Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Pharmacol Res Perspect ; 9(4): e00830, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34302721

RESUMEN

α2 -Adrenoceptor agonists such as clonidine and dexmedetomidine are used as adjuvants to local anesthetics in regional anesthesia. Fadolmidine is an α2 -adrenoceptor agonist developed especially as a spinal analgesic. The current studies investigate the effects of intrathecally administered fadolmidine with a local anesthetic, bupivacaine, on antinociception and motor block in conscious rats and dogs. The antinociceptive effects of intrathecal fadolmidine and bupivacaine alone or in combination were tested in the rat tail-flick and the dog's skin twitch models. The durations of motor block in rats and in dogs were also assessed. In addition, the effects on sedation, mean arterial blood pressure, heart rate, respiratory rate and body temperature were evaluated in telemetrized dogs. Concentrations of fadolmidine in plasma and spinal cord were determined after intrathecal and intravenous administration in rats. Co-administration of intrathecal fadolmidine with bupivacaine increased the magnitude and duration of the antinociceptive effects and prolonged motor block without hypotension. The interaction of the antinociceptive effect was synergistic in its nature in rats. Concentration of fadolmidine in plasma was very low after intrathecal dosing. Taken together, these studies show that fadolmidine as an adjuvant to intrathecal bupivacaine provides enhanced sensory-motor block and enables a reduction of the doses of both drugs. The results indicate that co-administration of fadolmidine with intrathecal bupivacaine was able to achieve an enhanced antinociceptive effect without hypotension and could thus represent a suitable combination for spinal anesthesia.


Asunto(s)
Adyuvantes Anestésicos/administración & dosificación , Agonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Analgésicos/administración & dosificación , Anestesia Raquidea , Anestésicos Locales , Bupivacaína , Imidazoles/administración & dosificación , Indanos/administración & dosificación , Adyuvantes Anestésicos/sangre , Adyuvantes Anestésicos/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Analgésicos/sangre , Analgésicos/farmacocinética , Animales , Presión Arterial/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Perros , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Imidazoles/sangre , Imidazoles/farmacocinética , Indanos/sangre , Indanos/farmacocinética , Masculino , Ratas Sprague-Dawley , Frecuencia Respiratoria/efectos de los fármacos , Prueba de Desempeño de Rotación con Aceleración Constante , Médula Espinal/metabolismo
3.
PLoS One ; 14(1): e0210391, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30640937

RESUMEN

Dexmedetomidine is an imidazole derivative, with high affinity for α2 adrenergic receptors, used for sedation, analgesia and adjuvant anaesthesia. In this study, an analytical method for the quantification of dexmedetomidine in dried blood spots was developed, validated and applied. The drug was extracted from dried blood spot by liquid extraction; the separation was carried out by ultra high-resolution liquid chromatography in reverse phase coupled to tandem mass spectrometry method. An X Select cyano 5 µm HSS column (2.1 X 150 mm, Waters) and a mobile phase composed of 0.1% formic acid: acetonitrile [50:50 v/v], were used. The test was linear over the concentration range of 50 to 2000 pg/mL. The coefficients of variation for the intra and interday trials were less than 15%. The drug was stable under the conditions tested. The method was successfully applied for the quantification of 6 patients, aged 0 to 2 years, with classification ASA I, who underwent ambulatory surgeries, receiving a dose of 1 µg/Kg dexmedetomidine IV. The drug concentrations in the different sampling times were in the range of 76 to 868 pg/mL.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/sangre , Dexmedetomidina/sangre , Pruebas con Sangre Seca/métodos , Agonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Agonistas de Receptores Adrenérgicos alfa 2/normas , Analgésicos no Narcóticos/administración & dosificación , Analgésicos no Narcóticos/sangre , Analgésicos no Narcóticos/normas , Preescolar , Cromatografía Líquida de Alta Presión/métodos , Dexmedetomidina/administración & dosificación , Dexmedetomidina/normas , Pruebas con Sangre Seca/normas , Pruebas con Sangre Seca/estadística & datos numéricos , Hematócrito , Humanos , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/sangre , Hipnóticos y Sedantes/normas , Lactante , Recién Nacido , Estándares de Referencia , Espectrometría de Masas en Tándem/métodos
4.
PLoS One ; 13(11): e0207427, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30427948

RESUMEN

OBJECTIVES: Dexmedetomidine, a highly selective central α2-agonist, undergoes mainly biotransformation in the liver. The pharmacokinetics of dexmedetomidine were significantly affected by hepatic insufficiency. The clearance of dexmedetomidine in patients with severe hepatic failure decreased by 50% compared with controls. We tested the hypothesis that the pharmacokinetics of dexmedetomidine would be affected by obstructive jaundice. The prospective registration number of clinical trial is ChiCTR-IPR-15007572. METHODS: 18 patients with obstructive jaundice and 12 non-jaundiced patient controls received dexmedetomidine, 1 µg/kg, over 10 min. Arterial blood samples were drawn before, during, and up to 5 h after the infusion. Plasma dexmedetomidine concentrations were determined by 1290 infinity high performance liquid chromatography coupled with 6470 tandem mass spectrometry. The relevant pharmacokinetic parameters were calculated by non-compartmental analysis using Phoenix WinNonlin 7.0. RESULTS: Plasma clearance of dexmedetomidine was decreased by 33.3% in the obstructive jaundice group as compared with the control group (0.0068±0.0017 vs. 0.0102±0.0033 L/kg/min; P = 0.002). Volume of distribution was decreased by 29.2% in the obstructive jaundice group as compared with the control group (1.43±0.58 vs. 2.02±0.84 L/kg; P = 0.041). CONCLUSIONS: This study demonstrates that the clearance and distribution volume of dexmedetomidine were decreased in patients with obstructive jaundice. It may be advisable to adjust the dose of dexmedetomidine in those patients.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Dexmedetomidina/administración & dosificación , Hipnóticos y Sedantes/administración & dosificación , Ictericia Obstructiva/tratamiento farmacológico , Agonistas de Receptores Adrenérgicos alfa 2/efectos adversos , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Adulto , Anciano , Bilirrubina/sangre , Dexmedetomidina/efectos adversos , Dexmedetomidina/sangre , Dexmedetomidina/farmacocinética , Femenino , Humanos , Hipnóticos y Sedantes/efectos adversos , Infusiones Intravenosas , Ictericia Obstructiva/sangre , Ictericia Obstructiva/patología , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Espectrometría de Masas en Tándem
5.
BMC Pharmacol Toxicol ; 19(1): 29, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29884231

RESUMEN

BACKGROUND: The Oslo Study of Clonidine in Elderly Patients with Delirium (LUCID) is an RCT investigating the effect of clonidine in medical patients > 65 years with delirium. To assess the dosage regimen and safety measures of this study protocol, we measured the plasma concentrations and hemodynamic effects of clonidine in the first 20 patients. METHODS: Patients were randomised to clonidine (n = 10) or placebo (n = 10). The treatment group was given a loading dose (75µg every 3rd hour up to a maximum of 4 doses) to reach steady state, and further 75µg twice daily until delirium free for 2 days, discharge or a maximum of 7 days. Blood pressure (BP) and heart rate (HR) were measured just before every dose. If the systolic BP was < 100 mmHg or HR < 50 beats per minute the next dose was omitted. Plasma concentrations of clonidine were measured 3 h after each drug intake on day 1, just before intake (day 2 and at steady state day 4-6) and 3 h after intake at steady state (Cmax). Our estimated pre-specified plasma concentration target range was 0.3-0.7µg/L. RESULTS: 3 h after the first dose of 75µg clonidine, plasma concentration levels rose to median 0.35 (range 0.24-0.40)µg/L. Median trough concentration (C0) at day 2 was 0.70 (0.47-0.96)µg/L. At steady state, median C0 was 0.47 (0.36-0.76)µg/L, rising to Cmax 0.74 (0.56-0.95)µg/L 3 h post dose. A significant haemodynamic change from baseline was only found at a few time-points during the loading doses within the clonidine group. There was however extensive individual BP and HR variation in both the clonidine and placebo groups, and when comparing the change scores (delta values) between the clonidine and the placebo groups, there were no significant differences. CONCLUSIONS: The plasma concentration of clonidine was at the higher end of the estimated therapeutic range. Hemodynamic changes during clonidine treatment were as expected, with trends towards lower blood pressure and heart rate in patients treated with clonidine, but with dose adjustments based on SBP this protocol appears safe. TRIAL REGISTRATION: ClinicalTrials.gov NCT01956604 , 09.25.2013. EudraCT Number: 2013-000815-26, 03.18.2013. Enrolment of first participant: 04.24.2014.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Antihipertensivos/farmacología , Clonidina/farmacología , Delirio/metabolismo , Delirio/fisiopatología , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Anciano , Anciano de 80 o más Años , Antihipertensivos/sangre , Presión Sanguínea/efectos de los fármacos , Clonidina/sangre , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Masculino
6.
Anesthesiology ; 129(1): 22-36, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29642080

RESUMEN

BACKGROUND: Differentiating drug-related changes and state-related changes on the electroencephalogram during anesthetic-induced unconsciousness has remained a challenge. To distinguish these, we designed a rigorous experimental protocol with two drugs known to have distinct molecular mechanisms of action. We hypothesized that drug- and state-related changes can be separated. METHODS: Forty-seven healthy participants were randomized to receive dexmedetomidine (n = 23) or propofol (n = 24) as target-controlled infusions until loss of responsiveness. Then, an attempt was made to arouse the participant to regain responsiveness while keeping the drug infusion constant. Finally, the concentration was increased 1.5-fold to achieve presumable loss of consciousness. We conducted statistical comparisons between the drugs and different states of consciousness for spectral bandwidths, and observed how drug-induced electroencephalogram patterns reversed upon awakening. Cross-frequency coupling was also analyzed between slow-wave phase and alpha power. RESULTS: Eighteen (78%) and 10 (42%) subjects were arousable during the constant drug infusion in the dexmedetomidine and propofol groups, respectively (P = 0.011 between the drugs). Corresponding with deepening anesthetic level, slow-wave power increased, and a state-dependent alpha anteriorization was detected with both drugs, especially with propofol. The slow-wave and frontal alpha activities were momentarily disrupted as the subjects regained responsiveness at awakening. Negative phase-amplitude coupling before and during loss of responsiveness frontally and positive coupling during the highest drug concentration posteriorly were observed in the propofol but not in the dexmedetomidine group. CONCLUSIONS: Electroencephalogram effects of dexmedetomidine and propofol are strongly drug- and state-dependent. Changes in slow-wave and alpha activity seemed to best detect different states of consciousness.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Dexmedetomidina/administración & dosificación , Electroencefalografía/efectos de los fármacos , Hipnóticos y Sedantes/administración & dosificación , Propofol/administración & dosificación , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Adulto , Anestésicos Intravenosos , Dexmedetomidina/sangre , Electroencefalografía/métodos , Humanos , Hipnóticos y Sedantes/sangre , Infusiones Intravenosas , Masculino , Propofol/sangre , Adulto Joven
7.
Drug Test Anal ; 9(5): 713-720, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27492623

RESUMEN

There is limited data describing xylazine serum concentrations in the horse and no reports of concentrations beyond 24 hours. The primary goal of the study reported here was to update the pharmacokinetics of xylazine following intravenous (IV) administration in order to assess the applicability of current regulatory recommendations. Pharmacodynamic parameters were determined using PK-PD modeling. Sixteen exercised adult Thoroughbred horses received a single IV dose of 200 mg of xylazine. Blood and urine samples were collected at time 0 and at various times for up to 96 hours and analyzed using liquid chromatography tandem mass spectrometry. Xylazine serum concentrations were best fit by a 3-compartment model. Mean ± SEM systemic clearance, volume of distribution at steady state, beta half-life and gamma half-life were 12.7 ± 0.735 mL/min/kg, 0.660 ± 0.053 L/kg, 2.79 ± 0.105 hours and 26.0 ± 1.9, respectively. Immediately following administration, horses appeared sedate as noted by a decrease in chin-to-ground distance, decreased locomotion and decreased heart rate (HR). Sedation lasted approximately 45 minutes. Glucose concentrations were elevated for 1-hour post administration. The EC50 (IC50) was 636.1, 702.2, 314.1 and 325.7 ng/mL for HR, atrioventricular block, chin-to-ground distance and glucose concentrations, respectively. The Emax (Imax) was 27.3 beats per minute, 47.5%, 42.4 cm and 0.28 mg/dL for HR, atrioventricular block, chin-to-ground distance and glucose concentrations, respectively. Pharmacokinetic parameters differ from previous reports and a prolonged detection time suggests that an extended withdrawal time, beyond current regulatory recommendations, is warranted to avoid inadvertent positive regulatory findings in performance horses. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/sangre , Agonistas de Receptores Adrenérgicos alfa 2/orina , Caballos/sangre , Caballos/orina , Xilazina/sangre , Xilazina/orina , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Animales , Área Bajo la Curva , Glucemia/metabolismo , Monitoreo de Drogas , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Caballos/fisiología , Locomoción/efectos de los fármacos , Masculino , Modelos Biológicos , Condicionamiento Físico Animal , Drogas Veterinarias/sangre , Drogas Veterinarias/farmacología , Drogas Veterinarias/orina , Xilazina/farmacología
8.
J Vet Pharmacol Ther ; 39(5): 460-8, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26896345

RESUMEN

This study characterized the pharmacokinetics of dexmedetomidine, MK-467, and their combination following intravenous bolus administration to cats. Seven 6- to-year-old male neutered cats, weighting 5.1 ± 0.7 kg, were used in a randomized, crossover design. Dexmedetomidine [12.5 (D12.5) and 25 (D25) µg/kg], MK-467 [300 µg/kg (M300)] or dexmedetomidine (25 µg/kg) and MK-467 [75, 150, 300 or 600 µg/kg-only the plasma concentrations in the 600 µg/kg group (D25M600) were analyzed] were administered intravenously, and blood was collected until 8 hours thereafter. Plasma drug concentrations were analyzed using liquid chromatography/mass spectrometry. A two-compartment model best fitted the data. Median (range) volume of the central compartment (mL/kg), volume of distribution at steady state (mL/kg), clearance (mL min/kg) and terminal half-life (min) were 342 (131-660), 829 (496-1243), 14.6 (9.6-22.7) and 48 (40-69) for D12.5; 296 (179-982), 1111 (908-2175), 18.2 (12.4-22.9) and 52 (40-76) for D25; 653 (392-927), 1595 (1094-1887), 22.7 (18.5-36.4) and 48 (35-60) for dexmedetomidine in D25M600; 117 (112-163), 491 (379-604), 3.0 (2.0-4.5) and 122 (99-139) for M300; and 147 (112-173), 462 (403-714), 2.8 (2.1-4.8) and 118 (97-172) for MK-467 in D25M600. MK-467 moderately but statistically significantly affected the disposition of dexmedetomidine, whereas dexmedetomidine minimally affected the disposition of MK-467.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Antagonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Dexmedetomidina/farmacocinética , Quinolizinas/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Antagonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Antagonistas de Receptores Adrenérgicos alfa 2/sangre , Animales , Gatos , Dexmedetomidina/administración & dosificación , Dexmedetomidina/sangre , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Inyecciones Intravenosas/veterinaria , Masculino , Quinolizinas/administración & dosificación , Quinolizinas/sangre
9.
J Vet Pharmacol Ther ; 39(4): 405-7, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26763140

RESUMEN

This study determined the unbound fraction of the peripheral α2 -adrenoceptor antagonist MK-467 alone and combined with medetomidine. MK-467 (0.1, 1 and 10 µm) was incubated in canine plasma with and without medetomidine (molar ratio 20:1), with human serum albumin (HSA) and with α1-acid glycoprotein (AGP). Rapid equilibrium dialysis was used for the measurement of protein binding. All samples were analysed by liquid chromatography and tandem mass spectrometry to obtain the unbound fraction (fu ) of MK-467. Unbound fractions (fu ) of MK-467 in canine plasma (mean ± standard deviation) were 27.6 ± 3.5%, 26.6 ± 0.9% and 42.4 ± 1.2% at 0.1, 1.0 and 10 µm concentrations, respectively. In the presence of medetomidine, fu were 27.5 ± 0.4%, 26.6 ± 0.9% and 41.0 ± 2.4%. The fu of MK-467 in HSA were 50.1 ± 2.5% at 0.1 µm, 49.4 ± 1.2% at 1.0 µm and 56.7 ± 0.5% at 10 µm. fu of MK-467 in AGP was 56.3 ± 3.7% at 0.1 µm, 54.6 ± 5.6% at 1.0 µm and 65.3 ± 0.4% at 10 µm. Protein binding of MK-467 was approximately 70% between 0.1 and 1.0 µm. Medetomidine had no apparent effect on the protein binding of MK-467.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Medetomidina/farmacología , Quinolizinas/metabolismo , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Antagonistas de Receptores Adrenérgicos alfa 2/sangre , Animales , Perros , Interacciones Farmacológicas , Masculino , Medetomidina/sangre , Orosomucoide/metabolismo , Unión Proteica/efectos de los fármacos , Quinolizinas/sangre , Albúmina Sérica/metabolismo
10.
Anesth Analg ; 122(5): 1556-66, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26218862

RESUMEN

BACKGROUND: Dexmedetomidine is a highly selective α2-agonist with hypnotic, analgesic, and anxiolytic properties. Despite off-label administration, dexmedetomidine has found a niche in critically ill neonates and infants with congenital heart disease because of its minimal effects on respiratory function at sedative doses, facilitating early extubation and fast-track postoperative care. There are little pharmacokinetic data regarding newborns who have immature drug metabolizing capacity and who are at risk for reduced dexmedetomidine clearance and drug toxicity. The aim of this study was to determine the pharmacokinetics of dexmedetomidine in neonates and infants after open heart surgery. This study included 23 evaluable neonates (age, 1 day-1 month) and 36 evaluable infants (age, 1 month-24 months) after open heart surgery. METHODS: Full-term neonates and infants requiring mechanical ventilation after open heart surgery received dexmedetomidine in a dose-escalation study. Dexmedetomidine was administered as a loading dose over 10 minutes followed by a continuous IV infusion up to 24 hours. Cohorts of 12 infants were enrolled sequentially to receive 0.35, 0.7, or 1 µg/kg dexmedetomidine followed by 0.25, 0.5, or 0.75 µg/kg/h dexmedetomidine, respectively. Cohorts of 9 neonates received 0.25, 0.35, or 0.5 µg/kg dexmedetomidine followed by 0.2, 0.3, or 0.4 µg/kg/h dexmedetomidine, respectively. Plasma dexmedetomidine concentrations were determined using a validated high-performance liquid chromatography-tandem mass spectrometry assay. A population nonlinear mixed effects modeling approach was used to characterize dexmedetomidine pharmacokinetics. RESULTS: Pharmacokinetic parameters of dexmedetomidine were estimated using a 2-compartment disposition model with weight allometrically scaled as a covariate on drug clearance, intercompartmental clearance, central and peripheral volume of distributions and age, total bypass time, and intracardiac shunting on clearance. Dexmedetomidine demonstrated a plasma drug clearance of 657 × (weight/70) mL/min, intercompartmental clearance of 6780 × (weight/70) mL/min, central volume of distribution of 88 × (weight/70) L and peripheral volume of distribution of 112 × (weight/70) L for a typical subject with age >1 month with a cardiopulmonary bypass time of 60 minutes and without right-to-left intracardiac shunt. Dexmedetomidine pharmacokinetics may be influenced by age during the neonatal period, weight, total bypass time, and presence of intracardiac shunt. CONCLUSIONS: Dexmedetomidine clearance is significantly diminished in full-term newborns and increases rapidly in the first few weeks of life. The dependence of clearance on age during the first few weeks of life reflects the relative immaturity of metabolic processes during the newborn period. Continuous infusions of up to 0.3 µg/kg/h in neonates and 0.75 µg/kg/h in infants were well tolerated after open heart surgery.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Analgésicos no Narcóticos/farmacocinética , Procedimientos Quirúrgicos Cardíacos , Dexmedetomidina/farmacocinética , Cardiopatías Congénitas/cirugía , Hipnóticos y Sedantes/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Agonistas de Receptores Adrenérgicos alfa 2/efectos adversos , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Factores de Edad , Analgésicos no Narcóticos/administración & dosificación , Analgésicos no Narcóticos/efectos adversos , Analgésicos no Narcóticos/sangre , Preescolar , Cromatografía Líquida de Alta Presión , Simulación por Computador , Dexmedetomidina/administración & dosificación , Dexmedetomidina/efectos adversos , Dexmedetomidina/sangre , Esquema de Medicación , Cálculo de Dosificación de Drogas , Monitoreo de Drogas/métodos , Cardiopatías Congénitas/diagnóstico , Humanos , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/efectos adversos , Hipnóticos y Sedantes/sangre , Lactante , Recién Nacido , Infusiones Intravenosas , Tasa de Depuración Metabólica , Modelos Biológicos , Método de Montecarlo , Dinámicas no Lineales , Respiración Artificial , Espectrometría de Masas en Tándem , Nacimiento a Término , Resultado del Tratamiento
11.
BMC Pediatr ; 15: 117, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26357864

RESUMEN

BACKGROUND: Chronic Fatigue Syndrome (CFS) is a common and disabling condition in adolescence with few treatment options. A central feature of CFS is orthostatic intolerance and abnormal autonomic cardiovascular control characterized by sympathetic predominance. We hypothesized that symptoms as well as the underlying pathophysiology might improve by treatment with the alpha2A-adrenoceptor agonist clonidine. METHODS: A total of 176 adolescent CFS patients (12-18 years) were assessed for eligibility at a single referral center recruiting nation-wide. Patients were randomized 1:1 by a computer system and started treatment with clonidine capsules (25 µg or 50 µg twice daily, respectively, for body weight below/above 35 kg) or placebo capsules for 9 weeks. Double-blinding was provided. Data were collected from March 2010 until October 2012 as part of The Norwegian Study of Chronic Fatigue Syndrome in Adolescents: Pathophysiology and Intervention Trial (NorCAPITAL). Effect of clonidine intervention was assessed by general linear models in intention-to-treat analyses, including baseline values as covariates in the model. RESULTS: A total of 120 patients (clonidine group n = 60, placebo group n = 60) were enrolled and started treatment. There were 14 drop-outs (5 in the clonidine group, 9 in the placebo group) during the intervention period. At 8 weeks, the clonidine group had lower plasma norepinephrine (difference = 205 pmol/L, p = 0.05) and urine norepinephrine/creatinine ratio (difference = 3.9 nmol/mmol, p = 0.002). During supine rest, the clonidine group had higher heart rate variability in the low-frequency range (LF-HRV, absolute units) (ratio = 1.4, p = 0.007) as well as higher standard deviation of all RR-intervals (SDNN) (difference = 12.0 ms, p = 0.05); during 20° head-up tilt there were no statistical differences in any cardiovascular variable. Symptoms of orthostatic intolerance did not change during the intervention period. CONCLUSIONS: Low-dose clonidine reduces catecholamine levels in adolescent CFS, but the effects on autonomic cardiovascular control are sparse. Clonidine does not improve symptoms of orthostatic intolerance. TRIAL REGISTRATION: Clinical Trials ID: NCT01040429, date of registration 12/28/2009.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Clonidina/administración & dosificación , Síndrome de Fatiga Crónica/tratamiento farmacológico , Síndrome de Fatiga Crónica/fisiopatología , Adolescente , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Clonidina/sangre , Creatinina/orina , Método Doble Ciego , Epinefrina/sangre , Epinefrina/orina , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Norepinefrina/sangre , Norepinefrina/orina , Intolerancia Ortostática/tratamiento farmacológico , Intolerancia Ortostática/fisiopatología , Pruebas de Mesa Inclinada
12.
Pediatr Res ; 77(6): 772-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25751572

RESUMEN

BACKGROUND: The α2-adrenergic agonist dexmedetomidine (DEX) is increasingly used for prolonged sedation of critically ill neonates, but there are currently no data evaluating possible consequences of prolonged neonatal DEX exposure. We evaluated the pharmacokinetics and histological consequences of neonatal DEX exposure. METHODS: DEX was administered (s.c.) to naive (uninjured) neonatal Lewis rats to provide acute (25 µg/kg, ×1) or prolonged (25 µg/kg three times daily, ×2 or ×4 d) exposure. Therapeutic hypothermia was simulated using a water-cooled blanket. Cranial temperatures were measured using an infrared thermometer. DEX concentrations were measured by LC-MS in plasma and homogenized brainstem tissue for pharmacokinetic analysis. Cortex, cerebellum, and brainstem were evaluated for evidence of inflammation or injury. RESULTS: Prolonged neonatal DEX exposure was not associated with renal or brain pathology or indices of gliosis, macrophage activation, or apoptosis in either hypothermic or control rats. Plasma and brain DEX concentrations were tightly correlated. DEX peaked within 15 min in brain and reduced cranial temperature from 32 to 30 °C within 30 min after injection in cooled rats. CONCLUSION: Prolonged DEX treatment in neonatal rats was not associated with abnormal brain histology. These data provide reassuring preliminary results for using DEX with therapeutic hypothermia to treat near-term brain injury.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Encéfalo/efectos de los fármacos , Dexmedetomidina/farmacocinética , Hipotermia/fisiopatología , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Animales , Animales Recién Nacidos , Temperatura Corporal/efectos de los fármacos , Encéfalo/patología , Encéfalo/fisiología , Cromatografía Liquida , Dexmedetomidina/sangre , Dexmedetomidina/farmacología , Evaluación Preclínica de Medicamentos , Espectrometría de Masas , Ratas , Ratas Endogámicas Lew
13.
Int J Clin Pharmacol Ther ; 53(4): 301-16, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25109412

RESUMEN

OBJECTIVES: Guanfacine extended- release (GXR) is approved for the treatment of attention-deficit/hyperactivity disorder in children and adolescents. As part of the clinical development of GXR, and to further explore the effect of guanfacine on QT intervals, a thorough QT study of guanfacine was conducted (ClinicalTrials. gov identifier: NCT00672984). METHODS: In this double-blind, 3-period, crossover trial, healthy adults (n = 83) received immediaterelease guanfacine (at therapeutic (4 mg) and supra-therapeutic (8 mg) doses), placebo, and 400 mg moxifloxacin (positive control) in 1 of 6 randomly assigned sequences. Continuous 12-lead electrocardiograms were extracted, and guanfacine plasma concentrations were assessed pre-dose and at intervals up to 24 hours post-dose. QT intervals were corrected using 2 methods: subject-specific (QTcNi) and Fridericia (QTcF). Time-matched analyses examined the largest, baseline-adjusted, drug-placebo difference in QTc intervals. RESULTS: In the QTcNi analysis, the largest 1-sided 95% upper confidence bound (UCB) through hour 12 was 1.94 ms (12 hours postdose). For the 12-hour QTcF analysis, the largest 1-sided 95% UCB was 10.34 ms (12 hours post-supratherapeutic dose), representing the only 1-sided 95% UCB > 10 ms. Following the supra-therapeutic dose, maximum guanfacine plasma concentration was attained at 5.0 hours (median) post-dose. Assay sensitivity was confirmed by moxifloxacin results. Among guanfacine-treated subjects, most treatment-emergent adverse events were mild (78.9%); dry mouth (65.8%) and dizziness (61.8%) were most common. CONCLUSIONS: Neither therapeutic nor supra-therapeutic doses of guanfacine prolonged QT interval after adjusting for heart rate using individualized correction, QTcNi, through 12 hours postdose. Guanfacine does not appear to interfere with cardiac repolarization of the form associated with pro-arrhythmic drugs.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/efectos adversos , Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Guanfacina/efectos adversos , Sistema de Conducción Cardíaco/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Potenciales de Acción , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Adulto , Estudios Cruzados , Método Doble Ciego , Electrocardiografía , Femenino , Guanfacina/sangre , Guanfacina/farmacocinética , Voluntarios Sanos , Sistema de Conducción Cardíaco/fisiología , Humanos , Masculino , Seguridad del Paciente , Medición de Riesgo , Factores de Riesgo , Adulto Joven
14.
AAPS J ; 16(6): 1237-46, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25135837

RESUMEN

Using a previously developed population pharmacokinetic model, an exposure-response (ER) model was successfully developed to describe guanfacine plasma concentrations and changes in heart rate (HR) and the QT interval. Guanfacine exposure was associated with small decreases in HR and a small prolongation of the population-corrected QT (QTcP) interval. Based on the final ER model for effect of guanfacine on HR, the estimated population typical decrease in HR would be 2.3% (2.1-2.7%) of the baseline circadian HR for every 1 ng/mL of guanfacine exposure. A QTcP was developed for the analysis using the sampled population. An effect of sex on baseline-corrected QT (BQTP) was the only covariate effect in the final ER model for QTcP, its inclusion resulting in a typical baseline QTcP estimate that is 9 (5-13) ms higher for females. There was no evidence of QT-RR hysteresis. A linear model was used to relate guanfacine plasma concentrations to QTcP. The typical (95% confidence interval) slope parameter was estimated to be 0.941 (0.62-1.25) ms/ng/mL. The final model predicted an approximate 1-ms increase from baseline for every 1 ng/mL of guanfacine in plasma. The main predictor of QTcP prolongation was guanfacine exposure, which decreased with body weight and increased with dose.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2 , Guanfacina , Frecuencia Cardíaca/efectos de los fármacos , Modelos Biológicos , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Factores de Edad , Niño , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Bases de Datos Factuales , Electrocardiografía , Femenino , Guanfacina/sangre , Guanfacina/farmacología , Humanos , Masculino , Análisis Multivariante , Factores Sexuales
15.
Acta Anaesthesiol Scand ; 58(6): 733-42, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24724965

RESUMEN

BACKGROUND: The highly selective α2 -adrenoreceptor agonist, dexmedetomidine, exerts neuroprotective, analgesic, anti-inflammatory and sympatholytic properties that may be beneficial for perinatal asphyxia. The optimal safe dose for pre-clinical newborn neuroprotection studies is unknown. METHODS: Following cerebral hypoxia-ischaemia, dexmedetomidine was administered to nine newborn piglets in a de-escalation dose study in combination with hypothermia (whole body cooling to 33.5°C). Dexmedetomidine was administered with a loading dose of 1 µg/kg and maintenance infusion at doses from 10 to 0.6 µg/kg/h. One additional piglet was not subjected to hypoxia-ischaemia. Blood for pharmacokinetic analysis was sampled pre-insult and frequently post-insult. A one-compartment linear disposition model was used to fit data. Population parameter estimates were obtained using non-linear mixed effects modelling. RESULTS: All dexmedetomidine infusion regimens led to plasma concentrations above those associated with sedation in neonates and children (0.4-0.8 µg/l). Seven out of the nine piglets with hypoxia-ischaemia experienced periods of bradycardia, hypotension, hypertension and cardiac arrest; all haemodynamic adverse events occurred in piglets with plasma concentrations greater than 1 µg/l. Dexmedetomidine clearance was 0.126 l/kg/h [coefficient of variation (CV) 46.6.%] and volume of distribution was 3.37 l/kg (CV 191%). Dexmedetomidine clearance was reduced by 32.7% at a temperature of 33.5°C. Dexmedetomidine clearance was reduced by 55.8% following hypoxia-ischaemia. CONCLUSIONS: Dexmedetomidine clearance was reduced almost tenfold compared with adult values in the newborn piglet following hypoxic-ischaemic brain injury and subsequent therapeutic hypothermia. Reduced clearance was related to cumulative effects of both hypothermia and exposure to hypoxia. High plasma levels of dexmedetomidine were associated with major cardiovascular complications.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Asfixia Neonatal/complicaciones , Dexmedetomidina/farmacocinética , Hipotermia Inducida , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Fármacos Neuroprotectores/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Agonistas de Receptores Adrenérgicos alfa 2/uso terapéutico , Animales , Dexmedetomidina/sangre , Dexmedetomidina/uso terapéutico , Modelos Animales de Enfermedad , Hipoxia-Isquemia Encefálica/etiología , Masculino , Tasa de Depuración Metabólica , Fármacos Neuroprotectores/sangre , Fármacos Neuroprotectores/uso terapéutico , Dinámicas no Lineales , Sus scrofa , Porcinos
16.
J Toxicol Sci ; 39(2): 251-62, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24646706

RESUMEN

The neuroprotective effects of dexmedetomidine have been reported by many investigators; however its underlying mechanism to reduce neuronal injury during a prolonged anesthesia remains unclear. In this study, we investigated the neurotoxic effects of dexmedetomidine in fetal monkey brains. In the present study, we compare the neurotoxic effects of dexmedetomidine and ketamine, a general anesthetic with a different mechanism of action, in fetal cynomolgus monkeys. Twenty pregnant monkeys at approximate gestation day 120 were divided into 4 groups: non-treatment controls (Group 1); ketamine at 20 mg/kg intramuscularly followed by a 12-hr infusion at 20-50 mg/kg/hr (Group 2); dexmedetomidine at 3 µg/kg intravenously (i.v.) over 10 min followed by a 12-hr infusion at the human equivalent dose (HED) of 3 µg/kg/hr (Group 3); and dexmedetomidine at 30 µg/kg i.v. over 10 min followed by a 12-hr infusion at 30 µg/kg/hr, 10 times HED (Group 4). Blood samples from both dams and fetuses were measured for concentration of dexmedetomidine. Each fetus was perfusion-fixed, serial sections were cut through the frontal cortex, and stained to detect for apoptosis (activated caspase 3 and TUNEL) and neurodegeneration (silver stain). In utero treatment with ketamine resulted in marked apoptosis and degeneration primarily in layers I and II of the frontal cortex. In contrast, fetal brains from animals treated with dexmedetomidine showed none to minimal neuroapoptotic or neurodegenerative lesions at both low- and high-dose treatments. Plasma levels confirmed systemic exposure of dexmedetomidine in both dams and fetuses. In conclusion, these results demonstrate that dexmedetomidine at both low-dose (HED) and high-dose (10 times HED) does not induce apoptosis in the frontal cortex (layers I, II, and III) of developing brain of cynomolgus monkeys.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/toxicidad , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Dexmedetomidina/toxicidad , Hipnóticos y Sedantes/toxicidad , Agonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Animales , Encéfalo/enzimología , Encéfalo/patología , Caspasa 3/metabolismo , Dexmedetomidina/administración & dosificación , Dexmedetomidina/sangre , Relación Dosis-Respuesta a Droga , Femenino , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/sangre , Ketamina/administración & dosificación , Ketamina/sangre , Ketamina/toxicidad , Macaca fascicularis , Intercambio Materno-Fetal , Enfermedades Neurodegenerativas/inducido químicamente , Corteza Prefrontal , Embarazo
17.
Br J Dermatol ; 171(1): 162-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24506775

RESUMEN

BACKGROUND: Persistent facial erythema is the most common primary pathological feature of rosacea, the only treatment for which is brimonidine tartrate (BT) gel. OBJECTIVES: To assess the relative bioavailability of topical BT gel in comparison with the ophthalmic BT solution. METHODS: A pharmacokinetic study was conducted to compare intraindividual systemic exposures after dermal application of BT gel (0·07%, 0·18% and 0·5%) under maximal use conditions in patients with moderate-to-severe facial erythema associated with rosacea, and administration of BT ophthalmic solution 0·2%. RESULTS: Patients who received BT ophthalmic solution 0·2% three times a day for 1 day had a mean Cmax of 54 ± 28 pg mL(-1) and a mean 0-24-h area under the curve (AUC0-24 h ) of 568 ± 277 pg h mL(-1) . Topical application of BT gel for 29 days resulted in quantifiable systemic exposure in 22%, 48%, 71% and 79% of patients who received BT gel 0·07% twice daily, 0·18% once daily, 0·18% twice daily and 0·5% once daily, respectively. The mean Cmax values for the BT gels ranged between 13 and 25 pg mL(-1) , and mean AUC0-24 h values ranged between 42 and 290 pg h mL(-1) . Systemic exposure increased with applied dose, with no drug accumulation for the duration of treatment. The systemic exposure observed with the highest dose of BT gel (0·5% once daily) was significantly lower than the systemic levels observed for the ophthalmic solution. 0·2% apply for all the concentrations. CONCLUSIONS: The systemic safety profile of BT gel may be considered better than that of the ophthalmic solution.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Dermatosis Facial/tratamiento farmacológico , Soluciones Oftálmicas/farmacocinética , Quinoxalinas/farmacocinética , Rosácea/tratamiento farmacológico , Administración Cutánea , Agonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Adulto , Área Bajo la Curva , Disponibilidad Biológica , Tartrato de Brimonidina , Esquema de Medicación , Dermatosis Facial/metabolismo , Femenino , Geles , Humanos , Masculino , Quinoxalinas/administración & dosificación , Quinoxalinas/sangre , Rosácea/metabolismo
18.
Biomed Chromatogr ; 27(12): 1708-13, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23832392

RESUMEN

A simple, practical, accurate and sensitive liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed and fully validated for the quantitation of guanfacine in beagle dog plasma. After protein precipitation by acetonitrile, the analytes were separated on a C18 chromatographic column by methanol and water containing 0.1% (v/v) formic acid with a gradient elution. The subsequent detection utilized a mass spectrometry under positive ion mode with multiple reaction monitoring of guanfacine and enalaprilat (internal standard) at m/z 246.2 → 159.0 and m/z 349.2 → 205.9, respectively. Good linearity was obtained over the concentration range of 0.1-20 ng/mL for guanfacine in dog plasma and the lower limit of quantification of this method was 0.1 ng/mL. The intra- and inter-day precisions were <10.8% relative standard deviation with an accuracy of 92.9-108.4%. The matrix effects ranged from 89.4 to 100.7% and extraction recoveries were >90%. Stability studies showed that both analytes were stable during sample preparation and analysis. The established method was successfully applied to an in vivo pharmacokinetic study in beagle dogs after a single oral dose of 4 mg guanfacine extended-release tablets.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/sangre , Cromatografía Liquida/métodos , Guanfacina/sangre , Espectrometría de Masas en Tándem/métodos , Agonistas de Receptores Adrenérgicos alfa 2/química , Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Animales , Perros , Femenino , Guanfacina/química , Guanfacina/farmacocinética , Modelos Lineales , Masculino , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
19.
Bioanalysis ; 4(12): 1445-56, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22793029

RESUMEN

BACKGROUND: Direct analysis of dried blood spot (DBS) samples was investigated using a prototype semi-automated robotic device that allows the direct elution of sample spots from a DBS paper card to an online SPE cartridge. The eluted SPE samples were analyzed with high-performance LC-MS/MS. RESULTS: A LLOQ of 0.01 ng/ml was achieved with a linear calibration range from 0.01 to 25 ng/ml. Optimal performance data were obtained from spotting the internal standard solution on the card before blood spotting. Internal standard addition from the system injector loop produced intra-assay inaccuracy of -9.0-7.3% and precision of 1.3-8.2%, and inter-assay inaccuracy of -3.5-3.9% and precision of 4.4-8.7%. CONCLUSION: Results demonstrated the feasibility of a semi-automated online rapid direct elution method that avoids manual extraction for DBS sample analysis using the online DBS-SPE system coupled to LC-MS/MS.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/sangre , Cromatografía de Fase Inversa/métodos , Pruebas con Sangre Seca/instrumentación , Pruebas con Sangre Seca/métodos , Guanfacina/sangre , Espectrometría de Masas en Tándem/métodos , Animales , Automatización de Laboratorios/métodos , Calibración , Humanos , Límite de Detección , Modelos Lineales , Estándares de Referencia , Extracción en Fase Sólida/métodos , Espectrometría de Masas en Tándem/instrumentación
20.
Vet J ; 193(2): 481-5, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22277719

RESUMEN

Six healthy laboratory Beagles were treated IV with 10 µg/kg dexmedetomidine (DEX) or 10 µg/kg dexmedetomidine combined with 500 µg/kg MK-467 in the same syringe (DMK) in a randomised cross-over design with a 14 day washout. Blood was collected immediately before treatment and 35, 60 and 120 min post-injection through a central venous catheter. The plasma concentrations of glucose, insulin, non-esterified free fatty acids (NEFAs), lactate and cortisol were determined. A repeated-measures ANOVA test was used to compare treatments and effects for each sample time point. Significant differences between treatments were found for plasma glucose (P=0.037) and insulin (P=0.009). DEX significantly increased plasma glucose at 120 min, but reduced plasma insulin at 35 and 60 min. NEFA decreased for both treatments at 35 min. This reduction was transient for DMK, whereas it persisted during the follow up period for DEX. Plasma lactate concentrations increased at 35 and 60 min with DEX. Neither treatment altered plasma cortisol concentrations. The addition of MK-467 to dexmedetomidine prevented or abolished most metabolic changes in healthy Beagles.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Antagonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Anestésicos Intravenosos/farmacocinética , Dexmedetomidina/farmacocinética , Quinolizinas/farmacocinética , Administración Intravenosa/veterinaria , Agonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Antagonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Antagonistas de Receptores Adrenérgicos alfa 2/sangre , Análisis de Varianza , Anestésicos Intravenosos/administración & dosificación , Anestésicos Intravenosos/sangre , Animales , Análisis Químico de la Sangre/veterinaria , Análisis de los Gases de la Sangre/veterinaria , Glucemia/análisis , Colorimetría/veterinaria , Estudios Cruzados , Dexmedetomidina/administración & dosificación , Dexmedetomidina/sangre , Perros , Combinación de Medicamentos , Femenino , Hidrocortisona/sangre , Insulina/sangre , Masculino , Modelos Biológicos , Quinolizinas/administración & dosificación , Quinolizinas/sangre , Radioinmunoensayo/veterinaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...