throbber
[CANCER RESEARCH 64, 7099 –7109, October 1, 2004]
`
`BAY 43-9006 Exhibits Broad Spectrum Oral Antitumor Activity and Targets the
`
`RAF/MEK/ERK Pathway and Receptor Tyrosine Kinases Involved in Tumor
`
`Progression and Angiogenesis
`
`Scott M. Wilhelm,1 Christopher Carter,1 LiYa Tang,1 Dean Wilkie,1 Angela McNabola,1 Hong Rong,1 Charles Chen,1
`Xiaomei Zhang,1 Patrick Vincent,1 Mark McHugh,1 Yichen Cao,1 Jaleel Shujath,1 Susan Gawlak,1 Deepa Eveleigh,1
`Bruce Rowley,1 Li Liu,1 Lila Adnane,1 Mark Lynch,1 Daniel Auclair,1 Ian Taylor,1 Rich Gedrich,1
`Andrei Voznesensky,1 Bernd Riedl,1 Leonard E. Post,2 Gideon Bollag,2 and Pamela A. Trail1
`
`1Bayer Pharmaceuticals Corporation, West Haven, Connecticut; and 2Onyx Pharmaceuticals, Richmond, California
`
`ABSTRACT
`
`The RAS/RAF signaling pathway is an important mediator of tumor
`cell proliferation and angiogenesis. The novel bi-aryl urea BAY 43-9006 is
`a potent inhibitor of Raf-1, a member of the RAF/MEK/ERK signaling
`pathway. Additional characterization showed that BAY 43-9006 sup-
`presses both wild-type and V599E mutant BRAF activity in vitro.
`In addition, BAY 43-9006 demonstrated significant activity against several
`receptor tyrosine kinases involved in neovascularization and tumor
`progression,
`including vascular endothelial growth factor receptor
`(VEGFR)-2, VEGFR-3, platelet-derived growth factor receptor ␤, Flt-3,
`and c-KIT. In cellular mechanistic assays, BAY 43-9006 demonstrated
`inhibition of the mitogen-activated protein kinase pathway in colon, pan-
`creatic, and breast tumor cell lines expressing mutant KRAS or wild-type
`or mutant BRAF, whereas non–small-cell lung cancer cell lines expressing
`mutant KRAS were insensitive to inhibition of the mitogen-activated
`protein kinase pathway by BAY 43-9006. Potent inhibition of VEGFR-2,
`platelet-derived growth factor receptor ␤, and VEGFR-3 cellular receptor
`autophosphorylation was also observed for BAY 43-9006. Once daily oral
`dosing of BAY 43-9006 demonstrated broad-spectrum antitumor activity
`in colon, breast, and non–small-cell lung cancer xenograft models. Immu-
`nohistochemistry demonstrated a close association between inhibition of
`tumor growth and inhibition of the extracellular signal-regulated kinases
`(ERKs) 1/2 phosphorylation in two of three xenograft models examined,
`consistent with inhibition of the RAF/MEK/ERK pathway in some but not
`all models. Additional analyses of microvessel density and microvessel
`area in the same tumor sections using antimurine CD31 antibodies dem-
`onstrated significant inhibition of neovascularization in all three of the
`xenograft models. These data demonstrate that BAY 43-9006 is a novel
`dual action RAF kinase and VEGFR inhibitor that targets tumor cell
`proliferation and tumor angiogenesis.
`
`INTRODUCTION
`
`Many of the processes involved in tumor growth, progression, and
`metastasis are mediated by signaling pathways initiated by activated
`receptor tyrosine kinases (RTKs; ref. 1). RAS functions downstream
`of several RTKs, and activation of RAS signaling pathways is an
`important mechanism by which human cancer develops (2). Consti-
`tutive activation of the RAS pathways occurs through mutational
`activation of the RAS oncogene or of downstream effectors of RAS
`(3). RAS activation can also be exploited by overexpression of a
`variety of RTKs, including those for the epidermal (EGFR), platelet-
`derived (PDGFR), or vascular-endothelial (VEGFR) growth factors
`(4 –9). In this way, the majority of human tumors, not just those with
`
`Received 4/26/04; revised 7/14/04; accepted 7/29/04.
`The costs of publication of this article were defrayed in part by the payment of page
`charges. This article must therefore be hereby marked advertisement in accordance with
`18 U.S.C. Section 1734 solely to indicate this fact.
`Requests for reprints: Scott M. Wilhelm, Dept. Cancer Research, Bayer Pharmaceu-
`ticals Corporation, 400 Morgan Lane, West Haven, CT 06516. Phone: (203) 812-2961;
`Fax: (203) 812-6923; E-mail: scott.wilhelm.b@bayer.com.
`©2004 American Association for Cancer Research.
`
`RAS mutations, depend on activation of the RAS signal transduction
`pathways to achieve cellular proliferation and survival (4).
`RAS regulates several pathways that synergistically induce cellular
`transformation, including the well-characterized RAF/MEK/ERK cas-
`cade. RAF kinases are serine/threonine protein kinases that function in
`this pathway as downstream effector molecules of RAS. RAS local-
`izes RAF to the plasma membrane, where RAF initiates a mitogenic
`kinase cascade that ultimately modulates gene expression via the
`phosphorylation of transcription factors (3), which can have profound
`effects on cellular proliferation and tumorigenesis.
`The RAF kinase family is composed of three members: ARAF,
`BRAF, and Raf-1 (also termed c-Raf). BRAF is reportedly mutated in
`70% of malignant melanomas (10), in 33% of papillary thyroid
`carcinomas (11), and in lower frequencies in other cancers (12). The
`V599E mutant form of BRAF activates the RAF/MEK/ERK pathway
`in human melanoma cells in vitro, and small interfering RNA silenc-
`ing of V599E BRAF, but not Raf-1, inhibits soft agar growth of these
`cells (13). In addition, transformation of a melanocyte cell line with
`V599E BRAF activates the mitogen-activated protein kinase (MAPK)
`pathway. BAY 43-9006, a RAF kinase and VEGFR-2 inhibitor, and
`U0126, a MAP kinase kinase (MEK) inhibitor, block MAPK activa-
`tion and inhibit cell proliferation in mutant BRAF- and KRAS-
`transformed melanocytes (14).
`Recent evidence suggests that Raf-1 and BRAF participate in the
`regulation of endothelial apoptosis and, therefore, angiogenesis, a
`process essential for tumor development and metastasis (15, 16).
`Selective delivery of mutant Raf-1 to tumor blood vessels induces
`endothelial cell apoptosis, which inhibits angiogenesis and results in
`regression of established tumors (16). Mice deficient in BRAF or
`Raf-1 die during embryogenesis because of severe vascular defects
`and increased apoptosis that could be due, in part, to effects on
`endothelial cell survival (17, 18).
`Angiogenesis is a tightly regulated multistep process that involves
`the interaction of multiple growth factors expressed as multiple iso-
`forms, including VEGFs, basic fibroblast growth factor, and PDGFs.
`VEGF also regulates vascular permeability. Vessel stabilization
`through pericyte recruitment and maturation is primarily driven by
`PDGF (19). Several antiangiogenic agents are currently being inves-
`tigated in clinical trials (20 –24); however, because of the complex
`interactions between tumor cells, the invading stroma, and new blood
`vessels, a therapeutic agent targeting a single molecular entity might
`have limited efficacy across a spectrum of tumor types (25, 26).
`BAY 43-9006 is a novel bi-aryl urea that has been previously
`shown to inhibit Raf-1 and tumor cell line proliferation and tumor
`growth in several human tumor xenograft models (27, 28). Here, we
`demonstrate that BAY 43-9006 inhibits another member of the RAF
`family, wild-type (wt) BRAF and V599E BRAF. In addition, BAY
`43-9006 demonstrates potent
`inhibition of certain proangiogenic
`RTKs, including VEGFR-2, PDGFR-␤, and VEGFR-3. BAY 43-9006
`also substantially inhibits tumor growth of several human tumor
`
`7099
`Amerigen Exhibit 1106
`Amerigen v. Janssen IPR2016-00286
`
`

`
`BAY 43-9006, A DUAL ACTING RAF KINASE AND VEGFR INHIBITOR
`
`xenograft models, even in the absence of MAPK pathway inhibition.
`Taken together, these data suggest that BAY 43-9006 functions as a
`novel dual action RAF kinase and VEGFR inhibitor targeting both
`the RAF/MEK/ERK pathway and RTKs that promote tumor angio-
`genesis.
`
`MATERIALS AND METHODS
`
`Preparation of BAY 43-9006
`
`The chemical name of BAY 43-9006 is N-(3-trifluoromethyl-4-chlorophenyl)-
`N‘-(4-(2-methylcarbamoyl pyridin-4-yl)oxyphenyl)urea, and the structural for-
`mula is shown in Table 1. For in vitro experiments, BAY 43-9006 was dissolved
`in DMSO. For in vivo experiments, BAY 43-9006 was dissolved in Cremophor
`EL/ethanol (50:50; Sigma Cremophor EL, 95% ethyl alcohol) at 4-fold (4⫻) of the
`highest dose, foil wrapped, and stored at room temperature. This 4⫻ stock solution
`was prepared fresh every 3 days. Final dosing solutions were prepared on the day
`of use by dilution of the stock solution to 1⫻ with water. Lower doses were
`prepared by dilution of the 1⫻ solution with Cremophor EL/ethanol/water (12.5:
`12.5:75).
`
`Biochemical Assays
`
`In vitro Assays with Recombinant Raf-1 (Residues 305– 648), BRAF
`(Residues 409 –765), V599E BRAF (Residues 409 –765), MEK-1, and Ex-
`tracellular Signal-Regulated Kinase (ERK)-1. COOH-terminal kinase do-
`mains of Raf-1 (residues 305– 648) and BRAF (residues 409 –765) were
`generated by PCR. The BRAF (residues 409 –765) V599E mutation was
`introduced using the QuikChange Site-directed Mutagenesis kit (Stratagene,
`
`Table 1 BAY 43-9006 inhibits the RAF/MEK/ERK pathway and receptor tyrosine
`kinases involved in tumor angiogenesis
`
`Biochemical assay†
`Raf-1‡
`BRAF wild-type§
`V599E BRAF mutant¶
`VEGFR-2
`mVEGFR-2 (flk-1)
`mVEGR-3
`mPDGFR-␤
`Flt-3
`c-KIT
`FGFR-1
`ERK-1, MEK-1, EGFR, HER-2, IGFR-1, c-met, PKB,
`PKA, cdk1/cyclinB, PKC␣, PKC␥, pim-1
`Cellular mechanism储
`MDA MB 231 MEK phosphorylation (human breast)
`MDA MB 231 ERK 1/2 phosphorylation (human breast)
`BxPC-3 ERK 1/2 phosphorylation (human pancreatic)
`LOX ERK 1/2 phosphorylation (human melanoma)
`VEGFR-2 phosphorylation (human, NIH 3T3 cells)
`VEGF–ERK 1/2 phosphorylation (human, HUVEC)
`PDGFR-␤ phosphorylation (human HAoSMC)
`mVEGFR-3 phosphorylation (mouse, HEK-293 cells)
`Flt-3 phosphorylation (human ITD, HEK-293 cells)
`Cellular proliferation
`MDA MB 231 (10% FCS)
`PDGF-BB HAoSMC (0.1% BSA)
`
`IC50
`(nmol/L) ⫾ SD (n)*
`
`6 ⫾ 3 (7)
`22 ⫾ 6 (7)
`38 ⫾ 9 (4)
`90 ⫾ 15 (4)
`15 ⫾ 6 (4)
`20 ⫾ 6 (3)
`57 ⫾ 20 (5)
`58 ⫾ 20 (3)
`68 ⫾ 21 (3)
`580 ⫾ 100 (3)
`⬎10,000
`
`40 ⫾ 20 (2)
`90 ⫾ 26 (7)
`1,200** ⫾ 165 (2)
`880** ⫾ 90 (2)
`30 ⫾ 21 (3)
`60** ⫾ 26 (2)
`80 ⫾ 40 (3)
`100 ⫾ 80 (2)
`20 ⫾ 10 (2)
`
`2,600 ⫾ 810 (3)
`280 ⫾ 140 (5)
`
`* IC50 mean ⫾ SD; (n ⫽ number of trials).
`† Kinase assay were carried out as described in Materials and Methods at ATP
`concentrations at or below Km (1 to 10 ␮mol/L).
`‡ Lck activated NH2-terminal–truncated Raf-1.
`§ NH2-terminal–truncated BRAF (wild-type).
`¶ NH2-terminal V599E-truncated BRAF (mutant).
`储 Cellular mechanism assays (RTK autophosphorylation and RAF/MEK/ERK path-
`way) were performed in 0.1% BSA using phospho-specific antibodies or 4G10 for
`VEGFR-3 as described in Materials and Methods.
`** Activated phospho-ERK 1/2 was quantitated with phospho-ERK 1/2 immunoassay
`(Bio-Plex; Bio-Rad, Inc.).
`
`La Jolla, CA) according to the manufacturer’s protocol. Recombinant baculo-
`viruses expressing Raf-1 (residues 305– 648), BRAF (residues 409 –765), and
`V599E BRAF (residues 409 –765) were purified as fusion proteins as de-
`scribed previously (29). Full-length human MEK-1 was generated by PCR and
`purified as a fusion protein from Escherichia coli lysates (30).
`To test compound inhibition against various RAF kinase isoforms, BAY
`43-9006 was added to a mixture of Raf-1 (80 ng), wt BRAF, or V599E BRAF
`(80 ng) with MEK-1 (1 ␮g) in assay buffer [20 mmol/L Tris (pH 8.2), 100
`mmol/L NaCl, 5 mmol/L MgCl2, and 0.15% ␤-mercaptoethanol] at a final
`concentration of 1% DMSO. The RAF kinase assay (final volume of 50 ␮L)
`was initiated by adding 25 ␮L of 10 ␮mol/L ␥-[33P]ATP (400 Ci/mol) and
`incubated at 32°C for 25 minutes. Phosphorylated MEK-1 was harvested by
`filtration onto a phosphocellulose mat, and 1% phosphoric acid was used to
`wash away unbound radioactivity. After drying by microwave heating, a
`␤-plate counter was used to quantify filter-bound radioactivity. Activated
`MEK-1 and ERK-1 were purchased from Upstate Biotechnology (UBI, Wal-
`tham, MA) and assayed according to manufacturer’s instructions.
`In vitro Assays
`for Murine (m)VEGFR-2 (flk-1), mVEGFR-3,
`mPDGFR-␤, Flt-3, c-KIT, EGFR, HER2, c-MET, c-yes, FGFR-1, and
`IGFR-1. mVEGFR-2 (flk-1; residues 785-1367), human VEGFR-2 (KDR)
`kinase domain, mPDGFR-␤ (residues 560-1098), mVEGFR-3 (residues 818-
`1363), EGFR (residues 669-1210), HER2/neu (residues 691-1255), and
`FGFR-1 (residues 398 – 882) were expressed and purified from Sf9 lysates as
`described previously (29, 31). Flt-3, c-KIT, insulin growth factor receptor
`(IGFR)-1, VEGFR-2, c-MET, and cdk-1/cyclin B were purchased from Pro-
`qinase (Freiburg, Germany). Activated protein kinase (PK)B, PKA, LCK, and
`c-yes were purchased from Calbiochem, Inc. (San Diego, CA) and assayed
`according to the manufacturer’s instructions. BAY 43-9006 was assayed
`against recombinant pim-1 at Proqinase and PKC␣ and PKC␥ at Pan Labs
`(Bothell, WA).
`Time-resolved fluorescence energy transfer assays for mVEGFR-2 (flk-1),
`mVEGFR-3, mPDGFR-␤, Flt-3, c-KIT, EGFR, HER2, c-MET, c-yes, LCK,
`and IGFR-1 were performed in 96-well opaque plates in the time-resolved
`fluorescence energy transfer format. Final reaction conditions were as follows:
`1 to 10 ␮mol/L ATP, 25 nmol/L poly GT-biotin, 2 nmol/L Europium-labeled
`phospho (p)-Tyr antibody (PY20; Perkin-Elmer, Wellesley, MA), 10 nmol/L
`APC (Perkin-Elmer), 1 to 7 nmol/L cytoplasmic kinase domain in final
`concentrations of 1% DMSO, 50 mmol/L HEPES (pH 7.5), 10 mmol/L MgCl2,
`0.1 mmol/L EDTA, 0.015% Brij-35, 0.1 mg/mL BSA, and 0.1% ␤-mercapto-
`ethanol. Reactions volumes were 100 ␮L and were initiated by addition of
`enzyme. Plates were read at both 615 and 665 nmol/L on a Perkin-Elmer
`VictorV Multilabel counter at ⬃1.5 to 2.0 hours after reaction initiation. Signal
`was calculated as a ratio: (665 nm/615 nmol/L) ⫻ 10,000 for each well. Signal
`to noise was generally 4 to 8-fold in each assay.
`For IC50 generation, compounds were added before the enzyme initiation. A
`50-fold stock plate was made with compounds serially diluted 1:3 in a 50%
`DMSO/50% distilled water solution. Final compound concentrations ranged
`from 10 ␮mol/L to 4.56 nmol/L in 1% DMSO. The data were expressed as
`percent inhibition ⫽ 100 ⫺ [(signal with inhibitor ⫺ background)/(signal
`without inhibitor ⫺ background)] ⫻ 100.
`
`Cellular Mechanistic Assays
`
`Tumor Cell Lines and Reagents. The MDA-MB-231 human mammary
`adenocarcinoma cell line was obtained from the National Cancer Institute.
`These cells were maintained in DMEM (Invitrogen, Inc., Carlsbad, CA),
`supplemented with 1% L-glutamine (Invitrogen, Inc.), 1% HEPES buffer
`(Invitrogen, Inc.), and 10% heat-inactivated fetal bovine serum. The Colo-205,
`HT-29, and DLD-1 human colon carcinomas and the NCI-H460 and A549
`human non–small-cell lung cancer (NSCLC) carcinoma lines were obtained
`from and propogated as recommended by the American Type Tissue Culture
`Collection Repository (Manassas, VA).
`Cellular MEK 1/2, ERK 1/2, and PKB Activation and Bio-Plex pERK
`Immunoassay. Tumor cell lines were plated at 2 ⫻ 105 cells per well in
`12-well tissue culture plates in DMEM growth media (10% heat-inactivated
`FCS) overnight. Cells were washed once with serum-free media and incubated
`in DMEM supplemented with 0.1% fatty acid-free BSA (Sigma, St. Louis,
`MO) containing various concentrations of BAY 43-9006 in 0.1% DMSO for
`120 minutes to measure changes in basal pMEK 1/2, pERK 1/2, or pPKB.
`
`7100
`
`

`
`BAY 43-9006, A DUAL ACTING RAF KINASE AND VEGFR INHIBITOR
`
`Cells were washed with cold PBS (PBS containing 0.1 mmol/L vanadate) and
`lysed in a 1% (v/v) Triton X-100 solution containing protease inhibitors.
`Lysates were clarified by centrifugation, subjected to SDS-PAGE, transferred
`to nitrocellulose membranes, blocked in TBS-BSA, and probed with anti-
`pMEK 1/2 (Ser217/Ser221; 1:1000), anti-MEK 1/2, anti-pERK 1/2 (Thr202/
`Tyr204; 1:1000), anti-ERK 1/2, anti-pPKB (Ser473; 1:1000), or anti-PKB pri-
`mary antibodies (Cell Signaling Technology, Beverly, MA). Blots were
`developed with horseradish peroxidase (HRP)-conjugated secondary antibod-
`ies and developed with Amersham ECL reagent on Amersham Hyperfilm.
`A 96-well pERK immunoassay, using the laser flow cytometry (Bio-Rad,
`Hercules, CA) platform, was developed to measure BAY 43-9006 –mediated
`inhibition of basal pERK 1/2 in tumor cell lines. MDA-MB-231, LOX, and
`BxPC-3 cells were plated at 50,000 cells per well. One day after plating, tumor
`cells in DMEM with 0.1% fatty acid-free BSA were incubated for 2 hours with
`BAY compounds diluted to a final concentration of 3 ␮mol/L to 12 nmol/L in
`0.1% DMSO. Cells were incubated washed, lysed, and directly transferred to
`assay plate or frozen at ⫺80°C until processed. Tumor cell lysates were
`incubated with ⬃2000 of 5-␮m Bio-Plex beads conjugated with an anti-ERK
`1/2 antibody. The next day, biotinylated pERK 1/2 sandwich immunoassay
`was performed, beads were washed three times during each incubation, and
`phycoerythrin-streptavidin was used as a develop reagent. The relative fluo-
`rescence units of pERK 1/2 were detected by counting 25 beads with Bio-Plex
`flow cell (probe) at high sensitivity. The IC50 was calculated by taking
`untreated cells as maximum and no cells (beads only) as background using an
`Excel spreadsheet-based program.
`VEGFR-2 Autophosphorylation and MAPK Phosphorylation in Hu-
`man Umbilical Vascular Endothelial Cells (HUVECs) and NIH 3T3
`VEGFR-2–Transfected Cells. Subconfluent HUVECs (ATCC or Cambrex)
`were cultured in growth factor deprived culture medium for 24 hours. Cells
`were serum starved by replacing media with basal media (EBM-2) containing
`0.2% BSA for 1 hour. BAY 43-9006 was added to the cells with serum-free
`media for 1 hour followed by VEGF165 treatment (final concentration of 30
`ng/mL) for 10 minutes.
`NIH 3T3 cells transfected with VEGFR-2 were obtained from Dr. Mas-
`abumi Shibuya (Institute of Medical Science, University of Tokyo, Tokyo,
`Japan) and plated at 1 ⫻ 106 cells/well in 6-well tissue culture plates in
`DMEM, 10% fetal bovine serum, and 1.5 mg/mL G418. After 6 hours, culture
`medium was changed to 2 mL per well of 0.2% BSA/DMEM and incubated for
`14 hours. Cells were preincubated with compound added in 0.1% BSA/PBS for
`30 minutes followed by stimulation with 30 ng/mL VEGF165 for 10 minutes.
`Cells were washed and lysed with buffer containing 0.3% Triton X-100 and
`protease inhibitors (Complete Protease Inhibitor Tablets). Twelve ␮g of pro-
`tein from control and treated cell lysates were electrophoresed under reducing
`conditions and transferred onto nitrocellulose membranes. Blots were probed
`with anti-pVEGFR-2 (pTyr-1054 and pY1059) antibodies (PC460; Biosource,
`Inc., Camarillo, CA) or anti-VEGFR-2 antibody (sc-315; Santa Cruz Biotech-
`nology, Santa Cruz, CA). Blots were developed with HRP-conjugated second-
`ary antibodies and developed with Amersham ECL reagent on Amersham
`Hyperfilm.
`To monitor the effects of BAY 43-9006 on VEGF and basic fibroblast
`growth factor-dependent MAPK activation, exponentially growing human
`endothelial cells (HUVECs; Cambrex, East Rutherford, NJ) were seeded at
`25,000 cells per well in 96-well plates in growth medium with (EBM-2 MV;
`Cambrex) and incubated at 37°C in 5% CO2. Sixteen hours after plating, the
`cells were changed to serum-free RPMI 1640 containing 0.1% fatty acid-free
`BSA, and cells were preincubated at different concentrations of BAY43-9006.
`Cells were stimulated for 10 minutes with 50 ng/mL of either VEGF or basic
`fibroblast growth factor and processed for as described above for Bio-Plex
`pERK 1/2 immunoassay from tumor cell lysates.
`PDGFR-␤ Autophosphorylation and Cell Proliferation in Human Aor-
`tic Smooth Muscle Cells (HAoSMCs). A total of 1 ⫻ 105 HAoSMCs (P3-P6;
`Clonetics) were plated in 12-well cluster plates in 1 mL volume per well of
`SGM-2 (Clonetics). Cells were rinsed the next day with D-PBS (Life Tech-
`nologies, Inc.), then serum starved in 500 ␮L of smooth muscle cell basal
`media (Clonetics) with 0.1% BSA (Sigma) overnight. Diluted compounds
`ranged from 10 ␮mol/L to 1 nmol/L in 0.1% DMSO. Media was removed, and
`100 ␮L of each dilution were added to cells for 1 hour at 37°C. Cells were then
`stimulated with 10 ng/mL PDGF BB ligand (Leinco) for 7 minutes at 37°C.
`The media was decanted and 150 ␮L of isotonic 25 mmol/L bicine pH 7.6 lysis
`
`buffer (M-PER from Pierce) with protease inhibitor tablet (Complete; EDTA-
`free), and 0.2 mmol/L Na vanadate were added. Cells were lysed, and 15 ␮L
`of agarose-conjugated anti-PDGFR-␤ antibody (sc-339; Santa Cruz Biotech-
`nology) were added. Next day, beads were rinsed, boiled in 1⫻ LDS sample
`buffer, run on 3 to 8% gradient Tris-Acetate gels (Invitrogen), and transferred
`onto nitrocellulose. Membranes were probed with anti-pPDGFR-␤ (Tyr857)
`antibody (sc-12907; Santa Cruz Biotechnology) and then the secondary goat
`antirabbit HRP IgG (Amersham). Positive bands were visualized using en-
`hanced chemiluminescence. Subsequently, membranes were stripped and rep-
`robed with SC-339 (Santa Cruz Biotechnology) for total PDGFR-␤.
`The PDGF-dependent bromodeoxyuridine incorporation assay measures the
`ability of compounds to inhibit the induction of DNA synthesis by PDGF in
`serum-starved HAoSMCs. For the assay, HAoSMCs (4 ⫻ 103/well) were
`plated in complete SMBM media in 96-well tissue culture plates, incubated
`overnight, and then serum starved for 16 hours in SMBM containing 0.1%
`BSA (SF-SMBM). Fresh SF-SMBM was then added to the cells. Dilutions of
`BAY 43-9006 in SF-SMBM were added in a dose range from 10 ␮mol/L to
`4.57 nmol/L 1 hour before the addition of 10 ng/mL PDGF-BB. Cells were
`incubated for 24 hours and processed using the BrdUrd ELISA kit from
`Amersham.
`mVEGFR-3 and Human Flt-3 (ITD) Receptor Autophosphorylation
`Assays. The human embryonic kidney (HEK-293)-Flt-3 (ITD) cells (CRL-
`1573; American Type Cell Culture) were plated at 2.5 to 5 ⫻ 105 cells/well in
`6-well plates (RPMI ⫹10% FBS). The following day, cells were treated with
`inhibitors (3 ␮mol/L to 10 nmol/L) for 2 hours in serum-free RPMI media.
`Cells were lysed, and 10 ␮g of proteins per lane were loaded on 3 to 8%
`Tris-Acetate NuPAGE gels (Invitrogen). Gels were transferred to nitrocellu-
`lose membranes, blocked, probed with anti-pFlt-3 monoclonal antibody (3466;
`Cell Signaling), washed, and probed with secondary HRP-conjugated anti-
`mouse IgG (Amersham). Membranes were washed, developed with ECL
`Western blot detection reagent (Amersham), and exposed on Hyperfilm ECL
`film (Amersham). For total Flt-3 quantification, membranes were stripped with
`RESTORE buffer
`(Pierce) and blotted as described above using anti-
`Flt-3 polyclonal antibodies (sc-479; Santa Cruz Biotechnology) and HRP-
`conjugated antirabbit IgG (Amersham).
`For mVEGFR-3 studies, HEK-293 cells were transiently transfected with
`pcDNA3.1 vector (Invitrogen) containing a full-length cDNA of murine
`VEGFR3. Two days after transfection, cells were exposed to test compounds
`(3 ␮mol/L to 10 nmol/L in 0.1% DMSO) for 30 minutes at 37°C. Cells were
`washed, lysed in Triton-lysis buffer, pelleted, and 10 ␮g of total protein were
`loaded on 3 to 8% Tris-Acetate NuPAGE gels (Invitrogen). Gels were trans-
`ferred to nitrocellulose membranes, which were probed with anti-mVEGFR-3
`(ADI) or anti-p-mVEGFR-3 (4G10; Upstate Biotechnology) antibodies and
`secondary HRP-conjugated antimouse or antirabbit IgG, respectively (Amer-
`sham).
`
`Tumor Cell Proliferation
`
`Tumor cells were trypsinized and plated in 96-well plates at 3000 cells per
`well in complete media with 10% FCS. Cells were incubated overnight at
`37°C, and the next day, compounds were added in complete growth media over
`a final concentration range of 10 ␮mol/L to 10 nmol/L in 0.1% DMSO. Cells
`were incubated with test compounds for 72 hours at 37°C in complete growth
`media, and cell number was quantitated using the Cell TiterGlo ATP Lumi-
`nescent assay kit (Promega). This assay measures the number of viable cells
`per well by measurement of luminescent signal based on amount of cellular
`ATP.
`
`Tumor Xenograft Experiments
`
`Female NCr-nu/nu mice (Taconic Farms, Germantown, NY) were used for
`all studies. The mice were housed and maintained in accordance with Bayer
`Institutional Animal Care and Use Committee and state and federal guidelines
`for the humane treatment and care of laboratory animals and received food and
`water ad libitum.
`Tumors for all but the DLD-1 model were generated by harvesting cells
`from mid-log phase cultures using Trypsin-EDTA (Invitrogen, Inc.). Three to
`five million cells were injected s.c. into the right flank of each mouse. DLD-1
`tumors were established and maintained as a serial in vivo passage of s.c.
`fragments (3 ⫻ 3 mm) implanted in the flank using a 12-gauge trocar. A new
`
`7101
`
`

`
`BAY 43-9006, A DUAL ACTING RAF KINASE AND VEGFR INHIBITOR
`
`generation of the passage was initiated every three weeks, and studies were
`conducted between generations 3 and 12 of this line.
`Treatment was initiated when tumors in all mice in each experiment ranged
`in size from 75 to 144 mg for antitumor efficacy studies and from 100 to 250
`mg for studies of microvessel density and ERK phosphorylation. All treatment
`was administered orally once daily for the duration indicated in each experi-
`ment. Tumor weight was calculated using the equation length ⫻ (width)2)/2.
`Treatments producing ⬎20% lethality and/or 20% net body weight loss were
`considered toxic.
`
`Detection of Tumor Microvessels and Activated ERK 1/2
`
`Immunostaining of paraffin sections of tumors with murine anti-CD31
`antibodies was performed on the Dako Autostainer, model LV (DakoCytoma-
`tion). Sections were deparaffinized and hydrated, and endogenous peroxidase
`activity was blocked with 3% H2O2. Antigen retrieval was performed using
`Dako Target Retrieval Solution (S1699, DakoCytomation). Sections were
`blocked with an avidin/biotin block (Vector Laboratories) and rabbit serum.
`Immunostaining was performed using a goat Vectastain ABC elite kit (Vector
`Laboratories) and 3,3⬘-diaminobenzidine as the chromagen (DakoCytoma-
`tion), according to the manufacturer’s protocol with the following modifica-
`tions: (a) 2 ⫻ 30-minute incubations were performed with primary antibody;
`and (b) after incubation with the secondary antibody, the slides were rinsed in
`buffer, distilled water, and then buffer again. Sections were incubated with
`anti-CD31 antibodies [PECAM-1 (M-20) SC-1506 goat polyclonal; Santa
`Cruz Biotechnology] diluted 1:750 in Dako Antibody Diluent (DakoCytoma-
`tion, Carpinteria, CA) or goat IgG (1:750; Jackson ImmunoResearch Labora-
`
`tories, Inc., West Grove, PA) as a negative control. Sections were counter-
`stained with Mayer’s hematoxylin for 1 minute and washed with water.
`Immunohisotchemical localization of activated ERK 1/2 was determined
`from paraffin sections. Sections were then placed in heated (95°C) 0.1M
`citrate buffer (pH 6.0) for 35 min, brought to RT for 30 min, and then
`blocked with 1.5% H2O2. Sections were stained using primary pERK 1/2
`antibody (phospho-p44/42 Cell Signaling) diluted 1:100 with Dako Anti-
`body Diluent. Staining was performed using the Envision Plus HRP (3,3⬘-
`diaminobenzidine) System from Dako (4011) according to the manufac-
`turer’s protocol. The slides were counterstained with Mayer’s hematoxylin
`for 1 min and washed with water.
`
`Quantification of Microvessels
`
`Histologic slides were blind-coded during the assessment. The tissue
`sections were viewed at ⫻100 magnification (⫻10 objective lens and ⫻10
`ocular lens; 0.644 mm2 per field). Tissue image was captured with a digital
`camera (Diagnostic Instruments, Inc., Sterling Heights, MI). Four fields per
`section were randomly analyzed, excluding peripheral surrounding connec-
`tive tissues and central necrotic tissues. Total tissue area analyzed in each
`section was 2.576 mm2.
`Area and number of CD31 positive objects were quantified using the software
`ImagePro Plus version 3.0 (Media Cybernetics, Silver Spring, MD). Percentage of
`microvessel area (MVA, %) in each field was calculated as [(area of CD31-
`positive objects/measured tissue area) ⫻ 100%]. Microvessel density (MVD,
`number/mm2) in each field was calculated as (number of CD31-positive objects/
`0.644 mm2). Mean values of MVA and MVD in each group were calculated from
`
`Fig. 1. BAY 43-9006 inhibits activation of the
`RAF/MEK/ERK pathway in most but not all human
`tumor cell lines. A. MDA-MB-231 cells were incu-
`bated with various concentrations of BAY 43-9006 or
`DMSO. Cell lysates were subjected to Western blot
`analysis for phosphorylated (p) and total (T) MEK 1/2
`(top box), ERK 1/2 (middle box), and PKB (bottom
`box). Changes in total MEK 1/2, ERK 1/2 levels, or
`PKB were not observed. The MEK inhibitor U0126
`(10 ␮mol/L) was used in all Western blot experiments
`as a control for detecting RAF/MEK/ERK pathway
`inhibition (Lane 1: MEK-1). B. Subconfluent human
`tumor cells were incubated with BAY 43-9006 for 2
`hours, lysed, and processed for Western blotting. Ac-
`tivated ERK 1/2 was detected with anti-pERK anti-
`bodies. Changes in total ERK 1/2 levels were not
`observed. U0126 was used at 10 ␮mol/L (Lane 1:
`MEK-1).
`
`7102
`
`

`
`BAY 43-9006, A DUAL ACTING RAF KINASE AND VEGFR INHIBITOR
`
`four tumor samples. Data were analyzed statistically with one-way ANOVA
`followed by Fisher’s PLSD (StatView, version 4.5; Abacus Concepts, Inc., Berke-
`ley, CA). P ⬍ 0.05 was considered significant.
`
`RESULTS
`
`BAY 43-9006 Inhibition of the MAPK Pathway. BAY 43-9006
`is a synthetic molecule that can be broadly defined as a bi-aryl urea
`(Table 1), which was originally identified through inhibition of Raf-1
`kinase biochemical and cellular mechanistic assays (27, 32). BAY
`43-9006 was additionally profiled against wt BRAF and V599E
`mutant BRAF (Table 1). Biochemical assays were performed in which
`varying concentrations of BAY 43-9006 were tested for the capacity
`to inhibit MEK-1 phosphorylation by the catalytic domains of Raf-1,
`BRAF, and V599E BRAF. As shown in Table 1, BAY 43-9006
`potently inhibited Raf-1 (IC50, 6 nmol/L), wt BRAF (IC50, 22 nmol/
`L), and V599E mutant BRAF (IC50, 38 nmol/L) but did not signifi-
`cantly inhibit MEK-1 or ERK-1 activity (IC50, ⬎10,000 nmol/L).
`
`The ability of BAY 43-9006 to block activation of the MAPK pathway
`was examined by measuring ERK 1/2 phosphorylation in several tumor
`cell lines by Western blot analysis or Bio-Plex pERK immunoassay.
`Genotyping of each cell line revealed mutations in KRAS (Mia PaCa 2,
`HCT 116, A549, and NCI-H460), V599E BRAF (LOX melanoma,
`HT-29), or both KRAS and G463V BRAF (MDA-MB-231), suggesting
`that transformation of these cells is driven, in part, by disruption of the
`MAPK pathway. Results show that BAY 43-9006 inhibits ERK phos-
`phorylation in most of these cell lines, independent of which mutation
`caused aberrant activation of the RAS/RAF pathway (Fig. 1).
`In MDA-MB-231 breast cancer cells, BAY 43-9006 completely
`blocked activation of the MAPK pathway (Fig. 1A). Cells were
`preincubated with BAY 43-9006 at concentrations ranging from 0.01
`to 3 ␮mol/L, and dose-dependent inhibition of basal MEK 1/2 and
`ERK 1/2 phosphorylation (IC50, 40 and 100 nmol/L, respectively) was
`observed (Fig. 1A). BAY 43-9006 had no effect on the PKB pathway
`in MDA-MB-231 cells, demonstrating selectivity for inhibition of the
`MAPK, but not the PKB, pathway in these cells (Fig. 1A).
`
`tyrosine kinases
`receptor
`Fig. 2. BAY 43-9006 targets
`(VEGFR-2 and PDGFR-␤) involved in tumor angiogenesis. A,
`VEGF-stimulated VEGFR-2 autophosphorylation in HUVECs. B,
`VEGF-stimulated VEGFR-2 autophosphorylation in NIH 3T3
`VEGFR-2 cells. C, PDGF-BB–stimulated PDGFR-␤ phosphoryla-
`tion in HAoSMCs. D, PDGF-BB–stimulated bromodeoxyuridine
`(BrdUrd) proliferation assay in HAoSMCs.
`
`7103
`
`

`
`BAY 43-9006, A DUAL ACTING RAF KINASE AND VEGFR INHIBITOR
`
`BAY 43-9006 inhibition of pERK was also observed for the human
`pancreatic (Mia PaCa 2) and colon (HCT 116 and HT-29) tumor cell
`lines by Western blot analysis (Fig. 1B) and in the human LOX
`melanoma and pancreatic BxPC-3 cell lines using the Bio-Plex pERK
`immunoassay (Table 1). However, BAY 43-9006, at concentrations as
`high as 10 ␮mol/L, had no effect on inhibition of ERK 1/2 phospho-
`rylation in the two NSCLC cell lines (Fig. 1B).
`BAY 43-9006 Targets Receptor Tyrosine Kinases Involved in
`Tumor Progression and Angiogenesis. Additional characterization
`of BAY 43-9006 in biochemical assays demonstrated potent inhibi-
`tion of several RTKs, including human and murine VEGFR-2 (IC50,
`90 and 15 nmol/L, respectively), mVEGFR-3 (IC50, 20 nmol/L),
`mPDGFR-␤ (IC50, 57 nmol/L), Flt-3 (IC50, 58 nmol/L), c-KIT (IC50,
`68 nmol/L), and FGFR-1 (IC50, 580 nmol/L; Table 1). By contrast,
`EGFR, IGFR-1, c-met, and HER-2 RTKs were not inhibited by BAY
`43-9006 (IC50, ⬎10,000 nmol/L). Other kinases tested included PKB,
`PKA, cdk1/cyclinB, PKC␣, PKC␥, and pim-1, which were all insen-
`sitive to inhibition by BAY 43-9006 (Table 1).
`Inhibition of VEGFR-2 autophosphorylation by BAY 43-9006 was
`examined in two cell culture systems, HUVECs, and NIH 3

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket