Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Br J Anaesth ; 121(4): 758-767, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30236238

ABSTRACT

BACKGROUND: We investigated potential for hypersensitivity reactions after repeated sugammadex administration and explored the mechanism of hypersensitivity. METHODS: In this double-blind, placebo-controlled study (NCT00988065), 448 healthy volunteers were randomised to one of three arms to receive three repeat i.v. administrations of either sugammadex 4 mg kg-1, 16 mg kg-1, or placebo. Primary endpoint was percentage of subjects with hypersensitivity (assessed by an independent adjudication committee). Secondary endpoint of anaphylaxis was classified per Sampson and Brighton criteria. Exploratory endpoints included skin testing, serum tryptase, anti-sugammadex antibodies [immunoglobulin (Ig) E/IgG], and other immunologic parameters. RESULTS: Hypersensitivity was adjudicated for 1/148 (0.7%), 7/150 (4.7%), and 0/150 (0.0%) subjects after sugammadex 4 mg kg-1, 16 mg kg-1, and placebo, respectively. After sugammadex 16 mg kg-1, one subject met Sampson criterion 1 and Brighton level 1 (highest certainty) anaphylaxis criteria; two met Brighton level 2 criteria. After database lock it was determined that certain protocol deviations could have introduced bias in the reporting of hypersensitivity signs/symptoms in a subject subset. Objective laboratory investigations indicated that potential underlying hypersensitivity mechanisms were unlikely to have been activated; the results suggest that most of the observed hypersensitivity reactions were unlikely IgE/IgG-mediated. CONCLUSION: Dose-dependent hypersensitivity or anaphylaxis reactions to sugammadex were observed when administered without prior neuromuscular blocking agent. Laboratory investigations do not suggest prevalent allergen-specific IgE/IgG-mediated immunologic hypersensitivity. Because it could not be fully excluded that estimates of hypersensitivity/anaphylaxis incidence were unbiased, an additional study was conducted to characterise the potential for hypersensitivity reactions and is described in a companion report. CLINICAL TRIAL REGISTRATION: http://www.clinicaltrials.gov NCT00988065; Protocol number P06042.


Subject(s)
Drug Hypersensitivity/immunology , Sugammadex/adverse effects , Administration, Intravenous , Adolescent , Adult , Anaphylaxis/immunology , Antibodies/immunology , Double-Blind Method , Female , Healthy Volunteers , Humans , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Male , Middle Aged , Safety , Skin Tests , Sugammadex/administration & dosage , Tryptases/blood , Young Adult
2.
Tijdschr Psychiatr ; 60(4): 221-230, 2018.
Article in Dutch | MEDLINE | ID: mdl-29638236

ABSTRACT

BACKGROUND: The FACTboard meeting structures the multidisciplinary meetings held by FACT teams, held for the 10-20% most care intensive patients. The FACT manual only provides a general outline for the FACTboard meeting, leaving out criteria specifying the methods to structure meetings. Precisely describing these criteria could improve the quality of these meetings. AIM: To develop a more detailed guideline on how to structure a FACTboard meeting by means of a Delphi study. METHOD: The panel of the Delphi study existed of 22 professions working in certified FACT teams and 8 experts in the field of FACT. Panel members individually assessed 113 items according to whether the statement should be included in the guideline. Statements rated important or essential by ≥80% of the panel members were included in the guideline. The panel members' commentary was used to shape and adjust the statements to clarify why they were regarded as important or unimportant. RESULTS: 54 statements were rated essential or important by ≥80% of the panel members. These statements pertained to the organization and structure of the FACTboard meeting and the roles and responsibilities of the team members. CONCLUSION: The developed guideline could be used by FACT and possibly ACT teams to structure the FACTboard meeting.


Subject(s)
Delphi Technique , Patient Care Team/organization & administration , Practice Guidelines as Topic , Psychiatry/organization & administration , Cooperative Behavior , Humans
3.
Clin Pharmacol Ther ; 98(2): 196-204, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25869840

ABSTRACT

Preclinical development of new biological entities (NBEs), such as human protein therapeutics, requires considerable expenditure of time and costs. Poor prediction of pharmacokinetics in humans further reduces net efficiency. In this study, we show for the first time that pharmacokinetic data of NBEs in humans can be successfully obtained early in the drug development process by the use of microdosing in a small group of healthy subjects combined with ultrasensitive accelerator mass spectrometry (AMS). After only minimal preclinical testing, we performed a first-in-human phase 0/phase 1 trial with a human recombinant therapeutic protein (RESCuing Alkaline Phosphatase, human recombinant placental alkaline phosphatase [hRESCAP]) to assess its safety and kinetics. Pharmacokinetic analysis showed dose linearity from microdose (53 µg) [(14) C]-hRESCAP to therapeutic doses (up to 5.3 mg) of the protein in healthy volunteers. This study demonstrates the value of a microdosing approach in a very small cohort for accelerating the clinical development of NBEs.


Subject(s)
Alkaline Phosphatase/administration & dosage , Alkaline Phosphatase/pharmacokinetics , Carbon Radioisotopes , Isoenzymes/administration & dosage , Isoenzymes/pharmacokinetics , Administration, Intravenous , Adolescent , Adult , Alkaline Phosphatase/adverse effects , Area Under Curve , Double-Blind Method , Drug Dosage Calculations , GPI-Linked Proteins/administration & dosage , GPI-Linked Proteins/adverse effects , GPI-Linked Proteins/pharmacokinetics , Half-Life , Healthy Volunteers , Humans , Isoenzymes/adverse effects , Linear Models , Male , Mass Spectrometry/methods , Metabolic Clearance Rate , Models, Biological , Netherlands , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Young Adult
4.
J Psychopharmacol ; 25(3): 353-60, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20142306

ABSTRACT

Arginine-vasopressin (AVP) is a physiological co-activator of the hypothalamus-pituitary-adrenal (HPA) axis, together with corticotrophin releasing hormone (CRH). A synthetic analogue of AVP, desmopressin (dDAVP), is often used as a pharmacological tool to assess co-activation in health and disease. The relation between dDAVP's neuroendocrine, cardiovascular, pro-coagulatory, anti-diuretic and non-specific stress effects has not been studied. A randomized, double-blind, placebo-controlled, three-way crossover study was performed in 12 healthy male and female volunteers (6 : 6). dDAVP was administered intravenously as a 10 µg bolus (over 1 min) or a 30 µg incremental infusion (over 60 min). Neuroendocrine, cardiovascular, pro-coagulatory, anti-diuretic effects and adverse events (AEs) were recorded, and autonomic nervous system (ANS) activation evaluated. The incremental infusion reached 1.8-fold higher dDAVP concentrations than the bolus. Neuroendocrine effects were similar for the 10 µg dDAVP bolus and the 30 µg incremental infusion, while cardiovascular and coagulatory effects were greater with the 30 µg dose. Osmolality and ANS activity remained uninfluenced. AEs corresponded to dDAVP's side-effect profile. In conclusion, the neuroendocrine effects of a 10 µg dDAVP bolus administered over 1 min are similar to those of a 30 µg incremental infusion administered over one hour, despite higher dDAVP concentrations after the infusion. Cardiovascular and coagulatory effects showed clear dose-related responses. A 10 µg dDAVP bolus is considered a safe vasopressinergic function test at which no confounding effects of systemic or autonomic stress were seen.


Subject(s)
Antidiuretic Agents/pharmacology , Deamino Arginine Vasopressin/pharmacology , Pituitary-Adrenal System/drug effects , Adolescent , Adult , Antidiuretic Agents/administration & dosage , Antidiuretic Agents/adverse effects , Autonomic Nervous System/drug effects , Autonomic Nervous System/metabolism , Blood Coagulation/drug effects , Cross-Over Studies , Deamino Arginine Vasopressin/administration & dosage , Deamino Arginine Vasopressin/adverse effects , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Hypothalamo-Hypophyseal System/metabolism , Infusions, Intravenous , Injections, Intravenous , Male , Pituitary-Adrenal System/metabolism , Young Adult
5.
J Psychopharmacol ; 25(3): 361-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20147570

ABSTRACT

Pharmacological function tests consisting of 100 µg hCRH (corticorelin) and 10 µg dDAVP (desmopressin) mimic endogenous hypothalamus-pituitary-adrenal (HPA) axis activation. However, physiological CRH concentrations preclude informative vasopressinergic co-activation (using dDAVP) and independent quantification of both corticotrophinergic (using hCRH) and vasopressinergic (using dDAVP) activation is limited due to administration on separate occasions. This randomized, double-blind, placebo-controlled, partial five-way crossover study in healthy males and females (six : six) examined whether (1) concomitant administration of dDAVP and hCRH provides more informative vasopressinergic co-activation than dDAVP alone; and (2) whether the administration of dDAVP followed two hours later by hCRH can quantify both vasopressinergic and corticotrophinergic activation on a single test day. Combining 10 µg dDAVP with 10 µg and 30 µg hCRH caused dose-related ACTH and cortisol release which was larger than with 10 µg dDAVP alone and respectively comparable to and greater than that induced by 100 µg hCRH. Using 10 µg dDAVP alone demonstrated limited ACTH release while the effects of 100 µg hCRH two hours later were three times as large. ACTH and cortisol released by 10 µg dDAVP returned to baseline prior to 100 µg hCRH administration and dDAVP did not influence the response to subsequent hCRH administration. Dose-related vasopressinergic co-activation of the HPA axis was induced by combining 10 µg dDAVP with 10 µg and 30 µg hCRH. Combining 10 µg dDAVP with 10 µg hCRH induced the potentially most informative vasopressinergic co-activation since it is not restricted by ceiling or flooring effects. The hCRH response was not affected by prior dDAVP, allowing for a practical function test examining both HPA activation routes on the same day.


Subject(s)
Corticotropin-Releasing Hormone/pharmacology , Deamino Arginine Vasopressin/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Adolescent , Adult , Corticotropin-Releasing Hormone/administration & dosage , Cross-Over Studies , Deamino Arginine Vasopressin/administration & dosage , Dose-Response Relationship, Drug , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Hypothalamo-Hypophyseal System/metabolism , Male , Pituitary-Adrenal System/metabolism , Young Adult
6.
Drug Metab Dispos ; 39(4): 580-90, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21177986

ABSTRACT

The metabolism and excretion of asenapine [(3aRS,12bRS)-5-chloro-2-methyl-2,3,3a,12b-tetrahydro-1H-dibenzo[2,3:6,7]-oxepino [4,5-c]pyrrole (2Z)-2-butenedioate (1:1)] were studied after sublingual administration of [(14)C]-asenapine to healthy male volunteers. Mean total excretion on the basis of the percent recovery of the total radioactive dose was ∼90%, with ∼50% appearing in urine and ∼40% excreted in feces; asenapine itself was detected only in feces. Metabolic profiles were determined in plasma, urine, and feces using high-performance liquid chromatography with radioactivity detection. Approximately 50% of drug-related material in human plasma was identified or quantified. The remaining circulating radioactivity corresponded to at least 15 very polar, minor peaks (mostly phase II products). Overall, >70% of circulating radioactivity was associated with conjugated metabolites. Major metabolic routes were direct glucuronidation and N-demethylation. The principal circulating metabolite was asenapine N(+)-glucuronide; other circulating metabolites were N-desmethylasenapine-N-carbamoyl-glucuronide, N-desmethylasenapine, and asenapine 11-O-sulfate. In addition to the parent compound, asenapine, the principal excretory metabolite was asenapine N(+)-glucuronide. Other excretory metabolites were N-desmethylasenapine-N-carbamoylglucuronide, 11-hydroxyasenapine followed by conjugation, 10,11-dihydroxy-N-desmethylasenapine, 10,11-dihydroxyasenapine followed by conjugation (several combinations of these routes were found) and N-formylasenapine in combination with several hydroxylations, and most probably asenapine N-oxide in combination with 10,11-hydroxylations followed by conjugations. In conclusion, asenapine was extensively and rapidly metabolized, resulting in several regio-isomeric hydroxylated and conjugated metabolites.


Subject(s)
Antipsychotic Agents/metabolism , Glucuronides/analysis , Heterocyclic Compounds, 4 or More Rings/metabolism , Adult , Antipsychotic Agents/blood , Antipsychotic Agents/chemistry , Antipsychotic Agents/urine , Area Under Curve , Dibenzocycloheptenes , Glucuronides/metabolism , Heterocyclic Compounds, 4 or More Rings/blood , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/urine , Humans , Hydroxylation , Male , Middle Aged , Radioligand Assay , Young Adult
7.
Br J Anaesth ; 100(3): 373-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18238834

ABSTRACT

BACKGROUND: Sugammadex rapidly reverses rocuronium- and vecuronium-induced neuromuscular block. To investigate the effect of combination of sugammadex and rocuronium or vecuronium on QT interval, it would be preferable to avoid the interference of anaesthesia. Therefore, this pilot study was performed to investigate the safety, tolerability, and plasma pharmacokinetics of single i.v. doses of sugammadex administered simultaneously with rocuronium or vecuronium to anaesthetized and non-anaesthetized healthy volunteers. METHODS: In this phase I study, 12 subjects were anaesthetized with propofol/remifentanil and received sugammadex 16, 20, or 32 mg kg(-1) combined with rocuronium 1.2 mg kg(-1) or vecuronium 0.1 mg kg(-1); four subjects were not anaesthetized and received sugammadex 32 mg kg(-1) with rocuronium 1.2 mg kg(-1) or vecuronium 0.1 mg kg(-1) (n=2 per treatment). Neuromuscular function was assessed by TOF-Watch SX monitoring in anaesthetized subjects and by clinical tests in non-anaesthetized volunteers. Sugammadex, rocuronium, and vecuronium plasma concentrations were measured at several time points. RESULTS: No serious adverse events (AEs) were reported. Fourteen subjects reported 23 AEs after study drug administration. Episodes of mild headache, tiredness, cold feeling (application site), dry mouth, oral discomfort, nausea, increased aspartate aminotransferase and gamma-glutamyltransferase levels, and moderate injection site irritation were considered as possibly related to the study drug. The ECG and vital signs showed no clinically relevant changes. Rocuronium/vecuronium plasma concentrations declined faster than those of sugammadex. CONCLUSIONS: Single-dose administration of sugammadex 16, 20, or 32 mg kg(-1) in combination with rocuronium 1.2 mg kg(-1) or vecuronium 0.1 mg kg(-1) was well tolerated with no clinical evidence of residual neuromuscular block, confirming that these combinations can safely be administered simultaneously to non-anaesthetized subjects. Rocuronium and vecuronium plasma concentrations decreased faster than those of sugammadex, reducing the theoretical risk of neuromuscular block developing over time.


Subject(s)
Androstanols/antagonists & inhibitors , Neuromuscular Nondepolarizing Agents/antagonists & inhibitors , Vecuronium Bromide/antagonists & inhibitors , gamma-Cyclodextrins/adverse effects , Adolescent , Adult , Androstanols/administration & dosage , Androstanols/blood , Anesthetics, Intravenous , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Male , Neuromuscular Blockade , Neuromuscular Junction/drug effects , Neuromuscular Junction/physiology , Neuromuscular Nondepolarizing Agents/administration & dosage , Neuromuscular Nondepolarizing Agents/blood , Propofol , Rocuronium , Sugammadex , Vecuronium Bromide/administration & dosage , Vecuronium Bromide/blood , gamma-Cyclodextrins/administration & dosage , gamma-Cyclodextrins/blood , gamma-Cyclodextrins/pharmacology
8.
Eur J Clin Pharmacol ; 58(6): 423-9, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12242602

ABSTRACT

OBJECTIVE: The objectives of this study were to assess the effect of mirtazapine on steady-state pharmacokinetics of phenytoin and vice versa and to assess tolerability and safety of the combined use of mirtazapine and phenytoin. METHODS: This was an open-label, randomised, parallel-groups, single-centre, multiple-dose pharmacokinetic study. Seventeen healthy, male subjects completed either treatment A [nine subjects: daily 200 mg phenytoin for 17 days plus mirtazapine (15 mg for 2 days continuing with 30 mg for 5 days) from day 11 to day 17] or treatment B [eight subjects: mirtazapine, daily 15 mg for 2 days continuing with 30 mg for 15 days plus phenytoin 200 mg from day 8 to day 17]. Serial blood samples were taken for kinetic profiling on the 10th and 17th days of treatment A and on the 7th and 17th days of treatment B. Induction of CYP 3A by phenytoin was evaluated by measuring the ratio of 6 beta-hydroxycortisol over cortisol on the 1st, 7th and 17th days of treatment B. RESULTS: Co-administration of mirtazapine had no effect on the steady-state pharmacokinetics of phenytoin, i.e. the area under the plasma concentration-time curve (AUC)(0-24) and peak plasma concentration (C(max)) remained unchanged. The addition of phenytoin to an existing daily administration of mirtazapine resulted in a mean (+/-SD) decrease of the AUC(0-24) from 576+/-104 ng h/ml to 305+/-81.6 ng h/ml and a mean decrease of C(max) from 69.7+/-17.5 ng/ml to 46.9+/-10.9 ng/ml. Induction of CYP 3A by phenytoin is confirmed by the significantly ( P=0.001) increased 6beta-hydroxycortisol/cortisol ratio from 1.74+/-1.00 to 2.74+/-1.64. CONCLUSION: Co-administration of mirtazapine did not alter the steady-state pharmacokinetics of phenytoin. The addition of phenytoin to an existing daily administration of mirtazapine results in a decrease of the plasma concentrations of mirtazapine by 46% on average, most likely due to induction of CYP 3A3/4.


Subject(s)
Anticonvulsants/pharmacokinetics , Antidepressive Agents, Tricyclic/pharmacokinetics , Mianserin/pharmacokinetics , Phenytoin/pharmacokinetics , Administration, Oral , Adolescent , Adult , Anticonvulsants/administration & dosage , Anticonvulsants/adverse effects , Antidepressive Agents, Tricyclic/administration & dosage , Antidepressive Agents, Tricyclic/adverse effects , Area Under Curve , Drug Interactions , Humans , Male , Mianserin/administration & dosage , Mianserin/adverse effects , Mianserin/analogs & derivatives , Middle Aged , Mirtazapine , Phenytoin/administration & dosage , Phenytoin/adverse effects
9.
Br J Clin Pharmacol ; 53(5): 492-500, 2002 May.
Article in English | MEDLINE | ID: mdl-11994055

ABSTRACT

AIMS: The antimalarial efficacy/pharmacodynamics and pharmacokinetics of intramuscular (i.m.) artemotil in Thai patients with acute uncomplicated falciparum malaria were studied to determine effective dose regimens and to compare these with the standard dose regimen of artemether. METHODS: In part I of the study three different artemotil dose regimens were explored in three groups of 6-9 patients for dose finding: 3.2 mg kg-1 on day 0 and 1.6 mg kg-1 on days 1-4 (treatment A), 1.6 mg kg-1 on day 0 and 0.8 mg kg-1 on days 1-4 (treatment B), 3.2 mg kg-1 on day 0 and 0.8 mg kg-1 on days 1-4 (treatment C). In part II of the study, artemotil treatments A and C were compared in three groups of 20-22 patients with standard i.m. artemether treatment: 3.2 mg kg-1 on day 0 and 0.8 mg kg-1 on days 1-4 (treatment R). RESULTS: Full parasite clearance was achieved in all patients in Part I, but parasite clearance time (PCT) and fever clearance time (FCT) tended to be longer in treatment B. Also the incidence of recrudescence before day 28 (RI) tended to be higher for treatment B. In part II, the mean PCT for each of the two artemotil treatments (52 and 55 h, respectively) was significantly longer than for artemether (43 h). The 95% CI for the difference A vs R was 0, 16 h (P=0.0408) and for difference C vs R it was 2, 19 h (P=0.0140). FCT was similar for the three treatments. The incidence of RI ranged from 5 out of 19 for treatment C to 3 out of 20 for treatment R. Plasma concentration-time profiles of artemotil indicated an irregular and variable rate of absorption after i.m. injection. A late onset of parasite clearance was associated with delayed absorption and/or very low initial artemotil plasma concentrations. Pharmacokinetic-pharmacodynamic evaluations supported a relationship between the rate of parasite clearance and exposure to artemotil during approximately the first 2 days of treatment, and suggested that artemotil has a slower rate of absorption than artemether. Safety assessment, including neurological and audiometric examinations showed no clinically relevant findings. Adverse events before and during treatment included headache, dizziness, nausea, vomiting and abdominal pain. These are characteristic of acute malaria infections and resolved during treatment. CONCLUSIONS: The optimum dose regimen for artemotil in this study was identical to the standard dose regimen of artemether. The findings that artemotil is more slowly absorbed from the i.m. injection site than artemether, and that early systemic availability may be insufficient for an immediate onset of parasite clearance contributed to the decision to choose a higher loading dose of artemotil (divided over two injection sites) and to omit the fifth dose in later studies. With this optimized dosing schedule, the more pronounced depot characteristics of i.m. artemotil can be an advantage, since it may allow shorter hospitalization.


Subject(s)
Antimalarials/therapeutic use , Artemisinins/therapeutic use , Malaria, Falciparum/drug therapy , Sesquiterpenes/therapeutic use , Acute Disease , Adolescent , Adult , Animals , Antimalarials/administration & dosage , Antimalarials/pharmacology , Artemether , Artemisinins/administration & dosage , Artemisinins/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Injections, Intramuscular , Malaria, Falciparum/blood , Male , Middle Aged , Sesquiterpenes/administration & dosage , Sesquiterpenes/pharmacology
10.
J Steroid Biochem Mol Biol ; 78(5): 471-80, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11738557

ABSTRACT

The metabolism of desogestrel (13-ethyl-11-methylene-18,19-dinor-17alpha-pregn-4-en-20-yn-17-ol), a progestagen used in oral contraceptives and hormone replacement therapy, was studied in vivo after a single oral administration of 150 microg [14C]-labeled desogestrel and 30 microg ethinylestradiol under steady state conditions to healthy postmenopausal women. After this oral administration, desogestrel was extensively metabolized. The dosed radioactivity was predominantly ( approximately 60%) excreted via urine, while about 35% was excreted via the feces. Desogestrel was metabolized mainly at the C3-, C5-, C6- and C13-CH(2)CH(3) positions. At the C3-position, the 3-keto moiety was found and in addition, 3beta-hydroxy and 3alpha-hydroxy groups were observed in combination with a reduced Delta(4)-double bond (5alpha-H). Hydroxy groups were introduced at the C6- (6beta-OH), the C13-ethyl (C13-CH(2)CH(2)OH) and possibly the C15- (15alpha-OH) position of desogestrel. Conjugation of the 3alpha-hydroxy moiety with sulfonic acid and conjugation with glucuronic acid were also major metabolic routes found for desogestrel in postmenopausal women. The 3-keto metabolite of desogestrel (the biologically active metabolite) was the major compound present in plasma at least up to 24 h after administration of the radioactive dose. Species comparison of the metabolic routes of desogestrel after oral administration indicates that in rats and dogs desogestrel is also mainly metabolized at the C3-position, similar to what is now found for postmenopausal women. Most other metabolic routes of desogestrel were found to differ between species. Finally, major metabolic routes found in the present study in postmenopausal women are in line with outcome of previous in vitro metabolism studies with human liver tissue (microsomes and postmitochondrial liver fractions) and intestinal mucosa.


Subject(s)
Desogestrel/metabolism , Desogestrel/pharmacokinetics , Menopause/metabolism , Progesterone Congeners/metabolism , Progesterone Congeners/pharmacokinetics , Administration, Oral , Animals , Biotransformation , Chromatography, High Pressure Liquid , Desogestrel/chemistry , Dogs , Estrogen Replacement Therapy , Feces/chemistry , Female , Glucuronides/chemistry , Glucuronides/metabolism , Glucuronides/urine , Humans , Hydroxylation , In Vitro Techniques , Intestinal Mucosa/metabolism , Liver/metabolism , Molecular Structure , Progesterone Congeners/chemistry , Rats , Species Specificity
11.
J Clin Pharmacol ; 40(12 Pt 1): 1312-22, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11185629

ABSTRACT

The study was conducted in healthy male volunteers to evaluate the absorption, metabolic pattern, and mode of elimination of telmisartan, a nonpeptide angiotensin II receptor antagonist. [14C]telmisartan was administered orally in solution as a single 40 mg dose to 5 subjects. A further 5 subjects received short-term intravenous infusion of [14C]telmisartan 40 mg. Measurement of total 14C radioactivity in plasma showed that about 50% was absorbed following oral administration, with maximum plasma concentration observed after 0.5 to 1 hour. Absolute bioavailability was 43%. On average, 84% of total radioactivity in plasma reflected the parent compound. The remainder of total radioactivity could be ascribed to the glucuronide conjugate of telmisartan, which represented the only metabolite in man. About 99.5% of telmisartan was bound to plasma protein, mainly to albumin and alpha-1-acid glycoprotein. Telmisartan was reversibly distributed into erythrocytes. More than 90% of administered dose was excreted within 120 hours, and the excretion balance was complete 144 hours after dosing. Radioactivity was almost exclusively (> 98%) excreted via the feces; urinary excretion accounted for < 1% of the dose, irrespective of the route of administration. In the small fraction excreted into urine, the glucuronide conjugate of telmisartan was predominant. Although some telmisartan glucuronide was detected in plasma, only unchanged drug was identified in the feces. No changes in vital signs, electrocardiogram, or clinical laboratory tests were detected following telmisartan administration, and adverse events, predominantly unrelated to treatment and of mild intensity, were infrequent. One subject fainted and, on another occasion, reported faintness; these events were probably due to the antihypertensive action of the intravenous study medication.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacokinetics , Benzimidazoles/pharmacokinetics , Benzoates/pharmacokinetics , Absorption , Administration, Oral , Adolescent , Adult , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Angiotensin-Converting Enzyme Inhibitors/blood , Angiotensin-Converting Enzyme Inhibitors/chemistry , Angiotensin-Converting Enzyme Inhibitors/urine , Benzimidazoles/adverse effects , Benzimidazoles/blood , Benzimidazoles/chemistry , Benzimidazoles/urine , Benzoates/adverse effects , Benzoates/blood , Benzoates/chemistry , Benzoates/urine , Carbon Radioisotopes , Humans , Infusions, Intravenous , Male , Middle Aged , Structure-Activity Relationship , Telmisartan
12.
Am J Trop Med Hyg ; 62(4): 524-9, 2000 Apr.
Article in English | MEDLINE | ID: mdl-11220772

ABSTRACT

The efficacy and safety of intramuscular artemotil (ARTECEF) was compared to intravenous quinine in African children with cerebral malaria. This prospective block randomized open-label study was conducted at two centers in Zambia. Subjects were children aged 0 to 10 years of age with cerebral malaria and a Blantyre Coma Score of 2 or less. Ninety two children were studied; 48 received artemotil and 44 quinine. No significant differences in survival, coma resolution time, neurologic sequelae, parasite clearance time, and fever resolution time were seen between the two regimens. Rates for negative malaria smears one month after therapy were similar in both groups. Artemotil was a well-tolerated drug in the 48 patients in this study. It appears to be at least therapeutically equivalent to quinine for the treatment of pediatric cerebral malaria. It has the advantage of being able to be given intramuscularly once daily for only five days.


Subject(s)
Antimalarials/therapeutic use , Artemisinins , Malaria, Cerebral/drug therapy , Sesquiterpenes/therapeutic use , Antimalarials/administration & dosage , Antimalarials/adverse effects , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Injections, Intramuscular , Malaria, Cerebral/mortality , Male , Prospective Studies , Quinine/adverse effects , Quinine/therapeutic use , Sesquiterpenes/administration & dosage , Sesquiterpenes/adverse effects , Survival Rate , Zambia/epidemiology
13.
Semin Thromb Hemost ; 25 Suppl 2: 51-4, 1999.
Article in English | MEDLINE | ID: mdl-10440424

ABSTRACT

The safety, and the pharmacodynamic and pharmacokinetic compatibility of clopidogrel, 75 mg daily, with the cardiac glycoside digoxin, were assessed in 12 healthy male subjects who took digoxin 0.25 mg once daily for 20 days and, in addition, clopidogrel 75 mg once daily from day 11 to day 20, so as to achieve steady-state conditions with both drugs. The drugs were taken after an overnight fast, and a standardized breakfast was served 30 minutes later. Blood samples for digoxin determination were drawn pre-dose on days 1, 8, 9, 10, 18, 19, and 20 of the schedule, and at 0.5, 1, 2, 3, 4, 5, 6, 8, 12, 16, and 24 hours post-dose on days 10 and 20. Urine samples were collected pre-dose and from 0-4, 4-8, 8-12, and 12-24 hours post-dose on days 10 and 20. Platelet aggregation studies were carried out using ADP at 5 micromol/L final concentration as an agonist. Establishment of steady-state plasma concentrations of digoxin on days 8-11 and 18-21 was confirmed by application of Dunnett's test on the trough plasma concentrations. The plasma pharmacokinetics and urinary excretions of digoxin for day 10 and day 20 were very similar: the day 20/day 10 ratios (90% Cl) were 1.1 (0.99; 1.24) for Cmax, 1.0 (0.92; 1.08) for Cmin, 1.02 (0.96; 1.07) for AUC(0-24), and 0.99 (0.94; 104) for urinary excretion. Mean inhibition of ADP-induced platelet aggregation at the end of the clopidogrel treatment period was 34%. The clinical, cardiac, and biological evidence from the study indicated that clopidogrel administration does not enhance digoxin's cardiac effects. Overall, the data indicated that there is no reason to anticipate an interaction when clopidogrel is added to digoxin for long-term management of patients with cardiac disease.


Subject(s)
Digoxin/pharmacology , Digoxin/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/pharmacokinetics , Ticlopidine/analogs & derivatives , Adolescent , Adult , Clopidogrel , Digoxin/adverse effects , Drug Combinations , Drug Interactions , Humans , Male , Platelet Aggregation Inhibitors/adverse effects , Ticlopidine/adverse effects , Ticlopidine/pharmacokinetics , Ticlopidine/pharmacology
14.
J Clin Pharmacol ; 39(5): 480-6, 1999 May.
Article in English | MEDLINE | ID: mdl-10234595

ABSTRACT

The pharmacokinetics and tolerability of the novel antiexcitatory agent, riluzole, were compared in 18 healthy elderly and 18 healthy gender- and weight-matched young volunteers. All participants received riluzole 50 mg twice daily (the recommended dosage for patients with amyotrophic lateral sclerosis), administered orally for 5 days. The pharmacokinetics of riluzole, determined on the morning of the 5th day of dosing, were not significantly affected by age or gender. The mean terminal elimination half-life (t1/2), however, was statistically significant between elderly and young subjects. Riluzole was well tolerated upon repeat dose administration. Headache was the most frequent adverse event reported, and there was no overt difference in the type, frequency, or severity of adverse events between elderly and young volunteers or between genders. In conclusion, these results indicate that no dosage adjustments of riluzole are required in the elderly.


Subject(s)
Aging/metabolism , Excitatory Amino Acid Antagonists/adverse effects , Excitatory Amino Acid Antagonists/pharmacokinetics , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacokinetics , Riluzole/adverse effects , Riluzole/pharmacokinetics , Adolescent , Adult , Aged , Aged, 80 and over , Excitatory Amino Acid Antagonists/administration & dosage , Female , Humans , Male , Neuroprotective Agents/administration & dosage , Riluzole/administration & dosage
15.
Eur J Drug Metab Pharmacokinet ; 23(1): 45-53, 1998.
Article in English | MEDLINE | ID: mdl-9625272

ABSTRACT

Losigamone ((+/-)-(R*,S*)-5-(2-chlorophenylhydroxymethyl)-4-methoxy-2 (5H)-furanone; AO-33) is a new potential antiepileptic drug undergoing clinical development. In a crossover study, 200 mg [14C]-labelled Losigamone, as well as 100 mg of each of the unlabelled enantiomers, was administered to 5 healthy volunteers as an oral suspension. The objectives of the study were to determine the mode of elimination, the excretion balance, metabolic profile, the in vitro and in vivo binding to plasma proteins and the pharmacokinetics of both enantiomers in plasma. From the plasma concentration-time profiles of [14C]-radioactivity and unchanged Losigamone it can be concluded that the absorption of Losigamone occurs very rapidly and the plasma concentration of the parent compound versus total radioactivity was consistently about 40%. An overall recovery of total radioactivity of about 97% with 85% in urine and 12% in faeces was found. Protein binding was 50%. Losigamone was extensively metabolized, with only traces of unchanged drug found in urine. The predominant metabolic pathways are hydroxylation and conjugation. After administration of the pure enantiomers, significant differences in pharmacokinetics were observed. The mean oral clearance of the (-)-enantiomer was 1863 ml/min and of the (+)-enantiomer was 171 ml/min. There was no chiral inversion after administration of the enantiomers.


Subject(s)
Anticonvulsants/pharmacokinetics , Furans/pharmacokinetics , Administration, Oral , Adult , Blood Proteins/metabolism , Carbon Radioisotopes , Cross-Over Studies , Furans/administration & dosage , Humans , Male , Protein Binding , Stereoisomerism
16.
Clin Pharmacol Ther ; 60(2): 124-37, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8823230

ABSTRACT

INTRODUCTION: Bosentan (Ro 47-0203) is a potent and mixed ETA-and ETB-receptor antagonist. Its activity has been studied in a variety of preclinical disease models. METHODS: Two double-blind placebo-controlled studies were performed to investigate the pharmacokinetics and pharmacodynamics of bosentan after single oral and intravenous doses in healthy volunteers; doses of 3, 10, 30, 100, 300, 600, 1200, and 2400 mg were given in a single ascending oral dose study, and doses of 10, 50, 250, 500, and 750 mg were given in a single ascending intravenous dose study (six subjects received active drug and two received placebo at each dose level). In an open-label crossover added to the second study, six subjects received a single oral dose of 600 mg and a single intravenous dose of 250 mg in randomized order. At regular intervals, blood pressure, pulse rate, and skin responses to intradermally injected endothelin-1 (ET-1) were recorded, and plasma levels of ET-1, proendothelin-1 (big ET-1), and ET-3, and drug and urinary levels of ET-1 and drug were determined. RESULTS: Systemic plasma clearance and volume of distribution decreased with increasing dose to limiting values of around 6 L/hr and 0.2 L/kg, respectively. The absolute bioavailability was 50% and appeared to decrease with doses above 600 mg. Plasma ET-1 increased maximally twofold (oral) and threefold (intravenous), and this increase was directly related to bosentan plasma concentrations according to an Emax model. Bosentan reversed the vasoconstrictor effect of ET-1 measured in the skin microcirculation. There was a tendency toward decreased blood pressure (approximately 5 mm Hg) and increased pulse rate (approximately 5 beats/min), neither was clearly dose dependent. Oral bosentan was well tolerated. Vomiting and local intolerability was observed at the higher intravenous doses. CONCLUSION: Bosentan is an orally bioavailable, well-tolerated, and active ET-1 antagonist with a low clearance and a moderate volume of distribution. Its intravenous use is limited because of local intolerability.


Subject(s)
Endothelin Receptor Antagonists , Endothelins/blood , Sulfonamides/pharmacokinetics , Administration, Oral , Adult , Analysis of Variance , Area Under Curve , Biological Availability , Blood Pressure/drug effects , Bosentan , Double-Blind Method , Endothelin-1/blood , Endothelin-3/blood , Endothelins/administration & dosage , Heart Rate/drug effects , Humans , Injections, Intradermal , Injections, Intravenous , Male , Protein Precursors/blood , Reference Values , Skin/drug effects , Sulfonamides/administration & dosage , Sulfonamides/chemistry , Sulfonamides/pharmacology
17.
Eur J Clin Pharmacol ; 51(2): 175-81, 1996.
Article in English | MEDLINE | ID: mdl-8911885

ABSTRACT

OBJECTIVES: A double-blind, placebo-controlled study was conducted to assess the pharmacokinetics and pharmacodynamics of the bisphosphonate tiludronic acid, administered once daily as sodium tiludronate 200, 400, 600 and 800 mg for 12 days. Four groups of ten subjects participated in the study, with a drug to placebo ratio of 4:1. METHODS: Pre-dose blood samples were taken on alternate days, starting on Day 1 and additional samples were collected over 144 h following the final dose on Day 12. Urine was collected over 24 h after the final dose. Indices of calcium homeostasis and biochemical markers of bone turnover were assessed during the study as pharmacodynamic parameters. Tolerability was evaluated with special emphasis on renal function and gastrointestinal irritation. Adverse experiences were assessed at regular time intervals. RESULTS AND CONCLUSIONS: Steady state was attained from Day 4 (200 mg) or from Day 6 (400, 600 and 800 mg). Following the final dose on Day 12, minimal plasma concentrations (Cmin) ranged between 0.19 and 1.5 mg.1(-1), and maximal plasma concentrations (Cmax) between 1.1 and 7.8 mg.1(-1) for the lowest and highest doses, respectively. A supra-proportional increase in Cmax, AUC24 and Ae24 with dose was observed. There was a linear relationship between the plasma tiludronic acid and its urinary excretion rate, so, the disproportional rise in Cmax and AUC24 with increasing dose could not be attributed to saturation of renal excretion. Certain indices of calcium homeostasis changed significantly during the study, but generally, became only prominent at the highest dose level of 800 mg. Total serum calcium and the urinary calcium/creatinine clearance ratio fell, indicating depression of osteoclastic bone resorption, which was not revealed by serum osteocalcin levels probably because of the brevity of the treatment (12 days). In response to the decline in serum calcium, serum 1,25-dihydroxyvitamin D3 and intact PTH (1-84) levels increased. None of the safety parameters raised any concerns about the safety of sodium tiludronate administered in this way.


Subject(s)
Diphosphonates/administration & dosage , Diphosphonates/pharmacokinetics , Adolescent , Adult , Bone Resorption , Calcium/metabolism , Diphosphonates/pharmacology , Double-Blind Method , Drug Administration Schedule , Humans , Male , Phosphates/metabolism
18.
Ann Allergy Asthma Immunol ; 75(2): 157-61, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7648380

ABSTRACT

BACKGROUND: There is a close relationship between improvement in airway function and the plasma concentration of theophylline, as well as between rapidly rising plasma theophylline concentrations and increased frequency of undesired effects. Development of pharmaceutical formulations and prescribed dosage intervals for theophylline dosage forms should therefore be directed toward providing the most stable plasma concentrations attainable. OBJECTIVE: To characterize the steady-state biopharmaceutic profile of Uni-Dur following once-daily or twice-daily administration. METHODS: Twenty-four adult male volunteers with average theophylline clearance (3.0 and 5.5 L.h-1) received three treatments on separate occasions: Uni-Dur 800 mg once-daily, Uni-Dur 400 mg twice-daily, and Uniphyl 800 mg once-daily. Treatments were taken after a meal for five days with at least 1 week washout between treatment periods. Trough blood samples were collected prior to the AM dose on days 3, 4, and 5, and at specified intervals up to 48 hours after the AM dose on day 5 for subsequent determination of theophylline concentrations in plasma. RESULTS: The area under the plasma concentration-time curve (AUC; microgram.mL-1.h) for theophylline over 24 hours on day 5 was 187 for Uni-Dur 800 mg once-daily, 187 for Uni-Dur 400 mg twice-daily, and 172 for Uniphyl 800 mg once-daily; the peak plasma concentrations were 10.4, 9.4, and 11.0 micrograms.mL-1 and the trough concentrations were 5.5, 7.2, and 3.5 micrograms.mL-1, respectively; fluctuation index (peak minus trough divided by trough) was 78%, 16%, and 231%, respectively. No further accumulation of theophylline occurred after day 3. No serious nor severe adverse events were reported during any treatment. CONCLUSIONS: Uni-Dur is an extended-release formulation that provides stable plasma concentrations of theophylline over a 24-hour period with less fluctuation than observed with a once-daily reference formulation. In subjects with normal theophylline clearance, Uni-Dur administered twice-daily provided remarkably stable theophylline plasma concentrations over a 24-hour period. Absorption of theophylline from Uni-Dur was not affected by food, and no evidence of dose-dumping was observed. Uni-Dur should provide efficacious theophylline therapy with minimal adverse events in patients with symptoms of asthma and reversible bronchospasm associated with chronic bronchitis and emphysema.


Subject(s)
Theophylline/pharmacokinetics , Adult , Cross-Over Studies , Delayed-Action Preparations , Drug Administration Schedule , Humans , Male , Metabolic Clearance Rate , Theophylline/administration & dosage
19.
J Clin Psychopharmacol ; 15(1): 41-8, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7714227

ABSTRACT

The combination of classic monoamine oxidase inhibitors (MAOIs) and tricyclic antidepressant drugs (TCAs) has been associated with a variety of adverse events. A switch in treatment from TCAs to moclobemide, a reversible and selective inhibitor of MAO-A, was investigated in a double-blind, placebo-controlled study in healthy volunteers. Two groups of 12 subjects were treated with either amitriptyline (75 mg/day) or clomipramine (100 mg/day) until steady-state conditions had been attained (14 days). Treatment with the TCAs was discontinued abruptly and switched to either a therapeutic dose regimen of moclobemide (300 mg/day) or placebo. The tolerability and safety pattern did not reveal any clinically relevant differences between moclobemide and placebo recipients, nor was there any sign of a pharmacokinetic interaction between the TCAs and moclobemide. In conclusion, the findings of this study suggest that therapeutic doses of moclobemide up to 300 mg daily can be given 24 hours after the last dose of treatment with either amitriptyline or clomipramine without major risks.


Subject(s)
Antidepressive Agents, Tricyclic/therapeutic use , Antidepressive Agents/therapeutic use , Benzamides/therapeutic use , Depressive Disorder/drug therapy , Monoamine Oxidase Inhibitors/therapeutic use , Adolescent , Adult , Antidepressive Agents/pharmacokinetics , Benzamides/pharmacokinetics , Female , Humans , Male , Moclobemide , Monoamine Oxidase Inhibitors/pharmacokinetics
20.
Aliment Pharmacol Ther ; 9(1): 51-6, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7766744

ABSTRACT

AIMS: To compare the effects on intragastric acidity of a single evening dose of either standard or effervescent formulations of ranitidine (300 mg) or cimetidine (800 mg). METHODS: Twelve healthy subjects were studied, using a four-period randomized cross-over design and an ambulatory intragastric pH monitoring technique. The subjects received a standard evening meal at 17.00 hours and one of the H2-receptor antagonist formulations was given at 23.00 hours. RESULTS: Both effervescent formulations caused a transient rapid increase in intragastric pH, reaching a maximum at about 3 min after ingestion. After both effervescent formulations a significantly higher pH was measured during the first 45 min after ingestion (P < 0.05), compared to the regular formulations. The onset of action of the H2-receptor antagonists was similar for both formulations of ranitidine and the effervescent cimetidine, but tended to be slower for the regular cimetidine (P = 0.06). Nocturnal intragastric pH was significantly increased by all four formulations, but more effectively so by the two ranitidine formulations. The duration of action (taken as time with pH > 4) of both ranitidine formulations was longer than that of both cimetidine formulations (P < 0.002). CONCLUSIONS: A single evening dose of 300 mg ranitidine produces a stronger decrease of nocturnal gastric acid secretion than 800 mg cimetidine. The effervescent formulations of both drugs offer the advantage of a rapid decrease (within minutes) of intragastric acidity, with preservation of the sustained systemic effect.


Subject(s)
Cimetidine/pharmacology , Gastric Acid/metabolism , Gastric Mucosa/drug effects , Ranitidine/pharmacology , Adolescent , Adult , Cimetidine/administration & dosage , Cross-Over Studies , Drug Delivery Systems , Female , Gastric Acidity Determination , Gastric Mucosa/metabolism , Humans , Hydrogen-Ion Concentration , Male , Ranitidine/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL