Physiologically-Based Pharmacokinetics And

  • Upload
    que12

  • View
    225

  • Download
    0

Embed Size (px)

Citation preview

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    1/35

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    2/35

    Xu, Eiseman, Egorin, and DArgenio186

    model proidedin vivoestimates of endogenous HSP70 and HSP90 turnoer. In modeling pharm-acokinetics and pharmacodynamics, Bayesian inference was employed to estimate the kinetic,

    physiological and molecular parameters when prior information was aailable.

    KEY WORDS: geldanamycin; physiologically-based pharmacokinetics; molecular pharmaco-dynamics; intrinsic clearance; Bayesian estimation; onco-protein; heat shock protein; auto-regulation.

    INTRODUCTION

    17-(allylamino)-17-demethoxygeldanamycin (17AAG) is a derivative of

    the benzoquinone ansamycin antibiotic geldanamycin and is currently in

    phase I testing (1). Geldanamycin is a potent antiproliferative agent that

    produces growth arrest in the G1 phase of most tumor cell lines (2); how-

    ever, it is also associated with dose-limiting hepatotoxicity.In itroand ani-

    mal studies have shown that while the antiproliferative activity and

    mechanism of action of 17AAG is similar to that of geldanamycin (35), it

    is considerably less hepatotoxic than its precursor (6).

    Preclinical pharmacology studies of 17AAG, including metabolism

    studies in mouse and human hepatic preparations, reveal several metabolites

    including the active CYP3A4 product 17-(amino)-17-demethoxygeldanamy-

    cin (17AG) (7). Murine pharmacokinetic studies of intravenous 17AAG

    show that both 17AAG and 17AG are widely distributed to tissues, where

    they are present in most tissues at substantial concentrations, with only

    small amounts of each compound detected in urine (8). Compartmental and

    non-compartmental analyses of plasma data reported in mice indicate linear

    kinetics over a dose range of 26.67 to 40 mgkg with a total systemic clear-ance of 17AAG in the range 54 to 74 mlminkg (8).

    Extensivein itrostudies indicate that the antiproliferative activity and

    mechanism of action of ansamycin antibiotics, including 17AAG, correlate

    with their ability to deplete oncoproteins such as p185erbB2, mutant p53 and

    Raf-1 (9,10). The proto-onco-proteins p185erbB2 and Raf-1 are essential for

    transmission and amplification of mitogenic signals from the cell surface to

    nucleus in normal cells and both are over-expressed in certain tumor cell

    lines. The specific mechanism of oncoprotein depletion by ansamycin anti-

    biotics is thought to be related to their ability to disrupt the molecular

    chaperone function of heat shock protein HSP90 and its homologue

    GRP94, causing the degradation of their client proteins that require the

    chaperones for maturation and stability (1015).

    The work reported in this paper builds on the previously reported pre-

    clinical pharmacology studies of 17AAG, known mechanisms of action of

    ansamycin antibiotics, and current understanding of pathways involved

    with the auto-regulation of heat shock proteins. The goals of the present

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    3/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 187

    study were: (1) use data from normal mice to develop a physiologically-

    based pharmacokinetic (PBPK) model of 17AAG and its primary active

    metabolite 17AG; (2) use data of 17AAG and 17AG concentrations in the

    tumors of nude mice bearing human breast cancer xenografts to develop a

    model for the uptake and distribution of these compounds in the tumor;

    (3) use measurements of p185erbB2, Raf-1 and heat shock proteins (HSP70

    and HSP90) in the tumor xenografts to develop molecular pharmaco-

    dynamic models relating the cellular concentrations of 17AAG and 17AG

    to the targeted onco-proteins and heat shock proteins based on the known

    molecular mechanisms of drug action; and (4) apply Bayesian estimation

    methods that allow the combination of both informative and non-informa-

    tive prior information in physiological pharmacokinetic and molecularpharmacodynamic modeling. The work culminates in a complete PBPKPD model for the investigational anticancer agent 17AAG and its metab-

    olite 17AG, and provides insights into the molecular mechanism of action

    of these compounds in io.

    METHODS

    Experimental Methods

    Animal Experiments

    In experiments on non-tumor-bearing mice, previously reported in (8),

    48 adult female CD2F1 mice were divided into 16 time groups (one group

    as vehicle control) for terminal sampling of plasma, red blood cells (RBC),

    lung, brain, heart, spleen, liver, kidney and skeletal muscle at 5, 10, 15, 30,

    45 min and 1, 1.5, 2, 3, 4, 6, 7, 16, 24, 48 hr following drug administration.

    In a subsequent set of experiments on tumor-bearing mice, 16 NCR SCID

    adult female mice (weighing 17.321.0 gm) with MDA-MB-453 humanbreast cancer xenografts in their flank were divided into 8 time groups (one

    group as vehicle control ) for terminal sampling of plasma, lung, heart,

    spleen, liver, kidney, and tumor at 2, 4, 7, 16, 24, 48, and 72 hr following

    drug administration. In both experiments, 17AAG was injected as an intra-

    venous bolus via lateral tail vein and at a dose of 40 mgkg fasted bodyweight. At each designated time point, the animals were euthanized with

    carbon dioxide and venous blood was collected by cardiac puncture until

    the liver became pale. Organstissues were dissected out, weighted andstored frozen until analyzed. Food was withheld until 4 hr after dosing.

    In both non-tumor-bearing and tumor-bearing experiments, 17AAG was

    supplied by the National Cancer Institute (8).

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    4/35

    Xu, Eiseman, Egorin, and DArgenio188

    Assays

    Plasma and tissue concentrations of 17AAG and 17AG were deter-

    mined by HPLC (see Ref. 8 for details). The concentrations of Raf-1,p185erbB2, HSP90 and HSP70 in tumor tissues were determined by western

    blot analysis. The western blot band density readings were first normalized

    to actin and then referenced to the average normalized reading of the con-

    trols. The measurement variances for each protein were determined by giv-

    ing a group of three animals control vehicle and measuring the levels of the

    protein in various organs.

    Blood Distribution and Protein Binding

    The concentration profiles of 17AAG and 17AG in venous RBC andplasma indicated that a rapid equilibrium was achieved between the two

    fractions in blood for both compounds. Accordingly, the RBC and plasma

    concentrations of 17AAG and 17AG from the non-tumor experiments were

    used to calculate the RBC-plasma partition coefficient (RGCRBCCplasma)for each compound: RAAGG2.47J1.23 (meanJSD, nG30) and RAGG

    5.86J2.88 (nG30). The unbound fractions of 17AAG and 17AG in plasma

    (fupGCplasma,free/Cplasma) were measured in a separate in itrostudy with the

    following results: fup,AAGG0.063J0.015 (nG10) and fup,AGG0.081J0.042

    (nG10) (8). It was found that RAAG, RAG , and fup,AAG were concentrationindependent. The unbound free fractions in blood thus can be calculated as

    follows:

    fuu,drugG1AHct

    (1AHct)CHctRdrugfup,drug (1)

    Using a hematocrit (Hct) of 0.45 yields,fub,AAGG0.0209 andfub,AGG0.0140.

    Concentrations of 17AAG and 17AG in venous blood were calculated from

    the plasma and RBC measurements as follows:

    CVen(t)G(1AHct)Cplasma (t)CHctCRBC(t) (2)

    No study of 17AAG or 17AG protein binding in the interstitial fluid

    or cells was conducted. Thus concentrations of both compounds in inter-

    stitial fluid and cells were taken to be homogenous. The exchange of each

    compound across the vascular wall was therefore assumed to occur between

    the free fraction in blood and the total in tissue.

    Physiologically-Based Pharmacokinetic Modeling

    Non-Tumor-Bearing Mice

    The PBPK model for 17AAG and 17AG in non-tumor-bearing mice

    was constructed through the following six steps: (1) fit sums of exponentials

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    5/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 189

    to the venous blood data of 17AAG and 17AG separately; (2) simul-

    taneously model the distribution of 17AAG and 17AG in the lung using

    the fitted venous exponential functions as input, and predict the arterial

    concentration-time profile for each compound; (3) in each non-eliminating

    organ, simultaneously model the distribution of 17AAG and 17AG using

    the predicted arterial concentration-time profiles as input; (4) simul-

    taneously model the liver distribution and metabolism (see below for

    detailed description); (5) construct a whole-body model from the foregoing

    results and model the distribution in the unsampled tissues (Misc.); and (6)

    simulate the resulting whole-body model to predict the concentration pro-

    files of 17AAG and 17AG in blood and all tissues.

    In modeling the individual organs, both perfusion-limited and dif-fusion-limited models were evaluated to describe the distribution of 17AAG

    or 17AG in each organ. See Appendix A for the diagrams, equations and

    parameter descriptions of the two model structures for non-eliminating and

    eliminating organs. The lists of fixed and estimated model parameters are

    given in Tables AI and AII. Different combinations of the perfusion- and

    diffusion-limited models for the distribution of 17AAG and 17AG in the

    same organ were evaluated by fitting simultaneously to the measurements

    of both compounds. The models of 17AAG and 17AG in the same organ

    shared the same set of anatomical and physiological parameters.The liver is the only eliminating organ of significance for 17AAG and

    17AG (8). No prior information is available on the intrinsic clearances of

    17AAG (CLintAAG), 17AG (CLintAG) in the liver or on the fraction of

    CLintAAG responsible for the formation of 17AG (fm). Furthermore, these

    parameters cannot be uniquely determined via individual organ model-

    based estimation using solely the measured liver concentration data. To

    solve this problem of unidentifiability of hepatic parameters, the following

    equation relating the systemic clearance to the intrinsic clearance of 17AAG

    in the liver was derived (see Appendix B for detailed derivation):

    CLsAAGGQVVfub,AAGkV,EV,AAG

    Q1CkEV,V,AAGCLintAAG VEVCVVfub,AAGkV,EV,AAG(3)

    where Q, VV, kV,EV,AAG , and kEV,V,AAG are parameters for liver. In deriving

    this systemic-intrinsic clearances relationship constraint, a diffusion-limited

    model was used for the distribution of 17AAG in the liver. The systemic

    clearance of 17AAG (CLsAAG) was calculated as dose divided by the area

    under the predicted arterial concentration-time profile of 17AAG. A similar

    constraint derivation for the metabolite 17AG is given in Appendix C, and

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    6/35

    Xu, Eiseman, Egorin, and DArgenio190

    leads to the following equation for the case of a perfusion-limited model:

    AUC(CArt,AG(t))GfmCLintAAGAUC(CEV,AAG(t))

    CLintAGRAG(4)

    The area under the arterial concentration-time curve of 17AG

    (AUC(CArt,AG )) was calculated from the predicted arterial concentration

    profile of 17AG. With these two additional relationship constraints, the

    drug-specific kinetic parameters of liver can be uniquely estimated for the

    composite liver model.

    The unsampled tissues, including the carcass, were modeled as a single

    tissue (labeledMisc.) arranged in parallel with the other systemic organs.

    The previously selected models for each of the measured non-eliminating

    organs and for the liver were included in a whole-body model with each of

    the four candidate models for Misc. Weight and blood flow of Misc.

    were calculated by subtracting the sum of the organ weights and blood flows

    of the measured organs from the total body weight and cardiac output.

    Other parameters ofMisc.were estimated for each of the four candidate

    whole-body models byfitting the model prediction of venous concentration

    profiles to the venous measurements of 17AAG and 17AG simultaneously.

    Tumor-Bearing Mice

    A whole-body model for the distribution of 17AAG and 17AG intumor-bearing mice was constructed by adding a parallel vascular bed

    representing the tumor to the whole-body model developed for the non-

    tumor-bearing mice. All kinetic and physiological parameters for normal

    organs were fixed at the values determined from the non-tumor model, and

    the anatomical parameters were replaced by those measured in the tumor

    animal experiments. It was assumed in this approach that the animal physi-

    ology and drug transport kinetics in tumor-bearing mice were the same as

    those in non-tumor-bearing mice.

    A number of different diffusion-limited models were evaluated todescribe the tumor distribution of 17AAG and 17AG, including a model

    with a vascular and an extra-vascular space, and a model with an extra-

    cellular and cellular space, as well as several models containing vascular,

    interstitial and cellular spaces. One of the tested models is shown in Fig. 1,

    with Eqs. (58) describing the drug concentration (17AAG or 17AG) in

    each of the tumor spaces and the measured tissue concentration.

    VVdCV

    dt

    GQ(CArtACV)APSV,IT(CVACIT) (5)

    VITdCIT

    dtGPSV,IT(CVACIT)AkIT,CCITVITCkC,ITCCVC (6)

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    7/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 191

    Fig. 1. Tumor Model. The subscripts V, IT, and C represent the vascular space,interstitial space and cellular space; PSV,IT represents the permeability surface-areaproduct of the vascular wall; kIT,Cand kC,ITare the transport rate constants acrossthe cell membrane.

    VCdCC

    dtGkIT,CCITVITAKC,ITCCVC (7)

    CtissueGCITVITCCCVC

    VITCVC(8)

    After incorporating the candidate tumor models into the whole-body

    model for the two compounds, the parameters of the tumor models were

    estimated by fitting the model predictions to the measured tumor tissue

    concentrations of 17AAG and 17AG simultaneously.

    Model Validation

    The organ concentrations of 17AAG and 17AG measured in the

    tumor-bearing animal experiments were used in model validation. The

    whole-body PBPK model with tumor was simulated to predict the organconcentrations of 17AAG and 17AG, which were then compared to the

    organ concentrations measured in the tumor experiments.

    Molecular Pharmacodynamic Modeling

    The concentration profiles of 17AAG and 17AG in the tumor cells

    predicated from the whole-body PBPK model with tumor were used to

    model the action of these compounds on the tumor cell onco-proteins

    (p185erbB2 and Raf-1) and heat shock proteins (HSP70 and HSP90). These

    ansamycins were shown to be equally potent in depleting p185erbB2 (3), and

    were assumed to also exhibit equal potency in their actions on Raf-1, HSP70

    and HSP90.

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    8/35

    Xu, Eiseman, Egorin, and DArgenio192

    Onco-Protein Responses

    The role of HSP90 in chaperoning Raf-1 is summarized in Fig. 2. After

    HSP90 binds to newly synthesized Raf-1, the HSP90-Raf-1 complex is trans-ported to the cell membrane where it is available for activation by Ras.

    Following activation, the HSP90-Raf-1 complex binds to unknown cyto-

    skeletal elements, activates down-stream kinases and promotes tumor

    growth (14). Ansamycins, including 17AAG, are thought to act by inducing

    the degradation of the HSP90-Raf-1 complex at each of its three sites equ-

    ally, and by inhibiting transport of the complex as shown in Fig. 2 (14).

    Since the net action of ansamycins is to enhance the proteasomal degra-

    dation of Raf-1 and since the cellular Raf-1 measured in our experiments is

    the sum of the protein at the three sites, the simplified indirect responsemodel (16) shown in Fig. 3 and Eq. (9) were postulated to explain the

    response of Raf-1 to 17AAG and 17AG. A similar model was used for the

    onco-protein p185erbB2.

    dO

    dtGRsynAkdg,b1C Emax (AAGCAG)EC50C(AAGCAG)O (9)

    Fig. 2. Molecular mechanism of Raf-1 depletion by ansamycin antibiotics (14). 1: The newlysynthesized Raf-1 protein complexes with HSP90; 2: The Raf-1-HSP90 complex in cytosol istransported to the cell membrane; 3: HSP90 remains bound to Raf-1 when Raf-1 is recruitedand activated by Ras; 4: HSP90 remains bound to Raf-1 when Raf-1 binds to unknown cyto-skeletal elements and activates downstream kinases. Ansamycins destabilize the cytosolic, Ras-associated and skeleton-associated Raf-1. The disappearances of Raf-1 from the three fractionsare equal. Raf-1 synthesis in ansamycin treated cells is elevated (approximately 3-fold) andansamycins inhibit the transportation of Raf-1-HSP90 complex.

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    9/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 193

    Fig. 3.Indirect response model for Raf-1 and erbB2.O represents the onco-proteins Raf-1 andp185erbB2 with the associated parameters unique to each onco-protein; Rsynrepresents the zero-order synthesis rate of the protein; kdg,b represents the basalfirst-order degradation rate of theprotein; (AAGCAG ) is the total concentration of 17AAG and 17AG in tumor cells.

    Heat Shock Protein Responses

    The molecular mechanism of heat shock protein auto-regulation and

    the responses of HSP70 and HSP90 following exposure to 17AAG and

    17AG are depicted in Fig. 4. The transcription of HSP genes and the

    resulting synthesis of HSPs are enhanced upon exposure of cells to proteo-

    toxic stress such as the presence of ansamycin antibiotics. This transcrip-

    tional enhancement is due to the activation of heat shock transcription

    factor 1 (HSF1), which is capable of specifically binding to the HSP genespromoter sequence and thereby enhancing the transcription of HSP genes.

    While the synthesis of HSPs is up regulated by active HSF1, the transcrip-

    tional activity of HSF1 is down regulated by direct binding of HSP70,

    HSP90 and other co-factors (1720). We have hypothesized that this auto-

    regulatory system is responsible for the heat shock response measured in

    the mouse tumor xenograft model following 17AAG administration. Based

    on the signaling pathway shown in Fig. 4, the feedback control model

    described in Fig. 5 was constructed for which the following equations can

    be written:

    dHSP70P1

    dtGR70syn HSF1Ak70 HSP70P1 (10)

    dHSP70Pi

    dtGk70 HSP70P(iA1)Ak70 HSP70Pi iG2, . . . ,n (11)

    dHSP70

    dt

    Gk70 HSP70PnAk70dg,b

    1C

    E70max (AAGCAG)

    EC70

    50C(AAGCAG )HSP70 (12)

    dHSF1

    dtGFactAkdeact A(HSP70CHSP90)HSF1 (13)

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    10/35

    Xu, Eiseman, Egorin, and DArgenio194

    Fig. 4. The auto-regulation of HSP70 and HSP90 through HSF1a gene-transcription-mediated feedback control system. HSF1 exists in the control state as inert monomers. Uponstress (such as exposure to 17AAG and 17AG), the concentration of HSP70 is reduced. HSF1loses contact with HSP70, and trimerizes. The transcriptionally active trimers of HSF1 bindto heat-shock protein genes and lead to the production of HSP70 and HSP90 proteins. Thetranscriptional activity of HSF1 is repressed by direct binding of HSP70 and HSP90 to theHSF1 trimer, and HSF1 eventually returns to its inert monomer state.

    A time-dependent transduction system (21) was adopted to model the timedelay in HSP70 gene expression (Eqs. (10), (11)). The initial values of

    HSP70pi (iG1, . . . , n) were assumed to be equal and were estimated. The

    initial values of HSP70 and HSF1 were set at one. The differential equations

    for HSP90 follow the same format as those for HSP70 (Eqs. (10)(12)). The

    activation function of HSF1, Factin Eq. (13), is depicted in detail in Fig. 5.

    It is a function of HSP70 and involves several parameters including the

    basal activation rate (Ract,b) and the amplification factor (A). The function

    Fact increases as HSP70 decreases below its baseline value (HSP70b). When

    HSP70 exceeds this baseline, the rate of activation of HSF1 reminds at its

    basal level (Ract,b). The rate of de-activation of HSF1 is a function of the

    HSP70 and HSP90 concentrations. The amplification factorA was assumed

    to be the same for activation and deactivation.

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    11/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 195

    Fig. 5. Model diagram of HSP auto-regulation upon treatment of 17AAG and 17AG. TheHSP70 and HSP90 represent the normalized concentration of the two heat-shock proteins;Rsyn

    is the basal zero-order synthesis rate and kdg,bis the basal first-order degradation rate; HSPP1HSPPn are the transit compartments representing the protein synthesis delay for HSP70 andHSP90; k70 and k90 are the transit rate constants where the total delay time Gnk (21); Factis the activation function of HSF1 transcriptional activity, in which Ract,bis the basal and zero-order activation rates, and HSP70b is the HSP70 concentration at control condition; kdeact isthe basal first-order de-activation rate of HSF1 transcriptional activity; A is the scalar rep-resenting the amplification effect of gene activation and deactivation.

    Estimation Methods and Software

    In the modeling steps described above, model parameters were esti-

    mated using either Maximum Likelihood estimation (ML) when no prior

    information was available for any of the model parameters, or Maximum a

    PosterioriBayesian estimation (MAP) when prior information was available

    for at least one of the model parameters. Both ML and MAP estimation

    were performed using the ADAPT pharmacokineticpharmacodynamic sys-tems analysis software (22). Model selection was guided using the Akiake

    information criterion (AIC) for ML estimation and generalized information

    criterion (GEN-IC) for MAP estimation (see Ref. 22).

    A version of MAP estimation has been used that allows estimation of

    parameters for which prior information (mean and covariance) is available

    as well as parameters with no prior information (i.e., parameters with non-

    informative priors). For example, for a perfusion-limited organ model, prior

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    12/35

    Xu, Eiseman, Egorin, and DArgenio196

    information was available for organ blood flow while no prior information

    was available for the 17AAG or 17AG partition coefficients. In general, if

    represents the vector of all parameters to be estimated, and 1 is the

    subset of parameters with known distribution p1(1) and 2 is an indepen-

    dent subset of parameters with distribution p2(2), then the MAP estimate

    of(MAP) is:

    MAPGarg max{lnl(z 1 , 2)Clnp1 (1)Clnp2 (2)} (14)

    wherez is the vector of measured data andl(z ) is the likelihood function.Ifp2(2) is non-informative, p2(2)Gconstant, then:

    MAPGarg max{lnl(z 1 , 2)Clnp1 (1)} (15)

    Equation (15) can be solved when the likelihood is normally distributed and

    p1(1) is either normally or log-normally (used herein) distributed.

    The following equation was used to model the variance of the normally

    distributed output error for 17AAG, 17AG, Raf-1, p185erbB2, HSP90, and

    HSP70: 2i(tj)G(interCslopeyi(tj))2. The values for inter and slope for

    17AAG and 17AG are 0.136 and 0.104. For the onco-proteins, slopeG

    0.167, and for the heat shock proteins, slopeG0.180, with interG0 for both

    the onco-proteins and heat shock proteins.

    RESULTS

    Physiologically-Based Pharmacokinetic Model

    Whole-Body Results

    The composite whole-body model for 17AAG-17AG in tumor-bearing

    mice is depicted in Fig. 6. The individual organ models for the uptake of

    17AAG incorporate diffusion-limited exchange for all organs, while per-

    fusion-limited models were selected to describe 17AG uptake in all organs.

    Estimates of the model parameters (with the exception of liver and tumor

    which are presented in the next sections) are given in Table I (physiological

    parameters) and Table II (drug-specific kinetic parameters). Table II lists

    the vascular-to-extra-vascular clearance (CLV,EVGkV,EVVVfub) and the

    extra-vascular-to-vascular clearance (CLEV,VGkEV,VVEV) for each diffusion-

    limited organ as well as Rthe partition coefficient value for each per-

    fusion-limited organ. Recall that while the protein binding of 17AAG and

    17AG was measured in the blood and incorporated in the model, it was

    assumed that all 17AAG and 17AG in tissue are available for extra-

    vascular-to-vascular transport. The predicted tissue concentrations from the

    whole-body model for non-tumor-bearing mice and the measured tissue

    concentrations for 17AAG and 17AG are shown in Figs. 7 and 8.

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    13/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 197

    Fig. 6. Composite whole-body model for 17AAG and 17AG intumor-bearing mice. The acronyms for the organs used in equa-tions are: LU: lung, BR: brain, HT: heart, SP: spleen, LI: liver,KI: kidney, MU: muscle, Art: arterial blood and Ven: venousblood.

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    14/35

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    15/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 199

    Fig. 7. Whole-body model simulation results for 17AAG.

    The estimated model parameters are listed in Table III and Fig. 10 shows

    the liver concentration-time profiles of 17AAG and 17AG predicted by the

    estimated whole-body non-tumor mouse model.

    The uptake of 17AAG in liver tissue was found to be diffusion-limited

    with no significant transport from the extra-vascular space to the vascular

    space (kEV,VG0). This result is supported by in itro studies of 17AAG

    metabolism in mice and human hepatic preparations, in which the metabolic

    activity was found to reside predominantly in microsomes with little or no

    metabolism of 17AAG in liver cytosol (7). Our model suggests that upon

    entering hepatocytes, 17AAG is either trapped in microsomes where it is

    metabolized or secreted into bile. Interestingly, with only the difference of

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    16/35

    Xu, Eiseman, Egorin, and DArgenio200

    Fig. 8. Whole-body model simulation results for 17AG.

    a propenyl group (CH2CHCH2) on the side chain of the ansamycin ring,

    17AG produced in microsomes can be released from microsomes and trans-

    ported out of hepatocytes into blood circulation. 17AG can be secreted into

    bile but cannot be further metabolized (7). A perfusion-limited model was

    found to adequately describe the distribution of 17AG in the liver. The

    hepatic metabolism of 17AAG is complex, with metabolites, including

    17AG, found in the plasma and bile of mice treated with 17AAG (7,8).However, neither the identity of the metabolites nor the metabolic or

    secretory pathways have been fully characterized. By simultaneously model-ing the distribution and elimination of 17AAG and 17AG in liver, we wereable to estimate that 40% of the total intrinsic clearance of 17AAG involvesthe formation of its metabolite 17AG.

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    17/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 201

    Fig. 9. Liver model diagram of 17AAG and 17AG.

    Table III.Liver Model Kinetic and Physiological ParameterEstimates

    Parameter Unit 17AAG 17AG

    kV,EV hr1 6132

    CLV,EV mlhr 27.45 R 4.87CLint mlhr 4.93 3.34

    fm 0.40 Q

    a % CO 18.9VV

    b % VLI 23.9

    aLiterature meanJSDG16.10J2.45 (23,24).bLiterature meanJSDG31.0J7.0 (25).

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    18/35

    Xu, Eiseman, Egorin, and DArgenio202

    Fig. 10. Whole-body model simulation result of 17AAG and 17AG concentrations in liver.

    Tumor Model

    Of the five different models postulated to describe the uptake of

    17AAG and 17AG in the human breast tumor xenografts, the model shown

    in Fig. 1 was selected for both 17AAG and 17AG. The resulting estimates

    of the vascular-interstitial permeability-surface area product (PSV,IT) as well

    as the interstitial-cellular exchange rate constants (kIT,Cand kC,IT) are listed

    in Table IV for both 17AAG and 17AG. Table V lists the model estimates

    of tumor vascular and interstitial volumes. Because the distribution of both

    17AAG and 17AG in tumor was diffusion-limited, there was no information

    Table IV. Tumor Model Kinetic Parameters

    Parameter Unit 17AAG 17AG

    PSV,IT mlhr 0.227 0.264kIT,C hr

    1 0.186 0.566kC,IT hr

    1 0.062 0.057

    Table V. Tumor Model Physiological Parameters

    PopulationParameter Unit Estimate meanJSD

    Q mlmingm tissue a 0.154J0.114a

    VV % tumor volume 6.5 5.5J2.1b

    VIT % tumor volume 54.3 43.4J3.5c

    aQ was fixed at the mean value in the model estimation. Mean and stan-dard deviation obtained from measurements of 31 human anaplastic car-cinomas (26).

    bLiterature values from Refs. 27 and 28 were pooled to generate the popu-lation mean and SD for the vascular volume.

    c Mean was calculated from the interstitial volumes reported for carci-nomas in Ref. 29, and the highest CV% reported in Ref. 29 was used toobtain the SD.

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    19/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 203

    Fig. 11. Tumor model results of PK16A. Model predictions (solid lines) and measurements(symbols) of 17AAG (left) and 17AG (right) concentrations in tumor tissue (interstitial and

    cellular). The dashed lines are the model predictions of the concentrations of 17AAG and17AG in tumor cells.

    in the data on tumor blood flow. Tumor blood flow thus was fixed at the

    population mean value and not estimated as the other physiological param-

    eters. The 17AAG and 17AG tumor tissue and cell concentrations predicted

    from the composite whole-body model with tumor are shown in Fig. 11

    together with the tumor tissue measurements.

    Model Validation

    The whole-body model constructed using the data from the non-tumor

    animal study was used to predict the 17AAG and 17AG concentrations

    measured in the organs of the tumor-bearing mice. Figure 12 shows the

    model predictions along with the measured concentrations for 17AAG in

    selected tissues, while the corresponding results for 17AG are displayed in

    Fig. 13.

    Molecular Pharmacodynamic ModelsThe Onco-Proteins Raf-1 and p185erbB2

    The model predictions and the measured values of Raf-1 and p185 erbB2

    in the tumor are shown in Fig. 14. Long-term model simulation predicts

    that the level of Raf-1 in tumor returns to its base value at approximately

    200 hr, while p185erbB2 returns to its pre-drug value at about around 120 hr

    (graphs not shown). Estimated model parameters are listed in Table VI and

    compared with corresponding in itro literature values. The half-lives of

    Raf-1 and p185erbB2 andEC50 of Raf-1 estimated from the model compare

    favorably to the values reported from in itroexperiments. The degradation

    rate of p185erbB2 was estimated to be linearly related to the concentration of

    17AAG and 17AG, so no EC50on p185erbB2 depletion was estimated.

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    20/35

    Xu, Eiseman, Egorin, and DArgenio204

    Fig. 12. Measured and predicted concentrations of 17AAG in organs in tumor-bearing mice.

    Fig. 13. Measured and predicted concentrations of 17AG in organs in tumor-bearing mice.

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    21/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 205

    Fig. 14. Onco-protein model predictions and measurements.

    Table VI. Onco-Protein Models Parameter Estimates and Literature Values

    Raf-1 p185erbB2

    Model Literature Model LiteratureParameter Unit estimation values estimation values

    t1/2a hr 22.6 11b, 17.5b 8.63 7c, 9.5d

    EC50 nM 3.47 3.4e f

    Rsyn norm. conc.hr 0.03 0.08 Emax 4.30 0.24

    f

    aHalf-life of endogenous protein turnover (t1/2Gln 2kdg,b from Fig. 3);b

    In

    itromeasurement reported in Ref. 14.cIn itro measurement reported in Ref. 30.dIn itromeasurement reported in Ref. 11.eThe value is derived from the in itro EC50 of geldanamycin reported in Ref. 14, given that

    17AAG is three-fold more potent than geldanamycin (3) and that the concentration of 17AAGin tumor cells is 2.3 times that in whole tissue.

    fFor p185erbB2, a linear model relating drug induced degradation to total drug concentrationwas used in the place of the Emaxmodel. In the table, Emax represents this scale factor.

    Heat Shock ProteinsHSP70 and HSP90

    The measurements and model predictions of HSP70 and HSP90 in

    tumor are shown in Fig. 15. Long-term model simulation predicts that the

    Fig. 15. Heat-shock protein model predictions and measurements.

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    22/35

    Xu, Eiseman, Egorin, and DArgenio206

    Table VII. Heat-Shock Protein Model Parameter Estimates

    HSP70 HSP90

    Model ModelParameter Unit estimation estimation

    t1/2a hr 4.51 40.4

    EC50 nM 0.90 38.7Rsyn hr

    1 0.15 0.02Emax 1.64 31.9 hr 13.6 0Ract,b 0.08kdeact 0.04A 331

    aHalf-life of endogenous protein turnover (t1/2Gln 2kdg,b from

    Fig. 5).

    level of HSP70 in tumor returns to its baseline value at approximately

    200 hr, while HSP90 returns to its pre-drug value at about around 300 hr

    (graphs not shown). Table VII lists the resulting estimates of the heat shock

    protein model parameters. There is no report in the literature from eitherin

    io or in itro studies that provide a basis for comparing the results from

    our analysis.

    DISCUSSION AND CONCLUSION

    Distribution of 17AAG and 17AG in Individual Organs

    The model and the data agree reasonably well for 17AAG and 17AG

    in venous blood and all organs, except for 17AG in brain and lung. The

    measured values of 17AG concentration in brain tissue fall mostly on the

    edge of the lower limit of quantification, and the measurements are associ-

    ated with large standard deviation. While there is no report on whether

    17AG can cross the bloodbrain barrier, the data and our model suggest

    there is little uptake in brain tissue. It is also likely that the measurements

    of 17AG in brain included drug in the residual blood remaining in the tissue

    sample. The lung 17AG data has a peak value at 5 min while the peak

    concentration of 17AG in venous blood is not reached until 30 min, which

    resulted in a poorfit regardless of the models tested. We could notfind any

    physiological evidence to explain the lung 17AG data. However, the particu-

    lar models used to describe 17AG distribution in brain and lung had little

    effect on the whole-body model predictions, including those in the tumor.

    The Bayesian estimates of all the physiological parameters are reason-

    able given the prior means, except for muscle blood flow. The relatively

    large variability in muscle concentration data, especially that of 17AG, may

    contribute to this discrepancy.

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    23/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 207

    Estimation of Intrinsic Clearances and 17AAG Metabolism in the Liver

    The intrinsic clearance of 17AAG (CLintAAG), the fraction ofCLintAAG

    associated with the formation of 17AG (fm) and the intrinsic clearanceof 17AG CLintAG in the liver are especially relevant to understanding the

    distribution, metabolism and hepatic toxicity of 17AAG. We have estab-

    lished a method to uniquely estimate these parameters at the stage of indi-

    vidual organ analysis in the model developing process (i.e., using only the

    liver measurements and the arterial concentration profiles). The relationship

    between intrinsic clearance and systemic clearance of 17AAG was derived

    from a simplified whole-body model by solving the model equations in

    Laplace domain assuming linear kinetics and only hepatic elimination (see

    Appendix B ). We have proved that the CLintAAGCLsAAG relationshipdepends only on the liver model (work not shown), when the remaining

    systemic organs are lumped into a single composite tissue. The resulting

    equation relatingCLintAAGand CLsAAGwas then included in the liver model

    as the additional constraint, thus allowing CLintAAG to be uniquely esti-

    mated as part of the individual liver modeling procedure. An equation rela-

    ting CLintAGand fm to the systemic distribution of 17AG was also derived

    in a similar fashion (see Appendix C) to uniquely estimate CLintAGandfm.

    The approach we developed for estimating liver clearance in PBPK

    models has the following advantages: (1) it allows intrinsic clearance andits related parameters (such as fm) to be estimated without constructing the

    whole-body model; (2) it makes it possible to model the unsampled tissues,

    when significant drug uptake is accounted in those unsampled tissues.

    Although the liver model describes the data reasonably well, there was

    a consistent overprediction of 17AAG in the early time period from 0

    15 min. The highest mean measured concentration of 17AAG in liver tissue

    was 89 mgml at 5 min, while the model prediction at 5 min is 215 gml.A number of factors might contribute to this discrepancy. Firstly, roughly

    75% of the blood entering the liver is the venous effluent from the small

    intestine, stomach, pancreas, and spleen, while the model assumes no uptake

    of 17AAG in the small intestine, stomach or pancreas. The uptake of

    17AAG to those tissues may have a significant effect on the drugs distri-

    bution. Secondly, in our investigation of different liver model structures, we

    found that a model consisting of three sub-organ spaces (vascular, inter-

    stitial, and cellular) did not reduce the over prediction. The systemic clear-

    ance, defined by the 17AAG data of venous blood and other organs,

    indicated a high-transport rate of 17AAG from blood to liver tissue, while

    the liver 17AAG measurements, especially at early times, are consistent with

    a lower transport rate independent of the liver model structure. This obser-

    vation suggests a discrepancy between the information imbedded in liver

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    24/35

    Xu, Eiseman, Egorin, and DArgenio208

    measurements and that in venous and other organ measurements. It is poss-

    ible that the continuous metabolism of 17AAG in liver tissue during the

    time of animal and organ processing contributes to this discrepancy. And

    lastly, the assumption of a constant fm may contribute to the difference

    between measured and predicted 17AAG liver concentrations as well.

    Tumor Xenograft Uptake of 17AAG and 17AG

    One of the attractive properties of geldanamycin derivatives as

    anticancer agents is their considerably longer half-lives in tumor compared

    to normal organstissues. We were able to describe this property by a three-component model that includes vascular, interstitial fluid and cellular spaces

    of the tumor. The model revealed that sustained concentration-time profilesof 17AAG and 17AG in tumor tissue was due to a relatively slow diffusion

    across the cell membrane, while in normal organs this process was estimated

    to be essentially instantaneous. The concentrations of 17AAG and 17AG in

    tumor cells themselves were estimated to be both 2.3 times higher than those

    in the tumor tissue (in extra-vascular space), showing the preferential uptake

    of both compounds in tumor cells.

    It was found from the data that although the concentration of 17AG

    in blood was generally less than that of 17AAG, the concentration of 17AG

    in tumor was significantly higher (1.5 to 3 fold) than that of 17AAG after2 hr following dosing. In the tumor models for 17AAG and 17AG, the per-

    meability surface-area product (PS) of the vascular wall was estimated to

    be similar for 17AAG (0.23 mlhr) and 17AG (0.26 mlhr), and the trans-port rate constants of the two compounds from cellular to interstitial space

    (kC,IT) were also estimated to be similar (0.062 hr1 for 17AAG and

    0.057 hr1 for 17AG). However, the transport rate constant of 17AG from

    interstitial space to the cellular space (kIT,C) was estimated to be significantly

    higher than that of 17AAG (0.57 hr1 for 17AG vs. 0.19 hr1 for 17AAG).

    The model estimates suggest that the preferential uptake of 17AG over17AAG in tumor tissue is due to a more effective uptake mechanism of

    17AG in the tumor cells rather than in the interstitial fluid of tumor tissue.

    Pharmacodynamic Model for the Onco-Proteins

    Mechanisms of Raf-1 synthesis, transport, activation, and depletion by

    ansamycins have been studied extensively at the sub-cellular and molecular

    level (see Fig. 2). Because the three forms of measurable Raf-1 protein were

    destabilized equally by ansamycins, they were modeled as one single pool

    of Raf-1 with an indirect response model. Although HSP90 plays a key role

    in the regulation of Raf-1 stabilization and degradation, it was not included

    in the Raf-1 model, for the following reasons: (1) HSP90 is present in the

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    25/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 209

    cells in excess (12% of total proteins in the cell is HSP90), so that all the

    Raf-1 molecules are chaperoned by HSP90; (2) the binding of HSP90 to

    newly synthesized Raf-1 is essentially instantaneous; and (3) the degradation

    of Raf-1 is assumed to be instantaneous following the disruption of the

    HSP90-Raf-1 complex by 17AAG and 17AG. Although the transport and

    activation pathways of p185erbB2 are different from those of Raf-1, the

    action of ansamycin antibiotics on the degradation of p185erbB2 is the same

    as that of Raf-1 and therefore was also described using the same indirect

    response model.

    In the model, the synthesis of the onco-proteins was assumed to remain

    constant following drug administration. However, it was reported that the

    synthesis of Raf-1 in geldanamycin treated cells was elevated approximately

    3-fold (14). In addition, in herbimycin A treated SKBr3 cells (0.35 mM,

    6 hr), it was reported that p185erbB2 mRNA levels were elevated by 30% and

    the total protein synthesis decreased by 16% (31). Although the synthesis of

    p185erbB2 protein was not measured directly, the above finding suggests the

    possible induction of p185erbB2 synthesis following ansamycin treatment.

    The mechanism of the synthesis inductions remains unknown, nor has there

    been any quantitative analysis of the process. With limited data and infor-

    mation, we were not able to model the synthesis induction. The discrepancy

    between measured and model predicted p185erbB2 concentrations might be a

    direct result of this inability. The data and model prediction suggest that

    after a period of depletion, the fast recovery of p185erbB2 is at least partially

    the result of the induced p185erbB2 synthesis.

    Pharmacodynamic Model of HSP Auto-Regulation

    The auto-regulation of the heat shock transcriptional response, dis-

    covered in the early 1990s is an example of a self-regulatory biological pro-

    cess. While the role of heat shock proteins as molecular chaperons is under

    intensive study, little quantitative information is available on the dynamicsof HSP auto-regulation. For example, there have been no published reports

    of the half-life of HSP70 or HSP90, thus the model-based estimates obtained

    from our modeling work represent the first reported HSP70 and HSP90

    turnover rates (HSP90 t1/2G40.43 hr and HSP70 t1/2G4.51 hr).

    There has been considerable debate regarding the pathways responsible

    for mediating the cellular stress signal in the heat shock auto-regulatory

    system (19). One hypothesis states that external stress alters HSF1 directly,

    while a second postulates that HSP70 serves as the intermediary between

    cell stress and HSF1 activation. The model developed herein incorporates

    the later hypothesis, and assumes that HSF1 activation increases linearly as

    HSP70 decreases below its control value with that it remains constant as

    HSP70 increases above the control value.

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    26/35

    Xu, Eiseman, Egorin, and DArgenio210

    The time course of HSPs concentration measurements in this study is

    not long enough to adequately reveal the dynamics of the processes associ-

    ated with the return of HSP70 or HSP90 to their baseline values. The esti-

    mated parameter values and the model prediction of HSPs recovery

    reported, await confirmation by further in itro and in io studies.

    Bayesian Analysis

    Recent advances in PBPK modeling have further established the rel-

    evance of this approach in drug development (e.g., Ref. 32). Bayesian

    methods have also been applied previously in PBPK modeling within a hier-

    archical population modeling framework (e.g., Refs. 33, 34). While the

    MAP estimation approach used herein does not involve a mixed effectsmodeling formulation, it does provide a simple-to-use method for incorpor-

    ating prior uncertainty in PBPK modeling, and can be implemented using

    most nonlinear regression software.

    The use of Bayesian inference in model development exposes all parts

    of the model, including the model structure and the prior distribution, to

    appropriate criticism. An example of how the model development in the

    work reported herein benefits from the model criticism function of Bayesian

    inference is as follows. One of the models considered for the tumor uptake

    of 17AAG and 17AG was a 2-compartment diffusion-limited model con-sisting of an extra-cellular compartment and an intra-cellular compartment,

    i.e., assuming drug exchange across the leaky tumor vascular wall is

    instantaneous. This model would have been selected as the best model based

    on the AIC selection criterion, if the physiological parameters were fixed at

    their prior values and the kinetic parameters were estimated by maximum

    likelihood estimation. However, the Bayesian analysis of this model resulted

    in unrealistic physiological parameter values, suggesting that this 2-com-

    partment model structure is inappropriate. Indeed, by including separate

    vascular and interstitial spaces, a 3-compartment model with reasonableparameter estimates was established for tumor. The exchange of the com-

    pounds across the vascular wall was determined by a permeability area-

    surface product estimated at 0.23 mlhr and 0.26 mlhr for 17AAG and17AG.

    SUMMARY

    In the present work, we have: (1) constructed a physiologically-based

    pharmacokinetic model for 17AAG and 17AG in nude mice bearing human

    tumor xenografts, to characterize the disposition of 17AAG and 17AG in

    normal mice organs and human breast tumor xenografts; (2) established

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    27/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 211

    the modeling procedures to uniquely estimate the intrinsic clearances and

    clearance related parameters in the liver at individual organ analysis level,

    and to uniquely identify the model of unsampled tissues; (3) constructed

    pharmacodynamic models, based on the molecular mechanism of action,

    for onco-protein depletion and heat shock protein auto-regulation, with the

    drug concentration-time profiles at the site of action predicted by the

    pharmacokinetic model; (4) applied Bayesian inference to characterize the

    kinetic and dynamic properties of the compounds, the physiological and

    anatomical properties of the animal and the molecular properties of the

    proteins involved in the drug response. From the modeling efforts, a number

    of insights having been gained into both the kinetic and dynamic character-

    istics of 17AAG and 17AG and their target molecules, and into the model-

    ing procedures and techniques for PBPKPD model development.

    ACKNOWLEDGMENTS

    This work was supported in part by NIH grant P41-RR01861 made to the

    BMSR (Biomedical Simulations Resource), and by the NCI, CM07106 and

    CA099168.

    APPENDIX

    Appendix A. Individual Organ Models

    Perfusion-Limited Model for Non-Eliminating Organs

    Fig. A1. Perfusion-limited model for non-eliminating organs. VT is the totalvolume of organ T; QTis the bloodflow to the organ; CT is the concentrationof the drug in the organ; RT is the partition coefficient; CArt is the concen-tration of drug in arterial blood; CT,Ven is the venous effluent drug concen-tration (CT,VenGCTRT). In this model, the measured tissue concentration isCT.

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    28/35

    Xu, Eiseman, Egorin, and DArgenio212

    Diffusion-Limited Model for Non-Eliminating Organs

    Fig. A2. Diffusion-limited model for non-eliminating organs. CT,V and VT,Vrepresent the drug concentration and volume for the organ vascular space;CT,EV and VT,EV represent the corresponding terms for the extra-vascularspace;kV,EVand kEV,Vrepresent the drug transport rates between the vascularand extra-vascular spaces; fub is the fraction of unbound drug in the vascularspace. In this model, the measured tissue concentration is CT,EV.

    Perfusion-Limited Model for Eliminating Organs

    Fig. A3. Perfusion-limited model for eliminating organ. CLintT represents theintrinsic clearance of the drug.

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    29/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 213

    Diffusion-Limited Model for Eliminating Organs

    Fig. A4.Diffusion-limited model for eliminating organs.CLintTrepresents the intrin-sic clearance of the drug.

    Table AI. Parameters Measured in Animal Studies

    Measured value (meanJSD)

    Parameter Units PK7 (non-tumor) PK16A (with tumor)

    BW gm 16.80J0.71 19.15J1.28Tumor weight gm 0.26J0.11VLU ml 0.16J0.05 0.16J0.02VBR ml 0.39J0.05 VHT ml 0.12J0.02 0.12J0.01VSP ml 0.06J0.01 0.04J0.01VLI

    a ml 0.67J0.05 0.93J0.10VKI ml 0.24J0.02 0.28J0.02

    VMUb

    ml 6.45 7.35VMisc

    c ml 7.02 8.08

    fub,AAG 0.0209fub,AG 0.0140

    aMean of the values before feeding only.bThe total muscle value was calculated from literature reported muscle frac-

    tional weight WfMUG0.384 (25), using the measured body weight of thestudied animals.

    cVolume of the unsampled tissues was calculated as follows, with literaturereported bone fractional weight WfboneG0.107, blood fractional weightWfbloodG0.076 and bone density equals 1.3:

    VMiscGBWAWiAWfblood BWAWfbone BW

    1C

    Wfbone BW

    1.3

    (istands for each measured organs).

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    30/35

    Xu, Eiseman, Egorin, and DArgenio214

    Table AII. Parameters Estimated in Model Development

    Parameter Units Priora Parameter Units Priora Parameter Units Priora

    QBRb,c

    % CO 3.35J

    1.04 VLU,Ve

    % VT 50J

    11 VMisc,Vh

    % VT 4.0J

    1.6QHT % CO 6.72J2.66 VBR,V % VT 3.0J1.2f VT,EV,V

    i % VT QSP % CO 0.42J0.32 VHT,V % VT 4.0J1.6

    g Rj NIQLI % CO 16.1J2.45 VSP,V % VT 17J1.0 kV,EV

    j hr1 NI

    QKI % CO 11.1J0.05 VLI,V % VT 31J7.0 kEV,Vj hr

    1 NIQMU % CO 18.0J8.85 VKI,V % VT 24J11 CLint

    k mlhr NIQMisc

    d % CO VMU,V % VT 4.0J1.4 fml NI

    aMeanJSD when prior information is available; NI for non-informative prior.bLiterature values from Refs. 23 and 24 were pooled to generate the population mean and SD

    for the organ blood flow QTs.cQTs can be converted to mlhr form by multiplying the values by CO (cardiac output), which

    is calculated as 30.82 (ml

    hr

    gm BW) BW (23). QLUGCO.

    dQMiscwas calculated as the difference between CO and the sum of the measured organ bloodflows.

    eThe organ fractional vascular volume VT,Vs were obtained from reference (25). VT,Vs canbe converted to ml form by multiplying the values by the corresponding values ofVT fromTable AI.

    fNo information was available onVBR,VSD, the highest CV% reported for the VT,Vs was thusadopted.

    gNo information was available. VHT,Vwas assumed to have a mean of 0.04 (same as that ofmuscle), and the highest CV% reported for the VT,Vs was adopted to obtain its SD.

    hNo information was available. Mean and SD were set to be the same as those for heart.iExtra-vascular volume for each organ was calculated as VTAVT,V.j

    Estimated for both 17AAG and 17AG for each of the eight organstissues.kHepatic intrinsic clearance estimated for both 17AAG and 17AG.lFraction of 17AAG converted to 17AG in liver cells.

    Parameter Categories and Parameter Values

    Tables AI and AII list all the parameters for the PBPK model. The

    parameters that were measured in the animal experiments are listed in Table

    AI along with their values. In model development, these parameters were

    fixed at their listed values. Table AII lists parameters that were estimated

    as part of the model development, including parameters for which priormean and standard deviation were available, as well as parameters for which

    no prior information was available (i.e., with non-informative prior).

    Appendix B. Derivation of 17AAG CLintCLs Relationship in the Liver

    Diffusion-Limited Model

    This Appendix presents the derivation of an equation relating the sys-

    temic clearance of 17AAG (CLs) to the hepatic intrinsic clearance (CLint)

    with a diffusion-limited model for hepatic distribution. Consider the simpli-

    fied PBPK model shown in Fig. B1, in which all the systemic organs except

    the lung are lumped into a single effective tissue Other. Assuming linear

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    31/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 215

    Fig. B1. Reduced 17AAG whole-body model for intrinsic clearance deri-vation.

    kinetics and the liver is the only eliminating organ, the five differential equa-

    tions describing this system can be expressed in Laplace domain as follows:

    sVLUCLU(s)GQCVen(s)ACLU(s)RLU (B1)

    sVVCV(s)GQLICLU(s)RLU ACV(s)AkV,EVfubVVCV(s)CkEV,VVEVCEV(s) (B2)

    sVEVCEV(s)GkV,EVfubCV(s)VVAkEV,VCEV(s)VEVACLintCEV(s) (B3)

    sVOCO(s)GQOCLU(s)RLU ACO(s)

    RM (B4)

    sVVenCVen(s)ADoseGQOCO(s)

    ROCQLICV(s)AQCVen(s) (B5)

    Equations (B1)(B5) can be solved for (CLU

    (s))RLU

    . Since

    CLsGDose

    AUC(CArt(t))(B6)

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    32/35

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    33/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 217

    sVOCO,AG (s)GQOCLU,AG(s)RLU,AG ACO,AG(s)

    RO,AG (C3)

    sVVenCVen,AG(s)GQOCO,AG(s)

    RO,AGCQLI

    CAG(s)

    RAGAQtotal CVen,AG(s) (C4)

    Equations (C1)(C4) can be solved for (CLU,AG(s))RLU,AG. Since

    AUC(CArt,AG(t))GAUCCLU,AG(t)RLU,AG Glims0CLU,AG(s)

    RLU,AGG

    CLU,AG (sG0)

    RLU,AG(C5)

    Therefore:

    AUC(CArt,AG(t))GfmCLintAAGAUC(CEV,AAG(t))

    CLintAGRAG(C6)

    AUC(CEV,AAG(t)) can be derived from Eqs. (B2) and (B3) in the 17AAG

    model for the case ofkEV,VG0, and is given as follows:

    AUC(CEV,AAG(t))GQLIVVkV,EV,AAGfubCArt,AAG(t)

    QLICLintAAGCVVkV,EV,AAGfubCLintAAG

    (C7)

    Fig. C1. Reduced 17AG whole-body model for intrinsic clearance deri-vation. All concentration and drug-specific parameters refer to AG exceptwhere noted.

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    34/35

    Xu, Eiseman, Egorin, and DArgenio218

    REFERENCES

    1. C. Erlichman, J. L. Grem, H. I. Scher, D. L. Trump, R. Ramanathan, M. J. Egorin, B.Blaylock, and P. Ivy. Phase I clinical trials of 17-allylamino-17-deoxygeldanamycin (17-AAG) (NSC#330507) sponsored by the national cancer institute, USA(NCI). 1st Inter-national Conference on the HSP90 Chaperone Machine. (2002)

    2. P. N. Munster, M. Srethapakdi, M. M. Moasser, and N. Rosen. Inhibition of heat shockprotein 90 function by ansamycins causes the morphological and functional differentiationof breast cancer cells. Cancer Res. 61: 29452952 (2001).

    3. R. C. Schnur, M. L. Corman, R. J. Gallaschun, B. A. Cooper, M. F. Dee, J. L. Doty,M. L. Muzzi, J. D. Moyer, C. I. DiOrio, E. G. Barbacci, P. E. Miller, A. T. OBrien,M. J. Morin, B. A. Foster, V. A. Pollack, D. M. Savage, D. E. Sloan, L. R. Pustilnik,and M. P. Moyer. Inhibition of the oncogene product p185 erbB-2 in vitro and in vivo bygeldanamycin and dihydrogeldanamycin derivatives. J. Med. Chem. 38: 38063812 (1995).

    4. R. C. Schnur, M. L. Corman, R. J. Gallaschun, B. A. Cooper, M. F. Dee, J. L. Doty,

    M. L. Muzzi, C. I. DiOrio, E. G. Barbacci, P. E. Miller, V. A. Pollack, D. M. Savage,D. E. Sloan, L. R. Pusitilnik, J. D. Mayer, and M. P. Moyer. erbB-2 oncogene inhibitionby geldanamycin derivatives: synthesis, mechanism of action, and structure-activityrelationships.J. Med. Chem. 38:38133820 (1995).

    5. T. W. Schulte and L. M. Neckers. The benzoquinone ansamycin 17-allylamino-17-demethoxygeldanamycin binds to HSP90 and shares important biologic activities with gel-danamycin.Cancer Chemother. Pharmacol. 42:273279 (1998).

    6. J. Page, J. Heath, R. Fulton, E. Yalkowsky, E. Tabibi, J. Tomzszewski, A. Smith, andL. Rodman. Comparison of geldanamycin (NSC-122750) and 17-allylaminogeldanamycin(NSC-330507D) toxicity in rats. Proc. Am. Assoc. Cancer Res. 38:308 (1997).

    7. M. J. Egorin, D. M. Rosen, J. H. Wolff, P. S. Callery, S. M. Musser, and J. L. Eiseman.Metabolism of 17-(allylamino)-17-demethoxygeldanamycin (NSC 330507) by murine and

    human hepatic preparations. Cancer Res. 58: 23852396 (1998).8. M. J. Egorin, E. G. Zuhowski, D. M. Rosen, D. L. Sentz, J. M. Covey, and J. L. Eiseman.

    Plasma pharmacokinetics and tissue distribution of 17-(allylamino)-17-demethoxy-geldanamycin (NSC 330507) in CD2F1 mice1. Cancer Chemother. Pharmacol.47:291302(2001).

    9. W. G. An, R. C. Schnur, L. M. Neckers, and M. V. Blagosklonny. Depletion of p185erbB2,Raf-1 and mutant p53 proteins by geldanamycin derivatives correlates with antiproliferat-ive activity. Cancer Chemother. Pharmacol. 40: 6064 (1997).

    10. L. M. Neckers, T. W. Schulte, and E. Mimnaugh. Geldanamycin as a potential anti-canceragent: its molecular target and biochemical activity. Inest.New Drugs 17:361373 (1999).

    11. C. Chavany, E. Mimnaugh, P. Miller, R. Bitton, P. Nguyen, J. Trepel, L. Whitesell, R.Schnur, J. Moyer, and L. Neckers. p185

    erbB2binds to GRP94 in vivo. Dissociation of

    the p185erbB2GRP94 heterocomplex by benzoquinone ansamycins precedes depletion ofp185erbB2. J. Biol. Chem. 271:49744977 (1996).

    12. E. G. Mimnaugh, C. Chavany, and L. Neckers. Polyubiquitination and proteasomal degra-dation of the p185c-erbB-2 receptor protein-tyrosine kinase induced by geldanamycin. J.Biol. Chem. 271:2279622801 (1996).

    13. C. Schneider, L. Sepp-Lorenzino, E. Nimmesgern, O. Ouerfelli, S. Danishefsky, N. Rosen,and F. U. Hartl. Pharmacologic shifting of a balance between protein refolding and degra-dation mediated by Hsp90. Proc. Natl. Acad. Sci. 93: 1453614541 (1996).

    14. T. W. Schulte, M. V. Blagosklonny, C. Ingui, and L. Neckers. Disruption of the Raf-1-Hsp90 molecular complex results in destabilization of Raf-1 and loss of Raf-1-Ras associ-ation.J. Biol. Chem. 270:2458524588 (1995).

    15. C. E. Stebbins, A. A. Russo, C. Schneider, N. Rosen, F. U. Hartl, and N. P. Pavletich.Crystal structure of an Hsp90-geldanamycin complex: targeting of a protein chaperone byan antitumor agent. Cell89:239250 (1997).

    16. A. Sharma and W. J. Jusko. Characterization of four basic models of indirect pharmaco-dynamic responses. J. Pharmacokinet. Biopharm. 24:611635 (1996).

  • 8/11/2019 Physiologically-Based Pharmacokinetics And

    35/35

    Physiologically-Based PK and Molecular PD in Tumor-Bearing Mice 219

    17. R. I. Morimoto. Cells in stress: transcriptional activation of heat shock genes. Science259:14091410 (1993).

    18. C. Jolly and R. I. Morimoto. Role of the heat shock response and molecular chaperonesin oncogenesis and cell death. J. Natl. Cancer Inst. 92:15641572 (2000).

    19. Y. Shi, D. D. Mosser, and R. I. Morimoto. Molecular chaperones as HSF1-specific tran-scriptional repressors. Genes De. 12: 654666 (1998).

    20. R. I. Morimoto. Regulation of the heat shock transcriptional response: cross talk betweena family of heat shock factors, molecular chaperones, and negative regulators.Genes De.12:37883796 (1998).

    21. D. E. Mager and W. J. Jusko. Pharmacodynamic modeling of time-dependent transductionsystems.Clin. Pharmacol. Ther. 70: 210216 (2001).

    22. D. Z. DArgenio and A. Schumitzky.ADAPT II Users Guide: PharmacokineticPharmac-odynamic Systems Analysis Software. Biomedical Simulations Resource, Los Angeles, 1997.

    23. P. Wang, Z. F. Ba, J. Burkhardt, and I. H. Chaudry. Trauma-hemorrhage and resusci-tation in the mouse: effects on cardiac output and organ blood flow. Am. J. Physiol.264:H11661173 (1993).

    24. R. W. Barbee, B. D. Perry, R. N. Re, and J. P. Murgo. Microsphere and dilution tech-niques for the determination of bloodflows and volumes in conscious mice.Am.J.Physiol.263:R728733 (1992).

    25. R. P. Brown, M. D. Delp, S. L. Lindstedt, L. R. Rhomberg, and R. P. Beliles. Physiologicalparameter values for physiologically based pharmacokinetic models. Toxicol. Ind. Health13: 407484 (1997).

    26. M. J. Mantyla. Regional bloodflow in human tumors. Cancer Res. 39:23042306 (1979).27. R. K. Jain. Determinants of tumor bloodflow: a review.Cancer Res.48:26412658 (1988).28. L. T. Baxter, H. Zhu, D. G. Mackensen, and R. K. Jain. Physiologically based pharmaco-

    kinetic model for specific and nonspecific monoclonal antibodies and fragments in normaltissues and human tumor xenografts in nude mice. Cancer Res. 54:15171528 (1994).

    29. R. K. Jain. Transport of molecules in the tumor interstitium: a review. Cancer Res.47: 30393051 (1987).

    30. W. Xu, E. Mimnaugh, M. F. Rosser, C. Nicchitta, M. Marcu, Y. Yarden, and L. Neckers.Sensitivity of mature P185erbB2 to geldanamycin is conferred by its kinase domain and ismediated by the chaperone protein Hsp90. J. Biol. Chem. 276:37023708 (2001).

    31. P. Miller, C. DiOrio, M. Moyer, R. C. Schnur, A. Bruskin, W. Cullen, and J. D. Moyer.Depletion of the erbB-2 gene product p185 by benzoquinoid ansamycins. Cancer Res.54:27242730 (1994).

    32. G. E. Blakey, I. A. Nestorov, P. A. Arundel, L. J. Aarons, and M Rowland. QualitativeStructure-Pharmacokinetics Relationships: I. Development of a Whole-Body Physiologi-cally Based Model to Characterize Changes in Pharmacokinetics Across a HomologousSeries of Barbuturates in the Rat. J. Pharmacokin. Biopharm. 25:277213 (1997).

    33. A. Gelman, F. Y. Bois, and J. Jiang. Physiological pharmacokinetic analysis using popu-lation modeling and informative prior distributions. J. Am. Stat. Assoc. 91:14001412(1996).

    34. F. Jonsson and G. Johanson. Physiologically based modeling of the inhalation kinetics ofstyrene in humans using a Bayesian population approach. Toxicol. Appl. Pharmacol.179:3549 (2002).