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










Publication year range
1.
J Clin Pharmacol ; 41(8): 811-22, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11504268

ABSTRACT

The Food and Drug Administration (FDA) has proposed replacing the 1992 average bioequivalence (ABE) with population and individual bioequivalence (PBE & IBE), as outlined in the preliminary draft guidance of December 1997, which was subsequently replaced by the draft guidances of August 1999 and resolved in the final guidance of October 2000. This has led to considerable public debate among regulatory, academic, and industry experts at numerous conferences (e.g., FDA/AAPS March 1998, FDA/AAPS August-September 1999, FDA Pharmaceutical Sciences Advisory Committee September 1999) and in the literature. The final guidance calls for ABE to remain as the primary criterion by which new formulations may be judged ready for access to the marketplace. In addition, the FDA recommends the use of replicate study designs for the specific drug classes of controlled-release formulations and highly variable drugs. The final guidance also alludes to the possibility of a sponsor requesting alternative criteria such as PBE and IBE following consultation with the FDA. This procedure amounts to a data collection period during which data suitable to evaluate the operating characteristics of PBE and IBE would be generated, analyzed, and discussed among interested parties. A comprehensive review of currently available databases is useful in determining the ultimate value of this data collection period. This report provides an update to the previous publication by the authors. In all, 28 data sets from 20 replicate cross-over bioequivalence studies have been analyzed (n = 12-96) using the statistical methodology in the most recent FDA draft guidance. The results are presented below. ABE Pass: ABE Fail: Total: AUC/Cmax AUC/Cmax AUC/Cmax AUC/Cmax Pass PBE & IBE 20/14 1/3 21/17 Pass IBE only 1/0 0/0 1/0 Fail PBE and IBE 0/2 0/1 0/3 Fail IBE only 2/3 4/5 6/8 Total 23/19 5/9 28/28 Review of the database reveals many interesting features, most notably the lack of consistent results within a given data set across all three criteria. The sensitivity of subject-by-formulation interaction to sample size and inherent variability of the compounds is further explored through simulation studies. It is concluded that additional simulation assessments must be considered when evaluating the value of a data collection period for PBE and IBE assessment. It will be shown that definitive conclusions regarding some of the operating characteristics of PBE and IBE can be achieved through a combination of data-driven hypotheses followed by simulation studies to further evaluate the hypotheses. Some recommendations for further data collection will be made.


Subject(s)
Therapeutic Equivalency , Data Collection , Humans , United States , United States Food and Drug Administration
2.
Eur J Clin Pharmacol ; 57(9): 663-70, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11791897

ABSTRACT

OBJECTIVE: To demonstrate average bioequivalence, the ninety-percent confidence intervals (CI) on the ratio of geometric means for area under the concentration-time curve (AUC) and maximum observed plasma concentration (Cmax) must lie within 0.80-1.25. Demonstration of average bioequivalence (ABE) for highly variable drug products requires large numbers of subjects in a standard, adequately powered, two-period crossover. METHODS: Application of non-traditional study designs can help to meet this hurdle. Study design and analysis for replicate and group sequential-replicate study designs are presented and illustrated using examples. It is demonstrated how to use such approaches to meet the difficult regulatory hurdle of average bioequivalence for a highly variable drug product. RESULTS: To illustrate, data are provided from three separate ABE studies for a highly variable drug product at three dosage strengths. In all three studies, a replicate study design was used to compensate for high intrasubject variation. Additionally, for the last study, a group sequential study design was imposed to provide early evidence of conclusive results. CONCLUSION: Replicate designs and group-sequential designs in bioequivalence should be used to demonstrate average bioequivalence for highly variable drug products or when uncertain of true intrasubject variability in order to ensure conclusive study results.


Subject(s)
Models, Statistical , Research Design , Therapeutic Equivalency , Area Under Curve , Cross-Over Studies , Humans
3.
Stat Med ; 19(20): 2811-20, 2000 Oct 30.
Article in English | MEDLINE | ID: mdl-11033577

ABSTRACT

The FDA has proposed replacing the 1992 average bioequivalence (ABE) with population and individual bioequivalence (PBE and IBE). This has led to considerable public discussion between regulatory, academic and industry experts. At the heart of the discussion has been the relatively modest amount of available data to examine the behaviour of the PBE and IBE criteria. A retrospective analysis of 22 data sets from 15 replicate cross-over bioequivalence studies has been conducted (n=12-74). AUC and C(max) parameters from these studies were analysed using ABE, PBE and IBE methods. Of the 22 data sets for AUC, 19 pass ABE, all pass PBE and 20 pass IBE. Of the three data sets that failed ABE, all passed PBE and one passed IBE. The results for C(max) are more variable. Of the 16 data sets where ABE is demonstrated, one data set failed both PBE and IBE. Of the six data sets that failed ABE, two passed both PBE and IBE, three passed PBE but not IBE and one failed all three criteria. There were five data sets that passed ABE and PBE but not IBE. Additional practical issues involving the behaviour of the new criteria and its expected impact on sample size for highly variable drug products will be presented. The characterization of key parameters and their interrelationships will also be discussed with particular emphasis on the subject by formulation term in the IBE criteria. It is concluded that more studies and simulations are desirable before full-scale implementation of PBE and IBE criteria.


Subject(s)
Models, Statistical , Therapeutic Equivalency , Area Under Curve , Cross-Over Studies , Humans , Research Design , Retrospective Studies , United States , United States Food and Drug Administration
4.
J Clin Pharmacol ; 40(6): 561-70, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10868305

ABSTRACT

The Food and Drug Administration (FDA) issued a second-draft guidance in August 1999 on the subject of in vivo bioequivalence, which is based on the concepts of individual and population bioequivalence (IBE and PBE, respectively). The intention of this guidance is to replace the 1992 guidance that requires that in vivo bioequivalence be demonstrated by average bioequivalence (ABE). Although the concepts of population and individual bioequivalence are intuitively reasonable, a detailed review of the literature has not uncovered clinical evidence to justify the additional burden to the innovator and generic companies as well as the consumer that the new guidelines would impose. The criteria for bioequivalence described in the draft guidance employ aggregate statistics that combine information about differences in bioavailability between formulation means and differences in bioavailability variation of formulations between and within subjects. The purely technical aspects of the statistical approach are reasonably sound. However, PhRMA believes that important operational issues remain that need to be resolved before any changes to current practice are implemented. PhRMA believes that the ideals of prescribability and switchability are intuitively reasonable, but it is uncertain of the extent to which the proposed guidance can achieve these goals. It is not clear whether the attainment of such goals is necessary in the evaluation of bioequivalence given the role this plays in drug development, and the lack of clinical evidence argues against a pressing need to change current practice. PhRMA is concerned that the trade-off offered by the aggregate criteria may ultimately represent more harm than good to the public interest. PhRMA recommends more rigorous evaluation of methods based on two-way crossover designs before moving to methods that require more complex designs. One such method is identified herein and contains procedures for estimating prescribability and switchability. The possibility of a phase-in or trial period to collect replicate crossover data to further evaluate IBE and PBE and possibly allow market access based on these criteria as they are being evaluated has been proposed. PhRMA believes this is unprecedented and will offer little additional information beyond that which can be obtained by simulation or has already been collected by the FDA. Simulation studies have the advantage of allowing evaluation of the sensitivity of various procedures to represent the data patterns as created within the simulation. Operating characteristics by which proposed criteria can be adequately judged have not yet been defined. The limitations of ABE for highly variable drugs and narrow therapeutic drugs are well appreciated and may be addressed by means other than a wholesale change in the current criteria.


Subject(s)
Therapeutic Equivalency , Humans , United States , United States Food and Drug Administration
6.
J Clin Pharmacol ; 39(8): 855-65, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10434239

ABSTRACT

The potential for nonprescription cimetidine (200 mg twice daily) to affect the pharmacokinetics of sustained-release (SR) theophylline was assessed in 26 male subjects, 13 smokers and 13 nonsmokers. This was a concentration-controlled drug interaction study in which the subjects were administered a dose of SR theophylline every 12 hours to provide a mean steady-state concentration between 8 and 15 micrograms/ml. To determine individual theophylline dose, a test dose of aminophylline was administered, and baseline theophylline pharmacokinetics were determined. Subjects remained on SR theophylline for 23 days and were treated in the following sequence: run-in phase (4 days), treatment 1 (7 days), washout (5 days), and treatment 2 (7 days). During the treatment phases, subjects received cimetidine (200 mg at approximately 08:00 and 12:00) or placebo for 7 days in a randomized crossover fashion. Theophylline pharmacokinetics were determined on days 1, 4, and 7 of both treatment phases. A large day-to-day variability in the oral clearance of theophylline was evident for the theophylline-placebo treatment and the theophylline-cimetidine treatment. Nonprescription strength cimetidine resulted in a mean 5% decrease in theophylline oral clearance on day 1 and a mean 12% decrease on days 4 and 7 combined. There were no significant differences in the cimetidine-theophylline interaction between smokers and nonsmokers. Oral clearance during the nighttime dosing interval was 13% greater than the daytime oral clearance for nonsmokers and 22% greater for smokers, showing a greater circadian rhythm for smokers. In summary, nonprescription doses of cimetidine (400 mg/day) have the potential to produce small changes in theophylline concentrations during steady-state dosing with SR theophylline; however, this effect appears less than changes that occur as a consequence of theophylline's intrasubject variability.


Subject(s)
Cimetidine/pharmacology , Enzyme Inhibitors/pharmacology , Theophylline/pharmacokinetics , Vasodilator Agents/pharmacokinetics , Adult , Aged , Area Under Curve , Circadian Rhythm , Cross-Over Studies , Dose-Response Relationship, Drug , Drug Interactions , Humans , Male , Metabolic Clearance Rate , Middle Aged , Single-Blind Method , Smoking , Theophylline/blood
7.
Br J Clin Pharmacol ; 46(3): 267-70, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9764969

ABSTRACT

AIMS: To compare the pharmacokinetics of eprosartan between young (18-45 years) and elderly (65 years) men and between young men and young, premenopausal women (18-45 years). METHODS: Twenty-four subjects (eight subjects/group) received a single 200 mg eprosartan oral dose followed by serial blood sampling over 24 h. RESULTS: Eprosartan was safe and well tolerated. There were no apparent differences in the pharmacokinetics of eprosartan between young females and young males or in the plasma protein binding of eprosartan (98%) for the three groups. On average, AUC (0,infinity) and Cmax values were approximately 2-fold higher in elderly men than young men [AUC (0,infinity) 95% CI: 1.22, 4.34; Cmax 95% CI: 0.98, 4.001. Similarly, unbound AUC (0,infinity) and Cmax values were, on average, approximately 2-fold higher in elderly men than young men [unbound AUC (0,infinity) 95% CI: 1.29, 4.44; unbound Cmax 95% CI: 1.02, 4.12]. tmax was delayed in the elderly men compared with young men, with a median difference of 2.5 h (95% CI: 1.00, 3.01 h). CONCLUSIONS: No gender differences were observed in the pharmacokinetics of eprosartan. There were approximately two fold higher AUC and Cmax values for eprosartan observed in elderly men as compared with young men, most likely due to increased bioavailability of eprosartan in the elderly. Based on the excellent safety profile in the elderly in Phase III clinical trials (doses up to 1200 mg eprosartan) eprosartan can be safely administered to elderly hypertensive patients without an initial dose adjustment. Subsequently, the dose of eprosartan, as for other antihypertensive agents, may be individualized based on tolerability/response.


Subject(s)
Acrylates/pharmacokinetics , Aging/metabolism , Antihypertensive Agents/pharmacokinetics , Imidazoles/pharmacokinetics , Thiophenes , Acrylates/metabolism , Adult , Aged , Antihypertensive Agents/metabolism , Area Under Curve , Female , Half-Life , Humans , Imidazoles/metabolism , Male , Middle Aged , Premenopause , Protein Binding , Sex Factors
8.
Biopharm Drug Dispos ; 19(6): 351-6, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9737815

ABSTRACT

Eighteen healthy males received a single 300 mg oral dose of eprosartan as the commercial wet granulation formulation under fasting conditions and following a high-fat breakfast and a single 20 mg intravenous (i.v.) dose. The pharmacokinetics of i.v. eprosartan (mean +/- S.D.) were characterized by a low systemic plasma clearance (131.8 +/- 36.2 mL min(-1)) and a small steady-state volume of distribution (12.6 +/- 2.6 L). Oral bioavailability averaged 13.1%, due to incomplete absorption. In vitro dynamic flow cell dissolution data showed that pH-dependent aqueous solubility of eprosartan is one factor which limits absorption. Eprosartan terminal half-life was shorter after i.v. (approximately 2 h) versus oral (approximately 5-7 h) administration, which may be due to detection of an additional elimination phase or absorption rate-limited elimination following oral administration. Oral administration of eprosartan following a high-fat meal compared with fasting conditions resulted in a similar extent of absorption (based on AUC), but a decreased absorption rate. Cmax was approximately 25% lower, and a median delay of 1.25 h in time to Cmax was observed when eprosartan was administered with food. These minor changes in exposure are unlikely to be of clinical consequence; therefore, eprosartan may be administered without regard to meal times.


Subject(s)
Acrylates/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Dietary Fats/pharmacology , Food-Drug Interactions , Imidazoles/pharmacokinetics , Thiophenes , Acrylates/administration & dosage , Administration, Oral , Adult , Analysis of Variance , Antihypertensive Agents/administration & dosage , Area Under Curve , Biological Availability , Cross-Over Studies , Dietary Fats/administration & dosage , Half-Life , Humans , Imidazoles/administration & dosage , Injections, Intravenous , Male
9.
J Clin Pharmacol ; 38(7): 649-53, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9702851

ABSTRACT

Eprosartan is an angiotensin II receptor antagonist being developed for the treatment of hypertension and heart failure. The effect of eprosartan on the steady-state anticoagulant activity of warfarin was evaluated in 18 healthy male volunteers. Each subject's daily warfarin dose was titrated over 9 days to achieve a stable international normalized ratio (INR) of 1.3 to 1.6 by day 14. After the 14-day warfarin titration phase, subjects were randomized to receive either eprosartan 300 mg or matching placebo twice a day for 7 days. All subjects continued to take the warfarin dose established during the 14-day titration phase. The anticoagulant activity of warfarin was statistically equivalent when coadministered with eprosartan or with placebo. No serious or unexpected adverse events suggestive of abnormal bleeding occurred during coadministration of eprosartan and warfarin. As measured by the INR, there is no apparent effect of eprosartan on the anticoagulant effect of warfarin.


Subject(s)
Acrylates/pharmacology , Anticoagulants/pharmacology , Antihypertensive Agents/pharmacology , Imidazoles/pharmacology , Thiophenes , Warfarin/pharmacology , Acrylates/adverse effects , Adult , Anticoagulants/adverse effects , Antihypertensive Agents/adverse effects , Double-Blind Method , Drug Interactions , Humans , Imidazoles/adverse effects , International Normalized Ratio , Male , Middle Aged , Warfarin/adverse effects
10.
J Ocul Pharmacol Ther ; 14(3): 203-16, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9671428

ABSTRACT

Recent studies have suggested that intravenous infusion of fenoldopam, a selective dopamine-1 receptor agonist, elevates intraocular pressure (IOP) in man. This study evaluated the effect of intravenous fenoldopam on IOP, aqueous humor outflow facility and gonioscopy in 12 healthy human subjects. Three doses (0.2, 0.5 and 1.0 microg/kg/min) were infused for 120 minutes in a double masked, placebo controlled, four-way crossover design. IOP was measured every 20 minutes in the supine position and every 40 minutes while sitting during the drug and placebo infusions. Tonography and gonioscopy were performed at baseline and after 120 minutes of infusion. Compared to placebo, IOP increased by 3.5 mm Hg (32%) for the lowest dose, 5.8 mm Hg (46%) for the intermediate dose, and 6.9 mm Hg (55%) for the highest dose (p<0.05 for all three doses). IOP returned to baseline within 30 minutes of stopping the infusion. The outflow facility decreased from baseline by 26% after 120 minutes of infusion for all drug doses. In contrast, outflow facility increased from baseline by 11% during placebo infusion. Compared to placebo, the fenoldopam induced changes in outflow were statistically significant (p<0.05). There was no change in the gonioscopic appearance of the anterior chamber angle during the infusion. This study shows that systemic administration of a selective dopamine-1 receptor agonist causes a significant dose-dependent increase in IOP that can be explained in part by diminished outflow facility. These results support a role for the dopamine-1 receptor in the modulation of IOP in general and suggest modulation of aqueous humor outflow by dopaminergic receptors.


Subject(s)
Dopamine Agonists/pharmacology , Fenoldopam/pharmacology , Intraocular Pressure/drug effects , Adult , Analysis of Variance , Aqueous Humor/physiology , Cross-Over Studies , Dopamine Agonists/administration & dosage , Dose-Response Relationship, Drug , Double-Blind Method , Fenoldopam/administration & dosage , Gonioscopy , Humans , Infusions, Intravenous , Male , Posture , Tonometry, Ocular
11.
Clin Pharmacol Ther ; 63(4): 471-81, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9585802

ABSTRACT

STUDY DESIGN: The effects of orally administered eprosartan on changes induced by angiotensin II in blood pressure, renal hemodynamics, and aldosterone secretion were evaluated in healthy men in this double-blind, randomized, single-dose, placebo-controlled crossover study, which was conducted in three parts. Part 1 (n = 12) assessed the onset and duration of the effect of eprosartan 350 mg or placebo; part 2 (n = 14) assessed the dose-response profile of placebo or 10, 30, 50, 70, 100 or 200 mg eprosartan; and part 3 (n = 5) assessed the duration of the effect of 50, 100, or 350 mg eprosartan. RESULTS: In part 1 of the study; 350 mg eprosartan caused complete inhibition of angiotensin II-induced pressor and renal blood flow hemodynamic effects (effects on effective renal plasma flow [ERPF]) and inhibited angiotensin II-induced stimulation of aldosterone secretion from 1 to 3 hours after administration. Eprosartan, 350 mg, inhibited the effects of exogenous angiotensin II by approximately 50% to 70% from 12 to 15 hours after dosing. Eprosartan had no angiotensin II agonistic activity and produced an increase in ERPF starting at 1 to 4 hours after dosing. In study part 2, at 3 hours after single doses of 10, 30, 50, 70, 100, and 200 mg, eprosartan inhibited angiotensin 11-induced decreases in ERPF by 39.1%, 49.9%, 33.0%, 56.0%, 71.0%, and 85.7%, respectively, compared with placebo. In study part 3, 50, 100, and 350 mg eprosartan produced measurable Inhibition of angiotensin II-induced decreases in ERPF from 12 to 15 hours after administration. In parts 2 and 3, the eprosartan angiotensin II antagonism on blood pressure response and aldosterone secretion mirrored the angiotensin II antagonism on ERPF.


Subject(s)
Acrylates/pharmacology , Aldosterone/metabolism , Angiotensin Receptor Antagonists , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Imidazoles/pharmacology , Renal Circulation/drug effects , Thiophenes , Acrylates/administration & dosage , Adult , Aldosterone/blood , Antihypertensive Agents/administration & dosage , Dose-Response Relationship, Drug , Humans , Imidazoles/administration & dosage , Male , Potassium, Dietary/administration & dosage , Reference Values , Sodium, Dietary/administration & dosage , Time Factors
12.
J Clin Pharmacol ; 38(2): 129-37, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9549643

ABSTRACT

This was an open-label, parallel group study to compare the pharmacokinetics of multiple oral doses of eprosartan in subjects with normal renal function (Clcr > 80 mL/min; n = 8) and patients with mild (Clcr 60-80 mL/min; n = 8), moderate (Clcr 30-59 mL/min; n = 15), or severe (Clcr < 30 mL/min; n = 3) renal insufficiency. Each subject received oral eprosartan 200 mg twice daily for 6 days and a single dose on day 7. Mean total maximum concentration (Cmax) and area under the concentration-time curve from 0 to 12 hours (AUC0-12) were similar for healthy subjects and those with mild renal impairment, but were an average of 25% to 35% and 51% to 55% greater for patients with moderate and severe renal impairment, respectively, compared with healthy subjects. Mean renal clearance (Clr), which was similar for healthy subjects and patients with mild renal impairment, was decreased an average of 41% and 95% in the groups with moderate and severe renal impairment, respectively, compared with normal subjects. Eprosartan was highly bound to plasma proteins in all groups; however, the unbound fraction was increased approximately two-fold in the group with severe renal impairment. Mean unbound Cmax and AUC0-12 were an average of 53% to 61% and 185% to 210% greater for the patients with moderate and severe renal impairment, respectively, compared with healthy subjects. Headache was the most common adverse experience reported in all subgroups. Eprosartan was safe and well tolerated regardless of degree of renal impairment. Cmax and AUC were increased and renal clearance decreased in patients with moderate to severe renal impairment in comparison to healthy subjects and patients with mild renal impairment. However, based on the moderate renal clearance and known safety profile of eprosartan, it is not necessary to adjust the dose of eprosartan in patients with renal insufficiency.


Subject(s)
Acrylates/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Imidazoles/pharmacokinetics , Renal Insufficiency/metabolism , Thiophenes , Acrylates/administration & dosage , Acrylates/blood , Adult , Aged , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/blood , Area Under Curve , Humans , Imidazoles/administration & dosage , Imidazoles/blood , Metabolic Clearance Rate , Middle Aged , Renal Insufficiency/blood
13.
Am J Health Syst Pharm ; 54(10): 1172-6, 1997 May 15.
Article in English | MEDLINE | ID: mdl-9161624

ABSTRACT

The cardiovascular effects of granisetron given as a 30-second i.v. bolus dose and of granisetron and ondansetron given by currently recommended methods were studied. Healthy adults 18 to 50 years of age were randomly assigned to one of four treatments during each of four study periods: granisetron 10 micrograms/kg (as the hydrochloride salt) i.v. over 5 minutes, granisetron 10 micrograms/kg i.v. over 30 seconds, ondansetron 32 mg (as the hydrochloride salt) i.v. over 15 minutes, and placebo. During each study period, the researchers gave each subject three sequential injections using a double-blind, double-dummy technique. Each subject was to receive all four regimens. Two resting 12-lead electrocardiograms (ECGs) were obtained before the regimen, and one was obtained at the end of each injection and at intervals up to 24 hours after the third injection. Sitting blood pressure and pulse were measured before treatments, immediately after the end of each injection, and at intervals up to 24 hours after injection 3. Safety data were analyzed for 13 subjects, and ECG interval data for 12 of them. The mean postdose QTc interval differed significantly among regimens. There were no other significant regimen-associated differences among the four mean results for any ECG interval. The mean post-dose QTc interval for ondansetron was significantly greater than that for each of the other regimens. The drug regimens were comparable in safety and tolerability. A total of 20 adverse effects, all mild to moderate, were reported in 10 subjects. Changes in vital signs were minimal. There were no clinically important cardiovascular changes associated with the i.v. administration of granisetron 10 micrograms/kg over 30 seconds, granisetron 10 micrograms/kg over 5 minutes, or ondansetron 32 mg over 15 minutes in healthy adults.


Subject(s)
Antiemetics/administration & dosage , Granisetron/administration & dosage , Heart/drug effects , Ondansetron/pharmacology , Adult , Antiemetics/adverse effects , Double-Blind Method , Drug Interactions , Electrocardiography/drug effects , Female , Granisetron/adverse effects , Headache/chemically induced , Heart/physiology , Humans , Infusions, Intravenous , Injections, Intravenous , Male , Ondansetron/adverse effects
14.
J Clin Pharmacol ; 37(4): 312-20, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9115057

ABSTRACT

Intravenous fenoldopam, a selective dopamine-1 receptor agonist, was compared with placebo in this randomized, double-blind, two-period crossover study to evaluate its effects on intraocular pressure, aqueous dynamics, and macular blood flow in patients with elevated intraocular pressure or primary open-angle glaucoma. Doses of fenoldopam were titrated up to a maximum of 0.5 microgram/kg/min. Intraocular pressure, measured by pneumotonometry, was the primary outcome variable. Other outcomes included macular blood flow assessed by blue field examination, visual field examined by automated perimetry, aqueous outflow facility measured by tonography, and aqueous humor production determined by fluorophotometry. During infusions of fenoldopam, intraocular pressure increased from a mean baseline level of 29.2 mmHg to a mean maximum level of 35.7 mmHg. During the placebo infusions, pressure increased from a mean baseline of 28.4 mmHg to a mean of 29.0 mmHg at the time point that corresponded to the mean maximum intraocular pressure on the day intravenous fenoldopam was administered, to yield a mean difference in pressure between study days of 6.7 mmHg (P < 0.05). There were no apparent changes in macular blood flow, visual fields, or production or outflow of aqueous humor associated with fenoldopam infusion. The increase in intraocular pressure seen in this population of patients with ocular hypertension during infusions of fenoldopam is consistent with fenoldopam-associated increases in intraocular pressure reported in previous studies of healthy volunteers and of patients with accelerated systemic hypertension. These results further suggest that dopamine-1 receptors play a role in the regulation of intraocular pressure.


Subject(s)
Fenoldopam/therapeutic use , Intraocular Pressure/drug effects , Ocular Hypertension/drug therapy , Vasodilator Agents/therapeutic use , Adult , Aged , Cross-Over Studies , Double-Blind Method , Female , Fenoldopam/adverse effects , Glaucoma, Open-Angle/drug therapy , Headache/chemically induced , Humans , Male , Middle Aged , Retinal Vessels/drug effects , Vasodilator Agents/adverse effects
15.
J Clin Pharmacol ; 36(8): 732-9, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8877678

ABSTRACT

To assess the pharmacokinetics of testosterone after application of one, two, or three testosterone transdermal delivery systems to hypogonadal patients, 12 hypogonadal men (mean age 46.6 +/- 10.5 years) were enrolled in an open-label, randomized, crossover study. Each application period comprised 4 days: a 2-day washout period with no exogenous testosterone therapy followed by 2 days of therapy with one, two, or three transdermal systems applied daily to the patient's back. On day 4 of each period, serial blood samples were collected for determination of total and non-sex hormone binding globulin (non-SHBG) bound serum testosterone concentrations. Serum concentrations of testosterone were determined using validated radioimmunoassay methods. Residual testosterone analysis of used transdermal systems was used to estimate testosterone delivery through the skin. In general, serum concentrations of testosterone rose in accordance with an increase in dose. Using a strict bioequivalence approach to dose proportionality, the increases in area under the concentration-time curve (AUC) and morning concentrations were proportional to the increase in dose from two to three transdermal systems, but somewhat less than proportional with an increase from one to two transdermal systems. Results from the non-SHBG bound serum testosterone concentrations closely paralleled those of total serum testosterone. Use of three transdermal systems yielded serum concentrations of testosterone that tended to be above the upper limit of the normal range. The AUC and cumulative release of testosterone were linearly related to the number of applied systems. If necessary, the standard recommended dose of two testosterone transdermal delivery systems can be modified to accommodate interindividual differences in testosterone requirements of hypogonadal men.


Subject(s)
Hypogonadism/metabolism , Testosterone/pharmacokinetics , Administration, Cutaneous , Adult , Area Under Curve , Dose-Response Relationship, Drug , Humans , Hypogonadism/drug therapy , Male , Metabolic Clearance Rate , Middle Aged , Testosterone/administration & dosage
16.
Biopharm Drug Dispos ; 17(3): 249-58, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8983399

ABSTRACT

The objective of the current investigation was to describe the pharmacokinetics and absolute oral bioavailability of epristeride. Twelve healthy male subjects (mean (SD) age, 27 (6.2) years) received a single oral dose of 5 mg and an intravenous infusion of 4.5 mg over 30 min in a crossover fashion. Blood samples were obtained over 72h for the determination of epristeride plasma concentrations using a sensitive high-performance liquid chromatography assay. The lower limit of quantification was 5 ng mL-1. Pharmacokinetic analysis of the plasma concentration-time data was performed by both non-compartmental and compartmental methods. Absolute bioavailability was determined using dose-normalized AUC values following oral and intravenous administration. Epristeride plasma concentrations declined in a biexponential fashion with secondary peaks evident around 24 h in a majority of subjects following both routes of administration. Maximal plasma concentrations were typically achieved approximately 4 h after oral dosing. The mean apparent terminal elimination half-life estimates were similar following intravenous and oral administration and were 27.3 and 26.2 h, respectively. The mean plasma clearance and steady-state volume of distribution were 0.33 (0.09) mL min-1 kg-1 and 0.54 (0.17) L kg-1, respectively. The mean absolute bioavailability was 93% (95% CI: 84%, 104%). Following compartmental analysis of the intravenous data, the mean (SD) lambda 1 and lambda 2 half-life estimates were 2.74 (0.48) and 31.8 (19.5) h, respectively. The % AUC associated with the lambda 2 exponential phase was approximately 68%. This long half-life allows for once-daily dosing of epristeride.


Subject(s)
Androstadienes/pharmacokinetics , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/administration & dosage , 5-alpha Reductase Inhibitors , Administration, Oral , Adult , Androstadienes/administration & dosage , Androstadienes/blood , Biological Availability , Cross-Over Studies , Humans , Infusions, Intravenous , Male
17.
J Clin Pharmacol ; 34(12): 1199-207, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7738216

ABSTRACT

The pharmacokinetic profile of penciclovir was determined after a single 500-mg dose of its oral precursor, famciclovir, in 9 healthy volunteers and in 14 patients with chronic hepatic disease. Plasma and urine samples were analyzed for concentrations of penciclovir and 6-deoxy-penciclovir using a reverse-phase high-performance liquid chromatography (HPLC) method. Famciclovir was not quantifiable in patients with hepatic disease, and 6-deoxy-penciclovir was quantifiable in only a limited number of specimens. The extent of systemic availability of penciclovir, as measured by AUC0-infinity, was similar in patients with hepatic disease and in healthy subjects. In contrast, Cmax was significantly lower (average decrease of 43%) in subjects with hepatic disease relative to healthy normal subjects. Median Tmax for subjects with hepatic disease was significantly increased (by 0.75 hours) compared with subjects with normal liver function. These data suggest a decrease in the rate, but not the extent, of systemic availability of penciclovir in patients with hepatic disease. It should be unnecessary to modify the dose of famciclovir for subjects with compensated hepatic disease and normal renal function.


Subject(s)
2-Aminopurine/analogs & derivatives , Acyclovir/analogs & derivatives , Liver Diseases/metabolism , Prodrugs/pharmacokinetics , 2-Aminopurine/administration & dosage , 2-Aminopurine/pharmacokinetics , Acyclovir/blood , Acyclovir/pharmacokinetics , Acyclovir/urine , Administration, Oral , Adolescent , Adult , Biological Availability , Chromatography, High Pressure Liquid , Chronic Disease , Famciclovir , Female , Guanine , Humans , Male , Metabolic Clearance Rate , Middle Aged , Prodrugs/administration & dosage
18.
J Clin Pharmacol ; 34(11): 1098-108, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7876402

ABSTRACT

Nonsteroidal antiinflammatory drugs differ with respect to their effects on prostaglandin metabolism in various tissues, a property that may be partly responsible for some of the differences in the pharmacologic activities and side-effect profiles that are associated with their use. The effects of nabumetone on urinary prostaglandin excretion have not been reported. Fourteen healthy females, age 21-43 years, were treated with nabumetone (NAB) 1000 mg daily, sulindac (SUL) 200 mg every 12 hours, and indomethacin (IND) 50 mg every 12 hours for 7 days in a randomized period-balanced crossover study. The effects of drug treatment on urinary prostaglandin excretion (PGE2, 6-keto-PGF1 alpha, PGF2 alpha, thromboxane [TX] B2) and platelet function (collagen-induced whole blood platelet aggregation [CIPA] and template bleeding time) were determined on day 1 and day 7. For each treatment regimen, mean baseline urinary PG excretion values were comparable for each prostanoid, but the pattern of excretion differed in response to each drug. Treatment with NAB significantly increased the urinary excretion rates of PGE2 and PGF2 alpha, but 6-keto-PGF1 alpha and TXB2 excretion were unchanged. IND treatment did not result in a significant change in PGE2 excretion but did significantly reduce urinary 6-keto-PGF1 alpha and TXB2 excretion rates. Reduced excretion of PGF2 alpha was observed on both study days during treatment with IND and SUL. SUL treatment also resulted in increased urinary PGE2 excretion while significantly reducing 6-keto-PGF1 alpha excretion on day 7. Significant differences were observed between the NAB and SUL regimens with respect to PGF2 alpha excretion and between the NAB and SUL regimens for PGE2, PGF2 alpha, 6-keto-PGF alpha 1 (on day 1 only) and TXB2 (on day 1 only). Neither NAB nor SUL caused inhibition of CIPA or bleeding time although platelet aggregation was inhibited during IND treatment. That NAB treatment was neither associated with alterations in platelet function nor decreases in the urinary excretion of the vasodilatory prostaglandins, PGE2 and 6-keto-PGF1 alpha, suggests that NAB possesses renal sparing properties.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blood Platelets/drug effects , Butanones/pharmacology , Indomethacin/pharmacology , Prostaglandins/urine , Sulindac/pharmacology , Adult , Bleeding Time , Butanones/pharmacokinetics , Cross-Over Studies , Female , Humans , Nabumetone , Platelet Aggregation/drug effects
19.
Thromb Haemost ; 72(4): 622-6, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7878642

ABSTRACT

SKF 107260 is a potent pentapeptide antagonist of the platelet membrane glycoprotein receptor GP IIb/IIIa. The in vitro platelet inhibitory effects of SKF 107260, acetylsalicylic acid (ASA), and their combination, on collagen-induced platelet aggregation and secretion (ATP release) were assessed in human whole blood. Additionally, the concentration-response relationships for these inhibitors were compared for males and females in order to explore gender differences in platelet responsiveness. SKF 107260 caused a concentration-dependent inhibition of platelet aggregation which was significant at concentrations > or = 30 nM. ASA also caused a concentration-dependent inhibition of platelet aggregation which was significant at concentrations > or = 1 mg/dl. The addition of ASA 1 mg/dl to increasing concentrations of SKF 107260 resulted in a more pronounced inhibition of platelet aggregation than when either agent was used alone. These data suggest a pharmacologic interaction, especially at SKF 107260 concentrations < or = 30 nM. Since ATP release was significantly inhibited at concentrations > or = 1 nM, platelet secretion appears to be more sensitive than aggregation to inhibition by SKF 107260. These data suggest that platelet secretion in response to collagen is dependent on the aggregation response mediated by GP IIb/IIIa. In conclusion, SKF 107260 is a potent inhibitor of both whole blood platelet aggregation and secretion and these anti-aggregatory effects may be augmented by concomitant ASA administration.


Subject(s)
Adenosine Triphosphate/metabolism , Aspirin/pharmacology , Blood Platelets/metabolism , Peptides, Cyclic/pharmacology , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/antagonists & inhibitors , Adenosine Triphosphate/blood , Adult , Blood Platelets/drug effects , Collagen/pharmacology , Drug Synergism , Female , Humans , Male , Sex Characteristics
20.
Clin Pharmacol Ther ; 55(4): 418-26, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8162668

ABSTRACT

OBJECTIVE: To characterize the pharmacokinetics of a single 500 mg oral dose of famciclovir in subjects with varying degrees of renal impairment. METHODS: Twenty-seven subjects were enrolled in an open-label parallel-group study. Eighteen patients had renal impairment (average age [ +/- SD], 49 +/- 12 years), and nine subjects were healthy volunteers (average age, 28 +/- 7 years). Patients with renal impairment were stratified into groups based on estimated creatinine clearance (CLCR): mild impairment (CLCR, 60 to 80 ml/min/1.73 m2), moderate impairment (CLCR, 30 to 59 ml/min/1.73 m2) and severe impairment (CLCR, 5 to 29 ml/min/1.73 m2). Plasma and urine specimens were analyzed for concentrations of penciclovir, the antivirally active metabolite of famciclovir, by reverse-phase HPLC. Plasma data were analyzed with use of model-independent methods. RESULTS: In subjects with normal renal function (CLCR > 80), the mean maximum plasma concentrations of penciclovir was 2.83 micrograms/ml (range, 1.30 to 3.82 micrograms/ml) and the mean time to reach maximum concentration was 0.89 hours (range, 1/2 to 1 1/2 hours). The mean apparent terminal elimination half-life was 2.15 hours (range, 1.56 to 2.87 hours). A linear relationship was observed between the plasma elimination rate constant and CLCR and between renal clearance and CLCR. Mean area under the plasma concentration-time curve from zero to infinity was approximately tenfold higher and the plasma elimination rate constant was approximately fourfold lower in patients with severe renal impairment than in subjects with normal renal function. CONCLUSION: Consideration should be given to modification of the dosing schedule of famciclovir from the usual 8-hour interval to a 12-hour interval for patients with moderate renal impairment (CLCR 30 to 59 ml/min/1.73 m2) or a 24-hour interval for patients with severe renal impairment (CLCR < 30 ml/min/1.73 m2).


Subject(s)
2-Aminopurine/analogs & derivatives , Kidney Diseases/metabolism , Prodrugs/pharmacokinetics , 2-Aminopurine/administration & dosage , 2-Aminopurine/blood , 2-Aminopurine/pharmacokinetics , 2-Aminopurine/urine , Administration, Oral , Adult , Aged , Chromatography, High Pressure Liquid , Creatinine/blood , Creatinine/urine , Famciclovir , Female , Humans , Male , Middle Aged , Regression Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...