activation status of the pregnane x receptor (pxr ......2015/09/11  · humanized for cyp3a4/3a7,...

34
Vemurafenib Pharmacokinetics in Humanized Mice 1 Title: Activation status of the Pregnane X Receptor (PXR) influences Vemurafenib availability in humanized mouse models Authors: A. Kenneth MacLeod, Lesley A. McLaughlin, Colin J. Henderson and C. Roland Wolf*. Affiliations: Medical Research Institute, University of Dundee, Dundee DD1 9SY, United Kingdom. Running title: Vemurafenib Pharmacokinetics in Humanized Mice Keywords: Vemurafenib, melanoma, humanized mice, pharmacology, pregnane X receptor Financial support: This work was supported by Cancer Research UK programme grant C4639/A12330. on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

Upload: others

Post on 27-Jan-2021

3 views

Category:

Documents


0 download

TRANSCRIPT

  • Vemurafenib Pharmacokinetics in Humanized Mice

    1

    Title:

    Activation status of the Pregnane X Receptor (PXR) influences Vemurafenib availability in humanized mouse models

    Authors:

    A. Kenneth MacLeod, Lesley A. McLaughlin, Colin J. Henderson and C. Roland Wolf*.

    Affiliations:

    Medical Research Institute, University of Dundee, Dundee DD1 9SY, United Kingdom.

    Running title:

    Vemurafenib Pharmacokinetics in Humanized Mice

    Keywords:

    Vemurafenib, melanoma, humanized mice, pharmacology, pregnane X receptor

    Financial support:

    This work was supported by Cancer Research UK programme grant C4639/A12330.

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    2

    * Corresponding author:

    C. Roland Wolf,

    Medical Research Institute,

    University of Dundee,

    Dundee DD1 9SY,

    United Kingdom.

    Tel +441382383134

    Email: [email protected]

    Conflict of interest statement:

    The authors disclose no potential conflicts of interest.

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    3

    Abstract

    Vemurafenib is a revolutionary treatment for melanoma, but the magnitude of therapeutic response

    is highly variable and the rapid acquisition of resistance is frequent. Here, we examined how

    vemurafenib disposition, particularly through cytochrome P450-mediated oxidation pathways, could

    potentially influence these outcomes using a panel of knockout and transgenic humanized mouse

    models. We identified CYP3A4 as the major enzyme involved in the metabolism of vemurafenib in in

    vitro assays with human liver microsomes. However, mice expressing human CYP3A4 did not process

    vemurafenib to a greater extent than CYP3A4 null animals, suggesting that other pregnane X

    receptor (PXR)-regulated pathways may contribute more significantly to vemurafenib metabolism in

    vivo. Activation of PXR, but not of the closely related constitutive androstane receptor (CAR),

    profoundly reduced circulating levels of vemurafenib in humanized mice. This effect was

    independent of CYP3A4 and was negated by co-treatment with the drug efflux transporter inhibitor,

    elacridar. Finally, vemurafenib strongly induced PXR activity in vitro, but only weakly induced PXR in

    vivo. Taken together, our findings demonstrate that vemurafenib is unlikely to exhibit a clinically

    significant interaction with CYP3A4, but that modulation of bioavailability through PXR-mediated

    regulation of drug transporters (for example by other drugs) has the potential to markedly influence

    systemic exposure and thereby therapeutic outcomes. Activation status of the Pregnane X Receptor

    (PXR) influences Vemurafenib availability in humanized mouse models.

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    4

    Introduction

    The BRAF gene is mutated in approximately 50% of all malignant melanomas (1). In three quarters

    of these cases, mutation results in a valine to glutamic acid substitution at codon 600 (V600E),

    conferring an enhancement of kinase activity that promotes cellular proliferation and tumorigenesis

    (1-4). Vemurafenib (RG7204, PLX4032) was the first therapeutic agent clinically approved for the

    treatment of BRAFV600E-positive malignant melanoma. A highly specific inhibitor of the mutant

    kinase, vemurafenib elicits an objective tumour response in 48% of all patients with BRAFV600E-

    positive metastatic melanoma when given as a single agent, with increases in both overall and

    progression-free survival when compared to conventional chemotherapy with dacarbazine (5-7).

    Vemurafenib is both a revolutionary treatment for melanoma and, along with the companion

    diagnostic assay (8), a widely-cited example of the potential for therapeutic stratification based on

    mutational analysis. However, as with many recently developed clinically-approved kinase inhibitors,

    drug resistance is a major problem. The magnitude of initial response to vemurafenib is highly

    variable (5-7). While approximately half of patients meet the RECIST (Response Evaluation in Criteria

    in Solid Tumours) threshold for an objective response after 14 weeks of therapy, the remainder

    occupy a spectrum from non-objective response to disease progression, suggestive of varying

    degrees of innate resistance (7). Moreover, the median duration of response is only 6.7 months due

    to the acquisition of resistance (6). A large number of potential mechanisms of both innate and

    acquired resistance have been identified, most of which invoke the plasticity of signalling networks

    within the tumour to circumvent blockade to BRAFV600E function (9, 10). The majority of these studies

    have been conducted in cultured cell lines and tumour biopsy samples. There has been

    comparatively little effort to investigate whether the high inter-individual variability in drug

    exposure at steady state, with coefficients of variation of both Cmax and AUC of 30 – 40%, is a

    determinant of therapeutic outcome and/or resistance, despite the trends towards an exposure-

    response relationship in both overall and progression-free survival identified by population

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    5

    pharmacokinetic (PK) analyses(11-13), is a determinant of therapeutic outcome and/or resistance,

    with the notable exception of a recent study in which low circulating vemurafenib concentrations

    were associated with disease progression (14).

    During pre-clinical development, vemurafenib was found to be a substrate and possible inducer of

    CYP3A4, which was the major enzyme involved in its metabolism in vitro (11, 12, 15). Moreover,

    CYP3A4-generated monohydroxy-vemurafenib (OH-vemurafenib) was detected in a mass balance

    analysis in patients at steady-state (11, 12, 15). In this latter study, OH-vemurafenib levels were low

    compared to the parental compound but, as the absolute bioavailability of vemurafenib is unknown,

    it is impossible to draw any conclusions as to the relative contribution of metabolism in general, and

    CYP3A4 in particular, to elimination of the drug. Therefore if only a small percentage of vemurafenib

    reaches the systemic circulation, metabolism may yet constitute a major route of elimination and an

    important determinant of drug exposure. In addition to these metabolic considerations, post-

    approval studies using knockout mouse lines have identified Phase III drug efflux transporters as

    critical determinants of vemurafenib bioavailability (16, 17).

    CYP3A4 shows high inter- and intra-individual variation in expression, with differences in excess of

    100-fold (18). The primary mode of CYP3A4 regulation is transcriptional, with activation of two

    nuclear hormone receptors in particular, PXR and CAR, by exogenous ligands, endogenous ligands, or

    upstream signalling pathways known to increase its levels (19). In the present study, we have

    assessed the role of these metabolic factors in mediating vemurafenib disposition in both in vitro

    assays and in humanized mouse models in vivo. We have compared the relative contributions of

    metabolic and efflux pathways to vemurafenib PK, and have investigated the potential of

    vemurafenib to influence its own disposition.

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    6

    Materials and Methods

    Chemicals and reagents

    Crystalline vemurafenib and PLX4720 were purchased from Selleck chemicals (Houston, TX, USA).

    Microprecipitated bulk powder (MBP) vemurafenib was obtained through the Genentech Research

    Projects and Reagents Program (Genentech, San Francisco, CA, USA). Klucel LF Pharm was obtained

    from Ashland Inc. (Tadworth, UK). Elacridar hydrochloride was purchased from Sequoia Research

    Products (Pangbourne, UK). NADPH was purchased from Melford laboratories (Ipswich, UK).

    Protease inhibitors (cOmplete ULTRA, EDTA-free) were obtained from Roche (Burgess Hill, UK). All

    other chemicals were purchased from Sigma-Aldrich (Poole, UK).

    Animal lines and husbandry

    The generation and characterisation of Cyp3aKO/Cyp3a13+/+, huCYP3A4/3A7,

    huPXR/huCAR/huCYP3A4/3A7 and huPXR/huCAR mice has been described previously (20, 21).

    Briefly, the Cyp3aKO/Cyp3a13+/+ line bears a deletion of seven of the eight murine Cyp3a genes and

    exhibits low and uninducible oxidative metabolism of the CYP3A4 probe substrates triazolam,

    dibenzylfluorescein and midazolam (20). The huCYP3A4/3A7 line carries a large genomic insertion of

    human CYP3A4 and CYP3A7 in place of the Cyp3a locus (20). In this line, CYP3A4 is maintained at a

    low basal level but can be upregulated following ligand-activated nuclear translocation of the

    transcription factor, Pxr, most effectively with a synthetic glucocorticoid, 5-pregnenolone-3β-ol-20-

    one-16α-carbonitrile (PCN) (20). The huPXR/huCAR/huCYP3A4/3A7 line has been similarly

    humanized for CYP3A4/3A7, with additional humanisations for Pxr/PXR and Car/CAR (20). In this

    line, CYP3A4 can be induced by rifampicin (RIF), a ligand activator of human PXR (20). The

    huPXR/huCAR line carries the human transcription factors but retains the murine Cyp3a cluster (21).

    All animals were maintained under standard animal house conditions, with free access to food (RM1

    diet, Special Diet Services, Essex, UK) and water, and a 12-h light/12-h dark cycle. All animal work

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    7

    was carried in accordance with the Animal Scientific Procedures Act (1986) and after local ethical

    review.

    Subcellular fractionation

    Livers were excised and snap-frozen in liquid nitrogen for storage at -80°C until processing. Briefly,

    samples were thawed by the addition of 3 volumes of KCl buffer (1.15% w/v potassium chloride,

    10mM potassium phosphate, pH 7.4) and homogenised by rotor-stator. Debris was pelleted by

    centrifugation (11 000 x g at 4°C for 15 minutes) and the supernatant withdrawn for

    ultracentrifugation (100 000 x g at 4°C for 60 minutes). After ultracentrifugation, the supernatant

    (cytosolic fraction) was retained and the pellet (microsomal fraction) was resuspended in KCl buffer

    containing 0.25 M sucrose. Protein content of microsomal and cytosolic fractions was quantified by

    Bradford assay (Bio-Rad, Hemel Hempstead UK).

    In vitro studies

    All in vitro analyses were carried out with crystalline vemurafenib in 50 mM HEPES pH 7.4 containing

    30 mM MgCl2. Incubations were initiated ay the addition of NADPH to a final concentration of 1mM

    and terminated by the addition of 1x volume of ice cold acetonitrile. Microsomal stability assays

    were performed in triplicate with 1 µM compound and 0.2 mg/mL pooled human liver microsomes

    (HLM, 150 donors, Life Technologies, Paisley, UK) with or without NADPH for a period of up to 50

    min. HLM and mouse liver microsome (MLM, pooled from 3 wild-type mice) incubations for

    formation of the OH-vemurafenib metabolite contained 0.4 mg/ml microsomal protein and 60 µM

    vemurafenib. Incubations with HLM from individual donors for Spearman’s rank correlation were

    performed with 50 µM vemurafenib and 0.1 mg/mL protein for 6 min. Initial incubations with

    recombinant P450s were carried out with 60 µM compound and 400 pmol/mL enzyme for 15 min.

    Assays to determine the apparent kinetic parameters of OH-vemurafenib formation in HLM and

    MLM were performed in triplicate under conditions of linearity for time and protein: 10 min at 0.2

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    8

    mg/mL for HLM, 15 min at 0.4 mg/mL for MLM, 6 min at 160 pmol/mL for recombinant CYP3A4, and

    9 min at 160 pmol/mL for CYP2C9. For inhibition studies, pooled HLM were incubated at 0.2 mg/mL

    with 60 µM vemurafenib for 10 min in the presence of 1 µM ketoconazole, 2 µM sulfaphenazole, or

    both together. Inhibition of 7-benzyloxy quinolone (BQ) turnover by vemurafenib was measured

    using a BQ concentration of 30 µM and 0.2 mg/ml HLM with increasing concentrations of

    vemurafenib, and the fluorescent product measured using a Fluoroskan Ascent FL fluorimeter

    (Thermo Fisher, UK)

    PXR-transactivation assay

    Cells stably transfected with the PXR gene and a reporter construct with luciferase under the control

    of PXR-responsive elements of the CYP3A4 promoter (DPX2 cells, Puracyp Inc., Carlsbad, CA, USA)

    were treated with a range of concentrations of vemurafenib (0.06, 0.17, 0.51, 1.54, 4.62, 13.89,

    41.67 and 125 µM) in triplicate for 48 hours and assayed for luminescence and fluorescence as per

    the manufacturer’s instructions. The top two concentrations of vemurafenib were highly toxic so

    data from these points were excluded from the analysis. PXR activation (fold change) was calculated

    by dividing luminescence by fluorescence and normalising to vehicle control.

    In vivo studies

    All animal work was carried out on 8- to 12-week-old male mice (except for Figure 7, where female

    mice were used). PCN was suspended in corn oil at 1 mg/mL for administration by intraperitoneal

    (i.p.) injection at 10 mg/kg (10 µL per g body weight). RIF was administered in the same way at 60

    mg/kg. Phenobarbital (PB) was dissolved in PBS for i.p. administration at 80 mg/kg. For

    pharmacokinetic analyses, crystalline vemurafenib was first dissolved in DMSO then further diluted

    in a mixture of Polysorbate 80, ethanol and water (20:13:67) for immediate administration by oral

    gavage at a final dose of 50 mg/kg, as described by Durmus et. al. (16). The same method was used

    for administration of elacridar at 100 mg/kg (16). For sample collection, 10 µL of whole blood was

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    9

    withdrawn from the tail vein at the indicated time points. Samples were immediately added to a

    tube containing heparin solution (10 µL, 15 IU/mL) and stored at -20°C until processing. For

    induction studies, MBP vemurafenib was suspended in water containing 2% Klucel LF Pharm which

    had been adjusted to pH 4.0 by the addition of hydrochloric acid, and administered twice daily (eight

    hours apart) by oral gavage, as described by Yang et. al. (22).

    Sample processing for LC-MS/MS

    100 µL of water containing 100 µg/mL internal standard (PLX4720) was added to in vitro and in vivo

    samples, followed by 500 µL of diethyl ether (Fisher Scientific, Loughborough, UK). This mixture was

    incubated at room temperature for 30 minutes with gentle mixing. Samples were centrifuged for 10

    minutes at 16 000 x g and the organic solvent phase withdrawn to a fresh tube for drying under a

    vacuum (20 minutes at 30°C). The dried extract was re-dissolved in 100 µL of acetonitrile (Sigma) at

    37°C with agitation for 5 minutes and samples were added to 96-well plates for LC-MS/MS.

    LC-MS/MS

    Analysis of in vitro incubation and in vivo blood PK samples was carried out on a Waters Acquity

    UPLC and Micromass Quattro Premier mass spectrometer (both Micromass, Machester, UK). LC

    separation was performed on a Kinetex 1.7µ C19 100A; 50 x 2.1 mm column (Phenomenex,

    Macclesfield, UK) at a temperature of 45°C with an injection volume of 5 µL and flow rate of 0.6

    mL/min. Mobile phases were water containing 0.1% (v/v) formic acid (A) and acetonitrile containing

    0.1% (v/v) formic acid (B). Isocratic elution was carried out at 35%/65% A/B for 1 minute. Multiple

    reaction monitoring data in ESI-positive mode were acquired for vemurafenib (490.10 > 255.04, cone

    voltage (CV) = 65 V, collision energy (CE) = 40 kV), OH-vemurafenib (505.99 > 255.24, CV = 65 V, CE =

    40 kV) and PLX4720 (414.20 >306.97, CV = 60 V, CE = 37 kV). Acquired data were analysed in

    Quanlynx relative to analyte standard curves spanning the range of concentrations under study.

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    10

    Data analysis

    In vitro data exhibited non-Michaelis–Menten kinetics and therefore was fitted with the Hill

    equation using GraFit version 5 (Erithacus Software, Horley, UK). Standard deviations given are from

    the fit of the calculated curve. Pharmacokinetic parameters of in vivo data were calculated with a

    simple non-compartmental model using WinNonLin software, v4.1 (Pharsight, Munich, Germany)

    and are shown with standard deviations. P values were calculated using an unpaired t-test.

    Western blotting

    Microsomal and cytosolic samples were adjusted to 1 mg/mL in LDS sample buffer (Life

    Technologies) for electrophoresis through 10% acrylamide gels for 1 hr at 150 V, followed by transfer

    onto nitrocellulose membranes. Primary antibodies used for immunoblotting included anti-CYP3A4

    (458234, BD Biosciences, Oxford, UK), Cyp1a, Cyp2b, Cyp2c, Cyp3a/CYP3A and Por (23, 24). Anti-

    GRP78 (ab21685, Abcam, Cambridge, UK) and anti-GAPDH (G9545, Sigma) were used as loading

    controls for microsomal and cytosolic fractions, respectively. In blots for Cyp3a/CYP3A and CYP3A4,

    pooled HLM material was the same as used in microsomal incubations, while low, medium and high

    expression individuals were taken from the panel of 15 characterised donors based on testosterone

    6β-hydroxylation activity.

    mRNA analysis

    Total RNA was isolated from snap-frozen small intestinal samples using TRIzol (Life Technologies).

    The purity of this RNA was increased by processing with the RNeasy Mini Kit (Qiagen, Crawley, UK)

    followed by treatment with RG1 DNase (Promega, Southampton, UK). Conversion to cDNA was

    carried out using the ImProm-II Reverse Transcription System with random primers (Promega). The

    relative levels of mRNA species were determined by TaqMan RT-PCR (Life Technologies) using the

    following assays: Abcb1a (Mm00440761_m1), Abcb1b (Mm00440736_m1), Abcg2

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    11

    (Mm00496364_m1) and CYP3A4 (Hs00604506_m1). Fold changes were calculated by the

    comparative Ct method with 18S rRNA as an endogenous control (4319413E, Life Technologies).

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    12

    Results

    Vemurafenib is metabolised by CYP3A4 in vitro

    Metabolic stability of vemurafenib was assessed in pooled HLM (Figure 1A). At 1 µM, the compound

    was highly stable in both the presence and absence of NADPH, however formation of OH-

    vemurafenib was detectable in the presence of NADPH (Figure 1B). Apparent kinetic parameters of

    OH-vemurafenib formation by HLM and MLM were found to be sigmoidal, therefore were fitted

    using the Hill equation. The Vmax for HLM was approximately 10-fold higher than that observed in

    MLM (4879 ± 108 vs. 467 ± 54 peak area/min/mg, respectively) while the S50 was similar (23.2 ±0.8

    vs. 23.8 ± 4.9 µM, respectively, Figure 1C). Hill coefficients are detailed in Supplementary Table S1,

    alongside parameters calculated using the Michaelis-Menten model. In a panel of HLM from 15

    donors which had been characterised by the vendor with regard to their activity with probe

    substrates for individual CYP isoforms, formation of OH-vemurafenib correlated most strongly with

    that of 6β-hydroxy-testosterone, the marker for CYP3A4 (Supplementary Table S2 and Figure 1D).

    There was also a strong correlation with CYP2A6 activity (Supplementary Table S2), but this was

    deemed artefactual as CYP3A4 and CYP2A6 probe activities within the HLM panel correlate (data not

    shown), an observation which has previously been made in a separate batch of samples (23). Indeed,

    in incubations with recombinant CYP containing high concentrations of both protein and substrate,

    OH-vemurafenib was formed by CYP3A4, CYP2C9 and, to a lesser extent, by CYP2C8 and CYP1A1, but

    not by CYP2A6 (Figure 1E). The in vitro apparent kinetics of OH-vemurafenib formation by CYP3A4

    and CYP2C9 were, similarly to HLM and MLM, sigmoidal in nature. CYP3A4 had a 2-fold higher

    affinity for vemurafenib (S50: 10.0 ± 0.9 vs. 20.4 ± 1.7 µM) and a 3.4-fold higher Vmax (11693 ± 471 vs.

    3481 ± 216 peak area/min/nmol) than CYP2C9. As with the hepatic microsomal preparations, Hill

    coefficients and Michaelis-Menten analyses for recombinant P450s are detailed in Supplementary

    Table S1. In pooled HLM, co-incubation with the CYP3A4 inhibitor, ketoconazole, significantly

    inhibited OH-vemurafenib formation (Figure 1G). The CYP2C9 inhibitor, sulfaphenazole, effected a

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    13

    partial decrease in OH-vemurafenib formation. As expected, co-incubation with both inhibitors

    completely ablated OH-vemurafenib formation. Also in pooled HLM, vemurafenib inhibited the

    activity of the CYP3A4 probe drug substrate BQ with an IC50 of 14.9 µM (Figure 1H).

    Single-dose vemurafenib is not extensively metabolised by CYP3A4 in vivo

    In order to study the contribution of CYP3A4 to metabolism of vemurafenib in vivo,

    Cyp3aKO/Cyp3a13+/+ and huCYP3A4/3A7 mice were administered either PCN or vehicle (CO) alone

    for three days prior to administration of vemurafenib, as described in Materials and Methods. 24

    hours after the third dose of PCN, CYP3A4 is expressed in the liver of huCYP3A4/3A7 at a level similar

    to that observed in pooled HLM (data not shown). As stated above, Cyp3aKO/Cyp3a13+/+ mice are

    essentially null for Cyp3a/CYP3A activity, both basal and in response to activation of Pxr. Therefore

    any difference in the pharmacokinetics of vemurafenib between huCYP3A4/3A7 PCN and

    Cyp3aKO/Cyp3a13+/+ PCN is attributable to the activity of CYP3A4. Although not statistically

    significant, there was a decrease in both Cmax and AUCall, and an increase in CL, in the huCYP3A4/3A7

    PCN group, relative to the Cyp3aKO/Cyp3a13+/+ PCN group (Figure 2A, Supplementary Table S3).

    More pronounced, however, were these same changes in the PCN groups of both genotypes,

    relative to their respective vehicle control groups. The fact that PCN treatment enhanced clearance

    equally well in Cyp3a null and huCYP3A4/3A7 animals suggests that PXR-regulated pathways other

    than CYP3A4 predominate in the clearance of vemurafenib in vivo. Immediately after the final blood

    sample collection, livers were harvested and Western blotting carried out to confirm that CYP3A4

    had been effectively induced by PCN in the huCYP3A4/3A7 mice (Figure 2B). Due to this harvest

    occurring 48 hours after the last dose of PCN, CYP3A4 expression was lower than in pooled HLM due

    to turnover of the enzyme.

    Exposure to vemurafenib is decreased following activation of Pxr/PXR, but unaffected by activation of

    CAR

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    14

    As PCN pre-treatment decreased the systemic concentration of vemurafenib independently of

    CYP3A4 (Figure 2A, Supplementary Table S3), we further investigated the role of nuclear hormone

    receptor activity in mediating disposition to vemurafenib. HuPXR/huCAR/huCYP3A4/3A7 mice were

    pre-treated with RIF, a potent activator of human PXR and thus inducer of CYP3A4, leading to a

    marked reduction in the AUCall of vemurafenib from 432 ± 169 to 121 ± 18 hr*µg/mL (p = 0.0106,

    Figure 3A, Supplementary Table S4). Western blotting confirmed induction of CYP3A4 in the liver

    and, due to the high initial magnitude of this induction, CYP3A4 levels remained several fold higher

    than pooled HLM (and equivalent to HLM expressing high CYP3A4) despite 48 hours having elapsed

    after administration of the final dose of RIF (Figure 3B). In addition, we observed a 4.7-fold increase

    in mRNA for Abcb1a, but no change for Abcb1b or Abcg2, in the small intestine of

    huPXR/huCAR/huCYP3A4/3A7 mice treated with RIF (Figure 3C). Activation of PXR in the

    huPXR/huCAR mouse line led to a similar decrease in AUCall of vemurafenib from 349 ± 24 to 75 ± 33

    hr*µg/mL (p = 0.0003, Figure 3A, Supplementary Table S4).

    In order to determine whether CAR might play a role in modulating vemurafenib disposition,

    huPXR/huCAR/huCYP3A4/3A7 mice were dosed for three days with PB, a CAR activator (25), prior to

    vemurafenib administration. This had no effect on any of the kinetic parameters assessed (Figure 4A,

    Supplementary Table S5). Western blotting of liver samples demonstrated induction of the well-

    characterised Car target protein, Cyp2b10, confirming the effective activation of CAR by PB (Figure

    4B).

    Activation of PXR reduces vemurafenib exposure through induction of efflux transporters

    Activation of PXR by RIF in huPXR/huCAR/huCYP3A4/3A7 mice decreased the AUCall of vemurafenib

    (Figures 3A, 4A and 5A, Supplementary Tables S3, S4 and S5). This decrease was almost completely

    ablated by pre-treatment of mice, 90 minutes before administration of vemurafenib, with elacridar,

    an effective inhibitor of Abcb1 and Abcg2, but not CYP3A4, at the dose used (100 mg/kg p.o., Figure

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    15

    5A) (16, 26, 27). Relative to mice dose with RIF alone, AUCall of vemurafenib increased from 90 ± 13

    to 330 ± 85 hr*µg/mL (p = 0.0085) in animals dosed with RIF then elacridar, while Cmax increased

    from 11.1 ± 1.8 to 21.3 ± 2.2 µg/mL (p = 0.0032, Supplementary Table S6). Moreover,

    huPXR/huCAR/huCYP3A4/3A7 mice which had been pre-dosed with vehicle (CO) alone were subject

    to a similar magnitude of increase in exposure to vemurafenib following elacridar pre-treatment,

    with AUCall increasing from 391 ± 112 to 684 ± 85 hr*µg/mL (p = 0.0224) and Cmax increasing from

    29.1 ± 4.3 to 37.8 ± 3.5 µg/mL (p = 0.0507, Figure 5A). Again, effective activation of PXR by RIF was

    confirmed by Western blot of CYP3A4 in liver samples (Figure 5B).

    Vemurafenib is a strong inducer of PXR in vitro

    We tested the capacity of crystalline vemurafenib to activate a PXR-driven luciferase reporter

    construct in the DPX2 cell line (Puracyp Inc.). The reported aqueous solubility of this form is in the

    range of 0.01 – 10 µg/mL (approximately 20 nM – 20 µM) at physiological pH (12). An EC50 of 8.13

    µM and an EMAX of 8.54-fold (Figure 6) was determined. The positive control, rifampicin, gave an EC50

    of 1.01 µM and an EMAX of 14.10-fold (data not shown).

    Vemurafenib is a weak inducer of PXR in vivo

    We administered the high bioavailability MBP form of vemurafenib at a dose of 100 mg/kg b.i.d. for

    four days to huPXR/huCAR/huCYP3A4/3A7 mice, as described in Materials and Methods. The PK

    parameters of a single 100 mg/kg dose (Supplementary Table S7) were in good agreement with

    those calculated in a previous study (22). Western blotting of liver proteins demonstrated weak

    induction of CYP3A4 and Cyp2b, and moderate induction of Cyp1a (Figure 7A). Analysis of small

    intestinal mRNA found no change in expression in CYP3A4, Abcb1a or Abcb1b (Figure 7B).

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    16

    Discussion

    Vemurafenib is a revolutionary treatment for advanced melanoma and, along with the companion

    diagnostic test for BRAF mutation (8), is a successful example of cancer therapy stratification based

    on molecular-level information. As with other oncogenic kinases, however, clinical response to

    inhibition of BRAFV600 is highly variable and the acquisition of resistance in patients initially

    responsive to therapy is usually rapid (5-7). A wide variety of mechanisms of innate and acquired

    resistance have been identified, most of which are a function of the redundancy of the pro-

    proliferative signalling cascade being targeted (9, 10, 28). Data generated during the pre-clinical

    development of vemurafenib suggested several points of interaction with proteins involved in

    xenobiotic metabolism. These have the capacity to alter PK parameters and hence the therapeutic

    effectiveness of this agent (11, 12).

    In agreement with the pre-clinical data (11, 12), we found that CYP3A4 was the major CYP isoform

    involved in vemurafenib metabolism in vitro, but that the rate of metabolism was low. We also

    found that vemurafenib has the capacity to inhibit CYP3A4, with an IC50 for BQ activity of 14.9 µM. In

    addition, and in conflict with the manufacturer’s findings (11, 12), we identified a minor role of

    CYP2C9 in generating OH-vemurafenib, evidenced by a small but statistically significant decrease in

    OH-vemurafenib in pooled HLM co-incubated with the 2C9-specific inhibitor, sulfaphenazole.

    In a previous study, mass balance analysis in seven patients at steady state found that, in serum

    samples withdrawn up to 48 hours after administration of a radiolabelled dose of vemurafenib, 95%

    of recovered radioactivity was in the parental form (11, 12, 15). There was an increase in (the

    CYP3A4-generated form of) OH-vemurafenib from 0.5 to ~4% over this time period. It took 18 days

    for 95% of the radioactive dose to be recovered; 94% in faeces, 1% in urine (15). Metabolic profiling

    was carried out on the 68.5% of the total dose which was recovered within the first 96 hours. Over

    the 0 – 48 hr period, ≥ 94% of the radioactivity recovered was in the parental form. Over the 48 – 96

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    17

    hr period, however, this value decreased to 55.5 ± 20.1%. OH-vemurafenib accounted for 13.7% of

    the radioactivity recovered from the 48 – 96 hr samples (3.4% of the total dose). Conjugated

    (glucosylated and glucuronidated) forms of vemurafenib were the only other metabolites definitively

    identified in these samples (11, 12, 15). Although the percentage of vemurafenib excreted in the

    form of metabolites was low relative to the parent compound, it was acknowledged that a large

    proportion of the parent compound eliminated during the first 48 hr might constitute drug which

    had not been absorbed, and a significant proportion of the vemurafenib excreted during 48 - 96 h

    post-dose might have been generated by hepatobiliary recirculation. Indeed, it was also reported

    that metabolites predominated over the parent compound in a bile sample from one of the patients

    receiving vemurafenib (15). As the absolute bioavailibility of vemurafenib is unknown, and hence the

    contribution of metabolism to its elimination cannot be calculated, Phase IV clinical trials into the

    effect of an inhibitor and an inducer of CYP3A4 into the pharmacokinetics of vemurafenib have been

    requested by both the FDA and EMA. At present, an inhibitor (ketoconazole) study (NCT01765556)

    has been withdrawn and an inducer (rifampicin) study (NCT01765543) is recruiting

    (clinicaltrials.gov). Despite the occurrence of CYP-dependent hydroxylation of vemurafenib in vitro,

    we found that the in vivo effect of CYP3A4 in modulating the systemic levels of a single dose of the

    compound in humanized mice was small. Far more apparent was a decrease in AUCall following

    activation of Pxr, which was independent of CYP3A4 as it occurred in both the Cyp3aKO/Cyp3a13+/+

    and huCYP3A4/3A7 lines. This decrease was even more pronounced following activation of PXR in

    the huPXR/huCAR and huPXR/huCAR/huCYP3A4/3A7 mouse lines with RIF. The likely explanation for

    the differing magnitudes of this decrease is that, at the doses used, PCN-mediated activation of mPxr

    (and thereby induction of the downstream effector proteins) is less effective than RIF-mediated

    activation of hPXR (20).

    Pre-incubation of vemurafenib-resistant clones of the BRAFV600E-positive A375 melanoma cell line

    with verapamil, an ABCB1 inhibitor, partially increased their sensitivity to the drug (29). Studies with

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    18

    the MDCK-ABCB1 cell line demonstrated that vemurafenib was both an ABCB1 substrate and

    inhibitor (12), and a post-authorisation report mentions evidence of an interaction with ABCG2 (30).

    These observations have been confirmed independently, and studies in mice nulled for these

    transporters either separately or in combination have found that they modulate both intestinal

    bioavailability and permeability at the blood-brain barrier (16, 17, 31). This synergistic interaction of

    Abcb1 and Abcg2 is a phenomenon which has been shown to modulate the bioavailability of other

    therapeutics (32). ABCB1 is a transcriptional target of PXR in human cells (33, 34), and this

    interaction is thought to be conserved for Abcb1a in the mouse (35, 36). There is some evidence that

    Pxr also regulates Abcg2 (37). Our measurements of Abcb1a, Abcb1b and Abcg2 mRNA levels in the

    small intestine of huPXR/huCAR/huCYP3A4/3A7 mice demonstrate that, of these, only the first was

    induced following RIF treatment. Activation of CAR by PB induced Cyp2b10 to the same extent as

    activation of PXR by RIF, while also weakly inducing CYP3A4, but this pre-treatment had no effect on

    the PK parameters of vemurafenib. Taken together, these observations suggest that the Pxr/PXR-

    dependent modulation of vemurafenib PK is likely to be due to the upregulation of Abcb1a, and

    hence a decrease in the bioavailability of vemurafenib, rather than to induction of drug metabolism

    enzymes. Although PXR regulates the transcription of other drug metabolising enzymes in

    humanized mice, such as UDP-glucuronsyltransferases (UGT) of the 1a subfamily (38), our

    contention that the observed effects are due to the modulation of transport are supported by the

    finding that administration of the Abcb1/Abcg2 inhibitor, elacridar, 90 minutes prior to

    administration of vemurafenib completely ablated the effect of RIF pre-treatment in

    huPXR/huCAR/huCYP3A4/3A7 mice. Furthermore, elacridar pre-treatment of uninduced (i.e. not

    pre-treated with RIF) huPXR/huCAR/huCYP3A4/3A7 mice increased AUCall of vemurafenib by a

    degree comparable to pre-treatment of induced mice, suggesting that inhibition of efflux

    transporters in the basal state can profoundly increase vemurafenib bioavailability. It should be

    noted that, although elacridar is often cited as a specific inhibitor of ABCB1/Abcb1 and

    ABCG2/Abcg2, this is not the case. At the dose used in this study, the predicted plasma

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    19

    concentration of elacridar is an order of magnitude below the IC50 for CYP3A4 and other P450s (26,

    27). But this compound is known to inhibit SLCO1B1 (39) and is highly likely to inhibit other

    transporters (40). Therefore, although we cannot conclusively state that PXR-directed modulation of

    Abcb1a level in the small intestine is the pivotal determinant of AUCall of vemurafenib, this remains

    the most likely explanation. In support of this, studies in the Abcb1a/b and Bcrp knockout mouse

    lines suggested a predominant role for the former transporter (16). Increased drug efflux mediated

    by ABC transporter proteins in the cancer cell is a mechanism of multidrug resistance which has

    been understood for nearly 40 years (41). As a class of therapeutics, tyrosine kinase inhibitors, such

    as vemurafenib, are no less vulnerable to this process than conventional chemotherapeutics (42).

    The demonstration that drug transporters can markedly affect in vivo vemurafenib pharmacokinetics

    implies that changes in their expression in the tumour itself will also be a factor in drug response.

    Surprisingly, to date, this possibility has not been investigated.

    During pre-clinical development it was posited that decreases in the steady state blood plasma

    concentrations of vemurafenib in dogs on a twice-daily dosing regimen between 29 and 92 days

    might be due to the induction of metabolism (11). Furthermore, in patients at steady state,

    vemurafenib decreased AUC0-last of the CYP3A4-specific probe, midazolam, by 39%, increased AUC0-

    last of the CYP1A2 probe, caffeine, by 160%, and increased AUC0-last of the CYP2D6 probe,

    dextromethorphan, by 47% (11, 12). This defined vemurafenib as an inducer of CYP3A4, a moderate

    inhibitor of CYP1A2 and a weak inhibitor of CYP2D6. In agreement with these findings, we observed

    strong induction of CYP3A4-promoter-driven luciferase activity in a cell-line based reporter assay for

    PXR with crystalline vemurafenib. During the Phase I clinical trial, vemurafenib was reformulated

    from a crystalline to an MBP form to improve bioavailability (5). With this formulation, AUC0-24 is

    proportional to doses of between 240 and 960 mg bid (2, 5, 43). In the present study, we

    administered MBP vemurafenib to huPXR/huCAR/huCYP3A4/3A7 mice to assess the drug-drug

    interaction potential of this compound in this mouse model in vivo. Moreover, the AUCall of 1142 ±

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    20

    242 hr*µM observed at this dose is approximately four times the AUC0-24 of 300 hr*µM cited as

    being necessary to effect regression of COLO205 xenografts (44) and, with twice daily dosing, was

    considered to be sufficiently representative of the steady state level of exposure in humans (AUC0-24

    = 1741 ± 639 hr*µM, (5)). In addition, the Cmax of this single dose, at 121.7 ± 19.7 µM, is above the

    mean value of 86 ± 32 µM reported in patients at steady state (5). With these parameters, in silico

    analysis predicted that steady state concentrations in mice would be achieved within approximately

    32 hours (i.e. with the fourth dose) on a schedule of two doses per day, 8 hours apart (data not

    shown). Under these conditions we observed induction of CYP3A4 in the liver of

    huPXR/huCAR/huCYP3A4/3A7 mice administered a high dose of the MBP formulation twice daily for

    four days, although the level of this induction was slight and highly variable. Nevertheless, our in

    vitro and in vivo data demonstrate that vemurafenib has the capacity to induce CYP3A4. In addition,

    we also observed induction of Cyp1a and Cyp2b proteins in huPXR/huCAR/huCYP3A4/3A7 mice.

    These effects may, in part, be due to activation of PXR, but are likely also due to the activation of

    one or more additional transcription factors, such as the aryl hydrocarbon receptor (45). In addition

    to possible overall effects on drug PK, the capacity of vemurafenib to activate transcription factors

    involved in drug disposition raises the possibility that it may induce its own metabolism and efflux in

    tumour cells. This could potentially be a significant factor in the acquisition of drug resistance.

    Similar effects might also be invoked by co-medications which activate these transcription factors.

    In conclusion, we have found that vemurafenib interacts with the P450 system in vitro, but that PXR-

    directed regulation of efflux transporters and the consequent modulation of bioavailability is likely

    to be a more important factor in defining disposition in vivo. In addition, we have demonstrated that

    vemurafenib has the capacity to activate PXR, and potentially other transcription factors, at

    clinically-relevant concentrations. Taken together, our observations support the case for therapeutic

    monitoring of vemurafenib levels in patients over the course of therapy to evaluate whether low

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    21

    systemic drug concentrations coincide with innate or acquired resistance, and to determine whether

    modulators of PXR/CYP3A4/ABCB1 activity influence the outcome of vemurafenib therapy.

    Acknowledgements

    We would like to thank Julia Carr, Catherine Meakin and Susanne van Schelven for assistance with

    animal work, and Cheryl Wood, Mairi Lennie and Rumen Kostov for additional technical support. We

    thank Dr. Yury Kapelyukh for critical reading of the manuscript. We would also like to acknowledge

    Taconic Biosciences for the supply of the mouse lines used in this study.

    References

    1. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature 2002;417: 949-54. 2. Bollag G, Tsai J, Zhang J, Zhang C, Ibrahim P, Nolop K, et al. Vemurafenib: the first drug approved for BRAF-mutant cancer. Nat Rev Drug Discov 2012;11: 873-86. 3. Long GV, Menzies AM, Nagrial AM, Haydu LE, Hamilton AL, Mann GJ, et al. Prognostic and clinicopathologic associations of oncogenic BRAF in metastatic melanoma. J Clin Oncol 2011;29: 1239-46. 4. Hoeflich KP, Gray DC, Eby MT, Tien JY, Wong L, Bower J, et al. Oncogenic BRAF is required for tumor growth and maintenance in melanoma models. Cancer Res 2006;66: 999-1006. 5. Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med 2010;363: 809-19. 6. Sosman JA, Kim KB, Schuchter L, Gonzalez R, Pavlick AC, Weber JS, et al. Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib. N Engl J Med 2012;366: 707-14. 7. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011;364: 2507-16. 8. Halait H, Demartin K, Shah S, Soviero S, Langland R, Cheng S, et al. Analytical performance of a real-time PCR-based assay for V600 mutations in the BRAF gene, used as the companion diagnostic test for the novel BRAF inhibitor vemurafenib in metastatic melanoma. Diagn Mol Pathol 2012;21: 1-8. 9. Bucheit AD, Davies MA. Emerging insights into resistance to BRAF inhibitors in melanoma. Biochem Pharmacol 2014;87: 381-9. 10. Hartsough E, Shao Y, Aplin AE. Resistance to RAF inhibitors revisited. J Invest Dermatol 2014;134: 319-25. 11. http://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/202429s000TOC.cfm. 12. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Public_assessment_report/human/002409/WC500124400.pdf. 13. Alvarez JC, Funck-Brentano E, Abe E, Etting I, Saiag P, Knapp A. A LC/MS/MS micro-method for human plasma quantification of vemurafenib. Application to treated melanoma patients. J Pharm Biomed Anal 2014;97: 29-32.

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    22

    14. Funck-Brentano E, Alvarez JC, Longvert C, Abe E, Beauchet A, Funck-Brentano C, et al. Plasma vemurafenib concentrations in advanced BRAFV600mut melanoma patients: impact on tumour response and tolerance. Ann Oncol 2015. 15. Goldinger SM, Rinderknecht J, Dummer R, Kuhn FP, Yang KH, Lee L, et al. A single-dose mass balance and metabolite-profiling study of vemurafenib in patients with metastatic melanoma. Pharmacol Res Perspect 2015;3: e00113. 16. Durmus S, Sparidans RW, Wagenaar E, Beijnen JH, Schinkel AH. Oral availability and brain penetration of the B-RAFV600E inhibitor vemurafenib can be enhanced by the P-GLYCOprotein (ABCB1) and breast cancer resistance protein (ABCG2) inhibitor elacridar. Mol Pharm 2012;9: 3236-45. 17. Mittapalli RK, Vaidhyanathan S, Sane R, Elmquist WF. Impact of P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) on the brain distribution of a novel BRAF inhibitor: vemurafenib (PLX4032). J Pharmacol Exp Ther 2012;342: 33-40. 18. Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther 2013;138: 103-41. 19. Stanley LA, Horsburgh BC, Ross J, Scheer N, Wolf CR. PXR and CAR: nuclear receptors which play a pivotal role in drug disposition and chemical toxicity. Drug Metab Rev 2006;38: 515-97. 20. Hasegawa M, Kapelyukh Y, Tahara H, Seibler J, Rode A, Krueger S, et al. Quantitative prediction of human pregnane X receptor and cytochrome P450 3A4 mediated drug-drug interaction in a novel multiple humanized mouse line. Mol Pharmacol 2011;80: 518-28. 21. Scheer N, Ross J, Rode A, Zevnik B, Niehaves S, Faust N, et al. A novel panel of mouse models to evaluate the role of human pregnane X receptor and constitutive androstane receptor in drug response. J Clin Invest 2008;118: 3228-39. 22. Yang H, Higgins B, Kolinsky K, Packman K, Go Z, Iyer R, et al. RG7204 (PLX4032), a selective BRAFV600E inhibitor, displays potent antitumor activity in preclinical melanoma models. Cancer Res 2010;70: 5518-27. 23. Forrester LM, Henderson CJ, Glancey MJ, Back DJ, Park BK, Ball SE, et al. Relative expression of cytochrome P450 isoenzymes in human liver and association with the metabolism of drugs and xenobiotics. Biochem J 1992;281 ( Pt 2): 359-68. 24. Smith GC, Tew DG, Wolf CR. Dissection of NADPH-cytochrome P450 oxidoreductase into distinct functional domains. Proc Natl Acad Sci U S A 1994;91: 8710-4. 25. Mutoh S, Sobhany M, Moore R, Perera L, Pedersen L, Sueyoshi T, et al. Phenobarbital indirectly activates the constitutive active androstane receptor (CAR) by inhibition of epidermal growth factor receptor signaling. Sci Signal 2013;6: ra31. 26. Englund G, Lundquist P, Skogastierna C, Johansson J, Hoogstraate J, Afzelius L, et al. Cytochrome p450 inhibitory properties of common efflux transporter inhibitors. Drug Metab Dispos 2014;42: 441-7. 27. Ward KW, Azzarano LM. Preclinical pharmacokinetic properties of the P-glycoprotein inhibitor GF120918A (HCl salt of GF120918, 9,10-dihydro-5-methoxy-9-oxo-N-[4-[2-(1,2,3,4-tetrahydro-6,7-dimethoxy-2-isoquino linyl)ethyl]phenyl]-4-acridine-carboxamide) in the mouse, rat, dog, and monkey. J Pharmacol Exp Ther 2004;310: 703-9. 28. Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, Chan E, et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature 2012;487: 505-9. 29. Su F, Bradley WD, Wang Q, Yang H, Xu L, Higgins B, et al. Resistance to selective BRAF inhibition can be mediated by modest upstream pathway activation. Cancer Res 2012;72: 969-78. 30. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Procedural_steps_taken_and_scientific_information_after_authorisation/human/002409/WC500137805.pdf. 31. Wu CP, Sim HM, Huang YH, Liu YC, Hsiao SH, Cheng HW, et al. Overexpression of ATP-binding cassette transporter ABCG2 as a potential mechanism of acquired resistance to vemurafenib in BRAF(V600E) mutant cancer cells. Biochem Pharmacol 2013;85: 325-34.

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    23

    32. Kodaira H, Kusuhara H, Ushiki J, Fuse E, Sugiyama Y. Kinetic analysis of the cooperation of P-glycoprotein (P-gp/Abcb1) and breast cancer resistance protein (Bcrp/Abcg2) in limiting the brain and testis penetration of erlotinib, flavopiridol, and mitoxantrone. J Pharmacol Exp Ther 2010;333: 788-96. 33. Synold TW, Dussault I, Forman BM. The orphan nuclear receptor SXR coordinately regulates drug metabolism and efflux. Nat Med 2001;7: 584-90. 34. Geick A, Eichelbaum M, Burk O. Nuclear receptor response elements mediate induction of intestinal MDR1 by rifampin. J Biol Chem 2001;276: 14581-7. 35. Cheng X, Klaassen CD. Regulation of mRNA expression of xenobiotic transporters by the pregnane x receptor in mouse liver, kidney, and intestine. Drug Metab Dispos 2006;34: 1863-7. 36. Gu L, Chen J, Synold TW, Forman BM, Kane SE. Bioimaging real-time PXR-dependent mdr1a gene regulation in mdr1a.fLUC reporter mice. J Pharmacol Exp Ther 2013;345: 438-45. 37. Anapolsky A, Teng S, Dixit S, Piquette-Miller M. The role of pregnane X receptor in 2-acetylaminofluorene-mediated induction of drug transport and -metabolizing enzymes in mice. Drug Metab Dispos 2006;34: 405-9. 38. Lee SY, Lee JY, Kim YM, Kim SK, Oh SJ. Expression of hepatic cytochrome P450s and UDP-glucuronosyltransferases in PXR and CAR double humanized mice treated with rifampicin. Toxicol Lett 2015;235: 107-15. 39. Oostendorp RL, van de Steeg E, van der Kruijssen CM, Beijnen JH, Kenworthy KE, Schinkel AH, et al. Organic anion-transporting polypeptide 1B1 mediates transport of Gimatecan and BNP1350 and can be inhibited by several classic ATP-binding cassette (ABC) B1 and/or ABCG2 inhibitors. Drug Metab Dispos 2009;37: 917-23. 40. Oostendorp RL, Buckle T, Beijnen JH, van Tellingen O, Schellens JH. The effect of P-gp (Mdr1a/1b), BCRP (Bcrp1) and P-gp/BCRP inhibitors on the in vivo absorption, distribution, metabolism and excretion of imatinib. Invest New Drugs 2009;27: 31-40. 41. Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nat Rev Drug Discov 2006;5: 219-34. 42. Deng J, Shao J, Markowitz JS, An G. ABC Transporters in Multi-Drug Resistance and ADME-Tox of Small Molecule Tyrosine Kinase Inhibitors. Pharm Res 2014. 43. Grippo JF, Zhang W, Heinzmann D, Yang KH, Wong J, Joe AK, et al. A phase I, randomized, open-label study of the multiple-dose pharmacokinetics of vemurafenib in patients with BRAF V600E mutation-positive metastatic melanoma. Cancer Chemother Pharmacol 2014;73: 103-11. 44. Bollag G, Hirth P, Tsai J, Zhang J, Ibrahim PN, Cho H, et al. Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma. Nature 2010;467: 596-9. 45. Okino ST, Pendurthi UR, Tukey RH. Phorbol esters inhibit the dioxin receptor-mediated transcriptional activation of the mouse Cyp1a-1 and Cyp1a-2 genes by 2,3,7,8-tetrachlorodibenzo-p-dioxin. J Biol Chem 1992;267: 6991-8.

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    24

    Figure Legends

    Figure 1. Vemurafenib is metabolised by CYP3A4 and CYP2C9 in vitro.

    (A) Microsomal stability of vemurafenib with HLM in the absence (open circles) and presence (filled

    circles) of NADPH. (B) Formation of OH-vemurafenib from vemurafenib when incubated with HLM in

    the absence (open circles) and presence (filled circles) of NADPH. (C) Vemurafenib kinetics with HLM

    (open circles) and MLM (filled circles) fitted with the Hill equation. (D) Correlation of conversion of

    6β-OH-testosterone formation in characterised liver microsomes from 15 individuals with formation

    of OH-vemurafenib from vemurafenib. (E) Generation of OH-vemurafenib by recombinant CYP. (F)

    Vemurafenib kinetics with recombinant CYP2C9 (filled circles) and CYP3A4 (open circles) fitted with

    the Hill equation. (G) Inhibition of OH-vemurafenib formation in pooled HLM by ketoconazole and

    sulfaphenazole. (H) Inhibition of BQ activity in pooled HLM by vemurafenib (IC50 = 14.9 µM). In all

    cases data comprise mean ± SD of triplicate incubations.

    Figure 2. CYP3A4 has a modest effect on the pharmacokinetic profile of a single dose of

    vemurafenib in vivo.

    (A) Pharmacokinetic profiles of vemurafenib administered to huCYP3A4/3A7 PCN (filled circles, n=5),

    Cyp3aKO/Cyp3a13+/+ PCN (filled squares, n=4), huCYP3A4/3A7 Vehicle (open circles, n=5), and

    Cyp3aKO/Cyp3a13+/+ Vehicle (open squares, n=4) mice. Mean data ± SEM are shown. (B) Western

    blot of hepatic microsomal fractions from Cyp3aKO/Cyp3a13+/+ and huCYP3A4/3A7 mice dosed with

    either PCN or CO vehicle alone, alongside pooled HLM.

    Figure 3. Activation of PXR reduces exposure to vemurafenib independently of CYP3A4 activity.

    (A) Pharmacokinetic profiles of vemurafenib in huPXR/huCAR/huCYP3A4/3A7 Vehicle (open circles,

    n=4), huPXR/huCAR/huCYP3A4/3A7 RIF (closed circles, n=4), huPXR/huCAR Vehicle (open squares,

    n=3) and huPXR/huCAR RIF (closed squares, n=3) mice. Mean data ± SEM are shown. (B) Western

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    25

    blot of hepatic microsomal fractions from huPXR/huCAR/huCYP3A4/3A7 mice dosed with either RIF

    or CO vehicle alone, alongside HLM preparations; pooled and low, medium and high CYP3A4 activity.

    (C) TaqMan RT-PCR analysis of Abcb1a, Abcb1b and Abcg2 mRNA levels in whole small intestine of

    huPXR/huCAR/huCYP3A4/3A7 mice dosed with vehicle or RIF.

    Figure 4. Activation of CAR does not alter vemurafenib pharmacokinetics.

    (A) Pharmacokinetic profiles of vemurafenib in huPXR/huCAR/huCYP3A4/3A7 Vehicle (open circles,

    n=3), PB (closed circles, n=3) and RIF (closed squares, n=3). Mean data ± SEM are shown. (B)

    Western blots of hepatic microsomal fractions from huPXR/huCAR/huCYP3A4/3A7 mice dosed with

    either vehicle (PBS), PB, or RIF alongside HLM preparations; pooled and low, medium and high

    CYP3A4 activity.

    Figure 5. PXR regulates vemurafenib bioavailability through the modulation of efflux transporters.

    (A) Pharmacokinetic profiles of vemurafenib in huPXR/huCAR/huCYP3A4/3A7 mice treated with

    vehicle/vehicle (open circles, n=3), vehicle/elacridar (filled circles, dashed line, n=3), RIF/vehicle

    (open squares, n=3) and RIF/elacridar (filled squares, dashed line, n=3). Mean data ± SEM are shown.

    (B) Western blots of hepatic microsomal fractions from huPXR/huCAR/huCYP3A4/3A7 mice dosed

    with either CO (vehicle) or RIF, then with either DMSO/Tween 80/ethanol/water (vehicle) or

    elacridar 90 mins prior to vemurafenib, alongside HLM preparations; pooled and low, medium and

    high CYP3A4 activity.

    Figure 6. Vemurafenib activates PXR in vitro.

    DPX2 cells were used to calculate the EC50 (8.13 µM) and EMAX (8.54-fold) for vemurafenib mediated

    trans-activation of PXR

    Figure 7. Vemurafenib activates PXR and possibly other transcription factors in vivo.

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Vemurafenib Pharmacokinetics in Humanized Mice

    26

    (A) Western blots of hepatic microsomal fractions from huPXR/huCAR/huCYP3A4/3A7 mice dosed

    with either vehicle (4% Klucel LF, pH 4.0, n = 4) or MBP vemurafenib (n = 4) twice daily (8 hours

    apart) for four days. (B) TaqMan RT-PCR of CYP3A4, Abcb1a and Abcb1b in duodenum (Duo),

    jejunum (Jej) and ileum (Ile) of huPXR/huCAR/huCYP3A4/3A7 mice dosed with either vehicle or

    vemurafenib. Data shown are mean of triplicate analyses ± SD.

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • A B

    100

    80

    0

    20

    40

    60

    % V

    em

    ura

    fen

    ib r

    em

    ain

    ing

    120

    Time (min)0 10 20 40 5030

    Time (min)0 10 20 40 5030

    100

    80

    0

    20

    40

    60

    OH

    -ve

    mu

    rafe

    nib

    (pe

    ak a

    rea

    /min

    /mg

    ) 120

    00

    CYP1A

    2

    CYP2A

    6

    CYP2B

    6

    CYP2C

    8

    CYP2C

    9

    CYP2D

    6

    CYP3A

    4

    CYP2E

    1

    CYP1A

    10

    00

    % B

    Q a

    ctivity

    No

    inhibito

    r

    Ket

    ocon

    azole

    Sulfa

    phen

    azole

    Sulfa

    phen

    azole

    +

    keto

    cona

    zole

    8000

    6000

    2000

    4000

    0 ***

    *

    ***

    H

    DC

    G

    FE

    0 20 40 60 80 1000

    5000

    10000

    15000

    0 20 40 60 80 1000

    2000

    4000

    6000

    OH

    -ve

    mu

    rafe

    nib

    (pe

    ak a

    rea

    /min

    /mg

    )

    OH

    -ve

    mu

    rafe

    nib

    (pe

    ak a

    rea

    /min

    /mg

    )O

    H-v

    em

    ura

    fen

    ib(p

    ea

    k a

    rea

    /min

    /nm

    ol)

    OH

    -ve

    mu

    rafe

    nib

    (pe

    ak a

    rea

    /min

    /nm

    ol)

    OH

    -ve

    mu

    rafe

    nib

    (pe

    ak a

    rea

    /min

    /mg

    )

    6β-OH-testosterone(pmol/min/mg)

    [Vemurafenib] (µM)

    [Vemurafenib] (µM)

    [Vemurafenib] (µM)

    1200

    900

    600

    300

    10000

    8000

    6000

    2000

    4000

    4000 80001200016000

    100

    80

    60

    40

    20

    10 100

    Figure 1

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • A

    B

    HLM

    Cyp3aKO/Cyp3a13+/+

    Cyp3a/CYP3A

    huCYP3A4/3A7

    Vem

    uraf

    enib

    (µg/

    mL)

    Vehicle PCN Vehicle PCN

    Time (hours)0 10 20 30

    0

    5

    10

    15

    20

    25

    Cyp3aKO/Cyp3a13+/+ Vehicle

    Cyp3aKO/Cyp3a13+/+ PCNhuCYP3A4/3A7 Vehicle

    huCYP3A4/3A7 PCN

    Figure 2

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • AVe

    mur

    afen

    ib (µ

    g/m

    L)

    B

    C

    HLMVehicle

    Cyp3a/CYP3A

    RIF Pool Low Med High

    Vehicle RIF

    Fold

    cha

    nge

    Time (hours)0 10 20 30

    0

    10

    20

    30

    40huPXR/huCAR/huCYP3A4/3A7 VehiclehuPXR/huCAR/huCYP3A4/3A7 RIFhuPXR/huCAR VehiclehuPXR/huCAR RIF

    0

    2

    4

    6

    8 Abcb1aAbcb1bAbcg2

    Figure 3

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • A

    RIFHLM

    Pool Low Med High

    CYP3A4

    Cyp1a

    Cyp2b

    Cyp2c

    GRP78

    PBVehicle

    Cyp3a/CYP3A

    [Vem

    uraf

    enib

    ] (µg

    /mL)

    B

    Time (hours)0 10 20 30

    0

    10

    20

    30

    40

    50

    60

    70VehiclePBRIF

    Figure 4

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • A

    HLM

    CYP3A4

    Cyp1a

    Cyp2b

    Cyp2c

    GRP78

    VehicleVehicle

    Cyp3a/CYP3A

    [Vem

    uraf

    enib

    ] (µg

    /mL)

    B VehicleElacridar

    RIFElacridar

    RIFVehicle

    Time (hours)0 10 20 30

    0

    10

    20

    30

    40 Vehicle, VehicleVehicle, ElacridarRIF, VehicleRIF, Elacridar

    Figure 5

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • log [Vemurafenib] (µM)

    Fold

    cha

    nge

    -2 -1 0 1 20

    2

    4

    6

    8

    Figure 6

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • CYP3A4

    Cyp1a

    Cyp2b

    Cyp2c

    Grp78

    Vehicle

    Cyp3a/CYP3A

    VemurafenibHLM

    Pool Low Med HighA

    B

    0

    1

    2

    3

    4Duo VehicleDuo VemurafenibJej VehicleJej VemurafenibIle VehicleIle Vemurafenib

    CYP3A4 Mdr1a Mdr1b

    Fold

    cha

    nge

    Figure 7

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/

  • Published OnlineFirst September 11, 2015.Cancer Res A. Kenneth MacLeod, Lesley A McLaughlin, Colin J Henderson, et al. Vemurafenib availability in humanized mouse modelsActivation status of the Pregnane X Receptor (PXR) influences

    Updated version

    10.1158/0008-5472.CAN-15-1454doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://cancerres.aacrjournals.org/content/suppl/2015/12/12/0008-5472.CAN-15-1454.DC1

    Access the most recent supplemental material at:

    Manuscript

    Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected] at

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://cancerres.aacrjournals.org/content/early/2015/09/11/0008-5472.CAN-15-1454To request permission to re-use all or part of this article, use this link

    on June 6, 2021. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 11, 2015; DOI: 10.1158/0008-5472.CAN-15-1454

    http://cancerres.aacrjournals.org/lookup/doi/10.1158/0008-5472.CAN-15-1454http://cancerres.aacrjournals.org/content/suppl/2015/12/12/0008-5472.CAN-15-1454.DC1http://cancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://cancerres.aacrjournals.org/content/early/2015/09/11/0008-5472.CAN-15-1454http://cancerres.aacrjournals.org/

    Article FileFigure 1Figure 2Figure 3Figure 4Figure 5Figure 6Figure 7