Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Achiel Van Peer is active.

Publication


Featured researches published by Achiel Van Peer.


Clinical Pharmacology & Therapeutics | 1993

Pharmacokinetics of the novel antipsychotic agent risperidone and the prolactin response in healthy subjects

M. L. Huang; Achiel Van Peer; R. Woestenborghs; Roland De Coster; Jos Heykants; Arno A I Jansen; Zbigniew Zylicz; Hendrik W Visscher; Jan H G Jonkman

The pharmacokinetics of a novel antipsychotic agent, risperidone, and the prolactin response were studied in 12 dextromethorphan‐phenotyped healthy men after administration of 1 mg risperidone intravenously, intramuscularly, and orally. The formation of the equipotent major metabolite, 9‐hydroxyrisperidone, exhibited CYP2D6‐related polymorphism. The plasma area under the concentration—time curve from time zero to infinity ratio of 9‐hydroxyrisperidone to risperidone averaged 3 (intravenous and intramuscular) and 6 (oral administration) in the extensive metabolizers and 0.2 in the poor metabolizers. Risperidone half‐life was about 3 hours in extensive metabolizers and 22 hours in poor metabolizers. Risperidone absolute oral bioavailability was 66%. The pharmacokinetics of the active moiety (risperidone plus 9‐hydroxyrisperidone) varied little among subjects (mean terminal half‐life, 20 ± 21/2 hours; absolute oral and intramuscular bioavailability, 100%). The prolactin response correlated best with the plasma active moiety, which showed little hysteresis. It is concluded that risperidone metabolic polymorphism on increased plasma prolactin is minimal and that the active moiety is clinically relevant.


Anesthesiology | 1995

Linearity of Pharmacokinetics and Model Estimation of Sufentanil

Elisabeth Gepts; Steven L. Shafer; F. Camu; Donald R. Stanski; R. Woestenborghs; Achiel Van Peer; Jos Heykants

Background The pharmacokinetic profiles of sufentanil available in the literature are conflicting because of methodologic differences. Length of sampling and assay sensitivity are key factors involved in accurately estimating the volumes of distribution, clearances, and elimination phase. The unit disposition function of increasing doses of sufentanil were investigated and the influence of dose administered on the linearity of pharmacokinetics was assessed.


European Journal of Pharmaceutical Sciences | 2014

In vivo methods for drug absorption - comparative physiologies, model selection, correlations with in vitro methods (IVIVC), and applications for formulation/API/excipient characterization including food effects.

Erik Sjögren; Bertil Abrahamsson; Patrick Augustijns; Dieter Becker; Michael B. Bolger; Marcus E. Brewster; Joachim Brouwers; Talia Flanagan; Matthew D. Harwood; Christian Heinen; René Holm; Hans-Paul Juretschke; Marlies Kubbinga; Anders Lindahl; Viera Lukacova; Uwe Münster; Sibylle Neuhoff; Mai Anh Nguyen; Achiel Van Peer; Christos Reppas; Amin Rostami Hodjegan; Christer Tannergren; Werner Weitschies; Clive G. Wilson; Patricia Zane; Hans Lennernäs; Peter Langguth

This review summarizes the current knowledge on anatomy and physiology of the human gastrointestinal tract in comparison with that of common laboratory animals (dog, pig, rat and mouse) with emphasis on in vivo methods for testing and prediction of oral dosage form performance. A wide range of factors and methods are considered in addition, such as imaging methods, perfusion models, models for predicting segmental/regional absorption, in vitro in vivo correlations as well as models to investigate the effects of excipients and the role of food on drug absorption. One goal of the authors was to clearly identify the gaps in todays knowledge in order to stimulate further work on refining the existing in vivo models and demonstrate their usefulness in drug formulation and product performance testing.


European Journal of Pharmaceutical Sciences | 2001

Role of modelling and simulation in Phase I drug development

Leon Aarons; Mats O. Karlsson; Ferdinand Rombout; Jean-Louis Steimer; Achiel Van Peer

Although the use of pharmacokinetic/pharmacodynamic modelling and simulation (M&S) in drug development has increased during the last decade, this has most notably occurred in patient studies using the population approach. The role of M&S in Phase I, although of longer history, does not presently have the same impact on drug development. However, trends such as the increased use of biomarkers and clinical trial simulation as well as adoption of the learn/confirm concept can be expected to increase the importance of modelling in Phase I. To help identify the role of M&S, its main advantages and the obstacles to its rational use, an expert meeting was organised by COST B15 in Brussels, January 10-11, 2000. This article presents the views expressed at that meeting. Although it is clear that M&S occurs in only a minority of Phase I clinical trials, it is used for a large number of different purposes. In particular, M&S is considered valuable in the following situations: censoring because of assay limitation, characterisation of non-linearity, estimating exposure-response relationship, combined analyses, sparse sampling studies, special population studies, integrating PK/PD knowledge for decision making, simulation of Phase II trials, predicting multiple dose profile from single dose, bridging studies and formulation development. One or more of the following characteristics of M&S activities are often present and severely impede its successful integration into clinical drug development: lack of trained personnel, lack of protocol and/or analysis plan, absence of pre-specified objectives, no timelines or budget, low priority, inadequate reporting, no quality assurance of the modelling process and no evaluation of cost-benefit. The early clinical drug development phase is changing and if these implementation aspects can be appropriately addressed, M&S can fulfill an important role in reshaping the early trials by more effective extraction of information from studies, better integration of knowledge across studies and more precise predictions of trial outcome, thereby allowing more informed decision making.


The Journal of Clinical Pharmacology | 1998

Fentanyl Delivery from an Electrotransport System: Delivery is a Function of Total Current, Not Duration of Current

Suneel K. Gupta; Keith J. Bernstein; Henk Noorduin; Achiel Van Peer; Gayatri Sathyan; Ron Haak

This open‐label, parallel study of 28 men was conducted to evaluate the pharmacokinetics and safety of fentanyl delivered by the E‐TRANS (fentanyl) electrotransport transdermal system (ALZA Corporation, Palo Alto, CA). The E‐TRANS (fentanyl) system provided electrically assisted, transdermal, continuous delivery of fentanyl. Treatments consisted of no current (group A); a constant current of 100 μA for 26 hours plus 4 additional doses at varying currents for varying times during hour 25 (groups B, C, D); a constant current of 100 μA for 26 hours plus 4 additional doses at 1,200 μA over 2.5 minutes during hour 1 (group E); or 500 μA for 0.5 hours and 100 μA for 3.5 hours (group F). No fentanyl was detected in serum when no current had been applied. Mean serum fentanyl concentrations were similar regardless of current duration during hour 25 (treatments B, C, D). Increases in mean serum fentanyl concentrations were significantly lower during additional dosing for treatment E compared with treatments B, C, and D. Serum fentanyl concentrations sufficient for analgesia (1–3 ng/mL) were attained in treatments using the E‐TRANS (fentanyl) system with basal current of 100 μA for 26 hours. There were no safety issues after treatment with E‐TRANS (fentanyl) system with concurrent opioid antagonist (naltrexone) administration. The only adverse event requiring treatment was a headache (n = 1). The majority of subjects had no or barely perceptible erythema at the application site 24 hours after system removal. Application of E‐TRANS (fentanyl) resulted in therapeutically significant serum fentanyl concentrations over a range of applied currents. Overall serum fentanyl concentrations were higher when the skin had been primed by constant‐current fentanyl delivery.


Biopharmaceutics & Drug Disposition | 2012

From preclinical to human – prediction of oral absorption and drug–drug interaction potential using physiologically based pharmacokinetic (PBPK) modeling approach in an industrial setting: a workflow by using case example

Vikash Sinha; Jan Snoeys; Nancy Van Osselaer; Achiel Van Peer; Claire Mackie; Donald Heald

A case example is presented in which the physiologically based modeling approach has been used to model the absorption of a lipophilic BCS Class II compound predominantly metabolized by CYP3A4, and to assess the interplay of absorption related parameters with the drug–drug interaction (DDI) potential.


Clinical Pharmacology & Therapeutics | 1984

Alfentanil kinetics in the elderly.

Henk Helmers; Achiel Van Peer; R. Woestenborghs; Henk Noorduin; Jos Heykants

Alfentanil disposition after an intravenous bolus of 50 µg/kg was followed in 15 elderly surgical patients and was compared to that in nine young adults. A two‐compartment open model described alfentanil disappearance from plasma. Apparent volumes of distribution of the central compartment (201 and 211 ml/kg; X̄), at steady state (460 and 543 ml/kg), and of the AUC (746 and 722 ml/kg) in young adults and in elderly subjects did not differ. Plasma clearance was lower in elderly subjects (4.4 ml/min/kg) than in young adults (6.5 ml/min/kg), whereas terminal plasma t½ was longer in the elderly patients (137 and 83 min). Alfentanil dosage should therefore be reduced in elderly patients when large single doses, multiple doses, or long‐term infusions are required.


Pharmaceutical Research | 2014

Mechanistic Understanding of Brain Drug Disposition to Optimize the Selection of Potential Neurotherapeutics in Drug Discovery

Irena Loryan; Vikash Sinha; Claire Mackie; Achiel Van Peer; Wilhelmus Drinkenburg; An Vermeulen; Denise Morrison; Mario Monshouwer; Donald Heald; Margareta Hammarlund-Udenaes

ABSTRACTPurposeThe current project was undertaken with the aim to propose and test an in-depth integrative analysis of neuropharmacokinetic (neuroPK) properties of new chemical entities (NCEs), thereby optimizing the routine of evaluation and selection of novel neurotherapeutics.MethodsForty compounds covering a wide range of physicochemical properties and various CNS targets were investigated. The combinatory mapping approach was used for the assessment of the extent of blood-brain and cellular barriers transport via estimation of unbound-compound brain (Kp,uu,brain) and cell (Kp,uu,cell) partitioning coefficients. Intra-brain distribution was evaluated using the brain slice method. Intra- and sub-cellular distribution was estimated via calculation of unbound-drug cytosolic and lysosomal partitioning coefficients.ResultsAssessment of Kp,uu,brain revealed extensive variability in the brain penetration properties across compounds, with a prevalence of compounds actively effluxed at the blood-brain barrier. Kp,uu,cell was valuable for identification of compounds with a tendency to accumulate intracellularly. Prediction of cytosolic and lysosomal partitioning provided insight into the subcellular accumulation. Integration of the neuroPK parameters with pharmacodynamic readouts demonstrated the value of the proposed approach in the evaluation of target engagement and NCE selection.ConclusionsWith the rather easily-performed combinatory mapping approach, it was possible to provide quantitative information supporting the decision making in the drug discovery setting.


Molecular Pharmaceutics | 2015

Molecular properties determining unbound intracellular and extracellular brain exposure of CNS drug candidates

Irena Loryan; Vikash Sinha; Claire Mackie; Achiel Van Peer; Wilhelmus Drinkenburg; An Vermeulen; Donald Heald; Margareta Hammarlund-Udenaes; Carola M. Wassvik

In the present work we sought to gain a mechanistic understanding of the physicochemical properties that influence the transport of unbound drug across the blood-brain barrier (BBB) as well as the intra- and extracellular drug exposure in the brain. Interpretable molecular descriptors that significantly contribute to the three key neuropharmacokinetic properties related to BBB drug transport (Kp,uu,brain), intracellular accumulation (Kp,uu,cell), and binding and distribution in the brain (Vu,brain) for a set of 40 compounds were identified using partial least-squares (PLS) analysis. The tailoring of drug properties for improved brain exposure includes decreasing the polarity and/or hydrogen bonding capacity. The design of CNS drug candidates with intracellular targets may benefit from an increase in basicity and/or the number of hydrogen bond donors. Applying this knowledge in drug discovery chemistry programs will allow designing compounds with more desirable CNS pharmacokinetic properties.


The Journal of Clinical Pharmacology | 2002

Galantamine pharmacokinetics, safety, and tolerability profiles are similar in healthy Caucasian and Japanese subjects

Qinying Zhao; Martin Brett; Nancy Van Osselaer; Fenglei Huang; Alain Raoult; Achiel Van Peer; Tom Verhaeghe; Rita Hust

The aim of this study was to compare the pharmacokinetics of galantamine in healthy Japanese and Caucasian subjects and assess the safety and tolerability of galantamine in both ethnic groups. Parallel groups of healthy Japanese (n = 13; 6 males and 7females) and Caucasian (n = 12; 6males and 6 females) subjects matched for weight and age received single oral doses of galantamine 4 mg, or galantamine 8 mg, or placebo in a double‐blind, three‐way crossover trial according to a randomized dosing schedule. Concentrations of galantamine and norgalantamine were determined in plasma and urine samples taken up to 48 and 24 hours after dosing, respectively. Safety and tolerability were monitored throughout the trial by recording adverse events, laboratoryr tests, and cardiovascular parameters. The mean plasma concentration‐time profiles of galantamine were very similar after single doses of galantamine (4 and 8 mg), and there was an approximate dose proportionality of galantamine pharmacokinetic parameters in both Caucasian and Japanese ethnic groups. The mean (± SD) pharmacokinetic parameters in the two ethnic groups did not show any clinically relevant differences. The ratios for the area under the plasma‐concentration curve from time zero to infinity (AUC0∞) in Japanese. Caucasian subjects with 4 and 8 mg doses were 103% (90% confidence interval [CI] = 92–116) and 107% (90% CI = 94–121), respectively. Ratios for maximum plasma concentration (Cmax) values were 107% (90% CI= 90–127) and 108% (90% CI =95–123), respectively These ratios and associated 90% CIs were within the 80% to 125% range limit of bioequivalence. Analysis of variance (ANCWA) showed that these ratio values demonstrated no statistically significant difference between the two ethnic groups. There was no overt difference in the adverse event profile in Japanese subjects compared with Caucasian subjects. There were no serious adverse events, and no subjects discontinued from the study because of adverse events. No consistent or clinically relevant pattern of blood chemistry, hematology, or cardiovascular changes was seen. These results suggest that the pharmacokinetic profiles of galantamine after single‐dose administration are not statistically significantly different between Caucasian and Japanese groups. Galantamine was well tolerated. The safety and tolerability of galantamine were very similar in the two ethnic groups.

Collaboration


Dive into the Achiel Van Peer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leon Aarons

University of Manchester

View shared research outputs
Researchain Logo
Decentralizing Knowledge