Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am Heart J ; 213: 97-104, 2019 07.
Article in English | MEDLINE | ID: mdl-31132584

ABSTRACT

BACKGROUND: The Fontan procedure is the final step of the 3-stage palliative procedure commonly performed in children with single ventricle physiology. Thrombosis remains an important complication in children after this procedure. To date, guideline recommendations for the type and duration of thromboprophylaxis after Fontan surgery are mainly based on extrapolation of knowledge gained from adults at risk for thrombosis in other clinical settings. Warfarin is being used off-label, and because of its multiple interactions with other drugs and food, a new alternative is highly desirable. Rivaroxaban, a direct Factor Xa inhibitor with a predictable pharmacokinetic profile, is a candidate to address this medical need. STUDY DESIGN: The UNIVERSE study is a prospective, open-label, active-controlled, multicenter study in children 2 to 8 years of age who have single ventricle physiology and had the Fontan procedure within the 4 months preceding enrollment. This study consists of 2 parts. In Part A, rivaroxaban pharmacokinetics, pharmacodynamics, safety, and tolerability are assessed to validate the pediatric dosing selected. In Part B, safety and efficacy of rivaroxaban versus acetylsalicylic acid are evaluated for thromboprophylaxis in children post-Fontan procedure. Children in each part will receive study drug for 12 months. Part A has been completed with 12 children enrolled. Enrollment into Part B is currently ongoing. CONCLUSIONS: The UNIVERSE study aims to provide dosing, pharmacokinetics/pharmacodynamics, safety, and efficacy information on the use of rivaroxaban, an oral anticoagulant, versus acetylsalicylic acid, an antiplatelet agent, in children with single ventricle physiology after the Fontan procedure.


Subject(s)
Aspirin/administration & dosage , Factor Xa Inhibitors/administration & dosage , Fibrinolytic Agents/administration & dosage , Fontan Procedure/adverse effects , Postoperative Complications/prevention & control , Randomized Controlled Trials as Topic , Rivaroxaban/administration & dosage , Thrombosis/prevention & control , Aspirin/pharmacokinetics , Child , Child, Preschool , Factor Xa Inhibitors/pharmacokinetics , Female , Fibrinolytic Agents/pharmacokinetics , Humans , Male , Multicenter Studies as Topic , Prospective Studies , Rivaroxaban/pharmacokinetics
2.
J Clin Psychopharmacol ; 39(6): 567-574, 2019.
Article in English | MEDLINE | ID: mdl-31688450

ABSTRACT

BACKGROUND: Pharmacokinetic-pharmacodynamic (PK/PD) models were developed to describe the relationship between the time course of paliperidone plasma concentrations and the risk of relapse of schizophrenia symptoms following administration of paliperidone palmitate 1-month (PP1M) and 3-month (PP3M) long-acting injectables, and to identify relevant covariates for relapse and dropout events. METHODS: Patient data from two global phase 3, relapse prevention studies comparing PP3M to placebo (study A) and PP3M to PP1M (study B) were analyzed. Dropout and relapse data were assessed using survival analysis as two separate single time-to-event models. Baseline covariates included age, sex, race/country, duration of illness, previous hospitalizations, prior use of long-acting injectables and use of multiple (≥2) antipsychotics at screening. RESULTS: The PK/PD analysis data set included 305 patients who were randomized to receive PP3M or placebo in the double-blind phase of study A and 1002 patients randomized to receive PP3M or PP1M in the double-blind phase of study B. Risk of relapse decreased with increasing paliperidone concentrations for both PP1M and PP3M, while it appeared to increase in patients with higher number of previous hospitalizations and/or with higher prerandomization (trough) paliperidone concentration (study A), and in patients on concomitant benzodiazepine medication and/or at Japan centers (study B). These findings are reflective of different illness severity in the population and of differences in medical practice for Japanese patients. In model-based simulations, PP3M and PP1M displayed similar relapse rates over time. CONCLUSIONS: This PK/PD analysis confirmed that PP1M and PP3M provide comparable efficacy in terms of relapse prevention, and that PP3M is superior to placebo. The PK/PD models presented here may as well be applied to studies with similar designs as either study A or B.


Subject(s)
Antipsychotic Agents/administration & dosage , Antipsychotic Agents/pharmacokinetics , Mental Disorders/drug therapy , Outcome Assessment, Health Care , Paliperidone Palmitate/administration & dosage , Paliperidone Palmitate/pharmacokinetics , Patient Dropouts , Secondary Prevention , Adult , Delayed-Action Preparations , Double-Blind Method , Female , Humans , Injections , Male , Middle Aged , Recurrence , Severity of Illness Index
3.
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
4.
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
5.
Clin Pharmacol Ther ; 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38519861

ABSTRACT

A recent industry perspective published in this journal describes the benefits received by drug companies from participation in the MIDD Pilot Program. Along with the primary objectives of supporting good decision-making in drug development, there were substantial savings in time and development costs. Furthermore, many sponsors reported qualitative benefits such as new learnings and clarity on MIDD strategies and methodology that could be applied to other development programs. Based on the success of the Pilot Program, the FDA recently announced the continuation of the MIDD Paired Meeting Program as part of the Prescription Drug User Fee Act (PDUFA VII). In this report, we describe the collective experiences of industry participants in the MIDD Program to date, including all aspects of the process from meeting request submission to follow-up actions. The purpose is to provide future participants with information to optimize the value of the MIDD Program.

6.
Br J Clin Pharmacol ; 75(1): 146-61, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22534009

ABSTRACT

AIM: The objective is to develop a semi-mechanistic disease progression model for mild cognitive impairment (MCI) subjects. The model aims to describe the longitudinal progression of ADAS-cog scores from the Alzheimer's disease neuroimaging initiative trial that had data from 198 MCI subjects with cerebrospinal fluid (CSF) information who were followed for 3 years. METHOD: Various covariates were tested on disease progression parameters and these variables fell into six categories: imaging volumetrics, biochemical, genetic, demographic, cognitive tests and CSF biomarkers. RESULTS: CSF biomarkers were associated with both baseline disease score and disease progression rate in subjects with MCI. Baseline disease score was also correlated with atrophy measured using hippocampal volume. Progression rate was also predicted by executive functioning as measured by the Trail B-test. CONCLUSION: CSF biomarkers have the ability to discriminate MCI subjects into sub-populations that exhibit markedly different rates of disease progression on the ADAS-cog scale. These biomarkers can therefore be utilized for designing clinical trials enriched with subjects that carry the underlying disease pathology.


Subject(s)
Biomarkers/cerebrospinal fluid , Cognitive Dysfunction/cerebrospinal fluid , Aged , Aged, 80 and over , Alzheimer Disease/cerebrospinal fluid , Apolipoproteins E/genetics , Cholesterol/blood , Disease Progression , Female , Humans , Male , Middle Aged , Neuroimaging
7.
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
8.
Alzheimers Dement ; 9(1 Suppl): S21-31, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23127469

ABSTRACT

BACKGROUND: The Alzheimer's Disease Assessment Scale-Cognitive (ADAS-Cog) has been used widely as a cognitive end point in Alzheimer's Disease (AD) clinical trials. Efforts to treat AD pathology at earlier stages have also used ADAS-Cog, but failure in these trials can be difficult to interpret because the scale has well-known ceiling effects that limit its use in mild cognitive impairment (MCI) and early AD. A wealth of data exists in ADAS-Cog from both historical trials and contemporary longitudinal natural history studies that can provide insights about parts of the scale that may be better suited for MCI and early AD trials. METHODS: Using Alzheimer's Disease Neuroimaging Initiative study data, we identified the most informative cognitive measures from the ADAS-Cog and other available scales. We used cross-sectional analyses to characterize trajectories of ADAS-Cog and its individual subscales, as well as other cognitive, functional, or global measures across disease stages. Informative measures were identified based on standardized mean of 2-year change from baseline and were combined into novel composite endpoints. We assessed performance of the novel endpoints based on sample size requirements for a 2-year clinical trial. A bootstrap validation procedure was also undertaken to assess the reproducibility of the standardized mean changes of the selected measures and the corresponding composites. RESULTS: All proposed novel endpoints have improved standardized mean changes and thus improved statistical power compared with the ADAS-Cog 11. Further improvements were achieved by using cognitive-functional composites. Combining the novel composites with an enrichment strategy based on cerebral spinal fluid beta-amyloid (Aß(1-42)) in a 2-year trial yielded gains in power of 20% to 40% over ADAS-Cog 11, regardless of the novel measure considered. CONCLUSION: An empirical, data-driven approach with existing instruments was used to derive novel composite scales based on ADAS-Cog 11 with improved performance characteristics for MCI and early AD clinical trials. Together with patient enrichment based on Aß(1-42) pathology, these modified endpoints may allow more efficient clinical trials in these populations and can be assessed without modifying current test administration procedures in ongoing trials.


Subject(s)
Alzheimer Disease/diagnosis , Cognitive Dysfunction/diagnosis , Early Diagnosis , Aged , Alzheimer Disease/psychology , Clinical Trials as Topic , Female , Humans , Male , Neuropsychological Tests
9.
Target Oncol ; 18(5): 667-684, 2023 09.
Article in English | MEDLINE | ID: mdl-37713090

ABSTRACT

BACKGROUND: Teclistamab, a B-cell maturation antigen × CD3 bispecific antibody, is approved in patients with relapsed/refractory multiple myeloma (RRMM) who have previously received an immunomodulatory agent, a proteasome inhibitor, and an anti-CD38 antibody. OBJECTIVE: We report the population pharmacokinetics of teclistamab administered intravenously and subcutaneously (SC) and exposure-response relationships from the phase I/II, first-in-human, open-label, multicenter MajesTEC-1 study. METHODS: Phase I of MajesTEC-1 consisted of dose escalation and expansion at the recommended phase II dose (RP2D; 1.5 mg/kg SC weekly, preceded by step-up doses of 0.06 and 0.3 mg/kg); phase II investigated the efficacy of teclistamab RP2D in patients with RRMM. Population pharmacokinetics and the impact of covariates on teclistamab systemic exposure were assessed using a 2-compartment model with first-order absorption for SC and parallel time-independent and time-dependent elimination pathways. Exposure-response analyses were conducted, including overall response rate (ORR), duration of response (DoR), progression-free survival (PFS), overall survival (OS), and the incidence of grade ≥ 3 anemia, neutropenia, lymphopenia, leukopenia, thrombocytopenia, and infection. RESULTS: In total, 4840 measurable serum concentration samples from 338 pharmacokinetics-evaluable patients who received teclistamab were analyzed. The typical population value of time-independent and time-dependent clearance were 0.449 L/day and 0.547 L/day, respectively. The time-dependent clearance decreased rapidly to < 10% after 8 weeks of teclistamab treatment. Patients who discontinue teclistamab after the 13th dose are expected to have a 50% reduction from Cmax in teclistamab concentration at a median (5th to 95th percentile) time of 15 days (7-33 days) after Tmax and a 97% reduction from Cmax in teclistamab concentration at a median time of 69 days (32-163 days) after Tmax. Body weight, multiple myeloma type (immunoglobulin G vs non-immunoglobulin G), and International Staging System (ISS) stage (II vs I and III vs I) were statistically significant covariates on teclistamab pharmacokinetics; however, these covariates had no clinically relevant effect on the efficacy of teclistamab at the RP2D. Across all doses, ORR approached a plateau at the concentration range associated with RP2D, and in patients who received the RP2D, a flat exposure-response curve was observed. No apparent relationship was observed between DoR, PFS, OS, and the incidence of grade ≥3 adverse events across the predicted exposure quartiles. CONCLUSION: Body weight, myeloma type, and ISS stage impacted systemic teclistamab exposure without any clinically relevant effect on efficacy. The exposure-response analyses for ORR showed a positive trend with increasing teclistamab systemic exposure, with a plateau at the RP2D, and there was no apparent exposure-response trend for safety or other efficacy endpoints. These analyses support the RP2D of teclistamab in patients with RRMM. CLINICAL TRIAL REGISTRATION: NCT03145181 (phase I, 09 May 2017); NCT04557098 (phase II, 21 September 2020).


Subject(s)
Antineoplastic Agents , Multiple Myeloma , Neutropenia , Humans , Multiple Myeloma/drug therapy , Proteasome Inhibitors , Body Weight
10.
BMC Psychiatry ; 12: 26, 2012 Mar 28.
Article in English | MEDLINE | ID: mdl-22455454

ABSTRACT

BACKGROUND: There are no previous reports of paliperidone palmitate's (PP) long term tolerability or pharmacokinetics of the highest dose in patients with schizophrenia. This study evaluates safety and tolerability, as well as pharmacokinetics, of the highest marketed dose of PP (150 mg eq. [234 mg]) in stable patients with schizophrenia over a 1-year period. METHODS: In this 1-year prospective study, eligible patients (aged 18-65 years; Positive and Negative Syndrome Scale's total score ≤ 70) received an initial deltoid injection of PP 150 mg eq. The second injection one week later and subsequent once-monthly injections were deltoid or gluteal. All injections were to be PP 150 mg eq. Patients willing to participate in intensive pharmacokinetic sampling were classified as Treatment A. Patients unwilling to undergo intensive pharmacokinetic sampling or unable to tolerate the 150 mg eq. dose (consequently receiving flexible doses of 50, 100 or 150 mg eq.) were classified as Treatment B. RESULTS: Of the 212 patients (safety analysis set), 73% were men; 45% white; 20% black; 34% Asians; mean (SD) age 41 (10.2) years, and mean (SD) baseline Positive and Negative Syndrome Scale total score 54.9 (9.03). A total of 53% (n = 113) patients completed the study and 104 received PP 150 mg eq. throughout. Mean (SD) mode dose of PP was 144.8 (19.58) mg eq. The dosing initiation regimen resulted in rapidly achieved and maintained therapeutic paliperidone levels over the study (average concentrations during the dosing interval were 34.7, 40.0, and 47.8 ng/mL after the 2nd, 8th, and 14th injection respectively). Most frequent (≥ 10%) treatment-emergent adverse events were nasopharyngitis (n = 37), insomnia (n = 32), injection-site pain (n = 32), headache (n = 28), and tachycardia (n = 27). Akathisia (n = 19) and tremor (n = 11) were the most common extrapyramidal adverse events. 33 patients had an SAE and 27 discontinued due to treatment-emergent adverse events. No deaths were reported. Mean (SD) weight change from baseline was 2.5 (5.41) kg at endpoint. Patients' psychoses remained stable. CONCLUSIONS: Safety results after one-year therapy with the highest available dose of once-monthly paliperidone palmitate were consistent with results from previous studies, with no new concerns noted. Plasma concentrations were within the expected range. TRIAL REGISTRATION NO: ClinicalTrials.gov: NCT01150448.


Subject(s)
Antipsychotic Agents/therapeutic use , Isoxazoles/therapeutic use , Palmitates/therapeutic use , Schizophrenia/drug therapy , Schizophrenic Psychology , Adolescent , Adult , Aged , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/adverse effects , Antipsychotic Agents/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Injections, Intramuscular/adverse effects , Injections, Intramuscular/statistics & numerical data , Isoxazoles/administration & dosage , Isoxazoles/adverse effects , Isoxazoles/pharmacokinetics , Male , Middle Aged , Paliperidone Palmitate , Palmitates/administration & dosage , Palmitates/adverse effects , Palmitates/pharmacokinetics , Prospective Studies , Psychiatric Status Rating Scales/statistics & numerical data , Schizophrenia/blood , Treatment Failure
11.
Clin Obes ; 11(2): e12432, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33475255

ABSTRACT

Individuals with obesity have a heightened risk of developing serious comorbidities, and pharmacological treatments for people with obesity are limited. This phase 2 study assessed the safety and efficacy of JNJ-64565111, a dual agonist of glucagon-like peptide-1 and glucagon receptors, in individuals with class II/III obesity without type 2 diabetes. In this randomized, double-blind, placebo-controlled and open-label active-controlled, parallel-group, multicentre study, participants aged 18 to 70 years with a body mass index of 35 to 50 kg/m2 and stable weight were randomly assigned in a 1:1:2:2:2 ratio to blinded treatment with placebo; JNJ-64565111 (5.0, 7.4 or 10.0 mg, each with no dose escalation), or open-label liraglutide 3.0 mg. The primary efficacy endpoint was percent change from baseline in body weight at week 26. Four-hundred seventy four participants were randomized and 343 (72.4%) completed treatment. At week 26, placebo-subtracted body weight changes (adjusted for multiplicity) were -6.8%, -8.1% and -10.0% for the JNJ-64565111 5.0 mg, 7.4 mg and 10.0 mg groups, respectively, and -5.8% for the liraglutide group. Incidence of treatment-emergent adverse events, especially nausea and vomiting, was higher in each JNJ-64565111 treatment group compared to placebo and liraglutide. JNJ-64565111 significantly reduced body weight in a dose-dependent manner vs placebo but was associated with greater incidence of treatment-emergent adverse events.


Subject(s)
Obesity , Adolescent , Adult , Aged , Blood Glucose , Diabetes Mellitus, Type 2/drug therapy , Double-Blind Method , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Glycated Hemoglobin , Humans , Hypoglycemic Agents , Liraglutide , Middle Aged , Obesity/drug therapy , Receptors, Glucagon , Treatment Outcome , Young Adult
12.
Clin Obes ; 11(2): e12433, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33475251

ABSTRACT

Weight loss has been shown to improve metabolic parameters and cardiovascular risk in people with type 2 diabetes mellitus (T2DM). This phase 2 study evaluated the safety and efficacy of JNJ-64565111, a dual agonist of GLP-1 and glucagon receptors, in individuals with T2DM and class II/III obesity. In this randomized, double-blind study, participants with T2DM (HbA1c 6.5%-9.5%), body mass index of 35 to 50 kg/m2 and stable weight were randomly assigned (1:1:1:1) to placebo or JNJ-64565111 (5.0 mg, 7.4 mg or 10.0 mg). The primary endpoint was percent change from baseline in body weight at week 12. Of 195 dosed participants, 144 (73.8%) completed treatment. At week 12, placebo-subtracted body weight changes were -4.6%, -5.9% and -7.2% with JNJ-64565111 5.0 mg, 7.4 mg and 10.0 mg, respectively. All JNJ-64565111 doses were associated with no change in HbA1c and slight numerical elevation of fasting insulin. Numerical increases in fasting plasma glucose were observed with JNJ-64565111 5.0 mg and 7.4 mg. Incidence of treatment-emergent adverse events, especially nausea and vomiting, was higher with JNJ-64565111 vs placebo. Overall, JNJ-64565111 significantly reduced body weight in a dose-dependent manner vs placebo but was associated with greater incidence of treatment-emergent adverse events, no HbA1c reductions, and increased fasting plasma glucose and fasting insulin.


Subject(s)
Diabetes Mellitus, Type 2 , Obesity , Blood Glucose , Diabetes Mellitus, Type 2/drug therapy , Double-Blind Method , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Glycated Hemoglobin , Humans , Hypoglycemic Agents/adverse effects , Obesity/complications , Obesity/drug therapy , Receptors, Glucagon
13.
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
14.
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
15.
Biopharm Drug Dispos ; 31(2-3): 162-77, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20213855

ABSTRACT

The objectives were to develop a translational model that will help select doses for Phase-3 trials based on abbreviated Phase-2 trials and understand the competitive landscape for oral anti-diabetics based on efficacy, tolerability and ability to slow disease progression. Data for eight anti-diabetics with temporal profiles for fasting plasma glucose (FPG) and hemoglobin A1c (HbA1c) from 12 publications were digitized. The monotherapy data consisted of FPG and HbA1c profiles that were modeled using a transit compartment model. Evaluation of the competitive landscape utilized HbA1c literature data for 11 drugs. For the safety metric, tolerability issues with anti-diabetic drug classes were tabulated. For disease progression, the coefficient of failure method was used for assessing data from two long-term trials. The transit rate constants were remarkably consistent across 12 trials and represent system-specific/drug-independent parameters. The competitive landscape analysis showed that the primary efficacy metric fell into two groups of DeltaHbA1c: >0.8% or approximately 0.8%. On the safety endpoints, older agents showed some tolerability issues while the new agents were relatively safe. Among the different drug classes, only the thiazolidinediones appeared to slow disease progression but may also increase heart failure risk. In conclusion, the ability of system-specific parameters to predict HbA1c provides a tool to predict the expected efficacy profile from abbreviated dose-finding trials. To be commercially viable, new drugs should improve DeltaHbA1c by about 0.8% or more and possess safety profiles similar to newer anti-diabetic agents. Thus, this study proposes a suite of simple yet powerful tools to guide type-2-diabetes drug development.


Subject(s)
Administration, Oral , Diabetes Mellitus, Type 2/drug therapy , Drug Delivery Systems/methods , Hypoglycemic Agents/therapeutic use , Blood Glucose , Clinical Trials, Phase III as Topic , Cost of Illness , Diabetes Mellitus, Type 2/blood , Disease Progression , Drug Administration Schedule , Humans , Hypoglycemic Agents/blood , Insulin/blood , Patient Compliance , Randomized Controlled Trials as Topic , Treatment Outcome
16.
Clin Pharmacol Drug Dev ; 9(2): 224-234, 2020 02.
Article in English | MEDLINE | ID: mdl-31876117

ABSTRACT

The paliperidone pharmacokinetics after intramuscular administration of once-monthly paliperidone palmitate in Japanese patients were studied in 3 phase 1 studies and in 2 phase 3 studies performed in Japan, Korea, and Taiwan. These data (Japanese, n = 509; Korean, n = 31; Taiwanese, n = 47) were used to describe the paliperidone palmitate pharmacokinetics in Japanese, to compare with non-Japanese, and to validate the historical population pharmacokinetic (Pop-PK) model for paliperidone palmitate, developed using data from studies in patients with schizophrenia outside Japan. The final historical Pop-PK model, including all significant patient covariates of Japanese studies, was used to simulate paliperidone plasma concentration-time data using nonlinear mixed effects, followed by comparison with actual data. Visual predictive checks displayed considerable overlap between predicted and actual plasma concentrations; the majority of observations were within the 90% prediction interval. Japanese, Korean, and Taiwanese patients had comparable plasma concentrations. Covariate distributions demonstrated comparatively lower median body mass index in Japanese, Korean, and Taiwanese patients versus rest-of-world population. Prediction errors for the data set used for external validation were within cutoff values, confirming accuracy/precision of the model. Paliperidone pharmacokinetics were adequately predicted for Japanese studies using the historical Pop-PK model, confirming its robustness. Pharmacokinetics in Japanese, Korean, and Taiwanese patients with schizophrenia were comparable with rest-of-world population.


Subject(s)
Antipsychotic Agents/pharmacokinetics , Paliperidone Palmitate/pharmacokinetics , Schizophrenia/drug therapy , Adult , Aged , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/blood , Antipsychotic Agents/therapeutic use , Body Mass Index , Case-Control Studies , Dopamine D2 Receptor Antagonists , Female , Half-Life , Humans , Injections, Intramuscular , Japan/epidemiology , Male , Middle Aged , Paliperidone Palmitate/administration & dosage , Paliperidone Palmitate/blood , Paliperidone Palmitate/therapeutic use , Predictive Value of Tests , Republic of Korea/epidemiology , Serotonin 5-HT2 Receptor Antagonists , Taiwan/epidemiology , Treatment Outcome
17.
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
18.
J Clin Pharmacol ; 49(3): 336-50, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19246731

ABSTRACT

Pegylated thrombopoietin mimetic peptide (PEG-TPOm) is a novel, potent thrombopoietin receptor agonist with low immunotoxicity potential that protects against chemotherapy-induced thrombocytopenia in preclinical animal models. The aim of this study was to develop a population pharmacokinetic and pharmacodynamic model of PEG-TPOm following single intravenous doses in healthy subjects. Data were obtained from a double-blind, randomized, placebo-controlled study. A model based on target-mediated drug disposition and precursor pool life spans was applied. Model evaluation was performed through predictive checks and bootstrap analysis. The half-life of PEG-TPOm ranged between 18 and 36 hours, and the estimated distributional volume was 5 L. The increase in platelet counts was observed after a 4-day delay, consistent with the megakaryocyte cell life span. The platelet life span was estimated to be 5 days. After maximum platelets counts were achieved on day 9, platelets returned back to baseline on day 29. Model-based simulations were undertaken to explore pharmacodynamic effects after multiple dosing. Weekly dosing produced a sustained pharmacodynamic response, whereas an interdosing interval >or=2 weeks resulted in fluctuating pharmacodynamic profiles. Thus, the mechanistic pharmacokinetic/pharmacodynamic model was suitable for describing the complex PEG-TPOm pharmacokinetics/pharmacodynamics, including target-mediated disposition, dose-dependent platelet stimulation, and mean life spans of thrombopoietic cell populations.


Subject(s)
Models, Biological , Polyethylene Glycols/pharmacology , Polyethylene Glycols/pharmacokinetics , Receptors, Thrombopoietin/agonists , Thrombopoietin/pharmacology , Thrombopoietin/pharmacokinetics , Adult , Computer Simulation , Double-Blind Method , Half-Life , Humans , Injections, Intravenous , Male , Platelet Count , Polyethylene Glycols/adverse effects , Thrombopoietin/adverse effects , Time Factors
19.
CNS Drugs ; 31(4): 273-288, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28258365

ABSTRACT

Paliperidone palmitate 3-month formulation (PP3M), a long-acting injectable atypical antipsychotic, was recently approved in the US and Europe for the treatment of schizophrenia in adult patients who have already been treated with paliperidone palmitate 1-month formulation (PP1M) for ≥4 months. This article reviews the pharmacokinetic rationale for the approved dosing regimens for PP3M, dosing windows, management of missed doses and treatment discontinuation, switching to other formulations, and dosing in special populations. Approved PP3M dosing regimens are based on the comparisons of simulations with predefined dosing regimens using paliperidone palmitate and oral paliperidone extended release (ER) population pharmacokinetic models (one-compartment model with two saturable absorption processes for PP3M; one-compartment model with parallel zero- and first-order absorption for PP1M; two-compartment model with sequential zero- and first-order absorption for ER) versus clinical trial data. Covariates were obtained by resampling subject covariates from the pharmacokinetics database for PP1M and PP3M. Simulation scenarios with varying doses and covariate values were generated. The population median and 90% prediction interval of the simulated concentration-time profiles were plotted for simulation outcomes evaluation. Simulations described in this paper provide (a) simulated plasma exposures for switching from PP1M to PP3M, (b) support for a once-every-3-months injection cycle, (c) information on dosing windows and managing missed doses of PP3M, (d) important guidance on PP3M dosing in special patient populations, and (e) key PP3M pharmacokinetic exposure metrics based on the population pharmacokinetic PP3M model. Population pharmacokinetics provided practical guidance to establish dosing regimens for PP3M.


Subject(s)
Antipsychotic Agents/administration & dosage , Paliperidone Palmitate/administration & dosage , Schizophrenia/drug therapy , Adult , Antipsychotic Agents/pharmacokinetics , Computer Simulation , Delayed-Action Preparations , Dose-Response Relationship, Drug , Humans , Models, Biological , Paliperidone Palmitate/pharmacokinetics
20.
Clin Pharmacokinet ; 56(4): 421-433, 2017 04.
Article in English | MEDLINE | ID: mdl-27743205

ABSTRACT

OBJECTIVES: Our objective was to characterize the population pharmacokinetics of paliperidone after intramuscular administration of its long-acting 3-month formulation palmitate ester at various doses and at different injection sites (deltoid and gluteal muscles). METHODS: This retrospective analysis included pooled data from 651 subjects from one phase I study (single injection of the 3-month formulation) and one phase III study (multiple injections of both 1- and 3-month formulations). A total of 8990 pharmacokinetic samples with valid concentration time points were available for this analysis. Nonlinear mixed-effects modelling of the pooled data was conducted using NONMEM software. Knowledge from a previously developed 1-month formulation model was used as a starting point to build the 3-month formulation model. RESULTS: The final model describing the plasma concentrations after administration of the 3-month formulation was a one-compartment model with first-order elimination and two saturable absorption processes (rapid and slow). The apparent volume of distribution estimated for the 3-month formulation was not the same as for the previously modelled 1-month formulation. Apparent clearance (CL), apparent volume of distribution (V), and fraction of the absorbed dose (F3) were estimated to be 3.84 l/h, 1960 L, and 20.9 %. For slow absorption, the maximum absorption rate constant (k a1 max), amount of paliperidone at the absorption site when half of the maximum absorption rate was achieved (k amt1 50), and Hill factor (γ) were estimated to be 90.4 µg/h, 120 mg, and 1.44, respectively. For rapid absorption, the maximum absorption rate constant (k a3 max) and amount of paliperidone at the absorption site when half of the maximum absorption rate was achieved (k amt3 50) were estimated to be 164 µg/h and 21.4 mg, respectively. CONCLUSION: The final model with two saturable absorption processes provided a good description of the pharmacokinetic characteristics of paliperidone after intramuscular administration of its long-acting 3-month formulation palmitate ester. In addition to the structural covariates (creatinine clearance on CL, body mass index on V, and injection volume on both absorption rates), injection site and sex were identified as covariates on k a max of the slow absorption process (k a1 max). Clinical trial registration numbers: NCT01559272, NCT01529515, and NCT01515423.


Subject(s)
Antipsychotic Agents/administration & dosage , Antipsychotic Agents/pharmacokinetics , Paliperidone Palmitate/administration & dosage , Paliperidone Palmitate/pharmacokinetics , Schizophrenia/drug therapy , Schizophrenia/metabolism , Adolescent , Adult , Aged , Double-Blind Method , Drug Administration Schedule , Drug Compounding , Female , Humans , Injections, Intramuscular , Male , Middle Aged , Retrospective Studies , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL