Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Clin Pharmacokinet ; 63(1): 27-42, 2024 01.
Article in English | MEDLINE | ID: mdl-38177893

ABSTRACT

Nirmatrelvir is a potent and selective inhibitor of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease that is used as an oral antiviral coronavirus disease 2019 (COVID-19) treatment. To sustain unbound systemic trough concentrations above the antiviral in vitro 90% effective concentration value (EC90), nirmatrelvir is coadministered with 100 mg of ritonavir, a pharmacokinetic enhancer. Ritonavir inhibits nirmatrelvir's cytochrome P450 (CYP) 3A4-mediated metabolism which results in renal elimination becoming the primary route of nirmatrelvir elimination when dosed concomitantly. Nirmatrelvir exhibits absorption-limited nonlinear pharmacokinetics. When coadministered with ritonavir in patients with mild-to-moderate COVID-19, nirmatrelvir reaches a maximum concentration of 3.43 µg/mL (11.7× EC90) in approximately 3 h on day 5 of dosing, with a geometric mean day 5 trough concentration of 1.57 µg/mL (5.4× EC90). Drug interactions with nirmatrelvir/ritonavir (PAXLOVIDTM) are primarily attributed to ritonavir-mediated CYP3A4 inhibition, and to a lesser extent CYP2D6 and P-glycoprotein inhibition. Population pharmacokinetics and quantitative systems pharmacology modeling support twice daily dosing of 300 mg/100 mg nirmatrelvir/ritonavir for 5 days, with a reduced 150 mg/100 mg dose for patients with moderate renal impairment. Rapid clinical development of nirmatrelvir/ritonavir in response to the emerging COVID-19 pandemic was enabled by innovations in clinical pharmacology research, including an adaptive phase 1 trial design allowing direct to pivotal phase 3 development, fluorine nuclear magnetic resonance spectroscopy to delineate absorption, distribution, metabolism, and excretion profiles, and innovative applications of model-informed drug development to accelerate development.


Subject(s)
COVID-19 , Lactams , Leucine , Nitriles , Proline , Ritonavir , Humans , Ritonavir/pharmacology , Ritonavir/therapeutic use , Pandemics , Drug Interactions , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Drug Combinations
2.
CPT Pharmacometrics Syst Pharmacol ; 12(12): 1897-1910, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37803876

ABSTRACT

Protease inhibitor nirmatrelvir coadministered with ritonavir as a pharmacokinetic enhancer (PAXLOVID™; Pfizer Inc) became the first orally bioavailable antiviral agent granted Emergency Use Authorization in the United States in patients ≥12 years old with mild to moderate coronavirus disease 2019 (COVID-19). This population pharmacokinetic analysis used pooled plasma nirmatrelvir concentrations from eight completed phase I and II/III studies to characterize nirmatrelvir pharmacokinetics when coadministered with ritonavir in adults with/without COVID-19. Influence of covariates (e.g., formulation, dose, COVID-19) was examined using a stepwise forward selection (α = 0.05) and backward elimination (α = 0.001) approach. Simulations with 5000 subjects for each age and weight group and renal function category were performed to support dosing recommendations of nirmatrelvir/ritonavir for adults with COVID-19 and guide dose adjustments for specific patient populations (e.g., renal insufficiency, pediatrics). The final model was a two-compartment model with first-order absorption, including allometric scaling of body weight and dose-dependent absorption (power function on relative bioavailability). Nirmatrelvir clearance (CL) increased proportionally to body surface area-normalized creatinine CL (nCLCR) up to 70 ml/min/1.73 m2 and was independent of nCLCR above the breakpoint. Significant covariates included carbamazepine or itraconazole coadministration as markers for drug interactions, COVID-19 on CL, formulation on relative bioavailability, and age on central volume of distribution. Simulation results support current dosing recommendations of nirmatrelvir/ritonavir 300/100 mg twice daily (b.i.d.) in adults with normal renal function or mild impairment and pediatrics (12 to <18 years) weighing ≥40 kg and nirmatrelvir/ritonavir 150/100 mg b.i.d. in adults with moderate renal impairment.


Subject(s)
COVID-19 , Ritonavir , Adult , Humans , Child , Antiviral Agents , Benzodiazepines
3.
Br J Clin Pharmacol ; 88(12): 5428-5433, 2022 12.
Article in English | MEDLINE | ID: mdl-36040430

ABSTRACT

Pharmacometric analyses of time series viral load data may detect drug effects with greater power than approaches using single time points. Because SARS-CoV-2 viral load rapidly rises and then falls, viral dynamic models have been used. We compared different modelling approaches when analysing Phase II-type viral dynamic data. Using two SARS-CoV-2 datasets of viral load starting within 7 days of symptoms, we fitted the slope-intercept exponential decay (SI), reduced target cell limited (rTCL), target cell limited (TCL) and TCL with eclipse phase (TCLE) models using nlmixr. Model performance was assessed via Bayesian information criterion (BIC), visual predictive checks (VPCs), goodness-of-fit plots, and parameter precision. The most complex (TCLE) model had the highest BIC for both datasets. The estimated viral decline rate was similar for all models except the TCL model for dataset A with a higher rate (median [range] day-1 : dataset A; 0.63 [0.56-1.84]; dataset B: 0.81 [0.74-0.85]). Our findings suggest simple models should be considered during pharmacodynamic model development.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Humans , Bayes Theorem , Viral Load
4.
Clin Pharmacol Ther ; 112(4): 892-900, 2022 10.
Article in English | MEDLINE | ID: mdl-35712797

ABSTRACT

Nirmatrelvir coadministered with ritonavir is highly efficacious in reducing the risk of coronavirus disease 2019 (COVID-19) adverse outcomes among patients at increased risk of progression to severe disease, including patients with chronic kidney disease. Because nirmatrelvir is eliminated by the kidneys when given with ritonavir, this phase I study evaluated the effects of renal impairment on pharmacokinetics, safety, and tolerability of nirmatrelvir/ritonavir. Participants with normal renal function (n = 10) or mild, moderate, or severe renal impairment (n = 8 each) were administered a single 100-mg nirmatrelvir dose with 100 mg ritonavir given 12 hours before, together with and 12 and 24 hours after the nirmatrelvir dose. Systemic nirmatrelvir exposure increased with increasing renal impairment, with mild, moderate, and severe renal impairment groups having respective adjusted geometric mean ratio areas under the plasma concentration-time profile from time 0 extrapolated to infinite time of 124%, 187%, and 304% vs. the normal renal function group. Corresponding ratios for maximum plasma concentration were 130%, 138%, and 148%. Apparent clearance was positively correlated with estimated glomerular filtration rate, and geometric mean renal clearance values were particularly lower for the moderate (47% decrease) and severe (80% decrease) renal impairment groups vs. the normal renal function group. Nirmatrelvir/ritonavir exhibited an acceptable safety profile; treatment-related adverse events were mild in severity, and there were no significant findings regarding laboratory measurements, vital signs, or electrocardiogram assessments. These findings led to a dose reduction recommendation for nirmatrelvir/ritonavir in patients with moderate renal impairment (150/100 mg nirmatrelvir/ritonavir instead of 300/100 mg twice daily for 5 days). NCT04909853.


Subject(s)
COVID-19 Drug Treatment , Renal Insufficiency , Antiviral Agents/adverse effects , Enzyme Inhibitors , Humans , Protease Inhibitors , Ritonavir/adverse effects
5.
Clin Pharmacol Ther ; 111(3): 635-645, 2022 03.
Article in English | MEDLINE | ID: mdl-34687548

ABSTRACT

Increasing prevalence of infections caused by antimicrobial-resistant gram-negative bacteria represents a global health crisis, and while several novel therapies that target various aspects of antimicrobial resistance have been introduced in recent years, few are currently approved for children. Ceftazidime-avibactam is a novel ß-lactam ß-lactamase inhibitor combination approved for adults and children 3 months and older with complicated intra-abdominal infection, and complicated urinary tract infection or hospital-acquired ventilator-associated pneumonia (adults only in the United States) caused by susceptible gram-negative bacteria. Extensive population pharmacokinetic (PK) data sets for ceftazidime and avibactam obtained during the adult clinical development program were used to iteratively select, modify, and validate the approved adult dosage regimen (2,000-500 mg by 2-hour intravenous (IV) infusion every 8 hours (q8h), with adjustments for renal function). Following the completion of one phase I (NCT01893346) and two phase II ceftazidime-avibactam studies (NCT02475733 and NCT02497781) in children, adult PK data sets were updated with pediatric PK data. This paper describes the development of updated combined adult and pediatric population PK models and their application in characterizing the population PK of ceftazidime and avibactam in children, and in dose selection for further pediatric evaluation. The updated models supported the approval of ceftazidime-avibactam pediatric dosage regimens (all by 2-hour IV infusion) of 50-12.5 mg/kg (maximum 2,000-500 mg) q8h for those ≥6 months to 18 years old, and 40-10 mg/kg q8h for those ≥3 to 6 months old with creatinine clearance > 50 mL/min/1.73 m2 .


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Azabicyclo Compounds/pharmacokinetics , Ceftazidime/pharmacokinetics , beta-Lactamase Inhibitors/pharmacokinetics , Adolescent , Anti-Bacterial Agents/therapeutic use , Azabicyclo Compounds/therapeutic use , Ceftazidime/therapeutic use , Child , Child, Preschool , Drug Combinations , Drug Resistance, Multiple, Bacterial/drug effects , Female , Gram-Negative Bacteria/drug effects , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , Humans , Infant , Intraabdominal Infections/drug therapy , Intraabdominal Infections/microbiology , Male , Pneumonia, Ventilator-Associated/drug therapy , Pneumonia, Ventilator-Associated/microbiology , Probability , Urinary Tract Infections/drug therapy , Urinary Tract Infections/microbiology , beta-Lactamase Inhibitors/therapeutic use
6.
CPT Pharmacometrics Syst Pharmacol ; 10(6): 551-563, 2021 06.
Article in English | MEDLINE | ID: mdl-33687148

ABSTRACT

A model-informed drug development approach was used to select ceftaroline fosamil high-dose regimens for pediatric patients with complicated skin and soft-tissue infections caused by Staphylococcus aureus with a ceftaroline minimum inhibitory concentration (MIC) of 2 or 4 mg/L. Steady-state ceftaroline concentrations were simulated using a population pharmacokinetics (PK) model for ceftaroline fosamil and ceftaroline including data from 304 pediatric subjects and 944 adults. Probability of target attainment (PTA) for various simulated pediatric high-dose regimens and renal function categories were calculated based on patients achieving 35% fT>MIC (S. aureus PK/pharmacodynamic target for 2-log10 bacterial killing). For extrapolation of efficacy, simulated exposures and PTA were compared to adults with normal renal function receiving high-dose ceftaroline fosamil (600 mg 2-h infusions every 8 h). For safety, predicted ceftaroline exposures were compared with observed pediatric and adult data. Predicted ceftaroline exposures for the approved pediatric high-dose regimens (12, 10, or 8 mg/kg by 2-h infusions every 8 h for patients aged >2 to <18 years with normal/mild, moderate, or severe renal impairment, respectively; 10 mg/kg by 2-h infusions every 8 h for patients aged ≥2 months to <2 years with normal renal function/mild impairment) were well matched to adults with normal renal function. Median predicted maximum concentration at steady state (Cmax,ss ) and area under the plasma concentration-time curve over 24 h at steady state pediatric to adult ratios were 0.907-1.33 and 0.940-1.41, respectively. PTAs (>99% and ≥81% for MICs of 2 and 4 mg/L, respectively) matched or exceeded the adult predictions. Simulated Cmax,ss values were below the maximum observed data in other indications, including a high-dose pediatric pneumonia trial, which reported no adverse events related to high exposure.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Cephalosporins/administration & dosage , Models, Biological , Skin Diseases, Infectious/drug therapy , Soft Tissue Infections/drug therapy , Staphylococcal Infections/drug therapy , Staphylococcus aureus , Adolescent , Adult , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/pharmacokinetics , Cephalosporins/blood , Cephalosporins/pharmacokinetics , Child , Child, Preschool , Clinical Trials as Topic , Computer Simulation , Female , Humans , Infant , Infant, Newborn , Male , Microbial Sensitivity Tests , Renal Insufficiency/blood , Renal Insufficiency/metabolism , Skin Diseases, Infectious/blood , Skin Diseases, Infectious/metabolism , Soft Tissue Infections/blood , Soft Tissue Infections/metabolism , Staphylococcal Infections/blood , Staphylococcal Infections/metabolism , Ceftaroline
7.
Clin Pharmacol Drug Dev ; 10(4): 420-427, 2021 04.
Article in English | MEDLINE | ID: mdl-33465279

ABSTRACT

The key pharmacokinetic/pharmacodynamic (PK/PD) efficacy index for ß-lactam antibiotics is the percentage of time that free drug concentrations exceed the minimum inhibitory concentration (MIC) of bacteria during each dosing interval (fT>MIC). Ceftaroline fosamil, the prodrug of the ß-lactam ceftaroline, was initially approved for administration as 60-minute intravenous (IV) infusions. Population PK analyses comparing exposure and PK/PD target attainment for 5-minute and 60-minute IV infusions, described here, have supported ceftaroline fosamil labeling updates to include variable infusion durations of 5 to 60 minutes in adults and children aged ≥2 months. A 2-compartment disposition PK model for ceftaroline fosamil and ceftaroline was used to predict steady-state ceftaroline exposures (maximum plasma concentrations [Cmax,ss ] and area under the plasma concentration-time curve over 24 hours [AUCss,0-24 ]) and probability of target attainment in simulated adult and pediatric patients with various degrees of renal function receiving standard doses of ceftaroline fosamil as 5-minute or 60-minute IV infusions. Across age groups and renal function categories, median ceftaroline AUCss,0-24 values were similar for 5-minute and 60-minute infusions, whereas Cmax,ss was up to 42% higher for 5-minute infusions. Both infusion durations achieved >99% probability of target attainment based on PK/PD targets for Staphylococcus aureus (35% fT>MIC) and Streptococcus pneumoniae (44% fT>MIC) at European Committee on Antimicrobial Susceptibility Testing/Clinical and Laboratory Standards Institute MIC breakpoints (1 mg/L and 0.25/0.5 mg/L, respectively). These findings support administration of standard ceftaroline fosamil doses over 5 to 60 minutes for adults and children aged ≥2 months, providing added flexibility to clinicians and patients.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Cephalosporins/administration & dosage , Models, Biological , Renal Insufficiency/physiopathology , Adolescent , Adult , Age Factors , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Area Under Curve , Cephalosporins/pharmacokinetics , Cephalosporins/pharmacology , Child , Child, Preschool , Computer Simulation , Female , Humans , Infant , Infusions, Intravenous , Male , Microbial Sensitivity Tests , Ceftaroline
8.
Clin Pharmacol Ther ; 110(1): 132-140, 2021 07.
Article in English | MEDLINE | ID: mdl-33280106

ABSTRACT

Pregabalin is approved in multiple countries as adjunctive therapy for adult patients with focal onset seizures (FOS; previously termed partial onset seizures). This study used population pharmacokinetic (PK) and exposure-response (E-R) analyses from pooled pregabalin concentration and efficacy data to compare pregabalin exposure and E-R relationships in pediatric and adult patients with FOS, to support pediatric dosage recommendations. A one-compartment disposition model was used, with first-order absorption and body surface area-normalized creatinine clearance on clearance. Individual pregabalin average steady-state concentrations were predicted and used in an E-R analysis of efficacy. The E-R relationship of pregabalin was similar in pediatric (4-16 years) and adult patients with FOS after accounting for differences in baseline natural log-transformed 28-day seizure rate and placebo effect. Population PK simulations showed that children aged 4-16 years and weighing ≥ 30 kg required pregabalin 2.5-10 mg/kg/day to achieve similar pregabalin exposure at steady-state to adult patients receiving the approved doses of 150-600 mg/day. For children 4-16 years weighing < 30 kg, a higher pregabalin dose of 3.5-14 mg/kg/day was required to achieve equivalent exposure at steady-state. The results support the dosage guidance provided in the pregabalin prescribing label, whereby pediatric patients (4-16 years) weighing < 30 kg should receive a 40% higher pregabalin dose (per kg of body weight) than patients weighing ≥ 30 kg to achieve similar exposure. Our combined modeling approach may provide guidance for future extrapolation assessment from adult to pediatric patients.


Subject(s)
Anticonvulsants/administration & dosage , Epilepsies, Partial/drug therapy , Models, Biological , Pregabalin/administration & dosage , Adolescent , Adult , Age Factors , Aged , Anticonvulsants/pharmacokinetics , Child , Child, Preschool , Creatinine/metabolism , Dose-Response Relationship, Drug , Female , Humans , Infant , Male , Middle Aged , Pregabalin/pharmacokinetics , Treatment Outcome , Young Adult
9.
Pediatr Infect Dis J ; 39(5): 411-418, 2020 05.
Article in English | MEDLINE | ID: mdl-32091493

ABSTRACT

BACKGROUND: With increasing antimicrobial resistance, antibiotic treatment options for neonatal late-onset sepsis (LOS) are becoming limited. Primary objective of this study was assessment of the safety of ceftaroline fosamil in LOS. METHODS: Eligible neonates and very young infants 7 to <60 days of age with LOS were enrolled in this phase 2, open-label, multicenter study (NCT02424734) and received ceftaroline fosamil 4 or 6 mg/kg every 8 hours by 1-hour intravenous infusion plus intravenous ampicillin and optional aminoglycoside for 48 hours-14 days. Safety was assessed through the final study visit (21-35 days after the last study therapy dose). Efficacy, assessed as clinical and microbiologic response, was evaluated at end-of-treatment and test-of-cure. Pharmacokinetic samples were collected via sparse-sampling protocol. RESULTS: Eleven patients [54.5% male, median (range) age 24 (12-53) days] were enrolled and received ceftaroline fosamil for a median (range) duration of 8 (3-15) days. Ten adverse events (AEs) occurred in 5 (45.5%) patients (safety population); most frequent AE was diarrhea (n = 2). All except 1 AE (diarrhea) were nontreatment-related. Predominant baseline pathogen was Escherichia coli. No patients were clinical failures at end-of-treatment/test-of-cure. Observed sparse steady-state pharmacokinetics data (19 samples) were comparable to previous pediatric data and generally within 90% model prediction intervals; neonatal probability of target attainment was >95% based on established pharmacokinetic/pharmacodynamic targets. CONCLUSIONS: Safety in neonates and very young infants was consistent with the known ceftaroline fosamil safety profile. These results support the use of ceftaroline fosamil (6 mg/kg every 8 hours) as a potential treatment option for LOS.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/therapeutic use , Cephalosporins/pharmacokinetics , Cephalosporins/therapeutic use , Neonatal Sepsis/drug therapy , Administration, Intravenous , Double-Blind Method , Female , Humans , Infant , Infant, Newborn , Internationality , Male , Ceftaroline
10.
Clin Transl Sci ; 13(1): 125-136, 2020 01.
Article in English | MEDLINE | ID: mdl-31550073

ABSTRACT

Modeling and simulation provides quantitative information on target coverage for dose selection. Optimal model selection often relies on fit criteria and is not necessarily mechanistically driven. One such case is discussed where healthy volunteer data of an anti-myostatin monoclonal antibody domagrozumab were used to develop different target-mediated drug disposition models; a quasi-steady state (QSS) rapid binding approximation model, a Michaelis-Menten (MM)-binding kinetics (MM-BK) model, and an MM-indirect response (MM-IDR) model. Whereas the MM-BK model was identified as optimal in fitting the data, with all parameters estimated with high precision, the QSS model also converged but was not able to capture the nonlinear decline. Although the least mechanistic model, MM-IDR, had the lowest objective function value, the MM-BK model was further developed as it provided a reasonable fit and allowed simulations regarding growth differentiation factor-8 target coverage for phase II dose selection with sufficient certainty to allow for testing of the underlying mechanistic assumptions.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Models, Biological , Myostatin/antagonists & inhibitors , Adult , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Clinical Trials, Phase II as Topic , Computer Simulation , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Healthy Volunteers , Humans , Infusions, Intravenous , Inhibitory Concentration 50 , Male , Myostatin/metabolism
11.
J Clin Pharmacol ; 59(1): 139-152, 2019 01.
Article in English | MEDLINE | ID: mdl-30192390

ABSTRACT

Maraviroc is a C-C chemokine receptor type-5 antagonist approved for the treatment of HIV-1. Previous studies show that cytochrome P450 3A5 (CYP3A5) plays a role in maraviroc metabolism. CYP3A5 is subject to a genetic polymorphism. The presence of 2 functional alleles (CYP3A5*1/*1) confers the extensive metabolism phenotype, which is rare in whites but common in blacks. The effect of CYP3A5 genotype on maraviroc and/or metabolite pharmacokinetics was evaluated in 2 clinical studies: a post hoc analysis from a phase 2b/3 study (NCT00098293) conducted in 494 HIV-1-infected subjects (study 1) in which the impact on maraviroc efficacy in 303 subjects was also assessed, and a study conducted in 47 healthy volunteers (study 2). In study 2 (NCT02625207), extensive metabolizers had 26% to 37% lower mean area under the concentration-time curve compared with poor metabolizers (no CYP3A5*1 alleles). This effect diminished to 17% in the presence of potent CYP3A inhibition. The effect of CYP3A5 genotype was greatest in the formation of the metabolite (1S,2S)-2-hydroxymaraviroc. In study 1, the CYP3A5*1/*1 genotype unexpectedly had higher maraviroc area under the curve predictions (20%) compared with those with no CYP3A5*1 alleles. The reason for this disparity remains unclear. The proportions of subjects with viral loads <50 and <400 copies/mL for maraviroc were comparable among all 3 CYP3A5 genotypes. In both studies maraviroc exposures were in the range of near-maximal viral inhibition in the majority of subjects. These results demonstrate that although CYP3A5 contributes to the metabolism of maraviroc, CYP3A5 genotype does not affect the clinical response to maraviroc in combination treatment of HIV-1 infection at approved doses.


Subject(s)
Cytochrome P-450 CYP3A/genetics , HIV Fusion Inhibitors/pharmacokinetics , HIV Fusion Inhibitors/therapeutic use , HIV Infections , HIV-1 , Maraviroc/pharmacokinetics , Maraviroc/therapeutic use , Adult , Double-Blind Method , Female , Genotype , HIV Fusion Inhibitors/blood , HIV Infections/drug therapy , HIV Infections/genetics , HIV Infections/metabolism , Healthy Volunteers , Humans , Male , Maraviroc/blood , Middle Aged , Polymorphism, Genetic , Treatment Outcome , Young Adult
12.
Br J Clin Pharmacol ; 73(1): 77-92, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21689140

ABSTRACT

AIM: To use non-linear mixed effects modelling and simulation techniques to predict whether PF-04878691, a toll-like receptor 7 (TLR7) agonist, would produce sufficient antiviral efficacy while maintaining an acceptable side effect profile in a 'proof of concept' (POC) study in chronic hepatitis C (HCV) patients. METHODS: A population pharmacokinetic-pharmacodynamic (PKPD) model was developed using available 'proof of pharmacology' (POP) clinical data to describe PF-04878691 pharmacokinetics (PK) and its relationship to 2',5'-oligoadenylate synthetase (OAS; marker of pharmacology) and lymphocyte levels (marker of safety) following multiple doses in healthy subjects. A second model was developed to describe the relationship between change from baseline OAS expressed as fold change and HCV viral RNA concentrations using clinical data available in HCV patients for a separate compound, CPG-10101 (ACTILON™), a TLR9 agonist. Using these models the antiviral efficacy and safety profiles of PF-04878691 were predicted in HCV patients. RESULTS: The population PKPD models described well the clinical data as assessed by visual inspection of diagnostic plots, visual predictive checks and precision of the parameter estimates. Using these relationships, PF-04878691 exposure and HCV viral RNA concentration was simulated in HCV patients receiving twice weekly administration for 4 weeks over a range of doses. The simulations indicated that significant reductions in HCV viral RNA concentrations would be expected at doses > 6 mg. However at these doses grade ≥ 3 lymphopenia was also predicted. CONCLUSIONS: The model simulations indicate that PF-04878691 is unlikely to achieve POC criteria and support the discontinuation of this compound for the treatment of HCV.


Subject(s)
2',5'-Oligoadenylate Synthetase/pharmacokinetics , Drug Design , Hepatitis C, Chronic/metabolism , Toll-Like Receptor 7/agonists , Viral Load/drug effects , Adult , Computer Simulation , Decision Support Techniques , Dose-Response Relationship, Drug , Female , Hepacivirus/drug effects , Humans , Male , Middle Aged , Models, Biological , Randomized Controlled Trials as Topic , Young Adult
13.
J Pharmacokinet Pharmacodyn ; 38(1): 41-61, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21088872

ABSTRACT

Using simulated viral load data for a given maraviroc monotherapy study design, the feasibility of different algorithms to perform parameter estimation for a pharmacokinetic-pharmacodynamic-viral dynamics (PKPD-VD) model was assessed. The assessed algorithms are the first-order conditional estimation method with interaction (FOCEI) implemented in NONMEM VI and the SAEM algorithm implemented in MONOLIX version 2.4. Simulated data were also used to test if an effect compartment and/or a lag time could be distinguished to describe an observed delay in onset of viral inhibition using SAEM. The preferred model was then used to describe the observed maraviroc monotherapy plasma concentration and viral load data using SAEM. In this last step, three modelling approaches were compared; (i) sequential PKPD-VD with fixed individual Empirical Bayesian Estimates (EBE) for PK, (ii) sequential PKPD-VD with fixed population PK parameters and including concentrations, and (iii) simultaneous PKPD-VD. Using FOCEI, many convergence problems (56%) were experienced with fitting the sequential PKPD-VD model to the simulated data. For the sequential modelling approach, SAEM (with default settings) took less time to generate population and individual estimates including diagnostics than with FOCEI without diagnostics. For the given maraviroc monotherapy sampling design, it was difficult to separate the viral dynamics system delay from a pharmacokinetic distributional delay or delay due to receptor binding and subsequent cellular signalling. The preferred model included a viral load lag time without inter-individual variability. Parameter estimates from the SAEM analysis of observed data were comparable among the three modelling approaches. For the sequential methods, computation time is approximately 25% less when fixing individual EBE of PK parameters with omission of the concentration data compared with fixed population PK parameters and retention of concentration data in the PD-VD estimation step. Computation times were similar for the sequential method with fixed population PK parameters and the simultaneous PKPD-VD modelling approach. The current analysis demonstrated that the SAEM algorithm in MONOLIX is useful for fitting complex mechanistic models requiring multiple differential equations. The SAEM algorithm allowed simultaneous estimation of PKPD and viral dynamics parameters, as well as investigation of different model sub-components during the model building process. This was not possible with the FOCEI method (NONMEM version VI or below). SAEM provides a more feasible alternative to FOCEI when facing lengthy computation times and convergence problems with complex models.


Subject(s)
Algorithms , Cyclohexanes/pharmacokinetics , HIV Fusion Inhibitors/pharmacokinetics , HIV Infections/metabolism , HIV Infections/virology , HIV/drug effects , Models, Statistical , Triazoles/pharmacokinetics , Computer Simulation , Cyclohexanes/pharmacology , Cyclohexanes/therapeutic use , HIV/physiology , HIV Fusion Inhibitors/pharmacology , HIV Fusion Inhibitors/therapeutic use , HIV Infections/drug therapy , Humans , Maraviroc , Research Design , Software , Triazoles/pharmacology , Triazoles/therapeutic use , Viral Load
14.
Br J Clin Pharmacol ; 65 Suppl 1: 76-85, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18333869

ABSTRACT

AIMS: To develop a population pharmacokinetic model for maraviroc, a noncompetitive CCR5 antagonist, after oral administration of tablets to healthy volunteers and asymptomatic HIV-infected subjects and to quantify the inherent variability and influence of covariates on the parameters of the model. METHODS: Rich pharmacokinetic data available from 15 studies in healthy volunteers (n = 365) and two studies in asymptomatic HIV-infected subjects (n = 48) were analysed using NONMEM. Maraviroc was administered as single or multiple oral tablet doses under fasted and fed conditions. Doses ranged from 100 to 1800 mg day(-1). RESULTS: A two-compartment model parameterized to separate out absorption and clearance components on bioavailability was used. Absorption was described by a lagged first-order process. A sigmoid E(max) model described the effect of dose on absorption. A visual predictive check and nonparametric bootstrap evaluation confirmed that the model was a good description of the data. Typical CL, V(c) and V(p) values for a 30-year-old non-Asian are 51.5 l h(-1), 132 l and 277 l, respectively. CONCLUSIONS: For the typical non-Asian subject, fasted bioavailability increased asymptotically with dose from 24% at 100 mg to 33% at 600 mg. A high-fat meal taken with maraviroc reduced exposure by 43% for a 100-mg dose to approximately 25% at doses of 600 mg. The typical Asian subject had a 26.5% higher AUC than the typical non-Asian subject irrespective of dose, a difference not considered to be clinically relevant. None of the other covariates tested had any clinically relevant effects on exposure.


Subject(s)
Anti-HIV Agents/pharmacokinetics , CCR5 Receptor Antagonists , Cyclohexanes/pharmacokinetics , HIV Infections/drug therapy , Triazoles/pharmacokinetics , Adolescent , Adult , Computer Simulation , Female , Humans , Male , Maraviroc , Middle Aged , Models, Biological , Models, Statistical , Randomized Controlled Trials as Topic
15.
Pharm Res ; 24(4): 791-802, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17308968

ABSTRACT

PURPOSE: To externally validate the model predictions of a DATATOP cohort analysis through application of clinical trial simulation with the study design of the ELLDOPA trial. METHODS: The stochastic pharmacokinetic-pharmacodynamic and disease progress model was developed from the large DATATOP cohort of patients followed for 8 years. ELLDOPA was designed to detect a difference between placebo and levodopa treated arms in the total Unified Parkinson's Disease Rating Scale (UPDRS) taken at baseline and following 2 weeks levodopa washout after 40 weeks of treatment. The total UPDRS response was simulated with different assumptions on levodopa effect (symptomatic with/without disease modifying capability) and washout speed of symptomatic effect. RESULTS: The observed results of ELLDOPA were similar to the model predictions assuming levodopa slows disease progression and has a slow washout of symptomatic effect. CONCLUSIONS: This simulation work confirmed the conclusion of the DATATOP analysis finding that levodopa slows disease progression. The simulation results also showed that a dose-related increased rate of progression in Parkinson's disease, obscured by symptomatic benefit, is very unlikely. Finally, the simulation results also shown that 2 weeks washout period was not adequate to completely eliminate the symptomatic benefits of levodopa.


Subject(s)
Antiparkinson Agents/therapeutic use , Computer Simulation , Levodopa/therapeutic use , Models, Biological , Parkinson Disease/drug therapy , Research Design , Antiparkinson Agents/adverse effects , Antiparkinson Agents/pharmacokinetics , Cohort Studies , Disease Progression , Follow-Up Studies , Humans , Levodopa/adverse effects , Levodopa/pharmacokinetics , Predictive Value of Tests , Reproducibility of Results , Time Factors , Treatment Outcome
16.
J Pharmacokinet Pharmacodyn ; 33(3): 281-311, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16625427

ABSTRACT

We have modelled the Unified Parkinson's Disease Rating Scale (UPDRS) scores collected in 800 subjects followed for 8 years. Newly diagnosed and previously untreated subjects were initially randomized to treatment with placebo, deprenyl, tocopherol or both and, when clinical disability required, received one or more dopaminergic agents (levodopa (carbidopa/levodopa), bromocriptine, or pergolide). Using models for disease progression and pharmacodynamic models for drug effects we have characterized the changes in UPDRS over time to determine the influence of the various drug treatments. We have confirmed and quantitated the relative symptomatic benefits of the dopaminergic agents and provide model-based evidence for slowing of disease progression.


Subject(s)
Levodopa/pharmacology , Models, Biological , Parkinson Disease/drug therapy , Algorithms , Antiparkinson Agents/pharmacokinetics , Antiparkinson Agents/pharmacology , Antiparkinson Agents/therapeutic use , Bayes Theorem , Bromocriptine/pharmacokinetics , Bromocriptine/pharmacology , Bromocriptine/therapeutic use , Disease Progression , Dopamine Agonists/pharmacokinetics , Dopamine Agonists/pharmacology , Dopamine Agonists/therapeutic use , Drug Therapy, Combination , Humans , Levodopa/pharmacokinetics , Levodopa/therapeutic use , Parkinson Disease/pathology , Pergolide/pharmacokinetics , Pergolide/pharmacology , Pergolide/therapeutic use , Randomized Controlled Trials as Topic , Selegiline/pharmacokinetics , Selegiline/pharmacology , Selegiline/therapeutic use , Treatment Outcome
17.
J Pharmacokinet Pharmacodyn ; 32(3-4): 307-31, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16320098

ABSTRACT

The purpose of the study was to describe the population pharmacokinetics of levodopa in patients with Parkinson's disease studied in 5 trials (10 occasions) over 4 years. Twenty previously untreated Parkinsonian patients were investigated. Each trial consisted of a 2-hr IV infusion of levodopa (1 mg/kg/h) with concomitant oral carbidopa given on two occasions separated by 72 hr with no levodopa in between. This trial design was repeated at 6, 12, 24 and 48 months. A two-compartment pharmacokinetic model with central volume (V1), peripheral volume (V2), clearance (CL) and inter-compartmental clearance (CL(ic)) was used to fit plasma levodopa concentrations. The model accounted for levodopa dosing prior to each trial and endogenous levodopa synthesis. Population parameter estimates (geometric mean) and population parameter variability (PPV; SD of normal distribution) were V1 11.4 l/70 kg (0.44), CL 30.9 l/h/70 kg (0.25), V2 27.3 l/70 kg (0.27), and CL(ic) 34.6 l/h/70 kg (0.48). PPV was partitioned into between subject variability (BSV) which was 0.12 V1, 0.13 CL, 0.15 V(2), 0.28 CL(ic), within trial variability (WTV) which was 0.16 V1, 0.13 CL, 0.08 V2, 0.18 CL(ic) and between trial variability (BTV) which was 0.40 V1, 0.17 CL, 0.21 V2, 0.34 CL(ic.) Neither structural nor random levodopa pharmacokinetic parameters were associated with the time course of development of fluctuation in motor response. Variability in levodopa pharmacokinetic parameters (particularly V1) may result in variability in plasma levodopa concentrations that could contribute to fluctuations in motor response.


Subject(s)
Dopamine Agents/pharmacokinetics , Levodopa/pharmacokinetics , Parkinson Disease/drug therapy , Administration, Oral , Adult , Aged , Aromatic Amino Acid Decarboxylase Inhibitors , Carbidopa/administration & dosage , Carbidopa/therapeutic use , Clinical Trials as Topic , Cohort Studies , Computer Simulation , Dopa Decarboxylase/metabolism , Dopamine Agents/administration & dosage , Dopamine Agents/therapeutic use , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , Female , Humans , Infusions, Intravenous , Levodopa/administration & dosage , Levodopa/therapeutic use , Longitudinal Studies , Male , Middle Aged , Models, Biological , Parkinson Disease/blood , Parkinson Disease/metabolism
18.
J Pharmacokinet Pharmacodyn ; 32(3-4): 459-84, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16320101

ABSTRACT

The purpose of this analysis is to describe how levodopa pharmacokinetic and pharmacodynamic parameters change over the first 4 years of long-term levodopa treatment in patients with Parkinson's disease. Twenty previously untreated Parkinsonian patients were admitted to the general clinical research center (GCRC) for 4 days at the beginning of long-term levodopa therapy and 6, 12, 24 and 48 months later. On each GCRC admission, patients received a 2 hr IV infusion of levodopa on day 1 and day 4 with no oral levodopa between the infusions. After the first GCRC admission patients were treated with oral levodopa dosed for optimal control of Parkinsonism. Motor function was measured by finger tapping rate. A pharmacokinetic-pharmacodynamic model incorporating 3 effect compartments was used to fit the individual plasma levodopa concentrations and tapping rates. Motor function before the first levodopa infusion (E0(1)) improved over the first 20 months and subsequently returned to the initial baseline at the start of the study. A similar pattern was seen in motor function before the second infusion (E0(2)) after the 3 days levodopa withdrawal, with a decline predicted to fall below the initial baseline at the start of the study by 6 years. Eight patients showed an increase in maximum tapping rate with levodopa (E(max)) approaching a steady state after 16 months. Ten patients showed an increase in E(max) with a peak at 31 months. One patient showed a linear decrease and another patient did not change over the 48 months. Longitudinal progress models were used to describe the time course of pharmacokinetic and pharmacodynamic parameters over 4 years. Peak treatment benefit, defined as the difference between E(max) and E0(1) or E0(2) (D(max)1 or D(max)2), increased with time particularly after the 3-day levodopa withdrawal. Deterioration of pre-dose motor function (E0) as disease progresses coupled with a greater amplitude of response due to levodopa (D(max)) could be a key factor contributing to motor fluctuations associated with long-term levodopa treatment.


Subject(s)
Dopamine Agents/pharmacology , Dopamine Agents/pharmacokinetics , Levodopa/pharmacology , Levodopa/pharmacokinetics , Parkinson Disease/metabolism , Administration, Oral , Adult , Aged , Dopamine Agents/administration & dosage , Humans , Infusions, Intravenous , Levodopa/administration & dosage , Middle Aged , Models, Biological , Motor Activity/drug effects , Parkinson Disease/drug therapy , Time Factors
19.
J Pharmacokinet Pharmacodyn ; 31(3): 243-68, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15518246

ABSTRACT

Clinicians recognize levodopa has a short-duration response (measured in hr) and a long-duration response (measured in days) in Parkinson's disease. In addition there is a diurnal pattern of motor function with better function in the morning. Previous pharmacokinetic-pharmacodynamic modeling has quantified only the short-duration response. We have developed a pharmacokinetic-pharmacodynamic model for the short- and long-duration responses to exogenous levodopa and the effects of residual endogenous levodopa synthesis in patients with Parkinson's disease. Thirteen previously untreated (de novo) patients with Parkinson's disease and twelve patients who had received levodopa orally for 9.7+/-4.0 years (chronic) were investigated. A 2 hr IV infusion of levodopa with concomitant oral carbidopa was given on two occasions separated by 3 days with no levodopa in between. A two compartment pharmacokinetic model was used to fit plasma levodopa concentrations. A sigmoid Emax model was used to relate concentrations from endogenous and exogenous sources to tapping rate (a measure of motor response). A model incorporating three effect compartments (fast equilibration (half life, Teqf). slow equilibration (Teqs) and dopa synthesis (Teqd)), yielded the most descriptive model for levodopa pharmacokinetics and pharmacodynamics. Baseline tapping rate reflected endogenous levodopa synthesis and the long-duration response. Partial loss of the long-duration response during the 3 days without levodopa in the chronic group lowered baseline tapping (36+/-7%, mean+/-SEM) and increased maximum levodopa induced response above baseline (112+/-31%). The maximum levodopa induced response after the drug holiday is a result of lowered baseline tapping due to the loss of long-duration response and not due to a change in levodopa pharmacokinetics or pharmacodynamics.


Subject(s)
Levodopa/administration & dosage , Levodopa/blood , Models, Biological , Parkinson Disease/blood , Parkinson Disease/drug therapy , Adult , Aged , Analysis of Variance , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Statistics, Nonparametric , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...