1
|
Alfosea-Cuadrado GM, Zarzoso-Foj J, Adell A, Valverde-Navarro AA, González-Soler EM, Mangas-Sanjuán V, Blasco-Serra A. Population Pharmacokinetic-Pharmacodynamic Analysis of a Reserpine-Induced Myalgia Model in Rats. Pharmaceutics 2024; 16:1101. [PMID: 39204446 PMCID: PMC11359992 DOI: 10.3390/pharmaceutics16081101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/11/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
(1) Background: Fibromyalgia syndrome (FMS) is a chronic pain condition with widespread pain and multiple comorbidities, for which conventional therapies offer limited benefits. The reserpine-induced myalgia (RIM) model is an efficient animal model of FMS in rodents. This study aimed to develop a pharmacokinetic-pharmacodynamic (PK-PD) model of reserpine in rats, linking to its impact on monoamines (MAs). (2) Methods: Reserpine was administered daily for three consecutive days at dose levels of 0.1, 0.5, and 1 mg/kg. A total of 120 rats were included, and 120 PK and 828 PD observations were collected from 48 to 96 h after the first dose of reserpine. Non-linear mixed-effect data analysis was applied for structural PK-PD model definition, variability characterization, and covariate analysis. (3) Results: A one-compartment model best described reserpine in rats (V = 1.3 mL/kg and CL = 4.5 × 10-1 mL/h/kg). A precursor-pool PK-PD model (kin = 6.1 × 10-3 mg/h, kp = 8.6 × 10-4 h-1 and kout = 2.7 × 10-2 h-1) with a parallel transit chain (k0 = 1.9 × 10-1 h-1) characterized the longitudinal levels of MA in the prefrontal cortex, spinal cord, and amygdala in rats. Reserpine stimulates the degradation of MA from the pool compartment (Slope1 = 1.1 × 10-1 h) and the elimination of MA (Slope2 = 1.25 h) through the transit chain. Regarding the reference dose (1 mg/kg) of the RIM model, the administration of 4 mg/kg would lead to a mean reduction of 65% (Cmax), 80% (Cmin), and 70% (AUC) of MA across the brain regions tested. (4) Conclusions: Regional brain variations in neurotransmitter depletion were identified, particularly in the amygdala, offering insights for therapeutic strategies and biomarker identification in FMS research.
Collapse
Affiliation(s)
- Gloria M. Alfosea-Cuadrado
- Department of Human Anatomy and Embryology, University of Valencia, 46010 Valencia, Spain; (G.M.A.-C.); (A.A.V.-N.); (A.B.-S.)
| | - Javier Zarzoso-Foj
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, 46100 Valencia, Spain;
- Interuniversity Research Institute for Molecular Recognition and Technological Development, Polytechnic University of Valencia, University of Valencia, 46100 Valencia, Spain
| | - Albert Adell
- Systems Neurobiology, Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC), Spanish National Research Council (CSIC), 39011 Santander, Spain;
- Biomedical Research Networking Centre for Mental Health (CIBERSAM), 39011 Santander, Spain
| | - Alfonso A. Valverde-Navarro
- Department of Human Anatomy and Embryology, University of Valencia, 46010 Valencia, Spain; (G.M.A.-C.); (A.A.V.-N.); (A.B.-S.)
| | - Eva M. González-Soler
- Department of Human Anatomy and Embryology, University of Valencia, 46010 Valencia, Spain; (G.M.A.-C.); (A.A.V.-N.); (A.B.-S.)
| | - Víctor Mangas-Sanjuán
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, 46100 Valencia, Spain;
- Interuniversity Research Institute for Molecular Recognition and Technological Development, Polytechnic University of Valencia, University of Valencia, 46100 Valencia, Spain
| | - Arantxa Blasco-Serra
- Department of Human Anatomy and Embryology, University of Valencia, 46010 Valencia, Spain; (G.M.A.-C.); (A.A.V.-N.); (A.B.-S.)
| |
Collapse
|
2
|
Kim S, Hooker AC, Shi Y, Kim GHJ, Wong WK. Metaheuristics for pharmacometrics. CPT Pharmacometrics Syst Pharmacol 2021; 10:1297-1309. [PMID: 34562342 PMCID: PMC8592519 DOI: 10.1002/psp4.12714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 08/06/2021] [Accepted: 09/07/2021] [Indexed: 12/22/2022] Open
Abstract
Metaheuristics is a powerful optimization tool that is increasingly used across disciplines to tackle general purpose optimization problems. Nature-inspired metaheuristic algorithms is a subclass of metaheuristic algorithms and have been shown to be particularly flexible and useful in solving complicated optimization problems in computer science and engineering. A common practice with metaheuristics is to hybridize it with another suitably chosen algorithm for enhanced performance. This paper reviews metaheuristic algorithms and demonstrates some of its utility in tackling pharmacometric problems. Specifically, we provide three applications using one of its most celebrated members, particle swarm optimization (PSO), and show that PSO can effectively estimate parameters in complicated nonlinear mixed-effects models and to gain insights into statistical identifiability issues in a complex compartment model. In the third application, we demonstrate how to hybridize PSO with sparse grid, which is an often-used technique to evaluate high dimensional integrals, to search for D -efficient designs for estimating parameters in nonlinear mixed-effects models with a count outcome. We also show the proposed hybrid algorithm outperforms its competitors when sparse grid is replaced by its competitor, adaptive gaussian quadrature to approximate the integral, or when PSO is replaced by three notable nature-inspired metaheuristic algorithms.
Collapse
Affiliation(s)
- Seongho Kim
- Department of OncologyWayne State UniversityDetroitMichiganUSA
| | | | - Yu Shi
- Department of BiostatisticsUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Grace Hyun J. Kim
- Department of BiostatisticsUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Weng Kee Wong
- Department of BiostatisticsUniversity of California Los AngelesLos AngelesCaliforniaUSA
| |
Collapse
|
3
|
Giráldez-Montero JM, Gonzalez-Lopez J, Campos-Toimil M, Lamas-Díaz MJ. Therapeutic drug monitoring of anti-tumour necrosis factor-α agents in inflammatory bowel disease: Limits and improvements. Br J Clin Pharmacol 2020; 87:2216-2227. [PMID: 33197071 DOI: 10.1111/bcp.14654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/28/2020] [Accepted: 11/08/2020] [Indexed: 11/27/2022] Open
Abstract
AIMS Since the publication of the American Gastroenterological Association's recommendations in 2017, there have been no significant changes in the biological monitoring recommendations in inflammatory bowel disease. Possible limitations are the lack of evidence to recommend proactive therapeutic drug monitoring (pTDM) over reactive TDM (rTDM), and the limited information about individualized dosing methods. This article aims to review the TDM strategy updates and the use of individualized dosing methods. METHODS For the analysis of the TDM strategies and individualized dosing method, a search was carried out in PubMed and Cochrane Central. In the TDM case, since August 2017. RESULTS A total of 263 publications were found, but only 7 related to proactive TDM. Five of these publications directly compared pTDM vs rTDM and 2 were randomized clinical trials. Six studies found benefits of pTDM and 1 found no differences. Regarding the individualized dosing method, 229 distinct results were found. Population pharmacokinetics was the most widely used method to develop individual dosage models and to analyse the influence of factors on drug concentrations (albumin concentration, weight, presence of anti-drug antibodies etc). CONCLUSION We have found no major changes in TDM strategies. There is a growing trend towards the use of pTDM because it has shown a longer duration of treatment response, lower rates of discontinuation and relapses. However, the available evidence is limited and of low quality. Despite the common use of population pharmacokinetic methods to analyse pharmacokinetic factors, they are not commonly used for personalized dosing.
Collapse
Affiliation(s)
- José María Giráldez-Montero
- Department of Pharmacy, Hospital Clínico Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Spain.,Clinical Pharmacology Group, Hospital Clínico Universitario de Santiago de Compostela (SERGAS), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Jaime Gonzalez-Lopez
- Department of Pharmacy, Hospital Clínico Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Spain.,Clinical Pharmacology Group, Hospital Clínico Universitario de Santiago de Compostela (SERGAS), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Manuel Campos-Toimil
- Group of Research on Physiology and Pharmacology of Chronic Diseases (FIFAEC), Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain.,Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - María Jesús Lamas-Díaz
- Clinical Pharmacology Group, Hospital Clínico Universitario de Santiago de Compostela (SERGAS), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| |
Collapse
|
4
|
Dumont C, Lestini G, Le Nagard H, Mentré F, Comets E, Nguyen TT. PFIM 4.0, an extended R program for design evaluation and optimization in nonlinear mixed-effect models. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2018; 156:217-229. [PMID: 29428073 DOI: 10.1016/j.cmpb.2018.01.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 12/22/2017] [Accepted: 01/10/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND AND OBJECTIVE Nonlinear mixed-effect models (NLMEMs) are increasingly used for the analysis of longitudinal studies during drug development. When designing these studies, the expected Fisher information matrix (FIM) can be used instead of performing time-consuming clinical trial simulations. The function PFIM is the first tool for design evaluation and optimization that has been developed in R. In this article, we present an extended version, PFIM 4.0, which includes several new features. METHODS Compared with version 3.0, PFIM 4.0 includes a more complete pharmacokinetic/pharmacodynamic library of models and accommodates models including additional random effects for inter-occasion variability as well as discrete covariates. A new input method has been added to specify user-defined models through an R function. Optimization can be performed assuming some fixed parameters or some fixed sampling times. New outputs have been added regarding the FIM such as eigenvalues, conditional numbers, and the option of saving the matrix obtained after evaluation or optimization. Previously obtained results, which are summarized in a FIM, can be taken into account in evaluation or optimization of one-group protocols. This feature enables the use of PFIM for adaptive designs. The Bayesian individual FIM has been implemented, taking into account a priori distribution of random effects. Designs for maximum a posteriori Bayesian estimation of individual parameters can now be evaluated or optimized and the predicted shrinkage is also reported. It is also possible to visualize the graphs of the model and the sensitivity functions without performing evaluation or optimization. RESULTS The usefulness of these approaches and the simplicity of use of PFIM 4.0 are illustrated by two examples: (i) an example of designing a population pharmacokinetic study accounting for previous results, which highlights the advantage of adaptive designs; (ii) an example of Bayesian individual design optimization for a pharmacodynamic study, showing that the Bayesian individual FIM can be a useful tool in therapeutic drug monitoring, allowing efficient prediction of estimation precision and shrinkage for individual parameters. CONCLUSION PFIM 4.0 is a useful tool for design evaluation and optimization of longitudinal studies in pharmacometrics and is freely available at http://www.pfim.biostat.fr.
Collapse
Affiliation(s)
- Cyrielle Dumont
- IAME, UMR 1137, INSERM and University Paris Diderot, Sorbonne Paris Cité, Paris, F-75018, France; University of Lille, EA 2694, Public Health: Epidemiology and Healthcare Quality, ILIS, Lille, F-59000, France
| | - Giulia Lestini
- IAME, UMR 1137, INSERM and University Paris Diderot, Sorbonne Paris Cité, Paris, F-75018, France
| | - Hervé Le Nagard
- IAME, UMR 1137, INSERM and University Paris Diderot, Sorbonne Paris Cité, Paris, F-75018, France
| | - France Mentré
- IAME, UMR 1137, INSERM and University Paris Diderot, Sorbonne Paris Cité, Paris, F-75018, France
| | - Emmanuelle Comets
- IAME, UMR 1137, INSERM and University Paris Diderot, Sorbonne Paris Cité, Paris, F-75018, France
| | - Thu Thuy Nguyen
- IAME, UMR 1137, INSERM and University Paris Diderot, Sorbonne Paris Cité, Paris, F-75018, France.
| |
Collapse
|
5
|
Individual Bayesian Information Matrix for Predicting Estimation Error and Shrinkage of Individual Parameters Accounting for Data Below the Limit of Quantification. Pharm Res 2017; 34:2119-2130. [DOI: 10.1007/s11095-017-2217-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/19/2017] [Indexed: 11/26/2022]
|
6
|
Wu M, Diez-Roux A, Raghunathan TE, Sánchez BN. FPCA-based method to select optimal sampling schedules that capture between-subject variability in longitudinal studies. Biometrics 2017; 74:229-238. [PMID: 28482120 DOI: 10.1111/biom.12714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 04/01/2017] [Accepted: 04/01/2017] [Indexed: 11/27/2022]
Abstract
A critical component of longitudinal study design involves determining the sampling schedule. Criteria for optimal design often focus on accurate estimation of the mean profile, although capturing the between-subject variance of the longitudinal process is also important since variance patterns may be associated with covariates of interest or predict future outcomes. Existing design approaches have limited applicability when one wishes to optimize sampling schedules to capture between-individual variability. We propose an approach to derive optimal sampling schedules based on functional principal component analysis (FPCA), which separately characterizes the mean and the variability of longitudinal profiles and leads to a parsimonious representation of the temporal pattern of the variability. Simulation studies show that the new design approach performs equally well compared to an existing approach based on parametric mixed model (PMM) when a PMM is adequate for the data, and outperforms the PMM-based approach otherwise. We use the methods to design studies aiming to characterize daily salivary cortisol profiles and identify the optimal days within the menstrual cycle when urinary progesterone should be measured.
Collapse
Affiliation(s)
- Meihua Wu
- Gilead Sciences, Inc., Foster City, California 94404, U.S.A
| | - Ana Diez-Roux
- Department of Epidemiology and Biostatistics, Drexel University, Philadelphia, Pennsylvania 19104, U.S.A
| | | | - Brisa N Sánchez
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan 48109, U.S.A
| |
Collapse
|
7
|
Nguyen TT, Bénech H, Delaforge M, Lenuzza N. Design optimisation for pharmacokinetic modeling of a cocktail of phenotyping drugs. Pharm Stat 2015; 15:165-77. [DOI: 10.1002/pst.1731] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Indexed: 12/24/2022]
Affiliation(s)
- Thu Thuy Nguyen
- CEA, LIST; Data Analysis and Systems Intelligence Laboratory; Gif-sur-Yvette France
| | | | | | - Natacha Lenuzza
- CEA, LIST; Data Analysis and Systems Intelligence Laboratory; Gif-sur-Yvette France
| |
Collapse
|
8
|
Mould DR, Walz AC, Lave T, Gibbs JP, Frame B. Developing Exposure/Response Models for Anticancer Drug Treatment: Special Considerations. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2015. [PMID: 26225225 PMCID: PMC4369756 DOI: 10.1002/psp4.16] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Anticancer agents often have a narrow therapeutic index (TI), requiring precise dosing to ensure sufficient exposure for clinical activity while minimizing toxicity. These agents frequently have complex pharmacology, and combination therapy may cause schedule-specific effects and interactions. We review anticancer drug development, showing how integration of modeling and simulation throughout development can inform anticancer dose selection, potentially improving the late-phase success rate. This article has a companion article in Clinical Pharmacology & Therapeutics with practical examples.
Collapse
Affiliation(s)
- D R Mould
- Projections Research Phoenixville, Pennsylvania, USA
| | - A-C Walz
- Roche Pharma Research and Early Development, Modeling & Simulation, Pharmaceutical Sciences, Roche Innovation Center Basel F. Hoffmann-La Roche, Basel, Switzerland
| | - T Lave
- Roche Pharma Research and Early Development, Modeling & Simulation, Pharmaceutical Sciences, Roche Innovation Center Basel F. Hoffmann-La Roche, Basel, Switzerland
| | - J P Gibbs
- Amgen Thousand Oaks, California, USA
| | - B Frame
- Projections Research Phoenixville, Pennsylvania, USA
| |
Collapse
|
9
|
Dumont C, Chenel M, Mentré F. Influence of covariance between random effects in design for nonlinear mixed-effect models with an illustration in pediatric pharmacokinetics. J Biopharm Stat 2014; 24:471-92. [PMID: 24697342 DOI: 10.1080/10543406.2014.888443] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Nonlinear mixed-effect models are used increasingly during drug development. For design, an alternative to simulations is based on the Fisher information matrix. Its expression was derived using a first-order approach, was then extended to include covariance and implemented into the R function PFIM. The impact of covariance on standard errors, amount of information, and optimal designs was studied. It was also shown how standard errors can be predicted analytically within the framework of rich individual data without the model. The results were illustrated by applying this extension to the design of a pharmacokinetic study of a drug in pediatric development.
Collapse
Affiliation(s)
- Cyrielle Dumont
- a Université Paris Diderot, Sorbonne Paris Cité , UMR 738, INSERM, Paris , France
| | | | | |
Collapse
|
10
|
Nguyen TT, Mentré F. Evaluation of the Fisher information matrix in nonlinear mixed effect models using adaptive Gaussian quadrature. Comput Stat Data Anal 2014. [DOI: 10.1016/j.csda.2014.06.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
11
|
Volaufova J. Approximate Small-Sample Tests of Fixed Effects in Nonlinear Mixed Models. COMMUN STAT-SIMUL C 2014. [DOI: 10.1080/03610918.2013.835407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
12
|
Lange MR, Schmidli H. Optimal design of clinical trials with biologics using dose-time-response models. Stat Med 2014; 33:5249-64. [DOI: 10.1002/sim.6299] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 07/31/2014] [Accepted: 08/20/2014] [Indexed: 12/23/2022]
Affiliation(s)
- Markus R. Lange
- Statistical Methodology, Development; Novartis Pharma AG; Basel Switzerland
- Hannover Medical School; Institute for Biometry; Hannover Germany
| | - Heinz Schmidli
- Statistical Methodology, Development; Novartis Pharma AG; Basel Switzerland
| |
Collapse
|
13
|
Dumont C, Chenel M, Mentré F. Two-stage Adaptive Designs in Nonlinear Mixed Effects Models: Application to Pharmacokinetics in Children. COMMUN STAT-SIMUL C 2014. [DOI: 10.1080/03610918.2014.930901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Cyrielle Dumont
- IAME, UMR 1137, INSERM, Paris, France
- Univ Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Marylore Chenel
- Division of Clinical Pharmacokinetics, Institut de Recherches Internationales Servier, Suresnes, France
| | - France Mentré
- IAME, UMR 1137, INSERM, Paris, France
- Univ Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
14
|
Dumont C, Mentré F, Gaynor C, Brendel K, Gesson C, Chenel M. Optimal sampling times for a drug and its metabolite using SIMCYP(®) simulations as prior information. Clin Pharmacokinet 2013; 52:43-57. [PMID: 23212609 DOI: 10.1007/s40262-012-0022-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Since 2007, it is mandatory for the pharmaceutical companies to submit a Paediatric Investigation Plan to the Paediatric Committee at the European Medicines Agency for any drug in development in adults, and it often leads to the need to conduct a pharmacokinetic study in children. Pharmacokinetic studies in children raise ethical and methodological issues. Because of limitation of sampling times, appropriate methods, such as the population approach, are necessary for analysis of the pharmacokinetic data. The choice of the pharmacokinetic sampling design has an important impact on the precision of population parameter estimates. Approaches for design evaluation and optimization based on the evaluation of the Fisher information matrix (M(F)) have been proposed and are now implemented in several software packages, such as PFIM in R. OBJECTIVES The objectives of this work were to (1) develop a joint population pharmacokinetic model to describe the pharmacokinetic characteristics of a drug S and its active metabolite in children after intravenous drug administration from simulated plasma concentration-time data produced using physiologically based pharmacokinetic (PBPK) predictions; (2) optimize the pharmacokinetic sampling times for an upcoming clinical study using a multi-response design approach, considering clinical constraints; and (3) evaluate the resulting design taking data below the lower limit of quantification (BLQ) into account. METHODS Plasma concentration-time profiles were simulated in children using a PBPK model previously developed with the software SIMCYP(®) for the parent drug and its active metabolite. Data were analysed using non-linear mixed-effect models with the software NONMEM(®), using a joint model for the parent drug and its metabolite. The population pharmacokinetic design, for the future study in 82 children from 2 to 18 years old, each receiving a single dose of the drug, was then optimized using PFIM, assuming identical times for parent and metabolite concentration measurements and considering clinical constraints. Design evaluation was based on the relative standard errors (RSEs) of the parameters of interest. In the final evaluation of the proposed design, an approach was used to assess the possible effect of BLQ concentrations on the design efficiency. This approach consists of rescaling the M(F), using, at each sampling time, the probability of observing a concentration BLQ computed from Monte-Carlo simulations. RESULTS A joint pharmacokinetic model with three compartments for the parent drug and one for its active metabolite, with random effects on four parameters, was used to fit the simulated PBPK concentration-time data. A combined error model best described the residual variability. Parameters and dose were expressed per kilogram of bodyweight. Reaching a compromise between PFIM results and clinical constraints, the optimal design was composed of four samples at 0.1, 1.8, 5 and 10 h after drug injection. This design predicted RSE lower than 30 % for the four parameters of interest. For this design, rescaling M(F) for BLQ data had very little influence on predicted RSE. CONCLUSION PFIM was a useful tool to find an optimal sampling design in children, considering clinical constraints. Even if it was not forecasted initially by the investigators, this approach showed that it was really necessary to include a late sampling time for all children. Moreover, we described an approach to evaluate designs assuming expected proportions of BLQ data are omitted.
Collapse
Affiliation(s)
- Cyrielle Dumont
- Division of Clinical Pharmacokinetics, Institut de Recherches Internationales Servier, Suresnes, France.
| | | | | | | | | | | |
Collapse
|
15
|
Saleh MI, Nalbant D, Widness JA, Veng-Pedersen P. Population pharmacodynamic analysis of erythropoiesis in preterm infants for determining the anemia treatment potential of erythropoietin. Am J Physiol Regul Integr Comp Physiol 2013; 304:R772-81. [PMID: 23485870 DOI: 10.1152/ajpregu.00173.2012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
A population pharmacokinetics/pharmacodynamic (PK/PD) model was developed to describe changes in erythropoiesis as a function of plasma erythropoietin (EPO) concentration over the first 30 days of life in preterm infants who developed severe anemia requiring red blood cell (RBC) transfusion. Several covariates were tested as possible factors influencing the responsiveness to EPO. Discarded blood samples in 27 ventilated preterm infants born at 24-29 wk of gestation were used to construct plasma EPO, hemoglobin (Hb), and RBC concentration-time profiles. The amount of Hb removed for laboratory testing and that transfused throughout the study period were recorded. A population PK/PD model accounting for the dynamic Hb changes experienced by these infants was simultaneously fitted to plasma EPO, Hb, and RBC concentrations. A covariate analysis suggested that the erythropoietic efficacy of EPO is increased for preterm infants at later gestational ages. The PD analysis showed a sevenfold difference in maximum Hb production rate dependent on gestational age and indicated that preterm infants, when stimulated by EPO, have the capacity to produce additional Hb that may result in a decrease in RBC transfusions. The present model has utility in clinical trial simulations investigating the treatment potential of erythropoietic stimulating agents in the treatment of anemia of prematurity.
Collapse
Affiliation(s)
- Mohammad I Saleh
- Division of Pharmaceutics, College of Pharmacy, The University of Iowa, Iowa City, IA 52212, USA
| | | | | | | |
Collapse
|
16
|
Nyberg J, Höglund R, Bergstrand M, Karlsson MO, Hooker AC. Serial correlation in optimal design for nonlinear mixed effects models. J Pharmacokinet Pharmacodyn 2012; 39:239-49. [PMID: 22415637 DOI: 10.1007/s10928-012-9245-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 02/26/2012] [Indexed: 10/28/2022]
Abstract
In population modeling two sources of variability are commonly included; inter individual variability and residual variability. Rich sampling optimal design (more samples than model parameters) using these models will often result in a sampling schedule where some measurements are taken at exactly the same time point, thereby maximizing the signal-to-noise ratio. This behavior is a result of not appropriately taking into account error generation mechanisms and is often clinically unappealing and may be avoided by including intrinsic variability, i.e. serially correlated residual errors. In this paper we extend previous work that investigated optimal designs of population models including serial correlation using stochastic differential equations to optimal design with the more robust, and analytic, AR(1) autocorrelation model. Further, we investigate the importance of correlation strength, design criteria and robust designs. Finally, we explore the optimal design properties when estimating parameters with and without serial correlation. In the investigated examples the designs and estimation performance differs significantly when handling serial correlation.
Collapse
Affiliation(s)
- Joakim Nyberg
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-751 24 Uppsala, Sweden.
| | | | | | | | | |
Collapse
|
17
|
Vong C, Bergstrand M, Nyberg J, Karlsson MO. Rapid sample size calculations for a defined likelihood ratio test-based power in mixed-effects models. AAPS JOURNAL 2012; 14:176-86. [PMID: 22350626 DOI: 10.1208/s12248-012-9327-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 01/27/2012] [Indexed: 11/30/2022]
Abstract
Efficient power calculation methods have previously been suggested for Wald test-based inference in mixed-effects models but the only available alternative for Likelihood ratio test-based hypothesis testing has been to perform computer-intensive multiple simulations and re-estimations. The proposed Monte Carlo Mapped Power (MCMP) method is based on the use of the difference in individual objective function values (ΔiOFV) derived from a large dataset simulated from a full model and subsequently re-estimated with the full and reduced models. The ΔiOFV is sampled and summed (∑ΔiOFVs) for each study at each sample size of interest to study, and the percentage of ∑ΔiOFVs greater than the significance criterion is taken as the power. The power versus sample size relationship established via the MCMP method was compared to traditional assessment of model-based power for six different pharmacokinetic and pharmacodynamic models and designs. In each case, 1,000 simulated datasets were analysed with the full and reduced models. There was concordance in power between the traditional and MCMP methods such that for 90% power, the difference in required sample size was in most investigated cases less than 10%. The MCMP method was able to provide relevant power information for a representative pharmacometric model at less than 1% of the run-time of an SSE. The suggested MCMP method provides a fast and accurate prediction of the power and sample size relationship.
Collapse
Affiliation(s)
- Camille Vong
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, 75124, Uppsala, Sweden.
| | | | | | | |
Collapse
|
18
|
Aliev A, Fedorov V, Leonov S, McHugh B, Magee M. PkStaMp Library for Constructing Optimal Population Designs for PK/PD Studies. COMMUN STAT-SIMUL C 2012. [DOI: 10.1080/03610918.2012.625273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
19
|
Nguyen TT, Bazzoli C, Mentré F. Design evaluation and optimisation in crossover pharmacokinetic studies analysed by nonlinear mixed effects models. Stat Med 2011; 31:1043-58. [PMID: 21965170 DOI: 10.1002/sim.4390] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 07/13/2011] [Accepted: 08/04/2011] [Indexed: 01/15/2023]
Abstract
Bioequivalence or interaction trials are commonly studied in crossover design and can be analysed by nonlinear mixed effects models as an alternative to noncompartmental approach. We propose an extension of the population Fisher information matrix in nonlinear mixed effects models to design crossover pharmacokinetic trials, using a linearisation of the model around the random effect expectation, including within-subject variability and discrete covariates fixed or changing between periods. We use the expected standard errors of treatment effect to compute the power for the Wald test of comparison or equivalence and the number of subjects needed for a given power. We perform various simulations mimicking crossover two-period trials to show the relevance of these developments. We then apply these developments to design a crossover pharmacokinetic study of amoxicillin in piglets and implement them in the new version 3.2 of the r function PFIM.
Collapse
|
20
|
Dubois A, Lavielle M, Gsteiger S, Pigeolet E, Mentré F. Model-based analyses of bioequivalence crossover trials using the stochastic approximation expectation maximisation algorithm. Stat Med 2011; 30:2582-600. [DOI: 10.1002/sim.4286] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Accepted: 04/12/2011] [Indexed: 11/08/2022]
Affiliation(s)
- Anne Dubois
- INSERM UMR738, University Diderot Paris 7; Paris France
| | | | - Sandro Gsteiger
- Modeling and Simulation Department Novartis Pharma AG; Basel Switzerland
| | - Etienne Pigeolet
- Modeling and Simulation Department Novartis Pharma AG; Basel Switzerland
| | - France Mentré
- INSERM UMR738, University Diderot Paris 7; Paris France
| |
Collapse
|
21
|
Ogungbenro K, Aarons L. Population Fisher information matrix and optimal design of discrete data responses in population pharmacodynamic experiments. J Pharmacokinet Pharmacodyn 2011; 38:449-69. [PMID: 21660504 DOI: 10.1007/s10928-011-9203-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 05/30/2011] [Indexed: 11/26/2022]
Affiliation(s)
- Kayode Ogungbenro
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Oxford Road, Manchester, UK.
| | | |
Collapse
|
22
|
Guedj J, Bazzoli C, Neumann AU, Mentré F. Design evaluation and optimization for models of hepatitis C viral dynamics. Stat Med 2011; 30:1045-56. [PMID: 21337592 DOI: 10.1002/sim.4191] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Revised: 12/06/2010] [Accepted: 12/14/2010] [Indexed: 01/04/2023]
Abstract
Mathematical modeling of hepatitis C viral (HCV) kinetics is widely used for understanding viral pathogenesis and predicting treatment outcome. The standard model is based on a system of five non-linear ordinary differential equations (ODE) that describe both viral kinetics and changes in drug concentration after treatment initiation. In such complex models parameter estimation is challenging and requires frequent sampling measurements on each individual. By borrowing information between study subjects, non-linear mixed effect models can deal with sparser sampling from each individual. However, the search for optimal designs in this context has been limited by the numerical difficulty of evaluating the Fisher information matrix (FIM). Using the software PFIM, we show that a linearization of the statistical model avoids most of the computational burden, while providing a good approximation to the FIM. We then compare the precision of the parameters that can be expected using five study designs from the literature. We illustrate the usefulness of rationalizing data sampling by showing that, for a given level of precision, optimal design could reduce the total number of measurements by up 50 per cent. Our approach can be used by a statistician or a clinician aiming at designing an HCV viral kinetics study.
Collapse
Affiliation(s)
- Jeremie Guedj
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
| | | | | | | |
Collapse
|
23
|
Savic RM, Mentré F, Lavielle M. Implementation and evaluation of the SAEM algorithm for longitudinal ordered categorical data with an illustration in pharmacokinetics-pharmacodynamics. AAPS JOURNAL 2010; 13:44-53. [PMID: 21063925 DOI: 10.1208/s12248-010-9238-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Accepted: 10/07/2010] [Indexed: 11/30/2022]
Abstract
Analysis of longitudinal ordered categorical efficacy or safety data in clinical trials using mixed models is increasingly performed. However, algorithms available for maximum likelihood estimation using an approximation of the likelihood integral, including LAPLACE approach, may give rise to biased parameter estimates. The SAEM algorithm is an efficient and powerful tool in the analysis of continuous/count mixed models. The aim of this study was to implement and investigate the performance of the SAEM algorithm for longitudinal categorical data. The SAEM algorithm is extended for parameter estimation in ordered categorical mixed models together with an estimation of the Fisher information matrix and the likelihood. We used Monte Carlo simulations using previously published scenarios evaluated with NONMEM. Accuracy and precision in parameter estimation and standard error estimates were assessed in terms of relative bias and root mean square error. This algorithm was illustrated on the simultaneous analysis of pharmacokinetic and discretized efficacy data obtained after a single dose of warfarin in healthy volunteers. The new SAEM algorithm is implemented in MONOLIX 3.1 for discrete mixed models. The analyses show that for parameter estimation, the relative bias is low for both fixed effects and variance components in all models studied. Estimated and empirical standard errors are similar. The warfarin example illustrates how simple and rapid it is to analyze simultaneously continuous and discrete data with MONOLIX 3.1. The SAEM algorithm is extended for analysis of longitudinal categorical data. It provides accurate estimates parameters and standard errors. The estimation is fast and stable.
Collapse
|
24
|
Bazzoli C, Retout S, Mentré F. Design evaluation and optimisation in multiple response nonlinear mixed effect models: PFIM 3.0. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2010; 98:55-65. [PMID: 19892427 DOI: 10.1016/j.cmpb.2009.09.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2009] [Revised: 09/16/2009] [Accepted: 09/18/2009] [Indexed: 05/28/2023]
Abstract
Nonlinear mixed effect models (NLMEM) with multiple responses are increasingly used in pharmacometrics, one of the main examples being the joint analysis of the pharmacokinetics (PK) and pharmacodynamics (PD) of a drug. Efficient tools for design evaluation and optimisation in NLMEM are necessary. The R functions PFIM 1.2 and PFIMOPT 1.0 were proposed for these purposes, but accommodate only single response models. The methodology used is based on the Fisher information matrix, developed using a linearisation of the model. In this paper, we present an extended version, PFIM 3.0, dedicated to both design evaluation and optimisation for multiple response models, using a similar method as for single response models. In addition to handling multiple response models, several features have been integrated into PFIM 3.0 for model specification and optimisation. The extension includes a library of classical analytical pharmacokinetics models and allows the user to describe more complex models using differential equations. Regarding the optimisation algorithm, an alternative to the Simplex algorithm has been implemented, the Fedorov-Wynn algorithm to optimise more practical D-optimal design. Indeed, this algorithm optimises design among a set of sampling times specified by the user. This R function is freely available at http://www.pfim.biostat.fr. The efficiency of this approach and the simplicity of use of PFIM 3.0 are illustrated with a real example of the joint PKPD analysis of warfarin, an oral anticoagulant, with a model defined by ordinary differential equations.
Collapse
|
25
|
|