Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
CPT Pharmacometrics Syst Pharmacol ; 12(1): 27-40, 2023 01.
Article in English | MEDLINE | ID: mdl-36385744

ABSTRACT

As formal causal inference begins to play a greater role in disciplines that intersect with pharmacometrics, such as biostatistics, epidemiology, and artificial intelligence/machine learning, pharmacometricians may increasingly benefit from a basic fluency in foundational causal inference concepts. This tutorial seeks to orient pharmacometricians to three such fundamental concepts: potential outcomes, g-formula, and directed acyclic graphs (DAGs).


Subject(s)
Artificial Intelligence , Biometry , Humans , Confounding Factors, Epidemiologic , Data Interpretation, Statistical , Causality
2.
Clin Pharmacokinet ; 58(12): 1577-1593, 2019 12.
Article in English | MEDLINE | ID: mdl-31104266

ABSTRACT

BACKGROUND AND OBJECTIVES: The thrombin inhibitor dabigatran is administered as the prodrug dabigatran etexilate, which is a substrate of esterases and P-glycoprotein (P-gp). Dabigatran is eliminated via renal excretion but is also a substrate of uridine 5'-diphospho (UDP)-glucuronosyltransferases (UGTs). The objective of this study was to build a physiologically based pharmacokinetic (PBPK) model comprising dabigatran etexilate, dabigatran, and dabigatran 1-O-acylglucuronide to describe the pharmacokinetics in healthy adults and renally impaired patients mechanistically. METHODS: Model development and evaluation were carried out using (i) physicochemical and absorption, distribution, metabolism, and excretion (ADME) parameter values of all three analytes; (ii) concentration-time profiles from 13 studies of healthy and renally impaired individuals after varying doses (0.1-300 mg), intravenous (dabigatran) and oral (dabigatran etexilate) administration, and different formulations of dabigatran etexilate (capsule, solution); and (iii) drug-drug interaction studies of dabigatran with the P-gp perpetrators rifampin (inducer) and clarithromycin (inhibitor). RESULTS: A PBPK model of dabigatran was successfully developed. The predicted area under the plasma concentration-time curve, trough concentration, and half-life values of the assessed clinical studies satisfied the two-fold acceptance criterion. Metabolic clearances of dabigatran etexilate and dabigatran were implemented using data on carboxylesterase 1/2 enzymes and UGT subtype 2B15. In severe renal impairment, the UGT2B15 metabolism and the P-gp transport in the model were reduced to 67% and 65% of the rates in healthy adults. CONCLUSION: This is the first implementation of a PBPK model for dabigatran to distinguish between the prodrug, active moiety, and main active metabolite. Following adjustment of the UGT2B15 metabolism and P-gp transport rates, the PBPK model accurately predicts the pharmacokinetics in renally impaired patients.


Subject(s)
Antithrombins/administration & dosage , Dabigatran/administration & dosage , Models, Biological , Renal Insufficiency/physiopathology , Adult , Antithrombins/pharmacokinetics , Dabigatran/pharmacokinetics , Glucuronides/chemistry , Humans
3.
CPT Pharmacometrics Syst Pharmacol ; 8(4): 230-239, 2019 04.
Article in English | MEDLINE | ID: mdl-30681293

ABSTRACT

Models were developed to characterize the relationship between afatinib exposure and diarrhea and rash/acne adverse event (AE) trajectories, and their predictive ability was assessed. Based on pooled data from seven phase II/III clinical studies including 998 patients, mixed-effects models for ordered categorical data were applied to describe daily AE severity. Clinical trial simulation aided by trial execution models was used for internal and external model evaluation. The final exposure-safety model consisted of longitudinal logistic regression models with first-order Markov elements for both AEs. Drug exposure was included as daily area under the concentration-time curve (AUC), and drug effects on the AEs were correlated. Clinical trial simulation allowed adequate prediction of maximum AE grades and AE severity time courses but overestimated the proportion of AE-dependent dose reductions and discontinuations. Both diarrhea and rash/acne were correlated with afatinib exposure. The developed modeling framework allows a prospective comparison of dosing strategies and study designs with respect to safety.


Subject(s)
Afatinib/adverse effects , Antineoplastic Agents/adverse effects , Diarrhea/chemically induced , Neoplasms/drug therapy , Skin Diseases/chemically induced , Acneiform Eruptions/chemically induced , Afatinib/therapeutic use , Antineoplastic Agents/therapeutic use , Area Under Curve , Clinical Decision-Making , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Computer Simulation , Exanthema/chemically induced , Humans , Logistic Models , Time Factors
4.
Thromb Haemost ; 118(9): 1625-1636, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30112751

ABSTRACT

BACKGROUND: Physiological age-related changes in the haemostatic and coagulation systems result in differing anticoagulant assay responses to standard anticoagulants. Therefore, we investigated the response of anticoagulant assays to dabigatran etexilate (DE) in children compared with adults. OBJECTIVE: This article assesses the relationship between plasma dabigatran concentration and coagulation assay results across age groups in children and adults. PATIENTS AND METHODS: Data from three clinical trials in which children received DE following standard of care for venous thromboembolism were compared with data from adult clinical trials. The effects of dabigatran concentration on diluted thrombin time (dTT), ecarin clotting time (ECT) and activated partial thromboplastin time (aPTT) were analysed graphically and with modelling. RESULTS: The concentration-dTT relationships were consistent in children across all ages and adults in the graphical analysis. For ECT and aPTT, relationships based on ratios over baseline were similar across all ages; absolute clotting times showed that the same exposure resulted in longer clotting times in some of the children aged < 1 year versus adults. Modelling showed concentration-clotting time relationships for all three assays were largely comparable between adults and children, except in those aged < 2 months, in whom there was a slight upward shift in ECT and aPTT relative to adults. CONCLUSION: Results suggest that developmental haemostatic changes will have little impact on response to DE. However, further paediatric clinical trials assessing the relationship between coagulation assay responses and clinical outcomes will be needed to confirm this finding.


Subject(s)
Anticoagulants/therapeutic use , Dabigatran/therapeutic use , Venous Thromboembolism/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Anticoagulants/pharmacokinetics , Blood Coagulation , Child , Dabigatran/pharmacokinetics , Female , Hemostasis , Humans , Infant , Male , Middle Aged , Partial Thromboplastin Time , Thrombin Time , Young Adult
5.
J Cardiovasc Pharmacol Ther ; 23(5): 399-406, 2018 09.
Article in English | MEDLINE | ID: mdl-29695165

ABSTRACT

INTRODUCTION: Dabigatran etexilate is an oral direct thrombin inhibitor. Dabigatran excretion is 80% renal, so exposure increases with severity of renal failure. The US Food and Drug Administration-approved dabigatran etexilate 75 mg twice daily (BID) for patients with nonvalvular atrial fibrillation (NVAF) having severely impaired renal function (creatinine clearance: 15-30 mL/min), based on post hoc pharmacokinetic modeling. We assessed dabigatran exposure at trough and peak levels in patients with NVAF and severely impaired renal function and compared with model predictions. METHODS: Patients received dabigatran etexilate (75 mg BID) for ≥7 days before blood sampling; Cpre,ss (steady-state predose concentration; trough) was taken 10 to 16 hours postdose (prior to next dose), and C2,ss (steady-state concentration; peak) was taken 2 hours (± 30 minutes) postdose. Pharmacodynamic parameters at baseline (Ebase), trough concentrations (Epre,ss), and peak concentrations (E2,ss) were assessed by established coagulation assays. RESULTS: Of the 150 patients screened, 60 were treated, of which 40% were male and 78.3% were white; median age was 84 years. Cpre,ss values (n = 51) were close to pharmacokinetic modeling predictions with a geometric mean (gMean) of 155 ng/mL, geometric coefficient of variation (gCV) of 76.9%, and range of 15.6 to 498 ng/mL. The C2,ss values (n = 59) had a gMean of 202 ng/mL, gCV of 70.6%, and range of 42.0 to 680 ng/mL. Pharmacodynamic effects on coagulation paralleled dabigatran concentrations. Eleven (18.3%) patients had ≥1 adverse event (AE); pharmacokinetic results for these patients versus those without AEs (n = 49) were Cpre, ss: gMean = 206 versus 145 ng/mL, gCV = 64.0% versus 78.3%; C2,ss: gMean = 243 versus 193 ng/mL, gCV = 68.9% versus 70.8%. All bleeding events (8 events in 5 patients) were considered minor by the investigators. CONCLUSION: Dabigatran exposure levels largely confirmed earlier pharmacokinetic predictions, supporting the use of dabigatran etexilate 75 mg BID in patients with NVAF and severely impaired renal function. Pharmacodynamic results were also in agreement with earlier studies. TRIAL REGISTRATION: ClinicalTrials.gov NCT01896297.


Subject(s)
Antithrombins/administration & dosage , Antithrombins/pharmacokinetics , Atrial Fibrillation/drug therapy , Dabigatran/administration & dosage , Dabigatran/pharmacokinetics , Kidney Diseases/physiopathology , Kidney/physiopathology , Aged , Aged, 80 and over , Antithrombins/adverse effects , Antithrombins/blood , Atrial Fibrillation/blood , Atrial Fibrillation/complications , Atrial Fibrillation/diagnosis , Biomarkers/blood , Creatinine/blood , Dabigatran/adverse effects , Dabigatran/blood , Drug Administration Schedule , Female , Hemorrhage/chemically induced , Humans , Kidney Diseases/blood , Kidney Diseases/complications , Kidney Diseases/diagnosis , Male , Models, Biological , Prospective Studies , Risk Factors , Severity of Illness Index , Treatment Outcome
6.
Thromb Haemost ; 117(11): 2168-2175, 2017 11.
Article in English | MEDLINE | ID: mdl-29202215

ABSTRACT

Venous thromboembolism (VTE) is more frequent in infants than in older children. Treatment guidelines in children are adapted from adult VTE data, but do not currently include direct oral anticoagulant use. Dabigatran etexilate (DE) use in the paediatric population with VTE therefore requires verification. We investigated the pharmacokinetic/pharmacodynamic (PK/PD) relationship, safety and tolerability of DE oral liquid formulation (OLF) in infants with VTE (aged < 12 months) who had completed standard anticoagulant treatment in an open-label, phase IIa study. Patients received a single-dose of DE OLF (based on an age- and weight-adjusted nomogram) yielding an exposure comparable to 150 mg in adults. The PK end point was plasma concentration of total dabigatran; PD end points included activated partial thromboplastin time (aPTT), ecarin clotting time (ECT) and diluted thrombin time (dTT). Safety end points included incidence of all bleeding and other adverse events (AEs). Ten patients were screened; eight entered the study (age range, 41-169 days). The geometric mean (gMean) total dabigatran plasma concentrations 2 hours post-dose (around peak concentrations) were 120 ng/mL with a geometric coefficient of variation (gCV) of 62.1%. The gMean at 12 hours post-dosing was 60.4 ng/mL (gCV 30%). PK/PD relationship was linear for ECT and dTT (R2 = 0.858 and 0.920, respectively), while total dabigatran concentration and aPTT showed a non-linear correlation. There were no deaths, treatment discontinuations or treatment-related AEs. In conclusion, DE was well tolerated without any treatment-related AEs in infants. The observed PK/PD relationships were comparable with the established profile in older patients with VTE.


Subject(s)
Antithrombins/administration & dosage , Antithrombins/pharmacokinetics , Blood Coagulation/drug effects , Dabigatran/administration & dosage , Dabigatran/pharmacokinetics , Venous Thromboembolism/drug therapy , Administration, Oral , Age Factors , Antithrombins/adverse effects , Antithrombins/blood , Blood Coagulation Tests , Dabigatran/adverse effects , Dabigatran/blood , Drug Compounding , Drug Monitoring/methods , Female , France , Hemorrhage/chemically induced , Humans , Infant , Linear Models , Male , Nonlinear Dynamics , Ontario , Pharmaceutical Solutions , Russia , Treatment Outcome , Venous Thromboembolism/blood , Venous Thromboembolism/diagnosis
8.
N Engl J Med ; 369(13): 1206-14, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-23991661

ABSTRACT

BACKGROUND: Dabigatran is an oral direct thrombin inhibitor that has been shown to be an effective alternative to warfarin in patients with atrial fibrillation. We evaluated the use of dabigatran in patients with mechanical heart valves. METHODS: In this phase 2 dose-validation study, we studied two populations of patients: those who had undergone aortic- or mitral-valve replacement within the past 7 days and those who had undergone such replacement at least 3 months earlier. Patients were randomly assigned in a 2:1 ratio to receive either dabigatran or warfarin. The selection of the initial dabigatran dose (150, 220, or 300 mg twice daily) was based on kidney function. Doses were adjusted to obtain a trough plasma level of at least 50 ng per milliliter. The warfarin dose was adjusted to obtain an international normalized ratio of 2 to 3 or 2.5 to 3.5 on the basis of thromboembolic risk. The primary end point was the trough plasma level of dabigatran. RESULTS: The trial was terminated prematurely after the enrollment of 252 patients because of an excess of thromboembolic and bleeding events among patients in the dabigatran group. In the as-treated analysis, dose adjustment or discontinuation of dabigatran was required in 52 of 162 patients (32%). Ischemic or unspecified stroke occurred in 9 patients (5%) in the dabigatran group and in no patients in the warfarin group; major bleeding occurred in 7 patients (4%) and 2 patients (2%), respectively. All patients with major bleeding had pericardial bleeding. CONCLUSIONS: The use of dabigatran in patients with mechanical heart valves was associated with increased rates of thromboembolic and bleeding complications, as compared with warfarin, thus showing no benefit and an excess risk. (Funded by Boehringer Ingelheim; ClinicalTrials.gov numbers, NCT01452347 and NCT01505881.).


Subject(s)
Anticoagulants/administration & dosage , Atrial Fibrillation/drug therapy , Benzimidazoles/administration & dosage , Heart Valve Prosthesis , Stroke/prevention & control , Thromboembolism/prevention & control , Warfarin/administration & dosage , beta-Alanine/analogs & derivatives , Aged , Anticoagulants/adverse effects , Benzimidazoles/adverse effects , Dabigatran , Female , Hemorrhage/chemically induced , Humans , Male , Middle Aged , Thromboembolism/chemically induced , Warfarin/adverse effects , beta-Alanine/administration & dosage , beta-Alanine/adverse effects
9.
Clin Pharmacokinet ; 47(2): 139-46, 2008.
Article in English | MEDLINE | ID: mdl-18193920

ABSTRACT

OBJECTIVE: The objective of this analysis was to simulate the performance of oral triptan formulations with varying absorption characteristics and their impact on the onset and magnitude of the antimigraine effect using a Markov model for migraine attacks. ANALYSIS: Sumatriptan pharmacokinetic data were obtained from clinical pharmacology studies in which marketed solid formulations were administered. Based on a population pharmacokinetic model, mean concentration-time profiles were generated by varying the absorption rate constant and lag time. Subsequently, the simulated profiles were evaluated in a disease model of migraine to predict the onset and duration of the effect (the pain-free, pain-relief response). RESULTS: Based on a therapeutic dose of 50 mg of sumatriptan, a maximum gain in the pain-free response of 12% was achieved with an increased absorption rate. This gain in the response was reached approximately 0.5 hours after administration. A decrease only in the lag time with respect to the currently available formulations (i.e. 0.24 hours) resulted in a maximum gain of 5% in the pain-free response, which in contrast may not be interpreted as clinically relevant. CONCLUSION: Model-based predictions suggest that increases in the absorption rate of the currently marketed oral formulation of sumatriptan result in a gain in the pain-free response that is both clinically and statistically relevant.


Subject(s)
Migraine Disorders/drug therapy , Serotonin Receptor Agonists/pharmacokinetics , Sumatriptan/pharmacokinetics , Administration, Oral , Biological Availability , Clinical Trials as Topic , Computer Simulation , Databases, Factual , Dose-Response Relationship, Drug , Humans , Intestinal Absorption , Markov Chains , Models, Biological , Serotonin 5-HT1 Receptor Agonists , Serotonin Receptor Agonists/administration & dosage , Serotonin Receptor Agonists/therapeutic use , Software , Sumatriptan/administration & dosage , Sumatriptan/therapeutic use , Time Factors , Treatment Outcome
10.
Stat Med ; 26(22): 4163-78, 2007 Sep 30.
Article in English | MEDLINE | ID: mdl-17385187

ABSTRACT

Markov-type models have been used in the analysis of disease progression. Although standard errors of model parameters are usually estimated, available software often does not permit the construction of confidence intervals around predictions of the dependent or response variable. A method is presented to calculate means and confidence intervals of model-predicted responses in time governed by a non-homogeneous hidden Markov model in continuous time. The Kolmogorov equations serve as the basis for the calculations. The method is realised in S-Plus and is applied to the prediction of headache responses in clinical studies of anti-migraine treatment. Means and confidence intervals are calculated by numerically solving differential equations that are non-linear in the explanatory variable. Results indicate that uncertainty on predicted drug responses is larger than that on predicted placebo responses and that pain-free responses are less precisely predicted than pain-relief responses. This is due to the uncertainty in the drug-specific parameters which is not present in predicted placebo responses.


Subject(s)
Markov Chains , Migraine Disorders , Biometry , Clinical Trials as Topic , Confidence Intervals , Humans , Migraine Disorders/classification , Migraine Disorders/physiopathology , Pain/drug therapy , Sumatriptan/therapeutic use
11.
Br J Clin Pharmacol ; 62(5): 591-600, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17061965

ABSTRACT

AIMS: Currently, direct comparisons between 5-HT(1B/d) receptor agonists are used to assess differences and similarities in antimigraine response. Such comparisons depend on the selected sampling time and do not allow evaluation of entire response profiles. A thorough evaluation of drug properties requires that the time course of the response be taken into account. In this investigation we show the advantages of a model-based approach to compare the efficacy of two triptans (sumatriptan vs. naratriptan). METHODS: A Markov model was used to describe the course of a migraine attack over three clinically identified stages. Drug effects were modelled as concentration-dependent increases in transition rates and were parameterised as potency (EC(50)) and maximum effect (E(max)). Parameters were estimated using headache measurements from efficacy studies. Model estimates were then used to compare the pharmacodynamics of the two drugs in a time-independent manner. RESULTS: Efficacy parameters could be derived, allowing for comparison between compounds. The potency ratio (EC50(suma)/EC50(nara)) for headache relief was 3.3 (0.9, 12). The ratio of maximum effects (Emax(suma)/Emax(nara)) for this endpoint was 0.74 (0.55, 0.97). To interpret these efficacy measures and explore their value for the development of antimigraine drugs, results were evaluated against the reported in vitro potency at 5-HT(1B) and 5-HT(1D) receptors. CONCLUSIONS: Comparison of the effects of two or more drugs based on preset sampling times does not allow proper assessment of the antimigraine properties in vivo. Disease dynamics must be considered to evaluate treatment response adequately and optimise the dosing regimen in migraine.


Subject(s)
Migraine Disorders/drug therapy , Piperidines/pharmacokinetics , Serotonin Receptor Agonists/pharmacokinetics , Sumatriptan/pharmacokinetics , Tryptamines/pharmacokinetics , Dose-Response Relationship, Drug , Humans , Markov Chains , Models, Biological , Piperidines/administration & dosage , Serotonin Receptor Agonists/administration & dosage , Sumatriptan/administration & dosage , Treatment Outcome , Tryptamines/administration & dosage
12.
J Clin Oncol ; 20(24): 4713-21, 2002 Dec 15.
Article in English | MEDLINE | ID: mdl-12488418

ABSTRACT

PURPOSE: To develop a semimechanistic pharmacokinetic-pharmacodynamic model describing chemotherapy-induced myelosuppression through drug-specific parameters and system-related parameters, which are common to all drugs. PATIENTS AND METHODS: Patient leukocyte and neutrophil data after administration of docetaxel, paclitaxel, and etoposide were used to develop the model, which was also applied to myelosuppression data from 2'-deoxy-2'-methylidenecytidine (DMDC), irinotecan (CPT-11), and vinflunine administrations. The model consisted of a proliferating compartment that was sensitive to drugs, three transit compartments that represented maturation, and a compartment of circulating blood cells. Three system-related parameters were estimated: baseline, mean transit time, and a feedback parameter. Drug concentration-time profiles affected the proliferation of sensitive cells by either an inhibitory linear model or an inhibitory E(max) model. To evaluate the model, system-related parameters were fixed to the same values for all drugs, which were based on the results from the estimations, and only drug-specific parameters were estimated. All modeling was performed using NONMEM software. RESULTS: For all investigated drugs, the model successfully described myelosuppression. Consecutive courses and different schedules of administration were also well characterized. Similar system-related parameter estimates were obtained for the different drugs and also for leukocytes compared with neutrophils. In addition, when system-related parameters were fixed, the model well characterized chemotherapy-induced myelosuppression for the different drugs. CONCLUSION: This model predicted myelosuppression after administration of one of several different chemotherapeutic drugs. In addition, with fixed system-related parameters to proposed values, and only drug-related parameters estimated, myelosuppression can be predicted. We propose that this model can be a useful tool in the development of anticancer drugs and therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Marrow/drug effects , Camptothecin/analogs & derivatives , Deoxycytidine/analogs & derivatives , Models, Biological , Paclitaxel/analogs & derivatives , Taxoids , Vinblastine/analogs & derivatives , Camptothecin/pharmacology , Deoxycytidine/pharmacology , Docetaxel , Etoposide/pharmacology , Humans , Irinotecan , Leukocytes/drug effects , Neutrophils/drug effects , Paclitaxel/pharmacology , Vinblastine/pharmacology
13.
J Pharmacol Exp Ther ; 303(3): 1130-7, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12438536

ABSTRACT

The objective of this investigation was to compare the in vivo potency and intrinsic activity of buspirone and its metabolite 1-(2-pyrimidinyl)-piperazine (1-PP) in rats by pharmacokinetic-pharmacodynamic modeling. Following intravenous administration of buspirone (5 or 15 mg/kg in 15 min) or 1-PP (10 mg/kg in 15 min), the time course of the concentrations in blood were determined in conjunction with the effect on body temperature. The pharmacokinetics of buspirone and 1-PP were analyzed based on a two-compartment model with metabolite formation. Differences in the pharmacokinetics of buspirone and 1-PP were observed with values for clearance of 13.1 and 8.2 ml/min and for terminal elimination half-life of 25 and 79 min, respectively. At least 26% of the administered dose of buspirone was converted into 1-PP. Complex hypothermic effects versus time profiles were observed, which were successfully analyzed on the basis of a physiological indirect response model with set-point control. Both buspirone and 1-PP behaved as partial agonists relative to R-(+)-8-hydroxy-2-(di-n-propylamino)tetralin (R-8-OH-DPAT) with values of the intrinsic activity of 0.465 and 0.312, respectively. Differences in the potency were observed with values of 17.6 and 304 ng/ml for buspirone and 1-PP, respectively. The results of this analysis show that buspirone and 1-PP behave as partial 5-hydroxytryptamine(1A) agonists in vivo and that following intravenous administration the amount of 1-PP formed is too small to contribute to the hypothermic effect.


Subject(s)
Buspirone/analogs & derivatives , Buspirone/pharmacology , Buspirone/pharmacokinetics , Models, Biological , Animals , Body Temperature/drug effects , Body Temperature/physiology , Buspirone/blood , Male , Models, Chemical , Rats , Rats, Wistar , Receptors, Serotonin/physiology , Receptors, Serotonin, 5-HT1
14.
J Pharmacol Exp Ther ; 300(1): 330-8, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11752133

ABSTRACT

The objective of this investigation was to characterize quantitatively the pharmacodynamic interaction between N-[2-[4-(2-methoxyphenyl)-1-piperazinyl]ethyl]-N-2-pyridinyl-cyclohexanecarboxamide (WAY-100,635) and R-(+)-8-hydroxy-2-(di-n-propylamino)tetralin (R-8-OH-DPAT) in vivo. The 8-OH-DPAT-induced change in body temperature was used as a pharmacodynamic endpoint. Four groups of rats each received 1 mg/kg 8-OH-DPAT in 5 min during computer-controlled infusions of physiological saline or WAY-100,635, targeted at steady-state concentrations of 20, 85, and 170 ng/ml. Body temperature was monitored continuously with a telemetric system, and frequent blood samples were obtained to determine the pharmacokinetics of both drugs. Large differences in pharmacokinetics were observed between WAY-100,635 and R-8-OH-DPAT, reflected in values of the terminal elimination half-life of 33 and 143 min, respectively. Infusion of WAY-100,635 had no influence on the pharmacokinetics of R-8-OH-DPAT. With regard to the pharmacodynamics, clear antagonism of the R-8-OH-DPAT-induced hypothermia was observed. The complex pharmacological effect versus time profiles of R-8-OH-DPAT were analyzed on the basis of an indirect physiological response model with set point control coupled to a competitive interaction model for an agonist and antagonist acting at a common receptor. This model converged, yielding precise estimates of the pharmacodynamic parameters of both WAY-100,635 and R-8-OH-DPAT, which were independent of the infusion rate of WAY-100,635. The estimated in vivo binding constant of WAY-100,635 was 0.98 ng/ml (2.3 nM), which is very similar to the reported value from in vitro receptor binding assays. The findings of this investigation show that, in contrast to earlier reports in the literature, WAY 100,635 behaves as a pure competitive antagonist at the 5-hydroxytryptamine(1A) receptor in vivo.


Subject(s)
8-Hydroxy-2-(di-n-propylamino)tetralin/antagonists & inhibitors , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Hypothermia/chemically induced , Piperazines/antagonists & inhibitors , Piperazines/pharmacology , Pyridines/antagonists & inhibitors , Pyridines/pharmacology , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Algorithms , Animals , Binding, Competitive/drug effects , Body Temperature/drug effects , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Drug Interactions , Infusions, Intravenous , Kinetics , Male , Models, Biological , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...