Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Diabetes Obes Metab ; 22(6): 969-977, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32009288

RESUMEN

AIMS: To investigate the impact on weight loss of the treatment changes in overweight or obese people that may be needed in case of gastrointestinal (GI) tolerability issues during escalation of the glucagon-like peptide-1 analogue liraglutide. MATERIALS AND METHODS: The individual longitudinal body weight data from the main trial periods of three phase II/III trials in overweight or obese patients (56-week treatment with once-daily liraglutide 1.2, 1.8, 2.4 or 3.0 mg or placebo, n = 4952) were analysed using a non-linear mixed-effect modelling approach. Individual pharmacokinetic profiles were derived based on published pharmacokinetic models. Baseline body weight, baseline glycated haemoglobin (HbA1c), age, gender, diabetes status (no diabetes, prediabetes or type 2 diabetes), race and trial region were investigated as covariates. As a form of external validation, the model was used to predict the weight regain after treatment cessation at week 56 (data not included in model development). RESULTS: A pharmacokinetic/pharmacodynamic model provided an adequate description of the weight loss trajectories for all studied doses. Gender and diabetes status were identified as the most influential covariates, and an underlying seasonal weight fluctuation was identified. Slower than that recommended, one-week dose-escalation algorithms led up to 2 weeks slower initial weight loss but similar long-term weight loss trajectories. CONCLUSIONS: The relationship between liraglutide systemic exposure and weight loss was successfully established in overweight or obese people. The model could predict the time course of weight regain after treatment cessation and suggests that GI tolerability can be mitigated by slower escalation with only minor impact on the weight loss trajectory.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hipoglucemiantes , Liraglutida , Pérdida de Peso , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Método Doble Ciego , Femenino , Hemoglobina Glucada/análisis , Humanos , Hipoglucemiantes/uso terapéutico , Liraglutida/uso terapéutico , Masculino , Resultado del Tratamiento
2.
Br J Anaesth ; 123(2): e204-e214, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30915992

RESUMEN

BACKGROUND: Naloxone, an opioid receptor antagonist, is used as a pharmacological tool to detect tonic endogenous activation of opioid receptors in experimental pain models. We describe a pharmacokinetic model linking naloxone pharmacokinetics to its main metabolite after high-dose naloxone infusion. METHODS: Eight healthy volunteers received a three-stage stepwise high-dose i.v. naloxone infusion (total dose 3.25 mg kg-1). Naloxone and naloxone-3-glucuronide (N3G) plasma concentrations were sampled from infusion onset to 334 min after infusion discontinuation. Pharmacokinetic analysis was performed using non-linear mixed effect models (NONMEM). The predictive performances of Dowling's and Yassen's models were evaluated, and target-controlled infusion simulations were performed. RESULTS: Three- and two-compartment disposition models with linear elimination kinetics described the naloxone and N3G concentration time-courses, respectively. Two covariate models were developed: simple (weight proportional) and complex (with the shallow peripheral volume of distribution linearly increasing with body weight). The median prediction error (MDPE) and wobble for Dowling's model were -32.5% and 33.4%, respectively. For Yassen's model, the MDPE and wobble were 1.2% and 19.9%, respectively. CONCLUSIONS: A parent-metabolite pharmacokinetic model was developed for naloxone and N3G after high-dose naloxone infusion. No saturable pharmacokinetics were observed. Whereas Dowling's model was inaccurate and over-predicted naloxone concentrations, Yassen's model accurately predicted naloxone pharmacokinetics. The newly developed covariate models may be used for high-dose TCI-naloxone for experimental and clinical practice. CLINICAL TRIALS REGISTRATION: NCT01992146.


Asunto(s)
Naloxona/farmacocinética , Antagonistas de Narcóticos/farmacocinética , Adolescente , Adulto , Humanos , Masculino , Adulto Joven
3.
Pharm Res ; 33(11): 2630-43, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27380190

RESUMEN

PURPOSE: The combination of morphine and gabapentin seems promising for the treatment of postoperative and neuropathic pain. Despite the well characterised pharmacodynamic interaction, little is known about possible pharmacokinetic interactions. The aim of this study was to evaluate whether co-administration of the two drugs leads to modifications of their pharmacokinetic profiles. METHODS: The pharmacokinetics of morphine, morphine-3-glucuronide and gabapentin were characterised in rats following subcutaneous injections of morphine, gabapentin or their combination. Non-linear mixed effects modelling was applied to describe the pharmacokinetics of the compounds and possible interactions. RESULTS: The plasma-concentration-time profiles of morphine and gabapentin were best described using a three- and a one-compartment disposition model respectively. Dose dependencies were found for morphine absorption rate and gabapentin bioavailability. Enterohepatic circulation of morphine-3-glucuronide was modelled using an oscillatory model. The combination did not lead to pharmacokinetic interactions for morphine or gabapentin but resulted in an estimated ~33% diminished morphine-3-glucuronide formation. CONCLUSIONS: The finding of a lack of pharmacokinetic interaction strengthens the notion that the combination of the two drugs leads to better efficacy in pain treatment due to interaction at the pharmacodynamic level. The interaction found between gabapentin and morphine-3-glucuronide, the latter being inactive, might not have any clinical relevance.


Asunto(s)
Aminas/química , Aminas/farmacocinética , Analgésicos/química , Ácidos Ciclohexanocarboxílicos/química , Ácidos Ciclohexanocarboxílicos/farmacocinética , Modelos Biológicos , Morfina/química , Morfina/farmacocinética , Ácido gamma-Aminobutírico/química , Ácido gamma-Aminobutírico/farmacocinética , Aminas/metabolismo , Analgésicos/metabolismo , Analgésicos/farmacocinética , Animales , Ácidos Ciclohexanocarboxílicos/metabolismo , Interacciones Farmacológicas , Circulación Enterohepática , Gabapentina , Glucurónidos/metabolismo , Morfina/metabolismo , Dinámicas no Lineales , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem , Ácido gamma-Aminobutírico/metabolismo
4.
Clin Pharmacokinet ; 60(9): 1217-1226, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33864240

RESUMEN

BACKGROUND AND OBJECTIVE: Once-daily injectable recombinant human growth hormone (GH) formulations (e.g. Norditropin®; Novo Nordisk A/S) are used to treat GH deficiency in children and adults, with much of the therapeutic effect mediated via the insulin-like growth factor-I (IGF-I) response. Despite a long history of use, there are few data on the pharmacokinetics and pharmacodynamics (serum IGF-I response) of this therapy, or of potential differences in the relationship of GH pharmacokinetic/pharmacodynamic (PK/PD) effects between children and adults. This study aimed to characterise the GH pharmacokinetics and IGF-I profile following daily subcutaneous GH in adults and children with GH deficiency. METHODS: A model was developed based on a population PK/PD modelling meta-analysis of data from three phase I clinical trials (two using Norditropin® as a comparator with somapacitan, and one as a comparator with a pegylated GH product). Sequential model building was performed, first developing a model that could describe GH pharmacokinetics. A PD model of IGF-I data was then developed using PK and PD data, and where all PK parameters were kept fixed to those estimated in the PK model. RESULTS: The model developed accurately describes and predicts GH pharmacokinetics and IGF-I response. Body weight was shown to have an important inversely correlated influence on GH exposure (and IGF-I standard deviation score), and this largely explained differences between adults and children. CONCLUSIONS: The pharmacokinetics/pharmacodynamics developed here can inform expectations about the PD effects of different doses of GH in patients with GH deficiency of different body weights, regardless of their age. CLINICAL TRIAL REGISTRATION: Pooled modelling analysis of data from ClinicalTrials.gov identifiers NCT01973244, NCT00936403 and NCT01706783. DATES OF REGISTRATION: NCT01973244: 22 October, 2013; NCT00936403: 9 July, 2009; NCT01706783: 11 October, 2012.


Asunto(s)
Enanismo Hipofisario , Hormona de Crecimiento Humana , Adulto , Peso Corporal , Niño , Hormona del Crecimiento , Humanos , Proteínas Recombinantes
5.
PLoS One ; 15(11): e0242169, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33180816

RESUMEN

Severe chronic postsurgical pain has a prevalence of 4-10% in the surgical population. The underlying nociceptive mechanisms have not been well characterized. Following the late resolution phase of an inflammatory injury, high-dose µ-opioid-receptor inverse agonists reinstate hypersensitivity to nociceptive stimuli. This unmasking of latent pain sensitization has been a consistent finding in rodents while only observed in a limited number of human volunteers. Latent sensitization could be a potential triggering venue in chronic postsurgical pain. The objective of the present trial was in detail to examine the association between injury-induced secondary hyperalgesia and naloxone-induced unmasking of latent sensitization. Healthy volunteers (n = 80) received a cutaneous heat injury (47°C, 420 s, 12.5 cm2). Baseline secondary hyperalgesia areas were assessed 1 h post-injury. Utilizing an enriched enrollment design, subjects with a magnitude of secondary hyperalgesia areas in the upper quartile ('high-sensitizers' [n = 20]) and the lower quartile ('low-sensitizers' [n = 20]) were selected for further study. In four consecutive experimental sessions (Sessions 1 to 4), the subjects at two sessions (Sessions 1 and 3) received a cutaneous heat injury followed 168 h later (Sessions 2 and 4) by a three-step target-controlled intravenous infusion of naloxone (3.25 mg/kg), or normal saline. Assessments of secondary hyperalgesia areas were made immediately before and stepwise during the infusions. Simple univariate statistics revealed no significant differences in secondary hyperalgesia areas between naloxone and placebo treatments (P = 0.215), or between 'high-sensitizers' and 'low-sensitizers' (P = 0.757). In a mixed-effects model, secondary hyperalgesia areas were significantly larger following naloxone as compared to placebo for 'high-sensitizers' (P < 0.001), but not 'low-sensitizers' (P = 0.651). Although we could not unequivocally demonstrate naloxone-induced reinstatement of heat injury-induced hyperalgesia, further studies in clinical postsurgical pain models are warranted.


Asunto(s)
Hiperalgesia/fisiopatología , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Nocicepción/efectos de los fármacos , Dolor/fisiopatología , Calor , Humanos , Infusiones Intravenosas , Masculino , Naloxona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Adulto Joven
6.
AAPS J ; 22(2): 32, 2020 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-31989328

RESUMEN

Exposure-response (ER) modeling for fixed-dose combinations (FDC) has previously been found to have an inflated false positive rate (FP), i.e., observing a significant effect of FDC components when no true effect exists. Longitudinal exposure-response (LER) analysis utilizes the time course of the data and is valid for several clinical endpoints for FDCs. The aim of the study was to investigate if LER is applicable for the validation of FDCs by demonstrating the contribution of each component to the overall effect without inflation of FP rates. FP and FN rates associated with ER and LER analysis were investigated using stochastic simulation and estimation. Four hundred thirty-two scenarios with varying numbers of patients, duration, sampling frequency, dose distribution, design, and drug activity were analyzed using a range of linear, log-linear, and non-linear models to asses FP and FN rates. Lastly, the impact of the clinical trial parameters was investigated. LER analyses provided well-controlled FP rates of the expected 5% or less; however, in low information clinical trials consisting of 30 patients, 4 samples, and 20 days, LER analyses lead to inflated FN rates. Parameter investigation showed that when the clinical trial includes sufficient patients, duration, samples, and an appropriate trial design, the FN rates are in general below the expected 5% for LER analysis. Based on the results, LER analysis can be used for the validation of FDCs and fixed ratio drug combinations. The method constitutes a new avenue for providing evidence that demonstrates the contribution of each component to the overall clinical effect.


Asunto(s)
Combinación de Medicamentos , Modelos Biológicos , Farmacocinética , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Humanos , Modelos Lineales , Estudios Longitudinales , Dinámicas no Lineales , Reproducibilidad de los Resultados
7.
AAPS J ; 21(5): 95, 2019 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-31359219

RESUMEN

Combinations of pharmacological treatments are increasingly being investigated for potentially higher clinical benefit, especially when the combined drugs are expected to act via synergistic interactions. The clinical development of combination treatments is particularly challenging, particularly during the dose-selection phase, where a vast range of possible combination doses exists. The purpose of this work was to evaluate the added value of using optimal design for guiding the dose allocation in drug combination dose-finding studies as compared with a typical drug-combination trial. Optimizations were performed using local [D(s)-optimality] and global [ED(s)-optimality] optimal designs to maximize the precision of model parameters in a number of potential exposure-response (E-R) surfaces. A compound criterion [D(s)/V-optimality] was used to optimize the precision of model predictions in specific parts of the E-R surfaces. Optimal designs provided unbiased estimates and significantly improved the accuracy of results relative to the typical design. It was possible to improve the efficiency and overall parameter precision up to 7832% and 96.6% respectively. When the compound criterion was used, the probability to accurately identify the optimal dose-combination increased from 71% for the typical design up to 91%. These results indicate that optimal design methodology in tandem with E-R analyses is a beneficial tool that can be used for appropriate dose allocation in dose-finding studies for drug combinations.


Asunto(s)
Combinación de Medicamentos , Diseño de Fármacos , Modelos Biológicos , Preparaciones Farmacéuticas/administración & dosificación , Relación Dosis-Respuesta a Droga , Desarrollo de Medicamentos/métodos , Humanos , Proyectos de Investigación
8.
Bioanalysis ; 11(3): 165-173, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30661370

RESUMEN

Aim: High-dose administration of the µ-opioid receptor inverse agonist naloxone (NX), has previously been demonstrated to reinstate nocifensive behavior in the late phase of inflammatory injuries. However, no current analytical methods can provide pharmacokinetic insight into the pharmacodynamic response of high-dose administration of NX. Materials & methods: Based on protein precipitation using 50 µl human plasma, NX and naloxone-ß-d-glucuronide (NXG) was analysed by UHPLC-MS/MS with 6 min cycle time. Results: A method for quantification of high-dose administered NX and NXG was developed and validated with intra- and interday precision and accuracy within ≤8.5% relative standard deviation (RSD) and -1.2-5.5% relative error (RE) for NX and ≤9.6% RSD and 0.6-6.5% RE for NXG. The method show excellent internal standard corrected matrix effects. Conclusion: A rapid UHPLC-MS/MS method was developed for quantification of NX and NXG in human plasma within 10-4000 ng/ml.

9.
AAPS J ; 20(3): 64, 2018 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-29687351

RESUMEN

The exposure-response relationship of combinatory drug effects can be quantitatively described using pharmacodynamic interaction models, which can be used for the selection of optimal dose combinations. The aim of this simulation study was to evaluate the reliability of parameter estimates and the probability for accurate dose identification for various underlying exposure-response profiles, under a number of different phase II designs. An efficacy variable driven by the combined exposure of two theoretical compounds was simulated and model parameters were estimated using two different models, one estimating all parameters and one assuming that adequate previous knowledge for one drug is readily available. Estimation of all pharmacodynamic parameters under a realistic, in terms of sample size and study design, phase II trial, proved to be challenging. Inaccurate estimates were found in all exposure-response scenarios, except for situations where no pharmacodynamic interaction was present, with the drug potency and interaction parameters being the hardest to estimate. When previous knowledge of the exposure-response relationship of one of the monocomponents is available, such information should be utilized, as it enabled relevant improvements in parameter estimation and in correct dose identification. No general trends for classification of the performance of the tested study designs across different scenarios could be identified. This study shows that pharmacodynamic interactions models can be used for the exposure-response analysis of clinical endpoints especially when accompanied by appropriate dose selection in regard to the expected drug potencies and appropriate trial size and if information regarding the exposure-response profile of one monocomponent is available.


Asunto(s)
Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Ensayos Clínicos Fase II como Asunto , Estudios de Factibilidad , Humanos , Reproducibilidad de los Resultados , Proyectos de Investigación , Tamaño de la Muestra , Procesos Estocásticos
10.
AAPS J ; 19(6): 1804-1813, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28853055

RESUMEN

The combination of morphine and gabapentin has shown to be promising for managing postoperative pain but finding the right dose for the combination has proven to be a challenge. The purpose of this study was to quantitatively characterize the pharmacodynamic interaction between the two drugs and to identify the optimal concentration-effect relationship of the combination. Information regarding plasma concentrations and von Frey withdrawal thresholds following incisional surgery on Sprague Dawley rats, after administration of morphine, gabapentin, or their combination was available from published studies. The combined pharmacodynamic effect of morphine and gabapentin was analyzed and linked to drug plasma concentrations via a response surface approach using non-linear mixed-effect modeling. Full reversal of withdrawal thresholds for the pain stimulation to presurgery values was estimated at morphine plasma concentration of 435.1 ng/mL. Co-administration of up to 40 µg/mL of gabapentin led to a reduction of the needed morphine concentration down to 307.5 ng/mL (~ 29% reduction). Combination of concentration ranges of gabapentin between 20 and 40 µg/mL with any morphine concentrations between 100 and 600 ng/mL were found to lead up to 50% increased effect relatively to the effect attained by morphine alone. This study highlights the importance of finding the right combination in multimodal analgesia and demonstrates the usefulness of the response surface approach for the study of pharmacodynamic interactions. The proposed pharmacokinetic-pharmacodynamic model may provide the basis for a rational clinical trial design with the aim to identify the optimal dose combination ratios in humans.


Asunto(s)
Aminas/farmacología , Analgésicos/farmacología , Ácidos Ciclohexanocarboxílicos/farmacología , Morfina/farmacología , Ácido gamma-Aminobutírico/farmacología , Animales , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Gabapentina , Dolor Postoperatorio/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley
11.
Eur J Pharm Sci ; 82: 97-105, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26610393

RESUMEN

Despite much evidence that combination of morphine and gabapentin can be beneficial for managing postoperative pain, the nature of the pharmacological interaction of the two drugs remains unclear. The aim of this study was to assess the interaction of morphine and gabapentin in range of different dose combinations and investigate whether co-administration leads to synergistic effects in a preclinical model of postoperative pain. The pharmacodynamic effects of morphine (1, 3 and 7mg/kg), gabapentin (10, 30 and 100mg/kg) or their combination (9 combinations in total) were evaluated in the rat plantar incision model using an electronic von Frey device. The percentage of maximum possible effect (%MPE) and the area under the response curve (AUC) were used for evaluation of the antihyperalgesic effects of the drugs. Identification of synergistic interactions was based on Loewe additivity response surface analyses. The combination of morphine and gabapentin resulted in synergistic antihyperalgesic effects in a preclinical model of postoperative pain. The synergistic interactions were found to be dose dependent and the increase in observed response compared to the theoretical additive response ranged between 26 and 58% for the synergistic doses. The finding of dose-dependent synergistic effects highlights that choosing the right dose-dose combination is of importance in postoperative pain therapy. Our results indicate benefit of high doses of gabapentin as adjuvant to morphine. If these findings translate to humans, they might have important implications for the treatment of pain in postoperative patients.


Asunto(s)
Aminas/administración & dosificación , Analgésicos/administración & dosificación , Ácidos Ciclohexanocarboxílicos/administración & dosificación , Hiperalgesia/tratamiento farmacológico , Morfina/administración & dosificación , Dolor Postoperatorio/tratamiento farmacológico , Ácido gamma-Aminobutírico/administración & dosificación , Aminas/farmacocinética , Aminas/uso terapéutico , Analgésicos/farmacocinética , Analgésicos/uso terapéutico , Animales , Ácidos Ciclohexanocarboxílicos/farmacocinética , Ácidos Ciclohexanocarboxílicos/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Gabapentina , Masculino , Morfina/farmacocinética , Morfina/uso terapéutico , Estimulación Física , Ratas Sprague-Dawley , Ácido gamma-Aminobutírico/farmacocinética , Ácido gamma-Aminobutírico/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA