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










Publication year range
1.
Nat Med ; 28(7): 1432-1438, 2022 07.
Article in English | MEDLINE | ID: mdl-35725922

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is a common chronic liver disease that may advance to fibrosis and lead to mortality; however, no pharmacotherapy is currently available. We tested the hypothesis that inhibition of both the sodium-glucose cotransporters 1 and 2 with licogliflozin would lead to improvement in NASH. A total of 107 patients with phenotypic or histologic NASH were randomized (1:2:2) to receive oral administration of either placebo (n = 21), licogliflozin 30 mg (n = 43) or 150 mg (n = 43) once daily for 12 weeks. Licogliflozin 150 mg showed a significant 32% (80% confidence interval (CI): 21-43%; P = 0.002) placebo-adjusted reduction in serum alanine aminotransferase after 12 weeks of treatment, the primary endpoint of the study. However, the 30 mg dose of licogliflozin did not meet the primary endpoint (placebo-adjusted reduction 21% (80% CI: 7-32%; P = 0.061)). Diarrhea occurred in 77% (33 of 43), 49% (21 of 43) and 43% (9 of 21) of patients treated with licogliflozin 150 mg, 30 mg and placebo, respectively, which was mostly mild in severity. No other major safety concerns were identified. Treatment with 150 mg licogliflozin led to reductions in serum alanine aminotransferase in patients with NASH. Studies of longer duration and in combination with drugs that have different mechanisms of action are needed to validate these findings and to define a role of licogliflozin as a therapeutic option for NASH. ClinicalTrials.gov identifier: NCT03205150.


Subject(s)
Non-alcoholic Fatty Liver Disease , Alanine Transaminase , Anhydrides/pharmacology , Anhydrides/therapeutic use , Double-Blind Method , Humans , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Sorbitol/analogs & derivatives , Sorbitol/pharmacology , Sorbitol/therapeutic use , Treatment Outcome
2.
Clin Transl Sci ; 15(1): 158-171, 2022 01.
Article in English | MEDLINE | ID: mdl-34498388

ABSTRACT

Chronic antihypertensive treatment often includes combination of two or more therapies with complementary mechanism of action targeting different blood pressure (BP) control system. If available, these components are recommended to be administered as a fixed-dose combination (FDC) to reduce tablet burden, improve adherence and thus BP control. A combination of ramipril (RAMI) and bisoprolol (BISO) is one of the options used in clinical practice and is supported by therapeutic guidelines. The clinical program for a novel BISO/RAMI FDC consisted of two randomized, open-label, bioequivalence (BE) studies and one drug-drug interaction (DDI) study. The BE was examined between two FDC strengths of BISO/RAMI (10/10 and 10/5 mg) and the individual reference products administered concomitantly at respective doses after a single oral dose under fasting conditions. In both BE studies, 64 healthy subjects were randomized according to a two-way crossover design. The DDI study evaluated a potential pharmacokinetic (PK) interaction between BISO 10 mg and RAMI 10 mg following their single or concomitant administrations in 30 healthy subjects under fasting condition. BE for BISO/RAMI 10/5 mg and absence of a clinically relevant PK DDI between BISO and RAMI was demonstrated as the 90% confidence intervals (CIs) of the geometric mean ratios (GMRs) for area under the concentration time curve (AUC) and maximum concentration (Cmax ) remained within the acceptance range of 80.00 to 125.00%. However, BE for BISO/RAMI 10/10 mg was not demonstrated, as the lower bound of the 90% CI of Cmax for RAMI was outside the acceptance range of BE. Both drugs administered alone or combined were well-tolerated. No PK interaction was observed between BISO and RAMI/ramiprilat, since the co-administration of BISO and RAMI 10 mg single doses resulted in comparable rate and extent of absorption for BISO and RAMI when compared to their individual products.


Subject(s)
Antihypertensive Agents/administration & dosage , Bisoprolol/administration & dosage , Drug Interactions , Drug Therapy, Combination , Ramipril/administration & dosage , Therapeutic Equivalency , Adolescent , Adult , Female , Humans , Male , Middle Aged , Young Adult
4.
Clin Drug Investig ; 38(9): 819-827, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30008052

ABSTRACT

BACKGROUND AND OBJECTIVE: Co-Crystal of Tramadol-Celecoxib (CTC), in development for the treatment of moderate to severe acute pain, is a first-in-class co-crystal containing a 1:1 molecular ratio of two active pharmaceutical ingredients; rac-tramadol·HCl and celecoxib. This randomised, open-label, crossover study compared the bioavailability of both components after CTC administration under fed and fasting conditions. METHODS: Healthy adults received single doses of 200 mg CTC under both fed and fasting conditions (separated by a 7-day washout). Each dose of CTC was administered orally as two 100 mg tablets, each containing 44 mg tramadol·HCl and 56 mg celecoxib. In the fed condition, a high-fat, high-calorie meal [in line with recommendations by the US Food and Drug Administration (FDA)] was served 30 min before CTC administration. Tramadol, O-desmethyltramadol and celecoxib plasma concentrations were measured pre- and post-dose up to 48 h. Pharmacokinetic parameters were calculated using non-compartmental analysis. Safety was also assessed. RESULTS: Thirty-six subjects (18 female/18 male) received one or both doses of CTC; 33 provided evaluable pharmacokinetic data under fed and fasting conditions. For tramadol and O-desmethyltramadol, fed-to-fasting ratios of geometric least-squares means and corresponding 90% confidence interval (CI) values for maximum plasma concentration (Cmax) and extrapolated area under the plasma concentration-time curve to infinity (AUC∞) were within the pre-defined range for comparative bioavailability (80-125%). For celecoxib, Cmax and AUC∞ fed-to-fasting ratios (90% CIs) were outside this range, at 130.91% (116.98-146.49) and 129.34% (121.78-137.38), respectively. The safety profile of CTC was similar in fed and fasting conditions. CONCLUSIONS: As reported for standard-formulation celecoxib, food increased the bioavailability of celecoxib from single-dose CTC. Food had no effect on tramadol or O-desmethyltramadol bioavailability. CLINICAL TRIAL REGISTRATION NUMBER: 152052 (registered with the Therapeutic Products Directorate of Health Canada).


Subject(s)
Analgesics, Opioid/pharmacokinetics , Celecoxib/pharmacokinetics , Cyclooxygenase 2 Inhibitors/pharmacokinetics , Food-Drug Interactions , Tramadol/pharmacokinetics , Adolescent , Adult , Analgesics, Opioid/adverse effects , Analgesics, Opioid/blood , Area Under Curve , Biological Availability , Celecoxib/adverse effects , Celecoxib/blood , Cross-Over Studies , Cyclooxygenase 2 Inhibitors/adverse effects , Cyclooxygenase 2 Inhibitors/blood , Drug Combinations , Fasting/metabolism , Female , Healthy Volunteers , Humans , Male , Middle Aged , Tramadol/adverse effects , Tramadol/analogs & derivatives , Tramadol/blood , Young Adult
5.
Diabetes Obes Metab ; 20(3): 646-653, 2018 03.
Article in English | MEDLINE | ID: mdl-29053231

ABSTRACT

AIMS: Nasal glucagon (NG) is a nasally-administered glucagon powder, absorbed through the nasal mucosa, designed for treatment of severe hypoglycaemia. This study evaluated the safety, pharmacokinetics (PK) and pharmacodynamics (PD) of NG in otherwise healthy participants with common colds and after recovery from cold symptoms, with and without concomitant nasal decongestant. MATERIALS AND METHODS: This was a single-centre, open-label study. Cohort 1 participants (N = 18) received 2 doses of NG: one while experiencing nasal congestion and another after recovery from cold symptoms. Cohort 2 participants (N = 18), who also had colds with nasal congestion, received a single dose of NG 2 hours after treatment with the decongestant oxymetazoline. Total symptoms score and other safety measures were assessed before and after NG administration. RESULTS: NG was well tolerated, without serious adverse events. Common adverse events (transient lacrimation, nasal discomfort, rhinorrhea and nausea) were more frequent in both Cohorts 1 and 2 during nasal congestion. Glucagon levels peaked 18 minutes post-dose and glucose levels peaked 30 to 42 minutes post-dose in all groups. Nasal congestion, with or without concomitant nasal decongestant, did not significantly affect PK of NG. Although glucose AUECs0-t was different between Cohort 1 with nasal congestion and Cohort 2, glucose concentrations at 30 minutes appeared similar in all groups. CONCLUSIONS: There were no clinically relevant differences in safety or PK/PD of NG associated with nasal congestion or concomitant administration of nasal decongestant, suggesting that NG can be used to treat severe hypoglycaemia in individuals experiencing nasal congestion.


Subject(s)
Common Cold/complications , Gastrointestinal Agents/administration & dosage , Glucagon/administration & dosage , Nasal Decongestants/pharmacology , Administration, Intranasal , Adult , Blood Glucose/drug effects , Blood Glucose/metabolism , Female , Gastrointestinal Agents/pharmacokinetics , Gastrointestinal Agents/pharmacology , Glucagon/pharmacokinetics , Glucagon/pharmacology , Healthy Volunteers , Humans , Hypoglycemia/drug therapy , Male , Nasal Obstruction/complications , Powders
6.
Br J Clin Pharmacol ; 84(1): 64-78, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28888220

ABSTRACT

AIM: We compared the pharmacokinetic (PK) profiles of co-crystal of tramadol-celecoxib (CTC) vs. each reference product (alone and in open combination) after single (first dose) and multiple dosing. METHODS: Healthy adults aged 18-50 years received, under fasted conditions, 15 twice-daily doses of the following treatments (separated by ≥14-day washout): 200 mg immediate-release (IR) CTC (equivalent to 88 mg tramadol and 112 mg celecoxib; treatment 1); 100 mg IR tramadol (treatment 2), 100 mg celecoxib (treatment 3); and 100 mg IR tramadol and 100 mg celecoxib (treatment 4). The treatment sequence was assigned by computer-generated randomization. PK parameters were calculated using non-compartmental analysis. Parameters for CTC were adjusted according to reference product dose. RESULTS: A total of 30 subjects (20 males, mean age 35 years) were included. Multiple-dose tramadol PK parameters for treatments 1, 2 and 4, respectively, were 551, 632 and 661 ng ml-1 [mean maximum plasma concentration (Cmax )]; 4796, 4990 and 5284 ng h ml-1 (area under the plasma concentration-time curve over the dosing interval at steady state); and 3.0, 2.0 and 2.0 h (median time to Cmax at steady state). For treatments 1, 3 and 4, multiple-dose celecoxib PK parameters were 445, 536 and 396 ng ml-1 ; 2803, 3366 and 2897 ng h ml-1 ; and 2.0, 2.0 and 3.0 h. Single-dose findings were consistent with multiple-dose data. Types of adverse events were consistent with known reference product safety profiles. CONCLUSION: After single (first dose) and multiple dosing, PK parameters for each active pharmaceutical ingredient in CTC were modified by co-crystallization compared with reference products alone or in open combination.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Celecoxib/pharmacokinetics , Cyclooxygenase 2 Inhibitors/pharmacokinetics , Drug Compounding/methods , Tramadol/pharmacokinetics , Adult , Analgesics, Opioid/chemistry , Analgesics, Opioid/therapeutic use , Area Under Curve , Celecoxib/chemistry , Celecoxib/therapeutic use , Cross-Over Studies , Crystallization/methods , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2 Inhibitors/therapeutic use , Drug Administration Schedule , Drug Combinations , Fasting , Female , Healthy Volunteers , Humans , Male , Pain/drug therapy , Tramadol/chemistry , Tramadol/therapeutic use
7.
Br J Clin Pharmacol ; 83(12): 2718-2728, 2017 12.
Article in English | MEDLINE | ID: mdl-28810061

ABSTRACT

AIMS: Co-crystal of tramadol-celecoxib (CTC) is a novel co-crystal molecule containing two active pharmaceutical ingredients under development by Esteve (E-58425) and Mundipharma Research (MR308). This Phase I study compared single-dose pharmacokinetics (PK) of CTC with those of the individual reference products [immediate-release (IR) tramadol and celecoxib] alone and in open combination. METHODS: Healthy adults aged 18-55 years were orally administered four treatments under fasted conditions (separated by 7-day wash-out period): 200 mg IR CTC (equivalent to 88 mg tramadol and 112 mg celecoxib; Treatment 1); 100 mg IR tramadol (Treatment 2); 100 mg celecoxib (Treatment 3); and 100 mg IR tramadol and 100 mg celecoxib (Treatment 4). Treatment sequence was assigned using computer-generated randomization. PK parameters were calculated using noncompartmental analysis with parameters for CTC adjusted according to reference product dose (100 mg). RESULTS: Thirty-six subjects (28 male, mean age 36 years) participated. Tramadol PK parameters for Treatments-1, -2 and -4, respectively, were 263, 346 and 349 ng ml-1 (mean maximum plasma concentration); 3039, 2979 and 3119 ng h ml-1 (mean cumulative area under the plasma concentration-time curve); and 2.7, 1.8 and 1.8 h (median time to maximum plasma concentration). For Treatments 1, 3 and 4, the respective celecoxib PK parameters were 313, 449 and 284 ng ml-1 ; 2183, 3093 and 2856 ng h ml-1 ; and 1.5, 2.3 and 3.0 h. No unexpected adverse events were reported. CONCLUSION: PK parameters of each API in CTC were modified by co-crystallization compared with marketed formulations of tramadol, celecoxib, and their open combination.


Subject(s)
Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Celecoxib/administration & dosage , Celecoxib/pharmacokinetics , Cyclooxygenase 2 Inhibitors/administration & dosage , Cyclooxygenase 2 Inhibitors/pharmacokinetics , Tramadol/administration & dosage , Tramadol/pharmacokinetics , Administration, Oral , Adolescent , Adult , Analgesics, Opioid/blood , Analgesics, Opioid/chemistry , Area Under Curve , Celecoxib/chemistry , Cross-Over Studies , Crystallization , Cyclooxygenase 2 Inhibitors/blood , Cyclooxygenase 2 Inhibitors/chemistry , Drug Administration Schedule , Drug Combinations , Drug Compounding , Female , Half-Life , Healthy Volunteers , Humans , Male , Metabolic Clearance Rate , Middle Aged , Models, Biological , Quebec , Tramadol/chemistry , Young Adult
8.
Hum Psychopharmacol ; 32(4)2017 07.
Article in English | MEDLINE | ID: mdl-28568608

ABSTRACT

OBJECTIVE: The objective of this study was to determine the next-day residual effects of acute and steady-state nighttime dosing of flibanserin on simulated driving performance and cognitive function in healthy premenopausal women. METHODS: In this randomized, double-blind, placebo-controlled, four-way crossover study, 72 subjects were treated with either acute oral doses of placebo, zopiclone 7.5 mg (positive control) or flibanserin 100 mg at bedtime (indicated therapeutic dose), or after chronic nightly oral doses of flibanserin 100 mg for 1 week followed by a single bedtime dose of flibanserin 200 mg (supratherapeutic dose). Simulated driving assessments were conducted 9 hr after dosing and cognitive function tests were administered immediately before or during the driving assessment. RESULTS: Zopiclone increased standard deviation of lateral position (≥3.1 cm; p < .0001) relative to placebo and impaired other parameters previously shown to be sensitive to sedation. No impairment was detected for flibanserin at either dose relative to placebo. Flibanserin 200 mg was similar to the 100-mg dose on cognitive testing and driving performance even though commonly reported adverse events for flibanserin were predictably increased at the higher dose. CONCLUSIONS: At both therapeutic and supratherapeutic doses, flibanserin did not impair next-day driving performance and cognitive function compared to placebo.


Subject(s)
Automobile Driving , Benzimidazoles/adverse effects , Cognition/drug effects , Hypnotics and Sedatives/adverse effects , Administration, Oral , Adult , Azabicyclo Compounds/adverse effects , Benzimidazoles/administration & dosage , Contraceptives, Oral, Hormonal/therapeutic use , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Hypnotics and Sedatives/administration & dosage , Piperazines/adverse effects , Premenopause , Reaction Time/drug effects , Sexual Dysfunctions, Psychological/drug therapy , User-Computer Interface
9.
Article in English | MEDLINE | ID: mdl-28052855

ABSTRACT

SYN-004 (ribaxamase) is a ß-lactamase designed to be orally administered concurrently with intravenous ß-lactam antibiotics, including most penicillins and cephalosporins. Ribaxamase's anticipated mechanism of action is to degrade excess ß-lactam antibiotic that is excreted into the small intestine. This enzymatic inactivation of excreted antibiotic is expected to protect the gut microbiome from disruption and thus prevent undesirable side effects, including secondary infections such as Clostridium difficile infections, as well as other antibiotic-associated diarrheas. In phase 1 clinical studies, ribaxamase was well tolerated compared to a placebo group and displayed negligible systemic absorption. The two phase 2a clinical studies described here were performed to confirm the mechanism of action of ribaxamase, degradation of ß-lactam antibiotics in the human intestine, and were therefore conducted in subjects with functioning ileostomies to allow serial sampling of their intestinal chyme. Ribaxamase fully degraded ceftriaxone to below the level of quantitation in the intestines of all subjects in both studies. Coadministration of oral ribaxamase with intravenous ceftriaxone was also well tolerated, and the plasma pharmacokinetics of ceftriaxone were unchanged by ribaxamase administration. Since ribaxamase is formulated as a pH-dependent, delayed-release formulation, the activity of ribaxamase in the presence of the proton pump inhibitor esomeprazole was examined in the second study; coadministration of these drugs did not adversely affect ribaxamase's ability to degrade ceftriaxone excreted into the intestine. These studies have confirmed the in vivo mechanism of action of ribaxamase, degradation of ß-lactam antibiotics in the human intestine (registered at ClinicalTrials.gov under NCT02419001 and NCT02473640).


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Ceftriaxone/pharmacokinetics , Dysbiosis/prevention & control , Inactivation, Metabolic , Protective Agents/pharmacokinetics , Recombinant Proteins/pharmacokinetics , beta-Lactamases/pharmacokinetics , Administration, Oral , Drug Administration Schedule , Humans , Ileostomy , Infusions, Intravenous , Intestinal Absorption , Intestinal Mucosa/metabolism , Intestines/drug effects
10.
Eur J Contracept Reprod Health Care ; 22(6): 429-438, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29336615

ABSTRACT

OBJECTIVE: To show the clinical development of Ornibel® (ExeltisHealthcare, Spain) a contraceptive vaginal ring manufactured with a new polymer composition and containing etonogestrel/ethinylestradiol, compared to Nuvaring® (MSD, Spain). SUBJECTS AND METHODS: Randomised, single dose, 2-period, 2-sequence, 2-stage crossover, comparative bioavailability study conducted in 40 healthy female subjects. All subjects received both treatments for 28 days in each of two periods, separated by a 28 days washout. Ornibel® contains etonogestrel/ethinylestradiol 11.00/3.47 mg and Nuvaring® contains etonogestrel/ethinylestradiol 11.7/2.7 mg, both rings delivering 120/15 µg/day. For the calculation of pharmacokinetic parameters, 37 blood samples were collected up to 840 h after each ring insertion to quantify plasma concentrations of etonogestrel and ethinylestradiol using a validated MS/MS-HPLC. Safety was assessed by adverse events recording, clinical laboratory and vital signs and tolerability by vaginal examination. Acceptability was investigated by a 5-point scale questionnaire. RESULTS: Bioequivalence was demonstrated in the first stage as the 94.12% Confidence Intervals of the primary parameters laid within the 80-125% acceptance range for both etonogestrel (Cmax: 96.81-112.20%; AUC0-504h: 98.71-108.61%; AUC0-t: 100.14-109.10%) and ethinylestradiol. (Cmax: 105.91-120.62%; AUC0-504h: 105.47-114.59%; AUC0-t: 108.31-117.61%). During the first day of use a burst effect was observed with Nuvaring®, with significantly higher level of ethinylestradiol (Cmax0-24h ratio: 78.34%, 94.12CI: 73.55-83.45%). Both products were well tolerated and accepted, without significant differences between them. CONCLUSION: Ornibel® is bioequivalent to Nuvaring® in terms of efficacy, safety, tolerability and acceptability. The new polymer composition provides Ornibel® with more stability and gradual hormonal release during the first day of use, particularly for ethinylestradiol.


Subject(s)
Contraceptive Agents, Female/pharmacokinetics , Contraceptive Devices, Female , Desogestrel/pharmacokinetics , Estrogens/pharmacokinetics , Ethinyl Estradiol/pharmacokinetics , Adult , Cross-Over Studies , Desogestrel/analogs & derivatives , Drug Combinations , Female , Healthy Volunteers , Humans , Polymers , Therapeutic Equivalency , Treatment Outcome
11.
Br J Clin Pharmacol ; 83(3): 540-553, 2017 03.
Article in English | MEDLINE | ID: mdl-27763682

ABSTRACT

AIMS: To compare the levodopa/carbidopa (LC) and levodopa/benserazide (LB) pharmacokinetic profiles following repeated doses of opicapone (OPC) administered apart from levodopa. METHODS: Two randomized, double blind, sex-balanced, placebo-controlled studies in four groups of 12 or 18 healthy subjects each. In each group, enrolled subjects received a once-daily morning (5, 15 and 30 mg) or evening (5, 15 and 50 mg) administration of OPC or placebo for up to 28 days. On the morning of Day 11, 12 h after the OPC or placebo evening dose, or the morning of Day 21, 1 h after the OPC or placebo dose, a single dose of immediate-release 100/25 mg LC was administered. Similarly, on Day 18 morning, 12 h after the OPC or placebo evening dose, or Day 28 morning, 1 h after the OPC or placebo dose, a single dose of immediate-release 100/25 mg LB was administered. RESULTS: All OPC treatments, in relation to the placebo group, presented a higher extent of exposure (AUC) to levodopa following either LC or LB doses. A relevant but not dose-dependent increase in the levodopa AUC occurred with all OPC dose groups in relation to placebo. All active treatments significantly inhibited both peak (Emax ) and extent (AUEC) of the catechol-O-methyltransferase activity in relation to placebo. The tolerability profile was favourable. CONCLUSION: Opicapone, as once-daily oral evening regimen and/or 1 h apart from levodopa therapy, increases the bioavailability of levodopa associated with its pronounced, long-lasting and sustained catechol-O-methyltransferase inhibition. The tolerability profile was favourable and similar between OPC and placebo.


Subject(s)
Benserazide/pharmacokinetics , Levodopa/pharmacokinetics , Oxadiazoles/pharmacology , Oxadiazoles/pharmacokinetics , Adult , Antiparkinson Agents/pharmacokinetics , Benserazide/adverse effects , Benserazide/blood , Benserazide/pharmacology , Biological Availability , Carbidopa/adverse effects , Carbidopa/pharmacology , Catechol O-Methyltransferase Inhibitors/adverse effects , Catechol O-Methyltransferase Inhibitors/blood , Catechol O-Methyltransferase Inhibitors/pharmacokinetics , Catechol O-Methyltransferase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Double-Blind Method , Drug Combinations , Female , Humans , Levodopa/adverse effects , Levodopa/blood , Levodopa/pharmacology , Male , Middle Aged , Oxadiazoles/adverse effects , Oxadiazoles/blood
12.
Br J Clin Pharmacol ; 82(4): 994-1001, 2016 10.
Article in English | MEDLINE | ID: mdl-27276421

ABSTRACT

AIMS: In light of the growing recognition of renal disease in thalassemia, it is important to understand the impact of renal impairment on the pharmacokinetics of iron chelators. This study evaluated the pharmacokinetics and safety of the iron chelator deferiprone (DFP) in subjects with renal impairment in comparison with healthy volunteers (HVs). METHODS: Thirty-two subjects were categorized into four groups based on degree of renal impairment: none, mild, moderate or severe, as determined by estimated glomerular filtration rate (eGFR). All subjects received a single oral dose of 33 mg kg(-1) DFP, provided serum and urine samples for pharmacokinetic assessment over 24 h and were monitored for safety. RESULTS: Renal clearance of DFP decreased as renal impairment increased. However, based on Cmax , AUC(0,t) and AUC(0,∞), there were no significant group differences in systemic exposure, because less than 4% of the drug was excreted unchanged in the urine. DFP is extensively metabolized to a renally excreted, pharmacologically inactive metabolite, deferiprone 3-O-glucuronide (DFP-G), which exhibited higher Cmax , AUC(0,t), AUC(0,∞) and longer tmax and t1/2 in the renally impaired groups compared with HVs. The Cmax and AUCs of DFP-G increased as eGFR decreased. Overall, 75%-95% of the dose was retrieved in urine, either as DFP or DFP-G, regardless of severity of renal impairment. With respect to safety, DFP was well tolerated. CONCLUSIONS: These data suggest that no adjustment of the DFP dosage regimen in patients with renal impairment is necessary, as there were no significant changes in the systemic exposure to the drug.


Subject(s)
Pyridones/pharmacokinetics , Renal Insufficiency/metabolism , Administration, Oral , Adult , Aged , Deferiprone , Female , Glomerular Filtration Rate , Humans , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/adverse effects , Iron Chelating Agents/pharmacokinetics , Male , Middle Aged , Pyridones/adverse effects , Pyridones/blood , Pyridones/urine
13.
Drugs R D ; 15(2): 195-201, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25939332

ABSTRACT

AIMS: This bioequivalence study was conducted to assess the bioequivalence of two formulations, test and reference, of pregabalin 300 mg hard capsules, under fasting conditions. METHODS: This was a single-center, randomized, single-dose, open-label, laboratory-blinded, two-way crossover study, with a minimum washout period of 7 days. Plasma samples were collected prior to and up to 36 h after dosing. Pregabalin plasma concentrations were determined, using a validated method, by reversed phase high performance liquid chromatography coupled to a tandem mass spectrometry detector (LC-MS-MS). Pharmacokinetic metrics used for bioequivalence assessment were the AUC(0-t) (area under the plasma concentration-time curve from time zero to time of last observed non-zero plasma concentration) and the C max (maximum observed plasma concentration). These parameters were determined from the pregabalin plasma concentration data using noncompartmental analysis. RESULTS: Forty healthy subjects, age ranging from 18 to 43 years old, were enrolled and randomized, of whom 39 completed the study. The ratio of geometric least square means for C max was 99.29 % (90 % confidence interval [CI] 93.29-105.67). The ratio of geometric least square means for AUC(0-t) was 101.54 % (90 % CI 100.13-102.98). The 90 % CIs were within the predefined range (80.00-125.00). CONCLUSIONS: Bioequivalence between test and reference formulations, under fasting conditions, was concluded both in terms of rate and extent of absorption.


Subject(s)
Capsules/administration & dosage , Capsules/pharmacokinetics , Pregabalin/administration & dosage , Pregabalin/pharmacokinetics , Adolescent , Adult , Area Under Curve , Chemistry, Pharmaceutical/methods , Cross-Over Studies , Fasting/physiology , Healthy Volunteers , Humans , Male , Therapeutic Equivalency , Young Adult
14.
J Hepatol ; 63(2): 320-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25733157

ABSTRACT

BACKGROUND & AIMS: GS-9620 is an oral agonist of toll-like receptor 7, a pattern-recognition receptor whose activation results in innate and adaptive immune stimulation. We evaluated the safety, pharmacokinetics, and pharmacodynamics of GS-9620 in patients with chronic hepatitis B. METHODS: In two double-blind, phase 1b trials of identical design, 49 treatment-naïve and 51 virologically suppressed patients were randomized 5:1 to receive GS-9620 (at doses of 0.3mg, 1mg, 2mg, 4mg) or placebo as a single dose or as two doses seven days apart. Pharmacodynamic assessment included evaluation of peripheral mRNA expression of interferon-stimulated gene 15 (ISG15), serum interferon gamma-induced protein 10 and serum interferon (IFN)-alpha. RESULTS: Overall, 74% of patients were male and 75% were HBeAg negative at baseline. No subject discontinued treatment due to adverse events. Fifty-eight percent experienced ⩾1 adverse event, all of which were mild to moderate in severity. The most common adverse event was headache. No clinically significant changes in HBsAg or HBV DNA levels were observed. Overall, a transient dose-dependent induction of peripheral ISG15 gene expression was observed peaking within 48 hours of dosing followed by return to baseline levels within seven days. Higher GS-9620 dose, HBeAg positive status, and low HBsAg level at baseline were independently associated with greater probability of ISG15 response. Most patients (88%) did not show detectable levels of serum IFN-alpha at any time point. CONCLUSIONS: Oral GS-9620 was safe, well tolerated, and associated with induction of peripheral ISG15 production in the absence of significant systemic IFN-alpha levels or related symptoms.


Subject(s)
Hepatitis B, Chronic/drug therapy , Pteridines/administration & dosage , Toll-Like Receptor 7/agonists , Administration, Oral , Adolescent , Adult , Aged , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , DNA, Viral/analysis , Dose-Response Relationship, Drug , Double-Blind Method , Female , Follow-Up Studies , Genotype , Hepatitis B Surface Antigens/analysis , Hepatitis B virus/genetics , Hepatitis B virus/immunology , Hepatitis B, Chronic/metabolism , Hepatitis B, Chronic/virology , Humans , Male , Middle Aged , Pteridines/pharmacokinetics , Retrospective Studies , Treatment Outcome , Young Adult
15.
J Hepatol ; 62(3): 533-40, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25450717

ABSTRACT

BACKGROUND & AIMS: Tenofovir alafenamide, a phosphonate prodrug of tenofovir with greater plasma stability than tenofovir disoproxil fumarate, provides efficient delivery of active drug to hepatocytes at reduced systemic tenofovir exposures. METHODS: Non-cirrhotic, treatment-naïve subjects with chronic hepatitis B were randomized (1:1:1:1:1) to receive tenofovir alafenamide 8, 25, 40, or 120 mg, or tenofovir disoproxil fumarate 300 mg for 28 days and assessed for safety, antiviral response, and pharmacokinetics, followed-up by off-treatment for 4 weeks. RESULTS: 51 subjects were randomized and all completed study treatment. Groups were generally well matched (67% male, 57% Asian, 53% HBeAg-negative, mean HBV DNA approximately 6.0 log10 IU/ml) with HBV genotypes reflective of the population. No subject experienced an adverse event that was serious or severe (grade 3/4). Across the tenofovir alafenamide groups, similar mean changes in serum HBV DNA were found at Week 4 (-2.81, -2.55, -2.19, and -2.76 log10 IU/ml for the 8, 25, 40, and 120 mg groups, respectively) which were also comparable to the control (-2.68 log10 IU/ml for tenofovir disoproxil fumarate 300 mg). Kinetics of viral decline were also similar among groups. Tenofovir alafenamide pharmacokinetics were linear and proportional to the dose; doses⩽25 mg were associated with ⩾92% reductions in mean tenofovir area under the curve relative to tenofovir disoproxil fumarate 300 mg. CONCLUSIONS: Tenofovir alafenamide was safe and well tolerated; declines in HBV DNA were similar to tenofovir disoproxil fumarate at all doses evaluated. Tenofovir alafenamide 25 mg has been selected for further hepatitis B clinical development.


Subject(s)
Adenine/analogs & derivatives , Antiviral Agents/therapeutic use , Hepatitis B, Chronic/drug therapy , Adenine/adverse effects , Adenine/pharmacokinetics , Adenine/therapeutic use , Adult , Alanine , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , DNA, Viral/blood , Female , Hepatitis B, Chronic/metabolism , Hepatitis B, Chronic/virology , Humans , Male , Middle Aged , Tenofovir/adverse effects , Tenofovir/pharmacokinetics , Tenofovir/therapeutic use , Time Factors , Treatment Outcome , Young Adult
16.
Drugs R D ; 13(2): 129-35, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23633146

ABSTRACT

BACKGROUND: Doxylamine succinate, an ethanolamine-based antihistamine, is used in the short-term management of insomnia because of its sedative effects. No data on the dose proportionality of the pharmacokinetics of doxylamine are available, although this drug has been marketed in European countries for more than 50 years. OBJECTIVE: The objective of this study was to evaluate and compare the dose proportionality between two marketed strengths (12.5 mg and 25 mg) of doxylamine hydrogen succinate after a single oral dose administration under fasting conditions in healthy human subjects. STUDY DESIGN: This was a single-center, randomized, single dose, laboratory-blinded, two-period, two-sequence, crossover study. SETTING: The study was conducted in a phase I clinical unit. SUBJECTS AND METHODS: A single oral dose of doxylamine hydrogen succinate of 12.5 mg (equivalent to 8.7 mg of doxylamine base) or 25 mg (equivalent to 17.4 mg of doxylamine base) was administered to healthy volunteers under fasting conditions in each study period. The drug administrations were separated by a wash-out period of 7 calendar days. Blood samples were collected for up to 60 h post-dose, and plasma doxylamine levels were determined by an ultra high-performance liquid chromatography method with tandem mass spectrometry detection. Pharmacokinetic parameters were calculated using non-compartmental analysis. Dose proportionality was assessed based on the parameter area under the concentration-time curve (AUCt normalized). Safety was evaluated through assessment of adverse events, standard laboratory evaluations, vital signs and 12-lead electrocardiogram (ECG). RESULTS: In total, 12 healthy volunteers (3 male; 9 female) were included in the study. Mean maximum observed plasma concentration (Cmax) and area under the concentration-time curve from time zero to time t (AUCt ) of doxylamine hydrogen succinate 12.5 mg and 25 mg tablets increased linearly and dose-dependently [12.5 mg: mean Cmax 61.94 ng/mL, coefficient of variation (CV) 23.2%; mean AUCt 817.33 ng·h/mL, CV 27.4%; and 25 mg: mean Cmax 124.91 ng/mL, CV 18.7%; mean AUCt 1630.85 ng·h/mL, CV 22.8%]. Mean AUCt normalized was 815.43 ng·h/mL, CV 22.8% for 25 mg. The dose-normalized geometric mean ratio (%, 12.5 mg/25 mg) of AUCt was 98.92 (90% CI: 92.46, 105.83). The most common adverse event was somnolence. CONCLUSIONS: Exposure to doxylamine was proportional over the therapeutic dose range of 12.5-25 mg in healthy volunteers. Based on the results, a predictable and linear increase in systemic exposure can be expected. Doxylamine hydrogen succinate was safe and well tolerated.


Subject(s)
Doxylamine/analogs & derivatives , Histamine H1 Antagonists/pharmacokinetics , Adult , Biological Availability , Cross-Over Studies , Dose-Response Relationship, Drug , Doxylamine/administration & dosage , Doxylamine/adverse effects , Doxylamine/blood , Doxylamine/pharmacokinetics , Fasting/blood , Female , Healthy Volunteers , Histamine H1 Antagonists/adverse effects , Histamine H1 Antagonists/blood , Humans , Male , Medication Adherence , Middle Aged , Tablets
17.
Drugs R D ; 12(4): 217-25, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23230999

ABSTRACT

BACKGROUND: Doxylamine succinate, an ethanolamine-based antihistamine, is used in the short-term management of insomnia because of its sedative effects. The data available on the pharmacokinetic profile of doxylamine in humans are limited, notwithstanding that this drug has been marketed in European countries for more than 50 years. In fact, no data on the effect of food on the pharmacokinetic parameters of doxylamine are available. OBJECTIVE: The objective of this study was to evaluate the pharmacokinetic parameters of doxylamine following a single oral dose of doxylamine hydrogen succinate 25 mg in healthy human subjects under fed and fasting conditions. STUDY DESIGN: This was a single-center, randomized, single-dose, laboratory-blinded, two-period, two-sequence, crossover study. SETTING: The study was conducted in a phase I clinical unit. SUBJECTS AND METHODS: A single oral dose of doxylamine hydrogen succinate 25 mg (equivalent to 17.4 mg of doxylamine base) was administered to healthy volunteers under either fed conditions (high-fat, high-calorie food intake) or fasting conditions in each study period. The drug administrations were separated by a wash-out period of seven calendar days. Plasma samples were collected for up to 60 hours postdose, and plasma doxylamine concentrations were determined by a high-performance liquid chromatography method with tandem mass spectrometry detection. Pharmacokinetic parameters were calculated using noncompartmental analysis. Safety was evaluated through assessment of adverse events, standard laboratory evaluations, vital signs, and 12-lead electrocardiography. RESULTS: In total, 24 healthy subjects (12 male and 12 female) were included in the study. Doxylamine succinate 25 mg tablets exhibited similar oral bioavailability of doxylamine in the fasting state (mean maximum plasma drug concentration [C(max)] 118.21 ng/mL, coefficient of variation [CV] 19.2%; mean area under the plasma concentration time curve from time zero to time t [AUC(t)] 1746.97 ng · h/mL, CV 31.6%) and in the fed state (mean C(max) 120.99 ng/mL, CV 15.0%; mean AUC(t) 1712.20 ng · h/mL, CV 26.7%). No statistically significant between-treatment differences were observed for any of the pharmacokinetic parameters under study. The fed : fasting ratios of the geometric least squares means with corresponding 90% confidence intervals for C(max) and AUC(t) were within the range of 80-125%. CONCLUSION: High-fat, high-calorie food intake does not affect the kinetics of doxylamine in healthy subjects. The drug was safe and well tolerated by the subjects in this study.


Subject(s)
Doxylamine/analogs & derivatives , Food-Drug Interactions , Tablets/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Biological Availability , Cross-Over Studies , Doxylamine/administration & dosage , Doxylamine/adverse effects , Doxylamine/pharmacokinetics , Fasting/metabolism , Female , Half-Life , Humans , Male , Middle Aged , Tablets/administration & dosage , Tablets/adverse effects , Tablets, Enteric-Coated , Young Adult
18.
Arzneimittelforschung ; 61(3): 153-9, 2011.
Article in English | MEDLINE | ID: mdl-21528639

ABSTRACT

BACKGROUND: Eplerenone (CAS 107724-20-9) prevents the binding of aldosterone, a key hormone in the renin-angiotensin-aldosterone-system (RAAS), which is involved in the regulation of blood pressure and the pathophysiology of cardiovascular disease and is indicated, in addition to standard therapy including beta-blockers, to reduce the risk of cardiovascular mortality and morbidity in stable patients with left ventricular dysfunction (LVEF < or = 40%) and clinical evidence of heart failure after recent myocardial infarction. OBJECTIVE: The aim of this study was to assess the bioequivalence of a new eplerenone 50 mg formulation (test formulation) vs. the reference product, as required by European regulatory authorities for the marketing of a generic product. METHODS: This was a single-center, randomized, single-dose, open-label, 2-way crossover study in healthy volunteers under fasting conditions. Plasma samples were collected up to 24 h post-dosing and plasma eplerenone levels were determined by reversed phase high performance liquid chromatography and by tandem mass spectrometry detection (ie, the LC-MS/MS method). Pharmacokinetic parameters were calculated using non-compartmental analysis. Area under the concentration-time curve from time zero to time of last non-zero concentration (AUClast) and maximum observed concentration (Cmax) were the main evaluation criteria. All of the above-mentioned pharmacokinetic parameters were analyzed using 90% geometric confidence interval of the ratio (T/R) of least-squares means from the ANOVA of the 1n-transformed parameter. Tolerability was monitored using physical examination, including vital sign measurements and laboratory analysis. RESULTS: According to the classical approach, the 90% geometric confidence intervals obtained by analysis of variance for AUClast and Cmax were within the predefined ranges (80.00-125.00%). CONCLUSION: Bioequivalence between test and reference formulations, both in terms of rate and extension of absorption, under fasting conditions was concluded according to European guidelines. Both formulations were well tolerated.


Subject(s)
Mineralocorticoid Receptor Antagonists/pharmacokinetics , Spironolactone/analogs & derivatives , Adolescent , Adult , Area Under Curve , Biological Availability , Chemistry, Pharmaceutical , Chromatography, High Pressure Liquid , Cross-Over Studies , Eplerenone , Fasting , Female , Half-Life , Humans , Male , Middle Aged , Mineralocorticoid Receptor Antagonists/adverse effects , Spironolactone/adverse effects , Spironolactone/pharmacokinetics , Tandem Mass Spectrometry , Therapeutic Equivalency , Young Adult
19.
Curr Med Res Opin ; 26(6): 1355-62, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20377319

ABSTRACT

OBJECTIVE: Combination therapy is frequently required in the management of epilepsy. The primary objective of this study was to investigate the pharmacokinetic interaction between eslicarbazepine acetate (ESL) 1200 mg once daily and topiramate (TPM) 200 mg once daily in healthy subjects. METHODS: Multiple-dose, open-label, one-sequence study in two parallel groups of 16 healthy male volunteers. After an 8-day treatment with ESL (Group A) or TPM (Group B), ESL and TPM were co-administered for 19 days. A bioequivalence approach based on a within-subject comparison was used to investigate a potential drug-drug interaction. End/start of treatment geometric mean ratios (GMR, %) and 90% confidence intervals (90% CI) were calculated for maximum plasma concentration (C(max)) and area under the plasma concentration-time curve over the dosing interval at steady-state (AUC(ss)) of eslicarbazepine (ESL major active metabolite), R-licarbazepine (ESL minor active metabolite) and TPM at Day 8 and Day 27. RESULTS: In Group A, eslicarbazepine GMR (90% CI) was 86.79% (81.06%; 92.94%) for C(max) and 92.70% (89.21%; 96.32%) for AUC(ss). In Group B, TPM GMR (90% CI) was 81.50% (77.48%; 85.89%) for C(max) and 81.81% (79.69%; 84.00%) for AUC(ss). The 90% CI of eslicarbazepine C(max) and AUC(ss) fell within the pre-specified bioequivalence range (80.00%; 125.00%), allowing it to be concluded that the extent of systemic exposure to eslicarbazepine was unaffected by the concomitant administration of TPM. The 90% CI for topiramate AUC(ss) was borderline in relation to the pre-specified bioequivalence range and topiramate C(max) fell outside the pre-specified bioequivalence range. Therefore, the extent of systemic exposure to TPM following co-administration with ESL was not formally bioequivalent to the extent of systemic exposure to TPM when TPM was administered alone. However, there was no difference between TPM elimination half-life following TPM co-administered with ESL and TPM administered alone (24.0 and 24.3 h, respectively). The bioavailability of R-licarbazepine was essentially bioequivalent. Two subjects discontinued due to adverse events. No clinical interaction appeared to be present in terms of adverse events when both drugs were given concomitantly. CONCLUSION: Concomitant administration of eslicarbazepine acetate 1200 mg once daily and topiramate 200 mg once daily showed no significant change in exposure to eslicarbazepine but an 18% decrease in exposure to topiramate, most likely caused by a reduced bioavailability of topiramate. No dose adjustment is required.


Subject(s)
Anticonvulsants/pharmacokinetics , Dibenzazepines/pharmacokinetics , Fructose/analogs & derivatives , Adult , Anticonvulsants/administration & dosage , Dibenzazepines/administration & dosage , Drug Interactions , Fructose/administration & dosage , Fructose/pharmacokinetics , Humans , Male , Middle Aged , Quebec , Topiramate , Young Adult
20.
Antimicrob Agents Chemother ; 53(7): 3081-7, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19414580

ABSTRACT

Shiga toxin (Stx)-producing Escherichia coli (STEC) causes hemorrhagic colitis and hemolytic-uremic syndrome (HUS). The rates of STEC infection and complications, including death, are highest among young children and elderly individuals. There are no causal therapies. Because Stx is the primary pathological agent leading to organ injury in patients with STEC disease, therapeutic antibodies are being developed to neutralize systemically absorbed toxin during the early phase of the infection. Two phase I, single-dose, open-label, nonrandomized studies were conducted to evaluate the safety and pharmacokinetics of the chimeric monoclonal antibodies (antitoxins) against Stx 1 and 2 (calphaStx1 and calphaStx2, respectively). In the first study, 16 volunteers received 1 or 3 mg/kg of body weight of calphaStx1 or calphaStx2 as a single, short (1-h) intravenous infusion (n = 4 per group). In a second study, 10 volunteers received a 1-h infusion of calphaStx1 and calphaStx2 combined at 1 or 3 mg/kg (n = 5 per group). Treatment-emergent adverse events were mild, resolved spontaneously, and were generally unrelated to the antibody infusion. No serious adverse events were observed. Human antichimeric antibodies were detected in a single blood sample collected on day 57. Antibody clearance was slightly greater for calphaStx1 (0.38 +/- 0.16 ml/h/kg [mean +/- standard deviation]) than for calphaStx2 (0.20 +/- 0.07 ml/h/kg) (P = 0.0013, t test). The low clearance is consistent with the long elimination half-lives of calphaStx1 (190.4 +/- 140.2 h) and calphaStx2 (260.6 +/- 112.4 h; P = 0.151). The small volume of distribution (0.08 +/- 0.05 liter/kg, combined data) indicates that the antibodies are retained within the circulation. The conclusion is that calphaStx1 and calphaStx2, given as individual or combined short intravenous infusions, are well tolerated. These results form the basis for future safety and efficacy trials with patients with STEC infections to ameliorate or prevent HUS and other complications.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Shiga Toxin 1/metabolism , Shiga Toxin 2/metabolism , Adult , Female , Humans , Male , Middle Aged , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...