Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.140
Filter
1.
Int J Nanomedicine ; 19: 4759-4777, 2024.
Article in English | MEDLINE | ID: mdl-38828199

ABSTRACT

Background: Opioids are irreplaceable analgesics owing to the lack of alternative analgesics that offer opioid-like pain relief. However, opioids have many undesirable central side effects. Restricting opioids to peripheral opioid receptors could reduce those effects while maintaining analgesia. Methods: To achieve this goal, we developed Tet1-LNP (morphine), a neural-targeting lipid nanoparticle encapsulating morphine that could specifically activate the peripheral opioid receptor in the dorsal root ganglion (DRG) and significantly reduce the side effects caused by the activation of opioid receptors in the brain. Tet1-LNP (morphine) were successfully prepared using the thin-film hydration method. In vitro, Tet1-LNP (morphine) uptake was assessed in differentiated neuron-like PC-12 cells and dorsal root ganglion (DRG) primary cells. The uptake of Tet1-LNP (morphine) in the DRGs and the brain was assessed in vivo. Von Frey filament and Hargreaves tests were used to assess the antinociception of Tet1-LNP (morphine) in the chronic constriction injury (CCI) neuropathic pain model. Morphine concentration in blood and brain were evaluated using ELISA. Results: Tet1-LNP (morphine) had an average size of 131 nm. Tet1-LNP (morphine) showed high cellular uptake and targeted DRG in vitro. CCI mice treated with Tet1-LNP (morphine) experienced prolonged analgesia for nearly 32 h compared with 3 h with free morphine (p < 0.0001). Notably, the brain morphine concentration in the Tet1-LNP (morphine) group was eight-fold lower than that in the morphine group (p < 0.0001). Conclusion: Our study presents a targeted lipid nanoparticle system for peripheral neural delivery of morphine. We anticipate Tet1-LNP (morphine) will offer a safe formulation for chronic neuropathic pain treatment, and promise further development for clinical applications.


Subject(s)
Analgesics, Opioid , Ganglia, Spinal , Morphine , Nanoparticles , Animals , Morphine/administration & dosage , Morphine/pharmacokinetics , Morphine/chemistry , Morphine/pharmacology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Nanoparticles/chemistry , Rats , PC12 Cells , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Male , Neuralgia/drug therapy , Mice , Lipids/chemistry , Proto-Oncogene Proteins/metabolism , Peripheral Nerves/drug effects , Mixed Function Oxygenases/metabolism , DNA-Binding Proteins , Liposomes
2.
Ups J Med Sci ; 1292024.
Article in English | MEDLINE | ID: mdl-38863729

ABSTRACT

Background: Standard dosages of analgesic and sedative drugs are given to intensive care patients. The resulting range of blood concentrations and corresponding clinical responses need to be better examined. The purpose of this study was to describe daily dosages, measured blood concentrations, and clinical responses in critically ill patients. The purpose was also to contribute to establishing whole blood concentration reference values of the drugs investigated. Methods: A descriptive study of prospectively collected data from 302 admissions to a general intensive care unit (ICU) at a university hospital. Ten drugs (clonidine, fentanyl, morphine, dexmedetomidine, ketamine, ketobemidone, midazolam, paracetamol, propofol, and thiopental) were investigated, and daily dosages recorded. Blood samples were collected twice daily, and drug concentrations were measured. Clinical responses were registered using Richmond agitation-sedation scale (RASS) and Numeric rating scale (NRS). Results: Drug dosages were within recommended dose ranges. Blood concentrations for all 10 drugs showed a wide variation within the cohort, but only 3% were above therapeutic interval where clonidine (57 of 122) and midazolam (38 of 122) dominated. RASS and NRS were not correlated to drug concentrations. Conclusion: Using recommended dose intervals for analgesic and sedative drugs in the ICU setting combined with regular monitoring of clinical responses such as RASS and NRS leads to 97% of concentrations being below the upper limit in the therapeutic interval. This study contributes to whole blood drug concentration reference values regarding these 10 drugs.


Subject(s)
Analgesics , Hypnotics and Sedatives , Intensive Care Units , Midazolam , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/pharmacokinetics , Hypnotics and Sedatives/blood , Analgesics/administration & dosage , Analgesics/pharmacokinetics , Analgesics/blood , Male , Female , Middle Aged , Aged , Prospective Studies , Adult , Midazolam/administration & dosage , Midazolam/pharmacokinetics , Midazolam/blood , Critical Care/methods , Dexmedetomidine/administration & dosage , Dexmedetomidine/pharmacokinetics , Dexmedetomidine/blood , Fentanyl/administration & dosage , Fentanyl/blood , Fentanyl/pharmacokinetics , Critical Illness , Propofol/administration & dosage , Propofol/pharmacokinetics , Propofol/blood , Clonidine/administration & dosage , Clonidine/pharmacokinetics , Clonidine/blood , Ketamine/administration & dosage , Ketamine/blood , Ketamine/pharmacokinetics , Morphine/administration & dosage , Morphine/blood , Morphine/pharmacokinetics , Aged, 80 and over , Dose-Response Relationship, Drug , Thiopental/administration & dosage , Thiopental/pharmacokinetics , Acetaminophen/administration & dosage , Acetaminophen/blood , Acetaminophen/pharmacokinetics
3.
Pharmacogenomics J ; 24(3): 18, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824169

ABSTRACT

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.


Subject(s)
Analgesics, Opioid , Cancer Pain , Delayed-Action Preparations , Morphine , Pharmacogenetics , Humans , Morphine/adverse effects , Morphine/pharmacokinetics , Morphine/administration & dosage , Male , Female , Cancer Pain/drug therapy , Cancer Pain/genetics , Middle Aged , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/adverse effects , Analgesics, Opioid/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Aged , Pharmacogenetics/methods , Polymorphism, Single Nucleotide/genetics , Morphine Derivatives/pharmacokinetics , Morphine Derivatives/adverse effects , Adult , Pharmacogenomic Variants , Toll-Like Receptor 2/genetics
4.
Int J Legal Med ; 138(3): 839-847, 2024 May.
Article in English | MEDLINE | ID: mdl-38231204

ABSTRACT

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.


Subject(s)
Burns , Propofol , Humans , Morphine/pharmacokinetics , Palliative Care , Diazepam , Burns/metabolism
5.
Ther Drug Monit ; 45(6): 777-785, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37296504

ABSTRACT

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.


Subject(s)
Chronic Pain , Neoplasms , Humans , Fentanyl/adverse effects , Oxycodone/therapeutic use , Oxycodone/pharmacokinetics , Analgesics, Opioid/adverse effects , Morphine/therapeutic use , Morphine/pharmacokinetics , Chronic Pain/drug therapy , Neoplasms/drug therapy
6.
Eur J Pharm Sci ; 187: 106482, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37247795

ABSTRACT

Morphine blood-brain barrier (BBB) transport is governed by passive diffusion, active efflux and saturable active influx. This may result in nonlinear plasma concentration-dependent brain extracellular fluid (brainECF) pharmacokinetics of morphine. In this study, we aim to evaluate the impact of nonlinear BBB transport on brainECF pharmacokinetics of morphine and its metabolites for different dosing strategies using a physiologically based pharmacokinetic simulation study. We extended the human physiologically based pharmacokinetic LeiCNS-PK3.0, model with equations for nonlinear BBB transport of morphine. Simulations for brainECF pharmacokinetics were performed for various dosing strategies: intravenous (IV), oral immediate (IR) and extended release (ER) with dose range of 0.25-150 mg and dosing frequencies of 1-6 times daily. The impact of nonlinear BBB transport on morphine CNS pharmacokinetics was evaluated by quantifying (i) the relative brainECF to plasma exposure (AUCu,brainECF/AUCu,plasma) and (ii) the impact on the peak-to-trough ratio (PTR) of concentration-time profiles in brainECF and plasma. We found that the relative morphine exposure and PTRs are dose dependent for the evaluated dose range. The highest relative morphine exposure value of 1.4 was found for once daily 0.25 mg ER and lowest of 0.1 for 6-daily 150 mg IV dosing. At lower doses the PTRs were smaller and increased with increasing dose and stabilized at higher doses independent of dosing frequency. Relative peak concentrations of morphine in relation to its metabolites changed with increasing dose. We conclude that nonlinearity of morphine BBB transport affects the relative brainECF exposure and the fluctuation of morphine and its metabolites mainly at lower dosing regimens.


Subject(s)
Blood-Brain Barrier , Morphine , Humans , Morphine/pharmacokinetics , Brain/metabolism , Biological Transport , Computer Simulation
7.
Genes (Basel) ; 13(10)2022 10 11.
Article in English | MEDLINE | ID: mdl-36292717

ABSTRACT

BACKGROUND: The increase in the medical use of cannabis has revealed a number of beneficial effects, a variety of adverse side effects and great inter-individual variability. Association studies connecting consumption, addiction and side effects related to recreational cannabis use have led to the identification of several polymorphic genes that may play a role in the pharmacodynamics and pharmacokinetics of cannabis. METHOD: In total, 600 patients treated with cannabis were genotyped for several candidate polymorphic genes (single-nucleotide polymorphism; SNP), encoding receptors CNR1 and TRPV1; for the ABCB1 transporter; for biotransformation, bioactivation and biosynthesis; and CYP3A4, COMT and UGT2B7 conjugation. RESULTS: Three polymorphic genes (ABCB1, TRPV1 and UGT2B7) were identified as being significantly associated with decline in pain after treatment with cannabis. Patients simultaneously carrying the most favourable allele combinations showed a greater reduction (polygenic effect) in pain compared to those with a less favourable combination. Considering genotype combinations, we could group patients into good responders, intermediate responders and poor or non-responders. Results suggest that genetic makeup is, at the moment, a significant predictive factor of the variability in response to cannabis. CONCLUSIONS: This study proves, for the first time, that certain polymorphic candidate genes may be associated with cannabis effects, both in terms of pain management and side effects, including therapy dropout. SIGNIFICANCE: Our attention to pharmacogenetics began in 2008, with the publication of a first study on the association between genetic polymorphisms and morphine action in pain relief. The study we are presenting is the first observational study conducted on a large number of patients involving several polymorphic candidate genes. The data obtained suggest that genetic makeup can be a predictive factor in the response to cannabis therapy and that more extensive and planned studies are needed for the opening of new scenarios for the personalization of cannabis therapy.


Subject(s)
Cannabis , Chronic Pain , Hallucinogens , Humans , Pharmacogenetics , Chronic Pain/drug therapy , Chronic Pain/genetics , Cannabis/genetics , Cytochrome P-450 CYP3A/genetics , Morphine/pharmacokinetics , Polymorphism, Single Nucleotide
8.
Basic Clin Pharmacol Toxicol ; 130(1): 93-102, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34599645

ABSTRACT

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.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Morphine/pharmacokinetics , Octamer Transcription Factor-1/genetics , Aged , Analgesics, Opioid/administration & dosage , Area Under Curve , Female , Genetic Variation , Genotype , Humans , Male , Middle Aged , Morphine/administration & dosage , Morphine Derivatives/pharmacokinetics , Pain, Postoperative/drug therapy , Time Factors
9.
CPT Pharmacometrics Syst Pharmacol ; 11(1): 82-93, 2022 01.
Article in English | MEDLINE | ID: mdl-34842366

ABSTRACT

Intravenous (i.v.) morphine is a safe, robust, and recommended treatment for severe pain using the titration principle. Despite its high efficacy, it is impacted by organizational constraints related to venous access. Nebulized (NEB) morphine may represent an alternative for titration but pharmacokinetic (PK) properties of short nebulization using routine devices need evaluation. Twenty-seven healthy volunteers were included to receive NEB or i.v. morphine administration using increasing amounts according to Dixon's reference method. Plasma morphine, morphine-3-glucuronide (M3G), and morphine-6-glucuronide (M6G) were quantified. PK modeling and simulations were performed using Monolix. Dixon's method exhibited a significantly higher morphine dose regimen in the NEB group versus the i.v. group (6.2 [5.3-7.1] vs. 3.0 [2.0-4.0] mg, p < 0.001). Morphine, M3G, and M6G dose-normalized exposure were significantly lower in the NEB group versus the i.v. group: morphine (19 [13-23] vs. 1044 [702-1266] µg min/L, p < 0.001), M3G (245 [162-287] vs. 3752 [2487-5165] µg min/L, p < 0.001) and M6G (28 [21-43] vs. 466 [370-723] µg min/L, p < 0.001). The model that best fitted the data consisted in a transit compartment for morphine absorption, three compartments for morphine distribution followed by multiple transit compartments (8.2 and 57.5-min transit time for M3G and M6G, respectively) and a first order elimination for M3G and M6G. Morphine bioavailability in the NEB group was 3.5% using the i.v. group as reference. Administration route and sex significantly influenced morphine and metabolite PKs. This work aimed to evaluate the PKs of NEB morphine compared with the i.v. route. Despite a bioavailability to improve, NEB morphine administration using a routine device is suitable to plan morphine titration.


Subject(s)
Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Morphine Derivatives/metabolism , Morphine/administration & dosage , Morphine/pharmacokinetics , Administration, Inhalation , Adult , Computer Simulation , Dose-Response Relationship, Drug , Female , Healthy Volunteers , Humans , Injections, Intravenous , Male , Middle Aged , Models, Biological , Nebulizers and Vaporizers , Sex Factors
10.
Life Sci ; 283: 119866, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34352257

ABSTRACT

AIMS: Morphine, a commonly used drug for anesthesia, affects lipid metabolism in different tissues, but the mechanism is currently unclear. Adipose triglyceride lipase (ATGL) is the rate-limiting enzyme responsible for the first step of triglyceride (TG) hydrolysis. Here we aim to investigate whether ATGL phosphorylation is involved in morphine-induced TG accumulation. MAIN METHODS: Oil red O staining and TG content analysis were used to detect the effect of morphine on lipid storage. A series of ATGL phosphoamino acid site mutant plasmids were constructed by gene synthesis and transfected to HL-1 cells to evaluate the phosphorylation levels of ATGL phosphoamino acid in morphine-treated HL-1 cells with immunoprecipitation and immunoblotting assay. KEY FINDINGS: Morphine acute treatment induced excessive accumulation of TG and decreased the phosphorylation level of ATGL Ser406 in HL-1 cells. Of note, the phosphorylation positive mutation of ATGL Ser406 to aspartic acid effectively reversed morphine-induced excessive accumulation of TG in HL-1 cells. SIGNIFICANCE: This discovery will help to fully understand the lipid regulation function of morphine in a new scope. In addition, it will expand the phosphorylation research of ATGL more comprehensively and provide powerful clues for lipid metabolism regulation.


Subject(s)
Lipase/metabolism , Morphine/pharmacology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Triglycerides/biosynthesis , Animals , Cell Line , Male , Mice , Morphine/pharmacokinetics , Myocardium/pathology , Myocytes, Cardiac/pathology , Phosphorylation/drug effects
11.
Clin Transl Sci ; 14(6): 2408-2419, 2021 11.
Article in English | MEDLINE | ID: mdl-34268884

ABSTRACT

The aim of the study was to investigate if there is a clinically relevant drug interaction between metformin and codeine. Volunteers were randomized to receive on four separate occasions: (A) orally administered metformin (1 g), (B) intravenously administered metformin (0.5 g), (C) five doses of tablet codeine 25 mg; the last dose was administered together with oral metformin (1 g), and (D) five doses of tablet codeine 25 mg; the last dose was administered together with metformin (0.5 g) intravenously. Blood samples were drawn for 24 h after administration of metformin, and for 6 h after administration of codeine and analyzed using liquid chromatography and tandem mass spectrometry. Healthy volunteers genotyped as CYP2D6 normal metabolizers (*1/*1) without known reduced function variants in the OCT1 gene (rs12208357, rs34130495, rs34059508, and rs72552763) were invited. The median absorption fraction of metformin was 0.31 and was not influenced by codeine intake. The median time to maximum concentration ( T max ) after oral intake of metformin was 2 h without, and 3 h with codeine (p = 0.06). The geometric mean ratios of the areas under the plasma concentration time-curve (AUCs) for morphine and its metabolites M3G and M6G for oral intake of metformin-to-no metformin were 1.21, 1.31, and 1.27, respectively, and for i.v. metformin-to-no metformin 1.28, 1.34, and 1.30, respectively. Concomitant oral and i.v. metformin increased the plasma levels of morphine, M3G and M6G. These small pharmacokinetic changes may well contribute to an increased risk of early discontinuation of metformin. Hence, a clinically relevant drug-drug interaction between metformin and codeine seems plausible.


Subject(s)
Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Codeine/administration & dosage , Healthy Volunteers , Morphine/administration & dosage , Morphine/pharmacokinetics , Adult , Algorithms , Cross-Over Studies , Humans , Young Adult
12.
J Clin Pharmacol ; 61(9): 1149-1155, 2021 09.
Article in English | MEDLINE | ID: mdl-33847389

ABSTRACT

Morphine is an opioid analgesic indicated in the treatment of acute and chronic moderate to severe pain. From a pharmacodynamic standpoint, morphine exerts its effects by agonizing mu-opioid receptors predominantly, resulting in analgesia and sedation. Pharmacokinetically, morphine is primarily metabolized in the liver via glucuronidation by the enzyme uridine diphosphate glucuronosyltransferase family 2 member B7 and encounters the transporter proteins organic cation transporter isoform 1 and P-glycoprotein (adenosine triphosphate-binding cassette subfamily B member 1) as it is being distributed throughout the body. The genes coding for the proteins impacting either the pharmacokinetics or pharmacodynamics of morphine may bear genetic variations, also known as polymorphisms, which may alter the function of the proteins in such a manner that an individual may have disparate treatment outcomes. The purpose of this review is to highlight some of the genes coding for proteins that impact morphine pharmacokinetics and pharmacodynamics and present some treatment considerations.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Pharmacogenetics , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Analgesics, Opioid/pharmacokinetics , Glucuronosyltransferase/genetics , Humans , Morphine/pharmacokinetics , Organic Cation Transporter 1/genetics , Polymorphism, Single Nucleotide , Receptors, Opioid, mu/genetics
13.
PLoS Comput Biol ; 17(3): e1008786, 2021 03.
Article in English | MEDLINE | ID: mdl-33661919

ABSTRACT

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.


Subject(s)
Analgesics, Opioid , Brain/metabolism , Models, Biological , Morphine Derivatives , Morphine , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adult , Age Factors , Analgesia , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/blood , Analgesics, Opioid/pharmacokinetics , Blood-Brain Barrier/metabolism , Child , Child, Preschool , Computational Biology , Female , Humans , Infant, Newborn , Male , Morphine/administration & dosage , Morphine/blood , Morphine/pharmacokinetics , Morphine Derivatives/administration & dosage , Morphine Derivatives/blood , Morphine Derivatives/pharmacokinetics
14.
Eur J Drug Metab Pharmacokinet ; 46(2): 249-263, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33547559

ABSTRACT

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).


Subject(s)
Analgesics, Opioid/pharmacokinetics , Models, Biological , Morphine Derivatives/pharmacokinetics , Morphine/pharmacokinetics , Pain, Postoperative/drug therapy , Adult , Aged , Analgesia, Patient-Controlled/methods , Analgesics, Opioid/administration & dosage , Cardiac Output/physiology , Cardiac Surgical Procedures/methods , Female , Half-Life , Humans , Male , Middle Aged , Morphine/administration & dosage , Tissue Distribution
15.
Arch Dis Child ; 106(10): 1002-1009, 2021 10.
Article in English | MEDLINE | ID: mdl-33461958

ABSTRACT

OBJECTIVE: To systematically review available paediatric literature on comparisons between morphine (Mo) and hydromorphone (Hm), to guide clinicians to rationally use these medications. DESIGN: Systematic review within four databases for all studies published from 1963 to July 2019. SETTING: All paediatric settings. ELIGIBILITY: All studies comparing Mo to Hm in individuals younger than 21 years. MAIN OUTCOME MEASURES: The primary outcome was to compare clinical efficacy and side effects of Mo and Hm. The secondary outcomes were the comparison of pharmacokinetic profiles and the description of predefined Mo to Hm conversion ratios used across the paediatric literature. RESULTS: Among 754 abstracts reviewed, 59 full-text articles met inclusion criteria and 24 studies were included in the analysis: 4 studies compared pharmacodynamics of Mo and Hm and 20 studies reported the use of a predefined Mo to Hm conversion ratio. Most studies had a poor methodological quality. Available evidence suggests that, when given intravenously, the equianalgesic ratio of Mo to Hm is 5:1. Intravenous administration with this ratio results in a similar rate of adverse effects, including pruritus and nausea. The epidural administration with a ratio of 10:1 results in more pruritus and urinary retention with Mo than Hm. Pharmacokinetic data were reported in only one study. A wide range of pre-established ratios for different routes of administration were reported, but few were based on evidence. CONCLUSION: Current literature does not permit a rational choice between Mo and Hm. A ratio of 5:1 seems adequate for intravenous administration and leads to a similar rate of adverse effects.


Subject(s)
Analgesics, Opioid/therapeutic use , Hydromorphone/therapeutic use , Morphine/therapeutic use , Pain/drug therapy , Administration, Intravenous , Adolescent , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacokinetics , Child , Child, Preschool , Humans , Hydromorphone/adverse effects , Hydromorphone/pharmacokinetics , Infant , Infant, Newborn , Morphine/adverse effects , Morphine/pharmacokinetics , Nausea/chemically induced , Pruritus/chemically induced , Urinary Retention/chemically induced
16.
AAPS J ; 23(1): 15, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33404848

ABSTRACT

Guidance regarding the effect of codeine and its metabolites on foetal development is limited by small studies and inconsistent findings. The primary objective was to use physiologically based pharmacokinetic modelling to investigate the impact of gestational stage and maternal CYP2D6 phenotype on foetal morphine exposure following codeine administration. Full body physiologically based pharmacokinetic models were developed and verified for codeine and morphine using Simcyp (version 19.1). The impact of gestational age and maternal CYP2D6 phenotype on foetal and maternal morphine and codeine exposure following oral codeine administration was modelled in a cohort of 250 pregnant females and foetuses at gestational weeks 0 (mothers only), 6, 12, 24 and 36. Consistent with the known effect on codeine metabolism, a clinically meaningful (> 1.65-fold) increase in foetal morphine AUC was observed in the CYP2D6 UM phenotype cohort compared to the CYP2D6 EM and PM phenotype cohorts. The mean (95% CI) foetal morphine AUC in the CYP2D6 UM cohort of 0.988 (0.902 to 1.073) ng/mL.h was 1.8-fold higher than the CYP2D6 EM cohort of 0.546 (0.492 to 0.600) ng/mL.h (p < 0.001). Despite a 2.8-fold increase in maternal CYP2D6 protein abundance between gestational weeks 6 and 36, the mean foetal morphine AUC in the CYP2D6 EM and UM phenotype cohorts reduced by 1.55- and 1.75-fold, respectively, over this period. Maternal CYP2D6 phenotype is a significant determinant of foetal morphine AUC. Simulations suggest that the greatest risk with respect to foetal morphine exposure is during the first trimester of pregnancy, particularly in CYP2D6 UM phenotype mothers.


Subject(s)
Analgesics, Opioid/administration & dosage , Codeine/administration & dosage , Cytochrome P-450 CYP2D6/genetics , Drug Dosage Calculations , Models, Biological , Administration, Oral , Analgesics, Opioid/pharmacokinetics , Area Under Curve , Codeine/pharmacokinetics , Cohort Studies , Computer Simulation , Cytochrome P-450 CYP2D6/metabolism , Female , Fetus/metabolism , Gestational Age , Humans , Low Back Pain/drug therapy , Male , Maternal-Fetal Exchange , Metabolic Clearance Rate , Morphine/administration & dosage , Morphine/pharmacokinetics , Pharmacogenomic Variants , Precision Medicine/methods , Pregnancy , Pregnancy Complications/drug therapy , Pregnancy Trimester, First/metabolism
17.
J Clin Pharmacol ; 61(7): 857-870, 2021 07.
Article in English | MEDLINE | ID: mdl-33382111

ABSTRACT

Chronic intrauterine exposure to psychoactive drugs often results in neonatal opioid withdrawal syndrome (NOWS). When nonpharmacologic measures are insufficient in controlling NOWS, morphine, methadone, and buprenorphine are first-line medications commonly used to treat infants with NOWS because of in utero exposure to opioids. Research suggests that buprenorphine may be the leading drug therapy used to treat NOWS when compared with morphine and methadone. Currently, there are no consensus or standardized treatment guidelines for medications prescribed for NOWS. Opioids used to treat NOWS exhibit large interpatient variability in pharmacokinetics (PK) and pharmacodynamic (PD) response in neonates. Organ systems undergo rapid maturation after birth that may alter drug disposition and exposure for any given dose during development. Data regarding the PK and PD of opioids in neonates are sparse. Pharmacometric methods such as physiologically based pharmacokinetic and population pharmacokinetic modeling can be used to explore factors predictive of some of the variability associated with the PK/PD of opioids in newborns. This review discusses the utility of pharmacometric techniques for enhancing precision dosing in infants requiring opioid treatment for NOWS. Applying these approaches may contribute to optimizing the outcome by reducing cumulative drug exposure, mitigating adverse drug effects, and reducing the burden of NOWS in neonates.


Subject(s)
Narcotics/pharmacokinetics , Narcotics/therapeutic use , Neonatal Abstinence Syndrome/drug therapy , Opiate Substitution Treatment/methods , Opioid-Related Disorders/drug therapy , Buprenorphine/pharmacokinetics , Buprenorphine/therapeutic use , Cytochrome P-450 CYP3A/metabolism , Dose-Response Relationship, Drug , Humans , Infant, Newborn , Methadone/pharmacokinetics , Methadone/therapeutic use , Models, Biological , Morphine/pharmacokinetics , Morphine/therapeutic use , Narcotics/administration & dosage , Narcotics/pharmacology
18.
Anesth Analg ; 132(3): 726-734, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33122543

ABSTRACT

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.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Cardiac Surgical Procedures/adverse effects , Models, Theoretical , Morphine/pharmacokinetics , Pain Management , Pain, Postoperative/drug therapy , Pain, Postoperative/etiology , Aged , Analgesics, Opioid/administration & dosage , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Morphine/administration & dosage , Pain, Postoperative/diagnosis , Risk Assessment , Risk Factors , Treatment Outcome
19.
Curr Opin Support Palliat Care ; 14(3): 177-181, 2020 09.
Article in English | MEDLINE | ID: mdl-32740277

ABSTRACT

PURPOSE OF REVIEW: Clinicians who seek to reduce the symptomatic burden of chronic breathlessness by initiating regular low-dose morphine has the choice of immediate or sustained-release formulations - which will be better for this often frail population, and which has the more robust evidence to inform its prescription? Both formulations can be used. RECENT FINDINGS: For chronic breathlessness, three factors consistently favour the use of regular, low-dose, sustained-release morphine over immediate-release formulations: SUMMARY: As the evidence base expands for the symptomatic reduction of chronic breathlessness, pharmacological interventions will play a part. Using the best available evidence underpins patient-centred approaches that seek to predictably maximize the net effect.As such, the weight of evidence in patient-centred clinical care favours the use of regular, low-dose sustained-release morphine for the symptomatic reduction of chronic breathlessness.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/therapeutic use , Dyspnea/drug therapy , Morphine/pharmacokinetics , Morphine/therapeutic use , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Automobile Driving , Chronic Disease , Delayed-Action Preparations , Dose-Response Relationship, Drug , Drug and Narcotic Control , Dyspnea/physiopathology , Humans , Meta-Analysis as Topic , Morphine/administration & dosage , Morphine/pharmacology , Patient Preference , Randomized Controlled Trials as Topic
20.
Drug Des Devel Ther ; 14: 2307-2317, 2020.
Article in English | MEDLINE | ID: mdl-32606600

ABSTRACT

INTRODUCTION: Transdermal drug delivery is gaining popularity as an alternative to traditional routes of administration. It can increase patient compliance because of its painless and noninvasive nature, aid compounds in bypassing presystemic metabolic effects, and reduce the likelihood of adverse effects through decreased systemic exposure. In silico physiological modeling is critical to predicting dermal exposure for a therapeutic and assessing the impact of different formulations on transdermal disposition. METHODS: The present study aimed at developing a physiologically based transdermal platform, "BIOiSIM", that could be globally applied to a wide variety of compounds to predict their transdermal disposition. The platform integrates a 16-compartment model of compound pharmacokinetics and was used to simulate and predict drug exposure of three chemically and biologically distinct drug-like compounds. Machine learning optimization was composed of two components: exhaustive search algorithm (coarse-tuning) and descent (fine-tuning) integrated with the platform used to quantitatively determine parameters influencing pharmacokinetics (eg permeability, kperm) of test compounds. RESULTS: The model successfully predicted drug exposure (AUC, Cmax and Tmax) following transdermal application of morphine, buprenorphine and nicotine in human subjects, mostly with less than two-fold absolute average fold error (AAFE). The model was further able to successfully characterize the relationship between observed systemic exposure and intended pharmacological effect. The predicted systemic concentration of morphine and plasma levels of endogenous pain biomarkers were used to estimate the effectiveness of a given therapeutic regimen. CONCLUSION: BIOiSIM marks a novel approach to in silico prediction that will enable leveraging of machine learning technology in the pharmaceutical space. The approach to model development outlined results in scalable, accurate models and enables the generation of large parameter/coefficient datasets from in vivo clinical data that can be used in future work to train quantitative structure activity relationship (QSAR) models for predicting likelihood of compound utility as a transdermally administered therapeutic.


Subject(s)
Buprenorphine/metabolism , Models, Biological , Morphine/metabolism , Nicotine/metabolism , Administration, Cutaneous , Buprenorphine/administration & dosage , Buprenorphine/pharmacokinetics , Computer Simulation , Humans , Morphine/administration & dosage , Morphine/pharmacokinetics , Nicotine/administration & dosage , Nicotine/pharmacokinetics , Permeability , Quantitative Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...