articulo hawwa

Upload: ossyneidee-gutierrez

Post on 13-Apr-2018

227 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/27/2019 Articulo Hawwa

    1/12

    Population pharmacokineticand pharmacogenetic

    analysis of 6-mercaptopurinein paediatric patients withacute lymphoblasticleukaemiaAhmed F. Hawwa, Paul S. Collier, Jeff S. Millership,

    Anthony McCarthy,1 Sid Dempsey,1 Carole Cairns1 &

    James C. McElnay

    Clinical and Practice Research Group, School of Pharmacy, Medical Biology Centre, Queens University

    Belfast and1Haematology and Oncology Outpatient Department, Royal Belfast Hospital for Sick

    Children,The Royal Hospitals, Belfast Health and Social Care Trust,Belfast, UK

    CorrespondenceProfessor Paul S. Collier,Clinical and

    Practice Research Group, School of

    Pharmacy, Medical Biology Centre,

    Queens University Belfast, 97 Lisburn

    Road,Belfast BT9 7BL, UK.

    Tel:+44 28 9097 2009

    Fax: +44 28 9024 7794E-mail: [email protected]

    ----------------------------------------------------------------------

    Keywords6-mercaptopurine, acute lymphoblastic

    leukaemia, NONMEM, pharmacogenetics,

    population pharmacokinetics

    ----------------------------------------------------------------------

    Received10 July 2008

    Accepted5 August 2008

    PublishedOnlineEarly24 September 2008

    WHAT IS ALREADY KNOWN ABOUT

    THIS SUBJECT

    The cytotoxic effects of 6-mercaptopurine (6-MP)

    were found to be due to drug-derived intracellular

    metabolites (mainly 6-thioguanine nucleotides and to

    some extent 6-methylmercaptopurine nucleotides)

    rather than the drug itself.

    Current empirical dosing methods for oral 6-MP result

    in highly variable drug and metabolite concentrations

    and hence variability in treatment outcome.

    WHAT THIS STUDY ADDS

    The first population pharmacokinetic model has been

    developed for 6-MP active metabolites in paediatric

    patients with acute lymphoblastic leukaemia and the

    potential demographic and genetically controlled

    factors that could lead to interpatient

    pharmacokinetic variability among this populationhave been assessed.

    The model shows a large reduction in interindividual

    variability of pharmacokinetic parameters when body

    surface area and thiopurine methyltransferase

    polymorphism are incorporated into the model as

    covariates.

    The developed model offers a more rational dosing

    approach for 6-MP than the traditional empirical

    method (based on body surface area) through

    combining it with pharmacogenetically guided

    dosing based on thiopurine methyltransferase

    genotype.

    AIMS

    To investigate the population pharmacokinetics of 6-mercaptopurine (6-MP) activemetabolites in paediatric patients with acute lymphoblastic leukaemia (ALL) and

    examine the effects of various genetic polymorphisms on the disposition of these

    metabolites.

    METHODS

    Data were collected prospectively from 19 paediatric patients with ALL (n = 75

    samples, 150 concentrations) who received 6-MP maintenance chemotherapy

    (titrated to a target dose of 75 mg m-2 day-1). All patients were genotyped for

    polymorphisms in three enzymes involved in 6-MP metabolism. Population

    pharmacokinetic analysis was performed with the nonlinear mixed effects

    modelling program (NONMEM) to determine the population mean parameter

    estimate of clearance for the active metabolites.

    RESULTS

    The developed model revealed considerable interindividual variability (IIV) in the

    clearance of 6-MP active metabolites [6-thioguanine nucleotides (6-TGNs) and6-methylmercaptopurine nucleotides (6-mMPNs)]. Body surface area explained a

    significant part of 6-TGNs clearance IIV when incorporated in the model (IIV

    reduced from 69.9 to 29.3%). The most influential covariate examined, however,

    was thiopurine methyltransferase (TPMT) genotype, which resulted in the greatest

    reduction in the models objective function (P< 0.005) when incorporated as a

    covariate affecting the fractional metabolic transformation of 6-MP into 6-TGNs.

    The other genetic covariates tested were not statistically significant and therefore

    were not included in the final model.

    CONCLUSIONS

    The developed pharmacokinetic model (if successful at external validation) would

    offer a more rational dosing approach for 6-MP than the traditional empirical

    method since it combines the current practice of using body surface area in 6-MP

    dosing with a pharmacogenetically guided dosing based on TPMT genotype.

    British Journal of ClinicalPharmacology

    DOI:10.1111/j.1365-2125.2008.03281.x

    826 / Br J Clin Pharmacol / 66:6 / 826837 2008 The AuthorsJournal compilation 2008 The British Pharmacological Society

    mailto:[email protected]:[email protected]
  • 7/27/2019 Articulo Hawwa

    2/12

    Introduction

    6-Mercaptopurine(6-MP) is a purine antimetabolite widely

    used in the maintenance chemotherapy of childhood

    acute lymphoblastic leukaemia (ALL), the most common

    cancer in children. 6-MP has been a key component of

    almost every successful therapeutic regimen for low- to

    mild-risk ALL. However, it was not until the early 1980s that6-MP cytotoxic effects were found to be due to drug-

    derived intracellular metabolite concentrations rather

    than the plasma level of 6-MP itself [1].

    Following oral administration, 6-MP undergoes exten-

    sive biotransformation by three enzymes, two of which are

    catabolic, xanthine oxidase (XO) and thiopurine S-methyl

    transferase (TPMT) and one anabolic, hypoxanthine phos-

    phoribosyl transferase (HPRT). XO metabolises 6-MP

    to 6-thiouric acid (6-TU),whereasTPMT methylates 6-MP to

    6-mMP. HPRT carries out the first anabolic step to produce

    6-thioinosine monophosphate (6-TIMP) and subsequently

    the active 6-thioguanine nucleotides (6-TGNs). 6-TIMPcan alternatively be methylated by TPMT, yielding

    6-methylmercaptopurine nucleotides (6-mMPNs) [2].

    Finally, it is hypothesized that 6-TIMP is converted succes-

    sively into 6-thioinosine diphosphate (6-TIDP) and triphos-

    phate (6-TITP) to form 6-TIMP once again by the action of

    the enzyme inosine triphosphatase (ITPA) [3].

    Cytotoxic effects of 6-MP are achieved primarily

    through the incorporation of 6-TGNs into the DNA of leu-

    cocytes, due to their structural similarity to the endog-

    enous purine based guanine [2,4]. Moreover, 6-mMPNs are

    strong inhibitors of purine de novo synthesis, which is a

    well-established protocol to achieve immunosuppression

    [5, 6]. Despite these facts, the pharmacokinetics of the

    active metabolites of 6-MP remain poorly explored. A

    better understanding of the disposition of 6-TGNs and

    6-mMPNs would therefore be immensely helpful in

    improving the design of dosing regimens for 6-MP.

    In this study, the pharmacokinetics of 6-TGNs and

    6-mMPNs in paediatric patients with ALL under 6-MP

    maintenance chemotherapy were examined prospectively

    and, for the first time,a population pharmacokinetic model

    for 6-MP active metabolites, 6-TGNs and 6-mMPNs, in pae-

    diatric patients with ALL was developed. In developing this

    model, potential factors that could lead to variability in

    6-MP cytotoxic metabolites that would be particularlyhelpful in improving the dosing guidelines for 6-MP were

    assessed.

    Methods

    Patients and data collectionThe study was approved by the National Health Service

    Office for Research Ethics Committees in Northern Ireland.

    Informed parental consent was obtained for each child

    before enrolment in the study. In addition, verbal assent

    was obtained from older children (>10 years) after provi-sion of a verbal description of the study and what it

    involved.

    Data were collected from 19 paediatric patients attend-

    ing the Haematology and Oncology Outpatient Depart-

    ment at the Royal Belfast Hospital for Sick Children and

    who had been diagnosed to be suffering from ALL. Blood

    samples were taken from children who had been oncontinuous/maintenance 6-MP therapy for at least 1

    month and who had received constant daily doses for at

    least 1 week. Patients who satisfied the above criteria but

    who had received intensification therapy or red blood cell

    (RBC) transfusion within the previous 2 months were

    excluded.

    Blood samples were obtained at a phase of treatment

    when children had an indwelling cannula for vincristine

    therapy and at least 12 h after the preceding 6-MP dose.

    (Note,the blood sample was taken prior to the administra-

    tion of vincristine.)

    Maintenance chemotherapy for ALL patients consistedof daily oral 6-MP and weeklymethotrexate.The 6-MP dose

    was titrated to the target protocol dose of 75 mg m-2 day-1,

    adjusted for each child according to leucocyte count and

    the presence of clinically relevant infections. Additionally, a

    monthly dose of intravenous vincristine was given to all

    children irrespective of blood count. Chemotherapy was

    administered usually for 2 or 3 years.The children had their

    full blood counts assessed at each clinic visit (every 2

    weeks) for bone marrow toxicity.

    The blood samples (1.5 ml) were collected in ethylene-

    diamine tetraaceticacid (EDTA) tubes and kept on ice until

    centrifuged at 1000gfor 10 min to separate plasma from

    RBC. Separated plasma was frozen at-20C in Eppendorftubes while RBC were washed twice with a balanced salt

    solution, then suspended at a density of 8 108 RBC per

    200 ml and kept frozen at -20C until required for further

    processing. These latter samples for the determination of

    metabolite content were taken on a maximum of five occa-

    sions (one sample per occasion), at monthly intervals, over

    the study period.

    A sample of blood (1 ml) from each patient, taken on

    one occasion only,was collected in an EDTA tube and kept

    at -20C without centrifugation for genotyping the

    enzymes of interest (200 ml of whole blood was sufficient

    for this purpose).In addition to information on dosing and times of sam-

    pling, the following data were collected for each child:age,

    weight, surface area, gender, ongoing pathology (e.g.renal

    and/or hepatic impairment), concomitant drug therapy,

    lab test results andrecords of any side-effects experienced.

    The demographic and clinical characteristics of the study

    participants are shown in Table 1.

    Assay of 6-MP metabolitesRBC concentrations of 6-MP active metabolites, 6-TGNs

    and 6-mMPNs, were measured by a reversed high-

    6-Mercaptopurine pharmacokinetics and pharmacogenetics

    Br J Clin Pharmacol / 66:6 / 827

  • 7/27/2019 Articulo Hawwa

    3/12

    performance liquid chromatography methodology that

    was developed earlier [7]. Intraday and interday coeffi-

    cients of variation (CV) were 3.1 and 4.3%, and the limitsof quantification were 13 and 95 pmol per 8 108 RBC,

    respectively.

    Genotyping of TMPT, ITPA and XOAll patients were screened for seven common polymor-

    phisms in three enzymes involved in 6-MP metabolism

    (XO,TPMT and ITPA) that are potentially linked to the phar-

    macodynamics, toxicity and treatment outcome of 6-MP;

    two polymorphisms in XO (A1936G and A2107G) andITPA (C94A and IVS2+21AC) and three polymorphismsin TPMT (TPMT*3A, TPMT*3B and TPMT*3C).

    Detection of the various single nucleotide polymor-phisms (SNPs) in the enzymes genetic loci was based on

    TaqMan genotyping assays (ABI, Foster City, CA, USA)

    using somatic cell DNA extracted from patient blood

    samples (QIAmp DNA Blood Mini kit; Qiagen, Hilden,

    Germany).The conditions used for polymerase chain reac-

    tion and subsequent detection of the genotypes were as

    described in the manufacturers instructions.

    Population pharmacokinetic modellingThe pharmacokinetics of 6-MP active metabolites, 6-TGNs

    and 6-mMPNs, were determined using a population

    approach in which concentrations from ALL patients were

    analysed simultaneously to produce estimates of the phar-

    macokinetic parameters. RBC concentrationtime profiles

    of 6-TGNs and 6-mMPNs were used for nonlinear mixed

    effect modelling by extended least squares regression

    usingNONMEM(version VI, level 1.1 with double precision)

    [8] installed on a personal computer in conjunction with

    DIGITAL Visual Fortran compiler (version 5.0.A). Patientswere assigned randomly to either an index group (n = 15)

    for the development of the pharmacokinetic model, or to

    the validation group (n = 4) for the purpose of assessing

    the predictive performance of the derived model.The first-

    order conditional estimation (FOCE) method with interac-

    tion was used to estimate population mean parameters,

    interindividual variability (IIV) in these parameters and

    residual variability between measured and predicted

    metabolite concentrations.

    The concentrationtime courses of 6-MP metabolites

    were described by using a one-compartment model with

    first-order absorption and elimination.The absorption rateconstant (ka) and the bioavailability factor (F) of the model

    were fixed at 1.3 h-1 and 22%,respectively,according to the

    literature [1,9].The pharmacokinetic parameters estimated

    from this model (implemented using PREDPP subroutine

    ADVAN6) were clearance (CL) for 6-TGNs and 6-mMPNs

    and the fractional metabolic transformation of 6-MP into

    6-TGNs.

    The relationship between the parent drug and its

    metabolites was defined according to the following differ-

    ential equations:

    d A 6-MP

    dtF A 6-MP

    Guta Gut

    ( )= ( )k

    d A 6-MP

    dtF A 6-MP A 6-MP

    Centrala Gut Central

    ( )= ( ) ( )k k20

    d A 6-TGNs

    dtFM A 6-MP

    CL C 6-TG

    CentralCentral

    -TGNs

    ( )= ( )

    3

    6

    kme

    NNs Central( )

    d A 6-mMPNs

    dtFM A 6-MP

    CL C 6-

    CentralCentral

    -mMPNs

    ( )= ( )

    4

    6

    kme

    mmMPNs Central( )

    where ka is the absorption rate constant of the parent drug,k20is the elimination rate constant of 6-MP (k20 = 0.53 h-

    1

    based on literature [1, 9]), kmeis the metabolic transforma-

    tion rate constant of the parent drug into either 6-TGNs or

    6-MPNs.FMiis the fractional metabolic transformationinto

    the metabolite i (6-TGNs are designated by the number

    3 and 6-mMPNs are designated by the number 4, FM4 =

    1 FM3), and A, C are the amount/concentration of the

    drug or metabolite at the time t.k20 = kme + kother (kother is

    the elimination rate constant of bioavailable 6-MP trans-

    formed by other metabolic processes, kother = 0.22 k20 =

    0.1166 h-1 based on the literature [9]).

    Table 1

    The demographic and clinical characteristics of the ALL population

    Parameter n= 19

    Gender (F : M) 6 : 13 (32% : 68%)

    Age [median (range) years] 10 (317)

    Weight [median (range) kg] 33.4 (13.277.5)Body surface area [median (range) m2] 1.14 (0.592)

    6-MP daily dose [median (range) mg] 50 (10100)

    6-MP daily dose [median (range) mg kg-1] 1.42 (0.163.46)

    6-MP daily dose [median (range) mg m-2] 40 (5.8876.47)

    Co-medication during 6-MP chemotherapy

    Methotrexate weekly dose [median (range) mg] 15 (525)

    Cotrimoxazole (b.d. twice weekly) [median

    (range) mg]

    360 (120480)

    Haematological parameters

    Hb [median (range) g dl-1] 12.9 (10.916.5)

    WBC [median (range) 109 l-1] 3.3 (1.29.1)

    PLT [median (range) 109 l-1] 282 (66648)

    ANC [median (range) 109 l-1] 1.68 (0.38.3)

    XO, TPMT, and ITPA genotypes

    XO A1936,

    G (heterozygotes/homozygotes) 1/0XO A2107,G 1/0

    TPMT*3A 1/0

    TPMT*3B

    TPMT*3C 2/0

    ITPA C94,A 3/0

    ITPA IVS2+21A,C 2/1

    ALL, acute lymphoblastic leukaemia; 6-MP, 6-mercaptopurine; WBC, white blood

    cells; PLT, platelets; ANC, absolute neutrophil count; XO, xanthine oxidase; TPMT,

    thiopurine methyltransferase; ITPA, inosine triphosphatase.

    A. F. Hawwa et al.

    828 / 66:6 / Br J Clin Pharmacol

  • 7/27/2019 Articulo Hawwa

    4/12

    An exponential error model was used to describe the

    deviations of the individuals clearance from the true (but

    unknown) population mean values:

    CL TVCL ei i CL= ,

    where CLiis the CL of the ith individual,TVCL is the typical

    population estimate for CL and h i,CLis a random variable

    that distinguish the ith individuals parameter from the

    population mean values as predicted by the regression

    model and is assumed to be normally distributed in the log

    domain with a geometric mean of zero and a variance ofCL

    2 . An exponential model was chosen for the IIV, since

    pharmacokinetic parameters are usually log-normally dis-

    tributed. In modelling this variability, the square root of the

    variance was interpreted as the CV.

    Residual variability, which describes the residual error

    between the measured and the predicted metabolite con-

    centrations,was modelled using additive, proportional and

    combined error structures. The additive error model,

    however, best described the residual variability. This vari-ability could arise from intra-individual variability in the

    pharmacokinetic parameters, inaccuracy in the timing of

    sample collection and dosage administration, assay error,

    model misspecification, or non-adherence to therapy.

    C Cij pred ij ij= +,

    Cij is the measured and Cpred,ij is the model predicted

    metabolite concentration of the ith individual at the jth sam-

    pling time and eij is the residual error term, which is a

    random variable with zero mean and variance ofs2. The

    magnitude of this residual error or variability was

    expressed as a standard deviation (SD).

    Regression modelThe initial analysis for the population pharmacokinetics of

    6-MP metabolites, 6-TGNs and 6-mMPNs, was conducted

    without including any patient covariates in the model

    (BASE model). The conditional estimates of hi,CL were

    obtained from this BASE model and then plotted against

    the following covariates:age, gender, weight, body surface

    area (BSA),TPMT, ITPA and XO genotypes in order to iden-

    tify any potential relationship between CL and the covari-

    ates. The influence of the identified covariates on CL was

    individually assessed by incorporating them in the BASEmodel (univariate analysis).The regression relationship for

    CL was modelled in a linear or nonlinear way for continu-

    ous covariates:

    Proportional: CL = qCL (1 + qCOV COV)

    Power: CL = qCL COVqCOV

    Exponential: qCL eqCOV COV

    where COV is a general continuous covariate and qCLand

    qCOV are the regression coefficients to be estimated by

    NONMEM.

    Categorical covariates (1 or 0) were examined using a

    multiplicative model:

    CL CL COVCOV=

    where qCL is the population value in the absence of the

    covariate (COV = 0) andqCOVis the fractional change when

    the covariate is present (COV = 1). Similar models wereused to investigate the effect of covariates on FM3.

    For each model,the improvement in the fit obtained on

    addition of a fixed effect variable (covariate) into the

    model was assessed using a likelihood ratio test. The

    change in the objective function value (DOBJF) produced

    by the inclusion of a covariate represents a statistic that is

    proportional to minus twice the log-likelihood of the data

    and approximates a c2 distribution with degrees of

    freedom equal to the difference in the number of struc-

    tural parameters (qs) between two models. A decrease in

    the OBJF 6.63 was considered statistically significant

    (P< 0.01, 1 degree of freedom) for the addition of one fixedeffect.The goodness of fit for each model was also assessed

    by examining the precision of parameter estimates (i.e.

    standard errors of the mean), the decrease in interindi-

    vidual and residual variability, and graphs of residuals

    (RES), weighted residuals (WRES) and measured metabo-

    lite concentrations plotted separately against predicted

    concentrations.

    As a result of the univariate analysis, each model with

    significant effect was ranked according to its DOBJF com-

    pared with the BASE model. The model with the largest

    DOBJF was designated as the INTERMEDIATE model and

    multiple regression analysis with forward selection wasperformed where covariates were incorporated into the

    INTERMEDIATE model one by one along the rank order

    established by univariate analysis until all significant cova-

    riates were included and no further statistically significant

    reduction in OBJF was obtained (FULL model). The FULL

    model was then subjected to stepwise, backward elimina-

    tion to obtain the FINAL model. An increase in the OBJF

    6.63 (P< 0.01, 1 degree of freedom) was required toretain the covariate in the FINAL model.

    Model evaluation

    Since external validation using a new dataset from anotherstudy is extremely difficult in paediatric studies, internal

    validation using data splitting or resampling techniques is

    an appropriate alternative [10]. In the present study, inter-

    nal validation using the data-splitting technique was per-

    formed to verify the predictive value of the population

    model.

    The predictive performance of the model was assessed

    in terms of bias (mean prediction error) and precision (root

    mean square predictionerror) by comparing the measured

    concentrations in the validation group (n = 4) with the cor-

    responding predicted values by the population model

    6-Mercaptopurine pharmacokinetics and pharmacogenetics

    Br J Clin Pharmacol / 66:6 / 829

  • 7/27/2019 Articulo Hawwa

    5/12

    usingpost hocBayesian forecasting. This was achieved by

    fixing the structural and variance model parameters to the

    values estimated in the final population model.

    The mean prediction error (ME) and root mean square

    prediction error (RMSE) were determined according to the

    following formulae [11]:

    MEN

    peii

    N

    ==

    11

    MSEN

    peii

    N

    ==

    1 2

    1

    RMSE MSE =

    The prediction error (pei) was calculated as:

    pei= C Cobs pred

    whereCobsandCpredrepresent the observed and predictedconcentrations, respectively.

    In addition,WRES and measured metabolite concentra-

    tions were plotted separately against the predicted con-

    centrations to assess visually the deviations of model

    predicted from observed metabolite concentrations in the

    validation group.

    In order to evaluatethe performance of the final model,

    obtained by fitting the full dataset (comprising index and

    validation groups combined together), a posterior visual

    predictive check was performed by simulating from

    the final estimates and comparing the distribution of

    the observations with the simulated distribution. The

    adequacy of the model was demonstrated by plotting thetime course of the observations along with the prediction

    interval for the simulated values.

    Results

    On oral administration of 6-MP, large interindividual differ-

    ences were observed as regards metabolite concentra-

    tions and the course of elimination as shown in Figure 1. It

    is apparent from the graph that patients with TPMT muta-

    tions had higher 6-TGN concentrations but compared withother patients they had relatively low 6-mMPN concentra-

    tions in erythrocytes.

    At certain time points after treatment with the drug,

    6-TU could also be identified in addition to the parent

    drug, 6-MP. However, their low levels (due to the sampling

    time chosen in this study as stated above) did not qualify

    them for incorporation into NONMEM analysis. NONMEM

    analysis in this pharmacokinetic study, therefore, was per-

    formed using the measured erythrocyte levels of 6-TGNs

    and 6-mMPNs in all samples (n = 75 samples, 150 concen-

    trations) obtained from 19 paediatric patients with ALL

    receiving 6-MP maintenance chemotherapy (a maximum

    of five samples was obtained per patient, one sample per

    occasion).

    Pharmacokinetic modellingIn the initial model, the data were described with a one-

    compartment model with first-order absorption and elimi-

    nation since there were no points to enable the accurate

    evaluation of the distribution phase. In addition,since mostof the kinetic data were collected in the post-absorption

    phase,ka and F(the bioavailability factor) could not be

    reliably estimated.Hence, their values were fixedaccording

    to the literature (1.3 h-1 and 22%, respectively) throughout

    the analysis [1, 9]. An additive error model best described

    the residual variability and was used in the basic pharma-

    cokinetic model (BASE) in order to be used for further

    analysis.

    The population estimates from the BASE model for FM3(the fractional metabolic transformation of 6-MP into

    6-TGNs), CL6-TGNs(6-TGN clearance) and CL6-mMPNs(6-mMPN

    0

    5000

    10000

    15000

    20000

    25000

    30000

    4321

    Time (in months)

    6-mMPNs(pmo

    l/8108R

    BCs)

    0

    200

    400

    600

    800

    1000

    1200

    4321Time (in months)

    6-TGNs(pmol/8108R

    BCs)A

    B

    Figure 1Individual 6-thioguanine nucleotide (6-TGN) (A) and

    6-methylmercaptopurine nucleotide (6-mMPN) (B) concentration plots.

    Patients having any mutation are highlighted and their corresponding

    types are displayed.The therapeutic lower and upper limits suggested in

    literature areindicated by thedashedlines.TPMT mutant();ITPAmutant

    (); TPMT and ITPA mutant ()

    A. F. Hawwa et al.

    830 / 66:6 / Br J Clin Pharmacol

  • 7/27/2019 Articulo Hawwa

    6/12

    clearance) were 0.028, 0.0125 l h-1 and 0.0231 l h-1 with

    an interindividual variability (%CV) of 69.9 and 38.1% for

    CL6-TGNsand CL6-mMPNs, respectively. The residual variability

    corresponded to a SD of 0.18 and 9.04 mg l-1 of packed

    RBC for 6-TGN and 6-mMPN metabolite levels,respectively.

    Regression modelsPlotting of the conditional estimates of hi,CL for the

    metabolites 6-TGNs and 6-mMPNs in paediatric patients

    with ALL from the BASE model vs. covariates showed some

    potential relationships. An example is shown in Figure 2.

    Univariate analyses showing covariates with significant

    effects on FM3, CL6-TGNs and CL6-mMPNs are presented in

    Table 2. The addition of several covariates resulted in a

    reduction of the OBJF of>6.63 (P< 0.01). These covariateswere TPMT mutations affecting FM3, CL6-TGNsand CL6-mMPNsparameters, along with weight (WT) and BSA, which

    affected CL6-TGNs.

    In the model that incorporated weight as a covariateaffecting CL6-TGNs, clearance was standardized using an allo-

    metric model that is based on theoretical and empirical

    evidence that CL is proportional to the 3/4power of weight

    [12].

    The covariate that caused the largest reduction in the

    OBJF was the dichotomous covariate model incorporating

    the effect of TPMT mutations on FM3. Therefore, it was

    declared as an INTERMEDIATE model, subsequent addition

    to which of the significant covariates was analysed. Multi-

    variate analysis incorporating both BSA and TPMT muta-

    tions as covariates showed that in the presence of the

    effect of TPMT mutations on FM3, the effect of BSA on

    CL6-TGNs remained significant. The results of multivariate

    analysis together with the various steps in building the

    FULL model are presented in Table 3.Backward elimination

    of any covariate from the FULL model increased the OBJF

    by>6.63.Therefore, both factors TPMT mutations and BSAwere retained in the FINAL model.

    Adequate correlations between predicted andobserved RBC concentrations of 6-TGNs and 6-mMPNs

    were observed in the FINAL model, as shown in Figures 3

    and 4.The scatterplots show that the population predicted

    and individual predicted concentrations of 6-TGNs and

    6-mMPNs were in reasonable agreement with the mea-

    sured concentrations around the line of identity, although

    some underprediction could be observed, particularly at

    higher concentrations.

    Model evaluationBias (ME) and precision (RMSE) of the predictive perfor-

    mance of the FINAL model were tested in the validationgroup (n = 4) between the measured and predicted

    metabolite concentrations. ME computed values for

    6-TGNs and 6-mMPNs were -0.047 and 1.67 mg l-1,respec-

    tively. The corresponding RMSE values were 0.16 and

    4.84 mg l-1, respectively. Both were less than the residual

    unexplained variability of the index group, which corre-

    sponded to a SD of 0.188 and 9.04 mg l-1 for 6-TGNs and

    6-mMPNs, respectively.

    The scatter plots of weighted residuals vs. model

    predicted RBC concentrations of 6-TGNs and 6-mMPNs

    (Figure 5) showed that both were randomly distributed

    and the weighted residuals lay within 2 units of the nullordinate of perfect agreement. Specific examples of the

    predictive capability of the final model are shown for two

    representative individual patients in the validation group

    in Figure 6, which shows the time course of measured and

    post hoc predicted RBC concentrations of 6-TGNs and

    6-mMPNs.

    Final population pharmacokinetic modelThe computed population structural parameter estimates,

    the interpatient variability (CV%) and the residual variabil-

    ity obtained by fitting the full dataset to FINAL model are

    presented in Table 4.The final population model for 6-TGNs and 6-mMPNs in

    RBC was:

    FM TPMT

    3 0 019 2 56= ( ). .

    CL h BSA6-TGNs1.16

    1 0 009141( ) = ( ).

    CL h6-mMPNs 1 0 02281( ) = .

    where BSA is body surface area in m2 andTPMT = 1 in case

    of mutation, otherwise = 0.

    1.50

    1.00

    0.50

    0.00

    0.50

    1.00

    1.50

    2.001.00 1.50

    Surface area (m2)

    hi,CL6-TGNs

    2.00

    Figure 2Plots of the conditional estimates ofh i,CL6-TGNsvs. body surface area.The

    solid line indicates the Lowess smooth line

    6-Mercaptopurine pharmacokinetics and pharmacogenetics

    Br J Clin Pharmacol / 66:6 / 831

  • 7/27/2019 Articulo Hawwa

    7/12

    For a hypothetical individual with population median

    value of BSA (i.e.1.14 m2),the model-predicted FM3, CL6-TGNs

    and CL6-mMPNs would be 0.019, 0.011 l h-1 and 0.0288 l h-1,respectively. The FM3would increase by 256% to 0.049 if

    the patient had a TPMT mutation. The model for CL 6-TGNsfound that CL6-TGNswas proportional to the 1.16 power of

    BSA, resulting in an estimated range of 0.00490.02 l h-1

    across the BSA range of 0.592.0 m2 among the study

    group.

    The median individual Bayesian estimates for FM3,

    CL6-TGNs, CL6-mMPNs and BSA normalized CL6-TGNs values

    obtained by fitting the full dataset from the study popula-

    tion to the FINAL model are presented in Table 5. These

    estimates are determined as a postprocessing step using

    the measured concentrations, in contrast to the populationparameter estimates, which are derived from covariate

    information. Since the individual Bayesian estimates are

    drawn from a distribution where the population estimates

    reflect the posterior mode of the marginal likelihood dis-

    tribution for that parameter, a possibility for differences in

    this summary parameter could be introduced. In order to

    evaluate the predictive performance of the final model

    obtained from the full dataset,a posterior predictive check

    was performed (Figure 7).

    The interpatient variability (CV%) for the population

    pharmacokinetic parameters of CL6-TGNsand CL6-mMPNswere

    33.6 and 33.2%, respectively. The interpatient variability of

    CL6-TGNswas reduced from 65.6 to 33.6% by the inclusion of

    BSA covariate, which explained most of the variability in

    CL6-TGNs between individuals.The residual unexplained vari-

    ability (SD) was 0.177 and 8.42 mg l-1 for 6-TGNs and

    6-mMPNs, respectively, which translates to a CV% of 24.3

    Table 2

    Summary of univariate analysis showing covariate models with significant effects on FM3, CL6-TGNsor CL6-mMPNsof 6-TGN and 6-mMPN metabolites

    Effect on Model DOBJF* Parameter estimate P-value

    FM3 FM TPMT 3 TPMT

    -8.69 FM3 = 0 129.

  • 7/27/2019 Articulo Hawwa

    8/12

    and 57.6% at the mean RBC concentrations of 6-TGNs and

    6-mMPNs measured in the full dataset (0.729 and

    14.61 mg l-1, respectively).

    Discussion

    Current empirical dosing methods for oral 6-MP result in

    highly variable drug and metabolite concentrations, asdemonstrated by the present study and other investiga-

    tors [13, 14]. The variability presumably arises in part from

    individual differences in bioavailability. Other factors,

    however, that could contribute to this variability were

    evaluated in the present study for paediatric patients with

    ALL using a population pharmacokinetic modelling

    approach. The different factors studied were age, gender,

    WT and BSA, along with various genetic factors such as

    polymorphisms in XO,TPMT and ITPA enzymes.

    Quantitativeevaluation of pharmacokineticparameters

    of 6-MP seems especially attractive and is a potentially

    important prognostic factor in cancer chemotherapy [15].

    In addition, the study of drug distribution is of particular

    relevance for paediatric patients, given the effect of matu-

    rational changes on organ function and body composition

    that can affect drug disposition [16]. For these reasons, we

    developed in this study, for the first time, a population

    pharmacokinetic model for 6-MP and its metabolites for

    paediatric patients with ALL.

    The pharmacokinetic model developed revealed con-

    siderable IIV in the clearance of both 6-MP metabolites

    investigated, 6-TGNs and 6-mMPNs. This variability,

    however, coincides with the highly variable RBC concen-trations of 6-TGNs and 6-mMPNs reported previously in

    children taking identical doses of 6-MP [13, 14]. The esti-

    mated IIV (CV%) of clearance in the BASE model, fitted to

    the index group, was 69.9 and 38.1% for 6-TGNs and

    6-mMPNs, respectively. The model, however, showed large

    reduction in the IIV of 6-TGN clearance (69.929.3%) and

    a significant reduction in the OBJF (7.95, P< 0.005) whenBSA alone was incorporated into the model as a covariate

    affecting 6-TGN clearance.This indicated that large part of

    the IIV in 6-TGN clearance was explained by differences in

    BSA.

    0 706050403020100

    10

    20

    30

    40

    50

    60

    70

    Population predicted 6-mMPN

    concentrations (mg/L)

    0 70605040302010

    Individual predicted 6-mMPN

    concentrations (mg/L)

    Obse

    rved6-mMPN

    concen

    trations(mg/L)s

    0

    10

    20

    30

    40

    50

    60

    70

    Observed6-mMPN

    concentra

    tions(mg/L)

    A

    B

    Figure 4Scatter plots of observedvs.population predicted (A) and individual pre-

    dicted(B) red blood cell (RBC)concentrations of 6-methylmercaptopurine

    nucleotides (6-mMPNs) in the index group of the FINAL model. The solid

    line represents the line of identity

    0 0.80.70.60.50.40.30.20.1

    0 252015105

    1.5

    1

    0.5

    0

    0.5

    1

    1.5

    Model predicted 6-TGN

    concentrations (mg/L)

    Weig

    htedresiduals

    0.6

    0.4

    0.2

    0

    0.2

    0.4

    0.6

    0.8

    1

    Model predicted 6-mMPN

    concentrations (mg/L)

    Weightedresiduals

    A

    B

    Figure 5Scatter plots of weighted residuals vs. predicted concentrations of

    6-thioguanine nucleotides (6-TGNs) (A) and 6-methylmercaptopurine

    nucleotides (6-mMPNs) (B) in red blood cells (RBC)

    6-Mercaptopurine pharmacokinetics and pharmacogenetics

    Br J Clin Pharmacol / 66:6 / 833

  • 7/27/2019 Articulo Hawwa

    9/12

    Both the WT and BSA had significant effects on 6-TGN

    clearance in this study. BSA, however, was found to be a

    better predictor of clearance than WT. The final model

    obtained by fitting the full dataset indicated that CL6-TGNsincreased by 116% per 1-m2 increase in the BSA resulting

    in an estimated range of 0.00490.02 l h-1 across the BSA

    range of 0.592.0 m2 among the study group.

    Clinical experience also confirms the effect of impaired

    renal function on the concentration of 6-MP metabolites

    and its clinical outcome. It has been shown that patients

    with impaired renal function have increased susceptibility

    to 6-MP side-effects. In the present study, none of the

    patients had clinically significant renal impairment and

    hence renal function was mainly proportional to patients

    BSA.

    The rationale behind the use of BSA as a criterion for

    dosing in anticancer chemotherapy was outlined about

    50 years ago [17], giving rise to the practice of using BSA

    for dosing anticancer therapy. The current approach for

    dosing anticancer drugs is to administer a standard dose,

    which is normalized to BSA,and then to adjust or individu-

    alize subsequent doses based on the severity of drug tox-

    icity. Even though this clearly has practical and economicimplications, its clinical value has, in recent years, been

    questioned [18]. One objection to the use of BSA in mea-

    suring drug dosage was the difficulty in measuring BSA.

    Apart from the inaccuracies inherent in methods for BSA

    calculation [19], they are dependent on the accuracy of

    weight and height measurements used in BSA calculation.

    Another alternative was the use of normograms to avoid

    inaccurate determination of BSA.However, the reliability of

    these normograms tends to differ [20].

    Another objection was that pharmacokinetic studies of

    anticancer drugs revealed substantial interpatient variabil-

    ity in plasma drug concentrations when the dose wasbased on BSA [13, 21].The potential consequences of this

    variability in systemic drug exposure are life-threatening

    toxic effects in patients who are exposed to excessive drug

    concentrations and tumour progression in patients who

    achieve subtherapeutic drug concentrations. The identifi-

    cation of specific factors that account for variability in drug

    disposition among patients can, therefore, lead to more

    rational dosing methods.

    In the present study, the most influential covariate

    examined was TPMT genotype on the fraction of meta-

    bolic transformation, FM3. Its inclusion in the model

    resulted in the greatest reduction in OBJF (DOBJF = -8.69,

    P< 0.005). Hence, it explained part of the variability in FM3between individuals. In addition,the final model predicted

    an increase in FM3 of 256% if the patient had a TPMT muta-

    tion. This would mean preferential 6-TGN production and

    6-mMPN underproduction if the patient had a TPMT muta-

    tion. This is in perfect agreement with previous studies,

    which demonstrated an inverse correlation between TPMT

    activity and the levels of 6-TGNs in RBC, suggesting that

    an inherited decrease in the methylation step (due to the

    TPMT mutation) results in shunting 6-MP metabolism

    away from 6-mMPNs towards overproduction of 6-TGNs

    [22, 23]. More importantly,the optimal dose of 6-MP, which

    wasdetermined in the standard empirical fashion,was alsorelated to TPMT genotype [22, 24].

    No prior studies, however, have provided specific 6-MP

    dosing guidelines based on TPMT genotype.In the present

    study, for the first time, the potential utility of TPMT geno-

    type in prospectively defining the percentage of 6-MP con-

    verted to 6-TGNs or 6-mMPNs as predicted by FM3 was

    illustrated.This would identify patients who are less toler-

    ant of 6-MP standard doses based on their capacity to

    metabolize the drug. One limitation of the developed

    model, however, was its inability to account for variation in

    TPMT activity among patients having the wild-type. This

    4500400035003000200015001000500 25000.0

    0.2

    0.4

    0.6

    0.8

    10

    15

    20

    25

    Concentr

    ation(mg/L)

    Time (hrs)

    4500400035003000200015001000500 2500

    Time (hrs)

    0.00.10.20.30.40.50.6

    6

    8

    10

    12

    14

    16

    18

    20

    Concentration(m

    g/L)

    A

    B

    Figure 6Longitudinal assessment of the predictive performance of the FINAL

    model in two representative patients from the validation dataset: (A)

    12-year-old boy and (B) 6-year-old girl. () Observed and () model-

    predicted 6-thioguanine nucleotide (6-TGN) concentrations. ()

    Observed and () predicted 6-methylmercaptopurine nucleotide

    (6-mMPN) concentrations

    A. F. Hawwa et al.

    834 / 66:6 / Br J Clin Pharmacol

  • 7/27/2019 Articulo Hawwa

    10/12

    could be the reason for underprediction observed in the

    model for 6-mMPN concentrations at higher levels. Higher

    concentrations of 6-mMPNs are probably due to high

    TPMT activity that could not be accounted for by the

    model. Phenotyping patients, however, by measuringTPMT enzymatic activity could resolve this limitation and

    allow for individualization of drug doses among the whole

    population.

    The other genetic covariates tested in this study (ITPA,

    XO mutations) were not statistically significant and there-

    fore not included in the FINAL model. It should be noted,

    however, that the IIV not explained by the model might

    have resulted, in part, from these covariates. It is also pos-

    sible that the small number of patients studied (n = 19)

    was not enough to identify more potential sources of IIV.

    Therefore, the developed population model will need to

    be tested and refined by larger prospective studies that

    have the potential to encounter and quantify more

    covariates.

    The reasonably large residual (unexplained) variability

    in the model might be due to large IIV in the pharmaco-

    kinetic parameters, interoccasional variability, assay errors,

    or model misspecification. Since the data in most patients

    were collected for a period of several months, this mighthave added to the variability within the same patient.

    Moreover, the significant IIV in drug absorption may

    explain some of the variability obtained with the orally

    administered 6-MP [25]. Non-adherence to therapy could

    also have contributed to this variability. Children with ALL

    who have reached remission always remain under a pro-

    longed and complex course of chemotherapy in spite of

    being practically asymptomatic. These factors are likely

    to contribute to missing some doses given the home

    therapy of 6-MP and the absence of obvious conse-

    quences at the time. Therefore, non-adherence is

    somehow expected in this group of patients and shouldbe considered.

    Although the robustness of the pharmacokinetic

    model developed in this study was validated internally

    using the data-splitting technique, it is highly recom-

    mended that the model be subjected to external valida-

    tion before using it to guide dosage adjustments. If

    successful, this would offer a more rational dosing

    approach than the traditional empirical method, since it

    would combine the current practice of using BSA in 6-MP

    dosing with a pharmacogenetically guided dosing based

    on TPMT genotype.

    Table 4

    Population pharmacokintics of 6-TGNs and 6-mMPNs, obtained by fitting the full dataset (19 children) to the BASE and FINAL models

    Parameter Symbol

    Base model Final model

    Estimate w(CV%) Estimate w(CV%) SE (%)

    FM3 FM3 0.0295 0.0191 64.1

    CL6-TGNs(l h-1

    ) CL6-TGNs

    0.0138 65.7 0.00914 33.6 56.8CL6-mMPNs(l h-1) CL6-mMPNs 0.0227 33.9 0.0228 33.1 14.2

    TPMT qTPMT 2.56 35.9

    BSA qBSA 1.16 49.0

    wCL6-TGNs 0.431 0.113 81.2

    wCL6-mMPNs 0.115 0.11 82.6

    s6-TGNs(mg l-1) 0.171 0.177 17.5

    s6-mMPNs(mg l-1) 9.04 8.42 19.3

    Structural models:

    TVFM FMTPMT

    3 3= TPMT

    TVCL BSA-TGNs -TGNs6 6= ( )

    CL

    BSA

    TVCL -mMPNs -mMPNs6 6= CL

    Random effects models:

    CL TVCL e-TGNsi 6-TGNsi CL6-TGNs

    6 = ,

    CL TVCL e- mMPN si 6- mMPN s i CL6-mMPNs6 = ,

    where FM3is the fractional transformation of 6-MP into 6-TGNs; TPMT, 1 if the patient had TPMT mutation and 0 otherwise; CL 6-TGNsis 6-TGN clearance; BSA is body surface area

    in m2; CL6-mMPNs is 6-mMPN clearance; w is the interindividual variability; h is squared w; s6-TGNs and s6-mMPNs are the residual variabilities of 6-TGNs and 6-mMPNs. 6-TGN,

    6-thioguanine nucleotide; 6-mMPN, 6-methylmercaptopurine nucleotide; TPMT, thiopurine methyltransferase.

    Table 5

    IndividualBayesianestimatesobtained from theFINAL populationmodel

    Parameter Median (P5, P95)

    FM3 (no TPMT mutation) 0.0191

    FM3 (with TPMT mutation) 0.0491

    CL6-TGNs(l h-

    1) 0.0112 (0.0046, 0.0235)CL6-mMPNs(l h-1) 0.0226 (0.0153, 0.0310)

    CL6-TGNs(l h-1 m-2)* 0.0082 (0.0059, 0.0151)

    P5, 5th percentile; P95, 95th percentile. *Calculated using the body surface area

    of each individual. 6-TGN, 6-thioguanine nucleotide; 6-mMPN, 6-

    methylmercaptopurine nucleotide; TPMT, thiopurine methyltransferase.

    6-Mercaptopurine pharmacokinetics and pharmacogenetics

    Br J Clin Pharmacol / 66:6 / 835

  • 7/27/2019 Articulo Hawwa

    11/12

    Competing interests

    None declared.

    REFERENCES

    1Lennard L, Keen D, Lilleyman JS. Oral 6-mercaptopurine in

    childhood leukemia: parent drug pharmacokinetics andactive metabolite concentrations. Clin Pharmacol Ther 1986;40: 28792.

    2Lennard L. The clinical pharmacology of 6-mercaptopurine.Eur J Clin Pharmacol 1992; 43: 32939.

    3Derijks LJ, Gilissen LP, Hooymans PM, Hommes DW. Reviewarticle: thiopurines in inflammatory bowel disease. AlimentPharmacol Ther 2006; 24: 71529.

    4Fairchild CR, Maybaum J, Kennedy KA.Concurrent unilateralchromatid damage and DNA strand breakage in response to

    6-thioguanine treatment. Biochem Pharmacol 1986; 35:

    353341.

    5Elion GB. The purine path to chemotherapy. Science 1989;244: 417.

    6Cara CJ, Pena AS, Sans M, Rodrigo L, Guerrero-Esteo M,Hinojosa J,Garcia-Paredes J,Guijarro LG.Reviewing themechanism of action of thiopurine drugs: towards a new

    paradigm in clinical practice. Med Sci Monit 2004; 10:RA24754.

    7Hawwa AF, Millership JS, Collier PS, McElnay JC.

    Improved HPLC methodology for the determination ofmercaptopurine and its metabolites in plasma and redblood cells. J Pharm Pharmacol 2006; 58 (Suppl. 1): A3.

    8Beal SL, Sheiner LB, Boeckmann AJ (eds). NONMEMU UsersGuides. Ellicott City, MD: Icon Development Solutions,19892006.

    9Chabner B, Amrein P, Druker B. Chemotherapy of neoplasticdiseases. In: Goodman & Gilmans the Pharmacological Basisof Therapeutics, 11th edn, eds Brunton L, Lazo J, Parker K.

    New York, NY: McGraw-Hill, 2006; 13151405.

    10Food and Drug Administration Guidance for Industry.Guidance for Industry: Population Pharmacokinetics.

    Rockville, MD: U.S. Department of Health and HumanServices, 1999.

    11Sheiner LB, Beal SL. Some suggestions for measuring

    predictive performance.J Pharmacokinet Biopharm 1981; 9:50312.

    12Holford NH. A size standard for pharmacokinetics.ClinPharmacokinet 1996; 30: 32932.

    13Lennard L, Lilleyman JS. Variable mercaptopurine

    metabolism and treatment outcome in childhoodlymphoblastic leukemia. J Clin Oncol 1989; 7: 181623.

    14Chrzanowska M, Kolecki P, Duczmal-Cichocka B, Fiet J.

    Metabolites of mercaptopurine in red blood cells: arelationship between 6-thioguanine nucleotides and6-methylmercaptopurine metabolite concentrations in

    children with lymphoblastic leukemia. Eur J Pharm Sci 1999;8: 32934.

    15Evans WE, Relling MV. Clinical

    pharmacokineticspharmacodynamics of anticancer drugs.Clin Pharmacokinet 1989; 16: 32736.

    16McLeod HL, Relling MV, Crom WR, Silverstein K, Groom S,

    Rodman JH, Rivera GK, Crist WM, Evans WE. Disposition ofantineoplastic agents in the very young child. Br J CancerSuppl 1992; 18: S239.

    17Pinkel D. The use of body surface area as a criterion of drugdosage in cancer chemotherapy. Cancer Res 1958; 18: 8536.

    18Kaestner SA, Sewell GJ. Chemotherapy dosing part I:scientific basis for current practice and use of body surfacearea. Clin Oncol (R Coll Radiol) 2007; 19: 2337.

    19Du Bois D, Du Bois EF.A formula to estimate theapproximate surface area if height and weight be known.1916. Nutrition 1989; 5: 303, 11; discussion 3123.

    45004000350030002000150010000 500 2500

    Time (hrs)

    45004000350030002000150010000 500 2500

    Time (hrs)

    1

    0

    1

    2

    3

    4

    5

    6-TGNconce

    ntrations(mg/L)

    30

    10

    10

    30

    50

    70

    90

    6-mMPNconcentrations(mg/L)

    A

    B

    Figure 7Visual predictive check of the final model fitted to the full dataset ( n = 19

    patients). A plot of the time course of the observed concentrations of6-thioguanine nucleotides (6-TGNs) (A) and 6-methylmercaptopurine

    nucleotides (6-mMPNs) (B) along with the median and 90% prediction

    intervals for the simulated values. Median prediction (------); 90% predic-

    tion interval (); Observed concentrations ()

    A. F. Hawwa et al.

    836 / 66:6 / Br J Clin Pharmacol

  • 7/27/2019 Articulo Hawwa

    12/12

    20Reilly JJ, Workman P. Normalisation of anti-cancer drug

    dosage using body weight and surface area: is itworthwhile? A review of theoretical and practicalconsiderations. Cancer Chemother Pharmacol 1993; 32:4118.

    21Zimm S, Collins JM, Riccardi R, ONeill D, Narang PK,Chabner B, Poplack DG. Variable bioavailability of oral

    mercaptopurine. Is maintenance chemotherapy in acutelymphoblastic leukemia being optimally delivered? N Engl J

    Med 1983; 308: 10059.

    22Evans WE, Horner M, Chu YQ, Kalwinsky D, Roberts WM.Altered mercaptopurine metabolism, toxic effects,and dosage requirement in a thiopurine

    methyltransferase-deficient child with acute lymphocytic

    leukemia. J Pediatr 1991; 119: 9859.

    23Wang L, Weinshilboum R. Thiopurine S-methyltransferasepharmacogenetics: insights, challenges and futuredirections. Oncogene 2006; 25: 162938.

    24Balis FM, Adamson PC. Application of pharmacogenetics to

    optimization of mercaptopurine dosing. J Natl Cancer Inst1999; 91: 19835.

    25Mawatari H, Unei K, Nishimura S, Sakura N, Ueda K.Comparative pharmacokinetics of oral 6-mercaptopurineand intravenous 6-mercaptopurine riboside in children.Pediatr Int 2001; 43: 6737.

    6-Mercaptopurine pharmacokinetics and pharmacogenetics

    Br J Clin Pharmacol / 66:6 / 837