Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.141
Filtrar
Mais filtros

Intervalo de ano de publicação
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.
Int J Legal Med ; 138(3): 839-847, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38231204

RESUMO

The evaluation of a morphine concentration in postmortem blood is routine for a forensic toxicologist. We here report three fatal cases where we found high morphine concentrations with 7.96, 4.30, and 5.82 mg/l in femoral blood that have to be estimated as unusually high. All these individuals died due to severe burn injuries and obtained morphine in the context of their palliative care in the last hours of their lives. According to the autopsy results, the cause of death in case 1 was burn disease with burns of about 90% of the body surface area (BSA), case 2 burn trauma, and case 3 burn shock. Besides morphine, propofol, fentanyl, sufentanil, midazolam, diazepam, lorazepam, cefazolin, and rocuronium were detected in femoral blood. The findings fitted well with the detailed clinical documentation. Further evidence of therapeutic concentrations of quetiapine, duloxetine, and melperone could be matched to preexisting medication of the individuals. Physiologically based pharmacokinetic modelling (PBPK) was applied, developed for the intravenous administration of morphine, to find an explanation for the high morphine concentrations in femoral blood. Quantification of morphine in body fluids and tissue was performed to calculate morphine tissue concentration ratios to the morphine concentration in femoral blood. The presented cases show that pharmacokinetic simulations can reflect decreased renal clearance and decreased hepatic metabolism in general. However, this prediction is not sufficient to explain the high morphine concentrations in femoral blood measured here. It can be assumed that burn shock in particular leads to altered pharmacokinetics, namely decreased distribution of morphine.


Assuntos
Queimaduras , Propofol , Humanos , Morfina/farmacocinética , Cuidados Paliativos , Diazepam , Queimaduras/metabolismo
3.
Ther Drug Monit ; 45(6): 777-785, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37296504

RESUMO

BACKGROUND: Interpreting opioid concentrations is challenging because of the lack of reference ranges. Therefore, the authors aimed to propose dose-specific concentration ranges in serum for oxycodone, morphine, and fentanyl in patients with chronic pain, based on concentration measurements from a large number of patients and supported by theoretical pharmacokinetic calculations and previously published concentrations. METHODS: The opioid concentrations in patients undergoing therapeutic drug monitoring (TDM) for various indications (TDM group) and patients with cancer (cancer group) were investigated. Patients were divided based on the daily opioid doses, and the 10th and 90th percentiles of the concentrations in each dose interval were evaluated. In addition, the expected average serum concentrations were calculated for each dose interval based on published pharmacokinetic data, and a targeted literature search for previously reported dose-specific concentrations was performed. RESULTS: The opioid concentrations in 1054 patient samples were included: 1004 in the TDM group and 50 in the cancer group. In total, 607 oxycodone, 246 morphine, and 248 fentanyl samples were evaluated. The authors proposed dose-specific concentration ranges based mainly on 10th-90th percentiles of the concentrations measured in patient samples, whereas the calculated average concentrations and previously published concentrations were used to adjust the ranges. In general, results from calculations and concentrations retrieved from previous literature were within the 10th-90th percentiles of concentrations from patient samples. However, the lowest calculated average concentrations of fentanyl and morphine were below the 10th percentiles of patient samples in all dose groups. CONCLUSIONS: The proposed dose-specific ranges may be useful for interpreting steady-state opioid serum concentrations in clinical and forensic settings.


Assuntos
Dor Crônica , Neoplasias , Humanos , Fentanila/efeitos adversos , Oxicodona/uso terapêutico , Oxicodona/farmacocinética , Analgésicos Opioides/efeitos adversos , Morfina/uso terapêutico , Morfina/farmacocinética , Dor Crônica/tratamento farmacológico , Neoplasias/tratamento farmacológico
4.
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
5.
Anesth Analg ; 132(3): 726-734, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33122543

RESUMO

BACKGROUND: Optimal analgesic treatment following cardiac surgery is crucial for both patient comfort and successful postoperative recovery. While knowledge of both the pharmacokinetics and pharmacodynamics of analgesics is required to predict optimal drug dosing, models quantifying the pharmacodynamics are scarce. Here, we quantify the pharmacodynamics of morphine by modeling the need for rescue morphine to treat unacceptable pain in 118 patients after cardiac surgery. METHODS: The rescue morphine event data were analyzed with repeated time-to-event (RTTE) modeling using NONMEM. Postoperative pain titration protocol consisted of continuous morphine infusions (median duration 20.5 hours) with paracetamol 4 times daily and rescue morphine in case of unacceptable pain (numerical rating scale ≥4). RESULTS: Patients had a median age of 73 years (interquartile range [IQR]: 63-77) and median bodyweight of 80 kg (IQR: 72-90 kg). Most patients (55%) required at least 1 rescue morphine dose. The hazard for rescue morphine following cardiac surgery was found to be significantly influenced by time after surgery, a day/night cycle with a peak at 23:00 (95% confidence interval [CI], 19:35-02:03) each day, and an effect of morphine concentration with 50% hazard reduction at 9.3 ng·mL-1 (95% CI, 6.7-16). CONCLUSIONS: The pharmacodynamics of morphine after cardiac surgery was successfully quantified using RTTE modeling. Future studies can be used to expand the model to better predict morphine's pharmacodynamics on the individual level and to include the pharmacodynamics of other analgesics so that improved postoperative pain treatment protocols can be developed.


Assuntos
Analgésicos Opioides/farmacocinética , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Modelos Teóricos , Morfina/farmacocinética , Manejo da Dor , Dor Pós-Operatória/tratamento farmacológico , Dor Pós-Operatória/etiologia , Idoso , Analgésicos Opioides/administração & dosagem , Esquema de Medicação , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Morfina/administração & dosagem , Dor Pós-Operatória/diagnóstico , Medição de Risco , Fatores de Risco , Resultado do Tratamento
6.
Ther Drug Monit ; 42(5): 778-786, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32427759

RESUMO

BACKGROUND: Pain control in infants is an important clinical concern, with potential long-term adverse neurodevelopmental effects. Intravenous morphine is routinely administered for postoperative pain management; however, its dose-concentration-response relationship in neonates and infants has not been well characterized. Although the current literature provides dosing guidelines for the average infant, it fails to control for the large unexplained variability in morphine clearance and response in individual patients. Bayesian estimation can be used to control for some of this variability. The authors aimed to evaluate morphine pharmacokinetics (PKs) and exposure in critically ill neonates and infants receiving standard-of-care morphine therapy and compare a population-based approach to the model-informed Bayesian techniques. METHODS: The PKs and exposure of morphine and its active metabolites were evaluated in a prospective opportunistic PK study using 221 discarded blood samples from 57 critically ill neonates and infants in the neonatal intensive care unit. Thereafter, a population-based PK model was compared with a Bayesian adaptive control strategy to predict an individual's PK profile and morphine exposure over time. RESULTS: Among the critically ill neonates and infants, morphine clearance showed substantial variability with a 40-fold range (ie, 2.2 to 87.1, mean 23.7 L/h/70 kg). Compared with the observed morphine concentrations, the population-model based predictions had an R of 0.13, whereas the model-based Bayesian predictions had an R of 0.61. CONCLUSIONS: Model-informed Bayesian estimation is a better predictor of morphine exposure than PK models alone in critically ill neonates and infants. A large variability was also identified in morphine clearance. A further study is warranted to elucidate the predictive covariates and precision dosing strategies that use morphine concentration and pain scores as feedbacks.


Assuntos
Morfina/farmacocinética , Morfina/uso terapêutico , Dor/tratamento farmacológico , Teorema de Bayes , Estado Terminal , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Dor/metabolismo , Estudos Prospectivos
7.
Anesth Analg ; 131(3): 876-884, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-31688081

RESUMO

BACKGROUND: Obesity increases susceptibility to chronic pain, increases metabolism, and is associated with obstructive sleep apnea syndrome (OSAS), all which can complicate perioperative pain management of patients. In addition, obesity and OSAS can cause elevation of the adipose-derived hormone leptin, which increases metabolism. We hypothesized that obesity along with sleep apnea and leptin independently enhance morphine pharmacokinetics. METHODS: Children 5-12 years of age who were presenting for surgery were administered a morphine dose of 0.05 mg/kg. Blood was collected at baseline and at subsequent preset times for pharmacokinetic analysis of morphine and its metabolites. Three groups were studied: a nonobese group with severe OSAS, an obese group with severe OSAS, and a control group. RESULTS: Thirty-four patients consisting of controls (n = 16), nonobese/OSAS (n = 8), and obese/OSAS (n = 10) underwent analysis. The obese/OSAS group had a higher dose-adjusted mean maximum morphine concentration (CMAX) over 540 minutes compared to the controls (P < .001) and those with only OSAS (P = .014). The obese/OSAS group also had lower volume of distribution (Vd) when compared to OSAS-only patients (P = .007). In addition, those in the obese/OSAS group had a higher morphine 3-glucuronide (M3G) maximum concentration (P = .012) and a higher ratio of M3G to morphine than did the control group (P = .011). Time to maximum morphine 6-glucuronide (M6G) concentration was significantly lower in both nonobese/OSAS and obese/OSAS groups than in the control group (P < .005). C-reactive protein (CRP), interleukin (IL)-10, and leptin were all higher in the obese/OSAS group than in controls (P = .004, 0.026, and <0.001, respectively), and compared to OSAS-only patients, CRP (P = .013) and leptin (P = .002) levels were higher in the obese/OSAS group. CONCLUSIONS: The combination of obesity and OSAS was associated with an increase in morphine metabolism compared with that in normal-weight controls. Our previous study in mice demonstrated that obesity from leptin deficiency decreased morphine metabolism, but that metabolism normalized after leptin replacement. Leptin may be a cause of the increased morphine metabolism observed in obese patients.


Assuntos
Analgésicos Opioides/farmacocinética , Morfina/farmacocinética , Obesidade Infantil/complicações , Apneia Obstrutiva do Sono/complicações , Procedimentos Cirúrgicos Operatórios , Fatores Etários , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/sangue , Biomarcadores/sangue , Biotransformação , Criança , Pré-Escolar , Cálculos da Dosagem de Medicamento , Feminino , Humanos , Leptina/sangue , Masculino , Modelos Biológicos , Morfina/administração & dosagem , Morfina/sangue , Obesidade Infantil/sangue , Obesidade Infantil/diagnóstico , Apneia Obstrutiva do Sono/sangue , Apneia Obstrutiva do Sono/diagnóstico
8.
Can J Physiol Pharmacol ; 98(11): 771-776, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32516551

RESUMO

Since both morphine and tadalafil have been proven to exert some of their analgesic activity through modulation of the NO-cGMP pathway, the aim of the current study is to evaluate the pharmacologic interaction between tadalafil and morphine to decrease the dose of morphine and subsequently its side effects. The assessment was carried out through isobolographic analysis relative to ED50s of both morphine and tadalafil obtained by tail-flick test on BALB/c mice. Morphine and tadalafil ED50s calculated from the dose-response curves were 8303 and 2080 µg/kg, respectively. The experimental ED50 values of morphine and tadalafil in their mixture were 4800 and 1210 µg/kg, respectively. Those results showed an additive interaction between morphine and tadalafil presented by a total fraction value for the mixture of 1160 µg/kg. This outcome can be interpreted by the fact that both drugs share common pathways, namely, NO-cGMP and opioid receptors. As a conclusion, the morphine and tadalafil combination showed an additive effect against acute pain, which is mediated through the central nervous system, thus providing a rationale for combining them to decrease morphine dose and thus minimizing its side effects.


Assuntos
Analgesia/métodos , Morfina/farmacocinética , Dor/tratamento farmacológico , Tadalafila/farmacocinética , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada/métodos , Humanos , Injeções Intraperitoneais , Masculino , Camundongos , Morfina/administração & dosagem , Dor/diagnóstico , Medição da Dor , Tadalafila/administração & dosagem
9.
J Vet Pharmacol Ther ; 43(2): 153-161, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31856324

RESUMO

The purpose of this study was to evaluate the pharmacokinetics of morphine in combination with dexmedetomidine and maropitant injected intramuscularly in dogs under general anaesthesia. Eight healthy dogs weighing 25.76 ± 3.16 kg and 3.87 ± 1.64 years of age were used in a crossover study. Dogs were randomly allocated to four groups: (1) morphine 0.6 mg/kg; (2) morphine 0.3 mg/kg + dexmedetomidine 5 µg/kg; (3) morphine 0.3 mg/kg + maropitant 1 mg/kg; (4) morphine 0.2 mg/kg + dexmedetomidine 3 µg/kg + maropitant 0.7 mg/kg. Blood samples were collected before, 15 and 30 min, and 1, 2, 3 4, 6 and 8 hr after injection of the test drugs. Plasma concentration of the drugs was determined by liquid chromatography-mass spectrometry. The elimination half-life (T1/2 ) of morphine was higher and the clearance rate (CL) was lower when combined with dexmedetomidine (T1/2  = 77.72 ± 20.27 min, CL = 119.41 ± 23.34 ml kg-1  min-1 ) compared to maropitant (T1/2  = 52.73 min ± 13.823 ml kg-1  min-1 , CL = 178.57 ± 70.55) or morphine alone at higher doses (T1/2  = 50.53 ± 12.55 min, CL = 187.24 ± 34.45 ml kg-1  min-1 ). Combining morphine with dexmedetomidine may increase the dosing interval of morphine and may have a clinical advantage.


Assuntos
Dexmedetomidina/farmacocinética , Cães/sangue , Halotano/farmacologia , Morfina/farmacocinética , Quinuclidinas/farmacocinética , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/sangue , Analgésicos Opioides/farmacocinética , Anestésicos Inalatórios/farmacologia , Animais , Antieméticos/administração & dosagem , Antieméticos/sangue , Antieméticos/farmacocinética , Área Sob a Curva , Estudos Cross-Over , Dexmedetomidina/administração & dosagem , Quimioterapia Combinada , Meia-Vida , Hipnóticos e Sedativos/administração & dosagem , Hipnóticos e Sedativos/sangue , Hipnóticos e Sedativos/farmacocinética , Injeções Intramusculares , Morfina/administração & dosagem , Quinuclidinas/administração & dosagem
10.
Crit Care Med ; 47(6): e485-e494, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30920410

RESUMO

OBJECTIVE: To develop a pharmacokinetic-pharmacogenomic population model of morphine in critically ill children with acute respiratory failure. DESIGN: Prospective pharmacokinetic-pharmacogenomic observational study. SETTING: Thirteen PICUs across the United States. PATIENTS: Pediatric subjects (n = 66) mechanically ventilated for acute respiratory failure, weight greater than or equal to 7 kg, receiving morphine and/or midazolam continuous infusions. INTERVENTIONS: Serial blood sampling for drug quantification and a single blood collection for genomic evaluation. MEASUREMENTS AND MAIN RESULTS: Concentrations of morphine, the two main metabolites, morphine-3-glucuronide and morphine-6-glucuronide, were quantified by high-performance liquid chromatography tandem mass spectrometry/mass spectroscopy. Subjects were genotyped using the Illumina HumanOmniExpress genome-wide single nucleotide polymorphism chip. Nonlinear mixed-effects modeling was performed to develop the pharmacokinetic-pharmacogenomic model. A two-compartment model with linear elimination and two individual compartments for metabolites best describe morphine disposition in this population. Our analysis demonstrates that body weight and postmenstrual age are relevant predictors of pharmacokinetic parameters of morphine and its metabolites. Furthermore, our research shows that a duration of mechanical ventilation greater than or equal to 10 days reduces metabolite formation and elimination upwards of 30%. However, due to the small sample size and relative heterogeneity of the population, no heritable factors associated with uridine diphosphate glucuronyl transferase 2B7 metabolism of morphine were identified. CONCLUSIONS: The results provide a better understanding of the disposition of morphine and its metabolites in critically ill children with acute respiratory failure requiring mechanical ventilation due to nonheritable factors. It also provides the groundwork for developing additional studies to investigate the role of heritable factors.


Assuntos
Analgésicos Opioides/sangue , Analgésicos Opioides/farmacocinética , Morfina/sangue , Morfina/farmacocinética , Respiração Artificial , Insuficiência Respiratória/terapia , Doença Aguda , Adolescente , Fatores Etários , Analgésicos Opioides/administração & dosagem , Peso Corporal , Criança , Pré-Escolar , Estado Terminal , Feminino , Genótipo , Glucuronosiltransferase/genética , Humanos , Lactente , Masculino , Morfina/administração & dosagem , Derivados da Morfina/sangue , Testes Farmacogenômicos , Estudos Prospectivos , Fatores de Tempo
11.
Br J Clin Pharmacol ; 85(1): 258-262, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30421550

RESUMO

ADVERSE EVENT: Repeated and prolonged episodes of central apnoea and hypoxia after receiving intravenous morphine for analgesia and ketamine for sedation. DRUG IMPLICATED: Intravenous morphine sulfate. THE PATIENT: Previously healthy 12-year-old male with no history of sleep apnoea who presented with distal tibia and fibula fracture. EVIDENCE THAT LINKS DRUG TO EVENT: Pharmacogenomic testing revealed that the patient was homozygous for the T allele at the rs887829 SNP in UGT1A1, an enzyme involved in the metabolism of morphine. This polymorphism is a loss-of-function variant, leading to impaired metabolism of morphine. MECHANISM: Morphine is metabolized by UDP-glucuronosyltransferase (UGT)-2B7 and UGT1A1 to form its major metabolites morphine-3-glucuronide (M3G) and morphine-6-glucuronide (M6G). Our patient was a poor metabolizer through UGT1A1, likely leading to increased respiratory depression as morphine has greater respiratory depressant effects compared to its metabolites. IMPLICATIONS: When appropriate, physicians should enquire about prior receipt of opioids, in both the patient and family, to be better prepared for potential adverse reactions. In the patient with excessive sedation or respiratory depression to standard doses of morphine, genetic testing may be warranted, especially if there is a family or past history that supports a metabolic defect in morphine metabolism and/or excretion.


Assuntos
Analgésicos Opioides/efeitos adversos , Apneia/induzido quimicamente , Morfina/efeitos adversos , Dor/tratamento farmacológico , Administração Intravenosa , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacocinética , Criança , Fíbula/lesões , Fraturas Múltiplas/complicações , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Humanos , Mutação com Perda de Função , Masculino , Morfina/administração & dosagem , Morfina/farmacocinética , Dor/etiologia , Testes Farmacogenômicos , Polimorfismo de Nucleotídeo Único , Fraturas da Tíbia/complicações
12.
J Pharmacokinet Pharmacodyn ; 46(6): 513-529, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31396799

RESUMO

The primary goal of this work was to develop a computational tool to enable personalized prediction of pharmacological disposition and associated responses for opioids and antidotes. Here we present a computational framework for physiologically-based pharmacokinetic (PBPK) modeling of an opioid (morphine) and an antidote (naloxone). At present, the model is solely personalized according to an individual's mass. These PK models are integrated with a minimal pharmacodynamic model of respiratory depression induction (associated with opioid administration) and reversal (associated with antidote administration). The model was developed and validated on human data for IV administration of morphine and naloxone. The model can be further extended to consider different routes of administration, as well as to study different combinations of opioid receptor agonists and antagonists. This work provides the framework for a tool that could be used in model-based management of pain, pharmacological treatment of opioid addiction, appropriate use of antidotes for opioid overdose and evaluation of abuse deterrent formulations.


Assuntos
Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/farmacocinética , Antídotos/efeitos adversos , Antídotos/farmacocinética , Analgésicos Opioides/administração & dosagem , Antídotos/administração & dosagem , Humanos , Masculino , Morfina/efeitos adversos , Morfina/farmacocinética , Naloxona/administração & dosagem , Naloxona/efeitos adversos , Naloxona/farmacocinética , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/efeitos adversos , Antagonistas de Entorpecentes/farmacocinética , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Dor/tratamento farmacológico , Receptores Opioides/metabolismo
13.
J Vet Pharmacol Ther ; 42(4): 401-410, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30919469

RESUMO

The objective of the current study was to describe and characterize the pharmacokinetics and selected pharmacodynamic effects of morphine and its two major metabolites in horses following several doses of morphine. A total of ten horses were administered a single intravenous dose of morphine: 0.05, 0.1, 0.2, or 0.5 mg/kg, or saline control. Blood samples were collected up to 72 hr, analyzed for morphine, and metabolites by LC/MS/MS, and pharmacokinetic parameters were determined. Step count, heart rate and rhythm, gastrointestinal borborygmi, fecal output, packed cell volume, and total protein were also assessed. Morphine-3 glucuronide (M3G) was the predominant metabolite detected, with concentrations exceeding those of morphine-6 glucuronide (M6G) at all time points. Maximal concentrations of M3G and M6G ranged from 55.1 to 504 and 6.2 to 28.4 ng/ml, respectively, across dose groups. The initial assessment of morphine pharmacokinetics was done using noncompartmental analysis (NCA). The volume of distribution at steady-state and systemic clearance ranged from 9.40 to 16.9 L/kg and 23.3 to 32.4 ml min-1  kg-1 , respectively. Adverse effects included signs of decreased gastrointestinal motility and increased central nervous excitation. There was a correlation between increasing doses of morphine, increases in M3G concentrations, and adverse effects. Findings from this study support direct administration of purified M3G and M6G to horses to better characterize the pharmacokinetics of morphine and its metabolites and to assess pharmacodynamic activity of these metabolites.


Assuntos
Analgésicos Opioides/farmacocinética , Cavalos/sangue , Derivados da Morfina/urina , Morfina/farmacocinética , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/urina , Animais , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Injeções Intravenosas , Masculino , Morfina/administração & dosagem , Morfina/urina
14.
Vet Anaesth Analg ; 46(4): 476-482, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31178413

RESUMO

OBJECTIVE: To determine if the combinations of morphine, dexmedetomidine and maropitant enhance the analgesic effect and decrease the dose of individual drugs in rats subjected to noxious thermal stimulation with hot-plate and tail-flick tests. STUDY DESIGN: Randomized, blinded, prospective experimental study. ANIMALS: A total of 96 male Sprague-Dawley rats. METHODS: The rats were randomly assigned to the following groups: 1) morphine (3 mg kg-1; Mor); 2) dexmedetomidine (10 µg kg-1; Dex); 3) maropitant (20 mg kg-1; Maro); 4) morphine (1.5 mg kg-1) + dexmedetomidine (5 µg kg-1; Mor + Dex); 5) dexmedetomidine (5 µg kg-1) + maropitant (10 mg kg-1; Dex + Maro); 6) morphine (1.5 mg kg-1) + maropitant (10 mg kg-1; Mor + Maro); 7) morphine (1 mg kg-1) + dexmedetomidine (3.5 µg kg-1) + maropitant (6.5 mg kg-1; Mor + Dex + Maro); and 8) normal saline (0.5 mL; saline), all injected intravenously. The tail-flick and hot-plate tests were performed before and 5, 15, 30, 45, 60, 90 and 120 minutes after the injection of the drugs. These variables were analysed with the effect-time area under the curve (AUC) analysis and a mixed linear model. RESULTS: Data were analysed in 94 rats. The rank order of the total analgesic effects of the treatment groups shown by AUC analysis was found to be Mor > Maro + Mor > Dex + Mor > Dex > Maro > Dex + Maro + Mor > Dex + Maro > saline for the hot-plate test, and Maro + Mor > Mor > Dex + Mor > Dex + Maro + Mor > Maro > Dex > Dex + Maro > saline for the tail-flick test. The mixed model analysis showed a significant difference between latencies of the group morphine + maropitant versus all other treatment groups in the tail-flick test (p < 0.0001) and morphine versus saline in the hot-plate test (p < 0.05). CONCLUSIONS AND CLINICAL RELEVANCE: Morphine and maropitant appeared to show a supra-additive effect for analgesia in the tail-flick test. Clinical trials should be conducted to establish its use in treating pain.


Assuntos
Dexmedetomidina/farmacologia , Morfina/farmacologia , Medição da Dor/veterinária , Dor/tratamento farmacológico , Quinuclidinas/farmacologia , Analgésicos não Narcóticos/administração & dosagem , Analgésicos não Narcóticos/farmacologia , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacologia , Animais , Antieméticos/administração & dosagem , Antieméticos/farmacologia , Dexmedetomidina/administração & dosagem , Dexmedetomidina/farmacocinética , Sinergismo Farmacológico , Quimioterapia Combinada , Masculino , Morfina/administração & dosagem , Morfina/farmacocinética , Quinuclidinas/administração & dosagem , Quinuclidinas/farmacocinética , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
15.
Drug Metab Dispos ; 46(7): 943-952, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29695616

RESUMO

To predict the impact of liver cirrhosis on hepatic drug clearance using physiologically based pharmacokinetic (PBPK) modeling, we compared the protein abundance of various phase 1 and phase 2 drug-metabolizing enzymes (DMEs) in S9 fractions of alcoholic (n = 27) or hepatitis C (HCV, n = 30) cirrhotic versus noncirrhotic (control) livers (n = 25). The S9 total protein content was significantly lower in alcoholic or HCV cirrhotic versus control livers (i.e., 38.3 ± 8.3, 32.3 ± 12.8, vs. 51.1 ± 20.7 mg/g liver, respectively). In general, alcoholic cirrhosis was associated with a larger decrease in the DME abundance than HCV cirrhosis; however, only the abundance of UGT1A4, alcohol dehydrogenase (ADH)1A, and ADH1B was significantly lower in alcoholic versus HCV cirrhotic livers. When normalized to per gram of tissue, the abundance of nine DMEs (UGT1A6, UGT1A4, CYP3A4, UGT2B7, CYP1A2, ADH1A, ADH1B, aldehyde oxidase (AOX)1, and carboxylesterase (CES)1) in alcoholic cirrhosis and five DMEs (UGT1A6, UGT1A4, CYP3A4, UGT2B7, and CYP1A2) in HCV cirrhosis was <25% of that in control livers. The abundance of most DMEs in cirrhotic livers was 25% to 50% of control livers. CES2 abundance was not affected by cirrhosis. Integration of UGT2B7 abundance in cirrhotic livers into the liver cirrhosis (Child Pugh C) model of Simcyp improved the prediction of zidovudine and morphine PK in subjects with Child Pugh C liver cirrhosis. These data demonstrate that protein abundance data, combined with PBPK modeling and simulation, can be a powerful tool to predict drug disposition in special populations.


Assuntos
Hepatite C/metabolismo , Inativação Metabólica/fisiologia , Cirrose Hepática Alcoólica/metabolismo , Cirrose Hepática/metabolismo , Fígado/metabolismo , Adulto , Idoso , Álcool Desidrogenase/metabolismo , Alcoólicos , Carboxilesterase/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Morfina/farmacocinética , Proteômica/métodos , Adulto Jovem , Zidovudina/farmacocinética
16.
Int J Legal Med ; 132(2): 415-424, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29196879

RESUMO

In forensic medicine, expert opinion is often required concerning dose and time of intake of a substance, especially in the context of fatal intoxications. In the present case, a 98-year-old man died 4 days after admission to a hospital due to a femur neck fracture following a domestic fall in his retirement home. As he had obtained high morphine doses in the context of palliative therapy and a confusion of his supplemental magnesium tablets with a diuretic by the care retirement home was suspected by the relatives, a comprehensive postmortem examination was performed. Forensic toxicological GC- and LC-MS analyses revealed, besides propofol, ketamine, and a metamizole metabolite in blood and urine, toxic blood morphine concentrations of approximately 3 mg/l in femoral and 5 mg/l in heart blood as well as 2, 7, and 10 mg/kg morphine in brain, liver, and lung, respectively. A physiologically based pharmacokinetic (PBPK) model was developed and applied to examine whether the morphine concentrations were (i) in agreement with the morphine doses documented in the clinical records or (ii) due to an excessive morphine administration. PBPK model simulations argue against an overdosing of morphine. The immediate cause of death was respiratory and cardiovascular failure due to pneumonia following a fall, femur neck fracture, and immobilization accompanied by a high and probably toxic concentration of morphine, attributable to the administration under palliative care conditions. The presented case indicates that PBPK modeling can be a useful tool in forensic medicine, especially in question of a possible drug overdosing.


Assuntos
Analgésicos Opioides/farmacocinética , Modelos Biológicos , Morfina/farmacocinética , Acidentes por Quedas , Idoso de 80 Anos ou mais , Analgésicos Opioides/análise , Química Encefálica , Cromatografia Líquida , Fraturas do Colo Femoral , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Fígado/química , Pulmão/química , Masculino , Morfina/análise , Cuidados Paliativos , Pneumonia
17.
Palliat Med ; 32(2): 554-558, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28631529

RESUMO

BACKGROUND: Children and infants with impaired swallow or compromised enteral absorption require alternative routes for administration of analgesia. Recent clinical guidance and practice for paediatric palliative care teams, who often treat such children, supports buccal morphine sulphate as a fast acting, effective and easily administered agent for pain relief. However, a consideration of the physicochemical properties and potency of morphine would suggest that it is not a suitable candidate for delivery via the transmucosal route, raising questions about its use in children and infants. AIM: To explore the permeability of buccal morphine sulphate in an established ex vivo porcine buccal mucosa as a necessary step in examining efficacy for use in children with life-limiting conditions and life-threatening illnesses. DESIGN: A permeation study conducted with morphine sulphate in an ex vivo porcine buccal tissue model. Flux values and pharmacokinetic data were used to calculate the plasma values of morphine that would result following buccal administration in a 20kg child. RESULTS: Results show that the estimated steady state plasma values of morphine sulphate following buccal administration in this model do not achieve minimum therapeutic concentration. CONCLUSION: These data strongly suggest that morphine sulphate is not suitable for buccal administration and that further research is needed to establish its efficacy in relief of pain in children with life-limiting conditions and life-threatening illnesses.


Assuntos
Administração Bucal , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacocinética , Morfina/administração & dosagem , Morfina/farmacocinética , Manejo da Dor/métodos , Doente Terminal , Animais , Criança , Transtornos de Deglutição , Humanos , Modelos Animais , Permeabilidade , Suínos
18.
Palliat Med ; 32(7): 1222-1232, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29724154

RESUMO

BACKGROUND: Morphine can cause central nervous system side effects which impair driving skills. The legal blood morphine concentration limit for driving is 20 µg/L in France/Poland/Netherlands and 80 µg/L in England/Wales. There is no guidance as to the morphine dose leading to this concentration. AIM: The in silico (computed) relationship of oral morphine dose and plasma concentration was modelled to provide dose estimates for a morphine plasma concentration above 20 and 80 µg/L in different patient groups. DESIGN: A dose-concentration model for different genders, ages and oral morphine formulations, validated against clinical pharmacokinetic data, was generated using Simcyp®, a population-based pharmacokinetic simulator. SETTING/PARTICIPANTS: Healthy Northern European population parameters were used with age, gender and renal function being varied in the different simulation groups. In total, 36,000 simulated human subjects (100 per modelled group of different ages and gender) received repeated simulated morphine dosing with modified-release or immediate-release formulations. RESULTS: Older age, women, modified-release formulation and worse renal function were associated with higher plasma concentrations. Across all groups, morphine doses below 20 mg/day were unlikely to result in a morphine plasma concentration above 20 µg/L; this was 80 mg/day with the 80 µg/L limit. CONCLUSION: This novel study provides predictions of the in silico (computed) dose-concentration relationship for international application. Individualised morphine prescribing decisions by clinicians must be informed by clinical judgement considering the individual patient's level of impairment and insight irrespective of the blood morphine concentration as people who have impaired driving will be breaking the law. Taking into account expected morphine concentrations enables improved individualised decision making.


Assuntos
Analgésicos Opioides/farmacocinética , Condução de Veículo/legislação & jurisprudência , Morfina/farmacocinética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Analgésicos Opioides/sangue , Europa (Continente) , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Morfina/sangue , Adulto Jovem
19.
Pediatr Crit Care Med ; 19(5): 459-467, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29547456

RESUMO

OBJECTIVES: To assess if morphine pharmacokinetics are different in children with Down syndrome when compared with children without Down syndrome. DESIGN: Prospective single-center study including subjects with Down syndrome undergoing cardiac surgery (neonate to 18 yr old) matched by age and cardiac lesion with non-Down syndrome controls. Subjects were placed on a postoperative morphine infusion that was adjusted as clinically necessary, and blood was sampled to measure morphine and its metabolites concentrations. Morphine bolus dosing was used as needed, and total dose was tracked. Infusions were continued for 24 hours or until patients were extubated, whichever came first. Postinfusion, blood samples were continued for 24 hours for further evaluation of kinetics. If patients continued to require opioid, a nonmorphine alternative was used. Morphine concentrations were determined using a unique validated liquid chromatography tandem-mass spectrometry assay using dried blood spotting as opposed to large whole blood samples. Morphine concentration versus time data was modeled using population pharmacokinetics. SETTING: A 16-bed cardiac ICU at an university-affiliated hospital. PATIENTS: Forty-two patients (20 Down syndrome, 22 controls) were enrolled. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: The pharmacokinetics of morphine in pediatric patients with and without Down syndrome following cardiac surgery were analyzed. No significant difference was found in the patient characteristics or variables assessed including morphine total dose or time on infusion. Time mechanically ventilated was longer in children with Down syndrome, and regarding morphine pharmacokinetics, the covariates analyzed were age, weight, presence of Down syndrome, and gender. Only age was found to be significant. CONCLUSIONS: This study did not detect a significant difference in morphine pharmacokinetics between Down syndrome and non-Down syndrome children with congenital heart disease.


Assuntos
Analgésicos Opioides/farmacocinética , Procedimentos Cirúrgicos Cardíacos , Síndrome de Down/complicações , Cardiopatias Congênitas/cirurgia , Morfina/farmacocinética , Dor Pós-Operatória/tratamento farmacológico , Adolescente , Analgésicos Opioides/sangue , Analgésicos Opioides/uso terapêutico , Estudos de Casos e Controles , Criança , Pré-Escolar , Feminino , Seguimentos , Cardiopatias Congênitas/complicações , Humanos , Lactente , Recém-Nascido , Infusões Intravenosas , Masculino , Morfina/sangue , Morfina/uso terapêutico , Estudos Prospectivos
20.
Paediatr Anaesth ; 28(11): 1015-1021, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30303265

RESUMO

BACKGROUND: Transition from the intrauterine to the extrauterine environment in neonates is associated with major changes in blood flow and oxygenation with consequent increases in metabolic functions. The additional impact of birth on renal function and drug metabolism above that predicted by postmenstrual age and allometry is uncertain. Increased clearance at birth could reduce analgesic effect attributable to a lowering of plasma concentration. These elimination processes can be described using the clearance concept. METHODS: Data from four publications that investigated the time course of glomerular filtration rate and clearance of paracetamol, morphine and tramadol were reanalyzed. The effect of birth, based on postnatal age, was used in conjunction with a theory-based allometric size scaling and maturation based on postmenstrual age. RESULTS: Postnatal age had a short-term effect on the time course of clearance distinguishable from the well-known slower maturation based on postmenstrual age. While elimination might be relatively reduced by 15%-45% at birth, there is a rapid increase in elimination for 1-3 weeks after birth to be 15% greater than that predicted by postmenstrual age alone. CONCLUSION: Birth is associated with a small increase in clearance in addition to that described by postmenstrual age for common analgesic drugs cleared by glucuronide conjugation (morphine, paracetamol) or by the P450 cytochrome oxidase (tramadol) and renal systems. While the increase is of biological interest, it would not be expected to have any clinically relevant impact on renal function or drug dosing. The processes of maturation described by these models are potentially applicable to any drug elimination process.


Assuntos
Acetaminofen/farmacocinética , Recém-Nascido/metabolismo , Rim/metabolismo , Morfina/farmacocinética , Parto/metabolismo , Tramadol/farmacocinética , Acetaminofen/urina , Taxa de Filtração Glomerular , Humanos , Recém-Nascido Prematuro/metabolismo , Taxa de Depuração Metabólica , Modelos Biológicos , Morfina/urina , Tramadol/urina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA