throbber
[CANCER RESEARCH 57, 4838-4848. November 1. 1997]
`
`Induction of Apoptosis and Cell Cycle Arrest by CP-358,774, an Inhibitor of
`
`Epidermal Growth Factor Receptor Tyrosine Kinasc
`
`James D. Moyer,’ Elsa G. Barbacci, Kenneth K. lwata, Lee Arnold, Bruce Boman, Ann Cunningham,
`Catherine DiOrio, Jonathan Doty, Michael J. Morin, Mikel P. Moyer, Mark Neveu, Vincent A. Pollack,
`Leslie R. Pustilnik, Margaret M. Reynolds, Don Sloan, April Thelenran, and Penny Miller
`Pfizer Central Research, Grown. Connecticut 06340 [J. D. M.. E. G. B.. L A.. A. C.. C. D., J. D.. M. J. M.. M. P. M., M. N., V. A. P.. L R. P.. D. 5.. P. M.I; Oncogene Science
`Inc, Uniorxdale, New York I 1553 (K. K. 1.. M. M. R. A. T. 1; and Storz Cancer Institute, Omaha. Nebraska 68105 (B. 8.]
`
`ABSTRACT
`
`The epidermal growth factor receptor (EGFR) is overexpressed in a
`significant percentage of carcinomas and contributes to the malignant
`phenotype. Cl’-358,774 is it directly acting inhibitor of human EGFR
`tyrosine kinase with an IC,o of 2 nM and reduces EGFR autophosphoryb
`atlon in intact tumor cells with an [C5, of 20 nM. This inhibition is selective
`for EGFR tyrosine kinase relative to other tyrosine kinases we have
`examined, both in ways of isolated kinases and whole cells. At doses of
`100 mg/kg, CP~358.7’74 completely prevents EGF-induced autophosphr»
`rylation of EGFR in human HN5 tumors growing as xenografts in athyrnic
`mice and of the hepatic EGFR of the treated mice. Cl‘-358,774 inhibits the
`proliferation of DiFl human colon tumor cells at subrnicromoiar concen-
`trations iu cell culture and blocks cell cycle progression at the G, phase.
`This inhibitor produces a marked accumulation of retinoblastoma protein
`in its underphosphorylated form and accumulation of p27"“" in DiFi
`cells, which may contribute to the cell cycle block. Inhibition of the EGFR
`also triggers apoptosis in these cells as determined by formation of DNA
`fragments and other criteria. These results indicate that Cl‘-358,774 has
`potential for the treatment of tumors that are dependent on the EGFR
`pathway for proliferation or survival.
`
`INTRODUCTION
`
`EGFR2 is a transmembrane glycoprotein with an external domain
`that binds activating ligands, such as EGF and tumor growth factor oz,
`and an intracellular tyrosine kinase domain that, upon activation,
`phosphorylates both the receptor itself and a variety of “effector"
`proteins such as SHC and phospholipase C7 (1). Activation of this
`signaling cascade triggers DNA synthesis in cells that express the
`EGFR.
`
`Many human tumors, especially squamous carcinomas of the lung
`or head and neck, express high levels of EGFR or tumor growth factor
`or relative to the corresponding normal tissue (2—4). Although over-
`expression of EGFR is the most prevalent alteration of this pathway in
`human tumors, constitutively active mutant EGFR has also been
`reported in gliomas (5), breast tumors (6) and lung tumors (7). This
`suggests that activation of the BGFR may drive the proliferation of
`these tumors and that inhibitors of EGFR may be of use as antitumor
`agents (2, 8——l0).
`Blockade of the EGFR pathway by several methods inhibits the
`proliferation of a variety of tumor cell lines. For example, down-
`regulation of EGFR by antisense expression reduces the proliferation
`
`and invasive properties of a human colon tumor cell Line (ii) and
`blocks proliferation of human rhabdomyosarcoma cells (12). More~
`over, the transformation of cells by overexpression of EGFR in the
`presence of EGF is reversed by expression of a dominant negative
`mutant EGFR (13). lnhibltion of EGFR as an antitumor approach has
`been further substantiated by studies that show that antibodies that
`block EGF binding to the EGFR inhibit tumor cell proliferation in cell
`cultures and tumor xenografts in athymic mice (2, 8, l4~l6). Impor-
`tantly, anti~EGFR antibodies have been mently shown to produce
`complete regressions of established human tumor xenografts in athy-
`rnic mice (17, 18). Clinical trials of a humanized monoclonal antibody
`against the EGFR for the treatment of patients with tumors overex-
`pressing EGFR are in progress (19). Finally, selective low molecular
`weight inhibitors of the EGFR kinase have been shown to inhibit
`EGF-dependent cell proliferation (20-22) and exhibit antitumor ac-
`tivity in a human tumor xenograft model (23). Thus, a subset of
`tumors are dependent, at least in pan, on the EGFR for proliferation.
`Although the evidence above suggests that inhibition of EGFR may
`block proliferation of some tumor cells, EGFR inhibitors must also be
`well tolerated in patients to provide a useful therapeutic index. Al-
`though an inhibitor of EGFR would not be expected to be cytotoxic in
`the manner of the most current chemotherapy, pharmacological inhi~
`bitlon of the EGFR could interfere with the physiological functions of
`EGF and other EGFR ligands. However, targeted “knockout" of the
`mouse EGFR permitted embryonic development and birth of mice
`that survived as long as l8 days (24. 25). This indicates that EGFR is
`dispensable for the proliferation of all critical cell types and essential
`physiological functions. However, various defects such as thin epi-
`dermis, distorted colonic and hepatic epithelium. and low body weight
`were observed in these animals. Mice with the waved-2 phenotype
`express mutant EGFR with markedly impaired klnase activity in vivo
`(26) and, thus, may represent a model of partial inhibition of EGFR.
`These mice are viable and fertile but have hair, skin, and eye abnor-
`malities. Furthermore, early results from clinical trials with the anti-
`EGFR antibody have not revealed any toxicities that would prevent
`further development (19). These results suggest that a partial inhibi-
`tion of EGFR may be reasonably well tolerated in adults or present a
`much different pattern of toxicity than does standard chemotherapy.
`However, many normal cells express EGFR, for example, skin, liver,
`and gastrointestinal epithelium, and may be affected by such an
`inhibitor.
`
`Received 6:26/97; accepted 9/l9I97.
`The costs of publication of this article were defrayed in part by the payment of page
`charges. This article must therefore be hereby marked advent:-amen: in accordance with
`18 U.S.C. Section l734 solely to indicate this fact.
`‘To whom requests for reprints should be addressed. at Pfiur Central Research.
`Eastern Point Road. Groton. CT 06340. Email: james_d_moyer@groton.pfizcr.com.
`2 The abbreviations used are: EGFR, epidermal growth factor receptor, EGF, epider-
`mal growth factor; IGF-I. insulin-like growth factor I; IGP-IR. IGF-l receptor: FBS. fetal
`bovine serum: FRE cells, Contact-inhibited Fischer rat embryo cells; POT. poly(glutamic
`scidztyrosine) 4:1: HRP. horseradish peroxidase; BrdUrd. bromodeoxyuridiue; PDGF.
`platcleuderived growth factor; IJFGF. basic fibroblast growth factor; pRB, rerinoblastomn
`gene protein; PARP. poly(ADP-ribosc) polymerase; PI, propidium iodide: TUNEL. ter~
`minal deoxynuclootidyl uansferasc dU'I'i’ nick end labeling;
`lRS—l.
`insulin receptor
`substrate-1; PS. phosphaddylscrine.
`
`Although treatment with anti-EGFR antibodies has produced re-
`gressions of established human tumors in some xenograft tumor
`models (17, 18, 27). inhibition of growth factor pathways may only
`produce a cytostatlc effect if tumor cells survive in a Go state when the
`EGFR pathway is blocked. However. an important recent report by
`Wu el al. (28) showed that an anti-EGFR antibody could trigger
`apoptosis in a human colorectal cell line, DiFi, that overexpresses
`EGFR. Addition of IGF~I was able to protect the cells from the
`apoptotic effect of the antibody. These results, together with recent
`studies of inhibition of lGF—lR function (29. 30), indicate that, in some
`tumors, inhibition of growth factor pathways may kill tumor cells
`4838
`
`Downloaded from cancerresaacrjournals.org on October 19, 2015. © 1997 American Association for Cancer
`Research.
`
`APOTEX EX. 1016-001
`
`

`
`APOPTOSIS AND CELL CYCLE ARREST BY CP-358,774
`
`rather than only produce cell cycle arrest. Here. we extend these
`findings with a novel EGFR tyrosine kinase inhibitor and demonstrate
`that this inhibitor blocks DiFi cell proliferation by a combination of
`cell cycle arrest and induction of apoptosis.
`
`MATERIALS AND METHODS
`
`Cl’-358,774. [6.7-Bis(2-methoxy—ethoxy)-quinazolin-4-yl]~(3-ethynylphe~
`nyl)amine. molecular formula C22H23N3O4. was prepared as described (CH).
`The structure is shown in Fig. IA. The monohydrocliloride salt form of this
`compound was used in the studies reported here.
`Cell Lines and Culture Conditions. DiFi is a human colorectal carcinoma
`
`cell line derived from a familial adenomatous polyposis patient, as described
`previously, that expresses 5 X 10° EGFRs/cell (I6, 32). DiFi cells were
`maintained in l:1 DMEM:Ham‘s F-12 with 10% FBS. HN5 human head and
`
`neck tumor cells that express L4 X 107 EGFRS/cell (l5, 33) were obtained
`from Dr. M}. O'llare of Haddow Laboratories, The Institute of Cancer
`Research, Sutton (Surry, United Kingdom), and grown in DMEM with 10%
`FBS. MDA—MB~468 human breast cancer cells that express l.5 X 10*’ EGFRs/
`cell (15) were obtained from the American Type Culture Collection (Bethesda,
`MD) and grown in DMEM with 5% FBS. RB cells (34) were maintained in
`DMEM with l0% FBS. All cells were cultured at 37°C in 5% carbon dioxide!
`
`95% air in the presence of 100 units/ml penicillin. l00 pg/ml streptomycin.
`and 2 mM glutamine. DiFi, MDA-MB-468. and HNS cells were tested and
`found to be mycoplasma free using the Gen—Probe kit (Fisher Scientific.
`Pittsburgh. PA).
`Kinase Assays. The EGFR kinase assay is similar to one described previ-
`ously (35). Nunc MaxiSorp 96-well plates were coated by incubation overnight
`at 37°C with l00 p.l per well of 0.25 mg/ml PGT (Sigma Chemical Co.. St.
`Louis, MO) in PBS. Excess PGT was removed by aspiration. and the plate was
`washed 3 times with wash buffer (0. l % Tween 20in PBS). The kinase reaction
`was performed in 50 pit of 50 mM HEPES (pll 7.3). containing l25 mM sodium
`chloride, 24 mM magnesium chloride. O.l mM sodium orthovanadate, 20 pm
`ATP. 1.6 pig/ml EGF. and i5 ng of EGFR. affinity purified from A43l cell
`membranes as described (36). The compound in DMSO was added to give a
`final DMSO concentration of 2.5%. Phosphorylation was initiated by addition
`of ATP and proceeded for 8 min at room temperature. with constant shaking.
`The lcinase reaction was tenninated by aspiration of the reaction mixture and
`was washed 4 times with wash buffer. Phosphorylated PGT was measured by
`25 min of incubation with 50 ptl per well HRP-conjugated PY54 (Oncogene
`Science Inc. Uniondale, NY) antiphosphotyrosine antibody, diluted to 0.2
`trg/ml in blocking buffer (3% BSA and 0.05% Tween 20 in PBS). Antibody
`was removed by aspiration. and the plate was washed 4 times with wash buffer.
`The colorimetric signal was developed by addition of TMB Microwell Perox-
`idase Substrate (Kirkegaard and Perry, Gaithersburg. MD). 50 pl per well. and
`stopped by the addition of 0.09 M sulfuric acid. 50 p.l per well. Phosphoty-
`rosine is estimated by measurement of absorbance at 450 nm. The signal for
`controls was typically 0.64.2 absorbance units. with essentially no back-
`ground in wells without ATP, EGFR, or PGT and was proportional to the time
`of incubation for l0 min.
`
`Conditions for selectivity assays were the same as those for the EGFR
`ltinase assay, except for the addition of l nuvt manganese chloride to the assay
`buffer and a final ATP concentration of l00 ;LM. The reaction was terminated
`by the addition of 50 id of 250 mM EDTA prior to aspiration. For experiments
`comparing inhibition of EGFR to v-abl or c—src kinase. recombinant bacterially
`expressed V-abl (3.4 ng/well) or purified human platelet c—src (l.2 units/well,
`Oncogene Science lnc.) was substituted for the EGFR. For experiments com-
`paring inhibition of EGFR to insulin receptor or lGF~lR. purified recombinant
`proteins were substituted for native EGFR. Baculovirus~expressed cytoplasmic
`domain of the insulin receptor B subunit (l0 units/well) was from Stratagene
`(La Jolla. CA). Recombinant EGFR kinase domain (2 ng).
`lGF~lR ltinase
`domain (3 ng), and web! ltinase were prepared as described below.
`Preparation of Recombinant Kinases. The complete intracellular domain
`of human EGFR (amino acids 6444186) was PCR amplified and subcloned
`into pAcG2T to generate a glutathione S-transferase fusion protein with a
`thrombin cleavage site. Plaque—purified recombinant baculovirus was used to
`infect Sf‘) insect cells for 60 h. The complete intracellular domain of human
`lGF—lR 8 subunit (amino acids 7l l~l377) was PCR amplified and subcloned
`into pAcG2’l“ to generate a glutathione S-transferase fusion protein with a
`thrombin cleavage site. Plaque-purified recombinant baculovirus was used to
`infect High V insect cells for 36 h. For both preparations. active kinase was
`purified using glutathione Sepharose followed by elution with free glutathione.
`The v—ahl was expressed as a Hiso-tagged protein and purified by affinity
`chromatography with the QlAexpness system (Qiagen).
`l.S X l0‘
`FRE Mltogenesis. FRE cells were plated in 96-well plates at
`cells/well in 100 ptl of DMEM with l0% F88. The next day. the medium was
`replaced with l00 (Ll of serum-free medium (RPMI 1640). After l2—20 h.
`growth factors and Brdljrd were added in the presence or absence of CP-
`358.774. The growth factors and their concentrations were as follows: murine
`EGF (Collaborative Biomedical Products). 0.5 ng/ml; PDGF (Genzyme,
`Cambridge, MA).
`l5 ng/ml;
`lGF—l
`(Genzyme). 50 ng/ml; and bFGF
`(Genzyme). 50 ng/ml. After overnight incubation, BrdUrd incorporation was
`measured using the Cell Proliferation Assay Kit (Amersham, Arlington
`Heights. IL) according to the supplier's instructions, except for substitution of
`mphenylenediamine (Pierce Chemical Co., Rockford, ll.) as the peroxidase
`substrate. The color reaction was stopped with 2 N sulfuric acid. and absorb-
`ance was read at 490 nm.
`For calculation of inhibition. all wells were corrected for the background
`signal obtained from cells incubated without Brdllrd. Percent inhibition was
`calculated as follows: I00 - l00l(treated - basal)l(control ~ basalll. where
`basal is signal from cells not stimulated with growth factor. control is signal
`4839
`
`HN
`
`A
`
`1 00
`90
`80
`70
`60
`50
`40
`30
`20
`‘l0
`
`%ofControl(tS.D.)
`
`0.01
`
`
`1 000
`0.1
`1
`10
`100
`[CP-358.774} (MA)
`
`Increasing
`[CP-358,774}
`
`4
`
`PhosphorglationI/V
`
`0
`
`0.0
`
`0.1
`
`0.2
`
`0.3
`
`1l{ATP]
`
`(nun)
`
`l. CP—358.774 inhibits purified EGFR kiuase and is competitive with ATP.
`Fig.
`Phosphorylation of PGT by purified EGFR was musured by immunoassay with antiphos—
`photyrosine antibodies as described in “Materials and Methods.“ A, data points. means of
`six determinations: bars. SD. This result is representative of four independent experi—
`ments. B, ATP competition experiments. CP-358,774 concentrations were: it) nm (0): 3
`nm (A); 1 nm (0); 0.3 nM (A); or 0 nM (Cl). The ATP concentrations tested were 5 mi.
`20 int. 80 p.M. 320 pm. and 3 mM. The K,,, for ATP under these conditions was 7 p.M.
`l‘-‘hosphorylation is measured in arbitrary units, and the data were fit by a linear least
`squares method. This experiment was repeated with identical results.
`
`Downloaded from cancerresaacrjournals.org on October 19, 2015. © 1997 American Association for Cancer
`Research.
`
`APOTEX EX. 1016-002
`
`

`
`APOPTOSIS AND CELL CYCLE ARREST BY CP~358,774
`
`Systems, Minneapolis. MN) and the supplier‘s protocol. Annexin V~FI'I‘C and
`Pl binding were analyzed by flow cytometry using a FACSort. Data were
`collected using logarithmic amplification of both the FL! (FITC) and FL2 (PI)
`channels. Quadrant analysis of coordinated dot plots was done using CellQuest
`software. Unstained cells were used to control for autofluorescence. Singly
`stained cells were used to adjust the photomultiplier voltages and compensa-
`tion settings to eliminate spectral overlap between the FLl and FL2 signals.
`Preparation of Tissue Extracts for Western Blot Analysis. Frozen tumor
`or liver samples were pulverized. extracted with boiling 2>< Laemmli sample
`buffer (37) with 2 mM sodium orthovanadate, placed in a boiling water bath for
`I0 min. and stored at ~80"C until analysis. After the tissue extracts of
`insoluble material were cleared by centrifugation. the protein content of the
`extracts was determined, and the samples were analyzed by Western blotting
`as described above.
`
`RESULTS
`
`from growth factor-stimulated cells. and treated is signal from Cl’~358,774-
`treated, growth factor-stimulated cells.
`Measurement of Phosphotyroslne, pRB Phosphorylation, p27'“"’
`Expression, and PARP Cleavage by Western Blotting. Cells (DiFi or HN5)
`were incubated in the presence or absence of CP~358,774 or cisplatin. After
`24 h. cells were washed with 50 mM Tris-l~lCl. 140 mM sodium chloride, 3.3
`mM potassium chloride. and 500 ,u.M sodium orthovanadate (pH 7.4) and lysed
`by boiling in 2X Laemmli sample buffer (37) with 2 mM sodium orthovanadate
`for 10 min. Cellular protein was determined using the BCA protein assay
`(Pierce Chemicals. Rockford. IL). Equal amounts of total protein (10-20 ug)
`were loaded onto 4—20% Tris-glycine minigels (Integrated Separated Systems,
`Natick. MA) for phosphotyrosine determinations. 7.5% Daiichi Tris-glycine
`minigels for pRB, 12.5% Daiichi Tris~glycine minigels for p27'“"‘. or 4~l2%
`Bis-Tris NuPage minigcls (Novex, San Diego. CA) for PARP. After electro
`phoresis. proteins were transferred to an lrnmobilon-P membrane (Miilipore,
`Bedford, MA) for 2 h at 250 mA. After transfer, membranes were blocked for
`l h or overnight with 5% BSA in TBST [50 mM Tris-I-lCl (pH 7.4). 150 mM
`NaCl. and 0.1% Tween 20} for antiphosphotyrosine blotting or with 4~5%
`nonfat dry milk in TBST. For phosphotyrosine determinations. blots were
`probed with HRP-conjugated antiphosphotyrosine PY20 (ICN, Costa Mesa.
`CA) or liRl’~l’Y54 (Oncogene Science). For pRB detenninations, blots were
`probed with l pig/ml monoclonal antibody G3-245 (Pharlvlingen, San Diego.
`CA). followed by HRP-conjugated goat antimouse (Pharhiingen; H000). The
`identification of the lower band as underphosphorylated pRB was confirmed
`by use of an antibody specific for this form (PharMingen). For p27‘“”’, blots
`were probed with 0.! pg/ml arrti—p27'“”’ Clone 57 (Transduction Labs. Lex-
`ington. KY) followed by HRP—goat antimouse. For PARP, blots were probed
`with anti-PARP. Clone C-2-l0 (Biornol, Plymouth Meeting. PA), followed by
`HR?-conjugated rabbit antimouse lgG (Pierce Chemicals). The M, ll6,000
`PAR? and its M, 85,000 proteolytic fragment were identified by comparison
`with lysates of untreated HL-60 cells or HL-60 cells induced to undergo
`apoptosis with etoposide (Biomol). All HRP-labeled antibodies were detected
`using enhanced chemiluminescence (Amersham) according to the supplier’s
`directions and quantitated by densitometry.
`Cell Cycle Analysis of DiFi Cells. Scmiconflucnt DiFi cells growing in
`six—well plates were incubated for 24 h in DMEM:l-larn's F12 and 0.5% FBS
`containing diluent (0.l25% DMSO). cisplatin. or CP—358.774 in the absence or
`presence of IGF-l. Cells were harvested and incubated in medium containing
`0.2% Triton X-100 and 50 ug/ml Pl for 30 min. according to published
`methods (38). Pl uptake was analyzed by flow cytometry using a FACSort
`(Becton Dickinson, San Jose. CA). Data were acquired using linear arnplili~
`cation of FL2 and analyzed using CellQuest softwa.re (Becton Dickinson). For
`Inhibition of Tyrosine Phosphorylation in Intact Cells. The
`measurement of BrdUrd incorporation, DiFi cells were treated for 24 h with
`addition of EGF to cells that express EGFR leads to a rapid autophos-
`diluent or 1 us: CP-358.774, pulse labeled with I0 ,u.M 8rdUrd, incubated with
`phorylation of the EGFR on tyrosine residues in the COOH terminus,
`FITC-conjugated anti-BrdUrd monoclonal antibody (Becton Dickinson). and
`counterstained with Pl.
`thus providing a facile assay for inhibition of the EGFR tyrosine
`Agarose Gel Analysis of DNA Fragmentation. DiFi cells were treated to
`kinase activity in intact cells. CP-358.774 potently inhibits EGFR
`induce apoptosis as described for cell cycle analysis. After trypsinization of the
`autophosphorylation in I-INS human head and neck tumor cells (Fig.
`cells, DNA was extracted using a TACS Apoptotic DNA Laddering Kit
`3). a cell line that expresses high levels of EGFR (15). Evaluation of
`according to the supplier‘s instructions (Trevigen. Gaithersburg, MD). Isolated
`these Westem blots by densitometry indicates an IC50 for inhibition of
`DNA was quantitated by absorbance at 260 nm. Samples (6 peg/lane) were
`EGFR phosphorylation of 20 nM. At the higher concentrations of the
`electrophorescd on a 1.5% agarose gel containing 0.5 pg/ml P! for 2 h at Ill)
`compound, the extent of autophosphorylation after EGF stimulation is
`V in TAB buffer (40 mM Tris~acetate. l0 mM EDTA, and 20 mM glacial acetic
`lower than that in the controls without EGF, indicating reduction of
`acid. pH 8.4). Gels were visualized on 8 UV transilluminator.
`basal EGFR activity, which may arise from autocrine stimulation.
`TUNEL Analysis. DiFi cells were treated to induce apoptosis as described
`for cell cycle analysis. Cells were harvested. and aliquots of 2 X I0” cells were
`Additional studies indicated that the inhibition of kinase is rapidly
`obtained (<10 min) on addition of CP-358,774 to cell medium and is
`fixed in l% paraformaldehyde. permeabilized in 70% ethanol. and stored at
`-20"C. 3'~OH DNA strand breaks were detected by the TUNEL technique
`rapidly reversed after the inhibitor is washed out and the cells
`using the ApopTag Plus Fluorcscein (FlTC) kit (Oncor, Gaithersburg. MD)
`are incubated in inhibitor-free medium (data not shown). Similarly
`according to the supplier's instructions. FITC-labeled cells were counter-
`potent
`inhibition of EGF-induced EGFR autophosphorylatiorr by
`stained with P1 and analyzed by flow cytometry using a FACSort. Excitation
`Cl’-358,774 was seen with DiFi human colon cancer cells and MDA-
`was at 488 nm with emission read in the FLI (BP 530/30) and FL2 (BP
`MB~468 human breast cancer cells (data not shown).
`SS5/42) channels. Data were acquired using logarithmic amplification of both
`The selectivity of CP-358,774 as an inhibitor of the phosphoryla—
`FL] and FL2 (the FITC and PI signals. respectively). Data were analyzed using
`tion of endogenous intracellular substrates by EGFR tyrosine ltinase
`CellQuest software. Regions of ill versus FL2 dot plots were set using
`can also be demonstrated in intact cells. The phosphorylation of the
`control (cells treated with diluent alone) samples (39).
`adaptor protein SHC (40, 41) upon addition of EGF serves as a
`Annexln V Binding. DiFi cells were treated to induce apoptosis as de-
`scribed for cell cycle. Cells were harvested, and aliquots of 2 X l05 cells were
`convenient physiological marker for EGFR activity. The EGF-
`treated with anrrexin V-FITC and PI using the Apoptosis Detection Kit (R&D
`induced tyrosine phosphorylation of SHC proteins is completely
`4840
`
`Inhibition of EGFR Tyrosine Kinasc by CP-358,774.
`CP-358,774 inhibits purified EGFR ltinase with an ICSO of 2 nM (Fig.
`IA). Kinetic analysis indicates that the inhibition is competitive with
`ATP. A Lineweaver—Burk plot of phosphorylation in the presence of
`varied ATP and inhibitor indicates that the inhibition is reduced at
`
`higher concentrations of ATP and can be restored to the uninhibited
`rate at high ATP concentrations (Fig. 18). The extent of inhibition
`may, therefore. be influenced by the intracellular ATP concentration.
`A replot of the Km“, versus concentration of CF‘-358,774 from the
`data of Fig. 28 indicated a K, for CP~358,774 of 2.7 nrvr. EGFR is more
`sensitive to inhibition by CP-358,774 than are the other tyrosine
`kinases we have examined, and it is >l000—fold more sensitive than
`human c—src or v-abl when compared under identical conditions (Fig.
`2A). Cl’-358.774 is also a potent inhibitor of the recombinant intra—
`cellular (kinase) domain of the EGFR, with an IC50 of I nm (Fig 2B),
`essentially identical to that observed with full-size EGFR (Fig. IA),
`which indicates that the inhibitor binding site is in the kinase domain.
`The kinase domains of the human insulin receptor and IGF—lR are
`much less sensitive to this inhibitor and are essentially unaffected at
`concentrations as high as 10 ptM (Fig. 28). Thus, Cl’-358,774 is a
`potent. selective, and directly acting inhibitor of the EGFR tyrosine
`kinase.
`
`Downloaded from cancerresaacrjournals.org on October 19, 2015. © 1997 American Association for Cancer
`Research.
`
`APOTEX EX. 1016-003
`
`

`
`APOPTOSIS AND CELL CYCLE ARREST BY Cl’-353,774
`
`s
`0'
`
`"
`*
`5
`s
`0

`{op-353,774}, not
`
`9
`
`100
`
`so
`
`E’ so
`
`§3
`
`2 40
`
`20
`
`o
`
`140
`
`120
`
`__1oo
`§ so
`3 so
`40
`
`20
`
`o
`
`A
`
`B
`
`a
`
`'*
`
`"
`
`‘-’
`

`
`C [
`
`s
`0-
`
`CP—358.774], nu
`
`Hg. 2. Selectivity of CP-358.774 for EGFR kinase. Inhibition of purified kinases was
`measured as described in “Materials and Methods." Data points. means of triplicate
`determinations; bars. SE. Similar results were observed in two experiments. A. selectivity
`for native human EGFR versus c-src and recombinant v-abl. I. El-‘GR: O. c-src: A, v—abl.
`B. selectivity for recombinant EGFR ltinase domain (I) versus insulin receptor kinase
`domain (A) and IGF-IR kinase domain (I). As a positive control.
`tyrphostin A25
`(Calbiochem. San Diego. CA) produced 50% inhibition of IGF-IR kinase in this assay at
`20 p.M.
`
`blocked by CP-358,774 (Fig. 4, Lane 9), whereas the insulin-induced
`phosphorylation of IRS-l, a prominent insulin receptor tyrosine ki-
`nase substrate, is unaffected (Fig. 4., Lane 4). Staurosporine, a rela-
`tively nonselective kinase inhibitor (42), completely blocked IRS-l
`phosphorylation in response to insulin (Fig. 4. Lane 5) and therefore
`served as a positive control. This experiment demonstrates that CP-
`358,774 markedly inhibits in situ phosphorylation of an endogenous
`substrate of activated EGFR at concentrations that have no effect on
`
`completely inhibited by pretreatment with 100 mg/kg CP~358,774 and
`is reduced by 89 i ll% (mean 1 SE; n = 4) and 61 1 7% at doses
`of 25 and l0 mg/kg, respectively, at l h after an i.p. treatment. The
`doses of CP-358.774 used here (10, 25, and 100 mg/kg) can be
`administered daily for at least 5 consecutive days without lethality in
`mice. These results indicate that CP-358,774 treatment of mice effec-
`tively inhibits both murine EGFR kinase in liver and human EGFR in
`transplanted tumors.
`Inhibition of Tumor Cell Proliferation. The DiFi human colon
`
`tumor cell line expresses high levels of EGFR and is inhibited by the
`anti~EGFR antibody (16, 28, 44). The proliferation of DiFi cells is
`strongly inhibited by CP-358.774 with an ICSO of I00 nM for an 8-day
`proliferation assay (Fig. 6). Cl’-358,774 did not produce a rapid loss
`of viability:
`the percentage of viable cells was 98 t 0.8%
`(mean 1: SD) in untreated cell cultures and 97 : l.7% in cultures
`exposed to l ;.tM CP-358,774 for 24 h. The I-{N5 human head and neck
`tumor cell line (15, 28. 33), which also expresses a very high level of
`the EGFR. is markedly inhibited by Cl’-358.774 at concentrations as
`low as 50 nM and is completely blocked at 250 nM (data not shown).
`In contrast,
`the proliferation of raf-transformed NIH-3T3 cells or
`ras-transformed FRE cells is much less sensitive, with IC,0s for
`proliferation of 7 and 3 p.M, respectively (data not shown).
`CP-358,774 is selective for the EGFR kinase pathway, as evaluated
`in cellular proliferation assays. Unlike tumor cell lines, FRE fibro-
`blasts can be rendered quiescent by incubation in scrum~free medium
`and then triggered to proliferate by addition of defined growth factors
`such as EGF, PDGF. IGF-I, or bFGF. Each of these growth factors
`acts through a cognate transmembrane receptor with tyrosine kinase
`activity (I); thus, FRE cells are a well-defined model, unambiguously
`dependent on the added factors. that can be used to evaluate selectivity
`of tyrosine kinase irmibitors. Cl’-358,774 inhibits EGF-stimulated
`mitogenesis with an IC,0 of 70 nM but only inhibits mitogenesis
`stimulated by the other factors at concentrations of >l p.M (Fig. 7).
`Thus, CP-358,774 is not simply indiscriminately cytotoxic because
`the FRE cells continue to undertake DNA synthesis when stimulated
`with mitogens other than EGF. This indicates that Cl’-358,774 is
`selective for EGFR ltinase relative to other tyrosine kinase-linked
`receptors and that, at concentrations that inhibit EGF-induced prolif-
`eration, it does not effectively inhibit any of the many ltinases or other
`enzymes that are necessary for mitogenesis in response to other
`rnitogens.
`Additional studies were done to further characterize the effects of
`
`Cl’-358.774 on DiFi cell proliferation. Analysis of the cell cycle
`distribution of these cells indicated that
`the cells were partially
`blocked in the G, phase of the cell cycle by this EGFR inhibitor (Fig.
`8 and Table 1). Although the changes were somewhat masked by the
`appearance of an apoptotic cell population discussed below, marked
`
`CP—358,774 [nM]
`EGF
`
`50
`
`phosphorylation of the major physiological substrate of another trans-
`membrane tyrosine kinase, the insulin receptor.
`Inhibition of Murine and Human EGFR in Vivo by CP-358,774.
`Intravenous administration of EGF to mice produces a marked auto-
`phosphorylation of EGFR in liver and other tissues (43), thus provid-
`ing a dynamic assay for EGFR inhibition in vivo. We examined the
`ability of CP-358,774 to block EGFR autophosphorylation in liver and
`HN5 tumors in athyrnic mice, as shown in Fig. 5. As reported, EGFR
`in mouse liver is hypophosphorylated but is rapidly tyrosine phos-
`Fig. 3. Effect of CP—358.774 on tyrosine phosphorylation in HN5 cells. as evaluated
`phorylated when mice receive EGF (Fig. 5B). This phosphorylation is
`with Western blots. HN5 cells U0’ cells/well in 24-well cell culture plates) were exposed
`to the indicated concentrations of CP-358.774-Ol for l h and then stimulated with 50
`inhibited 54% by pretreatment with 10 rnglltg CP-358,774 and is
`ng/ml EGF (Latter 2, 4, 6. 8, and I0) or not (tuner I, 3, 5. 7, and 9). Cell lysates were
`nearly completely inhibited (93%) at 100 mg/kg. Similarly, EGFR in
`prepared alter a 5~min exposure to EGF. and BGFR-associated phosphotyrosine was
`human HN5 tumors is rapidly autophosphorylated in response to EGF
`measured by Western blotting as described in “Materials and Methods." The Cl’-358.774
`(Fig. 5A). Analysis of a larger set of samples prepared as in Fig. 5
`was present at 0 nM (Lanes 1 and 2); 20 nM (Lanes 3 and 4); 50 nM (Lanes 5 and 6); 250
`nM (Lanes 7 and 8); or 1000 rm (Lanes 9 and I0).
`indicates that EGF~induced autophosphorylation of tumor EGFR is
`4841
`
`0
`
`20
`
`12345678910
`
`Downloaded from cancerresaacrjournals.org on October 19, 2015. © 1997 American Association for Cancer
`Research.
`
`APOTEX EX. 1016-004
`
`

`
`APOPTOSIS AND CELL CYCLE ARREST BY CP~358.774
`
`Fig. 4. CP-358,774 sclectivcly inhibits EGFR signaling in HN5
`cells. Following serum starvation for 2 it. HN5 cells were incubated
`for an additional hour in the presence of 012.5% DMSO (Lanes I~3
`and 6-8). l tm CP-358.774—0l (Lanes 4 and 9). or l0 p.M 513030-
`sporine (Lanes 5 and I0). Cells were then stimulated for 5 min with
`l00 nM insulin (Lanes 2-5) or 100 ng/ml EGF (Lanes 7-10) and
`lysed in 1% Triton X-l00 buffer. Ixmes l~5. lysates were subjected
`to immunoprecipitation with antibody to IRS- l. Lanes 6-10, lysates
`were subjected to immunoprecipitation with antibody to SHC. Im-
`munoprecipitates were resolved by SDS~PAGE. transferred to Im-
`rnobilon. and imrnunohlottcd with antiphosphotymsine antibodies as
`described in “Materials and Methods." Lanes 2 and 3. duplicates of
`the insulimtreated controls (no inhibitor); Lanes 7 and 8. duplicates
`of the EGF~treated controls (no inhibitor). Other conditions are
`single lanes.
`
`1234 5678910
`
`IRS-l ---> C O .
`
`EGFR -‘u- .
`
`}SHC
`
`decreases in the percentage of cycling cells in S phase and G2~M and
`an increase in the percentage of cells in G, were observed, indicating
`a G, block. Similar experiments were performed with the HN5 cells:
`48 h of exposure to 1
`;.LM CP-358,774 reduced the S-phase cells from
`55% of the total to 24% and increased the percentage of cells in G,
`from 24 to 56% (data not shown). Thus, for both DiFi and HN5 cells,
`
`inhibition of the EGFR ltinase by CP—358,774 leads to a partial G,
`arrest.
`
`The reduction in S~phase cells by Cl’-358.774 was confirmed
`by flow cytometric measurements of total DNA and incorporated
`BrdUrd, a more specific method for identifying cells in S—phase. The
`percentage of S-phase DiFi cells measured by this procedure de-
`
`A.
`CP358T74
`
`HN5 Tumor
`
`mglkg
`EGF
`
`O
`-
`
`0
`+
`
`1
`+
`
`10
`+
`
`100
`+
`
`EGFFl----
`
`B.
`
`Liver
`
`Fig. 5. CP~3S8.774 pretreatment inhibits EGF-
`i

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