Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
J Thromb Thrombolysis ; 50(1): 20-29, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32323192

ABSTRACT

Rivaroxaban exposure and patient characteristics may affect the rivaroxaban benefit-risk balance. This study aimed to quantify associations between model-predicted rivaroxaban exposure and patient characteristics and efficacy and safety outcomes in patients with non-valvular atrial fibrillation (NVAF), using data from the phase 3 ROCKET AF trial (NCT00403767). In ROCKET AF, 14,264 patients with NVAF were randomized to rivaroxaban (20 mg once daily [OD], or 15 mg OD if creatinine clearance was 30-49 mL/min) or dose-adjusted warfarin (median follow-up: 707 days); rivaroxaban plasma concentration was measured in a subset of 161 patients. In this post hoc exposure-response analysis, a multivariate Cox model was used to correlate individual predicted rivaroxaban exposures and patient characteristics with time-to-event efficacy and safety outcomes in 7061 and 7111 patients, respectively. There was no significant association between model-predicted rivaroxaban trough plasma concentration (Ctrough) and efficacy outcomes. Creatinine clearance and history of stroke were significantly associated with efficacy outcomes. Ctrough was significantly associated with the composite of major or non-major clinically relevant (NMCR) bleeding (hazard ratio [95th percentile vs. median]: 1.26 [95% confidence interval 1.13-1.40]) but not with major bleeding alone. The exposure-response relationship for major or NMCR bleeding was shallow with no clear threshold for an acceleration in risk. History of gastrointestinal bleeding had a greater influence on safety outcomes than Ctrough. These results support fixed rivaroxaban 15 mg and 20 mg OD dosages in NVAF. Therapeutic drug monitoring is unlikely to offer clinical benefits in this indication beyond evaluation of patient characteristics.


Subject(s)
Atrial Fibrillation , Drug Monitoring , Hemorrhage , Risk Adjustment/methods , Rivaroxaban , Stroke , Venous Thromboembolism , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Anticoagulants/blood , Anticoagulants/pharmacokinetics , Atrial Fibrillation/complications , Atrial Fibrillation/drug therapy , Biomarkers, Pharmacological/analysis , Chemoprevention/methods , Drug Monitoring/methods , Drug Monitoring/statistics & numerical data , Female , Hemorrhage/chemically induced , Hemorrhage/prevention & control , Humans , Male , Middle Aged , Models, Statistical , Prognosis , Prothrombin Time/methods , Risk Assessment , Rivaroxaban/administration & dosage , Rivaroxaban/adverse effects , Rivaroxaban/blood , Rivaroxaban/pharmacokinetics , Stroke/etiology , Stroke/prevention & control , Venous Thromboembolism/blood , Venous Thromboembolism/diagnosis , Venous Thromboembolism/etiology , Venous Thromboembolism/prevention & control
2.
Br J Clin Pharmacol ; 82(5): 1364-1370, 2016 11.
Article in English | MEDLINE | ID: mdl-27333588

ABSTRACT

AIMS: To prospectively select the dose of the paliperidone palmitate 3-month (PP3M) formulation, using a pharmacometric bridging strategy based on the paliperidone palmitate 1-month (PP1M) formulation previously approved for schizophrenia treatment. METHODS: Pharmacokinetic (PK) data from a 6-month interim analysis of a single dose PP3M Phase I clinical trial was integrated with a previously developed PP1M population-PK model. The model was updated to incorporate formulation as a covariate on absorption parameters and to explore the most critical design element of the Phase III study: the PP1M-to-PP3M dose multiplier for patients switching formulations. Plasma paliperidone concentrations were measured at predetermined intervals during Phase III, enabling comparison of the multiple-dose PK between PP1M and PP3M. Exposure matching was assessed graphically to determine whether paliperidone plasma concentrations from the two formulations overlapped. RESULTS: Prospective steady-state PK simulations revealed that a 3.5 multiple of the PP1M dose would yield a corresponding PP3M dose with comparable exposure. The prospective pharmacometric simulation and observed Phase III PK data agreed closely. Phase III results confirmed the hypothesis that efficacy of PP3M was noninferior to that of PP1M. The similarity in exposures between the two formulations was likely a key determinant of the equivalent efficacy between the two products observed in the Phase III study. CONCLUSIONS: Successful prospective PP3M Phase III clinical trial dose selection was achieved through the use of pharmacometric bridging, without conducting a Phase II study and using only limited Phase I data for PP3M. We estimate that this strategy reduced development time by 3-5 years and may be applicable to other drug development projects.


Subject(s)
Drug Administration Schedule , Drug Compounding , Paliperidone Palmitate/administration & dosage , Paliperidone Palmitate/pharmacokinetics , Adolescent , Adult , Aged , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Dosage Calculations , Female , Humans , Male , Middle Aged , Models, Biological , Paliperidone Palmitate/blood , Prospective Studies , Schizophrenia/blood , Young Adult
3.
Eur J Drug Metab Pharmacokinet ; 49(4): 477-489, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38750386

ABSTRACT

BACKGROUND AND OBJECTIVE: A model-informed drug development (MIDD) approach was implemented for paliperidone palmitate (PP) 6-month (PP6M) clinical development, using pharmacokinetics and pharmacokinetic/pharmacodynamic model-based simulations. METHODS: PP6M pharmacokinetics were simulated by extending the PP 3-month (PP3M) pharmacokinetic model to account for increased injection volume, and hence dose. Contribution of the MIDD approach to the design of the pivotal PP6M phase-3 study (PP6M/PP3M noninferiority study, NCT03345342) investigating schizophrenia relapse rates was twofold: (1) PP6M dose selection, and (2) hypothesis generation that lower trough concentrations (Ctrough) associated with PP6M, relative to PP3M, were not associated with lower efficacy, which was to be evaluated in the phase-3 study. Moreover, accompanied by an intense sampling scheme to adequately characterize paliperidone pharmacokinetics and to elucidate the potential relationship between concentration and safety/efficacy, the bridging strategy eliminated the need for additional phase-1/phase-2 clinical studies. RESULTS: Using a MIDD bridging strategy, PP6M doses were selected that, compared with PP3M, were expected to have a similar range of exposures and a noninferior relapse rate and safety profile. Clinical data from PP6M/PP3M noninferiority study confirmed that PP6M, compared with PP3M, had a similar range of exposures (T'jollyn et al. in Eur J Drug Metab Pharmacokinet 2024), as well as a noninferior relapse rate and safety profile (this manuscript). CONCLUSIONS: Consistency of the MIDD approach with observed clinical outcomes confirmed the hypothesis that lower Ctrough did not lead to increased relapse rates at the doses administered. Although higher paliperidone peak concentrations are achieved with corresponding doses of PP6M relative to PP3M in the phase-3 clinical study, types and incidences of treatment-related adverse events were comparable between PP6M and PP3M groups and no new safety concerns emerged for PP6M (Najarian et al. in Int J Neuropsychopharmacol 25(3):238-251, 2022).


Subject(s)
Antipsychotic Agents , Paliperidone Palmitate , Schizophrenia , Paliperidone Palmitate/pharmacokinetics , Paliperidone Palmitate/administration & dosage , Paliperidone Palmitate/adverse effects , Humans , Schizophrenia/drug therapy , Antipsychotic Agents/pharmacokinetics , Antipsychotic Agents/administration & dosage , Models, Biological , Drug Administration Schedule , Adult , Male , Female , Drug Development/methods , Computer Simulation
4.
Eur J Drug Metab Pharmacokinet ; 49(4): 491-506, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38769284

ABSTRACT

BACKGROUND AND OBJECTIVE: Paliperidone palmitate 6-month (PP6M) intramuscular (IM) injection is the longest-acting treatment available for patients with schizophrenia. A population pharmacokinetic (popPK) modeling and simulation approach was deployed to inform dosing strategies for PP6M. METHODS: The extensive analysis database included 15,932 paliperidone samples from 700 patients receiving gluteal paliperidone palmitate 3-month (PP3M) or PP6M injections in the double-blind phase of a phase-3 noninferiority study (NCT03345342). Exposure parameters for paliperidone appeared to increase dose-proportionally within each dosing schedule (PP3M/PP6M). The range of paliperidone exposures after IM administration of PP6M overlaps with that of corresponding doses of oral paliperidone extended release, PP 1-month (PP1M), and PP3M. Model-based simulations were performed to investigate paliperidone exposures in different PP6M dosing scenarios and relevant subpopulations. RESULTS: A dosing window of ≤ 2 weeks earlier and ≤ 3 weeks later than the target 6-month interval for maintenance treatment with PP6M dosing maintains paliperidone exposures at levels that are not expected to substantially impact its safety and efficacy. For missed-dose scenarios, tailored re-initiation regimens are proposed that should be applied before resuming PP6M maintenance dosing. Regarding subpopulations, PP6M 700 mg eq. is the highest dose recommended in mild renal-impairment patients; the paliperidone pharmacokinetics after PP6M administration is not affected by sex, body mass index, or age in a clinically meaningful way. CONCLUSION: Paliperidone concentration-time profiles after PP6M and PP3M dosing were adequately described by the popPK model. Model-based simulation results provide guidance for clinicians on initiating PP6M therapy, transitioning between paliperidone formulations, the dosing windows to use for maintenance dosing, and managing missed PP6M doses.


Subject(s)
Antipsychotic Agents , Models, Biological , Paliperidone Palmitate , Schizophrenia , Paliperidone Palmitate/pharmacokinetics , Paliperidone Palmitate/administration & dosage , Humans , Schizophrenia/drug therapy , Antipsychotic Agents/pharmacokinetics , Antipsychotic Agents/administration & dosage , Adult , Male , Female , Injections, Intramuscular , Middle Aged , Double-Blind Method , Computer Simulation , Drug Administration Schedule , Dose-Response Relationship, Drug , Young Adult , Delayed-Action Preparations/pharmacokinetics , Adolescent
5.
J Pharmacokinet Pharmacodyn ; 40(4): 537-44, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23645382

ABSTRACT

Beta regression models have been recommended for continuous bounded outcome scores that are often collected in clinical studies. Implementing beta regression in NONMEM presents difficulties since it does not provide gamma functions required by the beta distribution density function. The objective of the study was to implement mixed-effects beta regression models in NONMEM using Nemes' approximation to the gamma function and to evaluate the performance of the NONMEM implementation of mixed-effects beta regression in comparison to the commonly used SAS approach. Monte Carlo simulations were conducted to simulate continuous outcomes within an interval of (0, 70) based on a beta regression model in the context of Alzheimer's disease. Six samples per subject over a 3 years period were simulated at 0, 0.5, 1, 1.5, 2, and 3 years. One thousand trials were simulated and each trial had 250 subjects. The simulation-reestimation exercise indicated that the NONMEM implementation using Laplace and Nemes' approximations provided only slightly higher bias and relative RMSE (RRMSE) compared to the commonly used SAS approach with adaptive Gaussian quadrature and built-in gamma functions, i.e., the difference in bias and RRMSE for fixed-effect parameters, random effects on intercept, and the precision parameter were <1-3 %, while the difference in the random effects on the slope was <3-7 % under the studied simulation conditions. The mixed-effect beta regression model described the disease progression for the cognitive component of the Alzheimer's disease assessment scale from the Alzheimer's Disease Neuroimaging Initiative study. In conclusion, with Nemes' approximation of the gamma function, NONMEM provided comparable estimates to those from SAS for both fixed and random-effect parameters. In addition, the NONMEM run time for the mixed beta regression models appeared to be much shorter compared to SAS, i.e., 1-2 versus 20-40 s for the model and data used in the manuscript.


Subject(s)
Models, Statistical , Normal Distribution , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Computer Simulation , Disease Progression , Humans , Middle Aged , Monte Carlo Method , Regression Analysis
6.
Pharm Res ; 29(9): 2555-64, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22618801

ABSTRACT

PURPOSE: To understand the relationship between the risk of opioid-related gastrointestinal adverse effects (AEs) and exposure to tapentadol and oxycodone as well as its active metabolite, oxymorphone, using pharmacokinetic/pharmacodynamic models. METHODS: The analysis was based on a study in patients with moderate-to-severe pain following bunionectomy. Population PK modeling was conducted to estimate population PK parameters for tapentadol, oxycodone, and oxymorphone. Time to AEs was analyzed using Cox proportional-hazards models. RESULTS: Risk of nausea, vomiting, and constipation significantly increased with exposure to tapentadol or oxycodone/oxymorphone. However, elevated risk per drug exposure of AEs for tapentadol was ~3-4 times lower than that of oxycodone, while elevated AE risk per drug exposure of oxycodone was ~60 times lower than that for oxymorphone, consistent with reported in vitro receptor binding affinities for these compounds. Simulations show that AE incidence following administration of tapentadol IR is lower than that following oxycodone IR intake within the investigated range of analgesic noninferiority dose ratios. CONCLUSIONS: This PK/PD analysis supports the clinical findings of reduced nausea, vomiting and constipation reported by patients treated with tapentadol, compared to patients treated with oxycodone.


Subject(s)
Gastrointestinal Tract/drug effects , Oxycodone/administration & dosage , Phenols/administration & dosage , Female , Humans , Male , Middle Aged , Oxycodone/adverse effects , Oxycodone/pharmacokinetics , Oxycodone/pharmacology , Phenols/adverse effects , Phenols/pharmacokinetics , Phenols/pharmacology , Proportional Hazards Models , Receptors, Opioid, mu/antagonists & inhibitors , Tapentadol
7.
Pharm Stat ; 11(4): 280-6, 2012.
Article in English | MEDLINE | ID: mdl-22407972

ABSTRACT

Assessing dose-response from flexible-dose clinical trials (e.g., titration or dose escalation studies) is challenging and often problematic due to the selection bias caused by 'titration-to-response'. We investigate the performance of a dynamic linear mixed-effects (DLME) model and marginal structural model (MSM) in evaluating dose-response from flexible-dose titration clinical trials via simulations. The simulation results demonstrated that DLME models with previous exposure as a time-varying covariate may provide an unbiased and efficient estimator to recover exposure-response relationship from flexible-dose clinical trials. Although the MSM models with independent and exchangeable working correlations appeared to be able to recover the right direction of the dose-response relationship, it tended to over-correct selection bias and overestimated the underlying true dose-response. The MSM estimators were also associated with large variability in the parameter estimates. Therefore, DLME may be an appropriate modeling option in identifying dose-response when data from fixed-dose studies are absent or a fixed-dose design is unethical to be implemented.


Subject(s)
Controlled Clinical Trials as Topic/methods , Models, Statistical , Pharmaceutical Preparations/administration & dosage , Computer Simulation , Dose-Response Relationship, Drug , Humans , Linear Models , Selection Bias , Time Factors
8.
J Pharmacokinet Pharmacodyn ; 38(4): 423-32, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21626437

ABSTRACT

The objectives of the simulation study were to evaluate the impact of BQL data on pharmacokinetic (PK) parameter estimates when the incidence of BQL data is low (e.g. ≤10%), and to compare the performance of commonly used modeling methods for handling BQL data such as data exclusion (M1) and likelihood-based method (M3). Simulations were performed by adapting the method of a recent publication by Ahn et al. (J Phamacokinet Pharmacodyn 35(4):401-421, 2008). The BQL data in the terminal elimination phase were created at frequencies of 1, 2.5, 5, 7.5, and 10% based on a one- and a two-compartment model. The impact of BQL data on model parameter estimates was evaluated based on bias and imprecision. The simulations demonstrated that for the one-compartment model, the impact of ignoring the low percentages of BQL data (≤10%) in the elimination phase was minimal. For the two-compartment model, when the BQL incidence was less than 5%, omission of the BQL data generally did not inflate the bias in the fixed-effect parameters, whereas more pronounced bias in the estimates of inter-individual variability (IIV) was observed. The BQL data in the elimination phase had the greatest impact on the volume of distribution estimate of the peripheral compartment of the two-compartment model. The M3 method generally provided better parameter estimates for both PK models than the M1 method. However, the advantages of the M3 over the M1 method varied depending on different BQL censoring levels, PK models and parameters. As the BQL percentages decreased, the relative gain of the M3 method based on more complex likelihood approaches diminished when compared to the M1 method. Therefore, it is important to balance the trade-off between model complexity and relative gain in model improvement when the incidence of BQL data is low. Understanding the model structure and the distribution of BQL data (percentage and location of BQL data) allows selection of an appropriate and effective modeling approach for handling low percentages of BQL data.


Subject(s)
Data Interpretation, Statistical , Limit of Detection , Models, Biological , Pharmacokinetics , Computer Simulation
9.
Clin Pharmacokinet ; 60(4): 501-516, 2021 04.
Article in English | MEDLINE | ID: mdl-33128208

ABSTRACT

BACKGROUND: Esketamine nasal spray is approved for treatment-resistant depression. OBJECTIVE: The objective of this study was to characterize the pharmacokinetics of esketamine and noresketamine in healthy subjects and patients with treatment-resistant depression. METHODS: Esketamine and noresketamine were measured in > 9000 plasma samples collected from 820 individuals who received esketamine by the intranasal, intravenous, and oral routes. An open linear model for esketamine (three compartments) and noresketamine (two compartments) that included a hepato-portal compartment was developed using NONMEM® VII. The effects of covariates on esketamine pharmacokinetics and a model evaluation were performed using conventional methods. RESULTS: The fraction of a 28-mg intranasal dose absorbed through the nasal cavity (FRn) is 54% (100% of this fraction is completely absorbed); the remaining 46% is swallowed and undergoes intestinal and first-pass metabolism and 18.6% of the swallowed dose reaches the systemic circulation. The absolute bioavailability of 56 and 84 mg of intranasal esketamine is 54 and 51%, respectively. Esketamine volume at steady state and clearance were 752 L and 114 L/h, respectively. Noresketamine volume at steady state and apparent clearance were 185 L and 38 L/h, respectively. Relative to non-Asian subjects, Asian subjects showed a 64.0 and 19.4% decrease in the esketamine elimination rate constant and noresketamine apparent clearance, respectively. Japanese subjects exhibited a 34% increase in FRn vs other races. Hepatic blood flow decreased by 21.9 L/h for each decade in age in subjects aged > 60 years. These changes resulted in esketamine and noresketamine maximum concentration and area under the concentration-time curve after 24 h post-dose values that were up to 36% higher than those observed in other races or in younger adult subjects. CONCLUSIONS: Esketamine and noresketamine pharmacokinetics was successfully characterized in healthy subjects and patients with treatment-resistant depression. The model quantified esketamine absolute nasal and oral bioavailability, its hepatic flow-limited clearance and biotransformation to the major metabolite noresketamine, and the influence of intrinsic and extrinsic factors on esketamine pharmacokinetics. Clinical trials registration numbers of the studies included in the analysis: ESKETINTRD1001 (NCT01780259), ESKETINTRD1002 (NCT01980303), ESKETINTRD1003 (NCT02129088), ESKETINTRD1008 (NCT02846519), ESKETINTRD1009 (NCT02343289), ESKETINTRD1010 (NCT02568176), ESKETINTRD1012 (NCT02345148), 54135419TRD1015 (NCT02682225), ESKETINTRD2003 (NCT01998958), ESKETINSUI2001 (NCT02133001), ESKETINTRD3001 (NCT02417064), ESKETINTRD3002 (NCT02418585), and ESKETINTRD3005 (NCT02422186).


Subject(s)
Depression , Nasal Sprays , Adult , Antidepressive Agents , Healthy Volunteers , Humans , Ketamine , Middle Aged
10.
Antimicrob Agents Chemother ; 54(6): 2354-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20385854

ABSTRACT

A population pharmacokinetic model of doripenem was constructed using data pooled from phase 1, 2, and 3 studies utilizing nonlinear mixed effects modeling. A 2-compartment model with zero-order input and first-order elimination best described the log-transformed concentration-versus-time profile of doripenem. The model was parameterized in terms of total clearance (CL), central volume of distribution (V(c)), peripheral volume of distribution (V(p)), and distribution clearance between the central and peripheral compartments (Q). The final model was described by the following equations (for jth subject): CL(j) (liters/h) = 13.6.(CL(CR)(j)/98 ml/min)(0.659).(1 + CL(race)(j) [0 for Caucasian]); V(c)(j) (liters) = 11.6.(weight(j)/73 kg)(0.596); Q(j) (liters/h) = 4.74.(weight(j)/73)(1.06); and V(p)(j) (liters) = 6.04.(CL(CR)(j)/98 ml/min)(0.417).(weight(j)/73 kg)(0.840).(age(j)/40 years)(0.307). According to the final model, population mean parameter estimates and interindividual variability (percent coefficient of variation [% CV]) for CL (liters/h), V(c) (liters), V(p) (liters), and Q (liters/h) were 13.6 (19%), 11.6 (19%), 6.0 (25%), and 4.7 (42%), respectively. Residual variability, estimated using three separate additive residual error models, was 0.17 standard deviation (SD), 0.55 SD, and 0.92 SD for phase 1, 2, and 3 data, respectively. Creatinine clearance was the most significant predictor of doripenem clearance. Mean Bayesian clearance was approximately 33%, 55%, and 76% lower for individuals with mild, moderate, or severe renal impairment, respectively, than for those with normal renal function. The population pharmacokinetic model based on healthy volunteer data and patient data informs us of doripenem disposition in a more general population as well as of the important measurable intrinsic and extrinsic factors that significantly influence interindividual pharmacokinetic differences.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Carbapenems/pharmacokinetics , Adolescent , Adult , Aged , Aged, 80 and over , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/blood , Bacterial Infections/drug therapy , Bacterial Infections/metabolism , Bayes Theorem , Carbapenems/administration & dosage , Carbapenems/blood , Creatinine/metabolism , Critical Illness , Cross Infection/drug therapy , Cross Infection/metabolism , Doripenem , Female , Humans , Kidney/metabolism , Male , Metabolic Clearance Rate , Middle Aged , Models, Biological , Nonlinear Dynamics , Pneumonia, Bacterial/drug therapy , Pneumonia, Bacterial/metabolism , Pyelonephritis/drug therapy , Pyelonephritis/metabolism , Renal Insufficiency/metabolism , Urinary Tract Infections/drug therapy , Urinary Tract Infections/metabolism , Young Adult
11.
Antimicrob Agents Chemother ; 54(6): 2360-4, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20385857

ABSTRACT

The growing number of infections caused by multidrug-resistant pathogens has prompted a more rational use of available antibiotics given the paucity of new, effective agents. Monte Carlo simulations were utilized to determine the appropriateness of several doripenem dosing regimens based on the probability of attaining the critical drug exposure metric of time that drug concentrations remain above the drug MIC (T>MIC) for 35% (and lower thresholds) of the dosing interval in >80 to 90% of the population (T>MIC 35% target). This exposure level generally correlates with in vivo efficacy for carbapenems. In patients with creatinine clearance of >50 ml/min, a 500-mg dose of doripenem infused over 1 h every 8 h is expected to be effective against bacilli with doripenem MICs of < or =1 microg/ml based on a T>MIC 35% target and MICs of < or =2 microg/ml based on lower targets. A longer, 4-hour infusion time improved target attainment in most cases, such that the T>MIC was adequate for pathogens with doripenem MICs as high as 4 microg/ml. Efficacy is expected for infections caused by pathogens with doripenem MICs of < or =2 microg/ml in patients with moderate renal impairment (creatinine clearance, 30 to 50 ml/min) who receive doripenem at 250 mg infused over 1 h every 8 h and in patients with severe impairment (creatinine clearance between 10 and 29 ml/min) who receive doripenem at 250 mg, infused over 1 h or 4 h, every 12 h. Results of pharmacokinetics/pharmacodynamics (PK/PD) modeling can guide dose optimization, thereby potentially increasing the clinical efficacy of doripenem against serious Gram-negative bacterial infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/pharmacokinetics , Carbapenems/pharmacology , Carbapenems/pharmacokinetics , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/metabolism , Models, Biological , Anti-Bacterial Agents/administration & dosage , Carbapenems/administration & dosage , Creatinine/metabolism , Critical Illness , Doripenem , Drug Administration Schedule , Drug Resistance, Multiple, Bacterial , Gram-Negative Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/metabolism , Gram-Positive Bacterial Infections/microbiology , Humans , Infusions, Intravenous , Microbial Sensitivity Tests , Monte Carlo Method
12.
Antimicrob Agents Chemother ; 54(1): 375-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19858256

ABSTRACT

Levofloxacin was recently (May 2008) approved by the U.S. Food and Drug Administration as a treatment for children following inhalational exposure to anthrax. Given that no clinical trials to assess the efficacy of a chosen dose was conducted, the basis for the dose recommendation was based upon pharmacometric analyses. The objective of this paper is to describe the basis of the chosen pediatric dose recommended for the label. Pharmacokinetic (PK) data from 90 pediatric patients receiving 7 mg/kg of body weight levofloxacin and two studies of 47 healthy adults receiving 500 and 750 mg/kg levofloxacin were used for the pharmacometric analyses. Body weight was found to be a significant covariate for levofloxacin clearance and the volume of distribution. Consistently with developmental physiology, clearance also was found to be reduced in pediatric patients under 2 years of age due to immature renal function. Different dosing regimens were simulated to match adult exposure (area under the concentration-time curve from 0 to 24 h at steady state, maximum concentration of drug in serum at steady state, and minimum concentration of drug in serum at steady state) following the approved adult dose of 500 mg once a day. The recommended dose of 8 mg/kg twice a day was found to match the exposure of the dose approved for adults in a manner that permitted confidence that this dose in children would achieve efficacy comparable to that of adults.


Subject(s)
Anthrax/prevention & control , Anti-Bacterial Agents/administration & dosage , Levofloxacin , Ofloxacin/administration & dosage , Ofloxacin/therapeutic use , Adolescent , Aging/metabolism , Anthrax/microbiology , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/therapeutic use , Area Under Curve , Biological Availability , Body Weight/physiology , Child , Child, Preschool , Chromatography, High Pressure Liquid , Double-Blind Method , Female , Humans , Infant , Inhalation Exposure , Injections, Intravenous , Kidney Function Tests , Male , Ofloxacin/pharmacokinetics , Spores, Bacterial , Tablets
13.
Epilepsia ; 51(10): 1954-62, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20880232

ABSTRACT

PURPOSE: To identify and validate the efficacious monotherapy dosing regimen for topiramate in children aged 2 to <10 years with newly diagnosed epilepsy using pharmacokinetic-pharmacodynamic (PK-PD) modeling and simulation bridging. METHODS: Several models were developed in pediatric and adult populations to relate steady-state trough plasma concentrations (C(min)) of topiramate to the magnitude of clinical effect in monotherapy and adjunctive settings. These models were integrated to derive and support the monotherapy dosing regimen for pediatric patients. KEY FINDINGS: A two-compartmental population PK model with first-order absorption described the time course of topiramate C(min) as a function of dosing regimen. Disposition of topiramate was related to age, body weight, and use of various concomitant antiepileptic drugs. The PK-PD model for monotherapy indicated that the hazard of time to first seizure decreased with increasing C(min) and time since randomization. Higher baseline seizure frequency increased risk for seizures. Age did not significantly influence hazard of time to first seizure after randomization to monotherapy. For adjunctive therapy, the distribution of drug and placebo responses was not significantly different among age groups. Based on the available PK-PD modeling data, the dosing regimen expected to achieve a 65-75% seizure freedom rate after 1 year for pediatric patients age 2-10 years is approximately 6-9 mg/kg per day. SIGNIFICANCE: This analysis indicated no difference in PK-PD of topiramate between adult and pediatric patients. Effects of indication and body weight on PK were adequately integrated into the model, and monotherapy dosing regimens were identified for children 2-10 years of age.


Subject(s)
Anticonvulsants/administration & dosage , Anticonvulsants/pharmacokinetics , Epilepsy/drug therapy , Fructose/analogs & derivatives , Adolescent , Adult , Age Distribution , Age Factors , Aged , Aged, 80 and over , Anticonvulsants/therapeutic use , Body Weight , Child , Child, Preschool , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Therapy, Combination , Epilepsy/metabolism , Female , Fructose/administration & dosage , Fructose/pharmacokinetics , Fructose/therapeutic use , Humans , Male , Metabolic Clearance Rate , Middle Aged , Models, Biological , Randomized Controlled Trials as Topic/statistics & numerical data , Topiramate , Treatment Outcome
14.
AAPS J ; 22(5): 101, 2020 08 02.
Article in English | MEDLINE | ID: mdl-32743691

ABSTRACT

The concentration-QTc (C-QTc) analysis is often applied in the first-in-human (FIH) study to demonstrate the absence of a QTc effect in support of a TQT waiver. However, a C-QTc analysis without properly designed sensitivity could fail to conclude the absence of a QTc effect at high concentrations, even though the compound is QTc negative. This is because the 90% confidence interval (CI) of the model-derived ∆∆QTc grows wider with increasing concentration, and the upper-bound could cross the 10-ms threshold, even though the slope is close to 0. So far, there is no simple math formula to calculate the sensitivity/specificity of a C-QTc analysis. A PK/QTc trial simulation scheme was applied to optimize the design features of a C-QTc trial in FIH studies by evaluating the study's sensitivity over a wide concentration range, circumventing the problem of not knowing the target concentration during FIH studies. It was also used to ensure that the specificity of the trial was well-controlled. Simulation showed that the study sensitivity can be quantitatively gauged by optimizing the dose range, the number of samples per subjects or subject number, and by sampling around Tmax, and at steady-state. The specificity of the trial can also be evaluated with this approach, and it is important to combine model-derived ∆∆QTc and slope estimate in the evaluation. The trial simulation approach helps maximize the probability of success of C-QTc analyses in FIH studies intended to support a TQT waiver.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Dose-Response Relationship, Drug , Drug Evaluation/methods , Models, Theoretical , Humans , Prospective Studies , Research Design , Sensitivity and Specificity
15.
Clin Transl Sci ; 13(2): 309-317, 2020 03.
Article in English | MEDLINE | ID: mdl-31642608

ABSTRACT

Positron emission tomography (PET) provides useful information in target engagement or receptor occupancy in the brain for central nervous system (CNS) drug development, however, dose selection for human PET studies is challenging and largely empirical. Here, we describe a translational pharmacokinetic/pharmacodynamic (PK/PD) modeling work to inform dose selection for a human PET study of JNJ-54175446, a CNS-penetrating P2X7 receptor antagonist. Models were developed using data on monkey brain occupancy and plasma drug exposures from a monkey PET study and early human clinical studies that provided data on drug exposures and human ex vivo-stimulated peripheral interleukin (IL)-1ß release. The observed plasma PK of JNJ-54175446 in human was adequately described by a one-compartment model with parallel zero-order and first-order absorption and first-order elimination. An exposure-occupancy model was extrapolated from monkey to human assuming a similar unbound potency (all other model parameters remained unchanged). This model was then used to simulate human brain occupancy to guide human PET study dose selection, together with the human population PK model. The corroboration of model predicted occupancy by the observed occupancy data from the human PET study supports the use of a monkey as a predictive model for human PET target engagement. Potency estimate for brain occupancy was generally comparable to that for the suppression of the provoked peripheral IL-1ß release ex vivo, indicating that blood IL-1ß release may be used as a surrogate of central occupancy for JNJ-54175446. Translational PK/PD modeling approach could be used for selecting optimal doses for human PET and other clinical studies.


Subject(s)
Positron-Emission Tomography , Purinergic P2X Receptor Antagonists/pharmacokinetics , Pyridines/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Receptors, Purinergic P2X7/metabolism , Triazoles/pharmacokinetics , Adolescent , Adult , Aged , Animals , Brain/diagnostic imaging , Brain/metabolism , Clinical Trials, Phase I as Topic , Computer Simulation , Datasets as Topic , Dose-Response Relationship, Drug , Drug Development/methods , Female , Fluorine Radioisotopes/administration & dosage , Humans , Interleukin-1beta/metabolism , Macaca mulatta , Male , Middle Aged , Models, Biological , Purinergic P2X Receptor Antagonists/administration & dosage , Pyridines/administration & dosage , Radionuclide Imaging , Radiopharmaceuticals/administration & dosage , Randomized Controlled Trials as Topic , Triazoles/administration & dosage , Young Adult
16.
Antimicrob Agents Chemother ; 53(3): 1228-30, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19075059

ABSTRACT

Population pharmacokinetic analysis demonstrated that renal function, as assessed by creatinine clearance (CL(CR)), was the patient characteristic that had a clinically relevant impact on ceftobiprole pharmacodynamics. Dosing adjustments based on CL(CR) for subjects with renal impairment should provide ceftobiprole exposure similar to that in patients with normal renal function.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Cephalosporins/pharmacokinetics , Population , Skin Diseases, Infectious/drug therapy , Anti-Bacterial Agents/administration & dosage , Case-Control Studies , Cephalosporins/administration & dosage , Clinical Trials, Phase III as Topic , Computer Simulation , Creatinine/pharmacokinetics , Humans , Infusions, Intravenous , Kidney Function Tests , Male , Metabolic Clearance Rate , Microbial Sensitivity Tests , Models, Biological , Population/genetics , Software
17.
Antimicrob Agents Chemother ; 53(6): 2346-53, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19223624

ABSTRACT

Characterization of the time course and magnitude of enzyme induction due to multiple inducers is important for interpretation of clinical data from drug-drug interaction studies. A population interaction model was developed to quantify efavirenz autoinduction and further induction with concurrent carbamazepine coadministration. Efavirenz concentration data in the absence and presence of carbamazepine following single- and multiple-dose oral administrations in healthy subjects were used for model development. The proposed model was able to describe the time-dependent efavirenz autoinduction and the further induction with carbamazepine when the agents were combined. The estimated population averages of efavirenz oral clearance were 5.5, 9.4, 14.4, and 16.7 liters/h on days 1, 14, and 35 and at steady state for the interaction, respectively, for efavirenz monotherapy for 2 weeks followed by the coadministration of carbamazepine for 3 weeks. The estimated times to 50% of the steady state for efavirenz autoinduction and for the induction resulting from the concurrent administration of efavirenz and carbamazepine were similar (around 10 to 12 days). With this model-based analysis, efavirenz exposures can be projected prior to and at the steady state of induction, allowing a better understanding of the time course and magnitude of enzyme induction.


Subject(s)
Benzoxazines/pharmacology , Benzoxazines/pharmacokinetics , Carbamazepine/pharmacology , Adolescent , Adult , Alkynes , Area Under Curve , Aryl Hydrocarbon Hydroxylases/biosynthesis , Cyclopropanes , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP3A/biosynthesis , Drug Interactions , Enzyme Induction/drug effects , Female , Humans , Male , Middle Aged , Models, Biological , Oxidoreductases, N-Demethylating/biosynthesis
18.
Antimicrob Agents Chemother ; 53(8): 3371-4, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19528285

ABSTRACT

Ceftobiprole, a broad-spectrum cephalosporin with activity against methicillin (meticillin)-resistant staphylococci, was statistically noninferior to a combination of vancomycin plus ceftazidime in patients with complicated skin and skin structure infections (cSSSI). This analysis used data from this clinical trial to determine the relationship between therapeutic outcome and the percentage of time that the unbound ceftobiprole concentration exceeds the MIC (percent T>MIC). From the trial of ceftobiprole (500 mg every 8 h, 2-h infusion) for cSSSI due to gram-positive and/or gram-negative bacteria, data from 309 patients in the microbiological intent-to-treat analysis set with measured ceftobiprole concentrations and baseline MICs were used to assess the relationship between percent T>MIC and therapeutic outcome. Individual pharmacokinetic (PK) profiles were obtained from a three-compartment population PK model. The relationship between percent T>MIC and a clinical cure was determined. For the clinical trial dosing regimen, individual percent T>MICs were used to calculate fractional target attainment rates (TARs) for >or=30 and >or=50% T>MIC targets at various MICs. There was a statistically significant relationship between achieving a >or=30 or >or=50% T>MIC and a clinical cure (P = 0.003 and P = 0.007, respectively; Pearson's chi(2) test). The fractional TAR was greater than 90% at a MIC of MIC and a clinical cure with ceftobiprole was demonstrated. A ceftobiprole regimen of 500 mg every 8 h as a 2-h infusion has a high probability of achieving a target of >or=30 or >or=50% T>MIC for patients with cSSSI due to gram-positive and gram-negative pathogens.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Cephalosporins/therapeutic use , Skin Diseases, Infectious/drug therapy , Skin Diseases, Infectious/microbiology , Ceftazidime/therapeutic use , Female , Humans , Male , Microbial Sensitivity Tests , Treatment Outcome , Vancomycin/therapeutic use
19.
Ther Adv Cardiovasc Dis ; 13: 1753944719863641, 2019.
Article in English | MEDLINE | ID: mdl-31364490

ABSTRACT

BACKGROUND: This analysis aimed to evaluate the impact of rivaroxaban exposure and patient characteristics on efficacy and safety outcomes in patients with acute coronary syndrome (ACS) and to determine whether therapeutic drug monitoring might provide additional information regarding rivaroxaban dose, beyond what patient characteristics provide. METHODS: A post hoc exposure-response analysis was conducted using data from the phase III ATLAS ACS 2 Thrombolysis in Myocardial Infarction (TIMI) 51 study, in which 15,526 randomized ACS patients received rivaroxaban (2.5 mg or 5 mg twice daily) or placebo for a mean of 13 months (maximum follow up: 31 months). A multivariate Cox model was used to correlate individual predicted rivaroxaban exposures and patient characteristics with time-to-event clinical outcomes. RESULTS: For the incidence of myocardial infarction (MI), ischemic stroke, or nonhemorrhagic cardiovascular death, hazard ratios (HRs) for steady-state maximum plasma concentration (Cmax) in the 5th and 95th percentiles versus the median were statistically significant but close to 1 for both rivaroxaban doses. For TIMI major bleeding events, a statistically significant association was observed with Cmax [HR, 1.08; 95% CI, 1.06-1.11 (95th percentile versus median, 2.5 mg twice daily)], sex [HR, 0.56; 95% CI, 0.38-0.84 (female versus male)], and previous revascularization [HR, 0.62; 95% CI, 0.44-0.87 (no versus yes)]. CONCLUSIONS: The shallow slopes of the exposure-response relationships and the lack of a clear therapeutic window render it unlikely that therapeutic drug monitoring in patients with ACS would provide additional information regarding rivaroxaban dose beyond that provided by patient characteristics.


Subject(s)
Acute Coronary Syndrome/drug therapy , Factor Xa Inhibitors/administration & dosage , Models, Biological , Rivaroxaban/administration & dosage , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/diagnosis , Acute Coronary Syndrome/mortality , Aged , Brain Ischemia/mortality , Clinical Decision-Making , Clinical Trials, Phase III as Topic , Dose-Response Relationship, Drug , Drug Dosage Calculations , Drug Monitoring , Factor Xa Inhibitors/adverse effects , Factor Xa Inhibitors/pharmacokinetics , Female , Hemorrhage/chemically induced , Humans , Male , Middle Aged , Myocardial Infarction/mortality , Randomized Controlled Trials as Topic , Risk Assessment , Risk Factors , Rivaroxaban/adverse effects , Rivaroxaban/pharmacokinetics , Stroke/mortality , Treatment Outcome
20.
J Psychopharmacol ; 32(12): 1330-1340, 2018 12.
Article in English | MEDLINE | ID: mdl-30182786

ABSTRACT

BACKGROUND: Pharmacokinetics, pharmacodynamics and general safety and tolerability of JNJ-42847922, a selective orexin-2 receptor antagonist, were assessed in healthy subjects. METHODS: Five consecutive cohorts of healthy subjects were enrolled and received doses of 5-60 mg orally once daily over 10 days of JNJ-42847922 ( n=6) or placebo ( n=2). Concentrations of drug in plasma and urine were measured over 24 h after dosing on Days 1, 5 and 10. Observed- and self-reported somnolence was used to evaluate the principal pharmacodynamic effect of JNJ-42847922. A test battery to assess vigilance state, sedation and alertness was assessed at 4, 6 and 8 h after dosing. Safety assessments included recording of adverse events, vital signs, electrocardiograms, clinical laboratory assessments and suicidality per Columbia Suicide Severity Rating Scale. RESULTS: JNJ-42847922 was rapidly absorbed after the morning dose administration. The median tmax ranged from 0.5-1.5 h and mean t1/2 values from 2-3 h. At JNJ-42847922 dose levels ⩾20 mg, mean Cmax and mean area under the curve, values increased less than dose proportionally. At doses ⩾20 mg, JNJ-42847922 consistently induced somnolence on all study days. At four hours post-dose administration, dose levels >5 mg JNJ-42847922 were identified as sedating by the Addiction Research Center Inventory-49. Except for a mild decrease in attention (Bond and Lader Visual Analogue Scale Factor 1) at dose levels >10 mg at four hours post-dose, no clinically relevant changes in other central measures have been observed. JNJ-42847922 was well tolerated.


Subject(s)
Orexin Receptor Antagonists/administration & dosage , Orexin Receptors/drug effects , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , Sleepiness , Triazoles/administration & dosage , Administration, Oral , Adolescent , Adult , Area Under Curve , Dose-Response Relationship, Drug , Double-Blind Method , Female , Half-Life , Humans , Male , Middle Aged , Orexin Receptor Antagonists/pharmacokinetics , Orexin Receptor Antagonists/pharmacology , Orexin Receptors/metabolism , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Pyrroles/pharmacokinetics , Pyrroles/pharmacology , Time Factors , Triazoles/pharmacokinetics , Triazoles/pharmacology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL