Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Placenta ; 28(8-9): 868-77, 2007.
Article in English | MEDLINE | ID: mdl-17482262

ABSTRACT

Placental ATP-binding cassette (ABC) transporters limit fetal exposure to xenobiotics by regulating transplacental passage into the fetal circulation; their expression and function in fetal membranes, however, has not been studied. In the present study the expression, localisation and function of ABC transporters in human amnion was examined to explore their potential role in modulating amniotic fluid drug disposition in pregnancy. Single-assay oligo-microarrays were used to profile amnion gene expression, and drug transporters expressed at significant levels were identified and selected for further studies. The expression of ABCG2/breast cancer resistance protein (BCRP) and multidrug resistance-associated proteins (MRP) 1 (ABCC1), 2 (ABCC2) and 5 (ABCC5) was detected on the arrays, and verified by RT-PCR and immunoblotting. On confocal microscopy of fetal membrane cryosections, MRP1 and MRP5 were immunolocalised to both apical and basolateral surfaces of the amniotic epithelium, while MRP2 was expressed at low levels only in the apical membrane. BCRP in contrast showed cytoplasmic staining throughout the amniotic epithelium. In addition to the amnion, MRP1 and BCRP immunostaining was observed in the chorion and the decidua. Cell accumulation studies using selective MRP and BCRP inhibitors showed the transporters to be functionally active in amnion epithelial monolayer cultures. In contrast, transwell transport studies using intact amnion membranes did not show significant vectorial transport. These findings identify the amnion as a novel site of ABC drug transporter expression. Functional studies indicate that they may act primarily to prevent cellular xenobiotic accumulation, rather than to confer fetal protection through reduced accumulation in amniotic fluid.


Subject(s)
Amnion , Multidrug Resistance-Associated Proteins , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate , Amnion/metabolism , Humans , Membrane Transport Proteins , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics
2.
Placenta ; 28 Suppl A: S116-20, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17275084

ABSTRACT

Trophoblast cells undergo loss of plasma membrane lipid asymmetry during cell fusion without further progression to terminal phases of apoptosis. The nature of the anti-apoptotic mechanisms providing cell survival during this process is unknown. Using a BeWo cell model, we explored the role of the xenobiotic/lipid transporter ABCG2 in promoting cell survival during forskolin-induced differentiation. Suppression of ABCG2 expression by siRNA led to a marked increase in phosphatidylserine externalisation followed by accumulation of ceramides and increased apoptosis. Expression of markers of syncytial formation (beta-hCG and HERV-W) was decreased by ABCG2 silencing, although fusion was unaffected. These findings suggest that ABCG2 protects cells during the period of transient membrane instability associated with cell differentiation and fusion, highlighting a novel, previously unrecognised role of ABCG2 as a survival factor during the formation of the placental syncytium.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Cell Differentiation , Giant Cells/cytology , Neoplasm Proteins/physiology , Trophoblasts/cytology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , Apoptosis/genetics , Cell Survival/genetics , Cells, Cultured , Colforsin/pharmacology , Female , Giant Cells/metabolism , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Phosphatidylserines/metabolism , RNA, Small Interfering/pharmacology , Trophoblasts/metabolism , Xenobiotics/metabolism
3.
Cancer Chemother Pharmacol ; 59(5): 681-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17021822

ABSTRACT

PURPOSE: 5,6-Dimethylxanthenone-4-acetic acid (DMXAA) (AS1404) is a novel antitumour agent that selectively disrupts tumour vasculature and induces cytokines. The purpose of this study was to determine the pharmacokinetics (PK) of DMXAA in cancer patients enrolled in a phase I clinical trial. METHODS: DMXAA was administered as a 20-min i.v. infusion every 3 weeks and doses were escalated in cohorts of patients according to a predefined schema. PK samples were taken over the first 24 h of at least the first cycle. RESULTS: DMXAA was administered to 63 patients at 19 dose levels from 6 to 4,900 mg m(-2), and 3,700 mg m(-2) was established as the maximum tolerated dose. The PK observed over the dose range showed a non-linear fall in clearance from 16.1 to 1.42 l h(-1) m(-2) and resultant increase in the area under the concentration-time curve (AUC) from 1.29 to 12,400 microM h. In contrast, the increase in peak plasma concentrations from 2.17 to 1,910 microM approximated linearity. DMXAA was highly protein-bound to albumin (>99%) until saturation occurred at higher doses, leading to a rapid increase in the free fraction (up to 20%) and greater concentrations of DMXAA bound to non-albumin proteins. However, the main determinant of the non-linearity of the PK appeared to be sequential saturation of elimination mechanisms, which include hydroxylation, glucuronidation and perhaps hepatic transport proteins. This resulted in an exaggerated non-linear increase in free DMXAA plasma concentrations and AUC compared to total drug. CONCLUSIONS: The PK of DMXAA are well-defined, with a consistent degree of non-linearity across a very large dose range.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Xanthones/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Area Under Curve , Cohort Studies , Dose-Response Relationship, Drug , Half-Life , Humans , Linear Models , Nonlinear Dynamics , Xanthones/administration & dosage , Xanthones/adverse effects
4.
Xenobiotica ; 32(2): 87-107, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11868972

ABSTRACT

1. Mouse studies have indicated that the antitumour effects of 5,6-dimethylxanthenone-4-acetic acid (DMXAA) are dramatically potentiated in combination with other drugs, and it has been proposed that optimization of the therapeutic potential of DMXAA would exploit combination therapy. The aim was to identify the most appropriate animal model for further investigations of the pharmacokinetics of possible DMXAA-drug combinations and their extrapolation to patients. 2. Qualitatively, the metabolic profile for DMXAA in liver microsomes was similar in mouse, rat, rabbit and humans, with glucuronidation and 6-methylhydroxylation the two major metabolic pathways. In all species, the intrinsic clearance by glucuronidation was at least 2-fold that due to hydroxylation. There was significant variability in the in vitro kinetic parameters (Km, Vmax), with the mouse being the least efficient DMXAA metabolizer compared with the other species. 3. Mouse, rat and rabbit renal microsomes exhibited DMXAA glucuronidation activity, but only the rabbit showed 6-methylhydroxylation. For the total in vitro CL(int) (Vmax/Km) by glucuronidation and 6-methylhydroxylation, the ratio of kidney:liver was 0.67, 0.03 and 0.34 in the mouse, rat and rabbit respectively. However, taking into account the liver and kidney weight difference, it is apparent that the in vivo renal metabolism would not be a major contributor to the overall elimination of DMXAA. 4. The inhibitory profile for liver DMXAA glucuronidation was similar across species, but there was remarkable interspecies variability in the inhibition of liver DMXAA 6methylhydroxylation. 5. Extrapolation of in vitro intrinsic clearance to in vivo gave a significant underestimation of plasma clearance for all species. However, there was a significant allometric relationship for plasma clearance and volume of distribution, but not for maximum tolerated dose across species. 6. The results indicate that animal models may have a limited role in the extrapolation to patients of drug interactions with agents such as DMXAA that have immunomodulating activity that may vary widely between species.


Subject(s)
Antineoplastic Agents/metabolism , Xanthenes/metabolism , Xanthones , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Humans , In Vitro Techniques , Kidney/metabolism , Liver/metabolism , Mice , Microsomes/metabolism , Microsomes, Liver/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Rabbits , Rats , Species Specificity , Xanthenes/pharmacokinetics , Xanthenes/pharmacology
5.
Br J Clin Pharmacol ; 52(2): 129-36, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11488768

ABSTRACT

AIMS: To investigate the effects of various anticancer drugs on the major metabolic pathways (glucuronidation and 6-methylhydroxylation) of DMXAA in human liver microsomes. METHODS: The effects of various anticancer drugs at 100 and 500 microM on the formation of DMXAA acyl glucuronide (DMXAA-G) and 6-hydroxymethyl-5-methylxanthenone-4-acetic acid (6-OH-MXAA) in human liver microsomes were determined by high performance liquid chromatography (h.p.l.c.). For those anticancer drugs showing significant inhibition of DMXAA metabolism, the inhibition constants (Ki) were determined. The resulting in vitro data were extrapolated to predict in vivo changes in DMXAA pharmacokinetics. RESULTS: Vinblastine, vincristine and amsacrine at 500 microM significantly (P < 0.05) inhibited DMXAA glucuronidation (Ki = 319, 350 and 230 microM, respectively), but not 6-methylhydroxylation in human liver microsomes. Daunorubicin and N-[2-(dimethylamino)-ethyl]acridine-4-carboxamide (DACA) at 100 and 500 microM showed significant (P < 0.05) inhibition of DMXAA 6-methylhydroxylation (Ki = 131 and 0.59 microM, respectively), but not glucuronidation. Other drugs such as 5-fluoroucacil, paclitaxel, tirapazamine and methotrexate exhibited little or negligible inhibition of the metabolism of DMXAA. Pre-incubation of microsomes with the anticancer drugs (100 and 500 microM) did not enhance their inhibitory effects on DMXAA metabolism. Prediction of DMXAA-drug interactions in vivo based on these in vitro data indicated that all the anticancer drugs investigated except DACA appear unlikely to alter the pharmacokinetics of DMXAA, whereas DACA may increase the plasma AUC of DMXAA by 6%. CONCLUSIONS: These results indicate that alteration of the pharmacokinetics of DMXAA appears unlikely when used in combination with other common anticancer drugs. However, this does not rule out the possibility of pharmacokinetic interactions with other drugs used concurrently with this combination of anticancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Microsomes, Liver/drug effects , Xanthenes/metabolism , Xanthones , Antineoplastic Agents/antagonists & inhibitors , Antineoplastic Agents/metabolism , Chromatography, High Pressure Liquid , Drug Interactions , Glucuronides/metabolism , Humans , In Vitro Techniques , Microsomes, Liver/metabolism , Models, Biological , Xanthenes/antagonists & inhibitors , Xanthenes/chemistry , Xanthenes/pharmacokinetics
6.
Xenobiotica ; 31(5): 277-93, 2001 May.
Article in English | MEDLINE | ID: mdl-11491389

ABSTRACT

1. The novel anti-tumour agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) is extensively metabolized by glucuronidation and 6-methylhydroxylation, resulting in DMXAA acyl glucuronide (DMXAA-G) and 6-hydroxymethyl-5-methylxanthenone-4-acetic acid (6-OH-MXAA). 2. The major human urinary metabolite of DMXAA was isolated and purified by a solid-phase extraction (SPE) method. The isolated metabolite was hydrolysed to free DMXAA by strong base, and by beta-glucuronidase. Liquid chromatography-mass spectrometry (LC-MS) and spectral data indicated the presence of a molecular ion [M + 1]+ at m/z 459, which was consistent with the molecular weight of protonated DMXAA-G. 3. The glucuronide was unstable in buffer at physiological pH, plasma and blood with species variability in half-life. Hydrolysis and intramolecular migration were major degradation pathways. 4. In vitro and in vivo formation of DMXAA-protein adducts was observed. The formation of DMXAA-protein adducts in cancer patients receiving DMXAA was significantly correlated with plasma DMXAA-G concentration and maximum plasma DMXAA concentration. 5. At least five metabolites of DMXAA were observed in patient urine, with up to 60% of the total dose excreted as DMXAA-G, 5.5% as 6-OH-MXAA and 4.5% as the glucuronide of 6-OH-MXAA. 6. These data suggest that the major metabolite in patients' urine is DMXAA beta-1-glucuronide, which may undergo hydrolysis, molecular rearrangement and covalent binding to plasma protein. The reactive properties of DMXAA-G may have important implications for the pharmacokinetics, pharmacodynamics and toxicity of DMXAA.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Glucuronides/isolation & purification , Glucuronides/urine , Xanthenes/pharmacokinetics , Xanthones , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anticoagulants/pharmacology , Antineoplastic Agents/urine , Chromatography, High Pressure Liquid , Diazepam/pharmacology , Dose-Response Relationship, Drug , GABA Modulators/pharmacology , Gas Chromatography-Mass Spectrometry , Glucuronidase/metabolism , Humans , Hydrogen-Ion Concentration , Hydrolysis , Male , Mice , Mice, Inbred C57BL , Models, Chemical , Phenylbutazone/pharmacology , Protein Binding , Rabbits , Rats , Rats, Wistar , Spectrometry, Mass, Electrospray Ionization , Time Factors , Warfarin/pharmacology , Xanthenes/urine
7.
Cancer Chemother Pharmacol ; 47(6): 541-4, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11459209

ABSTRACT

PURPOSE: Coadministration of thalidomide, cyproheptadine or diclofenac has been shown to increase the area under the plasma concentration-time curve (AUC) of the novel antitumour agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) in mice. The aim of this study was to further investigate these pharmacokinetic DMXAA-drug interactions in the rat model. METHODS: The effects of coadministration of L-thalidomide, cyproheptadine or diclofenac on the pharmacokinetics of DMXAA were investigated in male Wistar Kyoto rats. The effects of L-thalidomide, cyproheptadine and diclofenac on microsomal metabolism and plasma protein binding of DMXAA were also investigated. RESULTS: No significant alteration in the plasma concentration profile for DMXAA was observed following L-thalidomide pretreatment in rats. In contrast, when combined with diclofenac or cyproheptadine, the plasma AUC of DMXAA was significantly (P<0.05) increased by 48% and 88% and the T1/2 by 36% and 107%, respectively, compared to controls. Both diclofenac and cyproheptadine at 500 microM caused a significant inhibition of DMXAA metabolism in rat liver microsomes. In contrast, L-thalidomide had no or little inhibitory effect on DMXAA metabolism in rat liver microsomes except for causing a 32% decrease in 6methylhydroxylation at 500 microM. None of the drugs had a significant effect on the plasma protein binding of DMXAA in the rat. CONCLUSION: These studies showed that coadministration of L-thalidomide did not alter the plasma DMXAA AUC in rats, in contrast to previous studies in mice, whereas diclofenac and cyproheptadine significantly reduced the plasma clearance of DMXAA in rats in a similar manner to their effect in mice. The cause of the species difference in the pharmacokinetic response to thalidomide by DMXAA is unknown, and indicates difficulties in predicting the outcome of such a combination in patients.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Thalidomide/pharmacology , Xanthenes/pharmacokinetics , Xanthones , Animals , Antineoplastic Agents/blood , Cyproheptadine/pharmacology , Diclofenac/pharmacology , Drug Interactions , Glucuronides/metabolism , Hydroxylation , Male , Mice , Microsomes, Liver/metabolism , Rats , Rats, Wistar , Thalidomide/administration & dosage , Xanthenes/blood
8.
J Chromatogr B Biomed Sci Appl ; 757(2): 343-8, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11417880

ABSTRACT

The reversed-phase HPLC methods were developed to determinate the covalently bound protein adducts of the novel anti-cancer drug 5,6-dimethylxanthenone-4-acetic acid (DMXAA) via its glucuronides after releasing aglycone by alkaline hydrolysis in human plasma and human serum albumin (HSA). An aliquot of 75 microl of the mixture was injected onto a Spherex C18 column (150x4.6 mm; 5 microm) at a flow-rate of 2.5 ml/min. The mobile phase comprising of acetonitrile:10 mM ammonium acetate buffer (24:76, v/v, pH 5.8) was used in an isocratic condition, and DMXAA was detected by fluorescence. The method was validated with respect to recovery, selectivity, linearity, precision, and accuracy. Calibration curves for DMXAA were constructed in the concentration range of 0.5-40 microM in washed blank human plasma or HSA prior to alkaline hydrolysis. The difference between the theoretical and calculated concentration and the relative standard deviation were less than 10% at all quality control (QC) concentrations. The limit of detection for the covalent adduct in human plasma or HSA is 0.20 microM. The methods presented good accuracy, precision and sensitivity for use in the preclinical and clinical studies.


Subject(s)
Antineoplastic Agents/blood , Chromatography, High Pressure Liquid/methods , Xanthenes/blood , Xanthones , Antineoplastic Agents/analysis , Calibration , Reproducibility of Results , Sensitivity and Specificity , Serum Albumin/chemistry , Xanthenes/analysis
9.
J Chromatogr B Biomed Sci Appl ; 757(2): 359-63, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11417882

ABSTRACT

The novel anti-tumour agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) is a highly protein bound drug with narrow therapeutic window. We report a simple HPLC method with fluorimetric detection for the determination of free DMXAA concentration in human plasma. Sample preparation involves the ultrafiltration of plasma by a Centrisart device for 30 min at 2000 g and extraction with acetonitrile: methanol mixture. The method was validated with respect to recovery, selectivity, linearity, precision, and accuracy. Calibration curves for DMXAA were constructed at the concentration range of 0.5-40 microM in blank plasma and phosphate buffer. The difference between the theoretical and calculated concentration and the relative standard deviation were less than 10% at all quality control (QC) concentrations. The HPLC method has been used for the analysis of preclinical studies.


Subject(s)
Antineoplastic Agents/blood , Chromatography, High Pressure Liquid/methods , Ultrafiltration/methods , Xanthenes/blood , Xanthones , Calibration , Humans , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Fluorescence
10.
Cancer Chemother Pharmacol ; 47(4): 319-26, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11345648

ABSTRACT

BACKGROUND: Previous studies have demonstrated that coadministration of L-thalidomide with the novel antitumour agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) results in an increased area under the plasma concentration-time curve (AUC) of DMXAA, suggesting an explanation for the observed increase in the antitumour activity. The aims of this study were to investigate the effects of L-thalidomide on the in vitro metabolism of DMXAA in mouse and human liver microsomes using diclofenac as positive control, to examine the effects of L-thalidomide and diclofenac on the plasma protein binding of DMXAA in vitro, and to investigate whether the in vivo interactions can be predicted from in vitro data, particularly in humans. METHODS: Mouse and human liver microsomes were used to investigate the effects of L-thalidomide and diclofenac on DMXAA metabolism. The resulting in vitro data were extrapolated to predict in vivo changes in DMXAA, which were then compared with the results of in vivo mouse pharmacokinetic interaction studies. The protein binding of DMXAA in mouse and human plasma was determined using ultrafiltration followed by HPLC. RESULTS: Diclofenac at 100 microM caused significant inhibition of glucuronidation (> 70%) and 6-methylhydroxylation (> 54%) of DMXAA in mouse and human liver microsomes. In vivo diclofenac (100 mg/kg i.p.) resulted in a 24% and 31% increase in the plasma DMXAA AUC, and a threefold increase in T1/2 (P < 0.05) in male and female mice, respectively. In contrast, L-thalidomide at 100 microM had no inhibitory effect on DMXAA metabolism in vitro in either species, except for a decrease of about 25% in 6-methylhydroxylation in mice. L-Thalidomide at 500 microM resulted in further significant decreases in 6-methylhydroxylation in mice (30-60%) and human (30%) microsomes. Coadministration of L-thalidomide in male mice resulted in a 23% increase in DMXAA AUC and a twofold increase in T1/2 (P < 0.05). Neither L-thalidomide nor diclofenac at 50 or 500 microM had any significant effect on the in vitro plasma protein binding of DMXAA (500 microM) in mouse or human plasma. Based on our in vitro inhibition studies, we predicted a 20% increase in DMXAA AUC in mice with concomitant diclofenac, but little or no effect (< 5%) with L-thalidomide. CONCLUSION: Both L-thalidomide and diclofenac increased the plasma DMXAA AUC in mice. In the case of diclofenac, this appeared to be due to direct competitive inhibition of DMXAA metabolism, but this mechanism does not appear to be appropriate for L-thalidomide. From the in vitro human inhibition studies, it appears unlikely that concurrent diclofenac will cause an increase in the plasma AUC of DMXAA in patients. However, the effect of L-thalidomide on DMXAA could not be readily predicted from the in vitro data. Our study demonstrated that a predictive model based on direct inhibition of metabolism is appropriate for diclofenac-DMXAA interactions, but is inappropriate for the prediction of L-thalidomide-DMXAA interactions in mice and humans in vivo.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Diclofenac/pharmacology , Diclofenac/pharmacokinetics , Teratogens/pharmacology , Teratogens/pharmacokinetics , Thalidomide/pharmacology , Thalidomide/pharmacokinetics , Xanthenes/pharmacology , Xanthenes/pharmacokinetics , Xanthones , Animals , Blood Proteins/metabolism , Chromatography, High Pressure Liquid , Drug Interactions , Female , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Protein Binding/drug effects
11.
J Pharm Pharmacol ; 53(4): 463-71, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11341362

ABSTRACT

The plasma protein binding and distribution in blood cells of the novel anti-tumour agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) has been investigated in-vitro using filtration and an HPLC method to measure DMXAA. DMXAA (500 microM) was extensively bound in plasma from all species with an unbound fraction (fu) of 4.61+/-1.10 (mouse), 2.59+/-0.32 (rat), 2.02+/-0.48 (rabbit) and 2.07+/-0.23% (human). The binding was concentration dependent with DMXAA concentrations > or = 1,000 microM markedly increasing the fu in the plasma from all species. The estimated number of binding sites in plasma were 2.4+/-0.2 (mouse), 1.7+/-0.2 (rat), 0.8+/-0.1 (rabbit) and 2.1+/- 0.2 (human). The major binding protein in human plasma was albumin, with negligible binding to gamma-globulin and alpha1-acid glycoprotein. There was a significant linear relationship between the bound:free DMXAA concentration ratio (Cb/Cu) and albumin concentration in human serum albumin solution (r = 0.955; P < 0.05) and in healthy human plasma (r = 0.998; P< 0.05), but not in plasma from cancer patients (n = 5), nor across species. In cancer patients (n = 5) DMXAA had a significantly higher (P < 0.05) fu (4.60+/- 0.42%) compared with healthy human plasma (2.07+/-0.23%). In human plasma, the fu of DMXAA (500 microM) was significantly reduced by 500 microM diazepam (P < 0.05), but not by warfarin, phenylbutazone, salicylic acid, ibuprofen or clofibric acid at that concentration. DMXAA significantly reduced the binding of dansylsarcosine (a Site-II binder) to HSA, but significantly increased the binding of dansylamide (a Site-I binder). Within species, the blood:plasma concentration ratio (CBL/CP) of DMXAA was relatively constant (mouse, 0.581+/-0.005; rat, 0.667+/-0.025; rabbit, 0.637+/-0.019; human, 0.673+/-0.103) over the range 50-1000 microM, but increased significantly at DMXAA concentrations > 1000 microM in all species except the rabbit. These results indicate that significant alterations in DMXAA plasma binding and distribution into blood cells occur with increasing concentrations of DMXAA in all species, and also that significant interspecies differences exist. It would be more appropriate to compare plasma unbound concentrations when assessing DMXAA exposure in cancer patients or when extrapolating across species.


Subject(s)
Antineoplastic Agents/metabolism , Xanthenes/metabolism , Xanthones , Animals , Antineoplastic Agents/pharmacokinetics , Blood Cells/chemistry , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Humans , Male , Neoplasms/drug therapy , Protein Binding , Rabbits , Rats , Rats, Wistar , Xanthenes/pharmacokinetics
12.
Drug Metab Dispos ; 28(12): 1449-56, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11095582

ABSTRACT

In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) isoenzyme involved in the 6-methylhydroxylation of 5, 6-dimethylxanthenone-4-acetic acid (DMXAA) by using a human liver library (n = 14). The metabolite 6-hydroxymethyl-5-methylxanthenone-4-acetic acid (6-OH-MXAA) was determined by HPLC with fluorescence detection. The metabolite formed in human liver microsomes and by cDNA-expressed CYP isoform was identified by liquid chromatography mass spectrometry as 6-OH-MXAA. In human liver microsomes (n = 14), 6-methylhydroxylation of DMXAA followed monophasic Michaelis-Menten kinetics, with a mean apparent K(m) of 21 +/- 5 microM and V(max) of 0.043 +/- 0.019 nmol/min/mg. An approximate 10-fold interindividual variation in the intrinsic clearance (V(max)/K(m)) of DMXAA 6-methylhydroxylation in human liver microsomes was observed. The involvement of CYP1A2 in DMXAA metabolism by human livers was demonstrated by the following: 1) the potent inhibition of DMXAA metabolism by furafylline (k(inact) = 0.23 +/- 0.04 min(-1), K'(app) = 15.6 +/- 6.7 microM) and alpha-naphthoflavone (K(i) = 0.036 microM), but not by cimetidine, ketoconazole, tolbutamide, quinidine, chlorzoxazone, diethyldithiocarbamate, troleandomycin, and sulfaphenazole; 2) when incubated with human lymphoblastoid cell microsomes containing cDNA-expressed CYP isoenzymes, DMXAA was metabolized only by CYP1A2, with an apparent K(m) of 6.2 +/- 1.5 microM and V(max) of 0.014 +/- 0.001 nmol/min/mg, but not by CYP2A6, CYP2B6, CYP2C9 (Arg(144)), CYP2C19, CYP2D6 (Val(374)), CYP2E1, and CYP3A4; 3) a significant correlation (r = 0.90; P <.001) between 6-methylhydroxylation of DMXAA and 7-ethoxyresorufin O-deethylation; and 4) a significant correlation (r = 0.75; P <.01) between the CYP1A protein level determined by Western blots and DMXAA 6-methylhydroxylation.


Subject(s)
Antineoplastic Agents/metabolism , Cytochrome P-450 Enzyme System/metabolism , Liver/enzymology , Xanthenes/metabolism , Xanthones , Adult , Aged , Blotting, Western , Chromatography, High Pressure Liquid , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/analysis , DNA, Complementary/biosynthesis , Female , Humans , Hydroxylation , In Vitro Techniques , Isoenzymes/analysis , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Male , Methylation , Microsomes, Liver/enzymology , Middle Aged , Proteins/metabolism , Subcellular Fractions/metabolism
13.
Drug Metab Dispos ; 28(10): 1184-6, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10997937

ABSTRACT

The fluorescent compound 4-methylumbelliferone (4MU) can be used to detect uridine diphosphate glucuronosyl transferase activity by observing the fall in fluorescence as the compound is converted to 4-methylumbelliferone glucuronide. A microplate assay has been developed that has improved sensitivity and is faster and cheaper than the historical extraction method. Activity is detectable with approximately 10% of the protein required in the extraction method. Absence of extraction and cleanup procedures and the ability to observe reaction rate directly are also of great advantage to the researcher. Michaelis-Menten kinetic data from one healthy female human liver is presented. The extraction method yielded a mean V(max) of 19.9 nmol/min/mg of protein and a mean K(m) of 652.5 microM on 1 day [n = 6, coefficients of variation (CV) 15 and 24%, respectively]. For the microplate method on 1 day, the mean V(max) was 36.21 +/- 1.3 nmol/min/mg of protein (CV = 3.7%), significantly (P <.0001) higher than for the extraction method. The mean K(m), 175. 4 +/- 24.2 microM (CV = 14.5%), was significantly lower (P <.0001) than observed in the extraction method. The assay was performed in replicates of six over 6 days; average intra- and interassay coefficients of variation were 9 and 22% for V(max) and 8 and 35% for K(m), respectively, for the microplate method. The microplate method has also detected activity in the placental trophoblast-derived cell lines JEG-3, JAr, and BeWo (5.5, 4.1, and 2. 6 nmol/min/mg of protein, respectively, at 200 microM 4MU concentration), indicating that placental cells may be capable of glucuronidating 4MU.


Subject(s)
Glucuronosyltransferase/metabolism , Microsomes, Liver/enzymology , Placenta/enzymology , Cell Line , Enzyme Stability , Female , Fluorescence , Fluorometry , Freezing , Glucuronic Acid/metabolism , Humans , Kinetics , Methods , Microsomes/enzymology , Middle Aged , Placenta/cytology , Sensitivity and Specificity
14.
Cancer Chemother Pharmacol ; 46(2): 135-41, 2000.
Article in English | MEDLINE | ID: mdl-10972483

ABSTRACT

BACKGROUND: 5,6-Dimethylxanthenone-4-acetic acid (DMXAA), an investigative drug currently in clinical trial, acts on tumour vasculature through the induction of cytokines. Coadministration of thalidomide, a modulator of cytokine production, potentiates the antitumour activity of DMXAA against the murine Colon 38 carcinoma in mice. We wished to determine whether alteration of the pharmacokinetics of DMXAA by thalidomide could provide an explanation for this potentiation. RESULTS: Coadministration of thalidomide to Colon 38 tumour-bearing mice significantly (P < 0.05) increased the elimination half-life (t1/2) of DMXAA in plasma (413 micromol/l), liver (132 micromol/l), and spleen (77 micromol/l), and significantly (P < 0.05) increased DMXAA concentrations in Colon 38 tumour tissue (0.25-4.5 h). L-Thalidomide had a greater effect on DMXAA elimination (P < 0.01) than did D-thalidomide or the racemate. Coadministration of thalidomide increased the area under the concentration-time curve (AUC) of DMXAA by 1.8-fold in plasma, liver and spleen, and by 3.0-fold in tumour. Bile from mice given thalidomide and DMXAA contained substantially lower amounts of the glucuronide metabolite of DMXAA (DMXAA-G) than did bile from mice given DMXAA alone. CONCLUSION: Glucuronidation is a major excretory pathway for DMXAA in the mouse. Thalidomide, probably as the L-form, decreases the rate of elimination of DMXAA from plasma, spleen, liver and tumour by altering the rate of glucuronidation. The reduction in the elimination of DMXAA by thalidomide may lead to a selective increase in exposure of tumour tissue to drug, providing a basis for its potentiation of antitumour activity.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Colonic Neoplasms/metabolism , Thalidomide/pharmacology , Xanthenes/pharmacokinetics , Xanthones , Animals , Antineoplastic Agents/blood , Area Under Curve , Bile/metabolism , Colonic Neoplasms/blood , Female , Half-Life , Mice , Mice, Inbred C57BL , Stereoisomerism , Time Factors , Tissue Distribution/drug effects , Xanthenes/blood
15.
Biochem Pharmacol ; 59(2): 161-5, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10810450

ABSTRACT

N-[(2'-Dimethylamino)ethyl]acridine-4-carboxamide (DACA) is a new anticancer agent currently undergoing clinical trials. The metabolism of DACA to acridone metabolites by aldehyde oxidase (AO) (EC 1.2.3.1) appears to play a major role in its elimination in human patients and rodents. The aim of this study was to compare the ability of human, guinea pig, and rat AO preparations to metabolise DACA, and to determine if either animal model was appropriate for predicting AO-mediated DACA-drug interactions in humans. Both human and rodent liver samples were homogenised in buffer before sequential centrifugation to produce the cytosol fraction. Human supernatant underwent an additional ammonium sulphate precipitation procedure, which produced a 2-fold increase in enzyme activity per milligram of protein. After incubations with DACA (range, 0-200 microM), DACA-9(10H)-acridone formation was determined by HPLC analysis. Michaelis-Menten parameters, Km and Vmax, were determined from the best fit curves by nonlinear regression. Three of the four human liver preparations had similar DACA intrinsic clearance values (Vmax/Km) ranging from 0.27 to 0.35 mL/min/mg protein, whereas both the rat and guinea pig had approximately 7- and 160-fold greater intrinsic clearances, due to lower Km values in rats (4.5 +/- 0.7 microM) and guinea pigs (0.15 +/- 0.1 microM) compared with humans (28.3 +/- 8.3 microM, N = 4). Amsacrine, menadione, and 7-hydroxy-DACA were potent inhibitors of DACA metabolism in all three species, but 10-fold differences in IC50 values were apparent between species. In addition, SKF-525A was a potent inhibitor of the metabolism of DACA in rat cytosol but caused minimal inhibition in the guinea pig or human preparations. These results suggest that neither rat nor guinea pig AO preparations are suitable for predicting AO-mediated DACA-drug interactions in humans.


Subject(s)
Acridines/pharmacology , Aldehyde Oxidoreductases/metabolism , Acridines/metabolism , Acridones , Adult , Aldehyde Oxidase , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Female , Guinea Pigs , Humans , Liver/drug effects , Liver/metabolism , Male , Middle Aged , Rats , Rats, Wistar , Species Specificity
16.
J Chromatogr B Biomed Sci Appl ; 734(1): 129-36, 1999 Oct 29.
Article in English | MEDLINE | ID: mdl-10574198

ABSTRACT

High-performance liquid chromatographic methods have been developed and validated for the glucuronidated and oxidative metabolites of the novel anti-tumour agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA), produced in human liver microsomal incubations. Calibration curves for DMXAA acyl glucuronide (DMXAA-Glu) and 6-hydroxymethyl-5-methylxanthenone-4-acetic acid (6-OH-MXAA) were constructed over the concentration ranges of 0.25 to 20 and 0.5 to 40 microM, respectively. Assay performance was determined by intra- and inter-day accuracy and precision of quality control (QC) samples. The difference between the theoretical and measured concentration, and the coefficient of variation, were less than 15% at low QC concentrations, and less than 10% at medium and high QC concentrations for both analytes. The methods presented good accuracy, precision and sensitivity for use in kinetic studies of the glucuronidated and oxidative metabolites of DMXAA in human liver microsomes.


Subject(s)
Antineoplastic Agents/metabolism , Chromatography, High Pressure Liquid/methods , Microsomes, Liver/metabolism , Xanthenes/metabolism , Xanthones , Humans , Hydrogen-Ion Concentration , Hydroxides/pharmacology , NADP/metabolism , Oxidation-Reduction , Phosphoric Acids/pharmacology , Potassium Compounds/pharmacology , Quality Control , Sensitivity and Specificity , Uridine Diphosphate Glucuronic Acid/metabolism
17.
Cancer Chemother Pharmacol ; 44(1): 45-50, 1999.
Article in English | MEDLINE | ID: mdl-10367748

ABSTRACT

DACA [N-[2-(dimethylamino)ethyl]acridine-4-carboxamide] is an acridine derivative with high activity against solid tumours in mice and a dual mode of cytotoxic action involving topoisomerases I and II. The plasma pharmacokinetics of DACA were studied in 28 patients with solid tumours in a phase I trial. A single dose was given every 3 weeks, being escalated from a starting dose of 18 mg/m2 (as the dihydrochloride trihydrate salt) to a maximal dose, limited by severe pain in the infusion arm, of 1000 mg/m2. Drug was given by constant intravenous infusion with a target delivery period of 3 h. Blood samples were taken from the contralateral arm before, during and for up to 72 h after the infusion. DACA was separated from plasma by solid-phase extraction and was analysed by reversed-phase high-performance liquid chromatography (C18 column) using fluorescence detection. A two-compartment pharmacokinetic model provided the best fit for the concentration-time profiles obtained for most patients showing clearance of 1.00+/-0.36 l h(-1) kg(-1), a volume of distribution of the central compartment of 0.72+/-0.55 l/kg, an initial half-life of 0.28+/-0.19 h and a terminal half-life of 2.04+/-0.94 h. All pharmacokinetic parameters were independent of dose, indicating first-order kinetics. As DACA binds strongly to alpha1-acid glycoprotein, plasma concentrations of this protein were determined and used to estimate free-drug fractions in plasma. Estimated values for the free fraction varied from 0.9% to 3.3% and were lower than those determined by equilibrium dialysis for mice and rats (15% and 16%, respectively). At the maximum tolerated dose (MTD) of 750 mg/m2, the area under the drug concentration-time curve (AUC) was 46.2+/-4.4 microM h, exceeding that obtained in mice treated at the MTD (23.4 microM h). On the other hand, the corresponding free-drug AUC was 0.92+/-0.03 microM h, much lower than the corresponding value (3.5 microM h) determined for mice. These results suggest that free-drug rather than total drug concentrations are more appropriate for interspecies dose comparisons when significant differences exist in the free plasma fraction.


Subject(s)
Acridines/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Acridines/administration & dosage , Adult , Aged , Antineoplastic Agents/administration & dosage , Area Under Curve , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Infusions, Intravenous , Male , Middle Aged , Neoplasms/drug therapy , Neoplasms/metabolism
18.
Cancer Chemother Pharmacol ; 44(1): 51-8, 1999.
Article in English | MEDLINE | ID: mdl-10367749

ABSTRACT

N-[2-(Dimethylamino)ethyl]acridine-4-carboxamide (DACA) is an experimental antitumour agent that has just completed phase I clinical trials in New Zealand and the United Kingdom. Urine (0-72 h) was analysed from 20 patients receiving DACA infused over 3 h (dose range 60-1000 mg/m2, the latter being the highest dose achieved in the trial). Aliquots were analysed for DACA and its metabolites by high-performance liquid chromatography (HPLC). Over 72 h, 44+/-5% (range 20-60%) of the dose was recovered in the urine, with 0.8+/-0.3% (range 0-3.1%) occurring as DACA. The major urinary metabolite was DACA-N-oxide-9(10H)acridone, accounting for 34+/-3% of the dose. Minor metabolites were identified as N-monomethyl-DACA-9(10H)acridone (2.0+/-0.5%), DACA-9(10H)acridone (3.3+/-0.5%), N-monomethyl-DACA (0.2+/-0.1%) and DACA-N-oxide (0.5+/-0.1%). No ring-hydroxylated metabolite was detected. The urinary excretion of metabolites was greatest over 0-6 h in most patients. The composition of urinary metabolites was also independent of the delivered dose. Plasma was sampled at intervals throughout the infusion and at time points up to 48 h post-administration. The major plasma metabolites observed were DACA-9(10H)acridone and DACA-N-oxide-9(10H)acridone. These results indicate that, based on urinary excreted metabolites, the major biotransformation reactions for DACA in humans involve N-oxidation of the tertiary amine side chain and acridone formation, both of which appear to be detoxication reactions.


Subject(s)
Acridines/metabolism , Antineoplastic Agents/metabolism , Acridines/administration & dosage , Acridines/pharmacokinetics , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Biotransformation , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Neoplasms/drug therapy
19.
Cancer Chemother Pharmacol ; 43(4): 323-30, 1999.
Article in English | MEDLINE | ID: mdl-10071984

ABSTRACT

5,6-Dimethylxanthenone-4-acetic acid (DMXAA), an experimental antitumour agent currently undergoing phase I clinical trial, has a maximum tolerated dose (MTD) in male BDF1 mice of 99 micromol/kg. We have found the male Sprague-Dawley rat and the New Zealand White rabbit to have greater tolerance to DMXAA, with MTDs being 990 and 330 micromol/kg, respectively. To investigate the causes of this difference, we measured plasma and urine DMXAA concentrations by high-performance liquid chromatography (HPLC) after single i.v. bolus injections of 99 and 990 micromol/kg in the rat and following a bolus dose of 99 micromol/kg and a 10-min infusion of 330 micromol/kg in the rabbit. Following administration of DMXAA at the MTD in the mouse, rat and rabbit the maximal concentrations were 600, 2,200 and 1,708 microM, respectively, whereas areas under the concentration-time curves were 2,400, 19,000 and 2,400 microMh, respectively, for unchanged DMXAA. Data obtained for mice and rabbits were satisfactorily fitted to a two-compartment model with Michaelis-Menten kinetics. DMXAA was highly bound to plasma proteins, with the highest degree of binding being found in the rabbit. A small proportion of the total dose (7.8%, 0.6% and 12.4%, respectively) was excreted unchanged in urine over 24 h. This proportion increased (to 11.6%, 3.5% and 72.4%, respectively) following alkaline hydrolysis, suggesting the presence of glucuronide metabolites. Examination of rat and mouse urine by HPLC revealed the presence of two metabolites, which were characterized by mass spectrometry and nuclear magnetic resonance to be the acyl glucuronide of DMXAA and 6-(hydroxymethyl)-5-methylxanthenone-4-acetic acid. Thus, both mice and rats metabolise DMXAA by similar pathways. The results demonstrate considerable interspecies variations in tolerance to DMXAA that cannot be explained by differences in pharmacokinetics.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Xanthenes/pharmacokinetics , Xanthones , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/urine , Blood Proteins/metabolism , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Infusions, Intravenous , Injections, Intravenous , Male , Metabolic Clearance Rate , Mice , Protein Binding , Rabbits , Rats , Rats, Sprague-Dawley , Species Specificity , Time Factors , Xanthenes/blood , Xanthenes/urine
20.
Clin Exp Pharmacol Physiol ; 22(11): 851-4, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8593743

ABSTRACT

1. The rationale for extrapolation or 'scaling' across animal species is based on their underlying anatomical, physiological and biochemical similarities. 2. Research carried out in the 19th and early 20th century resulted in Benedict's famous 'mouse-to-elephant' graph which showed that the log of the basal metabolic rate plotted against the log of bodyweight (W) produced a straight line with a slope of 0.76. Since then it has become apparent that a number of other physiological variables (Y) exhibit a similar relationship which can be represented by the general allometric equation, Y = alpha W beta; where beta is the slope of the log-log plot and alpha is the intercept on the y axis. 3. The major pharmacokinetic parameters such as clearance and volume of distribution of many drugs are also related to W in a similar manner. 4. This empirical approach does not require a strong mathematical background and offers a relatively simple method of predicting the kinetics of anti-cancer drugs in patients from pre-clinical animal data. 5. The occurrence of major qualitative and quantitative differences in the metabolism of drugs between species is probably the single greatest complicating factor in the use of animals as predictors of drug toxicity and kinetics in patients. 6. Despite this, the allometric approach is useful for allowing the estimation of a more appropriate starting dose for some drugs in a Phase I trial, which might result in potential savings in escalation steps and maximize the chance that the dose which an individual receives has the potential for therapeutic value.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Animals , Antineoplastic Agents/administration & dosage , Clinical Trials, Phase I as Topic , Dose-Response Relationship, Drug , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...