Quantitative prediction of formulation-specific food effects and their population variability from in vitro data with the physiologically-based ADAM model: A case study using the BCS/BDDCS Class II drug nifedipine

https://doi.org/10.1016/j.ejps.2013.09.006Get rights and content

Abstract

Quantitative prediction of food effects (FE) upon drug pharmacokinetics, including population variability, in advance of human trials may help with trial design by optimising the number of subjects and sampling times when a clinical study is warranted or by negating the need for conduct of clinical studies. Classification and rule-based systems such as the BCS and BDDCS and statistical QSARs are widely used to anticipate the nature of FE in early drug development. However, their qualitative rather than quantitative nature makes them less appropriate for assessing the magnitude of FE. Moreover, these approaches are based upon drug properties alone and are not appropriate for estimating potential formulation-specific FE on modified or controlled release products. In contrast, physiologically-based mechanistic models can consider the scope and interplay of a range of physiological changes after food intake and, in combination with appropriate in vitro drug- and formulation-specific data, can make quantitative predictions of formulation-specific FE including the inter-individual variability of such effects. Herein the Advanced Dissolution, Absorption and Metabolism (ADAM) model is applied to the prediction of formulation-specific FE for BCS/BDDCS Class II drug and CYP3A4 substrate nifedipine using as far as possible only in vitro data. Predicted plasma concentration profiles of all three studied formulations under fasted and fed states are within 2-fold of clinically observed profiles. The % prediction error (%PE) in fed-to-fasted ratio of Cmax and AUC were less than 5% for all formulations except for the Cmax of Nifedicron (%PE = −29.6%). This successful case study should help to improve confidence in the use of mechanistic physiologically-based models coupled with in vitro data for the anticipation of FE in advance of in vivo studies. However, it is acknowledged that further studies with drugs/formulations exhibiting a wide range of properties are required to further validate this methodology.

Introduction

Assessment of potential food effects (FE) on the rate and extent of absorption of orally dosed drugs is an important part of drug development particularly for poorly soluble drugs (Yasuji et al., 2012). As such the US FDA provides guidance for the consideration of FE in relation to bioavailability and bioequivalence studies (FDA, 2002). Understanding the impact of food on exposure to, and the efficacy of, a new drug as early as possible may avoid relabeling and safety issues at later stages of development. Classification systems such as the Biopharmaceutical Classification System (BCS) (Amidon et al., 1995, Custodio et al., 2008) and the Biopharmaceutical Drug Disposition Classification System (BDDCS) (Wu and Benet, 2005, Custodio et al., 2008), FeSSIF/FaSSIF solubility ratio- and Quantitative Structure–Activity Relationship (QSAR)-based approaches (Singh, 2005, Gu et al., 2007) are used to anticipate FEs during early development stages mainly because of simplicity of calculation and minimal requirements for input data. Such approaches are neither intended nor able to:

  • I.

    consider the full scope and interplay of postprandial physiological changes;

  • II.

    predict plasma concentration profiles in fasted and fed states;

  • III.

    provide quantitative information about population variability;

  • IV.

    quantitatively predict the changes in AUC, Cmax, and Tmax (with the exception of QSARs for predicting AUC change developed by Singh (2005)).

Furthermore these approaches are based upon characteristics of the drug alone. Hence they are applicable only to Immediate Release (IR) formulations where formulation-related properties are expected to have minimal impact on the kinetics of drug absorption. Thus, in general, these approaches cannot predict the nature and extent of FE for modified/controlled release (MR or CR) formulations which may be formulation-specific (Welling, 1996). In contrast, with appropriate in vitro data, population-based mechanistic models are able to integrate all available physiological and anatomical (or system) data, and drug- and formulation-specific information to predict pharmacokinetics (PK) and pharmacodynamics (PD) not only for an ‘average’ or ‘typical’ human’ but also for a population of individuals (Jamei et al., 2009a). A wide range of food-related system changes can be incorporated as model parameters to attempt to account for the clinically observed phenomena, viz. blood flow, gastric residence time, luminal pH, bile salt concentrations, fluid volumes, etc. (Jamei et al., 2009b, Jamei et al., 2009c).

Mechanistic PBPK models have at least two major advantages over empirical models. First, they can be used in advance of information from the clinic to anticipate drug pharmacokinetics. The models can be modified iteratively as further in vitro or clinical information becomes available and can be used to inform what and when additional data are collected or measured. Second, mechanistic models provide a rational framework within which to propagate the impact of the known inter-individual variability of parameters such as gut lumen pH and bile salt concentrations, fasted–fed status, transit times, enzyme abundances, and tissue volumes; covariate models for physiological parameters can be incorporated where such information is available to build as realistic virtual individuals as possible (Jamei et al., 2009a). Many applications related to age, sex, ethnicity and disease impact have been recently summarised by Rostami-Hodjegan (2012) while a particular example of the application of such models in the area of oral absorption was provided by Darwich et al., 2012, Darwich et al., 2013 who used the models to understand and demonstrate changes to drug bioavailability following bariatric surgery in morbidly obese patients.

Mechanistic PBPK models have also been used for the quantitative prediction of FE on the PK of IR formulations based upon measured biorelevant solubility/dissolution and other in vitro and in vivo disposition parameters (Jones et al., 2006, Turner et al., 2012, Heimbach et al., 2013). However, to our knowledge, there are no reports either on the use of mechanistic models to predict FE for CR formulations or on the prediction of formulation-specific differences in FE for BCS Class II drugs based exclusively upon in vitro data.

Herein the Advanced Dissolution, Absorption and Metabolism (ADAM) (Jamei et al., 2009c) model in conjunction with the 14 organ full Physiologically-Based Pharmacokinetic (PBPK) model of the Simcyp Simulator (Fig. 1) is used to predict FE on one IR (Procardia®, an immediate release soft gelatin capsule) (Reitberg et al., 1987), and two CR formulations ([i] ADALAT OROS®, an osmotically-driven Gastrointestinal Therapeutic System (GITS) and [ii] Nifedicron, encapsulated mini tablets) (Schug et al., 2002c) with different mechanisms of drug release; results are compared with those obtained using other methods. Nifedipine (NIF), a BCS Class II (poor solubility, high permeability) and BDDCS Class II (poor solubility, high metabolism) compound, was selected as a model drug for this case study because it is one of the most extensively studied drugs in the clinic for FE due to significant differences in the nature and extent of FE with different formulations (Challenor et al., 1987, Reitberg et al., 1987, Rimoy et al., 1989, Ueno et al., 1989, Armstrong et al., 1997, Toal et al., 1997, Toal et al., 2012, Schug et al., 2002a, Schug et al., 2002b, Schug et al., 2002c, Toal, 2004, Wonnemann et al., 2006, Wonnemann et al., 2008, Anschutz et al., 2010).

An IR formulation of NIF (Procardia) is reported to have significant reduction in Cmax, increase in Tmax, and negligible increase in AUC when given with food (Reitberg et al., 1987) while CR formulations tend to exhibit the opposite effect (increased Cmax, and increased AUC) (Schug et al., 2002c, Toal et al., 2012). Moreover, there are wide variations between different CR formulations of the rate and extent of absorption in the fasted state and even more so in the fed state (Toal et al., 2012). As a result concerns have been raised about switching between the innovator reference product Adalat OROS and other generic products without patient monitoring (Meredith, 2007, Toal et al., 2012).

The aim of this study is to provide an example of the use of mechanistic PBPK models in combination with in vitro data alone to predict variations in the PK of both the same formulation in the fasted and fed states as well as between different formulations; as far as possible clinical data were not used to parameterise the models rather only to assess model performance.

Section snippets

Software

Simulations were performed using the Simcyp Simulator (Version 12 Release 1, Simcyp Limited, Sheffield, UK). The Simulator platform provides population libraries consisting of system data and their covariates where available for ethnic, healthy or disease population (Jamei et al., 2009a).

Trial design

A clinical trial design is replicated by selecting an appropriate Simcyp population library based upon which a specified number of virtual subjects of the selected age range, gender, etc. are generated. In

Results

The predicted plasma drug concentration (Cp) profiles of NIF IR and CR formulations (OROS and Nifedicron) under fasted and fed conditions match the observed (clinical) values reasonably well (Fig. 3 and Table 3). Note that values of Cmax, Tmax and AUC vary significantly (Table 3) according to the statistical parameter reported (i.e., arithmetic mean, median, geometric mean) and it is important to compare the appropriate parameters when assessing predictions (Table 4). The two CR studies report

Discussion

NIF, a CYP3A4 substrate, is a BCS and BDDCS Class II drug with a FeSSIF v1/FaSSIF ratio of 2.54 (173/68 μM (Clarysse et al., 2009)); based on these data alone IR formulations of NIF are expected to exhibit positive or no FE on the extent of absorption (AUC) and prolongation of Tmax (Custodio et al., 2008). Such prediction approaches may allow qualitative anticipation of FE but cannot generally answer the ‘how much’ question. Likewise all three QSAR models (Eqs. (5), (6), (7)) predict

Conclusion

Mechanistic PBPK models in general are being increasingly applied as cost-effective tools in drug development and their use is being promoted by regulatory bodies (e.g., Zhang et al., 2011, Huang et al., 2013). This study focuses largely upon one component of such models viz. the quantitative prediction of the nature and magnitude of FE upon oral drug absorption during pre-clinical stages of drug development. The study was successful at predicting differential FE upon absorption from oral IR

Acknowledgements

We would like to thank Molecular Discovery Ltd. for providing a pre-release version of the MoKa pKa prediction software. The help of Mr. James Kay in preparing the manuscript is appreciated. The Simcyp Simulator is freely available, following completion of the training workshop, to approved members of academic institutions and other non-for-profit organizations for research and teaching purposes.

References (80)

  • L. Maggi et al.

    Supramicellar solutions of sodium dodecyl sulphate as dissolution media to study the in vitro release characteristics of sustained-release formulations containing an insoluble drug: nifedipine

    Int. J. Pharm.

    (1996)
  • S.Y. Oh et al.

    Diffusion coefficients of single bile salt and bile salt-mixed lipid micelles in aqueous solution measured by quasielastic laser light scattering

    Biochim. Biophys. Acta

    (1977)
  • A. Okazaki et al.

    Theoretical dissolution model of poly-disperse drug particles in biorelevant media

    J. Pharm. Sci.

    (2008)
  • B.S. Schug et al.

    Formulation-dependent food effects demonstrated for nifedipine modified-release preparations marketed in the European Union

    Eur. J. Pharm. Sci.

    (2002)
  • C.B. Toal et al.

    Nifedipine gastrointestinal therapeutic system (GITS) for hypertensive patients in a primary care setting: results of the Extended Release Adalat Canadian Trial (EXACT)

    Clin. Ther.

    (1997)
  • J. Wang et al.

    General solution for diffusion-controlled dissolution of spherical particles. 1. Theory

    J. Pharm. Sci.

    (1999)
  • M. Wonnemann et al.

    Comparison of two marketed nifedipine modified-release formulations: an exploratory clinical food interaction study

    Clin. Ther.

    (2008)
  • ACD Labs, 2013. ACD Labs v12.01. <http://www.acdlabs.com/home/> (accessed...
  • C.H. Ahsan et al.

    The influence of dose and ethnic origins on the pharmacokinetics of nifedipine

    Clin. Pharmacol. Ther.

    (1993)
  • G.L. Amidon et al.

    A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability

    Pharm. Res.

    (1995)
  • M. Anschutz et al.

    Differences in bioavailability between 60 mg of nifedipine osmotic push–pull systems after fasted and fed administration

    Int. J. Clin. Pharmacol. Ther.

    (2010)
  • J. Armstrong et al.

    The influence of two types of meal on the pharmacokinetics of a modified-release formulation of nifedipine (Adalat Retard)

    Eur. J. Clin. Pharmacol.

    (1997)
  • H. Bode et al.

    Investigation of nifedipine absorption in different regions of the human gastrointestinal (GI) tract after simultaneous administration of 13C- and 12C-nifedipine

    Eur. J. Clin. Pharmacol.

    (1996)
  • B. Carr et al.

    Characterization of the rhesus monkey CYP3A64 enzyme: species comparisons of CYP3A substrate specificity and kinetics using baculovirus-expressed recombinant enzymes

    Drug Metab. Dispos.

    (2006)
  • V.F. Challenor et al.

    Food and nifedipine pharmacokinetics

    Br. J. Clin. Pharmacol.

    (1987)
  • ChemAxon. Marvin, Calculator Plugin and Chemical Terms Demo. <http://www.chemaxon.com/marvin/sketch/index.php>...
  • Y.H. Choi et al.

    Pharmacokinetic and pharmacodynamic interaction between nifedipine and metformin in rats: competitive inhibition for metabolism of nifedipine and metformin by each other via CYP isozymes

    Xenobiotica

    (2012)
  • S. Clarysse et al.

    Postprandial changes in solubilizing capacity of human intestinal fluids for BCS class II drugs

    Pharm. Res.

    (2009)
  • R. Clayton et al.

    A Guide for IVIVE and PBPK/PD Modelling using the Simcyp Population-based Simulator. Version 12

    (2012)
  • A.S. Darwich et al.

    A mechanistic pharmacokinetic model to assess modified oral drug bioavailability post bariatric surgery in morbidly obese patients: interplay between CYP3A gut wall metabolism, permeability and dissolution

    J. Pharm. Pharmacol.

    (2012)
  • A.S. Darwich et al.

    Evaluation of an in silico PBPK post-bariatric surgery model through simulating oral drug bioavailability of atorvastatin and cyclosporine

    CPT: Pharmacometr. Syst. Pharmacol.

    (2013)
  • Dressman, J.B., 2012. Linking the lab to the patient: predicting plasma profiles from biorelevant dissolution tests...
  • C. Emoto et al.

    Enzymatic characteristics of CYP3A5 and CYP3A4: a comparison of in vitro kinetic and drug–drug interaction patterns

    Xenobiotica

    (2006)
  • C. Emoto et al.

    Approach to predict the contribution of cytochrome P450 enzymes to drug metabolism in the early drug-discovery stage: the effect of the expression of cytochrome b(5) with recombinant P450 enzymes

    Xenobiotica

    (2007)
  • FDA, 2002. Guidance for Industry: Food-effect Bioavailability and Fed Bioequivalence Studies. December 2002. Center for...
  • M. Gertz et al.

    Prediction of human intestinal first-pass metabolism of 25 CYP3A substrates from in vitro clearance and permeability data

    Drug Metab. Dispos.

    (2010)
  • A. Glomme et al.

    Predicting the intestinal solubility of poorly soluble drugs

  • C.H. Gu et al.

    Predicting effect of food on extent of drug absorption based on physicochemical properties

    Pharm. Res.

    (2007)
  • T. Heimbach et al.

    Case studies for practical food effect assessments across BCS/BDDCS class compounds using in silico, in vitro, and preclinical in vivo data

    AAPS J.

    (2013)
  • N. Holtbecker et al.

    The nifedipine–rifampin interaction. Evidence for induction of gut wall metabolism

    Drug Metab. Dispos.

    (1996)
  • Cited by (40)

    View all citing articles on Scopus
    View full text