Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Article in English | MEDLINE | ID: mdl-33246879

ABSTRACT

The aim of this study was to develop and to validate a UPLC-MS/MS method for the quantification of morphine, morphine-3-glucuronide, and morphine-6-glucuronide in mouse plasma and tissue homogenates to support preclinical pharmacokinetic studies. The sample preparation consisted of protein precipitation with cold (2-8 °C) methanol:acetonitrile (1:1, v/v), evaporation of the supernatant to dryness, and reconstitution of the dry-extracts in 4 mM ammonium formate pH 3.5. Separation was achieved on a Waters UPLC HSS T3 column (150 × 2.1 mm, 1.8 µm) maintained at 50 °C and using gradient elution with a total runtime of 6.7 min. Mobile phase A consisted of 4 mM ammonium formate pH 3.5 and mobile phase B of 0.1% formic acid in methanol:acetonitrile (1:1, v/v). Detection was carried out by tandem mass spectrometry with electrospray ionization in the positive ion mode. The method was validated within a linear range of 1-2,000 ng/mL, 10-20,000 ng/mL, and 0.5-200 ng/mL for morphine, morphine-3-glucuronide, and morphine-6-glucuronide, respectively. In human plasma, the intra- and inter-run precision of all analytes, including the lower limit of quantification levels, were ≤ 15.8%, and the accuracies were between 88.1 and 111.9%. It has been shown that calibration standards prepared in control human plasma can be used for the quantification of the analytes in mouse plasma and tissue homogenates. The applicability of the method was successfully demonstrated in a preclinical pharmacokinetic study in mice.


Subject(s)
Chromatography, High Pressure Liquid/methods , Morphine Derivatives/blood , Tandem Mass Spectrometry/methods , Animals , Linear Models , Mice , Morphine Derivatives/analysis , Morphine Derivatives/chemistry , Morphine Derivatives/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity
2.
Article in English | MEDLINE | ID: mdl-31352204

ABSTRACT

The first bioanalytical assay for tivozanib in human and mouse plasma, mouse tissue homogenates and culture medium was developed and validated over a linear dynamic range from 0.5 to 5000 ng/mL. The extended concentration range will cover the quantification of tivozanib in the majority of study samples, reducing the need for reanalysis which is often not possible due to limited amount of sample in preclinical studies. A simple and fast pretreatment method was used consisting of protein precipitation with acetonitrile followed by dilution of the supernatant. The final extract was injected onto an Ultra-Performance Liquid Chromatography (UPLC) BEH C18 column with gradient elution of formic acid in water and formic acid in acetonitrile mobile phase. Chromatographic separation was followed by detection with a triple-quadrupole mass spectrometer operating in the positive ion-mode. By simultaneously monitoring the sensitive conventional [M + H]+ isotopologue- product transition for quantification of low concentrations and a less abundant [M + H]++1 isotopologue- product transition to reduce the sensitivity for quantification of high concentrations, we were able to extend the overall linear dynamic range up to 0.5-5000 ng/mL. A full validation was performed in human plasma and a partial validation was executed for the other matrices. All results were within the acceptance criteria of the European Medicines Agency (EMA) guidelines and the US Food and Drug Administration (FDA) guidance, except for the carry-over. This was solved by the analysis of extra matrix blanks and by grouping study samples containing a high tivozanib concentration in the sample sequence. In this way carry-over did not impact the data integrity. We demonstrated that by measuring two multiple reaction monitoring (MRM) transitions for tivozanib, the linear dynamic range could be extended from two to four decades. The assay was successfully applied in pharmacokinetic studies in mice and a transport assay.


Subject(s)
Chromatography, High Pressure Liquid/methods , Culture Media/chemistry , Phenylurea Compounds/chemistry , Quinolines/chemistry , Tandem Mass Spectrometry/methods , Animals , Humans , Kidney/chemistry , Liver/chemistry , Lung/chemistry , Mice , Phenylurea Compounds/blood , Plasma/chemistry , Quinolines/blood , Spleen/chemistry
3.
J Pharm Biomed Anal ; 173: 169-175, 2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31146172

ABSTRACT

Galunisertib is an anti-cancer drug currently evaluated in phase I and II clinical trials. This study describes the development and validation of a bioanalytical assay to quantify galunisertib in human plasma using HPLC-MS/MS. Stable isotope labelled galunisertib was added as internal standard and the analyte and internal standard were extracted from the matrix by protein precipitation using acetonitrile-methanol (50:50, v/v). Final extracts were injected onto a C18 column, gradient elution was applied for chromatographic separation and detection was performed using a triple quadrupole mass spectrometer operating in the positive ion mode. The assay was linear over the range 0.05-10 ng/mL, with acceptable accuracy (bias ranging from -6.1 to 3.1%) and precision (below 5.7% C.V.) values. The applicability of the assay was demonstrated in a pharmacokinetic experiment in mice.


Subject(s)
Protein Kinase Inhibitors/blood , Pyrazoles/blood , Quinolines/blood , Tandem Mass Spectrometry/methods , Administration, Oral , Animals , Chromatography, High Pressure Liquid/methods , Drug Screening Assays, Antitumor , Humans , Male , Mice , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/administration & dosage , Pyrazoles/pharmacokinetics , Quinolines/administration & dosage , Quinolines/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Transforming Growth Factor beta/antagonists & inhibitors
4.
Arch Toxicol ; 93(3): 775-790, 2019 03.
Article in English | MEDLINE | ID: mdl-30701287

ABSTRACT

2-Amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) is an abundant dietary carcinogen, formed during high-temperature cooking of meat. In this study, we investigated whether clinically relevant ATP-binding cassette (ABC) efflux transporters can modulate PhIP-induced colorectal carcinogenesis in vivo using wild-type (WT), Bcrp1-/-; Mrp2-/-; Mrp3-/- and Bcrp1-/-; Mdr1a/b-/-; Mrp2-/- mice. We used a physiological mouse model of colorectal cancer; a combination of a single high-dose oral PhIP administration (200 mg/kg), followed by administering a colonic inflammatory agent, dextran sodium sulfate (DSS), in drinking water for 7 days. Pilot experiments showed that both knockout strains were more sensitive to DSS-induced colitis compared to WT mice. Lack of these transporters in mice also led to clearly altered disposition of activated PhIP metabolites after a high-dose oral PhIP administration. The results suggest that Mdr1a/1b, Bcrp1 and Mrp2 contributed to biliary excretion and Mrp3 to sinusoidal secretion of the pre-carcinogenic metabolite N2-OH-PhIP. The levels of a genotoxicity marker, PhIP-5-sulphate, were at least 4- and 17-fold reduced in the intestinal tissue and intestinal content of both knockout strains compared to WT mice. In line with these findings, the level of colon carcinogenesis was reduced by two- to four-fold in both knockout strains compared to WT mice when PhIP and DSS treatments were combined. Thus, perhaps counterintuitively, reduced activity of these ABC transporters may in part protect from PhIP-induced colon carcinogenesis. Collectively, these data suggest that ABC transporters are important in protecting the body from inflammatory agents such as DSS, in the disposition of carcinogenic metabolites, and in determining the sensitivity to dietary PhIP-induced carcinogenesis.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Dextran Sulfate/toxicity , Imidazoles/toxicity , Animals , Carcinogenesis , Carcinogens , Colitis/chemically induced , Colon/drug effects , Colonic Neoplasms/chemically induced , Mice
5.
Mol Psychiatry ; 20(7): 889-900, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25092247

ABSTRACT

Interactions between genetic and environmental factors, like exposure to stress, have an important role in the pathogenesis of mood-related psychiatric disorders, such as major depressive disorder. The polyspecific organic cation transporters (OCTs) were shown previously to be sensitive to the stress hormone corticosterone in vitro, suggesting that these transporters might have a physiologic role in the response to stress. Here, we report that OCT2 is expressed in several stress-related circuits in the brain and along the hypothalamic-pituitary-adrenocortical (HPA) axis. Genetic deletion of OCT2 in mice enhanced hormonal response to acute stress and impaired HPA function without altering adrenal sensitivity to adrenocorticotropic hormone (ACTH). As a consequence, OCT2(-/-) mice were potently more sensitive to the action of unpredictable chronic mild stress (UCMS) on depression-related behaviors involving self-care, spatial memory, social interaction and stress-sensitive spontaneous behavior. The functional state of the glycogen synthase kinase-3ß (GSK3ß) signaling pathway, highly responsive to acute stress, was altered in the hippocampus of OCT2(-/-) mice. In vivo pharmacology and western blot experiments argue for increased serotonin tonus as a main mechanism for impaired GSK3ß signaling in OCT2(-/-) mice brain during acute response to stress. Our findings identify OCT2 as an important determinant of the response to stress in the brain, suggesting that in humans OCT2 mutations or blockade by certain therapeutic drugs could interfere with HPA axis function and enhance vulnerability to repeated adverse events leading to stress-related disorders.


Subject(s)
Brain/physiopathology , Glycogen Synthase Kinase 3/metabolism , Organic Cation Transport Proteins/metabolism , Stress, Psychological/metabolism , Action Potentials/drug effects , Acute Disease , Animals , Brain/drug effects , Chronic Disease , Corticosterone/administration & dosage , Corticosterone/metabolism , Depression/metabolism , Glycogen Synthase Kinase 3 beta , Hormones/administration & dosage , Hormones/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Norepinephrine/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transporter 2 , Proto-Oncogene Proteins c-akt/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Resilience, Psychological , Serotonin/metabolism , Uncertainty
6.
Bioanalysis ; 6(7): 993-1010, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24806907

ABSTRACT

Since the isolation of paclitaxel and its approval for the treatment of breast cancer, various taxanes and taxane formulations have been developed. To date, almost 100 bioanalytical assays have been published with the method development and optimization often extensively discussed by the authors. This Review presents an overview of assays published between January 1970 and September 2013 that described method development and validation of assays used to quantify taxanes in biological matrices such as plasma, urine, feces and tissue samples. For liquid chromatography assays, sample pretreatment, chromatographic separation and assay performance are compared. Since this Review discusses the limitations of previously developed liquid chromatography assays and gives recommendations for future assay development, it can be used as a reference for future development of liquid chromatography assays for the quantification of taxanes in various biological matrices to support preclinical and clinical studies.


Subject(s)
Chromatography, High Pressure Liquid/methods , Taxoids/analysis , Chromatography, Micellar Electrokinetic Capillary , Limit of Detection , Solid Phase Extraction , Spectrophotometry, Ultraviolet , Tandem Mass Spectrometry
7.
Br J Cancer ; 110(11): 2669-76, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24781280

ABSTRACT

BACKGROUND: The intestinal uptake of the taxanes paclitaxel and docetaxel is seriously hampered by drug efflux through P-glycoprotein (P-gp) and drug metabolism via cytochrome P450 (CYP) 3A. The resulting low oral bioavailability can be boosted by co-administration of P-gp or CYP3A4 inhibitors. METHODS: Paclitaxel or docetaxel (10 mg/kg) was administered to CYP3A4-humanised mice after administration of the P-gp inhibitor elacridar (25 mg kg(-1)) and the CYP3A inhibitor ritonavir (12.5 mg kg(-1)). Plasma and brain concentrations of the taxanes were measured. RESULTS: Oral co-administration of the taxanes with elacridar increased plasma concentrations of paclitaxel (10.7-fold, P<0.001) and docetaxel (four-fold, P<0.001). Co-administration with ritonavir resulted in 2.5-fold (paclitaxel, P<0.001) and 7.3-fold (docetaxel, P<0.001) increases in plasma concentrations. Co-administration with both inhibitors simultaneously resulted in further increased plasma concentrations of paclitaxel (31.9-fold, P<0.001) and docetaxel (37.4-fold, P<0.001). Although boosting of orally applied taxanes with elacridar and ritonavir potentially increases brain accumulation of taxanes, we found that only brain concentrations, but not brain-to-plasma ratios, were increased after co-administration with both inhibitors. CONCLUSIONS: The oral availability of taxanes can be enhanced by co-administration with oral elacridar and ritonavir, without increasing the brain penetration of the taxanes.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Brain/metabolism , Acridines/administration & dosage , Administration, Oral , Animals , Area Under Curve , Docetaxel , Humans , Male , Mice , Mice, Knockout , Paclitaxel/administration & dosage , Ritonavir/administration & dosage , Taxoids/administration & dosage , Tetrahydroisoquinolines/administration & dosage , Tissue Distribution
8.
Rapid Commun Mass Spectrom ; 27(17): 1925-34, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23939959

ABSTRACT

RATIONALE: During drug development accurate quantification of metabolites in biological samples using mass spectrometry is often hampered by the lack of metabolites of chemically pure quality. However, quantification of metabolites can be useful for assessment and interpretation of (pre)clinical data. We now describe an approach to quantify docetaxel metabolites in human plasma by liquid chromatography/tandem mass spectrometry (LC/MS/MS) using docetaxel calibration standards. METHODS: Metabolites (M1/M3, M2 and M4) were generated using microsomal incubations. Retention times of docetaxel and its metabolites were assessed using an LC/UV assay and peak identification was performed by LC/MS(n). Samples containing isolated metabolites from human faeces were quantified by LC/UV and used as references for spiking human plasma samples. LC/MS/MS was applied to sensitively quantify docetaxel and its metabolites in human plasma using docetaxel calibration standards in a range of 0.25-500 ng/mL. RESULTS: Because ionisation of docetaxel and its metabolites differed, correction factors were established to quantify the metabolites using docetaxel calibration samples. During method validation, accuracy and precision of the metabolites were within ±7.7% and ≤17.6%, respectively, and within ±14.3% and ≤10.1%, respectively, for docetaxel. Metabolites were found to be unstable in human plasma at ambient temperature. After storage up to 1 year at -20 °C, recovered metabolite concentrations were within ±25%. CONCLUSIONS: Development and validation of an LC/MS/MS assay for the quantification of docetaxel and its metabolites M1/M3, M2 and M4 using docetaxel calibration standards is described. The same approach may be used for quantification of metabolites of other drugs by LC/MS/MS when chemically pure reference substances are unavailable.


Subject(s)
Antineoplastic Agents, Phytogenic/blood , Antineoplastic Agents, Phytogenic/metabolism , Chromatography, High Pressure Liquid/methods , Tandem Mass Spectrometry/methods , Taxoids/blood , Taxoids/metabolism , Docetaxel , Humans
9.
Pharmacol Res ; 76: 9-16, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23827160

ABSTRACT

CYT387 is an orally bioavailable, small molecule inhibitor of Janus family of tyrosine kinases (JAK) 1 and 2. It is currently undergoing Phase I/II clinical trials for the treatment of myelofibrosis and myeloproliferative neoplasms. We aimed to establish whether the multidrug efflux transporters P-glycoprotein (P-gp; MDR1; ABCB1) and breast cancer resistance protein (BCRP;ABCG2) restrict oral availability and brain penetration of CYT387. In vitro, CYT387 was efficiently transported by both human MDR1 and BCRP, and very efficiently by mouse Bcrp1 and its transport could be inhibited by specific MDR1 inhibitor, zosuquidar and/or specific BCRP inhibitor, Ko143. CYT387 (10 mg/kg) was orally administered to wild-type (WT), Bcrp1(-/-), Mdr1a/1b(-/-) and Bcrp1;Mdr1a/1b(-/-) mice and plasma and brain concentrations were analyzed. Over 8h, systemic exposure of CYT387 was similar between all the strains, indicating that these transporters do not substantially limit oral availability of CYT387. Despite the similar systemic exposure, brain accumulation of CYT387 was increased 10.5- and 56-fold in the Bcrp1;Mdr1a/1b(-/-) mice compared to the WT strain at 2 and 8h after CYT387 administration, respectively. In single Bcrp1(-/-) mice, brain accumulation of CYT387 was more substantially increased than in Mdr1a/1b(-/-) mice, suggesting that CYT387 is a slightly better substrate of Bcrp1 than of Mdr1a at the blood-brain barrier. These results indicate a marked and additive role of Bcrp1 and Mdr1a/1b in restricting brain penetration of CYT387, potentially limiting efficacy of this compound against brain (micro) metastases positioned behind a functional blood-brain barrier.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP-Binding Cassette Transporters/metabolism , Benzamides/pharmacokinetics , Janus Kinases/antagonists & inhibitors , Neoplasm Proteins/metabolism , Pyrimidines/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Animals , Benzamides/blood , Brain/metabolism , Cell Line , Female , Gene Deletion , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Mice , Pyrimidines/blood , ATP-Binding Cassette Sub-Family B Member 4
10.
Article in English | MEDLINE | ID: mdl-23542607

ABSTRACT

We present a sensitive validated LC-MS/MS assay for the simultaneous determination of cabazitaxel and docetaxel in human plasma, with calibration ranges of 1.0-150 ng/mL for cabazitaxel and 0.1-15 ng/mL for docetaxel. Sample pretreatment consisted of liquid-liquid extraction with tert-butyl methyl ether. Chromatographic separation was achieved on a Zorbax Extend C18 column using a gradient mixture of 10mM ammonium hydroxide and methanol. Mass detection was carried out by turbo ion spray ionization in positive ion multiple reaction monitoring mode. All inter-day accuracies and precisions were within ±15% of the nominal value and within ±20% at the lower limit of quantitation. Demethylations of cabazitaxel yielding the metabolites RPR112698 and RPR123142 were monitored semi-quantitatively and quantified as ng docetaxel equivalents. Plasma samples of a prostate cancer patient treated with cabazitaxel were analyzed to demonstrate the usefulness of the presented assay for clinical drug monitoring. In conclusion, this method can be applied to support clinical pharmacokinetic studies with the novel anticancer drug cabazitaxel.


Subject(s)
Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Taxoids/blood , Docetaxel , Drug Stability , Humans , Least-Squares Analysis , Reproducibility of Results , Sensitivity and Specificity , Taxoids/chemistry , Taxoids/metabolism
12.
J Clin Pharmacol ; 52(3): 370-80, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21505085

ABSTRACT

Intravenously administered docetaxel is approved for the treatment of various types of cancer. An oral regimen, in combination with ritonavir, is being evaluated in clinical trials. The pharmacokinetics of docetaxel are determined by the activity of the metabolizing enzyme cytochrome P450 3A (CYP3A) and the drug efflux transporter P-glycoprotein (P-gp). The effects of these proteins on the pharmacokinetics of docetaxel were investigated in different mouse models that lack 1 or both detoxifying systems. Docetaxel was given to these mice orally or intravenously with or without a strong CYP3A inhibitor, ritonavir. The data of these 2 preclinical studies were pooled and analyzed using nonlinear mixed-effects modeling. The results of the preclinical studies could be integrated successfully, with only a small difference in residual error (33% and 26%, respectively). Subsequently, the model was used to predict human exposure using allometric scaling and this was compared with clinical trial data. This model led to adequate predictions of docetaxel exposure in humans.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Taxoids/administration & dosage , Taxoids/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Docetaxel , Drug Evaluation, Preclinical , Female , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Models, Biological , Ritonavir/pharmacology
13.
J Chromatogr B Analyt Technol Biomed Life Sci ; 879(19): 1677-85, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21543272

ABSTRACT

A sensitive bioanalytical assay for the quantitative determination of tamoxifen and five of its phase I metabolites (N-desmethyltamoxifen, N-desmethyl-4-hydroxytamoxifen, N-desmethyl-4'-hydroxytamoxifen, 4-hydroxytamoxifen and 4'-hydroxytamoxifen) in serum is described. The method has been fully validated at ranges covering steady-state serum concentrations in patients receiving therapeutic dosages of tamoxifen. The bioanalytical assay is based on reversed phase liquid chromatography coupled with tandem mass spectrometry in the positive ion mode using multiple reaction monitoring for drug (-metabolite) quantification. The sample pretreatment consists of protein precipitation with acetonitrile using only 50 µL of serum. In the past, numerous assays have been developed by other groups for the quantification of tamoxifen and its phase I metabolites. However, the number of metabolites included in these studies is very limited and only very few of these assays have been fully validated. A liquid chromatography tandem mass spectrometry assay for the quantification of tamoxifen and four phase I metabolites in human serum that was previously developed by our group is now explicitly improved and described herein. Time of analysis has been reduced by 50% and sensitivity was increased by a reduction of the lower limit of quantification from 1.0 to 0.2 ng/mL for 4-hydroxytamoxifen and 4'-hydroxytamoxifen. Additionally, two phase I metabolites that have never been quantified in human serum hitherto, namely 4'-hydroxytamoxifen and N-desmethyl-4'-hydroxytamoxifen, were included in this assay. Validation results demonstrate an accurate and precise quantification of tamoxifen, N-desmethyltamoxifen, N-desmethyl-4-hydroxytamoxifen, N-desmethyl-4'-hydroxytamoxifen, 4-hydroxytamoxifen and 4'-hydroxytamoxifen in human serum. The applicability of the assay was demonstrated and it is now successfully used to support clinical studies in which patient-specific dose optimization is performed based on serum concentrations of tamoxifen metabolites.


Subject(s)
Chromatography, Reverse-Phase/methods , Tamoxifen/analogs & derivatives , Tamoxifen/blood , Tandem Mass Spectrometry/methods , Drug Stability , Humans , Least-Squares Analysis , Reproducibility of Results , Sensitivity and Specificity , Tamoxifen/chemistry , Tamoxifen/isolation & purification
14.
J Pharm Biomed Anal ; 55(3): 518-26, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21392921

ABSTRACT

A comprehensive overview is presented of currently known phase I metabolites of tamoxifen consisting of their systematic name and molecular structure. Reference standards are utilized to elucidate the MS(n) fragmentation patterns of these metabolites using a linear ion trap mass spectrometer. UV-absorption spectra are recorded and absorption maxima are defined. Serum extracts from ten breast cancer patients receiving 40mg tamoxifen once daily were qualitatively analyzed for tamoxifen phase I metabolites using a liquid chromatography-tandem mass spectrometry set-up. In total, 19 metabolites have been identified in these serum samples. Additionally a synthetic method for the preparation of the putative metabolite 3',4'-dihydroxytamoxifen is described.


Subject(s)
Antineoplastic Agents, Hormonal/metabolism , Chromatography, High Pressure Liquid/methods , Quinones/metabolism , Tamoxifen/analogs & derivatives , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/blood , Breast Neoplasms/blood , Breast Neoplasms/drug therapy , Female , Humans , Metabolic Detoxication, Phase I , Molecular Structure , Quinones/administration & dosage , Quinones/blood , Tamoxifen/administration & dosage , Tamoxifen/blood , Tamoxifen/metabolism , Tandem Mass Spectrometry
15.
J Chromatogr B Analyt Technol Biomed Life Sci ; 878(31): 3199-216, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21071289

ABSTRACT

The heterocyclic aromatic amine, 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), has been shown to be carcinogenic in rodents, mice and rats. Following phase I N-hydroxylation and phase II esterification PhIP exerts its carcinogenic effect by binding to DNA purines. Quantitative and qualitative analysis of its bioactivated metabolites as well as it detoxification products is important in studying its biological effects and inter- and intra-individual exposures. A review is presented with an extensive coverage of publications specifically reporting on the analysis of PhIP and its phase I and II metabolites in biological matrices, foodstuff and beverages. Analytical techniques such as liquid and gas chromatography coupled with various detection techniques (mass spectrometry, ultraviolet or fluorescence detection) were mostly applied. We conclude that since the initial identification of PhIP in 1986 a large set of assays has been developed for the analysis of PhIP and its phase I and phase II metabolites in a wide range of matrices, these included food products and biological samples such as plasma, urine and faeces. In addition, it was shown that numerous metabolites were recovered and identified. Thus, we conclude that liquid chromatography coupled to mass spectrometry is clearly the method of choice for sensitive qualitative as well as quantitative analysis with high selectivity and reaching lower quantification levels in the sub pg/mL range. The main aim of this review is that it can be used by other researchers as a resource for method development and optimization of analytical methods of PhIP and its carcinogenic or detoxification products.


Subject(s)
Food Analysis/methods , Imidazoles/analysis , Imidazoles/metabolism , Animals , Beverages/analysis , Body Fluids/chemistry , Carcinogens/analysis , Carcinogens/chemistry , Carcinogens/metabolism , Chromatography, Gas , Humans , Imidazoles/chemistry , Mass Spectrometry
16.
Anal Chim Acta ; 683(1): 21-37, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-21094378

ABSTRACT

The selective estrogen receptor modulator tamoxifen is used in the treatment of early and advanced breast cancer and in selected cases for breast cancer prevention in high-risk subjects. The cytochrome P450 enzyme system and flavin-containing monooxygenase are responsible for the extensive metabolism of tamoxifen into several phase I metabolites that vary in toxicity and potencies towards estrogen receptor (ER) alpha and ER beta. An extensive overview of publications on the determination of tamoxifen and its phase I metabolites in biological samples is presented. In these publications techniques were used such as capillary electrophoresis, liquid, gas and thin layer chromatography coupled with various detection techniques (mass spectrometry, ultraviolet or fluorescence detection, liquid scintillation counting and nuclear magnetic resonance spectroscopy). A trend is seen towards the use of liquid chromatography coupled to mass spectrometry (LC-MS). State-of-the-art LC-MS equipment allowed for identification of unknown metabolites and quantification of known metabolites reaching lower limit of quantification levels in the sub pg mL(-1) range. Although tamoxifen is also metabolized into phase II metabolites, the number of publications reporting on phase II metabolism of tamoxifen is scarce. Therefore the focus of this review is on phase I metabolites of tamoxifen. We conclude that in the past decades tamoxifen metabolism has been studied extensively and numerous metabolites have been identified. Assays have been developed for both the identification and quantification of tamoxifen and its metabolites in an array of biological samples. This review can be used as a resource for method transfer and development of analytical methods used to support pharmacokinetic and pharmacodynamic studies of tamoxifen and its phase I metabolites.


Subject(s)
Breast Neoplasms/blood , Chemistry Techniques, Analytical/methods , Estrogen Receptor alpha/blood , Estrogen Receptor beta/blood , Metabolic Detoxication, Phase I , Selective Estrogen Receptor Modulators/blood , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/blood , Tamoxifen/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/prevention & control , Estrogen Receptor alpha/drug effects , Estrogen Receptor beta/drug effects , Humans , Metabolic Detoxication, Phase II , Reproducibility of Results , Risk Factors , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/therapeutic use , Sensitivity and Specificity , Tamoxifen/chemistry , Tamoxifen/therapeutic use
17.
J Chromatogr B Analyt Technol Biomed Life Sci ; 878(25): 2353-62, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20708442

ABSTRACT

The development and validation of a bioanalytical assay is described for the simultaneous analysis of 2-amino-1-methyl-6-phenylimidazo[4-5-b]pyridine (PhIP) and its main metabolite 2-hydroxyamino-1-methyl-6-phenylimidazo[4,5-b]pyridine (N-OH-PhIP) in plasma, urine, faeces, bile, liver, kidney, testis, spleen, brain, as well as colon-, cecum- and small intestinal tissue and contents from mice. The effect of the matrix on the accuracy of the method was extensively investigated. The bioanalytical assay is based on reversed phase liquid chromatography coupled with tandem mass spectrometry in the positive ion mode using multiple reaction monitoring for analyte quantification. The assay is validated from 1 to 250 ng/mL and the sample pretreatment consists of protein precipitation with acetonitrile using only 100 microL matrix (plasma, bile diluted in 4% (m/v) BSA, intestinal contents, faeces and tissue samples homogenized in 4% (m/v) BSA). The measured concentrations of PhIP and N-OH-PhIP in homogenates were expressed in ng/mL. Based on the weight of the isolated intestinal contents, faeces or tissue the amount of PhIP and N-OH-PhIP per mass unit intestinal content, faeces or tissue was calculated. The validated range for PhIP in urine is from 10 to 1000 ng/mL using 20 microL urine. For N-OH-PhIP quantification, mouse urine was diluted 100 x in blank human urine to compensate for matrix effects. The developed method is simple, robust and reproducible. The applicability of the method was demonstrated and the assay could be successfully used to support in vivo toxicokinetics studies of PhIP and N-OH-PhIP in mice.


Subject(s)
Chromatography, High Pressure Liquid/methods , Feces/chemistry , Imidazoles/analysis , Pyridines/analysis , Tandem Mass Spectrometry/methods , Acetonitriles/chemistry , Animals , Bodily Secretions/chemistry , Digestive System/chemistry , Drug Stability , Humans , Imidazoles/blood , Imidazoles/urine , Linear Models , Mice , Pyridines/blood , Pyridines/urine , Reproducibility of Results
18.
Br J Pharmacol ; 160(5): 1224-33, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20590614

ABSTRACT

BACKGROUND AND PURPOSE: Lopinavir is extensively metabolized by cytochrome P450 3A (CYP3A) and is considered to be a substrate for the drug transporters ABCB1 (P-glycoprotein) and ABCC2 (MRP2). Here, we have assessed the individual and combined effects of CYP3A, ABCB1 and ABCC2 on the pharmacokinetics of lopinavir and the relative importance of intestinal and hepatic metabolism. We also evaluated whether ritonavir increases lopinavir oral bioavailability by inhibition of CYP3A, ABCB1 and/or ABCC2. EXPERIMENTAL APPROACH: Lopinavir transport was measured in Madin-Darby canine kidney cells expressing ABCB1 or ABCC2. Oral lopinavir kinetics (+/- ritonavir) was studied in mice with genetic deletions of Cyp3a, Abcb1a/b and/or Abcc2, or in transgenic mice expressing human CYP3A4 exclusively in the liver and/or intestine. KEY RESULTS: Lopinavir was transported by ABCB1 but not by ABCC2 in vitro. Lopinavir area under the plasma concentration - time curve (AUC)(oral) was increased in Abcb1a/b(-/-) mice (approximately ninefold vs. wild-type) but not in Abcc2(-/-) mice. Increased lopinavir AUC(oral) (>2000-fold) was observed in cytochrome P450 3A knockout (Cyp3a(-/-)) mice compared with wild-type mice. No difference in AUC(oral) between Cyp3a(-/-) and Cyp3a/Abcb1a/b/Abcc2(-/-) mice was observed. CYP3A4 activity in intestine or liver, separately, reduced lopinavir AUC(oral) (>100-fold), compared with Cyp3a(-/-) mice. Ritonavir markedly increased lopinavir AUC(oral) in all CYP3A-containing mouse strains. CONCLUSIONS AND IMPLICATIONS: CYP3A was the major determinant of lopinavir pharmacokinetics, far more than Abcb1a/b. Both intestinal and hepatic CYP3A activity contributed importantly to low oral bioavailability of lopinavir. Ritonavir increased lopinavir bioavailability primarily by inhibiting CYP3A. Effects of Abcb1a/b were only detectable in the presence of CYP3A, suggesting saturation of Abcb1a/b in the absence of CYP3A activity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cytochrome P-450 CYP3A/drug effects , HIV Protease Inhibitors/pharmacokinetics , Multidrug Resistance-Associated Proteins/metabolism , Pyrimidinones/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Biological Availability , Cytochrome P-450 CYP3A/genetics , Dogs , Drug Interactions , Humans , Intestinal Mucosa/metabolism , Intestines/enzymology , Liver/enzymology , Liver/metabolism , Lopinavir , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Ritonavir/pharmacology
19.
Clin Pharmacol Ther ; 86(4): 396-402, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19625999

ABSTRACT

Cisplatin is one of the most widely used anticancer agents for the treatment of solid tumors. The clinical use of cisplatin is associated with dose-limiting nephrotoxicity, which occurs in one-third of patients despite intensive prophylactic measures. Organic cation transporter 2 (OCT2) has been implicated in the cellular uptake of cisplatin, but its role in cisplatin-induced nephrotoxicity remains unknown. In mice, deletion of Oct1 and Oct2 resulted in significantly impaired urinary excretion of cisplatin without an apparent influence on plasma levels. Furthermore, the Oct1/Oct2-deficient mice were protected from severe cisplatin-induced renal tubular damage. Subsequently, we found that a nonsynonymous single-nucleotide polymorphism (SNP) in the OCT2 gene SLC22A2 (rs316019) was associated with reduced cisplatin-induced nephrotoxicity in patients. Collectively, these results indicate the critical importance of OCT2 in the renal handling and related renal toxicity of cisplatin and provide a rationale for the development of new targeted approaches to mitigate this debilitating side effect.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Kidney Diseases/chemically induced , Organic Cation Transport Proteins/metabolism , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/urine , Cisplatin/blood , Cisplatin/urine , Humans , Kidney Diseases/pathology , Male , Mice , Mice, Knockout , Organic Cation Transport Proteins/genetics , Organic Cation Transporter 1/genetics , Organic Cation Transporter 1/metabolism , Organic Cation Transporter 2 , Polymerase Chain Reaction , Polymorphism, Single Nucleotide
20.
J Chromatogr B Analyt Technol Biomed Life Sci ; 877(24): 2519-29, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19589736

ABSTRACT

The development and validation of a bioanalytical assay is described for the simultaneous analysis in human serum of tamoxifen, four of its main metabolites and three flavonoids, which are known constituents in alternative medicine and dietary supplements often used by breast cancer patients. The method has been fully validated at linear ranges covering steady-state serum concentrations in patients who receive therapeutic dosages of tamoxifen. The wide range also allows for quantification of large inter-patient fluctuations of flavonoid concentrations. The bioanalytical assay is based on reversed phase liquid chromatography coupled with tandem mass spectrometry in the positive ion mode using multiple reaction monitoring for drug (-metabolite) quantification. The sample pretreatment consists of a protein precipitation with acetonitrile using only 50 microL serum. The described method is simple, robust and reproducible with inter- and intra-assay accuracies within 85-115%. The applicability of the assay was demonstrated and it is now successfully used to study the in vivo pharmacokinetics of tamoxifen, its main metabolites and flavonoids in human serum of patients receiving tamoxifen.


Subject(s)
Antineoplastic Agents/blood , Chromatography, Liquid/methods , Isoflavones/blood , Tamoxifen/blood , Tandem Mass Spectrometry/methods , Antineoplastic Agents/metabolism , Humans , Neoplasms/blood , Neoplasms/drug therapy , Tamoxifen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...