clin. pharmaco

Upload: ferdinand-yuzon

Post on 08-Aug-2018

227 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/22/2019 Clin. Pharmaco

    1/13

    Pharmacokinetic-Pharmacodynamic Modelling of theAntihistaminic (H1) Effect of BilastineNerea Jauregizar,1 Leire de la Fuente,1 Maria Luisa Lucero,2 Ander Sologuren,2 Nerea Leal3 and Monica Rodrguez3

    1 PharmaDatum Data Analisis SL, Bermeo, Spain

    2 Clinical Department, Laboratorios FAES FARMA SA, Leioa, Spain

    3 DynaKin, SL Parque Tecnologico de Bizkaia, Derio, Spain

    Abstract Objective: To model the pharmacokinetic and pharmacodynamic relationship of bilastine, a new histamineH

    1

    receptor antagonist, from single- and multiple-dose studies in healthy adult subjects.

    Methods: The pharmacokinetic model was developed from different single-dose and multiple-dose studies.

    In the single-dose studies, a total of 183 subjects received oral doses of bilastine 2.5, 5, 10, 20, 50, 100, 120,

    160, 200 and 220 mg. In the multiple-dose studies, 127 healthy subjects received bilastine 10, 20, 40, 50, 80,

    100, 140 or 200 mg/day as multiple doses during a 4-, 7- or 14-day period.

    The pharmacokinetic profile of bilastine was investigated using a simultaneous analysis of all concentration-

    time data by means of nonlinear mixed-effects modelling population pharmacokinetic software NONMEM

    version 6.1.

    Plasma concentrations were modelled according to a two-compartment open model with first-order

    absorption and elimination.

    For the pharmacodynamic analysis, the inhibitory effect of bilastine (inhibition of histamine-induced

    wheal and flare) was assessed on a preselected time schedule, and the predicted typical pharmacokinetic pro-

    file (based on the pharmacokinetic model previously developed) was used. An indirect response model was

    developed to describe the pharmacodynamic relationships between flare or wheal areas and bilastine plasma

    concentrations.

    Finally, once values of the concentration that produced 50% inhibition (IC50) had been estimated for wheal

    and flare effects, simulations were carried out to predict plasma concentrations for the doses of bilastine 5, 10

    and 20 mg at steady state (7296 hours).

    Results: A non-compartmental analysis resulted in linear kinetics of bilastine in the dose range studied. Bilastine

    was characterized by two-compartmental kinetics with a rapid-absorption phase (first-order absorption rate

    constant = 1.50 h-1), plasma peak concentrations were observed at 1 hour following administration and the

    maximal response was observed at approximately 4 hours or later. Concerning the selected pharmacodynamic

    model to fit the data (type I indirect response model), this selection is attributable to the presence of inhibitory

    bilastine plasma concentrations that decrease the input response function, i.e. the production of the skin reac-

    tion. This model resulted in the best fit of wheal and flare data. The estimates (with relative standard errors

    expressed in percentages in parentheses) of the apparent zero-order rate constant for flare or wheal spontaneousappearance (kin), the first-order rate constant for flare or wheal disappearance (kout) and bilastine IC50 values

    were 0.44 ng/mL/h (14.60%), 1.09 h-1 (15.14%) and 5.15 ng/mL (16.16%), respectively, for wheal inhibition, and

    11.10 ng/mL/h (8.48%), 1.03 h-1 (8.35%) and 1.25 ng/mL (14.56%), respectively, for flare inhibition.

    The simulation results revealed that bilastine plasma concentrations do not remain over the IC50 value

    throughout the inter-dose period for doses of 5 and 10 mg. However, with a dose of 20 mg of bilastine

    administered every 24 hours, plasma concentrations remained over the IC50 value during the considered period

    for the flare effect, and up to 20 hours for the wheal effect.

    Conclusion: Pharmacokinetic and pharmacodynamic relationships of bilastine were reliably described with

    the use of an indirect response pharmacodynamic model; this led to an accurate prediction of the pharmaco-

    dynamic activity of bilastine.

    ORIGINAL RESEARCH ARTICLE Clin Pharmacokinet 2009; 48 (8): 543-50312-5963/09/0008-0543/$49.95 2009 Adis Data Information BV. All rights reserve

  • 8/22/2019 Clin. Pharmaco

    2/13

    Background

    Bilastine (F-96221-BM1, p-[2-[4-[1-(2-ethoxyethyl)-2-benz-

    imidazolyl]piperidino]ethyl]-a-methylhydratropic acid) is anew histamine H1 receptor antagonist (antihistamine) drug

    currently under clinical development by FAES FARMA, SA,

    Spain, for the treatment of allergic rhinoconjuntivitis (seasonal

    and/or perennial) and urticaria.

    The results of in vitro preclinical studies provide evidence

    that bilastine has antihistamine activity with high specificity for

    H1 receptors and poor or no affinity for other receptors.[1]

    The results of the in vivo pharmacological studies conducted

    in animal species corroborate those obtained in the in vitro

    receptor binding experiments carried out previously. In this

    sense, bilastine potently and selectively binds to H1 receptorswith behaviour similar to that of cetirizine and superior to that

    of fexofenadine.[2]

    Understanding of the pharmacokinetics and pharmaco-

    dynamics of this new H1 receptor antagonist provides the ob-

    jective basis for selection of an appropriate dose and dosage

    interval and the rationale for modification of the dosage regi-

    men as needed in special populations, including elderly

    patients, and those with hepatic or renal dysfunction.

    The in vivo test in which inhibition of the histamine-

    induced skin wheal and flare reactions is measured has been

    extensively used to study the antihistaminic activity of several H1receptor antagonists.[3-7] As was recently stated, this test may not

    be a valid surrogate measure for antihistaminic clinical efficacy

    and should not be used to compare different antihistamines.[8]

    However, it establishes the order of magnitude of the clinically

    efficacious dose. Indeed, phase III studies of bilastine have shown

    that the selected clinically efficacious regimen of 20 mg once daily

    was in accordance with the conclusions of phase I studies. [9]

    Little pharmacokinetic and pharmacodynamic modelling has

    been performed for antihistaminic agents. For most drugs, the

    relationship between exposure and effect is nonlinear and plasma

    drug concentrations cannot usually be (directly) related to the

    drug effect.[10] In the case of antihistaminic drugs, both the effectcompartment models for norebastine and levocabastine[7]and the

    indirect response models for mizolastine[11] and cetirizine[12] have

    been applied to account for the lag between peak plasma drug

    concentrations and pharmacodynamic effects.

    In the present study, plasma drug concentration-time pro-

    files of single and multiple oral doses of bilastine given to

    healthy subjects were described using an appropriate popula-

    tion pharmacokinetic model. The pharmacodynamic effect of

    bilastine was modelled by means of a physiological indirect

    response model (using the pharmacokinetic model previously

    developed). The estimated pharmacokinetic and pharmaco

    dynamic parameters of the model are summarized in this article

    Methods

    Subjects and Study Design

    A total of 310 healthy adult subjects (248 males an

    62 females) were included in the study. All participants gav

    signed informed consent, and ethical approval was obtained

    from the Queens University Research Ethics Committe

    (Belfast, Northern Ireland), the Northern Ireland Phase

    Research Ethics Committee from the Office for Researc

    Ethics Committees Northern Ireland (Belfast, Norther

    Ireland) or the Ethical Committee for Clinical Research oSanta Creu i Sant Pau Hospital (Barcelona, Spain).

    Bilastine plasma concentration-time data were availabl

    from 12 studies and antihistamine activity was quantified in

    two of these studies; all of the studieswere part of a phase I tria

    Study design details are summarized in table I. [13-18] All of th

    available data were analysed simultaneously.

    Blood samples from studies 1 and 2 were drawn at th

    following sampling times: predose (0 hour) and at 0.5, 1, 1.5, 2

    2.5, 3, 3.5, 4, 6, 8, 12, 16, 24, 36, 48, 72 and 96 hours post-dose

    Blood samples from studies 4, 5, 7, 8, 9 and 10 were collected a

    the same sampling times but only up to 48 hours post-dose andup to 72 hours post-dose for studies 11 and 12. Studies 3, 6 an

    10 were multiple-dose studies where samples were available o

    day 1 (samples were collected up to 24 hours post-dose) betwee

    96 and 144 hours for study 10, between 168 and 216 hours fo

    study 6 and on day 14 (240288 hours) for study 3.

    The antihistaminic activity of bilastine was evaluated b

    using the effect measurements (wheal and flare) from studie

    4 and 5. The inhibitory effect of bilastine (inhibition of histamine

    induced wheal and flare) was assessed at a preselected tim

    schedule: predose and at 1.5, 4, 8, 12 and 24 hours post-dose.

    Pharmacokinetics

    Blood samples (7 mL) from a suitable antecubital vein wer

    collected into green-top/sodium heparin Vacutainer tube

    predose and at different post-dose timepoints. Plasma sample

    were separated by centrifugation (approximately 2500 rpm 1

    minutes at 41C), split into two aliquots and stored in clearl

    labelled containers in a freezer set at or below -201C unt

    shipment to MDS Pharma Services in Zurich, Switzerland, fo

    analysis.

    544 Jauregizar et a

    2009 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2009; 48 (

  • 8/22/2019 Clin. Pharmaco

    3/13

    Bilastine concentrations were determined in heparinized hu-

    man plasma by a validated liquid chromatography-tandem mass

    spectrometry (LC-MS/MS) method using a solid-phase extrac-

    tion procedure. In brief, using 3-mL disposable glass tubes,

    500mL of each plasma sample was spiked with internal standard(100mL of 500ng/mL of trandolapril in water) and combined

    with 300mL of Millipore water and 100mL of 1% (v/v) aqueous

    trifluoroacetic acid. The sample was vortex-mixed and

    centrifuged for 1 minute at 3000 rpm. Solid-phase extraction

    cartridges (Isolute C8, EC; Biotage AB, Uppsala, Sweden) were

    solvated subsequently with 2 mL of methanol and 2 mL of 0.1%

    (v/v) aqueous trifluoroacetic acid. Samples were applied quanti-

    tatively, washed with 2 mL of 0.1% (v/v) aqueous trifluoroacetic

    acid and eluted with 2 mL of 5 mM ammonium acetate in 90%

    (v/v) aqueous methanol. The eluent was evaporated using a

    vacuum centrifuge (SpeedVac; Savant Instruments, Lif

    Sciences International, Runcorn, UK) and the residue wa

    reconstituted in 200mL of mobile phase (1% [v/v] formic acid i

    50% [v/v] aqueous acetonitrile), vortex-mixed for 1 minute an

    transferred into a polypropylene auto sampler vial.LC-MS/MS analysis was performed using a Perkin-Elme

    LC system combined with a Sciex API 3000 mass spectromete

    equipped with a Turbo Ion Spray source (GenTech Scientific

    Inc., Arcade, NY, USA). A 10 mL-aliquot of the reconstitute

    sample was injected using a reverse phase column (Water

    SymmetryShield RP18, 3.5 mm 2.1 50 mm; Waters Corpo

    ration, Milford, MA, USA) at a temperature of 501C and

    constant mobile phase flow of 0.15 mL/minute. The retentio

    times were 0.7 minutes for bilastine and 0.9 minutes fo

    trandolapril. Mass transitions of 464-272 for bilastine an

    Table I. Summary of phase I clinical studies, evaluating the pharmacokinetics of bilastine (BIL), included in the pharmacokinetic database. The pharmaco

    dynamic database was created with data from studies 4 and 5

    Study

    number

    Description Dosing regimen No. of healthy

    adult subjects

    1 Double-blind, ascending, single-dose study to evaluate the safety, tolerability and

    pharmacokinetics of BIL

    SOD: 5, 10, 50 and

    100mg

    36

    2 Pharmacokinetic study to assess the single-dose bioavailability of BIL under fed and

    fasted conditions

    SOD: 20 mg 12

    3[13] Randomized, multiple-dose study to evaluate the safety and tolerability and pharmacokinetics

    of BIL at escalating doses

    MOD: 10, 20, 50 and

    100mg/day for 14 days

    36

    4[14] Randomized, single-dose, placebo-controlled, four-period crossover study to evaluate the

    safety and tolerability, pharmacokinetics and antihistaminic activity of BIL at five dose levels

    compared with cetirizine

    SOD: 2.5, 5, 10, 20

    and 50 mg

    21

    5[15] Open-label study to assess the effects of age and gender on the pharmacokinetics and

    pharmacodynamics of BIL

    SOD: 20 mg 32

    6 Randomized, double-blind, crossover, placebo- and positive standard-controlled, single-centre

    clinical trial for evaluation of CNS effects of BIL at different doses after single and repeat

    oral administration

    MOD: 20, 40 and

    80mg/day for 7 days

    20

    7[16] Pharmacokinetic and safety study evaluating the potential interaction of erythromycin and BIL

    under steady-state conditions

    SOD: 20 mg 24

    8 Pharmacokinetic and safety study evaluating the potential interaction of ketoconazole and BIL

    under steady-state conditions

    SOD: 20 mg 24

    9[17] Randomized, double-blind, placebo-controlled, sequential group study to evaluate the safety,

    tolerability and pharmacokinetics of single, ascending doses of BIL and of multiple doses of BIL

    SOD: 120, 160, 200

    and 220 mg

    MOD: 140 and

    220mg/day for 7 days

    54

    10 Randomized, multiple-dose, double-blind, five-way crossover study of the ECG effects

    of bilastine

    MOD: 20 and 100 mg/day

    for 4 days

    30

    11 Randomized, open-label, two-way crossover study to evaluate the effect of grapefruit juice on

    the single-dose pharmacokinetics of BIL

    SOD: 20 mg 11

    12[18] Randomized, open-label, two-way crossover study to evaluate the effect of diltiazem on the

    single-dose pharmacokinetics of BIL

    SOD: 20 mg 11

    CNS = central nervous system; ECG =electrocardiographic; MOD=multiple oral doses; SOD= single oral dose.

    PK/PD Modelling of Bilastine 54

    2009 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2009; 48 (

  • 8/22/2019 Clin. Pharmaco

    4/13

    431-234 for trandolapril were selected for quantification in

    multiple-reaction monitoring mode.

    Signal quantification was performed by the peak area ratio.

    The assay was validated over a linear range (0.2200 ng/mL).The lower limit of quantification was 0.2 ng/mL.

    Antihistamine Activity

    Antihistamine activity was evaluated by measuring the sur-

    face areas of skin wheals and flares for 24 hours. In each

    treatment period, histamine skin-prick tests (100 mg/mL his-

    tamine in normal saline) were performed on the back of the

    subject at the following timepoints: predose and at 1.5, 4, 8, 12

    and 24 hours post-dose. At each timepoint, two skin-prick tests

    were performed on matching sites on opposing sides of the

    spine. Ten minutes after each skin-prick test, the wheal and

    flare areas induced by histamine were outlined directly on the

    back with a green marker and a red marker, respectively, and

    transferred onto acetate tracing paper using permanent marker

    pens. Calculation of the wheal and flare areas was performed

    manually. An average area of the two sites for each timepoint

    was calculated and used for analysis. Post-treatment, histamine

    time-response (wheal and flare areas expressed in square cen-

    timetres) curves were constructed.

    Data Analysis

    The first step was to perform a non-compartmental analysis of

    the data in order to evaluate linearity or non-linearity of the

    kinetics. The computational method used to measure areas was

    the trapezoidal rule. The analysis was performed simultaneously

    at the different dose levels. This analysis was carried out with the

    use of WinNonlin software (Pharsight Corporation, Cary, NC,

    USA). The estimated parameters for the different doses were

    compared by analysis of variance (ANOVA) with theuseof SPPS

    software version 14 (SPPS, Inc., Chicago, IL, USA).

    Afterwards, a population approach was used and the strat-

    egy was as follows: the pharmacokinetic model was built firstand, using the typical disposition characteristics of bilastine, a

    pharmacodynamic model describing the drug effect was

    then developed. All analyses were performed with nonlinear

    mixed-effects modelling software NONMEM version 6.1

    (Globomax LLC, Ellicott City, MD, USA).[19]

    For each of the pharmacokinetic or pharmacodynamic

    analyses, a basic population model was proposed. Based on a

    model that describes mean population and individual tenden-

    cies, observations are expressed as follows (equation 1):

    OBSij fyi; D;tj eij Eq: 1

    where OBSij is the jth observation (bilastine plasma concen

    tration or the measured wheal or flare effect) in the ith ind

    vidual, f represents the structural model, yi represents the set o

    parameters (pharmacokinetic or pharmacodynamic) for thith individual, D is the administered dose, tj is the time at whic

    the jth observation was recorded and eij represents the residua

    shift of the observation from model predictions; eij are random

    variables assumed to be symmetrically distributed around 0

    with variance denoted by s2. Although, in the previous expres

    sion, an additive model was used to relate observations t

    predictions, different models (i.e. the constant coefficient o

    variation [CV] slope/intercept) were also explored.

    For each of the elements ofyI, the following model was use

    (equation 2):

    Pi P eZiP Eq: 2

    This equation represents an exponential model to describ

    interindividual variability in the model parameters. Pi denote

    the individual parameter estimated forthe ith individual, P is th

    typical value of the parameter estimated for the population an

    Zi is the interindividual error for the ith individual, i.e. th

    difference between the estimated value of P and the typica

    value of P for the population. The random variable Zi is as

    sumed to be symmetrically distributed around 0 with varianc

    of O. Other models that were compared were the additiv

    model (constant variance) and the proportional model (th

    constant CV).

    Different pharmacokinetic models (in the absence of co

    variates) were fitted to the available data. The selection criteri

    used in the model-building and selection process include

    the minimum value of the objective function provided b

    NONMEM defined as -2 times log of maximum likelihood a

    a measure of goodness of fit, the Akaike Information Criterio

    (the value that is used to compare hierarchical and non

    hierarchical models with the same weighting scheme) and othe

    population-parameter values including standard errors of th

    estimates, the CV and confidence intervals. Residual plots ma

    also be used as diagnostic tools. In this sense, weighted annon-weighted residuals versus predicted concentrations and ver

    sus time were analysed since shaped patterns in these plots ma

    indicate problems with the error model (variance or weighting) o

    with the structural model. Other plots that were examined in

    cluded predicted versus observed concentration plots, as well a

    predicted and observed concentrations versus time.

    Individual concentration-time profiles were obtained b

    post hoc Bayesian estimation using model parameters that ha

    been previously obtained. Bayesian estimates of the individua

    pharmacokinetic parameters were obtained by standar

    546 Jauregizar et a

    2009 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2009; 48 (

  • 8/22/2019 Clin. Pharmaco

    5/13

    methods with the estimated values of the population fixed- and

    random-effect parameters.

    Visual predictive checks (VPCs) were used as a way to visua-

    lize how well the model predicted the profile and the vari-ability, assuming perfect precision (no uncertainty). VPCs were

    developed by simulating 5000 individual concentration-time

    profiles for the selected therapeutic dose (20 mg of bilastine

    given once daily), taking population estimates of fixed effects

    and sampling from random effects distribution to obtain indi-

    vidual profile prediction. Random draws from residual error

    distribution were added to each predicted value.

    Evaluation of the Relationship between Covariates

    and Model Parameters

    Theinfluence of covariates on themodel structural parameters

    was first assessed by plotting individual empirical Bayesian esti-

    mates of parameters versus all of the preselected potential co-

    variates including demographic data (age, bodyweight, height

    and sex), biochemical data (serum albumin, creatinine, bilirrubin,

    g-glutamyltransferase [GGT], aspartate aminotransferase [AST],

    alkaline phosphatase and blood urea nitrogen [BUN] con-

    centrations) and vital signs (pulse).

    Initially, plots of the interindividual variabilities of phar-

    macokinetic parameters versus each analysed covariate, as well

    as weighted residuals versus covariates, were analysed. Then a

    statistical analysis of covariates was carried out and the sta-

    tistically significant covariates were included in the base model

    by stepwise regressions with an inclusion criterion of p 0.05.

    Once the covariates to be included in the model were

    selected, this model with covariates was statistically compared

    with the base model.

    In the final model selection process, all of the described

    issues were considered, as well as the standard error of esti-

    mates, the extent of interindividual variability, and the corre-

    lation matrix of parameters and diagnostic plots (including

    covariance among parameters and distribution histograms).

    Pharmacokinetic Model

    A compartmental analysis showed that a two-compartment

    model with first-order absorption and elimination best de-

    scribed the kinetics of bilastine after oral administration.

    NONMEM subroutine ADVAN4, TRANS4 was used. The

    schematic illustration in figure 1 assumes first-order absorp-

    tion, where ka denotes the first-order absorption rate constant.

    This figure shows schematically the time-course of the drug

    in the central compartment (compartment 1) and peripheral

    compartment (compartment 2) after administration of an ora

    dose. The drug is eliminated from the central compartment

    where CL denotes drug clearance, and on the other hand, it i

    distributed from the central to the peripheral compartmentQ being the inter-compartmental clearance. Hence, V1 and V

    denote the central compartment volume of distribution and th

    peripheral compartment volume of distribution, respectively.

    Model building and final analysis were performed usin

    NONMEM with the first-order conditional estimatio

    (FOCE) method. After completing the model-building proces

    for the totality of plasma concentration-time data from th

    11 studies (single- and multiple-dose studies), population esti

    mates of the fixed effects (y) as well as their relative standar

    errors (ESy), expressed in percentages, were obtained.

    Individual concentration-time profiles were obtained byposhoc Bayesian estimation using the model parameters that ha

    been previously obtained. Bayesian estimates of individua

    pharmacokinetic parameters were obtained by standar

    methods, with estimated values of the population fixed- and

    random-effect parameters.

    Pharmacodynamic Model

    In order to obtain estimates of the pharmacodynami

    properties of bilastine, the entire population pharmacokinetic

    were estimated separately and the empirical parameter est

    mates, conditional on those subjects with pharmacodynami

    kin

    1V1

    2V2

    ka

    kout R

    D

    CL

    Q

    3 Pharmacodynamics

    Pharmacokinetics

    Inhibition

    Fig. 1. Schematic presentation of the pharmacokinetic-pharmacodynam

    model that best fitted the data (i.e. plasma concentration and wheal and flar

    data) on the time-course of the drug in the central compartment (compartmen

    1) and the peripheral compartment (compartment 2) after administration of a

    oral dose (D). The specified indirect response model is a type I model o

    inhibition of response build-up model; observed effects were coded as dat

    from compartment 3. CL=apparent total body clearance of the drug from

    plasma; ka= first-order absorption rate constant; kin= zero-order rate consta

    for productionof response; kout= first-orderrate constant for lossof response

    Q = intercompartmental clearance; R = response; V1= central compartme

    volume of distribution; V2=peripheral compartment volume of distribution.

    PK/PD Modelling of Bilastine 54

    2009 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2009; 48 (

  • 8/22/2019 Clin. Pharmaco

    6/13

    measurements, were used to drive the turnover model in

    $DES. In this sequential pharmacokinetic-pharmacodynamic

    modelling, the corresponding (proportional) sigma for the

    pharmacokinetic parameter was fixed to the pharmacokineticmodel estimate. This approach reduced run times drastically

    when compared with simultaneous fitting, which was very time

    intensive and did not converge.

    A physiological indirect response model with inhibition of

    response production[11] was used to describe bilastine pharma-

    codynamics. A schematic illustration of the indirect response

    model is presented in figure 1.

    In the population pharmacokinetic-pharmacodynamicmodel,

    the following differential equations defining the indirect res-

    ponse model were additionally introduced (equations 3 and 4):

    EFF IC50IC50 A1

    Eq: 3

    dA3

    dt kin EFF kout A3 Eq: 4

    where EFF corresponds to the effect.

    The specified indirect response model is a type I model or

    inhibition of response build-up model, where k in is the zero-

    order constant for response production, while kout represents

    the first-order constant for loss of response. IC50 can be

    defined as the drug concentration that achieves 50% of

    maximum inhibition. Note that in the NONMEM data file,

    the observed wheal or flare effects were coded as data from

    compartment 3 [A(3)].

    Initial values for the pharmacodynamic parameters of k in,

    kout, IC50 and maximum inhibition (Imax) were estimated as

    follows: kout was set to log2 divided by the effect half-life,

    roughly estimated from the data; kin was calculated taking into

    account that the baseline value of the response (the observed

    baseline value at time 0) equals k in/kout; the IC50 was set to the

    plasma concentration corresponding to the time of maximum

    effect; and Imax was set to 1.

    The FOCE method was applied in the pharmacokinetic-

    pharmacodynamic modelling process.

    Simulations

    Finally, computer simulations were carried out with the aim

    of selecting the optimal dose and dosing regimen of bilastine.

    The selected times to perform the simulations were between 72

    and 96 hours, i.e. those corresponding to day 4 in a multiple-

    dose regimen. This time selection was based on the value of the

    elimination half-life obtained previously, and simulated con-

    centrations were at steady state. Mean plasma concentration-

    time profiles of bilastine were simulated for daily doses of

    5, 10 and 20 mg. IC50 values, estimated separately for whea

    and flare, were used as thresholds for dose selection via simu

    lation. Proposed dosing regimens should maintain the con

    centration levels within the thresholds during the entire regimeinterval.

    Results

    Pharmacokinetic Study

    The non-compartmental pharmacokinetic analysis reveale

    that bilastine presents linear pharmacokinetics in the studie

    dose range (2.5220 mg/day). Both the maximum concentra

    tion and the area under the plasma concentration-time curv

    (AUC) of bilastine increased proportionally to the administered doses. The terminal elimination half-life as well as th

    other pharmacokinetic parameters of bilastine remained con

    stant over the entire studied dosing range. The ANOVA wit

    Scheffe post hoc confirmed that the pharmacokinetics of bi

    lastine are linear in the dose range of 2.5220 mg/day.

    After completing the model-building process for the totalit

    of the plasma concentration-time data from the different stud

    ies (single- and multiple-dose studies), population estimates o

    the fixed effects (y) as well as their ESy values, expressed i

    percentages, were obtained and are presented in table II. Thes

    standard errors denote the precision in parameter estimation

    concluding with an acceptable precision in estimation (

  • 8/22/2019 Clin. Pharmaco

    7/13

    shown) and high residual values that increased with predicted

    concentrations were observed. This situation led to a cone-

    shaped plot, suggesting the use of a weighting factor. The

    addition of a weight factor helped to overcome this effect. In

    fact, as the cone shape disappeared, residual values declined

    considerably, and residual distribution became random around

    the 0 line. In conclusion, this variance model is suitable. Addi-

    tionally, the time-course profiles of non-weighted and weighted

    residuals were studied (data not shown). The adequacy of the

    weighting scheme was again demonstrated. Residual values

    were acceptable, as well as random distribution of residual

    around the 0 line throughout the analysed time range.

    The VPC for the 20 mg dose is shown in figure 4.

    Pharmacodynamic Study

    The results of the population pharmacokinetic-pharmaco

    dynamic modelling for wheal and flare effects are summarized

    in table III. These ESy values are related to estimation preci

    sion, which were quite acceptable (14.60%, 15.14% and 16.16%

    for kin, kout and IC50, respectively).

    Model parameter Z values, expressed as CV%, are als

    presented in this table. It can be observed that the IC50 wa

    associated with a higher interindividual variability than th

    other parameters.Intra-individual variability (e) had a value of 0.011

    (ESy= 6.28%). As a result of the error model that was selecte

    (additive error), this e is defined as a standard deviation (in

    contrast to the CV% in the pharmacokinetic model) and ac

    counts for model mis-specification or measurement erro

    Hence, the obtained value is acceptable, as it is of the sam

    order as the available wheal effects.

    The model-predicted individual effect on histamine-induce

    wheal as a function of time was in close proximity to th

    time-course profile of the observed effects (figure 5). A simila

    congruence was observed for flare inhibition (figure 6).

    Pharmacodynamic parameter final estimates and their ES

    values for flare are also presented in table III. kin values wer

    different for flare and wheal; this can be explained by the fact tha

    Table II. Population pharmacokinetic-model fit to plasma concentration-

    time data from all available studies. Population pharmacokinetic-parameter

    estimates (with relative standard errorsa [ESy] expressed as %) and inter-

    individual variability expressed as the percentage of the coefficient of varia-tion (CV [%])

    Parameter Estimate y (ESy) CV% (ESy)

    CL (L/h) 18.1 (1.8) 29.0 (8.7)

    V1 (L) 59.2 (2.2) 35.4 (9.6)

    Q (L/h) 1.59 (3.9) 56.5 (10.3)

    V2 (L) 30.2 (5.1) 73.1 (9.6)

    ka (h-1) 1.50 (3.2) 35.4 (16.9)

    s (%) [ESy] 28.6 (6.1)

    a The relative standard error is the standard error divided by the parameter

    estimate.

    CL=

    apparent total body clearance of the drug from plasma; ka=

    first-orderabsorption rate constant; Q=apparent intercompartmental clearance;

    V1= central compartment volume of distribution; V2=peripheralcompartment

    volume of distribution.

    0

    1000

    2000

    3000

    4000

    5000

    6000

    7000

    8000

    0 2000 4000 6000 8000

    Observed concentration (ng/mL)

    Population-predictedco

    ncentration(ng/mL)

    Fig. 2. Population-predicted concentrations vs observed concentrations with the line of identity.

    PK/PD Modelling of Bilastine 54

    2009 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2009; 48 (

  • 8/22/2019 Clin. Pharmaco

    8/13

    flare areas were larger than wheal areas. The kout value was ap-

    proximately 1 h-1 for both wheal and flare, and the IC50 values

    (5.15 ng/mL for wheal and 1.25 ng/mL for flare) were lower for

    flare than for wheal by a ratio of approximately 4 and were low

    compared with the bilastine concentrations obtained after oral

    administration.

    Finally, the results of simulations of plasma concentrations

    corresponding to doses of 5, 10 and 20 mg and for a preselected

    time schedule that ranged from 72 to 96 hours were analysed

    (figure 7). The time selection was carried out under th

    assumption of being at steady state. Taking into account tha

    the estimated IC50 values for wheal and flare effects wer

    5.15 ng/mL and 1.25ng/mL, respectively, bilastine plasm

    concentrations reached after a dose of 20 mg remained over th

    IC50 value for the flare effect throughout the entire inter-dos

    period (24 hours). Bilastine plasma concentrations decrease

    to less than the mean estimated IC50 value for the whea

    effect between 20 and 24 hours only. This means that th

    0

    1000

    2000

    3000

    4000

    5000

    6000

    7000

    8000

    0 1000 2000 3000 4000 5000 6000 7000 8000

    Observed concentration (ng/mL)

    Individual-predictedconcentration(ng/mL)

    Fig. 3. Individual-predicted concentrations vs observed concentrations with the line of identity.

    0

    100

    200

    300

    400

    500

    600

    700

    800

    900

    1000

    0 2 4 6 8 10 12 14 16 18 20 22 24

    Time (h)

    Bilastineplasm

    aconcentration(ng/mL)

    Observed concentrationMedian predicted interquartile

    95% predicted interval

    Fig. 4. Predicted and observedconcentration-time profiles of bilastine after a single oral doseof 20 mg. The simulations usedfixed effects point estimates an

    were performed across, between and within subject variability.

    550 Jauregizar et a

    2009 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2009; 48 (

  • 8/22/2019 Clin. Pharmaco

    9/13

    main part of the antihistaminic activity lasts nearly for the

    entire duration between two once daily dosing intervals.

    However, for doses of 5 and 10 mg, plasma drug concentra-

    tions were below the wheal IC50 value for a few hours before

    administration of the next dose (10 hours for a 10 mg dose

    and 16 hours for a 5 mg dose). Hence it can be concluded

    that the optimal dosing regimen for bilastine is 20 mg every

    24 hours.

    Discussion

    The purpose of this study was to characterize the relation-

    ship between the pharmacokinetics and the antihistaminic ac-

    tivity of bilastine in healthy adult subjects. This is importantnot

    only to establish adequate dosage regimens,[20] but also

    to understand the actual mechanism of action involved in the

    in-vivo drug effect.[21]

    In this study, the pharmacokinetics and pharmacodynamics

    of bilastine after oral administration were studied using thepopulation pharmacokinetic-pharmacodynamic approach. The

    non-compartmental pharmacokinetic analysis revealed that

    bilastine displays linear pharmacokinetics in the studied dose

    range.

    Bilastine was characterized by two-compartmental kinetics

    with a rapid-absorption phase (ka= 1.50 h-1); peak plasma

    concentrations were observed at 1 hour following oral admin-

    istration and the elimination half-life was approximately

    14 hours. Bilastine and most orally administered new H1receptor antagonists are well absorbed and appear to be

    extensively distributed into body tissues; many are highly pro

    tein bound. Most new H1 receptor antagonists do not accu

    mulate in tissues during repeated administration.[3] The sam

    result was observed for bilastine: according to the ratio of thAUC obtained from multiple-dose studies (i.e. the accumula

    tion ratio between the AUC from 312 to 336 hours and th

    AUC from 0 to 24 hours = 1) [results not shown], it can b

    inferred that bilastine does not undergo accumulation after th

    studied dosing regimens.

    The effect of bilastine on raw wheal and flare areas, ex

    pressed as square centimetres, was modelled. Measurement o

    wheal and flare skin reactions to assess the pharmacodynamic

    of antihistaminic drugs has been extensively used. [4,10,11,22-24]

    Little pharmacokinetic and pharmacodynamic modellin

    has been performed for antihistaminic agents. The mechanismof action of bilastine corresponds to an inhibition of build-up

    turnover pharmacodynamic model,[25,26] and this model wa

    successfully applied in this study. In fact, indirect respons

    models have been successfully used to describe the pharmaco

    dynamics of antihistaminic drugs such as mizolastine[10] an

    cetirizine.[12]

    In this study, the peak plasma concentration was rapidl

    reached in approximately 1 hour and the maximal respons

    was observed later at approximately 4 hours or longer. Here

    the measurement site is the skin; therefore, the observe

    delay could be attributed to diffusion to the skin of the anti

    histamine. The indirect-effect model intrinsically addresses thi

    delay.[20]

    The pharmacokinetic-pharmacodynamic model that wa

    developed showed good performance in describing both th

    mean population tendency and individual effect profiles. Th

    0.1

    0

    0.1

    0.2

    0.3

    0.4

    0.5

    0.6

    0.7

    0 10 20 30

    Time (h)

    Predictedandobservedwhealareas(cm

    2)

    Observed wheal effect

    Predicted wheal effect

    Fig. 5. Predicted and observed wheal effects vs time for individuals wh

    participated in study 5.

    Table III. Pharmacokinetic-pharmacodynamic population model fit of the

    wheal and flare effects

    Parameter kin

    [ng/mL/h]

    kout

    [h-1]

    IC50

    [ng/mL]

    Wheal

    Estimatea 0.44 (14.60) 1.09 (15.14) 5.15 (16.16)

    Zb 29.36 (32.95) 14.04 (81.22) 55.95 (45.05)

    Flare

    Estimatea 11.10 (8.48) 1.03 (8.35) 1.25 (14.56)

    Zb 24.02 (45.41) 26.98 (26.65) 65.65 (29.93)

    a Values are expressed as estimate (%ESy). The ESy is the standard error

    divided by the parameter estimate.

    b Values are expressed as %CV (%ES).

    g = interindividual variability;ES = standard error; ESh= relative standarderror

    of the pharmacodynamic parameter; IC50= estimated concentration produ-cing 50% inhibition; kin= zero-order rate constant for production of response;

    kout= first-order rate constant for loss of response.

    PK/PD Modelling of Bilastine 55

    2009 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2009; 48 (

  • 8/22/2019 Clin. Pharmaco

    10/13

    pharmacodynamic parameters had reasonable estimates. Dif-

    ferences in kin values for flare and wheal, as well as lower IC50values for flare than for wheal, have also been observed with

    mizolastine,[11] cetirizine[12] and other antihistaminic drugs in

    an analysis in which an effect compartment model was used. [7]

    When using typical pharmacokinetic parameters, the inter-

    individual variability associated with pharmacodynamic

    parameters is likely to be overestimated, since variability of

    the absorption and disposition processes is also involved.

    However, the estimates of variability were not extremely

    high: 29.36% for kin, 14.04% for kout and 55.95% for IC50 forthe

    wheal effect; and 24.02%, 26.98% and 65.65%, respectively,

    for the flare effect.

    The NONMEM run, by itself, estimated population

    mean values and variances of parameter values within such a

    population, as well as residual error variance. Populatio

    means were then used as a priori estimates for individual pos

    hoc parameters by the Bayesian analysis in every iteration o

    minimizing the objective function. The population mixedeffects approach with Bayesian analysis is a powerful tool fo

    accounting for interindividual variability and, hence, allowin

    for the description of the time-course of the antihistamini

    effect for each individual. These results demonstrate tha

    pharmacokinetic-pharmacodynamic analysis, as stated b

    Levy,[20] can reflect the actual mechanism of action involved in

    the pharmacological response of a given drug.

    Finally, with the use of the population-pharmacokineti

    model, computer simulations were performed to obtain bilas

    tine plasma concentrations at steady state after the adminis

    tration of 5, 10 or 20 mg once daily. Bilastine concentrationreached after a dose of 20 mg are over the IC50 value for th

    flare effect throughout the entire inter-dose period (24 hours)

    Such concentrations decreased to less than the mean estimate

    IC50 value for wheal between 20 and 24 hours only. This implie

    that the main part of the antihistaminic activity lasts nearly fo

    the entire duration between two once-daily dosing intervals

    which suggests that the optimal dosing regimen for bilastine i

    20 mg every 24 hours. Pharmacokinetic and pharmacodynami

    relationships of bilastine were reliably described with the use o

    a physiological indirect response model; this led to an accurat

    prediction of bilastine pharmacodynamic activity. Further

    more, although this model has no predictive value for de

    termining the clinical superiority of one antihistaminic dru

    over another, it provides relevant information regarding which

    drug dosages, based on the drug concentration-response i

    terms of the rate and duration of inhibition, could be anti

    cipated to be below the desired pharmacodynamic response an

    1

    10

    100

    1000

    0 12 24 36 48 60 72 84 96

    Time (h)

    Simulatedbilastine

    plasma

    concentration(ng

    /mL)

    IC50 of wheal effect

    IC50 of flare effect

    Fig. 7. Simulation of population mean bilastine plasma concentrations corresponding to four consecutive 20 mg doses and, thus, being at steady state. Th

    estimated concentration producing 50% inhibition (IC50) values for wheal and flare effects (5.15 ng/mL and 1.25 ng/mL, respectively) are also shown. Plasm

    concentrations of bilastine after a dose of 20 mg are over the IC50 value for the flare effect throughout the inter-dose period (24 hours).

    10

    0

    10

    20

    30

    0 10 20 30

    Time (h)

    Predictedandobservedflareareas(cm

    2)

    Observed flare effect

    Predicted flare effect

    Fig. 6. Predicted and observed flare effects vs time for individuals who

    participated in study 5.

    552 Jauregizar et a

    2009 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2009; 48 (

  • 8/22/2019 Clin. Pharmaco

    11/13

    which dosage should be the potential candidate for further

    development in clinical trials. This should be determined on the

    basis of more clinically relevant endpoints.[26]

    Conclusion

    Bilastine displayed linear pharmacokinetics in the studied

    dose range (2.5220 mg/day). The population-pharmacokinetic

    model that resulted in the best fit of plasma concentration-time

    data was a two-compartmental model with first-order absorp-

    tion and elimination. No significant relationship could be

    detected between pharmacokinetic parameters and any of the

    covariates that were analysed (age, bodyweight, height, sex,

    serum albumin, creatinine, bilirubin, GGT, AST, BUN and

    alkaline phosphatase concentrations, as well as pulse). Ingeneral, no covariate had a substantial role in explaining the

    variability of the model parameters.

    The pharmacokinetic-pharmacodynamic model that yielded

    the best fit of time-effect data was a type I indirect response

    model, also known as a model for inhibition of response pro-

    duction. In fact, the selected population model fits success-

    fully both wheal-and flare-effect data. The estimated IC50values were 5.15 ng/mL for the wheal effect and 1.25 ng/mL

    for the flare effect. Therefore, with 20 mg of bilastine given

    every 24 hours, plasma drug concentrations at steady state

    would be maintained over the IC50 value (to inhibit thewheal and flare effects) throughout almost the entire dosing

    interval.

    Acknowledgements

    The authors would like to thank Deirdre McLaverty and A.J. Stewart

    (MDS Pharma Services Belfast, Northern Ireland), and M.J. Barbanoj,

    R.M. Antonijoan and C. Garca-Gea (Centre dInvestigacio de Medica-

    ments, Institut de Recerca de lHospital de la Santa Creu i Sant Pau,

    Barcelona, Spain) as the principalinvestigators of the different studies. The

    authors received no funding forthe conduct of this study.Nerea Jauregizar

    and Leire de la Fuente are employees of Pharma Datum Data Ana lisis SL;

    Nerea Leal and Mo nica Rodrguez are employees of DynaKin; andM. Luisa Lucero and Ander Sologuren are employees of FAES FARMA

    SA The authors have no other conflicts of interest that are directly relevant

    to the content of this study.

    References1. Corco stegui R, Labeaga L, Innera rity A, et al. Preclinical pharmaco-

    logy of bilastine, a new selective histamine H1 receptor antagonist:

    receptor selectivity and in vitro antihistaminic activity. Drugs 2005; 6:

    371-87

    2. Corco stegui R, Labeaga L, Innera rity A, et al. In vivo pharmacological

    characterisation of bilastine, a potent and selective histamine H1 receptor

    antagonist. Drugs 2006; 7: 219-31

    3. Simons FER, Simons KJ. Clinical pharmacology ofnew histamine H1 recepto

    antagonists. Clin Pharmacokinet 1999; 36: 329-52

    4. Rosenzweig P, Caplain H, Chaufour S, et al. Comparative wheal and flar

    study of mizolastine vs terfenadine, cetirizine, loratadine and placebo i

    healthy volunteers. Br J Clin Pharmacol 1995; 40: 459-65

    5. Harvey RP, Schocket AL. The effect of H1 and H2 blockade on cut

    neous histamine response in man. J Allergy Clin Inmunol 1980; 6

    136-9

    6. Simons FER, Mc Millan JL, Simons KJ. A double-blind, single dose, cros

    over comparison of cetirizine, terfenadine, loratadine, astemizole, and chlor

    pheniramine versus placebo: suppressive effects on histamine-induced whea

    and flares during 24 hours in healthy subjects. J Allergy Clin Inmunol 1990

    86: 540-7

    7. Heykants JJP, Snoeck E, Awouters F, et al. Antihistamines. In: Van Boxtel C

    Holford NHG, Danhof M, editors. The in vivo study of drug actio

    Amsterdam: Elsevier Science Publishers, 1992: 337-56

    8. Devillier P, Bousquet J. Inhibition of the histamine-induced weal and flar

    response: a valid surrogate measure for antihistamine clinical efficacy? Cli

    Exp Allergy 2007; 37: 400-14

    9. Kuna P, Nowacki Z, van Cauwenberge P, et al. A phase study com

    paring the efficacy and safety of once daily bilastine with cetirizine an

    placebo for the treatment of seasonal allergic rhinitis. Allergy 2007; 6

    132-3

    10. Holford NHG, Sheiner LB. Understanding the concentration-effect relation

    ship: clinical application of pharmacokinetic-pharmacodynamic model

    Clin Pharmacokinet 1981; 6: 429-53

    11. Deschamps C, Dubruc C, Mentre F, et al. Pharmacokinetic and pharmaco

    dynamic modeling of mizolastine in healthy volunteers with an indire

    response model. Clin Pharmacol Ther 2000; 68: 647-57

    12. Urien S, Tillement JP, Ganem B. A pharmacokinetic-pharmacodynam

    modelling of the antihistaminic (H1) effects of cetirizine. Int J Clin Pharmaco

    Ther 1999; 37: 499-502

    13. Roupe K, Sologuren A, Crean C, et al. The pharmacokinetics of bilastine aftsingle and 14 days once daily administration [abstract]. Basic Clin Pharmaco

    Toxicol 2007; 101 Suppl. 1: 148

    14. Sologuren A, Valiente R, Crean C, et al. Relationship of dose to inhibitio

    ofwhealandflarefor 5 dosesof bilastineand10 mgcetirizine[abstract no. 69

    J Clin Pharmacol 2007; 47 (9): 1198

    15. Roupe K, Sologuren A, Crean C, et al. Effect of age and gender on the pha

    macokinetics and pharmacodynamics of bilastine [abstract no. 70]. J Cli

    Pharmacol 2007; 47 (9): 1198

    16. Antonijoan RM, Garca-Gea C, Puntes M, et al. Estudio farmacocine tic

    y de actividad antihistamnica H1 de bilastina tras su administracio n unica

    repetida en voluntarios sanos. XIX Congress of the Spanish Society o

    Clinical Pharmacology; 2004 Oct 28-30; Santander

    17. Sologuren A, Crean C, Valiente R, et al. The drug-drug interaction o

    ketoconazole on bilastine pharmacokinetics [abstract plus poster no. 476

    Basic Clin Pharmacol Toxicol 2007; 101 Suppl. 1: 148

    18. Crean C, Valiente R, Sologuren A, et al. Effect of grapefruit juice on th

    pharmacokinetics of bilastine [abstract no. 71]. J Clin Pharmacol 2007; 47 (9

    1198

    19. Beal SL, Boeckmann AJ, Sheiner LB, editors. NONMEM user guides. Sa

    Francisco (CA): NONMEM Project Group, University of California, 1992

    20. Levy G. Mechanism-based pharmacodynamic modelling. Clin Pharmac

    Ther 1994; 56: 356-8

    21. Purohit A, Melac M, Pauli G, et al. Comparative activity of cetirizine an

    mizolastine on histamine-induced skin wheal and flare responses at 24 h. Br

    Clin Pharmacol 2002; 53: 250-4

    22. Bousquet J, Chanal I, Murrieta M, et al. Lack of sensitivity to mizolastine ove

    8-week treatment. Allergy 1996; 51: 251-6

    PK/PD Modelling of Bilastine 55

    2009 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2009; 48 (

  • 8/22/2019 Clin. Pharmaco

    12/13

    23. Pinquier JL, Caplain H, Cabanis MJ, et al. Inhibition of histamine-induced

    skin wheal and flare after 5 days of mizolastine. J Clin Pharmacol 1996; 36:

    72-8

    24. Devalia JL, De Vos C, Hanotte F, et al. A randomized, double-blind cross-

    over comparison among cetirizine, levocetirizine, and UCB 28557 on

    histamine-induced cutaneous responses in healthy adult volunteers. Allergy

    2001; 56: 50-7

    25. Dayneka NL, Garg V, Jusko WJ. Comparison of four basic models of indirect

    pharmacodynamic responses. J Pharmacokinet Biopharm 1993; 21: 457-78

    26. Sharma A, Jusko WJ. Characteristics of indirect pharmacodynamic mode

    and applications to clinical drug responses. Br J Clinical Pharmacol 1998; 45

    229-39

    Correspondence: Ms M. Luisa Lucero, FAES FARMA SA, Maxim

    Aguirre 14, 48940, Leioa (Vizcaya), Spain.

    E-mail: [email protected]

    554 Jauregizar et a

    2009 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2009; 48 (

  • 8/22/2019 Clin. Pharmaco

    13/13