throbber
[CANCER RESEARCH 61, 5106 –5115, July 1, 2001]
`
`Pharmacological Inhibitors of the Mitogen-activated Protein Kinase (MAPK)
`Kinase/MAPK Cascade Interact Synergistically with UCN-01 to Induce
`Mitochondrial Dysfunction and Apoptosis in Human Leukemia Cells1
`Yun Dai, Chunrong Yu, Victor Singh, Lin Tang, Zhiliang Wang, Robert McInistry, Paul Dent, and Steven Grant2
`Division of Hematology/Oncology [Y. D., C. Y., V. S., Z. W., S. G.], Departments of Pharmacology [S. G.], Biochemistry [S. G.], Microbiology [L. T., S. G.], and Radiation
`Oncology [R. M., P. D.], Medical College of Virginia, Richmond, Virginia 23298
`
`ABSTRACT
`
`Interactions between the checkpoint abrogator UCN-01 and several
`pharmacological
`inhibitors of
`the mitogen-activated protein kinase
`(MAPK) kinase (MEK)/MAPK pathway have been examined in a variety
`of human leukemia cell lines. Exposure of U937 monocytic leukemia cells
`to a marginally toxic concentration of UCN-01 (e.g., 150 nM) for 18 h
`resulted in phosphorylation/activation of p42/44 MAPK. Coadministra-
`tion of the MEK inhibitor PD184352 (10 mM) blocked UCN-01-induced
`MAPK activation and was accompanied by marked mitochondrial dam-
`age (e.g., cytochrome c release and loss of DCm), caspase activation, DNA
`fragmentation, and apoptosis. Similar interactions were noted in the case
`of other MEK inhibitors (e.g., PD98059; U0126) as well as in multiple
`other leukemia cell types (e.g., HL-60, Jurkat, CCRF-CEM, and Raji).
`Coadministration of PD184352 and UCN-01 resulted in reduced binding
`of the cdc25C phosphatase to 14-3-3 proteins, enhanced dephosphoryl-
`ation/activation of p34cdc2, and diminished phosphorylation of cyclic
`AMP-responsive element binding protein. The ability of UCN-01, when
`combined with PD184352, to antagonize cdc25C/14-3-3 protein binding,
`promote dephosphorylation of p34cdc2, and potentiate apoptosis was mim-
`icked by the ataxia telangectasia mutation inhibitor caffeine. In contrast,
`cotreatment of cells with UCN-01 and PD184352 did not substantially
`increase c-Jun-NH2-terminal kinase activation nor did it alter expression
`of Bcl-2, Bcl-xL, Bax, or X-inhibitor of apoptosis. However, coexposure of
`U937 cells to UCN-01 and PD184352 induced a marked increase in p38
`MAPK activation. Moreover, SB203580, which inhibits multiple kinases
`including p38 MAPK, partially antagonized cell death. Lastly, although
`UCN-01 6 PD184352 did not induce p21CIP1, stable expression of a
`p21CIP1 antisense construct significantly increased susceptibility to this
`drug combination. Together, these findings indicate that exposure of
`leukemic cells to UCN-01 leads to activation of the MAPK cascade and
`that interruption of this process by MEK inhibition triggers perturbations
`in several signaling and cell cycle regulatory pathways that culminate in
`mitochondrial injury, caspase activation, and apoptosis. They also raise
`the possibility that disrupting multiple signaling pathways, e.g., by com-
`bining UCN-01 with MEK inhibitors, may represent a novel antileukemic
`strategy.
`
`INTRODUCTION
`
`UCN-01 (7-hydroxystaurosporine) is a derivative of staurosporine
`that was originally developed as an inhibitor of PKC3 (1). However,
`UCN-01 has since been shown to inhibit other kinases, including
`
`Chk1, which is responsible for phosphorylation, binding to 14-3-3
`proteins, and subsequent degradation of the cdc25c phosphatase (2).
`Degradation of cdc25c results in phosphorylation and inactivation of
`CDKs such as CDK1 (p34cdc2), which are critically involved in cell
`cycle arrest after DNA damage and other insults (3). In this way,
`UCN-01 acts as a checkpoint abrogator, an action that may account
`for its ability to enhance the lethal actions of various cytotoxic agents,
`including cisplatin (4), mitomycin C (5), camptothecin (6), fludara-
`bine (7), gemcitabine (8), and 1-b-D-arabinofuranosylcytosine (9, 10),
`among others. When administered alone, UCN-01 induces arrest in
`G2M or G0G1, depending upon cell type, or, alternatively, the p53 or
`pRb status of the cell (11, 12). UCN-01 is also a potent inducer of
`apoptosis, particularly in hematopoietic cells, a phenomenon that
`appears to be more closely related to dephosphorylation of CDKs than
`to inhibition of PKC (13).
`Phase I and pharmacokinetic studies of UCN-01 have been initiated
`in humans and have shown that this compound exhibits a very long
`plasma half-life, presumably a consequence of extensive binding to a
`1
`acidic glycoprotein (14). Nevertheless, free plasma levels of UCN-01
`capable of inhibiting Chk 1 and abrogating checkpoint control events
`appear to be achievable (15, 16). In a preliminary study (16), combi-
`nation of UCN-01 with established cytotoxic agents was associated
`with evidence of clinical activity in a patient with advanced non-
`Hodgkin’s lymphoma, raising the possibility that UCN-01 may en-
`hance the in vivo activity of conventional chemotherapeutic drugs.
`Despite the intense interest in UCN-01 as an antineoplastic agent,
`the mechanism(s) by which it induces cell death remain(s) incom-
`pletely understood. Recently, considerable attention has focused on
`the role of signal transduction pathways in the regulation of cell
`survival, particularly those related to three parallel MAPK modules.
`Of these, the SAPK/JNK and p38 kinase are primarily induced by
`environmental insults (e.g., DNA damage or osmotic stress) and are
`generally associated with pro-apoptotic actions (17, 18). In contrast,
`p42/44 MAPKs (ERKs) are induced by mitogenic or differentiation-
`related stimuli and are most frequently (although not invariably)
`associated with pro-survival activity (19, 20). In fact, there is evidence
`that the relative outputs of the JNK and p42/44 MAPK cascades
`determine whether a cell lives or dies in response to a noxious
`stimulus (e.g., growth factor deprivation; Ref. 21). p42/44 MAPK lies
`downstream of a signaling pathway consisting of PKC, Raf-1, and
`MEK1 (22). Investigation of the functional role of p42/44 MAPK in
`cell death decisions, as well as other biological processes, has been
`greatly facilitated by the development of pharmacological MEK in-
`hibitors, including PD98059 (23), U0126 (24), and SL327 (25). Re-
`cently, Seybolt-Leopold et al. (26) described a novel MEK inhibitor,
`PD184352, which is able to block MAPK activation and to inhibit the
`in vivo growth of colon tumor cells in mice. Aside from their intrinsic
`antitumor activity, MEK inhibitors may also have a role as potentia-
`tors of established chemotherapeutic drug action (27).
`The relationship between UCN-01 actions and activity of the
`
`Received 1/10/01; accepted 4/20/01.
`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.
`1 Supported by Awards CA 63753, CA 77141, and DK 52825 from the NIH, and by
`Awards 6630-01 from the Leukemia and Lymphoma Society of America and BC980148
`from the Department of Defense.
`2 To whom requests for reprints should be addressed, at Division of Hematology/
`Oncology, Medical College of Virginia, MCV Station Box 230, Richmond, VA 23298.
`Phone: (804) 828-5211; Fax: (804) 828-8079; E-mail: stgrant@hsc.vcu.edu.
`3 The abbreviations used are: PKC, protein kinase C; CDK, cyclin-dependent
`kinase; MAPK, mitogen-activated protein kinase; MEK, MAPK kinase; SAPK, stress-
`activated protein kinase; JNK, c-Jun NH2-terminal kinase; ERK, extracellular regu-
`lated kinase; TUNEL,
`terminal deoxynucleotidyl
`transferase-mediated nick end
`merase; RIPA, radioimmunoprecipitation assay; CHX, cycloheximide; GFX, bisindo-
`labeling; DiOC6, 3,3-dihexyloxacarbocynine; BrdUrd, bromodeoxyuridine; CREB,
`lylmaleimide; PMA, phorbol 12-myristate 13-acetate.
`cyclic AMP-responsive element binding protein; PARP, poly(ADP-ribose) poly-
`5106
`
`
`
`Downloaded from on April 18, 2016. © 2001 American Association for Cancercancerres.aacrjournals.org
`
`
`Research.
`
`Page 1 of 11
`
`Fluidigm
`Exhibit 1008
`
`

`

`INDUCTION OF APOPTOSIS BY UCN-01 AND MEK INHIBITORS
`
`MATERIALS AND METHODS
`
`MAPK pathway is poorly understood. Given the fact that UCN-01 can
`function as a PKC inhibitor (1) and that it has been shown to mimic
`some of the actions of the PKC down-regulator bryostatin 1 as well as
`the kinase inhibitor staurosporine (25), the possibility that UCN-01
`might block the downstream PKC targets MEK1/2 and MAPK ap-
`peared plausible. To address this issue, we have examined the apo-
`ptotic actions of UCN-01 in relation to its effects on the MEK/MAPK
`cascade. Contrary to expectations, exposure of multiple myeloid and
`lymphoid cell
`lines to submicromolar concentrations of UCN-01
`potentiated, rather than reduced, MAPK phosphorylation/activation.
`Moreover, interference with this process by several pharmacological
`MEK inhibitors, including PD98059, U0126, and PD184352, resulted
`in a highly synergistic enhancement of mitochondrial damage, caspase
`activation, and apoptosis in these cells. Together,
`these findings
`suggest that exposure of human leukemia cells to UCN-01 elicits a
`cytoprotective MAPK response and raise the possibility that combin-
`ing this agent with pharmacological MEK inhibitors may effectively
`lower the apoptotic threshold.
`
`tained and fixed with 4% formaldehyde. The slides were treated with acetic
`acid/ethanol (1:2), stained with terminal transferase reaction mixture contain-
`ing 1 3 terminal transferase reaction buffer, 0.25 units/ml terminal transferase,
`2.5 mM CoCl2, and 2 pmol fluorescein-12-dUTP (Boehringer Mannheim,
`Indianapolis, IN), and visualized using fluorescence microscopy.
`Analysis of Mitochondrial Membrane Potential (DCm). Cells (2 3 105)
`were incubated with 40 nM DiOC6 (Molecular Probes Inc., Eugene, OR) in
`PBS at 37°C for 20 min and then analyzed by flow cytometry as described
`previously (29). The percentage of cells exhibiting a low level of DiOC6
`uptake, which reflects loss of mitochondrial membrane potential, was deter-
`mined using a Becton Dickinson FACScan (Becton Dickinson, San Jose, CA).
`Cell Cycle Analysis and S-phase Content. Cells (2 3 106) were pelleted
`at 4°C, resuspended, fixed at 4°C with 67% ethanol overnight, and treated on
`ice with a propidium iodide solution containing 3.8 mM Na citrate, 0.5 mg/ml
`RNase A (Sigma Chemical Co.), and 0.01 mg/ml propidium iodide (Sigma
`Chemical Co.) for 3 h. Cell cycle analysis was performed by flow cytometry
`using Verity Winlist software (Topsham, ME).
`Incorporation of BrdUrd was monitored to evaluate S-phase content. For
`each condition, 2 3 106 cells (cell density 5 5 3 105/ml) were incubated with
`10 mM BrdUrd for 30 min at 37°C. After washing twice with 1% BSA/PBS, the
`cells were resuspended in 70% ethanol and fixed for 30 min on ice. The
`BrdUrd-labeled cells were denatured and nuclei released by incubation with 2
`N HCl/0.5% Triton X-100 for 30 min at room temperature. After centrifuga-
`tion, the pellet was resuspended in 0.1 M Na2B4O4 (pH 8.5) to neutralize the
`Cells. U937, HL-60, Jurkat, CCRF-CEM, and Raji cells are human histi-
`acid. Cells (1 3 106)/100 ml in 0.5% Tween 20/1% BSA/PBS were incubated
`ocytic lymphoma, acute promyelocytic leukemia, acute T-cell leukemia, acute
`with FITC-conjugated anti-BrdUrd (1:10; mouse monoclonal; DAKO, Carpin-
`lymphoblastic leukemia, and Burkitt lymphoma cell lines, respectively. All of
`teria, CA) for 30 min at 4°C. After washing once with 0.5% Tween 20/1%
`the cells were derived by the American Type Culture Collection and main-
`BSA/PBS, the cells were resuspended in PBS containing 5 mg/ml propidium
`tained in RPMI 1640 medium containing 10% FBS, 200 units/ml penicillin,
`iodide and analyzed by flow cytometry. The percentage of S-phase cells was
`200 mg/ml streptomycin, minimal essential vitamins, sodium pyruvate, and
`determined by measuring BrdUrd FITC-positive part in a dot plot of FL-3 (red
`glutamine, as reported previously (28). U937/p21AS and U937/pREP4 cells
`fluorescence) against FL-1 (green fluorescence).
`were obtained by stable transfection of cells with plasmids containing anti-
`Immunoblot and Immunoprecipitation Analysis. Whole-cell pellets
`sense-oriented p21 cDNA or an empty vector (pREP4), and clones were
`were lysed by sonication in 1 3 sample buffer [62.5 mM Tris base (pH6.8), 2%
`selected with hygromycin (29).
`SDS, 50 mM DTT, 10% glycerol, 0.1% bromphenol blue, and 5 mg/ml each
`Drugs and Reagents. Selective MEK inhibitors (PD98059 and UO126),
`chymostatin, leupeptin, aprotinin, pepstatin, and soybean trypsin inhibitor] and
`selective PKC inhibitors (GF 109203X or GFX I and safingol), and specific
`boiled for 5 min. For analysis of phospho-proteins, 1 mM each Na vanadate and
`inhibitors of p38 MAPK (SB203580) were supplied by Calbiochem (San
`Na PPi was added to the sample buffer. Protein samples were collected from
`Diego, CA) as powder. The MEK inhibitor PD184352 was kindly provided by
`the supernatant after centrifugation of the samples at 12,800 3 g for 5 min, and
`Dr. Judith Sebolt-Leopold (Warner Lambert/Parke-Davis Co., Ann Arbor, MI).
`protein was quantified using Coomassie Protein Assay Reagent (Pierce, Rock-
`Materials were dissolved in sterile DMSO and stored frozen under light-
`ford, IL). Equal amounts of protein (30 mg) were separated by SDS-PAGE and
`protected conditions at 220°C. UCN-01 was kindly provided by Dr. Edward
`electrotransferred onto a nitrocellulose membrane. For blotting phospho-
`Sausville (Developmental Therapeutics Program/Cancer Treatment and Eval-
`proteins, no SDS was included in the transfer buffer. The blots were blocked
`uation Program (CTEP), National Cancer Institute). It was dissolved in DMSO
`at a stock concentration of 1 mM, stored at 220°C, and subsequently diluted
`with 5% milk in PBS-Tween 20 (0.1%) at room temperature for 1 h and probed
`with the appropriate dilution of primary antibody overnight at 4°C. The blots
`with serum-free RPMI medium before use. Caffeine (Alexis Co., San Diego,
`CA) was dissolved in chloroform and stored at 220°C. In all of the experi-
`were washed twice in PBS-Tween 20 for 15 min and then incubated with a
`1:2000 dilution of horseradish peroxidase-conjugated secondary antibody
`ments, the final concentration of DMSO or chloroform did not exceed 0.1%.
`(Kirkegaard & Perry, Gaithersburg, MD) in 5% milk/PBS-Tween 20 at room
`Caspase inhibitor (Z-VAD-fmk) and caspase 8 inhibitor (Z-IETD-fmk) were
`temperature for 1 h. After washing twice in PBS-Tween 20 for 15 min, the
`purchased from Enzyme System Products (Livermore, CA), dissolved in
`proteins were visualized by Western Blot Chemiluminescence Reagent (NEN
`DMSO, and stored at 4°C. Cycloheximide was purchased from Sigma Chem-
`Life Science Products, Boston, MA). For analysis of phospho-proteins, Tris-
`ical Co. (St. Louis, MO), stored frozen in DMSO, and diluted in RPMI 1640
`buffered saline was used instead of PBS throughout. Where indicated, the blots
`medium before use.
`were reprobed with antibodies against actin (Signal Transduction Laboratories)
`Experimental Format. All of the experiments were performed using log-
`arithmically growing cells (3–5 3 105 cells/ml). Cell suspensions were placed
`or tubulin (Calbiochem) to ensure equal loading and transfer of proteins. The
`in sterile 25 cm2 T-flasks (Corning, Corning, NY) and incubated with MEK or
`following antibodies were used as primary antibodies: phospho-p44/42 MAPK
`(Thr202/Tyr204) antibody (1:1000; rabbit polyclonal; NEB, Beverly, MA);
`PKC inhibitors for 30 min at 37°C. At the end of this period, UCN-01 (or in
`p44/42 MAPK antibody (1:1000; rabbit polyclonal; NEB); phospho-p38
`some cases, caffeine) was added to the suspension, and the flasks were placed
`MAPK (Thr180/Tyr182) antibody (1:1000; rabbit polyclonal; NEB); phospho-
`in 37°C/5% CO2 incubator at various intervals, generally 18 h. In some studies,
`SAPK/JNK (Thr183/Tyr185) antibody (1:1000; rabbit polyclonal; Cell Sig-
`the p38 MAP kinase inhibitor SB203580 was added concurrently with MEK
`naling Technology, Beverly, MA); SAPK/JNK antibody (1:1000; rabbit poly-
`inhibitors. After drug treatment, cells were harvested and subjected to further
`clonal; Cell Signaling Technology); anti-phospho-CREB (1:1000;
`rabbit
`analysis as described below.
`Analysis of Apoptosis. The extent of apoptosis was evaluated by assess-
`polyclonal; Upstate Biotechnology, Lake Placid, NY); phospho-cdc2 (Tyr15)
`antibody (1:1000; rabbit polyclonal; Cell Signaling Technology); anti-p21Cip/
`ment of Wright-Giemsa-stained preparation under light microscopy and scor-
`WAF1 (1:500; mouse monoclonal; Transduction Laboratories, Lexington,
`ing the number of cells exhibiting classic morphological features of apoptosis.
`KY); anti-p27kip1 (1:500; mouse monoclonal; PharMingen, San Diego, CA);
`For each condition, 5 to 10 randomly selected fields/condition were evaluated,
`MAP kinase phosphatase-1 (M-18; 1:200; rabbit polyclonal; Santa Cruz Bio-
`encompassing at least 500 cells (28). To confirm the results of morphological
`technology Inc., Santa Cruz, CA); MAP kinase phosphatase-3 (C-20; 1:100;
`analysis, in some cases cells were also evaluated by TUNEL staining (30) and
`goat polyclonal; Santa Cruz Biotechnology Inc.); antihuman Bcl-2 oncoprotein
`assessment of oligonucleosomal DNA fragmentation of total DNA. DNA
`fragmentation was analyzed by 1.8% agarose gel electrophoresis as described
`(1:2000; mouse monoclonal; DAKO, Carpinteria, CA); Bax (N-20; 1:2000;
`rabbit polyclonal; Santa Cruz Biotechnology Inc.); Bcl-xS/L (S-18; 1:500;
`previously (31). For TUNEL staining, cytocentrifuge preparations were ob-
`5107
`
`
`
`Downloaded from on April 18, 2016. © 2001 American Association for Cancercancerres.aacrjournals.org
`
`
`Research.
`
`Page 2 of 11
`
`Fluidigm
`Exhibit 1008
`
`

`

`INDUCTION OF APOPTOSIS BY UCN-01 AND MEK INHIBITORS
`
`RESULTS
`
`to the flasks, after which they were placed in the incubator for 24 h. At the end
`of this period, cytospin preparations were obtained and stained with Wright-
`Giemsa, and the cells were scored under light microscopy for the typical
`morphological features of apoptosis.
`Statistical Analysis. For morphological assessment of apoptotic cells, cell
`cycle analysis, S-phase content, cdk1/cdc2 kinase assay, analysis of DCm, and
`clonogenic and cell proliferation assays, experiments were repeated at least
`three times. Values represent the means 6 SD for at least three separate
`experiments performed in triplicate. The significance of differences between
`experimental variables was determined using the Student t test.
`
`The effects of combined exposure of human monocytic leukemia
`cells (U937) to UCN-01 and the MEK inhibitor PD184352 were first
`examined in relation to MAPK activation and apoptosis (Fig. 1).
`Unexpectedly, incubation with UCN-01 (150 nM) induced phospho-
`rylation (activation) of MAPK by 2 h, and this effect persisted over
`the ensuing 18 h (Fig. 1A). Coincubation of U937 cells with
`PD184532 (10 mM) attenuated induction of phospho-MAPK at 2 h,
`and inhibition of MAPK activation was essentially complete after
`18 h. To determine what impact this phenomenon had on cell fate, the
`extent of apoptosis was monitored in cells exposed to each agent
`individually and in combination. Whereas exposure to PD184352 or
`
`rabbit polyclonal; Santa Cruz Biotechnology Inc.); antihuman/mouse XIAP
`(1:500; rabbit polyclonal; R&D System, Minneapolis, MN); anti-caspase-3
`(1:1000; rabbit polyclonal; PharMingen); cleaved-caspase-3 (Mr 17,000) anti-
`body (1:1000; rabbit polyclonal; Cell Signaling Technology); anti-caspase-9
`(1:1000; rabbit polyclonal; PharMingen); anti-PARP (1:2500; mouse mono-
`clonal; Calbiochem); and cleaved PARP (Mr 89,000) antibody (1:1000; rabbit
`polyclonal; Cell Signaling Technology).
`Immunoprecipitation was performed to determine the extent of cdc25C
`activation (32). Briefly, 2 3 107 cells were lysed in RIPA buffer (1% NP40,
`0.5% Na deoxycholate, 1 mM phenylmethylsulfonyl fluoride, 1 mM Na vana-
`date, 5 mg/ml chymostatin, leupeptin, aprotinin, pepstatin, and soybean trypsin
`inhibitor, and 0.1% SDS in PBS) by syringing approximately 20 times with a
`23-gauge needle. Protein samples were centrifuged at 12,800 3 g for 30 min
`and quantified. Two-hundred mg of protein/condition were incubated under
`continuous shaking with 1 mg of anti-cdc25C (mouse monoclonal; PharMin-
`gen) overnight at 4°C. Twenty ml/condition of Dynabeads (goat antimouse
`IgG; Dynal, Oslo, Norway) were added and incubated for an additional 4 h.
`After washing three times with RIPA buffer, the bead-bound protein was
`eluted by vortexing and boiling in 20 ml of 13 sample buffer. The samples
`were separated by 12% SDS-PAGE and subjected to immunoblot analysis as
`described above. Anti-14-3-3b (rabbit polyclonal; Santa Cruz Biotechnology
`Inc.) was used as primary antibody at a dilution of 1:200.
`Analysis of Cytosolic Cytochrome c. Cells (2 3 106) were washed in PBS
`and lysed by incubating for 30 s in lysis buffer (75 mM NaCl, 8 mM Na2HPO4,
`1 mM NaH2PO4, 1 mM EDTA, and 350 mg/ml digitonin). The lysates were
`centrifuged at 12,000 3 g for 1 min, and the supernatant was collected and
`added to an equal volume of 2 3 sample buffer. The protein samples were
`quantified, separated by 15% SDS-PAGE, and subjected to immunoblot anal-
`ysis as described above. Anticytochrome c (mouse monoclonal; PharMingen)
`was used as primary antibody at a dilution of 1:500.
`Cdk1/cdc2 Kinase Assay. Cdk1/cdc2 Kinase Assay Kit (Upstate Biotech-
`nology) was used to determine the activity of cdk1/cdc2 kinase according to
`the manufacturer’s instructions. Briefly, 2 3 107 cells were lysed in RIPA
`buffer by sonication. Protein samples were centrifuged at 12,800 3 g for 30
`min and quantified. Fifty mg of protein/condition were incubated with 400
`mg/ml histone H1, 2 mCi of [g-32P]ATP, and 1:5 inhibitor cocktail in assay
`dilution buffer (total volume, 50 ml) at 30°C for 20 min. A 25-ml aliquot of
`reaction mixture was transferred onto P81 paper. After washing three times
`with 0.75% phosphoric acid and once with acetone, cpm of [g-32P] incorpo-
`rated into histone H1 was monitored using TRI-CARB 2100TR Liquid Scin-
`tillation Analyzer (Packard Instrument Co., Downers Grove, IL). In some
`cases, 10 ml of 23 sample buffer was added to 10 ml of the reaction mixture
`and boiled for 5 min. [g-32P]histone H1 was separated by 12% SDS-PAGE and
`visualized by exposure of the dried gels to X-ray film (KODAK) at 280°C
`for 1 h.
`Clonogenic Assay and Cell Proliferation Assays. Colony formation after
`drug treatment was evaluated using a soft agar cloning assay as described
`previously (33). Briefly, cells were washed three times with serum-free RPMI
`medium. Subsequently, 500 cells/well were mixed with RPMI medium con-
`taining 20% FBS and 0.3% agar and plated on 12-well plates (three wells/
`condition). The plates were then transferred to a 37°C/5% CO2, fully humid-
`ified incubator. After 10 days of incubation, colonies, consisting of groups of
`.50 cells, were scored using an Olympus Model CK inverted microscope, and
`Fig. 1. A, logarithmically growing U937 cells were incubated for the designated
`colony formation for each condition was calculated in relation to values
`intervals in the presence of 150 nM UCN-01 6 10 mM PD184352, after which cells were
`obtained for untreated control cells. For cell viability assays, CellTiter 96
`lysed, and proteins were separated by SDS-PAGE and probed with antibodies directed
`AQueous One Solution (Promega, Madison, WI) was used according to the
`against phospho-ERK, as described in “Materials and Methods.” Blots were subsequently
`manufacturer’s instructions, and the absorbance at 490 nm was recorded using
`stripped and reprobed with antibodies directed against total ERK. Two additional studies
`yielded equivalent results. B, cells were treated with PD184352 (PD) and/or UCN-01
`a 96-well plate reader (Molecular Devices, Sunnyvale, CA).
`(UCN; 61 mM CHX) as above for 18 h, after which Wright Giemsa-stained cytospin
`Normal Peripheral Blood Mononuclear Cells. Peripheral blood was ob-
`preparations were evaluated by light microscopy, and the percentage of cells exhibiting
`tained with informed consent from normal volunteers, diluted 1:3 with RPMI
`classic apoptotic features was determined by examining 5–10 randomly selected fields
`encompassing $500 cells. Values represent the means 6 SD for three separate experi-
`1640 medium, and layered over a cushion of 10 ml of Ficoll-Hypaque (specific
`ments performed in triplicate. C, cells were treated as in A, and the proteins were separated
`gravity, 1.077; Sigma Chemical Co.) in sterile 50-ml plastic centrifuge tubes.
`by SDS-PAGE and probed with antibodies directed against caspase-3, caspase-9, or
`These studies have been approved by the Human Investigations Committee of
`PARP. CF, cleavage fragment. Alternatively, cytosolic fractions were obtained as de-
`Virginia Commonwealth University. After centrifugation for 40 min at
`scribed in “Materials and Methods,” and expression of cytochrome c was monitored as
`400 3 g at room temperature, the interface layer, consisting of mononuclear
`above. Each lane was loaded with 30 mg of protein. Blots were stripped and reprobed with
`antibodies to actin or tubulin to ensure equal loading and transfer. D, cells were treated
`cells, was extracted with a sterile Pasteur pipette and diluted in fresh RPMI
`with UCN-01 6 PD184352 (6 1 mM cycloheximide) as above, after which the percentage
`medium. The cells were washed 32 in medium and resuspended in RPMI 1640
`of cells exhibiting reduced mitochondrial membrane potential (DCm) was determined by
`medium containing 10% FCS in 25-cm2 tissue culture flasks at a cell density
`monitoring DiOC6 uptake as described in “Materials and Methods.” Results represent the
`of 106 cells/ml. Various concentrations of UCN-01 6 PD 184352 were added
`means 6 SD for three separate experiments performed in triplicate.
`5108
`
`
`
`Downloaded from on April 18, 2016. © 2001 American Association for Cancercancerres.aacrjournals.org
`
`
`Research.
`
`Page 3 of 11
`
`Fluidigm
`Exhibit 1008
`
`

`

`INDUCTION OF APOPTOSIS BY UCN-01 AND MEK INHIBITORS
`
`investigated (23), and U0126 (20 mM), the affinity of which for the
`CDK ATP-binding site is significantly greater than that of PD98059
`(24). As noted in the case of PD184352, coadministration of mini-
`mally toxic concentrations of PD98059 or U0126 with 200 nM
`UCN-01 resulted in a marked potentiation of cell death, manifested by
`an increase in the morphological features of apoptosis (Fig. 3, A and
`C), PARP degradation, and release of cytochrome c into the cyto-
`plasm (Fig. 3, B and D). These findings demonstrated that multiple
`pharmacological MEK inhibitors are capable of substantially increas-
`ing the lethal actions of UCN-01 toward U937 cells.
`To establish whether the enhanced lethality of MEK inhibitors and
`UCN-01 was restricted to U937 cells or, instead, might be generalized
`to include other leukemia cell types, the effects of combined exposure
`to UCN-01 and PD184352 were examined in several additional leu-
`kemia cell lines (Fig. 4). Because the sensitivity of these cells to
`UCN-01 differed somewhat from that of U937 cells, slightly higher
`UCN-01 concentrations (e.g., 150 –300 nM) were used in some cases.
`On the basis of standard morphological criteria as well as evidence of
`PARP degradation, it can be seen that combined treatment with
`UCN-01 and PD184352, administered at concentrations that were
`marginally toxic by themselves, resulted in a dramatic increase in cell
`death in HL-60 promyelocytic leukemia cells, T-lymphoblastic
`CCRF-CEM and Jurkat cells, and B-lymphoblastic lymphoma Raji
`cells (Fig. 4, A and B). Qualitatively similar results were obtained
`when PD98059 and U0126 were used (data not shown). As in the case
`of U937 cells, UCN-01 treatment resulted in a substantial increase in
`MAPK activation in HL-60, CCRF-CEM, and in Jurkat cells (Fig.
`4C); moreover, this effect was blocked by PD184352 (5 mM). Thus,
`combined treatment with UCN-01 and MEK inhibitors prevented
`MAPK activation and produced a dramatic increase in apoptosis in a
`variety of myeloid and lymphoid cell lines.
`To investigate the hierarchy of events accompanying apoptosis
`induced by these agents, U937 cells were exposed to the combination
`of UCN-01 (150 nM) in conjunction with 10 mM PD184352 in the
`
`Fig. 2. Top panels, cells were exposed to UCN-01 (150 nM) 6 PD184352 (10 mM) as
`above for 18 h, after which cytospin preparations were obtained and TUNEL staining was
`performed as described in “Materials and Methods.” Cells were then viewed under
`fluorescence microscopy at 350 magnification. A, control; B, PD1843252; C, UCN-01; D,
`UCN-01 1 PD184352. Bottom panel, cells were treated as above after which cells were
`lysed, and DNA was extracted, separated by agarose gel electrophoresis, and stained with
`ethidium bromide as described in “Materials and Methods.” Lanes (20 mg of DNA each):
`M, molecular weight marker; A, control; B, PD184352; C, UCN-01; D, UCN-
`01 1 PD184352.
`
`150 nM UCN-01 alone was minimally toxic to these cells (,10%
`apoptosis in each case), combined treatment resulted in a dramatic
`increase in cell death (i.e., ;60%; Fig. 1B). Furthermore, this effect
`was not attenuated by coadministration of the protein synthesis inhib-
`itor CHX (1 mM). Consistent with these findings, combined treatment
`with UCN-01 and PD184352, but not individual exposure, induced
`marked cleavage of procaspases-3 and -9, PARP degradation, and
`cytochrome c release into the cytoplasmic S-100 fraction (Fig. 1C).
`Cotreatment of cells with UCN-01 and PD184352 also resulted in a
`marked increase in the number of cells exhibiting loss of the mito-
`chondrial membrane potential (e.g., Dc
`m; Fig. 1D), an action that was
`also not attenuated by CHX. TUNEL assays confirmed that a small
`number of cells exposed to UCN-01 or PD184352 alone for 18 h (Fig.
`2, B and C) displayed DNA breaks containing overhanging 39-OH
`ends, whereas coexposure resulted in a high percentage of TUNEL-
`positive cells. Similarly, agarose gel electrophoresis demonstrated a
`marked increase in oligonucleosomal DNA fragmentation in cells
`Fig. 3. A, U937 cells were exposed to UCN-01 (200 nM; UCN) 6 PD98059 (50 mM;
`PD98) for 24 h, after which the percentage of apoptotic cells was scored as described
`exposed to both agents (Fig. 2; bottom panel). Together, these find-
`above (Fig. 1). B, cells were exposed to UCN-01 (200 nM) 6 PD98059 (50 mM) for 24 h,
`ings indicate that coadministration of the MEK inhibitor PD184352
`after which cells were lysed, and proteins were separated by SDS-PAGE and probed for
`blocks MAPK activation and dramatically increases apoptosis in
`expression of PARP and cytosolic cytochrome c as described in Fig. 1. CF, cleavage
`fragment. C, cells were exposed to UCN-01 (200 nM) 6 20 mM U0126 (UO1) for 24 h,
`U937 cells exposed to a marginally toxic concentration of UCN-01.
`after which apoptosis was determined as in A. D, cells were exposed to UCN-01 6 U0126
`To determine whether these findings could be extended to other
`as above, after which expression of PARP and cytosolic cytochrome c were determined
`as above. For A and C, values represent the means 6 SD for three separate experiments
`known MEK inhibitors, U937 cells were incubated for 24 h with 200
`performed in triplicate. For B and D, each lane was loaded with 30 mg of protein. Blots
`nM UCN-01 either alone or in combination with PD98059 (50 mM), an
`were stripped and reprobed with antibodies directed against actin or tubulin to ensure
`aminoflavone that was among the earliest of the MEK inhibitors to be
`equal loading and transfer. Two additional studies yielded equivalent results.
`5109
`
`
`
`Downloaded from on April 18, 2016. © 2001 American Association for Cancercancerres.aacrjournals.org
`
`
`Research.
`
`Page 4 of 11
`
`Fluidigm
`Exhibit 1008
`
`

`

`INDUCTION OF APOPTOSIS BY UCN-01 AND MEK INHIBITORS
`
`Fig. 4. A, HL-60 promyelocytic leukemia cells, Jurkat and CCRF-
`CEM lymphoblastic leukemia cells, and Raji B-lymphoblastic leukemia
`cells were exposed to PD184352 (PD; 5 mM) 6 UCN-01 (UCN; 300 nM
`HL-60; 150 nM Jurkat; 200 nM CCRF; 200 nM Raji) for 24 h, after which
`the percentage of apoptotic cells was determined as described above.
`Values represent the means 6 SD for three separate experiments per-
`formed in triplicate. B, cells were treated as above, after which cells were
`lysed, and the lysates were separated by SDS-PAGE and probed with
`antibodies directed against PARP. Each lane was loaded with 30 mg of
`protein. Blots were subsequently stripped and reprobed with antibodies
`to tubulin or actin to ensure equivalent loading and transfer. CF, cleav-
`age fragment. C, cells were treated as above, after which Western
`analysis was performed to assess expression of phospho-ERK as de-
`scribed in “Materials and Methods.” For B and C, the results of a
`represen

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