a physiologically-based pharmacokinetic model for capreomycin

Upload: sukma-effendy

Post on 03-Apr-2018

220 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    1/59

    1

    Title: A Physiologically-Based Pharmacokinetic Model for Capreomycin1

    Running title: A PBPK Model for Capreomycin2

    Authors: B. Reisfeld*,1

    , C.P. Metzler1,

    , M.A. Lyons1, A.N. Mayeno

    1, E.J. Brooks

    2, M.A.3

    DeGroote24

    Key words: Mycobacterium tuberculosis, therapeutics, pharmacokinetics, computational5

    modeling, pharmacodynamics, pbpk, mouse, human, anti-tuberculosis agents.6

    Author affiliations:7

    1Department of Chemical and Biological Engineering; Colorado State University, Fort8

    Collins, CO 805239

    2Department of Microbiology, Immunology, and Pathology; Colorado State University, Fort10

    Collins, CO 8052311

    12

    13

    * Brad Reisfeld; Department of Chemical and Biological Engineering; Colorado State University; 1370 Campus

    Delivery; Fort Collins, CO 80523-1370; voice: 970-491-1019, fax: 970-491-7369, email:

    [email protected]

    current address Vertex Pharmaceuticals, Cambridge, MA

    Copyright 2011, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved.Antimicrob. Agents Chemother. doi:10.1128/AAC.05180-11AAC Accepts, published online ahead of print on 5 December 2011

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    2/59

    2

    Abstract14

    The emergence of multidrug-resistant tuberculosis (MDR-TB) has led to a renewed interest in15

    the use of second-line antibiotic agents. Unfortunately, there is currently a dearth of16

    information, data, and computational models that can be used to help design rational regimens17

    for administration of these drugs. To help fill this knowledge gap, an exploratory18

    physiologically-based pharmacokinetic (PBPK) model, supported by targeted experimental19

    data, was developed to predict the absorption, distribution, metabolism, and excretion20

    (ADME) of the second-line agent capreomycin, a cyclic peptide antibiotic often grouped with21

    the aminoglycoside antibiotics. To account for inter-individual variability, Bayesian inference22

    and Monte Carlo methods were used for model calibration, validation, and testing. Along with23

    the predictive PBPK model, the first for an anti-tuberculosis agent, this study has provided24

    estimates of various key pharmacokinetic parameter distributions and has supported a25

    hypothesized mechanism for capreomycin transport into the kidney.26

    27

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    3/59

    3

    Introduction28

    An estimated 500,000 cases of multi-drug resistant tuberculosis (MDR-TB) emerge each year29

    with 150,000 deaths (50). MDR-TB refers to strains that are resistant to at least isoniazid and30

    rifampicin. The World Health Organization (WHO) guidelines for treatment of MDR-TB31

    include regimens containing Group 2 injectable agents (51). One such agent is capreomycin32

    (CAP), a commonly used second line injectable drug with activity against many MDR-TB33

    strains. It is generally reserved for patients who have had prior exposure to or whose34

    isolates have documented resistance to kanamycin and streptomycin (51). Moreover, CAP35

    has unique effectiveness against both the dormant and active forms of tuberculosis (25).36

    However, the drug is nephrotoxic and ototoxic, especially in patients with renal impairment or37

    in geriatric patients (26).38

    CAP is a polypeptide antibiotic composed of four molecular analogs, IA, IIA, IB, and IIB. Its39

    mode of action, though not fully understood, involves ribosomal inhibition of protein40

    synthesis (22). Studies suggest that CAP binds to, and inhibits the function of, the 16S rRNA41

    molecule of the M. tuberculosis 30S ribosomal subunit, as supported by up-regulation of a42

    methyltransferase gene and the 16S rRNA processing protein gene (20). Due to similar43

    nomenclature, side effects, and mode of action, CAP is often compared to, and grouped with,44

    aminoglycoside antibiotics (AGAs) (19, 22, 24), despite its structural distinction.45

    CAP and AGAs are nephrotoxic (26, 38), with some of the administered dose being retained46

    in the epithelial cells of the kidney proximal tubules. The accumulation of AGAs is notable as47

    the concentration in the kidney is much higher than that in the serum (41). In the proximal48

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    4/59

    4

    tubule cells, AGAs must enter via apical membrane binding and endocytosis because the49

    charged drug molecule cannot freely cross the cellular membrane. Megalin, a glycoprotein50

    expressed in some specialized epithelial cells including in the renal tubule and inner ear51

    epithelium, is the proposed endocytic receptor for such drugs (13, 41, 49). As noted earlier,52

    CAP is known to exert both renal and ototoxicity, which supports the likelihood that megalin53

    is responsible for uptake (38). As further evidence that megalin is an important factor in AGA54

    uptake, a study comparing normal and genetically megalin-deficient mice was conducted by55

    Schmitz et al. (48). In these studies, after exposure to gentamicin, wild-type mice had56

    significant drug accumulation in the kidneys, whereas megalin-deficient mice did not.57

    Despite its long history as an antibiotic, there is limited experimental information and few58

    pharmacokinetic models available for the disposition of CAP. In the 1960s, Black et al.59

    measured the pharmacokinetics of CAP in humans (3, 4) and derived peak serum60

    concentrations and urinary excretion levels. Lee et al. (34) measured early time (up to two61

    hours) serum levels of two different formulations of CAP in rats and looked at the impact of62

    impurities on safety. In the present context, the most directly relevant experimental study to63

    date is that of Le Conte et al. (33), who measured the concentrations of free and liposomal64

    CAP in the blood, spleen, kidney, and lung of normal mice at various points (0.25, 0.5, 0.75,65

    1, 2, 4, and 6 hours) after administration. Notably, the concentration of CAP and the area66

    under the curve (AUC) in the kidney were found to be much higher than that in all the other67

    measured tissues at all time points. These investigators did not measure concentrations at later68

    time points so that peak kidney concentrations, and decays from these concentrations, were69

    not noted.70

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    5/59

    5

    Especially for second-line agents, where side effects and toxicity are inherent concerns, it is71

    critical to develop information and models that allow the examination of drug levels in72

    specific tissue types, such as the kidney. The classical data-driven pharmacokinetic model73

    assumes the body to be one homogenous, well-mixed vessel. To examine chemical74

    distribution in specific tissues, a more sophisticated approach is needed. Using75

    physiologically-based pharmacokinetic (PBPK) modeling, the body is divided into several76

    physiologically-representative compartments organs, blood, tissues with a mass-balance77

    for each compartment. With this approach, dose extrapolation, different routes of dosing, and78

    animal-to-animal extrapolation may all be performed by changing relevant physiological and79

    biochemical properties and by including appropriate allometric scaling laws (30, 31).80

    Although traditionally used for environmental toxicants, PBPK models are increasingly used81

    for the prediction of ADME for various drugs (45-47), including antibiotics (9, 10, 16, 32). A82

    relatively recent advance in PBPK modeling has been the incorporation of approaches for83

    accounting for inter-individual variability in anatomy, physiology, biochemistry, and84

    chemical exposure (1, 6, 8, 12, 36, 39). Among other things, these approaches allow a85

    rigorous incorporation of uncertainties, as well as predictions of chemical ADME in86

    susceptible subpopulations.87

    Overall, there is a knowledge gap in data and methods for predicting the ADME of second-88

    line agents for the treatment of MDR-TB, and until totally new regimens are approved,89

    optimized use of second-line drugs available to clinicians is a current research priority (42).90

    The present exploratory study is meant to help fill the knowledge gap by acquiring tissue91

    pharmacokinetic data and developing a PBPK model for CAP disposition. Although PK/PD92

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    6/59

    6

    models have been developed (23), to our knowledge, this is the first published PBPK model93

    for an anti-tuberculosis drug.94

    95

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    7/59

    7

    Materials and Methods96

    Experimental Study97

    The aim of the laboratory portion of this study was to determine the tissue and blood levels of98

    CAP in mice at specified time points following subcutaneous injection.99

    Mice. Female, six to eight week old C57BL/6 female mice were purchased from Jackson100

    Laboratories (Bar Harbor, ME) and were housed in the Painter Center at Colorado State101

    University. All experiments were approved by the Institutional Animal Care and Use102

    Committee. Mice were randomly assigned to three groups: low-dose (N=24), high dose103

    (N=24), and control (N=4).104

    Drug. Capreomycin sulfate was purchased from Sigma Chemical Co. (St Louis, MO).105

    Capreomycin solution was prepared by dissolving 92.5 mg (low-dose solution) and 231 mg106

    (high-dose solution) of capreomycin sulfate in 10 ml phosphate buffer saline (1X, pH 7.4)107

    (Fisher Scientific, Pittsburgh, PA). The vehicle solution was 1X PBS.108

    Pharmacokinetic studies. The dosing regimen was determined through a pilot study in mice109

    (TB Pharmacokinetic Laboratory of National Jewish Medical and Research Center, Dr. C.110

    Peloquin), in which CAP plasma levels were measured following a single dose.111

    Concentrations were adjusted to span doses that match human bioequivalence measures [Cmax112

    (maximum plasma concentration), t1/2 (half life)], leading to recommended dosing levels of113

    100 mg/kg and 250 mg/kg for the present study. At time zero, the drug or control solution was114

    administered subcutaneously to the mice. All mice received an injection of 0.2 ml of the115

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    8/59

    8

    relevant solution. This corresponded to 100 mg/kg, 250 mg/kg, and 0 mg/kg capreomycin116

    sulfate for mice in the low-dose, high-dose, and control groups, respectively. At each of the117

    time points (0.5, 1, 2, 6, and 20 h), four mice from both the low-dose and high-dose groups118

    were sacrificed via CO2 euthanasia followed by cervical dislocation. All of the mice in the119

    control group were sacrificed at the 1 hr time point. Immediately following sacrifice, blood120

    was collected by cardiac puncture, placed in a serum vial, put on ice for one hour after121

    collection, and spun down to collect the serum; and the kidneys, lungs, spleen, and liver were122

    harvested, weighed, and then flash frozen in cryovials at -80 C.123

    Capreomycin analyses. Capreomycin was quantified by LC/MS/MS (vide infra). The tissues124

    were prepared for analysis by adding water to give 100 mg tissue per ml, followed by125

    sonication. Sonication was performed in small bursts while the tissue remained on ice in order126

    to mitigate the effects of heat generated by the sonicator. In some cases, the larger organs127

    were subdivided. The organs were prepared as follows in order to improve homogeneity:128

    spleen (used in entirety due to the small size); kidneys (one kidney was used from each129

    mouse; it was assumed that there was no preferential clearance in one kidney or the other);130

    lung (portions of each lobe of the lung were removed and homogenized together); liver (after131

    removal of the gallbladder, the liver was diced into small pieces, mixed, and a random sample132

    of pieces was taken). Following sonication, 200 l of the tissue homogenate or 100 l of133

    serum were added to a microcentrifuge tube containing 10 l of capreomycin standard or 10134

    l of 50% acetonitrile. The mixtures were vortexed briefly. To the tissue homogenate, 150 l135

    of methanol + 1% formic acid was added, while 300 l of methanol + 1% formic acid was136

    added to the serum samples to induce protein precipitation. Each sample was then vortexed137

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    9/59

    9

    for 10 minutes, followed by centrifugation for 10 minutes to remove cell debris and protein138

    from the liquid portion. Supernatant was then transferred to plastic autosampler vials for139

    analysis. Plastic vials were used as capreomycin exhibits binding to glass (37). HPLC was140

    performed using a Waters Atlantis HILIC Silica, 5 m, 4.6 x 50 mm column with a141

    Phenomenex C18 guard cartridge. Standard curves for CAP (from 500 ng/mL to 50 g/mL)in142

    matrix (control serum or tissue homogenate) were generated, by adding capreomycin sulfate143

    standards prepared in 50% acetonitrile: 50% H2O with 0.1% acetic acid. The PK results for144

    CAP in the paper refer to capreomycin free base (the base form of the drug rather than the salt145

    form; "free" does not refer to unbound drug). Lower limits of quantitation (LLOQ) for146

    capreomycin in each tissue were determined to be as follows: kidney (50 ng/mg), serum (1147

    ng/mg), liver (10 ng/mg), lung (10 ng/mg), spleen (10 ng/mg). Further LC-MS/MS method148

    details are provided in the Appendix.149

    The use of tissue homogenates is appropriate for use in the PBPK modeling approach used150

    here because compartments are assumed to be well mixed and homogenous with respect to151

    drug concentration. This is in contrast to studies of drug activity and efficacy, in which results152

    derived from these types of samples could be misleading or erroneous (40).153

    PBPK Modeling Study154

    The aim of this study was to develop a PBPK model, calibrated and validated with155

    experimental data, to predict the time-dependent ADME of capreomycin in mice.156

    Model structure and equations. The structural model for capreomycin was based on a157

    generic whole body PBPK model (28) with subsequent modifications of the kidney158

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    10/59

    10

    compartment. This model comprises compartments for the lung, skin, fat, muscle, kidney,159

    brain, heart, bone, liver, spleen, gut, arterial blood, venous blood, and a carcass compartment160

    that contains all tissues not accounted for in other compartments. This flexible structure161

    allowed the prediction of drug concentrations in tissues relevant for examinations of toxicity162

    and pharmacological efficacy relevant to TB, as well as the possibility of assessing the impact163

    of different dosing routes. Some of the compartments are not directly relevant to the efficacy164

    and toxicity of CAP (e.g., heart and skin); however, they are included here because they165

    provide additional detail for scaling to other species and are often germane to studies in which166

    drug concentrations are measured in these tissues. The connection between compartments and167

    pathway of blood flow is shown in Figure 1.168

    Associated with each compartment is a mass balance for the drug. In all compartments, we169

    assumed flow-limited mass transfer, viz., the blood entering a tissue is quickly in equilibrium170

    with the tissue. The full set of governing equations for the model is given in the Appendix171

    (eqn A2 eqn A13).172

    Due to similarities in certain physicochemical and pharmacodynamic properties between173

    AGAs and capreomycin, we assumed that the mechanisms for capreomycin ADME are174

    related to those of other AGAs (52). As noted earlier, transport of AGAs is known to be175

    dependent on megalin endocytosis followed by lysosomal sequestration (41, 48, 49). To176

    account for the sequestration of capreomycin in the present model, we represented the kidney177

    by a system comprising two linked compartments: a shallow (S) and deep (D) compartment,178

    both of which were assumed to be well mixed (Figure 2). Although the correlation is not179

    exact, anatomically, the deep compartment would approximate the cells along the walls of the180

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    11/59

    11

    kidney tubules, while the shallow compartment would represent the remainder of the kidney181

    tissue.182

    The absorption for the kidney compartment was modeled using an equation describing183

    saturable kinetics, as seen in the literature for similar applications (2, 14, 18). For example,184

    Giuliano et al. (21) demonstrated that gentamicin and netilmicin accumulation in the kidney185

    could be described using a Michaelis-Menten (saturable kinetics) equation (eqn 1), which is186

    traditionally used to describe an enzyme-substrate reaction. For the present model, megalin187

    corresponds to the enzyme, v0 is the renal accumulation rate, vmax is the maximum renal188

    accumulation rate, [S] is the capreomycin concentration (in the shallow compartment), andKM189

    is the Michaelis constant.190

    ][

    ][max0

    SK

    Svv

    M +

    = (eqn 1)191

    Here, although capreomycin consists of 4 distinct molecules, this mixture was treated as a192

    lumped single chemical entity in this model.193

    Model parameters and accounting for variability. The following experimental inputs were194

    used directly in the model: body weight of the mice, and lung, liver, kidney, and spleen195

    weights. For the remaining organs, and for approximate blood flow through each organ,196

    values from Brown et al. (11) and Davies and Morris (17) were used. The tissue density for197

    all of the organ systems was assumed to be equal to that of water (11). With the exception of198

    that for the lung, all of the tissue:blood partition coefficients were set equal to one. The199

    lung:blood partition coefficient was set equal to two based on the observation that200

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    12/59

    12

    aminoglycosides are transported into the lungs by endocytosis (27), augmenting the baseline201

    thermodynamic partitioning. A lung:blood partition coefficient greater than one is consistent202

    with the observation that aminoglycosides are eliminated more slowly from the lung than203

    from the serum (15, 35). The list of model parameters used in the simulations is summarized204

    in Table A3 of the Appendix.205

    Owing to the unique nature of the model structure, and the lack of literature values for206

    physiological transport values for capreomycin, the model parameters listed in Table 1 were207

    determined using calibration simulations (vide infra).208

    Solution Method. Simulations were performed in two steps. First, a series of calibration209

    simulations were conducted using a Bayesian approach (7, 8, 35) to determine the unknown210

    parameters for the model. This approach rests on a relationship among probability211

    distributions involving unknown parameters and available data y given by Bayes theorem212

    ( ) ( ) ( )| |p y p y p . Here, the posterior distribution ( )|p y is obtained as the product213

    of the prior distribution

    ( )p and the likelihood

    ( )|p y . The model calibration involves the214

    identification of the parameters (Table 1) with those that are needed to complete the215

    specification of the PBPK model (Table A3). The data y corresponds to experimentally216

    obtained concentration-time profiles resulting from a known dose. The likelihood contains the217

    underlying PBPK model (eqn A2 - eqn A13) calculated with the parameters . Combining the218

    likelihood of the data with prior parameter distributions results in the posterior probability219

    distribution for the parameters conditioned on the data. To verify the robustness of the220

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    13/59

    13

    posterior estimates, several types of priors were used, including truncated-normal and uniform221

    distributions.222

    Following the calibration step, Monte Carlo (MC) simulations were performed, in which the223

    PBPK model equation system was solved using values sampled from the parameter224

    distributions found in the calibration step. In a typical MC simulation, 1000 model runs were225

    conducted, each containing a set of parameters randomly sampled from the distributions226

    found earlier. The result of these runs was families of time-dependent concentration profiles227

    of capreomycin in each model compartment.228

    All simulations were performed using GNU MCSim (5) (v. 5.2), a simulation package that is229

    useful in solving statistical and differential equation systems, performing Monte Carlo230

    stochastic simulations, and conducting Bayesian inference through Markov chain Monte231

    Carlo (MCMC) simulations. For the MCMC simulations, chains of length 10,000 were used232

    and convergence was assessed using visual inspection. All calculations were performed on a233

    PC workstation with a 2.8 GHz dual core Intel Pentium processor and 8 GB of RAM running234

    the Windows XP operating system.235

    236

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    14/59

    14

    Results237

    Experimental Study238

    The measured low-dose and high-dose concentrations of capreomycin in the organs of interest239

    are shown in Figure 3 (a-e). For all tissues except the kidney, the peak concentration of240

    capreomycin occurred at some time prior to 0.5 hours. Within the resolution of the data, the241

    concentrations were found to decay exponentially over time, consistent with a first-order242

    elimination process, with both low- and high-dose data following similar elimination kinetics.243

    The capreomycin was eliminated relatively rapidly, with concentrations falling below the244

    LLOQ in one to two hours for all tissues, besides the kidney.245

    For the kidney, peak concentrations for both low- and high-doses occurred around three hours246

    after administration and then decayed relatively slowly. Concentrations of capreomycin in the247

    kidney were still well above the LLOQ after 20 hours. The only other study in which tissue248

    concentrations for capreomycin were measured (33) focused on time points up to six hours,249

    and decay in concentration from the peak value was not seen. The kidney concentrations at all250

    time points are several times those in any other compartment, consistent with the transporter-251

    enhanced uptake mechanism and supporting data described earlier.252

    253

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    15/59

    15

    Modeling Study254

    Estimated Parameter Values255

    Based on the calibration simulations described earlier, probability distributions of the256

    unknown parameters were determined. Table 2 summarizes the means and standard257

    deviations for the values in these distributions, while Figure 4 shows the distributions for each258

    of these parameters. These parameters provide rough estimates for various transport259

    properties for CAP that are difficult to measure directly, including the kinetics of260

    accumulation in the kidney, the rate of renal clearance, and the rate of hepatic clearance.261

    However, as mentioned earlier, it is difficult to compare these values to others in the literature262

    because of the paucity of previous detailed pharmacokinetic and transport studies for263

    capreomycin.264

    265

    Comparisons Between Experimental Data and Model Predictions266

    The MC simulations produce a family of concentration profiles in each organ compartment267

    based on the animal and organ weights and sampling the distributions of model parameters268

    depicted in Figure 4. The fifth and ninety-fifth percentile curves for the low-dose MC269

    simulation results, along with the corresponding experimental data, are shown in Figure 6.270

    Similar comparisons for the high-dose case are shown in Figure 7.271

    For the low-dose case, agreement between simulations and experiments was generally good272

    for all organs, with the experimental points falling within the range of simulations in most273

    cases. The largest discrepancy appears in the peak serum concentrations for capreomycin. Part274

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    16/59

    16

    of this discrepancy arises because very early time data (

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    17/59

    17

    Discussion297

    Although a few studies have investigated the pharmacokinetics of capreomycin in blood (3, 4,298

    29, 34, 43, 44), only one study to date (33) has examined tissue concentrations of299

    capreomycin over time, and this study focused on relatively early-time data which do not300

    capture important late clearance events, especially for the kidney. To our knowledge no PBPK301

    models have been published for any anti-tuberculosis drugs. Moreover, even existing PBPK302

    models for antibiotics (9, 10, 16, 32) have not included considerations of inter-individual303

    variability, an important consideration in the interpretation of ADME predictions for these304

    drugs. The PBPK model for capreomycin disposition in mice developed in this study begins305

    to close this knowledge gap. It provides predictions that are generally in good agreement with306

    results from a corresponding experimental study and, through a Bayesian model calibration,307

    has provided distributions for several parameters related to capreomycin transport.308

    The approach used here has important advantages relative to classical PK or population-PK309

    approaches. Since classical models are not based upon the true anatomy, physiology, and310

    biochemistry of the species of interest, they cannot, in general, be used to generate reliable311

    predictions outside the range of doses, dose routes, and species used in the studies upon which312

    they were based. Such extrapolations, which are essential in estimating the dose-response of313

    chemicals, can be performed more accurately using PBPK modeling approaches.314

    Despite these advantages, there are limitations to the current study. Because of the relatively315

    small sample size, the model and results should be viewed as exploratory in nature. In316

    addition, very early time data were not collected in this study, making characterization of the317

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    18/59

    18

    peak concentration difficult and adding uncertainty to the analysis. Future studies could make318

    use of optimal sampling theory to design appropriate studies to capture these events in an319

    efficient manner.320

    Current work is focused on improving the model, principally through the inclusion of321

    additional data; the Bayesian approach used here provides for a straightforward means of322

    incorporating these data as they become available. Also, we are assessing the feasibility of323

    extrapolating the model to humans using appropriate physiological parameters from Brown et324

    al. (11) and Davies and Morris (17) and calibrating and validating using the human325

    pharmacokinetic data from Black et al. (3, 4). In addition, because of similarities between326

    CAP and AGAs as described earlier, the model is being extended to AGAs. Longer-term aims327

    are to use this model, along with appropriate pharmacodynamic and toxicity data, as part of a328

    predictive framework to help optimize drug regimens to treat MDR-TB.329

    330

    331

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    19/59

    19

    Acknowledgements332

    The authors thank Janet Gilliland for assistance in the animal studies, Dr. Ryan Hansen for333

    conducting the analytical chemistry analyses, and Dr. Daniel Gustafson for providing334

    resources for pharmacokinetic analyses and for helpful comments about capreomycin335

    pharmacokinetics. The authors also thank the anonymous reviewers for their valuable336

    comments and suggestions to improve the quality of the paper. We gratefully acknowledge337

    funding for this project from a Capacity Building Grant awarded by the Infectious Disease338

    Supercluster at Colorado State University.339

    340

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    20/59

    20

    Appendix341

    Analysis of Capreomycin IA and IB from Mouse Tissues by LC/MS/MS342

    Instrument: Shimadzu LC-20AD High Performance Liquid Chromatograph system343

    (Shimadzu Corporation, Kyoto, Japan). Column: Waters Atlantis HILIC Silica 5 m, 4.6 x344

    50 mm (part#186002028), protected by a Phenomenex C18 Guard Cartridge (and filter frits).345

    Vials: plastic. Injection Volume: 75 l. Loop: 100 l. Flow Rate: 800 l/min. Run Time: 4346

    min. Column Oven: RT.347

    LC Gradient Conditions:348

    Time (min) % Organic Phase % Aqueous Phase

    0.50 2% ACN 98% 0.1% Formic Acid in H2O

    2.5 90% ACN 10% 0.1% Formic Acid in H2O

    3.0 90% ACN 10% 0.1% Formic Acid in H2O

    3.2 2% ACN 98% 0.1% Formic Acid in H2O

    4.0 2% ACN 98% 0.1% Formic Acid in H2O

    349

    MS Conditions350

    Instrument: MDS Sciex 3200 Q-TRAP triple quadrupole mass spectrometer (Applied351

    Biosystems, Foster City, CA) with a TurboIonSpray source. MRM Positive Ion Mode.352

    Scan/Dwell Time: 300 msec. Transitions Monitored:353

    Capreomycin IA (669.30 507.00 amu) DP: 71.0; CE: 45; CEP: 30; CXP: 6354

    Capreomycin IB (653.3 491.3 amu) DP: 55.0; CE: 48; CEP: 33.8; CXP: 4355

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    21/59

    21

    Curtain (CUR) Gas: 20. Collision (CAD) Gas: 12 (High). Ion Spray Voltage: 2500. Source356

    Temperature: 550C. Ion Source Gas 1: 35. Ion Source Gas 2: 30. Entrance Potential (EP):357

    10. Ihe: on.358

    For analysis, both transitions were integrated as one. As capreomycin IA and IB are, by far,359

    the major constituents, only these two components were quantified. The weight of sulfate was360

    taken into account for quantitation, and the PK results for CAP in this paper refer to361

    capreomycin free base (the base form of the drug rather than the salt form).362

    363

    Governing Equations for PBPK Model364

    Although the model structure is applicable for both intravenous and subcutaneous dose, in this365

    section, we focus solely on the subcutaneous dose. Consistent with the structural model366

    (Figure 1), for all organs except the lung, blood, kidney, and liver, a capreomycin mass367

    balance may be written as368

    ( )organ organorgan

    organ

    d M CQ CAdt P

    =

    , (eqn A2)369

    where Morgan is the drug mass in the organ, Qorgan is blood flow to organ, CA is the drug370

    concentration in the arterial blood flow, Corgan is the drug concentration in the organ, and371

    Porgan is the tissue-blood partition coefficient. Organ flows are fractions of the cardiac output,372

    QC, which is allometrically-scaled to body weight (QCBW0.75

    ) (11, 17). The mass of drug373

    in the lung,MLU, is governed by374

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    22/59

    22

    ( )d MLU CLU QLU CA

    dt PLU

    =

    . (eqn A3)375

    A mass balance on the venous blood, considering the drug dose, is376

    ( )( ) organorgans organ

    organ

    C d MSC d MVQ QLU CV

    dt P dt

    =

    , (eqn A4)377

    whereMSCis the mass of drug in the subcutaneous compartment, given by378

    ( )_

    d MSC SC Decay MSC

    dt= . (eqn A5)379

    Here SC_Decay is the rate of drug movement from the subcutaneous compartment into the380

    venous blood. On the arterial side, the governing equation takes the following form:381

    ( )d MA CLU QLU CA

    dt PLU

    =

    . (eqn 6)382

    Based on the conceptual model for the kidney (Figure 2), the following series of equations383

    may be formulated:384

    ( )d MKE CLR CVKS

    dt= (eqn A7)385

    max( )

    m

    V CVKS d MKA

    dt K CVKS

    =

    +

    (eqn A8)386

    ( )d MKDECLRD CVKD

    dt= (eqn A9)387

    ( ) ( ) ( ) ( )( )

    d MKS d MKA d MKE d MKDE QK CA CVKS

    dt dt dt dt = + (eqn A10)388

    ( ) ( ) ( )d MKD d MKA d MKDE

    dt dt dt = (eqn A11)389

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    23/59

    23

    ( ) ( ) ( )d MK d MKS d MKD

    dt dt dt

    = + (eqn A12)390

    Here,MKEis the mass of drug excreted from the kidney,MKDEis the mass of drug excreted391

    from the deep compartment and returned to blood flow, MKA is the mass of drug392

    accumulating in the deep compartment of the kidney, MKSis the mass of drug in the shallow393

    compartment, CLR is the rate of capreomycin clearance from kidney blood flow, CLRD is the394

    rate of capreomycin clearance from the deep kidney compartment, CVKSis the capreomycin395

    concentration in the well-mixed blood in the kidney that is then mixed with the venous blood,396

    CVKD is the capreomycin concentration in the well-mixed deep kidney compartment, and397

    Vmax andKm are Michaelis-Menten parameters for accumulation.398

    Finally, for the liver compartment, we have399

    ( )d MLQLA CA QS CVS QG CVG QL CVL CLH CVL

    dt= + + , (eqn A13)400

    where CVL is the concentration of drug in the liver, and CLH is the hepatic clearance rate401

    (CLH= CLHCBW).402

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    24/59

    24

    403

    404

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    25/59

    25

    References405

    1. Allen, B. C., C. E. Hack, and H. J. Clewell. 2007. Use of Markov chain Monte Carlo406

    analysis with a physiologically-based pharmacokinetic model of methylmercury to407

    estimate exposures in US women of childbearing age. Risk Analysis 27:947-959.408

    2. Alonso, I. G., J. M. Lanao, M. C. Saez, A. A. Dominguez-Gil, and A. Dominguez-409

    Gil. 1987. Non-linear tissue binding of amikacin in rats: the effect of renal410

    impairment. Eur J Drug Metab Pharmacokinet 12:193-201.411

    3. Black, H. R., R. S. Griffith, and J. F. Brickler. 1963. Preliminary Laboratory412

    Studies with Capreomycin. Antimicrob Agents Chemother (Bethesda) 161:522-9.413

    4. Black, H. R., R. S. Griffith, and A. M. Peabody. 1966. Absorption, excretion and414

    metabolism of capreomycin in normal and diseased states. Ann N Y Acad Sci415135:974-82.416

    5. Bois, F. February 24, 2009 2009, posting date. MCSim manual, version 5.3.0. Free417

    Software Foundation, Inc. [Online.]418

    6. Bois, F. Y. 1999. Analysis of PBPK models for risk characterization. Uncertainty in419

    the Risk Assessment of Environmental and Occupational Hazards 895:317-337.420

    7. Bois, F. Y., A. Gelman, J. Jiang, D. R. Maszle, L. Zeise, and G. Alexeef. 1996.421

    Population toxicokinetics of tetrachloroethylene. Arch Toxicol 70:347-55.422

    8. Bois, F. Y., M. Jamei, and H. J. Clewell. 2010. PBPK modelling of inter-individual423

    variability in the pharmacokinetics of environmental chemicals. Toxicology 278:256-424

    267.425

    9. Brightman, F. A., D. E. Leahy, G. E. Searle, and S. Thomas. 2006. Application of a426

    generic physiologically based pharmacokinetic model to the estimation of xenobiotic427levels in rat plasma. Drug Metab Dispos 34:84-93.428

    10. Brocklebank, J. R., R. Namdari, and F. C. Law. 1997. An oxytetracycline residue429

    depletion study to assess the physiologically based pharmokinetic (PBPK) model in430

    farmed Atlantic salmon. Can Vet J 38:645-6.431

    11. Brown, R. P., M. D. Delp, S. L. Lindstedt, L. R. Rhomberg, and R. P. Beliles.432

    1997. Physiological parameter values for physiologically based pharmacokinetic433

    models. Toxicol Ind Health 13:407-84.434

    12. Chiu, W. A., M. S. Okino, and M. V. Evans. 2009. Characterizing uncertainty and435

    population variability in the toxicokinetics of trichloroethylene and metabolites in436

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    26/59

    26

    mice, rats, and humans using an updated database, physiologically based437

    pharmacokinetic (PBPK) model, and Bayesian approach. Toxicology and Applied438

    Pharmacology 241:36-60.439

    13. Christensen, E. I., and H. Birn. 2002. Megalin and cubilin: multifunctional440

    endocytic receptors. Nat Rev Mol Cell Biol 3:256-66.441

    14. Clewell, R. A., and H. J. Clewell, 3rd. 2008. Development and specification of442

    physiologically based pharmacokinetic models for use in risk assessment. Regul443

    Toxicol Pharmacol 50:129-43.444

    15. Craig, W. A., J. Redington, and S. C. Ebert. 1991. Pharmacodynamics of amikacin445

    in vitro and in mouse thigh and lung infections. J Antimicrob Chemother 27 Suppl446

    C:29-40.447

    16. Craigmill, A. L. 2003. A physiologically based pharmacokinetic model for448 oxytetracycline residues in sheep. J Vet Pharmacol Ther26:55-63.449

    17. Davies, B., and T. Morris. 1993. Physiological parameters in laboratory animals and450

    humans. Pharm Res 10:1093-5.451

    18. DeWoskin, R. S., and C. M. Thompson. 2008. Renal clearance parameters for PBPK452

    model analysis of early lifestage differences in the disposition of environmental453

    toxicants. Regul Toxicol Pharmacol 51:66-86.454

    19. Fiegel, J., L. Garcia-Contreras, M. Thomas, J. VerBerkmoes, K. Elbert, A.455

    Hickey, and D. Edwards. 2008. Preparation and in vivo evaluation of a dry powder456

    for inhalation of capreomycin. Pharm Res 25:805-11.457

    20. Fu, L. M., and T. M. Shinnick. 2007. Genome-wide exploration of the drug action of458capreomycin on Mycobacterium tuberculosis using Affymetrix oligonucleotide459

    GeneChips. J Infect 54:277-84.460

    21. Giuliano, R. A., G. A. Verpooten, L. Verbist, R. P. Wedeen, and M. E. De Broe. 461

    1986. In vivo uptake kinetics of aminoglycosides in the kidney cortex of rats. J462

    Pharmacol Exp Ther236:470-5.463

    22. Global Alliance for TB Drug Development 2008. Capreomycin. Tuberculosis464

    88:89-91.465

    23. Goutelle, S., L. Bourguignon, R. W. Jelliffe, J. E. Conte, Jr., and P. Maire. 466

    Mathematical modeling of pulmonary tuberculosis therapy: Insights from a prototype467

    model with rifampin. J Theor Biol 282:80-92.468

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    27/59

    27

    24. Hall, R. G., R. D. Leff, and T. Gumbo. 2009. Treatment of active pulmonary469

    tuberculosis in adults: current standards and recent advances. Insights from the Society470

    of Infectious Diseases Pharmacists. Pharmacotherapy 29:1468-81.471

    25. Heifets, L., J. Simon, and V. Pham. 2005. Capreomycin is active against non-472

    replicating M. tuberculosis. Ann Clin Microbiol Antimicrob 4:6.473

    26. Holdiness, M. R. 1984. Clinical pharmacokinetics of the antituberculosis drugs. Clin474

    Pharmacokinet 9:511-44.475

    27. Honeybourne, D. 1994. Antibiotic Penetration into Lung Tissues. Thorax 49:104-476

    106.477

    28. Jones, H. M., N. Parrott, K. Jorga, and T. Lave. 2006. A novel strategy for478

    physiologically based predictions of human pharmacokinetics. Clin Pharmacokinet479

    45:511-42.480

    29. Kanazawa, Y., and T. Kuramata. 1964. [Measurement of Capreomycin in Body481

    Fluids]. J Antibiot B 17:190-2.482

    30. Kirman, C. R., L. M. Sweeney, M. E. Meek, and M. L. Gargas. 2003. Assessing483

    the dose-dependency of allometric scaling performance using physiologically based484

    pharmacokinetic modeling. Regul Toxicol Pharmacol 38:345-67.485

    31. Knaak, J. B., M. A. al-Bayati, and O. G. Raabe. 1995. Development of partition486

    coefficients, Vmax and Km values, and allometric relationships. Toxicol Lett 79:87-487

    98.488

    32. Laplanche, R., G. M. Meno-Tetang, and R. Kawai. 2007. Physiologically based489

    pharmacokinetic (PBPK) modeling of everolimus (RAD001) in rats involving non-490linear tissue uptake. J Pharmacokinet Pharmacodyn 34:373-400.491

    33. Le Conte, P., F. Le Gallou, G. Potel, L. Struillou, D. Baron, and H. B. Drugeon.4921994. Pharmacokinetics, toxicity, and efficacy of liposomal capreomycin in493

    disseminated Mycobacterium avium beige mouse model. Antimicrob Agents494

    Chemother38:2695-701.495

    34. Lee, S. H., J. Shin, J. M. Choi, E. Y. Lee, D. H. Kim, J. W. Suh, and J. H. Chang. 496

    2003. The impurities of capreomycin make a difference in the safety and497

    pharmacokinetic profiles. Int J Antimicrob Agents 22:81-3.498

    35. Leggett, J. E., B. Fantin, S. Ebert, K. Totsuka, B. Vogelman, W. Calame, H.499

    Mattie, and W. A. Craig. 1989. Comparative antibiotic dose-effect relations at500

    several dosing intervals in murine pneumonitis and thigh-infection models. J Infect501Dis 159:281-92.502

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    28/59

    28

    36. Lyons, M. A., R. S. H. Yang, A. N. Mayeno, and B. Reisfeld. 2008. Computational503

    toxicology of chloroform: Reverse dosimetry using Bayesian inference, Markov chain504

    Monte Carlo simulation, and human biomonitoring data. Environmental Health505

    Perspectives 116:1040-1046.506

    37. Metzler, C. 2010. Masters Thesis: Physiologically based pharmacokinetic modeling507

    for prediction of pharmacokinetic parameters of capreomycin Colorado State508

    University Fort Collins509

    38. Mingeot-Leclercq, M. P., and P. M. Tulkens. 1999. Aminoglycosides:510

    nephrotoxicity. Antimicrob Agents Chemother43:1003-12.511

    39. Mork, A. K., and G. Johanson. 2010. Chemical-Specific Adjustment Factors for512

    Intraspecies Variability of Acetone Toxicokinetics Using a Probabilistic Approach.513

    Toxicological Sciences 116:336-348.514

    40. Mouton, J. W., U. Theuretzbacher, W. A. Craig, P. M. Tulkens, H. Derendorf,515

    and O. Cars. 2008. Tissue concentrations: do we ever learn? J Antimicrob Chemother516

    61:235-7.517

    41. Nagai, J., and M. Takano. 2004. Molecular aspects of renal handling of518

    aminoglycosides and strategies for preventing the nephrotoxicity. Drug Metab519

    Pharmacokinet 19:159-70.520

    42. NIAID Tuberculosis Working Group. 2007. NIAID Research Agenda Multidrug-521

    Resistant and Extensively Drug Resistant Tuberculosis.522

    43. Organick, A. B., and E. M. Wilson. 1968. Multiple drugs in retreatment of chronic523

    pulmonary tuberculosis. Results with capreomycin and ethambutol. Dis Chest 53:560-524

    70.525

    44. Peloquin, C. A. 1993. Pharmacology of the antimycobacterial drugs. Med Clin North526Am 77:1253-62.527

    45. Peters, S. A. 2012. Physiologically based pharmacokinetic (PBPK) modeling and528

    simulations : principles, methods, and applications in the pharmaceutical industry,529

    Wiley, Hoboken, N.J.530

    46. Reddy, M. B. 2005. Physiologically based pharmacokinetic modeling: science and531

    applications, Wiley-Interscience, Hoboken, N.J.532

    47. Reisfeld, B., A. N. Mayeno, M. A. Lyons, and R. S. H. Yang. 2007. Physiologically-533

    based pharmacokinetic and pharmacodynamic modeling p. xxii, 814 p. In S. Ekins534

    (ed.), Computational toxicology : risk assessment for pharmaceutical and535environmental chemicals. Wiley-Interscience, Hoboken, N.J.536

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    29/59

    29

    48. Schmitz, C., J. Hilpert, C. Jacobsen, C. Boensch, E. I. Christensen, F. C. Luft,537

    and T. E. Willnow. 2002. Megalin deficiency offers protection from renal538

    aminoglycoside accumulation. J Biol Chem 277:618-22.539

    49. Ward, D. T., S. J. McLarnon, and D. Riccardi. 2002. Aminoglycosides increase540

    intracellular calcium levels and ERK activity in proximal tubular OK cells expressing541

    the extracellular calcium-sensing receptor. J Am Soc Nephrol 13:1481-9.542

    50. World Health Organization. 2010 Multidrug and extensively drug-resistant TB543

    (M/XDR-TB): 2010 global report on surveillance and response.544

    WHO/HTM/TB/2010.3.545

    51. World Health Organization., and Stop TB Initiative (World Health546

    Organization). 2010. Treatment of Tuberculosis: guidelines for national programmes,547

    4th ed, World Health Organization, Geneva.548

    52. Zietse, R., R. Zoutendijk, and E. J. Hoorn. 2009. Fluid, electrolyte and acidbase549

    disorders associated with antibiotic therapy. Nature Reviews Nephrology 5:193-202.550

    551

    552

    553

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    30/59

    30

    Figure Captions554

    Figure 1. PBPK model structure555

    Figure 2. Conceptual model of the kidney556

    Figure 3. Low dose and high dose pharmacokinetic data in various tissues (N=4): (a) serum,557

    (b) kidney, (c) lung, (d) liver, and (e) spleen. Mean SD are shown. Scales for the abscissa558

    and ordinate differ for each plot. Data points below the LLOQ are not shown. LLOQ559

    (ng/mg): kidney (50.0), serum (1.0), liver (10.0), lung (10.0), spleen (10.0).560

    Figure 4. Parameter distributions found in the calibration simulations for (a) Km, (b) Vmax,561

    (c) CLRD, (d) CLR, (e) CLHC, (f) SC_Decay.562

    Figure 5. Measured and predicted serum concentration for the low-dose group based on (a)563

    using the measured peak concentration as the actual peak value, and (b) extrapolating to564

    determine the peak concentration565

    Figure 6. Low dose pharmacokinetic data and model predictions in various tissues: (a) serum,566

    (b) kidney, (c) lung, (d) liver, and (e) spleen. The experimental data are shown with symbols,567

    and the fifth and ninety-fifth percentile curves for the simulation data are shown with dashed568

    lines. Data points below the LLOQ are not shown.569

    Figure 7. High dose pharmacokinetic data and model predictions in various tissues: (a) serum,570

    (b) kidney, (c) lung, (d) liver, and (e) spleen. The experimental data are shown with symbols,571

    and the fifth and ninety-fifth percentile curves for the simulation results are shown with572

    dashed lines. Data points below the LLOQ are not shown.573

    574

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    31/59

    31

    Tables575

    576

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    32/59

    32

    577

    Table 1. Parameters to be determined through model calibration578

    Symbol Units Description (relevant compartment)

    KM ng/kg Michaelis constant in accumulation kinetics (kidney)

    Vmax ng/h Michaelis-Menten maximum accumulation rate (kidney)

    CLR l/h Overall renal clearance (kidney)

    CLRD l/h Deep renal compartment clearance (kidney)

    CLHC l h-1 kg-1 Hepatic clearance rate (liver)

    SC_Decay h-1Subcutaneous dose decay rate (subcutaneous); release into

    venous blood.

    579

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    33/59

    33

    580

    Table 2. Summary statistics for the parameters estimated in the calibration simulations581

    Parameter Mean SD

    Km(ng/kg) 5.55 x 108 0.46 x 108

    Vmax (ng/h) 1.04 x 106 0.08 x 106

    CLR (kg/h) 0.012 0.001

    CLRD (kg/h) 3.47 x 10-6 0.25 x 10-6

    CLHC(l hr-1 kg-1) 3.97 0.27

    SC_Decay (h-1) 0.902 0.164

    582

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    34/59

    34

    Table A3. Parameters for use in PBPK modeling583

    BW fromindividualmouse data

    partition coefficients

    BP (blood:plasma) 1 PSK (skin:blood) 1

    PLU (lung:blood) 2 PKS (shallow kidney:blood) 1

    PBR (brain:blood) 1 PS (spleen:blood) 1

    PF (fat:blood) 1 PG (gut:blood) 1

    PH (heart:blood) 1 PL (liver:blood) 1

    PM (muscle:blood) 1 PCR (carcass:blood) 1

    PB (bone:blood) 1

    Fractional tissue weights (17) Fractional tissue flows

    (fraction of cardiac output) (17)

    VLUC (lung) 0.0044 (exp) QLUC (lung) 1.0

    VBRC (brain) 0.018 QBRC (brain) 0.033

    VFC (fat) 0.070 QFC (fat) 0.043

    VHC (heart) 0.004 QHC (heart) 0.066

    VMC (muscle) 0.384 QMC (muscle) 0.159

    VBC (bone) 0.107 QBC (bone) 0.110

    VSKC (skin) 0.165 QSKC (skin) 0.058

    VKC (kidney) 0.014 (exp) QKSC (shallow kidney) 0.091

    VKSC (shallow kidney) 0.010 QSC (spleen) 0.01

    VKDC (deep kidney) 0.004 QGC (GI tract) 0.13

    VSpC (spleen) 0.0037 (exp) QLAC (hepatic artery) 0.02

    VGC (GI) 0.042 QCRC carcass 0.28

    VLC (liver) 0.05 (exp)

    VVC (venous blood) 0.0327

    VAC (arterial blood) 0.0163

    VRCR (carcass) 1 - sum of allothers

    584

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    35/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    36/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    37/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    38/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    39/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    40/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    41/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    42/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    43/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    44/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    45/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    46/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    47/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    48/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    49/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    50/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    51/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    52/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    53/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    54/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    55/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    56/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    57/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    58/59

  • 7/28/2019 A Physiologically-Based Pharmacokinetic Model for Capreomycin

    59/59