , Xantonas/administração & dosagem , Xantonas/sangue
4.
Artigo em Inglês | MEDLINE | ID: mdl-18926778

RESUMO

N-[2-(Dimethylamino)ethyl]-2,6-dimethyl-1-oxo-1,2-dihydrobenzo[b]-1,6-naphthyridine-4-carboxamide (SN 28049) is a potent topoisomerase II poison being developed to treat solid tumours. A reliable and sensitive LC-MS method has been developed and validated for the determination of SN 28049 in plasma using a structurally similar internal standard. This method had acceptable intra- and inter-assay accuracy (95-105%) and precision (R.S.D.<6.5%) over the range 0.062-2.5 microM (using a 100 microl sample), and had a lower limit of quantitation of 0.062 microM. Both aqueous and plasma solutions of SN 28049 were stable during short-term (24h at room temperature or 4 degrees C) and long-term storage (8 months at -80 degrees C), and following freezing and thawing (three cycles). The method was applied to study the pharmacokinetics of SN 28049 in mice after iv administration (8.9 mg/kg; n=3 mice per time point). The maximum plasma concentration achieved was 1.22+/-0.05 microM, and concentrations were measurable up to 12h post-administration. A bi-exponential concentration-time curve was observed with an elimination half-life of 2.3+/-0.2h (mean+/-S.E.), a volume of distribution of 34.5+/-2.2l/kg, and a plasma clearance of 12+/-0.5l/h/kg.


Assuntos
Antineoplásicos/sangue , Cromatografia Líquida/métodos , Espectrometria de Massas/métodos , Naftiridinas/sangue , Animais , Antineoplásicos/farmacocinética , Estabilidade de Medicamentos , Feminino , Meia-Vida , Humanos , Infusões Intravenosas , Camundongos , Naftiridinas/análise , Naftiridinas/metabolismo , Naftiridinas/farmacocinética , Padrões de Referência , Análise de Regressão , Sensibilidade e Especificidade
5.
Clin Cancer Res ; 9(17): 6545-50, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14695159

RESUMO

PURPOSE: 5,6-Dimethylxanthenone-4-acetic acid (DMXAA), a new anticancer drug that has recently completed Phase I clinical trial, is effective against transplantable murine tumors with established vasculature. We wished to determine the relationship between administration schedule and antitumor activity. EXPERIMENTAL DESIGN: C57Bl/6 mice with s.c. implanted Colon 38 tumors were used for determination of maximal tolerated doses and tumor growth delay. Plasma and tissue DMXAA concentrations were measured by high-performance liquid chromatography. RESULTS: Continuous infusion (30 mg/kg/day for 3 days) and daily i.p. administration schedules (7.5 mg/kg) were ineffective. A pharmacokinetically guided schedule was developed to increase tumor tissue drug concentrations without increasing the maximal plasma concentration. A schedule comprising a loading dose (25 mg/kg, i.p.) followed by supplementary doses (5 mg/kg after 4 and 8 h) provided a 1.6-fold increase in tumor tissue area under the concentration-time curve, no increased toxicity, and superior antitumor activity (100% cure rate, as compared with 55% for a single i.p. dose of 25 mg/kg). A similar strategy was developed for oral administration with a loading dose (30 mg/kg) and supplementary doses (15 mg/kg after 4 and 8 h). It provided a 90% cure rate, in contrast to a single oral dose (0% cure rate). CONCLUSIONS: The antitumor action of DMXAA is schedule dependent, and the achievement of an adequate tumor tissue DMXAA concentration above a threshold value appears to be critical for activity. The use of a pharmacokinetically guided schedule provides excellent oral activity against Colon 38 tumors and provides a basis for developing more effective administration schedules in clinical trials.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Xantonas/administração & dosagem , Xantonas/farmacologia , Administração Oral , Animais , Área Sob a Curva , Linhagem Celular Tumoral , Ensaios Clínicos como Assunto , Dose Máxima Tolerável , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Fatores de Tempo
6.
Clin Cancer Res ; 10(17): 5949-56, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15355928

RESUMO

PURPOSE: Thalidomide has a variety of biological effects that vary considerably according to the species tested. We sought to establish whether differences in pharmacokinetics could form a basis for the species-specific effects of thalidomide. EXPERIMENTAL DESIGN: Mice and rabbits were administered thalidomide (2 mg/kg) p.o. or i.v., and plasma concentrations of thalidomide were measured after drug administration using high performance liquid chromotography. Plasma samples from five multiple myeloma patients over 24 hours after their first dose of thalidomide (200 mg) were similarly analyzed and all data were fitted to a one-compartment model. Metabolites of thalidomide in plasma were identified simultaneously using liquid chromatography-mass spectrometry. RESULTS: Plasma concentration-time profiles for the individual patients were very similar to each other, but widely different pharmacokinetic properties were found between patients compared with those in mice or rabbits. Area under the concentration curve values for mice, rabbits, and multiple myeloma patients were 4, 8, and 81 micromol/L. hour, respectively, and corresponding elimination half-lives were 0.5, 2.2, and 7.3 hours, respectively. Large differences were also observed between the metabolite profiles from the three species. Hydrolysis products were detected for all species, and the proportion of hydroxylated metabolites was higher in mice than in rabbits and undetectable in patients. CONCLUSIONS: Our results show major interspecies differences in the pharmacokinetics of thalidomide that are related to the altered degree of metabolism. We suggest that the interspecies differences in biological effects of thalidomide may be attributable, at least in part, to the differences in its metabolism and hence pharmacokinetics.


Assuntos
Inibidores da Angiogênese/metabolismo , Inibidores da Angiogênese/farmacocinética , Mieloma Múltiplo/sangue , Talidomida/metabolismo , Talidomida/farmacocinética , Adulto , Idoso , Idoso de 80 Anos ou mais , Inibidores da Angiogênese/administração & dosagem , Animais , Área Sob a Curva , Cromatografia Líquida de Alta Pressão , Cromatografia Líquida , Feminino , Humanos , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Coelhos , Especificidade da Espécie , Talidomida/administração & dosagem
7.
Eur J Pharm Sci ; 24(5): 513-24, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15784341

RESUMO

5,6-Dimethylxanthenone-4-acetic acid (DMXAA), a potent cytokine inducer, exhibited marked antitumor activity when given as multiple oral doses in mice. The aim of this study was to examine the transport of DMXAA and its acyl glucuronide (DMXAA-G) using the human Caco-2 cells. DMXAA was minimally metabolized by Caco-2 cells and both DMXAA and DMXAA-G were taken up to a minor extent by the cells. The permeability coefficient (Papp) values of DMXAA over 10-500 microM were 4x10(-5) cm/s to 4.3x10(-5) cm/s for both apical (AP) to basolateral (BL) and BL-AP transport, while the Papp values for the BL to AP flux of DMXAA-G were significantly greater than those for the AP to BL flux, with Rnet values of 4.5-17.6 over 50-200 microM. The BL to AP active efflux of DMXAA-G followed Michaelis-Menten kinetics, with a Km of 83.5+/-5.5 microM, and Vmax of 0.022+/-0.001 nmol/min. The flux of DMXAA-G was energy and Na+-dependent and MK-571 significantly (P<0.05) inhibited its BL to AP flux, with an estimated Ki of 130 microM. These data indicate that the transport of DMXAA across Caco-2 monolayers was through a passive process, whereas the transport of DMXAA-G was mediated by MRP1/2.


Assuntos
Antineoplásicos/farmacocinética , Glucuronídeos/farmacocinética , Xantonas/farmacocinética , Trifosfato de Adenosina/antagonistas & inibidores , Transporte Biológico , Células CACO-2 , Estabilidade de Medicamentos , Humanos , Concentração de Íons de Hidrogênio , Temperatura , Xantonas/farmacologia
8.
Biochem Pharmacol ; 65(11): 1853-65, 2003 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-12781337

RESUMO

Cancer chemotherapy is characterized by significant interindividual variations in systemic clearance, therapeutic response, and toxicity. These variations are due mainly to genetic factors, leading to alterations in drug metabolism and/or target proteins. The aim of this study was to determine, using a human liver bank (N=14), the interindividual variations in the expression and activity of liver enzymes that metabolize the investigational anticancer drug 5,6-dimethylxanthenone-4-acetic acid (DMXAA), i.e cytochrome P450 (CYP1A2) and uridine diphosphate glucuronosyltransferase (UGT1A9/2B7). In addition, interindividual variations in enzyme inhibition, hydrolysis of DMXAA acyl glucuronide (DMXAA-G) by plasma and hepatic microsomes, and the binding of DMXAA by plasma proteins also were examined. The results indicated that there was approximately one order of magnitude of interindividual variation in the expression of CYP1A2 and UGT2B7, activity of the enzymes toward DMXAA, and inhibition potency (IC(50)) by diclofenac, cyproheptadine, and alpha-naphthoflavone. The enzyme activities toward DMXAA and IC(50) values were closely correlated with enzyme expression. There was a smaller (2- to 3-fold) variation in the enzyme-catalyzed hydrolysis of DMXAA acyl glucuronide in human plasma and liver microsomes (N=6) and in the binding of DMXAA by plasma proteins in humans. In conclusion, the interindividual variability of DMXAA disposition observed in vitro might reflect the greater elimination variability (>one order of magnitude) in Phase I cancer patients. The variability in DMXAA clearance in these cancer patients would be due mainly to differences in its metabolism and its metabolic inhibition by co-administered drugs. To a lesser extent, variability in the clearance of DMXAA could be due to the hydrolysis of its acyl glucuronide and/or its binding to plasma proteins. Further study is needed to examine the genotype-phenotype relationship, and the result, together with therapeutic drug monitoring may provide a useful strategy for optimizing DMXAA treatment.


Assuntos
Antineoplásicos/farmacocinética , Drogas em Investigação/farmacocinética , Taxa de Depuração Metabólica/fisiologia , Microssomos Hepáticos/metabolismo , Xantenos/farmacocinética , Xantonas , Antineoplásicos/metabolismo , Sítios de Ligação , Proteínas Sanguíneas/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Drogas em Investigação/metabolismo , Humanos , Hidrólise , Xantenos/metabolismo
9.
Biochem Pharmacol ; 65(1): 109-20, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12473385

RESUMO

It is important to determine the relative contribution of each metabolic pathway (f(p)) and of enzymes to the net metabolism of a drug. The aim of this study was to investigate, using a human liver bank, the f(p) of the anti-cancer drug 5,6-dimethylxanthenone-4-acetic acid (DMXAA) and the effects of various inhibitors and inducers on f(p). The mean apparent K(m) and V(max) values (N=14) were 21+/-5 microM and 0.04+/-0.02 nmol/min/mg, respectively, for 6-methylhydroxylation, and 143+/-79 microM and 0.71+/-0.52 nmol/min/mg, respectively, for acyl glucuronidation in human liver microsomes. 6-Methylhydroxylation and acyl glucuronidation contributed 26 and 74%, respectively, to DMXAA metabolism at 5 microM; values were 7 and 93% at 350 microM DMXAA. There was a significant relationship between the ratio of metabolic activity by Phase II and I reactions (R(II/I)) and uridine diphosphate glucuronosyltransferase (UGT2B7) protein level (r=0.605, P=0.022), whereas a reverse correlation between R(II/I) and cytochrome P450 (CYP1A) protein level was observed (r=-0.540, P=0.046). Various compounds inhibited either DMXAA glucuronidation or 6-methylhydroxylation, or both pathways. Pretreatment of rats with beta-naphthoflavone, but not phenobarbitone and cimetidine, increased the percentage of the contribution by 6-methylhydroxylation to 17% from 4% of control at 5 microM DMXAA. Our results indicate that the f(p) of DMXAA is subject to substrate concentration, inhibition, induction, and the protein levels of enzymes that biotransform DMXAA. However, clinical studies are important to verify the conclusions drawn from in vitro data.


Assuntos
Antineoplásicos/metabolismo , Microssomos Hepáticos/metabolismo , Xantenos/metabolismo , Xantonas , Animais , Antineoplásicos/farmacologia , Enzimas/metabolismo , Humanos , Masculino , Microssomos Hepáticos/enzimologia , Ratos , Ratos Endogâmicos WKY , Xantenos/farmacologia
10.
Biochem Pharmacol ; 67(5): 937-45, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15104247

RESUMO

The induction of haemorrhagic necrosis by 5,6-dimethylxanthenone-4-acetic acid (DMXAA) in transplantable murine tumours depends on the in situ synthesis of cytokines, particularly tumour necrosis factor (TNF). Since the in vivo action of DMXAA would be greatly clarified by the development of an in vitro model, we investigated whether DMXAA could induce cytokines in cultured murine splenocytes. DMXAA alone induced low amounts of TNF with an optimal concentration of 10 microg/mL and an optimal time of 4 hr. When combined with low concentrations of lipopolysaccharide, deactivated-lipopolysaccharide (dLPS) or phorbol-12-myristate-13-acetate that did not elicit TNF production alone, synergistic TNF production was obtained. DMXAA also induced interferon-gamma at an optimal dose of 300 microg/mL, but the addition of dLPS had no further effect. Decreasing culture pH, although not changing the optimal concentrations for stimulation, increased both TNF and interferon-gamma production in response to DMXAA. The major DMXAA metabolites, DMXAA-glucuronide and 6-hydroxy-5-methylxanthenone-4-acetic acid, did not induce either cytokine alone, in combination with dLPS or at low pH. The results indicate that DMXAA rather than a metabolite is responsible for cytokine induction and suggest that the microenvironment of the tumour may be responsible for the observed selective induction of cytokines in tumour tissue.


Assuntos
Interferon gama/metabolismo , Baço/citologia , Fator de Necrose Tumoral alfa/metabolismo , Xantonas/farmacologia , Animais , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Células Cultivadas , Citocinas/biossíntese , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Endogâmicos C57BL , Xantonas/metabolismo
11.
Cancer Chemother Pharmacol ; 49(1): 20-6, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11855749

RESUMO

PURPOSE: 5,6-Dimethylxanthenone-4-acetic acid (DMXAA), an anticancer drug with an antivascular action, has recently completed phase I clinical trials. Since oral administration has many advantages, we compared the biological activity and pharmacokinetics of DMXAA in mice following oral and intraperitoneal (i.p.) administration. METHODS: Growth delays of Colon 38 tumours were measured in C57Bl/6 mice. Plasma concentrations of DMXAA, 5-hydroxyindole-3-acetic acid (5HIAA) as a measure of serotonin production, and nitrate as a measure of nitric oxide production, were determined by high-performance liquid chromatography. Tumour necrosis factor (TNF) concentrations in serum and tumour tissues were measured by ELISA. RESULTS: The antitumour activity of DMXAA at the maximum tolerated oral dose (32.5 mg/kg) was low (4-day growth delay, no cures) compared to that (19-day growth delay, 40% cures) at the maximum tolerated i.p. dose (27.5 mg/kg). The pharmacokinetics of DMXAA in plasma, liver and tumour tissue indicated a bioavailability of 73%. Elevation of plasma 5HIAA, measured 4 h following i.p. administration of DMXAA, was linear with DMXAA dose, and the 5HIAA response to oral administration was consistent with its bioavailability. TNF concentrations increased following oral administration (30 mg/kg) and were particularly evident in tumour tissue, but were lower and less prolonged than those in response to i.p. administration at 25 mg/kg. Plasma nitrate levels were not increased following oral administration (30 mg/kg). CONCLUSIONS: DMXAA exhibits good bioavailability, and changes in serum TNF, tissue TNF, plasma 5HIAA and plasma nitrate, as markers of biological response, are consistent with this bioavailability. The low maximal plasma DMXAA concentration following oral administration, resulting in reduced retention of intratumoral TNF, may be responsible for the low antitumour activity.


Assuntos
Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/farmacocinética , Antineoplásicos/farmacologia , Antineoplásicos/farmacocinética , Neoplasias do Colo/tratamento farmacológico , Xantenos/farmacologia , Xantenos/farmacocinética , Xantonas , Administração Oral , Animais , Neoplasias do Colo/sangue , Neoplasias do Colo/patologia , Feminino , Meia-Vida , Ácido Hidroxi-Indolacético/sangue , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Óxido Nítrico/sangue , Fator de Necrose Tumoral alfa/metabolismo
12.
Cancer Chemother Pharmacol ; 49(2): 126-32, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11862426

RESUMO

PURPOSES: Marked gender differences in the pharmacokinetics of many drugs have been reported. For the investigational anticancer drug, 5,6-dimethylxanthenone-4-acetic acid (DMXAA), negligible gender differences in the plasma pharmacokinetics have been observed in mice. The gender effects on the plasma pharmacokinetics of DMXAA were further investigated using the rat model. In addition, the in vitro metabolism and plasma protein binding of DMXAA in male and female mice, rats and humans were investigated. METHODS: DMXAA was administered to male and female rats by intravenous injection. DMXAA and its major metabolites formed in liver microsomes were determined by HPLC. Unbound DMXAA in plasma was separated by ultrafiltration followed by HPLC determination. RESULTS: In vivo kinetic studies indicated that female rats had 60%, 55% and 73% higher area under the plasma concentration-time curve (AUC) of DMXAA (2413 +/- 188 vs 1505 +/- 312 microM x h, P<0.05), elimination half-life (2.40 +/- 0.45 vs 1.55 +/- 0.33 h) and maximal plasma concentration (Cmax) (1236 +/- 569 vs 716 +/- 280 micro M), but 61% lower plasma clearance than male rats. In vitro studies indicated that male rats had a 67% higher glucuronidation activity (0.75 +/- 0.03 nmol/min per mg) than female rats (0.45 +/- 0.01 nmol/min per mg), resulting in a 96% faster intrinsic clearance (CL(int)) in the males than the females (6.36 +/- 0.65 vs 3.24 +/- .42 ml/min per g, P< 0.05). In contrast, female rats had 25% higher 6-methylhydroxylation activity (0.045 +/- 0.003 nmol/min per mg) than male rats (0.036 +/- 0.002 nmol/min per mg), resulting in a 57% faster intrinsic clearance (CL(int)) in the females than males (0.36 +/- 0.06 vs 0.23 +/- 0.05 ml/min per g). Overall, total CL(int) by both glucuronidation and 6-methylhydroxylation in male rats was 83% higher than in female rats (6.59 +/- 2.11 vs 3.60 +/- 1.07 nmol/min per g). Men ( n=4) had a significantly lower ( P<0.05) CL(int) for glucuronidation than women ( n=10), but a higher CL(int) for 6-methylhydroxylation, resulting in significantly higher total CL(int) in women than men (5.63 vs 8.33 nmol/min per g). There was no significant difference in either the total plasma protein or albumin concentration between male and female mice, rats or humans. CONCLUSION: There were significant gender-related differences in the metabolism and pharmacokinetics in the rat, in contrast to the mouse. This indicates a limited usefulness of the rat as a model for the study of DMXAA metabolism in relation to gender differences, although the gender differences in the in vitro metabolic capacity for DMXAA may provide an explanation for the gender differences in the pharmacokinetics in rats. Data from human liver microsomes may allow the prediction of gender effects in the in vivo pharmacokinetics of DMXAA.


Assuntos
Antineoplásicos/metabolismo , Xantenos/metabolismo , Xantonas , Animais , Proteínas Sanguíneas/metabolismo , Feminino , Humanos , Masculino , Taxa de Depuração Metabólica , Camundongos , Camundongos Endogâmicos C57BL , Microssomos Hepáticos/metabolismo , Ligação Proteica , Ratos , Ratos Endogâmicos WKY , Fatores Sexuais
13.
Cancer Chemother Pharmacol ; 53(5): 377-83, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15060740

RESUMO

PURPOSE: There is considerable current interest in the use of thalidomide as a single agent or in combination with drugs such as cyclophosphamide in the treatment of multiple myeloma and other cancers. Our previous work has shown that thalidomide potentiates the antitumour activity of both cyclophosphamide and 5,6-dimethylxanthenone-4-acetic acid (DMXAA) against murine Colon 38 tumours. In both of these cases, thalidomide extends the half-life (t(1/2)) of the other drug. We wished to determine whether cyclophosphamide and DMXAA altered the t(1/2) of thalidomide. Since both thalidomide and DMXAA modulate tumour necrosis factor (TNF), we also wished to determine the role of TNF in this interaction. METHODS: Mice with Colon 38 tumours were treated with cyclophosphamide (220 mg/kg) and/or thalidomide (20 mg/kg) or DMXAA (25 mg/kg) and thalidomide (100 mg/kg), combinations that have previously demonstrated synergistic activity. Plasma and tumour tissue drug concentrations were analysed by high-performance liquid chromatography. To determine the role of TNF, similar experiments were performed using mice defective in the TNF gene (TNF(-/-)) or the TNF receptor-1 gene (TNFR1(-/-)). RESULTS: Coadministration of cyclophosphamide increased the thalidomide t(1/2) by 3.9- and 3.6-fold, respectively, in plasma and tumour tissue, with a corresponding increase in the concentration-time curve (AUC). The corresponding values following coadministration of DMXAA were 3.0- and 4.6-fold, respectively. Coadministration of cyclophosphamide had similar effects on thalidomide t(1/2) in C57Bl/6, TNF(-/-) and TNFR1(-/-) mice, while coadministration of DMXAA did not alter the t(1/2) or AUC in TNF(-/-) and TNFR1(-/-) mice. CONCLUSIONS: Both cyclophosphamide and DMXAA have a pharmacokinetic interaction with thalidomide, increasing t(1/2) and AUC. TNF mediates the effect of DMXAA on thalidomide pharmacokinetics but not that of cyclophosphamide.


Assuntos
Ciclofosfamida/farmacologia , Talidomida/farmacocinética , Xantonas/farmacologia , Animais , Área Sob a Curva , Interações Medicamentosas , Camundongos , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/fisiologia
14.
Cancer Chemother Pharmacol ; 50(3): 186-92, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12203100

RESUMO

PURPOSE: Thalidomide has recently shown significant promise in the treatment of some types of cancer, and trials in combination with conventional chemotherapy are being undertaken. We wished to determine whether thalidomide potentiated the effect of cyclophosphamide, a commonly used cytotoxic drug, in a murine tumour model. METHODS: C57Bl/6 mice implanted with subcutaneous Colon 38 tumours were treated with cyclophosphamide alone or together with thalidomide as a single intraperitoneal injection and tumour growth was measured. Concentrations of cyclophosphamide, 4-hydroxycyclophosphamide, 4-ketocyclophosphamide and 2-dechloroethylcyclophosphamide were determined in plasma, liver and tumour tissue using coupled high-performance liquid chromatography-mass spectrometry at different times after treatment. RESULTS: Cyclophosphamide alone (220 mg/kg) induced growth delays of 11-13 days with no cures, whereas cyclophosphamide together with thalidomide (100 mg/kg) cured mice of their tumours. Thalidomide at lower doses (1-20 mg/kg) also potentiated the antitumour effect. Coadministration of thalidomide (100 mg/kg) dramatically decreased the clearance of cyclophosphamide and its metabolites from plasma and tissue, with corresponding increases in the area under the concentration-time curves. The magnitude of the effect was dependent on the dose of thalidomide over the range 1-20 mg/kg with no further effect at a dose of 100 mg/kg. CONCLUSIONS: Coadministration of thalidomide and cyclophosphamide gave markedly greater activity against Colon 38 tumour compared with either drug alone. Investigation of the reason for this effect revealed thalidomide to possess the novel property of dramatically decreasing the clearance of cyclophosphamide and its metabolites.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/farmacocinética , Neoplasias do Colo/tratamento farmacológico , Ciclofosfamida/farmacologia , Ciclofosfamida/farmacocinética , Imunossupressores/farmacologia , Animais , Relação Dose-Resposta a Droga , Interações Medicamentosas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais
15.
Drugs Aging ; 19(2): 85-100, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11950376

RESUMO

There is increased interest in the treatment of cancer with thalidomide because of its antiangiogenic, immunomodulating and sedative effects. In animal models, the antitumour activity of thalidomide is dependent on the species, route of administration and coadministration of other drugs. For example, thalidomide has shown antitumour effects as a single agent in rabbits, but not in mice. In addition, the antitumour effects of the conventional cytotoxic drug cyclophosphamide and the tumour necrosis factor inducer 5,6-dimethylxanthenone-4-acetic acid (DMXAA) were found to be potentiated by thalidomide in mice bearing colon 38 adenocarcinoma tumours. Further studies have revealed that thalidomide upregulates intratumoral production of tumour necrosis factor-alpha 10-fold over that induced by DMXAA alone. Coadministration of thalidomide also significantly reduced the plasma clearance of DMXAA and cyclophosphamide. All these effects of thalidomide may contribute to the enhanced antitumour activity. Recent clinical trials of thalidomide have indicated that it has minimal anticancer activity for most patients with solid tumours when used as a single agent, although it was well tolerated. However, improved responses have been reported in patients with multiple myeloma. Palliative effects of thalidomide on cancer-related symptoms have also been observed, especially for geriatric patients with prostate cancer. Thalidomide also eliminates the dose-limiting gastrointestinal toxic effects of irinotecan. There is preliminary evidence indicating that the clearance of thalidomide may be reduced in the elderly. The exact role of thalidomide in the treatment of cancer and cancer cachexia in the elderly remains to be elucidated. However, it may have some value as part of a multimodality anticancer therapy, rather than as a single agent.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Talidomida/uso terapêutico , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Animais , Humanos , Camundongos , Pessoa de Meia-Idade , Coelhos , Ratos
16.
Artigo em Inglês | MEDLINE | ID: mdl-12138005

RESUMO

The experimental anti-cancer drug 5,6-dimethylxanthenone-4-acetic acid (DMXAA) is mainly metabolised by acyl glucuronidation and to a lesser degree by 6-methyl hydroxylation. Strain differences in the maximum tolerated dose (MTD) of DMXAA in mice have been observed. The aim of this study was to compare the kinetics of DMXAA acyl glucuronidation and 6-methylhydroxylation in five various mouse strains, and correlate the in vitro metabolism data with MTD observed. In all mouse strains studied, DMXAA acyl glucuronidation and 6-methylhydroxylation in the liver microsomes followed Michaelis-Menten kinetics. Significant strain variations in the kinetic parameters (K(m), V(max) and K(m)/V(max), i.e., CL(int)) for DMXAA acyl glucuronidation and 6-methylhydroxylation in mouse liver microsomes were observed. A 2-6-fold variation was spanned across strains for K(m), V(max) and CL(int), respectively, for DMXAA glucuronidation and 6-methylhydroxylation. The rank order for total CL(int) by glucuronidation and 6-methylhydroxylation was BDF1 (1.70 ml/min per g)>wild type of mice lacking IFN-gamma receptor (0.80 ml/min per g)>nude mice (0.70 ml/min per g)>Swiss CD mice (0.56 ml/min per g)>C57Bl/6 mice (0.46 ml/min per g), with a 4-fold variation between the mouse strain of the highest and lowest CL(int). There was no significant correlation between total CL(int) and MTD (r(2)=0.88, P>0.05), but the rank order for CL(int) was consistent with that for MTD. These results suggested that there were significant strain differences in DMXAA metabolism in mouse liver microsomes and the strain-related differences in the metabolism of DMXAA did not provide an explanation for the strain differences in the MTD.


Assuntos
Antineoplásicos/metabolismo , Microssomos Hepáticos/metabolismo , Xantenos/metabolismo , Xantonas , Animais , Antineoplásicos/farmacocinética , Cromatografia Líquida de Alta Pressão , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade da Espécie , Xantenos/farmacocinética
17.
Artigo em Inglês | MEDLINE | ID: mdl-11863291

RESUMO

By screening potential inhibitors of drug metabolism using the in vitro models, potential drug-drug interactions in vivo may be predicted with the use of appropriate pharmacokinetic principles. This study aimed to develop a rapid screening system using human liver microsomes to efficiently identify the potential inhibitors of DMXAA metabolism. Initial IC50 was estimated by using a two-point method, and then Ki values were determined if required and compared with those initial IC50 values. More than 100 compounds including known substrates and inhibitors of human uridine diphosphate glucuronosyltransferases (UGTs) and cytochrome P450 (CYP), anti-cancer drugs and xanthenone analogues were screened for their inhibitory effect on DMXAA glucuronidation and 6-methylhydroxylation in human liver microsomes. Both metabolites of DMXAA, DMXAA acyl glucuronide (DMXAA-G) and 6-hydroxymethyl-5-methylxanthenone-4-acetic acid (6-OH-MXAA), formed in human liver microsomes were quantitated by validated HPLC methods. The results indicated that there was a significant relationship (r2 = 0.966, P < 0.001) between the two-point IC50 values and the apparent Ki values for 20 compounds showing significant inhibitory effects on DMXAA metabolism, suggesting the usefulness of the two-point determination for the initial screening of compounds. This study has been completed using a strategy for rapid HPLC analysis and thus provided early access to detailed information for potential inhibitors of DMXAA metabolism and allows for further DMXAA-drug interaction studies.


Assuntos
Antineoplásicos/antagonistas & inibidores , Xantenos/antagonistas & inibidores , Xantonas , Antineoplásicos/farmacocinética , Cromatografia Líquida de Alta Pressão/métodos , Humanos , Microssomos Hepáticos/metabolismo , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Espectrometria de Fluorescência , Xantenos/farmacocinética
18.
Artigo em Inglês | MEDLINE | ID: mdl-15282097

RESUMO

5,6-Dimethylxanthenone-4-acetic acid (DMXAA) is a potent cytokine inducer, with a bioavailability of >70% in the mouse. The aim of this study was to develop and validate HPLC methods for the determination of DMXAA and DMXAA acyl glucuronide (DMXAA-G) in the human intestinal cell line Caco-2 monolayers. The developed HPLC methods were sensitive and reliable, with acceptable accuracy (85-115% of true values) and precision (intra- and inter-assay CV < 15%). The total running time was within 6.8 min, with acceptable separation of the compounds of interest. The limit of quantitation (LOQ) values for DMXAA and DMXAA-G were 14.2 and 24 ng/ml, respectively. The validated HPLC methods were applied to examine the epithelial transport of DMXAA and DMXAA-G by Caco-2 monolayers. The permeability coefficient (Papp) values (overall mean +/- S.D., n = 3-9) of DMXAA over 10-500 microM were independent of concentration for both apical (AP) to basolateral (BL) (4.0 +/- 0.4 x 10(-5)cm/s) and BL-AP (4.3 +/- 0.5 x 10(-5)cm/s) transport, and of similar magnitude in either direction, with net efflux ratio (Rnet) values of 1-1.3. However, the Papp values for the BL to AP transport of DMXAA-G were significantly greater than those for the AP to BL transport, with Rnet values of 17.6, 6.7 and 4.5 at 50, 100 and 200 microM, respectively. Further studies showed that the transport of DMXAA-G was Na+- and energy-dependent, and inhibited by MK-571 [a multidrug resistance associated protein (MRP) 1/2 inhibitor], but not by verapamil and probenecid. These data indicate that the HPLC methods for the determination of DMXAA and DMXAA-G in the transport buffer were simple and reliable, and the methods have been applied to the transport study of both compounds by Caco-2 monolayers. DMXAA across Caco-2 monolayers was through a passive transcellular process, whereas the transport of DMXAA-G was mediated by MRP1/2.


Assuntos
Antineoplásicos/análise , Cromatografia Líquida de Alta Pressão/métodos , Glucuronídeos/análise , Espectrometria de Fluorescência/métodos , Xantonas/análise , Células CACO-2 , Humanos , Sensibilidade e Especificidade
19.
Food Chem Toxicol ; 41(6): 785-92, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12738184

RESUMO

It has commonly been believed that increasing fibre in the diet should reduce the incidence of cancers, especially those of the colon and rectum. The earliest definitions of dietary fibre restricted the term to plant cell walls in which non-starch polysaccharides are key chemical components. However, new definitions encompass a wider range of materials, including starches resistant to digestion in the colon (resistant starches). Nevertheless, most definitions require that "dietary fibres" show physiological effects considered beneficial against cancer, including enhanced laxation and faecal bulking. On theoretical grounds, such properties might be expected to dilute the concentration of any carcinogen present and move it more rapidly through the colon, thereby reducing bioavailability. We have compared the properties of two dietary fibre preparations that are primarily non-starch polysaccharides with two resistant starch preparations for effects on carcinogen disposition in a rodent model. Although both preparations enhanced laxation and faecal bulking, only the non-starch polysaccharide preparation reduced carcinogen biovailability. Indeed, carcinogen biovailability was significantly enhanced by resistant starch. We suggest that there may be fundamental differences in the manner by which non-starch polysaccharides or resistant starches affect carcinogen disposition, and express concern that the events seen with the resistant starches [RS] are unlikely to be beneficial with respect to protection against cancer by exogenous carcinogens. Furthermore, the data reveal that the observation of enhanced laxation and faecal bulking does not necessarily imply a reduction in carcinogen bioavailability.


Assuntos
Anticarcinógenos/farmacologia , Carcinógenos/toxicidade , Polissacarídeos/farmacologia , Quinolinas/toxicidade , Amido/farmacologia , Animais , Disponibilidade Biológica , Carcinógenos/farmacocinética , Neoplasias Colorretais/prevenção & controle , Carboidratos da Dieta/farmacologia , Fibras na Dieta/farmacologia , Modelos Animais de Doenças , Fezes/química , Feminino , Trânsito Gastrointestinal/efeitos dos fármacos , Absorção Intestinal/efeitos dos fármacos , Quinolinas/farmacocinética , Ratos , Ratos Wistar
20.
J Pharm Pharmacol ; 54(7): 997-1003, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12162720

RESUMO

Total (added) drug concentrations other than unbound concentrations have been used to estimate the in-vitro enzyme kinetic parameters for 5,6-dimethylxanthenone-4-acetic acid (DMXAA), an experimental anti-cancer drug. This study aimed to investigate the non-specific binding of DMXAA to liver microsomes from variousspecies and to microsomesfrom human lymphoblastoid cells expressing drug-metabolising enzymes, and to examine the effect of the binding on the estimation of enzyme kinetic parameters for DMXAA in-vitro. The separation of unbound DMXAA was conducted by ultrafiltration and DMXAA concentrations were determined by validated HPLC. The results indicated that DMXAA was bound to liver microsomes and lymphoblastoid cell microsomes to a small extent (free fraction in microsomes, f(u(mic)) mostly > 0.85). Correction forthe unbound DMXAA concentration resulted in slightly lower apparent Michaelis-Menten constant (Km) values, but with the maximal velocity of reaction (Vmax) unchanged, leading to slightly higher unbound Vmax/Km values. These results indicate that the non-specific binding of DMXAA to microsomes is insignificant and has little impact on the enzyme kinetic estimation in-vitro.


Assuntos
Antineoplásicos/farmacocinética , Microssomos Hepáticos/efeitos dos fármacos , Xantenos/farmacocinética , Xantonas , Animais , Antineoplásicos/administração & dosagem , Sítios de Ligação , Cromatografia Líquida de Alta Pressão , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Coelhos , Ratos , Ratos Wistar , Xantenos/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA