Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Eur J Clin Pharmacol ; 59(1): 51-6, 2003 May.
Article in English | MEDLINE | ID: mdl-12682802

ABSTRACT

RATIONALE OBJECTIVE: To examine in vivo the effect of erythromycin on the pharmacokinetics of rosuvastatin [an inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase]. Erythromycin is a potent inhibitor of CYP3A4 that markedly increases circulating levels of some other HMG-CoA reductase inhibitors. METHODS: In this randomised, double-blind, two-way cross-over, placebo-controlled trial 14 healthy volunteers were given 500 mg erythromycin or placebo four times daily for 7 days. A single dose of 80 mg rosuvastatin was co-administered on day 4 of dosing. Plasma concentrations of rosuvastatin and active and total HMG-CoA reductase inhibitors were measured up to 96 h after dosing. RESULTS: Eleven volunteers had data available from both dosing periods. There was no increase in rosuvastatin plasma exposure following co-administration with erythromycin compared to placebo. In fact, following co-administration with erythromycin, rosuvastatin geometric least square mean AUC((0-t)) and C(max) were 20% and 31%, respectively, lower than with placebo. Individual treatment ratios for AUC((0-t)) ranged from 0.48 to 1.17, and for C(max) ranged from 0.33 to 2.19. Essentially all of the circulating active HMG-CoA reductase inhibitors and most (>94%) of the total inhibitors were accounted for by rosuvastatin. Erythromycin did not affect the proportion of circulating active or total inhibitors accounted for by circulating rosuvastatin. CONCLUSIONS: Erythromycin did not produce any increase in rosuvastatin plasma exposure. This indicates that CYP3A4 metabolism is not an important clearance mechanism for rosuvastatin, a result consistent with previous findings. The small decreases in rosuvastatin AUC((0-t)) and C(max) that occurred as a consequence of short-term treatment with erythromycin are unlikely to have relevance to long-term treatment with rosuvastatin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Erythromycin/pharmacology , Fluorobenzenes/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Pyrimidines , Sulfonamides , Adult , Area Under Curve , Cross-Over Studies , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Double-Blind Method , Drug Interactions , Half-Life , Humans , Male , Metabolic Clearance Rate , Rosuvastatin Calcium
2.
Eur J Clin Pharmacol ; 58(10): 669-75, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12610743

ABSTRACT

OBJECTIVE: To assess the effect of chronic hepatic impairment on rosuvastatin disposition, pharmacodynamic activity and tolerability. METHODS: This was an open-label, non-randomised, parallel-group trial. Six subjects were enrolled in each of three hepatic-function strata: Child-Pugh class A (CP-A, mild impairment), Child-Pugh class B (CP-B, moderate impairment) and normal hepatic function; the latter two strata were age, weight, race, sex and smoking history matched. All subjects were given rosuvastatin 10 mg for 14 days. RESULTS: In subjects with CP-A, and in four of six subjects with CP-B, rosuvastatin steady-state AUC(0-24) and C(max) were similar to subjects with normal hepatic function (geometric mean values 60.7 ng h/ml and 6.02 ng/ml, respectively). Two of six subjects with CP-B who had the highest CP scores (i.e. the highest degrees of hepatic impairment) had the highest AUC(0-24) (128 ng h/ml and 242 ng h/ml) and C(max) (23.4 ng/ml and 96.7 ng/ml) values. Low-density lipoprotein cholesterol (LDL-C) was decreased in all strata, but the response was more variable in the CP-B group. Rosuvastatin was well tolerated, and the safety profile was similar in subjects with hepatic impairment and normal hepatic function. CONCLUSION: In most subjects with mild-to-moderate hepatic impairment, the steady-state pharmacokinetics of rosuvastatin were similar to subjects with normal hepatic function (more extensive hepatic impairment may increase systemic exposure to rosuvastatin), and most had LDL-C reductions similar to subjects with normal hepatic function.


Subject(s)
Fluorobenzenes/pharmacology , Fluorobenzenes/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Liver Diseases/physiopathology , Pyrimidines , Sulfonamides , Adult , Area Under Curve , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Female , Fluorobenzenes/adverse effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Liver Diseases/etiology , Liver Diseases/metabolism , Liver Function Tests , Male , Middle Aged , Rosuvastatin Calcium
3.
Eur J Clin Pharmacol ; 58(8): 527-31, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12451430

ABSTRACT

OBJECTIVE: To examine the effect of fluconazole, a potent inhibitor of CYP2C9 and CYP2C19, on the pharmacokinetics of rosuvastatin in healthy volunteers. Significantly increased plasma concentrations of fluvastatin have been observed following co-administration with fluconazole. METHODS: This was a randomised, double-blind, two-way crossover, placebo-controlled trial. Healthy male volunteers ( n=14) were given fluconazole 200 mg or matching placebo once daily for 11 days; rosuvastatin 80 mg was co-administered on day 8 of dosing. Plasma concentrations of rosuvastatin, N-desmethyl rosuvastatin, and active and total 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors were measured up to 96 h post-dose. RESULTS: Following co-administration with fluconazole, rosuvastatin geometric least-square mean area under the plasma concentration-time curve (AUC(0-t)) and peak plasma concentration (C(max)) were increased by 14% and 9%, respectively, compared with placebo (90% confidence intervals for the treatment ratios: 0.967 to 1.341 and 0.874 to 1.355, respectively). Individual treatment ratios for AUC(0-t) ranged from 0.59 to 2.23, and for C(max) ranged from 0.52 to 2.28. The limited data available for the N-desmethyl metabolite show that geometric mean C(max) was decreased by approximately 25% compared with placebo. Rosuvastatin accounted for essentially all of the circulating active HMG-CoA reductase inhibitors and most (>90%) of the total inhibitors. Fluconazole did not affect the proportion of circulating active or total inhibitors accounted for by circulating rosuvastatin. CONCLUSIONS: Fluconazole produced only small increases in rosuvastatin AUC(0-t) and C(max), which were not considered to be of clinical relevance. The results support previous in-vitro findings that CYP2C9 and CYP2C19 metabolism is not an important clearance mechanism for rosuvastatin.


Subject(s)
Antifungal Agents/pharmacology , Fluconazole/pharmacology , Fluorobenzenes/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Pyrimidines , Sulfonamides , Administration, Oral , Adult , Antifungal Agents/administration & dosage , Area Under Curve , Cross-Over Studies , Double-Blind Method , Drug Interactions , Fluconazole/administration & dosage , Fluorobenzenes/administration & dosage , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Male , Middle Aged , Rosuvastatin Calcium
4.
J Clin Psychiatry ; 57(5): 204-6, 1996 May.
Article in English | MEDLINE | ID: mdl-8626351

ABSTRACT

BACKGROUND: Valproic acid is added to the lithium regimens of many patients with bipolar disorder, especially those with mania refractory to lithium treatment. METHOD: We evaluated the pharmacokinetic effects and safety of coadministration of lithium and valproate in 16 healthy volunteers in this randomized, placebo-controlled, two-period (12 days each) crossover trial. Valproate or placebo was given twice daily. On Days 6-10, lithium was added. Blood samples drawn on Days 5 and 10 were analyzed for valproate by high-pressure liquid chromatography (HPLC) and for lithium by atomic absorption spectrophotometry. RESULTS: Lithium pharmacokinetics were unchanged by valproate, but valproate C(max), C(min), and AUC rose slightly during lithium coadministration. Adverse events did not change significantly. CONCLUSION: Concomitant administration of lithium and valproate appears to be safe in patients with bipolar disorder.


Subject(s)
Lithium/adverse effects , Lithium/pharmacokinetics , Valproic Acid/adverse effects , Valproic Acid/pharmacokinetics , Adult , Bipolar Disorder/drug therapy , Chromatography, High Pressure Liquid , Cross-Over Studies , Drug Interactions , Drug Therapy, Combination , Headache/chemically induced , Humans , Lithium/blood , Male , Placebos , Spectrophotometry, Atomic , Valproic Acid/blood
5.
Antimicrob Agents Chemother ; 39(9): 2078-83, 1995 Sep.
Article in English | MEDLINE | ID: mdl-8540719

ABSTRACT

This study was conducted to determine (i) the effect of omeprazole on steady-state concentrations of clarithromycin and 14-(R)-hydroxyclarithromycin in plasma and gastric mucosa, (ii) the effect of clarithromycin on steady-state concentrations of omeprazole in plasma, and (iii) the effect of clarithromycin on the suppression of gastric acid secretion by omeprazole. Twenty healthy, Helicobacter pylori-negative male subjects completed this three-period, double-blind, randomized crossover study. In period 1, all subjects received 40 mg of omeprazole each morning for 6 days. Twenty-four-hour gastric pH monitoring took place on days -1 and 6. Pharmacokinetic sampling took place on day 6. In periods 2 and 3, subjects were randomly assigned to receive either 40 mg of omeprazole or omeprazole placebo daily for 6 days plus clarithromycin (500 mg) every 8 h for 5 days with a single 500-mg dose on day 6. Gastric tissue and mucus samples were obtained via endoscopy on day 5. Gastric pH monitoring and pharmacokinetic sampling took place on day 6. Two-week washout intervals separated the three study periods. Clarithromycin increased mean omeprazole area under the concentration-time curve from 0 to 24 h from 3.3 +/- 2.0 to 6.3 +/- 4.5 micrograms.h/ml (P < 0.05) and harmonic mean half-life from 1.2 to 1.6 h (P < 0.05) but did not significantly alter the effect of omeprazole on gastric pH. Mean clarithromycin area under the concentration-time curve from 0 to 8 h increased from 22.9 +/- 5.5 (placebo) to 26.4 +/- 5.7 micrograms.h/ml (omeprazole) (P < 0.05) when clarithromycin was administered with omeprazole. Analysis of variance revealed that mean concentrations of clarithromycin in tissue and mucus were statistically significantly higher when clarithromycin was given with omeprazole than when clarithromycin was given with placebo (P <0.001). Mean maximum observed concentrations of clarithromycin in the gastric fundus increased from 20.8 +/- 7.6 (placebo) to 24.3 +/- 6.4 micrograms/g (omeprazole), and those in the gastric mucous from 4.2 +/- 7.7 placebo to 39.3 +/- 32.8 micrograms/g (omeprazole). Similar increases were observed for the 14-(R)-hydroxyclarithromycin. These results show that omeprazole increases concentrations of clarithromycin in gastric tissue and mucus and may provide a mechanism for synergy between clarithromycin ad omeprazole that explains the excellent eradication of H. pylori seen in clinical trials.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Anti-Ulcer Agents/pharmacology , Clarithromycin/pharmacokinetics , Gastric Mucosa/metabolism , Omeprazole/pharmacology , Adolescent , Adult , Anti-Bacterial Agents/blood , Clarithromycin/blood , Cross-Over Studies , Double-Blind Method , Drug Interactions , Gastric Acidity Determination , Gastric Mucosa/drug effects , Humans , Male , Middle Aged , Mucus/metabolism , Stomach/drug effects
6.
Cancer Res ; 51(9): 2286-90, 1991 May 01.
Article in English | MEDLINE | ID: mdl-2015593

ABSTRACT

Monoclonal antibody (MoAb) conjugates have been used to treat a variety of malignancies. The majority of the MoAbs which have been used therapeutically are from murine sources. The infusion of these foreign proteins into humans can be expected to elicit anti-murine antibodies and may be one of the major limitations to the clinical use of murine MoAbs. In these studies, we report on the nature and specificity of the human anti-murine antibody (HAMA) response in patients given single and multiple infusions of the two Vinca alkaloid conjugates of the MoAb KS1/4, which recognizes tumor-associated antigens in a variety of adenocarcinomas. A HAMA response was induced in a majority of the patients receiving infusions of KS1/4 conjugates, regardless of the specific conjugate used or the number of infusions. The magnitude of the response did not appear to be dose related. Antibodies directed to the drug moieties of these conjugates, anti-Vinca alkaloids, were also induced in patients with HAMA responses. The magnitude of the anti-Vinca response paralleled that of the HAMA.


Subject(s)
Antibodies, Monoclonal/immunology , Immunotoxins/immunology , Vinblastine/analogs & derivatives , Antibodies, Monoclonal/administration & dosage , Antibody Formation , Drug Evaluation , Humans , Immunotoxins/administration & dosage , Infusions, Intravenous , Vinblastine/administration & dosage , Vinblastine/immunology
7.
Clin Pharmacol Ther ; 47(4): 499-503, 1990 Apr.
Article in English | MEDLINE | ID: mdl-2109665

ABSTRACT

Steady-state plasma nizatidine concentrations were related in a linear fashion to nizatidine infusion rate. Infusion rates of 2.5, 10, and 20 mg/hr resulted in mean plasma nizatidine concentrations of 69, 247, and 575 ng/ml. Basal acid secretion was inhibited by 50% and 90% at mean plasma nizatidine concentrations of 60 and 430 ng/ml. Protein-stimulated acid secretion was inhibited by 50% and 90% at mean plasma nizatidine concentrations of 75 and 490 ng/ml. The mean pH of basal gastric secretions was 1.6 during placebo infusion and 4.6 when the mean plasma nizatidine concentration was 575 ng/ml.


Subject(s)
Gastric Acid/metabolism , Thiazoles/blood , Adult , Depression, Chemical , Dietary Proteins/pharmacology , Humans , Hydrogen-Ion Concentration , Male , Metabolic Clearance Rate , Middle Aged , Nizatidine , Random Allocation , Reference Values , Thiazoles/pharmacokinetics , Thiazoles/pharmacology
8.
Article in English | MEDLINE | ID: mdl-2892261

ABSTRACT

In this report, the pharmacokinetic, pharmacodynamic, and hormonal effects of nizatidine are reviewed in healthy volunteers and in patients with renal or hepatic impairment. The absolute oral bioavailability of nizatidine exceeded 90%; the half-life (t1/2), plasma clearance (Clp), and volume of distribution (Vd) of iv nizatidine were 1.3 h, 461/h, and 1.21/kg, respectively. Within 16 h of dosing volunteers with nizatidine, more than 90% of the administered dose was recovered in urine as parent drug and metabolites; unchanged nizatidine accounted for 65 and 75% of the recovered substances, after oral and intravenous administration, respectively. Renal impairment decreased the elimination of nizatidine and dosage reductions are recommended for patients with creatinine clearance (Clcr) less than 50 ml/min/m2. Nizatidine suppressed gastric acid secretion produced by sham meals, protein meals, or pentagastrin; its antisecretory activity was dose and concentration dependent. Nizatidine was three times more potent than cimetidine. Nizatidine, after oral or iv administration, did not alter hormone concentrations in plasma.


Subject(s)
Histamine H2 Antagonists/pharmacokinetics , Thiazoles/pharmacokinetics , Adult , Aged , Female , Gastric Juice/drug effects , Gastric Juice/metabolism , Histamine H2 Antagonists/administration & dosage , Histamine H2 Antagonists/pharmacology , Hormones/blood , Humans , Male , Middle Aged , Nizatidine , Thiazoles/administration & dosage , Thiazoles/pharmacology
9.
Biopharm Drug Dispos ; 6(4): 433-40, 1985.
Article in English | MEDLINE | ID: mdl-4084668

ABSTRACT

Single oral doses of cibenzoline were administered to eight healthy volunteers on two different occasions, once at 8.00 am and once at 10.00 pm, in a randomized crossover design with at least one week separating treatments. A fast was maintained for 12 hours prior to and for 2 hours after the morning dose and the subjects did not lie down for at least 12 hours after dosing. A standard dinner was eaten 3 hours prior to the evening dose, and a fast was maintained for 10 hours after dosing; the subjects laid down 2 hours after dosing for at least 6 hours. Blood was collected at specific times for 72 hours and the total volume of urine voided was collected through 72 hours. Cibenzoline concentrations in plasma and urine were measured by HPLC. Cibenzoline absorption was slower in 7 of the 8 volunteers following the evening dose relative to the morning dose. Mean +/- S.D. tmax for the evening dose was 2.6 +/- 0.5 hours compared to 1.7 +/- 0.8 for the morning dose. The corresponding mean +/- S.D. Cmax following the morning dose was 446 +/- 124 ng ml-1 compared to 402 +/- 114 ng ml-1 after the evening dose. The mean +/- S.D. AUC was 3328 +/- 1101 ng . h . ml-1 after the morning dose and 3561 +/- 1430 ng . h . ml-1 after the evening dose. The harmonic mean half-life was 7.4 hours after both treatments. These data indicated that the total amount of drug absorbed and the elimination rate constant of the drug had not varied between treatments.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Anti-Arrhythmia Agents/administration & dosage , Imidazoles/administration & dosage , Administration, Oral , Adult , Anti-Arrhythmia Agents/blood , Anti-Arrhythmia Agents/urine , Circadian Rhythm , Drug Administration Schedule , Humans , Imidazoles/blood , Imidazoles/urine , Kinetics , Time Factors
11.
Lancet ; 2(8410): 1005-8, 1984 Nov 03.
Article in English | MEDLINE | ID: mdl-6149395

ABSTRACT

50 hypertensive patients and 50 normal controls were evaluated in the sleep laboratory for the presence of sleep apnoea or sleep apnoeic activity. Hypertensive patients were at high risk of sleep apnoea; 15 hypertensive patients (30%) had sleep apnoea and another 17 (34%) had sleep apnoeic activity. In contrast, none of the age-matched and sex-matched control subjects had sleep apnoea, and 24% had sleep apnoeic activity. The degree of oxygen desaturation was correlated with the duration as well as the number of apnoeic events. Presence of sleep apnoea in the patients was significantly correlated with higher blood pressure levels when they were initially seen in the clinic. Patients with the most severe sleep apnoea had the highest initial blood-pressure levels and were more refractory to treatment.


Subject(s)
Hypertension/complications , Sleep Apnea Syndromes/etiology , Adolescent , Adult , Age Factors , Aged , Female , Humans , Hypertension/drug therapy , Male , Middle Aged , Minoxidil/adverse effects , Oxygen/blood , Risk , Sex Factors , Sleep Apnea Syndromes/epidemiology , Time Factors
12.
Clin Pharmacol Ther ; 36(5): 584-7, 1984 Nov.
Article in English | MEDLINE | ID: mdl-6488678

ABSTRACT

Nadolol and propranolol effects on lidocaine elimination were followed in six healthy men and women. Each received three separate 30-hr infusions of lidocaine (2 mg/min): one alone, one after 3 days pretreatment with nadolol (160 mg daily), and one after 3 days pretreatment with propranolol (80 mg every 8 hr). Liver blood flow was determined by the systemic clearance of indocyanine green. Steady-state plasma lidocaine levels were increased by nadolol (2.1 +/- 0.2 to 2.7 +/- 0.3 micrograms/ml) and by propranolol (2.1 +/- 0.2 to 2.5 +/- 0.3 micrograms/ml). Lidocaine plasma clearance was decreased by nadolol (1030 +/- 81 to 850 +/- 82 ml/min) and by propranolol (1030 +/- 81 to 866 +/- 75 ml/min). Hepatic blood flow was decreased by nadolol (1275 +/- 77 to 902 +/- 102 ml/min) and propranolol (1275 +/- 77 to 957 +/- 119 ml/min). The hepatic extraction ratio for lidocaine was increased by nadolol (0.86 +/- 0.06 to 0.91 +/- 0.05) and by propranolol (0.86 +/- 0.06 to 0.90 +/- 0.06). Lidocaine intrinsic clearance was not changed by nadolol (8.19 +/- 1.87 to 9.52 +/- 2.36 l/min) or propranolol (8.19 +/- 1.87 to 9.50 +/- 3.13 l/min). Our data indicate that both nadolol and propranolol reduce lidocaine clearance by their effects on hepatic blood flow and not by inhibition of lidocaine metabolism.


Subject(s)
Lidocaine/metabolism , Liver/drug effects , Propanolamines/pharmacology , Propranolol/pharmacology , Adult , Drug Interactions , Female , Heart Rate/drug effects , Humans , Infusions, Parenteral , Lidocaine/blood , Liver/blood supply , Male , Nadolol
13.
Clin Pharmacol Ther ; 36(1): 105-15, 1984 Jul.
Article in English | MEDLINE | ID: mdl-6428793

ABSTRACT

Dazoxiben, a specific thromboxane synthetase inhibitor, was evaluated in 21 patients with Raynaud's phenomenon in a double-blind, placebo-controlled crossover experiment. Total fingertip blood flows were measured by plethysmography and capillary blood flows were measured by 133Xe disappearance rate. Subjects were studied in both a warm (28 degrees) and a cold (20 degrees) room. Arteriovenous (AV) shunt flow was estimated by subtraction of capillary flow from total flow. Ex vivo production of thromboxane B2 (TXB2) and 6-keto PGF1 alpha was determined by specific radioimmunoassay in serum from venous blood incubated for 1 hr (37 degrees). Plasma concentrations of TXB2 and 6-keto PGF1 alpha were also monitored. Dazoxiben (100 mg 4 times a day for 14 days) inhibited ex vivo TXB2 production (from 463.1 +/- 69.9 to 101.8 +/- 13.4 ng/ml/hr; (means +/- SE], enhanced ex vivo 6-keto PGF1 alpha production (from 1.38 +/- 0.05 to 3.76 +/- 0.18 ng/ml/hr), reduced plasma TXB2 concentration (from 88.1 +/- 13.9 to 38.8 +/- 5.9 pg/ml). There were no changes in plasma concentration of 6-keto PGF1 alpha. Dazoxiben did not improve total digital blood flow, capillary flow, AV shunt flow, or forearm blood flow at 28 degrees or 20 degrees. There was no subjective improvement in frequency or severity of Raynaud's attacks (assessed by patient diaries). It is concluded that dazoxiben is a potent and specific thromboxane synthetase inhibitor capable of altering arachidonic acid metabolism, but is of little or no benefit in the treatment of Raynaud's phenomenon.


Subject(s)
Imidazoles/therapeutic use , Raynaud Disease/drug therapy , 6-Ketoprostaglandin F1 alpha/blood , Adult , Aged , Arachidonic Acid , Arachidonic Acids/metabolism , Double-Blind Method , Drug Evaluation , Female , Forearm/blood supply , Humans , Male , Middle Aged , Plethysmography , Radioimmunoassay , Thromboxane B2/blood
14.
Clin Pharmacol Ther ; 35(4): 447-53, 1984 Apr.
Article in English | MEDLINE | ID: mdl-6705442

ABSTRACT

Five healthy subjects were given oral 14C-propranolol (10 microCi, 40 mg) alone and in combination with hydralazine, 25 and 50 mg. Hydralazine increased propranolol peak concentrations from 25 +/- 7 ng/ml to 61 +/- 10 and 85 +/- 11 ng/ml, reduced time to peak concentrations from 2.2 +/- 0.2 hr to 0.7 +/- 0.1 and 0.8 +/- 0.1 hr, and increased area under the propranolol concentration: time curves from 153 +/- 38 ng X ml-1 X hr to 246 +/- 64 and 324 ng X ml-1 X hr (in all cases P less than 0.05). Hydralazine did not change the fraction of the 14C-propranolol dose recovered in the urine as basic, acidic, and polar metabolites: 0.28 +/- 0.2, 0.27 +/- 0.03, and 0.44 +/- 0.03. The urinary excretion rate of radioactive metabolites of propranolol in acid, basic, and residue fractions increased in the 0 = to = 2-hr time interval after hydralazine but there was no change in the relative proportion of each metabolite fraction at any time. Similar results were obtained by HPLC. Studies with radioactive propranolol indicate that a major acid and basic metabolite remains to be defined in addition to unextracted polar metabolites. Our data indicate that hydralazine increases propranolol bioavailability by its hemodynamic actions rather than by inhibition of its metabolism.


Subject(s)
Hydralazine/pharmacology , Propranolol/metabolism , Administration, Oral , Adult , Biological Availability , Carbon Radioisotopes , Chromatography, High Pressure Liquid , Drug Interactions , Humans , Kinetics , Male , Propranolol/urine
15.
Clin Pharmacol Ther ; 35(1): 19-25, 1984 Jan.
Article in English | MEDLINE | ID: mdl-6317271

ABSTRACT

One gram ceftriaxone was injected at a constant rate in an intravenous infusion over 30 min to eight elderly subjects (mean age, 70.5 yr) and eight young subjects (mean age, 28.9 yr); the latter served as body weight-matched controls. Plasma and urine samples were collected in serial order for 48 hr and assayed for unchanged drug. Selected plasma samples were subjected to protein binding determinations by equilibrium dialysis. Statistical comparison of data for the old and young indicated no significant changes in means of (1) maximum plasma concentration (140 and 133 micrograms/ml); (2) elimination rate constant (0.078 and 0.093 hr-1) and elimination t1/2 (8.9 and 7.5 hr); (3) apparent volume of distribution (10.69 and 11.01 l); (4) plasma clearance (833 and 1023 ml/hr); (5) nonrenal clearance (515 and 606 ml/hr); and (6) percent dose excreted unchanged in urine (39.6 and 41.4). There was, however, a significant decrease in the renal clearance (318 and 416 ml/hr) and a significant increase in the plasma free fractions (0.157 and 0.136 at 100 micrograms/ml and 0.146 and 0.114 at 60 to 70 micrograms/ml) of ceftriaxone in elderly subjects. The 24% decrease in renal clearance in the elderly subjects corresponded to the 19% decrease in their creatinine clearance. Since the age-related changes in kinetics were relatively small, it is concluded that dosage adjustment is probably not necessary for elderly subjects requiring ceftriaxone.


Subject(s)
Aging , Cefotaxime/analogs & derivatives , Adult , Aged , Blood Proteins/metabolism , Cefotaxime/metabolism , Ceftriaxone , Chromatography, High Pressure Liquid , Female , Humans , Infusions, Parenteral , Kinetics , Male , Protein Binding/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...