Network


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

Hotspot


Dive into the research topics where Tahar van der Straaten is active.

Publication


Featured researches published by Tahar van der Straaten.


Journal of Clinical Oncology | 2009

Pharmacogenetic Pathway Analysis for Determination of Sunitinib-Induced Toxicity

Nielka P. van Erp; Karel Eechoute; Astrid A.M. van der Veldt; John B. A. G. Haanen; An Reyners; Ron H.J. Mathijssen; Epie Boven; Tahar van der Straaten; Renee Baak-Pablo; Judith A.M. Wessels; Henk-Jan Guchelaar; Hans Gelderblom

PURPOSE To identify genetic markers in the pharmacokinetic and pharmacodynamic pathways of sunitinib that predispose for development of toxicities: thrombocytopenia, leukopenia, mucosal inflammation, hand-foot syndrome, and any toxicity according to National Cancer Institute Common Toxicity Criteria higher than grade 2. PATIENTS AND METHODS A multicenter pharmacogenetic association study was performed in 219 patients treated with single-agent sunitinib. A total of 31 single nucleotide polymorphisms in 12 candidate genes, together with several nongenetic variants, were analyzed for a possible association with toxicity. In addition, genetic haplotypes were developed and related to toxicity. RESULTS The risk for leukopenia was increased when the G allele in CYP1A1 2455A/G (odds ratio [OR], 6.24; P = .029) or the T allele in FLT3 738T/C (OR, 2.8; P = .008) were present or CAG in the NR1I3 (5719C/T, 7738A/C, 7837T/G) haplotype (OR, 1.74; P = .041) was absent. Any toxicity higher than grade 2 prevalence was increased when the T allele of vascular endothelial growth factor receptor 2 1191C/T (OR, 2.39; P = .046) or a copy of TT in the ABCG2 (-15622C/T, 1143C/T) haplotype (OR, 2.63; P = .016) were present. The risk for mucosal inflammation was increased in the presence of the G allele in CYP1A1 2455A/G (OR, 4.03; P = .021) and the prevalence of hand-foot syndrome was increased when a copy of TTT in the ABCB1 (3435C/T, 1236C/T, 2677G/T) haplotype (OR, 2.56; P = .035) was present. CONCLUSION This exploratory study suggests that polymorphisms in specific genes encoding for metabolizing enzymes, efflux transporters, and drug targets are associated with sunitinib-related toxicities. A better understanding of genetic and nongenetic determinants of sunitinib toxicity should help to optimize drug treatment in individual patients.


Therapeutic Drug Monitoring | 2009

Explaining Variability in Tacrolimus Pharmacokinetics to Optimize Early Exposure in Adult Kidney Transplant Recipients

Rogier R. Press; Bart A. Ploeger; Jan den Hartigh; Tahar van der Straaten; Johannes van Pelt; Meindert Danhof; Johan W. de Fijter; Henk-Jan Guchelaar

To prevent acute rejection episodes, it is important to reach adequate tacrolimus (TRL) exposure early after kidney transplantation. With a better understanding of the high variability in the pharmacokinetics of TRL, the starting dose can be individualized, resulting in a reduction in dose adjustments to obtain the target exposure. A population pharmacokinetic analysis was performed to estimate the effects of demographic factors, hematocrit, serum albumin concentration, prednisolone dose, TRL dose interval, polymorphisms in genes coding for ABCB1, CYP3A5, CYP3A4, and the pregnane X receptor on TRL pharmacokinetics. Pharmacokinetic data were prospectively obtained in 31 de novo kidney transplant patients randomized to receive TRL once or twice daily, and subsequently, the data were analyzed by means of nonlinear mixed-effects modeling. TRL clearance was 1.5-fold higher for patients with the CYP3A5*1/*3 genotype compared with the CYP3A5*3/*3 genotype (5.5 ± 0.5 L/h versus 3.7 ± 0.3 L/h, respectively). This factor explained 30% of the interindividual variability in apparent clearance (exposure). Also, a relationship between the pregnane X receptor A+7635G genotype and TRL clearance was identified with a clearance of 3.9 ± 0.3 L/h in the A allele carriers versus 5.4 ± 0.6 L/h in the GG genotype. Finally, a concomitant prednisolone dose of more than 10 mg/d increased the TRL apparent clearance by 15%. In contrast, body weight was not related to TRL clearance in this population. Because patients are typically dosed per kilogram body weight, this might result in underexposure and overexposure in patients, with a low and high body weight, respectively. This integrated analysis shows that adult renal transplant recipients with the CYP3A5*1/*3 genotype require a 1.5 times higher, fixed, starting dose compared with CYP3A5*3/*3 to reach the predefined target exposure early after transplantation.


European Journal of Cancer | 2010

Correlation of FCGR3A and EGFR germline polymorphisms with the efficacy of cetuximab in KRAS wild-type metastatic colorectal cancer.

Jan Pander; Hans Gelderblom; Ninja Antonini; Jolien Tol; Johan H. J. M. van Krieken; Tahar van der Straaten; Cornelis J. A. Punt; Henk-Jan Guchelaar

BACKGROUND Next to KRAS mutation status, additional predictive markers are needed for the response to cetuximab in patients with metastatic colorectal cancer (mCRC). Previous studies indicated that germline polymorphisms in specific genes may predict efficacy and toxicity of cetuximab in mCRC patients. METHODS Germline DNA was isolated from 246 KRAS wild-type mCRC patients who were treated in the phase III CAIRO2 study with chemotherapy and bevacizumab alone or with the addition of cetuximab. Associations of epidermal growth factor (EGF) 61A>G, EGF receptor (EGFR) CA(14-22), cyclin D1 (CCND1) 932G>A, fragment-C gamma receptor (FCGR) 2A 535A>G and FCGR3A 818A>C polymorphisms with progression-free survival (PFS) and cetuximab-related skin toxicity were studied. RESULTS In cetuximab-treated patients, the FCGR3A 818C-allele was associated with decreased PFS compared with the FCGR3A 818AA genotype (median PFS, 8.2 [95%CI, 6.7-10.3] versus 12.8 [95%CI, 10.3-14.7] months, respectively; HR, 1.57 [95%CI, 1.06-2.34]; P=.025). The EGFR20 genotype was associated with decreased PFS compared with the EGFR<20 genotype (median PFS, 7.6 [95%CI, 6.7-10.0] versus 12.4 [95%CI, 10.3-13.4] months, respectively; HR, 1.58 [95%CI, 1.06-2.35]; P=.024). The FCGR3A and EGFR polymorphisms were not associated with PFS in patients treated without cetuximab. None of the polymorphisms were associated with the incidence of grades 2-3 skin toxicity. CONCLUSION EGFR and FCGR3A germline polymorphisms are associated with PFS in KRAS wild-type mCRC patients treated with cetuximab, bevacizumab and chemotherapy.


Clinical Cancer Research | 2011

Activation of tumor-promoting type 2 macrophages by EGFR-targeting antibody cetuximab.

Jan Pander; Moniek Heusinkveld; Tahar van der Straaten; Ekaterina S. Jordanova; Renee Baak-Pablo; Hans Gelderblom; Hans Morreau; Sjoerd H. van der Burg; Henk-Jan Guchelaar; Thorbald van Hall

Purpose: In a recent randomized phase III clinical trial in metastatic colorectal cancer patients, the addition of the anti-epidermal growth factor receptor (EGFR) monoclonal antibody (mAb) cetuximab to bevacizumab and chemotherapy resulted in decreased progression-free survival, in particular for patients with the high-affinity FcγRIIIA. Experimental Design: The presence of natural killer (NK) cells and type 2 (M2) macrophages in colorectal cancer was determined by immunohistochemistry, using antibodies to lineage-specific markers NKp46 and CD68 with CD163, respectively. Influence of tumor-bound cetuximab on M2 macrophages was carried out in vitro with EGFR-expressing tumor cells and short-term differentiated monocytes from blood donors, who were typed for the FcγRIIIA polymorphism (CD16). Results: Antibody-dependent cellular cytotoxicity by NK cells is generally proposed as one of the antitumor mechanisms of mAbs. We found that CD163-positive M2 macrophages are much more abundant in colorectal carcinomas. In vitro analysis of M2 macrophages revealed high levels of Fc-gamma receptors (FcγR) and PD-L1 and production of IL-10 and VEGF but not IL-12. These anti-inflammatory and tumor-promoting mediators were released upon coculture with EGFR-positive tumor cells loaded with low concentrations of cetuximab. Macrophage activation depended on EGFR expression on the tumor cells, FcγRs, target specificity of the mAb and mobility of antibody complexes. Cetuximab-induced macrophage responses were more pronounced for FCGR3A 158-Val (high-affinity) carriers. Conclusion: These results suggest that tumor-promoting M2 macrophages are activated by the therapeutic mAb cetuximab in the local tumor microenvironment and argue that this immune mechanism should be taken into account for the application of therapeutic antibodies. Clin Cancer Res; 17(17); 5668–73. ©2011 AACR.


Pharmacogenomics | 2010

Functional polymorphisms and methotrexate treatment outcome in recent-onset rheumatoid arthritis

Wouter M. Kooloos; Judith A.M. Wessels; Tahar van der Straaten; Cornelia F Allaart; Tom W J Huizinga; Henk-Jan Guchelaar

AIMS Clinical response to methotrexate (MTX) treatment differs among rheumatoid arthritis patients. Genetic variation can partly account for this phenomenon. In this study, functional polymorphisms in genes related to the mechanism of action of MTX or immunopathogenesis of rheumatoid arthritis were studied for association with treatment outcome in a Dutch cohort of patients with early rheumatoid arthritis. Furthermore, tests for replication of previous research on these genetic variants were performed according to reported end points. MATERIALS & METHODS Seven polymorphisms in seven genes were analyzed in 205 genotyped patients with active rheumatoid arthritis. All patients received standardized MTX treatment (< or =25 mg per week orally) combined with folic acid. MTX treatment outcome was evaluated by disease activity score criteria and adverse drug events. The following genetic variants were analyzed and correlated: ABCB1 3435C>T, ITPA IVS2 +21A>C, HLA-G (-14 bp >+14 bp), TGFB1 +869T>C and TLR4 +896A>G. In case of significant differences, regression analyses were applied. Since carriers of the minor alleles of the SNPs DHFR 829C>T and IMPDH2 +787C>T were not observed, no statistical analyses could be performed. RESULTS No significant associations or replications of these genetic variants with MTX efficacy were demonstrated. Regarding toxicity, patients carrying the ABCB1 3435T-allele and TLR4 +896G-allele were 2.5-times more likely to develop adverse drug events at 6 months (odds ratio: 2.6; 95% CI: 1.1-6.2, and odds ratio: 2.5; 95% CI: 1.1-6.1, respectively). Additionally, this chance increased almost fourfold in patients with the two unfavorable genotypes (odds ratio: 3.9; 95% CI: 1.5-10.3). However, none of these associations remained significant after correction for multiple testing (p < 0.004). CONCLUSION Our data indicate that MTX toxicity was potentially associated with ABCB1 3435C>T and TLR4 +896A>G. However, after correction, none of these associations remained significant. Furthermore, no significant associations or replications of these functional variants with efficacy were found.


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.


European Journal of Cancer | 2009

Glutathione-S-transferase pi (GSTP1) codon 105 polymorphism is not associated with oxaliplatin efficacy or toxicity in advanced colorectal cancer patients.

Dina M. Kweekel; Hans Gelderblom; Ninja Antonini; Tahar van der Straaten; Johan W.R. Nortier; Cornelis J. A. Punt; Henk-Jan Guchelaar

PURPOSE Oxaliplatin is detoxified by conjugation to glutathione via the enzyme Glutathione-S-transferase pi (GSTP1). The aim of this study is to investigate the association of GSTP1 Ile105Val genetic polymorphism with oxaliplatin efficacy and toxicity in advanced colorectal cancer (ACC) patients. EXPERIMENTAL DESIGN A total of 91 ACC patients received capecitabine and oxaliplatin (CAPOX) as a part of a multicentre phase-III study of the Dutch Colorectal Cancer Group. Tumour response was evaluated according to RECIST, toxicity was graded using CTC, and GSTP1 Ile105Val was determined by pyrosequencing. RESULTS Overall survival after CAPOX was similar for patients with the Ile/Ile (11.5 mo), Ile/Val (11.6 mo) and Val/Val (12.6 mo) genotypes (p=0.602). Likewise, there were no statistically significant differences in progression-free survival (p=0.252). Overall grades 3-4 toxicity was not related to genotype (p=0.313). There were no differences in any grade or grades 3-4 neurotoxicity amongst the patients who received > or =500 mg/m(2) of oxaliplatin (p-values of 0.376 and 0.772, respectively). CONCLUSIONS The results of this study indicate that the GSTP1 genotype is not predictive for progression-free survival or overall survival in ACC patients treated with CAPOX. Moreover, overall neurotoxicity and neurotoxicity in patients receiving 500 mg/m(2) of oxaliplatin was not associated with GSTP1 genotype.


The Journal of Clinical Endocrinology and Metabolism | 2008

Influence of the d3-Growth Hormone (GH) Receptor Isoform on Short-Term and Long-Term Treatment Response to GH Replacement in GH-Deficient Adults

Agatha A. van der Klaauw; Tahar van der Straaten; Renee Baak-Pablo; Nienke R. Biermasz; Henk-Jan Guchelaar; Alberto M. Pereira; Johannes W. A. Smit; Johannes A. Romijn

OBJECTIVE Recombinant human GH (rhGH) replacement in adults is aimed at improving signs and symptoms of the adult GH deficiency (GHD) syndrome. In children, a common polymorphism of the GH receptor (exon-3 deletion, d3GHR) increases the response to rhGH replacement. The aim of this study was to assess the effects of this polymorphism on the response to rhGH replacement in adults. DESIGN Prospective intervention with rhGH during 1 yr (n = 99) and in a subset during 5 yr (n = 53). PATIENTS AND METHODS The presence of the d3GHR variant was established in GHD patients and linked to short-term and long-term effects of rhGH replacement on IGF-I, lipid metabolism, anthropometric parameters, and bone mineral density. RESULTS Fifty-five patients had two wild-type alleles (56%), whereas 38 patients (38%) had one allele and six patients (6%) had two alleles coding the d3GHR isoform. During short-term rhGH replacement, the increase in IGF-I was higher in patients bearing at least one d3GHR allele, compared with those with two wild-type alleles (at an identical mean dose of rhGH). The decrease in total cholesterol and low-density lipoprotein cholesterol was lower in the group bearing at least one d3GHR allele, whereas the increase in high-density lipoprotein cholesterol was higher, compared with patients with the wild-type genotype. In contrast, these differential responses of GHR genotype could not be demonstrated during long-term rhGH replacement. CONCLUSION The d3GHR genotype contributes, at least for some parameters, to the interindividual differences in efficacy of short-term, but not long-term, rhGH replacement in adults with GHD.


Pharmacogenetics and Genomics | 2009

Gene-gene interactions in folate and adenosine biosynthesis pathways affect methotrexate efficacy and tolerability in rheumatoid arthritis.

Thierry Dervieux; Judith A.M. Wessels; Tahar van der Straaten; Nadia M. Penrod; Jason H. Moore; Henk-Jan Guchelaar; Joel M. Kremer

ObjectiveAs no single nucleotide polymorphism has emerged as pivotal to predict the lack of efficacy and dose-limiting toxicities to methotrexate (MTX), we evaluated the contribution of gene–gene interactions to the effects of this prodrug in rheumatoid arthritis. MethodsA total of 255 patients treated with MTX for at least 3 months were evaluated with efficacy assessed using the European League Against Rheumatism response criteria or a physicians assessment of patients response to MTX visual analog scale. Gastrointestinal and neurological idiosyncrasies were recorded in 158 patients. Fourteen single nucleotide polymorphisms in folate and adenosine biosynthesis pathways were measured and detection of gene–gene interactions was performed using multifactor-dimensionality reduction, a method that reduces high-dimensional genetic data into a single dimension of predisposing or risk–genotype combinations. ResultsEfficacy to MTX (53% responders) was associated with high-order epistasis among variants in inosine-triphosphate pyrophosphatase, aminoimidazole-carboxamide ribonucleotide transformylase, and reduced folate carrier genes. In the absence of predisposing genotype combinations, a 3.8-fold lower likelihood of efficacy was observed (vs. in their presence, 95% confidence interval: 2.2–6.4; P<0.001). Increasing MTX polyglutamate concentrations tended to partially overcome this selective disadvantage. Idiosyncrasies occurred in 29% of patients. In the presence of risk–genotype combinations among variants in methylene tetrahydrofolate reductase, &ggr;-glutamyl-hydrolase, thymidylate synthase, serine hydroxymethyltransferase, and inosine-triphosphate pyrophosphatase genes, an 8.9-fold higher likelihood to exhibit toxicities was observed (vs. in their absence, 95% confidence interval: 3.6–21.9; P<0.001). False-positive report probabilities were below 0.2, thereby indicating that true signals were likely detected in this cohort. ConclusionThese data indicate that gene–gene interactions impact MTX efficacy and tolerability in rheumatoid arthritis.


Clinical Pharmacokinectics | 2012

Population Pharmacokinetics and Pharmacogenetics of Everolimus in Renal Transplant Patients

Dirk Jan A.R. Moes; Rogier R. Press; Jan den Hartigh; Tahar van der Straaten; Johan W. de Fijter; Henk-Jan Guchelaar

Background and ObjectiveEverolimus is a novel macrolide immunosuppressant used in the prevention of acute and chronic rejection of solid organ transplants. Everolimus is being actively investigated worldwide as a non-nephrotoxic alternative for calcineurin inhibitors. Its highly variable pharmacokinetics and narrow therapeutic window make it difficult to maintain an adequate exposure to prevent serious adverse effects. The primary objective of this study was to improve prediction of everolimus systemic exposure in renal transplant patients by describing the pharmacokinetics of everolimus and identifying the influence of demographic factors and a selection of polymorphisms in genes coding for ABCB1, CYP3A5, CYP2C8 and PXR. The secondary objective of this study was to develop a limited sampling strategy to enable prediction of everolimus exposure in an efficient way and to compare it with the widely used trough blood concentration (Ctrough) monitoring.MethodsA total of 783 blood samples were obtained from 53 renal transplant patients who had been switched from a triple therapy of ciclosporin, mycophenolate mofetil and prednisolone to a calcineurin inhibitor-free dual therapy of everolimus (twice daily) and prednisolone. Everolimus blood concentrations were analysed in whole blood using liquid chromatography-tandem mass spectrometry during routine therapeutic drug monitoring targeting an area under the blood concentration-time curve from time zero to 12 hours (AUC12) of 120 μg · h/L. A population pharmacokinetic model was developed and demographic factors and genetic polymorphisms in genes coding for ABCB1, CYP3A5, CYP2C8 and PXR were included as covariates. In addition, a limited sampling strategy was developed.ResultsMaintaining everolimus systemic exposure at an AUC12 of 120 120 μg · h/L resulted in low rejection rates but considerable numbers of adverse events and toxicity. Everolimus pharmacokinetics were best described by a two-compartment model with lag-time (oral clearance= 17.9 L/h; volume of distribution of the central compartment after oral administration [V1/F]= 148 L and first-order absorption rate constant [ka] = 7.36 h−1). Ideal body weight was significantly related to V1/F. None of the selected polymorphisms in genes coding for enzymes involved in distribution and metabolism of everolimus had a significant influence on everolimus pharmacokinetics. The pharmacokinetic limited sampling model (Ctrough and whole blood drug concentration at 2 hours postdose [C2]) resulted in a significantly improved prediction of everolimus exposure compared with the widely used Ctrough monitoring.ConclusionA two-compartment pharmacokinetic model with lag-time describing the concentration-time profile of oral everolimus in renal transplant patients has been developed using pharmacokinetic modelling. Ideal body weight significantly influenced V1/F of everolimus; however, the selected polymorphisms in genes coding for ABCB1, CYP3A5, CYP2C8 and PXR had no clinically relevant effect on everolimus pharmacokinetics. Everolimus Ctrough and C2 as a limited sampling model can be used to accurately estimate everolimus systemic exposure, an improvement over the widely used Ctrough monitoring.

Collaboration


Dive into the Tahar van der Straaten's collaboration.

Top Co-Authors

Avatar

Henk-Jan Guchelaar

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Hans Gelderblom

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jesse J. Swen

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Judith A.M. Wessels

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Renee Baak-Pablo

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vincent O. Dezentjé

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hein Putter

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Caroline Seynaeve

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge