Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Article in English | MEDLINE | ID: mdl-31182538

ABSTRACT

Ibuprofen and indomethacin are commonly used to induce ductus arteriosus closure in preterm neonates. Our group previously reported that ibuprofen decreased vancomycin clearance by 16%. In this study, we quantified the impact of indomethacin coadministration on vancomycin clearance by extending our vancomycin population pharmacokinetic model with a data set containing vancomycin concentrations measured in preterm neonates comedicated with indomethacin. The modeling data set includes concentration-time data of vancomycin administered alone or in combination with either ibuprofen or indomethacin collected in the neonatal intensive care units of UZ Leuven (Leuven, Belgium) and São Francisco Xavier Hospital (Lisbon, Portugal). The derived vancomycin pharmacokinetic model was subsequently used to propose dose adjustments that yield effective vancomycin exposure (i.e., area under the concentration-time curve from 0 to 24 h [AUC0-24] between 300 to 550 mg·h/liter, with a probability of <0.1 of subtherapeutic exposure) in preterm neonates with patent ductus arteriosus. We found that indomethacin coadministration reduced vancomycin clearance by 55%. Model simulations showed that the most recent vancomycin dosing regimen, which was based on an externally validated model, requires 20% and 60% decreases of the loading and maintenance doses of vancomycin, respectively, when aiming for optimized exposure in the neonatal population. By analyzing vancomycin data from preterm neonates comedicated with indomethacin, we found a substantial decrease in vancomycin clearance of 55% versus a previously reported 16% for ibuprofen. This decrease in clearance impacts vancomycin dosing, and we anticipate that other drugs eliminated by glomerular filtration are likely to be affected to a similar extent as vancomycin.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Ductus Arteriosus, Patent/drug therapy , Ibuprofen/therapeutic use , Indomethacin/therapeutic use , Vancomycin/pharmacokinetics , Vancomycin/therapeutic use , Adult , Female , Humans , Infant, Newborn , Male , Monte Carlo Method , Pregnancy , Young Adult
2.
AAPS J ; 21(3): 38, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30850923

ABSTRACT

Previous research showed that scaling drug clearance from adults to children based on body weight alone is not accurate for all hepatically cleared drugs in very young children. This study systematically assesses the accuracy of scaling methods that, in addition to body weight, also take age-based variables into account for drugs undergoing hepatic metabolism in children younger than five years, namely scaling with (1) a body weight-based function using an age-dependent exponent (ADE) and (2) a body weight-based function with fixed exponent of 0.75 (AS0.75) combined with isoenzyme maturation functions (MFPBPK) similar to those implemented in physiologically based pharmacokinetic (PBPK) models (AS0.75 + MFPBPK). A PBPK-based simulation workflow was used, including hypothetical drugs with a wide range of properties and metabolized by different isoenzymes. Adult clearance values were scaled to seven typical children between one day and four years. Prediction errors of ± 50% were considered reasonably accurate. Isoenzyme maturation was found to be an important driver of changes in hepatic metabolic clearance in children younger than five years, which prevents the systematic accuracy of ADE scaling. AS0.75 + MFPBPK, when accounting for maturation of isoenzymes and microsomal protein per gram of liver (MPPGL), can reasonably accurately scale hepatic metabolic clearance for all low and intermediate extraction ratio drugs except for drugs binding to alpha-1-acid glycoprotein in neonates. As differences in the impact of changes in system-specific parameters on drugs with different properties yield differences in clearance ontogeny, it is unlikely that for the remaining drugs, scaling methods that do not take drug properties into account will be systematically accurate.


Subject(s)
Liver/metabolism , Metabolic Clearance Rate , Models, Biological , Patient-Specific Modeling , Adult , Age Factors , Body Weight , Child, Preschool , Humans , Infant , Infant, Newborn
3.
J Clin Pharmacol ; 59(6): 847-855, 2019 06.
Article in English | MEDLINE | ID: mdl-30633373

ABSTRACT

Children undergoing cardiac surgery often receive acetaminophen (paracetamol) as part of their postoperative pain treatment. To date, there is no information on the pharmacokinetics (PK) of acetaminophen in this special population, even though differences, as a result of altered hemodynamics and/or use of cardiopulmonary bypass, may be anticipated. Therefore, the aim of this study was to investigate the PK of intravenous acetaminophen in children after cardiac surgery with cardiopulmonary bypass. In the study, both children with and without Down syndrome were included. A population PK analysis, using NONMEM 7.2, was performed based on 161 concentrations of acetaminophen, acetaminophen sulfate, acetaminophen glucuronide, and oxidative metabolites from 17 children with Down syndrome and 13 children without Down syndrome of a previously published study (median age, 177 days [range, 92-944], body weight, 6.1 kg [4.0-12.9]). All children received 3 intravenous acetaminophen doses of 7.5 mg/kg (<10 kg) or 15 mg/kg (≥10 kg) at 8-hour intervals after cardiac surgery. For acetaminophen and its metabolites, 1-compartment models were identified. Clearance of acetaminophen and metabolites increased linearly with body weight. Acetaminophen clearance in a typical child of 6.1 kg is 0.96 L/h and volume of distribution 7.96 L. Down syndrome did not statistically significantly impact any of the PK parameters for acetaminophen, nor did any other remaining covariate. When comparing the PK parameters of acetaminophen in children after cardiac surgery with cardiopulmonary bypass with those from children of the same age following noncardiac surgery reported in the literature, clearance of acetaminophen was lower and volume of distribution higher.


Subject(s)
Acetaminophen/metabolism , Acetaminophen/pharmacokinetics , Analgesics, Non-Narcotic/metabolism , Analgesics, Non-Narcotic/pharmacokinetics , Pain, Postoperative/drug therapy , Acetaminophen/administration & dosage , Administration, Intravenous , Analgesics, Non-Narcotic/administration & dosage , Biological Variation, Population/drug effects , Body Weight , Cardiac Surgical Procedures , Cardiopulmonary Bypass , Child, Preschool , Down Syndrome , Female , Humans , Infant , Infusions, Intravenous , Male , Metabolic Clearance Rate , Models, Biological , Prospective Studies
4.
AAPS J ; 19(1): 172-179, 2017 01.
Article in English | MEDLINE | ID: mdl-27634384

ABSTRACT

In this study, we report the development of the first item response theory (IRT) model within a pharmacometrics framework to characterize the disease progression in multiple sclerosis (MS), as measured by Expanded Disability Status Score (EDSS). Data were collected quarterly from a 96-week phase III clinical study by a blinder rater, involving 104,206 item-level observations from 1319 patients with relapsing-remitting MS (RRMS), treated with placebo or cladribine. Observed scores for each EDSS item were modeled describing the probability of a given score as a function of patients' (unobserved) disability using a logistic model. Longitudinal data from placebo arms were used to describe the disease progression over time, and the model was then extended to cladribine arms to characterize the drug effect. Sensitivity with respect to patient disability was calculated as Fisher information for each EDSS item, which were ranked according to the amount of information they contained. The IRT model was able to describe baseline and longitudinal EDSS data on item and total level. The final model suggested that cladribine treatment significantly slows disease-progression rate, with a 20% decrease in disease-progression rate compared to placebo, irrespective of exposure, and effects an additional exposure-dependent reduction in disability progression. Four out of eight items contained 80% of information for the given range of disabilities. This study has illustrated that IRT modeling is specifically suitable for accurate quantification of disease status and description and prediction of disease progression in phase 3 studies on RRMS, by integrating EDSS item-level data in a meaningful manner.


Subject(s)
Disability Evaluation , Models, Theoretical , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Severity of Illness Index , Cladribine/therapeutic use , Clinical Trials, Phase III as Topic , Disease Progression , Humans , Immunosuppressive Agents/therapeutic use , Logistic Models , Multiple Sclerosis, Relapsing-Remitting/drug therapy
5.
Article in English | MEDLINE | ID: mdl-23887362

ABSTRACT

To develop a maturation function for drug glucuronidation in children, that can be used in population and physiologically based modeling approaches, the physiological and physicochemical basis of a semiphysiological glucuronidation function for children was untangled using Simcyp. The results show that using the currently available in vitro data, in vivo morphine and zidovudine clearances were under predicted by the physiologically based model in Simcyp. The maturation profile was similar to the clinically observed profile except for the first 2 weeks of life, and liver size and UGT2B7 ontogeny are the physiological drivers of the maturation of glucuronidation. Physicochemical drug parameters did not affect this maturation profile, although log P and pKa influenced the absolute value of clearance. The results suggest that the semiphysiological glucuronidation function for young children can be used to predict the developmental clearance profile of other UGT2B7 substrates, though scenarios with nonlinear kinetics and high-extraction ratios require further investigation.CPT: Pharmacometrics & Systems Pharmacology (2012) 1, e10; doi:10.1038/psp.2012.12; advance online publication 10 October 2012.

6.
Article in English | MEDLINE | ID: mdl-23887364

ABSTRACT

New approaches to expedite the development of safe and effective pediatric dosing regimens and first-in-child doses are urgently needed. Model-based approaches require quantitative functions on the maturation of different metabolic pathways. In this study, we directly incorporated a pediatric covariate model for the glucuronidation of morphine into a pediatric population model for zidovudine glucuronidation. This model was compared with a reference model that gave the statistically best description of the data. Both models had adequate goodness-of-fit plots and normalized prediction distribution errors (NPDE), similar population clearance values for each individual, and a Δobjective function value of 13 points (Δ2df). This supports our hypothesis that pediatric pharmacokinetic covariate models contain system-specific information that can be used as semiphysiological functions in pediatric population models. Further research should explore the validity of the semiphysiological function for other UDP-glucuronosyltransferase 2B7 substrates and patient populations and reveal how this function can be used for pediatric physiologically based pharmacokinetic models.CPT: Pharmacometrics & Systems Pharmacology (2012) 1, e9; doi:10.1038/psp.2012.11; advance online publication 3 October 2012.

7.
J Pharm Sci ; 99(5): 2511-20, 2010 May.
Article in English | MEDLINE | ID: mdl-20020526

ABSTRACT

In contrast to the impact of plasma protein binding on pharmacokinetics, no quantitative in vivo information is available on its impact on pharmacodynamics. The pharmacokinetic-pharmacodynamic relationship of the model drug S(-)-propranolol was evaluated using mechanism-based estimations of in vivo receptor affinity (K(B,vivo)), under conditions of altered plasma protein binding resulting from different levels of alpha-1-acid glycoprotein (AGP). Male Wistar Kyoto rats with isoprenaline-induced tachycardia received an intravenous infusion of S(-)-propranolol, on postsurgery day 2 (n = 7) and day 7 (n = 8) with elevated and normal plasma protein binding, respectively. Serial blood samples were taken in parallel to heart rate measurements. AGP concentrations at 2 and 7 days postsurgery were 708 +/- 274 and 176 +/- 111 microg/mL (mean +/- SE), respectively. Using nonlinear mixed effects modeling, AGP concentration was a covariate for intercompartmental clearance for the third compartment of the pharmacokinetic model of S(-)-propranolol. Individual values of AGP concentrations ranged between 110 and 1150 microg/mL, and were associated with K(B,vivo) values of S(-)-propranolol from 7.0 to 30 nM. Using the K(B,vivo) for S(-)-propranolol with correction for average values for normal and elevated plasma protein binding, nearly identical values were found. This confirms, strictly quantitative, earlier indications that plasma protein binding restricts the pharmacodynamics of S(-)-propranolol.


Subject(s)
Adrenergic beta-Antagonists/blood , Glycoproteins/blood , Heart Rate/drug effects , Propranolol/blood , Adrenergic beta-Antagonists/metabolism , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Animals , Blood Proteins/metabolism , Chromatography, High Pressure Liquid , Glycoproteins/metabolism , Infusions, Intravenous , Isoproterenol , Male , Orosomucoid , Propranolol/metabolism , Propranolol/pharmacology , Propranolol/therapeutic use , Protein Binding , Rats , Rats, Inbred WKY , Tachycardia/blood , Tachycardia/chemically induced , Tachycardia/drug therapy
8.
J Pharm Sci ; 98(10): 3816-28, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19117045

ABSTRACT

The objective of this investigation was to examine in a systematic manner the influence of plasma protein binding on in vivo pharmacodynamics. Comparative pharmacokinetic-pharmacodynamic studies with four beta blockers were performed in conscious rats, using heart rate under isoprenaline-induced tachycardia as a pharmacodynamic endpoint. A recently proposed mechanism-based agonist-antagonist interaction model was used to obtain in vivo estimates of receptor affinities (K(B,vivo)). These values were compared with in vitro affinities (K(B,vitro)) on the basis of both total and free drug concentrations. For the total drug concentrations, the K(B,vivo) estimates were 26, 13, 6.5 and 0.89 nM for S(-)-atenolol, S(-)-propranolol, S(-)-metoprolol and timolol. The K(B,vivo) estimates on the basis of the free concentrations were 25, 2.0, 5.2 and 0.56 nM, respectively. The K(B,vivo)-K(B,vitro) correlation for total drug concentrations clearly deviated from the line of identity, especially for the most highly bound drug S(-)-propranolol (ratio K(B,vivo)/K(B,vitro) approximately 6.8). For the free drug, the correlation approximated the line of identity. Using this model, for beta-blockers the free plasma concentration appears to be the best predictor of in vivo pharmacodynamics.


Subject(s)
Adrenergic beta-Antagonists/pharmacokinetics , Blood Proteins/metabolism , Receptors, Adrenergic, beta/metabolism , Adrenergic beta-Agonists/blood , Adrenergic beta-Agonists/pharmacokinetics , Adrenergic beta-Antagonists/blood , Adrenergic beta-Antagonists/pharmacology , Algorithms , Animals , Dose-Response Relationship, Drug , Heart Rate/drug effects , Isoproterenol/blood , Isoproterenol/pharmacokinetics , Male , Metoprolol/blood , Metoprolol/pharmacokinetics , Metoprolol/pharmacology , Models, Biological , Propranolol/blood , Propranolol/pharmacokinetics , Propranolol/pharmacology , Protein Binding , Rats , Rats, Inbred WKY , Timolol/blood , Timolol/pharmacokinetics , Timolol/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...