Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 243
Filter
Add more filters

Publication year range
1.
Therapie ; 70(4): 329-35, 2015.
Article in English | MEDLINE | ID: mdl-25679187

ABSTRACT

A two way, randomized cross-over bioequivalence study was conducted to analyse the rate and extent of absorption of atorvastatin after a single dose of 80 mg atorvastatin as atorvastatin calcium tablets. The study was carried out using healthy male volunteers (N = 24). A high performance liquid chromatography method was employed to determine the level of drug in human plasma. It was concluded that the test and the reference drug exhibited comparable values of pharmacokinetic parameters. It was also concluded that since there was no significant difference between the rate and extent of absorption of the drug from the test and the reference formulations: these two formulations could thus be declared bioequivalent.


Subject(s)
Heptanoic Acids/pharmacokinetics , Pyrroles/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Atorvastatin , Cross-Over Studies , Healthy Volunteers , Heptanoic Acids/administration & dosage , Humans , Male , Pakistan , Pyrroles/administration & dosage , Tablets , Therapeutic Equivalency , Young Adult
2.
Drug Metab Dispos ; 42(1): 182-92, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24194513

ABSTRACT

Although organic anion transporting polypeptide (OATP)-mediated hepatic uptake is generally conserved between rodents and humans at a gross pharmacokinetic level, the presence of three major hepatic OATPs with broad overlap in substrate and inhibitor affinity, and absence of rodent-human orthologs preclude clinical translation of single-gene knockout/knockin findings. At present, changes in pharmacokinetics and tissue distribution of pravastatin, atorvastatin, simvastatin, and carboxydichlorofluorescein were studied in oatp1a/1b-knockout mice lacking the three major hepatic oatp isoforms, and in knockout mice with liver-specific knockin of human OATP1B1 or OATP1B3. Relative to wild-type controls, oatp1a/1b-knockout mice exhibited 1.6- to 19-fold increased intravenous and 2.1- to 115-fold increased oral drug exposure, due to 33%-75% decreased clearance, 14%-60% decreased volume of distribution, and ≤74-fold increased oral bioavailability, with the magnitude of change depending on the contribution of oatp1a/1b to pharmacokinetics. Hepatic drug distribution was 4.2- to 196-fold lower in oatp1a/1b-knockout mice; distributional attenuation was less notable in kidney, brain, cardiac, and skeletal muscle. Knockin of OATP1B1 or OATP1B3 partially restored control clearance, volume, and bioavailability values (24%-142% increase, ≤47% increase, and ≤77% decrease vs. knockout, respectively), such that knockin pharmacokinetic profiles were positioned between knockout and wild-type mice. Consistent with liver-specific humanization, only hepatic drug distribution was partially restored (1.3- to 6.5-fold increase vs. knockout). Exposure and liver distribution changes in OATP1B1-humanized versus knockout mice predicted the clinical impact of OATP1B1 on oral exposure and contribution to human hepatic uptake of statins within 1.7-fold, but only after correcting for human/humanized mouse liver relative protein expression factor (OATP1B1 = 2.2, OATP1B3 = 0.30).


Subject(s)
Heptanoic Acids/pharmacokinetics , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/metabolism , Pravastatin/pharmacokinetics , Pyrroles/pharmacokinetics , Simvastatin/pharmacokinetics , Adolescent , Adult , Aged , Animals , Atorvastatin , Biological Availability , Humans , Liver/metabolism , Liver-Specific Organic Anion Transporter 1 , Mice , Mice, Knockout , Middle Aged , Solute Carrier Organic Anion Transporter Family Member 1B3 , Tissue Distribution/physiology , Young Adult
3.
Eur J Clin Pharmacol ; 70(6): 675-84, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24728182

ABSTRACT

PURPOSE: ACT-178882, a direct renin inhibitor, was used as a model compound in an elaborate drug-drug interaction study with atorvastatin and simvastatin to explore complex CYP3A4 inductive and inhibitory properties. METHODS: Thirty-two healthy male subjects received single doses of 20 mg atorvastatin and 20 mg simvastatin on days 1, 9, 31, and 41. On days 6 to 33, 500 mg ACT-178882 was administered once daily. Plasma concentrations of ACT-178882, simvastatin, and atorvastatin were measured by LC-MS/MS. Routine safety assessments were performed throughout the study. RESULTS: Exposure (as based on area under the curve) to simvastatin and 6ß-hydroxyacid simvastatin increased (90 % confidence interval) 4.63-fold (3.90, 5.50) and 3.71-fold (3.19, 4.32), respectively, when comparing day 9 and day 1. On day 9, exposure to atorvastatin was similar but Cmax decreased, while both variables decreased for ortho-hydroxy atorvastatin when compared to day 1. On day 31, after prolonged administration of ACT-178882, exposure to atorvastatin, ortho-hydroxy atorvastatin, simvastatin, and 6ß-hydroxyacid simvastatin decreased by 14, 19, 21, and 27 %, respectively, when compared to day 9. However, on this day, exposure to simvastatin and its metabolite was still markedly higher when compared to day 1. Effects of ACT-178882 had largely dissipated on day 41. CONCLUSIONS: This design enabled the study of complex time-dependent effects on CYP3A4 activity with clinically relevant substrates.


Subject(s)
Cyclopropanes/pharmacology , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Pyridines/pharmacology , Adolescent , Adult , Atorvastatin , Cyclopropanes/administration & dosage , Cyclopropanes/adverse effects , Cyclopropanes/blood , Cytochrome P-450 CYP3A/biosynthesis , Cytochrome P-450 CYP3A Inducers/administration & dosage , Cytochrome P-450 CYP3A Inducers/adverse effects , Cytochrome P-450 CYP3A Inducers/blood , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/adverse effects , Cytochrome P-450 CYP3A Inhibitors/blood , Drug Interactions , Heptanoic Acids/administration & dosage , Heptanoic Acids/blood , Heptanoic Acids/pharmacokinetics , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Male , Middle Aged , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/blood , Pyrroles/administration & dosage , Pyrroles/blood , Pyrroles/pharmacokinetics , Simvastatin/administration & dosage , Simvastatin/blood , Simvastatin/pharmacokinetics , Substrate Specificity , Young Adult
4.
J Thromb Thrombolysis ; 37(4): 427-34, 2014 May.
Article in English | MEDLINE | ID: mdl-23852152

ABSTRACT

Diabetes mellitus (DM) is associated with impaired platelet response to clopidogrel. In patients with high on-treatment platelet reactivity (HTPR) while on standard-dose clopidogrel, high-dose atorvastatin enhances the pharmacodynamic (PD) effects of double-dose clopidogrel. It is unknown if similar effects are achieved in patients with DM. This study compare the PD effects of high-dose atorvastatin associated with double dose clopidogrel in HTPR patients with and without DM undergoing elective percutaneous coronary intervention (PCI). This is a post hoc analysis of a prospective randomized PD study that compared double-dose (150 mg) clopidogrel associated with high-dose (80 mg) atorvastatin to double-dose clopidogrel alone in statin naïve patients with HTPR undergoing elective PCI. In this analysis, patients were divided in two groups according to DM (n = 27) and non-DM (n = 49) status. Platelet reactivity was evaluated immediately before PCI and at 30 days using the VerifyNow P2Y12 assay. HTPR was defined as P2Y12 reaction units (PRU) ≥235. Administering high-dose atorvastatin in addition to high-dose clipodogrel, the 30 days absolute PRU changes (106 ± 75 vs 100 ± 42, p = 0.7) and optimal response rates (83 vs 84%; p = 0.9) were similar in DM and non-DM patients. The baseline variables significantly associated with 30-day optimal response to high-dose clopidogrel were: atorvastatin treatment (OR = 7.5 [95% CI 1.19-47]; p = 0.032) in DM patients; PRU values (OR = 0.9 [95% CI 0.95-0.99]; p = 0.031) and creatinine clearance (OR = 1.07 [95% CI 1.008-1.13]; p = 0.025) in non-DM patients. High-dose atorvastatin significantly improved the PD effects of double-dose clopidogrel in DM patients with HTPR undergoing elective PCI.


Subject(s)
Diabetes Mellitus/blood , Heptanoic Acids , Percutaneous Coronary Intervention , Platelet Activation/drug effects , Platelet Aggregation Inhibitors , Pyrroles , Ticlopidine/analogs & derivatives , Aged , Atorvastatin , Clopidogrel , Female , Heptanoic Acids/administration & dosage , Heptanoic Acids/pharmacokinetics , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Male , Middle Aged , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacology , Prospective Studies , Pyrroles/administration & dosage , Pyrroles/pharmacokinetics , Thrombosis/blood , Thrombosis/etiology , Thrombosis/prevention & control , Ticlopidine/administration & dosage , Ticlopidine/pharmacokinetics
5.
Chem Pharm Bull (Tokyo) ; 62(6): 545-51, 2014.
Article in English | MEDLINE | ID: mdl-24881660

ABSTRACT

The aim of the present study was to investigate the effect of Soluplus® on the solubility of atorvastatin calcium and to develop a solid dispersion formulation that can improve the oral bioavailability of atorvastatin calcium. We demonstrated that Soluplus® increases the aqueous solubility of atorvastatin calcium. Several solid dispersion formulations of atorvastatin calcium with Soluplus® were prepared at various drug : carrier ratios by spray drying. Physicochemical analysis demonstrated that atorvastatin calcium is amorphous in each solid dispersion, and the 2 : 8 drug : carrier ratio provided the highest degree of sustained atorvastatin supersaturation. Pharmacokinetic analysis in rats revealed that the 2 : 8 dispersion significantly improved the oral bioavailability of atorvastatin. This study demonstrates that spray-dried Soluplus® solid dispersions can be an effective method for achieving higher atorvastatin plasma levels.


Subject(s)
Desiccation , Heptanoic Acids/chemistry , Heptanoic Acids/pharmacokinetics , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Polyvinyls/chemistry , Polyvinyls/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacokinetics , Administration, Oral , Animals , Atorvastatin , Biological Availability , Heptanoic Acids/administration & dosage , Heptanoic Acids/blood , Male , Pyrroles/administration & dosage , Pyrroles/blood , Rats , Rats, Sprague-Dawley , Solubility/drug effects , Water/chemistry
6.
Eur J Drug Metab Pharmacokinet ; 39(4): 311-20, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24346850

ABSTRACT

The effect of silymarin (SMN) on the pharmacokinetics of atorvastatin in diabetic rats was evaluated. Male Wistar rats were assigned into two major groups and then sub-grouped according to the purposes of the study. The first major group was subdivided into three groups (n = 6) including control, non-treated diabetic and SMN-treated diabetic animals. In the first major group, metabolism of testosterone by the hepatic microsomes was studied. The second major group also was divided to three groups including atorvastatin-treated non-diabetic, atorvastatin-treated diabetic and diabetic animals which received both atorvastatin and SMN. To study the pharmacokinetics of atorvastatin, serum samples were collected at 0, 3, 6, 12 and 24 h after the atorvastatin administration. Pharmacokinetic parameters were calculated using non-compartmental model. Streptozotocin-induced diabetes resulted in a remarkable induction of testosterone hydroxylation as the V max for 6ß-hydroxytestosterone production in the diabetic rats (77.3 ± 8.6 pM/min/mg) was significantly higher than that in the control animals (45.9 ± 5.9 pM/min/mg). Moreover, SMN-treated animals showed a significant (P < 0.05) reduction of V max (59.4 ± 6.1 pM/min/mg). Diabetes resulted in a significant reduction of AUC (control 6.98 ± 0.58 vs diabetic rats 4.35 ± 0.24 h mg/ml) and C max values (control 0.52 ± 0.03 vs diabetic group 0.33 ± 0.01 µg/ml), while the SMN-received group showed remarkable recovery of diabetes-reduced values of AUC and C max. These findings indicated that diabetes resulted in a significant up-regulation of microsomal enzyme activities. Moreover, as SMN could significantly regulate the enzyme activities and consequently the atorvastatin pharmacokinetics in diabetic rats, its regulative effect in a combination therapy is concluded.


Subject(s)
Heptanoic Acids/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Pyrroles/pharmacokinetics , Silymarin/pharmacology , Animals , Atorvastatin , Cytochrome P-450 CYP3A/physiology , Diabetes Mellitus, Experimental/metabolism , Drug Interactions , Liver/metabolism , Male , Rats , Rats, Wistar , Streptozocin , Testosterone/metabolism
7.
Eur Heart J Cardiovasc Pharmacother ; 10(4): 307-315, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38196131

ABSTRACT

AIMS: Objective methods to determine statin adherence are requested to improve lipid management. We have recently established a method to detect reduced adherence to atorvastatin therapy with cut-off values based on the sum of atorvastatin and its major metabolites in the blood. We aimed to validate this method in patients with and without cardiovascular disease, and optimize previous cut-off values. METHODS AND RESULTS: The pharmacokinetic study included 60 participants treated with atorvastatin 20 mg (N = 20), 40 mg (N = 20), and 80 mg (N = 20). Atorvastatin was then stopped and blood samples collected from day zero to day four. Quantification of the parent drug and its metabolites in blood plasma was performed with a liquid chromatography-tandem mass spectrometry assay. The cut-off values for reduced adherence were validated and optimized by calculating diagnostic sensitivity and specificity. Our candidate cut-off value of dose-normalized six-component sum of atorvastatin plus metabolites <0.10 nM/mg provided a sensitivity of 97% and a specificity of 93% for detecting ≥2 omitted doses. An optimized cut-off <0.062 nM/mg provided a sensitivity of 90% and a specificity of 100%. An alternative simplified two-component metabolite sum with a cut-off value <0.05 nM/mg provided a sensitivity of 98% and a specificity of 76%. An optimized cut-off <0.02 nM/mg provided a sensitivity of 97% and a specificity of 98%. CONCLUSION: This validation study confirms that our direct method discriminates reduced adherence from adherence to atorvastatin therapy with high diagnostic accuracy. The method may improve lipid management in clinical practice and serve as a useful tool in future studies.


Subject(s)
Atorvastatin , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Medication Adherence , Atorvastatin/pharmacokinetics , Atorvastatin/therapeutic use , Atorvastatin/blood , Humans , Male , Female , Middle Aged , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Aged , Heptanoic Acids/pharmacokinetics , Heptanoic Acids/administration & dosage , Heptanoic Acids/blood , Heptanoic Acids/therapeutic use , Pyrroles/pharmacokinetics , Pyrroles/blood , Pyrroles/administration & dosage , Tandem Mass Spectrometry , Chromatography, Liquid , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/blood , Cardiovascular Diseases/prevention & control , Reproducibility of Results , Dose-Response Relationship, Drug
8.
Adv Ther ; 41(7): 2808-2825, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38771476

ABSTRACT

INTRODUCTION: Fixed-dose combinations (FDCs) of angiotensin II receptor blockers, calcium channel blockers, and statins are conventional therapeutic interventions prescribed for cardiovascular diseases. This study aimed at drawing a comparison between the pharmacokinetics and safety of an FDC and the corresponding individual formulations in healthy subjects. METHODS: A randomized, open-label, single-dose, three-sequence, three-period, partially repeated crossover study was conducted with a cohort of healthy volunteers. A 14-day washout period was maintained between each of the three periods. In this study, candesartan cilexetil, amlodipine, and atorvastatin was administered orally as FDCs of 16/10/40 mg in study 1 and 16/5/20 mg in study 2. The maximum plasma concentration (Cmax) and area under the plasma concentration-time curve from time zero to the time of the last quantifiable concentration (AUClast) of candesartan, amlodipine, and atorvastatin were estimated as the geometric mean ratios (GMRs) and 90% confidence intervals (CIs) of the FDC to individual formulations. If the within-subject coefficient of variation (CVwr) of Cmax was greater than 0.3, the bioequivalence (BE) range calculated using the reference-scaled average bioequivalence was used to assess whether the 90% CI was within the BE range. RESULTS: The GMRs (90% CIs) for the AUClast for candesartan and amlodipine were 0.9612 (0.9158-1.0089)/0.9965 (0.9550-1.0397) and 1.0033 (0.9800-1.0271)/1.0067 (0.9798-1.0344), and the GMRs (90% CIs) for Cmax were 0.9600 (0.8953-1.0294)/0.9851 (0.9368-1.0359) and 1.0198 (0.9950-1.0453)/1.0003 (0.9694-1.0321) in studies 1 and 2, respectively. The extended BE ranges calculated from the CVwr of the Cmax of atorvastatin were 0.7814-1.2797 and 0.7415-1.3485, respectively. The GMRs (90% CIs) for the AUClast of atorvastatin were 1.0532 (1.0082-1.1003)/1.0252 (0.9841-1.0680), and the GMRs (90% CIs) for Cmax were 1.0630 (0.9418-1.1997)/0.9888 (0.8792-1.1120) in studies 1 and 2, respectively. CONCLUSION: The Cmax and AUClast values of candesartan cilexetil/amlodipine/atorvastatin 16/10/40 mg and 16/5/20 mg, respectively, were within the BE ranges. There were no clinically significant differences in safety between the two formulations. TRIAL REGISTRATION: ClinicalTrials.gov identifier, study 1: NCT04478097; study 2: NCT04627207.


Subject(s)
Amlodipine , Atorvastatin , Benzimidazoles , Biphenyl Compounds , Cross-Over Studies , Drug Combinations , Tetrazoles , Humans , Biphenyl Compounds/pharmacokinetics , Biphenyl Compounds/administration & dosage , Amlodipine/pharmacokinetics , Amlodipine/administration & dosage , Benzimidazoles/pharmacokinetics , Benzimidazoles/administration & dosage , Tetrazoles/pharmacokinetics , Tetrazoles/administration & dosage , Male , Adult , Female , Atorvastatin/pharmacokinetics , Atorvastatin/administration & dosage , Young Adult , Area Under Curve , Middle Aged , Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Calcium Channel Blockers/pharmacokinetics , Calcium Channel Blockers/administration & dosage , Therapeutic Equivalency , Antihypertensive Agents/pharmacokinetics , Antihypertensive Agents/administration & dosage , Heptanoic Acids/pharmacokinetics , Heptanoic Acids/administration & dosage , Healthy Volunteers
9.
Antimicrob Agents Chemother ; 57(6): 2582-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23529734

ABSTRACT

Boceprevir is a potent orally administered inhibitor of hepatitis C virus and a strong, reversible inhibitor of CYP3A4, the primary metabolic pathway for many 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors. Thus, the aim of the present study was to investigate drug-drug interactions between atorvastatin or pravastatin and boceprevir. We conducted a single-center, open-label, fixed-sequence, one-way-crossover study with 20 healthy adult volunteers. Subjects received single-dose atorvastatin (40 mg) or pravastatin (40 mg) on day 1, followed by boceprevir (800 mg three times daily) for 7 to 10 days. Repeat single doses of atorvastatin or pravastatin were administered in the presence of steady-state boceprevir. Atorvastatin exposure increased in the presence of boceprevir, with atorvastatin area under the concentration-time curve from time zero to infinity after single dosing (AUC(inf)) increasing 2.3-fold (90% confidence interval [CI], 1.85, 2.90) and maximum observed concentration in plasma (Cmax) 2.7-fold (90% CI, 1.81, 3.90). Pravastatin exposure was slightly increased in the presence of boceprevir, with pravastatin AUC(inf) increasing 1.63-fold (90% CI, 1.03, 2.58) and C(max) 1.49-fold (90% CI, 1.03, 2.14). Boceprevir exposure was generally unchanged when the drug was coadministered with atorvastatin or pravastatin. All adverse events were mild and consistent with the known safety profile of boceprevir. The observed 130% increase in AUC of atorvastatin supports the use of the lowest possible effective dose of atorvastatin when coadministered with boceprevir, without exceeding a maximum daily dose of 40 mg. The observed 60% increase in pravastatin AUC with boceprevir coadministration supports the initiation of pravastatin treatment at the recommended dose when coadministered with boceprevir, with close clinical monitoring.


Subject(s)
Drug Interactions , Hepacivirus/drug effects , Heptanoic Acids/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Pravastatin/pharmacokinetics , Proline/analogs & derivatives , Protease Inhibitors/pharmacokinetics , Pyrroles/pharmacokinetics , Adolescent , Adult , Area Under Curve , Atorvastatin , Cross-Over Studies , Female , Hepacivirus/enzymology , Heptanoic Acids/administration & dosage , Humans , Male , Middle Aged , Pravastatin/administration & dosage , Pravastatin/adverse effects , Proline/administration & dosage , Proline/adverse effects , Proline/pharmacokinetics , Protease Inhibitors/administration & dosage , Protease Inhibitors/adverse effects , Pyrroles/administration & dosage , Treatment Outcome , Young Adult
10.
Drug Metab Dispos ; 41(4): 836-43, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23364509

ABSTRACT

Incubational binding or the fraction of drug unbound in an in vitro incubation, fuinc, is an important parameter to predict or measure in the pursuit of accurate clearance predictions from in vitro data. Here we describe a method for fuinc determination directly in the hepatocyte intrinsic clearance (CLint) assay with emphasis on compounds that are actively transported into hepatocytes, hypothesizing that for such compounds the typical protocol of 1 million hepatocytes/ml systematically underestimates the maximum attainable unbound intracellular drug concentration. Using the transporter substrate atorvastatin as a test compound, incubations were performed and a mathematical model applied to describe metabolism, distribution, and binding at different hepatocyte concentrations. From these investigations it was evident that, since binding is more extensive intracellularly than in the medium, increased partitioning into the cellular volume, due to active uptake, increases the total amount of atorvastatin bound in the incubation. Consequently, a significant lowering of the hepatocyte concentration impacts the free drug concentration in the incubation and increases the observed rate of metabolism and therefore observed CLint (that is, when viewed from the media drug concentration). The applicability of the findings was tested for a series of 11 actively transported zwitterions for which standard rat hepatocyte metabolic CLint data (1 million cells/ml incubation) poorly predicted in vivo clearance (average fold error of 5.4). Using metabolic CLint determined at a lower hepatocyte concentration (0.125 million cells/ml) considerably improved clearance predictions (average fold error of 2.3).


Subject(s)
Hepatocytes/metabolism , Heptanoic Acids/pharmacokinetics , Metabolic Clearance Rate , Pyrroles/pharmacokinetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Atorvastatin , Biological Transport, Active , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , In Vitro Techniques , Male , Models, Biological , Rats
11.
Drug Metab Dispos ; 41(4): 897-905, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23378628

ABSTRACT

Lipophilic (logP > 1) and amphiphilic drugs (also known as cationic amphiphilic drugs) with ionizable amines (pKa > 6) can accumulate in lysosomes, a process known as lysosomal trapping. This process contributes to presystemic extraction by lysosome-rich organs (such as liver and lung), which, together with the binding of lipophilic amines to phospholipids, contributes to the large volume of distribution characteristic of numerous cardiovascular and central nervous system drugs. Accumulation of lipophilic amines in lysosomes has been implicated as a cause of phospholipidosis. Furthermore, elevated levels of lipophilic amines in lysosomes can lead to high organ-to-blood ratios of drugs that can be mistaken for active drug transport. In the present study, we describe an in vitro fluorescence-based method (using the lysosome-specific probe LysoTracker Red) to identify lysosomotropic agents in immortalized hepatocytes (Fa2N-4 cells). A diverse set of compounds with various physicochemical properties were tested, such as acids, bases, and zwitterions. In addition, the partitioning of the nonlysosomotropic atorvastatin (an anion) and the lysosomotropics propranolol and imipramine (cations) were quantified in Fa2N-4 cells in the presence or absence of various lysosomotropic or nonlysosomotropic agents and inhibitors of lysosomal sequestration (NH4Cl, nigericin, and monensin). Cellular partitioning of propranolol and imipramine was markedly reduced (by at least 40%) by NH4Cl, nigericin, or monensin. Lysosomotropic drugs also inhibited the partitioning of propranolol by at least 50%, with imipramine partitioning affected to a lesser degree. This study demonstrates the usefulness of immortalized hepatocytes (Fa2N-4 cells) for determining the lysosomal sequestration of lipophilic amines.


Subject(s)
Hepatocytes/metabolism , Heptanoic Acids/pharmacokinetics , Imipramine/pharmacokinetics , Lysosomes/metabolism , Propranolol/pharmacokinetics , Pyrroles/pharmacokinetics , Adrenergic beta-Antagonists/pharmacokinetics , Amines/metabolism , Ammonium Chloride/pharmacology , Antidepressive Agents, Tricyclic/pharmacokinetics , Atorvastatin , Cell Line, Transformed , Diuretics/pharmacokinetics , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Monensin/pharmacology , Nigericin/pharmacology
12.
Toxicol Appl Pharmacol ; 266(3): 452-8, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23200775

ABSTRACT

It has been hypothesized in the literature that intake of high-dosage vitamin E supplements might alter the expression of cytochrome P(450) enzymes (CYP), particularly CYP3A4, which may lead to adverse nutrient-drug interactions. Because previously published studies reported conflicting findings, we investigated the pharmacodynamics of the lipid-lowering drug atorvastatin (ATV), a CYP3A4 substrate, in response to high-dose α-tocopherol (αT) feeding and determined protein expression and activities of relevant CYP. Groups of ten female Dunkin-Hartley guinea pigs were fed a control (5% fat) or a high-fat control diet (HFC; 21% fat, 0.15% cholesterol) or the HFC diet fortified with αT (250 mg/kg diet), ATV (300 mg/kg diet) or both ATV+αT for 6 weeks. Relative to control, HFC animals had increased serum cholesterol concentrations, which were significantly reduced by ATV. High-dose αT feeding in combination with ATV (ATV+αT), albeit not αT feeding alone (αT), significantly lowered serum cholesterol relative to HFC, but did not alter the cholesterol-lowering activity of the drug compared to the ATV treated guinea pigs. Protein expression of CYP3A4, CYP4F2, CYP20A1 and OATP C was similar in all groups. Accordingly, no differences in plasma concentrations of phase I metabolites of ATV were observed between the ATV and ATV+αT groups. In conclusion, feeding guinea pigs high-doses of αT for 6 weeks did neither alter the hepatic expression of CYP, nor the pharmacodynamics and metabolism of ATV. High-dose αT intake is thus unlikely to change the efficacy of drugs metabolized by CYP enzymes, particularly by CYP3A4.


Subject(s)
Anticholesteremic Agents/pharmacology , Cytochrome P-450 CYP3A/metabolism , Heptanoic Acids/pharmacology , Liver/drug effects , Pyrroles/pharmacology , alpha-Tocopherol/pharmacology , Animals , Anticholesteremic Agents/blood , Anticholesteremic Agents/metabolism , Atorvastatin , Blotting, Western , Cholesterol/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Female , Guinea Pigs , Heptanoic Acids/blood , Heptanoic Acids/pharmacokinetics , Liver/enzymology , Liver/metabolism , Pyrroles/blood , Pyrroles/pharmacokinetics , Random Allocation
13.
Mol Pharm ; 10(11): 4038-45, 2013 Nov 04.
Article in English | MEDLINE | ID: mdl-24044638

ABSTRACT

The Biopharmaceutics Drug Disposition Classification System (BDDCS) predicts intestinal transporter effects to be clinically insignificant following oral dosing for highly soluble and highly permeable/metabolized drugs (class 1 drugs). We investigated the effect of inhibiting P-glycoprotein (P-gp) on the in vitro rat intestinal permeability (Papp) and metabolism of the class 1 drug verapamil. Jejunal segments from Sprague-Dawley rats fasted overnight were mounted in Ussing chambers filled with 10 mL of Krebs-Ringer buffer (KRB). For P-gp inhibition studies, GG918 0.5 µM was added to the KRB solution. The experiment started by the addition of verapamil (1 or 10 µM) to either apical or basolateral sides. Samples from verapamil donor and receiver compartments were collected at 30 s and 0.166, 0.5, 1, 1.83 and 3 h after the start of the experiment. Analysis of verapamil and its major metabolite, norverapamil, in the samples and intracellularly at 3 h was performed by HPLC. The same experiment was repeated with norverapamil 10 µM (verapamil metabolite), digoxin 100 nM (positive control for P-gp activity) and atorvastatin 1 and 10 µM (example of a class 2 drug). For 1 µM verapamil, efflux ratio (B to A Papp/A to B Papp) was 4.6 and markedly decreased by GG918 (efflux ratio = 1.1). For 10 µM verapamil efflux ratio was 4.1 (control) vs 1.8 (GG918), comparable to the change seen for digoxin 100 nM with an efflux ratio of 3.6 (control) vs 1.6 (with GG918) and atorvastatin (efflux ratio of 5.2 and 3.0 for atorvastatin 1.0 and 10 µM, respectively, changed to 1.0 and 0.65 with GG918). The changes observed in the norverapamil 10 µM experiment were also significant, where efflux ratio decreased from 13.5 (control) to 1.5 (GG918). The extraction ratio (ER) of 10 µM verapamil to norverapamil decreased from 0.41 after an apical dose to 0.21 after a basolateral dose, but was unaffected by the incubation with GG918. The results suggest that P-gp inhibition has an effect on class 1 drug verapamil and class 2 drug atorvastatin Papp in the rat intestine. Moreover, a stronger P-gp effect on the Papp of the more polar norverapamil metabolite was observed. Papp changes caused by the P-gp inhibitor GG918 do not affect the extent of verapamil metabolism.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Verapamil/metabolism , Verapamil/pharmacokinetics , Animals , Atorvastatin , Digoxin/metabolism , Digoxin/pharmacokinetics , Heptanoic Acids/metabolism , Heptanoic Acids/pharmacokinetics , Intestinal Absorption , Pyrroles/metabolism , Pyrroles/pharmacokinetics , Rats , Rats, Sprague-Dawley , Verapamil/analogs & derivatives
14.
Cardiovasc Drugs Ther ; 27(3): 211-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23400638

ABSTRACT

PURPOSE: Polymorphonuclear neutrophils, the first leukocytes to infiltrate the inflamed tissue, can make important contributions to vascular inflammatory processes driving the development of atherosclerosis. We herein investigated the effects of atorvastatin and NCX 6560 (a nitric oxide (NO)-donating atorvastatin derivative that has completed a successful phase 1b study) on neutrophilic inflammation in carotid arteries of normocholesterolemic rabbits subjected to perivascular collar placement. METHODS: Atorvastatin or NCX 6560 were administered orally (5 mg/kg/day or equimolar dose) to New Zealand White rabbits for 6 days, followed by collar implantation 1 h after the last dose. Twenty-four hours later carotids were harvested for neutrophil quantification by immunostaining. RESULTS: Treatment with NCX 6560 was associated with a lower neutrophil infiltration (-39.5 %), while atorvastatin did not affect neutrophil content. The result was independent of effects on plasma cholesterol or differences in atorvastatin bioavailability, which suggests an important role of NO-related mechanisms in mediating this effect. Consistent with these in vivo findings, in vitro studies showed that NCX 6560, as compared to atorvastatin, had greater inhibitory activity on processes involved in neutrophil recruitment, such as migration in response to IL-8 and IL-8 release by endothelial cells and by neutrophils themselves. Pretreatment with NCX 6560, but not with atorvastatin, reduced the ability of neutrophil supernatants to promote monocyte chemotaxis, a well-known pro-inflammatory activity of neutrophils. CONCLUSION: Experimental data suggest a potential role of NO-releasing statins in the control of the vascular inflammatory process mediated by polymorphonuclear neutrophils.


Subject(s)
Atherosclerosis/prevention & control , Cholesterol/blood , Heptanoic Acids/therapeutic use , Neutrophil Infiltration/drug effects , Nitric Oxide Donors/therapeutic use , Pyrroles/therapeutic use , Acute Disease , Animals , Atherosclerosis/blood , Atherosclerosis/immunology , Atherosclerosis/pathology , Atorvastatin , Carotid Arteries/drug effects , Carotid Arteries/pathology , Cell Survival/drug effects , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Heptanoic Acids/administration & dosage , Heptanoic Acids/pharmacokinetics , Heptanoic Acids/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Interleukin-8/immunology , Male , Molecular Structure , Monocytes/cytology , Monocytes/drug effects , Neutrophil Infiltration/immunology , Neutrophils/cytology , Neutrophils/drug effects , Nitric Oxide Donors/administration & dosage , Nitric Oxide Donors/pharmacokinetics , Nitric Oxide Donors/pharmacology , Pyrroles/administration & dosage , Pyrroles/pharmacokinetics , Pyrroles/pharmacology , Rabbits
15.
Eur J Clin Pharmacol ; 69(7): 1401-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23407853

ABSTRACT

OBJECTIVES: The objectives of this study were to determine if ABCB1 polymorphisms are associated with interindividual variability in sitagliptin pharmacokinetics and if atorvastatin alters the pharmacokinetic disposition of sitagliptin in healthy volunteers. METHODS: In this open-label, randomized, two-phase crossover study, healthy volunteers were prospectively stratified according to ABCB1 1236/2677/3435 diplotype (n = 9, CGC/CGC; n = 10, CGC/TTT; n = 10, TTT/TTT). In one phase, participants received a single 100 mg dose of sitagliptin; in the other phase, participants received 40 mg of atorvastatin for 5 days, with a single 100 mg dose of sitagliptin administered on day 5. A 24-h pharmacokinetic study followed each sitagliptin dose, and the study phases were separated by a 14-day washout period. RESULTS: Sitagliptin pharmacokinetic parameters did not differ significantly between ABCB1 CGC/CGC, CGC/TTT, and TTT/TTT diplotype groups during the monotherapy phase. Atorvastatin administration did not significantly affect sitagliptin pharmacokinetics, with geometric mean ratios (90 % confidence intervals) for sitagliptin maximum plasma concentration, plasma concentration-time curve from zero to infinity, renal clearance, and fraction of sitagliptin excreted unchanged in the urine of 0.93 (0.86-1.01), 0.96 (0.91-1.01), 1.02 (0.93-1.12), and 0.98 (0.90-1.06), respectively. CONCLUSIONS: ABCB1 CGC/CGC, CGC/TTT, and TTT/TTT diplotypes did not influence sitagliptin pharmacokinetics in healthy volunteers. Furthermore, atorvastatin had no effect on the pharmacokinetics of sitagliptin in the setting of ABCB1 CGC/CGC, CGC/TTT, and TTT/TTT diplotypes.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Heptanoic Acids/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Polymorphism, Single Nucleotide , Pyrazines/pharmacokinetics , Pyrroles/adverse effects , Triazoles/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adult , Atorvastatin , Biotransformation/drug effects , Cohort Studies , Colorado , Cross-Over Studies , Dipeptidyl-Peptidase IV Inhibitors/blood , Dipeptidyl-Peptidase IV Inhibitors/urine , Drug Interactions , Female , Genetic Association Studies , Half-Life , Heptanoic Acids/blood , Heptanoic Acids/pharmacokinetics , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Male , Metabolic Clearance Rate/drug effects , Middle Aged , Pyrazines/blood , Pyrazines/urine , Pyrroles/blood , Pyrroles/pharmacokinetics , Sitagliptin Phosphate , Triazoles/blood , Triazoles/urine , Young Adult
16.
Eur J Clin Pharmacol ; 69(3): 477-87, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22922682

ABSTRACT

PURPOSE: Interactions between ticagrelor and atorvastatin or simvastatin were investigated in two-way crossover studies. METHODS: Both studies were open-label for statin; the atorvastatin study was placebo-controlled for ticagrelor. For atorvastatin, volunteers (n = 24) received ticagrelor (loading dose 270 mg; 90 mg twice daily, 7 days) or placebo, plus atorvastatin calcium (80 mg; day 5). For simvastatin, volunteers (n = 24) received simvastatin 80 mg, or ticagrelor (loading dose 270 mg; 180 mg twice daily, 7 days) plus simvastatin (80 mg; day 5). In each study, volunteers received the alternate treatment after washout (≥ 7 days). RESULTS: Ticagrelor increased mean atorvastatin maximum plasma concentration (C(max)) and area under the plasma concentration-time curve from zero to infinity (AUC) by 23 % and 36 %, respectively. Simvastatin C(max) and AUC were increased by 81 % and 56 % with ticagrelor. Ticagrelor also increased C(max) and AUC of analysed atorvastatin metabolites by 13-55 % and 32-67 %, respectively, and simvastatin acid by 64 % and 52 %, respectively. Co-administration of ticagrelor with each statin was well tolerated. CONCLUSIONS: Exposure to ticagrelor and its active metabolite, AR-C124910XX, was generally unchanged by a single dose of either statin, except for a minor increase in ticagrelor C(max) in the presence of simvastatin. Effects of ticagrelor on atorvastatin pharmacokinetics were modest and unlikely clinically relevant, while with simvastatin, changes were slightly larger, and simvastatin doses >40 mg with ticagrelor should be avoided.


Subject(s)
Adenosine/analogs & derivatives , Heptanoic Acids/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacokinetics , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Pyrroles/pharmacokinetics , Simvastatin/pharmacokinetics , Adenosine/administration & dosage , Adenosine/adverse effects , Adenosine/pharmacokinetics , Adult , Area Under Curve , Atorvastatin , Biotransformation , Cross-Over Studies , Drug Administration Schedule , Drug Interactions , Drug Therapy, Combination , Female , Half-Life , Heptanoic Acids/administration & dosage , Heptanoic Acids/adverse effects , Heptanoic Acids/blood , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Least-Squares Analysis , Linear Models , Male , Metabolic Clearance Rate , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/adverse effects , Purinergic P2Y Receptor Antagonists/administration & dosage , Purinergic P2Y Receptor Antagonists/adverse effects , Pyrroles/administration & dosage , Pyrroles/adverse effects , Pyrroles/blood , Simvastatin/administration & dosage , Simvastatin/adverse effects , Simvastatin/blood , Ticagrelor
17.
Eur J Clin Pharmacol ; 69(6): 1235-45, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23334403

ABSTRACT

PURPOSE: To characterise further the previously observed cytochrome P450 3A4 (CYP3A4) interaction of the dual orexin receptor antagonist almorexant. METHODS: Pharmacokinetic interactions were investigated (n = 14 healthy male subjects in two treatment groups) between almorexant at steady-state when administered either concomitantly or 2 h after administration of single doses of simvastatin (40 mg) or atorvastatin (40 mg). RESULTS: Almorexant dose-dependently increased simvastatin exposure (AUC0-∞) when administered concomitantly [geometric mean ratios (90 % CI): 2.5 (2.1, 2.9) (100 mg), 3.9 (3.3, 4.6) (200 mg)], but not Cmax [3.7 (3.0, 4.5) for both doses]. Time-separated administration resulted in relevant reductions of the interaction [AUC0-∞: 1.4 (1.2, 1.7) (100 mg), 1.7 (1.5, 2.0) (200 mg); Cmax: 1.5 (1.3, 1.9) (100 mg), 1.9 (1.6, 2.4) (200 mg)]. Similar results were obtained for hydroxyacid simvastatin. Independent of almorexant dose and relative time of administration, AUC0-∞ and Cmax of atorvastatin increased (ratios ranged from 1.1 to 1.5). AUC0-∞ and Cmax of o-hydroxy atorvastatin decreased dose-independently [AUC0-∞: 0.8 (0.8, 0.9) (100 mg), 0.6 (0.5, 0.6) (200 mg); Cmax: 0.3 (0.3, 0.4) (100 mg), 0.2 (0.2, 0.3) (200 mg)] when atorvastatin was concomitantly administered. Cmax of o-hydroxy atorvastatin slightly decreased (0.8 for both doses) following time-separated administration; AUC0-∞ was unchanged. CONCLUSIONS: Whereas almorexant increased simvastatin exposure dose- and relative time of administration-dependently, atorvastatin exposure increased to a smaller extent and irrespective of dose and time. This suggests that the observed interaction of almorexant with simvastatin is mainly caused by intestinal CYP3A4 inhibition, whereas the interaction with atorvastatin is more due to hepatic CYP3A4 inhibition.


Subject(s)
Acetamides/administration & dosage , Cytochrome P-450 CYP3A Inhibitors , Enzyme Inhibitors/administration & dosage , Heptanoic Acids/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hypnotics and Sedatives/administration & dosage , Intestines/drug effects , Isoquinolines/administration & dosage , Liver/drug effects , Pyrroles/pharmacokinetics , Simvastatin/pharmacokinetics , Acetamides/blood , Acetamides/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Atorvastatin , Biotransformation , Cytochrome P-450 CYP3A/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Interactions , Germany , Half-Life , Heptanoic Acids/administration & dosage , Heptanoic Acids/blood , Humans , Hydroxylation , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Hypnotics and Sedatives/blood , Hypnotics and Sedatives/pharmacokinetics , Intestines/enzymology , Isoquinolines/blood , Isoquinolines/pharmacokinetics , Liver/enzymology , Male , Metabolic Clearance Rate , Pyrroles/administration & dosage , Pyrroles/blood , Simvastatin/administration & dosage , Simvastatin/blood , Young Adult
18.
Surg Endosc ; 27(6): 2094-101, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23247745

ABSTRACT

BACKGROUND: A previous study of 22 patients undergoing either gastric bypass or duodenal switch showed increased systemic exposure of atorvastatin acid 3-8 weeks after surgery in the majority of patients. This study aimed to investigate the long-term effects on systemic exposure of atorvastatin acid in the same group of patients. METHODS: An 8-h pharmacokinetic investigation was performed a median of 27 months (range 21-45 months) after surgery. Systemic exposure was measured as the area under the plasma concentration versus the time curve from 0 to 8 h postdose (AUC0-8). Linear mixed models with AUC0-8 as the dependent variable were implemented to assess the effect of time, surgical procedure, and body mass index (BMI) as explanatory variables. RESULTS: The study enrolled 20 patients. The systemic exposure of atorvastatin acid changed significantly over time (p = 0.001), albeit there was substantial variation between subjects. The effect of time was attenuated but remained significant after adjustment for surgical procedure and BMI (p = 0.048). The initial AUC0-8 increase seen in the majority of patients 3-8 weeks after surgery was normalized long term, with 7 of the 12 gastric bypass patients and 6 of the 8 duodenal switch patients showing decreased AUC0-8 compared with preoperative values. CONCLUSIONS: The systemic exposure of atorvastatin showed a significant change over time after bariatric surgery, albeit with large inter- and intraindividual variations. The findings indicate that patients using atorvastatin or drugs with similar pharmacokinetic properties should be monitored closely for both therapeutic effects and adverse events the first years after gastric bypass and duodenal switch.


Subject(s)
Biliopancreatic Diversion/adverse effects , Gastric Bypass/adverse effects , Heptanoic Acids/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Obesity, Morbid/surgery , Pyrroles/pharmacokinetics , Adult , Atorvastatin , Biological Availability , Female , Heptanoic Acids/adverse effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Male , Middle Aged , Prospective Studies , Pyrroles/adverse effects
19.
J Pharm Pharm Sci ; 16(4): 577-87, 2013.
Article in English | MEDLINE | ID: mdl-24210065

ABSTRACT

PURPOSE: Atorvastatin calcium (ATC) is classified as class II (low solubility and high permeability) compound according to the biopharmaceutical classification system. The amorphous form of ATC possesses higher solubility, dissolution rate, and bioavailability than its crystalline form. Coamorphous drug system is a new and emerging method to prepare stable amorphous forms, in this case leading to the improved stability of ATC in dissolution medium. METHODS: In this study, coamorphous form of ATC and nicotinamide (ATC-NIC) was prepared from solvent evaporation method and characterized using differential scanning calorimetry (DSC), Fourier transform infrared spectroscopy (FT-IR) and powder X-ray diffraction (PXRD). The intrinsic dissolution rate and solubility of ATC-NIC were determined along with plasma concentrations of ATC using HPLC after oral dosing in rats. RESULTS: The crystalline ATC was converted to coamorphous form revealing a molecular interaction between ATC and NIC. The intrinsic dissolution rate, solubility and plasma concentration of coamorphous ATC-NIC are higher than those of crystalline ATC. ATC-NIC coamorphous system showed greater solution stability than those reported in the literature for amorphous ATC. CONCLUSIONS: Coamorphous ATC-NIC has improved physicochemical and pharmacokinetic properties as compared to ATC. This article is open to POST-PUBLICATION REVIEW. Registered readers (see "For Readers") may comment by clicking on ABSTRACT on the issue's contents page.


Subject(s)
Heptanoic Acids/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Niacinamide/pharmacokinetics , Pyrroles/pharmacokinetics , Animals , Atorvastatin , Female , Heptanoic Acids/blood , Heptanoic Acids/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Niacinamide/blood , Niacinamide/chemistry , Pyrroles/blood , Pyrroles/chemistry , Rats , Rats, Wistar , Solubility
20.
Xenobiotica ; 43(11): 963-72, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23631744

ABSTRACT

1. 5-(N-(4-((4-ethylbenzyl)thio)phenyl)sulfamoyl)-2-methyl benzoic acid (CP-778875), an agonist of the peroxisome proliferator-activated receptor alpha, has been evaluated in the clinic to treat dyslipidemia and type 2 diabetes mellitus. Herein, we investigate the effect of CP-778875 on the pharmacokinetics of atorvastatin acid and its metabolites in humans. 2. The study incorporated a fixed-sequence design conducted in two groups. Group A was designed to estimate the effects of multiple doses of CP-778875 on the single dose pharmacokinetics of atorvastatin. Subjects in group A (n = 26) received atorvastatin (40 mg) on days 1 and 9 and CP-778875 (1.0 mg QD) on days 5-12. Group B was designed to examine the effects of multiple doses of atorvastatin on the single dose pharmacokinetics of CP-778875. Subjects in group B (n = 29) received CP-778875 (0.3 mg) on days 1 and 9 and atorvastatin (40 mg QD) on days 5-12. 3. Mean maximum serum concentration (Cmax) and area under the curve of atorvastatin were increased by 45% and 20%, respectively, upon co-administration with CP-778875. Statistically significant increases in the systemic exposure of ortho- and para-hydroxyatorvastatin were also observed upon concomitant dosing with CP-778875. CP-778875 pharmacokinetics, however, were not impacted upon concomitant dosing with atorvastatin. 4. Inhibition of organic anion transporting polypeptide 1B1 by CP-778875 (IC50 = 2.14 ± 0.40 µM) could be the dominant cause of the pharmacokinetic interaction as CP-778875 did not exhibit significant inhibition of cytochrome P450 3A4/3A5, multidrug resistant protein 1 or breast cancer resistant protein, which are also involved in the hepatobiliary disposition of atorvastatin.


Subject(s)
Benzoates/pharmacology , Benzoic Acid/pharmacology , Heptanoic Acids/pharmacology , PPAR alpha/agonists , Pyrroles/pharmacology , Sulfonamides/pharmacology , Animals , Atorvastatin , Benzoates/chemistry , Benzoic Acid/chemistry , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Dogs , Drug Interactions , HEK293 Cells , Heptanoic Acids/blood , Heptanoic Acids/pharmacokinetics , Humans , Hydroxylation/drug effects , Madin Darby Canine Kidney Cells , Membrane Transport Proteins/metabolism , Oxidation-Reduction/drug effects , Pyrroles/blood , Pyrroles/pharmacokinetics , Sulfonamides/chemistry , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL