Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
PLoS One ; 15(11): e0242169, 2020.
Article in English | MEDLINE | ID: mdl-33180816

ABSTRACT

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


Subject(s)
Hyperalgesia/physiopathology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nociception/drug effects , Pain/physiopathology , Hot Temperature , Humans , Infusions, Intravenous , Male , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Young Adult
2.
Haemophilia ; 26(1): 164-172, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31797491

ABSTRACT

INTRODUCTION: Monitoring of clinical effectiveness of bypassing agents in haemophilia patients is hampered by the lack of validated laboratory assays. Thromboelastography (TEG) and rotational thromboelastometry (ROTEM) have been evaluated for predicting clinical effectiveness of bypassing agents, however, with limited success. AIM: Application of a longitudinal model-based approach may allow for a quantitative characterization of the link between ROTEM parameters and the probability of bleeding events. METHODS: We analyse longitudinal data from haemophilia A rats receiving gene-based FVIIa prophylaxis in terms of total circulatory levels of FVII/FVIIa, clotting time (CT) measured using ROTEM and the probability of bleeding events. RESULTS: Using population pharmacokinetic-pharmacodynamic (PKPD) modelling, a PK-CT-repeated time-to-event (RTTE) model was developed composed of three submodels (a) a FVII/FVIIa PK model, (b) a PK-CT model describing the relationship between predicted FVIIa expression and CT and (c) a RTTE model describing the probability of bleeding events as a function of CT. The developed PK-CT-RTTE model accurately described the vector dose-dependent plasma concentration-time profile of total FVII/FVIIa and the exposure-response relationship between AAV-derived FVIIa expression and CT. Importantly, the developed model accurately described the occurrence of bleeding events over time in a quantitative manner, revealing a linear relationship between predicted change from baseline CT and the probability of bleeding events. CONCLUSION: Using PK-CT-RTTE modelling, we demonstrated that ROTEM parameters can accurately predict the probability of bleeding events in a translational animal model of haemophilia A.


Subject(s)
Factor VII/genetics , Hemophilia A/genetics , Hemophilia A/prevention & control , Hemorrhage/diagnosis , Probability , Rotation , Thrombelastography , Translational Research, Biomedical , Animals , Disease Models, Animal , Factor VII/pharmacokinetics , Hemophilia A/blood , Rats , Whole Blood Coagulation Time
3.
J Thromb Haemost ; 17(6): 964-974, 2019 06.
Article in English | MEDLINE | ID: mdl-30924607

ABSTRACT

Essentials Knowledge of the interplay between FVIII and VWF pharmacokinetics (PK) is lacking. We characterized the capacity-limited PK of FVIII and VWF. The PK model described the PK of FVIII and VWF over a broad range of rFVIII doses. High-dose rFVIII treatment can reduce the endogenous VWF levels. BACKGROUND: Understanding of the pharmacokinetics (PK) interplay between factor VIII (FVIII) and von Willebrand factor (VWF) following high-dose FVIII treatment is lacking. OBJECTIVES: To characterize the PK of recombinant FVIII (rFVIII), VWF, and the rFVIII:VWF complex in hemophilia A rats following intravenous administration of rFVIII using PK modeling. A second aim was to investigate the effect of high daily dosing and constant expression of rFVIII on VWF exposure using PK simulations. METHODS: We developed a population PK model based on the principles of target-mediated drug disposition modeling, using data on total rFVIII and VWF plasma concentrations, and the rFVIII:VWF complex luminescent oxygen channeling immunoassay signal in hemophilia A rats following intravenous administration of rFVIII (17.5, 100, 1000, and 5000 IU kg-1 ). Additionally, we evaluated the influence of high-dose rFVIII treatment on the exposure of VWF using PK simulations. RESULTS: The plasma concentration-time profiles of total rFVIII and VWF, and the luminescent oxygen channeling immunoassay signal-time profiles of the rFVIII:VWF complex were adequately described using a two-compartment quasi-steady-state target-mediated drug disposition model (Kss  = 0.14 nmol L-1 ). The elimination half-life of the rFVIII:VWF complex was dependent on the unbound plasma concentration of rFVIII. Additionally, we showed that high-dose rFVIII treatment may significantly reduce the endogenous VWF levels. CONCLUSIONS: We developed a population-based PK model describing the time-course of total rFVIII, total VWF, and the rFVIII:VWF complex over a broad range of rFVIII doses in hemophilia A rats.


Subject(s)
Factor VIII/pharmacokinetics , Hemophilia A/blood , Hemophilia A/drug therapy , von Willebrand Factor/pharmacokinetics , Administration, Intravenous , Animals , Disease Models, Animal , Factor VIII/administration & dosage , Female , Humans , Male , Models, Biological , Nonlinear Dynamics , Rats , Recombinant Proteins/administration & dosage , Recombinant Proteins/blood , Recombinant Proteins/pharmacokinetics , von Willebrand Factor/administration & dosage , von Willebrand Factor/metabolism
4.
Eur J Pharm Sci ; 123: 531-538, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-30077714

ABSTRACT

Historically, clinical trials of haemophilia with inhibitors (HwI) have been challenged by the small patient population. New approaches to clinical trial methodology and statistical modelling could potentially be used for study optimization. The aim of this work was to evaluate the impact of different trial designs and study conditions on the estimated drug potency and power, and compare traditional statistical methods with repeated time-to-event (RTTE) modelling in terms of power. Bleeding information from a clinical trial of 23 haemophilia patients with inhibitors treated on-demand was used to develop a baseline RTTE model using NONMEM. Clinical trial simulations for a hypothetical anti-haemophilic drug were performed, by adding a drug effect and a literature-derived placebo effect to the baseline RTTE model, using different trial designs (parallel-group, placebo-controlled parallel-group, crossover and placebo-controlled crossover designs) and study conditions, including sample size, study duration and doses. The precision and accuracy of the estimated drug potency (EC50) and power for different trial designs, study conditions and statistical methods (RTTE modelling, t-test and negative binomial regression) were evaluated. The developed baseline RTTE model accurately described the clinical data. The crossover designs displayed up to four-fold higher precision of the estimated EC50 and three-fold higher power relative to the parallel-group trial designs. Furthermore, RTTE modelling provided a higher power relative to the traditional statistical tests. We found that crossover designs in combination with RTTE modelling can reduce the required sample size and study duration, while ensuring high power and precise estimation of EC50, in clinical trials of HwI.


Subject(s)
Antibodies/blood , Clinical Trials as Topic/methods , Factor VIII/administration & dosage , Hemophilia A/drug therapy , Hemostatics/administration & dosage , Research Design , Adolescent , Adult , Antibodies/immunology , Clinical Trials as Topic/statistics & numerical data , Computer Simulation , Cross-Over Studies , Data Interpretation, Statistical , Factor VIII/adverse effects , Factor VIII/immunology , Hemophilia A/blood , Hemophilia A/diagnosis , Hemophilia A/immunology , Hemostatics/adverse effects , Hemostatics/immunology , Humans , Male , Middle Aged , Recombinant Proteins/administration & dosage , Research Design/statistics & numerical data , Sample Size , Time Factors , Treatment Outcome , Young Adult
5.
Eur J Pharm Sci ; 115: 196-203, 2018 Mar 30.
Article in English | MEDLINE | ID: mdl-29369801

ABSTRACT

Various experimental animal models are used in haemophilia research, however, little is known about how well the different species predict pharmacokinetic (PK) profiles in haemophilia patients. The aim of the current study was to describe the plasma concentration-time profile of recombinant activated factor VII (rFVIIa) and recombinant factor VIII (rFVIII) in several experimental animal models using population PK modelling, and apply a simulation-based approach to evaluate how well the developed animal population PK models predict human PK. PK models were developed for rFVIIa and rFVIII in mice, rats, monkeys, and dogs using nonlinear mixed-effects modelling, accounting for inter-individual variability, nonlinear kinetics and covariate effects. Three scaling principles were applied to predict human PK: proportional scaling to body weight from single species, scaling with fixed theory-based allometric exponents from single species, and allometric interspecies scaling with estimated allometric coefficients and exponents. The plasma concentration-time profile of rFVIIa and rFVIII in mice, rats, monkeys and dogs were accurately described by the developed species-specific PK models, accounting for nonlinear kinetics and gender-specific difference in clearance for rFVIII. The predictive performance of the animal population PK models of rFVIIa and rFVIII revealed significant species-variation. The developed PK models of rFVIIa and rFVIII in monkeys and dogs along with allometric interspecies scaling revealed high predictive performance for human PK, and may promote rational decision-making in future first-in-human trials for rFVIIa and rFVIII variants.


Subject(s)
Factor VIII/pharmacokinetics , Factor VIIa/pharmacokinetics , Hemophilia A/metabolism , Animals , Area Under Curve , Dogs , Female , Haplorhini , Humans , Male , Metabolic Clearance Rate/drug effects , Mice , Models, Animal , Rats , Recombinant Proteins/pharmacokinetics , Species Specificity
6.
AAPS J ; 19(6): 1804-1813, 2017 11.
Article in English | MEDLINE | ID: mdl-28853055

ABSTRACT

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


Subject(s)
Amines/pharmacology , Analgesics/pharmacology , Cyclohexanecarboxylic Acids/pharmacology , Morphine/pharmacology , gamma-Aminobutyric Acid/pharmacology , Animals , Dose-Response Relationship, Drug , Drug Interactions , Gabapentin , Pain, Postoperative/drug therapy , Rats , Rats, Sprague-Dawley
7.
J Pharmacokinet Pharmacodyn ; 44(4): 325-333, 2017 08.
Article in English | MEDLINE | ID: mdl-28389762

ABSTRACT

Inconsistent trial design and analysis is a key reason that few advances in postoperative pain management have been made from clinical trials analyzing opioid consumption data. This study aimed to compare four different approaches to analyze opioid consumption data. A repeated time-to-event (RTTE) model in NONMEM was used to simulate clinical trials of morphine consumption with and without a hypothetical adjuvant analgesic in doses equivalent to 15-62% reduction in morphine consumption. Trials were simulated with duration of 24-96 h. Monte Carlo simulation and re-estimation were performed to determine sample size required to demonstrate efficacy with 80% power using t test, Mann-Whitney rank sum test, time-to-event (TTE) modeling and RTTE modeling. Precision of efficacy estimates for RTTE models were evaluated in 500 simulations. A sample size of 50 patients was required to detect 37% morphine sparing effect with at least 80% power in a 24 h trial with RTTE modeling whereas the required sample size was 200 for Mann-Whitney, 180 for t-test and 76 for TTE models. Extending the trial duration from 24 to 96 h reduced the required sample size by 3.1 fold with RTTE modeling. Precise estimate of potency was obtained with a RTTE model accounting for both morphine effects and time-varying covariates on opioid consumption. An RTTE analysis approach proved better suited for demonstrating efficacy of opioid sparing analgesics than traditional statistical tests as a lower sample size was required due the ability to account for time-varying factors including PK.


Subject(s)
Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Clinical Trials as Topic/methods , Computer Simulation , Clinical Trials as Topic/statistics & numerical data , Computer Simulation/statistics & numerical data , Dose-Response Relationship, Drug , Humans , Morphine/administration & dosage , Morphine/pharmacokinetics , Sample Size , Time Factors
8.
Basic Clin Pharmacol Toxicol ; 120(1): 71-78, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27430990

ABSTRACT

The aim was to investigate systemic exposure after administration of a novel bupivacaine lozenge in healthy individuals with normal mucosa and in head and neck cancer (HNC) patients with oral mucositis. A lozenge containing 5, 10, 25 and 50 mg bupivacaine, respectively, was administered as single dose to 10 healthy individuals, and a lozenge containing 25 mg bupivacaine was administered as single dose to 10 HNC patients with oral mucositis and as multiple doses to five patients with HNC. Blood samples were collected for 6 hr from the healthy individuals and 3 hr from the patients with HNC, respectively, after administration. The plasma concentration-time profiles of bupivacaine were fitted to pharmacokinetic models using nonlinear mixed-effects modelling, evaluating demographics and health status as covariates. The population pharmacokinetics (PK) of bupivacaine lozenge was best described by a two-compartment distribution model with absorption transit compartments. All the observed plasma concentrations were well below the bupivacaine concentrations (2000-2250 ng/ml) which have caused toxic symptoms. The PK model suggested that relative bioavailability was two times higher in HNC patients with oral mucositis grade 1-2 and three times higher in HNC patients with oral mucositis grade 3-4 than in the healthy individuals. Simulations showed that the plasma concentrations would be below the toxic limit after repeated dosing every second hour with 25 mg bupivacaine for five days. The 25-mg bupivacaine lozenges were safe without systemic toxic levels of bupivacaine or risk of side effects. Based on PK simulations of repeated doses of 25 mg every two hours for 16 hr a day, the lozenges can be administered with minimum risk of exceeding the toxic limit.


Subject(s)
Anesthetics, Local/pharmacokinetics , Bupivacaine/pharmacokinetics , Models, Biological , Mouth Mucosa/drug effects , Mucositis/drug therapy , Oral Mucosal Absorption , Pain/prevention & control , Administration, Mucosal , Adult , Anesthetics, Local/administration & dosage , Anesthetics, Local/adverse effects , Anesthetics, Local/therapeutic use , Biological Availability , Bupivacaine/administration & dosage , Bupivacaine/adverse effects , Bupivacaine/therapeutic use , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Head and Neck Neoplasms/radiotherapy , Humans , Intestinal Absorption , Male , Metabolic Clearance Rate , Mouth Mucosa/metabolism , Mouth Mucosa/radiation effects , Mucositis/blood , Mucositis/metabolism , Mucositis/physiopathology , Pain/etiology , Radiation Injuries/drug therapy , Radiation Injuries/metabolism , Radiation Injuries/physiopathology , Severity of Illness Index
9.
Eur J Pharm Sci ; 92: 117-30, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27373670

ABSTRACT

Morphine is commonly used for pain management in preterm neonates. The aims of this study were to compare published models of neonatal pharmacokinetics of morphine and its metabolites with a new dataset, and to combine the characteristics of the best predictive models to design a meta-model for morphine and its metabolites in preterm neonates. Moreover, the concentration-analgesia relationship for morphine in this clinical setting was also investigated. A population of 30 preterm neonates (gestational age: 23-32weeks) received a loading dose of morphine (50-100µg/kg), followed by a continuous infusion (5-10µg/kg/h) until analgesia was no longer required. Pain was assessed using the Premature Infant Pain Profile. Five published population models were compared using numerical and graphical tests of goodness-of-fit and predictive performance. Population modelling was conducted using NONMEM® and the $PRIOR subroutine to describe the time-course of plasma concentrations of morphine, morphine-3-glucuronide, and morphine-6-glucuronide, and the concentration-analgesia relationship for morphine. No published model adequately described morphine concentrations in this new dataset. Previously published population pharmacokinetic models of morphine, morphine-3-glucuronide, and morphine-6-glucuronide were combined into a meta-model. The meta-model provided an adequate description of the time-course of morphine and the concentrations of its metabolites in preterm neonates. Allometric weight scaling was applied to all clearance and volume terms. Maturation of morphine clearance was described as a function of postmenstrual age, while maturation of metabolite elimination was described as a function of postnatal age. A clear relationship between morphine concentrations and pain score was not established.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Models, Biological , Morphine/pharmacokinetics , Analgesics, Opioid/blood , Analgesics, Opioid/therapeutic use , Female , Humans , Infant, Newborn , Infant, Premature , Male , Morphine/blood , Morphine/therapeutic use , Pain/blood , Pain/drug therapy
10.
Pharm Res ; 33(11): 2630-43, 2016 11.
Article in English | MEDLINE | ID: mdl-27380190

ABSTRACT

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


Subject(s)
Amines/chemistry , Amines/pharmacokinetics , Analgesics/chemistry , Cyclohexanecarboxylic Acids/chemistry , Cyclohexanecarboxylic Acids/pharmacokinetics , Models, Biological , Morphine/chemistry , Morphine/pharmacokinetics , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/pharmacokinetics , Amines/metabolism , Analgesics/metabolism , Analgesics/pharmacokinetics , Animals , Cyclohexanecarboxylic Acids/metabolism , Drug Interactions , Enterohepatic Circulation , Gabapentin , Glucuronides/metabolism , Morphine/metabolism , Nonlinear Dynamics , Rats, Sprague-Dawley , Tandem Mass Spectrometry , gamma-Aminobutyric Acid/metabolism
11.
AAPS J ; 18(4): 1013-22, 2016 07.
Article in English | MEDLINE | ID: mdl-27116023

ABSTRACT

Joint analysis of pain intensity and opioid consumption is encouraged in trials of postoperative pain. However, previous approaches have not appropriately addressed the complexity of their interrelation in time. In this study, we applied a non-linear mixed effects model to simultaneously study pain intensity and opioid consumption in a 4-h postoperative period for 44 patients undergoing percutaneous kidney stone surgery. Analysis was based on 748 Numerical Rating Scale (NRS) scores of pain intensity and 51 observed morphine and oxycodone dosing events. A joint model was developed to describe the recurrent pattern of four key phases determining the development of pain intensity and opioid consumption in time; (A) Distribution of pain intensity scores which followed a truncated Poisson distribution with time-dependent mean score ranging from 0.93 to 2.45; (B) Probability of transition to threshold pain levels (NRS ≥ 3) which was strongly dependent on previous pain levels ranging from 2.8-15.2% after NRS of 0-2; (C) Probability of requesting opioid when allowed (NRS ≥ 3) which was strongly correlated with the number of previous doses, ranging from 89.8% for requesting the first dose to 26.1% after three previous doses; (D) Reduction in pain scores after opioid dosing which was significantly related to the pain intensity at time of opioid request (P < 0.001). This study highlights the importance of analyzing pain intensity and opioid consumption in an integrated manner. Non-linear mixed effects modeling proved a valuable tool for analysis of interventions that affect pain intensity, probability of rescue dosing or the effect of opioids in the postoperative pain period.


Subject(s)
Analgesics, Opioid/administration & dosage , Pain, Postoperative/drug therapy , Humans , Morphine/administration & dosage , Oxycodone/administration & dosage , Pain Measurement
12.
Eur J Pharm Sci ; 86: 41-9, 2016 Apr 30.
Article in English | MEDLINE | ID: mdl-26946441

ABSTRACT

BACKGROUND: Polymorphisms in the opioid receptor genes may affect the pharmacodynamics (PD) of oxycodone and be part of the reason behind the diversity in clinical response. The aim of the analysis was to model the exposure-response profile of oxycodone for three different pain variables and search for genetic covariates. Model simulations were used to predict how population and effect-size impact the power to detect clinical significant SNPs. METHOD: The population pharmacokinetic-pharmacodynamic (PKPD) model of oral single-dosed oxycodone was based on pooled data from three published studies in healthy volunteers. Pain tolerance data from muscle pressure (n=36), visceral pressure (n=54) and skin pinch (n=34) were included. Genetic associations with 18 opioid-receptor SNPs were explored using a stepwise covariate approach. Model simulations were performed using the estimated model parameters. RESULTS: None of the selected SNPs were associated with analgesic response of oxycodone at P<0.001. Baseline response in muscle cuff pressure was associated with OPRK1 rs7016778 and rs7824175 (P<0.001). Simulations indicated that large differences in drug response between genotypes (>50% for similar population sizes) or large populations (n>200 for a 20% response difference) are necessary to identify clinical significant SNP effects due to high population variability. CONCLUSION: A population PKPD model has been developed for oral oxycodone using three different pain variables to explore impact of genetic covariates and study design. None of the selected polymorphisms were significantly associated with analgesic response of oxycodone, but an association of baseline response in muscle cuff pressure with two OPRK1 SNPs was identified.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Models, Biological , Oxycodone/pharmacokinetics , Pain/metabolism , Receptors, Opioid/genetics , Administration, Oral , Adult , Analgesics, Opioid/therapeutic use , Double-Blind Method , Female , Humans , Male , Middle Aged , Oxycodone/therapeutic use , Pain/drug therapy , Pain/genetics , Polymorphism, Single Nucleotide , Young Adult
13.
Eur J Pharm Sci ; 86: 29-33, 2016 Apr 30.
Article in English | MEDLINE | ID: mdl-26946443

ABSTRACT

The kidneys are thought to play an important role in the clearance of recombinant human growth hormone (rhGH), but the relative importance is not clear. Obtaining knowledge of clearance pathway is an important prerequisite for the development of new long acting growth hormone analogues targeted at treatment of patients with growth hormone disorders. The purpose of this study was to investigate the relative importance of the kidneys in the clearance of rhGH. The study employed a newly validated nephrectomy rat model and a population based pharmacokinetic approach to assess renal clearance of rhGH in non-anesthetized rats, anesthetized rats and in nephrectomized anesthetized rats. Clearance in non-anesthetized rats was 290 ml/h/kg. This was reduced to 185 ml/h/kg by anesthesia and further reduced to 18 ml/h/kg by nephrectomy. As nephrectomy was able to reduce clearance with 90%, we conclude that renal clearance plays a pivotal role in the elimination of rhGH in rats.


Subject(s)
Human Growth Hormone/pharmacokinetics , Kidney/metabolism , Animals , Human Growth Hormone/blood , Male , Nephrectomy , Rats, Sprague-Dawley
14.
Pharm Res ; 33(5): 1133-43, 2016 May.
Article in English | MEDLINE | ID: mdl-26780452

ABSTRACT

PURPOSE: Gabapentin displays non-linear drug disposition, which complicates dosing for optimal therapeutic effect. Thus, the current study was performed to elucidate the pharmacokinetic/pharmacodynamic (PKPD) relationship of gabapentin's effect on mechanical hypersensitivity in a rat model of CFA-induced inflammatory hyperalgesia. METHODS: A semi-mechanistic population-based PKPD model was developed using nonlinear mixed-effects modelling, based on gabapentin plasma and brain extracellular fluid (ECF) time-concentration data and measurements of CFA-evoked mechanical hyperalgesia following administration of a range of gabapentin doses (oral and intravenous). RESULTS: The plasma/brain ECF concentration-time profiles of gabapentin were adequately described with a two-compartment plasma model with saturable intestinal absorption rate (K m = 44.1 mg/kg, V max = 41.9 mg/h∙kg) and dose-dependent oral bioavailability linked to brain ECF concentration through a transit compartment. Brain ECF concentration was directly linked to a sigmoid E max function describing reversal of hyperalgesia (EC 50, plasma = 16.7 µg/mL, EC 50, brain = 3.3 µg/mL). CONCLUSIONS: The proposed semi-mechanistic population-based PKPD model provides further knowledge into the understanding of gabapentin's non-linear pharmacokinetics and the link between plasma/brain disposition and anti-hyperalgesic effects. The model suggests that intestinal absorption is the primary source of non-linearity and that the investigated rat model provides reasonable predictions of clinically effective plasma concentrations for gabapentin.


Subject(s)
Amines/pharmacology , Amines/pharmacokinetics , Analgesics/pharmacology , Analgesics/pharmacokinetics , Cyclohexanecarboxylic Acids/pharmacology , Cyclohexanecarboxylic Acids/pharmacokinetics , Hyperalgesia/drug therapy , gamma-Aminobutyric Acid/pharmacology , gamma-Aminobutyric Acid/pharmacokinetics , Amines/administration & dosage , Amines/therapeutic use , Analgesics/administration & dosage , Analgesics/therapeutic use , Animals , Biological Availability , Brain/metabolism , Computer Simulation , Cyclohexanecarboxylic Acids/administration & dosage , Cyclohexanecarboxylic Acids/therapeutic use , Gabapentin , Hyperalgesia/metabolism , Male , Models, Biological , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/administration & dosage , gamma-Aminobutyric Acid/therapeutic use
15.
Pharm Res ; 33(5): 1093-103, 2016 May.
Article in English | MEDLINE | ID: mdl-26753622

ABSTRACT

PURPOSE: To characterize the pharmacokinetic-pharmacodynamic (PK-PD) relationship between exposure of morphine and subsequent morphine consumption and to develop simulation tools for model validation. METHODS: Dose, formulation and time of morphine administration was available from a published study in 63 patients receiving intravenous, oral immediate release or oral controlled release morphine on request after hip surgery. The PK-PD relationship between predicted exposure of morphine and morphine consumption was modeled using repeated time to event (RTTE) modeling in NONMEM. To validate the RTTE model, a visual predictive check method was developed with simulated morphine consumption given the exposure of preceding morphine administration. RESULTS: The probability of requesting morphine was found to be significantly related to the exposure of morphine as well as night/day. Oral controlled release morphine was more effective than intravenous and oral immediate release formulations at equivalent average concentrations. Maximum effect was obtained for 8 h by oral controlled release doses ≥ 15 mg, where probability of requesting a new dose was reduced to 20% for a typical patient. CONCLUSION: This study demonstrates the first quantitative link between exposure of morphine and subsequent morphine consumption and introduces an efficient visual predictive check approach with simulation of adaptive dosing.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/therapeutic use , Morphine/pharmacokinetics , Morphine/therapeutic use , Pain, Postoperative/drug therapy , Administration, Intravenous , Administration, Oral , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Computer Simulation , Humans , Models, Biological , Morphine/administration & dosage , Morphine/pharmacology
16.
Eur J Pharm Sci ; 82: 97-105, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26610393

ABSTRACT

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


Subject(s)
Amines/administration & dosage , Analgesics/administration & dosage , Cyclohexanecarboxylic Acids/administration & dosage , Hyperalgesia/drug therapy , Morphine/administration & dosage , Pain, Postoperative/drug therapy , gamma-Aminobutyric Acid/administration & dosage , Amines/pharmacokinetics , Amines/therapeutic use , Analgesics/pharmacokinetics , Analgesics/therapeutic use , Animals , Cyclohexanecarboxylic Acids/pharmacokinetics , Cyclohexanecarboxylic Acids/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Gabapentin , Male , Morphine/pharmacokinetics , Morphine/therapeutic use , Physical Stimulation , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/pharmacokinetics , gamma-Aminobutyric Acid/therapeutic use
17.
Anesthesiology ; 123(6): 1411-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26495978

ABSTRACT

BACKGROUND: Reduction in consumption of opioid rescue medication is often used as an endpoint when investigating analgesic efficacy of drugs by adjunct treatment, but appropriate methods are needed to analyze analgesic consumption in time. Repeated time-to-event (RTTE) modeling is proposed as a way to describe analgesic consumption by analyzing the timing of consecutive analgesic events. METHODS: Retrospective data were obtained from 63 patients receiving standard analgesic treatment including morphine on request after surgery following hip fracture. Times of analgesic events up to 96 h after surgery were extracted from hospital medical records. Parametric RTTE analysis was performed with exponential, Weibull, or Gompertz distribution of analgesic events using NONMEM, version 7.2 (ICON Development Solutions, USA). The potential influences of night versus day, sex, and age were investigated on the probability. RESULTS: A Gompertz distribution RTTE model described the data well. The probability of having one or more analgesic events within 24 h was 80% for the first event, 55% for the second event, 31% for the third event, and 18% for fourth or more events for a typical woman of age 80 yr. The probability of analgesic events decreased in time, was reduced to 50% after 3.3 days after surgery, and was significantly lower (32%) during night compared with day. CONCLUSIONS: RTTE modeling described analgesic consumption data well and could account for time-dependent changes in probability of analgesic events. Thus, RTTE modeling of analgesic events is proposed as a valuable tool when investigating new approaches to pain management such as opioid-sparing analgesia.


Subject(s)
Analgesia/statistics & numerical data , Analgesics, Opioid/therapeutic use , Hip Fractures/surgery , Pain, Postoperative/drug therapy , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Morphine/therapeutic use , Periodicity , Retrospective Studies , Sex Factors , Time Factors , Young Adult
18.
Antimicrob Agents Chemother ; 59(11): 7018-26, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26349823

ABSTRACT

Antibiotic dosing in septic shock patients poses a challenge for clinicians due to the pharmacokinetic (PK) variability seen in this patient population. Piperacillin-tazobactam is often used for empirical treatment, and initial appropriate dosing is crucial for reducing mortality. Accordingly, we determined the pharmacokinetic profile of piperacillin (4 g) every 8 h, during the third consecutive dosing interval, in 15 patients treated empirically for septic shock. We developed a population pharmacokinetic model to assess empirical dosing and to simulate alternative dosing regimens and modes of administration. Time above the MIC (T>MIC) predicted for each patient was evaluated against clinical breakpoint MIC for Pseudomonas aeruginosa (16 mg/liter). Pharmacokinetic-pharmacodynamic (PK/PD) targets evaluated were 50% fT>4×MIC and 100% fT>MIC. A population PK model was developed using NONMEM, and data were best described by a two-compartment model. Central and intercompartmental clearances were 3.6 liters/h (relative standard error [RSE], 15.7%) and 6.58 liters/h (RSE, 16.4%), respectively, and central and peripheral volumes were 7.3 liters (RSE, 11.8%) and 3.9 liters (RSE, 9.7%), respectively. Piperacillin plasma concentrations varied considerably between patients and were associated with levels of plasma creatinine. Patients with impaired renal function were more likely to achieve predefined PK/PD targets than were patients with preserved or augmented renal function. Simulations of alternative dosing regimens showed that frequent intermittent bolus dosing as well as dosing by extended and continuous infusion increases the probability of attaining therapeutic plasma concentrations. For septic shock patients with preserved or augmented renal function, dose increment or prolonged infusion of the drug needs to be considered. (This study has been registered at ClinicalTrials.gov under registration no. NCT02306928.).


Subject(s)
Anti-Bacterial Agents/blood , Anti-Bacterial Agents/pharmacokinetics , Piperacillin/blood , Piperacillin/pharmacokinetics , Shock, Septic/blood , Shock, Septic/drug therapy , Aged , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Female , Humans , Male , Microbial Sensitivity Tests , Middle Aged , Monte Carlo Method , Penicillanic Acid/analogs & derivatives , Penicillanic Acid/blood , Penicillanic Acid/pharmacokinetics , Penicillanic Acid/pharmacology , Penicillanic Acid/therapeutic use , Piperacillin/pharmacology , Piperacillin/therapeutic use , Prospective Studies , Pseudomonas aeruginosa/drug effects , Tazobactam
19.
Eur J Pharm Sci ; 74: 45-62, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-25861720

ABSTRACT

Morphine is a widely used opioid for treatment of moderate to severe pain, but large interindividual variability in patient response and no clear guidance on how to optimise morphine dosage regimen complicates treatment strategy for clinicians. Population pharmacokinetic-pharmacodynamic models can be used to quantify dose-response relationships for the population as well as interindividual and interoccasion variability. Additionally, relevant covariates for population subgroups that deviate from the typical population can be identified and help clinicians in dose optimisation. This review provides a detailed overview of the published human population pharmacokinetic-pharmacodynamic studies for morphine analgesia in addition to basic drug disposition and pharmacological properties of morphine and its analgesic active metabolite, morphine-6-glucuronide, that may help identify future covariates. Furthermore, based on simulations from key pharmacokinetic-pharmacodynamic models, the contribution of morphine-6-glucuronide to the analgesic response in patients with renal insufficiency was investigated. Simulations were also used to examine the impact of effect-site equilibration half-life on time course of response. Lastly, the impact of study design on the likelihood of determining accurate pharmacodynamic parameters for morphine response was evaluated.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Models, Biological , Morphine Derivatives/pharmacokinetics , Morphine/pharmacokinetics , Precision Medicine , Acute Pain/complications , Acute Pain/drug therapy , Acute Pain/metabolism , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Animals , Biological Availability , Biotransformation , Half-Life , Humans , Morphine/adverse effects , Morphine/pharmacology , Morphine/therapeutic use , Morphine Derivatives/adverse effects , Morphine Derivatives/pharmacology , Morphine Derivatives/therapeutic use , Renal Insufficiency/complications , Tissue Distribution
20.
Antimicrob Agents Chemother ; 59(4): 2398-404, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25666151

ABSTRACT

When antimicrobials are used empirically, pathogen MICs equal to clinical breakpoints or epidemiological cutoff values must be considered. This is to ensure that the most resistant pathogen subpopulation is appropriately targeted to prevent emergence of resistance. Accordingly, we determined the pharmacokinetic (PK) profile of moxifloxacin at 400 mg/day in 18 patients treated empirically for community-acquired pneumonia. We developed a population pharmacokinetic model to assess the potential efficacy of moxifloxacin and to simulate the maximal MICs for which recommended pharmacokinetic-pharmacodynamic (PK-PD) estimates are obtained. Moxifloxacin plasma concentrations were determined the day after therapy initiation using ultra-high-performance liquid chromatography. Peak drug concentrations (Cmax) and area under the free drug concentration-time curve from 0 to 24 h (fAUC0-24) values predicted for each patient were evaluated against epidemiological cutoff MIC values for Streptococcus pneumoniae, Haemophilus influenzae, and Legionella pneumophila. PK-PD targets adopted were a Cmax/MIC of ≥12.2 for all pathogens, an fAUC0-24/MIC of >34 for S. pneumoniae, and an fAUC0-24/MIC of >75 for H. influenzae and L. pneumophila. Individual predicted estimates for Cmax/MIC and fAUC0-24/MIC as well as simulated maximal MICs resulting in target attainment for oral and intravenous administration of the drug were suitable for S. pneumoniae and H. influenzae but not for L. pneumophila. These results indicate that caution must be taken when moxifloxacin is used as monotherapy to treat community-acquired pneumonia caused by L. pneumophila. In conclusion, this report reveals key information relevant to the empirical treatment of community-acquired pneumonia while highlighting the robust and flexible nature of this population pharmacokinetic model to predict therapeutic success. (Clinical Trials Registration no. NCT01983839.).


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/therapeutic use , Community-Acquired Infections/drug therapy , Community-Acquired Infections/metabolism , Fluoroquinolones/pharmacokinetics , Fluoroquinolones/therapeutic use , Pneumonia/drug therapy , Pneumonia/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Area Under Curve , Chromatography, High Pressure Liquid , Community-Acquired Infections/microbiology , Computer Simulation , Female , Humans , Male , Microbial Sensitivity Tests , Middle Aged , Models, Statistical , Moxifloxacin , Pneumonia/microbiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...