Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
J Clin Pharmacol ; 58(7): 952-964, 2018 07.
Article in English | MEDLINE | ID: mdl-29505101

ABSTRACT

Nonclinical assays with JNJ-54861911, a ß-secretase 1 inhibitor have indicated that at high concentrations, it may delay cardiac repolarization. A 4-way crossover thorough QT (TQT) study was performed in 64 healthy subjects with 50 and 150 mg JNJ-54861911 once daily for 7 days, placebo, and 400 mg moxifloxacin. Retrospective high-precision QT (HPQT) analysis was performed on serial elecrocardiograms extracted from first-in-human single-ascending dose (SAD) and multiple-ascending dose (MAD) studies to evaluate if early studies could detect and predict QT effect. In the TQT study, a high therapeutic 50 mg dose did not cause QT prolongation, and an effect >10 milliseconds could be excluded at all postdose timepoints. QT prolongation with peak effect on placebo-corrected change from baseline QTcF of 15.5 milliseconds (90%CI, 12.9-18.1 milliseconds) was observed following a supratherapeutic dose (150 mg). No clinically relevant QT changes were observed in earlier studies. However, with SAD/MAD findings by HPQT, the slope of the exposure-response (ER) relationship in the SAD study (doses up to 150 mg) was similar to the TQT study slope, and the estimated QT effect was comparable at high plasma levels. In the MAD study, doses up to 90 mg once daily for 7 days resulted in JNJ-54861911 peak plasma concentrations (Cmax ) comparable to those in the SAD study (∼750 ng/mL), but ER by HPQT failed to detect a QT effect and resulted in negative estimations. Adding a higher dose cohort (150 mg; Cmax , 1125 ng/mL) demonstrated a QT effect, with a slightly lower ER slope than the TQT study. JNJ-54861911 (up to 50 mg) did not cause QT prolongation at clinically relevant plasma concentrations in any studies. Provided sufficiently high plasma concentrations were captured, mild QT prolongation observed postdose with a supratherapeutic dose could be detected (TQT study) and estimated in SAD/MAD studies. Based on population pharmacokinetic modeling and simulation, 5 and 25 mg doses are currently considered for further phase 3 studies and are expected not to cause any relevant QT prolongation.


Subject(s)
Heart Rate/drug effects , Heart/drug effects , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Thiazines/administration & dosage , Thiazines/pharmacokinetics , Adult , Aged , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Electrocardiography/drug effects , Female , Healthy Volunteers , Humans , Male , Middle Aged , Models, Biological , Moxifloxacin , Pyridines/blood , Retrospective Studies , Thiazines/blood
10.
J Clin Pharmacol ; 57(12): 1607-1615, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28679020

ABSTRACT

US prescribing guidelines recommend that 15- and 20-mg doses of rivaroxaban be administered with food for the treatment of deep vein thrombosis (DVT) and pulmonary embolism (PE) and for reduction in the risk of recurrence of DVT and PE. In addition, the US prescribing guidelines recommend these doses be administered with an evening meal to reduce the risk of stroke and systemic embolism in patients with nonvalvular atrial fibrillation (AF). The purpose of this model-based cross-study comparison was to examine the impact of food, with regard to both meal timing and content, on the pharmacokinetics (PK) of rivaroxaban, using data collected during its clinical development. Results of this analysis showed that a PK model built from pooled data in the AF population (for whom rivaroxaban was administered with an evening meal) and in the DVT population (for whom rivaroxaban was administered with a morning meal) can describe both data sets well. Furthermore, the PK model built from data in the AF population alone can adequately predict the PK profile of the DVT population and vice versa. This cross-study analysis also confirmed the findings from previous clinical pharmacology studies, which showed that meal content does not have a clinically relevant impact on the PK of rivaroxaban at 20 mg. Therefore, although the administration of rivaroxaban with food is necessary for maintaining high bioavailability, neither meal timing nor meal content appears to affect the PK of rivaroxaban.


Subject(s)
Factor Xa Inhibitors/administration & dosage , Factor Xa Inhibitors/pharmacokinetics , Food-Drug Interactions , Rivaroxaban/administration & dosage , Rivaroxaban/pharmacokinetics , Adult , Aged , Aged, 80 and over , Area Under Curve , Computer Simulation , Drug Administration Schedule , Female , Humans , Male , Meals , Middle Aged , Models, Biological
11.
Clin Pharmacokinet ; 56(1): 55-63, 2017 01.
Article in English | MEDLINE | ID: mdl-27324190

ABSTRACT

BACKGROUND AND OBJECTIVES: Recent analysis revealed strong associations between prostate-specific antigen (PSA) dynamics and overall survival (OS) in metastatic castration-resistant prostate cancer (mCRPC) and supported PSA dynamics as bridging surrogacy endpoints for clinical benefit from treatment with abiraterone acetate plus prednisone. This analysis aimed to investigate the abiraterone exposure-PSA dynamics relationship in mCRPC. METHODS: Abiraterone pharmacokinetics-PSA models were constructed using data from the COU-AA-301 (chemotherapy-pretreated) and COU-AA-302 (chemotherapy-naïve) trials comparing abiraterone acetate 1000 mg/day plus prednisone 5 mg twice daily with prednisone alone in mCRPC. The drug effect-PSA dynamics relationship was modeled as a function of selected pharmacokinetic measures. The influences of baseline demographic variables, laboratory values, and disease status on PSA dynamics were assessed. RESULTS: A tumor growth inhibition model best described PSA dynamics post-treatment with abiraterone acetate. Abiraterone acetate treatment in chemotherapy-pretreated and chemotherapy-naïve patients increased the PSA decay rate (k dec) to the same extent (1.28-fold, 95 % confidence interval [CI] 0.58-1.98; and 0.93-fold, 95 % CI 0.6-1.27, respectively). Lower baseline lactate dehydrogenase and higher baseline testosterone significantly increased k dec. Findings from our analysis suggest a maximum-effect relationship between abiraterone trough concentration and PSA dynamics in both patient populations. The majority of patients had a steady-state trough concentration greater than the estimated half maximal effective concentration. CONCLUSION: The model appropriately described the exposure-response relationship between abiraterone and PSA dynamics in chemotherapy-pretreated and chemotherapy-naïve patients following oral administration of abiraterone acetate.


Subject(s)
Abiraterone Acetate/pharmacokinetics , Models, Biological , Prednisone/pharmacokinetics , Prostate-Specific Antigen/drug effects , Prostatic Neoplasms, Castration-Resistant/drug therapy , Abiraterone Acetate/administration & dosage , Area Under Curve , Drug Therapy, Combination , Humans , L-Lactate Dehydrogenase/blood , Male , Prednisone/administration & dosage , Prednisone/pharmacology , Severity of Illness Index , Socioeconomic Factors , Testosterone/blood
12.
Clin Drug Investig ; 37(3): 273-283, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27853999

ABSTRACT

BACKGROUND AND OBJECTIVES: Tapentadol extended release (ER) is approved for the management of chronic and acute pain in adults. There has been no report of tapentadol ER pharmacokinetics in subjects with cancer pain. This analysis investigated tapentadol ER pharmacokinetics in Japanese patients with cancer pain and bridged it with the pharmacokinetics in Japanese healthy subjects and Caucasian patients with cancer pain. METHODS: Nonlinear mixed-effect pharmacokinetic modeling was conducted based on pooled tapentadol ER concentration data collected in five Phase 1 studies from 138 Japanese and Korean healthy subjects and in two Phase 3 studies from 215 Japanese and Korean subjects with cancer pain. Expected tapentadol exposure in subjects with different characteristics was assessed via simulation. Tapentadol ER exposures in Caucasian populations were compared with those in corresponding Japanese populations. RESULTS: Tapentadol ER pharmacokinetics in Japanese cancer-pain patients were adequately described by a time-invariant, one-compartment disposition model with two input functions and first-order elimination. Weight, age, and albumin were identified as statistically significant covariates, but do not warrant dose adjustment. Comparable pharmacokinetics were shown between Japanese healthy and Caucasian healthy subjects, and between Japanese cancer-pain patients and Caucasian cancer-pain patients. CONCLUSION: The apparent differences in the estimated individual pharmacokinetic parameters in Japanese healthy subjects and Japanese cancer-pain patients taking tapentadol ER were explained by covariates incorporated in a unified pharmacokinetic model. Population modeling was essential in this cross-population bridging analysis.


Subject(s)
Cancer Pain/drug therapy , Models, Biological , Phenols/administration & dosage , Adult , Aged , Aged, 80 and over , Delayed-Action Preparations , Female , Humans , Male , Middle Aged , Phenols/pharmacokinetics , Tapentadol , White People , Young Adult
13.
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
14.
Clin Drug Investig ; 36(3): 213-23, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26798023

ABSTRACT

BACKGROUND AND OBJECTIVE: Tapentadol is a centrally acting analgesic with two mechanisms of action, µ-opioid receptor agonism and noradrenaline reuptake inhibition. The objectives were to describe the pharmacokinetic behavior of tapentadol after oral administration of an extended-release (ER) formulation in healthy subjects and patients with chronic pain and to evaluate covariate effects. METHODS: Data were obtained from 2276 subjects enrolled in five phase I and nine phase II and III studies. Nonlinear mixed-effects modeling was conducted using NONMEM. RESULTS: The population estimates of apparent oral clearance and apparent central volume of distribution were 257 L/h and 1870 L, respectively. The complex absorption was described with a transit compartment for the first input. The second input function embraces saturable "binding" in the "absorption compartment", and a time-varying rate constant. Covariate evaluation demonstrated that age, aspartate aminotransferase, and health (painful diabetic neuropathy or not) had a statistically significant effect on apparent clearance, and bioavailability appeared to be dependent on body weight. The pcVPC indicted that the model provided a robust and unbiased fit to the data. CONCLUSIONS: A one-compartment disposition model with two input functions and first-order elimination adequately described the pharmacokinetics of tapentadol ER. The dose-dependency in the pharmacokinetics of tapentadol ER is adequately described by the absorption model. None of the covariates were considered as clinically relevant factors that warrant dose adjustments.


Subject(s)
Analgesics/pharmacokinetics , Chronic Pain/drug therapy , Models, Biological , Phenols/pharmacokinetics , Administration, Oral , Adult , Aged , Aged, 80 and over , Analgesics/administration & dosage , Biological Availability , Chemistry, Pharmaceutical , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Delayed-Action Preparations , Female , Humans , Male , Middle Aged , Phenols/administration & dosage , Tapentadol , Young Adult
15.
Curr Med Res Opin ; 31(11): 2043-54, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26306819

ABSTRACT

OBJECTIVE: This commentary summarizes recommended dosing strategies for a recently developed 3 monthly long-acting injectable 1 (LAI) formulation of paliperidone palmitate (PP3M) for the treatment of schizophrenia in adults. METHODS: Recommendations for different dosing scenarios are based on the pharmacokinetic, efficacy and safety outcomes from phase 1 and phase 3 studies, population pharmacokinetic models, and model based simulations. RESULTS: Switching to PP3M treatment is recommended only in patients previously treated with once monthly paliperidone palmitate LAI (PP1M) for at least 4 months. The first injection of PP3M (175 to 525 mg equivalent [eq.]) should be given at the time of next scheduled injection of PP1M as a 3.5-fold multiple of the last PP1M dose (50-150 mg eq.), with a dosing window of ± 1 week. Following that first injection of PP3M, once-every-three-months maintenance injections with PP3M are recommended, with a dosing window of ± 2 weeks. The doses of PP3M can be administered in either deltoid (≥ 90 kg: 1.5 inch 22 G needle; <90 kg: 1.0 inch 22 G needle) or gluteal muscles (1.5 inch 22 G needle regardless of weight). In patients with mild renal impairment (creatinine clearance: 50-80 mL/min), a 25% dose reduction in PP1M and subsequent switching to a corresponding 3.5-dose multiple of PP3M (but not exceeding 350 mg eq.) is recommended. Appropriate dosing is recommended in elderly patients with diminished renal function not exceeding mild renal impairment. Similarly to PP1M, PP3M is not recommended in patients with moderate/severe renal impairment. Like PP1M, no dosage adjustment is required in patients with mild or moderate hepatic impairment or elderly patients with normal renal function. CONCLUSIONS: These data provide clinical guidelines for the optimum use of PP3M in patients with schizophrenia previously treated with PP1M for at least 4 months. REGISTRATION: ClinicalTrials.gov identifier: NCT01559272 and NCT01529515.


Subject(s)
Antipsychotic Agents/administration & dosage , Paliperidone Palmitate/administration & dosage , Schizophrenia/drug therapy , Adult , Antipsychotic Agents/therapeutic use , Chemistry, Pharmaceutical , Humans , Injections, Intramuscular , Muscle, Skeletal
16.
Rev Sci Instrum ; 86(3): 033306, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25832222

ABSTRACT

The vacuum system of the Room Temperature (K = 130) Cyclotron of Variable Energy Cyclotron Centre is comprised of vacuum systems of main machine and Beam Transport System. The vacuum control system is upgraded to a PLC based Automated system from the initial relay based Manual system. The supervisory control of the vacuum system is implemented in Experimental Physics and Industrial Control System (EPICS). An EPICS embedded ARM based vacuum gauge controller is developed to mitigate the requirement of vendor specific gauge controller for gauges and also for seamless integration of the gauge controllers with the control system. A set of MS-Windows ActiveX components with embedded EPICS Channel Access interface are developed to build operator interfaces with less complex programming and to incorporate typical Windows feature, e.g., user authentication, file handling, better fonts, colors, mouse actions etc. into the operator interfaces. The control parameters, monitoring parameters, and system interlocks of the system are archived in MySQL based EPICS MySQL Archiver developed indigenously. In this paper, we describe the architecture, the implementation details, and the performance of the system.

17.
Clin Cancer Res ; 21(14): 3170-7, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25829400

ABSTRACT

PURPOSE: We constructed a biomarker-survival modeling framework to explore the relationship between prostate-specific antigen (PSA) kinetics and overall survival (OS) in metastatic castration-resistant prostate cancer (mCRPC) patients following oral administration of 1,000 mg/day of abiraterone acetate (AA). EXPERIMENTAL DESIGN: The PSA-survival modeling framework was based on data from two phase III studies, COU-AA-301 (chemotherapy pretreated, n = 1,184) and COU-AA-302 (chemotherapy naïve, n = 1,081), and included a mixed-effects tumor growth inhibition model and a Cox proportional hazards survival model. RESULTS: The effect of AA on PSA kinetics was significant (P < 0.0001) and comparable between the chemotherapy-naïve and -pretreated patients. PSA kinetics [e.g., PSA nadir, PSA response rate (≥30%, 50%, and 90%), time to PSA progression, PSA doubling time (PSADT)] were highly associated with OS in both populations. The model-based posttreatment PSADT had the strongest association with OS (HR ∼0.9 in both populations). The models could accurately predict survival outcomes. After adjusting for PSA kinetic endpoints, the treatment effect of AA on survival was no longer statistically significant in both studies, and the Prentice criteria of surrogacy were met for the PSA kinetic endpoints. A strong correlation was also observed between PSA and radiographic progression-free survival. CONCLUSIONS: The analysis revealed a consistent treatment effect of AA on PSA kinetics and strong associations between PSA kinetics and OS in chemotherapy-pretreated and -naïve patients, thereby providing a rationale to consider PSA kinetics as surrogacy endpoints to indicate clinical benefit in AA-treated patients with mCRPC regardless of chemotherapy treatment.


Subject(s)
Abiraterone Acetate/therapeutic use , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/blood , Prostate-Specific Antigen/blood , Prostatic Neoplasms, Castration-Resistant/drug therapy , Antineoplastic Agents, Hormonal/therapeutic use , Double-Blind Method , Drug Resistance, Neoplasm , Humans , Kaplan-Meier Estimate , Kinetics , Male , Proportional Hazards Models , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/mortality
18.
J Clin Pharmacol ; 55(3): 269-78, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25212288

ABSTRACT

The objective was to develop a quantitative model of disease progression of knee osteoarthritis over 6 years using the total WOMAC score from patients enrolled into the Osteoarthritis Initiative (OAI) study. The analysis was performed using data from the Osteoarthritis Initiative database. The time course of the total WOMAC score of patients enrolled into the progression cohort was characterized using non-linear mixed effect modeling in NONMEM. The effect of covariates on the status of the disease and the progression rate was investigated. The final model provided a good description of the experimental data using a linear progression model with a common baseline (19 units of the total WOMAC score). The WOMAC score decreased by 1.77 units/year in 89% of the population or increased by 1.74 units/year in 11% of the population. Multiple covariates were found to affect the baseline and the rate of progression, including BMI, sex, race, the use of pain medications, and the limitation in activity due to symptoms. A mathematical model to describe the disease progression of osteoarthritis in the studied population was developed. The model identified two sub-populations with increasing or decreasing total WOMAC score over time, and the effect of important covariates was quantified.


Subject(s)
Knee Joint , Osteoarthritis, Knee/diagnosis , Aged , Databases, Factual , Disability Evaluation , Disease Progression , Female , Follow-Up Studies , Health Status , Humans , Incidence , Knee Joint/pathology , Knee Joint/physiopathology , Linear Models , Male , Middle Aged , Multivariate Analysis , Nonlinear Dynamics , Osteoarthritis, Knee/epidemiology , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/physiopathology , Pain Measurement , Prospective Studies , Risk Factors , Severity of Illness Index , Surveys and Questionnaires , Time Factors
19.
Clin Pharmacokinet ; 53(12): 1149-60, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25204404

ABSTRACT

BACKGROUND AND OBJECTIVES: Abiraterone acetate, an androgen biosynthesis inhibitor, prolongs survival in men with metastatic castration-resistant prostate cancer (mCRPC) in the pre- and post-chemotherapy setting as demonstrated by the pivotal phase III studies COU-AA-301 and COU-AA-302. We performed population pharmacokinetic analyses to estimate pharmacokinetic parameters after oral administration of 1,000 mg/day of abiraterone acetate in patients with mCRPC, with or without prior chemotherapy, and after a single 1,000 mg dose in healthy volunteers. The study objectives were to determine consistency between patient populations and to characterize factors that may influence abiraterone pharmacokinetics. METHODS: Studies in this analysis included COU-AA-302 (chemotherapy naïve); COU-AA-301 and COU-AA-006 (chemotherapy pretreated); and COU-AA-008, COU-AA-009, and COU-AA-014 (healthy subjects). A total of 4,627 plasma concentrations from 359 subjects (62 healthy volunteers, 297 patients) were analyzed using non-linear mixed-effects modeling. RESULTS: An Erlang-type absorption model with first-order elimination and three-transit compartments following sequential zero- and first-order processes was used to characterize abiraterone pharmacokinetics. Absorption-related parameters were affected by food intake. Abiraterone pharmacokinetics were characterized by an extensive apparent clearance, which was lower in patients with mCRPC (1,550 L/h) versus healthy subjects (2,240 L/h), and by large apparent central (5,620 L) and peripheral (17,400 L) volumes of distribution. Abiraterone pharmacokinetics were similar in chemotherapy-pretreated and -naïve patients and were characterized by a high between- and within-subject variability [e.g., between-subject coefficient of variation (CV%) for relative bioavailability for the modified fasting state was 61.1% and the CV% for within-subject variability was 71.3%]. The fat content of food taken with abiraterone acetate affected the bioavailability of abiraterone. No factors beyond food intake and health status (healthy vs. mCRPC) impacted abiraterone pharmacokinetics. CONCLUSIONS: Based on the pharmacokinetics model, the recommended 1,000 mg/day of abiraterone acetate resulted in similar abiraterone exposure for patients with mCRPC regardless of prior chemotherapy. The fat content of food affected relative bioavailability of abiraterone, though the extent of this effect is dependent on health status.


Subject(s)
Androstenes/pharmacokinetics , Models, Biological , Prostatic Neoplasms, Castration-Resistant/metabolism , Adult , Aged , Aged, 80 and over , Androstenes/blood , Androstenes/therapeutic use , Antineoplastic Agents/therapeutic use , Biological Availability , Cross-Over Studies , Dietary Fats/administration & dosage , Docetaxel , Double-Blind Method , Fasting/metabolism , Food-Drug Interactions , Healthy Volunteers , Humans , Male , Middle Aged , Prostatic Neoplasms, Castration-Resistant/drug therapy , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Taxoids/therapeutic use , Young Adult
20.
AAPS J ; 16(6): 1271-81, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25165039

ABSTRACT

Mixed-effects beta regression (BR), boundary-inflated beta regression (ZOI), and coarsening model (CO) were investigated for analyzing bounded outcome scores with data at the boundaries in the context of Alzheimer's disease. Monte Carlo simulations were conducted to simulate disability assessment for dementia (DAD) scores using these three models, and each set of simulated data were analyzed by the original simulation model. One thousand trials were simulated, and each trial contained 250 subjects. For each subject, DAD scores were simulated at baseline, 13, 26, 39, 52, 65, and 78 weeks. The simulation-reestimation exercise showed that all the three models could reasonably recover their true parameter values. The bias of the parameter estimates of the ZOI model was generally less than 1%, while the bias of the CO model was mainly within 5%. The bias of the BR model was slightly higher, i.e., less than or in the order of 20%. In the application to real-world DAD data from clinical studies, examination of prediction error and visual predictive check (VPC) plots suggested that both BR and ZOI models had similar predictive performance and described the longitudinal progression of DAD slightly better than the CO model. In conclusion, the investigated three modeling approaches may be sensible choices for bounded outcome scores with data on the edges. Prediction error and VPC plots can be used to identify the model with best predictive performance.


Subject(s)
Alzheimer Disease/diagnosis , Disability Evaluation , Models, Statistical , Severity of Illness Index , Alzheimer Disease/psychology , Bias , Computer Simulation , Disease Progression , Humans , Regression Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...