Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Clin Pharmacol ; 77(6): 895-902, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33404754

RESUMO

PURPOSE: Tizanidine, an alpha-adrenergic substance with antinociceptive and antihypertensive effects, is extensively metabolized via cytochrome P450 (CYP) 1A2. Therefore, coadministration with potent CYP1A2 inhibitors, such as ciprofloxacin, is contraindicated. However, both drugs are broadly utilized in various countries. Their concomitant use bears an inherent high risk for clinically significant symptoms, especially in multimorbid patients experiencing polypharmacy. This study aims to investigate the impact of coadministration of tizanidine and ciprofloxacin using real-world pharmacovigilance data and to raise awareness of this potentially underestimated safety issue. METHODS: We conducted a retrospective study including Individual Case Safety Reports (ICSR) registered until March 1, 2017, in the World Health Organization (WHO) global database. Demographic data, drug administration information, the course of the adverse drug reaction (ADR), its severity, and outcomes were analyzed for cases reporting ciprofloxacin comedication. RESULTS: In 91 (2.0%) of the identified 4192 worldwide ICSR on tizanidine, coadministration of ciprofloxacin was reported. Most of the patients were female (n = 59, 64.8%) with a median age of 54 years (range 13-85 years). The countries contributing most reports were the USA (n = 54, 59.3%) and Switzerland (n = 16, 17.6%). ADRs reported most often affected the nervous system and the cardiac function, especially with large tizanidine doses or drugs with CNS and cardiovascular depressant effects. In two cases, a fatal outcome was reported. CONCLUSION: Despite the existing formal contraindication, the concomitant use of tizanidine and ciprofloxacin can be observed in real-world clinical practice. Reactions mainly affected the central nervous and the cardiovascular system resulting in potentially severe adverse effects. The concomitant use of tizanidine and ciprofloxacin should absolutely be avoided.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Antibacterianos/farmacocinética , Ciprofloxacina/farmacocinética , Clonidina/análogos & derivados , Adolescente , Agonistas de Receptores Adrenérgicos alfa 2/efeitos adversos , Adulto , Idoso , Idoso de 80 Anos ou mais , Antibacterianos/efeitos adversos , Área Sob a Curva , Ciprofloxacina/efeitos adversos , Clonidina/efeitos adversos , Clonidina/farmacocinética , Bases de Dados Factuais , Interações Medicamentosas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Farmacovigilância , Estudos Retrospectivos , Organização Mundial da Saúde , Adulto Jovem
2.
J Clin Psychopharmacol ; 39(2): 124-128, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30707118

RESUMO

BACKGROUND: Guanfacine is Food and Drug Administration approved for hypertension and attention-deficit hyperactivity disorder and has been used off-label for migraine prophylaxis, heroin withdrawal, and more recently smoking cessation. Previous studies have shown positive effects of 3 mg/d of immediate-release (IR) guanfacine on smoking outcomes, but the dose equivalency of the IR and extended-release (ER) formulations is unknown. PROCEDURES: A within-subject design was used to compare the pharmacokinetics and pharmacodynamics of 3 mg/d of IR, 4 mg/d of ER, and 6 mg/d of ER guanfacine in adult daily smokers (n = 5). Plasma medication levels, vital signs, cigarettes per day, tobacco craving, and adverse events were assessed. Medication was titrated to stable dosing after each laboratory day (3 mg/d IR, then 4 mg/d ER, then 6 mg/d ER). RESULTS: Plasma medication levels did not differ between the 3 mg/d of IR and 4 mg/d of ER doses after 24 hours from last dose and were highest at the 6 mg/d of ER dose (3 mg/d IR: M = 3.40 ng/mL, SE = 0.34 vs 4 mg/d ER: M = 3.46 ng/mL, SE = 0.67 vs 6 mg/d ER: M = 5.92 ng/mL, SE = 1.02). All doses of guanfacine decreased heart rate and blood pressure from baseline. Absolute values of cigarettes per day (6 mg/d ER) and tobacco craving (4 and 6 mg/d ER) were lowest with the ER formulations. Treatment-emergent adverse events were subject rated as minimal to mild, except dry mouth. CONCLUSIONS: We demonstrated similar pharmacokinetic profiles between 3 mg/d of IR guanfacine and 4 mg/d of ER guanfacine, as hypothesized. All doses of guanfacine were well tolerated.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Guanfacina/administração & dosagem , Abandono do Hábito de Fumar/métodos , Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Adulto , Pressão Sanguínea/efeitos dos fármacos , Preparações de Ação Retardada , Relação Dose-Resposta a Droga , Liberação Controlada de Fármacos , Feminino , Guanfacina/farmacocinética , Guanfacina/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento
3.
Anesth Analg ; 129(6): 1519-1528, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31743171

RESUMO

BACKGROUND: Dexmedetomidine is increasingly used off-label in infants and children with cardiac disease during cardiopulmonary bypass (CPB) and in the postoperative period. Despite its frequent use, optimal dosing of dexmedetomidine in the setting of CPB has not been identified but is expected to differ from dosing in those not supported with CPB. This study had the following aims: (1) characterize the effect of CPB on dexmedetomidine clearance (CL) and volume of distribution (V) in infants and young children; (2) characterize tolerance and sedation in patients receiving dexmedetomidine; and (3) identify preliminary dosing recommendations for infants and children undergoing CPB. We hypothesized that CL would decrease, and V would increase during CPB compared to pre- or post-CPB states. METHODS: Open-label, single-center, opportunistic pharmacokinetics (PK) and safety study of dexmedetomidine in patients ≤36 months of age administered dexmedetomidine per standard of care via continuous infusion. We analyzed dexmedetomidine PK data using standard nonlinear mixed effects modeling with NONMEM software. We compared model-estimated PK parameters to those from historical patients receiving dexmedetomidine before anesthesia for urologic, lower abdominal, or plastic surgery; after low-risk cardiac or craniofacial surgery; or during bronchoscopy or nuclear magnetic resonance imaging. We investigated the influence of CPB-related factors on PK estimates and used the final model to simulate dosing recommendations, targeting a plasma concentration previously associated with safety and efficacy (0.6 ng/mL). We used the Wilcoxon rank sum test to evaluate differences in dexmedetomidine exposure between infants with hypotension or bradycardia and those who did not develop these adverse events. RESULTS: We collected 213 dexmedetomidine plasma samples from 18 patients. Patients had a median (range) age of 3.3 months (0.1-34.0 months) and underwent CPB for 161 minutes (63-394 minutes). We estimated a CL of 13.4 L/h/70 kg (95% confidence interval, 2.6-24.2 L/h/70 kg) during CPB, compared to 42.1 L/h/70 kg (95% confidence interval, 38.7-45.8 L/h/70 kg) in the historical patients. No specific CPB-related factor had a statistically significant effect on PK. A loading dose of 0.7 µg/kg over 10 minutes before CPB, followed by maintenance infusions through CPB of 0.2 or 0.25 µg/kg/h in infants with postmenstrual ages of 42 or 92 weeks, respectively, maintained targeted concentrations. We identified no association between dexmedetomidine exposure and selected adverse events (P = .13). CONCLUSIONS: CPB is associated with lower CL during CPB in infants and young children compared to those not undergoing CPB. Further study should more closely investigate CPB-related factors that may influence CL.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Ponte Cardiopulmonar , Dexmedetomidina/farmacocinética , Hipnóticos e Sedativos/administração & dosagem , Hipnóticos e Sedativos/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Agonistas de Receptores Adrenérgicos alfa 2/efeitos adversos , Fatores Etários , Ponte Cardiopulmonar/efeitos adversos , Pré-Escolar , Estado de Consciência/efeitos dos fármacos , Dexmedetomidina/administração & dosagem , Dexmedetomidina/efeitos adversos , Cálculos da Dosagem de Medicamento , Feminino , Humanos , Hipnóticos e Sedativos/efeitos adversos , Lactente , Recém-Nascido , Masculino , Taxa de Depuração Metabólica , Modelos Biológicos , North Carolina , Uso Off-Label , Projetos Piloto
4.
Biopharm Drug Dispos ; 40(8): 282-293, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31313320

RESUMO

Guanfacine is used for the treatment of attention-deficit/hyperactivity disorder (ADHD). Using liquid chromatography-tandem mass spectrometry (LC-MS/MS), metabolite profiling of guanfacine was performed in plasma and urine collected from healthy Japanese adults following repeated oral administration of guanfacine extended-release formulation. Unchanged guanfacine was the most abundant component in both plasma and urine (from the MS signal intensity). In plasma, the M3 metabolite (a sulfate of hydroxy-guanfacine) was the prominent metabolite; the M2 metabolite (a glucuronide of a metabolite formed by monooxidation of guanfacine), 3-hydroxyguanfacine and several types of glucuronide at different positions on guanfacine were also detected. In urine, the M2 metabolite and 3-hydroxyguanfacine were the principal metabolites. From metabolite analysis, the proposed main metabolic pathway of guanfacine is monooxidation on the dichlorobenzyl moiety, followed by glucuronidation or sulfation. A minor pathway is glucuronidation at different positions on guanfacine. As the prominent metabolites in plasma were glucuronide and sulfate of hydroxyguanfacine, which have no associated toxicity concerns, further toxicity studies of the metabolites, for example in animals, were not deemed necessary.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Glucuronídeos/farmacocinética , Guanfacina/administração & dosagem , Sulfatos/farmacocinética , Administração Oral , Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Adulto , Cromatografia Líquida , Preparações de Ação Retardada , Guanfacina/farmacocinética , Humanos , Japão , Masculino , Comprimidos , Espectrometria de Massas em Tandem , Adulto Jovem
5.
Microsc Microanal ; 25(6): 1352-1366, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31018876

RESUMO

Brimonidine, an anti-glaucoma medicine, acts as an adrenergic agonist which decreases the synthesis of aqueous humour and increases the amount of drainage through Schlemm's canal and trabecular meshwork, but shows dose-dependent (0.2% solution thrice daily) toxicity. To reduce the side effects and improve the efficacy, brimonidine was nanoencapsulated on ultra-small-sized chitosan nanoparticles (nanobrimonidine) (28 ± 4 nm) with 39% encapsulation efficiency, monodispersity, freeze-thawing capability, storage stability, and 2% drug loading capacity. This nanocomplex showed burst, half, and complete release at 0.5, 45, and 100 h, respectively. Nanobrimonidine did not show any in vitro toxicity and was taken up by caveolae-mediated endocytosis. The nanobrimonidine-treated trabeculectomy tissue of glaucoma patients showed better dilation of the trabecular meshwork under the electron microscope. This is direct evidence for better bioavailability of nanobrimonidine after topical administration. Thus, the developed nanobrimonidine has the potential to improve the efficacy, reduce dosage and frequency, and improve delivery to the anterior chamber of the eye.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Tartarato de Brimonidina/administração & dosagem , Glaucoma/tratamento farmacológico , Nanocompostos/administração & dosagem , Malha Trabecular/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 2/efeitos adversos , Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Tartarato de Brimonidina/efeitos adversos , Tartarato de Brimonidina/farmacocinética , Quitosana/administração & dosagem , Portadores de Fármacos/administração & dosagem , Humanos , Modelos Teóricos , Resultado do Tratamento
6.
J Vet Pharmacol Ther ; 42(6): 738-744, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31584710

RESUMO

The alpha(α)2 -agonist detomidine is used for equine sedation with opioids such as methadone. We retrieved the data from two randomized, crossover studies where detomidine and methadone were given intravenously alone or combined as boli (STUDY 1) (Gozalo-Marcilla et al., 2017, Veterinary Anaesthesia and Analgesia, 2017, 44, 1116) or as 2-hr constant rate infusions (STUDY 2) (Gozalo-Marcilla et al., 2019, Equine Veterinary Journal, 51, 530). Plasma drug concentrations were measured with a validated tandem Mass Spectrometry assay. We used nonlinear mixed effect modelling and took pharmacokinetic (PK) data from both studies to fit simultaneously both drugs and explore their nonlinear kinetics. Two significant improvements over the classical mammillary two-compartment model were identified. First, the inclusion of an effect of detomidine plasma concentration on the elimination clearances (Cls) of both drugs improved the fit of detomidine (Objective Function Value [OFV]: -160) and methadone (OFV: -132) submodels. Second, a detomidine concentration-dependent reduction of distributional Cls of each drug further improved detomidine (OFV: -60) and methadone (OFV: -52) submodel fits. Using the PK data from both studies (a) helped exploring hypotheses on the nonlinearity of the elimination and distributional Cls and (b) allowed inclusion of dynamic effects of detomidine plasma concentration in the model which are compatible with the pharmacology of detomidine (vasoconstriction and reduction in cardiac output).


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Analgésicos Opioides/farmacocinética , Cavalos , Imidazóis/farmacocinética , Metadona/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Animais , Combinação de Medicamentos , Imidazóis/administração & dosagem , Metadona/administração & dosagem , Distribuição Tecidual
7.
Br J Clin Pharmacol ; 84(6): 1364-1372, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29495085

RESUMO

AIMS: Alpha-2 agonists are direct peripheral vasoconstrictors, which achieve these effects by activating vascular smooth muscle alpha-2 adrenoceptors. The impact of this response during dexmedetomidine infusion remains poorly quantified. Our goal was to investigate the pharmacokinetic (PK) and pharmacodynamic (PD, vasoconstriction) effects of a computer-controlled dexmedetomidine infusion in healthy volunteers. METHODS: After local ethics committee approval, we studied 10 healthy volunteers. To study the peripheral vasoconstrictive effect of dexmedetomidine without concurrent sympatholytic effects, sympathetic fibres were blocked with a brachial plexus block. Volunteers received a dexmedetomidine target-controlled infusion for 15 min, to a target concentration of 0.3 ng ml-1 . Arterial blood samples were collected during and for 60 min after dexmedetomidine infusion for PK analysis. Peripheral vasoconstriction (PD) was assessed using finger photoelectric plethysmography. PK/PD analysis was carried out using nonlinear mixed-effect models. RESULTS: We found that the computer-controlled infusion pump delivered mean concentrations greater than 0.3 ng ml-1 over the 15-min infusion duration. The peripheral vasoconstrictive effect correlated with dexmedetomidine plasma concentrations during and after the infusion. A three-compartment model provided a better fit to the data than a two-compartment model. CONCLUSIONS: We found that dexmedetomidine-induced vasoconstriction is concentration dependent over time. Dexmedetomidine PK were best estimated by a three-compartment model with allometric scaling. Our results may contribute to future modelling of dexmedetomidine-induced haemodynamic effects.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Dexmedetomidina/farmacocinética , Dedos/irrigação sanguínea , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Agonistas de Receptores Adrenérgicos alfa 2/efeitos adversos , Adulto , Dexmedetomidina/administração & dosagem , Dexmedetomidina/efeitos adversos , Relação Dose-Resposta a Droga , Feminino , Voluntários Saudáveis , Humanos , Infusões Intravenosas , Masculino , Modelos Biológicos , Dinâmica não Linear , Vasoconstritores/administração & dosagem , Vasoconstritores/efeitos adversos , Adulto Jovem
8.
Anesth Analg ; 127(3): 716-723, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29782406

RESUMO

BACKGROUND: Dexmedetomidine (DEX) is an α-2 adrenergic agonist with sedative and analgesic properties. Although not approved for pediatric use by the Food and Drug Administration, DEX is increasingly used in pediatric anesthesia and critical care. However, very limited information is available regarding the pharmacokinetics of DEX in children. The aim of this study was to investigate DEX pharmacokinetics and pharmacodynamics (PK-PD) in Mexican children 2-18 years of age who were undergoing outpatient surgical procedures. METHODS: Thirty children 2-18 years of age with American Society of Anesthesiologists physical status score of I/II were enrolled in this study. DEX (0.7 µg/kg) was administered as a single-dose intravenous infusion. Venous blood samples were collected, and plasma DEX concentrations were analyzed with a combination of high-performance liquid chromatography and electrospray ionization-tandem mass spectrometry. Population PK-PD models were constructed using the Monolix program. RESULTS: A 2-compartment model adequately described the concentration-time relationship. The parameters were standardized for a body weight of 70 kg by using an allometric model. Population parameters estimates were as follows: mean (between-subject variability): clearance (Cl) (L/h × 70 kg) = 20.8 (27%); central volume of distribution (V1) (L × 70 kg) = 21.9 (20%); peripheral volume of distribution (V2) (L × 70 kg) = 81.2 (21%); and intercompartmental clearance (Q) (L/h × 70 kg) = 75.8 (25%). The PK-PD model predicted a maximum mean arterial blood pressure reduction of 45% with an IC50 of 0.501 ng/ml, and a maximum heart rate reduction of 28.9% with an IC50 of 0.552 ng/ml. CONCLUSIONS: Our results suggest that in Mexican children 2-18 years of age with American Society of Anesthesiologists score of I/II, the DEX dose should be adjusted in accordance with lower DEX clearance.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Procedimentos Cirúrgicos Ambulatórios/métodos , Dexmedetomidina/farmacocinética , Hipnóticos e Sedativos/farmacocinética , Adolescente , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Criança , Pré-Escolar , Dexmedetomidina/administração & dosagem , Feminino , Hemodinâmica/efeitos dos fármacos , Hemodinâmica/fisiologia , Humanos , Hipnóticos e Sedativos/administração & dosagem , Infusões Intravenosas , Masculino
9.
J Vet Pharmacol Ther ; 40(5): 569-574, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28097665

RESUMO

Romifidine is an alpha-2 adrenergic agonist used for sedation and analgesia in horses. As it is a prohibited substance, its purported use at low doses in performance horses necessitates further study. The primary goal of the study reported here was to describe the serum concentrations and pharmacokinetics of romifidine following low-dose administration immediately prior to exercise, utilizing a highly sensitive liquid chromatography-tandem mass spectrometry assay that is currently employed in many drug testing laboratories. An additional objective was to describe changes in heart rate and rhythm following intravenous administration of romifidine followed by exercise. Eight adult Quarter Horses received a single intravenous dose of 5 mg (0.01 mg/kg) romifidine followed by 1 h of exercise. Blood samples were collected and drug concentrations measured at time 0 and at various times up to 72 h. Mean ± SD systemic clearance, steady-state volume of distribution and terminal elimination half-life were 34.1 ± 6.06 mL/min/kg and 4.89 ± 1.31 L/kg and 3.09 ± 1.18 h, respectively. Romifidine serum concentrations fell below the LOQ (0.01 ng/mL) and the LOD (0.005 ng/mL) by 24 h postadministration. Heart rate and rhythm appeared unaffected when a low dose of romifidine was administered immediately prior to exercise.


Assuntos
Administração Intravenosa/veterinária , Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Cavalos/metabolismo , Imidazóis/farmacocinética , Condicionamento Físico Animal/fisiologia , Animais , Relação Dose-Resposta a Droga , Frequência Cardíaca , Injeções Intravenosas
10.
Vet Anaesth Analg ; 44(1): 86-97, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27426730

RESUMO

OBJECTIVE: Propranolol has been suggested for anxiolysis in horses, but its sedation efficacy and side effects, both when administered alone and in combination with α2-adrenoceptor agonists, remain undetermined. This study aimed to document the pharmacokinetics and pharmacodynamics of propranolol, romifidine and their combination. STUDY DESIGN: Randomized, crossover study. ANIMALS: Six adult horses weighing 561 ± 48 kg. METHODS: Propranolol (1 mg kg-1; treatment P), romifidine (0.1 mg kg-1; treatment R) or their combination (treatment PR) were administered intravenously with a minimum of 1 week between treatments. Alertness, behavioral responsiveness (visual and tactile) and physiologic variables were measured before and up to 960 minutes after drug administration. Blood was collected for blood gas and acid-base analyses and measurement of plasma drug concentrations. Data were analyzed using repeated-measures analysis of variance or Friedman with Holm-Sidak and Wilcoxon rank-sum tests (p < 0.05). RESULTS: Systemic clearance significantly decreased and the area under the concentration-time curve significantly increased for both drugs in PR compared with P and R. Both PR and R decreased behavioral responsiveness and resulted in sedation for up to 240 and 480 minutes, respectively. Sedation was deeper in PR for the first 16 minutes. Heart rate significantly decreased in all treatments for at least 60 minutes, and PR significantly increased the incidence of severe bradycardia (<20 beats minute-1). CONCLUSIONS AND CLINICAL RELEVANCE: Although not associated with reduced behavioral responsiveness or sedation alone, propranolol augmented romifidine sedation, probably through alterations in romifidine pharmacokinetics, in horses administered PR. The occurrence of severe bradycardia warrants caution in the co-administration of these drugs at the doses studied.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Antagonistas Adrenérgicos beta/farmacocinética , Anestésicos Intravenosos/farmacocinética , Imidazóis/farmacocinética , Propranolol/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Agonistas de Receptores Adrenérgicos alfa 2/efeitos adversos , Antagonistas Adrenérgicos beta/administração & dosagem , Antagonistas Adrenérgicos beta/efeitos adversos , Análise de Variância , Anestésicos Intravenosos/administração & dosagem , Animais , Área Sob a Curva , Comportamento Animal/efeitos dos fármacos , Bradicardia/induzido quimicamente , Bradicardia/veterinária , Estudos Cross-Over , Feminino , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Doenças dos Cavalos/induzido quimicamente , Cavalos , Imidazóis/administração & dosagem , Imidazóis/efeitos adversos , Masculino , Propranolol/administração & dosagem , Propranolol/efeitos adversos
11.
Anesth Analg ; 122(5): 1556-66, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26218862

RESUMO

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.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Analgésicos não Narcóticos/farmacocinética , Procedimentos Cirúrgicos Cardíacos , Dexmedetomidina/farmacocinética , Cardiopatias Congênitas/cirurgia , Hipnóticos e Sedativos/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Agonistas de Receptores Adrenérgicos alfa 2/efeitos adversos , Agonistas de Receptores Adrenérgicos alfa 2/sangue , Fatores Etários , Analgésicos não Narcóticos/administração & dosagem , Analgésicos não Narcóticos/efeitos adversos , Analgésicos não Narcóticos/sangue , Pré-Escolar , Cromatografia Líquida de Alta Pressão , Simulação por Computador , Dexmedetomidina/administração & dosagem , Dexmedetomidina/efeitos adversos , Dexmedetomidina/sangue , Esquema de Medicação , Cálculos da Dosagem de Medicamento , Monitoramento de Medicamentos/métodos , Cardiopatias Congênitas/diagnóstico , Humanos , Hipnóticos e Sedativos/administração & dosagem , Hipnóticos e Sedativos/efeitos adversos , Hipnóticos e Sedativos/sangue , Lactente , Recém-Nascido , Infusões Intravenosas , Taxa de Depuração Metabólica , Modelos Biológicos , Método de Monte Carlo , Dinâmica não Linear , Respiração Artificial , Espectrometria de Massas em Tandem , Nascimento a Termo , Resultado do Tratamento
12.
Int J Clin Pharmacol Ther ; 54(3): 200-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26857784

RESUMO

OBJECTIVES: The objectives of this study were to construct a population pharmacokinetics model for dexmedetomidine used in Chinese adult patients with spinal anesthesia and to identify the key factors affecting the pharmacokinetics of dexmedetomidine. METHODS: A total of 34 subjects (elderly group: n = 15; young group: n = 19) undergoing spinal anesthesia received dexmedetomidine with a loading dose of 0.5 µg×kg(-1) for 10 minutes, followed by a maintenance dose of 0.5 µg×kg-1×h(-1) for 50 minutes. Blood samples were collected until 10 hours after dosing. Laboratory and respiratory parameters, and dexmedetomidine concentrations were measured. A population pharmacokinetic model for dexmedetomidine was constructed using a nonlinear mixed effects model (NONMEM). RESULTS: Pharmacokinetics of dexmedetomidine can be described by a three-compartment model. The respective typical values for clearance (CL), V1, V2, Q2, Q3, and V3 were 0.883 L×min(-1), 17.6 L, 51.5 L, 2.37 L×min(-1), 0.517 L×min(-1), and 44.00 L. Alanine aminotransferase (ALT), age, and body weight were key factors affecting CL, V1, and V2, respectively. CONCLUSIONS: A three-compartment model can be used to describe the pharmacokinetics processing of dexmedetomidine for Chinese adult patients during spinal anesthesia. The population pharmacokinetic of dexmedetomidine was generally in line with results from previous studies.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Raquianestesia , Dexmedetomidina/farmacocinética , Adolescente , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Análise de Regressão
13.
J Vet Pharmacol Ther ; 39(5): 460-8, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26896345

RESUMO

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.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Antagonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Dexmedetomidina/farmacocinética , Quinolizinas/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Agonistas de Receptores Adrenérgicos alfa 2/sangue , Antagonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Antagonistas de Receptores Adrenérgicos alfa 2/sangue , Animais , Gatos , Dexmedetomidina/administração & dosagem , Dexmedetomidina/sangue , Relação Dose-Resposta a Droga , Interações Medicamentosas , Injeções Intravenosas/veterinária , Masculino , Quinolizinas/administração & dosagem , Quinolizinas/sangue
14.
J Vet Pharmacol Ther ; 39(2): 122-30, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26228837

RESUMO

The dog CYP1A2 enzyme is likely an important contributor to the metabolism of veterinary drugs. Dog CYP1A2 is expressed in liver, plus it is inducible and polymorphic, creating the potential for intersubject differences in pharmacokinetics. Hence, the ability to probe dog CYP1A2 activity and inhibition is relevant toward veterinary drug development and drug-drug interaction assessment. Previous studies have relied on human probes with questionable specificity for CYP1A2, so it was hypothesized that recombinant CYP1A2 could be used to find a specific CYP1A2 substrate. Intrinsic clearance experiments demonstrated that tizanidine was a substrate of CYP1A2. Profiling of tizanidine metabolites generated by CYP1A2 identified the imidazole metabolite that was detectable in dog plasma. The imidazole metabolite was subsequently used to evaluate tizanidine as a CYP1A2 probe. Co-administration of the CYP1A inhibitor enrofloxacin with tizanidine significantly decreased (30%; n = 3) the formation of the imidazole metabolite vs. control experiments. As enrofloxacin is a weak inhibitor, further studies are required to confirm the sensitivity of tizanidine as an in vivo probe. However, tizanidine may be a more selective CYP1A2 probe than phenacetin when conducting in vitro studies due to the presence of other phenacetin-metabolizing enzymes in dog liver microsomes.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Clonidina/análogos & derivados , Inibidores do Citocromo P-450 CYP1A2/farmacocinética , Citocromo P-450 CYP1A2/metabolismo , Cães/metabolismo , Animais , Benzoflavonas/farmacocinética , Clonidina/farmacocinética , Citocromo P-450 CYP1A2/genética , Cães/sangue , Enrofloxacina , Feminino , Fluoroquinolonas/farmacocinética , Microssomos Hepáticos/metabolismo , Sondas Moleculares , Fenacetina , Especificidade por Substrato , Teofilina/análogos & derivados , Teofilina/farmacocinética
15.
Vet Anaesth Analg ; 43(6): 599-610, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26918855

RESUMO

OBJECTIVES: To investigate the effects of MK-467 on sedation quality, and cardiopulmonary and pharmacokinetic variables in horses sedated intravenously (IV) with romifidine. STUDY DESIGN: Experimental, randomized, crossover design. ANIMALS: Seven healthy mares. METHODS: Romifidine (80 µg kg-1 ; R) and MK-467 (200 µg kg-1 ; MK) were administered IV alone and in combination (R + MK). Levels of sedation and borborygmi were scored. Heart rate (HR), direct arterial blood pressure (ABP) and respiratory rate (fR ) were recorded. Arterial and venous blood gas analyses were performed and venous plasma drug concentrations were measured. Pharmacokinetic parameters were calculated. Linear mixed modelling for repeated measures, contrasts of least square means by Bonferroni correction tests, one-way anova for repeated measures with Bonferroni multiple comparison tests and paired Student's t-tests were used to compare results within and between treatments as appropriate. Significance was set at p < 0.05. RESULTS: After R, ABP increased and HR and fR decreased significantly. After R + MK, HR, fR , systolic and mean ABP decreased. MK alone increased both HR and fR . After R, ABP was significantly higher than after R + MK. HR and fR were significantly higher after MK than after R and R + MK. Areas under the curve for sedation time were similar after R and R + MK. Intestinal activity decreased markedly after R and less after R + MK. Volume of distribution and clearance of romifidine were significantly higher and area under the concentration time curve extrapolated to infinity significantly lower after R + MK than after R. CONCLUSIONS: Combined romifidine and MK-467 prevented the cardiovascular changes commonly seen with romifidine but did not affect sedation quality. CLINICAL RELEVANCE: Combined IV romifidine and MK-467 can be used to attenuate the cardiovascular effects of romifidine, such as in horses with colic or undergoing general anaesthesia.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Hipnóticos e Sedativos/farmacocinética , Imidazóis/farmacocinética , Quinolizinas/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Anestesia Intravenosa/veterinária , Anestésicos Combinados , Animais , Gasometria/veterinária , Sistema Cardiovascular/efeitos dos fármacos , Estudos Cross-Over , Sedação Profunda/veterinária , Feminino , Cavalos , Hipnóticos e Sedativos/farmacologia , Imidazóis/farmacologia , Quinolizinas/farmacologia , Respiração/efeitos dos fármacos
16.
Exp Eye Res ; 134: 141-7, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25701803

RESUMO

The effect of the dinucleotide P(1), P(4)-Di (adenosine-5') tetraphosphate (Ap4A) in improving adrenergic anti-glaucomatous delivery by modifying the tight junction proteins of the corneal epithelium was evaluated. Stratified human corneal epithelial cells (HCLE) were treated with Ap4A (100 µM) for 5 min and TJ protein levels and barrier function were analysed by western blotting and transepithelial electrical resistance (TEER), respectively. Western blot experiments showed a significant reduction at 2 h (45% reduction of ZO-1 and 65% reduction of occludin protein levels) as compared to non-treated (control) cells. Two hours after Ap4A treatment, TEER values were significantly reduced (65% as compared to control levels (p < 0.001)), indicating an increase in corneal barrier permeability. Topical application of Ap4A in New Zealand white rabbits two hours before the instillation of the hypotensor compounds (the α2-adrenergic receptor agonist, brimonidine and the ß-adrenergic receptor antagonist, timolol), improved the delivery of these compounds to the anterior chamber as well as their hypotensive action on the intraocular pressure. The results obtained showed that, when Ap4A was topically applied two hours before the adrenergic compounds, the concentration of brimonidine in the aqueous humour increased from 64.3 ± 5.3 nM to 240.6 ± 8.6 nM and from 58.9 ± 9.2 nM to 183.7 ± 6.8 nM in the case of timolol, which also produces a more profound effect on IOP. Therefore, Ap4A treatment results in a better entrance of adrenergic anti-glaucomatous compounds within the eye and consequently improved therapeutic efficiency by increasing corneal epithelial barrier permeability.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Antagonistas Adrenérgicos beta/farmacocinética , Anti-Hipertensivos/farmacocinética , Humor Aquoso/metabolismo , Fosfatos de Dinucleosídeos/farmacologia , Epitélio Corneano/efeitos dos fármacos , Junções Íntimas/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Antagonistas Adrenérgicos beta/administração & dosagem , Animais , Anti-Hipertensivos/administração & dosagem , Western Blotting , Tartarato de Brimonidina , Permeabilidade da Membrana Celular/efeitos dos fármacos , Células Cultivadas , Sistemas de Liberação de Medicamentos , Impedância Elétrica , Epitélio Corneano/metabolismo , Glaucoma/tratamento farmacológico , Humanos , Pressão Intraocular/efeitos dos fármacos , Masculino , Ocludina/metabolismo , Quinoxalinas/administração & dosagem , Coelhos , Junções Íntimas/metabolismo , Timolol/administração & dosagem , Proteína da Zônula de Oclusão-1/metabolismo
17.
Pediatr Res ; 77(6): 772-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25751572

RESUMO

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.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Encéfalo/efeitos dos fármacos , Dexmedetomidina/farmacocinética , Hipotermia/fisiopatologia , Agonistas de Receptores Adrenérgicos alfa 2/sangue , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Animais Recém-Nascidos , Temperatura Corporal/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/fisiologia , Cromatografia Líquida , Dexmedetomidina/sangue , Dexmedetomidina/farmacologia , Avaliação Pré-Clínica de Medicamentos , Espectrometria de Massas , Ratos , Ratos Endogâmicos Lew
18.
Int J Clin Pharmacol Ther ; 53(4): 301-16, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25109412

RESUMO

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.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/efeitos adversos , Transtorno do Deficit de Atenção com Hiperatividade/tratamento farmacológico , Guanfacina/efeitos adversos , Sistema de Condução Cardíaco/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Potenciais de Ação , Agonistas de Receptores Adrenérgicos alfa 2/sangue , Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Adulto , Estudos Cross-Over , Método Duplo-Cego , Eletrocardiografia , Feminino , Guanfacina/sangue , Guanfacina/farmacocinética , Voluntários Saudáveis , Sistema de Condução Cardíaco/fisiologia , Humanos , Masculino , Segurança do Paciente , Medição de Risco , Fatores de Risco , Adulto Jovem
19.
J Pharmacokinet Pharmacodyn ; 42(1): 45-65, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25373474

RESUMO

Guanfacine extended-release (GXR) is a selective α2A-adrenergic receptor agonist approved in the United States for once-daily administration for the treatment of attention-deficit hyperactivity disorder (ADHD) in children and adolescents ages 6-17 years old either as monotherapy or adjunctive to stimulant medications. This analysis integrates exposure-response, placebo, and dropout data from 10 clinical trials that used GXR in adolescents and children with ADHD. In these trials, the ADHD Rating Scale-IV (ADHD RS-IV) score was collected longitudinally within patients over the course of 6-13 weeks. Non-linear mixed effects models were developed and used to describe the exposure-response of the GXR and placebo time course. The OpenBUGS program was utilized to describe the dropout time course across the trials. Placebo time course was best described by an inverse Bateman function with a 3-group mixture model that allowed for the onset and offset of the placebo response. Dropout time modeling indicated a missing at random mechanism for dropouts which was best described by a Weibull distribution with an estimated percentage of non-dropout patients. A linear exposure-response model with an adolescent effect on maximum slope (SLPmax), and a time delay for reaching SLPmax, provided the best description of the GXR exposure-response time course. The GXR exposure-response model indicated that the typical (95 % confidence interval) decrease in ADHD RS-IV score from the placebo-response trajectory would be 37.1 % (32.2, 42.0 %) per 0.1 mg/kg of GXR exposure. There was little noticeable difference between the exposure-response in adolescents and children or across ADHD subtypes.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Transtorno do Deficit de Atenção com Hiperatividade/tratamento farmacológico , Simulação por Computador , Guanfacina/administração & dosagem , Modelos Biológicos , Adolescente , Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Transtorno do Deficit de Atenção com Hiperatividade/epidemiologia , Criança , Preparações de Ação Retardada , Guanfacina/farmacologia , Guanfacina/uso terapêutico , Humanos , Adesão à Medicação , Pacientes Desistentes do Tratamento , Ensaios Clínicos Controlados Aleatórios como Assunto/estatística & dados numéricos , Resultado do Tratamento
20.
J Vet Pharmacol Ther ; 38(1): 15-23, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25066475

RESUMO

The aim of the study was to describe the pharmacokinetics and selected pharmacodynamics of intravenous dexmedetomidine in horses. Eight adult horses received 5 µg/kg dexmedetomidine IV. Blood samples were collected before and for 10 h after drug administration to determine dexmedetomidine plasma concentrations. Pharmacokinetic parameters were calculated using noncompartmental analysis. Data from one outlier were excluded from the statistical summary. Behavioral and physiological responses were recorded before and for 6 h after dexmedetomidine administration. Dexmedetomidine concentrations decreased rapidly (elimination half-life of 8.03 ± 0.84 min). Time of last detection varied from 30 to 60 min. Bradycardia was noted at 4 and 10 min after drug administration (26 ± 8 and 29 ± 8 beats/min respectively). Head height decreased by 70% at 4 and 10 min and gradually returned to baseline. Ability to ambulate was decreased for 60 min following drug administration, and mechanical nociceptive threshold was increased during 30 min. Blood glucose peaked at 30 min (134 ± 24 mg/dL) and borborygmi were decreased for the first hour after dexmedetomidine administration. Dexmedetomidine was quickly eliminated as indicated by the rapid decrease in plasma concentrations. Physiological, behavioral, and analgesic effects observed after dexmedetomidine administration were of short duration.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Dexmedetomidina/farmacocinética , Cavalos/sangue , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Animais , Área Sob a Curva , Dexmedetomidina/administração & dosagem , Feminino , Injeções Intravenosas , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA