throbber
F O C U S
`
`Will mTOR inhibitors make it as cancer drugs?
`Charles L. Sawyers*
`
`Howard Hughes Medical Institute; Departments of Medicine, Molecular and Medical Pharmacology, and Urology and Jonsson Cancer
`Center, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California 90095
`*Correspondence: csawyers@mednet.ucla.edu
`
`Several pharmaceutical and biotechnology companies are
`actively pursuing the clinical development of inhibitors of the
`serine/threonine kinase mammalian target of rapamycin (mTOR)
`for cancer. Rapamycin, the original natural product compound
`first shown to inhibit mTOR, is already an approved drug for pre-
`vention of allograft rejection in recipients of organ transplants
`due to its potent inhibition of T cell activation. What is the logic
`behind the use of the same agent for cancer indications? This
`focus will review the background supporting the potential utility
`of mTOR inhibitors as anticancer agents, then compare and
`contrast two different approaches for its clinical development.
`The first approach is empiric, based on the traditional phase I
`design of escalation to maximum tolerated dose in a broad
`patient population, followed by larger trials focused on those
`tumor types that demonstrate hints of activity in the phase I set-
`ting. The second approach is mechanism based, building on
`knowledge of signaling pathways that activate mTOR, where
`the dose is selected by measuring target enzyme inhibition in
`tumor cells and patient eligibility is defined by molecular profil-
`ing studies. I will speculate on potential outcomes from both
`approaches as well as my view of the eventual role that mTOR
`inhibitors may play in the cancer drug armamentarium.
`
`mTOR: A central regulator of cell growth
`Rapamycin, a bacterially derived natural product, induces G1
`arrest in various cell types at low nanomolar concentrations.
`The mechanism was cleverly deciphered through yeast genetic
`screens that identified a serine/threonine kinase named target
`of rapamycin (TOR) (Heitman et al., 1991), which is a member
`of the larger phosphatidylinositol 3-kinase (PI3K) related family
`that includes PI3K, ATM, and ATR. Rapamycin exerts its action
`by first binding to the immunophilin FK506 binding protein
`(FKBP12). The FKBP12/rapamycin complex then binds mTOR,
`preventing phosphorylation of downstream targets such as S6
`kinase (S6K) and 4EBP1 (see Abraham, 2002; Schmelzle and
`Hall, 2000; Shamji et al., 2003).
`mTOR receives a diverse set of signaling inputs. Among the
`most relevant for a discussion of cancer is mTOR activation by
`growth factors like IGF-1, which activates the PI3K/Akt signaling
`pathway. Akt directly phosphorylates a number of proteins that
`impact cell survival and proliferation (reviewed in Vivanco and
`Sawyers, 2002), but the details defining the connection to
`mTOR were unclear until recently. Now a series of biochemical
`and genetic studies have established a pathway from Akt to
`mTOR involving the tuberous sclerosis complex proteins tuberin
`and hamartin, as well as the small Ras-like GTPase Rheb.
`Tuberous sclerosis complex 2 (TSC2) is a direct substrate of Akt
`(Inoki et al., 2002; Manning et al., 2002; Potter et al., 2002)
`(Figure 1). Unphosphorylated TSC2 is bound to TSC1 in a com-
`plex that blocks mTOR activation. Akt-mediated phosphory-
`lation of TSC2 disrupts the TSC1/TSC2 complex, allowing
`unrestrained mTOR kinase activity. Rheb (Ras homolog
`
`enriched in brain) functions in this pathway downstream of
`TSC2 and upstream of mTOR (Garami et al., 2003; Saucedo et
`al., 2003; Stocker et al., 2003; Zhang et al., 2003). Interestingly,
`Rheb is highly expressed in transformed cancer cell lines and
`functions as an oncogene in fibroblast transformation models,
`but can also block transformation by Ras or B-Raf (Clark et al.,
`1997; Im et al., 2002).
`A simple linear model of this pathway (Akt-TSC1/2-Rheb-
`TOR-S6K) cannot account for all experimental findings. For
`example, PI3K can activate S6K
`independently of Akt
`and mTOR through an alternative pathway involving PDK1
`(Radimerski et al., 2002). In addition, mTOR functions in a nutri-
`ent sensor pathway independent of PI3K and Akt. mTOR is
`inhibited during conditions of nutrient deprivation, which leads
`to a slowdown in cell growth (defined as cell size or mass as
`opposed to cell proliferation). This starvation response makes
`teleological sense because mTOR plays a key regulatory role in
`protein translation through modulation of S6K and 4EBP1
`action. The recently isolated mTOR binding protein Raptor pro-
`vides a potential mechanism for how mTOR regulates down-
`stream effectors (Hara et al., 2002; Kim et al., 2002). Because
`Raptor can also bind S6K and 4EBP1, it may function as a scaf-
`fold, keeping this signaling complex primed for rapid response
`to inputs from various pathways.
`Because S6K and 4EBP1 play crucial roles in regulating
`translation, there has been much interest in defining the down-
`stream mRNA targets of mTOR. The 5′ untranslated regions of
`cyclin D1 and c-Myc mRNAs both have CAP sequences, ren-
`dering them subject to regulation by 4EBP1. Cyclin D1 and c-
`Myc are regulated in part by mTOR since the levels of both
`proteins can fall in cells exposed to rapamycin in certain con-
`texts (Hosoi et al., 1998; Muise-Helmericks et al., 1998; Takuwa
`et al., 1999). Global transcriptome analyses using polysome
`fractions are beginning to define the range of mTOR-regulated
`mRNAs (Peng et al., 2002; Rajasekhar et al., 2003; Shamji et
`al., 2000). At a first approximation, these analyses appear to
`confirm important functional roles of c-Myc and cyclin D1 in
`rapamycin-induced growth arrest (Gera et al., 2003).
`
`Rapamycin has anticancer activity
`The natural products program at the National Cancer Institute
`identified rapamycin as a potential anticancer agent in the
`1970s (Douros and Suffness, 1981). Once the biochemical TOR
`was identified and more detailed activity profiles against a panel
`of human tumor cell lines were completed, some very interest-
`ing patterns emerged. Specifically, cell lines derived from differ-
`ent cancer types were noted to undergo G1 arrest when
`exposed to 1 nM rapamycin, a concentration which closely
`matches that required for biochemical inhibition of mTOR in
`cells. Notably, several other tumor cell lines that failed to
`respond to the 1 nM dose did undergo growth arrest at significantly
`higher concentrations (?1000 nM). While these phenotypic
`
`CANCER CELL : NOVEMBER 2003 • VOL. 4 • COPYRIGHT ' 2003 CELL PRESS
`
`343
`
`West-Ward Pharm.
`Exhibit 1034
`Page 001
`
`

`

`F O C U S
`
`Figure 1. Signaling pathways involving mTOR
`The diagram depicts the current view of mTOR regulation through the
`PI3K/Akt pathway based on biochemical and genetic studies. See text for
`more details.
`
`screening studies revealed the broad potential for rapamycin as
`an antiproliferative agent, they did not uncover the mechanism.
`Nonetheless, the results define two groups of rapamycin-sensi-
`tive cell lines—those whose response correlates with inhibition
`of mTOR (1 nM) (group 1) and those that require significantly
`higher concentrations (group 2). Of note, the failure of the group
`2 cell lines to respond to low “dose” (1 nM) rapamycin cannot be
`explained by insufficient blockade of mTOR kinase activity
`because 1 nM rapamycin was equally effective at inhibiting S6K
`and 4EBP1 phosphorylation in both groups (Neshat et al.,
`2001). These data support the notion that mTOR is the biologi-
`cally relevant target of rapamycin for the tumor lines in group 1,
`whereas other targets are relevant in group 2.
`
`Initial clinical development of mTOR inhibitors—The
`empiric strategy
`In the absence of any histologic subtype or molecular marker
`that can distinguish between group 1, group 2, and nonrespon-
`sive cell lines, the initial clinical development of mTOR inhibitors
`in cancer has proceeded empirically. Several pharmaceutical
`companies have compounds in clinical trials for this indication.
`Among the most advanced is CCI-779 from Wyeth. CCI-779 is
`an ester of rapamycin with comparable potency and specificity
`for mTOR but with a longer half-life. Phase I and phase II clinical
`trials of the intravenous formulation have been completed and
`show promising enough results to warrant a phase III random-
`ized trial that is underway. A brief review of the rationale and
`clinical details underlying this empiric approach is warranted to
`contrast with the molecularly driven approach described sub-
`sequently (see Dancey, 2002 for a comprehensive review of the
`clinical experience with mTOR inhibitors in cancer).
`Following the traditional strategy of defining the maximum
`
`tolerated dose (MTD), Wyeth has conducted two phase I dose
`escalation studies of CCI-779 in patients with solid tumors using
`two different delivery schedules—weekly versus daily for 5 days
`every 2 weeks (Hidalgo et al., 2000; Raymond et al., 2000).
`Toxicities such as low platelet counts and fatigue were observed
`at high doses, but the drug was generally well tolerated.
`Importantly, the doses tested in these cancer trials gave peak
`plasma concentrations well above that required for inhibition of
`mTOR, but the intermittent nature of the dosing allowed troughs
`to fall below mTOR inhibition levels. Clinical responses were
`observed in several patients with advanced stage kidney can-
`cer; therefore, a phase II trial was conducted in this disease.
`Using three different doses of CCI-779 given weekly, the objec-
`tive response rate was only 5%, but there was a higher rate of
`minor responses (29%) and stable disease (40%) (Atkin et al.,
`2002). Of note, responses were observed equally across all
`doses (all of which give peak serum levels that block mTOR).
`On this basis, a phase III randomized trial has been initiated.
`
`Deconvoluting the mechanism of response in kidney
`cancer
`Now that the empiric clinical development plan of CCI-779 has
`identified renal cell carcinoma as a potential mTOR-dependent
`cancer, it is interesting to speculate on possible mechanisms.
`Two scenarios come to mind based on the critical role of angio-
`genesis in these tumors due to expression of hypoxia inducible
`factor (HIF) (reviewed in (Kaelin, 2002). The first is based on
`recent evidence that mTOR inhibitors may be antiangiogenic
`agents. PI3K, Akt, and mTOR are all critical for vascular
`endothelial growth factor (VEGF)-mediated endothelial cell pro-
`liferation, survival, and migration (Yu and Sato, 1999). In fact,
`rapamycin-coated coronary artery stents prevent restenosis in
`patients with coronary artery disease who undergo angioplasty
`(Morice et al., 2002). Preclinical studies suggest that this effect
`of mTOR inhibitors on endothelial growth may also apply to
`tumor angiogenesis since rapamycin blocked the in vivo growth
`of tumor cells that were resistant to rapamycin in vitro (Guba et
`al., 2002). A recent antiangiogenesis trial in kidney cancer (not
`involving an mTOR inhibitor) demonstrated that anti-VEGF anti-
`body was not effective in causing tumors to shrink but caused
`significant delays in the time to tumor progression (Yang et al.,
`2003). This clinical outcome is strikingly reminiscent of the
`phase II results of CCI-779 in kidney cancer and begs the ques-
`tion of whether mTOR inhibitors may work by this mechanism.
`A second scenario to explain the activity of CCI-779 in kid-
`ney cancer is based on evidence that mTOR can regulate HIF
`expression through the PI3K/Akt pathway (Hudson et al., 2002;
`Zhong et al., 2000). Therefore, mTOR inhibitors could have
`direct effects on tumor cells by reducing HIF levels as well as
`indirect effects on the endothelial cells recruited for tumor
`angiogenesis.
`
`Rethinking the empiric clinical development strategy for
`mTOR inhibitors
`The usual justification for pursuing an empiric drug develop-
`ment approach is that the molecular target of the drug is
`unknown or its disease-specific role is poorly understood. As a
`result, drug dose is chosen on the basis of what can be tolerat-
`ed without inordinate toxicity. In this way, it is assumed that any
`clinical activity will not be missed due to insufficient drug levels.
`Once dose-limiting side effects are observed, the schedule of
`drug delivery is typically modified to allow recovery from any
`
`344
`
`CANCER CELL : NOVEMBER 2003
`
`West-Ward Pharm.
`Exhibit 1034
`Page 002
`
`

`

`Table 1. Potential mTOR-dependent cancers
`Molecular lesion
`Clinical disease
`Upstream of mTOR
`PTEN loss
`
`PI3K/Akt activation
`
`TSC1/2 loss
`
`Downstream of mTOR
`cyclin D1 overexpression
`
`Myc overexpression
`
`HIF overexpression
`
`Glioblastoma
`Prostate cancer
`Endometrial cancer
`Othersa
`Breast cancer (Her2+)
`Chronic myeloid leukemia (Bcr-Abl)
`Ovarian cancer
`(PI3K or Akt gene amplification)
`Othersa
`Tuberous sclerosis
`
`Mantle cell lymphoma
`Breast cancer
`Burkitt(cid:146)s lymphoma
`Other Myc-driven cancers?
`Kidney cancer
`Others?
`
`aFor more complete listings, see Vivanco and Sawyers, 2002.
`
`toxicities. Hence, an intermittent regimen using doses just
`below the toxic level is the typical outcome of phase I evalua-
`tions of novel agents. Based on this approach, the early phase
`CCI-779 studies converged on a weekly dosing regimen.
`However, mTOR inhibitors represent an unusual example
`because of the broad clinical experience with rapamycin as an
`immunosuppressive agent for patients receiving organ trans-
`plants. For this indication, the dose is based on that required to
`inhibit T cell activation, which correlates with mTOR inhibition in
`blood cells. These patients take rapamycin daily at relatively low
`doses (in comparison to the weekly “cancer” dose of CCI-779)
`with minimal side effects. A recent phase I clinical report of a
`second mTOR inhibitor, RAD-001 (Novartis), measured S6K in
`blood cells to guide dose selection in cancer patients
`(O’Donnell et al., 2003).
`If mTOR is the relevant target in cancer cells, why wasn’t
`this low dose, daily schedule evaluated initially? And why not
`select patients whose tumors are more likely to be mTOR
`dependent? Obviously, the details underlying the decision-
`making processes at pharmaceutical companies are not public
`knowledge, but I will speculate on a few potential reasons. First,
`a daily dosing schedule would lead to constitutive T cell sup-
`pression, a side effect that one would prefer not to have in can-
`cer patients. My personal view is that immune suppression is of
`secondary concern in the initial stages of developing an anti-
`cancer agent and should be factored into decision making only
`after the primary goal of tumor response has been achieved.
`Second, in vitro studies have defined a second population of
`rapamycin-sensitive tumor cell lines (group 2, see above) where
`the relevant target of drug action is not mTOR (perhaps another
`PIKK family kinase?). In hopes of capturing this second group of
`tumors, one would not want to restrict clinical evaluation to daily,
`low dose mTOR inhibitor because this would only be effective
`against those tumors presumed to be mTOR dependent (group
`1). Third, it would be difficult to convince upper management to
`
`CANCER CELL : NOVEMBER 2003
`
`F O C U S
`
`support a clinical development program focused exclusively on
`mTOR-dependent cancers without any knowledge of the size of
`the target population or the tools to identify the patients. Finally,
`marketing departments would presumably be concerned about
`the confusion generated by parallel use of the same drug for
`immune suppression and cancer. Reformulation into a new
`compound with a different schedule of administration could
`address this issue (albeit, cosmetically). I reiterate that my com-
`ments in this paragraph are highly speculative, but I hope that
`they help illustrate the complex set of scientific, economic, and
`regulatory considerations that influence decisions underlying a
`clinical development path.
`
`An alternative clinical development strategy using
`molecularly selected patients
`Studies of kinase inhibitors such as imatinib (also called
`Gleevec or STI571) indicate that these drugs can have tremen-
`dous clinical activity in appropriately selected patients. This
`experience has led to the notion that certain cancers are kinase
`dependent, typically due to fusions, point mutations, or amplifi-
`cation affecting the kinase gene that is targeted by the inhibitor
`(reviewed in Sawyers, 2003). In general, these genetic events
`enhance enzymatic activity of the kinase and serve as onco-
`genic events driving the growth of the cancer. Since mTOR is a
`kinase, can a similar paradigm of clinical development be
`applied here?
`Strict application of the imatinib paradigm is unlikely since
`there is currently no evidence that the mTOR gene is mutated or
`amplified in human cancer. However, preclinical observations
`suggest that tumors with primary genetic abnormalities affect-
`ing pathways that regulate mTOR are, in fact, dependent on
`mTOR. These abnormalities
`include upregulation of
`the
`PI3K/Akt pathway, directly or by loss of the tumor suppressor
`phosphatase PTEN, as well as mTOR upregulation by TSC2
`loss. PTEN null tumors are sensitive to mTOR inhibitors in sev-
`eral different human and murine preclinical models (Grunwald
`et al., 2002; Neshat et al., 2001; Podsypanina et al., 2001; Shi et
`al., 2002). Transformation induced by oncogenic alleles of Akt,
`but not Myc or Ras, is also reversed by mTOR inhibition (Aoki et
`al., 2001). Recent studies in conditional PTEN knockout or
`transgenic Akt mouse models confirm a role for mTOR in either
`aberrant cell growth (Kwon et al., 2003) or transformation
`through the PI3K/Akt pathway (E. Holland, personal communi-
`cation; W. Sellers, personal communication). Similarly, tumors
`caused by loss of TSC2 also show enhanced sensitivity to
`rapamycin in growth assays (Kenerson et al., 2002).
`The mTOR dependency of these tumors, whether induced
`by loss of PTEN or TSC2 or by activation of PI3K or Akt, shares
`conceptual similarity to synthetic lethal relationships originally
`described in yeast. Loss of PTEN or TSC2 seems to render
`mTOR essential in tumor cells but not in surrounding normal
`cells. However, genetic studies in worms and flies make it clear
`that TOR is essential for normal development (Long et al., 2002;
`Oldham et al., 2000; Zhang et al., 2000). Because TOR is highly
`conserved, it is likely that certain normal mammalian functions,
`including T cell function, will be mTOR dependent. Nonetheless,
`the clinical experience with rapamycin as an
`immuno-
`suppressive agent indicates that mTOR inhibitors are
`well tolerated.
`An alternative group of tumors that might also be mTOR
`dependent are those that express high levels of mTOR-regulat-
`ed mRNAs, such as cyclin D1 or Myc. Because both genes are
`
`345
`
`West-Ward Pharm.
`Exhibit 1034
`Page 003
`
`

`

`F O C U S
`
`also regulated by mTOR-independent mechanisms, it is difficult
`to anticipate how effective an mTOR inhibitor might be in reduc-
`ing the expression of either protein. This, as well as the mTOR
`dependency of PTEN or TSC2-deficient cancers, can be tested
`in clinical trials where patient selection is based on document-
`ing the relevant molecular pathway abnormality in tumor tissue.
`The greatest challenge in designing these clinical trials is
`identification of molecularly defined patient cohorts (Table 1).
`Traditional inclusion criteria using tumor histology and site of
`origin will fail miserably because the molecular phenotype can-
`not be discerned from the clinical phenotype (tuberous sclerosis
`may be an exception). The most appropriate tools for discerning
`these phenotypes in the context of a clinical trial have not been
`defined. Among the potential approaches are proteomic or gene
`expression profiling to recognize signatures of kinase depen-
`dency. A first iteration of this approach, through the use of
`phospho-specific antibodies against specific kinase targets or
`substrates, shows promise in initial immunohistochemical appli-
`cations (Choe et al., 2003). Limitations include the tricky perfor-
`mance characteristics of certain antibodies and the potential
`need for large numbers of optimized antibodies for comprehen-
`sive evaluation. It may also be possible to recognize kinase acti-
`vation through gene expression profiling (Allander et al., 2001;
`Shai et al., 2003). Tissue availability and tumor heterogeneity
`present additional obstacles. Nonetheless, several of these
`approaches are under evaluation in small clinical studies of
`mTOR inhibitors. One possibility is that pilot trials will identify
`biomarkers that can be incorporated more easily into large
`scale studies.
`
`Expectations
`mTOR inhibitors are now far along on the clinical development
`path as anticancer agents, but it remains unclear how the story
`will unfold. The empiric approach has uncovered a low but
`reproducible objective response rate in kidney cancer patients.
`There is the impression of a much larger rate of disease stabi-
`lization, but this must be confirmed by a randomized trial. mTOR
`inhibitors will also be combined empirically with other agents
`(like interferon in kidney cancer) in an effort to increase the
`response rate, but these trials will be conducted without molec-
`ular insight into the mechanism of response. Although we all
`hope for success, this strategy is strikingly similar to recent
`combination trials of EGFR inhibitors with chemotherapy in
`advanced stage lung cancer (reviewed in Dancey and Freidlin,
`2003). Single agent response rates with EGFR inhibitors in
`these patients are low but reproducible; the molecular basis of
`response is unknown; and four large randomized trials of EGFR
`inhibitors plus chemotherapy were all negative.
`The alternative approach of using molecular insights from
`preclinical work to select patients is just now being evaluated.
`Efforts to identify appropriate patients based on immunohisto-
`chemical staining of tumor biopsies have been convincing
`enough to launch exploratory trials, but the robustness of these
`assays in realtime clinical settings remains to be defined.
`If we assume that these assays are accurate and that con-
`tinuous daily dosing of mTOR inhibitors effectively blocks mTOR
`in tumor cells, what clinical outcomes might we expect? There
`are several issues to consider. First, mTOR inhibition in sensi-
`tive tumor cell lines typically causes G1 arrest rather than apop-
`tosis. Therefore, objective response rates may be low but
`disease stabilization could be high. Second, even though we
`have the tools to recognize tumors with loss of PTEN or activa-
`
`346
`
`tion of Akt, we do not know if this molecular abnormality repre-
`sents an early or late event in the history of that tumor. This
`issue could be critical because an inhibitor that blocks an initiat-
`ing oncogenic event is, presumably, more likely to induce a clin-
`ical response than one that blocks a later event involved in
`disease progression.
`No matter the outcome, it is likely that combination therapy
`will be required to fully evaluate the potential of mTOR inhibitors
`in order to maximize response rates and prevent drug resis-
`tance. To avoid mistakes of the past, we need to select combi-
`nations based on mechanistic insights into why certain patients
`respond and others do not. One possibility is an mTOR plus
`EGFR inhibitor combination, particularly in a disease like
`glioblastoma where PTEN loss and genome-based EGFR acti-
`vation can occur in the same tumor (Choe et al., 2003).
`Furthermore, perturbations in one signaling pathway may alter
`the cellular response to inhibition of another, as has been
`observed with PTEN and EGFR in laboratory models (Bianco et
`al., 2003; She et al., 2003). The good news is that we finally
`have a very nice selection of signaling pathway inhibitors, and
`we have the tools to select the patients. We just have to get to
`work and do the right clinical experiments.
`
`Acknowledgments
`
`I thank Ingo Mellinghoff and George Thomas for helpful discussions.Work in my
`lab is supported by the Howard Hughes Medical Institute, Doris Duke
`Charitable Foundation, National Cancer Institute, Leukemia and Lymphoma
`Society, Department of Defense, CaPCURE, and Accelerate Brain Cancer
`Cure.
`
`References
`
`Abraham, R.T. (2002). Identification of TOR signaling complexes: More
`TORC for the cell growth engine. Cell 111, 9–12.
`
`Allander, S.V., Nupponen, N.N., Ringner, M., Hostetter, G., Maher, G.W.,
`Goldberger, N., Chen, Y., Carpten, J., Elkahloun, A.G., and Meltzer, P.S.
`(2001). Gastrointestinal stromal tumors with KIT mutations exhibit a remark-
`ably homogeneous gene expression profile. Cancer Res. 61, 8624–8628.
`
`Aoki, M., Blazek, E., and Vogt, P.K. (2001). A role of the kinase mTOR in cel-
`lular transformation induced by the oncoproteins P3k and Akt. Proc. Natl.
`Acad. Sci. USA 98, 136–141.
`
`Atkin, M.B., Hidalgo, M., Stadler W., Logan, T., Dutcher, J.P., Hudes, G.,
`Park, Y., Marshall, B., Boni, J., and Dukart, G. (2002). A randomized double-
`blind phase 2 study of intravenous CCI-779 administered weekly to patients
`with advanced renal cell carcinoma. Proc. Am. Soc. Clin. Oncol. 21:10a;36A.
`
`Bianco, R., Shin, I., Ritter, C.A., Yakes, F.M., Basso, A., Rosen, N., Tsurutani,
`J., Dennis, P.A., Mills, G.B., and Arteaga, C.L.
`(2003). Loss of
`PTEN/MMAC1/TEP in EGF receptor-expressing tumor cells counteracts the
`antitumor action of EGFR tyrosine kinase inhibitors. Oncogene 22,
`2812–2822.
`
`Choe, G., Horvath, S., Cloughesy, T.F., Crosby, K., Seligson, D., Palotie, A.,
`Inge, L., Smith, B.L., Sawyers, C.L., and Mischel, P.S. (2003). Analysis of the
`phosphatidylinositol 3′-kinase signaling pathway in glioblastoma patients in
`vivo. Cancer Res. 63, 2742–2746.
`
`Clark, G.J., Kinch, M.S., Rogers-Graham, K., Sebti, S.M., Hamilton, A.D.,
`and Der, C.J. (1997). The Ras-related protein Rheb is farnesylated and
`antagonizes Ras signaling and transformation. J. Biol. Chem. 272,
`10608–10615.
`
`Dancey, J.E. (2002). Clinical development of mammalian target of rapamycin
`inhibitors. Hematol. Oncol. Clin. North Am. 16, 1101–1114.
`
`Dancey, J.E., and Freidlin, B. (2003). Targeting epidermal growth factor
`receptor–are we missing the mark? Lancet 362, 62–64.
`
`Douros, J., and Suffness, M. (1981). New antitumor substances of natural
`
`CANCER CELL : NOVEMBER 2003
`
`West-Ward Pharm.
`Exhibit 1034
`Page 004
`
`

`

`F O C U S
`
`origin. Cancer Treat. Rev. 8, 63–87.
`Garami, A., Zwartkruis, F.J., Nobukuni, T., Joaquin, M., Roccio, M., Stocker,
`H., Kozma, S.C., Hafen, E., Bos, J.L., and Thomas, G. (2003). Insulin activa-
`tion of Rheb, a mediator of mTOR/S6K/4E-BP signaling, is inhibited by TSC1
`and 2. Mol. Cell 11, 1457–1466.
`
`Gera, J.F., Mellinghoff, I.K., Shi, Y., Rettig, M.B., Tran, C., Hsu, J.H., Sawyers,
`C.L., and Lichtenstein, A.K. (2003). AKT activity determines sensitivity to
`mTOR
`inhibitors by
`regulating cyclin D1 and c-myc expression.
`J. Biol. Chem.,
`in press. Published online October 23, 2003.
`10.1074/jbc.M309999200.
`
`Grunwald, V., DeGraffenried, L., Russel, D., Friedrichs, W.E., Ray, R.B., and
`Hidalgo, M. (2002). Inhibitors of mTOR reverse doxorubicin resistance con-
`ferred by PTEN status in prostate cancer cells. Cancer Res. 62, 6141–6145.
`
`Guba, M., von Breitenbuch, P., Steinbauer, M., Koehl, G., Flegel, S.,
`Hornung, M., Bruns, C.J., Zuelke, C., Farkas, S., Anthuber, M., et al. (2002).
`Rapamycin inhibits primary and metastatic tumor growth by antiangiogene-
`sis: involvement of vascular endothelial growth factor. Nat. Med. 8, 128–135.
`
`Hara, K., Maruki, Y., Long, X., Yoshino, K., Oshiro, N., Hidayat, S., Tokunaga,
`C., Avruch, J., and Yonezawa, K. (2002). Raptor, a binding partner of target of
`rapamycin (TOR), mediates TOR action. Cell 110, 177–189.
`
`Heitman, J., Movva, N.R., and Hall, M.N. (1991). Targets for cell cycle arrest
`by the immunosuppressant rapamycin in yeast. Science 253, 905–909.
`
`Hidalgo, M., Rowinsky, E., Erlichman, C., Drengler, R., and Marshall, B.
`(2000). CCI-779, a rapamycin analog and multifaceted inhibitor of signal
`transduction: a phase I study. Proc. Am. Soc. Clin. Oncol. 19:10a:726A.
`
`Muise-Helmericks, R.C., Grimes, H.L., Bellacosa, A., Malstrom, S.E.,
`Tsichlis, P.N., and Rosen, N. (1998). Cyclin D expression is controlled post-
`transcriptionally via a phosphatidylinositol 3-kinase/Akt-dependent pathway.
`J. Biol. Chem. 273, 29864–29872.
`
`Neshat, M.S., Mellinghoff, I.K., Tran, C., Stiles, B., Thomas, G., Petersen, R.,
`Frost, P., Gibbons, J.J., Wu, H., and Sawyers, C.L. (2001). Enhanced sensi-
`tivity of PTEN-deficient tumors to inhibition of FRAP/mTOR. Proc. Natl. Acad.
`Sci. USA 98, 10314–10319.
`
`O’Donnell A., Faivre S., Judson I., Delbado C., Brock C., Lane H., Shand N.,
`Hazell K., Armand J.-P., and Raymond E. (2003). A phase I study of the oral
`mTOR inhibitor RAD001 as monotherapy to identify the optimal biologically
`effective dose using toxicity, pharmacokinetic (PK) and pharmacodynamic
`(PD) endpoints in patients with solid tumours. Proc. Am. Soc. Clin. Oncol. 22:
`200 (abstract 803).
`Oldham, S., Montagne, J., Radimerski, T., Thomas, G., and Hafen, E. (2000).
`Genetic and biochemical characterization of dTOR, the Drosophila homolog
`of the target of rapamycin. Genes Dev. 14, 2689–2694.
`
`Peng, T., Golub, T.R., and Sabatini, D.M. (2002). The immunosuppressant
`rapamycin mimics a starvation-like signal distinct from amino acid and glu-
`cose deprivation. Mol. Cell. Biol. 22, 5575–5584.
`
`Podsypanina, K., Lee, R.T., Politis, C., Hennessy, I., Crane, A., Puc, J.,
`Neshat, M., Wang, H., Yang, L., Gibbons, J., et al. (2001). An inhibitor of
`mTOR reduces neoplasia and normalizes p70/S6 kinase activity in Pten+/−
`mice. Proc. Natl. Acad. Sci. USA 98, 10320–10325.
`
`Potter, C.J., Pedraza, L.G., and Xu, T. (2002). Akt regulates growth by direct-
`ly phosphorylating Tsc2. Nat. Cell Biol. 4, 658–665.
`
`Hosoi, H., Dilling, M.B., Liu, L.N., Danks, M.K., Shikata, T., Sekulic, A.,
`Abraham, R.T., Lawrence, J.C., Jr., and Houghton, P.J. (1998). Studies on
`the mechanism of resistance to rapamycin in human cancer cells. Mol.
`Pharmacol. 54, 815–824.
`
`Radimerski, T., Montagne, J., Rintelen, F., Stocker, H., van der Kaay, J.,
`Downes, C.P., Hafen, E., and Thomas, G. (2002). dS6K-regulated cell growth
`is dPKB/dPI(3)K-independent, but requires dPDK1. Nat. Cell Biol. 4,
`251–255.
`
`Hudson, C.C., Liu, M., Chiang, G.G., Otterness, D.M., Loomis, D.C., Kaper,
`F., Giaccia, A.J., and Abraham, R.T. (2002). Regulation of hypoxia-inducible
`factor 1alpha expression and function by the mammalian target of
`rapamycin. Mol. Cell. Biol. 22, 7004–7014.
`
`Rajasekhar, V.K., Viale, A., Socci, N.D., Wiedmann, M., Hu, X., and Holland,
`E.C. (2003). Oncogenic Ras and Akt signaling contribute to glioblastoma for-
`mation by differential recruitment of existing mRNAs to polysomes. Mol. Cell
`12, 889–901.
`
`Im, E., von Lintig, F.C., Chen, J., Zhuang, S., Qui, W., Chowdhury, S., Worley,
`P.F., Boss, G.R., and Pilz, R.B. (2002). Rheb is in a high activation state and
`inhibits B-Raf kinase in mammalian cells. Oncogene 21, 6356–6365.
`
`Inoki, K., Li, Y., Zhu, T., Wu, J., and Guan, K.L. (2002). TSC2 is phosphorylat-
`ed and inhibited by Akt and suppresses mTOR signalling. Nat. Cell Biol. 4,
`648–657.
`
`Kaelin, W.G., Jr. (2002). Molecular basis of the VHL hereditary cancer syn-
`drome. Nat. Rev. Cancer 2, 673–682.
`
`Kenerson, H.L., Aicher, L.D., True, L.D., and Yeung, R.S. (2002). Activated
`mammalian target of rapamycin pathway in the pathogenesis of tuberous
`sclerosis complex renal tumors. Cancer Res. 62, 5645–5650.
`
`Kim, D.H., Sarbassov, D.D., Ali, S.M., King, J.E., Latek, R.R., Erdjument-
`Bromage, H., Tempst, P., and Sabatini, D.M. (2002). mTOR interacts with
`raptor to form a nutrient-sensitive complex that signals to the cell growth
`machinery. Cell 110, 163–175.
`
`Kwon, C.H., Zhu, X., Zhang, J., and Baker, S.J. (2003). mTor is required for
`hypertrophy of Pten-deficient neuronal soma in vivo. Proc. Natl. Acad. Sci.
`USA 100, 12923–12928. Published online October 8, 2003.
`
`Long, X., Spycher, C., Han, Z.S., Rose, A.M., Muller, F., and Avruch, J.
`(2002). TOR deficiency in C. elegans causes developmental arrest and
`intestinal atrophy by inhibition of mRNA translation. Curr. Biol. 12,
`1448–1461.
`
`Manning, B.D., Tee, A.R., Logsdon, M.N., Blenis, J., and Cantley, L.C.
`(2002). Identification of the tuberous sclerosis complex-2 tumor suppressor
`gene product tuberin as a target of the phosphoinositide 3-kinase/akt path-
`way. Mol. Cell 10, 151–162.
`
`Morice, M.C., Serruys, P.W., Sousa, J.E., Fajadet, J., Ban Hayashi, E., Perin,
`M., Colombo, A., Schuler, G., Barragan, P., Guagliumi, G., et al. (2002). A
`randomized comparison of a sirolimus-eluting stent with a standard stent for
`coronary revascularization. N. Engl. J. Med. 346, 1773–1780.
`
`Raymond, E., Alexandre, J., Depenbrock, H., Mekhaldi, S., Angevin, E.,
`Hanauske, A., Baudin, E., Escudier, B., Frisch, J., Boni, J., and Armand, J.P.
`(2000). CCI-779, a rapamycin analog wi

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