Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cells ; 12(8)2023 04 12.
Article in English | MEDLINE | ID: mdl-37190047

ABSTRACT

Enteroviruses are a leading cause of upper respiratory tract, gastrointestinal, and neurological infections. Management of enterovirus-related diseases has been hindered by the lack of specific antiviral treatment. The pre-clinical and clinical development of such antivirals has been challenging, calling for novel model systems and strategies to identify suitable pre-clinical candidates. Organoids represent a new and outstanding opportunity to test antiviral agents in a more physiologically relevant system. However, dedicated studies addressing the validation and direct comparison of organoids versus commonly used cell lines are lacking. Here, we described the use of human small intestinal organoids (HIOs) as a model to study antiviral treatment against human enterovirus 71 (EV-A71) infection and compared this model to EV-A71-infected RD cells. We used reference antiviral compounds such as enviroxime, rupintrivir, and 2'-C-methylcytidine (2'CMC) to assess their effects on cell viability, virus-induced cytopathic effect, and viral RNA yield in EV-A71-infected HIOs and cell line. The results indicated a difference in the activity of the tested compounds between the two models, with HIOs being more sensitive to infection and drug treatment. In conclusion, the outcome reveals the value added by using the organoid model in virus and antiviral studies.


Subject(s)
Enterovirus A, Human , Enterovirus Infections , Enterovirus , Humans , Antiviral Agents/pharmacology , Enterovirus A, Human/physiology , Enterovirus Infections/drug therapy , Organoids
2.
Stem Cell Reports ; 15(4): 983-998, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33053362

ABSTRACT

Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) are commercially available, and cardiac differentiation established routine. Systematic evaluation of several control hiPSC-CM is lacking. We investigated 10 different control hiPSC-CM lines and analyzed function and suitability for drug screening. Five commercial and 5 academic hPSC-CM lines were casted in engineered heart tissue (EHT) format. Spontaneous and stimulated EHT contractions were analyzed, and 7 inotropic indicator compounds investigated on 8 cell lines. Baseline contractile force, kinetics, and rate varied widely among the different lines (e.g., relaxation time range: 118-471 ms). In contrast, the qualitative correctness of responses to BayK-8644, nifedipine, EMD-57033, isoprenaline, and digoxin in terms of force and kinetics varied only between 80% and 93%. Large baseline differences between control cell lines support the request for isogenic controls in disease modeling. Variability appears less relevant for drug screening but needs to be considered, arguing for studies with more than one line.


Subject(s)
Drug Evaluation, Preclinical , Heart/physiology , Induced Pluripotent Stem Cells/cytology , Tissue Engineering , Calcium/metabolism , Cell Line , Extracellular Space/chemistry , Fluorescence , Gene Expression Regulation , Humans , Myocardial Contraction , Myocytes, Cardiac/cytology
3.
Toxicol Sci ; 176(1): 103-123, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32421822

ABSTRACT

Animal models are 78% accurate in determining whether drugs will alter contractility of the human heart. To evaluate the suitability of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) for predictive safety pharmacology, we quantified changes in contractility, voltage, and/or Ca2+ handling in 2D monolayers or 3D engineered heart tissues (EHTs). Protocols were unified via a drug training set, allowing subsequent blinded multicenter evaluation of drugs with known positive, negative, or neutral inotropic effects. Accuracy ranged from 44% to 85% across the platform-cell configurations, indicating the need to refine test conditions. This was achieved by adopting approaches to reduce signal-to-noise ratio, reduce spontaneous beat rate to ≤ 1 Hz or enable chronic testing, improving accuracy to 85% for monolayers and 93% for EHTs. Contraction amplitude was a good predictor of negative inotropes across all the platform-cell configurations and of positive inotropes in the 3D EHTs. Although contraction- and relaxation-time provided confirmatory readouts forpositive inotropes in 3D EHTs, these parameters typically served as the primary source of predictivity in 2D. The reliance of these "secondary" parameters to inotropy in the 2D systems was not automatically intuitive and may be a quirk of hiPSC-CMs, hence require adaptations in interpreting the data from this model system. Of the platform-cell configurations, responses in EHTs aligned most closely to the free therapeutic plasma concentration. This study adds to the notion that hiPSC-CMs could add value to drug safety evaluation.


Subject(s)
Dose-Response Relationship, Drug , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Pharmaceutical Preparations , Animals , Humans
4.
Article in English | MEDLINE | ID: mdl-29355722

ABSTRACT

Safety pharmacology studies that evaluate drug candidates for potential cardiovascular liabilities remain a critical component of drug development. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have recently emerged as a new and promising tool for preclinical hazard identification and risk assessment of drugs. Recently, Pluriomics organized its first User Meeting entitled 'Combining Pluricyte® Cardiomyocytes & MEA for Safety Pharmacology applications', consisting of scientific sessions and live demonstrations, which provided the opportunity to discuss the application of hiPSC-CMs (Pluricyte® Cardiomyocytes) in cardiac safety assessment to support early decision making in safety pharmacology. This report summarizes the outline and outcome of this Pluriomics User Meeting, which took place on November 24-25, 2016 in Leiden (The Netherlands). To reflect the content of the communications presented at this meeting we have cited key scientific articles and reviews.


Subject(s)
Action Potentials/drug effects , Drug Evaluation, Preclinical/methods , Myocytes, Cardiac/drug effects , Cardiotoxicity/prevention & control , Cell Line , Drug Evaluation, Preclinical/instrumentation , Drug Evaluation, Preclinical/standards , Electrodes , Guidelines as Topic , Humans , Induced Pluripotent Stem Cells/physiology , Myocardial Contraction/drug effects , Myocytes, Cardiac/physiology , Patch-Clamp Techniques/instrumentation , Patch-Clamp Techniques/methods
5.
Nucleic Acid Ther ; 27(4): 221-231, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28418733

ABSTRACT

Antisense oligonucleotides (AONs) are promising candidates for treatment of Duchenne muscular dystrophy (DMD), a severe and progressive disease resulting in premature death. However, more knowledge on the pharmacokinetics of new AON drug candidates is desired for effective application in the clinic. We assessed the feasibility of using noninvasive single-photon emission computed tomography-computed tomography (SPECT-CT) imaging to determine AON pharmacokinetics in vivo. To this end, a 2'-O-methyl phosphorothioate AON was radiolabeled with 123I or 111In, and administered to mdx mice, a rodent model of DMD. SPECT-CT imaging was performed to determine AON tissue levels, and the results were compared to data obtained with invasive analysis methods (scintillation counting and a ligation-hybridization assay). We found that SPECT-CT data obtained with 123I-AON and 111In-AON were qualitatively comparable to data derived from invasive analytical methods, confirming the feasibility of using SPECT-CT analysis to study AON pharmacokinetics. Notably, also AON uptake in skeletal muscle, the target tissue in DMD, could be readily quantified using SPECT-CT imaging, which was considered a particular challenge in mice, due to their small size. In conclusion, our results demonstrate that SPECT-CT imaging allows for noninvasive characterization of biodistribution and pharmacokinetics of AONs, thereby enabling quantitative comparisons between different radiolabeled AON drug candidates and qualitative conclusions about the corresponding unmodified parent AONs. This technology may contribute to improved (pre)clinical drug development, leading to drug candidates with optimized characteristics in vivo.


Subject(s)
Muscular Dystrophy, Duchenne/diagnostic imaging , Oligonucleotides, Antisense/pharmacokinetics , Phosphorothioate Oligonucleotides/pharmacokinetics , Animals , Femur/diagnostic imaging , Femur/metabolism , Iodine Radioisotopes/pharmacokinetics , Male , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/metabolism , Tissue Distribution , Tomography, Emission-Computed, Single-Photon
6.
Clin Pharmacokinet ; 55(1): 1-15, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26242381

ABSTRACT

Microdosing studies allow clinical investigation of pharmacokinetics earlier in drug development, before all high-dose safety concerns have been sorted out. Furthermore, microdosing allows inclusion of target groups that are inadmissible in high-dose phase I trials. A potential concern when considering a microdosing study is that a particular drug candidate may display non-linear pharmacokinetics. Saturation of, for example, membrane transport or metabolism at exposure levels between the microdose and therapeutic dose may limit the predictivity of high-dose pharmacokinetics from microdose observations. Guidance on the likelihood of appreciable non-linear pharmacokinetics based on preclinical information can be helpful in staging the clinical phase and the place of microdosing in it. We present a decision tree that evaluates concerns about non-linearities raised in the preclinical phase and their potential impact on the proportionality between microdose and intended therapeutic dose as predicted from preclinical information. The expected maximum concentrations at relevant sites are estimated by non-compartmental methods. These are compared with dissolution, Michaelis constants for active or enzymatic processes, and binding protein concentrations to assess the potential saturation of the processes below therapeutic doses. The decision tree was applied to ten published cases comparing microdose and therapeutic dose pharmacokinetics, for which concerns about non-linear pharmacokinetics were raised a priori. The decision tree was able to discriminate cases showing substantial non-linearities from cases displaying dose-proportional pharmacokinetics. The recommendations described in this paper may be useful in deciding whether a microdosing study is a sensible option to gain early insight in clinical pharmacokinetics of drug candidates.


Subject(s)
Clinical Trials as Topic/methods , Decision Trees , Pharmaceutical Preparations/administration & dosage , Pharmacokinetics , Dose-Response Relationship, Drug , Humans , Models, Biological , Nonlinear Dynamics
7.
Nucl Med Biol ; 42(11): 833-41, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26264927

ABSTRACT

INTRODUCTION: The efflux transporters P-glycoprotein (P-gp, ABCB1) and breast cancer resistance protein (BCRP, ABCG2) are expressed at the blood-brain barrier (BBB), and can limit the access of a wide range of drugs to the brain. In this study we developed a PET-CT imaging method for non-invasive, quantitative analysis of the effect of ABCB1 and ABCG2 on brain penetration of the anti-cancer drug gefitinib, and demonstrated the applicability of this method for identification and quantification of potential modulators of ABCB1 and ABCB2 using the dual inhibitor elacridar. METHODS: In vitro cellular accumulation studies with [(14)C]-gefitinib were conducted in LLC-PK1, MDCKII, and the corresponding ABCB1/Abcb1a and ABCG2/Abcg2 overexpressing cell lines. Subsequently, in vivo brain penetration of [(18)F]-gefitinib was quantified by PET-CT imaging studies in wild-type, Abcg2(-/-), Abcb1a/1b(-/-), and Abcb1a/1b;Abcg2(-/-) mice. RESULTS: In vitro studies showed that [(14)C]-gefitinib is a substrate of the human ABCB1 and ABCG2 transporters. After i.v. administration of [(18)F]-gefitinib (1mg/kg), PET-CT imaging showed 2.3-fold increased brain levels of [(18)F]-gefitinib in Abcb1a/1b;Abcg2(-/-) mice, compared to wild-type. Levels in single knockout animals were not different from wild-type, showing that Abcb1a/1b and Abcg2 together limit access of [(18)F]-gefitinib to the brain. Furthermore, enhanced brain accumulation of [(18)F]-gefitinib after administration of the ABCB1 and ABCG2 inhibitor elacridar (10 mg/kg) could be quantified with PET-CT imaging. CONCLUSIONS: PET-CT imaging with [(18)F]-gefitinib is a powerful tool to non-invasively assess potential ABCB1- and ABCG2-mediated drug-drug interactions (DDIs) in vivo. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE: This minimally-invasive, [(18)F]-based PET-CT imaging method shows the interplay of ABCB1 and ABCG2 at the BBB in vivo. The method may be applied in the future to assess ABCB1 and ABCG2 activity at the BBB in humans, and for personalized treatment with drugs that are substrates of ABCB1 and/or ABCG2.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP-Binding Cassette Transporters/metabolism , Blood-Brain Barrier/metabolism , Fluorine Radioisotopes , Positron-Emission Tomography , Quinazolines/metabolism , Tomography, X-Ray Computed , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Acridines/pharmacology , Animals , Biological Transport/drug effects , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/drug effects , Cell Line, Tumor , Drug Interactions , Gefitinib , Humans , Male , Mice , Quinazolines/pharmacokinetics , Tetrahydroisoquinolines/pharmacology , Tissue Distribution
8.
J Nucl Med ; 55(8): 1330-5, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24914056

ABSTRACT

UNLABELLED: The experimental autoimmune encephalomyelitis model is a model of multiple sclerosis that closely mimics the disease characteristics in humans. The main hallmarks of multiple sclerosis are neuroinflammation (microglia activation, monocyte invasion, and T-cell infiltration) and demyelination. PET imaging may be a useful noninvasive technique for monitoring disease progression and drug treatment efficacy in vivo. METHODS: Experimental autoimmune encephalomyelitis was induced by myelin-oligodendrocyte glycoprotein immunization in female Dark Agouti rats. Experimental autoimmune encephalomyelitis rats were imaged at baseline and at days 6, 11, 15, and 19 after immunization to monitor monocyte and microglia activation ((11)C-PK11195) and demyelination ((11)C-MeDAS) during normal disease progression and during treatment with dexamethasone. RESULTS: (11)C-PK11195 PET detected activation of microglia and monocytes in the brain stem and spinal cord during disease progression. The uptake of (11)C-PK11195 was elevated in dexamethasone-treated animals that had shown mild clinical symptoms that had resolved at the time of imaging. Demyelination was not detected by (11)C-MeDAS PET, probably because of the small size of the lesions (average, 0.13 mm). CONCLUSION: PET imaging of neuroinflammation can be used to monitor disease progression and the consequences of treatment in the experimental autoimmune encephalomyelitis rat model. PET imaging was more sensitive than clinical symptoms for detecting inflammatory changes in the central nervous system.


Subject(s)
Disease Progression , Encephalomyelitis, Autoimmune, Experimental/diagnostic imaging , Encephalomyelitis, Autoimmune, Experimental/therapy , Positron-Emission Tomography , Amides , Aniline Compounds , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Isoquinolines , Microglia/pathology , Monocytes/immunology , Myelin Sheath/metabolism , Rats , Stilbenes , Treatment Outcome
9.
Mol Pharmacol ; 85(3): 520-30, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24334255

ABSTRACT

The multidrug transporters breast cancer resistance protein (BCRP), multidrug-resistance protein 1 (MDR1), and multidrug-resistance-associated protein (MRP) 2 and 3 eliminate toxic compounds from tissues and the body and affect the pharmacokinetics of many drugs and other potentially toxic compounds. The food-derived carcinogen PhIP (2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine) is transported by BCRP, MDR1, and MRP2. To investigate the overlapping functions of Bcrp1, Mdr1a/b, and Mrp2 in vivo, we generated Bcrp1;Mdr1a/b;Mrp2(-/-) mice, which are viable and fertile. These mice, together with Bcrp1;Mrp2;Mrp3(-/-) mice, were used to study the effects of the multidrug transporters on the pharmacokinetics of PhIP and its metabolites. Thirty minutes after oral or intravenous administration of PhIP (1 mg/kg), the PhIP levels in the small intestine were reduced 4- to 6-fold in Bcrp1;Mdr1a/b;Mrp2(-/) (-) and Bcrp1;Mrp2;Mrp3(-/-) mice compared with wild-type mice. Fecal excretion of PhIP was reduced 8- to 20-fold in knockouts. Biliary PhIP excretion was reduced 41-fold in Bcrp1;Mdr1a/b;Mrp2(-/-) mice. Biliary and small intestine levels of PhIP metabolites were reduced in Bcrp1;Mrp2-deficient mice. Furthermore, in both knockout strains, kidney levels and urinary excretion of genotoxic PhIP-metabolites were significantly increased, suggesting that reduced biliary excretion of PhIP and PhIP metabolites leads to increased urinary excretion of these metabolites and increased systemic exposure. Bcrp1 and Mdr1a limited PhIP brain accumulation. In Bcrp1;Mrp2;Mrp3(-/-), but not Bcrp1;Mdr1a/b;Mrp(-/-) mice, the carcinogenic metabolites N2-OH-PhIP (2-hydroxyamino-1-methyl-6-phenylimidazo[4,5-b]pyridine) and PhIP-5-sulfate (a genotoxicity marker) accumulated in liver tissue, indicating that Mrp3 is involved in the sinusoidal secretion of these compounds. We conclude that Bcrp1, Mdr1a/b, Mrp2, and Mrp3 significantly affect tissue disposition and biliary and fecal elimination of PhIP and its carcinogenic metabolites and may affect PhIP-induced carcinogenesis as a result.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , ATP-Binding Cassette Transporters/genetics , Carcinogens/metabolism , Imidazoles/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Animals , Carcinogens/pharmacokinetics , Imidazoles/pharmacokinetics , Liver/metabolism , Male , Mice , Mice, Knockout , ATP-Binding Cassette Sub-Family B Member 4
10.
Appl Radiat Isot ; 70(1): 205-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21975107

ABSTRACT

In recent years, [(18)F]gefitinib PET has successfully been employed for a number of applications ranging from oncology to in vivo studies of drug transporter proteins. We here report a reliable, automated procedure for routine synthesis of this radiotracer on an Eckert and Ziegler modular system. The 3-step radiosynthesis followed by preparative HPLC-purification provided [(18)F]gefitinib in 17.2±3.3% (n=22) overall decay-corrected radiochemical yield with radiochemical purity >99% in a total synthesis time of about 2.5h.


Subject(s)
Chromatography, High Pressure Liquid/instrumentation , Fluorine Radioisotopes/chemistry , Isotope Labeling/instrumentation , Quinazolines/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Robotics/instrumentation , Equipment Design , Gefitinib
11.
Drug Metab Dispos ; 39(8): 1338-44, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21566011

ABSTRACT

The ATP-binding cassette (ABC) transporters ABCC2 [multidrug resistance-associated protein (MRP) 2], ABCC3 (MRP3), and ABCG2 (breast cancer resistance protein) are involved in the efflux of potentially toxic compounds from the body. We have shown before that ABCC2, ABCC3, and ABCG2 together influence the pharmacokinetics of the anticancer and antirheumatic drug methotrexate (MTX) and its toxic metabolite 7-hydroxymethotrexate (7OH-MTX) after intravenous MTX administration. We now have used Abcc2;Abcc3;Abcg2(-/-) and corresponding single and double knockout mice to investigate the relative impact of these transporters on MTX and 7OH-MTX pharmacokinetics after oral MTX administration (50 mg/kg). The plasma areas under the curve (AUC(plasma)) in Abcg2(-/-) and Abcc2;Abcg2(-/-) mice were 1.7- and 3.0-fold higher than those in wild-type mice, respectively, suggesting additive effects of Abcc2 and Abcg2 on oral MTX pharmacokinetics. However, the AUC(plasma) in Abcc2;Abcc3;Abcg2(-/-) mice was not different from that in wild-type mice, indicating that Abcc3 protein is necessary for increased MTX plasma concentrations in the absence of Abcc2 and/or Abcg2. Furthermore, 2 h after administration, MTX liver levels were increased in Abcg2-deficient strains and MTX kidney levels were 2.2-fold increased in Abcc2;Abcg2(-/-) mice compared with those in wild-type mice. The absence of Abcc2 and/or Abcg2 also led to significantly increased liver and kidney levels of 7OH-MTX. Our results suggest that inhibition of ABCG2 and/or ABCC2, genetic polymorphisms or mutations reducing expression or activity of these proteins may increase the oral availability of MTX. Such conditions may also present risk factors for increased MTX-related toxicity in patients treated with oral MTX.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Antimetabolites, Antineoplastic/pharmacokinetics , Methotrexate/analogs & derivatives , Multidrug Resistance-Associated Proteins/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Administration, Oral , Animals , Antimetabolites, Antineoplastic/blood , Antimetabolites, Antineoplastic/metabolism , Area Under Curve , Biological Availability , Female , Injections, Intravenous , Methotrexate/blood , Methotrexate/metabolism , Methotrexate/pharmacokinetics , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Organ Specificity , Tissue Distribution
12.
J Pharmacol Exp Ther ; 334(1): 78-86, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20388726

ABSTRACT

Cholyl-L-lysyl-fluorescein (CLF) is a fluorescent bile salt derivative that is being developed as an agent for determining in vivo liver function. However, the mechanisms of uptake and excretion by hepatocytes have not been rigorously studied. We have directly assessed the transport capacity of various hepatobiliary transporters for CLF. Uptake experiments were performed in Chinese hamster ovary cells transfected with human NTCP, OATP1B1, OATP1B3, and OATP2B1. Conversely, excretory systems were tested with plasma membrane vesicles from Sf21 insect cells expressing human ABCB11, ABCC2, ABCC3, and ABCG2. In addition, plasma clearance and biliary excretion of CLF were examined in wild-type, Abcc2(-/-), and Abcc3(-/-) mice. Human Na(+)-dependent taurocholic-cotransporting polypeptide (NTCP) and ATP-binding cassette B11 (ABCB11) were incapable of transporting CLF. In contrast, high-affinity transport of CLF was observed for organic anion-transporting polypeptide 1B3 (OATP1B3), ABCC2, and ABCC3 with K(m) values of 4.6 +/- 2.7, 3.3 +/- 2.0, and 3.7 +/- 1.0 microM, respectively. In Abcc2(-/-) mice biliary excretion of CLF was strongly reduced compared with wild-type mice. This resulted in a much higher hepatic retention of CLF in Abcc2(-/-) versus wild-type mice: 64 versus 1% of the administered dose (2 h after administration). In mice intestinal uptake of CLF was negligible compared with that of taurocholate. Our conclusion is that human NTCP and ABCB11 are incapable of transporting CLF, whereas OATP1B3 and ABCC2/Abcc2 most likely mediate hepatic uptake and biliary excretion of CLF, respectively. CLF can be transported back into the blood by ABCC3. Enterohepatic circulation of CLF is minimal. This renders CLF suitable as an agent for assessing in vivo liver function.


Subject(s)
Carrier Proteins/metabolism , Cholic Acids/pharmacokinetics , Fluoresceins/pharmacokinetics , Fluorescent Dyes/pharmacokinetics , Liver/metabolism , Animals , Bile/metabolism , Biological Transport , CHO Cells , Carrier Proteins/genetics , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetinae , Cricetulus , Humans , Insecta/cytology , Insecta/metabolism , Liver/drug effects , Male , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3 , Symporters/metabolism , Transfection
13.
Clin Cancer Res ; 16(1): 130-40, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20028753

ABSTRACT

PURPOSE: Despite the extensive use of etoposide for the treatment of different malignant neoplasms, its main pharmacokinetic determinants are not completely defined. We aimed to study the impact of P-glycoprotein (P-gp/ABCB1) and the multidrug resistance proteins ABCC2 (MRP2) and ABCC3 (MRP3) on the pharmacokinetics of etoposide. EXPERIMENTAL DESIGN: Abcb1a/1b(-/-), Abcc2(-/-), Abcc3(-/-), Abcb1a/1b;Abcc2(-/-), and Abcc2;Abcc3(-/-) mice were used to investigate the separate and combined impact of P-gp, Abcc2, and Abcc3 on the in vivo behavior of etoposide. RESULTS: P-gp restricted the oral (re)uptake of unchanged etoposide, and mediated its excretion across the gut wall. In contrast, hepatobiliary excretion was almost entirely dependent on Abcc2. Yet, complete loss of Abcc2 did not result in elevated liver or plasma concentrations of etoposide. Instead, Abcc2(-/-) mice displayed an increased hepatic formation of etoposide glucuronide, which was secreted via Abcc3 from the liver to the blood circulation and eliminated with the urine. Combination Abcc2;Abcc3(-/-) mice had highly increased accumulation of etoposide glucuronide in their livers, whereas both single knockouts did not, indicating that Abcc2 and Abcc3 provide alternative pathways for the hepatic elimination of etoposide glucuronide. CONCLUSIONS: P-gp, ABCC2, and ABCC3 significantly affect the pharmacokinetics of etoposide and/or etoposide glucuronide. Variation in transporter expression or activity may explain the high variation in oral availability of etoposide (25-80%) among cancer patients. However, despite the fact that substantial variations in transporter activity can occur, we believe that cancer patients are often relatively protected from etoposide toxicity due to overlapping functions of these transporters in the elimination of etoposide.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Etoposide/pharmacokinetics , Multidrug Resistance-Associated Proteins/metabolism , Animals , Etoposide/blood , Etoposide/urine , Feces/chemistry , Liver/metabolism , Male , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2
14.
Mol Cancer Ther ; 8(12): 3350-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19996279

ABSTRACT

The multidrug transporters ABCC2, ABCC3, and ABCG2 can eliminate potentially toxic compounds from the body and have overlapping substrate specificities. To investigate the overlapping functions of Abcc2, Abcc3, and Abcg2 in vivo, we generated and characterized Abcc3;Abcg2-/- and Abcc2;Abcc3;Abcg2-/- mice. We subsequently analyzed the relative impact of these transport proteins on the pharmacokinetics of the anticancer drug methotrexate (MTX) and its main, toxic, metabolite 7-hydroxymethotrexate (7OH-MTX) after i.v. administration of MTX (50 mg/kg). Whereas in single and double knockout mice, the plasma and liver concentrations of MTX and 7OH-MTX decreased rapidly after MTX administration, in the Abcc2;Abcc3;Abcg2-/- mice, they remained very high. One hour after administration, 67% of the MTX dose was still present in livers of Abcc2;Abcc3;Abcg2-/- mice as MTX or 7OH-MTX versus 7% in wild-type, showing dramatic liver accumulation of these toxic compounds when Abcc2, Abcc3, and Abcg2 were all absent. Furthermore, the urinary and fecal excretion of the nephrotoxic metabolite 7OH-MTX were increased 27- and 7-fold, respectively, in Abcc2;Abcc3;Abcg2-/- mice. Thus, Abcc2, Abcc3, and Abcg2 together mediate the rapid elimination of MTX and 7OH-MTX after i.v. administration and can to a large extent compensate for each other's absence. This may explain why it is still comparatively safe to use a toxic drug such as MTX in the clinic, as the risk of highly increased toxicity due to dysfunctioning of ABCC2, ABCC3, or ABCG2 alone is limited. Nevertheless, cotreatment with possible inhibitors of ABCC2, ABCC3, and ABCG2 should be done with utmost caution when treating patients with methotrexate.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Methotrexate/analogs & derivatives , Methotrexate/metabolism , Multidrug Resistance-Associated Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Alanine Transaminase/blood , Analysis of Variance , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/blood , Antimetabolites, Antineoplastic/pharmacokinetics , Area Under Curve , Blotting, Western , Chromatography, High Pressure Liquid , Creatinine/blood , Feces/chemistry , Female , Intestine, Small/metabolism , Kidney/metabolism , Liver/metabolism , Male , Metabolic Clearance Rate , Methotrexate/blood , Methotrexate/pharmacokinetics , Methotrexate/urine , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
15.
Mol Interv ; 9(3): 136-45, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19592674

ABSTRACT

A TP-binding cassette (ABC) multidrug transporters are cellular efflux pumps with broad and often widely overlapping substrate specificities. They can have a major impact on the pharmacokinetics and hence overall pharmacological behavior of many drugs. To study their separate roles and functional overlap, or complementarity, a collection of mice deficient in two or more ABC transporters has been generated. This review discusses recent findings obtained with these models, focusing on pharmacokinetic studies with a number of clinically relevant drugs. In addition, the characterization of these mice and some physiological aspects of ABC multidrug transporters are addressed.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Models, Animal , ATP-Binding Cassette Transporters/metabolism , Animals , Biological Transport , Humans , Mice , Mice, Knockout , Pharmaceutical Preparations/metabolism
16.
Drug Metab Dispos ; 37(8): 1698-702, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19443695

ABSTRACT

Ezetimibe lowers plasma cholesterol levels by inhibiting the uptake of cholesterol in the intestine. Because of the extensive enterohepatic circulation of ezetimibe, relatively low doses are required to be effective. In blood and bile the majority of ezetimibe is present as a glucuronide conjugate, which is formed in the enterocyte. Presently, it is not clear which mechanisms are responsible for this efficient enterohepatic circulation. Abcc2, Abcc3, and Abcg2 are ATP-binding cassette (ABC) transporters that are expressed in both liver and intestine and are capable of transporting glucuronidated compounds. The aim of this study was to investigate the contribution of these transporters in the enterohepatic cycling of ezetimibe glucuronide (Ez-gluc). Transport studies were performed in plasma membrane vesicles from ABCC2-, ABCC3-, and ABCG2-expressing Sf21 insect cells. Furthermore, intestinal explants from wild-type and Abcc3(-/-) mice were used to study vectorial transport in a Ussing chamber setup. Finally, biliary excretion of Ez-gluc was measured in vivo after duodenal delivery of ezetimibe in wild-type, Abcc3(-/-), Abcc2(-/-), Abcg2(-/-), and Abcg2(-/-)/Abcc2(-/-) mice. ABCC3-, ABCC2-, and ABCG2-mediated transport was dose dependently inhibited by Ez-gluc. In the Ussing chamber Ez-gluc recovered from the basolateral side was significantly reduced in duodenal (2.2%), in jejunal (23%), and in ileal (23%) tissue of Abcc3(-/-) mice compared with that in tissues of wild-type mice. Biliary excretion of Ez-gluc was significantly reduced in Abcc3(-/-) (34%), Abcc2(-/-) (56%), and Abcg2(-/-)/Abcc2(-/-) (2.5%) compared with that in wild-type mice. These data demonstrate that the enterohepatic circulation of Ez-gluc strongly depends on the joint function of Abcc3, Abcc2, and Abcg2.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Anticholesteremic Agents/pharmacokinetics , Azetidines/pharmacokinetics , Intestinal Absorption , Intestine, Small/metabolism , Multidrug Resistance-Associated Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Administration, Oral , Animals , Anticholesteremic Agents/administration & dosage , Azetidines/administration & dosage , Biological Transport , Biotransformation , Cell Line , Enterohepatic Circulation , Estradiol/analogs & derivatives , Ezetimibe , Glucuronides/pharmacokinetics , Intubation, Gastrointestinal , Methotrexate/metabolism , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/deficiency , Multidrug Resistance-Associated Proteins/genetics , Transfection
17.
Clin Cancer Res ; 15(9): 3084-93, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19383815

ABSTRACT

PURPOSE: ABCC2 (MRP2) and ABCG2 (BCRP) transport various endogenous and exogenous compounds, including many anticancer drugs, into bile, feces, and urine. We investigated the possibly overlapping roles of Abcg2 and Abcc2 in the elimination of the anticancer drug methotrexate (MTX) and its toxic metabolite 7-hydroxymethotrexate (7OH-MTX). EXPERIMENTAL DESIGN: We generated and characterized Abcc2;Abcg2(-/-) mice, and used these to determine the overlapping roles of Abcc2 and Abcg2 in the elimination of MTX and 7OH-MTX after i.v. administration of 50 mg/kg MTX. RESULTS: Compared with wild-type, the plasma areas under the curve (AUC) for MTX were 1.6-fold and 2.0-fold higher in Abcg2(-/-) and Abcc2(-/-) mice, respectively, and 3.3-fold increased in Abcc2;Abcg2(-/-) mice. The biliary excretion of MTX was 23-fold reduced in Abcc2;Abcg2(-/-) mice, and the MTX levels in the small intestine were dramatically decreased. Plasma levels of 7OH-MTX were not significantly altered in Abcg2(-/-) mice, but the areas under the curve were 6.2-fold and even 12.4-fold increased in Abcc2(-/-) and Abcc2;Abcg2(-/-) mice, respectively. This indicates that Abcc2 compensates for Abcg2 deficiency but that Abcg2 can only partly compensate for Abcc2 absence. Furthermore, 21-fold decreased biliary 7OH-MTX excretion in Abcc2;Abcg2(-/-) mice and substantial 7OH-MTX accumulation in the liver and kidney were seen. We additionally found that in the absence of Abcc2, Abcg2 mediated substantial urinary excretion of MTX and 7OH-MTX. CONCLUSIONS: Abcc2 and Abcg2 together are major determinants of MTX and 7OH-MTX pharmacokinetics. Variations in ABCC2 and/or ABCG2 activity due to polymorphisms or coadministered inhibitors may therefore substantially affect the therapeutic efficacy and toxicity in patients treated with MTX.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Biliary Tract/metabolism , Liver/metabolism , Methotrexate/analogs & derivatives , Methotrexate/pharmacokinetics , Multidrug Resistance-Associated Proteins/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Area Under Curve , Blotting, Western , Chromatography, High Pressure Liquid , Feces/chemistry , Female , Folic Acid Antagonists/pharmacokinetics , Folic Acid Antagonists/toxicity , Folic Acid Antagonists/urine , Male , Methotrexate/toxicity , Methotrexate/urine , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
18.
Article in English | MEDLINE | ID: mdl-19117812

ABSTRACT

The first bioanalytical assay for the simultaneous determination of sorafenib and sorafenib-glucuronide in mouse plasma and liver homogenate was developed and validated. In addition, the structure of the glucuronide metabolite was elucidated. The quantitative assay started with addition of isotopically labeled internal standards to a 20 microl sample volume and protein precipitation with acetonitrile, the supernatant was diluted with water and injected into the chromatographic system. A polar embedded reversed-phase column with gradient elution using formic acid in water-acetonitrile was used. The eluate was transferred into an electrospray interface with positive ionization and the analytes were detected and quantified using triple quadrupole mass spectrometry. The assay was validated in the ranges 10-5000 ng/ml for sorafenib and 1-500 ng/ml for sorafenib-glucuronide, the lowest levels of these ranges (10 and 1 ng/ml) being the lower limits of quantification (LLQ). Within day precisions were 2-8%, between day precisions 2-10% (both excluded the LLQ level of the glucuronide) and accuracies were between 89% and 106%. Both analytes were chemically stable under all relevant conditions. The assay was successfully applied in pilot in vivo pharmacokinetic studies with sorafenib in mice.


Subject(s)
Antineoplastic Agents/analysis , Benzenesulfonates/analysis , Chromatography, Liquid , Glucuronides/analysis , Liver/chemistry , Pyridines/analysis , Tandem Mass Spectrometry , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Benzenesulfonates/blood , Benzenesulfonates/chemistry , Benzenesulfonates/metabolism , Female , Glucuronides/blood , Glucuronides/chemistry , Glucuronides/metabolism , Liver/metabolism , Male , Mice , Niacinamide/analogs & derivatives , Phenylurea Compounds , Pilot Projects , Pyridines/blood , Pyridines/chemistry , Pyridines/metabolism , Reproducibility of Results , Sensitivity and Specificity , Sorafenib
19.
Adv Drug Deliv Rev ; 61(1): 14-25, 2009 Jan 31.
Article in English | MEDLINE | ID: mdl-19118589

ABSTRACT

The multidrug transporter ABCG2 (BCRP/MXR/ABCP) can actively extrude a broad range of endogenous and exogenous substrates across biological membranes. ABCG2 limits oral availability and mediates hepatobiliary and renal excretion of its substrates, and thus influences the pharmacokinetics of many drugs. Recent work, relying mainly on the use of Abcg2(-/-) mice, has revealed important contributions of ABCG2 to the blood-brain, blood-testis and blood-fetal barriers. Together, these functions indicate a primary biological role of ABCG2 in protecting the organism from a range of xenobiotics. In addition, several other physiological functions of ABCG2 have been observed, including extrusion of porphyrins and/or porphyrin conjugates from hematopoietic cells, liver and harderian gland, as well as secretion of vitamin B(2) (riboflavin) and possibly other vitamins (biotin, vitamin K) into breast milk. However, the physiological significance of these processes has been difficult to establish, indicating that there is still a lot to learn about this intriguing protein.


Subject(s)
ATP-Binding Cassette Transporters/pharmacology , ATP-Binding Cassette Transporters/physiology , Neoplasm Proteins/pharmacology , Neoplasm Proteins/physiology , Xenobiotics/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Biological Transport , Harderian Gland/metabolism , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Mice , Mice, Knockout , Milk/metabolism , Milk, Human/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phytoestrogens/pharmacokinetics , Phytoestrogens/pharmacology , Porphyrins/metabolism , Porphyrins/pharmacokinetics , Porphyrins/pharmacology , Vitamins/pharmacokinetics , Xenobiotics/pharmacology
20.
Clin Cancer Res ; 14(24): 8152-60, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19088030

ABSTRACT

PURPOSE: ATP-binding cassette sub-family C member 2 [ABCC2; multidrug resistance-associated protein 2 (MRP2)] and ABCC3 (MRP3) mediate the elimination of toxic compounds, such as drugs and carcinogens, and have a large overlap in substrate specificity. We investigated the roles of Abcc2 and Abcc3 in the elimination of the anticancer drug methotrexate (MTX) and its toxic metabolite 7-hydroxymethotrexate (7OH-MTX) in vivo. EXPERIMENTAL DESIGN: Abcc2;Abcc3(-/-) mice were generated, characterized, and used to investigate possibly overlapping or complementary roles of Abcc2 and Abcc3 in the elimination of MTX and 7OH-MTX after i.v. administration of 50 mg/kg MTX. RESULTS: Abcc2;Abcc3(-/-) mice were viable and fertile. In Abcc2(-/-) mice, the plasma area under the curve (AUCi.v.) for MTX was 2.0-fold increased compared with wild type, leading to 1.6-fold increased urinary excretion, which was not seen in Abcc2;Abcc3(-/-) mice. Biliary excretion of MTX was 3.7-fold reduced in Abcc2(-/-) but unchanged in Abcc2;Abcc3(-/-) mice. The plasma AUCi.v.s of 7OH-MTX were 6.0-fold and 4.3-fold increased in Abcc2(-/-) and Abcc2;Abcc3(-/-) mice, respectively, leading to increased urinary excretion. The biliary excretion of 7OH-MTX was 5.8-fold reduced in Abcc2(-/-) but unchanged in Abcc2;Abcc3(-/-) mice. 7OH-MTX accumulated substantially in the liver of Abcc2(-/-) and especially Abcc2;Abcc3(-/-) mice. CONCLUSIONS: Abcc2 is important for (biliary) excretion of MTX and its toxic metabolite 7OH-MTX. When Abcc2 is absent, Abcc3 transports MTX and 7OH-MTX back from the liver into the circulation, leading to increased plasma levels and urinary excretion. Variation in ABCC2 and/or ABCC3 activity may therefore have profound effects on the elimination and severity of toxicity of MTX and 7OH-MTX after MTX treatment of patients.


Subject(s)
Methotrexate/analogs & derivatives , Methotrexate/pharmacokinetics , Multidrug Resistance-Associated Proteins/physiology , Animals , Bile/metabolism , Female , Liver/metabolism , Male , Methotrexate/toxicity , Mice , Multidrug Resistance-Associated Protein 2
SELECTION OF CITATIONS
SEARCH DETAIL
...