Network


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

Hotspot


Dive into the research topics where Imke H. Bartelink is active.

Publication


Featured researches published by Imke H. Bartelink.


Clinical Pharmacokinectics | 2006

Guidelines on Paediatric Dosing on the Basis of Developmental Physiology and Pharmacokinetic Considerations

Imke H. Bartelink; Carin M. A. Rademaker; Alfred F. A. M. Schobben; John N. van den Anker

The approach to paediatric drug dosing needs to be based on the physiological characteristics of the child and the pharmacokinetic parameters of the drug. This review summarises the current knowledge on developmental changes in absorption, distribution, metabolism and excretion and combines this knowledge with in vivo and in vitro pharmacokinetic data that are currently available. In addition, dosage adjustments based on practical problems, such as child-friendly formulations and feeding regimens, disease state, genetic make-up and environmental influences are presented.Modification of a dosage based on absorption, depends on the route of absorption, the physico chemical properties of the drug and the age of the child. For oral drug absorption, a distinction should be made between the very young and children over a few weeks old. In the latter case, it is likely that practical considerations, like appropriate formulations, have much greater relevance to oral drug absorption.The volume of distribution (Vd) may be altered in children. Hydrophilic drugs with a high Vd in adults should be normalised to bodyweight in young children (age <2 years), whereas hydrophilic drugs with a low Vd in adults should be normalised to body surface area (BSA) in these children. For drugs that are metabolised by the liver, the effect of the Vd becomes apparent in children <2 months of age. In general, only the first dose should be based on the Vd subsequent doses should be determined by the clearance. Pharmacokinetic studies on renal and liver function clarify that a distinction should be made between maturation and growth of the organs. After the maturation process has finished, the main influences on the clearance of drugs are growth and changes in blood flow of the liver and kidney. Drugs that are primarily metabolised by the liver should be administered with extreme care until the age of 2 months. Modification of dosing should be based on response and on therapeutic drug monitoring. At the age of 2–6 months, a general guideline based on bodyweight may be used. After 6 months of age, BSA is a good marker as a basis for drug dosing. However, even at this age, drugs that are primarily metabolised by cytochrome P450 2D6 and uridine diphosphate glucuronosyltransferase should be normalised to bodyweight.In the first 2 years of life, the renal excretion rate should be determined by markers of renal function, such as serum creatinine and p-aminohippuric acid clearance. A dosage guideline for drugs that are significantly excreted by the kidney should be based on the determination of renal function in first 2 years of life. After maturation, the dose should be normalised to BSA.These guidelines are intended to be used in clinical practice and to form a basis for more research. The integration of these guidelines, and combining them with pharmacodynamic effects, should be considered and could form a basis for further study.


Biology of Blood and Marrow Transplantation | 2009

Association between Busulfan Exposure and Outcome in Children Receiving Intravenous Busulfan before Hematologic Stem Cell Transplantation

Imke H. Bartelink; Robert G.M. Bredius; Marit M. Suttorp; Marc Bierings; Catherijne A. J. Knibbe; Maarten Egeler; Arjan C. Lankester; Atoine C.G. Egberts; Juliette Zwaveling; Jaap Jan Boelens

Busulfan, combined with therapeutic drug monitoring-guided dosing, is associated with higher event-free survival (EFS) rates due to fewer graft failures/relapses and lower toxicity. The optimal target area under the curve (AUC) and dosing schedule of intravenous busulfan in children undergoing hematopoietic stem cell transplantation (HSCT) remain unclear, however. We conducted a retrospective analysis of the association between busulfan exposure and clinical outcome in 102 children age 0.2 to 21 years who received busulfan 1 or 4 times daily before undergoing HSCT (46 malignant and 56 nonmalignant indications). EFS and overall survival after a median of 2 years of follow-up were 68% and 72%, respectively. EFS was optimal when the exposure of busulfan (AUC) was 78 mg x h/L (95% confidence interval=74 to 82 mg x h/L). Acute graft-versus-host disease (aGVHD) grade II-IV occurred more frequently with greater busulfan exposure. The addition of melphalan was an independent risk factor; melphalan use combined with high busulfan exposure (AUC >74 mg x h/L) was associated with high incidences of aGVHD (58%), veno-occlusive disease (66%), and mucositis grade III-IV (26%). Dosing frequency (1 or 4 times daily) was not related to any outcome. In conclusion, dose targeting of busulfan to a narrow therapeutic range was found to increase EFS in children. Adding melphalan to optimal busulfan exposure is associated with a high incidence of toxicity.


Therapeutic Drug Monitoring | 2008

Glutathione S-transferase polymorphisms are not associated with population pharmacokinetic parameters of busulfan in pediatric patients.

Juliette Zwaveling; Rogier R. Press; Robbert G. M. Bredius; Tahar van der Straaten; Jan den Hartigh; Imke H. Bartelink; Jaap Jan Boelens; Henk-Jan Guchelaar

High busulfan exposure is associated with increased toxicity, for example veno-occlusive disease, whereas low exposure results in less efficacy such as lower engraftment rates. Despite adjusting dose to body weight, interindividual variability in pharmacokinetics and thus drug exposure remained rather large. In this report, the contribution of genetic polymorphisms in the glutathione-S-transferases (GST) isozymes GSTA1, GSTM1, GSTP1, and GSTT1 to the pharmacokinetics of busulfan is studied retrospectively. Seventy-seven children, undergoing myeloablative conditioning for allogeneic hematopoietic stem cell transplantation, were treated with busulfan (Busulvex) during 4 days, receiving busulfan either in one single dose or dived in four doses every 6 hours. Genetic variants of GSTA1, GSTM1, GSTP1, and GSTT1 were determined by pyrosequencing. Pharmacokinetic parameters were estimated by using nonlinear mixed-effect modeling (NONMEM). Subsequently, a combined population pharmacokinetic-pharmacogenetic model was developed describing the pharmacokinetics of busulfan taking into account the GST polymorphisms. In the presented pediatric population, body weight appeared to be the most important covariate and explained a major part of the observed variability in the pharmacokinetics of busulfan. None of the studied polymorphisms in the genes encoding GSTA1 GSTM1, GSTP1, and GSTT1 nor combinations of genotypes were significant covariates. It was concluded that in children, variability in pharmacokinetics of busulfan could not be related to polymorphisms in GST.


Digestive Diseases and Sciences | 2010

Intestinal Permeability in Irritable Bowel Syndrome Patients: Effects of NSAIDs

Angèle P. M. Kerckhoffs; L. M. A. Akkermans; Martin De Smet; Marc G. Besselink; Falco Hietbrink; Imke H. Bartelink; Wim Busschers; M. Samsom; Willem Renooij

Intestinal permeability and the effect of NSAIDs on permeability were investigated in 14 irritable bowel syndrome (IBS) patients and 15 healthy subjects. In the study, 24-h urinary recoveries of orally administered polyethylene glycols (PEGs 400, 1500, and 4000) were not significantly different in healthy subjects and IBS patients before or after NSAID ingestion. Lactulose mannitol ratios in healthy subjects and IBS patients were not significantly different. Only time-dependent monitoring of PEG excretion showed that NSAIDs enhanced intestinal permeability for PEG 4000 in healthy subjects (Pxa0=xa00.050) and for PEGs 400, 1500, and 4000 in IBS patients (Pxa0=xa00.012, Pxa0=xa00.041, and Pxa0=xa00.012, respectively). These results show that intestinal permeability in IBS patients is not different from that in healthy subjects; NSAIDs compromise intestinal permeability in IBS patients to a greater extent than in healthy subjects, which suggests that IBS is associated with an altered response of the intestinal barrier to noxious agents.


Antimicrobial Agents and Chemotherapy | 2013

Highly Variable Plasma Concentrations of Voriconazole in Pediatric Hematopoietic Stem Cell Transplantation Patients

Imke H. Bartelink; Tom F. W. Wolfs; Martine Jonker; Marjolein de Waal; Toine C. G. Egberts; Tessa F. T. Ververs; Jaap Jan Boelens; Marc Bierings

ABSTRACT Invasive fungal infections are of great concern in pediatric hematopoietic stem cell transplantation (HSCT) recipients. Voriconazole is usually the drug of first choice for treating or preventing invasive aspergillosis. Optimum trough levels (Ctroughs) are between 1 and 5 mg/liter. It is unclear whether these levels are reached with currently advised pediatric dosing schedules. Between 2007 and 2011, 11 patients <2 years of age, 31 between 2 and 12 years, and 20 between 12 and 20 years were (prophylactically or therapeutically) treated with voriconazole in the HSCT unit of UMC Utrecht. For children <2 years of age, the dosage recommended for 2 to 12 years was used. In 34% of children who started with the recommended dose, an adequate Ctrough was reached irrespective of age or administration route. After therapeutic drug monitoring (TDM)-based dose adjustments, adequate Ctroughs were reached in 80% of the patients at median doses of 31.5 (age, <2 years), 16 (age, 2 to 12 years), and 9.4 mg/kg of body weight/day (age, >12 years) (P = 0.034). The intrapatient variability in Ctrough ranged between 1 and 238%. Voriconazole was discontinued in six patients due to toxicity. These patients had a median Ctrough of 0.5 mg/liter at the initial dose (ranging from 0.5 to 2.6 mg/liter), and a medium maximal concentration of 4 mg/liter was reached. Inter- and intrapatient variability is a major concern in voriconazole treatment and necessitates therapeutic drug monitoring of dosing, especially in young children.


Therapeutic Drug Monitoring | 2012

Predictive Performance of a Busulfan Pharmacokinetic Model in Children and Young Adults

Imke H. Bartelink; Charlotte van Kesteren; Jaap Jan Boelens; Toine C. G. Egberts; Marc Bierings; Geoff D.E. Cuvelier; Robert Wynn; Mary Slatter; Robert Chiesa; Meindert Danhof; Catherijne A. J. Knibbe

Background: Recently a pediatric pharmacokinetic (PK) model was developed for busulfan to explain the wide variability in PK of busulfan in children, as this variability is known to influence the outcome of hematopoietic stem cell transplantation in terms of toxicity and event free survival. This study assesses the predictive performance of this busulfan PK model in a new, more diverse pediatric population, including data from patients with different underlying diseases, ethnicities, body weights, ages, and body mass indices, from 5 international pediatric transplant centers. Patients and Methods: The previously published (original) busulfan PK model was developed from data of 245 patients (0.1–26 years of age). To externally validate this model, data were collected from another 158 patients (0.1–35 years) who underwent hematopoietic stem cell transplantation in 5 international transplant centers. Observed versus predicted plots, normalized prediction distribution error analysis, refit of the model on the external (n = 158) and combined datasets (n = 403), and subpopulation analyses were evaluated. Results: The original busulfan PK model was found to be stable and parameter estimates precise. Concentrations predicted by this model were in good agreement with the observed concentrations from the 5 external datasets. Plasma concentrations in patients with different underlying diseases, ethnicities, body weights, ages, and body mass indices were adequately predicted. Conclusions: Our pediatric busulfan PK model has been externally validated. This model predicts busulfan concentrations in pediatric and young adult patients ranging between 3 and 86 kg without bias and with good precision, regardless of transplant center, underlying disease, ethnicity, body weight age, or body mass index. This busulfan PK model forms the basis for individualized busulfan dosing.


Oncotarget | 2017

GSTA1 diplotypes affect busulfan clearance and toxicity in children undergoing allogeneic hematopoietic stem cell transplantation: a multicenter study

Marc Ansari; Patricia Huezo-Diaz Curtis; Chakradhara Rao S. Uppugunduri; Mohammed Aziz Rezgui; Tiago Nava; Vid Mlakar; Laurence Lesne; Yves Théorêt; Yves Chalandon; L. Lee Dupuis; Tao Schechter; Imke H. Bartelink; Jaap Jan Boelens; Robbert G. M. Bredius; Jean-Hugues Dalle; Saba Azarnoush; Petr Sedlacek; Victor Lewis; Martin A. Champagne; Christina Peters; Henrique Bittencourt; Maja Krajinovic

Busulfan (BU) dose adjustment following therapeutic drug monitoring contributes to better outcome of hematopoietic stem cell transplantation (HSCT). Further improvement could be achieved through genotype-guided BU dose adjustments. To investigate this aspect, polymorphism within glutathione S transferase genes were assessed. Particularly, promoter haplotypes of the glutathione S transferase A1 (GSTA1) were evaluated in vitro, with reporter gene assays and clinically, in a pediatric multi-center study (N =138) through association with BU pharmacokinetics (PK) and clinical outcomes. Promoter activity significantly differed between the GSTA1 haplotypes (p<0.001) supporting their importance in capturing PK variability. Four GSTA1 diplotype groups that significantly correlated with clearance (p=0.009) were distinguished. Diplotypes underlying fast and slow metabolizing capacity showed higher and lower BU clearance (ml/min/kg), respectively. GSTA1 diplotypes with slow metabolizing capacity were associated with higher incidence of sinusoidal obstruction syndrome, acute graft versus host disease and combined treatment-related toxicity (p<0.0005). Among other GST genes investigated, GSTP1 313GG correlated with acute graft versus host disease grade 1-4 (p=0.01) and GSTM1 non-null genotype was associated with hemorrhagic cystitis (p=0.003). This study further strengthens the hypothesis that GST diplotypes/genotypes could be incorporated into already existing population pharmacokinetic models for improving first BU dose prediction and HSCT outcomes. (No Clinicaltrials.gov identifier: NCT01257854. Registered 8 December 2010, retrospectively registered).


Biology of Blood and Marrow Transplantation | 2008

Once-Daily Intravenous Busulfan with Therapeutic Drug Monitoring Compared to Conventional Oral Busulfan Improves Survival and Engraftment in Children Undergoing Allogeneic Stem Cell Transplantation

Imke H. Bartelink; Robbert G. M. Bredius; Tessa F. T. Ververs; Martine F. Raphael; Charlotte van Kesteren; Marc Bierings; Carin M. A. Rademaker; J. den Hartigh; Cuno S.P.M. Uiterwaal; Juliette Zwaveling; Jaap Jan Boelens


American Journal of Ophthalmology | 2006

A randomized, masked, cross-over trial of lisinopril for inflammatory macular edema

Bram van Kooij; Rob Fijnheer; Joke H. de Boer; Ninette H. ten Dam-van Loon; Imke H. Bartelink; Mark Roest; Aniki Rothova


Biology of Blood and Marrow Transplantation | 2014

Population Pharmacokinetic Modeling of Thymoglobulin in Children Receiving Allogeneic-Hematopoietic Cell Transplantation (HCT): Towards Individualized Dosing to Improve Survival

Rick Admiraal; Charlotte van Kesteren; Cornelia M. Jol-van der Zijde; Maarten J. D. van Tol; Imke H. Bartelink; Robbert G. M. Bredius; Jaap-Jan Boelens; Catherijne A. J. Knibbe

Collaboration


Dive into the Imke H. Bartelink's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robbert G. M. Bredius

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Juliette Zwaveling

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christina Peters

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge