129 results on '"Greupink, R."'
Search Results
102. Microbial Glucuronidase Inhibition Reduces Severity of Diclofenac-Induced Anastomotic Leak in Rats.
- Author
-
Yauw STK, Arron M, Lomme RMLM, van den Broek P, Greupink R, Bhatt AP, Redinbo MR, and van Goor H
- Subjects
- Animals, Disease Models, Animal, Male, Rats, Wistar, Treatment Outcome, Anastomotic Leak pathology, Anastomotic Leak prevention & control, Anti-Inflammatory Agents, Non-Steroidal adverse effects, Diclofenac adverse effects, Enzyme Inhibitors administration & dosage, Glucuronidase antagonists & inhibitors
- Abstract
Background: The non-steroidal anti-inflammatory drug diclofenac has been associated with intestinal anastomotic leakage, although the underlying pathophysiology is unclear. Previous data suggest that reactivation of biliary diclofenac metabolites by microbial β-glucuronidases in the gut plays a role in harming the intestinal mucosa, and that microbiome-targeted glucuronidase inhibition prevents this damage. Here, the microbial glucuronidase inhibitor Inh1 was examined for its ability to reduce diclofenac-induced anastomotic leakage in rats., Methods: Ninety male Wistar rats were allocated to five groups. In the two diclofenac groups, group DCF received diclofenac (3 mg/kg per day) and group DCF-Inh1 additionally received 800 mcg/kg per day of glucuronidase inhibitor Inh1 solution orally. In non-diclofenac groups, animals received either Inh1 (800 mcg/kg per day; group Inh1) solution, the vehicle (methylcellulose; group Veh), or no solution (group Ctrl). All solutions were provided from the day of surgery until sacrifice on day three. Plasma concentrations of diclofenac were determined. Outcomes were anastomotic leakage, leak severity, and anastomotic strength., Results: Anastomotic leak rates were 89% in group DCF and 44% in group DCF-Inh1 (p = 0.006). Leak severity was reduced in group DCFic-Inh1 (p = 0.029). In non-diclofenac cohorts, mostly minor leakage signs were observed in 25% in group Ctrl, 39% in group Inh1 (0.477), and 24% in group Veh (p = 1.000). Bursting pressure and breaking strength were not significantly different. Plasma concentrations of diclofenac were not changed by Inh1., Conclusion: Microbial glucuronidase inhibitor reduces diclofenac-induced anastomotic leakage severity, which suggests a harmful effect of diclofenac metabolite reactivation in the gut. This finding improves the understanding of the pathogenesis of anastomotic leakage.
- Published
- 2018
- Full Text
- View/download PDF
103. Development of a mechanistic biokinetic model for hepatic bile acid handling to predict possible cholestatic effects of drugs.
- Author
-
Notenboom S, Weigand KM, Proost JH, van Lipzig MMH, van de Steeg E, van den Broek PHH, Greupink R, Russel FGM, and Groothuis GMM
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 11 metabolism, Biological Transport drug effects, Cell Line, HEK293 Cells, Humans, Kinetics, Membrane Transport Proteins metabolism, Organic Anion Transporters, Sodium-Dependent metabolism, Symporters metabolism, Bile Acids and Salts metabolism, Chemical and Drug Induced Liver Injury metabolism, Cholestasis chemically induced, Cholestasis metabolism, Liver metabolism, Pharmaceutical Preparations metabolism
- Abstract
Drug-induced liver injury (DILI) is a common reason for drug withdrawal from the market. An important cause of DILI is drug-induced cholestasis. One of the major players involved in drug-induced cholestasis is the bile salt efflux pump (BSEP; ABCB11). Inhibition of BSEP by drugs potentially leads to cholestasis due to increased (toxic) intrahepatic concentrations of bile acids with subsequent cell injury. In order to investigate the possibilities for in silico prediction of cholestatic effects of drugs, we developed a mechanistic biokinetic model for human liver bile acid handling populated with human in vitro data. For this purpose we considered nine groups of bile acids in the human bile acid pool, i.e. chenodeoxycholic acid, deoxycholic acid, the remaining unconjugated bile acids and the glycine and taurine conjugates of each of the three groups. Michaelis-Menten kinetics of the human uptake transporter Na
+ -taurocholate cotransporting polypeptide (NTCP; SLC10A1) and BSEP were measured using NTCP-transduced HEK293 cells and membrane vesicles from BSEP-overexpressing HEK293 cells. For in vitro-in vivo scaling, transporter abundance was determined by LC-MS/MS in these HEK293 cells and vesicles as well as in human liver tissue. Other relevant human kinetic parameters were collected from literature, such as portal bile acid levels and composition, bile acid synthesis and amidation rate. Additional empirical scaling was applied by increasing the excretion rate with a factor 2.4 to reach near physiological steady-state intracellular bile acid concentrations (80μM) after exposure to portal vein bile acid levels. Simulations showed that intracellular bile acid concentrations increase 1.7 fold in the presence of the BSEP inhibitors and cholestatic drugs cyclosporin A or glibenclamide, at intrahepatic concentrations of 6.6 and 20μM, respectively. This simplified model provides a tool for a first indication whether drugs at therapeutic concentrations might cause cholestasis by inhibiting BSEP., (Copyright © 2018 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
104. Drug Dosing in Pregnant Women: Challenges and Opportunities in Using Physiologically Based Pharmacokinetic Modeling and Simulations.
- Author
-
Ke AB, Greupink R, and Abduljalil K
- Subjects
- Animals, Dose-Response Relationship, Drug, Female, Humans, Maternal-Fetal Exchange, Milk, Human metabolism, Pharmaceutical Preparations metabolism, Pregnancy, Models, Biological, Pharmaceutical Preparations administration & dosage, Pharmacokinetics
- Abstract
The unmet medical need of providing evidence-based pharmacotherapy for pregnant women is recognized by the regulatory bodies. Physiologically based pharmacokinetic (PBPK) modeling offers an attractive platform to quantify anticipated changes in the pharmacokinetics (PKs) of drugs during pregnancy. Recent publications applying a pregnancy PBPK module to the prediction of maternal and fetal exposure of drugs are summarized. Future opportunities to use PBPK models to predict breast milk exposure and assess human fetotoxicity risks are presented., (© 2018 The Authors CPT: Pharmacometrics & Systems Pharmacology published by Wiley Periodicals, Inc. on behalf of American Society for Clinical Pharmacology and Therapeutics.)
- Published
- 2018
- Full Text
- View/download PDF
105. Free dug concentrations in pregnancy: Bound to measure unbound?
- Author
-
Schalkwijk S, Greupink R, and Burger D
- Subjects
- Female, Humans, Pregnancy, Protein Binding, Serum Albumin
- Published
- 2017
- Full Text
- View/download PDF
106. Rat precision-cut liver slices predict drug-induced cholestatic injury.
- Author
-
Starokozhko V, Greupink R, van de Broek P, Soliman N, Ghimire S, de Graaf IAM, and Groothuis GMM
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 11 genetics, Animals, Bile Acids and Salts metabolism, Cholestasis chemically induced, Gene Expression Regulation drug effects, Liver metabolism, Liver pathology, Male, Organic Anion Transporters, Sodium-Dependent genetics, Rats, Wistar, Symporters genetics, Chemical and Drug Induced Liver Injury etiology, Liver drug effects, Organ Culture Techniques methods, Toxicity Tests methods
- Abstract
Drug-induced cholestasis (DIC) is one of the leading manifestations of drug-induced liver injury (DILI). As the underlying mechanisms for DIC are not fully known and specific and predictive biomarkers and pre-clinical models are lacking, the occurrence of DIC is often only reported when the drug has been approved for registration. Therefore, appropriate models that predict the cholestatic potential of drug candidates and/or provide insight into the mechanism of DIC are highly needed. We investigated the application of rat precision-cut liver slices (PCLS) to predict DIC, using several biomarkers of cholestasis: hepatocyte viability, intracellular accumulation of total as well as individual bile acids and changes in the expression of genes known to play a role in cholestasis. Rat PCLS exposed to the cholestatic drugs chlorpromazine, cyclosporine A and glibenclamide for 48 h in the presence of a 60 μM physiological bile acid (BA) mix reflected various changes associated with cholestasis, such as decrease in hepatocyte viability, accumulation and changes in the composition of BA and changes in the gene expression of Fxr, Bsep and Ntcp. The toxicity of the drugs was correlated with the accumulation of BA, and especially DCA and CDCA and their conjugates, but to a different extent for different drugs, indicating that BA toxicity is not the only cause for the toxicity of cholestatic drugs. Moreover, our study supports the use of several biomarkers to test drugs for DIC. In conclusion, our results indicate that PCLS may represent a physiological and valuable model to identify cholestatic drugs and provide insight into the mechanisms underlying DIC.
- Published
- 2017
- Full Text
- View/download PDF
107. Therapeutic effects of the mitochondrial ROS-redox modulator KH176 in a mammalian model of Leigh Disease.
- Author
-
de Haas R, Das D, Garanto A, Renkema HG, Greupink R, van den Broek P, Pertijs J, Collin RWJ, Willems P, Beyrath J, Heerschap A, Russel FG, and Smeitink JA
- Subjects
- Animals, Brain ultrastructure, Chromans chemistry, Diffusion Tensor Imaging methods, Gait drug effects, Mice, Mice, Knockout, Mitochondrial Diseases drug therapy, Neuroimaging, Reactive Oxygen Species metabolism, Brain diagnostic imaging, Chromans therapeutic use, Electron Transport Complex I genetics, Leigh Disease drug therapy
- Abstract
Leigh Disease is a progressive neurometabolic disorder for which a clinical effective treatment is currently still lacking. Here, we report on the therapeutic efficacy of KH176, a new chemical entity derivative of Trolox, in Ndufs4
-/- mice, a mammalian model for Leigh Disease. Using in vivo brain diffusion tensor imaging, we show a loss of brain microstructural coherence in Ndufs4-/- mice in the cerebral cortex, external capsule and cerebral peduncle. These findings are in line with the white matter diffusivity changes described in mitochondrial disease patients. Long-term KH176 treatment retained brain microstructural coherence in the external capsule in Ndufs4-/- mice and normalized the increased lipid peroxidation in this area and the cerebral cortex. Furthermore, KH176 treatment was able to significantly improve rotarod and gait performance and reduced the degeneration of retinal ganglion cells in Ndufs4-/- mice. These in vivo findings show that further development of KH176 as a potential treatment for mitochondrial disorders is worthwhile to pursue. Clinical trial studies to explore the potency, safety and efficacy of KH176 are ongoing.- Published
- 2017
- Full Text
- View/download PDF
108. Editor's Highlight: PlacentalDisposition and Effects of Crizotinib: An Ex Vivo Study in the Isolated Dual-Side Perfused Human Cotyledon.
- Author
-
Eliesen GAM, van den Broek P, van den Heuvel JJ, Bilos A, Pertijs J, van Drongelen J, Russel FGM, and Greupink R
- Subjects
- Biological Transport, Cell Line, Tumor, Cell Membrane drug effects, Cell Membrane metabolism, Cell Survival drug effects, Crizotinib, Female, HEK293 Cells, Humans, In Vitro Techniques, Maternal-Fetal Exchange drug effects, Models, Biological, Perfusion, Placenta drug effects, Pregnancy, Time Factors, Tissue Distribution, Placenta metabolism, Protein Kinase Inhibitors pharmacokinetics, Protein Kinase Inhibitors toxicity, Pyrazoles pharmacokinetics, Pyrazoles toxicity, Pyridines pharmacokinetics, Pyridines toxicity
- Abstract
Tyrosine kinase inhibitors (TKIs) play an important role in cancer pharmacotherapy, yet there is limited data on their use during pregnancy. We studied placental disposition and placental toxicity of crizotinib, a TKI used to treat nonsmall cell lung cancer. Term placentas were perfused for 3 h with crizotinib (1 µM) using the ex vivo dual-side cotyledon perfusion technique. Interference of TKIs with trophoblast viability was studied using BeWo cells. Expression of P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP) in placental tissue was assessed by immunohistochemistry and inhibition of these transporters was determined in vitro by transport studies with membrane vesicles overexpressing human P-gp or BCRP. We found that crizotinib rapidly and strongly accumulates in cotyledon perfusion experiments, reaching a concentration of 3.1 ± 0.4 µM in placental tissue. Final drug concentrations in the maternal and foetal reservoirs were 0.2 ± 0.05 and 0.08 ± 0.01 µM, respectively. Furthermore, crizotinib inhibited BeWo cell viability (IC50: 234 nM, 95% CI: 167-328 nM) 10 times more potently than other TKIs tested. In vitro transport studies revealed that crizotinib is a potent inhibitor of the transport activities of BCRP (IC50: 5.7 µM, 95% CI: 2.7-11.8 µM) and P-gp (IC50: 7.8 µM, 95% CI: 3.4-18.0 µM). In conclusion, crizotinib strongly accumulated in placental tissue at clinically relevant concentrations. IC50 values for transporter inhibition and trophoblast cell viability were similar to the tissue concentrations reached, suggesting that crizotinib can inhibit placental BCRP and P-gp function and possibly affect trophoblast viability., (© The Author 2017. Published by Oxford University Press on behalf of the Society of Toxicology. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2017
- Full Text
- View/download PDF
109. Substantially lowered dolutegravir exposure in a treatment-experienced perinatally HIV-1-infected pregnant woman.
- Author
-
Schalkwijk S, Feiterna-Sperling C, Weizsäcker K, Colbers A, Bührer C, Greupink R, Russel FG, and Burger D
- Subjects
- Female, Humans, Oxazines, Piperazines, Pregnancy, Pyridones, HIV Infections drug therapy, HIV Integrase Inhibitors administration & dosage, HIV-1, Heterocyclic Compounds, 3-Ring administration & dosage, Pregnancy Complications, Infectious drug therapy
- Published
- 2016
- Full Text
- View/download PDF
110. First reported use of elvitegravir and cobicistat during pregnancy.
- Author
-
Schalkwijk S, Colbers A, Konopnicki D, Greupink R, Russel FG, and Burger D
- Subjects
- Adult, Anti-HIV Agents pharmacokinetics, Cobicistat pharmacokinetics, Female, Humans, Plasma chemistry, Pregnancy, Quinolones pharmacokinetics, Anti-HIV Agents therapeutic use, Cobicistat therapeutic use, HIV Infections drug therapy, Pregnancy Complications, Infectious drug therapy, Quinolones therapeutic use
- Published
- 2016
- Full Text
- View/download PDF
111. Physiologically Based Modelling of Darunavir/Ritonavir Pharmacokinetics During Pregnancy.
- Author
-
Colbers A, Greupink R, Litjens C, Burger D, and Russel FG
- Subjects
- Adult, Cytochrome P-450 CYP3A Inhibitors pharmacology, Darunavir blood, Female, HIV Infections drug therapy, HIV Infections metabolism, HIV Protease Inhibitors blood, HIV Protease Inhibitors pharmacology, Humans, Liver metabolism, Ritonavir pharmacology, Young Adult, Darunavir pharmacokinetics, HIV Protease Inhibitors pharmacokinetics, Models, Biological, Pregnancy metabolism, Ritonavir pharmacokinetics
- Abstract
Pregnant women are usually excluded from clinical trials. Physiologically based pharmacokinetic (PBPK) modelling may provide a method to predict pharmacokinetics in pregnant women, without the need to perform extensive in vivo clinical trials. Here, we used mechanistic modelling to delineate the potential impact of drug transporters on darunavir pharmacokinetics and to identify current knowledge gaps that limit accurate PBPK modelling of darunavir/ritonavir (darunavir/r) exposure in pregnancy. Simcyp (version 13.2) was used for PBPK modelling, using physicochemical and in vitro pharmacokinetic parameters of darunavir and ritonavir from the literature. The Michaelis-Menten constant (K m) and the maximum rate of metabolite formation (V max) for cytochrome P450 3A4-mediated darunavir biotransformation and inhibition by ritonavir were determined experimentally, while the contributions of hepatocyte influx and efflux transporters were assessed by sensitivity analysis. The simulations were compared with previously published clinical pharmacokinetic data. We found that use of a well-stirred liver model overestimated darunavir exposure substantially. A permeability-limited liver model, including hepatic uptake and efflux transporters and an efficient enterohepatic circulation step, resulted in an acceptable description of darunavir/r exposure. For the 600/100 mg darunavir/r twice-daily dose and the 800/100 mg once-daily dose, the estimated pharmacokinetic parameters were within a 2-fold range of the reported data. The predicted decreases in the area under the concentration-time curve (AUC) values during pregnancy for the twice- and once-daily doses were 27 and 41%, respectively, which were in line with the observed decreases of 17-22 and 33%. In conclusion, our data support a clinically relevant role of hepatic transporters in darunavir pharmacokinetics. By including them in our model, we successfully approximated the increase in darunavir exposure mediated by ritonavir co-administration and the decrease in darunavir exposure observed during pregnancy.
- Published
- 2016
- Full Text
- View/download PDF
112. Placental transfer of the HIV integrase inhibitor dolutegravir in an ex vivo human cotyledon perfusion model.
- Author
-
Schalkwijk S, Greupink R, Colbers AP, Wouterse AC, Verweij VG, van Drongelen J, Teulen M, van den Oetelaar D, Burger DM, and Russel FG
- Subjects
- Chromatography, Liquid, Female, Humans, Oxazines, Perfusion, Piperazines, Pregnancy, Pyridones, Tandem Mass Spectrometry, Cotyledon metabolism, HIV Integrase Inhibitors pharmacokinetics, Heterocyclic Compounds, 3-Ring pharmacokinetics, Models, Biological, Placenta metabolism
- Abstract
Objectives: Data on fetal exposure to antiretroviral agents during pregnancy are important to estimate their potential for prevention of mother-to-child transmission (PMTCT) and possible toxicity. For the recently developed HIV integrase inhibitor dolutegravir, clinical data on fetal disposition are not yet available. Dual perfusion of a single placental lobule (cotyledon) provides a useful ex vivo model to predict the in vivo maternal-to-fetal transfer of this drug. The aim of this study was to estimate the transfer of dolutegravir across the human term placenta, using a dual-perfusion cotyledon model., Methods: After cannulation of the cotyledons (n = 6), a fetal circulation of 6 mL/min and maternal circulation of 12 mL/min were initiated. The perfusion medium consisted of Krebs-Henseleit buffer (pH = 7.2-7.4) supplemented with 10.1 mM glucose, 30 g/L human serum albumin and 0.5 mL/L heparin 5000IE. Dolutegravir was administered to the maternal circulation (∼ 4.2 mg/L) and analysed by UPLC-MS/MS., Results: After 3 h of perfusion, the mean ± SD fetal-to-maternal (FTM) concentration ratio of dolutegravir was 0.6 ± 0.2 and the mean ± SD concentrations in the maternal and fetal compartments were 2.3 ± 0.4 and 1.3 ± 0.3 mg/L, respectively., Conclusions: Dolutegravir crosses the blood-placental barrier with a mean FTM concentration ratio of 0.6. Compared with other antiretroviral agents, placental transfer of dolutegravir is moderate to high. These data suggest that dolutegravir holds clinical potential for pre-exposure prophylaxis and consequently PMTCT, but also risk of fetal toxicity., (© The Author 2015. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
113. Convallatoxin: a new P-glycoprotein substrate.
- Author
-
Gozalpour E, Greupink R, Bilos A, Verweij V, van den Heuvel JJ, Masereeuw R, Russel FG, and Koenderink JB
- Subjects
- Animals, Biological Transport physiology, Brain metabolism, Cell Line, Digoxin metabolism, HEK293 Cells, Humans, Kidney Cortex metabolism, Male, Membrane Transport Proteins metabolism, Rats, Rats, Wistar, ATP Binding Cassette Transporter, Subfamily B metabolism, Strophanthins metabolism
- Abstract
Digitalis-like compounds (DLCs), such as digoxin and digitoxin that are derived from digitalis species, are currently used to treat heart failure and atrial fibrillation, but have a narrow therapeutic index. Drug-drug interactions at the transporter level are frequent causes of DLCs toxicity. P-glycoprotein (P-gp, ABCB1) is the primary transporter of digoxin and its inhibitors influence pharmacokinetics and disposition of digoxin in the human body; however, the involvement of P-gp in the disposition of other DLCs is currently unknown. In present study, the transport of fourteen DLCs by human P-gp was studied using membrane vesicles originating from human embryonic kidney (HEK293) cells overexpressing P-gp. DLCs were quantified by liquid chromatography-mass spectrometry (LC-MS). The Lily of the Valley toxin, convallatoxin, was identified as a P-gp substrate (Km: 1.1±0.2 mM) in the vesicular assay. Transport of convallatoxin by P-gp was confirmed in rat in vivo, in which co-administration with the P-gp inhibitor elacridar, resulted in increased concentrations in brain and kidney cortex. To address the interaction of convallatoxin with P-gp on a molecular level, the effect of nine alanine mutations was compared with the substrate N-methyl quinidine (NMQ). Phe343 appeared to be more important for transport of NMQ than convallatoxin, while Val982 was particularly relevant for convallatoxin transport. We identified convallatoxin as a new P-gp substrate and recognized Val982 as an important amino acid involved in its transport. These results contribute to a better understanding of the interaction of DLCs with P-gp., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
114. Interaction of digitalis-like compounds with liver uptake transporters NTCP, OATP1B1, and OATP1B3.
- Author
-
Gozalpour E, Greupink R, Wortelboer HM, Bilos A, Schreurs M, Russel FG, and Koenderink JB
- Subjects
- Animals, Biological Transport physiology, CHO Cells, Cell Line, Cricetinae, Cricetulus, HEK293 Cells, Humans, Liver-Specific Organic Anion Transporter 1, Membrane Transport Proteins metabolism, Solute Carrier Organic Anion Transporter Family Member 1B3, Digitalis metabolism, Liver metabolism, Organic Anion Transporters metabolism, Organic Anion Transporters, Sodium-Dependent metabolism, Organic Anion Transporters, Sodium-Independent metabolism, Symporters metabolism
- Abstract
Digitalis-like compounds (DLCs) such as digoxin, digitoxin, and ouabain, also known as cardiac glycosides, are among the oldest pharmacological treatments for heart failure. The compounds have a narrow therapeutic window, while at the same time, DLC pharmacokinetics is prone to drug-drug interactions at the transport level. Hepatic transporters organic anion transporting polypeptide (OATP) 1B1, OATP1B3, and Na(+)-dependent taurocholate co-transporting polypeptide (NTCP) influence the disposition of a variety of drugs by mediating their uptake from blood into hepatocytes. The interaction of digoxin, digitoxin, and ouabain with hepatic uptake transporters has been studied before. However, here, we systematically investigated a much wider range of structurally related DLCs for their capability to inhibit or to be transported by these transporters in order to better understand the relation between the activity and chemical structure of this compound type. We studied the uptake and inhibitory potency of a series of 14 structurally related DLCs in Chinese hamster ovary cells expressing NTCP (CHO-NTCP) and human embryonic kidney cells expressing OATP1B1 and OATP1B3 (HEK-OATP1B1 and HEK-OATP1B3). The inhibitory effect of the DLCs was measured against taurocholic acid (TCA) uptake in CHO-NTCP cells and against uptake of β-estradiol 17-β-d-glucuronide (E217βG) in HEK-OATP1B1 and HEK-OATP1B3 cells. Proscillaridin A was the most effective inhibitor of NTCP-mediated TCA transport (IC50 = 22 μM), whereas digitoxin and digitoxigenin were the most potent inhibitors of OATP1B1 and OAPTP1B3, with IC50 values of 14.2 and 36 μM, respectively. Additionally, we found that the sugar moiety and hydroxyl groups of the DLCs play different roles in their interaction with NTCP, OATP1B1, and OATP1B3. The sugar moiety decreases the inhibition of NTCP and OATP1B3 transport activity, whereas it enhances the inhibitory potency against OATP1B1. Moreover, the hydroxyl group at position 12 reinforces the inhibition of NTCP but decreases the inhibition of OATP1B1 and OATP1B3. To investigate whether DLCs can be translocated, we quantified their uptake in transporter-expressing cells by LC-MS. We demonstrated that convallatoxin, ouabain, dihydroouabain, and ouabagenin are substrates of OATP1B3. No transport was observed for the other compounds in any of the studied transporters. In summary, this work provides a step toward an improved understanding of the interaction of DLCs with three major hepatic uptake transporters. Ultimately, this can be of use in the development of DLCs that are less prone to transporter-mediated drug-drug interactions.
- Published
- 2014
- Full Text
- View/download PDF
115. In silico identification and in vitro validation of potential cholestatic compounds through 3D ligand-based pharmacophore modeling of BSEP inhibitors.
- Author
-
Ritschel T, Hermans SM, Schreurs M, van den Heuvel JJ, Koenderink JB, Greupink R, and Russel FG
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 11, Computer Simulation, HEK293 Cells, Humans, Ligands, Models, Molecular, Taurocholic Acid metabolism, ATP-Binding Cassette Transporters antagonists & inhibitors, ATP-Binding Cassette Transporters metabolism, Drug Design
- Abstract
Drug-induced cholestasis is a frequently observed side effect of drugs and is often caused by an unexpected interaction with the bile salt export pump (BSEP/ABCB11). BSEP is the key membrane transporter responsible for the transport of bile acids from hepatocytes into bile. Here, we developed a pharmacophore model that describes the molecular features of compounds associated with BSEP inhibitory activity. To generate input and validation data sets, in vitro experiments with membrane vesicles overexpressing human BSEP were used to assess the effect of compounds (50 μM) on BSEP-mediated (3)H-taurocholic acid transport. The model contains two hydrogen bond acceptor/anionic features, two hydrogen bond acceptor vector features, four hydrophobic/aromatic features, and exclusion volumes. The pharmacophore was validated against a set of 59 compounds, including registered drugs. The model recognized 9 out of 12 inhibitors (75%), which could not be identified based on general parameters, such as molecular weight or SlogP, alone. Finally, the model was used to screen a virtual compound database. A number of compounds found via virtual screening were tested and displayed statistically significant BSEP inhibition, ranging from 13 ± 1% to 67 ± 7% of control (P < 0.05). In conclusion, we developed and validated a pharmacophore model that describes molecular features found in BSEP inhibitors. The model may be used as an in silico screening tool to identify potentially harmful drug candidates at an early stage in drug development.
- Published
- 2014
- Full Text
- View/download PDF
116. Pharmacological considerations on the use of antiretrovirals in pregnancy.
- Author
-
Colbers A, Greupink R, and Burger D
- Subjects
- Anti-Retroviral Agents adverse effects, Drug Therapy, Combination, Female, HIV Infections metabolism, HIV Infections transmission, HIV Integrase Inhibitors pharmacokinetics, HIV Protease Inhibitors pharmacokinetics, Humans, Maternal-Fetal Exchange, Pregnancy, Pregnancy Complications, Infectious metabolism, Reverse Transcriptase Inhibitors pharmacokinetics, Anti-Retroviral Agents pharmacokinetics, HIV Infections drug therapy, Infectious Disease Transmission, Vertical prevention & control, Pregnancy Complications, Infectious drug therapy
- Abstract
Purpose of Review: Treatment with combination antiretroviral therapy during pregnancy reduces the chance of mother to child transmission of HIV. Physiological changes during pregnancy can lead to lower exposure to antiretrovirals, possibly resulting in virological failure. For most antiretrovirals, data on exposure during pregnancy and transplacental passage are limited. This review summarizes the most recent information on pharmacokinetics (including transplacental passage), efficacy, as well as the safety of antiretrovirals during pregnancy., Recent Findings: Intensive-sampling pharmacokinetic studies as well as observational studies using sparse sampling were performed to explore the exposure to antiretrovirals during pregnancy. Transplacental passage, efficacy (viral load at delivery and infection status of the newborn) and safety information were evaluated for several antiretrovirals., Summary: For most nucleoside/nucleotide reverse transcriptase inhibitors and protease inhibitors, recent research shows a decreased exposure during pregnancy. However, the advantage of a general dose increase during pregnancy still remains unclear. For newer compounds and efavirenz, limited or no data on pharmacokinetics during pregnancy or transplacentally are available, while the mechanisms of transplacental passage also remain unknown. For safety reasons, it will be important to monitor pregnancy outcomes in resource-limited settings during the implementation of the WHO guidelines (including the use of efavirenz during pregnancy).
- Published
- 2013
- Full Text
- View/download PDF
117. Interaction of immunosuppressive drugs with human organic anion transporter (OAT) 1 and OAT3, and multidrug resistance-associated protein (MRP) 2 and MRP4.
- Author
-
El-Sheikh AA, Greupink R, Wortelboer HM, van den Heuvel JJ, Schreurs M, Koenderink JB, Masereeuw R, and Russel FG
- Subjects
- Biological Transport, Active drug effects, Drug Interactions, HEK293 Cells, Humans, Immunosuppressive Agents administration & dosage, Immunosuppressive Agents pharmacokinetics, Kidney Tubules, Proximal drug effects, Kidney Tubules, Proximal metabolism, Methotrexate pharmacokinetics, Multidrug Resistance-Associated Protein 2, Multidrug Resistance-Associated Proteins metabolism, Organic Anion Transport Protein 1 metabolism, Organic Anion Transporters, Sodium-Independent metabolism, Translational Research, Biomedical, Immunosuppressive Agents pharmacology, Multidrug Resistance-Associated Proteins antagonists & inhibitors, Organic Anion Transport Protein 1 antagonists & inhibitors, Organic Anion Transporters, Sodium-Independent antagonists & inhibitors
- Abstract
Renal proximal tubule transporters can play a key role in excretion, pharmacokinetic interactions, and toxicity of immunosuppressant drugs. Basolateral organic anion transporters (OATs) and apical multidrug resistance-associated proteins (MRPs) contribute to the active tubular uptake and urinary efflux of these drugs, respectively. We studied the interaction of 12 immunosuppressants with OAT1- and OAT3-mediated [(3)H]-methotrexate (MTX) uptake in cells, and adenosine triphosphate-dependent [(3)H]-MTX transport in membrane vesicles isolated from human embryonic kidney 293 cells overexpressing human MRP2 and MRP4. Our results show that at a clinically relevant concentration of 10 μM, mycophenolic acid inhibited both OAT1- and OAT3-mediated [(3)H]-MTX uptake. Cytarabine, vinblastine, vincristine, hydrocortisone, and mitoxantrone inhibited only OAT1, whereas tacrolimus, azathioprine, dexamethasone, cyclosporine, and 6-mercaptopurine had no effect on both transporters. Cyclophosphamide stimulated OAT1, but did not affect OAT3. With regard to the apical efflux transporters, mycophenolic acid, cyclophosphamide, hydrocortisone, and tacrolimus inhibited MRP2 and MRP4, whereas mitoxantrone and dexamethasone stimulated [(3)H]-MTX transport by both transporters. Cyclosporine, vincristine, and vinblastine inhibited MRP2 only, whereas 6-mercaptopurine inhibited MRP4 transport activity only. Cytarabine and azathioprine had no effect on either transporter. In conclusion, we charted comprehensively the differences in inhibitory action of various immunosuppressive agents against the 4 key renal anion transporters, and we provide evidence that immunosuppressant drugs can modulate OAT1-, OAT3-, MRP2-, and MRP4-mediated transport of MTX to different extents. The data provide a better understanding of renal mechanisms underlying drug-drug interactions and nephrotoxicity concerning combination regimens with these compounds in the clinic., (Copyright © 2013 Mosby, Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
118. Semi-mechanistic physiologically-based pharmacokinetic modeling of clinical glibenclamide pharmacokinetics and drug-drug-interactions.
- Author
-
Greupink R, Schreurs M, Benne MS, Huisman MT, and Russel FG
- Subjects
- Animals, CHO Cells, Clarithromycin administration & dosage, Computer Simulation, Cricetulus, Cytochrome P-450 Enzyme System metabolism, Drug Interactions, Fluconazole administration & dosage, Glyburide administration & dosage, Glyburide blood, HEK293 Cells, Humans, Hypoglycemic Agents administration & dosage, Hypoglycemic Agents blood, Rifampin administration & dosage, Anti-Infective Agents administration & dosage, Glyburide pharmacokinetics, Hypoglycemic Agents pharmacokinetics, Models, Biological
- Abstract
We studied if the clinical pharmacokinetics and drug-drug interactions (DDIs) of the sulfonylurea-derivative glibenclamide can be simulated via a physiologically-based pharmacokinetic modeling approach. To this end, a glibenclamide PBPK-model was build in Simcyp using in vitro physicochemical and biotransformation data of the drug, and was subsequently optimized using plasma disappearance data observed after i.v. administration. The model was validated against data observed after glibenclamide oral dosing, including DDIs. We found that glibenclamide pharmacokinetics could be adequately modeled if next to CYP metabolism an active hepatic uptake process was assumed. This hepatic uptake process was subsequently included in the model in a non-mechanistic manner. After an oral dose of 0.875 mg predicted Cmax and AUC were 39.7 (95% CI:37.0-42.7)ng/mL and 108 (95% CI: 96.9-120)ng/mLh, respectively, which is in line with observed values of 43.6 (95% CI: 37.7-49.5)ng/mL and 133 (95% CI: 107-159)ng/mLh. For a 1.75 mg oral dose, the predicted and observed values were 82.5 (95% CI:76.6-88.9)ng/mL vs 91.1 (95% CI: 67.9-115.9) for Cmax and 224 (95% CI: 202-248) vs 324 (95% CI: 197-451)ng/mLh for AUC, respectively. The model correctly predicted a decrease in exposure after rifampicin pre-treatment. An increase in glibenclamide exposure after clarithromycin co-treatment was predicted, but the magnitude of the effect was underestimated because part of this DDI is the result of an interaction at the transporter level. Finally, the effects of glibenclamide and fluconazol co-administration were simulated. Our simulations indicated that co-administration of this potent CYP450 inhibitor will profoundly increase glibenclamide exposure, which is in line with clinical observations linking the glibenclamide-fluconazol combination to an increased risk of hypoglycemia. In conclusion, glibenclamide pharmacokinetics and its CYP-mediated DDIs can be simulated via PBPK-modeling. In addition, our data underline the relevance of modeling transporters on a full mechanistic level to further improve pharmacokinetic and DDI predictions of this sulfonylurea-derivative., (Copyright © 2013 Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
119. Interaction of digitalis-like compounds with p-glycoprotein.
- Author
-
Gozalpour E, Wittgen HG, van den Heuvel JJ, Greupink R, Russel FG, and Koenderink JB
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, Blotting, Western, HEK293 Cells, Humans, Kinetics, Mutagenesis, Site-Directed, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Cardiac Glycosides metabolism, Digitalis metabolism
- Abstract
Digitalis-like compounds (DLCs), or cardiac glycosides, are produced and sequestered by certain plants and animals as a protective mechanism against herbivores or predators. Currently, the DLCs digoxin and digitoxin are used in the treatment of cardiac congestion and some types of cardiac arrhythmia, despite a very narrow therapeutic index. P-glycoprotein (P-gp; ABCB1) is the only known ATP-dependent efflux transporter that handles digoxin as a substrate. Ten alanine mutants of human P-gp drug-binding amino acids-Leu(65), Ile(306), Phe(336), Ile(340), Phe(343), Phe(728), Phe(942), Thr(945), Leu(975), and Val(982)-were generated and expressed in HEK293 cells with a mammalian baculovirus system. The uptake of [(3)H]-N-methyl-quinidine (NMQ), the P-gp substrate in vesicular transport assays, was determined. The mutations I306A, F343A, F728A, T945A, and L975A abolished NMQ transport activity of P-gp. For the other mutants, the apparent affinities for six DLCs (cymarin, digitoxin, digoxin, peruvoside, proscillaridin A, and strophanthidol) were determined. The affinities of digoxin, proscillaridin A, peruvoside, and cymarin for mutants F336A and I340A were decreased two- to fourfold compared with wild type, whereas that of digitoxin and strophanthidol did not change. In addition, the presence of a hydroxyl group at position 12β seems to reduce the apparent affinity when the side chain of Phe(336) and Phe(942) is absent. Our results showed that a δ-lactone ring and a sugar moiety at 3β of the steroid body are favorable for DLC binding to P-gp. Moreover, DLC inhibition is increased by hydroxyl groups at positions 5β and 19, whereas inhibition is decreased by those at positions 1β, 11α, 12β, and 16β. The understanding of the P-gp-DLC interaction improves our insight into DLCs toxicity and might enhance the replacement of digoxin with other DLCs that have less adverse drug effects.
- Published
- 2013
- Full Text
- View/download PDF
120. In silico identification of potential cholestasis-inducing agents via modeling of Na(+)-dependent taurocholate cotransporting polypeptide substrate specificity.
- Author
-
Greupink R, Nabuurs SB, Zarzycka B, Verweij V, Monshouwer M, Huisman MT, and Russel FG
- Subjects
- Animals, CHO Cells, Computer Simulation, Cricetinae, Cricetulus, Humans, Male, Organic Anion Transporters, Sodium-Dependent chemistry, Rats, Rats, Sprague-Dawley, Structure-Activity Relationship, Substrate Specificity, Cholestasis chemically induced, Models, Biological, Organic Anion Transporters, Sodium-Dependent metabolism, Peptides metabolism, Taurocholic Acid metabolism
- Abstract
Na(+)-dependent taurocholate cotransporting polypeptide (NTCP, SLC10A1) is the main transporter facilitating the hepatic uptake of bile acids from the circulation. Consequently, the interaction of xenobiotics, including therapeutic drugs, with the bile acid binding pocket of NTCP could lead to impairment of hepatic bile acid uptake. We pursued a 3D-pharmacophore approach to model the NTCP substrate and inhibitor specificity and investigated whether it is possible to identify compounds with intrinsic NTCP inhibitory properties. Based on known endogenous NTCP substrates, a 3D-pharmacophore model was built, which was subsequently used to screen two virtual libraries together containing the structures of 10 million compounds. Studies with Chinese hamster ovary cells overexpressing human NTCP, human hepatocytes, ex vivo perfused rat livers, and bile duct-cannulated rats were conducted to validate the activity of the virtual screening hits. Modeling yielded a 3D-pharmacophore, consisting of two hydrogen bond acceptors and three hydrophobic features. Six out of 10 structurally diverse compounds selected in the first virtual screening procedure significantly inhibited taurocholate uptake in the NTCP overexpressing cells. For the most potent inhibitor identified, an anthraquinone derivative, this finding was confirmed in human hepatocytes and perfused rat livers. Subsequent structure and activity relationship studies with analogs of this derivative indicated that an appropriate distance between hydrogen bond acceptor features and presence of one or two negative charges appear critical for a successful NTCP interaction. In conclusion, pharmacophore modeling was successfully used to identify compounds that inhibit NTCP. Our approach represents an important first step toward the in silico flagging of potential cholestasis-inducing molecules.
- Published
- 2012
- Full Text
- View/download PDF
121. Exploiting transport activity of p-glycoprotein at the blood-brain barrier for the development of peripheral cannabinoid type 1 receptor antagonists.
- Author
-
Wittgen HG, Greupink R, van den Heuvel JJ, van den Broek PH, Dinter-Heidorn H, Koenderink JB, and Russel FG
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, ATP Binding Cassette Transporter, Subfamily G, Member 2, ATP-Binding Cassette Transporters metabolism, Acridines pharmacology, Animals, Biological Transport drug effects, Blood-Brain Barrier, Blotting, Western, Cannabinoid Receptor Antagonists metabolism, Cell Line, Humans, Kinetics, Male, Neoplasm Proteins metabolism, Piperidines pharmacology, Pyrazoles pharmacology, Quinidine pharmacology, Rats, Rats, Wistar, Rimonabant, Tandem Mass Spectrometry, Tetrahydroisoquinolines pharmacology, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Cannabinoid Receptor Antagonists pharmacology
- Abstract
Although the CB1 receptor antagonist/inverse agonist rimonabant has positive effects on weight loss and cardiometabolic risk factors, neuropsychiatric side effects have prompted researchers to develop peripherally acting derivatives. Here, we investigated for a series of 3,4-diarylpyrazoline CB1 receptor antagonists if transport by the brain efflux transporter P-gp could be used as a selection criterion in the development of such drugs. All 3,4-diarylpyrazolines and rimonabant inhibited P-gp transport activity in membrane vesicles isolated from HEK293 cells overexpressing the transporter, but only the 1,1-dioxo-thiomorpholino analogue 23 exhibited a reduced accumulation (-38 ± 2%) in these cells, which could be completely reversed by the P-gp/BCRP inhibitor elacridar. In addition, 23 appeared to be a BCRP substrate, whereas rimonabant was not. In rats, the in vivo brain/plasma concentration ratio of 23 was significantly lower than for rimonabant (0.4 ± 0.1 vs 6.2 ± 1.6, p < 0.001). Coadministration of elacridar resulted in an 11-fold increase of the brain/plasma ratio for 23 (p < 0.01) and only 1.4-fold for rimonabant (p < 0.05), confirming the involvement of P-gp and possibly BCRP in limiting the brain entrance of 23 in vivo. In conclusion, these data support the conception that efflux via transporters such as P-gp and BCRP can limit the brain penetration of CB1 receptor antagonists, and that this property could be used in the development of peripheral antagonists.
- Published
- 2012
- Full Text
- View/download PDF
122. Interaction of fluvastatin with the liver-specific Na+ -dependent taurocholate cotransporting polypeptide (NTCP).
- Author
-
Greupink R, Dillen L, Monshouwer M, Huisman MT, and Russel FG
- Subjects
- Animals, Binding, Competitive, Biological Transport drug effects, CHO Cells, Cells, Cultured, Cricetinae, Estradiol analogs & derivatives, Estradiol metabolism, Fatty Acids, Monounsaturated pharmacology, Fluvastatin, Hepatocytes drug effects, Hepatocytes metabolism, Humans, Hydroxymethylglutaryl-CoA Reductase Inhibitors metabolism, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Indoles pharmacology, Inhibitory Concentration 50, Kinetics, Liver-Specific Organic Anion Transporter 1, Molecular Structure, Organic Anion Transporters antagonists & inhibitors, Organic Anion Transporters genetics, Organic Anion Transporters metabolism, Organic Anion Transporters, Sodium-Dependent genetics, Rifampin pharmacology, Simvastatin metabolism, Simvastatin pharmacology, Sodium deficiency, Sodium metabolism, Symporters genetics, Taurocholic Acid metabolism, Transfection, Fatty Acids, Monounsaturated metabolism, Indoles metabolism, Organic Anion Transporters, Sodium-Dependent metabolism, Symporters metabolism
- Abstract
It has been reported that polymorphisms in the organic anion transporting polypeptide 1B1 (OATP1B1, SLCO1B1) result in decreased hepatic uptake of simvastatin carboxy acid, the active metabolite of simvastatin. This is not the case for fluvastatin and it has been hypothesized that for this drug other hepatic uptake pathways exist. Here, we studied whether Na(+)-dependent taurocholate co-transporting polypeptide (NTCP, SLC10A1) can be an alternative hepatic uptake route for fluvastatin. Chinese Hamster Ovary cells transfected with human NTCP (CHO-NTCP) were used to investigate the inhibitory effect of fluvastatin and other statins on [(3)H]-taurocholic acid uptake ([(3)H]-TCA). Statin uptake by CHO-NTCP and cryopreserved human hepatocytes was assessed via LC-MS/MS. Fluvastatin appeared to be a potent and competitive inhibitor of [(3)H]-TCA uptake (IC(50) of 40μM), pointing to an interaction at the level of the bile acid binding pocket of NTCP. The inhibitory action of other statins was also studied, which revealed that statin inhibitory potency increased with molecular descriptors of lipophilicity: calculated logP (r(2)=0.82, p=0.034), logD(7.4) (r(2)=0.77, p=0.0001). Studies in CHO-NTCP cells showed that fluvastatin was indeed an NTCP substrate (K(m) 250±30μM, V(max) 1340±50ng/mg total cell protein/min). However, subsequent studies revealed that at clinically relevant plasma concentrations, NTCP contributed minimally to overall accumulation in human hepatocytes. In conclusion, fluvastatin interacts with NTCP at the level of the bile acid binding pocket and is an NTCP substrate. However, under normal conditions, NTCP-mediated uptake of this drug seems not to be a significant hepatocellular uptake pathway., (Copyright © 2011 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
123. Evaluation of a 99mTc-labeled AnnexinA5 variant for non-invasive SPECT imaging of cell death in liver, spleen and prostate.
- Author
-
Greupink R, Sio CF, Ederveen A, and Orsel J
- Subjects
- Animals, Cell Death, Cysteine chemistry, Male, Prostate pathology, Radionuclide Imaging, Rats, Rats, Wistar, Spleen pathology, Annexin A5 chemistry, Annexin A5 pharmacokinetics, Liver diagnostic imaging, Organotechnetium Compounds pharmacokinetics, Prostate diagnostic imaging, Spleen diagnostic imaging
- Abstract
Purpose: We investigate radio-labeling and pharmacokinetics of a new AnnexinA5 variant (HYNIC-cys-AnxA5) and then assess its utility for the non-invasive detection of cell death in liver, spleen and prostate., Methods: AnnexinA5 binds to phosphatidylserine expressed on the surface of apoptotic and necrotic cells. Contrary to other AnnexinA5 variants, the new cys-AnxA5 allows for site-specific conjugation of a hydrazinonicotinamide-maleimide moiety and subsequent radio-labeling with (99m)Tc at a position not involved in the AnxA5-phosphatidylserine interaction. Distribution of (99m)Tc-HYNIC-cys-AnxA5 was studied in rats, both invasively and via SPECT/CT. Cycloheximide was used to induce cell death in liver and spleen, whereas apoptosis in the prostate was induced by castration., Results: HYNIC-cys-AnxA5 was efficiently and reproducibly labeled with (99m)Tc. Blood clearance of radioactivity after iv-injection was adequately described by a two-compartment model, the renal cortex representing the main site of accumulation. Cycloheximide treatment resulted in increased accumulation of intravenous-injected (99m)Tc-HYNIC-cys-AnxA5 in liver and spleen over controls, which correlated well with TUNEL staining for cell death in corresponding tissue sections. However, the increase in TUNEL-positive prostate epithelial cells observed following castration was not paralleled by greater (99m)Tc-HYNIC-cys-AnxA5 accumulation., Conclusion: (99m)Tc-HYNIC-cys-AnxA5 appears a suitable tracer for assessment of cell death in liver and spleen, but not prostate.
- Published
- 2009
- Full Text
- View/download PDF
124. Pharmacokinetics of a hepatic stellate cell-targeted doxorubicin construct in bile duct-ligated rats.
- Author
-
Greupink R, Reker-Smit C, Proost JH, van Loenen Weemaes AM, de Hooge M, Poelstra K, and Beljaars L
- Subjects
- Animals, Antineoplastic Agents metabolism, Antineoplastic Agents pharmacokinetics, Bile Ducts physiopathology, Disease Models, Animal, Doxorubicin metabolism, Drug Carriers metabolism, Drug Delivery Systems, Hepatocytes cytology, Liver cytology, Male, Rats, Rats, Wistar, Tissue Distribution, Bile Ducts metabolism, Doxorubicin pharmacokinetics, Hepatocytes metabolism, Ligation
- Abstract
Background/aims: Inhibition of hepatic stellate cell (HSC) proliferation is a relevant strategy to inhibit liver fibrosis. Coupling of antiproliferative drugs to the HSC-selective drug carrier mannose-6-phosphate-modified human serum albumin (M6PHSA) may lead to cell-selective inhibition of HSC proliferation. We coupled the antiproliferative drug doxorubicin (DOX) to this drug carrier and investigated the pharmacokinetics of this construct in a rat model of liver fibrosis, as well as in cultured HSC., Methods/results: M6PHSA-DOX was cleared from the plasma in a biphasic manner. Upon i.v. injection of 4 microg kg(-1) (tracer), 2 and 20 mg kg(-1), the clearance in the distribution phase of drug disposition (CL(d)) significantly decreased from 9.7+/-0.7 to 4.7+/-2.3 and 1.0+/-0.1 ml kg(-1)min(-1), respectively. This indicates that saturation of clearance mechanisms occurs in this phase of drug disposition, likely reflecting saturable receptor-mediated uptake in the target cells. Gamma-camera studies revealed that the majority of the conjugate accumulated in the liver within 5 min, and immunohistochemical double-staining of liver sections demonstrated co-localization of the construct with HSC-markers. Simulation of the release of DOX from the carrier, after cellular uptake by HSC, showed that a gradual release of the drug takes place over a 9h period. Studies in cultured HSC illustrated that after 24h incubation with the conjugate, DOX was associated with the cell nucleus., Conclusions: The rapid distribution of M6PHSA-DOX from the blood to HSC, in combination with the expected gradual release of DOX within these cells, make this construct a promising tool for achieving sustained and selective inhibition of HSC proliferation.
- Published
- 2007
- Full Text
- View/download PDF
125. Targeting 15d-prostaglandin J2 to hepatic stellate cells: two options evaluated.
- Author
-
Hagens WI, Mattos A, Greupink R, de Jager-Krikken A, Reker-Smit C, van Loenen-Weemaes A, Gouw IA, Poelstra K, and Beljaars L
- Subjects
- Animals, Apoptosis, Apoptosis Regulatory Proteins metabolism, Enzyme-Linked Immunosorbent Assay, Humans, Immunohistochemistry, Liver cytology, Liver Cirrhosis metabolism, Liver Cirrhosis pathology, Liver Cirrhosis, Experimental chemically induced, Liver Cirrhosis, Experimental metabolism, Liver Cirrhosis, Experimental pathology, Male, Mannosephosphates chemistry, Mannosephosphates metabolism, Peptides chemistry, Peptides metabolism, Prostaglandin D2 administration & dosage, Prostaglandin D2 chemistry, Prostaglandin D2 pharmacokinetics, Rats, Rats, Wistar, Receptor, Platelet-Derived Growth Factor beta chemistry, Receptor, Platelet-Derived Growth Factor beta metabolism, Reproducibility of Results, Serum Albumin chemistry, Serum Albumin metabolism, Drug Delivery Systems methods, Liver metabolism, Prostaglandin D2 analogs & derivatives
- Abstract
Purpose: Delivery of apoptosis-inducing compounds to hepatic stellate cells (HSC) may be an effective strategy to reverse liver fibrosis. The aim of this study was therefore to examine the selective targeting of the apoptosis-inducing drug 15-deoxy-delta12,14-prostaglandin J2 (15dPGJ2) with two different HSC-carriers: human serum albumin modified with the sugar mannose-6-phosphate (M6PHSA) or albumin modified with PDGF-receptor recognizing peptides (pPBHSA)., Methods and Results: After chemical conjugation of 15dPGJ2 to the carriers, the constructs displayed pharmacological activity and specific receptor-mediated binding to HSC in vitro. Unlike 15dPGJ2-pPBHSA, the cellular binding of 15dPGJ2-M6PHSA was reduced by a scavenger receptor antagonist. In vivo, both conjugates rapidly accumulated in fibrotic livers. Intrahepatic analysis revealed that 15dPGJ2-M6PHSA mainly accumulated in HSC, and to a lesser extent in Kupffer cells. 15dPGJ2-pPBHSA also predominantly accumulated in HSC with additional uptake in hepatocytes. Assessment of target receptors in human cirrhotic livers revealed that M6P/IGFII-receptor expression was present in fibrotic areas. PDGF-P receptor expression was abundantly expressed on human fibroblasts., Conclusions: These studies show that 15dPGJ2 coupled to either M6PHSA or pPBHSA is specifically taken up by HSC and is highly effective within these cells. Both carriers differ with respect to receptor specificity, leading to differences in intrahepatic distribution. Nevertheless, both carriers can be used to deliver the apoptosis-inducing drug 15dPGJ2 to HSC in vivo.
- Published
- 2007
- Full Text
- View/download PDF
126. Mannose-6-phosphate/insulin-Like growth factor-II receptors may represent a target for the selective delivery of mycophenolic acid to fibrogenic cells.
- Author
-
Greupink R, Bakker HI, van Goor H, de Borst MH, Beljaars L, and Poelstra K
- Subjects
- 3T3 Cells, Animals, Antibiotics, Antineoplastic administration & dosage, Binding, Competitive drug effects, Capillaries drug effects, Capillaries metabolism, Cell Proliferation drug effects, Cell Survival drug effects, Common Bile Duct physiology, DNA biosynthesis, Dose-Response Relationship, Drug, Drug Delivery Systems, Electrophoresis, Polyacrylamide Gel, Fibroblasts drug effects, Fibroblasts metabolism, Immunohistochemistry, Ligation, Male, Mice, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular metabolism, Mycophenolic Acid administration & dosage, RNA, Messenger biosynthesis, RNA, Messenger genetics, Rats, Rats, Wistar, Renal Circulation drug effects, Reverse Transcriptase Polymerase Chain Reaction, Antibiotics, Antineoplastic pharmacology, Liver Cirrhosis pathology, Mannosephosphates metabolism, Mycophenolic Acid pharmacology, Receptor, IGF Type 2 metabolism
- Abstract
Purpose: The insulin-like growth factor axis plays an important role in fibrogenesis. However, little is known about mannose-6-phosphate/Insulin-like growth factor-II receptor (M6P/IGF-IIR) expression during fibrosis. When expressed preferentially on fibrogenic cells, this receptor may be used to selectively deliver drugs to these cells., Methods: We investigated M6P/IGF-IIR expression in livers of bile duct-ligated (BDL) rats and in renal vascular walls of renin transgenic TGR(mRen2)27 rats. Both models are characterized by fibrogenic processes. Furthermore, we studied whether drug delivery via M6P/IGF-II-receptor-mediated uptake is possible in fibroblasts., Results: M6P/IGF-IIR mRNA expression was investigated 3, 7 and 10 days after BDL. At all time-points hepatic M6P/IGF-IIR expression was significantly increased compared to healthy controls. Moreover, immunohistochemical staining revealed that alpha-sma-positive cells were M6P/IGF-IIR-positive. In kidneys of TGR(mRen2)27 rats, the number of M6P/IGF-IIR-positive arteries per microscopic field was increased 5.5 fold over healthy controls. To examine whether M6P/IGF-IIRs could be used as a port of entry for drugs, we coupled mycophenolic acid (MPA) to mannose-6-phosphate-modified human serum albumin (M6PHSA). M6PHSA-MPA inhibited 3T3-fibroblast proliferation dose-dependently, which was reversed by co-incubation with excess M6PHSA, but not by HSA., Conclusions: M6P/IGF-IIRs are expressed by fibrogenic cells and may be used for receptor-mediated intracellular delivery of the antifibrogenic drug MPA.
- Published
- 2006
- Full Text
- View/download PDF
127. The antiproliferative drug doxorubicin inhibits liver fibrosis in bile duct-ligated rats and can be selectively delivered to hepatic stellate cells in vivo.
- Author
-
Greupink R, Bakker HI, Bouma W, Reker-Smit C, Meijer DK, Beljaars L, and Poelstra K
- Subjects
- Animals, Binding, Competitive, Cells, Cultured, Disease Models, Animal, Doxorubicin administration & dosage, Doxorubicin pharmacokinetics, Drug Carriers pharmacokinetics, Liver cytology, Liver metabolism, Male, Mannose pharmacokinetics, Mannosephosphates pharmacokinetics, Rats, Rats, Wistar, Serum Albumin pharmacokinetics, Tissue Distribution, Cell Proliferation drug effects, Cholestasis complications, Doxorubicin therapeutic use, Liver drug effects, Liver Cirrhosis, Experimental prevention & control
- Abstract
Hepatic stellate cell (HSC) proliferation is a key event in liver fibrosis; therefore, pharmacological intervention with antiproliferative drugs may result in antifibrotic effects. In this article, the antiproliferative effect of three cytostatic drugs was tested in cultured rat HSC. Subsequently, the antifibrotic potential of the most potent drug was evaluated in vivo. As a strategy to overcome drug-related toxicity, we additionally studied how to deliver this drug specifically to HSC by conjugating it to the HSC-selective drug carrier mannose-6-phosphate-modified human serum albumin (M6PHSA). We investigated the effect of cisplatin, chlorambucil, and doxorubicin (DOX) on 5-bromo-2'-deoxyuridine incorporation in cultured HSC and found DOX to be the most potent drug. Treatment of bile duct-ligated (BDL) rats with daily i.v. injections of 0.35 mg/kg DOX from day 3 to 10 after BDL reduced alpha-smooth muscle actin-stained area in liver sections from 8.5 +/- 0.8 to 5.1 +/- 0.9% (P < 0.01) and collagen-stained area from 13.1 +/- 1.3 to 8.9 +/- 1.5% (P < 0.05). DOX was coupled to M6PHSA, and the organ distribution of this construct (M6PHSA-DOX) was investigated. Twenty minutes after i.v. administration, 50 +/- 6% of the dose was present in the livers, and colocalization of M6PHSA-DOX with HSC markers was observed. In addition, in vitro studies showed selective binding of M6PHSA-DOX to activated HSC. Moreover, M6PHSA-DOX strongly attenuated HSC proliferation in vitro, indicating that active drug is released after uptake of the conjugate. DOX inhibits liver fibrosis in BDL rats, and HSC-selective targeting of this drug is possible. This may offer perspectives for the application of antiproliferative drugs for antifibrotic purposes.
- Published
- 2006
- Full Text
- View/download PDF
128. Selective targeting of pentoxifylline to hepatic stellate cells using a novel platinum-based linker technology.
- Author
-
Gonzalo T, Talman EG, van de Ven A, Temming K, Greupink R, Beljaars L, Reker-Smit C, Meijer DK, Molema G, Poelstra K, and Kok RJ
- Subjects
- Actins analysis, Animals, Biological Availability, Caspase 3, Caspases metabolism, Cell Survival drug effects, Collagen Type I analysis, Drug Delivery Systems methods, Enzyme Activation drug effects, Liver chemistry, Liver cytology, Liver Cirrhosis metabolism, Liver Cirrhosis prevention & control, Male, Mice, Muscle, Smooth chemistry, NIH 3T3 Cells, Pentoxifylline chemistry, Pentoxifylline pharmacology, Phosphoric Monoester Hydrolases chemistry, Rats, Rats, Wistar, Technology, Pharmaceutical methods, Tissue Distribution, Chemistry, Pharmaceutical methods, Liver metabolism, Organoplatinum Compounds chemistry, Pentoxifylline pharmacokinetics
- Abstract
Targeting of antifibrotic drugs to hepatic stellate cells (HSC) is a promising strategy to block fibrotic processes leading to liver cirrhosis. For this purpose, we utilized the neo-glycoprotein mannose-6-phosphate-albumin (M6PHSA) that accumulates efficiently in HSC during liver fibrosis. Pentoxifylline (PTX), an antifibrotic compound that inhibits HSC proliferation and activation in vitro, was conjugated to M6PHSA. We employed a new type of platinum-based linker, which conjugates PTX via coordination chemistry rather than via covalent linkage. When incubated in plasma or in the presence of thiol compounds, free PTX was released from PTX-M6PHSA at a sustained slow rate. PTX-M6PHSA displayed pharmacological activity in cultured HSC as evidenced by changes in cell morphology and reduction of collagen I production. PTX-M6PHSA and platinum coupled PTX did not induce platinum-related toxicity (Alamar Blue viability assay) or apoptosis (caspase activation and TUNEL staining). In vivo distribution studies in fibrotic rats demonstrated specific accumulation of the conjugate in nonparenchymal cells in the fibrotic liver. In conclusion, we have developed PTX-M6PHSA employing a novel type of platinum linker, which allows sustained delivery of the drug to HSC in the fibrotic liver.
- Published
- 2006
- Full Text
- View/download PDF
129. Studies on the targeted delivery of the antifibrogenic compound mycophenolic acid to the hepatic stellate cell.
- Author
-
Greupink R, Bakker HI, Reker-Smit C, van Loenen-Weemaes AM, Kok RJ, Meijer DK, Beljaars L, and Poelstra K
- Subjects
- Animals, Antibiotics, Antineoplastic chemistry, Antibiotics, Antineoplastic metabolism, Antibiotics, Antineoplastic therapeutic use, Collagen metabolism, Crystallins genetics, Crystallins metabolism, Desmin metabolism, Drug Carriers, Fibrosis drug therapy, Fibrosis pathology, Glial Fibrillary Acidic Protein metabolism, Hepatocytes cytology, Humans, Liver drug effects, Liver metabolism, Male, Mannosephosphates chemistry, Mannosephosphates metabolism, Molecular Structure, Mycophenolic Acid chemistry, Mycophenolic Acid metabolism, Mycophenolic Acid therapeutic use, Protein Isoforms genetics, Protein Isoforms metabolism, Rats, Rats, Wistar, Reactive Oxygen Species metabolism, Tissue Distribution, Antibiotics, Antineoplastic pharmacology, Cell Proliferation drug effects, Hepatocytes drug effects, Liver pathology, Mycophenolic Acid pharmacology
- Abstract
Background/aims: Hepatic stellate cell (HSC) activation and proliferation are key events in the pathology of liver fibrosis. Inhibiting these parameters, therefore, is a relevant option to treat liver fibrosis pharmacologically. The immunosuppressive drug mycophenolic acid (MPA) has been shown to inhibit proliferation and activation of various types of fibroblasts. In an effort to circumvent the immunosuppression and at the same time enhance this antifibrotic effect, we coupled MPA to the HSC-selective drug carrier mannose-6-phosphate modified human serum albumin and evaluated this conjugate for its specificity and antifibrotic activity., Methods/results: We found that MPA inhibited proliferation of HSC in vitro. The drug coupled to the drug carrier bound specifically to HSC and reduced HSC proliferation in vitro. In vivo studies demonstrated that our conjugate accumulated selectively in the liver with significant uptake in HSC apart from Kupffer and endothelial cells, whereas primary and secondary lymphoid tissues were avoided. Treatment of bile duct-ligated rats with this conjugate reduced hepatic inflammation and hepatic alpha-beta-Crystallin mRNA expression, a marker for HSC activation., Conclusions: This study shows that targeted delivery of MPA to HSC results in a decrease in HSC activation, making it the first drug that is successfully delivered to this cell type.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.