Network


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

Hotspot


Dive into the research topics where Bart A. W. Jacobs is active.

Publication


Featured researches published by Bart A. W. Jacobs.


Bioanalysis | 2015

Improved pharmacodynamic assay for dihydropyrimidine dehydrogenase activity in peripheral blood mononuclear cells

Dick Pluim; Bart A. W. Jacobs; Maarten J. Deenen; Anneloes Em Ruijter; Robin Van Geel; Artur M. Burylo; Didier Meulendijks; Jos H. Beijnen; Jan H. M. Schellens

BACKGROUND Dihydropyrimidine dehydrogenase (DPD) activity determination in peripheral blood mononuclear cells of DPD deficient patients was hitherto inaccurate due to hemoglobin (Hb) contamination. We developed an improved method for accurate measurement of DPD activity in patients. RESULTS DPD activity was determined by HPLC with online radioisotope detection using liquid scintillation counting. Hb was determined spectrophotometrically. Method accuracy and precision were significantly improved by using cumulative area of all peaks as IS. Peripheral blood mononuclear cell lysates from DPD deficient patients were highly contaminated with on average 23.3% (range 2.7-51%) of Hb resulting in up to twofold underestimated DPD activity. DPD activities were corrected for Hb contamination. The method was validated and showed good long-term sample stability. CONCLUSION This method has increased specificity allowing accurate identification of DPD deficient patients.


British Journal of Cancer | 2017

Pretreatment serum uracil concentration as a predictor of severe and fatal fluoropyrimidine-associated toxicity

Didier Meulendijks; Linda M. Henricks; Bart A. W. Jacobs; Abidin Aliev; Maarten J. Deenen; Niels de Vries; Hilde Rosing; Erik van Werkhoven; Anthonius de Boer; Jos H. Beijnen; Caroline M. Mandigers; M. Soesan; Annemieke Cats; Jan H. M. Schellens

Background:We investigated the predictive value of dihydropyrimidine dehydrogenase (DPD) phenotype, measured as pretreatment serum uracil and dihydrouracil concentrations, for severe as well as fatal fluoropyrimidine-associated toxicity in 550 patients treated previously with fluoropyrimidines during a prospective multicenter study.Methods:Pretreatment serum concentrations of uracil and dihydrouracil were measured using a validated LC-MS/MS method. The primary endpoint of this analysis was global (any) severe fluoropyrimidine-associated toxicity, that is, grade ⩾3 toxicity according to the NCI CTC-AE v3.0, occurring during the first cycle of treatment. The predictive value of uracil and the uracil/dihydrouracil ratio for early severe fluoropyrimidine-associated toxicity were compared. Pharmacogenetic variants in DPYD (c.2846A>T, c.1679T>G, c.1129-5923C>G, and c.1601G>A) and TYMS (TYMS 5′-UTR VNTR and TYMS 3′-UTR 6-bp ins/del) were measured and tested for associations with severe fluoropyrimidine-associated toxicity to compare predictive value with DPD phenotype. The Benjamini-Hochberg false discovery rate method was used to control for type I errors at level q<0.050 (corresponding to P<0.010).Results:Uracil was superior to the dihydrouracil/uracil ratio as a predictor of severe toxicity. High pretreatment uracil concentrations (>16 ng ml−1) were strongly associated with global severe toxicity (OR 5.3, P=0.009), severe gastrointestinal toxicity (OR 33.7, P<0.0001), toxicity-related hospitalisation (OR 16.9, P<0.0001), as well as fatal treatment-related toxicity (OR 44.8, P=0.001). None of the DPYD variants alone, or TYMS variants alone, were associated with severe toxicity.Conclusions:High pretreatment uracil concentration was strongly predictive of severe, including fatal, fluoropyrimidine-associated toxicity, and is a highly promising phenotypic marker to identify patients at risk of severe fluoropyrimidine-associated toxicity.


British Journal of Clinical Pharmacology | 2016

Pronounced between‐subject and circadian variability in thymidylate synthase and dihydropyrimidine dehydrogenase enzyme activity in human volunteers

Bart A. W. Jacobs; Maarten J. Deenen; Dick Pluim; J. G. Coen van Hasselt; Martin D. Krähenbühl; Robin Van Geel; Niels de Vries; Hilde Rosing; Didier Meulendijks; Artur M. Burylo; Annemieke Cats; Jos H. Beijnen; Alwin D. R. Huitema; Jan H. M. Schellens

AIMS The enzymatic activity of dihydropyrimidine dehydrogenase (DPD) and thymidylate synthase (TS) are important for the tolerability and efficacy of the fluoropyrimidine drugs. In the present study, we explored between-subject variability (BSV) and circadian rhythmicity in DPD and TS activity in human volunteers. METHODS The BSVs in DPD activity (n = 20) in peripheral blood mononuclear cells (PBMCs) and in plasma, measured by means of the dihydrouracil (DHU) and uracil (U) plasma levels and DHU : U ratio (n = 40), and TS activity in PBMCs (n = 19), were examined. Samples were collected every 4 h throughout 1 day for assessment of circadian rhythmicity in DPD and TS activity in PBMCs (n = 12) and DHU : U plasma ratios (n = 23). In addition, the effects of genetic polymorphisms and gene expression on DPD and TS activity were explored. RESULTS Population mean (± standard deviation) DPD activity in PBMCs and DHU : U plasma ratio were 9.2 (±2.1) nmol mg(-1) h(-1) and 10.6 (±2.4), respectively. Individual TS activity in PBMCs ranged from 0.024 nmol mg(-1) h(-1) to 0.596 nmol mg(-1) h(-1) . Circadian rhythmicity was demonstrated for all phenotype markers. Between 00:30 h and 02:00 h, DPD activity in PBMCs peaked, while the DHU : U plasma ratio and TS activity in PBMCs showed trough activity. Peak-to-trough ratios for DPD and TS activity in PBMCs were 1.69 and 1.62, respectively. For the DHU : U plasma ratio, the peak-to-trough ratio was 1.43. CONCLUSIONS BSV and circadian variability in DPD and TS activity were demonstrated. Circadian rhythmicity in DPD might be tissue dependent. The results suggested an influence of circadian rhythms on phenotype-guided fluoropyrimidine dosing and supported implications for chronotherapy with high-dose fluoropyrimidine administration during the night.


Journal of Pharmaceutical and Biomedical Analysis | 2016

Development and validation of a rapid and sensitive UPLC–MS/MS method for determination of uracil and dihydrouracil in human plasma

Bart A. W. Jacobs; Hilde Rosing; Niels de Vries; Didier Meulendijks; Linda M. Henricks; Jan H. M. Schellens; Jos H. Beijnen

Quantification of the endogenous dihydropyrimidine dehydrogenase (DPD) substrate uracil (U) and the reaction product dihydrouracil (UH2) in plasma might be suitable for identification of patients at risk of fluoropyrimidine-induced toxicity as a result of DPD deficiency. In this paper, we describe the development and validation of a rapid and sensitive ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) assay for quantification of U and UH2 in human plasma. Analytes were extracted by protein precipitation, chromatographically separated on an Acquity UPLC(®) HSS T3 column with gradient elution and analyzed with a tandem mass spectrometer equipped with an electrospray ionization source. U was quantified in the negative ion mode and UH2 in the positive ion mode. Stable isotopes for U and UH2 were used as internal standards. Total chromatographic run time was 5min. Validated concentration ranges for U and UH2 were from 1 to 100ng/mL and 10 to 1000ng/mL, respectively. Inter-assay bias and inter-assay precision for U were within ±2.8% and ≤12.4%. For UH2, inter-assay bias and inter-assay precision were within ±2.9% and ≤7.2%. Adequate stability of U and UH2 in dry extract, final extract, stock solution and plasma was demonstrated. Stability of U and UH2 in whole blood was only satisfactory when stored up to 4hours at 2-8°C, but not at ambient temperatures. An accurate, precise and sensitive UPLC-MS/MS assay for quantification of U and UH2 in plasma was developed. This assay is now applied to support clinical studies with fluoropyrimidine drugs.


Analytical and Bioanalytical Chemistry | 2013

Correction of peripheral blood mononuclear cell cytosolic protein for hemoglobin contamination

Dick Pluim; Bart A. W. Jacobs; Martin D. Krähenbühl; Anna E. M. Ruijter; Jos H. Beijnen; Jan H. M. Schellens

Pharmacodynamic (PD) analysis requires accurate and precise quantification of enzyme activity targeted by anticancer agents in surrogate cells like peripheral blood mononuclear cells (PBMCs). Enzyme activity is normally reported per mass unit of protein input. However, high and fluctuating hemoglobin (Hb) contamination strongly influences the protein content of PBMC cytosolic lysate. We present the development and validation of a spectrophotometrical Hb quantification method to correct for this contamination. The applicability of Hb correction was demonstrated by determination of the dihydropyrimidine dehydrogenase enzyme activity in PBMC cytosolic lysates.


Pediatric Transplantation | 2016

Pharmacokinetics and target attainment of mycophenolate in pediatric renal transplant patients

Lisa C. Martial; Bart A. W. Jacobs; Elisabeth A. M. Cornelissen; Anton F.J. De Haan; Birgit C. P. Koch; David M. Burger; Rob E. Aarnoutse; Michiel F. Schreuder; Roger J. M. Brüggemann

MPA is an immunosuppressive agent used to prevent graft rejection after renal transplantation. MPA shows considerable inter‐ and intraindividual variability in exposure in children and has a defined therapeutic window, and TDM is applied to individualize therapy. We aimed to study the exposure to MPA measured as the AUC in pediatric renal transplant patients, to identify factors influencing exposure and to assess target attainment. Children transplanted between 1998 and 2014 in a single center were included. Two groups were identified: Group 1 (AUC <3 wk post‐transplantation) and Group 2 (AUC >18 months post‐transplantation). Therapeutic targets were set at: AUC0–12h of 30–60 mg h/L. A total of 39 children were included in Group 1 (median age 13.3 yr) vs. 14 in Group 2 (median age 13.4 yr). AUC0–12h was 29.7 mg h/L in Group 1 and 56.6 mg h/L in Group 2, despite a lower dosage in Group 2 (584 and 426 mg/m2, respectively). About 46% of patients reached the target AUC0–12h in Group 1. Time since transplantation and serum creatinine were significantly associated with MPA exposure (p < 0.001), explaining 36% of the variability. Individualization of the mycophenolate dose by more intense and more early TDM could improve target attainment.


British Journal of Clinical Pharmacology | 2018

Food‐effect study on uracil and dihydrouracil plasma levels as marker for dihydropyrimidine dehydrogenase activity in human volunteers

Linda M. Henricks; Bart A. W. Jacobs; Didier Meulendijks; Dick Pluim; Daan van den Broek; Niels de Vries; Hilde Rosing; Jos H. Beijnen; Alwin D. R. Huitema; Henk-Jan Guchelaar; Annemieke Cats; Jan H. M. Schellens

This study aimed to determine the effect of food intake on uracil and dihydrouracil plasma levels. These levels are a promising marker for dihydropyrimidine dehydrogenase activity and for individualizing fluoropyrimidine anticancer therapy.


International Journal of Cancer | 2016

Increased risk of severe fluoropyrimidine‐associated toxicity in patients carrying a G to C substitution in the first 28‐bp tandem repeat of the thymidylate synthase 2R allele

Didier Meulendijks; Bart A. W. Jacobs; Abidin Aliev; Dick Pluim; Erik van Werkhoven; Maarten J. Deenen; Jos H. Beijnen; Annemieke Cats; Jan H. M. Schellens


Cancer Chemotherapy and Pharmacology | 2016

Patients homozygous for DPYD c.1129-5923C>G/haplotype B3 have partial DPD deficiency and require a dose reduction when treated with fluoropyrimidines

Didier Meulendijks; Linda M. Henricks; André B.P. van Kuilenburg; Bart A. W. Jacobs; Abidin Aliev; Lisette Rozeman; Judith Meijer; Jos H. Beijnen; Hiltje de Graaf; Annemieke Cats; Jan H. M. Schellens


Analytical and Bioanalytical Chemistry | 2013

Pharmacodynamic assay of thymidylate synthase activity in peripheral blood mononuclear cells.

Dick Pluim; Kim A. A. Schilders; Bart A. W. Jacobs; Daniëlle Vaartjes; Jos H. Beijnen; Jan H. M. Schellens

Collaboration


Dive into the Bart A. W. Jacobs's collaboration.

Top Co-Authors

Avatar

Jan H. M. Schellens

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Jos H. Beijnen

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Dick Pluim

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Hilde Rosing

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Didier Meulendijks

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Niels de Vries

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Alwin D. R. Huitema

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Annemieke Cats

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Maarten J. Deenen

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Linda M. Henricks

Netherlands Cancer Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge