Network


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

Hotspot


Dive into the research topics where O. van Tellingen is active.

Publication


Featured researches published by O. van Tellingen.


Cell | 1994

Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs

Alfred H. Schinkel; J.J.M. Smit; O. van Tellingen; Jos H. Beijnen; Els Wagenaar; L van Deemter; Carla A. A. M. Mol; M.A. van der Valk; Els C. Robanus-Maandag; H te Riele; Anton Berns; Piet Borst

We have generated mice homozygous for a disruption of the mdr1a (also called mdr3) gene, encoding a drug-transporting P-glycoprotein. The mice were viable and fertile and appeared phenotypically normal, but they displayed an increased sensitivity to the centrally neurotoxic pesticide ivermectin (100-fold) and to the carcinostatic drug vinblastine (3-fold). By comparison of mdr1a (+/+) and (-/-) mice, we found that the mdr1a P-glycoprotein is the major P-glycoprotein in the blood-brain barrier and that its absence results in elevated drug levels in many tissues (especially in brain) and in decreased drug elimination. Our findings explain some of the side effects in patients treated with a combination of carcinostatics and P-glycoprotein inhibitors and indicate that these inhibitors might be useful in selectively enhancing the access of a range of drugs to the brain.


British Journal of Cancer | 1997

Enhanced oral bioavailability of paclitaxel in mice treated with the P-glycoprotein blocker SDZ PSC 833

J. van Asperen; O. van Tellingen; A. Sparreboom; Alfred H. Schinkel; Piet Borst; Willem J. Nooijen; Jos H. Beijnen

Inhibition of intestinal P-glycoprotein might enhance the absorption of orally administered P-glycoprotein substrate drugs. We show here a 10-fold increased oral bioavailability of paclitaxel in mice treated with the P-glycoprotein blocker SDZ PSC 833. These results encourage further research on the development of a clinically useful oral formulation of paclitaxel.


European Journal of Cancer | 1998

Intraperitoneal cisplatin with regional hyperthermia in advanced ovarian cancer: pharmacokinetics and cisplatin–DNA adduct formation in patients and ovarian cancer cell lines

P.J.M. van de Vaart; N. van der Vange; F.A.N. Zoetmulder; A.R. van Goethem; O. van Tellingen; W.W. ten Bokkel Huinink; Jos H. Beijnen; Harry Bartelink; Adrian C. Begg

The purpose of this study was to investigate the influence of hyperthermia on cisplatin pharmacokinetics and DNA adduct formation. The latter was investigated both in tumour cell lines in vitro and in tumour cells and buccal cells from cancer patients. The patients had advanced ovarian carcinoma and were entered into a phase I study for cytoreductive surgery followed by hyperthermia in combination with intraperitoneal cisplatin administration. The cisplatin-DNA modifications in vivo and in vitro were studied by an immunocytochemical method with the polyclonal antiserum NKI-A59. The patient samples for pharmacokinetic determinations were analysed by flameless atomic absorption spectrometry. In vitro, the combination of hyperthermia and cisplatin enhanced cell killing compared with either treatment alone, such that the cisplatin-resistant ovarian cell line A2780/DDP became almost as sensitive as the parent A2780 cell line (resistance factor reduced from 30 to 2 at the IC50). In addition, increased cisplatin-DNA adducts were observed in the resistant cell line after the combined treatment compared with cisplatin alone. A good correlation was found between nuclear staining density and surviving fraction for all groups, indicating that the DNA adducts generated are an important determinant of toxicity and that the mechanism by which hyperthermia enhances kill is by increasing adduct levels. In the patients, the ratio of drug concentration in the peritoneal perfusate compared with that in plasma was found to be approximately 15, indicating a favourable pharmacokinetic ratio. Cisplatin-DNA adduct formation in tumour cells from patients was higher than in buccal cells, reflecting this higher drug exposure, i.e. local plus systemic versus systemic only. In addition, the tumour cells but not buccal cells were exposed to hyperthermia. The higher number of tumour adducts also suggests that a favourable therapeutic ratio could be achieved. Platinum-DNA adduct formation was found to decrease with distance from the surface of the tumour nodules. However, at a distance of 3-5 mm, the nuclear staining density levels were still measurable and higher than in buccal cells. In conclusion, the combined pharmacokinetic and adduct data in patients support the advantages of the intraperitoneal route for drug administration, and the addition of heat.


British Journal of Cancer | 1999

Cremophor EL causes (pseudo-) non-linear pharmacokinetics of paclitaxel in patients.

O. van Tellingen; M. T. Huizing; V R Nannan Panday; Jan H. M. Schellens; Willem J. Nooijen; Jos H. Beijnen

SummaryThe non-linear plasma pharmacokinetics of paclitaxel in patients has been well established, however, the exact underlying mechanism remains to be elucidated. We have previously shown that the non-linear plasma pharmacokinetics of paclitaxel in mice results from Cremophor EL. To investigate whether Cremophor EL also plays a role in the non-linear pharmacokinetics of paclitaxel in patients, we have established its pharmacokinetics in patients receiving paclitaxel by 3-, 24- or 96-h intravenous infusion. The pharmacokinetics of Cremophor EL itself was non-linear as the clearance (Cl) in the 3-h schedules was significantly lower than when using the longer 24- or 96-h infusions (Cl175–3 h = 42.8 ± 24.9 ml h–1 m–2; Cl175–24 h = 79.7 ± 24.3; P = 0.035 and Cl135–3 h = 44.1 ± 21.8 ml h–1 m–1; Cl140–96 h = 211.8 ± 32.0; P < 0.001). Consequently, the maximum plasma levels were much higher (0.62%) in the 3-h infusions than when using longer infusion durations. By using an in vitro equilibrium assay and determination in plasma ultrafiltrate we have established that the fraction of unbound paclitaxel in plasma is inversely related with the Cremophor EL level. Despite its relatively low molecular weight, no Cremophor EL was found in the ultrafiltrate fraction. Our results strongly suggest that entrapment of paclitaxel in plasma by Cremophor EL, probably by inclusion in micelles, is the cause of the apparent nonlinear plasma pharmacokinetics of paclitaxel. This mechanism of a (pseudo-)non-linearity contrasts previous postulations about saturable distribution and elimination kinetics and means that we must re-evaluate previous assumptions on pharmacokinetics–pharmacodynamics relationships.


Journal of Pharmacology and Experimental Therapeutics | 2006

Carcinogen and Anticancer Drug Transport by Mrp2 in Vivo: Studies Using Mrp2 (Abcc2) Knockout Mice

Maria L. H. Vlaming; Karin Mohrmann; Els Wagenaar; D.R. de Waart; R.P.J. Oude Elferink; Jurjen S. Lagas; O. van Tellingen; Liia D. Vainchtein; Hilde Rosing; Jos H. Beijnen; Jan H. M. Schellens; Alfred H. Schinkel

The ATP-binding-cassette (ABC) transporter multidrug resistance protein (MRP) 2 (ABCC2) forms a natural barrier and efflux system for various (conjugates of) drugs, other xenotoxins, and endogenous compounds. To obtain insight in the pharmacological and physiological functions of Mrp2, we generated Mrp2 knockout mice, which were viable and fertile but suffered from mild hyperbilirubinemia due to impaired excretion of bilirubin monoglucuronides into bile. The mice also had an 80-fold decreased biliary glutathione excretion and a 63% reduced bile flow. Levels of Mrp3 (Abcc3) in liver and Mrp4 (Abcc4) in kidney of Mrp2-/- mice were approximately 2-fold increased. After oral administration of the food-derived carcinogens [14C]PhIP (2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine) and [14C]IQ (2-amino-3-methylimidazo[4,5-f]quinoline) plasma values were 1.9- and 1.7-fold higher in Mrp2-/- mice versus wild-type mice, respectively, demonstrating the role of Mrp2 in restricting exposure to these compounds. At a high dose of 50 mg/kg of the drug [3H]methotrexate, the plasma area under the curve for i.v. administration was 1.8-fold higher in Mrp2-/- mice (1345 ± 207 versus 734 ± 81 min ·μg/ml). No clear plasma concentration difference arose at low dose (1 mg/kg). Subsequently, Mdr1a/b/Mrp2 knockout mice were generated. Their biliary excretion of doxorubicin after i.v. administration (5 mg/kg) was 54-fold decreased (0.32 ± 0.13 versus 17.30 ± 6.59 nmol/g liver in wild type), and a role for both Mdr1a/b and Mrp2 in this process was revealed. Our results demonstrate that the Mrp2-/- mouse provides a valuable tool for studies of the impact of Mrp2 on behavior of drugs and other toxins, especially when combined with other ABC transporter knockout mice.


British Journal of Cancer | 1999

Increased accumulation of doxorubicin and doxorubicinol in cardiac tissue of mice lacking mdr1a P-glycoprotein

J. van Asperen; O. van Tellingen; F Tijssen; Alfred H. Schinkel; Jos H. Beijnen

SummaryTo gain more insight into the pharmacological role of endogenous P-glycoprotein in the metabolism of the widely used substrate drug doxorubicin, we have studied the plasma pharmacokinetics, tissue distribution and excretion of this compound in mdr1a(–/–) and wild-type mice. Doxorubicin was administered as an i.v. bolus injection at a dose level of 5 mg kg–1. Drug and metabolite concentrations were determined in plasma, tissues, urine and faeces by high-performance liquid chromatography. In comparison with wild-type mice, the terminal half-life and the area under the plasma concentration–time curve of doxorubicin in mdr1a(–/–) mice were 1.6- and 1.2-fold higher respectively. The retention of both doxorubicin and its metabolite doxorubicinol in the hearts of mdr1a(–/–) mice was substantially prolonged. In addition, a significantly increased drug accumulation was observed in the brain and the liver of mdr1a(–/–) mice. The relative accumulation in most other tissues was not or only slightly increased. The differences in cumulative faecal and urinary excretion of doxorubicin and metabolites between both types of mice were small. These experiments demonstrate that the absence of mdr1a P-glycoprotein only slightly alters the plasma pharmacokinetics of doxorubicin. Furthermore, the substantially prolonged presence of both doxorubicin and doxorubicinol in cardiac tissue of mdr1a(–/–) mice suggests that a blockade of endogenous P-glycoprotein in patients, for example by a reversal agent, may enhance the risk of cardiotoxicity upon administration of doxorubicin.


Fresenius Journal of Analytical Chemistry | 1995

Validated method for the determination of carboplatin in biological fluids by Zeeman atomic absorption spectrometry

L. J. C. van Warmerdam; O. van Tellingen; R. A. A. Maes; Jos H. Beijnen

We have developed and validated a rapid, simple and sensitive analytical method, based on Zeeman atomic absorption spectrometry, for the quantitative analysis of carboplatin in human plasma, plasma-ultrafiltrate (UF), saliva and urine. The sample pretreatment is simple and only involves dilution of the samples with a solution containing 0.15 mol/L NaCl and 0.2 mol/L HCl in water (diluent). The caroplatin concentrations in the different biological matrices could be quantified using a standard curve constructed in plasma UF: diluent (1:10; v/v). The validated range of determination was 0.24 to 242 μmol/L of carboplatin in plasma UF, 0.84 to 242 μmol/L in plasma, 0.12 to 3.64 μmol/L in saliva, and 2.02 μmol/L to 12.1 mmol/L in urine. Matrix-effects were not observed. The stability of carboplatin (determined as Pt) in biological samples stored at −20°C was checked for a period of 8 months; no losses occurred during this period. The performance of the method, in terms of accuracy and precision, fitted well within the generally accepted criteria for validation of analytical methodologies. The proposed method was utilized in a pharmacokinetic study in one patient to demonstrate the usefulness of the assay.


Cancer Research | 2009

Absence of Both Cytochrome P450 3A and P-glycoprotein Dramatically Increases Docetaxel Oral Bioavailability and Risk of Intestinal Toxicity

R. A. B. van Waterschoot; Jurjen S. Lagas; Els Wagenaar; C. M.M. van der Kruijssen; A. E. van Herwaarden; Ji-Ying Song; Rogier W. Rooswinkel; O. van Tellingen; Hilde Rosing; Jos H. Beijnen; Alfred H. Schinkel

Docetaxel is one of the most widely used anticancer drugs. A major problem with docetaxel treatment, however, is the considerable interpatient variability in docetaxel exposure. Another disadvantage of the drug is that it has a very low oral bioavailability and can therefore only be administered i.v. The drug-metabolizing enzyme cytochrome P450 3A (CYP3A) and the drug transporter P-glycoprotein (P-gp; MDR1) are considered to be major determinants of docetaxel pharmacokinetics. It has been hypothesized that CYP3A and P-gp work synergistically in limiting the systemic exposure to many orally ingested drugs. However, it has been difficult to examine this interplay in vivo. We therefore generated mice lacking all CYP3A and P-gp genes. Although missing two primary detoxification systems, Cyp3a/Mdr1a/1b(-/-) mice are viable, fertile, and without spontaneous abnormalities. When orally challenged with docetaxel, a disproportionate (>70-fold) increase in systemic exposure was observed compared with the increases in single Cyp3a(-/-) (12-fold) or Mdr1a/1b(-/-) (3-fold) mice. Unexpectedly, although CYP3A and P-gp collaborated extremely efficiently in lowering docetaxel exposure, their individual efficacy was not dependent on activity of the other protein. On reflection, this absence of functional synergism makes biological sense, as synergism would conflict with a robust detoxification defense. Importantly, the disproportionate increase in docetaxel exposure in Cyp3a/Mdr1a/1b(-/-) mice resulted in dramatically altered and lethal toxicity, with severe intestinal lesions as a major cause of death. Simultaneous inhibition of CYP3A/P-gp might thus be a highly effective strategy to improve oral drug bioavailability but with serious risks when applied to drugs with narrow therapeutic windows.


British Journal of Cancer | 2001

The co-solvent Cremophor EL limits absorption of orally administered paclitaxel in cancer patients

Mirte M. Malingré; Jan H. M. Schellens; O. van Tellingen; Mariët Ouwehand; Heleen A. Bardelmeijer; Hilde Rosing; Franciska J. Koopman; Margaret Schot; W.W. ten Bokkel Huinink; Jos H. Beijnen

The purpose of this study was to investigate the effect of the co-solvents Cremophor EL and polysorbate 80 on the absorption of orally administered paclitaxel. 6 patients received in a randomized setting, one week apart oral paclitaxel 60 mg m–2 dissolved in polysorbate 80 or Cremophor EL. For 3 patients the amount of Cremophor EL was 5 ml m–2, for the other three 15 ml m–2. Prior to paclitaxel administration patients received 15 mg kg–1 oral cyclosporin A to enhance the oral absorption of the drug. Paclitaxel formulated in polysorbate 80 resulted in a significant increase in the maximal concentration (Cmax) and area under the concentration–time curve (AUC) of paclitaxel in comparison with the Cremophor EL formulations (P = 0.046 for both parameters). When formulated in Cremophor EL 15 ml m–2, paclitaxel Cmax and AUC values were 0.10 ± 0.06 μM and 1.29 ± 0.99 μM h–1, respectively, whereas these values were 0.31 ± 0.06 μM and 2.61 ± 1.54 μM h–1, respectively, when formulated in polysorbate 80. Faecal data revealed a decrease in excretion of unchanged paclitaxel for the polysorbate 80 formulation compared to the Cremophor EL formulations. The amount of paclitaxel excreted in faeces was significantly correlated with the amount of Cremophor EL excreted in faeces (P = 0.019). When formulated in Cremophor EL 15 ml m–2, paclitaxel excretion in faeces was 38.8 ± 13.0% of the administered dose, whereas this value was 18.3 ±15.5% for the polysorbate 80 formulation. The results show that the co-solvent Cremophor EL is an important factor limiting the absorption of orally administered paclitaxel from the intestinal lumen. They highlight the need for designing a better drug formulation in order to increase the usefulness of the oral route of paclitaxel


Brain Imaging and Behavior | 2013

Chemotherapy-related cognitive dysfunction: current animal studies and future directions

R. Seigers; Sanne B. Schagen; O. van Tellingen; J. Dietrich

Cognitive impairment is a potential long-term side effect of adjuvant chemotherapy that can have a major impact on the quality of life of cancer survivors. There is a growing number of preclinical studies addressing this issue, thereby extending our knowledge of the mechanisms underlying chemotherapy-induced neurotoxicity. In this review, we will summarize the recent advances and important findings presented in these studies. Emerging challenges, such as the development of neuroprotective strategies, and the role of the blood-brain barrier on cognitive impairment will be described and future directions in this field of investigation will be outlined.

Collaboration


Dive into the O. van Tellingen's collaboration.

Top Co-Authors

Avatar

Jos H. Beijnen

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Jan H. M. Schellens

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Hilde Rosing

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

W.J. Nooyen

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Alfred H. Schinkel

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

R. van Gijn

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

E.M. Kemper

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

H.R. van der Woude

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Tessa Buckle

Netherlands Cancer Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge