Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Neurochem ; 2024 May 12.
Article in English | MEDLINE | ID: mdl-38735840

ABSTRACT

The blood-brain barrier (BBB) is a structure mainly formed by brain capillary endothelial cells (BCEC) whose role is to regulate the exchange of compounds between the blood and the brain. In this process efflux and uptake transporters play a key role. Aim of this study was to compare the two previously established cell lines hCMEC/D3 and hBMEC as BBB cell models for the application of an adenoviral system to transiently express OATP2B1 and Pgp. Comparison of hCMEC/D3 and hBMEC mRNA and protein levels of BBB markers showed a unique expression pattern for each cell line. While showing similar expression of the efflux transporter BCRP, transferrin receptor (TFRC) and of the tight junctions proteins Occludin and ZO-1, hCMEC/D3 displayed higher levels of the endothelial marker PECAM1, VE-cadherin, Von Willebrand Factor (VWF) and of the efflux transporter Pgp. Moreover, measuring integrity of the monolayer by determining the Trans-Endothelial Electrical Resistance (TEER), electrical capacitance (CCl), and inulin apparent permeability coefficient (Papp) revealed higher TEER and lower CCl for hBMEC but comparable Papp in the two cell lines. Following adenoviral infection, enhanced OATP2B1 and Pgp expression and functionality could be observed only in hBMEC. Importantly, the adenoviral expression system did not affect expression of BBB markers and permeability in both cell lines. Taken together, our results provide first evidence that hBMEC is an applicable human BBB cell model in which adenoviral infection can be used to transiently express and investigate transporters of interest.

2.
J Pharmacol Exp Ther ; 389(1): 87-95, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38448247

ABSTRACT

The organic anion transporting polypeptide (OATP)2B1 [(gene: solute carrier organic anion transporter family member 2B1 (SLCO2B1)] is an uptake transporter that facilitates cellular accumulation of its substrates. Comparison of SLCO2B1+/+ knockin and rSlco2b1-/- knockout rats showed a higher expression of rCYP3A1 in the humanized animals. We hypothesize that humanization of OATP2B1 not only affects cellular uptake but also metabolic activity. To further investigate this hypothesis, we used SLCO2B1+/+ and rSlco2b1-/ - rats and the OATP2B1 and rCYP3A1 substrate erlotinib, which is metabolized to OSI-420, for in vivo and ex vivo experiments. One hour after administration of a single dose of erlotinib, the knockin rats exhibited significantly lower erlotinib serum levels, but no change was observed in metabolite concentration or the OSI-420/erlotinib ratio. Similar results were obtained for liver tissue levels comparing SLCO2B1+/+ and rSlco2b1-/- rats. Liver microsomes isolated from the erlotinib-treated animals were characterized ex vivo for rCYP3A activity using testosterone, showing higher activity in the knockin rats. The contrary was observed when microsomes isolated from treatment-naïve animals were assessed for the metabolism of erlotinib to OSI-420. The latter is in contrast to the higher rCYP3A1 protein amount observed by western blot analysis in rat liver lysates and liver microsomes isolated from untreated rats. In summary, rats humanized for OATP2B1 showed higher expression of rCYP3A1 in liver and reduced serum levels of erlotinib but no change in the OSI-420/erlotinib ratio despite a lower OSI-420 formation in isolated liver microsomes. Studies with CYP3A-specific substrates are warranted to evaluate whether humanization affects not only rCYP3A1 expression but also metabolic activity in vivo. SIGNIFICANCE STATEMENT: Humanization of rats for the organic anion transporting polypeptide (OATP)2B1 increases rCYP3A1 expression and activity in liver. Using the OATP2B1/CYP3A-substrate erlotinib to assess the resulting phenotype, we observed lower erlotinib serum and liver concentrations but no impact on the liver/serum ratio. Moreover, there was no difference in the OSI-420/erlotinib ratio comparing humanized and knockout rats, suggesting that OSI-420 is not applicable to monitor differences in rCYP3A1 expression as supported by data from ex vivo experiments with rat liver microsomes.


Subject(s)
Cytochrome P-450 CYP3A , Organic Anion Transporters , Rats , Animals , Erlotinib Hydrochloride/pharmacology , Cytochrome P-450 CYP3A/metabolism , Quinazolines/pharmacology , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism
3.
Mol Pharmacol ; 105(1): 14-22, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37863663

ABSTRACT

The pregnane X receptor (PXR) is a ligand-activated regulator of cytochrome P450 (CYP)3A enzymes. Among the ligands of human PXR is hyperforin, a constituent of St John's wort (SJW) extracts and potent inducer of human CYP3A4. It was the aim of this study to compare the effect of hyperforin and SJW formulations controlled for its content on CYP3A23-3A1 in rats. Hyperiplant was used as it contains a high hyperforin content and Rebalance because it is controlled for a low hyperforin content. In silico analysis revealed a weak hyperforin-rPXR binding affinity, which was further supported in cell-based reporter gene assays showing no hyperforin-mediated reporter activation in presence of rPXR. However, cellular exposure to Hyperiplant and Rebalance transactivated the CYP3A reporter 3.8-fold and 2.8-fold, respectively, and they induced Cyp3a23-3a1 mRNA expression in rat hepatoma cells compared with control 48-fold and 18-fold, respectively. In Wistar rats treated for 10 days with 400 mg/kg of Hyperiplant, we observed 1.8 times the Cyp3a23-3a1 mRNA expression, a 2.6-fold higher CYP3A23-3A1 protein amount, and a 1.6-fold increase in activity compared with controls. For Rebalance we only observed a 1.8-fold hepatic increase of CYP3A23-3A1 protein compared with control animals. Even though there are differing effects on rCyp3a23-3a1/CYP3A23-3A1 in rat liver reflecting the hyperforin content of the SJW extracts, the modulation is most likely not linked to an interaction of hyperforin with rPXR. SIGNIFICANCE STATEMENT: Treatment with St John's wort (SJW) has been reported to affect CYP3A expression and activity in rats. Our comparative study further supports this finding but shows that the pregnane X receptor-ligand hyperforin is not the driving force for changes in rat CYP3A23-3A1 expression and function in vivo and in vitro. Importantly, CYP3A induction mimics findings in humans, but our results suggest that another so far unknown constituent of SJW is responsible for the expression- and function-modifying effects in rat liver.


Subject(s)
Antineoplastic Agents , Hypericum , Rats , Humans , Animals , Cytochrome P-450 CYP3A/metabolism , Pregnane X Receptor , Hypericum/metabolism , Ligands , Rats, Wistar , RNA, Messenger , Plant Extracts/pharmacology , Plant Extracts/chemistry
4.
J Pharmacol Sci ; 153(3): 170-174, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37770158

ABSTRACT

Coproporphyrin I (CPI) and III (CPIII) are discussed as biomarkers for organic anion transporting polypeptides (OATPs). We report on CPI and CPIII levels in wildtype, rSlco2b1-knockout, and SLCO2B1-humanized rats at baseline and after administration of atorvastatin, an inhibitor of the CPIII-specific rOATP2B1/hOATP2B1 and the CPI/CPIII-transporting rOATP1B2. OATP-inhibition by atorvastatin leads to significantly increased CPI and CPIII serum levels. However, basal CP serum levels in rSlco2b1-knockout animals were significantly lower (CPI), or unaffected (CPIII). In the presence of atorvastatin, this genotype effect was abolished. In conclusion, our results indicate an unexpected impact of OATP2B1 on CP serum levels in rats.

5.
Br J Clin Pharmacol ; 88(4): 1885-1896, 2022 02.
Article in English | MEDLINE | ID: mdl-34648192

ABSTRACT

AIMS: Metamizole (dipyrone) is a prodrug not detectable in serum or urine after oral ingestion. The primary metabolite, 4-methylaminoantipyrine (4-MAA), can be N-demethylated to 4-aminoantipyrine (4-AA) or oxidized to 4-formylaminoantipyrine (4-FAA) by cytochrome P450 (CYP)-dependent reactions. We aimed to identify the CYPs involved in 4-MAA metabolism and to quantify the effect of CYP inhibition on 4-MAA metabolism. METHODS: We investigated the metabolism of 4-MAA in vitro using CYP expressing supersomes and the pharmacokinetics of metamizole in the presence of CYP inhibitors in male subjects. RESULTS: The experiments in supersomes revealed CYP1A2 as the major CYP for 4-MAA N-demethylation and 4-FAA formation with CYP2C19 and CYP2D6 contributing to N-demethylation. In the clinical study, we investigated the influence of ciprofloxacin (CYP1A2 inhibitor), fluconazole (CYP2C19 inhibitor) and the combination ciprofloxacin/fluconazole on the pharmacokinetics of metamizole in n = 12 male subjects in a randomized, placebo-controlled, double-blind study. The geometric mean ratios for the area under the concentration-time curve of 4-MAA after/before treatment were 1.17 (90% CI 1.09-1.25) for fluconazole, 1.51 (90% CI 1.42-1.60) for ciprofloxacin and 1.92 (90% CI 1.81-2.03) for ciprofloxacin/fluconazole. Fluconazole increased the half-life of 4-MAA from 3.22 hours by 0.47 hours (95% CI 0.13-0.81, P < .05), ciprofloxacin by 0.69 hours (95% CI 0.44-0.94, P < .001) and fluconazole/ciprofloxacin by 2.85 hours (95% CI 2.48-3.22, P < .001). CONCLUSION: CYP1A2 is the major CYP for the conversion of 4-MAA to 4-AA and 4-FAA. The increase in 4-MAA exposure by the inhibition of CYP1A2 and by the combination CYP1A2/CYP2C19 may be relevant for dose-dependent adverse reactions of 4-MAA.


Subject(s)
Cytochrome P-450 CYP1A2 , Dipyrone , Ciprofloxacin , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2C19 , Cytochrome P-450 Enzyme System/metabolism , Dipyrone/analogs & derivatives , Dipyrone/metabolism , Fluconazole/pharmacology , Humans , Male
6.
Eur J Pediatr ; 181(3): 1221-1231, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34739591

ABSTRACT

In daily paediatrics, drugs are commonly used off-label, as they are not approved for children. Approval is lacking because the required clinical studies were limited to adults in the past. Without clinical studies, evidence-based recommendations for drug use in children are limited. Information on off-label drug dosing in children can be found in different handbooks, databases and scientific publications but the dosing recommendations can differ considerably. To improve safety and efficacy of drugs prescribed to children and to assist the prescribers, stakeholders in Swiss paediatrics started a pilot project, supported by the Federal Office of Public Health, with the aim to create a database, providing healthcare professionals with so called "harmonised" dosage recommendations based on the latest available scientific evidence and best clinical practice. A standardised process for dosage harmonisation between paediatric experts was defined, guided and documented in an electronic tool, developed for this purpose. As proof of principle, a total of 102 dosage recommendations for 30 different drugs have been nationally harmonised in the pilot phase considering the current scientific literature and the approval of the most experienced national experts in the field.Conclusion: This approach paved the way for unified national dosage recommendations for children. Reaching the project's milestones fulfilled the prerequisites for funding and starting regular operation of SwissPedDose in 2018. Since then, the database was extended with recommendations for 100 additional drugs. What is Known: • Prescribing off-label is a common practice among paediatricians, as many drugs are still not authorised for use in children. • Some countries developed national drug formularies providing off-label dosage recommendations. What is New: • Comparison of published dosage recommendations in known drug handbooks and online databases show substantial differences and heterogeneity, revealing the need for harmonisation. • The design of a tool for standardised harmonisation of dosage recommendations, based on information collected on currently applied dosages, latest scientific evidence and the approval of experts.


Subject(s)
Databases, Pharmaceutical , Off-Label Use , Pediatrics , Adult , Child , Drug Labeling , Humans , Pilot Projects , Switzerland
7.
Planta Med ; 88(2): 152-162, 2022 Feb.
Article in English | MEDLINE | ID: mdl-33511622

ABSTRACT

Herbal medication used in the treatment of sleep disorders and anxiety often contain extracts of Valeriana officinalis or Passiflora incarnata. Valerenic acid in V. officinalis and apigenin, orientin, and vitexin in P. incarnata are thought to contribute to their therapeutic effect. It was the aim of this study to test whether these constituents of herbal extracts are interacting with the uptake of estrone 3-sulfate, pregnenolone sulfate, and dehydroepiandrosterone sulfate mediated by the uptake transporters organic anion transporting polypeptide 2B1 (OATP2B1) or organic anion transporting polypeptide 1A2 (OATP1A2). Madin-Darby canine kidney cells overexpressing OATP2B1 or OATP1A2 were used to determine the influence of the constituents on the cellular accumulation of the sulfated steroids. Subsequently, competitive counterflow experiments were applied to test whether identified inhibitors are also substrates of the transporters. Valerenic acid only interacted with OATP2B1, whereas apigenin, orientin, and vitexin interacted with OATP2B1 and OATP1A2. Competitive counterflow revealed that orientin is a substrate of both transporters, while apigenin was transported by OATP1A2 and vitexin by OATP2B1. In a next step, commercially available P. incarnata preparations were assessed for their influence on the transporters, revealing inhibition of transporter-mediated estrone 3-sulfate uptake. HPLC-UV-MS analysis confirmed the presence of orientin and vitexin in these preparations, thereby suggesting that these constituents are involved in the interaction. Our data indicate that constituents of P. incarnata may alter the function of OATP2B1 and OATP1A2, which could affect the uptake of other compounds relying on uptake mediated by the transporters.


Subject(s)
Organic Anion Transporters , Passiflora , Phytochemicals/pharmacology , Valerian , Animals , Biological Transport , Dogs , Organic Anion Transporters/metabolism , Passiflora/chemistry , Peptides , Valerian/chemistry
8.
Int J Mol Sci ; 22(12)2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34204223

ABSTRACT

We report two cases of patients who developed severe adverse drug reactions including persistent movement disorders, nausea, and vertigo during treatment with quetiapine at maximum daily doses ranging between 300 and 400 mg. The extensive hepatic metabolism of quetiapine is mainly attributed to cytochrome P450 3A4 (CYP3A4). However, there is recent evidence supporting the idea of CYP2D6 playing a role in the clearance of the quetiapine active metabolite norquetiapine. Interestingly, both patients we are reporting of are carriers of the CYP2D6*4 variant, predicting an intermediate metabolizer phenotype. Additionally, co-medication with a known CYP2D6 inhibitor and renal impairment might have further affected quetiapine pharmacokinetics. The herein reported cases could spark a discussion on the potential impact of a patient's pharmacogenetic predisposition in the treatment with quetiapine. However, further studies are warranted to promote the adoption of pharmacogenetic testing for the prevention of drug-induced toxicities associated with quetiapine.


Subject(s)
Cytochrome P-450 CYP2D6/genetics , Drug-Related Side Effects and Adverse Reactions/diagnosis , Drug-Related Side Effects and Adverse Reactions/etiology , Pharmacogenomic Variants , Quetiapine Fumarate/adverse effects , Alleles , Antipsychotic Agents/adverse effects , Antipsychotic Agents/chemistry , Antipsychotic Agents/metabolism , Cytochrome P-450 CYP2D6/metabolism , Drug-Related Side Effects and Adverse Reactions/metabolism , Genetic Association Studies , Genetic Variation , Genotype , Humans , Male , Middle Aged , Pharmacogenetics , Phenotype , Quetiapine Fumarate/administration & dosage , Quetiapine Fumarate/chemistry , Quetiapine Fumarate/metabolism , Severity of Illness Index
9.
Bioconjug Chem ; 31(3): 781-793, 2020 03 18.
Article in English | MEDLINE | ID: mdl-31894970

ABSTRACT

The concept of triggered drug release offers a possibility to overcome the toxic side effects of chemotherapeutics in cancer treatment by reducing systemic exposure to the active drug. In the present work, the concept foresees the use of the extracellular enzyme MMP9 as an enzymatic trigger for drug release in the proximity of tumor cells. METHODS: A paclitaxel-hemisuccinate-peptide conjugate as a building block for self-assembling nanoparticles was synthesized using standard conjugation approaches. The building block was purified via preparative HPLC and analyzed by LC-MS. Nanoparticles were formed using the nanoprecipitation method and characterized. For selection of a suitable in vitro model system, common bioanalytical methods were used to determine mRNA expression, enzyme amount, and activity of MMP9. RESULTS: The MMP9-labile prodrug was synthesized and characterized. Nanoparticles were formed out of MMP9-labile conjugate-building blocks. The nanoparticle's diameter averaged at around 120 nm and presented a spherical shape. LN-18 cells, a glioblastoma multiforme derived cell line, were chosen as an in vitro model based on findings in cancer tissue and cell line characterization. The prodrug showed cytotoxicity in LN-18 cells, which was reduced by addition of an MMP9 inhibitor. CONCLUSION: taken together, we confirmed increased MMP9 in several cancer tissues (cervical, esophageal, lung, and brain) compared to healthy tissue and showed the effectiveness of MMP9-labile prodrug in in vitro tests.


Subject(s)
Drug Design , Extracellular Space/enzymology , Matrix Metalloproteinase 9/metabolism , Paclitaxel/chemistry , Cell Line, Tumor , Cell Survival , Chemistry Techniques, Synthetic , Humans , Matrix Metalloproteinase 9/genetics , Paclitaxel/metabolism , Particle Size , Peptides/chemistry , Polyethylene Glycols/chemistry , RNA, Messenger/genetics
10.
Mol Pharm ; 17(6): 1987-1995, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32343897

ABSTRACT

Interaction with the dopaminergic system in the central nervous system is either therapeutically intended or it is a side effect. In both cases, dopamine-receptor agonists (DRA) like the ergoline derivative bromocriptine and dopamine-receptor antagonists (DRAn) like metoclopramide have to cross the blood-brain barrier (BBB). The organic anion transporting polypeptides (OATP) 1A2 and 2B1 are cellular uptake carriers for a variety of endogenous and xenobiotic compounds. As both transporters are expressed in endothelial cells of the BBB, the aim of the present study was to determine whether the DRA bromocriptine, cabergoline, and pergolide and the DRAn metoclopramide and domperidone are interacting with OATP1A2 and 2B1 and could therefore be candidate genes modifying wanted and unwanted effects of these drugs. Localization of both transporters in the brain was confirmed using LC-MS/MS and immunofluorescence stainings. For the functional studies, MDCKII cells stably expressing OATP1A2 or 2B1 were used. Initial interaction studies with the well-characterized transporter substrate estrone 3-sulfate revealed that all tested compounds except pergolide inhibit the transport function of both proteins with the most potent effect for bromocriptine (IC50 = 2.2 µM (OATP1A2) and IC50 = 2.5 µM (OATP2B1)). Further studies using the indirect competitive counterflow method identified bromocriptine, cabergoline, and domperidone as substrates of both transporters, whereas metoclopramide was only transported by OATP1A2. These findings were verified for domperidone by direct measurements using its tritium-labeled form as a tracer. Moreover, the transporter-mediated uptake of this compound was sensitive to the OATP1A2 and OATP2B1 inhibitor naringin. In conclusion, this study suggests that OATP1A2 and 2B1 may play a role in the uptake of DR agonists and antagonists into the brain.


Subject(s)
Dopamine Agonists/metabolism , Dopamine Antagonists/metabolism , Organic Anion Transporters/metabolism , Animals , Brain/metabolism , Bromocriptine/metabolism , Cell Line , Dogs , Domperidone/metabolism , Dopamine , Humans , Pituitary Gland, Anterior/metabolism , Tandem Mass Spectrometry
11.
Pharmacol Res ; 161: 105092, 2020 11.
Article in English | MEDLINE | ID: mdl-32758634

ABSTRACT

The impact of the C-X-C receptor (CXCR) 7 and its close co-player CXCR4 in different physiological and pathophysiological processes has been extensively investigated within the last decades. Following activation by their shared ligand C-X-C ligand (CXCL) 12, both chemokine receptors can induce various routes of cell signaling and/or scavenge CXCL12 from the extracellular environment. This contributes to organ development and maintenance of homeostasis. Alterations of the CXCR4/CXCR7-CXCL12 axis have been detected in diseases such as cancer, central nervous system and cardiac disorders, and autoimmune diseases. These alterations include changes of the expression pattern, distribution, or downstream effects. The progression of the diseases can be regulated in preclinical models by the use of various modulators suggesting that this axis serves as a promising therapeutic target. It is therefore of great interest to investigate CXCR4/CXCR7/CXCL12 modulators in clinical development, with several CXCR4 and CXCL12 modulators such as plerixafor, ulocuplumab, balixafortide, and olaptesed pegol having already reached this stage. An overview is presented of the most important diseases whose outcomes can be positively or negatively regulated by the CXCR4/CXCR7-CXCL12 axis and summarizes preclinical and clinical data of modulators of that axis. Contrary to CXCR4 and CXCL12 modulators, CXCR7 modulators have, thus far, not been extensively studied. Therefore, more (pre)clinical investigations are needed.


Subject(s)
Central Nervous System Diseases/metabolism , Chemokine CXCL12/metabolism , Neoplasms/metabolism , Receptors, CXCR4/metabolism , Receptors, CXCR/metabolism , Animals , Antineoplastic Agents/therapeutic use , Central Nervous System Agents/therapeutic use , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/pathology , Central Nervous System Diseases/physiopathology , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/physiopathology , Signal Transduction
12.
Pharmacol Res ; 161: 105155, 2020 11.
Article in English | MEDLINE | ID: mdl-32818652

ABSTRACT

The family of Organic Anion Transporting Polypeptides are known to facilitate the transmembrane transport. OATP1B3-1B7 is a novel member of the OATP1B-subfamily, and is encoded by SLCO1B3-SLCO1B7 readthrough deriving from the genes SLCO1B3 and SLCO1B7 on chromosome 12. The resulting protein is expressed in the smooth endoplasmatic reticulum of hepatocytes, is functional, and transports dehydroepiandrosterone-sulfate (DHEAS). In the gene area encoding for the 1B7-part of the protein, there are coding polymorphisms. It was the aim of this study to test the frequency and the impact of these genetic variants on transport activity. The minor allele frequency (MAF) of the coding polymorphisms was determined in a cohort of 192 individuals. DHEAS transport function was determined by applying the vTF-7 based heterologous expression system using plasmids encoding for OATP1B3-1B7 or the respective variants. The genetic variants 641 T (MAF 0.021), 1073 G (MAF 0.169) and 1775 A (MAF 0.013) significantly reduced DHEAS accumulation in cells transfected with OATP1B3-1B7, albeit without significantly influencing expression of the transporter as determined by Western blot analysis and immunofluorescence after heterologous expression. Genotyping revealed complete linkage of the variants 884A, 1073 G and 1501C. Presence of the haplotype abolished the DHEAS-transport function of OATP1B3-1B7. Naturally and frequently occurring genetic variants located within the gene region of SLCO1B7 encoding for the 1B7-part of OATP1B3-1B7 influence the in vitro function of this member of the OATP1B-family. With their functional characterisation, we provide the basis for pharmacogenetic studies, which may help to understand the in vivo relevance of this transporter.


Subject(s)
Dehydroepiandrosterone Sulfate/metabolism , Organic Anion Transporters/genetics , Pharmacogenomic Variants , Polymorphism, Single Nucleotide , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics , Solute Carrier Proteins/genetics , Biological Transport , Databases, Genetic , Gene Frequency , Haplotypes , HeLa Cells , Humans , Kinetics , Organic Anion Transporters/metabolism , Phenotype , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Solute Carrier Proteins/metabolism
13.
Mol Pharmacol ; 96(2): 128-137, 2019 08.
Article in English | MEDLINE | ID: mdl-31127008

ABSTRACT

Drug transporters play a crucial role in pharmacokinetics. One subfamily of transporters with proven clinical relevance are the OATP1B transporters. Recently we identified a new member of the OATP1B family named OATP1B3-1B7 (LST-3TM12). This functional transporter is encoded by SLCO1B3 and SLCO1B7 OATP1B3-1B7 is expressed in hepatocytes and is located in the membrane of the smooth endoplasmic reticulum (SER). One aim of this study was to test whether OATP1B3-1B7 interacts with commercial drugs. First, we screened a selection of OATP1B substrates for inhibition of OATP1B3-1B7-mediated transport of dehydroepiandrosterone sulfate and identified several inhibitors. One such inhibitor was ezetimibe, which not only inhibited OATP1B3-1B7 but is also a substrate, as its cellular content was significantly increased in cells heterologously expressing the transporter. In humans, ezetimibe is extensively metabolized by hepatic and intestinal uridine-5'-diphospho-glucuronosyltransferases (UGTs), the catalytic site of which is located within the SER lumen. After verification of OATP1B3-1B7 expression in the small intestine, we determined in microsomes whether SER access can be modulated by inhibitors of OATP1B3-1B7. We were able to show that these compounds significantly reduced accumulation in small intestinal and hepatic microsomes, which influenced the rate of ezetimibe ß-D-glucuronide formation as determined in microsomes treated with bromsulphthalein. Notably, this molecule not only inhibits the herein reported transporter but also other transport systems. In conclusion, we report that multiple drugs interact with OATP1B3-1B7; for ezetimibe, we were able to show that SER access and metabolism is significantly reduced by bromsulphthalein, which is an inhibitor of OATP1B3-1B7. SIGNIFICANCE STATEMENT: OATP1B3-1B3 (LST-3TM12) is a transporter that has yet to be fully characterized. We provide valuable insight into the interaction potential of this transporter with several marketed drugs. Ezetimibe, which interacted with OATP1B3-1B7, is highly metabolized by uridine-5'-diphospho-glucuronosyltransferases (UGTs), whose catalytic site is located within the smooth endoplasmic reticulum (SER) lumen. Through microsomal assays with ezetimibe and the transport inhibitor bromsulphthalein we investigated the interdependence of SER access and the glucuronidation rate of ezetimibe. These findings led us to the hypothesis that access or exit of drugs to the SER is orchestrated by SER transporters such as OATP1B3-1B7.


Subject(s)
Endoplasmic Reticulum, Smooth/chemistry , Ezetimibe/pharmacokinetics , Organic Anion Transporters/metabolism , Solute Carrier Proteins/metabolism , Sulfobromophthalein/pharmacology , Biological Transport , Catalytic Domain , Glucuronosyltransferase/chemistry , HeLa Cells , Humans , Intestine, Small/metabolism , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism
14.
Mol Pharmacol ; 95(3): 313-323, 2019 03.
Article in English | MEDLINE | ID: mdl-30573512

ABSTRACT

The herbal remedy St. John's wort (SJW) is used in the treatment of mild depressive symptoms and is known for its drug-drug interaction potential when enhanced expression of CYP3A4 modifies clearance of concomitantly applied substrate drugs. Hyperforin is one constituent of SJW that alters CYP3A4 expression by activation of the nuclear receptor pregnane X receptor (PXR). However, little is known about the transmembrane transport of hyperforin. One membrane protein that modulates cellular entry of drugs is the organic anion-transporting polypeptide (OATP) 2B1. It was the aim of this study to test whether hyperforin interacts with this transport protein. Transport inhibition studies and competitive counterflow experiments suggested that hyperforin is a substrate of OATP2B1. This notion was validated by showing that the presence of OATP2B1 enhanced the hyperforin-induced PXR activation in cell-based luciferase assays. Moreover, in Caco-2 cells transcellular transport of the known OATP2B1 substrate atorvastatin was changed in the presence of hyperforin, resulting in an increased efflux ratio. Eleven commercially available SJW formulations were assessed for their influence on OATP2B1-mediated transport of estrone 3-sulfate and for their impact on CYP3A4 promoter transactivation. The correlation between effect size and the hyperforin content as determined by high-performance liquid chromatography with ultraviolet detection suggested that hyperforin is the major determinant. Our results indicate an interaction between hyperforin and OATP2B1, which is not only known to contribute to hepatocellular uptake but also to intestinal absorption of its substrates. These findings extend the complexity of mechanisms that should be considered when evaluating the interaction potential of SJW preparations.


Subject(s)
Organic Anion Transporters/metabolism , Phloroglucinol/analogs & derivatives , Pregnane X Receptor/metabolism , Terpenes/pharmacology , Biological Transport/drug effects , Caco-2 Cells , Cell Line, Tumor , Cytochrome P-450 CYP3A/metabolism , Drug Interactions/physiology , Estrone/analogs & derivatives , Estrone/metabolism , HeLa Cells , Hep G2 Cells , Humans , Intestinal Absorption/drug effects , Phloroglucinol/pharmacology , Promoter Regions, Genetic/drug effects
15.
Am J Physiol Gastrointest Liver Physiol ; 317(6): G751-G762, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31509437

ABSTRACT

Organic anion transporting polypeptide (OATP) 1B3-1B7 (LST-3TM12) is a member of the OATP1B [solute carrier organic anion transporter (SLCO) 1B] family. This transporter is not only functional but also expressed in the membrane of the smooth endoplasmic reticulum of hepatocytes and enterocytes. OATP1B3-1B7 is a splice variant of SLCO1B3 in which the initial part is encoded by SLCO1B3, whereas the rest of the mRNA originates from the gene locus of SLCO1B7. In this study, we not only showed that SLCO1B3 and the mRNA encoding for OATP1B3-1B7 share the 5' untranslated region but also that silencing of an initial SLCO1B3 exon lowered the amount of SLCO1B3 and of SLCO1B7 mRNA in Huh-7 cells. To validate the assumption that both transcripts are regulated by the same promoter we tested the influence of the bile acid sensor farnesoid X receptor (FXR) on their transcription. Treatment of Huh-7 and HepaRG cells with activators of this known regulator of OATP1B3 not only increased SLCO1B3 but also OATP1B3-1B7 mRNA transcription. Applying a heterologous expression system, we showed that several bile acids interact with OATP1B3-1B7 and that taurocholic acid and lithocholic acid are OATP1B3-1B7 substrates. As OATP1B3-1B7 is located in the smooth endoplasmic reticulum, it may grant access to metabolizing enzymes. In accordance are our findings showing that the OATP1B3-1B7 inhibitor bromsulphthalein significantly reduced uptake of bile acids into human liver microsomes. Taken together, we report that OATP1B3-1B7 transcription can be modulated with FXR agonists and antagonists and that OATP1B3-1B7 transports bile acids.NEW & NOTEWORTHY Our study on the transcriptional regulation of the novel organic anion transporting polypeptide (OATP) 1B3-1B7 concludes that the promoter of solute carrier organic anion transporter (SLCO) 1B3 governs SLCO1B3-1B7 transcription. Moreover, the transcription of OATP1B3-1B7 can be modulated by farnesoid X receptor (FXR) agonists and antagonists. FXR is a major regulator in bile acid homeostasis that links OATP1B3-1B7 to this physiological function. Findings in transport studies with OATP1B3-1B7 suggest that this transporter interacts with the herein tested bile acids.


Subject(s)
Bile Acids and Salts/physiology , Isoxazoles/pharmacology , Organic Anion Transporters , Receptors, Cytoplasmic and Nuclear , Solute Carrier Organic Anion Transporter Family Member 1B3 , Solute Carrier Proteins , Antineoplastic Agents/pharmacology , Biological Transport/physiology , Gene Expression Regulation , Gene Regulatory Networks/physiology , HeLa Cells , Hep G2 Cells , Humans , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Solute Carrier Proteins/genetics , Solute Carrier Proteins/metabolism , Transcription Factors , Transcriptional Activation
16.
Nephrol Dial Transplant ; 34(4): 692-702, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30189012

ABSTRACT

BACKGROUND: The pharmacokinetics of oxycodone in patients with end-stage renal disease (ESRD) requiring haemodialysis are largely unknown. Therefore, we investigated the pharmacokinetics of oxycodone/naloxone prolonged release and their metabolites in patients with ESRD during and between haemodialysis sessions. METHODS: Single doses of oxycodone/naloxone (5/2.5 or 10/5 mg) were administered in nine patients with ESRD using a cross-over design on the day of dialysis and on a day between dialysis sessions. Plasma, dialysate and urine concentrations of oxycodone, naloxone and their metabolites were determined up to 48 h post-dosing using a liquid chromatography-tandem mass spectrometry system. RESULTS: Haemodialysis performed 6-10 h after dosing removed ∼10% of the administered dose of oxycodone predominantly as unconjugated oxycodone and noroxycodone or conjugated oxymorphone and noroxymorphone. The haemodialysis clearance of oxycodone based on its recovery in dialysate was (mean ± SD) 8.4 ± 2.1 L/h. The geometric mean (coefficient of variation) plasma elimination half-life of oxycodone during the 4-h haemodialysis period was 3.9 h (39%) which was significantly shorter than the 5.7 h (22%) without haemodialysis. Plasma levels of the active metabolite oxymorphone in its unconjugated form were very low. CONCLUSIONS: Oxycodone is removed during haemodialysis. The pharmacokinetics including the relatively short half-life of oxycodone in patients with ESRD with or without haemodialysis and the absence of unconjugated active metabolites indicate that oxycodone can be used at usual doses in patients requiring dialysis.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Kidney Failure, Chronic/drug therapy , Naloxone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Oxycodone/pharmacokinetics , Renal Dialysis/methods , Adult , Aged , Analgesics, Opioid/administration & dosage , Cross-Over Studies , Female , Humans , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/therapy , Kidney Function Tests , Male , Middle Aged , Morphinans/administration & dosage , Morphinans/pharmacokinetics , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Oxycodone/administration & dosage , Oxymorphone/administration & dosage , Oxymorphone/pharmacokinetics , Prognosis , Tissue Distribution
17.
Mol Pharmacol ; 94(1): 700-712, 2018 07.
Article in English | MEDLINE | ID: mdl-29735582

ABSTRACT

Levothyroxine replacement therapy forms the cornerstone of hypothyroidism management. Variability in levothyroxine oral absorption may contribute to the well-recognized large interpatient differences in required dose. Moreover, levothyroxine-drug pharmacokinetic interactions are thought to be caused by altered oral bioavailability. Interestingly, little is known regarding the mechanisms contributing to levothyroxine absorption in the gastrointestinal tract. Here, we aimed to determine whether the intestinal drug uptake transporter organic anion transporting polypeptide 2B1 (OATP2B1) may be involved in facilitating intestinal absorption of thyroid hormones. We also explored whether thyroid hormones regulate OATP2B1 gene expression. In cultured Madin-Darby Canine Kidney II/OATP2B1 cells and in OATP2B1-transfected Caco-2 cells, thyroid hormones were found to inhibit OATP2B1-mediated uptake of estrone-3-sulfate. Competitive counter-flow experiments evaluating the influence on the cellular accumulation of estrone-3-sulfate in the steady state indicated that thyroid hormones were substrates of OATP2B1. Additional evidence that thyroid hormones were OATP2B1 substrates was provided by OATP2B1-dependent stimulation of thyroid hormone receptor activation in cell-based reporter assays. Bidirectional transport studies in intestinal Caco-2 cells showed net absorptive flux of thyroid hormones, which was attenuated by the presence of the OATP2B1 inhibitor, atorvastatin. In intestinal Caco-2 and LS180 cells, but not in liver Huh-7 or HepG2 cells, OATP2B1 expression was induced by treatment with thyroid hormones. Reporter gene assays revealed thyroid hormone receptor α-mediated transactivation of the SLCO2B1 1b and the SLCO2B1 1e promoters. We conclude that thyroid hormones are substrates and transcriptional regulators of OATP2B1. These insights provide a potential mechanistic basis for oral levothyroxine dose variability and drug interactions.


Subject(s)
Organic Anion Transporters/metabolism , Thyroid Hormones/metabolism , Transcription, Genetic/physiology , Animals , Atorvastatin/pharmacology , Biological Transport/drug effects , Biological Transport/physiology , Caco-2 Cells , Cell Line , Cell Line, Tumor , Dogs , Drug Interactions/physiology , Estrone/analogs & derivatives , Estrone/pharmacology , Genes, Reporter/drug effects , Genes, Reporter/physiology , HeLa Cells , Hep G2 Cells , Humans , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Madin Darby Canine Kidney Cells , Organic Anion Transporters/antagonists & inhibitors , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , Transcription, Genetic/drug effects
18.
Mol Pharm ; 15(11): 4884-4897, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30274515

ABSTRACT

Cytotoxic compounds used to treat cancer are often associated with adverse events. The development of formulations activated by tumor-specific triggers would allow a reduction of systemic exposure while maintaining therapeutic concentrations in the tumor. One enzyme with proteolytic activity reported to be involved in tumor progression and assumed to be enhanced in the tumor environment is the matrix metalloproteinase 9 (MMP-9). In our study, we aimed to develop surface-modified PDMS-PMOXA polymersomes able to release their cytotoxic payload upon digestion by MMP-9. To test the applicability of such a system in breast cancer, this tumor entity was assessed for MMP-9 expression, supporting breast cancer as a potential target. The surface-modified polymersomes were synthesized and formulated resulting in paclitaxel-loaded particles of about 320 ± 153.15 nm in size with a surface potential of 0.04 ± 0.007 mV. After the expression and activity of MMP-9 in MCF7 cells were verified, this cell line was used for further analysis. Treatment of MCF7 cells with the polymersomes significantly reduced cell viability, this effect was abolished after addition of MMP-inhibitors, suggesting proteolytic activation. In zebrafish embryos, the polymersomes were observed in the circulation with some enrichment in liver and agglomerates in the caudal veins. Importantly, in zebrafish embryos xenografted with mKate2-expressing MCF7 cells, the amount of tumor cells, quantified by detecting the copies of the heterologously expressed fluorescent protein, significantly decreased after treatment with PDMS-PMOXA-SRL-paclitaxel polymersomes. Taken together, our data suggest that polymersomes modified with an MMP-9 labile peptide and loaded with paclitaxel can be formulated, and that these particles exert pharmacological activity upon enzymatic digestion.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Breast Neoplasms/drug therapy , Drug Carriers/chemical synthesis , Matrix Metalloproteinase 9/metabolism , Oxazoles/chemical synthesis , Polymers/chemical synthesis , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Breast Neoplasms/pathology , Drug Liberation , Embryo, Nonmammalian , Female , Humans , MCF-7 Cells , Nanoparticles/chemistry , Paclitaxel/administration & dosage , Paclitaxel/pharmacokinetics , Treatment Outcome , Xenograft Model Antitumor Assays , Zebrafish
19.
Mol Pharm ; 15(12): 5501-5513, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30380886

ABSTRACT

The organic anion transporting polypeptide (OATP) 2B1 is ubiquitously expressed and known to facilitate cellular entry. It is widely accepted that transport proteins play a pivotal role in pharmacokinetics. Consequently, testing for interaction with drug transporters became an important part in the assessment of new molecular entities in order to predict and prevent drug-drug interactions. Recently, competitive counterflow (CCF), an indirect method allowing the identification of substrates, was successfully applied to the organic cation transporter 2. It was the aim of this study to test whether CCF can be used to identify substrates of OATP2B1. A protocol for CCF experiments using estrone 3-sulfate (E1S) as the driven compound in expression-verified MDCKII-OATP2B1 cells was established. The protocol was tested using a substance library, which was prior screened for inhibition of OATP2B1-mediated transport accounting for both E1S-binding sites. In CCF experiments, all previously reported OATP2B1 substrates significantly reduced the amount of E1S in equilibrium, classifying them as substrates. In addition, we identified and verified novel substrates of OATP2B1, namely, astemizole and domperidone. Results of the CCF were complemented with cytotoxicity assays or cell-based reporter gene assays to validate the finding of etoposide and teniposide or hyperforin being substrates of OATP2B1, respectively. Our study indicates that the method of CCF can be used to identify substrates of OATP2B1, irrespective, whether interacting with binding site A or A and B, but is limited by solubility issues or the amount of transporter that is expressed in the used cellular system.


Subject(s)
Chemistry, Pharmaceutical/methods , Drug Interactions , Organic Anion Transporters/metabolism , Small Molecule Libraries/pharmacokinetics , Animals , Chemistry, Pharmaceutical/instrumentation , Dogs , Estrone/analogs & derivatives , Estrone/pharmacokinetics , HeLa Cells , Hep G2 Cells , Humans , Madin Darby Canine Kidney Cells , Organic Anion Transporters/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
20.
Eur J Clin Pharmacol ; 74(3): 275-283, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29198060

ABSTRACT

PURPOSE: Methylenedioxymethamphetamine (MDMA, ecstasy) is used recreationally and frequently leads to sympathomimetic toxicity. MDMA produces cardiovascular and subjective stimulant effects that were shown to partially depend on the norepinephrine transporter (NET)-mediated release of norepinephrine and stimulation of α1-adrenergic receptors. Genetic variants, such as single-nucleotide polymorphisms (SNPs), of the NET gene (SLC6A2) may explain interindividual differences in the acute stimulant-type responses to MDMA in humans. METHODS: We characterized the effects of common genetic variants of the SLC6A2 gene (rs168924, rs47958, rs1861647, rs2242446, and rs36029) on cardiovascular and subjective stimulation after MDMA administration in 124 healthy subjects in a pooled analysis of eight double-blind, placebo-controlled studies. RESULTS: Carriers of the GG genotype of the SLC6A2 rs1861647 SNP presented higher elevations of heart rate and rate-pressure product after MDMA than subjects with one or no G alleles. Subjects with a C allele in the SLC6A2 rs2242446 SNP presented higher elevations of the heart rate after MDMA administration compared with the TT genotype. Subjects with the AA genotype of the SLC6A2 rs36029 SNP presented higher elevations of mean arterial pressure and rate pressure product after MDMA administration than carriers of the G allele. The SLC6A2 rs168924 and rs47958 SNPs did not alter the response to MDMA. CONCLUSIONS: Genetic polymorphisms of the SLC6A2 gene weakly moderated the acute cardiovascular response to MDMA in controlled studies and may play a minor role in adverse cardiovascular events when MDMA is used recreationally.


Subject(s)
Adrenergic Uptake Inhibitors/toxicity , Cardiovascular Physiological Phenomena/drug effects , Illicit Drugs/toxicity , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Norepinephrine Plasma Membrane Transport Proteins/genetics , Polymorphism, Single Nucleotide , Psychotropic Drugs/toxicity , Adolescent , Adult , Alleles , Blood Pressure/drug effects , Cardiotoxins/toxicity , Cross-Over Studies , Double-Blind Method , Female , Gene Frequency , Genetic Association Studies , Heart Rate/drug effects , Heterozygote , Humans , Male , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Pharmacogenetics/methods , Switzerland , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL