Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where John A. Pieper is active.

Publication


Featured researches published by John A. Pieper.


Pharmacogenetics | 2002

Cytochrome P450 2C9 polymorphisms: a comprehensive review of the in-vitro and human data.

Craig R. Lee; Joyce A. Goldstein; John A. Pieper

The discovery of six distinct polymorphisms in the genetic sequence encoding for the cytochrome P450 2C9 (CYP2C9) protein has stimulated numerous investigations in an attempt to characterize their population distribution and metabolic activity. Since the CYP2C9*1, *2 and *3 alleles were discovered first, they have undergone more thorough investigation than the recently identified *4, *5 and *6 alleles. Population distribution data suggest that the variant *2 and *3 alleles are present in approximately 35% of Caucasian individuals; however, these alleles are significantly less prevalent in African-American and Asian populations. In-vitro data have consistently demonstrated that the CYP2C9*2 and *3 alleles are associated with significant reductions in intrinsic clearance of a variety of 2C9 substrates compared with CYP2C9*1; however, the degree of these reductions appear to be highly substrate-dependent. In addition, multiple in-vivo investigations and clinical case reports have associated genotypes expressing the CYP2C9*2 and *3 alleles with significant reductions in both the metabolism and daily dose requirements of selected CYP2C9 substrates. Individuals expressing these variant genotypes also appear to be significantly more susceptible to adverse events with the narrow therapeutic index agents warfarin and phenytoin, particularly during the initiation of therapy. These findings have subsequently raised numerous questions regarding the potential clinical utility of genotyping for CYP2C9 prior to initiation of therapy with these agents. However, further clinical investigations evaluating the metabolic consequences in individuals expressing the CYP2C9*2, *3, *4, *5, or *6 alleles are required before large-scale clinical genotyping can be recommended.


Clinical Pharmacokinectics | 2001

Clinical pharmacokinetics of fluvastatin.

Charity D. Scripture; John A. Pieper

Fluvastatin, the first fully synthetic HMG-CoA reductase inhibitor, has been shown to reduce cholesterol in patients with hyperlipidaemia, to prevent subsequent coronary events in patients with established coronary heart disease, and to alter endothelial function and plaque stability in animal models.Fluvastatin is relatively hydrophilic, compared with the semisynthetic HMG-CoA reductase inhibitors, and, therefore, it is extensively absorbed from the gastrointestinal tract. After absorption, it is nearly completely extracted and metabolised in the liver to 2 hydroxylated metabolites and an N-desisopropyl metabolite, which are excreted in the bile. Approximately 95% of a dose is recovered in the faeces, with 60% of a dose recovered as the 3 metabolites. The 6-hydroxy and N-desisopropyl fluvastatin metabolites are exclusively generated by cytochrome P450 (CYP) 2C9 and do not accumulate in the blood. CYP2C9, CYP3A4, CYP2C8 and CYP2D6 form the 5-hydroxy fluvastatin metabolite. Because of its hydrophilic nature and extensive plasma protein binding, fluvastatin has a small volume of distribution with minimal concentrations in extrahepatic tissues. The pharmacokinetics of fluvastatin are not influenced by renal function, due to its extensive metabolism and biliary excretion; limited data in patients with cirrhosis suggest a 30% reduction in oral clearance. Age and gender do not appear to affect the disposition of fluvastatin.CYP3A4 inhibitors (erythromycin, ketoconazole and itraconazole) have no effect on fluvastatin pharmacokinetics, in contrast to other HMG-CoA reductase inhibitors which are primarily metabolised by CYP3A and are subject to potential drug interactions with CYP3A inhibitors. Coadministration of fluvastatin with gastrointestinal agents such as cholestyramine, and gastric acid regulating agents (H2 receptor antagonists and proton pump inhibitors), significantly alters fluvastatin disposition by decreasing and increasing bioavailability, respectively. The nonspecific CYP inducer rifampicin (rifampin) significantly increases fluvastatin oral clearance.In addition to being a CYP2C9 substrate, fluvastatin demonstrates inhibitory effects on this isoenzyme in vitro and in vivo. In human liver microsomes, fluvastatin significantly inhibits the hydroxylation of 2 CYP2C9 substrates, tolbutamide and diclofenac. The oral clearances of the CYP2C9 substrates diclofenac, tolbutamide, glibenclamide (glyburide) and losartan are reduced by 15 to 25% when coadministered with fluvastatin. These alterations have not been shown to be clinically significant. There are inadequate data evaluating the potential interaction of fluvastatin with warfarin and phenytoin, 2 CYP2C9 substrates with a narrow therapeutic index, and caution is recommended when using fluvastatin with these agents. Fluvastatin does not appear to have a significant effect on other CYP isoenzymes or P-glycoprotein-mediated transport in vivo.


Clinical Pharmacology & Therapeutics | 1998

Effects of erythromycin or rifampin on losartan pharmacokinetics in healthy volunteers

Kristin M. Williamson; J. Herbert Patterson; Robert H. McQueen; Kirkwood F. Adams; John A. Pieper

Losartan is metabolized by CYP2C9 and CYP3A4 to an active metabolite, E3174, which has greater antihypertensive activity than the parent compound. Coadministered drugs that inhibit or induce metabolic processes may therefore alter the pharmacokinetics and pharmacologic response of losartan and E3174.


Pharmacotherapy | 1998

Optimal management of amiodarone therapy: Efficacy and side effects

Daniel E. Hilleman; Mary Ann Miller; Robert B. Parker; Paul L. Doering; John A. Pieper

Objectives. To review management and dosing guidelines for amiodarone therapy, and discuss the drugs adverse event profile.


Clinical Pharmacology & Therapeutics | 2002

Evaluation of cytochrome P4502C9 metabolic activity with tolbutamide in CYP2C9*1 heterozygotes

Craig R. Lee; John A. Pieper; Alan L. Hinderliter; Joyce Blaisdell; Joyce A. Goldstein

Multiple single‐nucleotide polymorphisms in the gene encoding cytochrome P450 (CYP) 2C9 have been identified, but the functional significance of the various putative defective genotypes in humans merits further study.


The Journal of Clinical Pharmacology | 2001

Lack of gender differences and large intrasubject variability in cytochrome P450 activity measured by phenotyping with dextromethorphan

Jeannine S. McCune; Celeste Lindley; Jodi L. Decker; Kristin M. Williamson; Amy M. Meadowcroft; Donald W. Graff; William T. Sawyer; David K. Blough; John A. Pieper

Gender‐based differences in cytochrome P450 (CYP) activity may occur due to endogenous hormonal fluctuations with the menstrual cycle, which are altered by oral contraceptives. This study assessed the average activity and within‐subject variability in CYP3A4 and CYP2D6 in men, women taking Triphasil®, and regularly menstruating women not receiving oral contraceptives. Thirty‐three healthy volunteers participated in this 28‐day pilot study (12 women receiving Triphasil®) (OCs), 11 regularly menstruating women not on exogenous progesterone or estrogen (no OCs), and 10 men. CYP3A4 and CYP2D6 activities were phenotyped with dextromethorphan (DM) on study days 7,14,21, and 28 using urinary ratios of DM:3‐methoxymorphinan (3MM) and DM:dextrorphan (DX), respectively. Serial blood concentrations of estrogen and progesterone and menstrual diaries were used to determine menstrual phase in both groups of women. Average urinary DM:3MM and DM:DXin the 28 extensive metabolizers of CYP2D6 did not differ between the three study populations (p = 0.86 and 0.93, respectively). Post hoc power analysis indicated that more than 1000 subjects would be needed for 80% power (a = 0.05) to detect a ± 15% difference from the population mean in the urinary ratios of dextromethorphan and its metabolites 3MM and DX. Variability in CYP3A4 and CYP2D6 activity, characterized by intrasubject standard deviation, also did not differ. The varying doses of levonorgesterol and ethinyl estradiol in Triphasil®, fluctuations in estrogen and progesterone, and menstrual phase did not influence CYP3A4 or CYP2D6 activity. It was concluded that CYP3A4 and CYP2D6 activity and intrasubject variability were not different in the three study populations, and thus a clinically important difference between men, women on Triphasil®, and women not receiving oral contraceptives is unlikely. High inter‐ and intrasubject variability in DM:3MM and DM:DX were clearly demonstrated and limit the use of dextromethorphan to phenotype endogenous CYP3A4 and CYP2D6 activity.


The Journal of Clinical Pharmacology | 1999

The effects of fluvastatin, a CYP2C9 inhibitor, on losartan pharmacokinetics in healthy volunteers.

Amy M. Meadowcroft; Kristin M. Williamson; J. Herbert Patterson; Alan L. Hinderliter; John A. Pieper

Losartan is an angiotensin II receptor antagonist that is metabolized by CYP2C9 and CYP3A4 to a more potent antihypertensive metabolite, E3174. Interaction studies with inhibitors of CYP3A4 have not demonstrated significant changes in the pharmacokinetics of losartan or E3174. The authors assessed the steady‐state pharmacokinetics of losartan and E3174 when administered alone and concomitantly with fluvastatin, a specific CYP2C9 inhibitor. A prospective, open‐label, crossover study was conducted in 12 healthy volunteers with losartan alone and in combination with fluvastatin. The baseline phase was 7 days of losartan (50 mg QAM), and the inhibition phase was 14 total days of fluvastatin (40 mg QHS), with the final 7 days including losartan. The authors found that flvastatin did not significantly change the steady‐state AUC0–24 or half‐life of losartan or E3174. Losartan apparent oral clearance was not affected by fluvastatin. Inhibition of losartan metabolism appears to require both CYP2C9 and CYP3A4 inhibition.


The Journal of Clinical Pharmacology | 2001

Effect of fluoxetine on carvedilol pharmacokinetics, CYP2D6 activity, and autonomic balance in heart failure patients

Donald W. Graff; Kristin M. Williamson; John A. Pieper; Stanley W. Carson; Kirkwood F. Adams; Wayne E. Cascio; J. Herbert Patterson

The objective of this study was to examine the pharmacokinetic and pharmacodynamic consequences of concomitant administration of fluoxetine and carvedilol in heart failure patients. Fluoxetine (20 mg) or matching placebo was administered in a randomized, double‐blind, two‐period crossover study to 10 patients previously identified as extensive metabolizers of CYP2D6 substrates. Patients were maintained on a carvedilol dose of 25 or 50 mg bid and given fluoxetine/placebo for a minimum of 28 days. Plasma was collected over the 12‐hour carvedilol dosing interval, and the concentrations of the R(+) and S(−) enantiomers of carvedilol were measured. CYP2D6 phenotype was assessed during each study period using dextromethorphan (30 mg). Changes in autonomic modulation between study periods were measured by heart rate variability in the time and frequency domains using ambulatory electrocardiographic monitoring. Compared to placebo, fluoxetine coadministration resulted in a 77% increase in mean (± SD) R(+) enantiomer AUC0–12 (522 ± 413 vs. 927 ± 506 ng•h/mL, p = 0.01) and a nonsignificant increase in S(−) enantiomer AUC (244 ± 185 vs. 330 ±179 ng•h/mL, p = 0.17). Mean apparent oral clearance for both enantiomers decreased significantly with fluoxetine administration (R(+): 10.3 ± 7.2 vs. 4.5 ± 2.2 mL/min/kg; S(−): 22.5 ± 12.3 vs. 12.6 ± 7.4 mL/min/kg; p = 0.004 and 0.03, respectively). No differences in adverse effects, blood pressure, or heart rate were noted between treatment groups, and there were no consistent changes in heart rate variability parameters. In conclusion, fluoxetine administration resulted in a stereospecific inhibition of carvedilol metabolism, with the R(+) enantiomer increasing to a greater extent than the S(−) enantiomer. However, this interaction was of little clinical significance in our sample population.


Clinical Pharmacology & Therapeutics | 2002

Evaluation of potential losartan‐phenytoin drug interactions in healthy volunteers*

Tracy L. Fischer; John A. Pieper; Donald W. Graff; Jo E. Rodgers; Jeffrey D. Fischer; Kimberly J. Parnell; Joyce A. Goldstein; Robert S. Greenwood; J. Herbert Patterson

Phenytoin, a cytochrome P450 (CYP) 2C9 substrate, has a narrow therapeutic index and nonlinear pharmacokinetics. Therefore there is the potential for significant concentration‐related adverse effects when phenytoin is coadministered with other CYP2C9 substrates. Losartan, an antihypertensive agent, is also a substrate for CYP2C9.


Pharmacotherapy | 2003

Losartan and E3174 pharmacokinetics in cytochrome P450 2C9*1/*1, *1/*2, and *1/*3 individuals

Craig R. Lee; John A. Pieper; Alan L. Hinderliter; Joyce Blaisdell; Joyce A. Goldstein

Study Objective. To determine if differences in the pharmacokinetics of losartan and its pharmacologically active E3174 metabolite exist among individuals expressing the cytochrome P450 (CYP) 2C9*1/*1, *1/*2, and *1/*3 genotypes.

Collaboration


Dive into the John A. Pieper's collaboration.

Top Co-Authors

Avatar

Craig R. Lee

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Joyce A. Goldstein

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kristin M. Williamson

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Alan L. Hinderliter

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Donald W. Graff

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

J. Herbert Patterson

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Joyce Blaisdell

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kirkwood F. Adams

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Amy M. Meadowcroft

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

J.H. Patterson

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge