Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Br J Dermatol ; 182(1): 180-189, 2020 01.
Article in English | MEDLINE | ID: mdl-30916381

ABSTRACT

BACKGROUND: We evaluated antidrug antibody (ADA) development in patients with chronic plaque psoriasis from three clinical trials of tildrakizumab, a humanized anti-interleukin-23p19 monoclonal antibody (P05495, reSURFACE 1 and reSURFACE 2). OBJECTIVES: To determine the effects of immunogenicity on the pharmacokinetics, efficacy and safety of tildrakizumab. METHODS: In 1400 (weeks 12-16) and 780 (weeks 52-64) evaluable patients randomized to tildrakizumab 100 or 200 mg, treatment-emergent ADA-positive (TE-POS) patients were identified and characterized for neutralizing antibodies (NAbs). Pharmacokinetics, safety and efficacy were evaluated by ADA status. RESULTS: In patients treated with tildrakizumab 100 or 200 mg continuously, < 7% were inconclusive at 52-64 weeks. In long-term data through 52-64 weeks, the incidence of TE-POS was 6·5% (100 mg) and 8·2% (200 mg) and the incidence of TE-POS NAb-POS was 2·5% (100 mg) and 3·2% (200 mg). TE-POS NAb-POS patients had modestly increased median tildrakizumab clearance (36·5%) compared with ADA-NEG patients. Percentage Psoriasis Area and Severity Index improvements in TE-POS NAb-POS vs. ADA-NEG patients on continuous treatment through week 52 were 76% (n = 10) vs. 91% (n = 342) for 100 mg and 77% (n = 12) vs. 87% (n = 299) for 200 mg. The incidence of potential immunogenicity-related adverse events did not indicate a clear trend in any positive ADA patient category compared with ADA-NEG patients through weeks 52-64. The effects of ADA on pharmacokinetics, efficacy and safety at 12-16 weeks were also summarized. CONCLUSIONS: ADA development with tildrakizumab treatment for 52-64 weeks was low; around 3% of patients developed TE-POS NAb-POS ADAs and showed lower serum concentrations and corresponding reduced efficacy. No relationship between ADAs and safety was observed. What's already known about this topic? Unwanted immune responses - for example immunogenicity and antidrug antibodies (ADAs) - have been observed with therapeutic monoclonal antibodies and can affect efficacy and safety. Tildrakizumab is a humanized monoclonal antibody targeting interleukin-23 and is currently approved for patients with plaque psoriasis. What does this study add? ADA development in tildrakizumab-treated patients with psoriasis over 52 weeks was low. The small proportion of patients who had treatment-emergent ADAs and had neutralizing antibodies experienced lower serum tildrakizumab concentrations and reduced efficacy. No relationship between ADAs and safety events was observed.


Subject(s)
Antibodies, Monoclonal, Humanized , Psoriasis , Antibodies, Monoclonal/adverse effects , Antibodies, Neutralizing , Humans , Psoriasis/drug therapy , Treatment Outcome
2.
CPT Pharmacometrics Syst Pharmacol ; 5(3): 93-122, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27069774

ABSTRACT

This document was developed to enable greater consistency in the practice, application, and documentation of Model-Informed Drug Discovery and Development (MID3) across the pharmaceutical industry. A collection of "good practice" recommendations are assembled here in order to minimize the heterogeneity in both the quality and content of MID3 implementation and documentation. The three major objectives of this white paper are to: i) inform company decision makers how the strategic integration of MID3 can benefit R&D efficiency; ii) provide MID3 analysts with sufficient material to enhance the planning, rigor, and consistency of the application of MID3; and iii) provide regulatory authorities with substrate to develop MID3 related and/or MID3 enabled guidelines.


Subject(s)
Guidelines as Topic , Technology, Pharmaceutical/standards , Documentation , Drug Design , Technology, Pharmaceutical/methods
3.
Clin Pharmacol Ther ; 65(1): 10-20, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9951426

ABSTRACT

BACKGROUND: The use of pimozide is associated with prolongation of the QT interval and fatal ventricular arrhythmia. We recently reported 2 fatal cases in patients taking pimozide and clarithromycin and we have shown that clarithromycin inhibits CYP3A-mediated metabolism of pimozide in vitro. In this study, we examined the effect of clarithromycin on pimozide pharmacokinetics and QT interval changes in a total of 12 healthy subjects (7 men and 5 women), documented as extensive metabolizers or poor metabolizers of CYP2D6. METHODS: In a randomized, double-blind placebo-controlled crossover design, subjects were given a single 6-mg oral dose of pimozide after 5 days of treatment with clarithromycin (500 mg twice a day) or a placebo pill. Blood samples were obtained before and for 96 hours after pimozide administration, and plasma pimozide and clarithromycin concentrations were measured by HPLC. Electrocardiograms for the analysis of the QTc intervals were recorded immediately before each blood sample. RESULTS: Pimozide significantly lengthened QTc interval in the first 20 hours in both the placebo-treated groups (delta QTcmax = 13.3 +/- 5.3 ms; P = .003) and clarithromycin-treated groups (delta QTcmax = 15.7 +/- 9.5 ms; P = .005) compared with baseline values. This is consistent with an effect of the parent drug. Clarithromycin caused a significant increase in the peak plasma concentration (P = .015), terminal elimination half-life (P = .003), and area under the plasma concentration-time curve (P = .024) and a decrease in the clearance (P = .029) of pimozide. Mean QTcmax observed within 20 hours of pimozide administration was significantly greater in the clarithromycin-treated group (23.8 +/- 12.2 ms; P = .0397) than in the placebo-treated group (16.8 +/- 6 ms). There was no significant effect of CYP2D6 or gender on the pharmacokinetics or pharmacodynamics of pimozide. CONCLUSIONS: A single 6-mg oral dose of pimozide resulted in measurable QT interval changes. Clarithromycin inhibited CYP3A-mediated pimozide metabolism and the resulting elevation in plasma concentrations may increase the risk of pimozide cardiotoxicity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antipsychotic Agents/pharmacokinetics , Clarithromycin/pharmacology , Cytochrome P-450 CYP2D6/metabolism , Dopamine Antagonists/pharmacokinetics , Pimozide/pharmacokinetics , Protein Synthesis Inhibitors/pharmacology , Adult , Anti-Bacterial Agents/administration & dosage , Antipsychotic Agents/administration & dosage , Clarithromycin/administration & dosage , Cross-Over Studies , Cytochrome P-450 CYP2D6/drug effects , Dopamine Antagonists/administration & dosage , Double-Blind Method , Drug Administration Schedule , Electrocardiography/drug effects , Female , Heart Conduction System/drug effects , Humans , Male , Middle Aged , Pimozide/administration & dosage , Protein Synthesis Inhibitors/administration & dosage , Reference Values , Sex Factors , Time Factors
4.
Clin Pharmacol Ther ; 70(2): 132-41, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11503007

ABSTRACT

BACKGROUND: The autoinducible metabolic transformation of the anticancer agent ifosfamide involves activation through 4-hydroxyifosfamide to the ultimate cytotoxic ifosforamide mustard and deactivation to 2- and 3-dechloroethylifosfamide with concomitant release of the neurotoxic chloroacetaldehyde. Activation is mediated by cytochrome P450 (CYP) 3A4 and deactivation by CYP3A4 and CYP2B6. The aim of this study was to investigate modulation of the CYP-mediated metabolism of ifosfamide with ketoconazole, a potent inhibitor of CYP3A4, and rifampin (INN, rifampicin), an inducer of CYP3A4/CYP2B6. METHODS: In a double-randomized, 2-way crossover study a total of 16 patients received ifosfamide 3 g/m(2) per 24 hours intravenously, either alone or in combination with 200 mg ketoconazole twice daily (1 day before treatment and 3 days of concomitant administration) or 300 mg rifampin twice daily (3 days before treatment and 3 days of concomitant administration). Plasma pharmacokinetics and urinary excretion of ifosfamide, 2- and 3-dechloroethylifosfamide, and 4-hydroxyifosfamide were assessed in both courses. Data analysis was performed with a population pharmacokinetic model with a description of autoinduction of ifosfamide. RESULTS: Rifampin increased the clearance of ifosfamide at the start of therapy at 102%. The fraction of ifosfamide metabolized to the dechloroethylated metabolites was increased, whereas exposure to the metabolites was decreased as a result of increased elimination. The fraction metabolized and the exposure to 4-hydroxyifosfamide were not significantly influenced. Ketoconazole did not affect the fraction metabolized or the exposure to the dechloroethylated metabolites, whereas both parameters were reduced with 4-hydroxyifosfamide. CONCLUSIONS: Coadministration of ifosfamide with ketoconazole or rifampin did not produce changes in the pharmacokinetics of the parent or metabolites that may result in an increased benefit of ifosfamide therapy.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/pharmacology , Ifosfamide/pharmacokinetics , Ketoconazole/pharmacology , Rifampin/pharmacology , Adult , Aged , Antibiotics, Antitubercular/pharmacology , Antifungal Agents/pharmacology , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/blood , Area Under Curve , Bayes Theorem , Cross-Over Studies , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/biosynthesis , Drug Administration Schedule , Enzyme Induction/drug effects , Enzyme Inhibitors/administration & dosage , Female , Humans , Ifosfamide/administration & dosage , Ifosfamide/blood , Ketoconazole/administration & dosage , Male , Middle Aged , Mixed Function Oxygenases/drug effects , Mixed Function Oxygenases/metabolism , Oxidoreductases, N-Demethylating/drug effects , Oxidoreductases, N-Demethylating/metabolism , Rifampin/administration & dosage , Time Factors
5.
Clin Pharmacokinet ; 40(8): 615-25, 2001.
Article in English | MEDLINE | ID: mdl-11523727

ABSTRACT

OBJECTIVE: To assess the feasibility of a sparse sampling approach for the determination of the population pharmacokinetics of ifosfamide, 2- and 3-dechloroethyl-ifosfamide and 4-hydroxy-ifosfamide in children treated with single-agent ifosfamide against various malignant tumours. DESIGN: Pharmacokinetic assessment followed by model fitting. PATIENTS: The analysis included 32 patients aged between 1 and 18 years receiving a total of 45 courses of ifosfamide 1.2, 2 or 3 g/m2 in 1 or 3 hours on 1, 2 or 3 days. METHODS: A total of 133 blood samples (median of 3 per patient) were collected. Plasma concentrations of ifosfamide and its dechloroethylated metabolites were determined by gas chromatography. Plasma concentrations of 4-hydroxy-ifosfamide were measured by high-performance liquid chromatography. The models were fitted to the data using a nonlinear mixed effects model as implemented in the NONMEM program. A cross-validation was performed. RESULTS: Population values (mean +/- standard error) for the initial clearance and volume of distribution of ifosfamide were estimated at 2.36 +/- 0.33 L/h/m2 and 20.6 +/- 1.6 L/m2 with an interindividual variability of 43 and 32%, respectively. The enzyme induction constant was estimated at 0.0493 +/- 0.0104 L/h2/m2. The ratio of the fraction of ifosfamide metabolised to each metabolite to the volume of distribution of that metabolite, and the elimination rate constant, of 2- and 3-dechloroethyl-ifosfamide and 4-hydroxy-ifosfamide were 0.0976 +/- 0.0556, 0.0328 +/- 0.0102 and 0.0230 +/- 0.0083 m2/L and 3.64 +/- 2.04, 0.445 +/- 0.174 and 7.67 +/- 2.87 h(-1), respectively. Interindividual variability of the first parameter was 23, 34 and 53%, respectively. Cross-validation indicated no bias and minor imprecision (12.5 +/- 5.1%) for 4-hydroxy-ifosfamide only. CONCLUSIONS: We have developed and validated a model to estimate ifosfamide and metabolite concentrations in a paediatric population by using sparse sampling.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Ifosfamide/pharmacokinetics , Models, Biological , Neoplasms/drug therapy , Adolescent , Adult , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/therapeutic use , Area Under Curve , Child , Child, Preschool , Feasibility Studies , Female , Humans , Ifosfamide/metabolism , Ifosfamide/therapeutic use , Infant , Male , Metabolic Clearance Rate
6.
Clin Pharmacokinet ; 40(1): 41-62, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11236809

ABSTRACT

This review discusses several issues in the clinical pharmacology of the antitumour agent ifosfamide and its metabolites. Ifosfamide is effective in a large number of malignant diseases. Its use, however, can be accompanied by haematological toxicity, neurotoxicity and nephrotoxicity. Since its development in the middle of the 1960s, most of the extensive metabolism of ifosfamide has been elucidated. Identification of specific isoenzymes responsible for ifosfamide metabolism may lead to an improved efficacy/toxicity ratio by modulation of the metabolic pathways. Whether ifosfamide is specifically transported by erythrocytes and which activated ifosfamide metabolites play a key role in this transport is currently being debated. In most clinical pharmacokinetic studies, the phenomenon of autoinduction has been observed, but the mechanism is not completely understood. Assessment of the pharmacokinetics of ifosfamide and metabolites has long been impaired by the lack of reliable bioanalytical assays. The recent development of improved bioanalytical assays has changed this dramatically, allowing extensive pharmacokinetic assessment, identifying key issues such as population differences in pharmacokinetic parameters, differences in elimination dependent upon route and schedule of administration, implications of the chirality of the drug and interpatient pharmacokinetic variability. The mechanisms of action of cytotoxicity, neurotoxicity, urotoxicity and nephrotoxicity have been pivotal issues in the assessment of the pharmacodynamics of ifosfamide. Correlations between the new insights into ifosfamide metabolism, pharmacokinetics and pharmacodynamics will rationalise the further development of therapeutic drug monitoring and dose individualisation of ifosfamide treatment.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Cyclophosphamide/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Ifosfamide/pharmacokinetics , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/adverse effects , Area Under Curve , Cyclophosphamide/administration & dosage , Cytochrome P-450 Enzyme System/drug effects , Enzyme Inhibitors , Fanconi Syndrome/chemically induced , Humans , Ifosfamide/administration & dosage , Ifosfamide/adverse effects , Methylene Blue/therapeutic use , Neurotoxicity Syndromes/drug therapy , Stereoisomerism
7.
Cancer Chemother Pharmacol ; 46(2): 119-27, 2000.
Article in English | MEDLINE | ID: mdl-10972481

ABSTRACT

PURPOSE: Cyclophosphamide and thioTEPA are frequently used simultaneously in high-dose chemotherapy regimens. During a pharmacokinetic study of 31 courses in 20 patients of cyclophosphamide and its activated metabolite 4-hydroxycyclophosphamide given in the combination cyclophosphamide thioTEPA carboplatin, a sharp decrease in 4-hydroxycyclophosphamide concentration was observed immediately after the start of the thioTEPA infusion. A drug-drug interaction was suspected. This putative interaction was investigated in this study. METHODS: Possible sequence dependency, due to inhibition of the formation of 4-hydroxycyclophosphamide by thioTEPA, was investigated by altering the sequence of infusion in three patients (four courses) receiving high-dose chemotherapy with cyclophosphamide (1,000 or 1,500 mg/m2 per day), thioTEPA (80 or 120 mg/m2 per day) and carboplatin (265 or 400 mg/m2 per day) in short infusions for four consecutive days. The pharmacokinetics of cyclophosphamide and 4-hydroxycyclophosphamide were established. Possible inhibition of the metabolism of cyclophosphamide and thioTEPA was investigated in human microsomes. RESULTS: A striking sequence dependency of the pharmacokinetics of 4-hydroxycyclophosphamide was observed. Administration of thioTEPA 1 h prior to cyclophosphamide resulted in decreased Cmax (-62%) and AUC (-26%) values of 4-hydroxycyclophosphamide compared to those of thioTEPA administered 1 h after cyclophosphamide. In human microsomes an inhibition of the conversion of cyclophosphamide to 4-hydroxycyclophosphamide by thioTEPA was observed at clinically relevant concentrations with an IC50 of 23 microM. No inhibition of the formation of TEPA by cyclophosphamide was observed. CONCLUSIONS: ThioTEPA strongly inhibits the bioactivation of cyclophosphamide and this may decrease both efficacy and toxicity. Our results seriously question the practice of the simultaneous continuous infusion of cyclophosphamide and thioTEPA and suggest that the sequencing and scheduling of these two agents in high-dose chemotherapy regimens may be of critical importance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Cyclophosphamide/pharmacokinetics , Neoplasms/drug therapy , Thiotepa/pharmacokinetics , Biotransformation , Cyclophosphamide/administration & dosage , Cyclophosphamide/analogs & derivatives , Cyclophosphamide/blood , Drug Administration Schedule , Hematopoietic Stem Cell Transplantation , Humans , Kinetics , Microsomes/metabolism , Neoplasms/blood , Thiotepa/administration & dosage , Time Factors
8.
Cancer Chemother Pharmacol ; 48(1): 53-61, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11488525

ABSTRACT

The aim of this study was to develop a population pharmacokinetic model that could describe the pharmacokinetics of ifosfamide. 2- and 3-dechloroethylifosfamide and 4-hydroxyifosfamide, and calculate their plasma exposure and urinary excretion. A group of 14 patients with small-cell lung cancer received a 1-h intravenous infusion of 2.0 or 3.0 g/m2 ifosfamide over 1 or 2 days in combination with 175 mg/m2 paclitaxel and carboplatin at AUC 6. The concentration-time profiles of ifosfamide were described by an ifosfamide concentration-dependent development of autoinduction of ifosfamide clearance. Metabolite compartments were linked to the ifosfamide compartment enabling description of the concentration-time profiles of 2- and 3-dechloroethylifosfamide and 4-hydroxyifosfamide. The Bayesian estimates of the pharmacokinetic parameters were used to calculate the systemic exposure to ifosfamide and its metabolites for the four ifosfamide schedules. Fractionation of the dose over 2 days resulted increased metabolite formation, especially of 2-dechloroethylifosfamide, probably due to increased autoinduction. Renal recovery was only minor with 6.6% of the administered dose excreted unchanged and 9.8% as dechloroethylated metabolites. In conclusion, ifosfamide pharmacokinetics were described with an ifosfamide concentration-dependent development of autoinduction and allowed estimation of the population pharmacokinetics of the metabolites of ifosfamide. Fractionation of the dose resulted in increased exposure to 2-dechloroethylifosfamide, probably due to increased autoinduction.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Carcinoma, Small Cell/drug therapy , Ifosfamide/pharmacokinetics , Lung Neoplasms/drug therapy , Adult , Aged , Area Under Curve , Drug Resistance, Neoplasm , Female , Humans , Male , Middle Aged
9.
Clin Pharmacol Ther ; 96(1): 101-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24682029

ABSTRACT

To support the development of a fixed-dose combination (FDC) of ezetimibe and atorvastatin for the treatment of dyslipidemia, bioequivalence (BE) studies were conducted across a combined dose range (10/10, 10/20, 10/40, and 10/80 mg of ezetimibe/atorvastatin). In the BE trials, all parameters met traditional BE bounds except for atorvastatin peak plasma concentration (Cmax) at two intermediate doses. Literature-based metadata analysis predicted that the observed difference in Cmax between an ezetimibe+atorvastatin FDC and coadministration of these agents translates directly into a non-clinically significant change of <1.2% absolute difference in the percentage lowering of low-density-lipoprotein cholesterol . Both FDC doses were confirmed to be clinically equivalent to coadministration in the subsequent clinical equivalence trials. These data suggest that modeling of dose-response relationships may be useful in predicting clinical equivalence, lowering cost/timelines through effective powering of studies, and predicting the effectiveness of new dosage formulations without the need for additional clinical efficacy trials in regulatory settings.


Subject(s)
Heptanoic Acids/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Models, Biological , Pyrroles/pharmacokinetics , Atorvastatin , Azetidines/pharmacokinetics , Azetidines/pharmacology , Cholesterol, LDL/blood , Drug Therapy, Combination , Ezetimibe , Heptanoic Acids/pharmacology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Meta-Analysis as Topic , Pyrroles/pharmacology , Therapeutic Equivalency
10.
CPT Pharmacometrics Syst Pharmacol ; 3: e142, 2014 Oct 22.
Article in English | MEDLINE | ID: mdl-25338195

ABSTRACT

Quantitative and systems pharmacology concepts and tools are the foundation of the model-informed drug development paradigm at Merck for integrating knowledge, enabling decisions, and enhancing submissions. Rigorous prioritization of modeling and simulation activities has enabled key drug development decisions and led to a high return on investment through significant cost avoidance. Critical factors for the successful implementation, examples on impact on decision making with associated return of investment, and drivers for continued success are discussed.

11.
Clin Pharmacol Ther ; 86(1): 84-91, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19387434

ABSTRACT

Modeling and simulation were utilized to characterize the efficacy dose response of sublingual asenapine in patients with schizophrenia and to understand the outcomes of six placebo-controlled trials in which placebo responses and dropout rates varied. The time course of total Positive and Negative Syndrome Scale (PANSS) scores was characterized for placebo and asenapine treatments in a pharmacokinetic-pharmacodynamic model in which the asenapine effect was described by an E(max) model, increasing linearly over the 6-week study period. A logistic regression model described the time course of dropouts, with previous PANSS value being the most important predictor. The last observation carried forward (LOCF) time courses were well described in simulations from the combined PANSS + dropout model. The observed trial outcomes were successfully predicted for all the placebo arms and the majority of the treatment arms. Although simulations indicated that the post hoc probability of success of the performed trials was low to moderate, these analyses demonstrated that 5 and 10 mg twice-daily (b.i.d.) doses of asenapine have similar efficacy.


Subject(s)
Computer Simulation , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Models, Biological , Patient Dropouts/psychology , Schizophrenia/drug therapy , Schizophrenic Psychology , Acute Disease , Controlled Clinical Trials as Topic/methods , Dibenzocycloheptenes , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Humans , Schizophrenia/metabolism , Time Factors
13.
Pharm Stat ; 5(2): 135-44, 2006.
Article in English | MEDLINE | ID: mdl-17080770

ABSTRACT

We discuss the practical and clinical considerations encountered when planning a Phase IIa trial in chronic obstructive pulmonary disease (COPD). Various adaptive strategies for reducing the cost of the trial and the statistical implications of these are explored. Use of the EAST software to evaluate the properties of the study designs with one or more interim analyses for futility, efficacy or either is described. We emphasize the rationale for choosing between alternative designs and the relationship between the clinical and statistical considerations.


Subject(s)
Clinical Trials, Phase II as Topic/methods , Pulmonary Disease, Chronic Obstructive/drug therapy , Research Design , Data Interpretation, Statistical , Humans , Sample Size
14.
J Chromatogr B Biomed Sci Appl ; 694(1): 163-8, 1997 Jun 20.
Article in English | MEDLINE | ID: mdl-9234859

ABSTRACT

A method is described for the measurement of pimozide in human plasma using HPLC with fluorescence detection. The method is specific and sensitive in the range of concentrations seen in human plasma after conventional dosing (1-15 ng/ml) with a limit of quantification of 1 ng/ml. The calibration curves are linear for concentrations between 1 and 50 ng/ml. Within-day and inter-day coefficients of variation are less than 7.4% and 15.5%, respectively, at three concentrations of pimozide (2, 10 and 20 ng/ml). Intra-day and inter-day bias are less than 18.5% and 12.5%, respectively. A pharmacokinetic study conducted in a healthy volunteer administered 6 mg of pimozide orally demonstrates the utility of this method.


Subject(s)
Antipsychotic Agents/blood , Pimozide/blood , Antipsychotic Agents/pharmacokinetics , Chromatography, High Pressure Liquid , Humans , Pimozide/pharmacokinetics , Sensitivity and Specificity , Spectrometry, Fluorescence
15.
J Pharmacol Exp Ther ; 285(2): 428-37, 1998 May.
Article in English | MEDLINE | ID: mdl-9580580

ABSTRACT

Using human liver microsomes (HLMs) and recombinant human cytochrome P450 (CYP450) isoforms, we identified the major route of pimozide metabolism, the CYP450 isoforms involved, and documented the inhibitory effect of pimozide on CYP450 isoforms. Pimozide was predominantly N-dealkylated to 1,3-dihydro-1-(4-piperidinyl)-2H-benzimidazol-2-one (DHPBI). The formation rate of DHPBI showed biphasic kinetics in HLMs, which suggests the participation of at least two activities. These were characterized as high-affinity (K(m1) and Vmax1) and low-affinity (K(m2) and Vmax2) components. The ratio of Vmax1 (14 pmol/min/mg protein)/K(m1) (0.73 microM) was 5.2 times higher than the ratio of Vmax2 (244 pmol/min/mg protein)/K(m2) (34 microM). K(m2) was 91 times higher than K(m1). The formation rate of DHPBI from 25 microM pimozide in nine human livers correlated significantly with the catalytic activity of CYP3A (Spearman r = 0.79, P = .028), but not with other isoforms. Potent inhibition of DHPBI formation from 10 microM pimozide was observed with ketoconazole (88%), troleandomycin (79%), furafylline (48%) and a combination of furafylline and ketoconazole (96%). Recombinant human CYP3A4 catalyzed DHPBI formation from 10 microM pimozide at the highest rate (V = 2.2 +/- 0.89 pmol/min/pmol P450) followed by CYP1A2 (V = 0.23 +/- 0.08 pmol/min/pmol P450), but other isoforms tested did not. The K(m) values derived with recombinant CYP3A4 and CYP1A2 were 5.7 microM and 36.1 microM, respectively. Pimozide itself was a potent inhibitor of CYP2D6 in HLMs when preincubated for 15 min (Ki = 0.75 +/- 0.98 microM) and a moderate inhibitor of CYP3A (Ki = 76.7 +/- 34.5 microM), with no significant effect on other isoforms tested. Our results suggest that pimozide metabolism is catalyzed mainly by CYP3A, but CYP1A2 also contributes. Pimozide metabolism is likely to be subject to interindividual variability in CYP3A and CYP1A2 expression and to drug interactions involving these isoforms. Pimozide itself may inhibit the metabolism of drugs that are substrates of CYP2D6.


Subject(s)
Antipsychotic Agents/metabolism , Cytochrome P-450 Enzyme System/physiology , Isoenzymes/physiology , Pimozide/metabolism , Cytochrome P-450 CYP2D6/physiology , Cytochrome P-450 CYP3A , Humans , Mixed Function Oxygenases/physiology
16.
J Pharmacol Exp Ther ; 284(3): 1095-103, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9495871

ABSTRACT

The purpose of our investigation was to characterize the relationships between the pharmacodynamics of synthetic opioids in vivo and the interaction at the mu-opioid receptor. The pharmacokinetics and pharmacodynamics were determined in vivo after a single i.v. infusion of 3.14 mg/kg alfentanil (A), 0.15 mg/kg fentanyl (F) or 0.030 mg/kg sufentanil (S) in rats. Amplitudes in the 0.5 to 4.5 Hz frequency band of the electroencephalogram (EEG) was used as pharmacodynamic endpoint. The EEG effect intensity was related to the (free) concentration in blood (A) or in a hypothetical effect compartment (F, S) on basis of the sigmoidal Emax pharmacodynamic model. The interaction at the mu-opioid receptor was determined in vitro on basis of the displacement of [3H]-naloxone binding in washed rat brain membranes. The value of the sodium shift was used as a measure of in vitro intrinsic efficacy. For the EEG effect the in vivo potencies based on free drug concentrations (EC50,u) were 4.62 +/- 0.66 ng/ml (A), 0.69 +/- 0.05 ng/ml (F) and 0.29 +/- 0.06 ng/ml (S). In the receptor binding studies the affinities at the mu-opioid receptor (Kl) were 47.4 +/- 6.6 nM (A), 8.6 +/- 4.1 nM (F) and 2.8 +/- 0.2 nM (S). For each opioid the ratio between EC50,u and Kl was the same with a value of 0.23-0.25, indicating the existence of receptor reserve for the EEG effect. The intrinsic activity (Emax) of the three opioids in vivo was similar with values of 111 +/- 10 microV (A), 89 +/- 11 microV (F) and 104 +/- 4 microV (S). However, the values of the sodium shift varied between 2.8 (S) and 19.1 (A). Further analysis of the in vivo pharmacodynamic data on basis of an operational model of agonism provided evidence for a large receptor reserve, which explains why compounds with different values of the sodium shift all behave as full agonists in vivo.


Subject(s)
Analgesics, Opioid/pharmacology , Electroencephalography/drug effects , Receptors, Opioid, mu/drug effects , Alfentanil/pharmacology , Analgesics, Opioid/pharmacokinetics , Animals , Dose-Response Relationship, Drug , Fentanyl/pharmacology , Male , Models, Biological , Naloxone/metabolism , Rats , Rats, Wistar , Sodium Chloride/pharmacology , Sufentanil/pharmacology
17.
J Chromatogr B Biomed Sci Appl ; 716(1-2): 177-86, 1998 Sep 25.
Article in English | MEDLINE | ID: mdl-9824231

ABSTRACT

A sensitive assay for the simultaneous determination of N,N',N"-triethylenethiophosphoramide (thioTEPA), its metabolite N,N',N"-triethylenephosphoramide (TEPA), cyclophosphamide (CP) and its metabolite 2-dechloroethylcyclophosphamide (2-DCE-CP) in plasma has been developed and validated. The analytes were determined using gas chromatography with nitrogen/phosphorus selective detection after liquid-liquid extraction with chloroform using 100 microl of plasma. Diphenylamine (for TEPA, thioTEPA and 2-DCE-CP) and imipramine (for CP) were used as internal standards. The limits of quantitation for thioTEPA, TEPA, CP and 2-DCE-CP were 5, 5, 50 and 250 ng/ml, respectively. Linear calibration curves were observed over two decades of concentration. Accuracy, within-day and between-day precision were less than 13% for all analytes. Stability of the analytes proved to be satisfactory for at least 1 month, stored at -70 degrees C. Analysis of samples obtained from patients receiving cyclophosphamide, thioTEPA and carboplatin in a high-dose regimen demonstrated the applicability of the assay.


Subject(s)
Antineoplastic Agents, Alkylating/blood , Chromatography, Gas/methods , Cyclophosphamide/blood , Thiotepa/blood , Cyclophosphamide/analogs & derivatives , Drug Stability , Humans , Quality Control , Sensitivity and Specificity , Triethylenephosphoramide/blood
18.
Eur J Clin Pharmacol ; 57(6-7): 467-77, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11699611

ABSTRACT

OBJECTIVE: The population pharmacokinetics and pharmacodynamics of the cytostatic agent ifosfamide and its main metabolites 2- and 3-dechloroethylifosfamide and 4-hydroxyifosfamide were assessed in patients with soft tissue sarcoma. METHODS: Twenty patients received 9 or 12 g/m2 ifosfamide administered as a 72-h continuous intravenous infusion. The population pharmacokinetic model was built in a sequential manner, starting with a covariate-free model and progressing to a covariate model with the aid of generalised additive modelling. RESULTS: The addition of the covariates weight, body surface area, albumin, serum creatinine, serum urea, alkaline phosphatase and lactate dehydrogenase improved the prediction errors of the model. Typical pretreatment (mean +/- SEM) initial clearance of ifosfamide was 3.03 +/- 0.18 l/h with a volume of distribution of 44.0 +/- 1.8 l. Autoinduction, dependent on ifosfamide levels, was characterised by an induction half-life of 11.5 +/- 1.0 h with 50% maximum induction at 33.0 +/- 3.6 microM ifosfamide. Significant pharmacokinetic-pharmacodynamic relationships (P = 0.019) were observed between the exposure to 2- and 3-dechloroethylifosfamide and orientational disorder, a neurotoxic side-effect. No pharmacokinetic-pharmacodynamic relationships between exposure to 4-hydroxyifosfamide and haematological toxicities could be observed in this population.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Cyclophosphamide/analogs & derivatives , Ifosfamide/analogs & derivatives , Ifosfamide/pharmacokinetics , Sarcoma/metabolism , Adult , Aged , Algorithms , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Agents, Alkylating/urine , Cyclophosphamide/blood , Cyclophosphamide/urine , Female , Humans , Ifosfamide/blood , Ifosfamide/therapeutic use , Ifosfamide/urine , Infusions, Intravenous , Male , Metabolic Clearance Rate , Middle Aged , Models, Biological , Sarcoma/drug therapy
19.
Ther Drug Monit ; 22(5): 613-20, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11034269

ABSTRACT

A comparison was made between methods for determining ifosfamide (IF), 2- (2DCE) and 3-dechloroethylifosfamide (3DCE) using gas chromatography with nitrogen-phosphorus detection (GC-NPD) versus positive ion electron-impact ion-trap mass spectrometry (GC-MS2). Sample pretreatment involved liquid-liquid extraction with ethyl acetate after adding trofosfamide as internal standard and alkalinization. The GC-NPD was linear, specific, and sensitive for all analytes in the range of 0.0500-100 microg/mL with lower limits of quantification (LLQ) of 0.0500 microg/mL using a 50-microgL plasma sample. The GC-MS2 was linear, specific, and sensitive for IF, 2DCE, and 3DCE in the ranges of 0.250-100, 0.500-25.0, and 0.500-25.0 microg/mL, respectively, with LLQs of 0.250, 0.500, and 0.500 microg/mL. The ranges of accuracy, within-day precision, and between-day precision for analysis of all compounds with GC-NPD did not exceed 93.3% to 105.4%, 8.0% and 9.8%, respectively. The ranges of accuracy, within-day precision, and between-day precision for analysis of all compounds with GC-MS2 did not exceed 86.5% to 99.0%, 9.0% and 12.7%, respectively. In conclusion, GC-NPD proved to be superior to GC-MS2 in sensitivity, detection range, accuracy, and precisions. Therefore GC-NPD is the method of choice for fast un-derivatized determination of IF, 2DCE, and 3DCE in human plasma, and it can readily be used for clinical pharmacokinetic studies and routine monitoring of IF-treated patients in a hospital setting.


Subject(s)
Antineoplastic Agents, Alkylating/analysis , Cyclophosphamide/analogs & derivatives , Cyclophosphamide/analysis , Drug Monitoring/standards , Ifosfamide/analogs & derivatives , Ifosfamide/analysis , Antineoplastic Agents, Alkylating/pharmacokinetics , Chromatography, Gas/standards , Cyclophosphamide/pharmacokinetics , Gas Chromatography-Mass Spectrometry/standards , Humans , Ifosfamide/pharmacokinetics , Quality Control , Sensitivity and Specificity
20.
Drug Metab Dispos ; 29(7): 967-75, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11408362

ABSTRACT

The anticancer drug ifosfamide is a prodrug requiring activation through 4-hydroxyifosfamide to ifosforamide mustard, to exert cytotoxicity. Deactivation of ifosfamide leads to 2- and 3-dechloroethylifosfamide and the release of potentially neurotoxic chloracetaldehyde. The aim of this study was to quantify and to compare the pharmacokinetics of ifosfamide, 2- and 3-dechloroethylifosfamide, 4-hydroxyifosfamide, and ifosforamide mustard in short (1-4 h), medium (24-72 h), and long infusion durations (96-240 h) of ifosfamide. An integrated population pharmacokinetic model was used to describe the autoinducible pharmacokinetics of ifosfamide and its four metabolites in 56 patients. The rate by which autoinduction of the metabolism of ifosfamide developed was found to be significantly dependent on the infusion schedule. The rate was 52% lower with long infusion durations compared with short infusion durations. This difference was, however, comparable with its interindividual variability (22%) and was, therefore, considered to be of minor clinical importance. Autoinduction caused a less than proportional increase in the area under the ifosfamide plasma concentration-time curve (AUC) and more than proportional increase in metabolite exposure with increasing ifosfamide dose. During long infusion durations dose-corrected exposures (AUC/D) were significantly decreased for ifosfamide and increased for 3-dechloroethylifosfamide compared with short infusion durations. No differences in dose-normalized exposure to ifosfamide and metabolites were observed between short and medium infusion durations. This study demonstrates that the duration of ifosfamide infusion influences the exposure to the parent and its metabolite 3-dechloroethylifosfamide. The observed dose and infusion duration dependence should be taken into account when modeling ifosfamide metabolism.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Ifosfamide/pharmacokinetics , Antineoplastic Agents, Alkylating/administration & dosage , Area Under Curve , Bayes Theorem , Chromatography, Gas , Dose-Response Relationship, Drug , Drug Administration Schedule , Humans , Ifosfamide/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL