Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 189
Filtrar
Más filtros

Intervalo de año de publicación
1.
J Vet Pharmacol Ther ; 43(6): 533-537, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32557697

RESUMEN

Different structurally related phenylpiperidine opioids exhibit different isoflurane-sparing effects in cats. Because minimum alveolar concentration (MAC) in cats is affected only by very high plasma concentrations of some phenylpiperidine opioids, we hypothesized these effects are caused by actions on nonopioid receptors. Using a prospective, randomized, crossover design, six cats were anesthetized with isoflurane, intubated, ventilated, and instrumented. Isoflurane MAC was measured in triplicate using a tail-clamp and bracketing technique. A computer-controlled intravenous infusion using prior pharmacokinetic models targeted plasma concentrations of 60 ng/ml fentanyl, 10 ng/ml sufentanil, or 500 ng/ml alfentanil, and isoflurane MAC was measured in duplicate. Next, naltrexone 0.6 mg/kg was administered to cats hourly during the opioid infusion, and isoflurane MAC was measured in duplicate. Blood was collected during MAC determinations to measure opioid concentrations. Responses were analyzed using repeated measures ANOVA with significance at p < .05. Alfentanil and sufentanil decreased isoflurane MAC by 16.4% and 6.4%, respectively, and these effects were completely reversed by naltrexone. Fentanyl had no significant effect on isoflurane MAC. Alfentanil and sufentanil modestly reduce isoflurane MAC via agonist effects on opioid receptors. However, these effects are too small to justify clinical use of phenylpiperidine opioids as single agents to reduce MAC in cats.


Asunto(s)
Alfentanilo/farmacocinética , Fentanilo/farmacocinética , Isoflurano/farmacocinética , Sufentanilo/farmacocinética , Alfentanilo/administración & dosificación , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacocinética , Anestesia por Inhalación/veterinaria , Anestésicos por Inhalación/administración & dosificación , Anestésicos por Inhalación/farmacocinética , Animales , Presión Sanguínea/efectos de los fármacos , Gatos , Estudios Cruzados , Interacciones Farmacológicas , Fentanilo/administración & dosificación , Frecuencia Cardíaca/efectos de los fármacos , Infusiones Intravenosas/veterinaria , Isoflurano/administración & dosificación , Isoflurano/farmacología , Sufentanilo/administración & dosificación
2.
J Formos Med Assoc ; 118(1 Pt 2): 291-298, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29803320

RESUMEN

BACKGROUND: Sedation for esophagogastroduodenoscopy (EGD) and colonoscopy is characterized by rapid patient induction and emergence. The drugs midazolam and alfentanil have long been used for procedural sedation; however, the relationship between plasma or effect-site concentrations (Cp or Ce, respectively) and emergence remains unclear. The aim of this study is to develop patient wake-up prediction models for both Cp and Ce using response surface modeling, a pharmacodynamics tool for assessing patients' responses. METHODS: The Observer's Alertness/Sedation (OAA/S) score was used to monitor sedation depth during the examinations. Concentration pairs of midazolam and alfentanil were calculated for each of Cp and Ce using pharmacokinetic simulation software. Response surface models were developed using the Greco construct. Temporal analysis was done by comparing model-predicted wake-up time with true patient wake-up time. RESULTS: Thirty-three patients with an average body mass index of 21.85 ± 2.3 kg/m2 were pooled for analysis. The average duration of examination were 2.9 ± 1.4 min for EGD and 6.6 ± 2.7 min for colonoscopy. Seventy-five concentration pairs of midazolam and alfentanil were obtained for each Cp and Ce. The Cp-based Greco response surface model showed significant synergy between midazolam and alfentanil and was a better predictor of patient wake-up time, with an average deviation of 1.0 ± 3.9 min, while the Ce model show time deviation greater than 20 min. CONCLUSION: The early phases of drug distribution are unique and complicated by nonsteady-state concentrations, and our study revealed that Ce-based wake-up time prediction is more difficult under these circumstances.


Asunto(s)
Alfentanilo/administración & dosificación , Sedación Consciente/métodos , Endoscopía Gastrointestinal/métodos , Midazolam/administración & dosificación , Adulto , Alfentanilo/farmacocinética , Anestésicos Intravenosos/administración & dosificación , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Midazolam/farmacocinética , Persona de Mediana Edad , Modelos Biológicos , Pronóstico
3.
Br J Anaesth ; 120(6): 1209-1218, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29793588

RESUMEN

BACKGROUND: The non-linear mixed amount with zero amounts response surface model can be used to describe drug interactions and predict loss of response to noxious stimuli and respiratory depression. We aimed to determine whether this response surface model could be used to model sedation with the triple drug combination of midazolam, alfentanil and propofol. METHODS: Sedation was monitored in 56 patients undergoing gastrointestinal endoscopy (modelling group) using modified alertness/sedation scores. A total of 227 combinations of effect-site concentrations were derived from pharmacokinetic models. Accuracy and the area under the receiver operating characteristic curve were calculated. Accuracy was defined as an absolute difference <0.5 between the binary patient responses and the predicted probability of loss of responsiveness. Validation was performed with a separate group (validation group) of 47 patients. RESULTS: Effect-site concentration ranged from 0 to 108 ng ml-1 for midazolam, 0-156 ng ml-1 for alfentanil, and 0-2.6 µg ml-1 for propofol in both groups. Synergy was strongest with midazolam and alfentanil (24.3% decrease in U50, concentration for half maximal drug effect). Adding propofol, a third drug, offered little additional synergy (25.8% decrease in U50). Two patients (3%) experienced respiratory depression. Model accuracy was 83% and 76%, area under the curve was 0.87 and 0.80 for the modelling and validation group, respectively. CONCLUSION: The non-linear mixed amount with zero amounts triple interaction response surface model predicts patient sedation responses during endoscopy with combinations of midazolam, alfentanil, or propofol that fall within clinical use. Our model also suggests a safety margin of alfentanil fraction <0.12 that avoids respiratory depression after loss of responsiveness.


Asunto(s)
Sedación Consciente/métodos , Hipnóticos y Sedantes/administración & dosificación , Modelos Biológicos , Adulto , Anciano , Alfentanilo/administración & dosificación , Alfentanilo/efectos adversos , Alfentanilo/farmacocinética , Esquema de Medicación , Combinación de Medicamentos , Sinergismo Farmacológico , Endoscopía Gastrointestinal/métodos , Femenino , Humanos , Hipnóticos y Sedantes/efectos adversos , Hipnóticos y Sedantes/farmacocinética , Masculino , Midazolam/administración & dosificación , Midazolam/efectos adversos , Midazolam/farmacocinética , Persona de Mediana Edad , Propofol/administración & dosificación , Propofol/efectos adversos , Propofol/farmacocinética , Insuficiencia Respiratoria/inducido químicamente
4.
Anesth Analg ; 123(2): 299-308, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27192475

RESUMEN

BACKGROUND: Selecting an effective dose of sedative drugs in combined upper and lower gastrointestinal endoscopy is complicated by varying degrees of pain stimulation. We tested the ability of 5 response surface models to predict depth of sedation after administration of midazolam and alfentanil in this complex model. The procedure was divided into 3 phases: esophagogastroduodenoscopy (EGD), colonoscopy, and the time interval between the 2 (intersession). METHODS: The depth of sedation in 33 adult patients was monitored by Observer Assessment of Alertness/Scores. A total of 218 combinations of midazolam and alfentanil effect-site concentrations derived from pharmacokinetic models were used to test 5 response surface models in each of the 3 phases of endoscopy. Model fit was evaluated with objective function value, corrected Akaike Information Criterion (AICc), and Spearman ranked correlation. A model was arbitrarily defined as accurate if the predicted probability is <0.5 from the observed response. RESULTS: The effect-site concentrations tested ranged from 1 to 76 ng/mL and from 5 to 80 ng/mL for midazolam and alfentanil, respectively. Midazolam and alfentanil had synergistic effects in colonoscopy and EGD, but additivity was observed in the intersession group. Adequate prediction rates were 84% to 85% in the intersession group, 84% to 88% during colonoscopy, and 82% to 87% during EGD. The reduced Greco and Fixed alfentanil concentration required for 50% of the patients to achieve targeted response Hierarchy models performed better with comparable predictive strength. The reduced Greco model had the lowest AICc with strong correlation in all 3 phases of endoscopy. Dynamic, rather than fixed, γ and γalf in the Hierarchy model improved model fit. CONCLUSIONS: The reduced Greco model had the lowest objective function value and AICc and thus the best fit. This model was reliable with acceptable predictive ability based on adequate clinical correlation. We suggest that this model has practical clinical value for patients undergoing procedures with varying degrees of stimulation.


Asunto(s)
Alfentanilo/administración & dosificación , Estado de Conciencia/efectos de los fármacos , Endoscopía Gastrointestinal/efectos adversos , Hipnóticos y Sedantes/administración & dosificación , Midazolam/administración & dosificación , Modelos Biológicos , Umbral del Dolor/efectos de los fármacos , Dolor/prevención & control , Adulto , Alfentanilo/farmacocinética , Colonoscopía/efectos adversos , Técnicas de Apoyo para la Decisión , Cálculo de Dosificación de Drogas , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Humanos , Hipnóticos y Sedantes/farmacocinética , Masculino , Midazolam/farmacocinética , Persona de Mediana Edad , Dolor/etiología , Dolor/fisiopatología , Reproducibilidad de los Resultados
5.
J Vet Pharmacol Ther ; 37(1): 13-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23895731

RESUMEN

The aim of this study was to compare the pharmacokinetics of fentanyl, alfentanil, and sufentanil in isoflurane-anesthetized cats. Six adult cats were used. Anesthesia was induced and maintained with isoflurane in oxygen. End-tidal isoflurane concentration was set at 2% and adjusted as required due to spontaneous movement. Fentanyl (10 µg/kg), alfentanil (100 µg/kg), or sufentanil (1 µg/kg) was administered intravenously as a bolus, on separate days. Blood samples were collected immediately before and for 8 h following drug administration. Plasma drug concentration was determined using liquid chromatography/mass spectrometry. Compartment models were fitted to concentration-time data. A 3-compartment model best fitted the concentration-time data for all drugs, except for 1 cat in the sufentanil group (excluded from analysis). The volume of the central compartment and the volume of distribution at steady-state (L/kg) [mean ± SEM (range)], the clearance (mL/min/kg) [harmonic mean ± pseudo-SD (range)], and the terminal half-life (min) [median (range)] were 0.25 ± 0.04 (0.09-0.34), 2.18 ± 0.16 (1.79-2.83), 18.6 ± 5.0 (15-29.8), and 151 (115-211) for fentanyl; 0.10 ± 0.01 (0.07-0.14), 0.89 ± 0.16 (0.68-1.83), 11.6 ± 2.6 (9.2-15.8), and 144 (118-501) for alfentanil; and 0.06 ± 0.01 (0.04-0.10), 0.77 ± 0.07 (0.63-0.99), 17.6 ± 4.3 (13.9-24.3), and 54 (46-76) for sufentanil. Differences in clearance and volume of distribution result in similar terminal half-lives for fentanyl and alfentanil, longer than for sufentanil.


Asunto(s)
Alfentanilo/farmacocinética , Anestésicos Intravenosos/farmacocinética , Gatos/sangre , Fentanilo/farmacocinética , Sufentanilo/farmacocinética , Alfentanilo/administración & dosificación , Alfentanilo/sangre , Anestesia por Inhalación , Anestésicos por Inhalación , Anestésicos Intravenosos/administración & dosificación , Animales , Área Bajo la Curva , Gatos/metabolismo , Interacciones Farmacológicas , Fentanilo/administración & dosificación , Fentanilo/sangre , Semivida , Isoflurano , Sufentanilo/administración & dosificación , Sufentanilo/sangre
6.
Br J Anaesth ; 111(2): 197-208, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23512864

RESUMEN

BACKGROUND: In open TCI and anaesthesia display systems, the choice of pharmacokinetic (PK) parameter sets of opioids is clinically relevant. Accuracy and bias of the PK models may be affected by administration mode and the co-administered hypnotic drug. We retrospectively evaluated the performance of eight PK parameter sets for alfentanil in two data sets (infusion and bolus application). METHODS: With the dosing history from two studies in orthopaedic patients anaesthetized with propofol or inhalation anaesthetics the alfentanil plasma concentration over time was calculated with eight PK parameter sets. Median absolute performance error (MDAPE), log accuracy, median performance error (MDPE), log bias, Wobble, and Divergence were computed. Mann-Whitney rank test with Bonferroni correction was used for comparison between bolus and infusion data, repeated measures analysis of variance on ranks was used for comparison among parameter sets. RESULTS: The parameters by Scott (original and weight adjusted) and Fragen had a MDAPE ≤30% and a median log accuracy <0.15 independent of the administration mode, while MDPE was within ±20% and log bias nearly within ±0.1, respectively. The sets by Maitre and Lemmens were within these limits only in the bolus data. All other parameter sets were outside these limits. CONCLUSIONS: In healthy orthopaedic patients, the PK parameters by Scott and by Maitre were equally valid when alfentanil was given as repeated boluses. When given as infusion, the Maitre parameters were less accurate and subject to a significant bias. We cannot exclude that the difference between bolus and infusion is partially because of the different hypnotics used.


Asunto(s)
Alfentanilo/farmacocinética , Anestesia Intravenosa/métodos , Anestésicos Intravenosos/farmacocinética , Terminales de Computador , Adulto , Anciano , Anciano de 80 o más Años , Alfentanilo/administración & dosificación , Alfentanilo/sangre , Anestésicos Intravenosos/administración & dosificación , Anestésicos Intravenosos/sangre , Femenino , Humanos , Bombas de Infusión , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados , Estudios Retrospectivos , Adulto Joven
7.
Br J Anaesth ; 109(4): 551-60, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22732112

RESUMEN

BACKGROUND: Available alfentanil pharmacokinetic (PK) sets for target-controlled infusion (TCI) were derived from populations with normal BMI. The performance and accuracy of the models devised by Maitre and colleagues and Scott and colleagues were evaluated in a population including morbidly obese patients. METHODS: Alfentanil TCI using Maitre and colleagues' model was administered to 10 obese and six non-obese women (BMI 19.5-57.4 kg m(-2)) undergoing laparoscopic surgery. The initial effect-site target concentration was 100 ng ml(-1). Alfentanil arterial plasma concentrations were sampled from TCI onset to 220 min after its termination. Stanpump(®) software calculated predicted alfentanil concentrations. Data were analysed with a non-linear mixed-effect model (NONMEM, version 7.2), including calculations of the median performance error (MDPE) and the median absolute performance error (MDAPE). Scott and colleagues' model was evaluated retrospectively. RESULTS: Using Maitre and colleagues' model, MDPE and MDAPE (range) for the whole population were 13.3% and 23.9%, respectively. With Scott and colleagues' model, MDPE and MDAPE were -30.7% and 50.1%, respectively. We created a three-compartment model with BMI as the covariate (CL), yielding MDPE 1.1% and MDAPE 30.6%. CONCLUSIONS: Maitre and colleagues' PK set underestimated the predicted concentrations in our mixed-weighted population, but its bias and accuracy were acceptable for clinical application. Scott and colleagues' model was inaccurate. The NONMEM model seemed to be more accurate during the infusion and for high concentrations, but it needs to be validated in a larger population.


Asunto(s)
Alfentanilo/farmacocinética , Anestesia Intravenosa/métodos , Anestésicos Intravenosos/farmacocinética , Obesidad Mórbida/metabolismo , Adolescente , Adulto , Anciano , Alfentanilo/administración & dosificación , Algoritmos , Anestésicos Intravenosos/administración & dosificación , Cirugía Bariátrica , Índice de Masa Corporal , Femenino , Humanos , Infusiones Intravenosas , Laparoscopía , Persona de Mediana Edad , Modelos Estadísticos , Dinámicas no Lineales , Población , Estudios Prospectivos , Reproducibilidad de los Resultados
8.
Acta Anaesthesiol Scand ; 54(2): 132-40, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19817719

RESUMEN

In animal models of vomiting, mu-opioid (MOP, OP(3)) receptors mediate both emesis and anti-emesis. mu-receptors within the blood-brain barrier, mediating anti-emesis, are more rapidly accessible to lipid-soluble mu-opioid receptor agonists such as fentanyl than to morphine, and fentanyl has broad-spectrum anti-emetic effects in a number of species. Whether a similar situation exists in humans is not known. A search was performed for clinical studies comparing the emetic side effects of opioids administered peri-operatively in an attempt to identify differences between morphine and more lipid-soluble mu-receptor-selective agonists such as fentanyl. Overall, the evidence appears to suggest that fentanyl and other phenylpiperidines are associated with less nausea and vomiting than morphine, but not all studies support this, and fentanyl-like drugs are associated with nausea and vomiting per se. Good evidence, however, exists to show that fentanyl and alfentanil do not cause more nausea and vomiting than the ultra fast-acting remifentanil. Because remifentanil is cleared rapidly post-operatively, such trials suggest that the emetic side effects of fentanyl and alfentanil are minimal. The clinical evidence, although limited, is at least consistent with the possibility that central mu-opioid receptors may mediate anti-emesis in humans. It is possible that the role of mu-opioid agonists in anti-emesis may become clearer in the future as a result of the use of peripheral mu-opioid receptor antagonists.


Asunto(s)
Antieméticos/uso terapéutico , Receptores Opioides mu/agonistas , Alfentanilo/efectos adversos , Alfentanilo/farmacocinética , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/farmacocinética , Animales , Barrera Hematoencefálica/metabolismo , Fentanilo/efectos adversos , Fentanilo/farmacocinética , Humanos , Modelos Animales , Morfina/efectos adversos , Morfina/farmacocinética , Piperidinas/efectos adversos , Piperidinas/farmacocinética , Náusea y Vómito Posoperatorios/inducido químicamente , Náusea y Vómito Posoperatorios/prevención & control , Receptores Opioides mu/metabolismo , Receptores Opioides mu/fisiología , Remifentanilo , Vómitos/etiología
9.
Twin Res Hum Genet ; 13(5): 412-25, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20874462

RESUMEN

Opioids are the cornerstone medication for the treatment of moderate to severe pain. However, analgesic opioid requirements and the propensity to suffer from aversive opioid effects, including fatal respiratory depression and addiction, vary widely among patients. The factors underlying the substantial response variance remain largely unknown and need clarification for using opioids more effectively in appropriately selected patients. This ongoing study takes advantage of the twin paradigm to estimate the genetic and environmental contributions to inter-individual differences in opioid responses. Evidence of significant heritability will justify more detailed and extensive genomic studies. The enrollment target is 80 monozygotic and 45 dizygotic twin pairs who undergo a target-controlled infusion of the opioid alfentanil and saline placebo in sequential but randomized order. In a laboratory-type setting, well-defined pharmacodynamic endpoints are measured to quantify pain sensitivity, analgesic opioid effects, and aversive opioid effects including respiratory depression, sedation and reinforcing affective responses. First results obtained in 159 participants provide evidence for the feasibility and utility of this interventional study paradigm to estimate familial aggregation and heritability components of relevant drug effects. Areas highlighted in this report include recruitment strategies, required infrastructure and personnel, selection of relevant outcome measures, drug infusion algorithm minimizing pharmacokinetic variability, and considerations for optimizing data quality and quantity without hampering feasibility. Applying the twin paradigm to complex and potentially harmful studies comprehensively characterizing pharmacological response profiles is without much precedent. Methods and first results including heritability estimates for heat and cold pain sensitivity should be of interest to investigators considering similar studies.


Asunto(s)
Analgésicos Opioides/farmacocinética , Farmacogenética/métodos , Gemelos Dicigóticos/genética , Gemelos Dicigóticos/metabolismo , Gemelos Monocigóticos/genética , Gemelos Monocigóticos/metabolismo , Adulto , Alfentanilo/efectos adversos , Alfentanilo/farmacocinética , Alfentanilo/farmacología , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/farmacología , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Dolor/tratamiento farmacológico , Dolor/genética , Dolor/metabolismo , Dimensión del Dolor , Percepción del Dolor , Adulto Joven
10.
Clin Pharmacokinet ; 59(4): 501-518, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31587145

RESUMEN

BACKGROUND: Preterm neonates are usually not part of a traditional drug development programme, however they are frequently administered medicines. Developing modelling and simulation tools, such as physiologically based pharmacokinetic (PBPK) models that incorporate developmental physiology and maturation of drug metabolism, can be used to predict drug exposure in this group of patients, and may help to optimize drug dose adjustment. OBJECTIVE: The aim of this study was to assess and verify the predictability of a preterm PBPK model using compounds that undergo diverse renal and/or hepatic clearance based on the knowledge of their disposition in adults. METHODS: A PBPK model was developed in the Simcyp Simulator V17 to predict the pharmacokinetics (PK) of drugs in preterm neonates. Drug parameters for alfentanil, midazolam, caffeine, ibuprofen, gentamicin and vancomycin were collated from the literature. Predicted PK parameters and profiles were compared against the observed data. RESULTS: The preterm PBPK model predicted the PK changes of the six compounds using ontogeny functions for cytochrome P450 (CYP) 1A2, CYP2C9 and CYP3A4 after oral and intravenous administrations. For gentamicin and vancomycin, the maturation of renal function was able to predict the exposure of these two compounds after intravenous administration. All PK parameter predictions were within a twofold error criteria. CONCLUSION: While the developed preterm model for the prediction of PK behaviour in preterm patients is not intended to replace clinical studies, it can potentially help with deciding on first-time dosing in this population and study design in the absence of clinical data.


Asunto(s)
Alfentanilo/farmacocinética , Gentamicinas/farmacocinética , Recién Nacido/metabolismo , Midazolam/farmacocinética , Vancomicina/farmacocinética , Administración Intravenosa , Administración Oral , Alfentanilo/administración & dosificación , Antibacterianos/administración & dosificación , Antibacterianos/farmacocinética , Cafeína/administración & dosificación , Cafeína/farmacocinética , Simulación por Computador , Inhibidores de la Ciclooxigenasa/administración & dosificación , Inhibidores de la Ciclooxigenasa/farmacocinética , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP3A/metabolismo , Femenino , Gentamicinas/administración & dosificación , Edad Gestacional , Humanos , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/farmacocinética , Ibuprofeno/administración & dosificación , Ibuprofeno/farmacocinética , Recién Nacido/fisiología , Riñón/metabolismo , Hígado/metabolismo , Masculino , Tasa de Depuración Metabólica/fisiología , Midazolam/administración & dosificación , Modelos Biológicos , Narcóticos/administración & dosificación , Narcóticos/farmacocinética , Valor Predictivo de las Pruebas , Nacimiento Prematuro , Vancomicina/administración & dosificación
11.
Clin Pharmacol Ther ; 84(4): 506-12, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19238656

RESUMEN

Ritonavir diminishes methadone plasma concentrations, an effect attributed to CYP3A induction, but the actual mechanisms are unknown. We determined short-term (2-day) and steady-state (2-week) ritonavir effects on intestinal and hepatic CYP3A4/5 (probed with intravenous (IV) and oral alfentanil (ALF) and with miosis) and P-glycoprotein (P-gp) (fexofenadine), and on methadone pharmacokinetics and pharmacodynamics in healthy volunteers. Acute ritonavir increased the area under the concentration-time curve (AUC)(0-infinity)/dose ratio (ritonavir/control) for oral ALF 25-fold. Steady-state ritonavir increased the AUC(0-Infinity)/dose ratio for IV and oral ALF 4- and 10-fold, respectively; reduced hepatic extraction (from 0.26 to 0.07) and intestinal extraction (from 0.51 to 0); and increased bioavailability (from 37 to 95%). Acute ritonavir inhibits first-pass CYP3A > 96%. Chronic ritonavir inhibits hepatic CYP3A (> 70%) and first-pass CYP3A (> 90%). Acute and steady-state ritonavir increased the fexofenadine AUC(0-infinity) 2.8- and 1.4-fold, respectively, suggesting P-gp inhibition. Steady-state compared with acute ritonavir caused mild apparent induction of P-gp and hepatic CYP3A, but net inhibition still predominated. Ritonavir inhibited both intestinal and hepatic CYP3A and drug transport. ALF miosis noninvasively determined CYP3A inhibition by ritonavir.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Citocromo P-450 CYP3A/fisiología , Inhibidores de la Proteasa del VIH/farmacología , Metadona/farmacocinética , Narcóticos/farmacocinética , Ritonavir/farmacología , Adulto , Alfentanilo/administración & dosificación , Alfentanilo/sangre , Alfentanilo/farmacocinética , Área Bajo la Curva , Disponibilidad Biológica , Estudios Cruzados , Inhibidores del Citocromo P-450 CYP3A , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Femenino , Inhibidores de la Proteasa del VIH/farmacocinética , Humanos , Intestinos/enzimología , Hígado/enzimología , Masculino , Metadona/farmacología , Narcóticos/farmacología , Pupila/efectos de los fármacos , Ritonavir/farmacocinética , Estereoisomerismo , Terfenadina/administración & dosificación , Terfenadina/análogos & derivados , Terfenadina/sangre , Terfenadina/farmacocinética
12.
Clin Pharmacokinet ; 47(11): 743-52, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18840029

RESUMEN

BACKGROUND: Liver cirrhosis is a progressive disease characterized by loss of functional hepatocytes with concomitant connective tissue and nodule formation in the liver. The morphological and physiological changes associated with the disease substantially affect drug pharmacokinetics. Whole-body physiologically based pharmacokinetic (WB-PBPK) modelling is a predictive technique that quantitatively relates the pharmacokinetic parameters of a drug to such (patho-)physiological conditions. OBJECTIVE: To extend an existing WB-PBPK model, based on the physiological changes associated with liver cirrhosis, which allows for prediction of drug pharmacokinetics in patients with liver cirrhosis. METHODS: The literature was searched for quantitative measures of the physiological changes associated with the presence of Child-Pugh class A through C liver cirrhosis. The parameters that were included were the organ blood flows, cardiac index, plasma binding protein concentrations, haematocrit, functional liver volume, hepatic enzymatic activity and glomerular filtration rate. Predictions of pharmacokinetic profiles and parameters were compared with literature data for the model compounds alfentanil, lidocaine (lignocaine), theophylline and levetiracetam. RESULTS: The predicted versus observed plasma concentration-time profiles for alfentanil and lidocaine were similar, such that the pharmacokinetic changes associated with Child-Pugh class A, B and C liver cirrhosis were adequately described. The theophylline elimination half-life was greatly increased in Child-Pugh class B and C patients compared with controls, as predicted by the model. Levetiracetam urinary excretion was consistently reduced with disease progression and very closely resembled observed values. CONCLUSION: Consideration of the physiological differences between healthy individuals and patients with liver cirrhosis was important for the simulation of drug pharmacokinetics in this compromised group. The WB-PBPK model was altered to incorporate these physiological differences with the result of adequate simulation of drug pharmacokinetics. The information provided in this study will allow other researchers to further validate this liver cirrhosis model within a WB-PBPK model.


Asunto(s)
Alfentanilo/farmacocinética , Lidocaína/farmacocinética , Cirrosis Hepática/metabolismo , Piracetam/análogos & derivados , Teofilina/farmacocinética , Humanos , Levetiracetam , Modelos Biológicos , Piracetam/farmacocinética
13.
Clin Pharmacokinet ; 57(2): 125-149, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28688027

RESUMEN

Fentanyl and its derivatives sufentanil, alfentanil, and remifentanil are potent opioids. A comprehensive review of the use of fentanyl and its derivatives in the pediatric population was performed using the National Library of Medicine PubMed. Studies were included if they contained original pharmacokinetic parameters or models using established routes of administration in patients younger than 18 years of age. Of 372 retrieved articles, 44 eligible pharmacokinetic studies contained data of 821 patients younger than 18 years of age, including more than 46 preterm infants, 64 full-term neonates, 115 infants/toddlers, 188 children, and 28 adolescents. Underlying diagnoses included congenital heart and pulmonary disease and abdominal disorders. Routes of drug administration were intravenous, epidural, oral-transmucosal, intranasal, and transdermal. Despite extensive use in daily clinical practice, few studies have been performed. Preterm and term infants have lower clearance and protein binding. Pharmacokinetics was not altered by chronic renal or hepatic disease. Analyses of the pooled individual patients' data revealed that clearance maturation relating to body weight could be best described by the Hill function for sufentanil (R 2 = 0.71, B max 876 mL/min, K 50 16.3 kg) and alfentanil (R 2 = 0.70, B max (fixed) 420 mL/min, K 50 28 kg). The allometric exponent for estimation of clearance of sufentanil was 0.99 and 0.75 for alfentanil clearance. Maturation of remifentanil clearance was described by linear regression to bodyweight (R 2 = 0.69). The allometric exponent for estimation of remifentanil clearance was 0.76. For fentanyl, linear regression showed only a weak correlation between clearance and bodyweight in preterm and term neonates (R 2 = 0.22) owing to a lack of data in older age groups. A large heterogeneity regarding study design, clinical setting, drug administration, laboratory assays, and pharmacokinetic estimation was observed between studies introducing bias into the analyses performed in this review. A limitation of this review is that pharmacokinetic data, based on different modes of administration, dosing schemes, and parameter estimation methods, were combined.


Asunto(s)
Analgésicos Opioides/farmacocinética , Fentanilo/farmacocinética , Adolescente , Alfentanilo/administración & dosificación , Alfentanilo/farmacocinética , Analgésicos Opioides/administración & dosificación , Peso Corporal , Niño , Preescolar , Fentanilo/administración & dosificación , Humanos , Lactante , Recién Nacido , Modelos Lineales , Modelos Biológicos , Remifentanilo/administración & dosificación , Remifentanilo/farmacocinética , Sufentanilo/administración & dosificación , Sufentanilo/farmacocinética
14.
J Clin Pharmacol ; 58(7): 877-884, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29489016

RESUMEN

Allometric approaches are widely used for interspecies scaling for the prediction of pharmacokinetic (PK) parameters during drug development. The concept of allometry can also be extended to predict PK parameters from adults to children. Three methods for extrapolating pediatric clearance were developed and evaluated using the clearance values of 4 drugs. The first method was established using a simple allometric (SA) model with estimated coefficient and exponent based on data ranging from children older than 2 years to adult. Then we developed a unified multistep single-exponent (MSE) and multistep body-weight-dependent exponent (MBDE) models. The major steps in these 2 new methods include generating pseudopredicted clearance for unobserved new populations such as preterm neonates, term neonates, and infants. Subsequent steps involve incorporating the pseudopredicted clearance with the actual PK data from older children and adults. All 3 models were then used to predict drug clearance in children ≤2 years old (N = 278). Drug clearance was predicted with mean absolute error of 29.6, 14.2, and 12.9 using SA, MSE, MBDE, respectively. The root mean square error was 65.9, 29.8, 24.7 for SA, MSE, MBDE, respectively. Approximately 41%, 72%, and 74% of the children's clearance data were within 0.5 to 1.5-fold of the observed values when drug clearance was extrapolated using SA, MSE, and MBDE models, respectively. The present multistep unified extrapolation approaches improved the prediction of clearance from preterm neonates to 2 years of age and may have practical use for first-in-pediatric dose selection.


Asunto(s)
Alfentanilo/farmacocinética , Busulfano/farmacocinética , Recién Nacido/metabolismo , Tasa de Depuración Metabólica/efectos de los fármacos , Modelos Biológicos , Sufentanilo/farmacocinética , Teofilina/farmacocinética , Administración Intravenosa , Adolescente , Adulto , Factores de Edad , Peso Corporal , Niño , Preescolar , Humanos , Lactante , Adulto Joven
15.
Clin Pharmacol Ther ; 82(4): 410-26, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17554244

RESUMEN

The hepatic and first-pass cytochrome P4503A (CYP3A) probe alfentanil (ALF) is also metabolized in vitro by CYP3A5. Human hepatic microsomal ALF metabolism is higher in livers with at least one CYP3A5*1 allele and higher CYP3A5 protein content, compared with CYP3A5*3 homozygotes with little CYP3A5. The influence of CYP3A5 genotype on ALF pharmacokinetics and pharmacodynamics was studied, and compared to midazolam (MDZ), another CYP3A probe. Healthy volunteers (58 men, 41 women) were genotyped for CYP3A5 *1, *3, *6, and *7 alleles. They received intravenous MDZ then ALF, and oral MDZ and ALF the next day. Plasma MDZ and ALF concentrations were determined by mass spectrometry. Dark-adapted pupil diameters were determined coincident with blood sampling. In CYP3A5(*)3/(*)3 (n=62), (*)1/(*)3 (n=28), and (*)1/(*)1 (n=8) genotypes, systemic clearances of ALF were 4.6+/-1.8, 4.8+/-1.7, and 3.9+/-1.7 ml/kg/min and those of MDZ were 7.8+/-2.3, 7.7+/-2.3, and 6.0+/-1.4 ml/kg/min, respectively (not significant), and apparent oral clearances were 11.8+/-7.2, 13.3+/-6.1, and 12.6+/-8.2 ml/kg/min for ALF and 35.2+/-19.0, 36.4+/-15.7, and 29.4+/-9.3 ml/kg/min for MDZ (not significant). Clearances were not different between African Americans (n=25) and Whites (n=68), or between CYP3A5 genotypes within African Americans. ALF pharmacodynamics was not different between CYP3A5 genotypes. There was consistent concordance between ALF and MDZ, in clearances and extraction ratios. Thus, in a relatively large cohort of healthy subjects with constitutive CYP3A activity, CYP3A5 genotype had no effect on the systemic or apparent oral clearances, or pharmacodynamics, of the CYP3A probes ALF and MDZ, despite affecting their hepatic microsomal metabolism.


Asunto(s)
Alfentanilo/farmacocinética , Sistema Enzimático del Citocromo P-450/genética , Midazolam/farmacocinética , Polimorfismo Genético , Administración Oral , Adulto , Negro o Afroamericano/genética , Alfentanilo/administración & dosificación , Alfentanilo/efectos adversos , Alfentanilo/sangre , Biomarcadores/metabolismo , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/metabolismo , Femenino , Frecuencia de los Genes , Genotipo , Hispánicos o Latinos/genética , Humanos , Inyecciones Intravenosas , Masculino , Midazolam/administración & dosificación , Midazolam/sangre , Persona de Mediana Edad , Miosis/inducido químicamente , Fenotipo , Pupila/efectos de los fármacos , Valores de Referencia , Especificidad por Sustrato , Población Blanca/genética
16.
Clin Pharmacokinet ; 46(3): 261-70, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17328584

RESUMEN

OBJECTIVES: (i) To evaluate the pupillary response to alfentanil as a surrogate measure of alfentanil pharmacokinetics in cirrhotic patients and to compare the data observed in cirrhotic patients with those found in healthy volunteers (historical control group); and (ii) to compare this test with other liver function tests in cirrhotic patients. METHODS: Six patients with mild cirrhosis (Child-Pugh grade A) and six patients with moderate cirrhosis (Child-Pugh grade B) were studied after a single 15 microg/kg bolus of alfentanil. Alfentanil plasma concentrations were measured by liquid chromatography-tandem mass spectrometry, and pupillary responses were measured with a Pupilscan II pupillometer. Alfentanil pharmacokinetics (plasma concentration, area under the plasma concentration-time curve from time zero to infinity [AUC(infinity(p))] and from time zero to 2 hours [AUC(2(p))], apparent volume of distribution at steady state, clearance and terminal elimination half-life [t((1/2)(p))]) and miosis pseudo-kinetic parameters [AUC(infinity)((miosis)), AUC(2)((miosis)), t((1/2))((miosis))] were determined using a noncompartmental analysis method. In six patients (three Child-Pugh grade A and three Child-Pugh grade B), antipyrine (measure of liver intrinsic activity) and D-sorbitol (measure of liver blood flow) tests were performed. RESULTS: A significant correlation was found between the alfentanil AUC(infinity(p)) and AUC(infinity)((miosis)) (r = 0.6, p < 0.05) in cirrhotic patients. This correlation was even more significant if AUC determinations were limited to the first 2 hours after alfentanil administration (r = 0.9, p < 0.01). Statistically significant differences in pharmacokinetics and miosis pseudo-kinetic parameters were observed between cirrhotic patients and healthy volunteers from our previous experiment (historical control group). The correlations were significant between alfentanil clearance and antipyrine clearance (n = 6, r = 0.9, p < 0.05), alfentanil clearance and steady-state hepatic blood clearance [CL(H(b))] measured by the D-sorbitol test (n = 6, r = 0.9, p < 0.05). CONCLUSION: Alfentanil pharmacokinetic parameters were correlated with miosis pseudo-kinetic parameters in cirrhotic patients. There was a significant decrease in pharmacokinetics and miosis pseudo-kinetics in cirrhotic patients compared with volunteers from the historical control group. Alfentanil-induced miosis has the advantage of being noninvasive and can be limited to miosis measurements during the first 2 hours after alfentanil administration in cirrhotic patients. We thus propose to substitute the AUC(2(miosis)) for alfentanil pharmacokinetics in cirrhosis.


Asunto(s)
Alfentanilo/farmacocinética , Analgésicos Opioides/farmacocinética , Cirrosis Hepática/metabolismo , Pruebas de Función Hepática , Miosis/inducido químicamente , Miosis/fisiopatología , Adulto , Anciano , Antiinflamatorios no Esteroideos , Antipirina , Biomarcadores , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/metabolismo , Diuréticos , Relación Dosis-Respuesta a Droga , Electroencefalografía/efectos de los fármacos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Sorbitol
17.
Clin Pharmacol Ther ; 102(1): 115-122, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28001306

RESUMEN

Doxapram is an analeptic that induces ventilatory stimulation and increases blood pressure and cardiac output (CO). Its mechanism of action is the blockade of background K+ -channels expressed on type 1 carotid body cells. In the randomized controlled trial, the authors explored the role of the increase in CO by doxapram (plasma concentration (Cp) 1,000-3,500 ng/mL) on the pharmacokinetics (PKs) and pharmacodynamics (PDs) of the potent opioid alfentanil (Cp 100-200 ng/mL). Population PK-PD analyses were performed on the doxapram PK-CO data and the alfentanil PK-antinociception data. The analyses showed that the doxapram-induced increase in CO explained the increase in alfentanil distribution and elimination clearances causing a significant reduction in plasma alfentanil Cp and antinociception. This novel approach in which one PK-PD model effectively drives another PK-PD model highlights the importance of physiological influences on PK and PD of a potent opioid with rapid onset of effect and low clinical margin of safety.


Asunto(s)
Alfentanilo , Doxapram , Insuficiencia Respiratoria , Adulto , Alfentanilo/efectos adversos , Alfentanilo/sangre , Alfentanilo/farmacocinética , Analgésicos/administración & dosificación , Analgésicos/farmacocinética , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/sangre , Analgésicos Opioides/farmacocinética , Presión Sanguínea/efectos de los fármacos , Gasto Cardíaco/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/administración & dosificación , Estimulantes del Sistema Nervioso Central/farmacocinética , Doxapram/administración & dosificación , Doxapram/farmacocinética , Voluntarios Sanos , Humanos , Masculino , Tasa de Depuración Metabólica , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/prevención & control , Fármacos del Sistema Respiratorio/administración & dosificación , Fármacos del Sistema Respiratorio/farmacocinética , Resultado del Tratamiento
18.
Clin Pharmacol Ther ; 80(5): 502-8, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17112806

RESUMEN

OBJECTIVE: Alfentanil is a short-acting synthetic opioid analgesic, which is extensively metabolized, mainly by hepatic cytochrome P450 (CYP) 3A enzymes. Concomitant administration of alfentanil and CYP3A inhibitors may lead to clinically important drug interactions. We investigated the possible interactions between alfentanil and orally administered voriconazole and terbinafine. METHODS: A randomized crossover study design in 3 phases was used. Twelve healthy volunteers were given 20 microg/kg intravenous alfentanil without pretreatment (control), after oral voriconazole administration (400 mg twice on the first day and 200 mg twice on the second day), or after oral terbinafine administration (250 mg once daily for 3 days). Plasma concentrations of alfentanil were measured for 10 hours, and the pharmacokinetic parameters were calculated by use of noncompartmental methods. RESULTS: Voriconazole decreased the mean plasma clearance of intravenous alfentanil by 85%, from the control value of 4.4+/-2.4 mL.min-1.kg-1 to 0.67+/-0.27 mL.min-1.kg-1 (P<.001), and prolonged its elimination half-life from 1.5+/-0.49 hours to 6.6+/-1.8 hours (P<.001). The area under the alfentanil plasma concentration-time curve was increased by 6-fold by voriconazole (P<.001). Terbinafine had no statistically significant effect on the pharmacokinetics of alfentanil. Alfentanil administration caused nausea in 5 volunteers and vomiting in 2. These side effects all occurred in volunteers in the voriconazole phase. CONCLUSION: Oral voriconazole, but not terbinafine, markedly inhibited the metabolism of alfentanil. Caution should be exercised when alfentanil is given to patients receiving voriconazole. It is reasonable to assume that patients receiving voriconazole require 70% to 90% less alfentanil for the maintenance of analgesia than patients who are not receiving concomitant CYP3A inhibitors.


Asunto(s)
Alfentanilo/farmacocinética , Naftalenos/farmacología , Pirimidinas/farmacología , Triazoles/farmacología , Administración Oral , Adulto , Alfentanilo/administración & dosificación , Alfentanilo/sangre , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/farmacocinética , Antifúngicos/administración & dosificación , Antifúngicos/efectos adversos , Antifúngicos/farmacología , Área Bajo la Curva , Defectos de la Visión Cromática/inducido químicamente , Estudios Cruzados , Inhibidores del Citocromo P-450 CYP3A , Interacciones Farmacológicas , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/farmacología , Femenino , Semivida , Humanos , Inyecciones Intravenosas , Masculino , Tasa de Depuración Metabólica , Naftalenos/administración & dosificación , Naftalenos/efectos adversos , Fotofobia/inducido químicamente , Pirimidinas/administración & dosificación , Pirimidinas/efectos adversos , Factores Sexuales , Terbinafina , Triazoles/administración & dosificación , Triazoles/efectos adversos , Vómitos/inducido químicamente , Voriconazol
19.
Clin Pharmacokinet ; 45(1): 1-11, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16430308

RESUMEN

Correct dosing of drugs in neonates, infants and children is hampered by a general lack of knowledge about drug disposition in this population. Suggested methods to improve our knowledge without performing conventional full-scale investigations include population pharmacokinetic studies, allometric scaling of drug disposition according to bodyweight and in silico prediction of pharmacokinetics. The last method entails scaling of pharmacokinetic parameters according to age-dependent changes in drug absorption and elimination capacity, plasma protein binding and physiological characteristics of the subjects. Maturation (or ontogeny) of the drug-metabolising part of the cytochrome P450 (CYP) enzyme system is thus an important factor in the calculations for most drugs. The aim of this commentary is to test and critically examine the proposed methods to estimate hepatic clearance (CL) as a function of age (0-20 years), with CYP3A-mediated metabolism as the case in point. Midazolam and alfentanil were used as model drugs. Allometric scaling failed to predict the CL of midazolam and alfentanil in neonates. Calculations using in vitro findings on CYP maturation gave better estimates for neonates but very divergent ones for older infants and children. This was chiefly due to very different data on CYP3A4/5 ontogeny in three published studies. In the age range where full adult CYP activity per gram of liver could be assumed, allometric scaling and in silico predictions gave similar results. These predictions were also in approximate agreement with clinical data.The findings with the two model drugs can very probably be generalised to most drugs cleared by CYP-dependent hepatic metabolism. Allometric scaling accounts for development of body size and function but not for the fact that the drug-metabolising capacity of the liver is generally low at birth. The crucial question in the prediction of CL is thus when the activity of the applicable CYP isoform(s) attains adult levels. There are still not enough data on this, particularly when different studies even on the same CYP isoform have given very divergent results. It may also be pointed out that CYP ontogeny is an area where we have at least some information. There are several other important developmental changes about which we know practically nothing. Thus, while allometric scaling is generally unreliable for prediction in neonates and infants, the alternative method of in silico prediction can at present be used only to obtain tentative initial estimates of drug CL. Neither of the methods can be used as a substitute for actual clinical studies.


Asunto(s)
Envejecimiento/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Hígado/metabolismo , Modelos Biológicos , Farmacocinética , Alfentanilo/farmacocinética , Niño , Preescolar , Citocromo P-450 CYP3A , Humanos , Lactante , Recién Nacido , Hígado/enzimología , Tasa de Depuración Metabólica , Midazolam/farmacocinética , Valor Predictivo de las Pruebas
20.
Eur J Pain ; 10(8): 733-41, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16414295

RESUMEN

Musculoskeletal pain is a major clinical problem. By using various experimental models in humans, the understanding of the basic mechanisms behind muscle pain can increase, thereby giving hope for new and optimized treatment. Opioids are increasingly often used to treat muscle pain. There are, however, a limited number of previous studies on opioids and muscle pain, most of them using a relative low, single dose. Therefore, we wanted to further study the effect of two rather high doses of alfentanil (25 and 75ng/ml) and morphine (0.14 and 0.28mg/kg) in human volunteers. The study consisted of two parallel studies with morphine and alfentanil, respectively, and was conducted as randomized, double-blinded, placebo-controlled, 3-way cross-over. We used intramuscular infusion of hypertonic saline and intramuscular electrical stimulation to induce experimental pain. Visual analog scale (VAS)-score, intramuscular electrical pain thresholds and pain area (local and referred) were measured. Both alfentanil and morphine at their highest doses induced a 6 to 7-fold increase in pain thresholds to single and repetitive (5 stimulations, 2Hz) electrical stimulation. Alfentanil and morphine also reduced VAS score about 4 to 5-fold during suprathreshold electric stimulation and during infusion of hypertonic saline. None of the drugs decreased referred pain. There were no apparent differences between the drugs, in terms of effect or adverse reactions. In conclusion, this is the first study to compare two high doses of alfentanil and morphine on experimental muscle pain in humans. Both alfentanil and morphine reduced experimental muscle pain. There were no indications of any true pharmacodynamic differences between the two drugs.


Asunto(s)
Alfentanilo/administración & dosificación , Analgésicos Opioides/administración & dosificación , Morfina/administración & dosificación , Músculo Esquelético , Dolor/tratamiento farmacológico , Adulto , Alfentanilo/farmacocinética , Analgésicos Opioides/farmacocinética , Estudios Cruzados , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Estimulación Eléctrica , Femenino , Humanos , Masculino , Persona de Mediana Edad , Morfina/farmacocinética , Dolor/etiología , Umbral del Dolor/efectos de los fármacos , Solución Salina Hipertónica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA