Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 208
Filtrar
1.
Pharmacogenomics J ; 24(3): 18, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38824169

RESUMO

The aim was to determine if opioid neuroimmunopharmacology pathway gene polymorphisms alter serum morphine, morphine-3-glucuronide and morphine-6-glucuronide concentration-response relationships in 506 cancer patients receiving controlled-release oral morphine. Morphine-3-glucuronide concentrations (standardised to 11 h post-dose) were higher in patients without pain control (median (interquartile range) 1.2 (0.7-2.3) versus 1.0 (0.5-1.9) µM, P = 0.006), whereas morphine concentrations were higher in patients with cognitive dysfunction (40 (20-81) versus 29 (14-60) nM, P = 0.02). TLR2 rs3804100 variant carriers had reduced odds (adjusted odds ratio (95% confidence interval) 0.42 (0.22-0.82), P = 0.01) of opioid adverse events. IL2 rs2069762 G/G (0.20 (0.06-0.52)), BDNF rs6265 A/A (0.15 (0.02-0.63)) and IL6R rs8192284 carrier (0.55 (0.34-0.90)) genotypes had decreased, and IL6 rs10499563 C/C increased (3.3 (1.2-9.3)), odds of sickness response (P ≤ 0.02). The study has limitations in heterogeneity in doses, sampling times and diagnoses but still suggests that pharmacokinetics and immune genetics co-contribute to morphine pain control and adverse effects in cancer patients.


Assuntos
Analgésicos Opioides , Dor do Câncer , Preparações de Ação Retardada , Morfina , Farmacogenética , Humanos , Morfina/efeitos adversos , Morfina/farmacocinética , Morfina/administração & dosagem , Masculino , Feminino , Dor do Câncer/tratamento farmacológico , Dor do Câncer/genética , Pessoa de Meia-Idade , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/administração & dosagem , Preparações de Ação Retardada/farmacocinética , Idoso , Farmacogenética/métodos , Polimorfismo de Nucleotídeo Único/genética , Derivados da Morfina/farmacocinética , Derivados da Morfina/efeitos adversos , Adulto , Variantes Farmacogenômicos , Receptor 2 Toll-Like/genética
2.
Acta Pharmacol Sin ; 45(8): 1752-1764, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38570601

RESUMO

Morphine and morphine-6-glucuronide (M6G) produce central nervous system (CNS) effects by activating mu-opioid receptors, while naloxone is used mainly for the reversal of opioid overdose, specifically for the fatal complication of respiratory depression, but also for alleviating opioid-induced side effects. In this study we developed a physiologically-based pharmacokinetic-pharmacodynamic (PBPK-PD) model to simultaneously predict pharmacokinetics and CNS effects (miosis, respiratory depression and analgesia) of morphine as well as antagonistic effects of naloxone against morphine. The pharmacokinetic and pharmacodynamic parameters were obtained from in vitro data, in silico, or animals. Pharmacokinetic and pharmacodynamic simulations were conducted using 39 and 36 clinical reports, respectively. The pharmacokinetics of morphine and M6G following oral or intravenous administration were simulated, and the PBPK-PD model was validated using clinical observations. The Emax model correlated CNS effects with free concentrations of morphine and M6G in brain parenchyma. The predicted CNS effects were compared with observations. Most clinical observations fell within the 5th-95th percentiles of simulations based on 1000 virtual individuals. Most of the simulated area under the concentration-time curve or peak concentrations also fell within 0.5-2-fold of observations. The contribution of morphine to CNS effects following intravenous or oral administration was larger than that of M6G. Pharmacokinetics and antagonistic effects of naloxone on CNS effects were also successfully predicted using the developed PBPK-PD model. In conclusion, the pharmacokinetics and pharmacodynamics of morphine and M6G, antagonistic effects of naloxone against morphine-induced CNS effects may be successfully predicted using the developed PBPK-PD model based on the parameters derived from in vitro, in silico, or animal studies.


Assuntos
Modelos Biológicos , Morfina , Naloxona , Antagonistas de Entorpecentes , Naloxona/farmacocinética , Naloxona/farmacologia , Humanos , Morfina/farmacocinética , Morfina/administração & dosagem , Morfina/farmacologia , Antagonistas de Entorpecentes/farmacocinética , Antagonistas de Entorpecentes/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Animais , Derivados da Morfina/farmacocinética , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/administração & dosagem , Masculino , Simulação por Computador , Administração Oral , Adulto , Administração Intravenosa , Feminino
3.
Anesth Analg ; 138(3): 598-606, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-37733945

RESUMO

BACKGROUND: Subanesthetic ketamine may reduce perioperative consumption of opioids. We studied whether intravenous S-ketamine alters the pharmacokinetics of oral morphine in healthy volunteers. METHODS: In this paired, randomized, double-blind, crossover trial, 12 participants under a 2-hour intravenous S-ketamine (0.57 mg/kg/h) or placebo infusion received oral morphine (0.2 mg/kg) at 30 minutes. Plasma concentrations of ketamine, morphine, and their major metabolites were quantified for 24 hours. The primary end point was area under the curve (AUC) 0-24 of morphine. Other pharmacokinetic variables for morphine and its metabolites were studied as secondary end points. The data were analyzed as between-phase comparisons for each participant using Wilcoxon matched-pairs signed-rank tests ( tmax ) or paired t -tests on log-transformed variables (other variables). RESULTS: While the AUC 0-24 was similar between the 2 phases, S-ketamine reduced the AUC 0-1.5 of oral morphine by 69% (ratio to control, 0.31; 90% confidence interval [CI], 0.15-0.65; P = .0171) and increased its tmax from 0.5 (range, 0.50-1.5) to 1.0 hour (range, 0.50-4.0; P = .010). The AUC 0-1.5 of morphine-6-glucuronide (M6G) was reduced by 84% (0.16; 90% CI, 0.07-0.37; P = .0025) and maximum plasma concentration ( Cmax ) by 43% (0.57; 90% CI, 0.40-0.81; P = .0155), while its tmax was increased from 1.5 (range, 1.0-2.0) to 4.0 (range, 1.0-8.0; P = .0094) hours by S-ketamine. Similarly, the AUC 0-1.5 of morphine-3-glucuronide (M3G) was reduced by 85% (0.15; 90% CI, 0.05-0.43; P = .0083), and tmax increased from 1.0 (range, 0.5-1.5) to 4.0 hours (range, 1.0-8.0; P = .0063). In addition, the M6G-to-morphine and M3G-to-morphine metabolic AUC ratios were decreased by 47% (0.53; 90% CI, 0.39-0.71; P = .0033) and 52% (0.48; 90% CI, 0.27-0.85; P = .0043) during 0 to 1.5 hours and by 15% (0.85; 90% CI, 0.78-0.92; P = .0057) and 10% (0.90; 90% CI, 0.83-0.98; P = .0468) during 0 to 24 hours, respectively. One participant was excluded from the analyses due to vomiting in the S-ketamine phase. CONCLUSIONS: Intravenous S-ketamine inhibited the metabolism of oral morphine and delayed its absorption, resulting in a net reduction in the exposure to morphine during the first 1.5 hours. Intravenous S-ketamine may delay the absorption and impair the efficacy of orally administered analgesics and other drugs.


Assuntos
Ketamina , Humanos , Voluntários Saudáveis , Morfina , Derivados da Morfina/farmacocinética , Analgésicos Opioides
4.
Pharmacol Ther ; 241: 108312, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36423714

RESUMO

Morphine prescribed for analgesia has caused drug-related deaths at an estimated incidence of 0.3% to 4%. Morphine has pharmacological properties that make it particularly difficult to assess the causality of morphine administration with a patient's death, such as its slow transfer between plasma and central nervous sites of action and the existence of the active metabolite morphine-6-glucuronide with opioid agonistic effects, Furthermore, there is no well-defined toxic dose or plasma/blood concentration for morphine. Dosing is often adjusted for adequate pain relief. Here, we summarize reported deaths associated with morphine therapy, including associated morphine exposure and modulating patient factors such as pharmacogenetics, concomitant medications, or comorbidities. In addition, we systematically analyzed published numerical information on the stability of concentrations of morphine and its relevant metabolites in biological samples collected postmortem. A medicolegal case is presented in which the causality of morphine administration with death was in dispute and pharmacokinetic modeling was applied to infer the administered dose. The results of this analytical review suggest that (i) inference from postmortem blood concentrations to the morphine dose administered has low validity and (ii) causality between a patient's death and the morphine dose administered remains a highly context-dependent and collaborative assessment among experts from different medical specialties.


Assuntos
Analgésicos Opioides , Morfina , Humanos , Morfina/efeitos adversos , Analgésicos Opioides/farmacologia , Ciência de Dados , Derivados da Morfina/farmacocinética , Derivados da Morfina/uso terapêutico , Dor/tratamento farmacológico
5.
Vet Anaesth Analg ; 49(6): 634-644, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35999165

RESUMO

OBJECTIVE: To describe the pharmacokinetics, behavioral and physiologic effects and effects on thermal thresholds of morphine, morphine 6-glucuronide (M6G) and morphine 3-glucuronide (M3G) following administration to horses. STUDY DESIGN: Randomized balanced crossover study. ANIMALS: A total of seven University-owned horses, five mares and two geldings, aged 3-6 years. METHODS: Horses were treated with a single intravenous dosage of saline, morphine (0.2 mg kg-1), M6G (0.01 mg kg-1) and M3G (0.03 mg kg-1). Blood was collected prior to (baseline) and at several times post administration. Drug and metabolite concentrations were determined by liquid chromatography-mass spectrometry, and plasma pharmacokinetics were calculated. Behavioral observations and physiologic variables (heart rate, step counts, packed cell volume, total plasma protein and gastrointestinal sounds) were determined at baseline and for up to 6 hours. The effects on thermal nociception were determined and thermal excursion was calculated. RESULTS: The volumes of distribution were 4.75-10.5, 0.244-0.295 and 0.215-0.356 L kg-1 for morphine, M6G and M3G, respectively. Systemic clearances were 26.8-39.6, 3.16-3.88 and 1.46-2.13 mL minute-1 kg-1 for morphine, M6G and M3G, respectively. Morphine administration resulted in signs of excitation as evidenced by an increase in step counts and subjective behavioral observations, whereas M6G and M3G, based on the same criteria, appeared to cause sedative-like effects. Significant effects on thermal nociception were observed until 4 hours post morphine administration, 1 hour post M6G administration and at various times post M3G administration. CONCLUSIONS AND CLINICAL RELEVANCE: Results of this study provide additional information regarding the use of morphine in horses. Less locomotor excitation and gastrointestinal adverse effects, compared with morphine, coupled with favorable effects on thermal nociception are encouraging for further study of the pharmacodynamics of both M6G and M3G in horses.


Assuntos
Glucuronídeos , Nociceptividade , Cavalos , Animais , Masculino , Feminino , Estudos Cross-Over , Derivados da Morfina/farmacocinética , Morfina
6.
Can J Vet Res ; 86(3): 172-180, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35794968

RESUMO

Although morphine has demonstrated antinociceptive effects in horses, its administration has been associated with dose-dependent adverse effects. In humans and rats, part of the analgesic effect of morphine has been attributed to the active metabolite, morphine-6-glucuronide (M6G). Although morphine can cause several undesirable effects, M6G has a more favorable safety profile. The objective of this study was to characterize the pharmacokinetics, tissue distribution, and behavioral and select physiological effects of M6G following intravenous administration to a small group of horses. In Part 1 of the study, 3 horses received a single intravenous administration of saline, 0.5 mg/kg body weight (BW) M6G, or 0.5 mg/kg BW morphine in a 3-way crossover design. Blood samples were collected up to 96 hours post-administration, concentrations of drug and metabolites measured, and pharmacokinetics determined. Behavioral and physiological effects were then recorded. In Part 2 of the study, 2 horses scheduled to be euthanized for other reasons, were administered 0.5 mg/kg BW M6G. Blood, cerebrospinal fluid (CSF), and various tissue samples were collected post-administration and concentrations of drug were determined. The clearance of M6G was more rapid and the volume of distribution at steady state was smaller for M6G compared to morphine. A reaction characterized by head shaking, pawing, and slight ataxia was observed immediately following administration of both morphine and M6G to horses. After M6G administration, these behaviors subsided rapidly and were followed by a longer period of sedation. Following administration, M6G was detected in the kidney, liver, CSF, and regions of the brain. Results of this study encourage further investigation of M6G in order to assess its clinical feasibility as an analgesic in horses.


Bien que la morphine ait démontré des effets antinociceptifs chez les chevaux, son administration a été associée avec des effets non-désirés d'une manière dose-dépendante. Chez les humains et les rats, une partie de l'effet analgésique de la morphine a été attribuée au métabolite actif, morphine-6-glucuronide (M6G). Bien que la morphine puisse causer plusieurs effets indésirables, M6G a un profil de sécurité plus favorable. L'objectif de cette étude était de caractériser la pharmacocinétique, la distribution tissulaire, et le comportement et sélectionner des effets physiologiques de M6G suivant son administration intraveineuse à un petit groupe de chevaux. Dans la Partie 1 de l'étude, trois chevaux ont reçu l'administration intraveineuse d'une dose unique de saline, 0,5 mg/kg de poids corporel (BW) de M6G, ou 0,5 mg/kg BW de morphine selon un essai croisé à trois voies. Des échantillons sanguins ont été prélevés jusqu'à 96 h post-administration, les concentrations de drogues et de métabolites mesurées, et les pharmacocinétiques déterminées. Les effets physiologiques et sur le comportement ont par la suite été notés. Dans la Partie 2 de l'étude, deux chevaux devant être euthanasiés pour d'autres raisons, ont reçu 0,5 mg/kg BW de M6G. Du sang, du liquide céphalo-rachidien (CSF), et différents échantillons de tissu ont été prélevés post-administration et les concentration de drogue furent déterminées. La clairance de M6G a été plus rapide et le volume de distribution à l'état d'équilibre était plus petit pour M6G comparativement à la morphine. Une réaction caractérisée par le tremblement de la tête, du piaffage, et une légère ataxie a été observée immédiatement à la suite de l'administration soit de morphine ou de M6G aux chevaux. Après administration de M6G, ces comportements diminuèrent rapidement et furent suivis par une période plus longue de sédation. À la suite de l'administration, M6G a été détecté dans les reins, le foie, le CSF, et des régions du cerveau. Les résultats de cette étude incitent à réaliser des études additionnelles sur M6G afin d'évaluer son potentiel clinique comme analgésique chez les chevaux.(Traduit par Docteur Serge Messier).


Assuntos
Analgésicos Opioides , Glucuronídeos , Administração Intravenosa/veterinária , Animais , Cavalos , Morfina/farmacologia , Derivados da Morfina/farmacocinética , Ratos , Distribuição Tecidual
7.
BMC Vet Res ; 18(1): 196, 2022 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-35614473

RESUMO

BACKGROUND: In humans, codeine is a commonly prescribed analgesic that produces its therapeutic effect largely through metabolism to morphine. In some species, analgesic effects of morphine have also been attributed to the morphine-6-glucuronide (M6G) metabolite. Although an effective analgesic, administration of morphine to horses produces dose-dependent neuroexcitation at therapeutic doses. Oral administration of codeine at a dose of 0.6 mg/kg has been shown to generate morphine and M6G concentrations comparable to that observed following administration of clinically effective doses of morphine, without the concomitant adverse effects observed with morphine administration. Based on these results, it was hypothesized that codeine administration would provide effective analgesia with decreased adverse excitatory effects compared to morphine. Seven horses received a single oral dose of saline or 0.3, 0.6 or 1.2 mg/kg codeine or 0.2 mg/kg morphine IV (positive control) in a randomized balanced 5-way cross-over design. Blood samples were collected up to 72 hours post administration, codeine, codeine 6-glucuronide, norcodeine morphine, morphine 3-glucuronide and M6G concentrations determined by liquid chromatography- mass spectrometry and pharmacokinetic analysis performed. Pre- and post-drug related behavior, locomotor activity, heart rate and gastrointestinal borborygmi were recorded. Response to noxious stimuli was evaluated by determining thermal threshold latency. RESULTS: Morphine concentrations were highest in the morphine dose group at all times post administration, however, M6G concentrations were significantly higher in all the codeine dose groups compared to the morphine group starting at 1 hour post drug administration and up to 72-hours in the 1.2 mg/kg group. With the exception of one horse that exhibited signs of colic following administration of 0.3 and 0.6 mg/kg, codeine administration was well tolerated. Morphine administration, led to signs of agitation, tremors and excitation. There was not a significant effect on thermal nociception in any of the dose groups studied. CONCLUSIONS: The current study describes the metabolic profile and pharmacokinetics of codeine in horses and provides information that can be utilized in the design of future studies to understand the anti-nociceptive and analgesic effects of opioids in this species with the goal of promoting judicious and safe use of this important class of drugs.


Assuntos
Codeína , Glucuronídeos , Nociceptividade , Analgésicos Opioides , Animais , Codeína/efeitos adversos , Codeína/farmacocinética , Glucuronídeos/efeitos adversos , Glucuronídeos/farmacocinética , Cavalos , Morfina , Derivados da Morfina/efeitos adversos , Derivados da Morfina/farmacocinética
8.
Basic Clin Pharmacol Toxicol ; 130(1): 93-102, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34599645

RESUMO

We investigated the impact of genetic variants in OCT1 (SLC22A1) on morphine, morphine-3-glucuronide (M3G) and morphine-6-glucuronide (M6G) pharmacokinetics in adult patients scheduled for major surgery. Blood samples were taken before and 5, 10, 15, 30, 45, 60 and 90 min after a bolus of morphine (0.15 mg/kg). Patients were genotyped for the genetic variants (rs12208357, rs34059508, rs72552763 and rs34130495) in OCT1. Eighty-six patients completed the trial. The mean difference (95% confidence interval) for dose adjusted morphine, M3G and M6G AUC was 0.9 (-0.7-2.4), -5.9 (-11.8 to -0.03) and -1.1 (-2.5-0.4) h/L*10-6 , respectively, in patients with two reduced function alleles compared to patients with no reduced function alleles in OCT1. Accordingly, the (AUCM3G/Dose )/(AUCmorphine/Dose ) and (AUCM6G/Dose )/(AUCmorphine/Dose ) ratio was reduced, -1.8 (-3.2 to -0.4) and -0.4 (-0.7 to -0.03), respectively, when comparing the same groups. OCT1 variants had no influence on the experience of pain, adverse events or the number of PCA doses used. In conclusion, genetic variants in OCT1 had a small and clinically unimportant impact on the exposure of morphine after intravenous administration. Our results do not support pre-emptive genotyping for OCT1 prior to morphine administration in patients scheduled for major surgery.


Assuntos
Analgésicos Opioides/farmacocinética , Morfina/farmacocinética , Fator 1 de Transcrição de Octâmero/genética , Idoso , Analgésicos Opioides/administração & dosagem , Área Sob a Curva , Feminino , Variação Genética , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Morfina/administração & dosagem , Derivados da Morfina/farmacocinética , Dor Pós-Operatória/tratamento farmacológico , Fatores de Tempo
9.
PLoS Comput Biol ; 17(3): e1008786, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33661919

RESUMO

Morphine is a widely used opioid analgesic, which shows large differences in clinical response in children, even when aiming for equivalent plasma drug concentrations. Age-dependent brain disposition of morphine could contribute to this variability, as developmental increase in blood-brain barrier (BBB) P-glycoprotein (Pgp) expression has been reported. In addition, age-related pharmacodynamics might also explain the variability in effect. To assess the influence of these processes on morphine effectiveness, a multi-compartment brain physiologically based pharmacokinetic/pharmacodynamic (PB-PK/PD) model was developed in R (Version 3.6.2). Active Pgp-mediated morphine transport was measured in MDCKII-Pgp cells grown on transwell filters and translated by an in vitro-in vivo extrapolation approach, which included developmental Pgp expression. Passive BBB permeability of morphine and its active metabolite morphine-6-glucuronide (M6G) and their pharmacodynamic parameters were derived from experiments reported in literature. Model simulations after single dose morphine were compared with measured and published concentrations of morphine and M6G in plasma, brain extracellular fluid (ECF) and cerebrospinal fluid (CSF), as well as published drug responses in children (1 day- 16 years) and adults. Visual predictive checks indicated acceptable overlays between simulated and measured morphine and M6G concentration-time profiles and prediction errors were between 1 and -1. Incorporation of active Pgp-mediated BBB transport into the PB-PK/PD model resulted in a 1.3-fold reduced brain exposure in adults, indicating only a modest contribution on brain disposition. Analgesic effect-time profiles could be described reasonably well for older children and adults, but were largely underpredicted for neonates. In summary, an age-appropriate morphine PB-PK/PD model was developed for the prediction of brain pharmacokinetics and analgesic effects. In the neonatal population, pharmacodynamic characteristics, but not brain drug disposition, appear to be altered compared to adults and older children, which may explain the reported differences in analgesic effect.


Assuntos
Analgésicos Opioides , Encéfalo/metabolismo , Modelos Biológicos , Derivados da Morfina , Morfina , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Adulto , Fatores Etários , Analgesia , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/sangue , Analgésicos Opioides/farmacocinética , Barreira Hematoencefálica/metabolismo , Criança , Pré-Escolar , Biologia Computacional , Feminino , Humanos , Recém-Nascido , Masculino , Morfina/administração & dosagem , Morfina/sangue , Morfina/farmacocinética , Derivados da Morfina/administração & dosagem , Derivados da Morfina/sangue , Derivados da Morfina/farmacocinética
10.
Eur J Drug Metab Pharmacokinet ; 46(2): 249-263, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33547559

RESUMO

BACKGROUND AND OBJECTIVE: Morphine is a standard analgesic drug for postoperative pain therapy. This study aimed to evaluate the pharmacokinetics of morphine and its active metabolite morphine-6-glucuronide (M6G) in cardiac surgery  patients during postoperative analgesia. METHODS: Twenty-five adult patients undergoing cardiac surgery received postoperative pain therapy by patient-controlled analgesia with intravenous bolus doses of morphine. Plasma concentrations of morphine and M6G were determined from arterial samples. Population pharmacokinetic parameters were estimated using nonlinear mixed-effects modeling. RESULTS: Data from twenty-one patients (aged 44-79 years) were analyzed. Pharmacokinetics were best described by a three-compartment model for morphine and a two-compartment model for M6G, linked by a transit compartment. Mean (±SD) population estimates for morphine were: clearance (CL) = 1.35±0.40 L/min, central volume of distribution (V1) = 8.1±2.2 L, steady-state volume of distribution (Vss) = 207±83 L, terminal elimination half-life (T1/2γ) = 177±50 min. Clearance of morphine was proportional to cardiac output. Mean (±SD) population estimates for M6G were: CL = 0.098±0.037 L/min, V1 = 5.5±0.8 L, Vss = 15.8±0.8 L, T1/2ß = 227±74 min. The time to peak concentration of M6G after a bolus dose of morphine was 53±20 min. Clearance of M6G was proportional to estimated glomerular filtration rate. CONCLUSIONS: The pharmacokinetics of morphine and M6G in pain therapy of cardiac surgery  patients could be well described by standard compartmental models. Cardiac output was identified as a significant covariate for morphine clearance, whereas renal function was identified as the most significant covariate for clearance of M6G. These effects should be particularly considered if morphine is administered as a continuous infusion. The developed pharmacokinetic model also enables patient-controlled target-controlled infusion for pain therapy with morphine. TRIAL REGISTRATION: Clinical Trials NCT02483221 (June 26, 2015).


Assuntos
Analgésicos Opioides/farmacocinética , Modelos Biológicos , Derivados da Morfina/farmacocinética , Morfina/farmacocinética , Dor Pós-Operatória/tratamento farmacológico , Adulto , Idoso , Analgesia Controlada pelo Paciente/métodos , Analgésicos Opioides/administração & dosagem , Débito Cardíaco/fisiologia , Procedimentos Cirúrgicos Cardíacos/métodos , Feminino , Meia-Vida , Humanos , Masculino , Pessoa de Meia-Idade , Morfina/administração & dosagem , Distribuição Tecidual
11.
Clin Pharmacol Ther ; 109(3): 676-687, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32897538

RESUMO

Nonalcoholic steatohepatitis (NASH), the progressive form of nonalcoholic fatty liver disease, is increasing in prevalence. NASH-related alterations in hepatic protein expression (e.g., transporters) and in overall physiology may affect drug exposure by altering drug disposition and elimination. The aim of this study was to build a physiologically-based pharmacokinetic (PBPK) model to predict drug exposure in NASH by incorporating NASH-related changes in hepatic transporters. Morphine and morphine-3-glucuronide (M3G) were used as model compounds. A PBPK model of morphine with permeability-limited hepatic disposition was extended to include M3G disposition and enterohepatic recycling (EHR). The model captured the area under the plasma concentration-time curve (AUC) of morphine and M3G after intravenous morphine administration within 0.82-fold and 1.94-fold of observed values from 3 independent clinical studies for healthy adult subjects (6, 10, and 14 individuals). When NASH-related changes in multidrug resistance-associated protein 2 (MRP2) and MRP3 were incorporated into the model, the predicted M3G mean AUC in NASH was 1.34-fold higher compared to healthy subjects, which is slightly lower than the observed value (1.63-fold). Exploratory simulations on other physiological changes occurring in NASH (e.g., moderate decreases in glomerular filtration rate and portal vein blood flow) revealed that the effect of transporter changes was most prominent. Additionally, NASH-related transporter changes resulted in decreased morphine EHR, which could be important for drugs with extensive EHR. This study is an important first step to predict drug disposition in complex diseases such as NASH using PBPK modeling.


Assuntos
Analgésicos Opioides/farmacocinética , Fígado/metabolismo , Modelos Biológicos , Derivados da Morfina/farmacocinética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Administração Intravenosa , Analgésicos Opioides/administração & dosagem , Estudos de Casos e Controles , Simulação por Computador , Humanos , Desintoxicação Metabólica Fase II , Derivados da Morfina/administração & dosagem , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo
12.
Artigo em Inglês | MEDLINE | ID: mdl-33246879

RESUMO

The aim of this study was to develop and to validate a UPLC-MS/MS method for the quantification of morphine, morphine-3-glucuronide, and morphine-6-glucuronide in mouse plasma and tissue homogenates to support preclinical pharmacokinetic studies. The sample preparation consisted of protein precipitation with cold (2-8 °C) methanol:acetonitrile (1:1, v/v), evaporation of the supernatant to dryness, and reconstitution of the dry-extracts in 4 mM ammonium formate pH 3.5. Separation was achieved on a Waters UPLC HSS T3 column (150 × 2.1 mm, 1.8 µm) maintained at 50 °C and using gradient elution with a total runtime of 6.7 min. Mobile phase A consisted of 4 mM ammonium formate pH 3.5 and mobile phase B of 0.1% formic acid in methanol:acetonitrile (1:1, v/v). Detection was carried out by tandem mass spectrometry with electrospray ionization in the positive ion mode. The method was validated within a linear range of 1-2,000 ng/mL, 10-20,000 ng/mL, and 0.5-200 ng/mL for morphine, morphine-3-glucuronide, and morphine-6-glucuronide, respectively. In human plasma, the intra- and inter-run precision of all analytes, including the lower limit of quantification levels, were ≤ 15.8%, and the accuracies were between 88.1 and 111.9%. It has been shown that calibration standards prepared in control human plasma can be used for the quantification of the analytes in mouse plasma and tissue homogenates. The applicability of the method was successfully demonstrated in a preclinical pharmacokinetic study in mice.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Derivados da Morfina/sangue , Espectrometria de Massas em Tandem/métodos , Animais , Modelos Lineares , Camundongos , Derivados da Morfina/análise , Derivados da Morfina/química , Derivados da Morfina/farmacocinética , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
13.
Adv Ther ; 36(9): 2394-2401, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31278694

RESUMO

INTRODUCTION: Food can alter the pharmacokinetics of certain abuse-deterrent formulations. Morphine ARER is an oral abuse-deterrent formulation of ER morphine sulfate tablets formulated with physical and chemical properties that contribute to the abuse-deterrent aspects of the drug. This study compared the relative bioavailability of Morphine ARER in the presence and absence of food. METHODS: This was a randomized, single-dose, two-treatment, crossover study in which healthy adults received Morphine ARER 100 mg under fasting and fed conditions. Subjects were given naltrexone 50 mg to limit opioid effects. Plasma concentrations of morphine and its active metabolite morphine-6-glucuronide (M6G) were obtained up to 48 h post-dose; area under the plasma concentration-time curve (AUC) from time 0 extrapolated to infinity (AUC0-∞), maximum observed plasma concentration (Cmax) and time to Cmax (Tmax) were calculated. Safety was evaluated by observation or report of adverse events, which were monitored during the treatment periods. RESULTS: Of 28 enrolled subjects, 27 completed all treatments; 1 subject in the fasted group withdrew voluntarily. Under fed conditions, the Cmax for morphine was 33% higher (44.78 vs. 33.30 ng/ml for fed and fasted conditions, respectively) and the median Tmax was 30 min longer than under fasted conditions. The overall morphine exposure (AUC0-∞) was similar for fed (440.6 ng · h/ml) vs. fasted conditions (395.1 ng · h/ml). For M6G, the Cmax and AUC0-∞ were similar under both conditions, and the median Tmax for M6G was 60 min longer under fed conditions. Common adverse events were somnolence and nausea. CONCLUSION: Morphine ARER can be administered without regard to food. Plain language summary available for this article. FUNDING: Inspirion Delivery Sciences, LLC.


Food alters how the body processes some currently available opioids. How the opioid is formulated in the final commercial product can impact this effect. Morphine ARER is a new oral abuse-deterrent formulation of extended-release morphine created with properties to make it more difficult to abuse via the intranasal and intravenous routes. To better understand how food affects Morphine ARER bioavailability, we compared the amount of morphine in the blood when 100 mg of Morphine ARER was given with or without food, in random order, to 27 healthy volunteers. Plasma samples were collected up to 48 h after dosing to measure the concentrations of morphine and its active metabolite morphine-6-glucuronide. We measured the amount of drug absorbed by using the area under the plasma concentration-time curve (AUC) and the rate of drug absorption by looking at the highest amount of drug observed in the blood using the maximum observed plasma concentration (Cmax) and time to Cmax (Tmax). When subjects were fed, the Cmax for morphine was 33% higher (44.78 ng/ml) than when they fasted (33.30 ng/ml). The median Tmax was 30 min longer when subjects were fed. Total morphine exposure (AUC0­∞) was similar when subjects were fed (440.6 ng · h/ml) or when they fasted (395.1 ng · h/ml). Safety was evaluated throughout the treatment periods by adverse events, either observed by the clinician or reported by subjects. The most common adverse events noted were somnolence (e.g., sleepiness) and nausea. Our findings show that Morphine ARER has similar bioavailability when taken with or without food.


Assuntos
Analgésicos Opioides/farmacocinética , Derivados da Morfina/farmacocinética , Morfina/farmacocinética , Naltrexona/farmacocinética , Formulações de Dissuasão de Abuso , Adulto , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/sangue , Área Sob a Curva , Disponibilidade Biológica , Estudos Cross-Over , Preparações de Ação Retardada/farmacocinética , Jejum/sangue , Feminino , Interações Alimento-Droga , Humanos , Masculino , Morfina/administração & dosagem , Morfina/sangue , Derivados da Morfina/sangue , Naltrexona/administração & dosagem , Naltrexona/sangue , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/sangue , Antagonistas de Entorpecentes/farmacocinética , Período Pós-Prandial
14.
Drug Test Anal ; 11(2): 292-304, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30156385

RESUMO

The abuse of heroin (diamorphine) and heroin-related deaths are increasing around the world. The interpretation of the toxicological results from suspected heroin-related deaths is notoriously difficult, especially in cases where there may be limited samples. To help forensic practitioners with heroin interpretation, we determined the concentration of morphine (M), morphine-3-glucuronide (M3G), and morphine-6-glucuronide (M6G) in blood (femoral and cardiac), brain (thalamus), liver (deep right lobe), bone marrow (sternum), skeletal muscle (psoas), and vitreous humor in 44 heroin-related deaths. The presence of 6-monoacetylmorphine (6-MAM) in any of the postmortem samples was used as confirmation of heroin use. Quantitation was carried out using a validated liquid chromatography-tandem mass spectrometry (LC-MS/MS) method with solid-phase extraction. We also determined the presence of papaverine, noscapine and codeine in the samples, substances often found in illicit heroin and that may help determine illicit heroin use. The results of this study show that vitreous is the best sample to detect 6-MAM (100% of cases), and thus heroin use. The results of the M, M3G, and M6G quantitation in this study allow a degree of interpretation when samples are limited. However in some cases it may not be possible to determine heroin/morphine use as in four cases in muscle (three cases in bone marrow) no morphine, M3G, or M6G were detected, even though they were detected in other case samples. As always, postmortem cases of suspected morphine/heroin intoxication should be interpreted with care and with as much case knowledge as possible.


Assuntos
Heroína/toxicidade , Derivados da Morfina/farmacocinética , Morfina/farmacocinética , Adulto , Idoso , Medula Óssea/metabolismo , Encéfalo/metabolismo , Codeína/farmacocinética , Feminino , Toxicologia Forense , Humanos , Fígado/metabolismo , Masculino , Pessoa de Meia-Idade , Morfina/sangue , Derivados da Morfina/sangue , Músculo Esquelético/metabolismo , Noscapina/farmacocinética , Papaverina/farmacocinética , Corpo Vítreo/metabolismo , Adulto Jovem
15.
Biomed Chromatogr ; 32(2)2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28833311

RESUMO

A robust ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method for the determination of morphine-6-d-glucuronide (M6G), morphine-3-d-glucuronide (M3G) and morphine (MOR) in human plasma and urine has been developed and validated. The analytes of interest were extracted from plasma by protein precipitation. The urine sample was prepared by dilution. Both plasma and urine samples were chromatographed on an Acquity UPLC HSS T3 column using gradient elution. Detection was performed on a Xevo TQ-S tandem mass spectrometer in multiple reaction monitoring mode using positive electrospray ionization. Matrix interferences were not observed at the retention time of the analytes and internal standard, naloxone-D5. The lower limits of quantitation of plasma and urine were 2/0.5/0.5 and 20/4/2 ng/mL for M6G/M3G/MOR, respectively. Calibration curves were linear over the concentration ranges of 2-2000/0.5-500/0.5-500 and 20-20,000/4-4000/2-2000 ng/mL for M6G/M3G/MOR in plasma and urine samples, respectively. The precision was <7.14% and the accuracy was within 85-115%. Furthermore, stability of the analytes at various conditions, dilution integrity, extraction recovery and matrix effect were assessed. Finally, this quantitative method was successfully applied to the pharmacokinetic study of M6G injection in Chinese noncancer pain patients.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Derivados da Morfina/sangue , Derivados da Morfina/urina , Espectrometria de Massas em Tandem/métodos , Humanos , Limite de Detecção , Derivados da Morfina/química , Derivados da Morfina/farmacocinética , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
16.
In Vivo ; 31(5): 811-817, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28882946

RESUMO

We investigated the in vivo dynamics and analgesic effect of morphine using an adjuvant-induced arthritis (AA) rat as a model of chronic inflammation. Morphine generally binds to µ-opioid receptors in the brain to exert its effects. After several minutes, it is metabolized by glucuronidation via a UDP-glucuronosyltransferase (UGT). Here, we showed that in AA rats, UGT activity in liver microsomes was reduced. Morphine-free serum fractions in AA rats were also decreased (control, 84.9%; AA, 63.9%) and the expression of ATP-binding cassette, sub-family B (MDR/TAP), member 1 (ABCB1), which plays a crucial role in morphine bile excretion, decreased to 23.0% that of the control group. However, we observed no significant difference between the AA and control groups regarding blood concentrations of morphine and morphine-3-glucuronide. In contrast, the analgesic effect of morphine increased 4-fold in AA rats. Our results showed that the pharmacokinetics of morphine is not changed, but the pharmacodynamics of morphine is enhanced in chronic inflammation.


Assuntos
Analgésicos Opioides/farmacocinética , Artrite Experimental/tratamento farmacológico , Morfina/farmacocinética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Artrite Experimental/etiologia , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Biomarcadores , Proteínas Sanguíneas , Modelos Animais de Doenças , Adjuvante de Freund/efeitos adversos , Glucuronosiltransferase/metabolismo , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Microssomos Hepáticos/metabolismo , Derivados da Morfina/farmacocinética , Ligação Proteica , Ratos
17.
Eur J Pharm Sci ; 109S: S132-S139, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28558980

RESUMO

The pharmacokinetics and pharmacodynamics of drugs are influenced by daily fluctuations in physiological processes. The aim of this study was to determine the effect of dosing time on the pharmacokinetics and brain distribution of morphine. To this end, 4mg/kg morphine was administered intravenously to Wistar rats that were either pre-treated with vehicle or tariquidar and probenecid to inhibit processes involved in the active transport of morphine. Non-linear mixed effects modelling was used to describe the concentration-time profiles of morphine and its metabolite M3G in plasma and brain tissue. We found that the concentrations of morphine in the brain and of M3G in plasma depended on the time of day, which could be quantified by a 24-hour rhythm in the efflux of morphine from brain tissue back into the circulation, with the lowest efflux during the two light-dark phase transitions with a difference between peak and trough of 20%. The active processes involved in the clearance of morphine and its metabolite M3G from plasma also showed 24-hour variation with the highest value in the middle of the dark phase being 54% higher than the lowest value at the start of the light phase. Hence, time of day presents a considerable source of variation in the pharmacokinetics of morphine, which could be used to optimize the dosing strategy of morphine.


Assuntos
Analgésicos Opioides/farmacocinética , Encéfalo/metabolismo , Morfina/farmacocinética , Animais , Masculino , Taxa de Depuração Metabólica/fisiologia , Derivados da Morfina/farmacocinética , Probenecid/administração & dosagem , Quinolinas/administração & dosagem , Ratos , Ratos Wistar
18.
J Mol Histol ; 48(4): 285-292, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28550642

RESUMO

Heroin is an illicit opioid drug which is commonly abused and leads to dependence and addiction. Heroin is considered a pro-drug and is rapidly converted to its major active metabolite 6-monoacetylmorphine (6-MAM) which mediates euphoria and reward through the stimulation of opioid receptors in the brain. The aim of this study was to investigate the distribution and localization of 6-MAM in the healthy Sprague Dawley rat brain following intraperitoneal (i.p) administration of heroin (10 mg/kg), using matrix-assisted laser desorption/ionization mass spectrometric imaging (MALDI-MSI), in combination with quantification via liquid chromatography mass spectrometry (LC-MS/MS). These findings revealed that 6-MAM is present both in plasma and brain tissue with a Tmax of 5 min (2.8 µg/mL) and 15 min (1.1 µg/mL), respectively. MSI analysis of the brain showed high intensities of 6-MAM in the thalamus-hypothalamus and mesocorticolimbic system including areas of the cortex, caudate putamen, and ventral pallidum regions. This finding correlates with the distribution of opioid receptors in the brain, according to literature. In addition, we report a time-dependent distribution in the levels of 6-MAM, from 1 min with the highest intensity of the drug observed at 15 min, with sparse distribution at 45 min before decreasing at 60 min. This is the first study to use MSI as a brain imaging technique to detect a morphine's distribution over time in the brain.


Assuntos
Encéfalo/metabolismo , Heroína/metabolismo , Derivados da Morfina/farmacocinética , Animais , Química Encefálica , Cromatografia Líquida , Heroína/administração & dosagem , Ratos , Ratos Sprague-Dawley , Receptores Opioides , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem , Fatores de Tempo , Distribuição Tecidual
19.
Clin Pharmacokinet ; 56(12): 1577-1587, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28510797

RESUMO

INTRODUCTION: Obesity is associated with many pathophysiological changes that may result in altered drug metabolism. The aim of this study is to investigate the influence of obesity on the pharmacokinetics of morphine, morphine-3-glucuronide (M3G), and morphine-6-glucuronide (M6G) through a combined analysis in morbidly obese patients and non-obese healthy volunteers. METHODS: In this analysis, data from 20 morbidly obese patients [mean body mass index 49.9 kg/m2 (range 37.6-78.6 kg/m2) and weight 151.3 kg (range 112-251.9 kg)] and 20 healthy volunteers [mean weight 70.6 kg (range 58-85 kg)] were included. Morbidly obese patients received 10 mg of intravenous (I.V.) morphine after gastric bypass surgery, with additional morphine I.V. doses as needed. Healthy volunteers received an I.V. bolus of morphine of 0.1 mg/kg followed by an infusion of 0.030 mg kg-1 h-1 for 1 h. Population pharmacokinetic modeling was performed using NONMEM 7.2. RESULTS: In morbidly obese patients, elimination clearance of M3G and M6G was decreased substantially compared with healthy volunteers (p < 0.001). Regarding glucuronidation, only a slight decrease in the formation of M6G and a delay in the formation of M3G was found (both p < 0.001). Obesity was also identified as a covariate for the peripheral volume of distribution of morphine (p < 0.001). CONCLUSION: Metabolism of morphine is not altered in morbidly obese patients. However, decreased elimination of both M3G and M6G is evident, resulting in a substantial increase in exposure to these two metabolites. A rational explanation of this finding is that it results from alterations in membrane transporter function and/or expression in the liver. ClinicalTrials.gov identifier: NCT01097148.


Assuntos
Derivados da Morfina/farmacocinética , Morfina/farmacocinética , Obesidade Mórbida/fisiopatologia , Administração Intravenosa , Adulto , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacocinética , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Morfina/administração & dosagem , Dinâmica não Linear , Estudos Prospectivos , Distribuição Tecidual , Adulto Jovem
20.
J Forensic Sci ; 62(6): 1559-1574, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28105644

RESUMO

Sampling site, technique, and time influence postmortem drug concentrations. In 57 cases, we studied drug concentration differences as follows: subclavian vein-dissection/clamping versus blind stick, femoral vein-dissection/clamping versus blind stick, right cardiac chamber, and popliteal vein-dissection and clamping only. Cases were distributed in group #1 (all cases with both techniques), group #2 (dissection/clamping), and group #3 (blind stick). Sampled drugs were diazepam, methadone, morphine, and their metabolites. To assess PMR, mean concentrations and ratios were calculated for each group. Time-dependent variations of blood concentrations and ratios were also assessed. Results indicate that site, method, and time may influence postmortem distribution interpretation in different ways. Popliteal blood seems less subject to PMR. In conclusion, our study is the first to evaluate concurrently three main aspects of PMR and confirms that the popliteal vein may represent a site that is more resistant to the changes seen as a result of PMR.


Assuntos
Coleta de Amostras Sanguíneas/métodos , Diazepam/sangue , Metadona/sangue , Morfina/sangue , Adulto , Coleta de Amostras Sanguíneas/instrumentação , Cromatografia Líquida , Diazepam/farmacocinética , Feminino , Veia Femoral , Toxicologia Forense , Humanos , Masculino , Metadona/farmacocinética , Pessoa de Meia-Idade , Morfina/farmacocinética , Derivados da Morfina/sangue , Derivados da Morfina/farmacocinética , Nordazepam/sangue , Nordazepam/farmacocinética , Oxazepam/sangue , Oxazepam/farmacocinética , Veia Poplítea , Mudanças Depois da Morte , Veia Subclávia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA