throbber
[CANCER RESEARCH 62, 5645–5650, October 15, 2002]
`
`Advances in Brief
`
`Activated Mammalian Target of Rapamycin Pathway in the Pathogenesis of
`Tuberous Sclerosis Complex Renal Tumors1
`
`Heidi L. Kenerson, Lauri D. Aicher, Lawrence D. True, and Raymond S. Yeung2
`Departments of Surgery [H. L. K., L. D. A., R. S. Y.] and Pathology [L. D. T.], University of Washington, Seattle, Washington 98195
`
`Abstract
`
`Disruption of the TSC1 or TSC2 gene leads to the development of
`tumors in multiple organs, most commonly affecting the kidney, brain,
`lung, and heart. Recent genetic and biochemical studies have identified a
`role for the tuberous sclerosis gene products in phosphoinositide 3-kinase
`signaling. On growth factor stimulation, tuberin, the TSC2 protein, is
`phosphorylated by Akt, thereby releasing its inhibitory effects on p70S6K.
`Here we demonstrate that primary tumors from tuberous sclerosis com-
`plex (TSC) patients and the Eker rat model of TSC expressed elevated
`levels of phosphorylated mammalian target of rapamycin (mTOR) and its
`effectors: p70S6K, S6 ribosomal protein, 4E-BP1, and eIF4G. In the Eker
`rat, short-term inhibition of mTOR by rapamycin was associated with a
`significant tumor response, including induction of apoptosis and reduction
`in cell proliferation. Surprisingly, these changes were not accompanied by
`significant alteration in cyclin D1 and p27 levels. Our data provide in vivo
`evidence that the mTOR pathway is aberrantly activated in TSC renal
`pathology and that treatment with rapamycin appears effective in the
`preclinical setting.
`
`Introduction
`
`TSC3 is an autosomal dominant syndrome associated with the
`multiorgan development of benign and occasional malignant tumors
`most commonly affecting the central nervous system, kidney, and skin
`(1). Lesions such as cortical tubers, subependymal giant cell astrocy-
`toma, cardiac rhabdomyomas, and renal AML often exhibit abnormal
`patterns of differentiation along with deregulated cell growth and
`proliferation. The biochemical bases of these pathological alterations
`are not well understood, but genetic studies in Drosophila indicate
`that the two genes implicated in tuberous sclerosis, TSC1 and TSC2,
`participate in the control of cell size via the insulin/p70S6K pathway
`(reviewed in Ref. 2). Epistasis experiments demonstrate that dTsc1
`and dTsc2 act upstream of dS6K and downstream of dAkt. Recent
`biochemical analyses confirmed that tuberin, the TSC2 gene product,
`is a substrate of Akt and can modulate PI3K-dependent activation of
`p70S6K (3, 4). Phosphorylation of tuberin by Akt reduces the stability
`of tuberin and thereby releases its inhibitory function on p70S6K
`signaling. It has also been shown that disease-causing TSC2 mutations
`can produce a reduced state of tuberin phosphorylation that causes it
`to interact less stably with the TSC1 product, hamartin (5). Beyond
`this, TSC1 and TSC2 have been implicated in other molecular path-
`ways, including regulation of low-molecular weight GTPases (Rap1,
`
`Rab5, Rho), p27 stability, and steroid-dependent transcription (re-
`viewed in Ref. 6). As a large molecular complex, TSC1-TSC2 could
`potentially mediate multiple pathways related to cell growth, prolif-
`eration, differentiation, and migration, all of which are relevant to
`TSC biology.
`In this study, we examined the relevance of the PI3K/TSC2/S6K
`pathway in the pathogenesis of TSC renal manifestations. p70S6K is
`a key effector of the PI3K pathway whose activity is regulated by
`sequential phosphorylation by multiple upstream kinases (7). Phos-
`phorylation of the critical residue, Thr229, in the activation loop of
`p70S6K is mediated by PDK1 and is most efficient after prephospho-
`rylation at Thr389 by mTOR and at the COOH-terminal autoinhibitory
`domain by various kinases (7). On activation, p70S6K phosphorylates
`S6 ribosomal protein to regulate translation of 5⬘-TOP mRNA and
`ribosome biogenesis. Binding of mRNA with the 40S ribosomal
`subunit is also under the control of the eIF4F complex, consisting of
`eIF4E, eIF4A, and eIF4G. Stimulation of protein synthesis by amino
`acids releases eIF4E from its inhibitory partner, 4E-BP1, on phospho-
`rylation by mTOR (8). The latter cooperates with the PI3K pathway to
`coordinate cellular responses to growth factors, nutrients, and energy
`sources. Conserved through evolution, TOR has been shown to con-
`trol cell size by regulating protein synthesis mediated through down-
`stream targets, p70S6K and 4E-BP1.
`The importance of the mTOR pathway in human pathology is
`reflected in the overexpression of p70S6K in a subset of breast
`cancers and its correlation with a poor prognosis (9). Moreover, recent
`clinical studies have reported antitumor response to rapamycin and its
`ester derivatives. Rapamycin is a microbial product that binds the
`intracellular receptor FKBP12 to specifically inhibit mTOR activity
`(10). We propose that the loss of tuberin function leads to activation
`of the mTOR pathway in TSC-related renal tumors and that inhibition
`of mTOR signaling brings about reversal of the tumor phenotype. In
`primary renal tumors derived from TSC patients and the Eker rat
`model of TSC, multiple mTOR effectors and mTOR itself were found
`to be highly phosphorylated. Treatment with rapamycin in the Eker rat
`elicited a significant biochemical and histological tumor response in
`keeping with the hypothesis that mTOR is a relevant target for
`therapeutic intervention in TSC patients.
`
`Materials and Methods
`
`Received 8/1/02; accepted 9/3/02.
`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 This work was supported by NIH Grant CA77882 and the Tuberous Sclerosis
`Alliance.
`2 To whom requests for reprints should be addressed, at Department of Surgery,
`University of Washington, 1959 NE Pacific Street, Box 356410, Seattle, WA 98195.
`Phone: (206) 616-6405; Fax: (206) 616-6406; E-mail: ryeung@u.washington.edu.
`3 The abbreviations used are: TSC, tuberous sclerosis complex; AML, angiomyoli-
`poma; PI3K, phosphoinositide 3-kinase; mTOR, mammalian target of rapamycin; IHC,
`immunohistochemistry; PCNA, proliferating cell nuclear antigen; RCC, renal cell carci-
`noma; LOH,
`loss of heterozygosity; TUNEL,
`terminal deoxynucleotidyl
`transferase
`(TdT)-mediated nick end labeling.
`
`Antibodies and Chemicals. The antibody for PHAS-I (4E-BP1) was pur-
`chased from Zymed (San Francisco, CA), Kip1 (p27) was from Transduction
`Laboratories (Los Angeles, CA), cyclin D1 was from Rockland (Gilbertsville,
`PA), tuberin C20 was from Santa Cruz Biotechnology (Santa Cruz, CA), actin
`was from Sigma (St. Louis, MO), and PCNA was from DAKO (Carpinteria,
`CA). Antigelsolin antibody was a gift of David Kwiatkowski (Brigham and
`Women’s Hospital, Boston, MA). All other antibodies were purchased from
`Cell Signaling (Beverly, MA). Rapamycin was purchased from Calbiochem
`(La Jolla, CA). An In-Situ Cell Death Detection Kit (peroxidase) with a
`3,3⬘-diaminobenzidine substrate was obtained from Roche Diagnostics (Indi-
`anapolis, IN). Secondary antibodies and electrochemiluminescence reagents
`were purchased from Amersham Pharmacia Biotech (Piscataway, NJ). The
`5645
`
`
`
`Downloaded from on June 4, 2017. © 2002 American Association for Cancer Research. cancerres.aacrjournals.org
`
`
`
`Breckenridge Exhibit 1108
`Kenerson - Page 001
`
`

`

`RAPAMYCIN INHIBITS TSC TUMORS IN VIVO
`
`Elite ABC kits, 3,3⬘-diaminobenzidine, and Hematoxylin QS were purchased
`from Vector Laboratories (Burlingame, CA). Eosin was obtained from Richard
`Allen Scientific (Kalamazoo, MI).
`Animals. The Eker rat strain harboring a germ-line TSC2 mutation was as
`described previously (11). Fischer male carriers were identified by genotyping
`and housed and fed ad libitum until the age of 12 months, at which point
`rapamycin was injected i.p. once daily for 3 consecutive days. The control
`animal was given the same volume and concentration of DMSO-vehicle, and
`the treated rats were given rapamycin at three dose levels (0.16, 0.4, and 1
`mg/kg). Animals were sacrificed 24 h after the last injection, and tissues were
`procured for IHC and Western blot analysis. All work related to animals was
`in accordance with the protocol approved by the Animal Care Committee,
`University of Washington, Seattle.
`IHC. Kidney samples were fixed in formalin and paraffin embedded.
`Five-␮m sections were deparaffinized, rehydrated, and washed with PBS.
`After antigen retrieval in 10 mM sodium citrate (pH 6.0) and quenching of
`endogenous peroxidase activity with 1% H2O2, samples were blocked with 5%
`normal goat serum before incubation with primary antibodies overnight at 4°C.
`Negative controls were treated with 5% normal goat serum without the primary
`antibodies. Signals were processed according to the supplied protocol (Elite
`ABC Kit). Slides were counterstained with Hematoxylin QS, dehydrated, and
`mounted using Permount (Fischer Scientific, Santa Clara, CA). For the cell
`proliferation index, PCNA⫹ tumor cells were counted from 10 random, non-
`overlapping high-power fields within the tumors, and the results were ex-
`pressed as a percentage of the total number of tumor cells counted in the same
`fields.
`Western Blotting. Tissues were homogenized in ice-cold radioimmuno-
`precipitation assay buffer [1% NP40, 1% sodium deoxycholate, 0.1% SDS,
`0.15 M NaCl, 10 mM Tris (pH 7.2), 0.025 M ␤-glycophosphate (pH 7.2), 2 mM
`EDTA, and 50 mM sodium fluoride] with protease and kinase inhibitors [0.05
`mM 4-(2-aminoethyl)benzenesulfonyl fluoride, 10 ␮g/ml aprotinin, 10 ␮g/ml
`pepstatin, 1 mM orthovanadate, 10 ␮g/ml leupeptin, 1 mM microcystin LR].
`The protein concentration was measured using the BCA Protein Assay (Pierce,
`Rockford, IL). Equal amounts of protein were separated by SDS-PAGE,
`transferred to Immobilon-P membranes (Millipore, Bedford, MA), and blotted
`with antibodies according to the manufacturer’s recommendations, as de-
`scribed previously (5).
`
`ponents of the tumor (Fig. 1B, left). However, vessels with discrete
`walls were found to stain minimally for phospho-p70S6K (Fig. 1B,
`right). The latter structures may represent preexisting or reactive
`vessels. A sporadic AML was also found to aberrantly express phos-
`pho-p70S6K (data not shown). This is consistent with the identifica-
`tion of LOH at the TSC2 locus in sporadic AMLs and suggests a
`common pathogenic mechanism of deregulated p70S6K activity in
`both the sporadic and familial forms of AML (16).
`Within the normal kidney, discrete expression of phospho-p70S6K
`was detected in the distal tubules and some collecting ducts; the
`proximal tubules and glomeruli lacked immunoreactivity (Fig. 1C).
`Approximately 5% of the cells in the normal kidney expressed de-
`tectable levels of phospho-p70S6K. Because the latter has been im-
`plicated in cell size control by regulating 5⬘-TOP translation, it was
`unexpected to find that phospho-p70S6K-positive distal tubular cells
`were consistently smaller than those in adjacent proximal tubules.
`Collectively, IHC analyses of human TSC renal pathology showed
`overexpression of activated mTOR and p70S6K. To further investi-
`gate the functional role of this pathway in tumorigenesis, we exam-
`ined spontaneous renal tumors derived from the Eker rat strain, a
`well-characterized animal model of TSC (11).
`Expression of mTOR Effectors in Primary RCC of the Eker
`Rat. The Eker rat carries a germline mutation of the TSC2 gene as a
`result of a retrotransposition of an endogenous IAP element. As such,
`the Eker TSC2 allele behaves as a null mutation. Spontaneous renal
`cortical epithelial tumors in these animals have been shown to possess
`biallelic inactivation of TSC2 through LOH or nonsense or missense
`mutations (reviewed in 17). In addition to p70S6K, mTOR also
`regulates 4E-BP1 in controlling cap-dependent translation (8). Using
`antibodies for 4E-BP1 and phospho-S6, a substrate for p70S6K, we
`analyzed the expression of these proteins in a panel of five primary
`tumor lysates by Western blotting. Compared with normal kidney, all
`tumors showed marked phosphorylation of ribosomal protein S6 and
`4E-BP1 (Fig. 2A). These tumors also have elevated levels of cyclin
`D1 accompanied by minimal expression of p27 and phospho-Akt. Of
`the five tumors, three (tumors 2, 4, and 5) had lost expression of
`Expression of Phospho-p70S6K in Human TSC Renal Pathol-
`tuberin, and the remaining two were expected to possess aberrant
`ogy. Patients with TSC are predisposed to the development of two
`forms of the protein as a result of missense mutations. Our results
`suggest that effectors downstream of mTOR were activated in the
`forms of renal tumors: RCC and AML (1). To determine the status of
`Eker renal lesions, consistent with the IHC findings in human TSC
`the p70S6K pathway in these tumors, IHC was performed using
`tumors.
`antibodies that specifically recognize phosphorylated p70S6K at
`Thr389, a modification that is necessary for activation of the protein,
`To examine the distribution of these proteins in the Eker rat kidney,
`and phosphorylated mTOR at Ser2448, a site that has been associated
`immunostaining was performed using phospho-specific antibodies in
`paraffin-embedded tissues. Intense, uniform staining for phospho-S6
`with activity (7, 12). Three cases of RCC derived from TSC patients
`was found in tumors of all sizes compared with adjacent renal paren-
`were examined, all demonstrating elevated levels of phospho-mTOR
`chyma (Fig. 2B). Lesions in their earliest form with only a few cells
`and phospho-p70S6K expression in the tumor cells compared with
`were also decorated with phospho-S6 immunoreactivity (Fig. 2F).
`adjacent uninvolved kidney tissue (Fig. 1A). The staining pattern
`This suggests that the mTOR pathway is activated in the primary
`of p70S6K was cytoplasmic, whereas the mTOR signal could be
`stages of renal tumorigenesis in the Eker rat. Other downstream
`seen in the cytoplasm and nucleus, consistent with previous local-
`targets of mTOR, including p70S6K, 4E-BP1, and eIF4G, were also
`ization studies (13, 14). We did not find elevated levels of phos-
`expressed in their phosphorylated forms, although their patterns
`pho-p70S6K in a few sporadic RCCs examined (data not shown);
`showed greater degree of intratumoral heterogeneity (Fig. 2, C–E).
`sporadic clear-cell RCCs are known to involve pathways other than
`Conversely, pathways such as that of mitogen-activated protein kinase
`TSC1 and TSC2 (15).
`did not appear affected, as shown by the absence of phospho-Erk
`The second and more common form of renal pathology in TSC
`expression in the tumors based on IHC (Fig. 2G) and Western blotting
`patients is AML. These tumors contain variable proportions of three
`(data not shown). The staining pattern of phospho-S6 in the nontumor
`histological components: adipocytes, smooth muscle cells, and vas-
`portion of the Eker rat kidney was specific for cells within the distal
`cular structures. In five of six AMLs from TSC patients, we found
`tubules, similar to that found in wild-type kidneys in rats (data not
`robust expression of phospho-p70S6K compared with adjacent tissues
`shown).
`(Fig. 1B). Specifically, staining was uniformly identified in the
`Inhibition of the mTOR Pathway in RCC. Recent studies have
`smooth muscle and lipomatous components, whereas the vascular
`shown that elevated levels of phospho-p70S6K and phospho-S6 in
`structures showed a heterogeneous pattern. Many endothelial lined
`cells lacking TSC1 or TSC2 can be inhibited by rapamycin in vitro
`structures were surrounded by phospho-p70S6K-positive smooth
`(18, 19). Similarly, we have observed that rat embryo fibroblasts in
`muscle-like cells that were contiguous with the myolipomatous com-
`5646
`
`Results
`
`
`
`Downloaded from on June 4, 2017. © 2002 American Association for Cancer Research. cancerres.aacrjournals.org
`
`
`
`Breckenridge Exhibit 1108
`Kenerson - Page 002
`
`

`

`RAPAMYCIN INHIBITS TSC TUMORS IN VIVO
`
`Fig. 1. Expression of mTOR and p70S6K in
`human TSC tumors and normal kidney. A, papillary
`RCC from a TSC patient stained with phospho-
`specific antibodies for p70S6K (Thr389) and mTOR
`(Ser2448). Note negatively stained stromal cells (S)
`and adjacent kidney tissue (N) compared with
`brown-stained tumor cells (magnification, ⫻400
`for left panel, ⫻200 for right panel). B, TSC-
`related AMLs stained with phospho-p70S6K anti-
`body. Left panel shows uniform staining in all three
`cellular components, including the central vessel
`(V; magnification, ⫻200). Right panel shows
`negative-staining vessels (arrows; magnification,
`⫻400). A, adipocyte; SM, smooth muscle. C, nor-
`mal human kidney stained with phospho-p70S6K
`antibody. Arrow indicates prominently stained dis-
`tal tubule (magnification, ⫻200). G, glomerulus.
`
`change in the overall expression of S6 in the renal tumors. To rule out
`the absence of tuberin have constitutive activation of mTOR effectors,
`nonspecific response to rapamycin, serial sections were stained with
`including 4E-BP1, eIF4G, S6K, and S6, as seen in the Eker rat renal
`gelsolin, an actin-binding protein that is expressed in intercalated cells
`tumors; all of these effectors can be down-regulated by rapamycin and
`of the distal tubules and has been shown previously to be a marker for
`LY294002 but not by Wortmannin or PD98059 (data not shown).
`TSC-related pathology (20). The level of gelsolin immunoreactivity
`To investigate the dependence of in vivo tumor growth on the
`did not change significantly after rapamycin treatment (data not
`mTOR pathway, we treated animals with rapamycin and monitored
`shown). In the normal kidney adjacent to the tumors, phospho-S6
`tumor responses. After three daily i.p. doses of rapamycin at three
`immunostaining of the distal tubules was also decreased with rapa-
`dose levels, none of the animals appeared ill or behaved abnormally.
`mycin administration.
`On the fourth day, the Eker rats were sacrificed and the kidney tumors
`To determine tumor response to rapamycin, we examined the
`were analyzed for phosphorylation of the mTOR effectors by use
`histology, cell proliferation, and apoptosis in the renal tumors. Vehicle
`of phospho-specific antibodies. At
`the highest
`rapamycin dose
`control-treated lesions were indistinguishable from those that were
`(1 mg/kg), we did not find tumors large enough for tissue homoge-
`untreated (data not shown). However, rapamycin-treated tumors
`nization. On Western blotting, the level of phospho-S6 in the tumors
`showed significantly more condensed, fragmented, and pyknotic nu-
`treated with vehicle only was highly elevated compared with the
`clei compared with vehicle-treated lesions (Fig. 3B, H&E). TUNEL
`adjacent kidney tissue (Fig. 3A). Treatment with rapamycin dramati-
`staining confirmed the greater extent of apoptosis in the treated
`cally reduced phospho-S6 expression in the tumors even at the lowest
`tumors compared with control (Fig. 3B). With increasing doses of
`dose (0.16 mg/kg). Phosphorylation of 4E-BP1 was partially sup-
`rapamycin, the degree of apoptosis decreased as tumor necrosis in-
`pressed by rapamycin as shown by the increasing intensity of the
`creased. Lymphoid infiltration did not change significantly with ra-
`faster mobility band with higher doses. Surprisingly, the levels of
`pamycin dose, but did correlate in a direct manner with tumor size.
`cyclin D1 and p27 expression, when corrected for protein loading, did
`The percentage of PCNA⫹ nuclei was used as an index of cell
`not change significantly with rapamycin treatment (Fig. 3A). These
`findings correlated well with IHC. Fig. 3B shows specific reduction of
`proliferation (Table 1). Rapamycin-treated tumors were associated
`with a smaller proportion of PCNA⫹ cells. There was a highly
`phospho-S6 reactivity with rapamycin treatment without significant
`5647
`
`
`
`Downloaded from on June 4, 2017. © 2002 American Association for Cancer Research. cancerres.aacrjournals.org
`
`
`
`Breckenridge Exhibit 1108
`Kenerson - Page 003
`
`

`

`RAPAMYCIN INHIBITS TSC TUMORS IN VIVO
`
`Fig. 2. mTOR effectors in the Eker renal tumors. A, Western blot analysis of primary tumor lysates showing expression of the indicated proteins. Actin was used as loading control.
`B–G, immunohistochemical staining of renal tumors with phospho-specific antibodies: B, S6 (Ser235/236; magnification, ⫻100); C, p70S6K (Thr389; magnification, ⫻100); D, 4E-BP1
`(Thr70; magnification, ⫻100); E, eIF4G (Ser1108; magnification, ⫻100); F, S6 (Ser235/236) staining of an early lesion (magnification, ⫻400); G, Erk (Thr202/Tyr204), T, tumor tissue
`(magnification, ⫻200).
`
`significant inverse correlation between the rapamycin dose and the
`percentage of PCNA⫹ cells.
`Overall, rapamycin, even at the lowest dose administered, produced
`profound inhibition of p70S6K activity, which correlated with in-
`creased tumor cell death and necrosis. Importantly, rapamycin did not
`cause any significant histological alteration to the nontumor portions
`of the kidneys. If these observed effects are sustained, rapamycin is
`expected to be an effective therapy for the renal manifestations
`of TSC.
`
`phenotype. Clear-cell and papillary RCCs are known to involve path-
`ways independent of TSC1 or TSC2 (15). Interestingly, sporadic
`AMLs exhibit evidence of increased phospho-S6 expression consist-
`ent with earlier reports of TSC1/2 LOH in these lesions (16). Immuno-
`staining with the phospho-specific antibodies used in this study may
`aid in the classification of sporadic AMLs and RCCs with respect
`to their underlying pathogenesis. In the setting of TSC, abnormal
`“tumor” cells can be recognized by their expression of phosphorylated
`p70S6K, S6, or 4E-BP1. As potential surrogate markers of TSC
`pathology, further studies are needed to address their specificity.
`The kidney tumors in the Eker rat have an immunophenotype
`similar to the human lesions, thus further validating the use of the
`In this study, we showed that tumors associated with TSC gene
`Eker rat as a model of human TSC. Importantly, the fact that rapa-
`mutations are accompanied by activation of the mTOR pathway,
`mycin treatment can down-regulate mTOR effectors and induce tumor
`including p70S6K, 4E-BP1, and eIF4G. In humans, RCC is an infre-
`response in vivo points to the biological dependence of renal lesions
`quent component of TSC, whereas AML is a common manifestation.
`on the effects of the activated mTOR pathway. Of note, adjacent
`Both types of tumors were shown to express phosphorylated mTOR/
`normal kidneys showed minimal cellular toxicity from rapamycin
`p70S6K and their substrates. These alterations appear specific to TSC
`because sporadic RCCs do not share the same immunohistological
`treatment. This is in agreement with the vast clinical experience in
`5648
`
`Discussion
`
`
`
`Downloaded from on June 4, 2017. © 2002 American Association for Cancer Research. cancerres.aacrjournals.org
`
`
`
`Breckenridge Exhibit 1108
`Kenerson - Page 004
`
`

`

`RAPAMYCIN INHIBITS TSC TUMORS IN VIVO
`
`Fig. 3. Effects of rapamycin on Eker renal tumors. A, Western blot analysis of primary tumor lysates showing biochemical responses to i.p. rapamycin. The tumor samples were
`compared with adjacent normal kidney tissue for each dose level. Actin served as loading control. B, representative sections of renal tumors from animals treated with various doses
`of rapamycin stained for total S6, phospho-S6 (Ser235/236), H&E, and apoptosis using the TUNEL kit. N, normal kidney.
`
`alone. A recent study showed that p70S6K signals cell survive by
`using rapamycin to prevent rejection in renal transplant patients. The
`inactivating BAD through phosphorylation of Ser136 (21). Hence,
`“selective” antitumoral effects on the Eker renal tumors seen after a
`short exposure to rapamycin suggest mechanisms in addition to reg-
`acute down-regulation of p70S6K in tumor cells is expected to pro-
`ulation of protein synthesis. The rapid induction of apoptosis/necrosis
`mote apoptosis. Furthermore, rapamycin has been shown to be a
`in the tumors was unexpected on the basis of inhibition of translation
`potent cell cycle inhibitor by down-regulating cyclin D (22). Given
`5649
`
`
`
`Downloaded from on June 4, 2017. © 2002 American Association for Cancer Research. cancerres.aacrjournals.org
`
`
`
`Breckenridge Exhibit 1108
`Kenerson - Page 005
`
`

`

`RAPAMYCIN INHIBITS TSC TUMORS IN VIVO
`
`Table 1 PCNA staining in Eker renal tumors
`
`No. of cells counted
`
`Rapamycin
`
`Positive
`
`Negative
`
`% positivea
`
`53
`238
`268
`Control
`24
`307
`97
`0.16 mg/kg
`13
`581
`88
`0.4 mg/kg
`8
`683
`59
`1.0 mg/kg
`a ␹2 ⫽ 353; P ⬍ 0.0001. There is a highly significant trend between the proportion of
`PCNA⫹ cells and rapamycin dose.
`
`that the half-life of the cyclin D family of proteins is short (⬃2 h), the
`relatively stable levels of cyclin D and p27 in the treated renal tumors
`do not support this mechanism as the cause of reduced cell prolifer-
`ation in this in vivo model. Alternatively, rapamycin may induce
`tumor response through an antiangiogenesis pathway. Both hypoxia-
`induced vascular proliferation and insulin-dependent stimulation of
`HIF-1 have been shown to be dependent on mTOR (23, 24). In
`addition, in murine models, rapamycin inhibited vascular endothelial
`growth factor response, angiogenesis, and tumor growth (25). Collec-
`tively, our findings implicate the mTOR pathway as a biologically
`relevant target in TSC-related tumors. Short-term pharmacological
`manipulation of mTOR activity can bring about significant antitu-
`moral effects by promoting cell death and reducing cell proliferation
`by a cyclin D-independent pathway.
`As a sensor of nutrients, growth factors, and ATP, mTOR serves a
`critical role in regulating the translational machinery and, in doing so,
`affects cellular responses to growth, proliferation, and differentiation,
`all of which are abnormally manifested in TSC lesions. To date, little
`is known about the upstream regulators of mTOR. Protein kinase B,
`also known as Akt, has been shown to phosphorylate mTOR Ser2448
`in vitro, but its biological relevance remains unclear because disrup-
`tion of this site does not affect mTOR signaling and deletion of this
`region enhances its kinase activity (26). However, insulin-induced
`phosphorylation of 4E-BP1 was shown to be Akt-mediated and de-
`pendent on mTOR activity (27). Furthermore, a functional link be-
`tween Ser2448 phosphorylation and muscle hypertrophy/atrophy was
`noted in vivo (12). In this study, sustained phosphorylation of mTOR
`(Ser2448) and its downstream targets in TSC pathology support a role
`of TSC2 in regulating mTOR downstream of Akt. Consistent with this
`model, expression of tuberin was shown in a recent study to suppress
`mTOR activation of p70S6K and to modulate the level of Ser2448
`mTOR expression (28). However, current data do not distinguish
`between a model where tuberin acts upstream of mTOR versus one
`that converges on a common downstream target.
`In summary, activated mTOR signaling in TSC renal pathology
`provides evidence that this pathway, among others, is relevant to their
`pathogenesis. At least some of the classic TSC cellular phenotype
`(e.g., abnormal cell size) can now be explained by this mechanism.
`Encouragingly, induction of an in vivo response to short-term rapa-
`mycin treatment in spontaneous renal tumors of the rat model lend
`support to its use in the clinical setting.
`
`Acknowledgments
`
`We thank Jean Campbell and members of Dr. Yeung’s laboratory for critical
`reading and suggestions. We are grateful to David Ewalt, Lisa Henske, and
`Laura Finn for providing TSC-related pathology. This work is dedicated to Dr.
`Alfred Knudson, Jr. on his 80th birthday.
`
`References
`
`1. Gomez, M. R. Tuberous Sclerosis, 3rd Ed., pp. 10 –23. New York: Oxford University
`Press, 1999.
`2. Montagne, J., Radimerski, T., and Thomas, G. Insulin signaling: lessons from the
`Drosophila tuberous sclerosis complex, a tumor suppressor. Sci. STKE, 105: PE36,
`2001.
`
`3. Manning, B., Tee, A., Logsdon, M., Blenis, J., and Cantley, L. Identification of the
`tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the
`phosphoinositide 3-kinase/Akt pathway. Mol. Cell, 10: 151–162, 2002.
`4. Dan, H., Sun, M., Yang, L., Feldman, R., Sui, X., Yeung, R., Halley, D., Nicosia, S.,
`Pledger, W., and Cheng, J. PI3K/AKT pathway regulates TSC tumor suppressor
`complex by phosphorylation of tuberin. J. Biol. Chem., 277: 35364 –35370, 2002.
`5. Aicher, L. D., Campbell, J. S., and Yeung, R. S. Tuberin phosphorylation regulates its
`interaction with hamartin. Two proteins involved in tuberous sclerosis. J. Biol. Chem.,
`276: 21017–21021, 2001.
`6. Hengstschlager, M., Rodman, D. M., Miloloza, A., Hengstschlager-Ottnad, E.,
`Rosner, M., and Kubista, M. Tuberous sclerosis gene products in proliferation control.
`Mutat. Res., 488: 233–239, 2001.
`7. Templeton, D. J. Protein kinases: getting NEKed for S6K activation. Curr. Biol., 11:
`R596 –R599, 2001.
`8. Rohde, J., Heitman, J., and Cardenas, M. E. The TOR kinases link nutrient sensing to
`cell growth. J. Biol. Chem., 276: 9583–9586, 2001.
`9. Barlund, M., Forozan, F., Kononen, J., Bubendorf, L., Chen, Y., Bittner, M. L.,
`Torhorst, J., Haas, P., Bucher, C., Sauter, G., Kallioniemi, O. P., and Kallioniemi, A.
`Detecting activation of ribosomal protein S6 kinase by complementary DNA and
`tissue microarray analysis. J. Natl. Cancer Inst. (Bethesda), 92: 1252–1259, 2000.
`10. Heitman, J., Movva, N. R., and Hall, M. N. Targets for cell cycle arrest by the
`immunosuppressant rapamycin in yeast. Science (Wash. DC), 253: 905–909, 1991.
`11. Yeung, R. S., Xiao, G. H., Jin, F., Lee, W. C., Testa, J. R., and Knudson, A. G.
`Predisposition to renal carcinoma in the Eker rat is determined by germ-line mutation
`of the tuberous sclerosis 2 (TSC2) gene. Proc. Natl. Acad. Sci. USA, 91: 11413–
`11416, 1994.
`12. Reynolds, T. H., Bodine, S. C., and Lawrence, J. C., Jr. Control of Ser2448 phos-
`phorylation in the mammalian target of rapamycin by insulin and skeletal muscle
`load. J. Biol. Chem., 277: 17657–17662, 2002.
`13. Johanson, S. O., Naccache, P. A., and Crouch, M. F. A p85 subunit-independent
`p110␣ PI 3-kinase colocalizes with p70 S6 kinase on actin stress fibers and regulates
`thrombin-stimulated stress fiber formation in Swiss 3T3 cells. Exp. Cell Res., 248:
`223–233, 1999.
`14. Zhang, X., Shu, L., Hosoi, H., Murti, K. G., and Houghton, P. J. Predominant nuclear
`localization of mammalian target of rapamycin in normal and malignant cells in
`culture. J. Biol. Chem., 277: 28127–28134, 2002.
`15. Kondo, K., and Kaelin, W. G., Jr. The von Hippel-Lindau tumor suppressor gene.
`Exp. Cell Res., 264: 117–125, 2001.
`16. Smolarek, T. A., Wessner, L. L., McCormack, F. X., Mylet, J. C., Menon, A. G., and
`Henske, E. P. Evidence that lymphangiomyomatosis is caused by TSC2 mutations:
`chromosome 16p13 loss of heterozygosity in angiomyolipomas and lymph nodes
`from women with lymphangiomyomatosis. Am. J. Hum. Genet., 62: 810 – 815, 1998.
`17. Kajino, K., and Hino, O. TSC1 and TSC2 gene mutations in human kidney tumors.
`Contrib. Nephrol., 128: 45–50, 1999.
`18. Kwiatkowski, D. J., Zhang, H., Bandura, J. L., Heiberger, K. M., Glogauer, M.,
`el-Hashemite, N., and Onda, H. A mouse model of TSC1 reveals sex-dependent
`lethality from liver hemangiomas, and up-regulation of p70S6 kinase activity in Tsc1
`null cells. Hum. Mol. Genet., 11: 525–534, 2002.
`19. Goncharova, E. A., Goncharov, D. A., Eszterhas, A., Hunter, D. S., Glassberg, M. K.,
`Yeung, R. S., Walker, C. L., Noonan, D., Kwiatkowski, D. J., Chou, M. M.,
`Panettieri, R. A., Jr., and Krymskaya, V. P. Tuberin regulates p70 S6 kinase activation
`and ribosomal protein S6 phosphorylation: a role for the TSC2 tumor suppressor gene
`in pulmonary lymphangioleiomyomatosis (LAM). J. Biol. Chem., 277: 30958 –
`30967, 2002.
`20. Onda, H., Lueck, A., Marks, P. W., Warren, H. B., and Kwiatkowski, D. J.
`Tsc2(⫹/⫺) mice develop tumors in multiple sites that express gelsolin and are
`influenced by genetic background. J. Clin. Investig., 104: 687– 695, 1999.
`21. Harada, H., Andersen, J. S., Mann, M., Terada, N., and Korsmeyer, S. J. p70S6 kinase
`signals cell survival as well as growth, inactivating the pro-apoptotic molecule BAD.
`Proc. Natl. Acad. Sci. USA, 98: 9666 –9670, 2001.
`22. Muise-Helmericks, R. C., Grimes, H. L., Bellacosa, A., Malstrom, S. E., Tsichlis,
`P. N., and Rosen, N. Cyclin D expression is controlled post-transcriptionally via a
`phosphatidylinositol 3-kinase/Akt-dependent pathway. J. Biol. Chem., 273: 29864 –
`29872, 1998.
`23. Humar, R., Kiefer, F. N., Berns, H., Resink, T. J., and Battegay, E. J. Hypoxia
`enhances vascular cell proliferation and angiogenesis in vitro via rapamycin (mTOR)-
`dependent signaling. FASEB J., 16: 771–780, 2002.
`24. Treins, C., Giorgetti-Peraldi, S., Murdaca, J., Semenza, G. L., and Van Obberghen,

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