in vitro evaluation of hepatic transporter-mediated clinical drug … · 2012. 3. 1. · ms/ms...

41
DMD #43489 1 In vitro evaluation of hepatic transporter-mediated clinical drug-drug interactions: hepatocyte model optimization and retrospective investigation Yi-an Bi, Emi Kimoto, Samantha Sevidal, Hannah M. Jones, Hugh A. Barton, Sarah Kempshall, Kevin M. Whalen, Hui Zhang, Chengjie Ji, Katherine S Fenner, Ayman F. El- Kattan, and Yurong Lai Pharmacokinetics, Dynamics & Metabolism, World Wide Research & Development, Pfizer, Inc. Groton Laboratories, CT 06340 (Y.B, E.K, H.A.B, K.M.W, H.Z, C.J, A.F.E, and Y. L); La Jolla Laboratories, CA 92121 (S.S); Sandwich Laboratories, Kent, UK (H.M.J, S.K, K.S.F) DMD Fast Forward. Published on March 1, 2012 as doi:10.1124/dmd.111.043489 Copyright 2012 by the American Society for Pharmacology and Experimental Therapeutics. This article has not been copyedited and formatted. The final version may differ from this version. DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489 at ASPET Journals on July 3, 2021 dmd.aspetjournals.org Downloaded from

Upload: others

Post on 12-Feb-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

  • DMD #43489

    1

    In vitro evaluation of hepatic transporter-mediated clinical

    drug-drug interactions: hepatocyte model optimization and

    retrospective investigation

    Yi-an Bi, Emi Kimoto, Samantha Sevidal, Hannah M. Jones, Hugh A. Barton, Sarah

    Kempshall, Kevin M. Whalen, Hui Zhang, Chengjie Ji, Katherine S Fenner, Ayman F. El-

    Kattan, and Yurong Lai

    Pharmacokinetics, Dynamics & Metabolism, World Wide Research & Development,

    Pfizer, Inc. Groton Laboratories, CT 06340 (Y.B, E.K, H.A.B, K.M.W, H.Z, C.J, A.F.E,

    and Y. L); La Jolla Laboratories, CA 92121 (S.S); Sandwich Laboratories, Kent, UK

    (H.M.J, S.K, K.S.F)

    DMD Fast Forward. Published on March 1, 2012 as doi:10.1124/dmd.111.043489

    Copyright 2012 by the American Society for Pharmacology and Experimental Therapeutics.

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    2

    Running title page

    Running title: In vitro evaluation of OATP-mediated DDI

    Corresponding Author:

    Yurong Lai, PhD

    Department of Pharmacokinetics, Dynamics, and Metabolism, MS 8220-2475,

    Pfizer World Wide Research and Development, Pfizer Inc., Groton, CT 06340;

    Phone: 860-715-6448; Fax: 860-686-5364; Email: [email protected];

    Number of text pages:

    Number of tables: 4

    Number of Figures: 3

    Number of references: 53

    Number of words in Abstract: 206

    Number of words in Introduction: 743

    Number of words in Discussion: 1563

    Abbreviation: SCHH: Sandwich cultured human hepatocyte; OAT, organic

    anion transporter; OATP, organic anion transporting polypeptide; OCT, organic

    cation transporter; NTCP, sodium taurocholate cotransporting polypeptide. DDI,

    drug-drug interaction

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    3

    Abstract:

    To assess the feasibility of using sandwich cultured human hepatocytes (SCHH)

    as a model for characterizing transport kinetics for in vivo pharmacokinetic prediction,

    the expression of OATP proteins in SCHH, along with biliary efflux transporters (Li et al.,

    2009), were quantitatively confirmed by LC-MS/MS. Rifamycin SV (Rif SV), which was

    shown to completely block the function of OATP transporters, was selected as an

    inhibitor for assessing the initial rates of active uptake. The optimized SCHH model was

    applied in a retrospective investigation of compounds with known clinically significant

    OATP-mediated uptake and was further applied to explore drug-drug interactions (DDIs).

    Greater than 50% inhibition of active uptake by Rif SV was found to be associated with

    clinically significant OATP-mediated DDIs. We propose that the in vitro active uptake

    value could therefore serve as a cutoff for class 3 and 4 compounds of biopharmaceutics

    drug disposition classification system (BDDCS), which could be integrated into the

    International Transporter Consortium (ITC) decision tree recommendations (Giacomini et

    al., 2010) to trigger clinical evaluations for potential DDI risks. Furthermore, kinetics of

    in vitro hepatobiliary transport obtained from SCHH, along with protein expression

    scaling factors, offer an opportunity to predict complex in vivo processes using

    mathematical models, such as physiologically-based pharmacokinetics models (PBPK).

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    4

    Introduction

    Hepatobiliary elimination are primary elimination routes for many endogenous

    and exogenous xenobiotics. Hepatic uptake and efflux transporters respectively located

    on sinusoidal or canalicular membranes, contribute to the vectorial transport of drugs and

    their metabolites from systemic circulation to bile (Meier et al., 1997; Kullak-Ublick et

    al., 2000). Two classes of hepatic uptake transporters, sodium-dependent and sodium-

    independent transporters, co-exist on the sinusoidal membrane of hepatocytes with

    overlapping substrate specificities. For example, the sodium-dependent transporter,

    sodium-taurocholate co-transporting polypeptide (NTCP), is shown to transport the

    organic anion transporting polypeptide (OATP) substrates atorvastatin and rosuvastatin

    (Ho et al., 2006; Choi et al., 2011). OATP1B1/1B3, organic anion transporter 2 (OAT2)

    and organic cation transporter 1 (OCT1) are specifically expressed in the liver and

    transport structurally diverse substrates in a sodium-independent manner. In the last

    decade, significant advances toward the prediction of in vivo NME clearance using in

    vitro models have been well-documented. Indeed, in vitro human liver microsomes and

    isolated hepatocytes were shown to be important tools in drug discovery and

    development to confidently predict in vivo human drug metabolism for compounds

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    5

    predominately eliminated by cytochrome P-450 (Obach, 1999; McGinnity et al., 2004).

    However, human PK prediction remains very challenging for compounds where drug

    transporters are involved in the clearance mechanism. In 2010, the International

    Transporter Consortium (ITC) recommended decision trees using in vitro systems to

    assess the risk of in vivo transporter mediated drug-drug interactions (DDIs) (Giacomini

    et al., 2010). For OATP transporters, the investigation cascade is initiated by the criteria

    of active hepatic uptake if hepatic clearance is an important route of elimination, e.g >0.3

    of total clearance (Giacomini et al., 2010). However, the models and the extent of active

    hepatic uptake that would trigger the investigation cascade remain undetermined and

    suitable in vitro tools to assess the active/passive hepatic uptake need to be further

    validated. We hypothesized that clinically relevant OATP mediated DDI is associated

    with significant active uptake in vitro.

    Recently, a physiologically-based pharmacokinetic (PBPK) model, in which

    physiological compartments representing organs/tissues are connected with blood flow,

    was developed to predict in vivo clearance and time profiles of drug elimination using in

    vitro models such as suspension hepatocytes and canalicular membrane vesicles

    (Watanabe et al., 2009). The approach is generally accepted as a useful tool to predict

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    6

    tissue concentration, drug-drug interaction, and the effects of enzyme/transporter genetic

    polymorphisms on drug exposure. Practically, parameters for distinct clearance processes

    including passive diffusion, transporter-mediated hepatic uptake (sinusoidal), metabolism,

    and transporter-mediated efflux (canalicular) into the bile could be determined in vitro

    and used to predict in vivo human.

    The sandwich cultured hepatocyte model (SCH) built by culturing hepatocytes

    between two layers of gelled matrix in a sandwich configuration, forms a bile canalicular

    network and provides the three-dimensional orientation and proper localization of

    hepatobiliary transporters that mimic in vivo conditions and allow the vectorial transport

    of xenobiotics (Bi et al., 2006). In 2006, Turncliff et al. evaluated the hepatobiliary

    disposition of metabolites of terfenadine generated in sandwich cultured rat hepatocytes

    (SCRH) (Turncliff et al., 2006). The advancement of in vitro models for the clearance

    prediction in human has further reflected an increase in the mechanistic understanding of

    the hepatic vectorial elimination process (Kotani et al., 2011). For example, by using LC-

    MS/MS quantification methods, the expression of biliary efflux transporters in sandwich

    cultured rat hepatocytes (SCRH) were determined and used to improve the extrapolation

    from in vitro to in vivo for the compounds excreted from bile (Li et al., 2009; Li et al.,

    2010). Meanwhile, a decrease in functional uptake in SCRH was observed raising

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    7

    concerns for its suitability as a model for active hepatic uptake evaluations (Li et al.,

    2010; Kotani et al., 2011). However, unlike SCRH, based on our in house unpublished

    results and the recently published data (Kotani et al., 2011), active uptake functions in

    sandwich cultured human hepatocytes (SCHH) are well-maintained and the system

    appears promising as a single in vitro model for evaluation of candidate drug uptake and

    biliary excretion. The objectives of this research are three folds; first, confirm the

    maintenance of hepatic-specific uptake transporter function and expression in SCHH;

    second, optimize the experimental conditions needed to define the active hepatic uptake

    and biliary excretion to obtain in vitro parameters that could be used as inputs for

    mathematical PBPK model for in vivo prediction (Jones et al., 2012); and third, obtain an

    in vitro cutoff values for active uptake in SCHH that would be appropriate to trigger the

    clinical investigations recommended by the ITC.

    Materials and Methods

    Reagent and hepatocytes

    HPLC grade acetonitrile, water and methanol were purchased from Burdick &

    Jackson (Muskegon, MI) and EMD Chemicals, Inc (Gibbstown, NJ), respectively. Hanks

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    8

    balanced salt solution (HBSS) was purchased from Invitrogen (Carlsbad, CA).

    Rosuvastatin, atorvastatin, pitavastatin valsartan, fluvastatin, pravastatin, cerivastatin,

    buprenorphine, and midazolam were obtained from Sequoia Research Products Ltd.

    (Oxford, UK). Rifamycin SV (Rif SV) was purchased from Sigma-Aldrich (St. Louis,

    MO). Ammonium acetate was obtained from Mallinckrodt (Phillipsburg, NJ).

    [3H]Taurocholate (TC, 4.6 Ci/mmol) was purchased from PerkinElmer Life Sciences

    (Boston, MA). The BCA kit was purchased from Pierce Biotechnology (Rockford, IL).

    The ProteoExtract® native membrane protein extraction kit was purchased from

    Calbiochem (Temecula, CA). Trypsin was purchased from Promega (Madison, WI).

    MatrigelTM (phenol red free) and collagen coated 24 well plates were obtained from BD

    Biosciences (Franklin Lakes, NJ). In Vitro GRO-HT, In Vitro GRO-CP and In Vitro

    GRO-HI hepatocytes media were purchased from Celsis In Vitro Technologies

    Inc.(Baltimore, MD). Cryopreserved human hepatocyte (lot Hu4165) was purchased from

    CellzDirect (Pittsboro, NC).

    Sandwich cultured human hepatocytes (SCHH)

    Cryopreserved hepatocytes were thawed and plated as described previously (Bi et

    al., 2006). In brief, hepatocytes were thawed in a water bath at 37°C, and then

    immediately poured into 50 mL in pre-warmed In Vitro GRO-HT medium in a conical

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    9

    tube. The cells were then centrifuged at 50 × g for 3 minutes and resuspended 0.85 × 106

    cells/mL in In Vitro GRO-CP medium. Cell viability was determined by trypan blue

    exclusion. On day 1, hepatocyte suspensions were seeded in BioCoat 24-well plates in a

    volume of 0.5 mL/well. After incubation overnight at 37ºC, the hepatocytes were overlaid

    with 0.5 mL IN VITRO GRO-HI medium with MatrigelTM (0.25 mg/mL). The IN VITRO

    GRO-HI media were refreshed every 24 hr.

    Extraction and digestion of membrane protein

    At day 5 post culture, SCHH were detached from cell culture plates and washed

    with HBSS. Along with suspension hepatocytes, the membrane protein fraction of

    hepatocytes was extracted as described previously (Li et al., 2008) using the

    ProteoExtract® native membrane protein extraction kit (Calbiochem). Briefly, hepatocyte

    pellets were homogenized in extraction buffer I of the kit containing a protease inhibitor

    cocktail followed by incubation at 4 oC for 10 minutes with gently rocking. The

    homogenate was centrifuged at 16,000 x g for 15 minutes at 4 oC. The supernatant

    containing cytosolic proteins was discarded and the pellets were re-suspended in

    extraction buffer II of the kit containing a protease inhibitor cocktail. After 60 minutes of

    incubation at 4 oC with gentle rocking, the suspension was centrifuged at 16,000 x g for

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    10

    15 minutes at 4 oC. The protein concentration in the membrane fractions was determined

    using the BCA protein assay kit (Pierce Biotechnology, Inc. Rockford, IL).

    LC-MS/MS quantitative measurement of OATP1B1, 1B3 and 2B1 transporters

    Proteotypic peptides and stable isotope label (SIL) peptides of OATP1B1, 1B3,

    and 2B1 were selected (Table 1) and synthesized as surrogate analytes for the

    corresponding protein quantification by LC-MS/MS (Li et al., 2008). LC-MS

    quantification for human OATP proteins and two of six peptides identified

    (NVTGFFQSFK and SSPAVEQQLLVSGPGK) were also reported by Ji et al (Ji et al.,

    2012) . The digestion condition were optimized by our previous report (Balogh et al.,

    2012). Briefly, 80 μg of membrane fraction protein was reduced with 6 mM dithiothreitol

    and alkylated with iodoacetamide in 25 mM ammonium bicarbonate digestion buffer

    containing 10% sodium deoxycholate monohydrate (DOC), and then digested by trypsin

    (the final concentration of DOC during digestion was 1%). At the end of digestion,

    samples were acidified with an equal amount of 0.2% formic acid in water with SIL

    internal standards, and centrifuged at 14,000 x g for 5 minutes. The supernatants were

    transferred to a new plate and dried down. The samples were reconstituted with 0.1%

    formic acid in water and analyzed by LC-MS/MS.

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    11

    The calibration curve was prepared using the synthetic proteotypic peptide with a

    fixed concentration of each SIL peptide as the internal standard. Sample quantification

    was conducted by coupling a triple quadrupole mass spectrometer (TQ-MS, API4000,

    Applied Biosystem, Foster City, CA) to a Shimadzu High-performance liquid

    chromatography (LC) system (SLC-10A, WoolDale, IL) and HTS PAL Leap autosampler

    (Carrboro, NC). A Phenomenex Kinetex 2.6 µm C18 100A column (3.0 × 100 mm) was

    used for peptide chromatography. A linear gradient elution program was conducted to

    achieve chromatographic separation with mobile phase A (0.1% formic acid in HPLC

    Grade Water), and mobile phase B (0.1% formic acid in acetonitrile). A sample volume

    of 10 μl was injected onto the LC column at a flow rate of 0.5 mL/min. The parent-to-

    product transitions for the proteotypic peptide generally represent the doubly charged

    parent ion to the single charged product y ions for each peptide (Table 1). Data were

    processed by integrating the appropriate peak areas for the analyte peptides and the SIL

    internal standard peptides in Analyst 1.4.2 (Applied Biosystems, Foster City, CA).

    Determination of hepatic uptake in SCHH

    SCHH were rinsed twice with 0.5 ml of 37°C regular HBSS buffer or Ca2+/Mg2+-

    free HBSS containing 1 mM EGTA, and then replaced with fresh regular HBSS buffer or

    Ca2+/Mg2+-free HBSS containing 1 mM EGTA. The disruption of the bile canalicular

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    12

    network was achieved by pre-incubation with Ca2+/Mg2+-free HBSS containing 1 mM

    EGTA buffer for 10 minutes. For determination of the effects of various inhibitors on

    rosuvastatin uptake in SCHH, parallel incubations were conducted in the presence of the

    uptake transporter inhibitors, Rif SV (100 μΜ), Cyclosporin A (CsA, 10 μΜ), and

    gemfibrozil (30 μΜ). SCHH uptake was initiated by the addition of 500 µL containing

    substrate at a concentration indicated in the figure legends, with or without inhibitors.

    Reactions were terminated at designated time points by quickly washing the hepatocytes

    three times with ice-cold HBSS buffer. The cells were either lysed with 0.5% Triton-100

    (radiolabeled compounds) or 100% methanol containing internal standard (non-

    radiolabeled compounds). Samples were analyzed either by LC-MS/MS, respectively.

    LC-MS/MS Analysis of Probe Substrates

    LC-MS/MS analysis of probe substrates was conducted with an API 4000 triple

    quadruple mass spectrometer (PE Sciex, Ontario, ON, Canada) coupled with a turbo ion

    spray interface in positive ion mode, and connected with a Shimadzu LC (SLC-10A)

    system (WoolDale, IL) and Gilson 215 autosampler. The mass spectrometer was

    controlled by Analyst 1.4.2 software (Applied Biosystems). The Gilson autosampler was

    independently controlled by Gilson 735 software and synchronized to Analyst via contact

    closure. The HPLC method consisted of a step gradient with 25 uL samples loaded onto

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    13

    a 1.5 x 5 mm Showadenko (Tokyo, Japan) ODP 13 µm particle size column using 95%

    2mM ammonium acetate/5% 50/50 methanol/acetonitrile. Incubation samples and

    standard curve samples were eluted with 10% 2mM ammonium acetate in 50/50 of

    methanol/acetonitrile. Peak area counts of analyte compound and internal standard (IS)

    were integrated using DiscoveryQuant Analyze as an add-on to Analyst 1.4.2. The

    obtained values underwent data analysis by averaging the analyte area divided by the

    internal standard analyte for each concentration.

    Data Analysis

    The apparent in vitro biliary clearance (CLbile) (Liu et al., 1999) and percent active

    uptake were determined by equations 1 and 2, respectively. The hepatic uptake of test

    compounds was estimated from the initial uptake phase (0.5 to 1 min) while biliary

    excretion was assessed at 10 minutes. The data represent the results from a single study

    run in triplicate or duplicate and a minimum of two experiments were performed. The

    standard deviations or coefficient variation (CV%) was listed in the legends of figure or

    table.

    )(

    ),2/2(),(

    *_ medium

    FreeMgCaHBSSStd

    ionconcentrattimeIncubation

    onAccumulationAccumulatiCLbile

    ++−= Eq1

    % active uptake = 100-(slope+Rif SV)/(slope-Rif SV) Eq 2

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    14

    Results

    Protein expression of OATP1B1, 1B3, and 2B1 in suspension hepatocytes and SCHH

    As functional active uptake has been shown previously in SCHH both in house

    and in the literature (Kotani et al., 2011), in the present study, the protein expression of

    OATP1B1, 1B3, and 2B1 in SCHH was measured by LC-MS/MS and compared with that

    in suspension hepatocytes. Peptides proteolytically released from the target OATP

    proteins were quantified using external synthetic peptide calibration curves. The peptide

    fragments were monitored using multiple reactions monitoring (MRM) (Table 1). Sample

    preparation, digestion, and detection limits of OATP protein quantification were

    previously evaluated (Balogh et al., 2012). As shown in Table 2, while OATP1B3 and

    2B1 expression levels were reduced to approximately half of that in suspension

    hepatocytes, OATP1B1 expression in SCHH was slightly higher (1.5 fold) than that

    found in suspension hepatocytes. The results provide support for OATP-mediated active

    hepatic uptake in SCHH, which indicates that SCHH could be a suitable tool for the

    assessment of active OATP uptake. In addition, the protein expression obtained by LC-

    MS/MS measurement could be integrated into a mathematical model as a component of

    scaling factors for in vivo extrapolation from in vitro.

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    15

    Inhibition of active uptake in SCHH

    The active component of hepatic uptake can be determined by the total hepatic

    uptake (PSuptake) minus the passive diffusion (PSpassive) in an in vitro hepatocyte model.

    Generally, passive diffusion in hepatocytes can be obtained by either co-incubation with

    an OATP inhibitor or by conducting the uptake experiment at low temperature, e. g at 4ºC.

    We previously reported that the lack of uptake might be an artificial effect of a rigid cell

    membrane at 4ºC and the uptake in the hepatocyte model determined at 37ºC and 4ºC

    might not truly reflect the functional active uptake (Kimoto et al., 2011). To select a

    suitable inhibitor blocking carrier-mediated active hepatic uptake in SCHH, time-

    dependent accumulation of rosuvastatin, a known substrate of OATP proteins, was

    investigated in the presence or absence of known OATP inhibitors, rifamycin SV (Rif SV,

    100 μM), cyclosporin A (CsA, 10 μM) and gemfibrozil (30 μM). As depicted in Figure 1,

    rosuvastatin was actively transported into SCHH and the uptake was linear up to 1.5

    minutes. It is worthwhile to note that a positive Y-intercept was obtained by extending the

    trendline of rosuvastatin uptake. Non-specific binding on hepatocyte surface has been

    proposed to contribute the intercept and was not included here for the calculation of

    initial uptake rate. The initial uptake rates of rosuvastatin estimated from 0.5 to 1 minute

    were inhibited by all three OATP inhibitors, Rif SV, CsA and gemfibrozil, by 95, 80, and

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    16

    78%, respectively. The active uptake of rosuvastatin was almost completely inhibited in

    the presence of Rif SV at 100 μΜ.

    Hepatic uptake and biliary excretion for compounds that are the substrates of phase

    I/II metabolizing enzymes or hepatic uptake transporters in SCHH

    As mentioned above, rosuvastatin uptake in SCHH was nearly abolished by co-

    incubation with 100 μΜ Rif SV. The results agree with previous reports that Rif SV (100

    μM) totally blocks OATP functions in OATP transfected cell lines (Vavricka et al., 2002).

    To determine if Rif SV also affects metabolizing enzymes, biliary excretion, and active

    hepatic uptake by transporters other than OATPs, initial uptake and biliary excretion of

    various compounds were further tested in SCHH. Buprenorphine and midazolam, which

    are metabolized by UGT1A1 and CYP3A4, respectively, were used as compounds that

    penetrate the hepatocytes via passive diffusion. TC were selected as probe substrates of

    NTCP transporters. Rosuvastatin as a known OATP substrate was again tested in SCHH

    for characterizing the initial uptake and biliary excretion kinetics. In addition,

    rosuvastatin and taurocholate (TC) are excreted from bile through biliary efflux

    transporters such as multidrug resistant protein 2 (MRP2), breast cancer resistant protein

    (BCRP) or bile salt exporting pump (BSEP) (Ito et al., 2010). The uptake (PSuptake and

    PSpassive) and biliary excretion (CLbile) in SCHH of the compounds are shown in Figure 2

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    17

    and Table 3. As expected, Rif SV (100 μΜ) inhibited approximately 93% and 83% of the

    uptake rate of rosuvastatin and TC in SCHH, respectively. Following a decrease of uptake

    into SCHH caused by the inhibition of Rif SV, biliary excretion of rosuvastatin and TC

    were also substantially decreased (Table 3). Although significant increases in the

    intracellular concentration of buprenorphine or midazolam in SCHH were detected in the

    presence of Rif SV (Figure 2), the initial uptake rates of the compounds were not

    significantly altered. The intracellular accumulation of buprenorphine or midazolam by

    Rif SV might be due to the inhibition of CYP3A4 and UGT1A1 activities in SCHH.

    Since no biliary excretion was detected for burperenophine and midazolam (Table 3) both

    in the absence and presence of Rif SV, the results revealed that Rif SV did not change the

    elimination profile by switching elimination pathways between metabolizing enzyme-

    mediated clearance and transporter-mediated biliary excretion.

    Hepatic uptake in SCHH for the compounds that undergo clinically significant DDIs

    with OATP inhibitors

    As noted above, SCHH has been characterized as a suitable model to assess in

    vitro hepatic uptake (active and passive) and biliary excretion. To evaluate the risk of in

    vivo OATP-mediated DDIs from the in vitro SCHH model, a literature review was

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    18

    conducted to compile the OATP substrates that undergo significant DDIs in the clinic (>2

    fold increase in AUC) when co-administered with OATP inhibitors. The substrates are

    listed in Table 4 and we conducted uptake and biliary excretion assays in SCHH to

    determine the in vitro active uptake (PSactive) and CLbile. To avoid potential saturation of

    active uptake transporters, the substrate concentrations selected either were below the

    OATP Km known from literature reports or lack of saturation was confirmed by our

    preliminary experiments. As indicated in Table 4, initial uptake rates into SCHH for the

    compounds ranged from 2 to 45 uL/min/mg protein. Rif SV inhibitable active hepatic

    uptake ranged from 55% to 84% in SCHH for the compounds reported to undergo OATP-

    mediated DDIs. Although the Rif SV inhibitable active uptake (%) did not correlate with

    the AUC changes that are observed in clinic (Figure 3A), these compounds were actively

    uptaken into hepatocyte to the extent of greater than 50% (Figure 3A). On the other hand,

    an increase of passive diffusion into hepatocytes tended to diminish the fold increase in

    AUC change caused by OATP inhibitors (Figure 3B), suggesting OATP mediated DDI

    risk could be low for high permeable compounds.

    Discussion

    Adverse clinically significant DDIs represent major challenges in drug discovery

    and development. In the last two decades, preclinical in vitro /in vivo models were used to

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    19

    effectively predict human pharmacokinetics. However, the predictions were successful

    for the compounds that are mainly eliminated renally or via cytochrome P-450

    metabolizing enzymes (Obach, 2009). Predicting hepatic transporter mediated clearance

    continues to be challenging due to large interspecies differences in hepatic transporter

    homology or expression, and lack of validated in vitro tools (Lai, 2009). Hepatic drug

    elimination is generally composed of a series of processes that include: entrance into

    hepatocyte via passive diffusion and active uptake mediated by hepatic transporters;

    metabolic elimination through phase I and/or phase II enzymes; and excretion to bile

    and/or back to systemic circulation by the efflux transporters. To assess processes

    involved in hepatic uptake and biliary excretion for in vivo prediction, the development of

    an in vitro model that mimics the complexity of the hepatic transport system has become

    imperative.

    Human hepatocyte in vitro models are widely accepted as a valuable tool for

    investigating drug metabolic liability and gene induction and toxicity, as well as serving

    as effective screening tools for NMEs. The cellular polarity that allows vectorial transport

    in vivo is disrupted rapidly when cells are isolated from the intact organ. Therefore, the

    restoration of the bile canalicular network in the cultured hepatocyte model is desired for

    investigating the vectorial transport of drug candidates. SCHH has been generally

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    20

    accepted as a good model to aid in predicting biliary clearance in humans (Bi et al., 2006).

    Following the previous efforts to understand the expression of hepatobiliary efflux

    transporters in SCHH (Li et al., 2009), in the present study, we report, for the first time,

    the maintenance in SCHH of uptake hepatic transporters, OATP1B1, 1B3 and 2B1 as

    measured by LC-MS/MS. This provides molecular evidence to support SCHH as a

    model for obtaining functional hepatic uptake parameters for in vivo prediction.

    Moreover, to better define the uptake clearance in SCHH, ideally an inhibitor that can

    block all known hepatic uptake transporters with minimum effect on passive diffusion

    and biliary excretion is needed. To meet these criteria, we measured rosuvastatin uptake

    in SCHH co-incubated with several known OATP transporter inhibitors. Under the

    concentrations applied, these inhibitors can completely block OATP1B1 or 1B3 activity

    in OATP gene overexpressing systems (Vavricka et al., 2002; Yamazaki et al., 2005;

    Letschert et al., 2006). As a result, Rif SV (100 μΜ) was shown to completely knock

    down rosuvastatin uptake in SCHH. The inhibitor selected provided the ability in

    assessing the sum of active hepatic uptakes contributed by OATP transporters expressed

    in hepatocytes. In contrast, Rif SV (100 μΜ) had no effect on the passive diffusion of

    midazolam and buprenorphine into hepatocytes. Rif SV also increased the intracellular

    accumulation of burprenorphine and midazolam through the inhibition of metabolizing

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    21

    enzymes, such as UGT1A1 and CYP3A4. These inhibitory effects were minimized

    through optimization of experimental conditions by calculating the initial uptake rate to

    avoid the artificial effect on hepatic uptake caused by the inhibitory effects of Rif SV on

    metabolizing enzymes. Moreover, the reduced biliary excretion is observed followed the

    decreases of hepatic uptake. Since the intracellular accumulation of the compounds was

    not observed (Table 3, 4 and Figure 2) with or without Ca2+, we speculate that the

    reduced biliary excretion was caused by the inhibitory effects on hepatic uptake.

    However, direct inhibitory effects on hepatobiliary efflux transporters still remain to be

    further investigated as Rif SV appears as an inhibitor of efflux transporters including bile

    salt export pump (BSEP) (Wang et al., 2003).

    Three isoforms (OATP1B1, OATP1B3 and OATP2B1) are considered to play a

    pivotal role in the hepatic uptake of xenobiotics and endogenous compounds on the

    sinusoidal membrane of hepatocytes (Giacomini et al., 2010). The OATPs transport drugs

    from a wide range of therapeutic classes including the 3-hydroxymethylglutaryl-CoA

    reductase inhibitors (statins), angiotensin II receptor antagonists (e.,g., olmesartan and

    valsartan), angiotensin-coverting enzyme inhibitors (enalapril and temocaprilat), the H1-

    receptor antagonist fexofenadine, and the endothelin receptor antagonist, bosentan

    (Giacomini et al., 2010). As the OATP proteins are poorly conserved evolutionarily as

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    22

    evidenced by the lack of human orthologues in rodents, extrapolation of in vivo human

    from rodent models remains limited. The ITC recommends that the clinical investigation

    cascade should be initiated when active hepatic transport is involved in the liver

    clearance pathway and outlines decision trees for in vitro evaluation of hepatic uptake

    using a hepatocyte model to predict the potential risk of clinical DDIs. The investigation

    cascades are considered to be similar to the in vitro studies of drug metabolism and

    interaction (Giacomini et al., 2010). As a result, developing in vitro models that can

    predict OATP mediated DDIs is an efficient and inexpensive approach that could reduce

    or eliminate the need for further clinical investigation. However, the extent of in vitro

    active hepatic uptake necessary to trigger the clinical assessment of DDI was not

    provided (Giacomini et al., 2010). In addition, the performance of the in vitro hepatocyte

    models should be evaluated to increase confidence in obtaining outcomes through

    retrospective analysis. With this in mind, a literature review was conducted and the

    compounds with reported OATP-mediated clinical DDI were tested in our optimized

    SCHH model. As expected, active hepatic uptake of the compounds with clinically

    significant OATP DDIs was observed. The contribution of the active portion to overall

    hepatic uptake was high, being 55 to 84% of the total uptake rate. As hepatocyte lot

    differences in transporter expression exist, it is important that we confirmed the OATP

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    23

    expression in SCHH, and compared the active hepatic uptake in multiple hepatocyte lots

    (Supplemental Table 1).These results suggest that 50% active hepatic uptake in SCHH

    could be the in vitro cutoff to trigger the clinical risk assessment of hepatic transporter-

    mediated DDIs.

    The relationship among solubility, passive permeability, and the effects of drug

    transporter and metabolizing enzymes on drug disposition has been well addressed by the

    biopharmaceutics drug disposition classification system (BDDCS) (Wu and Benet, 2005).

    As depicted in Figure 3B, the fold increase of AUC tended to decrease as the PSpassive

    in SCHH increased. The results suggest that low permeability drugs that rely primarily on

    transporter uptake for entry are prone to clinical DDIs mediated by OATP transporters.

    When applying the classification system, transporter effects are predicted to be minimal

    for high permeability/high solubility Class 1 compounds (Wu and Benet, 2005). In the

    gastrointestinal space, good solubility is essential for saturation of efflux transporters to

    obtain good absorption. Once the compounds are absorbed (into the systemic space), high

    permeability/low solubility Class 2 compounds could behave similarly to Class 1

    compounds, in that the high permeability can overcome the transporter effects, and

    therefore reduce the risk of transporter mediated DDIs by competing drug entering into

    hepatocytes. This simple categorization under BDDCS suggests that the high vs low

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    24

    permeability designation reflects the differences in freedom of access to phase I and/or

    phase II metabolizing enzymes within hepatocytes. In addition to the factors described

    here, hepatic blood flow needs to be considered as under flow limited conditions the

    impacts of inhibition of uptake transport could be reduced.

    It is interesting to note that atorvastatin is categorized as class 2 in the previous

    report (Wu and Benet, 2005). However, atorvastatin was shown to be have limited

    diffusion into SCHH in present study, and could be categorized as a class 4 compound

    (9.3 uL/min/mg of atorvastatin vs. 91 ul/min/mg of propranolol, data not shown). In this

    regard, additional investigations are proposed to better understand the passive diffusion

    cutoff using cellular uptake model, e.g SCHH. Some exceptions were also found in the

    relationship between passive permeability and AUC increase observed in clinic. While

    cerivastatin was a high permeable compound in SCHH (15 μL/min/mg), a significant

    AUC increase (4.8 and 5.6 fold) was reported when cerivastatin was co-administered

    with the OATP inhibitors, CsA and gemfibrozil, respectively (Figure 3B, circle marked).

    Cerivastatin interacted with OATP proteins and is also extensively metabolized by

    CYP2C8 and CYP3A4 in liver via demethylation of the benzylic methyl ether moiety

    (Muck, 2000). CsA and Gemfibrozil are potent inhibitors of OATP transporters, as well

    as inhibiting CYP metabolizing enzymes. These clinical observations could be explained

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    25

    by the inhibition of multi-clearance pathways resulting in a sum of effects on overall

    hepatic clearance. Due to lack of exposure data, it is also worthwhile to note that outliers

    were anticipated with co-administration of CsA, as the inhibition of OATP transporters

    may have been insufficient in vivo (Figure 3 below the dotline). Collectively, caution

    should be raised in that the interplay between transporters and metabolizing enzymes

    could generate a multi-level complexity in predicting clinical DDI from an in vitro

    system.

    In conclusion, SCHH maintains hepatic transporter expression and functional

    activity, and appears to be a well-characterized in vitro model for prediction of in vivo

    human PK. Rif SV inhibited the active uptake of OATP transporters, demonstrating it was

    an OATP inhibitor useful to estimate the extents of active and passive uptake in SCHH.

    The optimization of the experimental design minimized the impacts of inhibitory effects

    on metabolizing enzymes. Retrospective analysis for the compounds that undergo

    clinically significant hepatic transporter-mediated DDIs suggested that 50% or greater

    active hepatic uptake in SCHH with low passive permeability would have a potential risk

    of clinical DDIs and this value could serve as a cutoff to trigger the clinical investigation

    cascade for DDI risk assessment.

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    26

    Acknowledgements:

    We would like to acknowledge Dr. Larissa M Balogh for the help on OATP protein

    quantification and the editing of manuscript. We also thank Andrea Clouser-Roche for

    bioanalytical support and Drs Theodore E. Liston, Larry M. Tremaine, Bo Feng,

    Manthena V. Varma, John Litchfield, and Hendrik Neubert for their helpful scientific

    comments and suggestions.

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    27

    Authorship contributions

    Participated in research design: Y.B., E.K., H.A.B., H.M.J., Y.L

    Conducted experiments: Y.B., E.K., S.S., S.K., K.M.W., H.Z

    Contributed new reagents or analytic tools: Y.B., E.K.

    Performed data analysis: Y.B., E.K., S.S., H.M. J., H.A.B., S.K., K.M.W., H.Z., C.J., K.S.F.,

    A.F.E., Y.L

    Wrote or contributed to the writing of the manuscript: Y.B., E.K., S.S., H.M J., H.A.B.,

    S.K., K.M.W., H.Z., C.J., K.S.F., A.F.E., Y.L

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    28

    Reference:

    Asberg A, Hartmann A, Fjeldsa E, Bergan S and Holdaas H (2001) Bilateral

    pharmacokinetic interaction between cyclosporine A and atorvastatin in renal

    transplant recipients. Am J Transplant 1:382-386.

    Backman JT, Kyrklund C, Neuvonen M and Neuvonen PJ (2002) Gemfibrozil greatly

    increases plasma concentrations of cerivastatin. Clin Pharmacol Ther 72:685-691.

    Balogh LM, Kimoto E, Chupka J, Zhang H and Lai Y (2012) Membrane Protein

    Quantification by Peptide-Based Mass Spectrometry Approaches: Studies on the

    Organic Anion-Transporting Polypeptide Family. J. Proteomics and

    Bioinformatics in press.

    Bi YA, Kazolias D and Duignan DB (2006) Use of cryopreserved human hepatocytes in

    sandwich culture to measure hepatobiliary transport. Drug Metab Dispos

    34:1658-1665.

    Binet I, Wallnofer A, Weber C, Jones R and Thiel G (2000) Renal hemodynamics and

    pharmacokinetics of bosentan with and without cyclosporine A. Kidney Int

    57:224-231.

    Busti AJ, Bain AM, Hall RG, 2nd, Bedimo RG, Leff RD, Meek C and Mehvar R (2008)

    Effects of atazanavir/ritonavir or fosamprenavir/ritonavir on the pharmacokinetics

    of rosuvastatin. J Cardiovasc Pharmacol 51:605-610.

    Choi MK, Shin HJ, Choi YL, Deng JW, Shin JG and Song IS (2011) Differential effect of

    genetic variants of Na(+)-taurocholate co-transporting polypeptide (NTCP) and

    organic anion-transporting polypeptide 1B1 (OATP1B1) on the uptake of HMG-

    CoA reductase inhibitors. Xenobiotica 41:24-34.

    Deng S, Chen XP, Cao D, Yin T, Dai ZY, Luo J, Tang L and Li YJ (2009) Effects of a

    concomitant single oral dose of rifampicin on the pharmacokinetics of pravastatin

    in a two-phase, randomized, single-blind, placebo-controlled, crossover study in

    healthy Chinese male subjects. Clin Ther 31:1256-1263.

    Dingemanse J, van Giersbergen PL, Patat A and Nilsson PN (2010) Mutual

    pharmacokinetic interactions between bosentan and lopinavir/ritonavir in healthy

    participants. Antivir Ther 15:157-163.

    Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, Chu X, Dahlin A,

    Evers R, Fischer V, Hillgren KM, Hoffmaster KA, Ishikawa T, Keppler D, Kim

    RB, Lee CA, Niemi M, Polli JW, Sugiyama Y, Swaan PW, Ware JA, Wright SH,

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    29

    Yee SW, Zamek-Gliszczynski MJ and Zhang L (2010) Membrane transporters in

    drug development. Nat Rev Drug Discov 9:215-236.

    Goldberg R and Roth D (1996) Evaluation of fluvastatin in the treatment of

    hypercholesterolemia in renal transplant recipients taking cyclosporine.

    Transplantation 62:1559-1564.

    He YJ, Zhang W, Chen Y, Guo D, Tu JH, Xu LY, Tan ZR, Chen BL, Li Z, Zhou G, Yu BN,

    Kirchheiner J and Zhou HH (2009) Rifampicin alters atorvastatin plasma

    concentration on the basis of SLCO1B1 521T>C polymorphism. Clin Chim Acta

    405:49-52.

    Hedman M, Neuvonen PJ, Neuvonen M, Holmberg C and Antikainen M (2004)

    Pharmacokinetics and pharmacodynamics of pravastatin in pediatric and

    adolescent cardiac transplant recipients on a regimen of triple immunosuppression.

    Clin Pharmacol Ther 75:101-109.

    Hermann M, Asberg A, Christensen H, Holdaas H, Hartmann A and Reubsaet JL (2004)

    Substantially elevated levels of atorvastatin and metabolites in cyclosporine-

    treated renal transplant recipients. Clin Pharmacol Ther 76:388-391.

    Ho RH, Tirona RG, Leake BF, Glaeser H, Lee W, Lemke CJ, Wang Y and Kim RB (2006)

    Drug and bile acid transporters in rosuvastatin hepatic uptake: function,

    expression, and pharmacogenetics. Gastroenterology 130:1793-1806.

    Holdaas H, Hagen E, Asberg A, Lund K, Hartman A, Vaidyanathan S, Prasad P, He YL,

    Yeh CM, Bigler H, Rouilly M and Denouel J (2006) Evaluation of the

    pharmacokinetic interaction between fluvastatin XL and cyclosporine in renal

    transplant recipients. Int J Clin Pharmacol Ther 44:163-171.

    Ito S, Kusuhara H, Kuroiwa Y, Wu C, Moriyama Y, Inoue K, Kondo T, Yuasa H,

    Nakayama H, Horita S and Sugiyama Y (2010) Potent and specific inhibition of

    mMate1-mediated efflux of type I organic cations in the liver and kidney by

    pyrimethamine. J Pharmacol Exp Ther 333:341-350.

    Ji C, Tschantz WR, Pfeifer ND, Ullah M and Sadagopan N (2012) Development of a

    multiplex UPLC-MRM MS method for quantification of human membrane

    transport proteins OATP1B1, OATP1B3 and OATP2B1 in in vitro systems and

    tissues. Anal Chim Acta 717:67-76.

    Jones HM, Barton HA, Lai Y, Bi Y-a, Kimoto E, Kempshall S, Tate SC, El-Kattan A,

    Houston JB, Galetin A and Fenner KS (2012) Mechanistic pharmacokinetic

    modelling for the prediction of transporter-mediated disposition in human from

    sandwich culture human hepatocyte data Drug Metab Dispos Published online

    before print February 16, 2012, doi: 10.1124/dmd.111.042994

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    30

    Kajosaari LI, Niemi M, Neuvonen M, Laitila J, Neuvonen PJ and Backman JT (2005)

    Cyclosporine markedly raises the plasma concentrations of repaglinide. Clin

    Pharmacol Ther 78:388-399.

    Kimoto E, Chupka J, Xiao Y, Bi YA and Duignan DB (2011) Characterization of digoxin

    uptake in sandwich-cultured human hepatocytes. Drug Metab Dispos 39:47-53.

    Kotani N, Maeda K, Watanabe T, Hiramatsu M, Gong LK, Bi YA, Takezawa T, Kusuhara

    H and Sugiyama Y (2011) Culture period-dependent changes in the uptake of

    transporter substrates in sandwich-cultured rat and human hepatocytes. Drug

    Metab Dispos 39:1503-1510.

    Kullak-Ublick GA, Beuers U and Paumgartner G (2000) Hepatobiliary transport. J

    Hepatol 32:3-18.

    Kyrklund C, Backman JT, Neuvonen M and Neuvonen PJ (2003) Gemfibrozil increases

    plasma pravastatin concentrations and reduces pravastatin renal clearance. Clin

    Pharmacol Ther 73:538-544.

    Lai Y (2009) Identification of interspecies difference in hepatobiliary transporters to

    improve extrapolation of human biliary secretion. Expert Opin Drug Metab

    Toxicol 5:1175-1187.

    Lau YY, Huang Y, Frassetto L and Benet LZ (2007) Effect of OATP1B transporter

    inhibition on the pharmacokinetics of atorvastatin in healthy volunteers. Clin

    Pharmacol Ther 81:194-204.

    Lemahieu WP, Hermann M, Asberg A, Verbeke K, Holdaas H, Vanrenterghem Y and

    Maes BD (2005) Combined therapy with atorvastatin and calcineurin inhibitors:

    no interactions with tacrolimus. Am J Transplant 5:2236-2243.

    Letschert K, Faulstich H, Keller D and Keppler D (2006) Molecular characterization and

    inhibition of amanitin uptake into human hepatocytes. Toxicol Sci 91:140-149.

    Li N, Bi YA, Duignan DB and Lai Y (2009) Quantitative expression profile of

    hepatobiliary transporters in sandwich cultured rat and human hepatocytes. Mol

    Pharm 6:1180-1189.

    Li N, Nemirovskiy OV, Zhang Y, Yuan H, Mo J, Ji C, Zhang B, Brayman TG, Lepsy C,

    Heath TG and Lai Y (2008) Absolute quantification of multidrug resistance-

    associated protein 2 (MRP2/ABCC2) using liquid chromatography tandem mass

    spectrometry. Anal Biochem 380:211-222.

    Li N, Singh P, Mandrell KM and Lai Y (2010) Improved extrapolation of hepatobiliary

    clearance from in vitro sandwich cultured rat hepatocytes through absolute

    quantification of hepatobiliary transporters. Mol Pharm 7:630-641.

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    31

    Liu X, Chism JP, LeCluyse EL, Brouwer KR and Brouwer KL (1999) Correlation of

    biliary excretion in sandwich-cultured rat hepatocytes and in vivo in rats. Drug

    Metab Dispos 27:637-644.

    LIVALO Label of LIVALO

    http://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?id=31751#s38.

    McGinnity DF, Soars MG, Urbanowicz RA and Riley RJ (2004) Evaluation of fresh and

    cryopreserved hepatocytes as in vitro drug metabolism tools for the prediction of

    metabolic clearance. Drug Metab Dispos 32:1247-1253.

    Meier PJ, Eckhardt U, Schroeder A, Hagenbuch B and Stieger B (1997) Substrate

    specificity of sinusoidal bile acid and organic anion uptake systems in rat and

    human liver. Hepatology 26:1667-1677.

    Muck W (2000) Clinical pharmacokinetics of cerivastatin. Clin Pharmacokinet 39:99-

    116.

    Muck W, Mai I, Fritsche L, Ochmann K, Rohde G, Unger S, Johne A, Bauer S, Budde K,

    Roots I, Neumayer HH and Kuhlmann J (1999) Increase in cerivastatin systemic

    exposure after single and multiple dosing in cyclosporine-treated kidney

    transplant recipients. Clin Pharmacol Ther 65:251-261.

    Obach RS (1999) Prediction of human clearance of twenty-nine drugs from hepatic

    microsomal intrinsic clearance data: An examination of in vitro half-life approach

    and nonspecific binding to microsomes. Drug Metab Dispos 27:1350-1359.

    Obach RS (2009) Predicting drug-drug interactions from in vitro drug metabolism data:

    challenges and recent advances. Curr Opin Drug Discov Devel 12:81-89.

    Park JW, Siekmeier R, Lattke P, Merz M, Mix C, Schuler S and Jaross W (2001)

    Pharmacokinetics and pharmacodynamics of fluvastatin in heart transplant

    recipients taking cyclosporine A. J Cardiovasc Pharmacol Ther 6:351-361.

    Park JW, Siekmeier R, Merz M, Krell B, Harder S, Marz W, Seidel D, Schuler S and

    Gross W (2002) Pharmacokinetics of pravastatin in heart-transplant patients

    taking cyclosporin A. Int J Clin Pharmacol Ther 40:439-450.

    Pham PA, la Porte CJ, Lee LS, van Heeswijk R, Sabo JP, Elgadi MM, Piliero PJ,

    Barditch-Crovo P, Fuchs E, Flexner C and Cameron DW (2009) Differential

    effects of tipranavir plus ritonavir on atorvastatin or rosuvastatin

    pharmacokinetics in healthy volunteers. Antimicrob Agents Chemother 53:4385-

    4392.

    Regazzi MB, Iacona I, Campana C, Raddato V, Lesi C, Perani G, Gavazzi A and Vigano

    M (1993) Altered disposition of pravastatin following concomitant drug therapy

    with cyclosporin A in transplant recipients. Transplant Proc 25:2732-2734.

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    32

    Schneck DW, Birmingham BK, Zalikowski JA, Mitchell PD, Wang Y, Martin PD,

    Lasseter KC, Brown CD, Windass AS and Raza A (2004) The effect of

    gemfibrozil on the pharmacokinetics of rosuvastatin. Clin Pharmacol Ther

    75:455-463.

    Simonson SG, Raza A, Martin PD, Mitchell PD, Jarcho JA, Brown CD, Windass AS and

    Schneck DW (2004) Rosuvastatin pharmacokinetics in heart transplant recipients

    administered an antirejection regimen including cyclosporine. Clin Pharmacol

    Ther 76:167-177.

    Turncliff RZ, Hoffmaster KA, Kalvass JC, Pollack GM and Brouwer KL (2006)

    Hepatobiliary disposition of a drug/metabolite pair: Comprehensive

    pharmacokinetic modeling in sandwich-cultured rat hepatocytes. J Pharmacol

    Exp Ther 318:881-889.

    Vavricka SR, Van Montfoort J, Ha HR, Meier PJ and Fattinger K (2002) Interactions of

    rifamycin SV and rifampicin with organic anion uptake systems of human liver.

    Hepatology 36:164-172.

    Wang EJ, Casciano CN, Clement RP and Johnson WW (2003) Fluorescent substrates of

    sister-P-glycoprotein (BSEP) evaluated as markers of active transport and

    inhibition: evidence for contingent unequal binding sites. Pharm Res 20:537-544.

    Watanabe T, Kusuhara H, Maeda K, Shitara Y and Sugiyama Y (2009) Physiologically

    based pharmacokinetic modeling to predict transporter-mediated clearance and

    distribution of pravastatin in humans. J Pharmacol Exp Ther 328:652-662.

    Wu CY and Benet LZ (2005) Predicting drug disposition via application of BCS:

    transport/absorption/ elimination interplay and development of a

    biopharmaceutics drug disposition classification system. Pharm Res 22:11-23.

    Yamazaki M, Li B, Louie SW, Pudvah NT, Stocco R, Wong W, Abramovitz M, Demartis

    A, Laufer R, Hochman JH, Prueksaritanont T and Lin JH (2005) Effects of

    fibrates on human organic anion-transporting polypeptide 1B1-, multidrug

    resistance protein 2- and P-glycoprotein-mediated transport. Xenobiotica 35:737-

    753.

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    33

    Footnotes:

    The authors were all employees of Pfizer during this research and declare no conflicts of

    interest. YB and EK are equal contributors.

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    34

    Figure Legends

    Figure 1. Inhibitory effect of CsA , gemfibrozil and Rif SV on rosuvastatin uptake in

    SCHH. The uptake of rosuvastatin (1µM) was measured at 37 ºC in the presence and

    absence of CsA (10 µM), gemfibrozil (30 µM) and rif SV (100 µM). Data are presented

    as mean ± SD.

    Figure 2. Hepatic uptake and biliary excretion of several compounds in SCHH. The

    hepatic uptake was investigated at 37 ºC in the presence and absence of Rif SV (100 µM)

    or in the buffer with/without Ca2+/Mg2+. A: midazolam (1µM), B: buprenorphine (0.2µM),

    C: TC (1µM) and D: rosuvastatin (1µM). Data are presented from single studies run in

    duplicate or triplicate. A minimum of two experiments were performed on different day

    to verify coefficient of variation (CV%)

  • DMD #43489

    35

    TABLES

    Table 1. Target peptides and MRM transitions monitored for human OATP proteins

    m/z

    Protein Peptide Q1 Q3

    OATP1B1 NVTGFFQSFK 587.8 961.4, 860.4, 656.3

    NVTGFFQSFK* 591.8 969.4, 868.4, 664.3

    OATP1B3 NVTGFFQSLK 570.8 927.4, 826.4, 622.3

    NVTGFFQSL*K 574.3 934.4, 833.5, 629.3

    OATP2B1 SSPAVEQQLLVSGPGK 798.9 711.9, 445.2, 1155.6

    SSPAVEQQLLVSGPGK* 802.9 715.9, 453.2, 1163.6

    m/z: mass to charge ratio of the ion. *, stable isotope labeled amino acid.

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • DMD #43489

    36

    Table 2. Quantification of OATP1B1, 1B3 and 2B1 in suspension hepatocytes and SCHH

    of lot Hu4165. The protein expression of OATP1B1, 1B3, and 2B1 in SCHH was

    measured by LC-MS/MS and compared with that in suspension hepatocytes. * P

  • DMD #43489

    37

    Table 3. Comparison of initial rate of hepatocyte uptake in absence or presence of Rif SV.

    Substrates were incubated in the presence and absence of the inhibitor Rif SV (100 µM)

    for passive and active uptake, or in the buffer with/without Ca2+ for in vitro biliary

    clearance (CLbile). Data are presented from single studies run in duplicate or triplicate. A

    minimum of two experiments were performed on different day to verify coefficient of

    variation (CV%)

  • DMD #43489

    38

    Table 4. OATP related drug-drug interactions and hepatic active uptake in SCHH.

    Clinical substrates were incubated in the presence and absence of the inhibitor Rif SV

    (100 µM) for passive and active uptake, or in the buffer with/without Ca2+ for in vitro

    biliary clearance (CLbile). Data are presented as the mean of duplicates or as the mean

    (SD) of 3~5 experiments.

    Inhibitors Substrates

    Control Rif SV (100 μΜ) Active uptake (%)

    AUC changes (Fold)

    Cmax changes (Fold)

    Reference Substrate concentrat

    ion (μM)

    PSuptake CLbil

    e PSpassive CLbile

    uL/min/mg protein

    CsA

    Atorvastatin

    1 22(5.9) 1.4 9.3(1.3) ~0 58 (6)

    14.3 12.6 (Lemahieu et al., 2005)

    Atorvastatin 7.7 9.65 (Hermann et al., 2004)

    Atorvastatin 6.44 5.59 (Asberg et al., 2001)

    Bosentan 1 45 1.9 8 ~0 82 0.97 0.67 (Binet et al., 2000)

    Cerivastatin 1 31.8(5.0) 0.2 15(1.7) ~0 55 (3) 3.75 5.0 (Muck et al., 1999)

    Fluvastatin

    1 31(8.4) 3.3 11(7.5) ~0 72(7.1)

    2.10 5.0 (Park et al., 2001)

    Fluvastatin 2.55 3.11 (Park et al., 2001)

    Fluvastatin 0.94 0.3 (Goldberg and Roth,

    1996)

    Fluvastatin 0.89 1.65 (Holdaas et al., 2006)

    Pitavastatin 1 36.5(4.2) 0.4 11.9(0.7) 67(5.6) 3.6 5.6 (LIVALO)

    pravastatin 1 2.4 (0.91) 0.4 0.5 (0.35) 0.1 79.1(18)

    12.2 7.4 (Park et al., 2002)

    pravastatin 8.93 6.78 (Hedman et al., 2004) Pravastatin 21.8 6.9 (Regazzi et al., 1993)

    Rosuvastatin 1 8.3 (1.5) 2.6 1.5(1.0) 0.5 84(14) 6.08 9.6 (Simonson et al., 2004)

    Repaglinide 0.2 61(38) 0.2 24(6.4) 0.4 55(17.7) 1.44 0.75 (Kajosaari et al., 2005)

    Erythromycin Pitavastatin 1 36.5(4.2) 0.4 11.9(0.7) 67(5.6) 1.8 2.6 (LIVALO)

    Gemfibrozil

    Cerivastatin 1 31.8(5.0) 0.2 15(1.7) ~0 55 (3) 4.59 2.07 (Backman et al., 2002)

    Pravastatin 1 2.4 (0.91) 0.4 0.5 (0.35) 0.1 79.1(18) 1.02 0.81 (Kyrklund et al., 2003)

    Rosuvastatin 1 8.3 (1.5) 2.6 1.5(1.0) 0.5 84(14) 0.88 1.21 (Schneck et al., 2004)

    lopinavir and ritonavir

    Atorvastatin 1 22(5.9) 1.4 9.3(1.3) ~0 58 (6) 6.82 (2.4) 11.7 (7.6) (Lau et al., 2007)

    Bosentan 1 45 1.9 8 ~0 82 (16) 4.22 5.12 (Dingemanse et al., 2010)

    Rifampin

    Atorvastatin 1 22(5.9) 1.4 9.3(1.3) ~0 58 (6) 7.52 13.85 (He et al., 2009)

    Pravastatin 1 2.4 (0.91) 0.5 (0.35) 79.1(18) 1.33 1.73 (Deng et al., 2009)

    Atorvastatin 1 22(5.9) 1.4 9.3(1.3) ~0 58 (6) 8.36 7.61 (Pham et al., 2009)

    tipranavir and ritonavir

    Rosuvastatin 1 8.3 (1.5) 2.6 1.5(1.0) 0.5 84(14)

    0.37 1.33 (Pham et al., 2009)

    atazanavir and ritonavir

    Rosuvastatin 1.13 5.00 (Busti et al., 2008)

    This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/

  • This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on March 1, 2012 as DOI: 10.1124/dmd.111.043489

    at ASPE

    T Journals on July 3, 2021

    dmd.aspetjournals.org

    Dow

    nloaded from

    http://dmd.aspetjournals.org/