Physiologically based pharmacokinetic modeling for assessing the clinical drug–drug interaction of alisporivir

https://doi.org/10.1016/j.ejps.2014.06.021Get rights and content

Abstract

Alisporivir is a novel cyclophilin-binding molecule with potent anti-hepatitis C virus (HCV) activity. In vitro data from human liver microsomes suggest that alisporivir is a substrate and a time-dependent inhibitor (TDI) of CYP3A4. The aim of the current work was to develop a novel physiologically based pharmacokinetic (PBPK) model to quantitatively assess the magnitude of CYP3A4 mediated drug–drug interactions with alisporivir as the substrate or victim drug. Towards that, a Simcyp PBPK model was developed by integrating in vitro data with in vivo clinical findings to characterize the clinical pharmacokinetics of alisporivir and further assess the magnitude of drug–drug interactions. Incorporated with absorption, distribution, elimination, and TDI data, the model accurately predicted AUC, Cmax, and tmax values after single or multiple doses of alisporivir with a prediction deviation within ±32%. The model predicted an alisporivir AUC increase by 9.4-fold and a decrease by 86% when alisporivir was co-administrated with ketoconazole (CYP3A4 inhibitor) or rifampin (CYP3A4 inducer), respectively. Predictions were within ±20% of the observed changes. In conclusion, the PBPK model successfully predicted the alisporivir PK and the magnitude of drug–drug interactions.

Introduction

Allometry has long been used in predicting human pharmacokinetics (PK) utilizing data from preclinical species (Mahmood, 1999) However, due to notable interspecies differences, especially with metabolizing enzymes and drug transporters, alternative approaches have been proposed over the years to obtain improved predictions. Unlike allometry and other empirical pharmacokinetic models, physiologically based pharmacokinetic (PBPK) models provide mechanistic time-based profiles by integrating drug-dependent and physiology-dependent parameters, in the process of predicting human pharmacokinetics. In addition to allowing human PK predictions at the discovery stage, PBPK modeling is also useful in early and late development to predict drug exposure for various purposes including drug–drug interactions (DDIs), PK in organ dysfunction, different age and genotype populations (Sinha et al., 2012). The US FDA recommends the use of PBPK models to quantitatively predict the magnitude of DDIs in various clinical situations; furthermore, this approach may offer useful information to facilitate dedicated clinical study designs (US Food and Drug Administration, 2012).

Pharmacokinetic DDIs, an important issue in healthcare, mostly result from the modulations of the drug-metabolizing enzymes, particularly P450 enzymes, which are present in the liver and extra-hepatic tissues. CYP3A4 is the most abundantly expressed P450 enzyme in the liver and gut, and it is involved in the metabolism of more than half of the drugs used clinically (Wienkers and Heath, 2005). A number of important drugs have been identified as substrates, inducers, and/or inhibitors of CYP3A4, and the assessment of the potential for CYP3A4-mediated DDIs is an important part of the clinical development program for any new chemical entity. The Simcyp™ population based ADME simulator, which is commercially available software for PBPK modeling, integrates inter-individual variability into PBPK modeling for the prediction of drug disposition and DDIs in virtual populations. By combining information on physiology, genetic and demography/ethnicity with in vitro data, Simcyp™ simulator can perform extrapolation to in vivo situations and virtual populations (Jamei et al., 2009).

Alisporivir (alisporivir), a hepatitis C antiviral agent, is currently in phase III clinical development. Alisporivir, a structural analog of cyclosporine, has physicochemical properties similar to those of cyclosporine. In vitro data described in this manuscript suggested that alisporivir is a CYP3A4 substrate and a time-dependent inhibitor (TDI) of CYP3A4. Hence, there is a potential for clinically significant DDI when alisporivir is co-administered with inhibitors or inducers of CYP3A4 or other drugs that are substrates of CYP3A4. The objective of the present study is to develop a PBPK model to characterize PK of alisporivir following single and multiple oral administration and assess the DDI effects of a CYP3A4 inhibitor (ketoconazole) and a CYP3A4 inducer (rifampin) on its exposure. Specifically, the metabolic and DDI properties for alisporivir, observed in in vitro results, were incorporated in the model for quantitative prediction of mechanism-based drug disposition and interactions.

Section snippets

Chemicals and reagents

Isotope labelled [14C]alisporivir, [2H9]-1′-hydroxybufuralol, and [13C6]-4′-hydroxydiclofenac were synthesized by the DMPK Isotope Laboratory (Novartis, East Hanover, NJ) with chemical and radiochemical purity was greater than 99%. [2H4]-Acetaminophen and [2H4]-1′-hydroxymidazolam were obtained from Cerilliant (Round Rock, TX). Pooled human liver microsomes, S9, and cytosol from the same donor pool (n = 150 donors, mixed gender), recombinant human CYP enzymes, flavin monooxygenase enzymes,

In vitro metabolism and permeability assessment results

The transport of [14C] labelled alisporivir across Caco-2 cells were performed using two concentrations (0.40, 3.5, and 10 μM) in the apical-to-basolateral (Ap  Bl) direction and basolateral-to-apical (Bl  Ap) directions. The apparent Ap  Bl permeability (Papp) and efflux permeability ratios (i.e., Papp,Bl→Ap/Papp,Ap→Bl) appeared to be concentration-dependent. The Papp values of alisporivir were 0.25 × 10−6, 0.96 × 10−6 and 8.46 × 10−6 cm s−1 at 0.40 μM, 3.5 μM, and 10 μM, respectively, while the efflux

Discussion

This report demonstrated the application of PBPK modeling and simulation for predicting the PK outcomes and assessing the DDI risk for alisporivir, a novel hepatitis C antiviral compound. The metabolic fate and DDI liability of alisporivir were investigated by in vitro ADME assays and in vivo PK studies. Alisporivir is a sensitive substrate of CYP3A and is susceptible to clinically relevant DDI’s if co-administered with CYP3A inhibitors and inducers. On the other hand, alisporivir is also a

Conclusions

In conclusion, the application of PBPK model plays a central role in describing the nonlinear PK behavior caused by auto-inactivation of P450 as well as investigation of the magnitude of TDI effects on other compounds. In the recent published draft guidance of drug-interaction studies, the regulatory agency encouraged the sponsors to utilize the PBPK model for better DDI study design and quantitative prediction of the magnitude of DDI (US Food and Drug Administration, 2012). Alisporivir, as a

References (29)

  • S.S. De Buck et al.

    Prediction of human pharmacokinetics using physiologically based modeling: a retrospective analysis of 26 clinically tested drugs

    Drug Metab. Dispos.

    (2007)
  • U. Fagerholm et al.

    Comparison between permeability coefficients in rat and human jejunum

    Pharmaceut. Res.

    (1996)
  • A.R. Hilgers et al.

    Predicting oral absorption of drugs: a case study with a novel class of antimicrobial agents

    Pharmaceut. Res.

    (2003)
  • Y.C. Huang et al.

    Pharmacokinetics and dose proportionality of ketoconazole in normal volunteers

    Antimicrob. Agents Chemother.

    (1986)
  • Cited by (11)

    • Physiologically based pharmacokinetic modeling of altered tizanidine systemic exposure by CYP1A2 modulation: Impact of drug-drug interactions and cigarette consumption

      2021, Drug Metabolism and Pharmacokinetics
      Citation Excerpt :

      Physiologically based pharmacokinetic (PBPK) modeling is widely used in drug development to predict the magnitude of anticipated DDIs depending on the enzyme-specificity for the substrate metabolism. This approach facilitates the design of clinical DDI investigations and provides the relevant information on drug-drug interactions in product labeling [15–20]. The US FDA and the European Medical Agency (EMA) continues to show interest in PBPK models to quantitatively predict the magnitude of DDIs in various clinical situations as part of model informed drug development with the possibility of study waivers under certain conditions [21,22].

    • Gold nanoparticles based sensor for in vitro analysis of drug-drug interactions using imipramine and isoniazid drugs: A proof of concept approach

      2017, Sensors and Actuators, B: Chemical
      Citation Excerpt :

      However, certain DDIs that comprises multiple enzymes based in vitro models, mechanisms, transporters could not be evaluated by employing modeling and simulation techniques solely without elucidation of underlying mechanisms [3,13–16]. Physiological based pharmacokinetic (PBPK) modeling [15,17] has also been introduced as a recent tool to evaluate potential DDIs that employs in vitro drug data simulation through a mathematical description to pharmacokinetics of a drug [18]. In this regards, much attention has been paid for the exploration of DDIs involving pharmacokinetics and pharmacodynamics routes [19–21] minor consideration has been given to ones involving pharmaceutical processes [9,22].

    View all citing articles on Scopus
    View full text