Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Pharm Res ; 40(8): 1885-1899, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37344602

ABSTRACT

BACKGROUND & PURPOSE: Heroin (diacetylmorphine; diamorphine) is a highly addictive opioid prodrug. Heroin prescription is possible in some countries for chronic, treatment-refractory opioid-dependent patients and as a potent analgesic for specific indications. We aimed to study the pharmacokinetic interactions of heroin and its main pharmacodynamically active metabolites, 6-monoacetylmorphine (6-MAM) and morphine, with the multidrug efflux transporters P-glycoprotein/ABCB1 and BCRP/ABCG2 using wild-type, Abcb1a/1b and Abcb1a/1b;Abcg2 knockout mice. METHODS & RESULTS: Upon subcutaneous (s.c.) heroin administration, its blood levels decreased quickly, making it challenging to detect heroin even shortly after dosing. 6-MAM was the predominant active metabolite present in blood and most tissues. At 10 and 30 min after heroin administration, 6-MAM and morphine brain accumulation were increased about 2-fold when mouse (m)Abcb1a/1b and mAbcg2 were ablated. Fifteen minutes after direct s.c. administration of an equimolar dose of 6-MAM, we observed good intrinsic brain penetration of 6-MAM in wild-type mice. Still, mAbcb1 limited brain accumulation of 6-MAM and morphine without affecting their blood exposure, and possibly mediated their direct intestinal excretion. A minor contribution of mAbcg2 to these effects could not be excluded. CONCLUSIONS: We show that mAbcb1a/1b can limit 6-MAM and morphine brain exposure. Pharmacodynamic behavioral/postural observations, while non-quantitative, supported moderately increased brain levels of 6-MAM and morphine in the knockout mouse strains. Variation in ABCB1 activity due to genetic polymorphisms or environmental factors (e.g., drug interactions) might affect 6-MAM/morphine exposure in individuals, but only to a limited extent.


Subject(s)
Heroin , Morphine , Mice , Animals , Heroin/metabolism , Heroin/pharmacology , Morphine/metabolism , Analgesics, Opioid/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Neoplasm Proteins/metabolism , Brain/metabolism , Morphine Derivatives/metabolism , Morphine Derivatives/pharmacology , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Mice, Knockout
2.
Biomed Pharmacother ; 164: 114956, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37267638

ABSTRACT

Mammalian carboxylesterase 1 enzymes can hydrolyze many xenobiotic chemicals and endogenous lipids. We here identified and characterized a mouse strain (FVB/NKI) in which three of the eight Ces1 genes were spontaneously deleted, removing Ces1c and Ces1e partly, and Ces1d entirely. We studied the impact of this Ces1c/d/e deficiency on drug and lipid metabolism and homeostasis. Ces1c/d/e-/- mice showed strongly impaired conversion of the anticancer prodrug irinotecan to its active metabolite SN-38 in plasma, spleen and lung. Plasma hydrolysis of the oral anticancer prodrug capecitabine to 5-DFCR was also profoundly reduced in Ces1c/d/e-/- mice. Our findings resolved previously unexplained FVB/NKI pharmacokinetic anomalies. On a medium-fat diet, Ces1c/d/e-/- female mice exhibited moderately higher body weight, mild inflammation in gonadal white adipose tissue (gWAT), and increased lipid load in brown adipose tissue (BAT). Ces1c/d/e-/- males showed more pronounced inflammation in gWAT and an increased lipid load in BAT. On a 5-week high-fat diet exposure, Ces1c/d/e deficiency predisposed to developing obesity, enlarged and fatty liver, glucose intolerance and insulin resistance, with severe inflammation in gWAT and increased lipid load in BAT. Hepatic proteomics analysis revealed that the acute phase response, involved in the dynamic cycle of immunometabolism, was activated in these Ces1c/d/e-/- mice. This may contribute to the obesity-related chronic inflammation and adverse metabolic disease in this strain. While Ces1c/d/e deficiency clearly exacerbated metabolic syndrome development, long-term (18-week) high-fat diet exposure overwhelmed many, albeit not all, observed phenotypic differences.


Subject(s)
Carboxylic Ester Hydrolases , Metabolic Syndrome , Prodrugs , Animals , Female , Male , Mice , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Inflammation , Irinotecan , Lipids , Mammals , Obesity/metabolism
3.
Acta Pharm Sin B ; 13(2): 618-631, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36873183

ABSTRACT

The mammalian carboxylesterase 1 (Ces1/CES1) family comprises several enzymes that hydrolyze many xenobiotic chemicals and endogenous lipids. To investigate the pharmacological and physiological roles of Ces1/CES1, we generated Ces1 cluster knockout (Ces1 -/- ) mice, and a hepatic human CES1 transgenic model in the Ces1 -/- background (TgCES1). Ces1 -/- mice displayed profoundly decreased conversion of the anticancer prodrug irinotecan to SN-38 in plasma and tissues. TgCES1 mice exhibited enhanced metabolism of irinotecan to SN-38 in liver and kidney. Ces1 and hCES1 activity increased irinotecan toxicity, likely by enhancing the formation of pharmacodynamically active SN-38. Ces1 -/- mice also showed markedly increased capecitabine plasma exposure, which was moderately decreased in TgCES1 mice. Ces1 -/- mice were overweight with increased adipose tissue, white adipose tissue inflammation (in males), a higher lipid load in brown adipose tissue, and impaired blood glucose tolerance (in males). These phenotypes were mostly reversed in TgCES1 mice. TgCES1 mice displayed increased triglyceride secretion from liver to plasma, together with higher triglyceride levels in the male liver. These results indicate that the carboxylesterase 1 family plays essential roles in drug and lipid metabolism and detoxification. Ces1 -/- and TgCES1 mice will provide excellent tools for further study of the in vivo functions of Ces1/CES1 enzymes.

4.
Bioanalysis ; 12(19): 1405-1425, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32975434

ABSTRACT

Therapeutic monoclonal antibodies (mAbs) are rapidly taking over the treatment of many malignancies, and an astonishing number of mAbs is in development. This causes a high demand for quantification of mAbs in biomatrices both for measuring therapeutic mAb concentrations and to support pharmacokinetics and pharmacodynamics studies. Conventionally, ligand-binding assays are used for these purposes, but LC-MS is gaining popularity. Although intact (top-down) and subunit (middle-down) mAb quantification is reported, signature peptide (bottom-up) quantification is currently most advantageous. This review provides an overview of the reported bottom-up mAb quantification methods in biomatrices as well as general recommendations regarding signature peptide and internal standard selection, reagent use and optimization of digestion in bottom-up quantification methods.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/therapeutic use , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Antineoplastic Agents/pharmacokinetics , Humans
5.
Anal Bioanal Chem ; 411(20): 5331-5345, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31209549

ABSTRACT

A novel method was developed and validated for the quantification of the three approved CDK4/6 inhibitors (abemaciclib, palbociclib, and ribociclib) in both human and mouse plasma and mouse tissue homogenates (liver, kidney, spleen, brain, and small intestine) using liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS). For all matrices, pretreatment was performed using 50 µL of sample by protein precipitation with acetonitrile, followed by dilution of the supernatant. Chromatographic separation of the analytes was done on a C18 column using gradient elution. A full validation was performed for human plasma, while a partial validation was executed for mouse plasma and mouse tissue homogenates. The method was linear in the calibration range from 2 to 200 ng/mL, with a correlation coefficient (r) ≥0.996 for each analyte. For both human and mouse plasma, the accuracy and precision were within ±15% and ≤15%, respectively, for all concentrations, except for the lower limit of quantification, where they were within ±20% and ≤20%, respectively. A fit-for-purpose strategy was followed for tissue homogenates, and the accuracy and precision were within ±20% and ≤20%, respectively, for all concentrations. Stability of all analytes in all matrices at different processing and storage conditions was tested; ribociclib and palbociclib were unstable in most tissue homogenates and conditions were modified to increase the stability. The method was successfully applied for the analysis of mouse samples from preclinical studies. A new ribociclib metabolite was detected in mouse plasma samples with the same m/z transition as the parent drug.


Subject(s)
Aminopyridines/analysis , Benzimidazoles/analysis , Chromatography, Liquid/methods , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Piperazines/analysis , Protein Kinase Inhibitors/analysis , Purines/analysis , Pyridines/analysis , Tandem Mass Spectrometry/methods , Aminopyridines/pharmacology , Animals , Benzimidazoles/pharmacology , Humans , Mice , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Pyridines/pharmacology
6.
J Pharm Anal ; 6(4): 268-275, 2016 Aug.
Article in English | MEDLINE | ID: mdl-29403992

ABSTRACT

Many anticancer drugs have an impaired bioavailability and poor brain penetration because they are substrates to drug efflux pumps such as P-glycoprotein and Breast Cancer Resistance Protein. Elacridar is a strong inhibitor of these two drug efflux pumps and therefore has great potential to improve oral absorption and brain penetration of many anticancer drugs. Currently, a clinical formulation of elacridar is unavailable and therefore the pharmaceutical development of a drug product is highly warranted. This also necessitates the availability of an analytical method for its quality control. A reverse-phase high-performance liquid chromatographic method with ultraviolet detection was developed for the pharmaceutical quality control of products containing elacridar as the active pharmaceutical ingredient. The analytical method was validated for linearity, accuracy, precision, selectivity, carry-over, stability of stock and reference solutions, stability of the final extract, stability-indicating capability and impurity testing. We found that elacridar is unstable in aqueous solutions that are exposed to light because a hydroxylation product of elacridar is formed. Therefore, sample solutions with elacridar must be protected from light.

7.
J Pharm Biomed Anal ; 110: 58-66, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-25804433

ABSTRACT

5-Fluorouracil (5-FU) and its oral prodrug capecitabine are among the most widely used chemotherapeutics. For cytotoxic activity, 5-FU requires cellular uptake and intracellular metabolic activation. Three intracellular formed metabolites are responsible for the antineoplastic effect of 5-FU: 5-fluorouridine 5'-triphosphate (FUTP), 5-fluoro-2'-deoxyuridine 5'-triphosphate (FdUTP) and 5-fluoro-2'-deoxyuridine 5'-monophosphate (FdUMP). In this paper, we describe the development of an LC-MS/MS assay for quantification of these active 5-FU nucleotides in peripheral blood mononuclear cells (PBMCs). Because the intracellular 5-FU nucleotide concentrations were very low, maximization of the release from the cell matrix and minimization of interference were critical factors. Therefore, a series of experiments was performed to select the best method for cell lysis and nucleotide extraction. Chromatography was optimized to obtain separation from endogenous nucleotides, and the effect of different cell numbers was examined. The assay was validated for the following concentration ranges in PBMC lysate: 0.488-19.9 nM for FUTP, 1.66-67.7 nM for FdUTP and 0.748-30.7 nM for FdUMP. Accuracies were between -2.2 and 7.0% deviation for all analytes, and the coefficient of variation values were ≤ 4.9%. The assay was successfully applied to quantify 5-FU nucleotides in PBMC samples from patients treated with capecitabine and patients receiving 5-FU intravenously. FUTP amounts up to 3054 fmol/10(6) PBMCs and FdUMP levels up to 169 fmol/10(6) PBMCs were measured. The FdUTP concentrations were below the lower limit of quantification. To our knowledge, this is the first time that 5-FU nucleotides were quantified in cells from patients treated with 5-FU or capecitabine without using a radiolabel.


Subject(s)
Antineoplastic Agents/metabolism , Chromatography, Liquid , Drug Monitoring/methods , Fluorouracil/metabolism , Leukocytes, Mononuclear/metabolism , Tandem Mass Spectrometry , Antineoplastic Agents/pharmacokinetics , Biological Transport , Biotransformation , Calibration , Chromatography, Liquid/standards , Deoxyuracil Nucleotides/metabolism , Drug Monitoring/standards , Floxuridine/analogs & derivatives , Floxuridine/metabolism , Fluorodeoxyuridylate/metabolism , Fluorouracil/pharmacokinetics , Humans , Linear Models , Reference Standards , Reproducibility of Results , Tandem Mass Spectrometry/standards , Uridine Triphosphate/analogs & derivatives , Uridine Triphosphate/metabolism
8.
J Pharm Biomed Anal ; 90: 7-14, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24317024

ABSTRACT

Azacitidine is a cytidine analog used in the treatment of myelodysplastic syndromes, chronic myelomonocytic leukemia and acute myeloid leukemia. The pharmacological effect of azacitidine arises after incorporation into the DNA and RNA. To this end, the drug first has to be converted into its triphosphate forms. This paper describes the development of an assay for quantitative determination of azacitidine triphosphate (aza-CTP) in peripheral blood mononuclear cells (PBMCs). To quantify aza-CTP, separation from the endogenous nucleotides cytidine triphosphate (CTP) and uridine triphosphate (UTP) is required. This was a challenge as the structures of these nucleotides are highly similar and the monoisotopic molecular masses of aza-CTP, UTP and the naturally occurring [(13)C]- and [(15)N]-isotopes of CTP differ less than 0.02 Da. Efforts to select a specific MS(2)-fragment for aza-CTP using a triple quadrupole mass spectrometer remained without success. Therefore, we investigated the feasibility to separate these highly resembling nucleotides based on accurate mass spectrometry using a linear trap quadrupole (LTQ) coupled with an Orbitrap. The LTQ-Orbitrap was able to differentiate between aza-CTP and the endogenous nucleotides UTP and [(13)C]-CTP. There was no baseline resolution between aza-CTP and [(15)N]-CTP, but the [(15)N]-CTP interference was low. For quantification, extracted ion chromatograms were obtained for the accurate m/z window of the aza-CTP product ion. The assay was able to determine aza-CTP concentrations in PBMC lysate from 40.7 to 281 nM. Assuming that an average cell suspension extracted from 16 mL blood contains 10 to 42 million PBMCs per mL, this range corresponds with 2.58/10.9-17.8/74.9 pmol aza-CTP per million PBMCs. Intra-assay accuracies were between -1.1 and 9.5% deviation and coefficient of variation values were ≤13.2%. The assay was successfully applied to quantify aza-CTP in samples from two patients treated with azacitidine. Aza-CTP concentrations up to 19.0 pmol per million PBMCs were measured. This is the first time that aza-CTP concentrations were quantified in PBMCs from patients treated with azacitidine.


Subject(s)
Antimetabolites, Antineoplastic/analysis , Azacitidine/analysis , Chromatography, Liquid/methods , Mass Spectrometry/methods , Aged , Antimetabolites, Antineoplastic/metabolism , Azacitidine/metabolism , Feasibility Studies , Female , Humans , Leukocytes, Mononuclear/metabolism , Male , Middle Aged
9.
Article in English | MEDLINE | ID: mdl-23270936

ABSTRACT

Capecitabine is the oral prodrug of the anticancer drug 5-fluorouracil (5-FU). The purpose of this study was to quantify capecitabine and its metabolites including 5'-deoxy-5-fluorocytidine (5'-dFCR), 5'-deoxy-5-fluorouridine (5'-dFUR), 5-FU, dihydro-5-fluorouracil (FUH(2)), α-fluoro-ureidopropionic acid (FUPA) and fluoro-ß-alanine (FBAL) in human plasma using liquid chromatography coupled to electrospray tandem mass spectrometry. To this end two individual assays were developed: one for the simultaneous quantification of capecitabine, 5'-dFCR and 5'-dFUR using reversed phase chromatography and gradient elution, and one assay for 5-FU, FUH(2), FUPA and FBAL using hydrophilic interaction chromatography and isocratic elution. Both assays were fully validated according to current FDA guidelines. Total run time for the capecitabine assay was 9.0min, and of the 5-FU assay 5.0min. Analyte extraction was performed by protein precipitation. Stable labeled isotopes for each of the analytes were used as internal standards. The linear ranges of the analytes were 50-6000ng/mL for the capecitabine assay and 50-5000ng/mL for the 5-FU assay. Validation results demonstrate that capecitabine and its metabolites can be rapidly, accurately, precisely and robustly quantified in human plasma with the presented methods. Both assays are currently in extensive use in support of pharmacokinetic studies in patients treated with capecitabine or 5-FU.


Subject(s)
Chromatography, High Pressure Liquid/methods , Deoxycytidine/analogs & derivatives , Fluorouracil/analogs & derivatives , Prodrugs/chemistry , Tandem Mass Spectrometry/methods , Capecitabine , Deoxycytidine/blood , Deoxycytidine/chemistry , Deoxycytidine/metabolism , Deoxycytidine/pharmacokinetics , Drug Stability , Fluorouracil/blood , Fluorouracil/chemistry , Fluorouracil/metabolism , Fluorouracil/pharmacokinetics , Humans , Linear Models , Mass Spectrometry/methods , Prodrugs/metabolism , Prodrugs/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity
10.
Int J Cancer ; 130(1): 223-33, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21351087

ABSTRACT

Sunitinib is an orally active, multitargeted tyrosine kinase inhibitor which has been used for the treatment of metastatic renal cell carcinoma and imatinib-resistant gastrointestinal stromal tumors. We aimed to investigate the in vivo roles of the ATP-binding cassette drug efflux transporters ABCB1 and ABCG2 in plasma pharmacokinetics and brain accumulation of oral sunitinib, and the feasibility of improving sunitinib kinetics using oral coadministration of the dual ABCB1/ABCG2 inhibitor elacridar. We used in vitro transport assays and Abcb1a/1b(-/-) , Abcg2(-/-) and Abcb1a/1b/Abcg2(-/-) mice to study the roles of ABCB1 and ABCG2 in sunitinib disposition. In vitro, sunitinib was a good substrate of murine (mu)ABCG2 and a moderate substrate of human (hu)ABCB1 and huABCG2. In vivo, the systemic exposure of sunitinib after oral dosing (10 mg kg(-1) ) was unchanged when muABCB1 and/or muABCG2 were absent. Brain accumulation of sunitinib was markedly (23-fold) increased in Abcb1a/b/Abcg2(-/-) mice, but only slightly (2.3-fold) in Abcb1a/b(-/-) mice, and not in Abcg2(-/-) mice. Importantly, a clinically realistic coadministration of oral elacridar and oral sunitinib to wild-type mice resulted in markedly increased sunitinib brain accumulation, equaling levels in Abcb1a/1b/Abcg2(-/-) mice. This indicates complete inhibition of the blood-brain barrier (BBB) transporters. High-dose intravenous sunitinib could saturate BBB muABCG2, but not muABCB1A, illustrating a dose-dependent relative impact of the BBB transporters. Brain accumulation of sunitinib is effectively restricted by both muABCB1 and muABCG2 activity. Complete inhibition of both transporters, leading to markedly increased brain accumulation of sunitinib, is feasible and safe with a clinically realistic oral elacridar/sunitinib coadministration.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , ATP-Binding Cassette Transporters/physiology , Acridines/administration & dosage , Antineoplastic Agents/administration & dosage , Brain/metabolism , Indoles/administration & dosage , Pyrroles/administration & dosage , Tetrahydroisoquinolines/administration & dosage , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Biological Transport , Blood-Brain Barrier/drug effects , Brain/drug effects , Dogs , Drug Resistance, Multiple , Humans , Indoles/blood , Indoles/pharmacokinetics , Male , Mice , Mice, Knockout , Pyrroles/blood , Pyrroles/pharmacokinetics , Sunitinib , Tissue Distribution
11.
J Chromatogr B Analyt Technol Biomed Life Sci ; 879(28): 2984-90, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21920826

ABSTRACT

A combined assay for the determination of paclitaxel, docetaxel and ritonavir in human plasma is described. The drugs were extracted from 200 µL human plasma using liquid-liquid extraction with tertiar-butylmethylether, followed by high performance liquid chromatography analysis using 10 mM ammonium hydroxide pH 10:methanol (3:7, v/v) as mobile phase. Chromatographic separation was obtained using a Zorbax Extend C(18) column. Labelled analogues of the analytes are used as internal standards. For detection, positive ionization electrospray tandem mass spectrometry was used. Method development including optimisation of the mass transitions and response, mobile phase optimisation and column selection are discussed. The method was validated according to FDA guidelines and the principles of Good Laboratory Practice (GLP). The validated range was 0.5-500 ng/mL for paclitaxel and docetaxel and 2-2000 ng/mL for ritonavir. For quantification, quadratic calibration curves were used (r(2)>0.99). The total runtime of the method is 9 min and the assay combines analytes with differences in ionisation and desired concentration range. Inter-assay accuracy and precision were tested at four concentration levels and were within 10% and less than 10%, respectively, for all analytes. Carry-over was less than 6% and endogenous interferences or interferences between analytes and internal standards were less than 20% of the response at the lower limit of quantification level. The matrix factor and recovery were determined at low, mid and high concentration levels. The matrix factor was around 1 for all analytes and total recovery between 77.5 and 104%. Stability was investigated in stock solutions, human plasma, dry extracts, final extracts and during 3 freeze/thaw cycles. The described method was successfully applied in clinical studies with oral administration of docetaxel or paclitaxel in combination with ritonavir.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/blood , Chromatography, High Pressure Liquid/methods , Paclitaxel/blood , Ritonavir/blood , Tandem Mass Spectrometry/methods , Taxoids/blood , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Docetaxel , Humans , Neoplasms/blood , Neoplasms/drug therapy , Paclitaxel/administration & dosage , Ritonavir/administration & dosage , Sensitivity and Specificity , Spectrometry, Mass, Electrospray Ionization , Taxoids/administration & dosage
12.
Hum Exp Toxicol ; 30(9): 1399-403, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21056950

ABSTRACT

Morphine-6-glucuronide, the active metabolite of morphine, and to a lesser extent morphine itself are known to accumulate in patients with renal failure. A number of cases on non-lethal morphine toxicity in patients with renal impairment report high plasma concentrations of morphine-6-glucuronide, suggesting that this metabolite achieves sufficiently high brain concentrations to cause long-lasting respiratory depression, despite its poor central nervous system penetration. We report a lethal morphine intoxication in a 61-year-old man with sickle cell disease and renal impairment, and we measured concentrations of morphine and morphine-6-glucuronide in blood, brain and cerebrospinal fluid. There were no measurable concentrations of morphine-6-glucuronide in cerebrospinal fluid or brain tissue, despite high blood concentrations. In contrast, the relatively high morphine concentration in the brain suggests that morphine itself was responsible for the cardiorespiratory arrest in this patient. Given the fatal outcome, we recommend to avoid repeated or continuous morphine administration in renal failure.


Subject(s)
Anemia, Sickle Cell/drug therapy , Morphine/toxicity , Renal Insufficiency/drug therapy , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/complications , Brain/drug effects , Brain/metabolism , Fatal Outcome , Heart Arrest/chemically induced , Humans , Male , Middle Aged , Morphine/blood , Morphine/cerebrospinal fluid , Morphine/therapeutic use , Morphine Derivatives/blood , Morphine Derivatives/cerebrospinal fluid , Renal Insufficiency/blood , Renal Insufficiency/complications
13.
Clin Cancer Res ; 15(7): 2344-51, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19276246

ABSTRACT

PURPOSE: Imatinib, a BCR-ABL tyrosine kinase inhibitor, is a substrate of the efflux transporters P-glycoprotein (P-gp; ABCB1) and ABCG2 (breast cancer resistance protein), and its brain accumulation is restricted by both transporters. For dasatinib, an inhibitor of SCR/BCR-ABL kinases, in vivo interactions with P-gp and ABCG2 are not fully established yet. EXPERIMENTAL DESIGN: We used Abcb1a/1b(-/-), Abcg2(-/-), and Abcb1a/1b;Abcg2(-/-) mice to establish the roles of P-gp and ABCG2 in the pharmacokinetics and brain accumulation of dasatinib. RESULTS: We found that oral uptake of dasatinib is limited by P-gp. Furthermore, relative brain accumulation, 6 hours after administration, was not affected by Abcg2 deficiency, but absence of P-gp resulted in a 3.6-fold increase after oral and 4.8-fold higher accumulation after i.p. administration. Abcb1a/1b;Abcg2(-/-) mice had the most pronounced increase in relative brain accumulation, which was 13.2-fold higher after oral and 22.7-fold increased after i.p. administration. Moreover, coadministration to wild-type mice of dasatinib with the dual P-gp and ABCG2 inhibitor elacridar resulted in a similar dasatinib brain accumulation as observed for Abcb1a/1b;Abcg2(-/-) mice. CONCLUSIONS: Brain accumulation of dasatinib is primarily restricted by P-gp, but Abcg2 can partly take over this protective function at the blood-brain barrier. Consequently, when both transporters are absent or inhibited, brain uptake of dasatinib is highly increased. These findings might be clinically relevant for patients with central nervous system Philadelphia chromosome-positive leukemia, as coadministration of an inhibitor of P-gp and ABCG2 with dasatinib might result in better therapeutic responses in these patients.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Acridines/pharmacology , Antineoplastic Agents/pharmacokinetics , Brain/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , Tetrahydroisoquinolines/pharmacology , Thiazoles/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1 , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/metabolism , Biological Transport , Cell Line , Dasatinib , Dogs , Mice , Mice, Knockout , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/metabolism , Pyrimidines/blood , Pyrimidines/metabolism , Thiazoles/blood , Thiazoles/metabolism
14.
J Chromatogr B Analyt Technol Biomed Life Sci ; 865(1-2): 55-62, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18325856

ABSTRACT

A sensitive and specific liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay for the quantification of miltefosine is presented. A 250 microL human EDTA plasma aliquot was spiked with miltefosine and extracted by a solid-phase extraction method. Separation was performed on a Gemini C18 column (150 mm x 2.0 mm I.D., 5 microm) using an alkaline eluent. Detection was performed by positive ion electrospray ionization followed by triple-quadrupole mass spectrometry. The assay has been validated for miltefosine from 4 to 2000 ng/mL using 250 microL human EDTA plasma samples. Results from the validation demonstrate that miltefosine can be accurately and precisely quantified in human plasma. At the lowest level, the intra-assay precision was lower than 10.7%, the inter-assay precision was 10.6% and accuracies were between 95.1 and 109%. This assay is successfully used in a clinical pharmacokinetic study with miltefosine.


Subject(s)
Antiprotozoal Agents/blood , Chromatography, Liquid/methods , Phosphorylcholine/analogs & derivatives , Tandem Mass Spectrometry/methods , Antiprotozoal Agents/pharmacokinetics , Humans , Phosphorylcholine/blood , Phosphorylcholine/pharmacokinetics , Sensitivity and Specificity
15.
Rapid Commun Mass Spectrom ; 21(15): 2505-14, 2007.
Article in English | MEDLINE | ID: mdl-17610214

ABSTRACT

For the quantification of all currently approved non-nucleoside reverse transcriptase inhibitors and protease inhibitors, including the new protease inhibitor darunavir and the active nelfinavir metabolite M8, an assay was developed, using liquid chromatography coupled with tandem mass spectrometry. The sample pretreatment consisted of a protein precipitation with a mixture of methanol and acetonitrile using only 100 microL plasma. Chromatographic separation was performed on a reversed-phase C18 column (150 x 2.0 mm, particle size 5 microm) with a quick stepwise gradient using an acetate buffer (pH 5) and methanol, at a flow rate of 0.25 mL/min. The analytical run time was only 10 min. The triple quadrupole mass spectrometer was operated in the positive ion mode and multiple reaction monitoring was used for drug quantification. The method was validated over a range of 0.1 to 20 microg/mL for amprenavir, atazanavir, efavirenz, indinavir, lopinavir, nelfinavir, the active nelfinavir metabolite M8, nevirapine and ritonavir, a range of 0.05 to 10 microg/mL for saquinavir and darunavir and a range of 0.5 to 100 microg/mL for tipranavir, based on observed concentration ranges in patients treated with these drugs. D5-squinavir, D6-indinavir, 13C6-efavirenz and dibenzepine were used as internal standards. The method was proven to be specific, accurate, precise and robust. Accuracies ranged from 88.5% to 102.2% and all precisions were less than 9.5%. Furthermore, the assay demonstrates a high sensitivity for all analytes and the stepwise gradient allows addition of new analytes into the same method. The method is now successfully applied for routine therapeutic drug monitoring and pharmacokinetic studies in HIV-infected patients.


Subject(s)
Chromatography, High Pressure Liquid , Drug Monitoring/methods , HIV Protease Inhibitors/blood , Spectrometry, Mass, Electrospray Ionization/methods , Sulfonamides/blood , Darunavir , Humans , Reproducibility of Results , Sensitivity and Specificity
16.
Mol Pharmacol ; 72(2): 387-94, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17485564

ABSTRACT

Glucuronidation is a major hepatic detoxification pathway for endogenous and exogenous compounds, resulting in the intracellular formation of polar metabolites that require specialized transporters for elimination. Multidrug resistance proteins (MRPs) are expressed in the liver and can transport glucuronosyl-conjugates. Using morphine as a model aglycone, we demonstrate that morphine-3-glucuronide (M3G), the predominant metabolite, is transported in vitro by human MRP2 (ABCC2), a protein present in the apical membrane of hepatocytes. Loss of biliary M3G secretion in Mrp2(-/-) mice results in its increased sinusoidal transport that can be attributed to Mrp3. Combined loss of Mrp2 and Mrp3 leads to a substantial accumulation of M3G in the liver, from which it is transported across the sinusoidal membrane at a low rate, resulting in the prolonged presence of M3G in plasma. Our results show that murine Mrp2 and Mrp3 provide alternative routes for the excretion of a glucuronidated substrate from the liver in vivo.


Subject(s)
Liver/metabolism , Membrane Transport Proteins/physiology , Morphine Derivatives/metabolism , Multidrug Resistance-Associated Proteins/physiology , Animals , Cell Line , Glucuronosyltransferase/physiology , Humans , Mice , Multidrug Resistance-Associated Protein 2 , Spodoptera
17.
Am J Physiol Cell Physiol ; 292(6): C2204-12, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17314268

ABSTRACT

Proper regulation of intracellular levels of protoporphyrin IX (PPIX), the direct precursor of heme, is important for cell survival. A deficiency in ferrochelatase, which mediates the final step in heme biosynthesis, leads to erythropoietic protoporphyria (EPP), a photosensitivity syndrome caused by the accumulation of PPIX in the skin. We have previously shown that mice with a deficiency in the ABC transporter Bcrp1/Abcg2 display a novel type of protoporphyria. This protoporphyria is mild compared with ferrochelatase-dependent EPP, and in itself not sufficient to cause phototoxicity, but it might exacerbate the consequences of other porphyrias. In this study, we identified the mouse harderian gland as a novel expression site of Bcrp1. Because of its pronounced role in porphyrin secretion, the harderian gland presents a useful tool to study the mechanism of Bcrp1-related protoporphyria and transport of porphyrins. Bcrp1(-/-) harderian gland displayed a highly increased accumulation of PPIX glycoconjugates, and a similar shift was seen in Bcrp1(-/-) liver. Tear- and hepatobiliary excretion data suggest that Bcrp1 controls intracellular levels of PPIX by mediating high affinity transport of its glycoconjugates and possibly low-affinity transport of unconjugated PPIX. This mechanism may allow cells to prevent or reduce cytotoxicity of PPIX under excess conditions, without spillage under physiological conditions where PPIX is needed.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Biological Transport, Active/physiology , Harderian Gland/metabolism , Protoporphyrins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Animals , Bile/metabolism , Female , Gene Deletion , Gene Expression Regulation , Harderian Gland/anatomy & histology , Liver/metabolism , Male , Mice , Molecular Structure , Protoporphyrins/chemistry
18.
Cancer Chemother Pharmacol ; 59(6): 825-37, 2007 May.
Article in English | MEDLINE | ID: mdl-16988825

ABSTRACT

PURPOSE: Trabectedin (ET-743, Yondelis) is a novel anti-cancer drug currently undergoing phase II-III evaluation, that has shown remarkable activity in pre-treated patients with soft tissue sarcoma. Despite extensive pharmacokinetic studies, the human disposition and metabolism of trabectedin remain largely unknown. We aimed to determine the metabolic profile of trabectedin and to identify its metabolites in humans. METHODS: We analysed urine and faeces (the major excretory route) from eight cancer patients after a 3 or 24 h intravenous administration of [14C]trabectedin. Using liquid chromatography with tandem quadrupole mass spectrometric detection (LC-MS/MS) and radiochromatography with off-line radioactivity detection by liquid scintillation counting (LC-LSC), we characterised the metabolic profile in 0-24 h urine and 0-120 h faeces. RESULTS: By radiochromatography, a large number of trabectedin metabolites were detected. Incubation with beta-glucuronidase indicated the presence of a glucuronide metabolite in urine. Trabectedin, ET-745, ET-759A, ETM-259, ETM-217 (all available as reference compounds) and a proposed new metabolite coined ET-731 were detected using LC-MS/MS. The inter-individual differences in radiochromatographic profiles were small and did not correlate with polymorphisms in drug-metabolising enzymes (CYP2C9, 2C19, 2D6, 2E1, 3A4, GST-M1, P1, T1 and UGT1A1 2B15) as determined by genotyping. CONCLUSIONS: Trabectedin is metabolically converted to a large number of compounds that are excreted in both urine and faeces. In urine and faeces we have confirmed the presence of trabectedin, ET-745, ET-759A, ETM-259, ETM-217 and ETM-204. In addition we have identified a putative new metabolite designated ET-731. Future studies should be aimed at further identification of possible metabolites and assessment of their activity.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Dioxoles/pharmacokinetics , Neoplasms/drug therapy , Tetrahydroisoquinolines/pharmacokinetics , Chromatography , Dioxoles/chemistry , Dioxoles/urine , Feces/chemistry , Glucuronides/analysis , Humans , Mass Spectrometry , Neoplasms/metabolism , Polymorphism, Genetic , Tetrahydroisoquinolines/chemistry , Tetrahydroisoquinolines/urine , Trabectedin
19.
J Mass Spectrom ; 41(10): 1268-76, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16981212

ABSTRACT

A sensitive and specific LC-MS/MS assay for the quantitative determination of EO9 and its metabolite EO5a is presented. A 200-microl human plasma aliquot was spiked with a mixture of deuterated internal standards EO9-d3 and EO5a-d4 and extracted with 1.25 ml ethyl acetate. Dried extracts were reconstituted in 0.1 M ammonium acetate-methanol (7 : 3, v/v) and 25 microl-volumes were injected into the HPLC system. Separation was achieved on a 150 x 2.1 mm C18 column using an alkaline eluent (1 mM ammonium hydroxide-methanol (gradient system)). Detection was performed by positive ion electrospray followed by tandem mass spectrometry. The assay quantifies a range from 5 to 2500 ng/ml for EO9 and from 10 to 2500 ng/ml EO5a using 200 microl of human plasma samples. Validation results demonstrate that EO9 and EO5a concentrations can be accurately and precisely quantified in human plasma. This assay will be used to support clinical pharmacologic studies with EO9.


Subject(s)
Antineoplastic Agents, Alkylating/blood , Aziridines/blood , Indolequinones/blood , Calibration , Chromatography, High Pressure Liquid , Humans , Quality Control , Reference Standards , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization
20.
Radiother Oncol ; 80(2): 207-13, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16914220

ABSTRACT

PURPOSE: Perifosine is an orally applicable, membrane-targeted alkylphosphocholine analogue with antitumour activity and radiosensitising properties in preclinical models. The purpose of this phase I study was to determine the feasibility and tolerability of concurrent daily perifosine and radiation in patients with advanced cancer. PATIENTS AND METHODS: Starting dose of perifosine was 50 mg/day; dose escalation was in steps of 50mg. Daily administration commenced 2 days before radiotherapy and was continued throughout the radiation treatment. At least three patients were entered at each dose level; at the 150 mg/day level 10 patients were included. Pharmacokinetic sampling was performed weekly pre-dosing. Twenty-one patients were entered. Tumour types included NSCLC (n=17), prostate, oesophageal, colon and bladder cancer. Most patients (16/21) had received prior chemotherapy; none radiotherapy. Median number of daily perifosine administrations was 31 (range 24-53). Mean radiation dose (BED(10)) was 59.8 Gy (range 50.7-87.5 Gy in 13-28 fractions). RESULTS: Major drug-related toxicities according to CTC criteria were nausea in 57%, fatigue in 48%, vomiting in 38%, diarrhoea in 38% and anorexia in 19%. No bone marrow toxicity was observed. DLT (nausea/vomiting) was encountered in two of five patients at the 200mg/day dose level. Dose-dependent steady-state plasma levels were reached after 1 week. Major radiotherapy-related acute toxicity consisted of dysphagia in 38% and pneumonitis in 29%. CONCLUSION: Perifosine can be safely combined with fractionated radiotherapy. A dosage of 150 mg/day, to be started at least 1 week prior to radiotherapy, is recommended for phase II evaluation.


Subject(s)
Neoplasms/drug therapy , Neoplasms/radiotherapy , Phosphorylcholine/analogs & derivatives , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Neoplasms/metabolism , Phosphorylcholine/administration & dosage , Phosphorylcholine/adverse effects , Phosphorylcholine/pharmacokinetics , Radiation-Sensitizing Agents/administration & dosage , Radiation-Sensitizing Agents/adverse effects , Radiation-Sensitizing Agents/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...