51
|
Feng B, Varma MV, Costales C, Zhang H, Tremaine L. In vitroandin vivoapproaches to characterize transporter-mediated disposition in drug discovery. Expert Opin Drug Discov 2014; 9:873-90. [DOI: 10.1517/17460441.2014.922540] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
52
|
Understanding the pharmacological properties of a metabolic PET tracer in prostate cancer. Proc Natl Acad Sci U S A 2014; 111:7254-9. [PMID: 24785505 DOI: 10.1073/pnas.1405240111] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Generally, solid tumors (>400 mm(3)) are inherently acidic, with more aggressive growth producing greater acidity. If the acidity could be targeted as a biomarker, it would provide a means to gauge the pace of tumor growth and degree of invasiveness, as well as providing a basis for predicting responses to pH-dependent chemotherapies. We have developed a (64)Cu pH (low) insertion peptide (pHLIP) for targeting, imaging, and quantifying acidic tumors by PET, and our findings reveal utility in assessing prostate tumors. The new pHLIP version limits indiscriminate healthy tissue binding, and we demonstrate its targeting of extracellular acidification in three different prostate cancer models, each with different vascularization and acid-extruding protein carbonic anhydrase IX (CAIX) expression. We then describe the tumor distribution of this radiotracer ex vivo, in association with blood perfusion and known biomarkers of acidity, such as hypoxia, lactate dehydrogenase A, and CAIX. We find that the probe reveals metabolic variations between and within tumors, and discriminates between necrotic and living tumor areas.
Collapse
|
53
|
Soars MG, Barton P, Elkin LL, Mosure KW, Sproston JL, Riley RJ. Application of anin vitroOAT assay in drug design and optimization of renal clearance. Xenobiotica 2014; 44:657-65. [DOI: 10.3109/00498254.2013.879625] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
54
|
Kunze A, Huwyler J, Poller B, Gutmann H, Camenisch G. In vitro-in vivo extrapolation method to predict human renal clearance of drugs. J Pharm Sci 2014; 103:994-1001. [PMID: 24549735 DOI: 10.1002/jps.23851] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 12/19/2013] [Accepted: 12/20/2013] [Indexed: 11/07/2022]
Abstract
Renal clearance is a key determinant of the elimination of drugs. To date, only few in vitro-in vivo extrapolation (IVIVE) approaches have been described to predict the renal organ clearance as the net result of glomerular filtration, tubular secretion, and tubular reabsorption. In this study, we measured in LLC-PK1 cells the transport of 20 compounds that cover all four classes of the Biopharmaceutical Drug Disposition System. These data were incorporated into a novel kidney model to predict all renal clearance processes in human. We showed that filtration and secretion were main contributors to the renal organ clearance for all compounds, whereas reabsorption was predominant for compounds assigned to classes 1 and 2. Our results suggest that anionic drugs were not significantly secreted in LLC-PK1 cells, resulting in under-predicted clearances. When all study compounds were included a high overall correlation between the reported and predicted renal organ clearances was obtained (R² = 0.83). The prediction accuracy in terms of percentage within twofold and threefold error was 70% and 95%, respectively. In conclusion, our novel IVIVE method allowed to predict the human renal organ clearance and the contribution of each underlying process.
Collapse
Affiliation(s)
- Annett Kunze
- Division of Drug Metabolism and Pharmacokinetics, Drug-Drug Interactions Section, Novartis Institutes for BioMedical Research, Basel, CH-4056, Switzerland; Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Basel, CH-4056, Switzerland
| | | | | | | | | |
Collapse
|
55
|
Fan J, de Lannoy IA. Pharmacokinetics. Biochem Pharmacol 2014; 87:93-120. [DOI: 10.1016/j.bcp.2013.09.007] [Citation(s) in RCA: 169] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 09/06/2013] [Accepted: 09/09/2013] [Indexed: 11/29/2022]
|
56
|
Heger M, van Golen RF, Broekgaarden M, Michel MC. The molecular basis for the pharmacokinetics and pharmacodynamics of curcumin and its metabolites in relation to cancer. Pharmacol Rev 2013; 66:222-307. [PMID: 24368738 DOI: 10.1124/pr.110.004044] [Citation(s) in RCA: 363] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review addresses the oncopharmacological properties of curcumin at the molecular level. First, the interactions between curcumin and its molecular targets are addressed on the basis of curcumin's distinct chemical properties, which include H-bond donating and accepting capacity of the β-dicarbonyl moiety and the phenylic hydroxyl groups, H-bond accepting capacity of the methoxy ethers, multivalent metal and nonmetal cation binding properties, high partition coefficient, rotamerization around multiple C-C bonds, and the ability to act as a Michael acceptor. Next, the in vitro chemical stability of curcumin is elaborated in the context of its susceptibility to photochemical and chemical modification and degradation (e.g., alkaline hydrolysis). Specific modification and degradatory pathways are provided, which mainly entail radical-based intermediates, and the in vitro catabolites are identified. The implications of curcumin's (photo)chemical instability are addressed in light of pharmaceutical curcumin preparations, the use of curcumin analogues, and implementation of nanoparticulate drug delivery systems. Furthermore, the pharmacokinetics of curcumin and its most important degradation products are detailed in light of curcumin's poor bioavailability. Particular emphasis is placed on xenobiotic phase I and II metabolism as well as excretion of curcumin in the intestines (first pass), the liver (second pass), and other organs in addition to the pharmacokinetics of curcumin metabolites and their systemic clearance. Lastly, a summary is provided of the clinical pharmacodynamics of curcumin followed by a detailed account of curcumin's direct molecular targets, whereby the phenotypical/biological changes induced in cancer cells upon completion of the curcumin-triggered signaling cascade(s) are addressed in the framework of the hallmarks of cancer. The direct molecular targets include the ErbB family of receptors, protein kinase C, enzymes involved in prostaglandin synthesis, vitamin D receptor, and DNA.
Collapse
Affiliation(s)
- Michal Heger
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
57
|
Feng B, Hurst S, Lu Y, Varma MV, Rotter CJ, El-Kattan A, Lockwood P, Corrigan B. Quantitative Prediction of Renal Transporter-Mediated Clinical Drug–Drug Interactions. Mol Pharm 2013; 10:4207-15. [DOI: 10.1021/mp400295c] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Bo Feng
- Department of Pharmacokinetics and Drug Metabolism, Pfizer Global Research & Development, Groton, Connecticut 06340, United States
| | - Susan Hurst
- Department of Pharmacokinetics and Drug Metabolism, Pfizer Global Research & Development, Groton, Connecticut 06340, United States
| | - Yasong Lu
- CV/Met Pharmacometrics, Department of Exploratory Clinical & Translational Research, Bristol-Myers Squibb, Lawrenceville, New Jersey 08540, United States
| | - Manthena V. Varma
- Department of Pharmacokinetics and Drug Metabolism, Pfizer Global Research & Development, Groton, Connecticut 06340, United States
| | - Charles J. Rotter
- Department of Pharmacokinetics and Drug Metabolism, Pfizer Global Research & Development, Groton, Connecticut 06340, United States
| | - Ayman El-Kattan
- Department of Pharmacokinetics and Drug Metabolism, Pfizer Global Research & Development, Groton, Connecticut 06340, United States
| | - Peter Lockwood
- Department of Clinical Pharmacology, Pfizer Global Research & Development, Groton, Connecticut 06340, United States
| | - Brian Corrigan
- Department of Clinical Pharmacology, Pfizer Global Research & Development, Groton, Connecticut 06340, United States
| |
Collapse
|
58
|
Krajcsi P. Drug-transporter interaction testing in drug discovery and development. World J Pharmacol 2013; 2:35-46. [DOI: 10.5497/wjp.v2.i1.35] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 10/25/2012] [Accepted: 01/30/2013] [Indexed: 02/06/2023] Open
Abstract
The human body consists of several physiological barriers that express a number of membrane transporters. For an orally absorbed drug the intestinal, hepatic, renal and blood-brain barriers are of the greatest importance. The ATP-binding cassette (ABC) transporters that mediate cellular efflux and the solute carrier transporters that mostly mediate cellular uptake are the two superfamilies responsible for membrane transport of vast majority of drugs and drug metabolites. The total number of human transporters in the two superfamilies exceeds 400, and about 40-50 transporters have been characterized for drug transport. The latest Food and Drug Administration guidance focuses on P-glycoprotein, breast cancer resistance protein, organic anion transporting polypeptide 1B1 (OATP1B1), OATP1B3, organic cation transporter 2 (OCT2), and organic anion transporters 1 (OAT1) and OAT3. The European Medicines Agency’s shortlist additionally contains the bile salt export pump, OCT1, and the multidrug and toxin extrusion transporters, multidrug and toxin extrusion protein 1 (MATE1) and MATE2/MATE2K. A variety of transporter assays are available to test drug-transporter interactions, transporter-mediated drug-drug interactions, and transporter-mediated toxicity. The drug binding site of ABC transporters is accessible from the cytoplasm or the inner leaflet of the plasma membrane. Therefore, vesicular transport assays utilizing inside-out vesicles are commonly used assays, where the directionality of transport results in drugs being transported into the vesicle. Monolayer assays utilizing polarized cells expressing efflux transporters are the test systems suggested by regulatory agencies. However, in some monolayers, uptake transporters must be coexpressed with efflux transporters to assure detectable transport of low passive permeability drugs. For uptake transporters mediating cellular drug uptake, utilization of stable transfectants have been suggested. In vivo animal models complete the testing battery. Some issues, such as in vivo relevance, gender difference, age and ontogeny issues can only be addressed using in vivo models. Transporter specificity is provided by using knock-out or mutant models. Alternatively, chemical knock-outs can be employed. Compensatory changes are less likely when using chemical knock-outs. On the other hand, specific inhibitors for some uptake transporters are not available, limiting the options to genetic knock-outs.
Collapse
|
59
|
Comparison of brain capillary endothelial cell-based and epithelial (MDCK-MDR1, Caco-2, and VB-Caco-2) cell-based surrogate blood–brain barrier penetration models. Eur J Pharm Biopharm 2012; 82:340-51. [DOI: 10.1016/j.ejpb.2012.07.020] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 05/14/2012] [Accepted: 07/31/2012] [Indexed: 12/18/2022]
|
60
|
Abstract
Sodium glucose cotransporter 2 (SGLT2) inhibition is a novel and promising treatment for diabetes under late-stage clinical development. It generally is accepted that SGLT2 mediates 90% of renal glucose reabsorption. However, SGLT2 inhibitors in clinical development inhibit only 30-50% of the filtered glucose load. Why are they unable to inhibit 90% of glucose reabsorption in humans? We will try to provide an explanation to this puzzle in this perspective analysis of the unique pharmacokinetic and pharmacodynamic profiles of SGLT2 inhibitors in clinical trials and examine possible mechanisms and molecular properties that may be responsible.
Collapse
Affiliation(s)
- Jiwen (Jim) Liu
- Medicinal Chemistry, Amgen, Inc., South San Francisco, California
- Corresponding authors: Jiwen (Jim) Liu, , and Ralph A. DeFronzo,
| | - TaeWeon Lee
- Metabolic Disorders, Amgen, Inc., South San Francisco, California
| | - Ralph A. DeFronzo
- Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Corresponding authors: Jiwen (Jim) Liu, , and Ralph A. DeFronzo,
| |
Collapse
|
61
|
Feng B, El‐Kattan AF, Radi ZA. Renal Transporters in Drug Disposition, Drug‐Drug Interactions, and Nephrotoxicity. ACTA ACUST UNITED AC 2012; Chapter 23:Unit 23.3.1-15. [DOI: 10.1002/0471140856.tx2303s53] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Bo Feng
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development Groton Connecticut
| | - Ayman F. El‐Kattan
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development Groton Connecticut
| | - Zaher A. Radi
- Drug Safety Research and Development, Pfizer Worldwide Research and Development Cambridge Massachusetts
| |
Collapse
|
62
|
Ballard P, Brassil P, Bui KH, Dolgos H, Petersson C, Tunek A, Webborn PJH. The right compound in the right assay at the right time: an integrated discovery DMPK strategy. Drug Metab Rev 2012; 44:224-52. [DOI: 10.3109/03602532.2012.691099] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
63
|
Varma MV, Gardner I, Steyn SJ, Nkansah P, Rotter CJ, Whitney-Pickett C, Zhang H, Di L, Cram M, Fenner KS, El-Kattan AF. pH-Dependent solubility and permeability criteria for provisional biopharmaceutics classification (BCS and BDDCS) in early drug discovery. Mol Pharm 2012; 9:1199-212. [PMID: 22489626 DOI: 10.1021/mp2004912] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The Biopharmaceutics Classification System (BCS) is a scientific framework that provides a basis for predicting the oral absorption of drugs. These concepts have been extended in the Biopharmaceutics Drug Disposition Classification System (BDDCS) to explain the potential mechanism of drug clearance and understand the effects of uptake and efflux transporters on absorption, distribution, metabolism, and elimination. The objective of present work is to establish criteria for provisional biopharmaceutics classification using pH-dependent passive permeability and aqueous solubility data generated from high throughput screening methodologies in drug discovery settings. The apparent permeability across monolayers of clonal cell line of Madin-Darby canine kidney cells, selected for low endogenous efflux transporter expression, was measured for a set of 105 drugs, with known BCS and BDDCS class. The permeability at apical pH 6.5 for acidic drugs and at pH 7.4 for nonacidic drugs showed a good correlation with the fraction absorbed in human (Fa). Receiver operating characteristic (ROC) curve analysis was utilized to define the permeability class boundary. At permeability ≥ 5 × 10(-6) cm/s, the accuracy of predicting Fa of ≥ 0.90 was 87%. Also, this cutoff showed more than 80% sensitivity and specificity in predicting the literature permeability classes (BCS), and the metabolism classes (BDDCS). The equilibrium solubility of a subset of 49 drugs was measured in pH 1.2 medium, pH 6.5 phosphate buffer, and in FaSSIF medium (pH 6.5). Although dose was not considered, good concordance of the measured solubility with BCS and BDDCS solubility class was achieved, when solubility at pH 1.2 was used for acidic compounds and FaSSIF solubility was used for basic, neutral, and zwitterionic compounds. Using a cutoff of 200 μg/mL, the data set suggested a 93% sensitivity and 86% specificity in predicting both the BCS and BDDCS solubility classes. In conclusion, this study identified pH-dependent permeability and solubility criteria that can be used to assign provisional biopharmaceutics class at early stage of the drug discovery process. Additionally, such a classification system will enable discovery scientists to assess the potential limiting factors to oral absorption, as well as help predict the drug disposition mechanisms and potential drug-drug interactions.
Collapse
Affiliation(s)
- Manthena V Varma
- Pfizer Global Research and Development, Pfizer Inc, Groton, Connecticut 06340, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Park EJ, Pai MP, Dong T, Zhang J, Ko CW, Lawrence J, Crentsil V, Zhang L, Xu NN. The Influence of Body Size Descriptors on the Estimation of Kidney Function in Normal Weight, Overweight, Obese, and Morbidly Obese Adults. Ann Pharmacother 2012; 46:317-28. [DOI: 10.1345/aph.1q374] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background: Dosing adjustments for patients with impaired kidney function are often based on estimated creatinine clearance (eCrCI) because measuring kidney (unction is not always possible for dose adjustment. However, there is no consensus on the body size descriptor that should be used in the estimation equations. Objective: To compare the use of alternative body size descriptors (ABSDs) on the performance of kidney function estimation equations compared with measured CrCI (mCrCI). Methods: We combined 2 data sources with mCrCI: a Food and Drug Administration clinical trial database that includes subjects with body mass index (BMI) less than 40 kg/m2 and published data from those 40 kg/m2 or more. The 3 parent equations (Cockcroft-Gault [CG], Modification of Diet in Renal Disease [MDRD], Chronic Kidney Disease-Epidemiology Collaboration [CKDEPI]), and 14 ABSD-modilied equations were compared with mCrCI for accuracy, bias, agreement, goodness of (it (R2). and prediction error. These equations were also compared across mCrCI and BMI strata. Results: Subjects (n = 590) were aged 19–80 years; 33.9% were female and BMI ranged from 17.2 to 95.6 kg/m2. Compared with mCrCI, the use of total weight in the CG equation yielded low accuracy (12.5%) and significant bias (–107 mL/min) in the morbidly obese group. In contrast, the use of lean body weights (BMI ≥40 kg/m2) and total ± adjusted weights (BMI <40 kg/m2) with the CG equation yielded higher accuracy, greater than or equal to 60.7% across all BMI strata, and was unbiased. Transforming the MDRD or CKDEPI equations with body surface area improved accuracy only at mCrCI of 30–80 mL/min and increased the overall prediction error. Conclusions: No kidney function equation was consistently accurate and unbiased across weight, mCrCI. and estimate ranges. The accuracy and over-estimation bias of the CG equation in obese subjects was improved through the selective use of total, adjusted, and lean body weight by BMI strata.
Collapse
Affiliation(s)
- Eun Jung Park
- Division of Cardiovascular and Renal Products, Office of New Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD
| | - Manjunath P Pai
- Department of Pharmacy Practice, Albany College of Pharmacy and Health Sciences, Albany, NY
| | - Ting Dong
- Division of Cardiovascular and Renal Products, Office of New Drugs, Center for Drug Evaluation and Research, Food and Drug Administration
| | - Jialu Zhang
- Office of Biostatistics, Center for Drug Evaluation and Research, Food and Drug Administration
| | - Chia-Wen Ko
- Office of Biostatistics, Center for Drug Evaluation and Research, Food and Drug Administration
| | - John Lawrence
- Office of Biostatistics, Center for Drug Evaluation and Research, Food and Drug Administration
| | - Victor Crentsil
- Division of Psychiatry Products, Office of New Drugs, Center for Drug Evaluation and Research, Food and Drug Administration
| | - Lei Zhang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration
| | - Nancy N Xu
- Division of Cardiovascular and Renal Products, Office of New Drugs, Center for Drug Evaluation and Research, Food and Drug Administration
| |
Collapse
|
65
|
Grabowski T, Jaroszewski JJ, Gad SC, Feder M. Correlation between in silico physicochemical characteristics of drugs and their mean residence time in human and dog. Int J Toxicol 2012; 31:25-33. [PMID: 22215410 DOI: 10.1177/1091581811429865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The correlation between 52 physicochemical parameters and mean residence time (MRT) for 27 drugs used in human and dog were investigated. The physicochemical parameter values calculated provided a basis for deriving a series of arithmetic expressions, which were used to build a mathematical model describing the relationship between them and the MRT values. From the entire set of analyzed parameters, a subset of 14 was identified that contributed to the derivation of an arithmetic expression: Log(PSA - WPSA + ACID) x [XlogP - (LogKp - EAxLn(Caco2 + AMINE + SAF))] + (AMIDE + IP - FG) - Ln(MW + PISA) the value of which is highly correlated with the MRT value in dogs (P < .001) and allowed prediction of the MRT predicted (MRT(pred)). In humans, no correlation was found that allowed the calculation of MRT(pred). These results indicate that predicting the pharmacokinetics of any specific drug for humans based on pharmacokinetic data obtained in the dog should be undertaken with knowledge of the inherent limitations.
Collapse
Affiliation(s)
- Tomasz Grabowski
- Centre of Pharmacokinetics Research FILAB, Ravimed, Polna 54, 05-119 Łajski, Poland.
| | | | | | | |
Collapse
|
66
|
Naud J, Michaud J, Beauchemin S, Hébert MJ, Roger M, Lefrancois S, Leblond FA, Pichette V. Effects of chronic renal failure on kidney drug transporters and cytochrome P450 in rats. Drug Metab Dispos 2011; 39:1363-9. [PMID: 21525170 DOI: 10.1124/dmd.111.039115] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Chronic renal failure (CRF) leads to decreased drug renal clearance due to a reduction in the glomerular filtration rate. However, little is known about how renal failure affects renal metabolism and elimination of drugs. Because both depend on the activity of uptake and efflux by renal transporters as well as enzymes in tubular cells, the purpose of this study was to investigate the effects of CRF on the expression and activity of select renal drug transporters and cytochrome P450. Two groups of rats were studied: control and CRF (induced by 5/6 nephrectomy). Compared with control rats, we observed reductions in the expression of both protein and mRNA of Cyp1a, sodium-dependent phosphate transport protein 1, organic anion transporter (Oat)1, 2, and 3, OatK1/K2, organic anion-transporting polypeptide (Oatp)1 and 4c1, P-glycoprotein, and urate transporter 1, whereas an induction in the protein and mRNA expression of Mrp2, 3, and 4 and Oatp2 and 3 was observed. Cyp3a expression remained unchanged. Similar results were obtained by incubating a human proximal tubule cell line (human kidney-2) with sera from CRF rats, suggesting the presence of uremic modulators. Finally, the renal elimination of [(3)H]digoxin and [(14)C]benzylpenicillin was decreased in CRF rats, compared with controls, as shown by a 4- and 9-fold accumulation, respectively, of these drugs in kidneys of rats in CRF. Our results demonstrate that CRF affects the expression and activity of several kidney drug transporters leading to the intrarenal accumulation of drugs and reduced renal clearance that could, at least partially, explain the tubular toxicity of many drugs.
Collapse
MESH Headings
- Animals
- Biological Transport
- Blotting, Western
- Cell Line
- Culture Media
- Cytochrome P-450 Enzyme System/metabolism
- Disease Models, Animal
- Gene Expression
- Humans
- Kidney/enzymology
- Kidney/metabolism
- Kidney Failure, Chronic/enzymology
- Kidney Failure, Chronic/metabolism
- Kidney Failure, Chronic/physiopathology
- Kidney Function Tests
- Kidney Tubules, Proximal/cytology
- Kidney Tubules, Proximal/metabolism
- Male
- Membrane Transport Proteins/genetics
- Membrane Transport Proteins/metabolism
- Pharmaceutical Preparations/blood
- Pharmaceutical Preparations/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Rhodamine 123/blood
- Rhodamine 123/pharmacokinetics
- Uremia/blood
- Uremia/metabolism
Collapse
Affiliation(s)
- Judith Naud
- Service de Néphrologie et Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Kido Y, Matsson P, Giacomini KM. Profiling of a prescription drug library for potential renal drug-drug interactions mediated by the organic cation transporter 2. J Med Chem 2011; 54:4548-58. [PMID: 21599003 DOI: 10.1021/jm2001629] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Drug-drug interactions (DDIs) are major causes of serious adverse drug reactions. Most DDIs have a pharmacokinetic basis in which one drug reduces the elimination of a second drug, leading to potentially toxic drug levels. As a major organ of drug elimination, the kidney represents an important site for DDIs. Here, we screened a prescription drug library against the renal organic cation transporter OCT2/SLC22A2, which mediates the first step in the renal secretion of many cationic drugs. Of the 910 compounds screened, 244 inhibited OCT2. Computational analyses revealed key properties of inhibitors versus noninhibitors, which included overall molecular charge. Four of six potential clinical inhibitors were transporter-selective in follow-up screens against additional transporters: OCT1/SLC22A1, MATE1/SLC47A1, and MATE2-K/SLC47A2. Two compounds showed different kinetics of interaction with the common polymorphism OCT2-A270S, suggesting a role of genetics in modulating renal DDIs.
Collapse
Affiliation(s)
- Yasuto Kido
- Department of Bioengineering and Therapeutic Sciences, University of California-San Francisco, San Francisco, California 94143, United States
| | | | | |
Collapse
|
68
|
Kell DB, Dobson PD, Oliver SG. Pharmaceutical drug transport: the issues and the implications that it is essentially carrier-mediated only. Drug Discov Today 2011; 16:704-14. [PMID: 21624498 DOI: 10.1016/j.drudis.2011.05.010] [Citation(s) in RCA: 148] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 04/04/2011] [Accepted: 05/11/2011] [Indexed: 01/04/2023]
Abstract
All cells necessarily contain tens, if not hundreds, of carriers for nutrients and intermediary metabolites, and the human genome codes for more than 1000 carriers of various kinds. Here, we illustrate using a typical literature example the widespread but erroneous nature of the assumption that the 'background' or 'passive' permeability to drugs occurs in the absence of carriers. Comparison of the rate of drug transport in natural versus artificial membranes shows discrepancies in absolute magnitudes of 100-fold or more, with the carrier-containing cells showing the greater permeability. Expression profiling data show exactly which carriers are expressed in which tissues. The recognition that drugs necessarily require carriers for uptake into cells provides many opportunities for improving the effectiveness of the drug discovery process.
Collapse
Affiliation(s)
- Douglas B Kell
- School of Chemistry, The University of Manchester, 131 Princess St, Manchester, M1 7DN, UK.
| | | | | |
Collapse
|
69
|
Morrish GA, Pai MP, Green B. The effects of obesity on drug pharmacokinetics in humans. Expert Opin Drug Metab Toxicol 2011; 7:697-706. [DOI: 10.1517/17425255.2011.570331] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|