Using Bayesian-PBPK modeling for assessment of inter-individual variability and subgroup stratification

PurposeInter-individual variability in clinical endpoints and occurrence of potentially severe adverse effects represent an enormous challenge in drug development at all phases of (pre-)clinical research. To ensure patient safety it is important to identify adverse events or critical subgroups within the population as early as possible. Hence, a comprehensive understanding of the processes governing pharmacokinetics and pharmacodynamics is of utmost importance. In this paper we combine Bayesian statistics with detailed mechanistic physiologically-based pharmacokinetic (PBPK) models. On the example of pravastatin we demonstrate that this combination provides a powerful tool to investigate inter-individual variability in groups of patients and to identify clinically relevant homogenous subgroups in an unsupervised approach. Since PBPK models allow the identification of physiological, drug-specific and genotype-specific knowledge separately, our approach supports knowledge-based extrapolation to other drugs or populations.MethodsPBPK models are based on generic distribution models and extensive collections of physiological parameters and allow a mechanistic investigation of drug distribution and drug action. To systematically account for parameter variability within patient populations, a Bayesian-PBPK approach is developed rigorously quantifying the probability of a parameter given the amount of information contained in the measured data. Since these parameter distributions are high-dimensional, a Markov chain Monte Carlo algorithm is used, where the physiological and drug-specific parameters are considered in separate blocks.ResultsConsidering pravastatin pharmacokinetics as an application example, Bayesian-PBPK is used to investigate inter-individual variability in a cohort of 10 patients. Correlation analyses infer structural information about the PBPK model. Moreover, homogeneous subpopulations are identified a posteriori by examining the parameter distributions, which can even be assigned to a polymorphism in the hepatic organ anion transporter OATP1B1.ConclusionsThe presented Bayesian-PBPK approach systematically characterizes inter-individual variability within a population by updating prior knowledge about physiological parameters with new experimental data. Moreover, clinically relevant homogeneous subpopulations can be mechanistically identified. The large scale PBPK model separates physiological and drug-specific knowledge which allows, in combination with Bayesian approaches, the iterative assessment of specific populations by integrating information from several drugs.

[1]  J. Hampe,et al.  A Mechanistic, Model-Based Approach to Safety Assessment in Clinical Development , 2012, CPT: pharmacometrics & systems pharmacology.

[2]  R. Milo,et al.  Variability and memory of protein levels in human cells , 2006, Nature.

[3]  P. Neuvonen,et al.  SLCO1B1 polymorphism and sex affect the pharmacokinetics of pravastatin but not fluvastatin , 2006, Clinical pharmacology and therapeutics.

[4]  Walter Schmitt,et al.  PK-Sim®: a physiologically based pharmacokinetic ‘whole-body’ model , 2003 .

[5]  J S Evans,et al.  The effect of neglecting correlations when propagating uncertainty and estimating the population distribution of risk. , 1992, Risk analysis : an official publication of the Society for Risk Analysis.

[6]  F. Theil,et al.  Prediction of adipose tissue: plasma partition coefficients for structurally unrelated drugs. , 2001, Journal of pharmaceutical sciences.

[7]  A. Serajuddin,et al.  Relative lipophilicities, solubilities, and structure-pharmacological considerations of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors pravastatin, lovastatin, mevastatin, and simvastatin. , 1991, Journal of pharmaceutical sciences.

[8]  P. Georgopoulos,et al.  A Bayesian population PBPK model for multiroute chloroform exposure , 2010, Journal of Exposure Science and Environmental Epidemiology.

[9]  S. Willmann,et al.  Risk to the Breast‐Fed Neonate From Codeine Treatment to the Mother: A Quantitative Mechanistic Modeling Study , 2009, Clinical pharmacology and therapeutics.

[10]  W. Schmitt,et al.  A Physiologic Model for Simulating Gastrointestinal Flow and Drug Absorption in Rats , 2003, Pharmaceutical Research.

[11]  S. Singhvi,et al.  Biotransformation of pravastatin sodium in humans. , 1991, Drug metabolism and disposition: the biological fate of chemicals.

[12]  Malcolm Rowland,et al.  Physiologically-based pharmacokinetics in drug development and regulatory science. , 2011, Annual review of pharmacology and toxicology.

[13]  J Schüttler,et al.  Population Pharmacokinetics of Propofol: A Multicenter Study , 2000, Anesthesiology.

[14]  M. Niemi,et al.  Influence of Drug Transporter Polymorphisms on Pravastatin Pharmacokinetics in Humans , 2007, Pharmaceutical Research.

[15]  Ivan Nestorov,et al.  Whole-body physiologically based pharmacokinetic models , 2007, Expert opinion on drug metabolism & toxicology.

[16]  S. Singhvi,et al.  Disposition of pravastatin sodium, a tissue-selective HMG-CoA reductase inhibitor, in healthy subjects. , 1990, British journal of clinical pharmacology.

[17]  B. Carlin,et al.  Diagnostics: A Comparative Review , 2022 .

[18]  Walter Schmitt,et al.  Development of a Physiology-Based Whole-Body Population Model for Assessing the Influence of Individual Variability on the Pharmacokinetics of Drugs , 2007, Journal of Pharmacokinetics and Pharmacodynamics.

[19]  S. Shapiro,et al.  An Analysis of Variance Test for Normality (Complete Samples) , 1965 .

[20]  J. Rosenthal,et al.  On adaptive Markov chain Monte Carlo algorithms , 2005 .

[21]  Stefan Willmann,et al.  Pharmacogenomics of codeine, morphine, and morphine-6-glucuronide: model-based analysis of the influence of CYP2D6 activity, UGT2B7 activity, renal impairment, and CYP3A4 inhibition. , 2012, Molecular diagnosis & therapy.

[22]  L. Kuepfer,et al.  Using Expression Data for Quantification of Active Processes in Physiologically Based Pharmacokinetic Modeling , 2012, Drug Metabolism and Disposition.

[23]  A. Doucet,et al.  Particle Markov chain Monte Carlo methods , 2010 .

[24]  T. Hatanaka Clinical Pharmacokinetics of Pravastatin , 2000, Clinical pharmacokinetics.

[25]  F Y Bois,et al.  Statistical issues in toxicokinetic modeling: a bayesian perspective. , 2000, Environmental health perspectives.

[26]  Walter Schmitt,et al.  Physiology-based pharmacokinetic modeling: ready to be used. , 2004, Drug discovery today. Technologies.

[27]  K. Sandy Pang,et al.  Physiologically-based pharmacokinetic modeling for absorption, transport, metabolism and excretion , 2010, Journal of Pharmacokinetics and Pharmacodynamics.

[28]  Wolfgang Weiss,et al.  A Computational Systems Biology Software Platform for Multiscale Modeling and Simulation: Integrating Whole-Body Physiology, Disease Biology, and Molecular Reaction Networks , 2011, Front. Physio..

[29]  Henk-Jan Guchelaar,et al.  Translating Pharmacogenomics: Challenges on the Road to the Clinic , 2007, PLoS medicine.

[30]  H Derendorf,et al.  Basic concepts of pharmacokinetic/pharmacodynamic (PK/PD) modelling. , 1997, International journal of clinical pharmacology and therapeutics.

[31]  Donald Geman,et al.  Stochastic relaxation, Gibbs distributions, and the Bayesian restoration of images , 1984 .

[32]  Stefan Willmann,et al.  Pharmacogenomics of Codeine, Morphine, and Morphine-6-Glucuronide , 2012, Molecular Diagnosis & Therapy.

[33]  Heikki Haario,et al.  Componentwise adaptation for high dimensional MCMC , 2005, Comput. Stat..

[34]  M. Jamei,et al.  PBPK modelling of inter-individual variability in the pharmacokinetics of environmental chemicals. , 2010, Toxicology.

[35]  N. Metropolis,et al.  Equation of State Calculations by Fast Computing Machines , 1953, Resonance.

[36]  Adrian F. M. Smith,et al.  Sampling-Based Approaches to Calculating Marginal Densities , 1990 .

[37]  H. Haario,et al.  An adaptive Metropolis algorithm , 2001 .

[38]  W. K. Hastings,et al.  Monte Carlo Sampling Methods Using Markov Chains and Their Applications , 1970 .

[39]  A. Gelman,et al.  Physiological Pharmacokinetic Analysis Using Population Modeling and Informative Prior Distributions , 1996 .

[40]  P. Sorger,et al.  Non-genetic origins of cell-to-cell variability in TRAIL-induced apoptosis , 2009, Nature.

[41]  M. Rowland,et al.  Physiologically based pharmacokinetic modeling 1: predicting the tissue distribution of moderate-to-strong bases. , 2005, Journal of pharmaceutical sciences.

[42]  Lars Kuepfer,et al.  Multiscale mechanistic modeling in pharmaceutical research and development. , 2012, Advances in experimental medicine and biology.

[43]  Nando de Freitas,et al.  An Introduction to MCMC for Machine Learning , 2004, Machine Learning.

[44]  M. Rowland,et al.  Physiologically based pharmacokinetic modelling 2: predicting the tissue distribution of acids, very weak bases, neutrals and zwitterions. , 2006, Journal of pharmaceutical sciences.

[45]  Steffen Borchers,et al.  Integrating Cellular Metabolism into a Multiscale Whole-Body Model , 2012, PLoS Comput. Biol..

[46]  Stefan Willmann,et al.  Physiology-Based Simulations of a Pathological Condition , 2008, Clinical pharmacokinetics.

[47]  Ivelina Gueorguieva,et al.  Diazepam Pharamacokinetics from Preclinical to Phase I Using a Bayesian Population Physiologically Based Pharmacokinetic Model with Informative Prior Distributions in Winbugs , 2006, Journal of Pharmacokinetics and Pharmacodynamics.

[48]  G. Roberts,et al.  Adaptive Markov Chain Monte Carlo through Regeneration , 1998 .

[49]  G. Johanson,et al.  Physiologically based modeling of the inhalation kinetics of styrene in humans using a bayesian population approach. , 2002, Toxicology and applied pharmacology.

[50]  Gareth O. Roberts,et al.  Examples of Adaptive MCMC , 2009 .

[51]  A. Bowman,et al.  Applied smoothing techniques for data analysis : the kernel approach with S-plus illustrations , 1999 .

[52]  Walter Schmitt,et al.  A physiological model for the estimation of the fraction dose absorbed in humans. , 2004, Journal of medicinal chemistry.

[53]  William M. Bolstad,et al.  Understanding Computational Bayesian Statistics: Bolstad/Understanding , 2009 .

[54]  R. Collins,et al.  SLCO1B1 variants and statin-induced myopathy--a genomewide study. , 2008, The New England journal of medicine.