Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Clin Transl Sci ; 14(1): 62-74, 2021 01.
Article in English | MEDLINE | ID: mdl-32770726

ABSTRACT

Biometric monitoring technologies (BioMeTs) are becoming increasingly common to aid data collection in clinical trials and practice. The state of BioMeTs, and associated digitally measured biomarkers, is highly reminiscent of the field of laboratory biomarkers 2 decades ago. In this review, we have summarized and leveraged historical perspectives, and lessons learned from laboratory biomarkers as they apply to BioMeTs. Both categories share common features, including goals and roles in biomedical research, definitions, and many elements of the biomarker qualification framework. They can also be classified based on the underlying technology, each with distinct features and performance characteristics, which require bench and human experimentation testing phases. In contrast to laboratory biomarkers, digitally measured biomarkers require prospective data collection for purposes of analytical validation in human subjects, lack well-established and widely accepted performance characteristics, require human factor testing, and, for many applications, access to raw (sample-level) data. Novel methods to handle large volumes of data, as well as security and data rights requirements add to the complexity of this emerging field. Our review highlights the need for a common framework with appropriate vocabulary and standardized approaches to evaluate digitally measured biomarkers, including defining performance characteristics and acceptance criteria. Additionally, the need for human factor testing drives early patient engagement during technology development. Finally, use of BioMeTs requires a relatively high degree of technology literacy among both study participants and healthcare professionals. Transparency of data generation and the need for novel analytical and statistical tools creates opportunities for precompetitive collaborations.


Subject(s)
Biomedical Technology/methods , Biometry/methods , Data Collection/methods , Monitoring, Physiologic/methods , Remote Sensing Technology/methods , Big Data , Biomedical Technology/trends , Data Collection/instrumentation , Humans , Monitoring, Physiologic/instrumentation , Remote Sensing Technology/trends , Research Design
2.
Clin Podiatr Med Surg ; 36(3): 457-468, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31079610

ABSTRACT

Surgical bone reconstructive procedures of the foot and ankle in diabetic patients must be considered when performing evaluation of a diabetic foot for patients with preulcerative lesions and preexisting wounds. Preventive deformity correction can reduce the potential risk of ulceration, infection, and possible amputation in a patient with an at-risk foot type. It can also expedite wound healing and prevent further breakdown in a patient with lower extremity ulcerations. This article discusses different types of surgical bone reconstructive procedures as preventive and prophylactic deformity corrections to reduce osseous deformity, minimize preulcerative lesions, and increase limb-salvage rates in the compromised patient.


Subject(s)
Diabetic Foot/surgery , Foot Bones/surgery , Achilles Tendon/surgery , Bunion, Tailor's/surgery , Elective Surgical Procedures , Humans , Limb Salvage , Osteotomy
3.
Am J Obstet Gynecol ; 213(5): 735.e1-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26366665

ABSTRACT

BACKGROUND: Vaginal delivery is a risk factor in pelvic floor disorders. We previously described changes in the pelvic floor associated with pregnancy and parturition in the squirrel monkey, a species with a humanlike pattern of spontaneous age- and parity-associated pelvic organ prolapse. OBJECTIVE: The potential to prevent or diminish these changes with scheduled cesarean delivery (CD) has not been evaluated. In a randomized, controlled trial, we compared female squirrel monkeys undergoing spontaneous vaginal delivery with those undergoing scheduled primary CD for pelvic floor muscle volumes, muscle contrast changes, and dynamic effects on bladder neck position. STUDY DESIGN: Levator ani, obturator internus, and coccygeus (COC) muscle volumes and contrast uptake were assessed by magnetic resonance imaging in 20 nulliparous females examined prior to pregnancy, a few days after delivery, and 3 months postpartum. The position of bladder neck relative to bony reference line also was assessed with abdominal pressure using dynamic magnetic resonance imaging. RESULTS: Baseline measurements of 10 females randomly assigned to scheduled primary CD were not different from those of 10 females assigned to spontaneous vaginal delivery. Levator ani and obturator internus muscle volumes did not differ between groups, while volumes were reduced (P < .05) in the observation immediately after pregnancy. The COC muscles increased (P < .05) immediately after delivery for females in the spontaneous vaginal delivery group, but not for females in the scheduled CD group. Position of the bladder neck descended (P < .05) by 3 months postpartum in both groups. CONCLUSION: Scheduled CD diminishes changes in COC muscle volume and contrast reported to be associated with spontaneous vaginal delivery in squirrel monkeys. However, pelvic support of the bladder was not protected by this intervention suggesting that effects of pregnancy and delivery are not uniformly prevented by this procedure.


Subject(s)
Delivery, Obstetric , Muscle Strength , Muscle, Smooth/anatomy & histology , Pelvic Floor/anatomy & histology , Animals , Cesarean Section , Female , Magnetic Resonance Imaging , Random Allocation , Saimiri
4.
Clin Pharmacol Drug Dev ; 3(2): 151-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-27128460

ABSTRACT

In order to evaluate the potential for CYP3A4 induction by moxidectin, midazolam pharmacokinetic (PK) parameters were compared before and after moxidectin administration. Healthy subjects received a single 8 mg dose of moxidectin and 3 single 7.5 mg doses of midazolam 3 days before, and 7 and 89 days after the moxidectin. Blood samples were taken for 24 hours to measure midazolam and metabolites in plasma, and for 89 days to measure moxidectin in plasma after dose administration. Noncompartmental PK analyses were performed for each analyte. Analysis of variance was performed on log-transformed midazolam parameters with treatment day as a fixed effect. Adverse events were recorded and laboratory tests, physical examinations, pulse oximetry monitoring, vital sign measurement, and electrocardiograms performed. Thirty-nine subjects were enrolled in the study; PK data were available for 37 subjects. Moxidectin PK parameters were similar to previous studies. There were no significant changes in PK for midazolam or its metabolites 7 or 89 days after moxidectin administration. Adverse events were generally mild and there were no relevant changes in safety assessments. Thus, 8 mg moxidectin does not induce CYP3A4 activity and other CYP3A4 substrates are unlikely to be affected by moxidectin co-administration.

5.
Eur J Clin Pharmacol ; 69(4): 835-42, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23052408

ABSTRACT

PURPOSE: Sirolimus and tacrolimus are immunosuppressive compounds that have been used concomitantly in renal transplant patients. Both drugs are dosed orally and have common intestinal and hepatic metabolism and intestinal transport mechanisms. As such, there is a potential for pharmacokinetic drug interaction. METHODS: A single-dose, open-label, four-period, four-treatment, randomized crossover study was conducted in 27 healthy fasting volunteers. Each subject received a 15-mg oral dose of sirolimus alone, a 10-mg oral dose of tacrolimus alone, sirolimus and tacrolimus administered simultaneously, and tacrolimus administered 4 h before sirolimus. Whole blood and plasma samples for sirolimus and tacrolimus testing were analyzed by liquid chromatography/tandem mass spectrometry. Pharmacokinetic parameters were assessed using noncompartmental methods and were compared using analysis of variance (ANOVA). RESULTS: The geometric mean ratio and 90 % confidence interval (CI) area under the concentration-time curve from time 0 to infinity (AUCinf) for sirolimus administered simultaneously with tacrolimus versus sirolimus alone were 97 and 89-106, respectively, and, when administered in a staggered approach versus sirolimus alone, 107 and 98-117, respectively. The geometric mean ratio (%) and 90 % CI AUCinf for tacrolimus administered simultaneously with sirolimus versus tacrolimus alone were 92 and 82-102, respectively, and, when administered in a staggered approach versus tacrolimus alone, 94 and 84-105, respectively. CONCLUSIONS: The results of this study demonstrate a lack of any clinically important drug interaction between sirolimus and tacrolimus in healthy subjects after single-dose administration. However, due to the complexity of anti-rejection immunosuppressive therapy dosing, we suggest that sirolimus and tacrolimus concentration monitoring be performed when changes in dosing are made for either drug regimen.


Subject(s)
Immunosuppressive Agents/pharmacokinetics , Sirolimus/pharmacokinetics , Tacrolimus/pharmacokinetics , Administration, Oral , Adolescent , Adult , Analysis of Variance , Brazil , Cross-Over Studies , Drug Administration Schedule , Drug Interactions , Fasting , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/blood , Kidney Transplantation , Male , Middle Aged , Sirolimus/administration & dosage , Sirolimus/adverse effects , Sirolimus/blood , Tacrolimus/administration & dosage , Tacrolimus/adverse effects , Tacrolimus/blood , Young Adult
6.
Am J Trop Med Hyg ; 86(1): 122-125, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22232462

ABSTRACT

The objective of this study was to assess the effect of a high-fat meal on the pharmacokinetics of moxidectin. Healthy male subjects were randomized to receive single oral 8 mg doses of moxidectin after an overnight fast or high-fat breakfast. In fasted subjects (N = 27), mean [SD] parameters were C(max): 58.9 [12.5] ng/mL; t(max): 3.7 [1.5] h; area under concentration-time curve (AUC): 3,387 [1,328] ng/h/mL; Vλ(z)/F: 2,829 [1,267] L; CL/F: 2.76 [1.28] L/h; and t(1/2): 784 [347] h. Compared with fasted subjects, fed subjects (N = 27) exhibited a 34% increase in C(max), delay in t(max) to 5.3 [2.1] h, 44% increase in AUC, 40% decrease in Vλ(z)/F, and a 35% decrease in CL/F. There was no significant change in t(1/2). The changes are consistent with an increase in moxidectin bioavailability following administration with food. There were no clinically relevant changes in vital signs, laboratory tests, or electrocardiograms.


Subject(s)
Antinematodal Agents/pharmacokinetics , Dietary Fats/pharmacology , Food-Drug Interactions , Administration, Oral , Adult , Antinematodal Agents/administration & dosage , Area Under Curve , Biological Availability , Dietary Fats/administration & dosage , Fasting , Humans , Macrolides/administration & dosage , Macrolides/pharmacokinetics , Male , Middle Aged , Young Adult
7.
Clin Pharmacol Drug Dev ; 1(1): 32-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-27206144

ABSTRACT

The antiparasitic agent moxidectin is under development for the treatment of onchocerciasis. As the first-in-human study of moxidectin used a liquid formulation but other trials used tablets, a study was performed to determine the relative bioavailability of the 2 formulations and to gain more information about the pharmacokinetics of moxidectin. Fifty-eight healthy male participants were randomized to receive open-label moxidectin (10 mg) as a tablet (n = 29) or liquid (n = 29) formulation. The mean ± SD pharmacokinetic parameters observed following administration of the tablet were peak concentration (Cmax) 67.1 ± 27.4 ng/mL, time to peak concentration (tmax) 3.2 ± 1.4 hours, area under the concentration time curve (AUC) 4403 ± 2360 ng·h/mL, apparent volume of distribution 3635 ± 1720 L, oral clearance 2.83 ± 1.25 L/h, and elimination half-life 1032 ± 502 hours. The Cmax and AUC observed following administration of the liquid formulation were 28.6% and 28.8% higher, respectively, and tmax 0.9 hours shorter compared with tablets. No serious adverse events (AEs) were observed. The most commonly reported AEs were headache, infection, diarrhea, asthenia, myalgia, and dizziness during the inpatient phase and flu syndrome, headache, and infection during the 6-month outpatient phase. There was no difference in reporting of these AEs between formulations.

8.
Antimicrob Agents Chemother ; 55(11): 5200-4, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21896908

ABSTRACT

Moxidectin, registered worldwide as a veterinary antiparasitic agent, is currently under development for humans for the treatment of onchocerciasis in collaboration with the World Health Organization. The objective of this study was to assess the pharmacokinetics of moxidectin in healthy lactating women, including the excretion into breast milk. Twelve women, ages 23 to 38 years, weighing 54 to 79 kg, all more than 5 months postpartum, were enrolled, following their plan to wean their infants and provision of informed consent. A single 8-mg, open-label dose was administered orally after consumption of a standard breakfast. Complete milk collection was done for approximately 28 days, and plasma samples were collected for 90 days. Moxidectin concentrations were measured by high-performance liquid chromatography (HPLC) with fluorescence detection, with a validated range of 0.08 to 120 ng/ml. Noncompartmental pharmacokinetic methods were used to find the following results: peak concentration in plasma (C(max)), 87 ± 25 ng/ml; time to C(max) (t(max)), 4.18 ± 1.59 h; terminal-phase elimination half-life (t(1/2)), 832 ± 321 h; total area under the concentration-time curve (AUC), 4,046 ± 1,796 ng · h/ml; apparent oral dose clearance (CL/F), 2.35 ± 1.07 l/h; ratio of CL/F to the terminal-phase disposition rate constant, λ(z) (Vλ(z)/F), 2,526 ± 772 liters; percentage of maternal dose excreted in milk, 0.701 ± 0.299%; absolute amount excreted in milk, 0.056 ± 0.024 mg; relative infant dose, 8.73 ± 3.17% of maternal dose assuming complete absorption; clearance in milk (CL(milk)), 0.016 ± 0.009 liter/h. Nine of 12 subjects reported adverse events, all of which were considered treatment emergent but not drug related and were mostly reported during the long outpatient period 8 to 90 days after dose administration. The most frequently reported adverse events were headache and nausea (n = 4), oropharyngeal pain (n = 2), rhinitis, viral pharyngitis, and viral upper respiratory tract infection (n = 2).


Subject(s)
Lactation/metabolism , Milk, Human/metabolism , Adult , Chromatography, High Pressure Liquid , Female , Humans , Macrolides/metabolism , Macrolides/pharmacokinetics , Young Adult
9.
J Am Assoc Lab Anim Sci ; 49(5): 560-3, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20858355

ABSTRACT

Owl monkeys are New World primates frequently used in biomedical research. Despite the historical difficulty of breeding owl monkeys in captivity, several productive owl monkey breeding colonies exist currently. The animals in the colony we describe here are not timed-pregnant, and determination of gestational age is an important factor in prenatal care. Gestational age of human fetuses is often determined by using transabdominal measurements of fetal biparietal diameter. The purpose of this study was to correlate biparietal diameter measurements with gestational age in owl monkeys. We found that biparietal diameter can be used to accurately predict gestational age in owl monkeys.


Subject(s)
Acaricides/therapeutic use , Anthropometry/methods , Aotidae/growth & development , Gestational Age , Ultrasonography, Prenatal/veterinary , Animals , Aotidae/anatomy & histology , Female , Fetus/anatomy & histology , Head/diagnostic imaging , Head/embryology , Pregnancy , Ultrasonography, Prenatal/standards
10.
J Med Primatol ; 39(3): 143-50, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20149027

ABSTRACT

BACKGROUND: Cardiovascular disease, especially cardiomyopathy, was the major cause of death among owl monkeys (Aotus sp.) at a major colony and threatened colony sustainability. For this study, echocardiography (echo) and electrocardiography (ECG) normal values were established, and cardiomyopathy animals identified. METHODS: Forty-eight owl monkeys were studied, 30 older than 10 years of age ('aged') and 8 of age 5 years ('young'). Eight aged owl monkeys had cardiomyopathy. RESULTS AND CONCLUSIONS: Aged Aotus had increased left ventricular posterior wall thickness over young animals. Left ventricular diameter and ejection fraction appeared to be the best identifying measurements for cardiomyopathy. There were no differences in the ECG.


Subject(s)
Aotidae/anatomy & histology , Cardiomyopathies/veterinary , Echocardiography , Heart/physiology , Monkey Diseases/diagnostic imaging , Animals , Aotidae/physiology , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/physiopathology , Case-Control Studies , Electrocardiography , Female , Male , Monkey Diseases/physiopathology
11.
Br J Clin Pharmacol ; 67(3): 299-308, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19523013

ABSTRACT

AIMS: To determine the safety, pharmacokinetics (PK) and pharmacodynamics (PD) of oral immediate release (IR) lecozotan in healthy young and elderly subjects. METHODS: Three randomized, double-blind, placebo-controlled, sequential, ascending dose Phase I studies of lecozotan were conducted. In a single-dose study, ascending doses of 2, 5 and 10 mg were administered to cohorts of eight young subjects. In the two ascending 14-day multiple-dose studies, 41 young subjects received 0.1, 0.25, 0.5, 1 and 5 mg q12h of lecozotan or placebo and 24 elderly received 0.5 mg and 5 mg q12h of lecozotan or placebo. Assessments included safety evaluations, a complete PK profile and PD. RESULTS: Lecozotan was safe and well tolerated at steady state up to 5 mg q12. The maximum tolerated dose after multiple doses was >10 mg (5 mg q12). In the single-dose study, the maximum tolerated dose was 10 mg. Dose-limiting mild-to-moderate adverse events included paraesthesia, dizziness and visual disturbances peaking at t(max) and disappearing concomitantly with plasma concentrations. No clinically relevant changes in vital signs, ECG intervals or routine laboratory tests occurred. Lecozotan did not significantly change cognitive function, EEG or hormone levels. PK was characterized by rapid absorption, dose proportionality, extensive distribution and rapid elimination. The mean CL/F was approximately 35% lower in the elderly. CONCLUSIONS: Lecozotan IR was safe and well tolerated after administration of multiple oral doses up to 5 mg q12h in young and elderly subjects. These results support the development of lecozotan in patients with Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Dioxanes/adverse effects , Nootropic Agents/adverse effects , Piperazines/adverse effects , Serotonin 5-HT1 Receptor Antagonists , Serotonin Antagonists/adverse effects , Administration, Oral , Adolescent , Adult , Aged , Dioxanes/administration & dosage , Dioxanes/pharmacokinetics , Dioxanes/pharmacology , Double-Blind Method , Female , Humans , Male , Nootropic Agents/pharmacokinetics , Nootropic Agents/pharmacology , Piperazines/administration & dosage , Piperazines/pharmacokinetics , Piperazines/pharmacology , Serotonin Antagonists/pharmacokinetics , Serotonin Antagonists/pharmacology , Treatment Outcome , Young Adult
12.
J Clin Pharmacol ; 48(3): 285-92, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18218785

ABSTRACT

Nine subjects with severe hepatic impairment (Child-Pugh grade C) and 9 healthy matched control subjects were given a single 15-mg dose of sirolimus by oral solution. Increases (P < or = .002) in mean whole-blood sirolimus t(1/2) (168%), AUC(0-infinity) (210%), and MRT(oral) (261%), together with a decrease (P = .001) in CL/F (-67%), were observed in subjects with severe hepatic impairment compared with healthy matched controls. Sirolimus pharmacokinetic data in Child-Pugh grade A (n = 13, mild) and B (n = 5, moderate) subjects from a previous identically designed study were available for an inter-study comparison. Overall, mean t(1/2), weight-normalized AUC, and MRT(oral) increased steadily, whereas mean CL/F decreased steadily, with increasing degrees of hepatic impairment. CL/F showed large intersubject variabilities within subject types and extensive overlap among the subject types. The results of this study suggest that an initial sirolimus dose reduction of approximately 60% is appropriate in patients with acute severe hepatic impairment; this should be followed by further dose adjustment, based on therapeutic drug monitoring, until the trough concentrations have stabilized at sirolimus levels existing prior to the onset of acute liver failure.


Subject(s)
Liver Diseases/drug therapy , Sirolimus/pharmacokinetics , Administration, Oral , Aged , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Area Under Curve , Aspartate Aminotransferases/blood , Aspartate Aminotransferases/metabolism , Bilirubin/metabolism , Case-Control Studies , Cholecystitis/chemically induced , Creatinine/blood , Female , Half-Life , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/pharmacokinetics , Liver Diseases/metabolism , Liver Diseases/pathology , Male , Middle Aged , Severity of Illness Index , Sirolimus/adverse effects , Sirolimus/blood
13.
J Am Assoc Lab Anim Sci ; 46(4): 74-6, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17645301

ABSTRACT

This case report describes the ultrasonographic findings during an idiopathic spontaneous abortion in an owl monkey. The female owl monkey presented for a transabdominal ultrasonogram to evaluate her pregnancy. This evaluation is a routine monitoring procedure in our owl monkey breeding colony. Although the fetus and placenta appeared normal at the initial scan, no fetal heartbeat could be detected. We followed the abortion with serial ultrasonographic scans and documented complete involution of the uterus post-abortion.


Subject(s)
Abortion, Veterinary/diagnostic imaging , Aotidae , Monkey Diseases/diagnostic imaging , Animals , Female , Pregnancy , Ultrasonography, Prenatal/veterinary , Uterus/diagnostic imaging
14.
J Clin Pharmacol ; 44(11): 1244-51, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15496642

ABSTRACT

Etanercept, a soluble recombinant human tumor necrosis factor receptor (TNFr), is effective and well tolerated in the treatment of rheumatoid arthritis, juvenile rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, and psoriasis. The primary objective of this study was to investigate the potential pharmacokinetic and pharmacodynamic interaction between digoxin and etanercept at steady state. In a crossover, open-label, nonrandomized, 3-period study, 12 healthy male subjects received loading oral doses of digoxin 0.5 mg every 12 hours on day 1 and 0.25 mg every 12 hours on day 2, followed by a daily maintenance dose of 0.25 mg for a total of 27 days. Etanercept was administered as a twice-weekly 25-mg subcutaneous dose beginning on day 9 and continuing up to day 37 for a total of 9 doses. All ratios of maximum plasma concentration (C(max)) and area under the plasma concentration versus time curve (AUC) for pharmacokinetics of digoxin fell within the confidence interval of 0.8 to 1.25. Although not considered clinically relevant, the mean C(max) and AUC of etanercept were 4.2% and 12.5% lower, respectively, when etanercept was given with digoxin than when administered alone. There were no clinically relevant changes in the electrocardiogram (ECG) parameters, and adverse events did not increase when both drugs were combined. In conclusion, there is no clinically relevant interaction between etanercept and digoxin, and both drugs can be safely coadministered without the need for a dosage adjustment.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antirheumatic Agents/pharmacokinetics , Cardiotonic Agents/pharmacokinetics , Digoxin/pharmacokinetics , Immunoglobulin G/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Adult , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Antirheumatic Agents/adverse effects , Antirheumatic Agents/metabolism , Area Under Curve , Cardiotonic Agents/adverse effects , Cross-Over Studies , Digoxin/adverse effects , Drug Interactions , Electrocardiography , Etanercept , Humans , Immunoglobulin G/adverse effects , Male
15.
J Clin Pharmacol ; 44(5): 543-50, 2004 May.
Article in English | MEDLINE | ID: mdl-15102876

ABSTRACT

Etanercept, a soluble recombinant human tumor necrosis factor receptor (TNFr) fusion protein, is effective and well tolerated in the treatment of rheumatoid arthritis (RA), juvenile rheumatoid arthritis (JRA), psoriatic arthritis (PsA), and ankylosing spondylitis (AS). The primary objective of this study was to investigate the potential pharmacokinetic and pharmacodynamic interaction between a single dose of R- and S-enantiomers of warfarin and multiple doses of etanercept after administration of warfarin and etanercept alone and together. In a nonrandomized, three-period study, 12 healthy male subjects received a single oral 25-mg dose of warfarin after an overnight fast, followed by twice-weekly 25-mg subcutaneous doses of etanercept for seven doses. The last dose of etanercept was administered concurrently with a second dose of warfarin. Serial blood samples for plasma warfarin concentration measurement and international normalized ratio (INR) assessment were collected before and up to 144 hours after dose administration. Serial blood samples for serum etanercept concentration measurement were collected before and up to 60 hours after the sixth dose and 264 hours after the seventh dose. Etanercept did not affect the pharmacokinetics and pharmacodynamics of warfarin. All ratios of maximum serum concentration (C(max)) and area under the serum concentration versus time curve (AUC) for pharmacokinetics (R- and S-enantiomers of warfarin) and INR fell within the confidence interval of 0.8 to 1.25. Warfarin also did not cause a clinically significant alteration in the pharmacokinetics of etanercept. In conclusion, coadministration of etanercept and warfarin would not be expected to change the pharmacokinetics of either medication; therefore, no dosage adjustment is needed in cases in which warfarin and etanercept are coadministered.


Subject(s)
Drug Interactions , Immunoglobulin G/blood , Receptors, Tumor Necrosis Factor/blood , Recombinant Fusion Proteins/pharmacokinetics , Warfarin/pharmacokinetics , Administration, Oral , Adult , Anticoagulants/administration & dosage , Anticoagulants/blood , Anticoagulants/pharmacokinetics , Area Under Curve , Chromatography, High Pressure Liquid , Clinical Trials as Topic , Drug Administration Schedule , Drug Interactions/physiology , Drug Therapy, Combination , Enzyme-Linked Immunosorbent Assay , Etanercept , Half-Life , Humans , Immunoglobulin G/administration & dosage , Injections, Subcutaneous , International Normalized Ratio , Male , Receptors, Tumor Necrosis Factor/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/blood , Stereoisomerism , Time Factors , Warfarin/blood , Warfarin/chemistry
16.
Br J Clin Pharmacol ; 56(1): 39-45, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12848774

ABSTRACT

AIMS: To evaluate potential pharmacokinetic interactions between phenobarbitone and retigabine, a new antiepileptic drug. METHODS: Fifteen healthy men received 200 mg of retigabine on day 1. On days 4-32, phenobarbitone 90 mg was administered at 22.00 h. On days 26-32, increasing doses of retigabine were given to achieve a final dose of 200 mg every 8 h on day 32. The pharmacokinetics of retigabine were determined on days 1 and 32, and those for phenobarbitone on days 25 and 31. RESULTS: After administration of a single 200 mg dose, retigabine was rapidly absorbed and eliminated with a mean terminal half-life of 6.7 h, a mean AUC of 3936 ng x ml(-1) x h and a mean apparent clearance of 0.76 l x h(-1) x kg(-1). Similar exposure to the partially active acetylated metabolite (AWD21-360) of retigabine was observed. After administration of phenobarbitone dosed to steady-state, the pharmacokinetics of retigabine at steady-state were similar (AUC of 4433 ng x ml(-1) x h and t1/2 of 8.5 h) to those of retigabine alone. The AUC of phenobarbitone was 298 mg x l(-1) x h when administered alone and 311 mg x ml(-1) x h after retigabine administration. The geometric mean ratios and 90% confidence intervals of the AUC were 1.11 (0.97, 1.28) for retigabine, 1.01 (0.88, 1.06) for AWD21-360 and 1.04 (0.96, 1.11) for phenobarbitone. Individual and combined treatments were generally well tolerated. One subject was withdrawn from the study on day 10 due to severe abdominal pain. Headache was the most commonly reported adverse event. No clinically relevant changes were observed in the electrocardiograms, vital signs or laboratory measurements. CONCLUSIONS: There was no pharmacokinetic interaction between retigabine and phenobarbitone in healthy subjects. No dosage adjustment is likely to be necessary when retigabine and phenobarbitone are coadministered to patients.


Subject(s)
Anticonvulsants/pharmacokinetics , Carbamates/pharmacokinetics , Phenobarbital/pharmacokinetics , Phenylenediamines/pharmacokinetics , Adult , Anticonvulsants/administration & dosage , Anticonvulsants/adverse effects , Carbamates/administration & dosage , Carbamates/adverse effects , Dose-Response Relationship, Drug , Drug Interactions , Half-Life , Humans , Male , Middle Aged , Phenobarbital/administration & dosage , Phenobarbital/adverse effects , Phenylenediamines/administration & dosage , Phenylenediamines/adverse effects , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...