31 results on '"Porras, AG"'
Search Results
2. The effects of modifying in vivo cytochrome P450 3A (CYP3A) activity on etoricoxib pharmacokinetics and of etoricoxib administration on CYP3A activity.
- Author
-
Agrawal NGB, Matthews CZ, Mazenko RS, Woolf EJ, Porras AG, Chen X, Miller JL, Michiels N, Wehling M, Schultz A, Gottlieb AB, Kraft WK, Greenberg HE, Waldman SA, Curtis SP, and Gottesdiener KM
- Abstract
To investigate the influence of modifying in vivo cytochrome P450 3A (CYP3A) activity on the pharmacokinetics of etoricoxib, a selective inhibitor of cyclooxygenase-2, and of etoricoxib administration on CYP3A activity, a 3-part, randomized, crossover study was conducted in 3 panels of healthy volunteers. In part I, 8 subjects were administered a single dose of 60 mg etoricoxib alone and following daily doses of 400 mg ketoconazole, a known strong inhibitor of CYP3A. In part II, 8 different subjects were administered a single dose of 60 mg etoricoxib alone and following daily doses of 600 mg rifampin, a known strong inducer of CYP3A. In parts I and II, plasma samples were collected following each etoricoxib dose and analyzed for etoricoxib. In part III, 8 different subjects were administered 120 mg etoricoxib or placebo once daily for 11 days, and the erythromycin breath test was administered on day 11 of each period. Coadministration of etoricoxib with daily doses of ketoconazole resulted in an average 43% increase in etoricoxib AUC; based on previous studies, this increase would not be expected to have any clinically meaningful effect. In contrast, coadministration of etoricoxib with daily doses of rifampin had a potentially clinically important effect on etoricoxib pharmacokinetics (average 65% decrease in etoricoxib AUC). Etoricoxib had no effect on hepatic CYP3A activity, as assessed by the erythromycin breath test. [ABSTRACT FROM AUTHOR]
- Published
- 2004
3. Pharmacokinetics of etoricoxib in patients with renal impairment.
- Author
-
Agrawal NGB, Matthews CZ, Mazenko RS, Kline WF, Woolf EJ, Porras AG, Geer LA, Wong PH, Cho M, Cote J, Marbury TC, Moncrief JW, Alcorn H Jr., Swan S, Sack MR, Robson RA, Petty KJ, Schwartz JI, and Gottesdiener KM
- Abstract
The effect of renal insufficiency on the pharmacokinetics of etoricoxib, a selective inhibitor of cyclooxygenase-2, was examined in 23 patients with varying degrees of renal impairment (12 moderate [creatinine clearance between 30 and 50 mL/min/1.73 m2], 5 severe [creatinine clearance below 30 mL/min/1.73 m2], and 6 with end-stage renal disease requiring hemodialysis) following administration of single 120-mg oral doses of etoricoxib. Even the most severe renal impairment was found to have little effect on etoricoxib pharmacokinetics. The low recovery of etoricoxib in dialysate (less than 6% of the dose) supports that hemodialysis also has little effect on etoricoxib pharmacokinetics, and binding of etoricoxib to plasma proteins was generally unaffected by renal disease. Single doses of etoricoxib were generally well tolerated by patients with renal impairment. Based on pharmacokinetic considerations, dosing adjustments are not necessary for patients with any degree of renal impairment. However, because patients with advanced renal disease (creatinine clearance below 30 mL/min/1.73 m2) are likely to be very sensitive to any further compromise of renal function, and there is no long-term clinical experience in these patients, the use of etoricoxib is not recommended in patients with advanced renal disease. [ABSTRACT FROM AUTHOR]
- Published
- 2004
4. Pharmacokinetics of etoricoxib in patients with hepatic impairment.
- Author
-
Agrawal NGB, Rose MJ, Matthews CZ, Woolf EJ, Porras AG, Geer LA, Larson PJ, Cote J, Dilzer SC, Lasseter KC, Alam I, Petty KJ, and Gottesdiener KM
- Abstract
The effect of hepatic insufficiency on the pharmacokinetics of etoricoxib, a selective inhibitor of cyclooxygenase-2, was investigated following administration of single and multiple oral doses to mild hepatic insufficiency patients (Child-Pugh score of 5 to 6), multiple oral doses to moderate hepatic insufficiency patients (Child-Pugh score of 7 to 9), and single intravenous doses to both mild and moderate hepatic insufficiency patients. A trend of decreasing systemic clearance with increasing hepatic impairment was observed. Absorption of etoricoxib was unaffected by hepatic impairment. Binding of etoricoxib to plasma proteins was also found to be unaffected by hepatic disease. Etoricoxib was generally well tolerated by patients with mild and moderate hepatic insufficiency. Together, these results support a 60-mg once-daily dosing regimen for mild hepatic insufficiency patients and a 60-mg every-other-day dosing regimen for moderate hepatic insufficiency patients. There are no clinical or pharmacokinetic data in patients with severe hepatic insufficiency (Child-Pugh score > 9). [ABSTRACT FROM AUTHOR]
- Published
- 2003
5. An evaluation of the dose-dependent inhibitions of CYP1A2 by rofecoxib using theophylline as a CYP1A2 probe.
- Author
-
Bachmann K, White D, Jauregui L, Schwartz JI, Agrawal NGB, Mazenko R, Larson PJ, and Porras AG
- Abstract
This study was undertaken to determine whether rofecoxib can interfere with CYP1A2 activity in humans using theophylline as a probe substrate. Single oral doses of theophylline were administered to each of three panels of 12 healthy subjects receiving daily doses of rofecoxib for 7 days to examine the effect of rofecoxib administration on the absorption and disposition of theophylline. Each panel was administered doses of 12.5, 25, or 50 mg of rofecoxib or a matching placebo in a two-way, randomized, crossover fashion and administered a single oral 300-mg dose of theophylline on day 7 of rofecoxib or placebo administration. Plasma concentrations of theophylline were monitored for 48 hours postdose to assess differences in pharmacokinetics. All three commercially marketed doses of rofecoxib were found to slow the clearance of theophylline with no detectable effect on absorption. CL/F values for theophylline were estimated from AUC infinity and by point estimates from the concentrations of drug in plasma at 12 and 24 hours postdose. The point estimates of CL/F were found to be in agreement with those derived from AUC. [ABSTRACT FROM AUTHOR]
- Published
- 2003
6. Single- and multiple-dose pharmacokinetics of etoricoxib, a selective inhibitor of cyclooxygenase-2, in man.
- Author
-
Agrawal NGB, Porras AG, Matthews CZ, Rose MJ, Woolf EJ, Musser BJ, Dynder AL, Mazina KE, Lasseter KC, Hunt TL, Schwartz JI, McCrea JB, and Gottesdiener KM
- Abstract
The single- and multiple-dose pharmacokinetics of etoricoxib, a selective inhibitor of cyclooxygenase-2, were examined in two clinical studies. Single-dose pharmacokinetics--including dose proportionality, absolute bioavailability of the highest dose-strength (120-mg) tablet, and the effect of a high-fat meal on the bioavailability of that tablet--were investigated in a two-part, open, balanced crossover study in two panels of healthy subjects (12 per panel). Steady-state pharmacokinetics were investigated in an open-label study in which 24 healthy subjects were administered 120-mg single and multiple (once daily for 10 days) oral doses of etoricoxib tablets. The pharmacokinetics of etoricoxib were found to be consistent with linearity through doses at least twofold greater than the highest anticipated clinical dose of 120 mg. Etoricoxib administered as a tablet was rapidly and completely absorbed and available; the absolute bioavailability was estimated to be 100%. A high-fat meal decreased the rate of absorption without affecting the extent of absorption of etoricoxib; therefore, etoricoxib can be dosed irrespective of food. Steady-state pharmacokinetics of etoricoxib, achieved following 7 days of once-daily dosing, were found to be reasonably predicted from single doses. The accumulation ratio averaged 2.1, and the corresponding accumulation t1/2 averaged 22 hours, supporting once-daily dosing. Etoricoxib was generally well tolerated. [ABSTRACT FROM AUTHOR]
- Published
- 2003
7. Effect of rofecoxib on prednisolone and prednisone pharmacokinetics in healthy subjects.
- Author
-
Schwartz JI, Mukhopadhyay S, Porras AG, Viswanathan-Aiyer K, Adcock S, Ebel DL, and Gertz BJ
- Abstract
Patients receiving nonsteroidal anti-inflammatory drug therapy may also require administration of corticosteroids, particularly patients with rheumatoid arthritis. To investigate the effect of rofecoxib on the single-dose pharmacokinetics of oral prednisone and intravenous prednisolone, the authors conducted a randomized, double-blind, placebo-controlled crossover study in 12 healthy subjects. Oral rofecoxib (250.0 mg/day for 14 days) failed to influence prednisone or prednisolone pharmacokinetics after intravenous prednisolone or oral prednisone administration. The geometric mean ratio (GMR) (90% confidence interval) of prednisolone AUC infinity (rofecoxib/placebo) following intravenous and oral corticosteroid was 0.97 (0.94, 1.01) and 0.99 (0.91, 1.08), respectively. Similarly, the prednisone AUC infinity GMRs (rofecoxib/placebo) after intravenous and oral corticosteroid were 1.03 (0.95, 1.11) and 1.08 (0.92, 1.28), respectively. The absence of an effect of rofecoxib on the pharmacokinetics of oral prednisone or intravenous prednisolone indicates that no adjustment in dose of this corticosteroid is necessary when administered concurrently with rofecoxib. [ABSTRACT FROM AUTHOR]
- Published
- 2003
8. Safety, tolerability, and pharmacokinetics of escalating high doses of ivermectin in healthy adult subjects.
- Author
-
Guzzo CA, Furtek CI, Porras AG, Chen C, Tipping R, Clineschmidt CM, Sciberras DG, Hsieh JY, and Lasseter KC
- Abstract
Safety and pharmacokinetics (PK) of the antiparasitic drug ivermectin, administered in higher and/or more frequent doses than currently approved for human use, were evaluated in a double-blind, placebo-controlled, dose escalation study. Subjects (n = 68) were assigned to one of four panels (3:1, ivermectin/placebo): 30 or 60 mg (three times a week) or 90 or 120 mg (single dose). The 30 mg panel (range: 34 7-594 microg/kg) also received a single dose with food after a 1-week washout. Safety assessments addressed both known ivermectin CNS effects and general toxicity. The primary safety endpoint was mydriasis, accurately quantitated by pupillometry. Ivermectin was generally well tolerated, with no indication of associated CNS toxicity for doses up to 10 times the highest FDA-approved dose of 200 microg/kg. All dose regimens had a mydriatic effect similar to placebo. Adverse experiences were similar between ivermectin and placebo and did not increase with dose. Following single doses of 30 to 120 mg, AUC and Cmax were generally dose proportional, with t(max) approximately 4 hours and t1/2 approximately 18 hours. The geometric mean AUC of 30 mg ivermectin was 2.6 times higher when administered with food. Geometric mean AUC ratios (day 7/day 1) were 1.24 and 1.40 for the 30 and 60 mg doses, respectively, indicating that the accumulation of ivermectin given every fourth day is minimal. This study demonstrated that ivermectin is generally well tolerated at these higher doses and more frequent regimens. [ABSTRACT FROM AUTHOR]
- Published
- 2002
9. Effect of rofecoxib on the pharmacokinetics of chronically administered oral contraceptives in healthy female volunteers.
- Author
-
Schwartz JI, Wong PH, Porras AG, Ebel DL, Hunt TR, and Gertz BJ
- Abstract
The effect of rofecoxib, a highly selective cyclooxygenase (COX)-2 inhibitor, on the pharmacokinetics of ethinyl estradiol (EE) and norethindrone (NET), two common components of a combination oral contraceptive product, was examined. A double-blind, two-period crossover study was conducted in 18 healthy women who received ORTHO-NOVUM 1/35, a combination of EE (35 microg) and NET (1 mg), concurrently for 14 days with either 175 mg rofecoxib or matching placebo during two consecutive menstrual cycles. Plasma was sampled for EE, NET, sex hormone binding globulin (SHBG), and albumin. The AUC(0-24 h) geometric mean ratio (GMR: rofecoxib/placebo) with corresponding 90% confidence interval (CI) of EE and NET was 1.13 (1.06, 1.19) and 1.18 (1.13, 1.24), respectively. The Cmax GMR of EE and NET was 1.06 (0.98, 1.16) and 1.04 (0.99, 1.09), respectively. In each case, the 90% CIs satisfied the predefined bioequivalence limits of (0.80, 1.25). Measures of SHBG and albumin and routine clinical and laboratory safety parameters showed no clinically meaningful changes. The addition of rofecoxib to the oral contraceptive was not associated with any clinically important changes in EE or NET pharmacokinetics and thus would not be anticipated to influence the efficacy of this contraceptive regimen. [ABSTRACT FROM AUTHOR]
- Published
- 2002
10. Lack of pharmacokinetic interaction between rofecoxib and methotrexate in rheumatoid arthritis patients.
- Author
-
Schwartz JI, Agrawal NGB, Wong PH, Bachmann KA, Porras AG, Miller JL, Ebel DL, Sack MR, Holmes GB, Redfern JS, and Gertz BJ
- Abstract
Rofecoxib is a highly selective and potent inhibitor of cyclooxgenase-2 (COX-2). Methotrexate is a disease-modifying agent with a narrow therapeutic index frequently prescribed for the management of rheumatoid arthritis. The objective of this study was to investigate the influence of clinical doses of rofecoxib on the pharmacokinetics of methotrexate in patients with rheumatoid arthritis. This was a randomized, double-blind, placebo-controlled study in 25 rheumatoid arthritis patients on stable doses of methotrexate. Patients received oral methotrexate (7.5 to 20 mg) on days -1, 7, 14, and 21. Nineteen patients received rofecoxib 12.5, 25, and 50 mg once daily on days 1 to 7, 8 to 14, and 15 to 21, respectively. Six patients received placebo on days 1 to 21 only to maintain a double-blinded design for assessment of adverse experiences. Plasma and urine samples were analyzed for methotrexate and its major although inactive metabolite, 7-hydroxymethotrexate. The AUC(0-infinity) geometric mean ratios (GMR) and their 90% confidence intervals (90% CI) (rofecoxib + methotrexate/methotrexate alone) for day 7/day -1, day 14/day -1, and day 21/day -1, for rofecoxib 12.5, 25, and 50 mg, were 1.03 (0.93, 1.14), 1.02 (0.92, 1.12), and 1.06 (0.96, 1.17), respectively (p > 0.2 for all comparisons to day -1). All AUC(0-infinity), GMR and Cmax GMR 90% CIs fell within the predefined comparability limits of (0.80, 1.25). Similar results were observed for renal clearance of methotrexate and 7-hydroxymethotrexate at the highest dose of rofecoxib tested (50 mg). It was concluded that rofecoxib at doses of 12.5, 25, and 50 mg once daily has no effect on the plasma concentrations or renal clearance (tested at the highest dose of rofecoxib) of methotrexate in rheumatoid arthritis patients. [ABSTRACT FROM AUTHOR]
- Published
- 2001
11. Dose proportionality of oral etoricoxib, a highly selective cyclooxygenase-2 inhibitor, in healthy volunteers.
- Author
-
Agrawal NGB, Porras AG, Matthews CZ, Woolf EJ, Miller JL, Mukhopadhyay S, Neu DC, and Gottesdiener KM
- Abstract
To assess dose proportionality of etoricoxib across the anticipated clinical dose range, a single panel of 12 healthy subjects was administered single oral doses of etoricoxib of 5, 10, 20, 40, and 120 mg in an open, two-part, five-period crossover study. Plasma samples were collected aftereach dose and analyzed for etoricoxib concentrations. The pharmacokinetics of etoricoxib appear to be linear over the entire dose range examined, from 5 to 120 mg. Etoricoxib was found to be well tolerated across the 5 to 120 mg dose range. [ABSTRACT FROM AUTHOR]
- Published
- 2001
12. Effect of food on the pharmacokinetics of extended release diltiazem
- Author
-
Cerchio, K, Seibold, J, Porras, AG, Chiou, R, McNamara, M, Laskin, O, and Polvino, WJ
- Published
- 1995
- Full Text
- View/download PDF
13. D5 - Effect of food on the pharmacokinetics of extended release diltiazem
- Author
-
Cerchio, K, Seibold, J, Porras, AG, Chiou, R, McNamara, M, Laskin, O, and Polvino, WJ
- Published
- 1995
- Full Text
- View/download PDF
14. Positional quality assessment of orthophotos obtained from sensors onboard multi-rotor UAV platforms.
- Author
-
Mesas-Carrascosa FJ, Rumbao IC, Berrocal JA, and Porras AG
- Abstract
In this study we explored the positional quality of orthophotos obtained by an unmanned aerial vehicle (UAV). A multi-rotor UAV was used to obtain images using a vertically mounted digital camera. The flight was processed taking into account the photogrammetry workflow: perform the aerial triangulation, generate a digital surface model, orthorectify individual images and finally obtain a mosaic image or final orthophoto. The UAV orthophotos were assessed with various spatial quality tests used by national mapping agencies (NMAs). Results showed that the orthophotos satisfactorily passed the spatial quality tests and are therefore a useful tool for NMAs in their production flowchart.
- Published
- 2014
- Full Text
- View/download PDF
15. Bioavailability and short-term tolerability of alendronate in glucocorticoid-treated children.
- Author
-
Nakhla M, Denker AE, Connor JD, Carpenter TO, Walson PD, Porras AG, Matthews CZ, Larson P, Freeman A, Wagner JA, and Ward LM
- Subjects
- Administration, Oral, Adolescent, Alendronate adverse effects, Alendronate therapeutic use, Alendronate urine, Biological Availability, Bone Density Conservation Agents administration & dosage, Bone Density Conservation Agents adverse effects, Bone Density Conservation Agents urine, Child, Child, Preschool, Cross-Over Studies, Drug Administration Schedule, Female, Fractures, Bone chemically induced, Glucocorticoids adverse effects, Humans, Injections, Intravenous, Least-Squares Analysis, Male, Middle Aged, Time Factors, Alendronate pharmacokinetics, Bone Density Conservation Agents pharmacokinetics, Fractures, Bone prevention & control, Glucocorticoids therapeutic use
- Abstract
Background: Children receiving glucocorticoids (GCs) are at an increased risk of fragility fractures. Conservative measures may be inadequate in treating low bone mass, giving rise to fractures in this population; as such, attention has turned to the use of bisphosphonates., Objective: The goal of this study was to evaluate the bioavailability and single-dose tolerability of alendronate (ALN) in children receiving a stable dose of GCs., Methods: Children (ages 4-17 years) receiving GC treatment for their chronic illnesses received intravenous (125 μg) and oral (35 mg) ALN in a 2-period, randomized crossover study, with doses separated by at least a 7-day washout period. Urine was collected for either 8 or 24 hours after drug administration to determine urinary excretion of ALN and bioavailability. Tolerability was assessed by continuous collection of adverse events reported during the study. The main outcome measures were total urinary excretion rates, oral bioavailability of ALN, and adverse events., Results: There were 12 patients in the 4- to 11-year-old group (mean age, 8.1 years; 5 girls) and 12 patients in the 12- to 17-year-old group (mean age, 14.3 years; 5 girls). The least-squares mean bioavailability (90% CI) for children aged 4 to 11 years (n = 12) was 0.43% (0.27-0.67) and for children aged 12 to 17 years (n = 12) it was 0.39% (0.26-0.60). The least-squares mean bioavailability for all ages combined was 0.41% (0.30-0.56), with no statistical difference between the 2 age groups. The total urinary excretion of ALN after the intravenous dose was similar between groups. Fifteen patients reported a total of 36 transient clinical nonserious adverse events, all of which were mild or moderate in intensity; the most common were headache (n = 13), abdominal pain (n = 3), limb, neck, or facial pain (n = 6), and ankle or knee swelling (n = 3)., Conclusions: The mean oral bioavailability of ALN was similar to previous pharmacokinetic studies in children with osteogenesis imperfecta and slightly lower than that observed in historical adult controls. Alendronate was generally well tolerated, with minor adverse events that resolved uneventfully. Elucidation of the full adverse-effect profile of this agent was limited by the single-dose nature of this study, and robust comparisons of the pharmacokinetics of ALN in different age groups may need a larger number of patients., (Copyright © 2011 Elsevier HS Journals, Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
16. Pharmacokinetic considerations in determining the terminal elimination half-lives of bisphosphonates.
- Author
-
Lasseter KC, Porras AG, Denker A, Santhanagopal A, and Daifotis A
- Abstract
Background and Objective: Bisphosphonates are commonly used to treat and prevent osteoporosis. These compounds have unusual pharmacokinetic characteristics because they bind strongly to bone, and a portion becomes buried under newly formed bone. Once incorporated into bone tissue, the subsequent release during bone remodeling is probably the rate-limiting step in the terminal elimination of bisphosphonates. Because of this unique property of bisphosphonates, pharmacokinetic studies with insufficient lengths of follow-up might entirely miss the true terminal elimination phase. A terminal half-life (t((1/2)gamma)) of approximately 11 years, similar to that of calcium and other minerals in bone, was reported from an 18-month study of alendronic acid in postmenopausal women with osteoporosis. We are not aware of any other published reports in which the elimination of a bisphosphonate has been followed for more than a few weeks post-dose. The purpose of the present study was to reanalyse the alendronic acid data to examine the effect of truncating the length of follow-up on the calculated t((1/2)gamma)., Patients and Methods: Twenty-one postmenopausal women with osteoporosis (mean age 66 years) received intravenous alendronic acid 30mg over 4 consecutive days (7.5 mg/day), and urinary excretion of alendronic acid was monitored over the following 18-24 months. Terminal elimination half-life was originally calculated by log-linear regression of the percentage retained versus time curve between days 240 and 540 and substituting the slope of the regression line into the equation, t((1/2)gamma) = -log 2/slope. These data were reanalysed based on the period up to 30 days., Results: Data were sufficient for analysis of pharmacokinetics in 11 patients. A mean t((1/2)gamma) of approximately 11 years was reported previously, based on analysis of data between days 240 and 540. Recalculating the 'terminal' half-life of alendronic acid using only data from the first 30 days resulted in an 'observed' half-life of only 11 days., Conclusion: This analysis illustrates the importance of sufficient length of follow-up to accurately characterise the true terminal elimination half-life of bisphosphonates. The relatively short (expressed in units of days rather than years) terminal elimination half-lives reported for some bisphosphonates based on only 30 days of follow-up or less are likely to substantially underestimate the true terminal elimination half-lives.
- Published
- 2005
- Full Text
- View/download PDF
17. Pharmacokinetic evaluation of rofecoxib : comparison of tablet and suspension formulations.
- Author
-
Schwartz JI, Larson PJ, Porras AG, Viswanathan-Aiyer KJ, Agrawal NG, Lasseter KC, Mazenko RS, Merschman SA, and Gertz BJ
- Abstract
Objective: Rofecoxib suspension is a formulation developed to increase the convenience of rofecoxib therapy for patients who have difficulty swallowing tablets. This open-label, two-part study compared the single-dose pharmacokinetics of rofecoxib tablets and rofecoxib suspension in healthy subjects., Design and Study Participants: Part I was a two-period crossover study that assessed the bioequivalence of the 12.5mg/5mL rofecoxib suspension and the 12.5mg rofecoxib tablet in 24 healthy subjects (12 men and 12 women). Part II was a crossover study in 24 additional healthy subjects (12 men and 12 women) that determined the bioequivalence of the rofecoxib 25mg/5mL suspension and the 25mg rofecoxib tablet., Results: No clinically meaningful differences between rofecoxib tablet and suspension were apparent with respect to the rofecoxib area under the concentration-time curve from time zero to infinity (AUC(0-infinity)) and maximum plasma concentration (C(max)), the primary measures of bioequivalence. At the 12.5mg and 25mg doses, the 90% CI for the geometric mean ratio (suspension/tablet) of both AUC(0-infinity) and C(max) fell within the prespecified interval for bioequivalence (0.80-1.25)., Conclusions: The rofecoxib suspension is bioequivalent to the rofecoxib tablet at single oral doses of 12.5mg and 25mg in healthy volunteers. The convenience and ease of administration of rofecoxib suspension may translate into increased compliance with therapy compared with a conventional solid tablet formulation, particularly for elderly patients.
- Published
- 2003
- Full Text
- View/download PDF
18. The disposition and metabolism of rofecoxib, a potent and selective cyclooxygenase-2 inhibitor, in human subjects.
- Author
-
Halpin RA, Porras AG, Geer LA, Davis MR, Cui D, Doss GA, Woolf E, Musson D, Matthews C, Mazenko R, Schwartz JI, Lasseter KC, Vyas KP, and Baillie TA
- Subjects
- Administration, Oral, Bile chemistry, Biotransformation, Chromatography, High Pressure Liquid, Chromatography, Liquid, Cyclooxygenase 2, Cyclooxygenase 2 Inhibitors, Cyclooxygenase Inhibitors metabolism, Cyclooxygenase Inhibitors urine, Dose-Response Relationship, Drug, Feces chemistry, Humans, Lactones metabolism, Lactones urine, Magnetic Resonance Spectroscopy, Mass Spectrometry, Membrane Proteins, Prostaglandin-Endoperoxide Synthases, Sulfones, Tissue Distribution, Cyclooxygenase Inhibitors pharmacokinetics, Isoenzymes antagonists & inhibitors, Lactones pharmacokinetics
- Abstract
The disposition and metabolism of rofecoxib, a selective cyclooxygenase-2 inhibitor, were examined in healthy human subjects and in cholecystectomy patients. After oral administration of [(14)C]rofecoxib (125 mg, 100 micro Ci) to healthy subjects, the mean concentrations of total radioactivity and rofecoxib in plasma as a function of time indicated that the t(max) was achieved at 9 h postdose. After t(max), levels of both radioactivity and rofecoxib decreased in a parallel, exponential fashion (effective t(1/2) approximately equal 17 h). A similar result was obtained after oral administration of [(14)C]rofecoxib (142 mg, 100 micro Ci) to cholecystectomy patients equipped with an L-tube. In healthy subjects, radioactivity was recovered predominantly from the urine (71.5% of dose), with a small amount excreted in feces (14.2%). In patients with an L-tube, half the radioactive dose was recovered in feces, with a lesser amount excreted in urine (28.8%) and a negligible fraction in bile (1.8%). Rofecoxib underwent extensive metabolism in humans, and very little parent drug was recovered unchanged in urine (<1%). Products resulting from both oxidative and reductive pathways were identified by a combination of (1)H NMR and liquid chromatography-tandem mass spectrometry analyses, and included rofecoxib-3',4'-trans-dihydrodiol, 4'-hydroxyrofecoxib-O-beta-D-glucuronide, diastereomeric 5-hydroxyrofecoxib-O-beta-D-glucuronide conjugates, 5-hydroxyrofecoxib, rofecoxib-erythro-3,4-dihydrohydroxy acid, and rofecoxib-threo-3,4-dihydrohydroxy acid. Interconversion of rofecoxib and 5-hydroxyrofecoxib appeared not to be a quantitatively important pathway of rofecoxib disposition in human subjects, in contrast to previous findings in rats.
- Published
- 2002
- Full Text
- View/download PDF
19. Effect of rofecoxib on the pharmacokinetics of digoxin in healthy volunteers.
- Author
-
Schwartz JI, De Smet M, Larson PJ, Verbesselt R, Ebel DL, Lins R, Lens S, Porras AG, and Gertz BJ
- Subjects
- Administration, Oral, Adult, Cardiotonic Agents blood, Cardiotonic Agents urine, Cross-Over Studies, Digoxin blood, Digoxin urine, Double-Blind Method, Drug Interactions, Female, Humans, Male, Middle Aged, Sulfones, Cardiotonic Agents pharmacology, Cyclooxygenase Inhibitors pharmacology, Digoxin pharmacokinetics, Lactones pharmacology
- Abstract
The authors examined the effect of the cyclooxygenase-2 (COX-2) inhibitor, rofecoxib, at steady state on the pharmacokinetics of digoxin following a single dose in healthy subjects. Each healthy subject (N = 10) received rofecoxib (75 mg once daily) or placebo for 11 days in a double-blind, randomized, balanced, two-period crossover study. A single 0.5 mg oral dose of digoxin elixir was administered on the 7th day of each 11-day period. Each treatment period was separated by 14 to 21 days. Samples for plasma and urine immunoreactive digoxin concentrations were collected through 120 hours following the digoxin dose. No statistically significant differences between treatment groups were observed for any of the calculated digoxin pharmacokinetic parameters. For digoxin AUC(0-infinity), AUC(0-24), and Cmax, the geometric mean ratios (90% confidence interval) for (rofecoxib + digoxin/placebo + digoxin) were 1.04 (0.94, 1.14), 1.02 (0.94, 1.09), and 1.00 (0.91, 1.10), respectively. The digoxin median tmax was 0.5 hours for both treatments. The harmonic mean elimination half-life was 45.7 and 43.4 hours for rofecoxib + digoxin and placebo + digoxin treatments, respectively. Digoxin is eliminated renally. The mean (SD) cumulative urinary excretion of immunoreactive digoxin after concurrent treatment with rofecoxib or placebo was 228.2 (+/- 30.8) and 235.1 (+/- 39.1) micrograms/120 hours, respectively. Transient and minor adverse events occurred with similar frequency on placebo and rofecoxib treatments, and no treatment-related pattern was apparent. Rofecoxib did not influence the plasma pharmacokinetics or renal elimination of a single oral dose of digoxin.
- Published
- 2001
- Full Text
- View/download PDF
20. The effect of rofecoxib on the pharmacodynamics and pharmcokinetics of warfarin.
- Author
-
Schwartz JI, Bugianesi KJ, Ebel DL, De Smet M, Haesen R, Larson PJ, Ko A, Verbesselt R, Hunt TL, Lins R, Lens S, Porras AG, Dieck J, Keymeulen B, and Gertz BJ
- Subjects
- Adult, Anticoagulants pharmacokinetics, Cross-Over Studies, Dose-Response Relationship, Drug, Double-Blind Method, Drug Administration Schedule, Drug Interactions, Female, Humans, Male, Prothrombin Time, Sulfones, Warfarin pharmacokinetics, Anticoagulants pharmacology, Cyclooxygenase Inhibitors pharmacology, Lactones pharmacology, Warfarin pharmacology
- Abstract
Objective: The objective of this study was to examine the effect of 3 doses of rofecoxib (12.5, 25, and 50 mg) on the pharmacodynamics and pharmacokinetics of warfarin., Methods: Two single-dose (12.5 or 50 mg of rofecoxib with 25 mg or 30 mg of oral warfarin, respectively, on day 7 of each period) trials (N = 12 men) and 1 steady-state warfarin trial (25 mg rofecoxib; N = 15, 13 men and 2 women) were completed as two-period, randomized, balanced, crossover, double-blind designs. The prothrombin time international normalized ratio (INR) and S(-) and R(+) warfarin enantiomers were assessed during 144 hours after the single warfarin doses. In the steady-state warfarin trial, after the attainment of a stable INR (1.4-1.7), the stable warfarin dose was co-administered with rofecoxib (25 mg) and placebo over two 21-day periods. After the dose of warfarin on day 21, INR and S(-) and R(+) warfarin were assessed during 24 hours., Results: Compared with placebo, rofecoxib slightly increased the INR by approximately 5% (90% confidence interval on the geometric ratio, 1.03, 1.08) and 11% (1.04, 1.19) for the two single-dose warfarin trials with 12.5 and 50 mg of rofecoxib, respectively. In the steady-state warfarin study with 25 mg of rofecoxib, the INR was increased by 8% (1.02, 1.15). Rofecoxib had no significant effect (versus placebo) on the pharmacokinetics of S(-) warfarin. However, in the 3 studies, treatment with 12.5, 25, and 50 mg of rofecoxib was associated with a 27%, 38%, and 40% increase in the area under the plasma concentration-time curve of the biologically less active R(+) warfarin., Conclusions: Rofecoxib increased plasma concentrations of the biologically less active R(+) warfarin, which accounted for a small increase in INR. The approximately 8% increase in INR at steady state with warfarin co-administered with 25 mg of rofecoxib is not likely to be clinically important in most patients taking warfarin. However, standard monitoring of INR values should be conducted when therapy with rofecoxib is initiated or changed, particularly in the first few days, for patients receiving warfarin.
- Published
- 2000
- Full Text
- View/download PDF
21. Grapefruit juice has minimal effects on plasma concentrations of lovastatin-derived 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors.
- Author
-
Rogers JD, Zhao J, Liu L, Amin RD, Gagliano KD, Porras AG, Blum RA, Wilson MF, Stepanavage M, and Vega JM
- Subjects
- Adult, Analysis of Variance, Anticholesteremic Agents administration & dosage, Area Under Curve, Cross-Over Studies, Food-Drug Interactions, Gas Chromatography-Mass Spectrometry, Humans, Hydroxymethylglutaryl-CoA Reductase Inhibitors administration & dosage, Lovastatin administration & dosage, Male, Reference Values, Anticholesteremic Agents blood, Beverages, Citrus, Hydroxymethylglutaryl-CoA Reductase Inhibitors blood, Lovastatin blood
- Abstract
Objective: To evaluate the effect of regular-strength grapefruit juice, a cytochrome P4503A4 (CYP3A4) inhibitor, on the pharmacokinetics of a commonly prescribed regimen of oral lovastatin., Methods: In a randomized crossover study, 16 healthy subjects received a single 40 mg dose of lovastatin in the evening after each consumed an 8-ounce glass of regular-strength grapefruit juice or water with breakfast for 3 consecutive days. The effect of the same grapefruit juice and water regimen on the pharmacokinetics of midazolam (2 mg oral dose given 1 hour after the third day of grapefruit juice and water) was used as a positive control in the same subjects. Plasma concentrations of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors were determined by an enzyme inhibition assay, and concentrations of lovastatin, lovastatin acid, and midazolam were determined by liquid chromatography-tandem mass spectrometry., Results: The area under the plasma concentration-time profiles (AUC) and maximum plasma concentrations (Cmax) of HMG-CoA reductase inhibitors increased slightly (-30% for each) after consumption of grapefruit juice. Similar effects on AUC and Cmax (approximately 40% increase for each) were noted after analysis of samples of hydrolyzed plasma (which converts inactive lactones to active hydroxy acid species). The AUC and Cmax values for lovastatin approximately doubled in the presence of grapefruit juice, whereas the same parameters for lovastatin acid increased 1.6-fold. Grapefruit juice caused the AUC for midazolam to increase by a factor of approximately 2.4., Conclusions: Daily consumption of a glass of regular-strength grapefruit juice has a minimal effect on plasma concentrations of HMG-CoA reductase inhibitors (approximately 30% to 40% increase) after a 40 mg evening dose of lovastatin.
- Published
- 1999
- Full Text
- View/download PDF
22. Pharmacokinetics of alendronate.
- Author
-
Porras AG, Holland SD, and Gertz BJ
- Subjects
- Alendronate pharmacology, Alendronate therapeutic use, Animals, Biological Availability, Bone and Bones metabolism, Female, Humans, Intestinal Absorption, Male, Metabolic Clearance Rate, Tissue Distribution, Alendronate pharmacokinetics, Osteoporosis, Postmenopausal drug therapy
- Abstract
Alendronate (alendronic acid; 4-amino-1-hydroxybutylidene bisphosphonate) has demonstrated effectiveness orally in the treatment and prevention of postmenopausal osteoporosis, corticosteroid-induced osteoporosis and Paget's disease of the bone. Its primary mechanism of action involves the inhibition of osteoclastic bone resorption. The pharmacokinetics and pharmacodynamics of alendronate must be interpreted in the context of its unique properties, which include targeting to the skeleton and incorporation into the skeletal matrix. Preclinically, alendronate is not metabolised in animals and is cleared from the plasma by uptake into bone and elimination via renal excretion. Although soon after administration the drug distributes widely in the body, this transient state is rapidly followed by a nonsaturable redistribution to skeletal tissues. Oral bioavailability is about 0.9 to 1.8%, and food markedly inhibits oral absorption. Removal of the drug from bone reflects the underlying rate of turnover of the skeleton. Renal clearance appears to involve both glomerular filtration and a specialised secretory pathway. Clinically, the pharmacokinetics of alendronate have been characterised almost exclusively based on urinary excretion data because of the extremely low concentrations achieved after oral administration. After intravenous administration of radiolabelled alendronate to women, no metabolites of the drug were detectable and urinary excretion was the sole means of elimination. About 40 to 60% of the dose is retained for a long time in the body, presumably in the skeleton, with no evidence of saturation or influence of one intravenous dose on the pharmacokinetics of subsequent doses. The oral bioavailability of alendronate in the fasted state is about 0.7%, with no significant difference between men and women. Absorption and disposition appear independent of dose. Food substantially reduces the bioavailability of oral alendronate; otherwise, no substantive drug interactions have been identified. The pharmacokinetic properties of alendronate are evident pharmacodynamically. Alendronate treatment results in an early and dose-dependent inhibition of skeletal resorption, which can be followed clinically with biochemical markers, and which ultimately reaches a plateau and is slowly reversible upon discontinuation of the drug. These findings reflect the uptake of the drug into bone, where it exerts its pharmacological activity, and a time course that results from the long residence time in the skeleton. The net result is that alendronate corrects the underlying imbalance in skeletal turnover characteristic of several disease states. In women with postmenopausal osteoporosis, for example, alendronate treatment results in increases in bone mass and a reduction in fracture incidence, including at the hip.
- Published
- 1999
- Full Text
- View/download PDF
23. Pharmacokinetics of intravenous alendronate.
- Author
-
Cocquyt V, Kline WF, Gertz BJ, Van Belle SJ, Holland SD, DeSmet M, Quan H, Vyas KP, Zhang KE, De Grève J, and Porras AG
- Subjects
- Adult, Aged, Alendronate adverse effects, Alendronate urine, Animals, Area Under Curve, Carbon Radioisotopes, Cricetinae, Dose-Response Relationship, Drug, Female, Fever chemically induced, Headache chemically induced, Humans, Infusions, Intravenous, Metabolic Clearance Rate, Middle Aged, Nausea chemically induced, Postmenopause, Alendronate pharmacokinetics
- Abstract
Alendronate is a potent bisphosphonate that has been studied for the treatment of osteoporosis and Paget's disease of the bone. To examine the pharmacokinetics of this drug, several groups of postmenopausal women were dosed intravenously in several studies. Twelve patients with metastatic bone disease were administered an intravenous dose of 10 mg of 14C-labeled alendronate (approximately 26 muCi), and plasma, feces, and urine samples were collected for 72 hours. Radioactivity was excreted almost exclusively in urine, and all of it was accounted for by alendronate. Overall recovery accounted for 47% of dose, with the remainder presumed to be retained in bone. Metabolism of alendronate was not observed. Renal clearance of alendronate was 71 mL/min. An additional 10 subjects were given repeated i.v. administrations of alendronate to demonstrate that previous exposure does not alter the pharmacokinetic behavior of the drug. Examination of the findings from these and other studies in which alendronate was administered intravenously revealed that disposition of single doses is linear in the range of 0.125 to 10 mg. With the possible exception of a somewhat greater skeletal retention of a systemically administered dose, the pharmacokinetics of i.v. alendronate were found to be similar to those of other bisphosphonates.
- Published
- 1999
- Full Text
- View/download PDF
24. Elimination and biochemical responses to intravenous alendronate in postmenopausal osteoporosis.
- Author
-
Khan SA, Kanis JA, Vasikaran S, Kline WF, Matuszewski BK, McCloskey EV, Beneton MN, Gertz BJ, Sciberras DG, Holland SD, Orgee J, Coombes GM, Rogers SR, and Porras AG
- Subjects
- Aged, Alendronate administration & dosage, Alkaline Phosphatase blood, Bone Density drug effects, Calcium urine, Female, Half-Life, Humans, Hydroxyproline urine, Injections, Intravenous, Lumbar Vertebrae, Middle Aged, Osteocalcin blood, Osteoporosis, Postmenopausal blood, Osteoporosis, Postmenopausal urine, Parathyroid Hormone blood, Alendronate pharmacokinetics, Alendronate therapeutic use, Osteoporosis, Postmenopausal drug therapy
- Abstract
Postmenopausal women with established vertebral osteoporosis were studied for 2 years to determine the terminal elimination half-life and the duration of response to treatment with intravenous alendronate (30 mg) given over 4 days. The urinary excretion of alendronate followed a multiexponential decline. Approximately 50% of the total dose was excreted over the first 5 days, and a further 17% was excreted in the succeeding 6 months. Thereafter, there was a much slower elimination phase with an estimated mean terminal half-life of greater than 10 years (n = 11). Urinary excretion of hydroxyproline and calcium decreased significantly from pretreatment values by day 3, reaching a nadir by 1 week (40% and 67% decrease, respectively). Thereafter, hydroxyproline remained suppressed for the following 2 years. In contrast, urinary calcium excretion returned gradually toward pretreatment values over the first year and during the second year was comparable to pretreatment values. Serum activity of alkaline phosphatase activity decreased over 3 months (23% reduction), increased gradually thereafter, and returned to pretreatment values at month 24. Bone mineral density measured at the spine increased by approximately 5% during the first year and remained significantly higher than pretreatment values at 2 years. We conclude that a short course of high doses of intravenous alendronate is associated with a prolonged skeletal retention of the agent. This open study also suggests that this regimen has a sustained effect on bone turnover persisting for at least 1 year.
- Published
- 1997
- Full Text
- View/download PDF
25. Studies of the oral bioavailability of alendronate.
- Author
-
Gertz BJ, Holland SD, Kline WF, Matuszewski BK, Freeman A, Quan H, Lasseter KC, Mucklow JC, and Porras AG
- Subjects
- Achlorhydria chemically induced, Achlorhydria metabolism, Administration, Oral, Adult, Aged, Alendronate, Analysis of Variance, Beverages, Biological Availability, Calcium pharmacology, Cross-Over Studies, Diphosphonates administration & dosage, Diphosphonates urine, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Food, Gastric Mucosa metabolism, Histamine H2 Antagonists pharmacology, Humans, Hydrogen-Ion Concentration, Intestinal Absorption, Male, Middle Aged, Osteoporosis drug therapy, Osteoporosis, Postmenopausal drug therapy, Ranitidine pharmacology, Diphosphonates pharmacokinetics
- Abstract
Clinical studies were performed to examine the oral bioavailability of alendronate (4-amino-1-hydroxy-butylidene-1,1-bisphosphonate monosodium). All studies, with the exception of one performed in men, involved postmenopausal women. Short-term (24 to 36 hours) urinary recovery of alendronate after an intravenous dose of 125 to 250 micrograms averaged about 40% in both men and women. In women, oral bioavailability of alendronate was independent of dose (5 to 80 mg) and averaged (90% confidence interval) 0.76% (0.58, 0.98) when taken with water in the fasting state, followed by a meal 2 hours later. Bioavailability was similar in men [0.59%, (0.43, 0.81)]. Taking alendronate either 60 or 30 minutes before a standardized breakfast reduced bioavailability by 40% relative to the 2-hour wait. Taking alendronate either concurrently with or 2 hours after breakfast drastically (> 85%) impaired availability. Black coffee or orange juice alone, when taken with the drug, also reduced bioavailability (approximately 60%). Increasing gastric pH, by infusion of ranitidine, was associated with a doubling of alendronate bioavailability. A practical dosing recommendation, derived from these findings and reflective of the long-term nature of therapy for a disease such as osteoporosis, is that patients take the drug with water after an overnight fast and at least 30 minutes before any other food or beverage.
- Published
- 1995
- Full Text
- View/download PDF
26. Determination of L-691,121, a new class III antiarrhythmic, and its principal metabolite in plasma by differential radioimmunoassay.
- Author
-
Greber TF, Olah TV, Gilbert JD, Porras AG, and Hichens M
- Subjects
- Animals, Antibody Specificity, Cross Reactions, Humans, Indicators and Reagents, Ligands, Rabbits immunology, Radioimmunoassay, Anti-Arrhythmia Agents blood, Piperidones blood, Spiro Compounds blood
- Abstract
A sensitive and specific method based on radioimmunoassay (RIA) has been developed for the analysis of L-691,121, a new antiarrhythmic agent, and its major metabolite, L-692,199, in plasma. Two RIAs using immunogens and radioligands prepared from different derivatives of L-691,121 were used in conjunction to determine both parent compound and metabolite concentrations by solving simultaneous equations, since neither assay alone was adequately specific. Variable cross-reactivity factors were incorporated into the calculations to correct for non-parallel drug and metabolite displacement curves. The direct assay using 30 microliters of plasma is sensitive to 0.1 ng ml-1 and has sufficient precision, accuracy and specificity for the analysis of clinical samples.
- Published
- 1994
- Full Text
- View/download PDF
27. Clinical pharmacology of alendronate sodium.
- Author
-
Gertz BJ, Holland SD, Kline WF, Matuszewski BK, and Porras AG
- Subjects
- Alendronate, Animals, Bone and Bones metabolism, Diphosphonates administration & dosage, Diphosphonates urine, Female, Half-Life, Humans, Osteoporosis, Postmenopausal urine, Diphosphonates pharmacokinetics, Osteoporosis, Postmenopausal drug therapy
- Abstract
Clinical studies have been performed to investigate the pharmacokinetics and pharmacodynamics of alendronate, an inhibitor of bone resorption for the treatment of osteoporosis. Alendronate is one of the most potent bisphosphonates currently undergoing clinical investigation (> 100-fold more potent than etidronate in vivo). The pharmacokinetics of alendronate are similar to those of other bisphosphonates. After a 2-h intravenous infusion, plasma concentrations of alendronate decline rapidly to approximately 5% of initial values within 6 h. About 50% of a systemic dose is excreted unchanged in the urine in the 72 h following administration. By analogy to its behavior in animals the remainder is assumed to be taken up by the skeleton. After sequestration into bone, the elimination of alendronate is very prolonged. The terminal half-life was estimated to be greater than 10 years. Despite prolonged skeletal residence, the biological effects of alendronate begin to diminish post-treatment, since the duration of effect reflects factors besides dose and cumulative drug exposure. When taken after an overnight fast, 2 h before breakfast, the oral bioavailability of alendronate averages approximately 0.75% of dose with substantial variability (coefficient of variation 55%-75%) both between and within subjects. Reducing the wait before food from 2 h to 1 h, or even 30 min, produces a mean reduction in absorption of 40%. Since the clinical efficacy of alendronate is indistinguishable whether it is given 30 min, 1h, or 3 h before a meal, the observed variability in bioavailability within this range is of little consequence. Dosing up to at least 2 h after a meal dramatically reduces absorption (80%-90%).
- Published
- 1993
- Full Text
- View/download PDF
28. Pharmacokinetics and dose proportionality of D2-agonist MK-458 (HPMC) in parkinsonism.
- Author
-
Cutler NR, Reines SA, McLean LF, Sramek JJ, Porras AG, and Hand EL
- Subjects
- Administration, Oral, Aged, Antiparkinson Agents adverse effects, Antiparkinson Agents blood, Delayed-Action Preparations, Dopamine Agents adverse effects, Dopamine Agents blood, Dose-Response Relationship, Drug, Female, Humans, Lactose adverse effects, Lactose blood, Lactose pharmacokinetics, Male, Methylcellulose adverse effects, Methylcellulose blood, Methylcellulose pharmacokinetics, Middle Aged, Oxazines, Parkinson Disease blood, Tablets, Antiparkinson Agents pharmacokinetics, Dopamine Agents pharmacokinetics, Lactose analogs & derivatives, Methylcellulose analogs & derivatives, Parkinson Disease metabolism
- Abstract
To investigate the pharmacokinetic profile, bioavailability, and dose proportionality of the D2-agonist MK-458 (hydroxypropylmethylcellulose tablet, a sustained release formulation), a 4-period crossover study was conducted in 10 patients with mild to moderate Parkinson's disease (mean age = 63 y; 1 woman, 9 men). Following a titration phase to induce tolerance, each patient was given single oral doses of 6, 12 and 18 mg and a single intravenous 40 micrograms dose (5 micrograms/h over 8h). The maximum concentrations of MK-458 observed in plasma after oral administration were 139, 240 and 344 ng/L for the 6, 12 and 18 mg doses, respectively, and occurred after 8.0, 9.0 and 5.5 h, respectively. Mean areas under the plasma concentration-time curves were 1728, 2849 and 5484 ng/L.h, respectively. The mean plasma half-life was 3.8 h and mean plasma clearance was 3390 ml/min (203.4 L/h). The bioavailability (approximately 5%) was very similar for the 3 tablet formulations tested. The disposition of MK-458 was independent of the dose over the range of doses studied.
- Published
- 1992
- Full Text
- View/download PDF
29. The reaction of reduced xanthine oxidase with oxygen. Kinetics of peroxide and superoxide formation.
- Author
-
Porras AG, Olson JS, and Palmer G
- Subjects
- Animals, Cattle, Cytochrome-c Peroxidase metabolism, Female, Kinetics, Milk enzymology, Oxidation-Reduction, Oxygen, Peroxides, Superoxides, Xanthine Oxidase metabolism
- Abstract
Product formation during the oxidation of xanthine oxidase has been examined directly by using cytochrome c peroxidase as a trapping agent for hydrogen peroxide and the reduction of cytochrome c as a measure of superoxide formation. When fully reduced enzyme is mixed with high concentrations of oxygen, 2 molecules of H2O2/flavin are produced rapidly, while 1 molecule of O2-/flavin is produced rapidly and another produced much more slowly. Time courses for superoxide formation and those for the absorbance changes due to enzyme oxidation were fitted successfully to the mechanism proposed earlier (Olson, J. S., Ballou, D. P., Palmer, G., and Massey, V. (1974) J. Biol. Chem. 249, 4363-4382). In this scheme, each oxidative step is initiated by the very rapid and reversible formation of an oxygen.FADH2 complex (the apparent KD = 2.2 X 10(-4) M at 20 degrees C, pH 8.3). In the cases of 6- and 4-electron-reduced enzyme, 2 electrons are transferred rapidly (ke = 60 s-1) to generate hydrogen peroxide and partially oxidized xanthine oxidase. In the case of the 2-electron-reduced enzyme, only 1 electron is transferred rapidly and superoxide is produced. The remaining electron remains in the iron-sulfur centers and is removed slowly by a second order process (ks = 1 X 10(4) M-1 s-1). When the pH is decreased from 9.9 to 6.2, both the apparent KD for oxygen binding and the rapid rate of electron transfer are decreased about 20-fold. This result is suggestive of uncompetitive inhibition and implies that proton binding to the enzyme-flavin active site affects primarily the rate of electron transfer, not the formation of the initial oxygen complex.
- Published
- 1981
30. Ligand binding to heme proteins. An evaluation of distal effects.
- Author
-
Mims MP, Porras AG, Olson JS, Noble RW, and Peterson JA
- Subjects
- Animals, Binding Sites, Carbon Monoxide metabolism, Hemoglobins metabolism, Humans, Kinetics, Myoglobin metabolism, Oxygen metabolism, Hemeproteins metabolism, Ligands metabolism
- Abstract
The O2, CO, and alkyl isocyanide-binding properties of a variety of vertebrate and invertebrate heme proteins have been compared in detail to those of protoheme mono-3-(1-imidazoyl)-propylamide monomethyl ester in aqueous suspensions of soap micelles. The proteins examined include: cytochrome P-450cam from Pseudomonas putida, beef heart cytochrome c oxidase, yeast cytochrome c peroxidase, alpha and beta subunits of human hemoglobin, sheep hemoglobin, carp hemoglobin, sperm whale myoglobin, horse heart myoglobin, a monomeric hemoglobin from Glycera dibranchiata, erythrocruorin from Chironomusthummii, soybean leghemoglobin, and several hemoglobins that lack distal histidines. The smallest bimolecular rates were observed for cytochrome P-450 containing bound camphor, cytochrome c oxidase, and cytochrome c peroxidase. In the case of P-450, the extremely low isonitrile binding rates (approximately 1 M-1 S-1 at 20 degrees C) are due to steric exclusion by bound camphor molecules. For the oxidase and peroxidase, inhibition of CO and isonitrile binding appears to be due to the polar nature of the active sites. In the cases of animal hemoglobins and myoglobins, the sixth coordination positions appear to be designed to accommodate diatomic molecules with no steric hindrance by distal protein residues. Protein resistance to the diffusion of CO and O2 does not limit the observed association rate constants. In contrast, ligands containing three or more atoms are sterically hindered both in their final bound positions and during diffusion to the active site. The magnitude of this hindrance (greater than or equal to 2 kcal/mol) exhibits a complex dependence on ligand size and shape. The most important protein residue appears to be His E7. In addition to restricting the size of the sixth coordination position, the distal histidine is also capable of forming a hydrogen bond with bound oxygen molecules. The strength of this hydrogen bond was estimated to be -2 and -1 kcal/mol for mammalian myoglobins and hemoglobins, respectively, and accounts for the smaller CO/O2 partition constants (M values) observed for these proteins in comparison to the constants observed for pentacoordinate model heme compounds.
- Published
- 1983
31. The room temperature potentiometry of xanthine oxidase. pH-dependent redox behavior of the flavin, molybdenum, and iron-sulfur centers.
- Author
-
Porras AG and Palmer G
- Subjects
- Animals, Cattle, Female, Hydrogen-Ion Concentration, Kinetics, Milk enzymology, Molybdenum, Oxidation-Reduction, Potentiometry, Temperature, Flavin-Adenine Dinucleotide, Iron-Sulfur Proteins metabolism, Metalloproteins metabolism, Xanthine Oxidase metabolism
- Abstract
A series of potentiometric titrations of xanthine oxidase have been performed at room temperature in the pH range 6.1-9.9. Reduction of the two Fe/S centers was monitored by CD, and that of the FAD and Mo center by EPR. The Fe/S centers behave as centers having a protonable group whose pKa changes with reduction state (E = -344 mV, pKo = 6.4, and pKr = 8.1 for Fe/S I; E = -249 mV, pKo = 6.4, and pKr = 8.0 for Fe/S II). The flavin and the two types of molybdenum centers show varying behavior, but, in all cases, electron addition is accompanied by protonation. The sequence for FAD is reduction, protonation, reduction, protonation with E1 = -398 mV, E2 = -240 mV, pK1 = 9.5, pK2 = 7.4. For "rapid" molybdenum, the sequence is protonation, reduction, protonation, reduction with E1 = -369 mV, E2 = -301 mV, pK1 = 7.9, pK2 = 8.4; and for slow molybdenum, protonation, reduction, protonation with E1 = 320 mV, E2 = -477 mV, pK1 = 7.5, pK2 = 9.5. Comparison to data obtained previously at cryogenic temperatures (Cammack, R., Barber, M. J., and Bray, R. C. (1976) Biochem. J. 157, 469-468 and Barber, M. J., and Seigel, L. M. (1982) in Flavins and Flavoproteins (Massey, V., and Williams, C. H., eds) pp. 796-804, Elsevier/North-Holland, New York) showed the centers to have significant temperature dependence, which calls for a re-evaluation of conclusions reached using cryogenic techniques (e.g. rapid-freeze). The optical absorbance characteristics of the enzyme were also investigated and a possible absorbance for molybdenum was suggested.
- Published
- 1982
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.