Network


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

Hotspot


Dive into the research topics where Rolf W. Sparidans is active.

Publication


Featured researches published by Rolf W. Sparidans.


British Journal of Cancer | 2000

Vinblastine and sulfinpyrazone export by the multidrug resistance protein MRP2 is associated with glutathione export

R. Evers; M. De Haas; Rolf W. Sparidans; Jos H. Beijnen; Peter R. Wielinga; Jan Lankelma; Piet Borst

The multidrug resistance proteins MRP1 and MRP2 are members of the same subfamily of ATP-binding cassette transporters. Besides organic molecules conjugated to negatively charged ligands, these proteins also transport cytotoxic drugs for which no negatively charged conjugates are known to exist. In polarized MDCKII cells, MRP1 routes to the lateral plasma membrane, and MRP2 to the apical plasma membrane. In these cells MRP1 transports daunorubicin, and MRP2 vinblastine; both transporters export reduced glutathione (GSH) into the medium. We demonstrate that glutathione transport in MDCKII-MRP1 cells is inhibited by the inhibitors of organic anion transporters sulfinpyrazone, indomethacin, probenecid and benzbromarone. In MDCKII-MRP2 cells, GSH export is stimulated by low concentrations of sulfinpyrazone or indomethacin, whereas export is inhibited down to control levels at high concentrations. We find that unmodified sulfinpyrazone is a substrate for MRP2, also at concentrations where GSH export is inhibited. We also show that GSH export in MDCKII-MRP2 cells increases in the presence of vinblastine, and that the stochiometry between drug and GSH exported is between two and three. Our data indicate that transport of sulfinpyrazone and vinblastine is associated with GSH export. However, at high sulfinpyrazone concentrations this compound is transported without GSH. Models of MRP action are discussed that could explain these results.


Journal of Chromatography B | 2008

Quantitative bioanalysis of peptides by liquid chromatography coupled to (tandem) mass spectrometry

Irene van den Broek; Rolf W. Sparidans; Jan H. M. Schellens; Jos H. Beijnen

With the growing interest for peptides and proteins in different kinds of fields, e.g. pharmacy, clinical diagnostics or food industry, the quantification of these compounds is becoming more and more important. Quantitative analysis of these analytes in biological matrices, however, remains a challenging task, due to the complexity of both the matrix and the analytical characteristics of these large bio-molecules. Liquid chromatography coupled to (tandem) mass spectrometry (LC-MS or LC-MS/MS) is the preferred analytical technique for peptide analysis as it allows very selective and sensitive measurements. This article summarizes the numerous published LC-MS applications for the quantification of peptides in biological matrices and discusses all different issues herewith concerned. This includes chromatographic aspects as the selection and effects of mobile and stationary phase, flow rate and temperature, as well as mass spectrometric characteristics such as ionization and detection modes, collision-induced dissociation of peptides and factors influencing the mass spectrometric response. For both techniques the main properties of all described methods have been listed, creating a comprehensive overview with the peptide analytes divided into different classes. Likewise, all other issues concerned with quantitative bioanalysis have been evaluated in detail, including extensive consideration of several different applied sample pre-treatment techniques and reflection of subjects as the choice for an internal standard and assay validation. Furthermore, several issues which are of particular interest for the quantitative bioanalysis of peptide compounds like peptide adsorption and degradation have been regarded.


Molecular Cancer Therapeutics | 2010

Breast Cancer Resistance Protein and P-glycoprotein Limit Sorafenib Brain Accumulation

Jurjen S. Lagas; Robert A.B. van Waterschoot; Rolf W. Sparidans; Els Wagenaar; Jos H. Beijnen; Alfred H. Schinkel

Sorafenib is a second-generation, orally active multikinase inhibitor that is approved for the treatment of patients with advanced renal cell carcinoma and patients with unresectable hepatocellular carcinoma. We studied active transport of sorafenib in MDCK-II cells expressing human P-glycoprotein (P-gp/ABCB1) or ABCG2 (breast cancer resistance protein) or murine Abcg2. Sorafenib was moderately transported by P-gp and more efficiently by ABCG2 and Abcg2. Because sorafenib is taken orally, we orally administered sorafenib to wild-type, Abcb1a/1b−/−, Abcg2−/−, and Abcb1a/1b;Abcg2−/− mice, completely lacking functional Abcb1a/1b, Abcg2, or both, respectively, and we studied plasma pharmacokinetics and brain accumulation. The systemic exposure on oral administration was not different among all strains. However, brain accumulation was 4.3-fold increased in Abcg2−/− mice and 9.3-fold increased in Abcb1a/1b;Abcg2−/− mice. Moreover, when wild-type mice were treated with sorafenib in combination with the dual P-gp and ABCG2 inhibitor elacridar, brain accumulation was similar to that observed for Abcb1a/1b;Abcg2−/− mice. These results show that the brain accumulation of sorafenib is primarily restricted by ABCG2. This contrasts with previous studies using shared ABCG2 and P-gp substrates, which all suggested that P-gp dominates at the blood-brain barrier, and that an effect of ABCG2 is only evident when both transporters are absent. Interestingly, for sorafenib, it is the other way around, that is, ABCG2, and not P-gp, plays the dominant role in restricting its brain accumulation. Clinically, our findings may be relevant for the treatment of renal cell carcinoma patients with central nervous system relapses, as a dual ABCG2 and P-gp inhibitor might improve the central nervous system entry and thereby the therapeutic efficacy of sorafenib. Mol Cancer Ther; 9(2); 319–26


Molecular Pharmacology | 2007

Midazolam metabolism in cytochrome P450 3A knockout mice can be attributed to up-regulated CYP2C enzymes.

Robert A.B. van Waterschoot; Antonius E. van Herwaarden; Jurjen S. Lagas; Rolf W. Sparidans; Els Wagenaar; Cornelia M.M. van der Kruijssen; Joyce A. Goldstein; Darryl C. Zeldin; Jos H. Beijnen; Alfred H. Schinkel

The cytochrome P450 3A (CYP3A) enzymes represent one of the most important drug-metabolizing systems in humans. Recently, our group has generated cytochrome P450 3A knockout mice to study this drug-handling system in vivo. In the present study, we have characterized the Cyp3a knockout mice by studying the metabolism of midazolam, one of the most widely used probes to assess CYP3A activity. We expected that the midazolam metabolism would be severely reduced in the absence of CYP3A enzymes. We used hepatic and intestinal microsomal preparations from Cyp3a knockout and wild-type mice to assess the midazolam metabolism in vitro. In addition, in vivo metabolite formation was determined after intravenous administration of midazolam. We were surprised to find that our results demonstrated that there is still marked midazolam metabolism in hepatic (but not intestinal) microsomes from Cyp3a knockout mice. Accordingly, we found comparable amounts of midazolam as well as its major metabolites in plasma after intravenous administration in Cyp3a knockout mice compared with wild-type mice. These data suggested that other hepatic cytochrome P450 enzymes could take over the midazolam metabolism in Cyp3a knockout mice. We provide evidence that CYP2C enzymes, which were found to be up-regulated in Cyp3a knockout mice, are primarily responsible for this metabolism and that several but not all murine CYP2C enzymes are capable of metabolizing midazolam to its 1′-OH and/or 4-OH derivatives. These data illustrate interesting compensatory changes that may occur in Cyp3a knockout mice. Such flexible compensatory interplay between functionally related detoxifying systems is probably essential to their biological role in xenobiotic protection.


International Journal of Cancer | 2014

Increased oral availability and brain accumulation of the ALK inhibitor crizotinib by coadministration of the P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) inhibitor elacridar

Seng Chuan Tang; Luan N. Nguyen; Rolf W. Sparidans; Els Wagenaar; Jos H. Beijnen; Alfred H. Schinkel

Crizotinib is an oral tyrosine kinase inhibitor approved for treating patients with non‐small cell lung cancer (NSCLC) containing an anaplastic lymphoma kinase (ALK) rearrangement. We used knockout mice to study the roles of P‐glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) in plasma pharmacokinetics and brain accumulation of oral crizotinib, and the feasibility of improving crizotinib kinetics using coadministration of the dual ABCB1/ABCG2 inhibitor elacridar. In vitro, crizotinib was a good transport substrate of human ABCB1, but not of human ABCG2 or murine Abcg2. With low‐dose oral crizotinib (5 mg/kg), Abcb1a/1b−/− and Abcb1a/1b;Abcg2−/− mice had an approximately twofold higher plasma AUC than wild‐type mice, and a markedly (∼40‐fold) higher brain accumulation at 24 hr. Also at 4 hr, crizotinib brain concentrations were ∼25‐fold, and brain‐to‐plasma ratios ∼14‐fold higher in Abcb1a/1b−/− and Abcb1a/1b;Abcg2−/− mice than in wild‐type mice. High‐dose oral crizotinib (50 mg/kg) resulted in comparable plasma pharmacokinetics between wild‐type and Abcb1a/1b−/− mice, suggesting saturation of intestinal Abcb1. Nonetheless, brain accumulation at 24 hr was still ∼70‐fold higher in Abcb1a/1b−/− than in wild‐type mice. Importantly, oral elacridar coadministration increased the plasma and brain concentrations and brain‐to‐plasma ratios of crizotinib in wild‐type mice, equaling the levels in Abcb1a/1b;Abcg2−/− mice. Our results indicate that crizotinib oral availability and brain accumulation were primarily restricted by Abcb1 at a non‐saturating dose, and that coadministration of elacridar with crizotinib could substantially increase crizotinib oral availability and delivery to the brain. This principle might be used to enhance therapeutic efficacy of crizotinib against brain metastases in NSCLC patients.


Molecular Pharmaceutics | 2012

Oral availability and brain penetration of the B-RAFV600E inhibitor vemurafenib can be enhanced by the P-GLYCOprotein (ABCB1) and breast cancer resistance protein (ABCG2) inhibitor elacridar.

Selvi Durmus; Rolf W. Sparidans; Els Wagenaar; Jos H. Beijnen; Alfred H. Schinkel

Vemurafenib (PLX4032) is a novel tyrosine kinase inhibitor that has clinical efficacy against metastatic melanoma harboring a BRAF(V600E) mutation. We aimed to establish whether oral availability and brain penetration of vemurafenib could be restricted by the multidrug efflux transporters P-glycoprotein (P-gp/ABCB1) and breast cancer resistance protein (BCRP/ABCG2), as these might limit therapeutic efficacy, especially against brain metastases. In vitro, vemurafenib was efficiently transported by both human ABCB1 and ABCG2, and very efficiently by mouse Abcg2, but not by mouse Abcc2. Upon oral administration of vemurafenib (5 mg/kg), Abcb1a/1b(-/-) mice had a 1.6-fold increased, Abcg2(-/-) mice a 2.3-fold increased, and Abcb1a/1b(-/-);Abcg2(-/-) mice a 6.6-fold increased plasma AUC, respectively, compared to wild-type (WT) mice, indicating a marked and additive role of these transporters in limiting vemurafenib oral availability. Brain-to-plasma ratios of vemurafenib (oral, 25 mg/kg) were not increased in Abcg2(-/-) mice, only 1.7-fold in Abcb1a/1b(-/-) mice, but 21.4-fold in Abcb1a/1b(-/-);Abcg2(-/-) mice, indicating pronounced overlapping functions of these transporters in reducing vemurafenib brain accumulation. Oral coadministration of the dual ABCB1 and ABCG2 inhibitor elacridar almost completely eliminated the roles of Abcb1 and Abcg2 in restricting oral availability and brain accumulation of vemurafenib. As predicted by previously described pharmacokinetic modeling, halving the amount of active efflux transport at the WT blood-brain barrier by testing heterozygous Abcb1a/1b(+/-);Abcg2(+/-) mice had little impact on vemurafenib brain accumulation. Our data suggest that elacridar coadministration may be considered to improve the therapeutic efficacy of vemurafenib, especially for brain metastases located behind a functional blood-brain barrier.


Journal of Chromatography B: Biomedical Sciences and Applications | 1999

Quantitative determination of efavirenz (DMP 266), a novel non-nucleoside reverse transcriptase inhibitor, in human plasma using isocratic reversed-phase high-performance liquid chromatography with ultraviolet detection.

Agnes I. Veldkamp; Rolf W. Sparidans; Richard M. W. Hoetelmans; Jos H. Beijnen

Efavirenz is a novel non-nucleoside reverse transcriptase inhibitor for the treatment of HIV-1-infected individuals. A simple and rapid high-performance liquid chromatographic method for the quantification of efavirenz in human plasma suitable for therapeutic drug monitoring in plasma is described. Sample pretreatment consists of protein precipitation with acetonitrile and subsequent evaporation of the extract to concentrate the analyte. The drug is separated from endogenous compounds by isocratic reversed-phase high-performance liquid chromatography with ultraviolet detection at 246 nm. The method has been validated over the range of 10 to 10,000 ng/ml using a volume of 250 microl of plasma. The assay is linear over this concentration range as indicated by the F-test for lack of fit. Within- and between-day precisions are less than 4.3% for all quality control samples. The lower limit of quantitation is 10 ng/ml and the recovery of efavirenz from human plasma is 106.4% (+/- 1.8%). Frequently co-administered drugs did not interfere with the described methodology. Efavirenz is stable under various relevant storage conditions, for example when stored for 24 h at room temperature. This validated assay is suited for use in pharmacokinetic studies with efavirenz and can readily be implemented in the setting of a hospital laboratory for the monitoring of efavirenz concentrations.


Pharmacy World & Science | 1998

BISPHOSPHONATES IN BONE DISEASES

Rolf W. Sparidans; Irene M. Twiss; Stephanie Talbot

Bisphosphonates are a class of drugs which are strongly attracted to the bone where they influence the calcium metabolism, mainly by inhibition of the osteoclast-mediated bone resorption. This property makes these compounds suited for the treatment of several diseases of the bone. In Pagets disease, several bisphosphonates can reduce bone pain and decrease the bone turnover 60‐70%. Cyclical oral etidronate and daily oral alendronate both proved to reduce the vertebral fracture rate for postmenopausal osteoporotic woman, while most investigated bisphosphonates can increase spinal bone mass in osteoporosis. Bisphosphonates can help lowering serum calcium and reverse skeletal complications in malignancy mediated bone diseases. Oral and intravenous administration of therapeutic doses is relatively safe. In general, gastrointestinal disturbances are described most often and the oldest, least potent, bisphosphonate etidronate can induce osteomalacia. The various characteristics of bisphosphonates: physicochemical, biological, therapeutic and toxicological, vary greatly depending on the structure of the individual bisphosphonate. Even small changes in the structure can lead to enormous differences in potency. Overall, this class of drugs offers several prospects for the future.


Gut | 2014

Collagen degradation and neutrophilic infiltration: a vicious circle in inflammatory bowel disease

Pim J. Koelink; Saskia A. Overbeek; Saskia Braber; Mary E. Morgan; P. A. J. Henricks; Mojtaba Abdul Roda; Hein W. Verspaget; Simone C. Wolfkamp; Anje A. te Velde; Caleb Jones; Patricia L. Jackson; J. Edwin Blalock; Rolf W. Sparidans; John A. W. Kruijtzer; Johan Garssen; Gert Folkerts; Aletta D. Kraneveld

Objective Proline–glycine–proline (PGP) has been shown to have chemotactic effects on neutrophils via CXCR2 in several lung diseases. PGP is derived from collagen by the combined action of matrix metalloproteinase (MMP) 8 and/or MMP9 and prolyl endopeptidase (PE). We investigated the role of PGP in inflammatory bowel disease (IBD). Design In intestinal tissue from patients with IBD and mice with dextran sodium sulfate (DSS)-induced colitis, MMP8, MMP9 and PE were evaluated by ELISA, immunoblot and immunohistochemistry. Peripheral blood polymorphonuclear cell (PMN) supernatants were also analysed accordingly and incubated with collagen to assess PGP generation ex vivo. PGP levels were measured by mass spectrometry, and PGP neutralisation was achieved with a PGP antagonist and PGP antibodies. Results In the intestine of patients with IBD, MMP8 and MMP9 levels were elevated, while PE was expressed at similar levels to control tissue. PGP levels were increased in intestinal tissue of patients with IBD. Similar results were obtained in intestine from DSS-treated mice. PMN supernatants from patients with IBD were far more capable of generating PGP from collagen ex vivo than healthy controls. Furthermore, PGP neutralisation during DSS-induced colitis led to a significant reduction in neutrophil infiltration in the intestine. Conclusions The proteolytic cascade that generates PGP from collagen, as well as the tripeptide itself, is present in the intestine of patients with IBD and mice with DSS-induced colitis. PGP neutralisation in DSS-treated mice showed the importance of PGP-guided neutrophilic infiltration in the intestine and indicates a vicious circle in neutrophilic inflammation in IBD.


Drug Metabolism and Disposition | 2011

Differential Impact of P-Glycoprotein (ABCB1) and Breast Cancer Resistance Protein (ABCG2) on Axitinib Brain Accumulation and Oral Plasma Pharmacokinetics

Birk Poller; Dilek Iusuf; Rolf W. Sparidans; Els Wagenaar; Jos H. Beijnen; Alfred H. Schinkel

The second-generation tyrosine kinase inhibitor and anticancer drug axitinib is a potent, orally active inhibitor of the vascular endothelial growth factor receptors 1, 2, and 3. Axitinib has clinical activity against solid tumors such as metastatic renal cell carcinoma and advanced pancreatic cancer. We studied axitinib transport using Madin-Darby canine kidney II cells overexpressing human ABCB1 or ABCG2 or murine Abcg2. Axitinib was a good substrate of ABCB1 and Abcg2, whereas transport activity by ABCG2 was moderate. These transporters may therefore contribute to axitinib resistance in tumor cells. Upon oral administration of axitinib, Abcg2(−/−) and Abcb1a/1b;Abcg2(−/−) mice displayed 1.7- and 1.8-fold increased axitinib areas under the plasma concentration-time curve from 0 to 4 compared with those of wild-type mice. Plasma concentrations in Abcb1a/1b(−/−) mice were not significantly increased. In contrast, relative brain accumulation of axitinib in Abcb1a/1b(−/−) and Abcb1a/1b;Abcg2(−/−) mice was, respectively, 6.8- and 13.9-fold higher than that in wild-type mice at 1 h and 4.9- and 20.7-fold at 4 h after axitinib administration. In Abcg2(−/−) mice, we found no significant differences in brain accumulation compared with those in wild-type mice. Thus, Abcb1 strongly restricts axitinib brain accumulation and completely compensates for the loss of Abcg2 at the blood-brain barrier, whereas Abcg2 can only partially take over Abcb1-mediated axitinib efflux. Hence, Abcg2 has a stronger impact on axitinib oral plasma pharmacokinetics, whereas Abcb1 is the more important transporter at the blood-brain barrier. These findings illustrate that in vitro transport data for ABCB1 and ABCG2 cannot always be simply extrapolated to the prediction of the relative impact of these transporters on oral availability versus brain penetration.

Collaboration


Dive into the Rolf W. Sparidans'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

Alfred H. Schinkel

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Els Wagenaar

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Jan den Hartigh

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Selvi Durmus

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pieter Vermeij

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alwin D. R. Huitema

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge