throbber
Endocrine-Related Cancer (2001) 8 249–258
`
`mTOR, a novel target in breast cancer: the
`effect of CCI-779, an mTOR inhibitor, in
`preclinical models of breast cancer
`
`K Yu, L Toral-Barza, C Discafani, W-G Zhang, J Skotnicki, P Frost
`and J J Gibbons
`Wyeth-Ayerst Research, Department of Oncology, 401 North Middletown Road, Pearl River, New York 10965, USA
`(Requests for offprints should be addressed to J J Gibbons Jr; Email: gibbonj@war.wyeth.com)
`
`Abstract
`
`The mammalian target of rapamycin (mTOR) is a central regulator of G1 cell cycle protein synthesis
`that precedes commitment to normal cellular replication. We have studied the effect of cell cycle
`inhibitor-779 (CCI-779), a rapamycin ester that inhibits mTOR function, on the proliferation of a panel
`of breast cancer cell lines. Six of eight lines studied were sensitive (IC50압 50 nM) and two lines were
`resistant (IC50>1.0 µM) to CCI-779. Sensitive lines were estrogen dependent (MCF-7, BT-474,
`T-47D), or lacked expression of
`the tumor suppressor PTEN (MDA-MB-468, BT-549), and/or
`overexpressed the Her-2/neu oncogene (SKBR-3, BT-474). Resistant
`lines (MDA-MB-435,
`MDA-MB-231) shared none of these properties. CCI-779 (50 nM) inhibited mTOR function in both a
`sensitive and a resistant line. In nu/nu mouse xenografts, CCI-779 inhibited growth of MDA-MB-468
`(sensitive) but not MDA-MB-435 resistant tumors. Treatment of sensitive lines with CCI-779 resulted
`in a decrease in D-type cyclin and c-myc levels and an increase in p27kip-1 levels. There was good
`correlation between activation of
`the Akt pathway and sensitivity to CCI-779. Amplification of
`mTOR-regulated p70S6 kinase, which is downstream of Akt, may also have conferred CCI-779
`sensitivity to MCF-7 cells. Taken together, the data suggest that mTOR may be a good target for
`breast cancer therapy, especially in tumors with Akt activation resulting from either growth factor
`dependency or loss of PTEN function.
`Endocrine-Related Cancer (2001) 8 249–258
`
`Introduction
`
`Cell cycle inhibitor-779 (CCI-779) is an ester derivative of
`the natural product
`rapamycin that was developed for
`intravenous use for cancer chemotherapy. Rapamycin is a
`macrolide antibiotic with anti-fungal, immunosuppressive,
`and anti-tumor properties (Sehgal et al. 1994). Genetic
`studies in yeast showed that rapamycin inhibited cell growth
`by blocking the function of the proteins TOR1 and TOR2
`(targets of rapamycin 1 and 2) (Heitman et al. 1991). The
`TOR proteins are members of
`the phosphatidylinositol
`3-kinase (PI3-K)-related family of kinases and regulate
`several cellular functions (Schmelzle & Hall 2000). In order
`to inhibit TOR function, rapamycin initially binds to the
`cytoplasmic immunophilin FKBP-12 and the complex then
`inhibits TOR (Brown et al. 1994).
`A mammalian homolog of the yeast TOR proteins has
`been cloned independently by several groups and will be
`referred to in this report as mTOR (Sabers et al. 1995), but
`
`is also known as FRAP (Brown et al. 1994), RAFT
`it
`(Sabatini et al. 1994), and RAPT (Chiu et al. 1994). The
`mTOR protein regulates cell cycle progression, in part, by
`enhancing translation initiation and/or the stability of cell
`cycle
`regulatory
`proteins
`such
`as D-type
`cyclins
`(Hashemolhosseini et al. 1998, Muise-Helmericks et al.
`1998), c-myc (West et al. 1998), and p27kip-1 (Nourse et al.
`1994) among others. At least two direct targets of mTOR,
`p70 S6 kinase and 4E-BP1/PHAS-1, have been suggested to
`mediate the effect of mTOR on protein translation (Brunn et
`al. 1997, Thomas & Hall 1997, Burnett et al. 1998). 4E-BP1
`(eIF-4E binding protein-1) binds
`to the mRNA cap
`recognition element of the translation initiation complex
`protein eIF-4E (eukaryotic initiation factor 4E) and thereby
`inhibits translation initiation (Beretta et al. 1996). mTOR
`phosphorylation of 4E-BP1 causes it
`to dissociate from
`eIF-4E,
`thus enhancing the translation initiation complex
`interactions with the mRNA 5′ cap. The kinase p70 S6K is
`phosphorylated
`and
`activated
`by mTOR,
`and
`then
`
`Endocrine-Related Cancer (2001) 8 249–258
`1351-0088/01/008–249  2001 Society for Endocrinology Printed in Great Britain
`
`Online version via http://www.endocrinology.org
`
`Roxane Labs., Inc.
`Exhibit 1035
`Page 001
`
`

`
`Yu et al.: mTOR, a novel target in breast cancer
`
`phosphorylates the S6 protein of the 40S ribosomal complex.
`Phosphorylation of S6 results in enhanced translation of
`in the 5′
`proteins that contain a polypyrimidine tract
`untranslated region (Jeffries et al. 1997, Volarevic & Thomas
`2000). In addition to regulating protein translation, mTOR
`can also regulate the stability of some cell cycle regulatory
`proteins such as D-type cyclins and p27kip-1. Activation of
`mTOR
`appears
`to
`stabilize
`D-type
`cyclins
`(Hashemolhosseini et al. 1998) and to destabilize the
`(Nourse et al.
`cyclin-dependent kinase inhibitor p27kip-1
`1994).
`Several of the cell cycle targets that are regulated by
`mTOR have been reported to be dysregulated in human
`breast cancer,
`including eIF-4E (Kerekatte et al. 1995),
`D-type cyclins
`(Weinstat-Saslow et al. 1995), p27kip-1
`(Fredersdorf et al. 1997), and c-myc (Liao & Dickson 2000).
`Therefore, we have begun to study the effect of the mTOR
`inhibitor CCI-779 in models of human breast cancer. Cell
`growth in culture revealed that 6 of 8 breast cancer lines
`studied were inhibited by CCI-779 with IC50s in the low nM
`range. Two lines, however, were found to be markedly
`resistant (IC50>1 µM). Cell lines sensitive to CCI-779 were
`estrogen receptor positive, or overexpressed Her-2/neu, or
`had lost
`the
`tumor
`suppressor gene product PTEN
`(phosphatase related to tensin and deleted on chromosome
`10) (Li et al. 1997). Sensitive lines contained higher levels
`of the activated form of Akt, suggesting that this PI3-K
`downstream target may be a common link between growth
`factor-dependent
`lines
`(estrogen,
`Her-2/neu)
`and
`PTEN-deleted lines that are sensitive to mTOR inhibition.
`The potential for therapeutic use of an mTOR inhibitor is
`discussed in terms of the rationale provided by known
`genetic alterations in human breast cancer cells.
`
`Materials and methods
`
`Chemicals and cell culture methods
`
`All chemicals were obtained from Sigma-Aldrich (St Louis,
`MO, USA). CCI-779 was synthesized at Wyeth-Ayerst
`Research. Cell
`lines
`of MDA-MB-468
`(MDA-468),
`MDA-MB-435 (MDA-435), MDA-MB-231 (MDA-231),
`MCF-7, T-47D, SKBR-3 and BT-474 were obtained from
`the American Type Culture Collection (ATCC) (Rockville,
`MD, USA). BT-474G is a sub-clone derived from BT-474.
`All cell lines were cultured in Minimum Essential Medium
`(MEM) containing 10% fetal bovine serum (FBS) and 1 mM
`MEM sodium pyruvate in a 37°C incubator containing 5%
`CO2. All cell culture reagents were purchased from
`Gibco-BRL (Grand Island, NY, USA).
`
`Proliferation assay
`Cells were plated in 96-well culture plates at about 3000 cells
`per well. One day following plating, drugs were added to
`
`cells. Three days after drug treatment, viable cell densities
`were determined by measuring metabolic conversion (by
`viable cells) of the dye MTS, a previously established cell
`proliferation assay. Stock solutions of MTS and PMS were
`purchased from Promega Corp. (Madison, WI, USA). For
`each assay, MTS and PMS stocks were freshly thawed and
`mixed (MTS/PMS, 20:1). The MTS/PMS mixture was then
`added to 96-well cell plates at 20 µl/well, and plates were
`incubated for 1–2 h in cell culture incubator. MTS assay
`results were read in a 96-well
`format plate reader by
`measuring absorbance at 490 nm. The effect of each drug
`treatment was calculated as a percentage of control cell
`growth obtained from vehicle-treated cells grown in the same
`culture plate.
`
`In vivo tumor inhibition
`Xenograft model athymic nu/nu female mice, 5–6 weeks of
`age, were obtained from Charles River Laboratories,
`Wilmington, MA, USA and maintained in a barrier facility
`in accordance with Institutional Animal Care and Use
`Committee (IACUC) regulations. Animals were injected s.c.
`with either 6 × 106 MDA-MB468 cells or 6 × 106
`MDA-MB435 cells. When tumors reached a weight of
`between 80 and 120 mg, animals were randomized into
`treatment groups (5 mice/group). Animals were treated
`intraperitoneally (i.p.) for 5 consecutive days with 40, 20, or
`10 mg/kg CCI-779 prepared in 2% ethanol, 8% cremophor
`el, (Sigma, St Louis, MO, USA), or vehicle alone. Tumor
`mass ([length × width2]/2) was determined on days 7, 14, 21
`and 28 post staging. The data were analyzed via Student’s
`t-test. A P-value <0.05 indicates a statistically significant
`reduction in relative tumor growth of the treated group
`compared with that of the vehicle control group.
`
`Protein lysates and immunoblotting
`For immunoblotting experiments, cells were plated in 10-cm
`dishes or 6-well plates. Depending on the study, after the
`cells had completely attached, they were either serum-starved
`or incubated in growth medium overnight. Treatment with
`various inhibitors ranged from 2 to 16 h. After drug
`pretreatment,
`the cells were rinsed once with cold PBS
`(phosphate buffered saline without Mg++ and Ca++) and then
`lysed in cold gentle lysis buffer (25 mM Hepes, pH 7.55,
`100 mM NaCl, 20 mM β-glycerophosphate, 1.5 mM MgCl2,
`0.5 mM EGTA, 0.25 mM EDTA, 1% NP-40, 10 mM
`Na3VO4, 10 µg/ml aprotinin, 10 µg/ml leupeptin, 1 mM
`phenylmethylsulfonylfluoride, 1 µM microcystin LR and
`0.1% 2–mercaptoethanol). In some experiments, cells from
`6-well plates were lysed in NuPAGE-LDS sample buffer
`(Invitrogen, Carlsbad, CA, USA). The crude lysates were
`briefly sonicated and then clarified by centrifugation for 15
`min at 14 000 r.p.m. Cleared lysates (20–50 µg) were
`subjected to SDS-PAGE electrophoresis using the NuPAGE
`system (Invitrogen, Carlsbad, CA, USA) and transferred to
`
`250
`
`www.endocrinology.org
`
`Roxane Labs., Inc.
`Exhibit 1035
`Page 002
`
`

`
`a nitrocellulose membrane. The sources of various primary
`antibodies are as follows. Phospho-AKT (Ser473), AKT,
`phospho-p70 S6
`kinase
`(Thr389),
`p70 S6
`kinase,
`phospho-4E-BP1
`(Thr-37, Thr-46
`and Ser-65) were
`purchased from Cell Signaling Technology (Beverly, MA,
`USA). Antibodies against cyclin D3, c-myc and 4E-BP1
`were from Santa Cruz Biotechnology (Santa Cruz, CA,
`USA). Anti-p27kip1 was from Transduction Laboratories (San
`Diego, CA, USA) and phospho-FKHRL-1 (Thr32) was from
`Upstate Biotechnology
`(Lake
`Placid, NY, USA).
`Immunoblots were blocked for 1 h with blocking buffer
`TBST (20 mM Tris, pH 7.5, 500 mM NaCl2, 0.1%-
`Tween-20) and 5% non-fat milk. After washing, they were
`at 4°C
`incubated with primary antibodies overnight
`according to the manufacturer’s suggestions, washed and
`treated
`with
`appropriate
`secondary
`antibodies.
`Immunoreactive proteins were detected using enhanced
`chemiluminescence (ECL) (Amersham Pharmacia Biotech,
`Piscataway, NJ, USA).
`
`Results
`
`The effect of the mTOR inhibitor CCI-779
`on the growth of human breast cancer lines
`in vitro
`
`The growth inhibitory properties of CCI-779 were studied in
`vitro on a panel of 8 human breast cancer cell lines (Table 1).
`The MCF-7, BT-474, and T-47D cell lines are all estradiol
`responsive (Yarden et al. 1996) and all were strongly growth
`inhibited by CCI-779 (IC50 low nM). Similarly, growth of lines
`BT-549 and MDA-MB-468 which contain deletions of the
`PTEN tumor suppressor gene (Lu et al. 1999) was highly
`sensitive
`to treatment with CCI-779. The Her-2/neu
`overexpressing (Chen et al. 2000) SKBR-3 line and ER
`positive, Her-2 overexpressing BT-474 cells were also
`inhibited at low nM concentrations of CCI-779. Two lines,
`MDA-MB-435 and MDA-MB-231, were resistant to treatment
`with CCI-779 (IC50s at low µM concentrations). These lines do
`
`Table 1 Effect of CCI-779 on the growth of human breast
`cancer lines in vitro.
`
`Cell
`line
`
`MCF-7
`BT-474
`T-47D*
`BT-549*
`MDA-MB-468
`SKBR-3
`MDA-MB-435
`MDA-MB-231
`
`IC50
`(nM)
`
`10–50
`0.6
`<10.0
`<10.0
`0.7
`0.7
`1600
`5900
`
`Estrogen
`receptor α Her-2/Neu PTEN−/−
`+
`−
`−
`+
`+
`−
`+
`−
`−
`+
`+
`−
`−
`−
`
`−
`−
`−
`
`+
`−
`−
`
`Endocrine-Related Cancer (2001) 8 249–258
`
`not respond to estradiol, do not overexpress Her-2/neu, and are
`wild-type for the tumor suppressor, PTEN.
`
`The mTOR pathway is activated in CCI-779
`sensitive MDA-MB-468 cells and minimally
`activated in CCI-779 resistant MDA-MB-435
`cells
`
`There was a marked difference in the ability of CCI-779 to
`inhibit MDA-468 (PTEN−/−) cells compared with MDA-435
`(PTEN+/+) cells (Fig. 1A). The PTEN−/−, CCI-779 sensitive
`MDA-468 line showed evidence of Akt activation, as has
`been reported by others (Lu et al. 1999, Weng et al. 1999).
`A comparison of Western blots using a phospho-specific
`antibody for
`the activated form of Akt shows marked
`activation relative to total Akt protein in the MDA-468 cells
`compared with the MDA-435 cells (Fig. 1B). The forkhead
`transcription factor (FKHRL−1), a downstream target of Akt
`(Biggs et al. 1999) is also highly phosphorylated, confirming
`that the Akt signaling is activated in these cells. p70 S6K, a
`direct target of mTOR, is also highly phosphorylated in the
`MDA-468 cells, suggesting that
`the mTOR pathway is
`activated in the PTEN−/− cells. Both the Akt and mTOR
`pathways were only minimally activated in the MDA-435
`PTEN wild-type cells.
`
`Inhibition of mTOR function inhibits growth in
`xenografts of MDA-468 (PTEN−/−) cells but not
`MDA-435 (PTEN+/+) cells
`
`CCI-779 produced a similar differential effect on the growth
`of MDA-468 cells compared with MDA-435 cells in vivo
`(Fig. 2). The PTEN mutant MDA-468 cells were sensitive
`whereas the PTEN wild-type MDA-435 cells were not. In
`these experiments, nu/nu mice were injected in the flank with
`tumor cells and tumors were allowed to grow to a size of
`100 mg. Staged mice were then randomized into treatment
`groups and treated with CCI-779 by i.p. injection at 10, 20,
`or 40 mg/kg or with vehicle for 5 consecutive days. Although
`the MDA-468 cells did not grow as well as the MDA-435
`cells in nude mice, there was a clear regression of tumor size
`in MDA-468–treated tumors at doses of 40 and 20 mg/kg.
`Even at the low dose of 10 mg/kg, the delay in growth of
`MDA-468 tumors extended 10 days beyond the last dose of
`CCI-779 on day 5. The growth of MDA-435 tumors was not
`affected by CCI-779 at any dose. The effect of CCI-779 in
`vivo has not been limited to slow growing tumors as we have
`seen similar growth inhibitory effects in the fast growing
`U87 MG glioblastoma (data not shown).
`
`Treatment with CCI-779 inhibits mTOR
`function in both sensitive (MDA-468) and
`resistant (MDA-435) cells
`
`*Data from National Cancer Institute 60 cell panel screen
`(Monks et al. 1991).
`
`MDA-468 cells were sensitive to growth inhibition by the
`mTOR inhibitor CCI-779 in vitro and in vivo while
`
`www.endocrinology.org
`
`251
`
`Roxane Labs., Inc.
`Exhibit 1035
`Page 003
`
`

`
`Yu et al.: mTOR, a novel target in breast cancer
`
`Figure 1 Loss of PTEN tumor suppressor in MDA-468 cells results in an elevated sensitivity to growth inhibition by CCI-779.
`(A) Growth inhibition curves. Cells were plated in 96-well cell culture plates at 3000 cells per well overnight before treatment
`with CCI-779 for 3 days. Cell growth assays were performed by standard MTS assay as described in Materials and methods.
`(B) Elevated AKT and mTOR signaling in MDA-468 cells. Cells were plated in 10-cm culture plates overnight and were then
`serum-starved for 24 h with culture medium containing 0.1% serum. Total cellular lysates were prepared using the gentle lysis
`buffer described in Materials and methods. Equal amounts (50 µg) of total proteins were analyzed by immunoblotting with
`antibodies of AKT, phospho (P)-AKT (S473), phospho (P)-FKHRL-1 (T32) and phospho (P)-p70 S6K (T389) as described in
`Materials and methods.
`
`Figure 2 Nu/nu mice (5 mice/group) were injected in the flank with either MDA-MB-468 or MDA-MB-435 cells (6×106/mouse).
`Mice were randomized into treatment groups after tumors reached a size of about 100 mg. Treatment with CCI-779 or vehicle
`was for days 1–5 after staging and mice were not treated thereafter.
`
`252
`
`www.endocrinology.org
`
`Roxane Labs., Inc.
`Exhibit 1035
`Page 004
`
`

`
`MDA-435 cells were not. We studied by Western blot the
`effect of CCI-779 on mTOR function in these cells by
`looking at the phosphorylation status of the mTOR targets
`p70 S6K and 4E-BP1 (Fig. 3). Cells were treated in vitro for
`16 h with CCI-779 at a concentration (50 nM) that inhibited
`growth of MDA-468 cells by about 50% and had no effect
`on growth of MDA-435 cells. The level of phosphorylated
`p70 S6K was higher in the MDA-468 cells, presumably due
`to loss of PTEN and activation of Akt (Fig. 1B), and
`CCI-779 treatment resulted in complete dephosphorylation of
`p70 S6K without affecting the total protein levels of p70
`S6K (not shown). There was considerably less but detectable
`phosphorylated p70 S6K in the PTEN+/+ MDA-435 cells;
`however, treatment with CCI-779 also resulted in complete
`dephosphorylation of p70 S6K in these cells (Fig. 3). Similar
`results have been reported for
`rhabdomyosarcoma lines
`sensitive or resistant to rapamycin (Hosoi et al. 1998). Using
`an antibody specific for threonine 46 (T46) on 4E-BP1, a site
`directly phosphorylated by mTOR (Gingras et al. 1999), we
`
`Figure 3 Differential effects of CCI-779 on cellular mTOR
`targets in CCI-sensitive and CCI–resistant cells. MDA-435
`and MDA-468 cells were plated in complete growth medium
`in 6-well culture plates and treated with CCI-779 for 16 h.
`Total cellular proteins were prepared by NuPAGE-LDS
`sample buffer. Immunoblotting assays with antibodies of
`phospho (P)-p70 S6K (T389), phospho (P)-4E-BP1 (T46),
`4E-BP1, c-Myc, cyclin D3 and p27kip1 were performed using
`the NuPAGE system. Cont, control.
`
`Endocrine-Related Cancer (2001) 8 249–258
`
`observed that CCI-779 treatment in MDA-468 cells resulted
`in a shift to a faster migrating species, suggesting inhibition
`of phosphorylation of other residues due to mTOR inhibition
`(Fig. 3). We were surprised to find that
`the resistant
`MDA-435 cells markedly overexpressed 4E-BP1 relative to
`MDA-468 cells. Nevertheless, there was also a shift to faster
`migrating species of 4E-BP1 after CCI-779 treatment of
`MDA-435 cells. This shift was more easily observed with an
`antibody that recognized total 4E-BP1 levels where a more
`condensed faster migrating band was seen after CCI-779
`treatment. Thus treatment with CCI-779 resulted in mTOR
`inhibition in both a sensitive and a resistant line as evidenced
`by a decrease in phosphorylation of the direct mTOR targets
`p70 S6K and 4E-BP1.
`We also looked at downstream cell cycle regulatory
`proteins that are reported to be modulated by mTOR (Fig.
`3). In the sensitive MDA-468 cells, we observed decreases
`in total c-myc and cyclin D3 protein levels after treatment
`with CCI-779 but not
`in the resistant MDA-435 cells.
`Similarly, we observed an increase in p27kip-1 levels in the
`sensitive MDA-468 cells, but not in the resistant cells. These
`data suggest
`that mTOR function is inhibited in both
`sensitive
`and
`resistant
`lines
`but
`the
`downstream
`in PTEN−/− MDA-468 cells,
`consequences are greater
`suggesting they are more dependent on mTOR function.
`Alternatively, there may be other targets of mTOR inhibition
`besides p70 S6K and 4E-BP-1 that are operative in sensitive
`lines but not in resistant lines.
`
`The effect of mTOR inhibition on the growth
`of estrogen-dependent MCF-7 cells
`
`MCF-7 cells do not contain a PTEN mutation or deletion but
`are growth inhibited by about 50% by treatment with 50 nM
`CCI-779. We looked by Western blot at
`the effect of
`CCI-779 treatment on proximal (p70 S6K, 4E-BP1) and
`downstream (c-myc, cyclin D3, p27kip-1) targets in these cells
`(Fig.
`4A). We
`found
`that MCF-7
`cells markedly
`overexpressed the mTOR target p70 S6K (data not shown).
`There were high levels of mTOR-dependent phosphorylated
`p70 S6K in these cells that were completely inhibited by
`CCI-779. Similarly, there was a shift to faster migrating
`species of 4E-BP1 in CCI-779-treated MCF-7 cells. These
`results suggest that phosphorylation of two specific targets of
`mTOR (p70 S6K and 4E-BP1) is inhibited by the drug. We
`also observed a slight decrease in c-myc and cyclin D3 levels
`in CCI-779-treated MCF-7 cells. The levels of p27kip-1 appear
`unchanged after CCI-779 treatment, although very high
`levels of p27kip-1 were seen in untreated MCF-7 cells making
`small changes difficult to detect. We have also looked at the
`status of other targets regulated by Akt signaling (Brunet et
`al. 1999). Phosphorylation of the forkhead transcription
`factor FKHRL-1 is highly elevated (Fig. 4B) compared with
`MDA-435 and MDA-231 cells. Similarly, we also observed
`
`www.endocrinology.org
`
`253
`
`Roxane Labs., Inc.
`Exhibit 1035
`Page 005
`
`

`
`Yu et al.: mTOR, a novel target in breast cancer
`
`Figure 4 Biochemical characterizations of cellular mTOR and AKT targets in MCF-7 cells. (A) Effects of CCI-779 on cellular
`mTOR targets. Cells were plated, treated and analyzed in a similar manner as described in Fig. 3. (B) Cells of the indicated cell
`lines were plated in complete growth medium in 10-cm culture plates. Total cell lysates were made from exponentially growing
`cells in the gentle lysis buffer. Fifty micrograms total proteins per lane were immunoblotted for phospho (P)-AKT (S473) and
`phospho(P)-FKHRL-1 (T32) in a similar manner as described in Fig. 1B.
`
`an increased phosphorylation of the Akt target GSK-3β (data
`not shown). Since MCF-7 cells do not have a high level of
`active Akt-1, the mechanism for elevated phosphorylation of
`FKHRL-1 and GSK-3β remains to be identified. It is also
`possible that deregulation of these targets may contribute to
`its sensitive response to inhibition of mTOR function.
`
`Cells resistant to mTOR inhibition contain
`lower levels of activated Akt than sensitive
`lines
`
`The phosphorylation status of 4E-BP1 is also affected by Akt
`(Gingras et al. 1999) and several laboratories have suggested
`that mTOR is either downstream of PI3-K씮Akt activation
`or activated in parallel with PI3-K/Akt to collaborate on the
`regulation of 4E-BP1 and p70 S6K. Therefore, we compared
`the levels of Akt phosphorylation in a panel of breast cancer
`lines containing cells sensitive or resistant to the mTOR
`
`inhibitor CCI-779 (Fig. 5). Cells were grown in 10% serum
`and harvested prior
`to achieving confluency. The two
`resistant lines, MDA-231 and MDA-435, showed the least
`phosphorylation of Akt relative to total Akt
`levels. The
`highest levels of phospho-Akt, as expected, were seen in the
`PTEN−/− MDA-468 cells. In the Her-2 overexpressing and
`ER positive BT-474
`cells, Akt was
`also
`highly
`phosphorylated and only slightly less so in SKBR-3 cells
`which also overexpress Her-2/neu. The estrogen responsive
`cells MCF-7 and T-47D were intermediate in the level of
`Akt phosphorylation.
`
`Discussion
`
`We have studied the effect of the mTOR inhibitor CCI-779
`on cell growth and cell signal transduction in a panel of
`human breast cancer cell lines. We found that most breast
`cancer
`lines were responsive to CCI-779. While the
`
`254
`
`www.endocrinology.org
`
`Roxane Labs., Inc.
`Exhibit 1035
`Page 006
`
`

`
`Endocrine-Related Cancer (2001) 8 249–258
`
`Figure 5 Survey of the phosphorylated AKT proteins in breast tumor lines. Indicated cell lines were plated in 10-cm dishes in
`complete growth medium. Lysates were prepared in the gentle lysis buffer and immunoblotted for phospho (P)-AKT (S473) and
`AKT in a similar manner as described in Fig. 1B.
`
`molecular basis of responsiveness to mTOR inhibition is not
`known, some observations from the literature regarding the
`responsive cells may be pertinent. Cells with known
`dependence on the hormonal growth factor estradiol or with
`aberrant expression of a growth factor receptor (Her-2/neu)
`were sensitive to in vitro growth inhibition by the mTOR
`inhibitor. Similarly, cells that had lost PTEN, a negative
`regulator of growth factor signaling through PI3-K, were also
`sensitive to CCI-779. These observations are consistent with
`the known effect of mTOR regulation
`on growth
`factor-induced proliferation (Sehgal et al. 1994, Wiederrecht
`et al. 1995). Cell lines in which growth was not inhibited
`by the mTOR inhibitor did not respond to estrogen, did not
`overexpress Her-2/neu, and were wild-type for PTEN.
`We chose one sensitive (MDA-MB-468) and one
`resistant (MDA-MB-435) line to study in vivo in nu/nu
`mouse
`xenografts
`and
`for
`further
`biochemical
`characterization of mTOR inhibition. In vivo, MDA-468
`tumors were inhibited by treatment with CCI-779 (10, 20, or
`40 mg/kg). At the higher doses there was regression of the
`staged tumors and even at the low dose of 10 mg/kg, the
`delay in growth extended 10 days beyond the last dose given.
`The MDA-435 line was not inhibited in the xenograft model
`at any of the doses tested, similar to the refractory phenotype
`observed in vitro.
`By Western blot we showed that the serine threonine
`kinase Akt was highly activated in exponentially growing
`MDA-468 cells but only minimally in MDA-435 cells.
`Activation of Akt in these cells results from loss of function of
`the PTEN tumor suppressor gene (Lu et al. 1999, Weng et al.
`1999) which negatively regulates PI3-K activation of Akt. In
`addition to Akt activation, p70 S6K (a downstream target of
`mTOR) was also highly phosphorylated on Thr-389 in
`MDA-468 cells but only marginally in MDA-435 cells,
`
`suggesting activation of the mTOR pathway in the sensitive
`but not in the resistant line. Treatment of both cell lines with
`CCI-779 resulted in complete dephosphorylation of p70 S6K
`at the mTOR-dependent site Thr-389. With respect to the other
`mTOR target 4E-BP1, treatment with CCI-779 resulted in a
`shift to faster migrating species in both sensitive and resistant
`lines, suggesting that mTOR-dependent phosphorylation of
`4E-BP1 was inhibited in both lines. The level of expression of
`4E-BP1 was markedly higher in the resistant MDA-435 cells
`and although there was a shift to the more dephosphorylated
`form after CCI-779 treatment, constitutive levels of the
`unphosphorylated form were high, suggesting a possible
`mechanism for MDA-435 cell resistance to mTOR regulation.
`This could occur if mTOR was able to phosphorylate only a
`portion of the 4E-BP1,
`leaving high residual
`levels of
`unphosphorylated 4E-BP1 bound to eIF-4E. Additional
`studies to determine if the unphosphorylated 4E-BP1 caused a
`difference in free 4E levels in growing MDA-435 versus
`MDA-468 cells will be necessary to address this possibility.
`Nevertheless, we have shown that
`the difference in
`responsiveness to CCI-779 in these two breast cancer lines is
`not due to the failure of CCI-779 to inhibit mTOR function in
`the resistant cells. Hosoi et al. (1998) have reported the same
`observation
`in
`rapamycin
`sensitive
`and
`resistant
`rhabdomyosarcoma cell lines.
`Although CCI-779 inhibited mTOR function in both
`sensitive and resistant cells, downstream of mTOR the
`response to CCI-779 was different in MDA-468 compared
`with MDA-435 cells (Fig. 3).
`Inhibition of mTOR by
`CCI-779 in MDA-468 cells resulted in decreased cyclin D3
`and c-myc levels and an increase in p27kip-1 levels. These
`effects were not seen in the resistant MDA-435 cells. Lu et
`(1999)
`reported that
`transfection of PTEN into the
`al.
`PTEN−/− MDA-468 cells decreased phosphorylation of p70
`
`www.endocrinology.org
`
`255
`
`Roxane Labs., Inc.
`Exhibit 1035
`Page 007
`
`

`
`Yu et al.: mTOR, a novel target in breast cancer
`
`S6K on the same residues as the mTOR inhibitor rapamycin.
`PTEN transfection also resulted in increased p27kip-1 levels in
`the MDA-468 cells. Thus, PTEN, like the mTOR inhibitor
`CCI-779, inhibited p70 S6K and increased p27kip-1. These
`data suggest that the growth advantage of cells that have lost
`PTEN may, at least in part, be mediated by mTOR. Our data
`showing an increase in p27kip-1 after CCI-779 treatment in
`MDA-468 cells support
`this contention and suggest
`that
`is regulated by PTEN in an mTOR-dependent
`p27kip-1
`manner. The effect of mTOR inhibition on p27kip-1 levels in
`normal T cells has long been known (Nourse et al. 1994).
`Taken together, these data suggest that loss of PTEN results
`in activation of mTOR and that mTOR inhibition may be
`therapeutically effective in breast tumors lacking PTEN.
`D-type cyclins have been reported to be overexpressed
`in human breast cancer (Weinstat-Saslow et al. 1995). The
`mTOR pathway can regulate both the stability and translation
`of D-type
`cyclins
`(Hashemolhosseini
`1998,
`et
`al.
`Muise-Helmericks et al. 1998). Reduction of cyclin D3 in
`the MDA-468 cells by CCI-779, coupled with the increase
`of p27kip-1 may result in redistribution of p27kip-1 towards
`cyclin E–CDK-2 complexes, thereby preventing entry into
`the S phase of the cell cycle.
`Breast lines that are wild-type for PTEN but dependent
`on estrogen were also sensitive to CCI-779. Western analysis
`of MCF-7 cells
`showed that
`these
`cells markedly
`overexpressed p70 S6K and had high levels of
`the
`mTOR-dependent phosphorylated form of p70 S6K when
`grown in 10% serum. The increased p70 S6K expression is
`a function of gene amplification (Barlund et al. 2000, Wu et
`al. 2000) and has been reported to occur in as many as 9%
`of primary breast cancers. Treatment of MCF-7 cells with
`CCI-779 completely inhibited phosphorylation of p70 S6K.
`Similarly, CCI-779 caused a nearly complete shift of 4E-BP1
`to the under-phosphorylated fast migrating species. Cyclin
`D3 levels in MCF-7 cells were reduced by CCI-779 by about
`50% and similar results were seen for cyclin D1 (data not
`shown). It has recently been shown that as little as 30–40%
`reduction in cyclin D1 levels by anti-estrogens in MCF-7
`cells is sufficient to induce a shift in p21cip1 molecules to
`cyclin E–Cdk2 complexes, causing inhibition of progression
`from G1씮S phase of the cell cycle (Carroll et al. 2000).
`Inasmuch as anti-estrogens inhibit D-type cyclin production
`at the transcriptional level and mTOR inhibition decreases
`D-type cyclins at the translational and/or protein stability
`level, there is a strong rationale for combining CCI-779 with
`anti-estrogen therapy.
`In experiments
`to be
`reported
`elsewhere, one of us (P Frost) has found that CCI-779 acted
`synergistically in combination with an anti-estrogen to inhibit
`proliferation of MCF-7 cells in vitro and also potentiated the
`effect of anti-estrogens in vivo in a MCF-7 mouse-xenograft
`model.
`mTOR inhibition also effectively inhibited proliferation
`of Her-2/neu-expressing cells BT-474 and SKBR-3 (Table
`
`1). Lee et al. (2000) recently showed that neu-dependent
`transformation requires cyclin D1 and is induced through an
`E2F-dependent signaling pathway. Although we did not
`directly study the effects of CCI-779 on D-type cyclins in
`the Her-2/neu overexpressing lines, the amply demonstrated
`effect of mTOR inhibition on D-type cyclin levels suggests
`a plausible mechanism for sensitivity of Her-2/neu positive
`tumors to mTOR inhibition.
`The upstream activator(s) of mTOR is not well
`characterized. Studies in yeast (Cardenas et al. 1999) and
`more recently in mammalian cells suggest that mTOR may
`act as a sensor to ensure appropriate nutritional status before
`the cells commit
`to division (Schmelzle & Hall 2000).
`Activation of Akt appears to be upstream of mTOR
`activation in that Akt has been shown to phosphorylate
`mTOR (Sekulic et al. 2000). However, mutation of the site
`on mTOR phosphorylated by Akt did not inhibit downstream
`signaling to p70 S6K or 4E-BP1 (Sekulic et al. 2000). This
`has led to the hypothesis that Akt and mTOR are activated
`by parallel pathways and converge to activate downstream
`targets (Gingras et al. 1999). Nevertheless, it appears that
`Akt and mTOR are activated by growth factors in a
`coordinated if not
`linear fashion and suggests that Akt
`activation may be a marker for enhanced mTOR dependency
`in tumor cells. We found good correlation between Akt
`activation and responsiveness to CCI-779 in the breast cancer
`lines studied. The two resistant lines, MDA-MB-231 and
`MDA-MB-435, showed the least activation of Akt as
`evidenced by phosphorylation of Akt and its downstream
`targets FKHRL-1 (Fig. 4B) and GSK-3β (data not shown).
`One exception was the MCF-7 line, which was sensitive to
`CCI-779 but did not show evidence of strong activation of
`Akt. MCF-7 cells did overexpress highly activated
`mTOR-dependent p70 S6K, suggesting that mTOR may be
`activated in an Akt-independent manner in these cells. The
`resistant line MDA-MB-231 has been shown to overexpress
`Akt-3 (Nakatani et al. 1999). There are three Akt isozymes,
`Akt 1–3, that are reported to be regulated similarly. Our data
`for MDA-MB-231 show no evidence of phosphorylation of
`downstream targets of Akt such as FKHRL-1 and GSK-3β,
`suggesting that even though Akt-3 is overexpressed, it does
`not appear to be constitutively active in these cells. This
`differs from PTEN-deficient cells where it has been shown
`the Akt
`is constitutively active,
`suggesting a greater
`dependency of these cells on the Akt and mTOR pathwa

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