throbber
Enhanced sensitivity of PTEN-deficient tumors to
`inhibition of FRAPymTOR
`
`Mehran S. Neshat*†, Ingo K. Mellinghoff*, Chris Tran*, Bangyan Stiles‡, George Thomas§, Roseann Petersen¶,
`Philip Frost¶, James J. Gibbons¶, Hong Wu‡i, and Charles L. Sawyers*†**
`Departments of *Medicine, ‡Medical and Molecular Pharmacology, §Pathology, †Molecular Biology Institute, and iHoward Hughes Medical Institute,
`University of California School of Medicine, Los Angeles, CA 90095; and ¶Wyeth Ayerst Research, Pearl River, NY 10965
`
`Edited by Richard D. Klausner, National Institutes of Health, Bethesda, MD, and approved June 26, 2001 (received for review February 15, 2001)
`collective evidence supports a role for mTOR in PI3-kinaseyAkt
`Recent evidence places the FRAPymTOR kinase downstream of the
`phosphatidyl inositol 3-kinaseyAkt-signaling pathway, which is
`function, but the relationship is more complex than that of a
`up-regulated in multiple cancers because of loss of the PTEN tumor
`linear signaling pathway (1).
`suppressor gene. We performed biological and biochemical studies
`Genomic amplification of either PI3-kinase or Akt has been
`to determine whether PTEN-deficient cancer cells are sensitive to
`reported in cervical, ovarian, and pancreatic cancers (23–25). In
`pharmacologic inhibition of FRAPymTOR by using the rapamycin
`addition, mutations in the tumor suppressor phosphatase gene
`PTEN, which regulates signaling through the PI3-kinaseyAkt-
`derivative CCI-779. In vitro and in vivo studies of isogenic PTEN1y1
`and PTEN2y2 mouse cells as well as human cancer cells with defined
`signaling pathway, occur commonly in prostate, glioblastoma,
`PTEN status showed that the growth of PTEN null cells was blocked
`and endometrial tumors (26–30). Because mTOR may function
`preferentially by pharmacologic FRAPymTOR inhibition. Enhanced
`in the PI3-kinaseyAkt pathway, we examined the potential
`tumor growth caused by constitutive activation of Akt in PTEN1y1
`antitumor properties of mTOR inhibitors in PTEN null tumors.
`PTEN2y2 mouse cells and human tumor lines lacking PTEN
`cells also was reversed by CCI-779 treatment,
`indicating that
`FRAPymTOR functions downstream of Akt in tumorigenesis. Loss
`were more sensitive than wild-type PTEN cells to the growth-
`of PTEN correlated with increased S6 kinase activity and phosphor-
`inhibitory effects of CCI-779, an ester of rapamycin. Transfor-
`ylation of ribosomal S6 protein, providing evidence for activation
`mation mediated by expression of activated Akt in cells with an
`of the FRAPymTOR pathway in these cells. Differential sensitivity
`intact PTEN gene also was reversed by CCI-779 treatment.
`to CCI-779 was not explained by differences in biochemical block-
`Biochemical analysis of the mTOR target S6 kinase showed
`ade of the FRAPymTOR pathway, because S6 phosphorylation was
`CCI-779-dependent constitutive activation in PTEN null cells,
`inhibited in sensitive and resistant cell lines. These results provide
`indicating up-regulation of the mTOR pathway. These results
`rationale for testing FRAPymTOR inhibitors in PTEN null human
`suggest that drugs that target mTOR may have clinical utility in
`cancers.
`human cancers lacking PTEN.
`
`Rapamycin is a natural product macrolide that induces G1
`
`growth arrest in yeast, Drosophila, and mammalian cells (1).
`Genetic and biochemical studies have established that the target
`of rapamycin is the protein kinase FRAPymTOR (hereafter
`called mTOR) (1–5), an evolutionarily conserved member of the
`phosphoinositide kinase-related kinase family that includes
`DNA-PK, ATM, and ATR (6). Rapamycin forms a complex with
`the immunophilin prolyl isomerase FKBP12, which binds spe-
`cifically to mTOR and inhibits its ability to phosphorylate
`substrates such as S6 kinase and 4E-BP1. Clinically, rapamycin
`is an approved immunosuppressive agent, based on its ability to
`block T cell activation (7, 8). Because rapamycin also can induce
`growth arrest and apoptosis in certain tumor cells (9), it is under
`investigation as a potential anticancer drug.
`Studies in yeast and mammalian cells suggest that mTOR
`functions as part of a nutrient-sensing mechanism, regulating the
`cellular response to starvation conditions such as amino acid
`deprivation (1, 10). This function is consistent with its biochem-
`ical activity in regulating S6 kinase and 4E-BP1, two mTOR
`targets that play fundamental roles in ribosome biogenesis and
`cap-dependent translation, respectively (11, 12). S6 kinase and
`4E-BP1 are also regulated, in part, through the phosphatidyl
`inositol 3-kinase (PI3-kinase)yAkt-signaling pathway (13, 14).
`That mTOR is phosphorylated by Akt (15) raises the possibility
`of a direct signaling pathway from PI3-kinaseyAkt to mTOR.
`Genetic studies in Drosophila are consistent with this hypothesis,
`as dTOR is downstream and epistatic to the PI3-kinaseyAkt
`pathway (16, 17). However, dTOR loss gives a more severe
`phenotype than PI3-kinase, Akt, or S6 kinase loss (18–21). In
`addition, the Akt phosphorylation site on mTOR is not required
`for S6 kinase activation (15). Finally, only membrane-targeted
`alleles of Akt are sufficient to activate S6 kinase, whereas
`4E-BP1 phosphorylation appears to be Akt-dependent (22). The
`
`Methods
`Protein Analysis. S6 kinase activity was measured by in vitro
`immune complex assay by using an artificial consensus peptide
`as substrate as described (31). Phosphorylated (Ser-235y236)
`and pan-S6 antibodies were provided by Cell Signaling Tech-
`nology (Beverly, MA). Akt and MAPK activation was measured
`by immunoblot assay by using phosphospecific antibodies with
`controls for total Akt and MAPK protein as described (32).
`Pan-4E-BP1 antibody was provided by N. Sonenberg (McGill,
`Montreal, Canada). The level of 4E-BP1 bound to eIF4E was
`measured by precipitation of eIF4E by using 7methyl-GTP
`Sepharose (Amersham Pharmacia), followed by 4E-BP1 immu-
`noblot as described (14). Cyclin D1 and actin antibodies were
`obtained from Santa Cruz Biotechnology. eIF4E antibody was
`obtained from Signal Transduction Laboratories (San Diego).
`
`Tissue Culture Experiments. PTEN1y1 and PTEN2y2 embryonic
`stem (ES) cells and mouse embryo fibroblasts (MEFs) were
`derived as described previously (33). 9L rat glioblastoma cells
`were provided by L. Liau (University of California at Los
`Angeles). All other cell lines were from American Type Culture
`
`This paper was submitted directly (Track II) to the PNAS office.
`Abbreviations: FRAPymTOR, mammalian target of rapamycin; PI3-kinase, phosphatidyl
`inositol 3-kinase; MEF, mouse embryo fibroblasts; ES, embryonic stem; SCID, severe com-
`bined immunodeficient.
`
`See commentary on page 10031.
`
`**To whom reprint requests should be addressed at: University of California Los Angeles,
`Hematology–Oncology, 11–934 Factor Building, 10833 Le Conte Avenue, Los Angeles, CA
`90095-1678. E-mail: csawyers@mednet.ucla.edu.
`
`The publication costs of this article were defrayed in part by page charge payment. This
`article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C.
`§1734 solely to indicate this fact.
`
`10314 –10319 u PNAS u August 28, 2001 u vol. 98 u no. 18
`
`www.pnas.orgycgiydoiy10.1073ypnas.171076798
`
`Breckenridge Exhibit 1093
`Neshat
`Page 001
`
`

`

`MEDICALSCIENCES
`
`PTEN null cells have enhanced sensitivity to mTOR inhibition. Isogenic MEFs derived from PTEN knockout mice were cultured for 72 h in the presence
`Fig. 1.
`of vehicle alone (open bars), 1.0 nM CCI-779 (hatched bars), or 10 nM CCI-779 (solid bars). Cell growth was measured by cell counts determined by Trypan blue
`staining. The data are plotted as the number of cells relative to vehicle control for three experiments. The IC50 for growth inhibition of the cell lines in the table
`was determined by 3H-uptake studies. (*IC50 for 9L was obtained based on lack of growth inhibition by up to 10 nM of CCI-779 as measured by direct cell count.)
`Examples are shown for U87MG and MDA-435. The PTEN status of these lines has been reported previously (26, 35) with the exception of SF-268, SF-295, and
`SF-539, which were kindly provided by R. Parsons (personal communication).
`
`Collection. Cell growth was measured by [3H]thymidine uptake
`andyor cell counts, which were determined by Trypan blue
`staining. CCI-779 was obtained from Wyeth Ayerst Laboratories
`(Marietta, PA).
`
`Mouse Experiments. PTEN1y1 or PTEN2y2 ES cells were in-
`jected s.c. into nude mice. LAPC-4 and LAPC-9 prostate cancer
`xenografts were maintained by serial passage in male severe
`combined immunodeficient (SCID) mice as described and in-
`jected as single cell suspensions (34). Tumor growth was mea-
`sured daily, and mice were randomized to CCI-779 vs. vehicle
`when tumors reached the size indicated in each experiment.
`Treatment was given by i.p. injection for 5 consecutive days.
`Serum prostate-specific antigen levels were measured by ELISA
`as described (34). Hematoxylinyeosin, Ki-67 antibody, and ter-
`minal deoxynucleotidyltransferase-mediated UTP end-labeling
`staining of tumor tissue was performed as described previously
`(34). Cell size (in m2) was measured by using Microcomp
`SCIMEASURE analytical software, calibrated at 3200 magnifica-
`tion. Statistical analysis was performed by using Student’s t test
`and the ANOVA statistical model (Stata Software, College
`Station, TX).
`
`Results
`Enhanced Sensitivity of PTEN Null Cells to mTOR Inhibition. To
`examine the effect of mTOR pathway inhibition on the growth
`of PTEN wild-type vs. PTEN-deficient cells, we exposed mouse
`and human cells with defined PTEN status to varying doses of
`CCI-779 in vitro. CCI-779 is an ester of rapamycin with equiv-
`alent activity and specificity for mTOR.†† First, we examined this
`question in a genetically defined background by comparing the
`growth of PTEN1y1 and PTEN2y2 MEFs that contain a tar-
`geted deletion of the PTEN phosphatase domain (33). The
`growth of PTEN2y2 MEFs was inhibited 40% and 60% by 1.0
`nM and 10 nM CCI-779, respectively, whereas PTEN1y1 MEFs
`
`††Gibbons, J. J., Discafani, C., Peterson, R., Hernandez, R., Skotnicki, J. & Fruman, D. (1999)
`Proc. Am. Assoc. Cancer Res. 40, 301 (abstr.).
`
`were unaffected (Fig. 1 Left). This result cannot be explained by
`an increased growth rate of PTEN null cells because the
`doubling times of PTEN null and PTEN wild-type cells were
`comparable in the absence of CCI-779 (data not shown). We
`expanded the analysis to a panel of 10 tumor cell lines with
`defined PTEN status. The concentration of CCI-779 required
`for 50% growth inhibition (IC50) was less than 10 nM for all of
`the PTEN null tumor cell lines (Fig. 1; see table and [3H]thy-
`midine uptake curve of U87MG vs. MDA-435). It is important
`to note that DU145 cells, which have an intact PTEN gene, are
`relatively sensitive to CCI-779 compared with MDA-435 or
`SF-268, thereby providing an exception to this correlation. One
`explanation might be the elevated Akt3 expression and consti-
`tutive mTOR phosphorylation reported in these cells (15, 36),
`suggesting that either PTEN loss or Akt activation may enhance
`sensitivity to CCI-779. Alternatively, there may be mechanisms
`independent of the PTENyAkt pathway that also affect sensi-
`tivity to CCI-779.
`
`Enhanced S6 Kinase Activation in PTEN Null Tumor Lines. Because
`PTEN-deficient cells have elevated Akt activation, we asked
`whether these cells also have enhanced mTOR activity. We
`explored this question by examining S6 kinase, a downstream
`target of mTOR. Upon activation by mTOR, S6 kinase phos-
`phorylates the 40S ribosomal protein S6, which functions in
`translation of mRNAs with a 59 terminal oligopolypyrimidine
`(TOP) sequence (37, 38). Because S6 mRNA itself contains a 59
`TOP sequence, both translation and phosphorylation of S6
`protein are measures of S6 kinase activity. Under serum star-
`vation conditions, S6 protein level and phosphorylation were
`increased in PTEN null U87MG cells compared with PTEN
`wild-type 9L cells. After serum challenge, S6 protein levels and
`phosphorylation increased in both cell lines and were mTOR-
`dependent, because treatment with CCI-779 reduced S6 levels
`and phosphorylation to undetectable levels (Fig. 2A). These data
`(and data from additional models shown in Figs. 3C and 6)
`provide evidence that loss of PTEN is accompanied by an
`increase in mTOR-dependent S6 kinase activity.
`
`Neshat et al.
`
`PNAS u August 28, 2001 u vol. 98 u no. 18 u 10315
`
`Breckenridge Exhibit 1093
`Neshat
`Page 002
`
`

`

`Enhanced inVivoSensitivity of PTEN-Deficient Tumors to CCI-779. To
`assess the role of mTOR in tumor growth mediated by PTEN
`loss, we examined the effect of CCI-779 on the growth of
`isogenic PTEN1y1 or PTEN2y2 tumors produced by s.c. injec-
`tion of murine ES cells into immunodeficient nude mice. When
`tumors reached 200 mm3 in size, mice were randomly assigned
`to treatment with CCI-779 or vehicle. The growth of PTEN1y1
`ES tumors was slowed during the 5 days of drug administration
`but resumed after the drug was stopped (Fig. 3A). In contrast,
`the growth of PTEN2y2 tumors was completely blocked for the
`duration of the experiment, providing evidence that PTEN-
`deficient tumors derived from a defined genetic background
`have enhanced dependence on mTOR for growth.
`We explored the potential clinical utility of this observation by
`using human tumors grown in immunodeficient SCID mice.
`LAPC-4 (PTEN wild type) and LAPC-9 (PTEN null) (39) are
`androgen-dependent human prostate cancer xenografts that
`grow in male SCID mice with defined kinetics and secrete the
`tumor marker prostate-specific antigen into the serum (34).
`Because phase I clinical studies of CCI-779 suggest that doses in
`the 3-mgykg range are well tolerated in humans and give serum
`levels well above that required to inhibit mTOR,‡‡ we examined
`a range of CCI-779 doses. Male mice were injected s.c. with
`LAPC-4 or LAPC-9 cells and randomly assigned to treatment
`with three different doses of CCI-779 (0.1 mgykg, 4.0 mgykg, or
`40 mgykg) or vehicle when tumors reached ’200 mm3 in size.
`CCI-779 impaired the growth of wild-type PTEN LAPC-4
`tumors in a dose-dependent fashion, with minimal activity at the
`0.1-mgykg dose, partial activity at 4.0 mgykg, and nearly com-
`plete growth suppression at 40 mgykg. In contrast, all three doses
`were equally effective at completely blocking PTEN null
`LAPC-9 tumor growth in three independent experiments (Fig.
`3B). These results from isogenic mouse tumors and human
`tumor lines establish an increase in sensitivity of PTEN null
`tumors to mTOR inhibition.
`We examined the effect of CCI-779 on S6 kinase inhibition by
`measuring S6 levels and phosphorylation in tumors harvested on
`day 5 of treatment with the 0.1-mgykg dose (Fig. 3C). In
`vehicle-treated mice, both the level and phosphorylation state of
`S6 protein were increased in PTEN null LAPC-9 tumors com-
`pared with PTEN wild-type LAPC-4 tumors, consistent with the
`notion that PTEN loss leads to up-regulation of S6 kinase
`activity. In the CCI-779-treated animals, S6 phosphorylation was
`almost completely blocked in LAPC-9 tumors and the level of S6
`protein was reduced, indicating effective inhibition of mTOR. It
`is difficult to make similar conclusions for the LAPC-4 tumors
`because basal S6 phosphorylation is low. The possibility of
`differential inhibition of mTOR in PTEN wild type vs. PTEN
`null cells is addressed further in Fig. 6.
`To determine the effect of CCI-779 treatment on growth and
`apoptosis, we compared proliferation (as determined by anti-
`Ki-67 staining) and apoptosis (as determined by apoptotic bodies
`and terminal deoxynucleotidyltransferase-mediated UTP end-
`labeling staining) in LAPC-4 and LAPC-9 tumors by examining
`histologic sections obtained after 5 days of treatment with
`vehicle or 0.1 mgykg CCI-779. Measurements were based on
`examination of 10 slides per treatment group and 12–18 high-
`power fields per slide. The number of Ki-67-positive nuclei fell
`1.4-fold in LAPC-4 cells vs. 2.6-fold in LAPC-9 cells, and the
`number of apoptotic cells increased 3.4-fold in LAPC-4 cells and
`1.9-fold in LAPC-9 cells (Fig. 4). Statistical analysis indicated
`that the decrease in proliferation was significantly greater in the
`
`‡‡Raymond, E., Alexandre, J., Depenbrock, H., Mekhaldi, S., Angevin, E., Hanauske, A.,
`Baudin, E., Escudier, B., Frisch, J., Boni, J., et al. Am. Soc. Clin. Oncol. 36th Annual Meeting,
`May 19 –23, 2000, New Orleans, LA, 728 (abstr.).
`
`Enhanced S6 kinase activation in PTEN null tumor lines. (A) Phos-
`Fig. 2.
`phorylated (Ser-235y236) and total S6 protein and actin were measured by
`immunoblot in 9L and U87MG cells treated with vehicle or doses of 0.1, 1.0,
`and 10 nM CCI-779 for 7 h. Serum challenge was for 15 min. Equal amounts of
`protein were loaded per lane as determined by Bio-Rad DC protein assay. (B)
`S6 kinase activity was measured by in vitro kinase assay (31) in MDCK cells
`stably transfected with vector or wild-type PTEN (Left) and in LAPC-4 prostate
`cancer cells stably infected with retrovirus expressing myr-Akt or vector con-
`trol (Right). MDCK cells were serum-starved overnight, pretreated with vehi-
`cle (open bars) or 10 nM of CCI-779 (solid bars), and then challenged with
`serum for 15 min. LAPC-4 cells were treated identically except that no serum
`challenge was given. Results from two experiments are plotted relative to
`serum-starved, untreated cells. Expression of PTEN and Akt in the transfec-
`tants was confirmed by immunoblot (data not shown). (C) Levels of total and
`activated Akt and MAPK were measured by immunoblot in lysates of PTEN
`wild-type (DU145) and PTEN null (PC3) cells by using specified antibodies after
`2 h of pretreatment with vehicle or 10 nM CCI-779.
`
`To directly test the role of the PTENyAkt pathway in regu-
`lating S6 kinase, we engineered PTEN or Akt overexpression in
`cells with wild-type endogenous PTEN. PTEN overexpression
`blunted serum-induced S6 kinase activity, as measured by using
`an immunoprecipitation kinase assay (Fig. 2B Left). Conversely,
`expression of a constitutively active, membrane-bound allele of
`Akt (myr-Akt) was sufficient to activate S6 kinase in the absence
`of serum, and this activation was mTOR-dependent (Fig. 2b
`Right). The effects of CCI-779 appear to be specific to the mTOR
`pathway, because no inhibition of Akt or MAPK activation was
`observed (Fig. 2C). Taken together, these data support the
`hypothesis that S6 kinase is activated in PTEN null cells.
`
`10316 u www.pnas.orgycgiydoiy10.1073ypnas.171076798
`
`Neshat et al.
`
`Breckenridge Exhibit 1093
`Neshat
`Page 003
`
`

`

`PTEN null cells have enhanced sensitivity to mTOR inhibition in vivo. (A) PTEN1y1 or PTEN2y2 ES cells were injected s.c. into nude mice at a dose of 5 3
`Fig. 3.
`105 cells per mouse (n 5 20). When tumor volume reached 200 mm3, mice were randomized to treatment with vehicle or 40 mgykg CCI-779. The fold change
`in tumor volume in response to treatment was plotted. (B) Single-cell suspensions of LAPC-4 or LAPC-9 prostate cancer xenografts were injected s.c. into male
`SCID mice (n 5 80) at a dose of 106 cells per mouse. When tumors became palpable, mice were randomized (arrow) to treatment with vehicle, 0.1 mgykg, 4 mgykg,
`or 40 mgykg CCI-779. The fold change in tumor volume from two independent experiments is plotted. (C) Tumors were harvested from mice after 5 days of
`treatment with vehicle or 0.1 mgykg CCI-779 and lysed by boiling in 2% SDS buffer. Immunoblots were performed by using antibodies for phosphorylated S6
`(Ser-235y236), total S6, and actin.
`
`PTEN null LAPC-9 tumor line (P 5 0.03), whereas the increase
`in apoptotic bodies was comparable in both xenografts (P 5 0.24).
`In addition to growth and apoptosis, a third parameter that
`can affect tumor volume is cell size. Genetic analysis has
`established that PI3-kinase, dPTEN, dAkt, and S6 kinase reg-
`ulate cell size in Drosophila (18–21). Similarly, S6 kinase-
`deficient mice have insulin insufficiency because of small islet
`
`cells in the pancreas (40). To determine whether mTOR inhi-
`bition affects cell size in tumors, we performed morphometric
`analysis of 20 tissue sections, in which 1,400 cells were analyzed.
`CCI-779 treatment led to a 1.4-fold reduction in cell size in
`PTEN null LAPC-9 tumors but had no effect in PTEN wild-type
`LAPC-4 tumors (P 5 0.01). These findings indicate that CCI-779
`can affect at least three parameters that influence tumor volume:
`growth, apoptosis, and cell size. Although further work is needed
`to fully account for its antitumor activity, the major difference
`in the effect of CCI-779 on PTEN null and PTEN wild-type cells
`appears to be growth suppression and reduction in cell size.
`
`Akt-Mediated Tumor Growth Is mTOR-Dependent. Because Akt is a
`major effector of transformation caused by PTEN loss, we asked
`whether introduction of constitutively active Akt into wild-type
`PTEN cells would affect CCI-779 sensitivity. To test this hy-
`pothesis, we introduced a constitutively active allele of Akt
`(myr-Akt) in wild-type PTEN LAPC-4 cells, which activated S6
`kinase in an mTOR-dependent fashion (refer to Fig. 2B). Male
`SCID mice were injected with LAPC-4ypuro or LAPC-4ymyr-
`Akt cells, tumors were allowed to reach 200 mm3 in size, and
`mice were randomly assigned to treatment with 0.1 mgykg
`
`MEDICALSCIENCES
`
`Akt-mediated growth in PTEN wild-type tumor cells is mTOR-
`Fig. 5.
`dependent. LAPC-4ypuro or LAPC-4ymyr-Akt cells were injected s.c. into male
`SCID mice (n 5 20) at a dose of 106 cells per mouse. When tumor volume
`reached 50 –200 mm3, mice were randomized to treatment with vehicle or 0.1
`mgykg CCI-779, given by i.p. injection for 5 consecutive days in weeks 1 and 4.
`The fold changes in tumor volume (Left) and in serum prostate-specific
`antigen (PSA) (Right) are plotted.
`
`Effect of CCI-779 on cell size, proliferation, and apoptosis. Photomi-
`Fig. 4.
`crographs (3200) of hematoxylinyeosin- and Ki-67-stained sections of LAPC-4
`and LAPC-9 tumors are shown after 5 days of treatment with vehicle or
`CCI-779. Cell size was measured by morphometry (1,200 total cells on 20
`sections). Proliferation and apoptosis were measured by counting the number
`of Ki-67-positive nuclei or apoptotic bodies [terminal deoxynucleotidyltrans-
`ferase-mediated UTP end labeling (TUNEL)-positive] divided by high-power
`field (hpf) (12–18 hpfytumor, 5 tumorsygroup). All values are mean 6 SEM.
`Both cell size and proliferation were reduced significantly in CCI-779-treated
`LAPC-9 tumors but not LAPC-4 tumors (LAPC-4 size: P 5 0.95; LAPC-9 size: P 5
`0.001; LAPC-4 proliferation: P 5 0.212; LAPC-9 proliferation: P 5 0.01, Stu-
`dent’s t test). Apoptosis was increased significantly in treated LAPC-4 and
`LAPC-9 tumors (P 5 0.01 and P 5 0.005, respectively, Student’s t test). The
`relative effect of CCI-779 treatment on size, proliferation, and apoptosis in
`LAPC-4 vs. LAPC-9 was compared by using an ANOVA model, with P values
`shown above the bar graphs.
`
`Neshat et al.
`
`PNAS u August 28, 2001 u vol. 98 u no. 18 u 10317
`
`Breckenridge Exhibit 1093
`Neshat
`Page 004
`
`

`

`PTEN2y2 cells (Fig. 6 Bottom). Treatment with CCI-779 caused
`a dose-dependent increase in eIF4E-bound 4E-BP1 in PTEN1y1
`cells and a dose-dependent decrease in hyperphosphorylated as
`well as total 4E-BP1 in PTEN2y2 cells, indicating that 4E-BP1
`phosphorylation is, in part, mTOR-dependent in both genetic
`backgrounds. Surprisingly, the decrease in 4E-BP1 phosphory-
`lation in PTEN2y2 cells was not accompanied by a detectable
`increase in eIF4E-bound 4E-BP1. A full explanation of this
`result and the apparent increase in total levels of 4E-BP1 in
`PTEN2y2 cells require further analysis. We also examined cyclin
`D1, a potential downstream parameter of 4E-BP1 phosphory-
`lation whose translation is dependent on eIF4E function (and is
`rapamycin-sensitive) because of complex secondary structure of
`the 59 untranslated region of the mRNA (44, 45). Cyclin D1
`protein levels were elevated in PTEN2y2 compared with
`PTEN1y1 MEFs and were reduced by CCI-779 treatment in
`both cell lines (Fig. 6). Because cyclin D1 is also regulated by
`transcriptional and posttranslationalydegradative mechanisms
`independent of mTOR (46, 47), we cannot make definitive
`conclusions about how each of these variables influences cyclin
`D1 levels in PTEN2y2 cells. Nevertheless, the collective evi-
`dence from analysis of S6, 4E-BP1, and cyclin D1 indicates that
`doses of CCI-779 (10 nM) that cause selective growth inhibition
`in PTEN2y2 cells block mTOR signaling in PTEN1y1 and
`PTEN2y2 cells.
`Discussion
`Our studies in human tumor lines and cells from PTEN
`knockout mice demonstrate that PTEN-deficient cells are
`sensitive to growth inhibition caused by pharmacologic mTOR
`blockade. We also show that a membrane-targeted Akt allele,
`which induces mTOR pathway activation in PTEN wild-type
`cells, promotes tumor growth in a fashion that is reversed by
`mTOR inhibition. Although targeted mTOR gene deletion is
`required to conclusively establish a genetic relationship be-
`tween PTEN, Akt, and mTOR in tumorigenesis, our data
`provide pharmacologic evidence that mTOR is an important
`effector of transformation mediated by perturbation of the
`PTENyAkt pathway. This conclusion is consistent with recent
`work in chicken embryo fibroblasts showing that the mTOR
`inhibitor rapamycin specifically blocks focus formation in-
`duced by oncogenic alleles of PI3-kinase or Akt, but not by Src,
`Ras, Myc, or other oncogenes (48). Because of evidence from
`other studies that PI3-kinaseyAkt- and mTOR-mediated sig-
`nals to S6 kinase and 4E-BP1 can be separated (15, 22), it
`important to stress that the relationship between these signal-
`ing pathways is not strictly linear.
`Why does loss of PTEN sensitize tumors to mTOR inhibition?
`Signaling through the PI3-kinaseyAkt pathway normally is
`tightly regulated, such that PIP3 levels are maintained at low
`concentrations and rise transiently in response to specific growth
`factor signals. In contrast, PIP3 levels are constitutively elevated
`in tumors lacking PTEN, raising the possibility that sustained
`activation of signaling molecules downstream of PIP3 renders
`cells more dependent on this pathway for growth. If this is the
`case, cells with PTEN loss could be more sensitive to the
`biological effects of mTOR inhibition than PTEN wild-type
`cells, even though biochemical inhibition of the mTOR pathway
`occurs efficiently in both cell types. Our data showing efficient
`blockade of S6 phosphorylation in all cell lines tested, regardless
`of their biological sensitivity to CCI-779, provide support for this
`hypothesis. A similar conclusion was drawn in the analysis of
`rapamycin-sensitive and rapamycin-resistant rhabdomyosarco-
`mas (49). In that study, elevated c-myc expression was correlated
`with rapamycin resistance, raising the possibility of parallel
`pathways that can rescue cells from mTOR dependence. In
`preliminary studies, we were unable to demonstrate a similar
`correlation in PTEN1y1 and PTEN2y2 MEFs (M.S.N. and
`
`Fig. 6. Analysis of S6, 4E-BP1, and cyclin D1 in PTEN1y1 and PTEN2y2 MEFs.
`PTEN1y1 or PTEN2y2 MEFs were treated with the indicated concentrations of
`CCI-779 for 6 h, lysed by three freezeythaw cycles in 50 mM Tris, pH 7.5y150
`mM KCl mM EDTAy1 mM EGTAy1 mM DTTy50 mM 2-mercaptoethanol, sup-
`plemented with protease and phosphatase inhibitors, and probed with anti-
`bodies to phosphorylated S6, total S6, total 4E-BP1, or actin. The level of
`4E-BP1 bound to eIF4E was measured by precipitation of eIF4E by using
`7methyl-GTP Sepharose, followed by 4E-BP1 immunoblot. Comparable pre-
`cipitation of eIF4E was confirmed by immunoblot.
`
`CCI-779 or vehicle for 5 days. In the absence of treatment,
`LAPC-4ymyr-Akt tumors grew more quickly than LAPC-4y
`puro tumors,
`indicating that activation of the Akt pathway
`enhances tumor growth in this model. The enhanced growth of
`LAPC-4ymyr-Akt tumors was completely inhibited by CCI-779
`treatment, whereas this dose had no discernible effect on
`LAPC-4ypuro tumors (Fig. 5 Left). Similar results were obtained
`by using serum prostate-specific antigen as the treatment
`endpoint (Fig. 5 Right). These results establish that mTOR
`is required for the enhanced tumor growth caused by Akt
`activation.
`
`Analysis of S6 and 4E-BP1 in PTEN1y1 and PTEN2y2 MEFs. One
`potential explanation for the differential sensitivity of PTEN
`wild-type and PTEN null tumor cells to CCI-779 might be a
`difference in biochemical inhibition of the mTOR pathway. We
`explored this issue by comparing S6 protein levels and phos-
`phorylation in PTEN1y1 and PTEN2y2 MEFs after exposure to
`concentrations of CCI-779 ranging from 0.05 to 10 nM. As
`observed previously in tumor cell lines (Fig. 2 A) and xenografts
`(Fig. 3C), the level and phosphorylation state of S6 were
`markedly elevated in PTEN2y2 cells (Fig. 6 Top). S6 phosphor-
`ylation and protein level were inhibited in a dose-dependent
`fashion in PTEN2y2 cells. In PTEN1y1 MEFs, S6 protein level
`was also reduced, but this effect was observed only at the 10-nM
`dose (S6 phosphorylation was too low in untreated PTEN1y1
`MEFs to measure any changes).
`We examined this issue further by characterizing the status of
`4E-BP1, a protein that blocks translation of 59 cap mRNAs by
`binding the initiation factor eIF4E (41). 4E-BP1 phosphoryla-
`tion, which is modulated by PI3-kinaseyAkt and mTOR as well
`as other pathways, occurs on multiple residues and prevents
`complex formation with eIF4E (22, 42, 43). The level of hyper-
`phosphorylated 4E-BP1 (detected as a slow-mobility isoform in
`SDSyPAGE) was elevated in PTEN2y2 relative to PTEN1y1
`MEFs (Fig. 6 Middle). Conversely, the level of 4E-BP1 (unphos-
`phorylated) bound to eIF4E was increased in PTEN1y1 vs.
`
`10318 u www.pnas.orgycgiydoiy10.1073ypnas.171076798
`
`Neshat et al.
`
`Breckenridge Exhibit 1093
`Neshat
`Page 005
`
`

`

`C.L.S, unpublished observations). Identification of these path-
`ways in PTEN1y1 vs. PTEN2y2 cells requires further investiga-
`tion. However,
`it remains possible that loss of PTEN may
`predispose cells to a greater degree of biochemical inhibition of
`the mTOR pathway at very low doses of CCI-779, as suggested
`by our analysis of S6 protein levels in MEFs. If so, this could have
`important implications with regard to the degree of mTOR
`inhibition in clinical situations in which drug levels vary depend-
`ing on the frequency of dosing.
`Aberrant activation of the PI3-kinaseyAkt pathway occurs in
`a large number of human cancers—primarily through PTEN
`loss, but also through PI3-kinase or Akt gene amplification (23,
`50). Phase I clinical trials of mTOR inhibitors in various cancers
`currently are underway and have shown preliminary evidence of
`
`antitumor activity.‡‡ Our findings suggest that PTEN null can-
`cers are one group of human tumors in which mTOR inhibitors
`may be effective. Because factors other than the PTENyAkt
`pathway also can affect mTOR activation, it will be important
`not to exclude patients with wild-type PTEN tumors from
`clinical trials.
`
`We are grateful to Peter Houghton for sharing data before publication,
`Nahum Sonenberg for providing 4E-BP1 antibody, Fred Dorey for help
`with statistics, Ramon Parsons for providing data on PTEN status of cell
`lines, and Lisa Rose for manuscript preparation. This work was sup-
`ported by grants from the National Institutes of Health, Department of
`Defense, and CaP CURE.
`
`1. Schmelzle, T. & Hall, M. N. (2000) Cell 103, 253–262.
`2. Brown, E. J., Albers, M. W., Shin, T. B., Ichikawa, K., Keith, C. T., Lane, W. S.
`& Schreiber, S. L. (1994) Nature (London) 369, 756–758.
`3. Sabatini, D. M., Erdjument-Bromage, H., Lui, M., Tempst, P. & Snyder, S. H.
`(1994) Cell 78, 35–43.
`4. Chiu, M. I., Katz, H. & Berlin, V. (1994) Proc. Natl. Acad. Sci. USA 91,
`12574–12578.
`5. Sabers, C. J., Martin, M. M., Brunn, G. J., Williams, J. M., Dumont, F. J.,
`Wiederrecht, G. & Abraham, R. T. (1995) J. Biol. Chem. 270, 815–822.
`6. Keith, C. T. & Schreiber, S. L. (1995) Science 270, 50–51.
`7. Schreiber, S. L. (1991) Science 251, 283–287.
`8. Kuo, C. J., Chung, J., Fiorentino, D. F., Flanagan, W. M., Blenis, J. & Crabtree,
`G. R. (1992) Nature (London) 358, 70–73.
`9. Dilling, M. B., Dias, P., Shapiro, D. N., Germain, G. S., Johnson, R. K. &
`Houghton, P. J. (1994) Cancer Res. 54, 903–907.
`10. Hara, K., Yonezawa, K., Weng, Q.-P., Kozlowski, M. T., Belham, C. & Avruch,
`J. (1998) J. Biol. Chem. 273, 14484–14494.
`11. Brown, E. J., Beal, P. A., Keith, C. T., Chen, J., Shin, T. B. & Schreiber, S. L.
`(1995) Nature (London) 377, 441–446.
`12. Brown, E. J. & Schreiber, S. L. (1996) Cell 86, 517–520.
`13. Brunn, G. J., Williams, J., Sabers, C., Wiederrecht, G., Lawrence, J. C., Jr., &
`Abraham, R. T. (1996) EMBO J. 15, 5256–5267.
`14. Gingras, A. C., Kennedy, S. G., O’Leary, M. A., Sonenberg, N. & Hay, N.
`(1998) Genes Dev. 12, 502–513.
`15. Sekulic, A., Hudson, C. C., Homme, J. L., Yin, P., Otterness, D. M., Karnitz,
`L. M. & Abraha

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