Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
CPT Pharmacometrics Syst Pharmacol ; 13(5): 812-822, 2024 05.
Article in English | MEDLINE | ID: mdl-38436514

ABSTRACT

Item response theory (IRT) models are usually the best way to analyze composite or rating scale data. Standard methods to evaluate covariate or treatment effects in IRT models do not allow to identify item-specific effects. Finding subgroups of patients who respond differently to certain items could be very important when designing inclusion or exclusion criteria for clinical trials, and aid in understanding different treatment responses in varying disease manifestations. We present a new method to investigate item-specific effects in IRT models, which is based on inspection of residuals. The method was investigated in a simulation exercise with a model for the Epworth Sleepiness Scale. We also provide a detailed discussion as a guidance on how to build a robust covariate IRT model.


Subject(s)
Models, Statistical , Humans , Computer Simulation
2.
CPT Pharmacometrics Syst Pharmacol ; 13(5): 880-890, 2024 05.
Article in English | MEDLINE | ID: mdl-38468601

ABSTRACT

Obstructive sleep apnea (OSA) is a sleep disorder which is linked to many health risks. The gold standard to evaluate OSA in clinical trials is the Apnea-Hypopnea Index (AHI). However, it is time-consuming, costly, and disregards aspects such as quality of life. Therefore, it is of interest to use patient-reported outcomes like the Epworth Sleepiness Scale (ESS), which measures daytime sleepiness, as surrogate end points. We investigate the link between AHI and ESS, via item response theory (IRT) modeling. Through the developed IRT model it was identified that AHI and ESS are not correlated to any high degree and probably not measuring the same sleepiness construct. No covariate relationships of clinical relevance were found. This suggests that ESS is a poor choice as an end point for clinical development if treatment is targeted at improving AHI, and especially so in a mild OSA patient group.


Subject(s)
Sleep Apnea, Obstructive , Humans , Sleep Apnea, Obstructive/diagnosis , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/physiopathology , Male , Female , Middle Aged , Sleepiness , Quality of Life , Patient Reported Outcome Measures , Severity of Illness Index , Disorders of Excessive Somnolence/diagnosis , Adult , Aged
3.
J Am Coll Cardiol ; 83(6): 669-678, 2024 02 13.
Article in English | MEDLINE | ID: mdl-38325992

ABSTRACT

BACKGROUND: Phase II trials of asundexian were underpowered to detect important differences in bleeding. OBJECTIVES: The goal of this study was to obtain best estimates of effects of asundexian vs active control/placebo on major and clinically relevant nonmajor (CRNM) and all bleeding, describe most common sites of bleeding, and explore association between asundexian exposure and bleeding. METHODS: We performed a pooled analysis of 3 phase II trials of asundexian in patients with atrial fibrillation (AF), recent acute myocardial infarction (AMI), or stroke. Bleeding was defined according to the International Society on Thrombosis and Hemostasis (ISTH) criteria. RESULTS: In patients with AF (n = 755), both asundexian 20 mg and 50 mg once daily vs apixaban had fewer major/CRNM events (3 of 249; incidence rate [IR] per 100 patient-years 5.47 vs 1 of 254 [IR: not calculable] vs 6 of 250 [IR: 11.10]) and all bleeding (12 of 249 [IR: 22.26] vs 10 of 254 [IR: 18.21] vs 26 of 250 [IR: 50.56]). In patients with recent AMI or stroke (n = 3,409), asundexian 10 mg, 20 mg, and 50 mg once daily compared with placebo had similar rates of major/CRNM events (44 of 840 [IR: 7.55] vs 42 of 843 [IR: 7.04] vs 56 of 845 [IR: 9.63] vs 41 of 851 [IR: 6.99]) and all bleeding (107 of 840 [IR: 19.57] vs 123 of 843 [IR: 22.45] vs 130 of 845 [IR: 24.19] vs 129 of 851 [IR: 23.84]). Most common sites of major/CRNM bleeding with asundexian were gastrointestinal, respiratory, urogenital, and skin. There was no significant association between asundexian exposure and major/CRNM bleeding. CONCLUSIONS: Analyses of phase II trials involving >500 bleeds highlight the potential for improved safety of asundexian compared with apixaban and similar safety compared with placebo. Further evidence on the efficacy of asundexian awaits the results of ongoing phase III trials.


Subject(s)
Atrial Fibrillation , Myocardial Infarction , Stroke , Humans , Anticoagulants/adverse effects , Hemorrhage/chemically induced , Hemorrhage/epidemiology , Hemorrhage/complications , Pyridones/adverse effects , Stroke/etiology , Stroke/prevention & control , Atrial Fibrillation/complications , Myocardial Infarction/drug therapy , Myocardial Infarction/epidemiology
4.
ERJ Open Res ; 8(4)2022 Oct.
Article in English | MEDLINE | ID: mdl-36329798

ABSTRACT

Introduction: The European Sleep Apnea Database was used to identify distinguishable obstructive sleep apnoea (OSA) phenotypes and to investigate the clinical outcome during positive airway pressure (PAP) treatment. Method: Prospective OSA patient data were recruited from 35 sleep clinics in 21 European countries. Unsupervised cluster analysis (anthropometrics, clinical variables) was performed in a random sample (n=5000). Subsequently, all patients were assigned to the clusters using a conditional inference tree classifier. Responses to PAP treatment change in apnoea severity and Epworth sleepiness scale (ESS) were assessed in relation to baseline patient clusters and at short- and long-term follow-up. Results: At baseline, 20 164 patients were assigned (mean age 54.1±12.2 years, 73% male, median apnoea-hypopnoea index (AHI) 27.3 (interquartile range (IQR) 14.1-49.3) events·h-1, and ESS 9.8±5.3) to seven distinct clusters based on anthropometrics, comorbidities and symptoms. At PAP follow-up (median 210 [IQR 134-465] days), the observed AHI reduction (n=1075) was similar, whereas the ESS response (n=3938) varied: largest reduction in cluster 3 (young healthy symptomatic males) and 6 (symptomatic males with psychiatric disorders, -5.0 and -5.1 units, respectively (all p<0.01), limited reduction in clusters 2 (obese males with systemic hypertension) and 5 (elderly multimorbid obese males, -4.2 (p<0.05) and -3.7 (p<0.001), respectively). Residual sleepiness in cluster 5 was particularly evident at long-term follow-up (p<0.05). Conclusion: OSA patients can be classified into clusters based on clinically identifiable features. Importantly, these clusters may be useful for prediction of both short- and long-term responses to PAP intervention.

5.
J Thromb Haemost ; 20(7): 1684-1695, 2022 07.
Article in English | MEDLINE | ID: mdl-35490404

ABSTRACT

BACKGROUND: Bleeding is a clinically significant issue with all current anticoagulants. Safer antithrombotic strategies are required. OBJECTIVES: To investigate the safety, pharmacodynamics, and pharmacokinetics of BAY 1831865, a humanized, factor XI (FXI)-directed monoclonal antibody, after single intravenous (i.v.) or subcutaneous (s.c.) doses in healthy volunteers. PATIENTS/METHODS: In a first-in-human, phase I study, 70 volunteers were randomly assigned (4:1) to receive single-dose BAY 1831865 (3.5, 7, 17, 35, 75, or 150 mg i.v. or 150 mg s.c.) or placebo. Adverse events, pharmacodynamics, and pharmacokinetics were evaluated. RESULTS: In this study, no hemorrhage, or hypersensitivity or infusion-/injection-related reactions were reported. Drug-related adverse events occurred in 3 (5.4%) of 56 volunteers; all were mild and self-limited. Dose-dependent prolongation of activated partial thromboplastin time (aPTT) and inhibition of FXI clotting activity was observed with BAY 1831865 i.v. (geometric mean maximum ratio-to-baseline: aPTT, range, 1.09-3.11 vs. 1.05 with placebo; FXI, range, 0.70-0.04 vs. 0.91 with placebo). Onset of effect was rapid after i.v. administration, with duration of effect (up to 55 days) determined by dose. BAY 1831865 s.c. had similar pharmacodynamic effects but a slower onset of action. Terminal half-life increased continuously with increasing i.v. dose (range, 28-208 h), leading to strong and continuous increases in systemic exposure to BAY 1831865. Absolute bioavailability of BAY 1831865 s.c. was 47.2% (95% confidence interval, 30.2-73.7). CONCLUSIONS: BAY 1831865 i.v. or s.c. was well tolerated, with no evidence of bleeding in healthy volunteers. BAY 1831865 exhibited pronounced, sustained dose-dependent prolongation of aPTT and duration of FXI inhibition.


Subject(s)
Anticoagulants , Factor XIa , Antibodies, Monoclonal, Humanized/pharmacology , Anticoagulants/adverse effects , Dose-Response Relationship, Drug , Double-Blind Method , Hemorrhage/chemically induced , Hemorrhage/drug therapy , Humans , Male , Partial Thromboplastin Time
6.
J Thromb Haemost ; 19(10): 2407-2416, 2021 10.
Article in English | MEDLINE | ID: mdl-34192419

ABSTRACT

BACKGROUND: Coagulation factor XI (FXI) contributes to the development of thrombosis but appears to play a minor role in hemostasis and is, therefore, an attractive anticoagulant drug target. OBJECTIVES: To evaluate the safety, pharmacokinetic, and pharmacodynamic properties of BAY 2433334, an orally administered small molecule targeting activated FXI (FXIa), in healthy men. PATIENTS/METHODS: This phase 1 study was conducted in two parts. In part 1, 70 volunteers were randomized 4:1 to receive a single oral dose of BAY 2433334 (5-150 mg as oral solution or immediate-release tablets) or placebo. In part 2, 16 volunteers received a single oral dose of five BAY 2433334 5-mg tablets with or without a high-calorie breakfast in a randomized crossover study design. Adverse events, pharmacokinetic parameters, and pharmacodynamic parameters were assessed up to 72 h after drug administration. Volunteers were followed up after 7 to 14 days. RESULTS: BAY 2433334 demonstrated favorable safety and tolerability with a dose-dependent increase in exposure and a terminal half-life of 14.2 to 17.4 h. A high-calorie breakfast reduced mean maximum plasma concentration and exposure by 31% and 12.4%, respectively. AY 2433334 was associated with a dose-dependent inhibition of FXIa activity and an increase in activated partial thromboplastin time. Bleeding times in volunteers who had received BAY 2433334 were similar to those in volunteers who had received placebo. CONCLUSIONS: These data indicate that BAY 2433334 is a promising development candidate for once-daily oral anticoagulation; it is being evaluated in phase 2 dose-finding studies in patients at risk of thrombosis.


Subject(s)
Anticoagulants , Factor XIa , Administration, Oral , Area Under Curve , Cross-Over Studies , Double-Blind Method , Humans , Male , Partial Thromboplastin Time
7.
Res Pract Thromb Haemost ; 3(2): 242-253, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31011708

ABSTRACT

BACKGROUND: Coagulation factor XI (FXI) contributes to the development of thrombosis but appears to play only a minor role in hemostasis and is therefore an attractive anticoagulant drug target. OBJECTIVES: To evaluate the safety, pharmacodynamic, and pharmacokinetic properties of BAY 1213790, a fully human immunoglobulin (Ig) G1 antibody targeting activated coagulation FXI (FXIa), in healthy men. METHODS: In this phase 1, single-blind, parallel-group, placebo-controlled, dose-escalation study, 83 healthy Caucasian men were randomized 4:1 to receive a single 60-minute intravenous infusion of BAY 1213790 (0.015-10 mg/kg) or placebo. Adverse events, pharmacodynamic parameters (including activated partial thromboplastin time [aPTT]) and pharmacokinetic parameters were determined. Volunteers were followed up for 150 days. RESULTS: BAY 1213790 demonstrated favorable safety and tolerability; there were no observed cases of bleeding or clinically relevant antidrug antibody formation. One volunteer (1.2%) experienced an infusion reaction. Following intravenous administration of BAY 1213790, dose-dependent increases in aPTT (maximal mean increase relative to baseline: 1.85 [conventional method] and 2.17 [kaolin-triggered method]) and rotational thromboelastometry whole blood clotting time were observed, as well as dose-dependent reductions in FXI activity. Bleeding times did not increase following administration of BAY 1213790 and were similar for all dose cohorts, including placebo. Measurable and dose-dependent increases in systemic exposure were detected for all doses of BAY 1213790 of 0.06 mg/kg or higher. CONCLUSIONS: Based on these safety, pharmacodynamic, and pharmacokinetic results, further evaluation of BAY 1213790 in patients with, or at risk of, thrombosis is warranted.

8.
Pain Med ; 19(6): 1206-1218, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29228247

ABSTRACT

Objectives: To evaluate the analgesic/antihyperalgesic effect of ASP8477. Design: Randomized, double-blind, double-dummy, cross-over, placebo- and active comparator-controlled study. Setting: HPR Dr. Schaffler GmbH, Munich, Germany. Subjects: Healthy female subjects aged 18-65 years. Methods: Eligible subjects were randomly assigned to one of six treatment sequences and received multiple ascending doses of ASP8477, duloxetine, and placebo over three treatment periods (each consisting of 21-day dosing separated by 14-day washout periods). On the last day of each dose level, laser evoked potentials (LEPs) and visual analog scales (VAS pain) on capsaicin-treated skin at baseline and at multiple postdose time points were assessed. The primary end point was the difference in LEP N2-P2 peak-to-peak (PtP) amplitudes for ASP8477 100 mg vs placebo. Results: Twenty-five subjects were randomized. In all subjects, LEP N2-P2 PtP amplitudes were numerically lower for ASP8477 100 mg vs placebo (P = 0.0721); in subjects who demonstrated positive capsaicin skin effects, a greater mean difference of -2.24 µV (P = 0.0146) was observed. Across all doses, LEP N2-P2 PtP amplitudes were lower for duloxetine compared with ASP8477 (mean difference -3.80 µV; P < 0.0001) or placebo (mean difference -5.21 µV; P < 0.0001). The effect of ASP8477 (all doses) on down-scoring the VAS pain score was significant compared with placebo (mean difference -2.55%; P < 0.0007). Conclusions: ASP8477 was well tolerated in this study. Analysis of all subjects did not demonstrate a significant difference in LEP for ASP8477 100 mg over placebo but did in subjects who demonstrated positive capsaicin skin effects.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Analgesics/pharmacology , Pain Management , Piperidines/pharmacology , Pyridines/pharmacology , Adult , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Healthy Volunteers , Humans , Pain Measurement
9.
J Clin Pharmacol ; 54(9): 1006-15, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24676942

ABSTRACT

First-dose-in-children relies on the prediction of clearance from adults for which little information is available on the accuracy of the scaling-approaches applied. For CYP3A-metabolized compounds, scaling of clearance is further challenged by different isoforms and by the CYP3A7 to CYP3A4 switch at young ages. This investigation aimed to evaluate the accuracy of two frequently used scaling approaches and to gain insights into the ontogeny of CYP3A. Hence, a literature database was compiled containing 203 clearance values from term-neonates to adults for 18 CYP3A-metabolized compounds. The clearances in adults were scaled to children using (i) allometric scaling plus maturation function and (ii) a mechanistic approach based on the well-stirred model. Three maturation functions were separately evaluated. In children >3 months, all approaches were interchangeable heeding the maturation function applied and biases were mostly observed in children <3 months. The results from a sensitivity analysis indicate that these biases are possibly caused by disregarding the CYP3A7 activity which could account for up to 86% of the metabolism in term-neonates. Only the mechanistic approach using an overall-CYP3A maturation function led to unbiased predictions of clearances across all ages. The current investigation adds to the predictions of the first-dose-in-children of compounds (partially) metabolized by CYP3A.


Subject(s)
Aging/metabolism , Cytochrome P-450 CYP3A/metabolism , Models, Biological , Pharmaceutical Preparations/metabolism , Adolescent , Adult , Algorithms , Body Weight , Child , Child, Preschool , Hepatic Artery/physiology , Humans , Infant , Infant, Newborn , Liver/blood supply , Pharmaceutical Preparations/blood , Pharmacokinetics , Portal Vein/physiology , Regional Blood Flow
10.
J Pharmacokinet Pharmacodyn ; 40(3): 401-18, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23197248

ABSTRACT

The current gap between animal research and clinical development of analgesic drugs presents a challenge for the application of translational PK-PD modeling and simulation. First, animal pain models lack predictive and construct validity to accurately reflect human pain etiologies and, secondly, clinical pain is a multidimensional sensory experience that can't always be captured by objective and robust measures. These challenges complicate the use of translational PK-PD modeling to project PK-PD data generated in preclinical species to a plausible range of clinical doses. To date only a few drug targets identified in animal studies have shown to be successful in the clinic. PK-PD modeling of biomarkers collected during the early phase of clinical development can bridge animal and clinical pain research. For drugs with novel mechanism of actions understanding of the target pharmacology is essential in order to increase the success of clinical development. There is a specific interest in the application of human pain models that can mimic different aspects of acute/chronic pain symptoms and serves as link between animal and clinical pain research. In early clinical development the main objective of PK-PD modeling is to characterize the relationship between target site binding and downstream biomarkers that have a potential link to the clinical endpoint (e.g. readouts from the human pain models) so as to facilitate the selection of doses for proof of concept studies. In patient studies, the role of PK-PD modeling and simulation is to characterize and confirm patient populations in terms of responder profiles with the aim to find the right dose for the right patient.


Subject(s)
Analgesics , Models, Biological , Pain/drug therapy , Analgesics/pharmacokinetics , Analgesics/pharmacology , Analgesics/therapeutic use , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Animals , Computer Simulation , Humans , Pain/blood , Tissue Distribution , Treatment Outcome
11.
J Pain Res ; 5: 51-9, 2012.
Article in English | MEDLINE | ID: mdl-22536092

ABSTRACT

Treatment of chronic pain is associated with high variability in the response to pharmacological interventions. A mathematical pharmacodynamic model was developed to quantify the magnitude and onset/offset times of effect of a single capsaicin 8% patch application in the treatment of painful diabetic peripheral neuropathy in 91 patients. In addition, a mixture model was applied to objectively match patterns in pain-associated behavior. The model identified four distinct subgroups that responded differently to treatment: 3.3% of patients (subgroup 1) showed worsening of pain; 31% (subgroup 2) showed no change; 32% (subgroup 3) showed a quick reduction in pain that reached a nadir in week 3, followed by a slow return towards baseline (16% ± 6% pain reduction in week 12); 34% (subgroup 4) showed a quick reduction in pain that persisted (70% ± 5% reduction in week 12). The estimate of the response-onset rate constant, obtained for subgroups 1, 3, and 4, was 0.76 ± 0.12 week(-1) (median ± SE), indicating that every 0.91 weeks the pain score reduces or increases by 50% relative to the score of the previous week (= t½). The response-offset rate constant could be determined for subgroup 3 only and was 0.09 ± 0.04 week(-1) (t½ 7.8 weeks). The analysis allowed separation of a heterogeneous neuropathic pain population into four homogenous subgroups with distinct behaviors in response to treatment with capsaicin. It is argued that this model-based approach may have added value in analyzing longitudinal chronic pain data and allows optimization of treatment algorithms for patients suffering from chronic pain conditions.

12.
J Pharmacokinet Pharmacodyn ; 39(2): 195-203, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22311388

ABSTRACT

Dose selection for "first in children" trials often relies on scaling of the pharmacokinetics from adults to children. Commonly used approaches are physiologically-based pharmacokinetic modeling (PBPK) and allometric scaling (AS) in combination with maturation of clearance for early life. In this investigation, a comparison of the two approaches was performed to provide insight into the physiological meaning of AS maturation functions and their interchangeability. The analysis focused on the AS maturation functions established using paracetamol and morphine paediatric data after intravenous administration. First, the estimated AS maturation functions were compared with the maturation functions of the liver enzymes as used in the PBPK models. Second, absolute clearance predictions using AS in combination with maturation functions were compared to PBPK predictions for hypothetical drugs with different pharmacokinetic properties. The results of this investigation showed that AS maturation functions do not solely represent ontogeny of enzyme activity, but aggregate multiple pharmacokinetic properties, as for example extraction ratio and lipophilicity (log P). Especially in children younger than 1 year, predictions using AS in combination with maturation functions and PBPK were not interchangeable. This highlights the necessity of investigating methodological uncertainty to allow a proper estimation of the "first dose in children" and assessment of its risk and benefits.


Subject(s)
Acetaminophen/pharmacokinetics , Drug Discovery/trends , Morphine/pharmacokinetics , Acetaminophen/administration & dosage , Adolescent , Age Factors , Child , Child, Preschool , Dose-Response Relationship, Drug , Drug Discovery/methods , Humans , Infant , Infant, Newborn , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/physiology , Morphine/administration & dosage
13.
Expert Rev Clin Pharmacol ; 4(6): 719-28, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22111858

ABSTRACT

In acute and chronic pain, the objective of pharmacokinetic-pharmacodynamic (PKPD) modeling is the development and application of mathematical models to describe and/or predict the time course of the pharmacokinetics (PK) and pharmacodynamics (PD) of analgesic agents and link PK to PD. Performing population PKPD modeling using nonlinear mixed effects modeling allows, apart from the estimation of fixed effects (the PK and PD model estimates), the quantification of random effects as within- and between-subject variability. Effect-compartment models and mechanism-based biophase distribution models that incorporate drug-association and -dissociation kinetics are applied in PKPD modeling of pain treatment. Mechanism-based models enable the quantification of the rate-limiting factors in drug effect owing to drug distribution versus receptor kinetics (since receptor kinetics are nonlinear they are discernable from the linear effect-compartment kinetics). It is a helpful technique in understanding the complex behavior of specific analgesics, such as buprenorphine, but also morphine and its active metabolite morphine-6-glucuronide, especially with respect to the reversal of opioid-induced side effects, most importantly life-threatening respiratory depression. One approach in chronic pain studies is the application of mixture models. Mixture models do not necessarily need to take PK data into account and allow the objective differentiation of measured responses to analgesics into specific response subgroups, and as such, may play an important role in analyzing Phase I and II analgesia studies. Appropriate application of PKPD modeling leads to the improvement of current therapeutics with respect to dose design and outcome, understanding the interaction of analgesics within complex chronic pain disease processes and may play an important role in drug development. In the current article, novel observations using the aforementioned techniques on opioids, NSAIDs, epidural analgesia, ketamine and GABA-ergic drugs in acute and chronic pain are discussed.


Subject(s)
Acute Pain/drug therapy , Analgesics/pharmacology , Analgesics/pharmacokinetics , Chronic Pain/drug therapy , Models, Biological , Acute Pain/metabolism , Analgesia/methods , Analgesics/administration & dosage , Animals , Chronic Pain/metabolism , Humans , Models, Chemical
14.
Drug Metab Dispos ; 39(7): 1278-87, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21487054

ABSTRACT

The application of model-based drug development in special populations becomes increasingly important for clinical trial optimization, mostly by providing a rationale for dose selection and thereby aiding risk-benefit assessment. In this article, a semiphysiological approach is presented, enabling the extrapolation of the pharmacokinetics from healthy subjects to patients with different disease conditions. This semiphysiological approach was applied to solifenacin, using clinical data on total and free plasma and urine concentrations in healthy subjects. The analysis was performed using nonlinear mixed-effects modeling and relied on the use of a general partitioning framework to account for binding to plasma proteins and to nonplasma tissues together with principles from physiology that apply to the main pharmacokinetic process, i.e., bioavailability, distribution, and elimination. Application of these physiology principles allowed quantification of the impact of key physiological parameters (i.e., body composition, glomerular function, liver enzyme capacity, and liver blood flow) on the pharmacokinetics of solifenacin. The prediction of the time course of the drug concentration in liver- and renal-impaired patients only required adjustment of the physiological parameters that are known to change upon liver and renal dysfunction without modifying the pharmacokinetic model structure and/or its respective parameter estimates. Visual predictive checks showed that the approach applied was able to adequately predict the pharmacokinetics of solifenacin in liver- and renal-impaired patients. In addition, better insight into the pharmacokinetic properties of solifenacin was obtained. In conclusion, the proposed semiphysiological approach is attractive for prediction of altered pharmacokinetics of compounds influenced by liver and renal disease conditions.


Subject(s)
Kidney Diseases/drug therapy , Liver Diseases/drug therapy , Models, Biological , Pharmacokinetics , Humans
15.
Pharm Res ; 25(1): 183-93, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17914664

ABSTRACT

OBJECTIVE: Respiratory depression is a serious and potentially life-threatening side-effect of opioid therapy. The objective of this investigation was to characterize the relationship between buprenorphine or fentanyl exposure and the effectiveness and safety outcome in rats. METHODS: Data on the time course of the antinociceptive and respiratory depressant effect were analyzed on the basis of population logistic regression PK-PD models using non-linear mixed effects modeling software (NONMEM). The pharmacokinetics of buprenorphine and fentanyl were described by a three- and two-compartment model, respectively. A logistic regression model (linear logit model) was used to characterize the relationship between drug exposure and the binary effectiveness and safety outcome. RESULTS: For buprenorphine, the odds ratios (OR) were 28.5 (95% CI, 6.9-50.1) and 2.10 (95% CI, 0.71-3.49) for the antinociceptive and respiratory depressant effect, respectively. For fentanyl these odds ratios were 3.03 (95% CI, 1.87-4.21) and 2.54 (95% CI, 1.26-3.82), respectively. CONCLUSION: The calculated safety index (OR(antinociception)/OR(respiratory depression)) for fentanyl of 1.20 suggests that fentanyl has a low safety margin, implicating that fentanyl needs to be titrated with caution. For buprenorphine the safety index is 13.54 suggesting that buprenorphine is a relatively safe opioid.


Subject(s)
Buprenorphine/pharmacology , Buprenorphine/pharmacokinetics , Fentanyl/pharmacology , Fentanyl/pharmacokinetics , Narcotic Antagonists/pharmacology , Narcotic Antagonists/pharmacokinetics , Algorithms , Animals , Buprenorphine/adverse effects , Depression, Chemical , Fentanyl/adverse effects , Linear Models , Logistic Models , Male , Models, Statistical , Narcotic Antagonists/adverse effects , Odds Ratio , Pain Measurement/drug effects , Plethysmography, Whole Body , Rats , Rats, Wistar , Regression Analysis , Respiratory Function Tests , Respiratory Mechanics/drug effects
16.
Eur J Pain ; 12(4): 403-11, 2008 May.
Article in English | MEDLINE | ID: mdl-17869146

ABSTRACT

Morphine-6-glucuronide (M6G) is morphine's active metabolite acting at the mu-opioid receptor. Recent experimental human studies and 5 of 6 randomized clinical trials indicate that M6G causes adequate and long lasting pain relief comparable to morphine. There are various observations that M6G is associated with a reduction in the severity of side effects normally associated with opioid use, such as reduced postoperative nausea and vomiting (PONV) and reduced respiratory depression. The present drug profile provides a review of the pharmacological properties of M6G, the clinical evidence relating to its efficacy and safety, and discusses its future role in the treatment of postoperative pain.


Subject(s)
Analgesics, Opioid/therapeutic use , Morphine Derivatives/therapeutic use , Pain, Postoperative/drug therapy , Humans
17.
Clin Pharmacokinet ; 46(11): 965-80, 2007.
Article in English | MEDLINE | ID: mdl-17922561

ABSTRACT

BACKGROUND AND OBJECTIVE: Respiratory depression is a potentially life-threatening adverse effect of opioid therapy. It has been postulated that the difficulty of reversing buprenorphine-induced respiratory depression is caused by slow receptor association-dissociation kinetics at the opioid mu receptor. The aim of this study was to characterise the pharmacodynamic interaction between buprenorphine and naloxone in healthy volunteers. METHODS: A competitive pharmacodynamic interaction model was proposed to describe and predict the time course of naloxone-induced reversal of respiratory depression. The model was identified using data from an adaptive naloxone dose-selection trial following intravenous administration of buprenorphine 0.2mg/70kg or 0.4mg/70kg. RESULTS: The pharmacokinetics of naloxone and buprenorphine were best described by a two-compartment model and a three-compartment model, respectively. A combined biophase equilibration-receptor association-dissociation pharmacodynamic model described the competitive interaction between buprenorphine and naloxone at the opioid mu receptor. For buprenorphine, the values of the rate constants of receptor association (k(on)) and dissociation (k(off)) were 0.203 mL/ng/min and 0.0172 min(-)(1), respectively. The value of the equilibrium dissociation constant (K(D)) was 0.18 nmol/L. The half-life (t((1/2))) of biophase equilibration was 173 minutes. These estimates of the pharmacodynamic parameters are similar to values obtained in the absence of naloxone co-administration. For naloxone, the half-life of biophase distribution was 6.5 minutes. CONCLUSIONS: Because of the slow receptor association-dissociation kinetics of buprenorphine in combination with the fast elimination kinetics of naloxone, naloxone is best administered as a continuous infusion for reversal of buprenorphine-induced respiratory depression.


Subject(s)
Buprenorphine/adverse effects , Models, Biological , Naloxone/pharmacokinetics , Respiratory Insufficiency/prevention & control , Adolescent , Adult , Algorithms , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Buprenorphine/administration & dosage , Dose-Response Relationship, Drug , Drug Interactions , Female , Half-Life , Humans , Infusions, Intravenous , Injections, Intravenous , Male , Naloxone/administration & dosage , Naloxone/therapeutic use , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacokinetics , Narcotic Antagonists/therapeutic use , Respiratory Function Tests , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/metabolism
18.
Clin Pharmacokinet ; 46(5): 433-47, 2007.
Article in English | MEDLINE | ID: mdl-17465641

ABSTRACT

OBJECTIVES: This investigation describes the interspecies scaling of the pharmacokinetics and pharmacodynamics of buprenorphine. METHODS: Data on the time course of the antinociceptive and respiratory depressant effects of buprenorphine in rats and in humans were simultaneously analysed on the basis of a mechanism-based pharmacokinetic-pharmacodynamic model. RESULTS: An allometric three-compartment pharmacokinetic model described the time course of the concentration in plasma. The value of the allometric coefficient for clearance was 35.2 mL/min (relative standard error [RSE] = 5.6%) and the value of the allometric exponent was 0.76 (RSE 5.61%). A combined biophase distribution-receptor association/dissociation model with a linear transduction function described hysteresis between plasma concentration and effect. The values of the drug-specific pharmacodynamic parameters were identical in rats and in humans. For the respiratory depressant effect, the values of the second-order rate constant of receptor association (k(on)) and the first-order rate constant of receptor dissociation (k(off)) were 0.23 mL/ng/min (RSE = 15.8%) and 0.014 min(-1) (RSE = 27.7%), respectively, and the value of the equilibrium dissociation constant (K(diss)) was 0.13 nmol/L. The value of the intrinsic activity alpha was 0.52 (RSE = 3.4%). For the antinociceptive effect, the values of the k(on) and k(off) were 0.015 mL/ng/min (RSE = 18.3%) and 0.053 min(-1) (RSE = 23.1%), respectively. The value of the K(diss) was 7.5 nmol/L. An allometric equation described the scaling of the system-specific parameter, the first-order distribution rate constant (k(e0)). The value of the allometric coefficient for the k(e0) was 0.0303 min(-1) (RSE = 11.3%) and the value of the exponent was -0.28 (RSE = 9.6%). CONCLUSIONS: The different values of the drug-specific pharmacodynamic parameters are consistent with the different opioid mu receptor subtypes involved in the antinociceptive and respiratory depressant effects.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/therapeutic use , Buprenorphine/pharmacokinetics , Buprenorphine/therapeutic use , Analgesics, Opioid/adverse effects , Animals , Buprenorphine/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Routes , Humans , Models, Biological , Rats , Reproducibility of Results , Respiratory Insufficiency/chemically induced
19.
Expert Opin Drug Saf ; 6(2): 125-32, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17367258

ABSTRACT

Naloxone is a non-selective, short-acting opioid receptor antagonist that has a long clinical history of successful use and is presently considered a safe drug over a wide dose range (up to 10 mg). In opioid-dependent patients, naloxone is used in the treatment of opioid-overdose-induced respiratory depression, in (ultra)rapid detoxification and in combination with buprenorphine for maintenance therapy (to prevent intravenous abuse). Risks related to naloxone use in opioid-dependent patients are: i) the induction of an acute withdrawal syndrome (the occurrence of vomiting and aspiration is potentially life threatening); ii) the effect of naloxone may wear off prematurely when used for treatment of opioid-induced respiratory depression; and iii) in patients treated for severe pain with an opioid, high-dose naloxone and/or rapidly infused naloxone may cause catecholamine release and consequently pulmonary edema and cardiac arrhythmias. These risks warrant the cautious use of naloxone and adequate monitoring of the cardiorespiratory status of the patient after naloxone administration where indicated.


Subject(s)
Naloxone/adverse effects , Naloxone/therapeutic use , Narcotic Antagonists/adverse effects , Narcotic Antagonists/therapeutic use , Opioid-Related Disorders/drug therapy , Humans , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Opioid-Related Disorders/diagnosis , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/physiology , Risk Factors
20.
J Pharmacol Exp Ther ; 321(2): 598-607, 2007 May.
Article in English | MEDLINE | ID: mdl-17283225

ABSTRACT

The objective of this investigation was to characterize the pharmacokinetic-pharmacodynamic (PK-PD) correlation of buprenorphine's active metabolite norbuprenorphine for the effect on respiration in rats. Following i.v. administration in rats (dose range 0.32-1.848 mg), the time course of the concentration in plasma was determined in conjunction with the effect in ventilation as determined with a novel whole-body plethysmography technique. The PK of norbuprenorphine was best described by a three-compartment PK model with nonlinear elimination. A saturable biophase distribution model with a power PD model described the PK-PD relationship best. No saturation of the effect at high concentrations was observed, indicating that norbuprenorphine acts as a full agonist with regard to respiratory depression. Moreover, analysis of the hysteresis based on the combined receptor association-dissociation biophase distribution model yielded high values of the rate constants for receptor association and dissociation, indicating that these processes are not rate-limiting. In a separate analysis, the time course of the plasma concentrations of buprenorphine and norbuprenorphine following administration of both the parent drug and the metabolite were simultaneously analyzed based on a six-compartment PK model with nonlinear elimination of norbuprenorphine. This analysis showed that following i.v. administration, 10% of the administered dose of buprenorphine is converted into norbuprenorphine. By simulation it is shown that following i.v. administration of buprenorphine, the concentrations of norbuprenorphine reach values that are well below the values causing an effect on respiration.


Subject(s)
Buprenorphine/analogs & derivatives , Respiration/drug effects , Animals , Buprenorphine/pharmacokinetics , Buprenorphine/pharmacology , Male , Models, Biological , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...