Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

DTD 5

ARTICLE IN PRESS
TRENDS in Molecular Medicine Vol.xx No.xx Monthxxxx

TRMOME 269

Opinion

An expanding role for mTOR in cancer


David A. Guertin1 and David M. Sabatini1,2
1 2

Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02141, USA Massachusetts Institute of Technology, Department of Biology, Cambridge, MA 02141, USA

Rapamycin, a valuable drug with diverse clinical applications, inhibits mTOR (mammalian target of rapamycin), which is a protein kinase that controls cell growth by regulating many cellular processes, including protein synthesis and autophagy. The sensitivity of select tumor cells to rapamycin has ignited considerable excitement over its potential as an anti-cancer therapeutic. Recent ndings identied a rapamycin-insensitive function of mTOR in regulating a cell-survival pathway that is hyperactive in many cancers, particularly those with elevated PtdIns3K signaling or harboring mutations in the tumor suppressor PTEN (phosphatase and tensin homolog deleted on chromosome 10). These new ndings suggest that targeting this function of mTOR might have broader applications in cancer therapy. In this article, we re-evaluate mTOR signaling, suggesting a more central role for mTOR in cancers with defective PtdIns3KPTEN signaling and conceptually discuss these implications in the context of drug discovery. New horizons for mTOR signaling Advances in our understanding of cancer have made clear that even tumors of the same pathological classication can have markedly different signaling-pathway proles and gene-expression patterns. Effective treatments will probably require the use of molecular markers to decipher these signatures on a patient-to-patient basis and the custom design of appropriate treatment strategies. Biomarkers indicate that the mTOR (mammalian target of rapamycin) growth pathway is hyperactive in certain cancers, suggesting mTOR as an attractive target for cancer therapy [1,2]. The general but unproven belief is that activated mTOR provides certain tumor cells with a growth advantage by promoting protein synthesis, which is the best-described physiological function of mTOR signaling. Recently, a new function for mTOR in regulating the AKT (also known as protein kinase B or PKB) kinase was described [3]. AKT, which is a crucial downstream effector in the phosphatidylinositol 3-kinase (PtdIns3K)PTEN (phosphatase and tensin homolog deleted on chromosome 10) pathway, controls cell proliferation and survival, thus expanding our view of the role of mTOR in signal transduction and the molecular pathology of cancer. Because hyperactive AKT is implicated in the etiology of cancers with defective PtdIns3KPTEN signaling, targeting this function of mTOR might have therapeutic potential.
Corresponding author: Sabatini, D.M. (sabatini@wi.mit.edu).

Here, we discuss evidence suggesting that the inhibition of mTOR by rapamycin is a promising anti-cancer strategy. The prevailing view is that mTOR lies downstream of and is activated by AKT. This model predicts that the aberrant activation of AKT upregulates mTOR growth signaling, sensitizing transformed cells to rapamycin treatment. Although this model is still controversial, adding to the debate is new evidence implicating mTOR as a direct AKT activator, placing mTOR on both sides of the AKT signaling hub. These differential functions of mTOR can be explained by the existence of at least two distinct mTOR complexes containing unique interacting proteins. We discuss each mTOR complex with respect to pathway specicity and conclude with a comment on the value of mTOR inhibitors for the treatment of cancer. mTOR signaling in cancer The best-described genetic deciencies commonly found in human cancer and impinging upon mTOR signaling are mutations in the PTEN gene [4]. PTEN mutations are associated with a spectrum of cancers, including prostate, breast, lung, bladder, melanoma, endometrial, thyroid, brain, renal carcinomas and others, making it one of the most frequently mutated tumor-suppressor genes (Table 1) [57]. PTEN, a lipid phosphatase, counteracts the lipid-kinase activity of class I PtdIns3Ks (Figure 1). Following growth-factor stimulation, activated receptor tyrosine kinases, or intermediaries such as the insulinreceptor substrates IRS-1 and IRS-2, recruit PtdIns3K to the membrane. PtdIns3K phosphorylates phosphatidylinositol (4,5) bisphosphate [PtdIns(4,5)P2] at the membrane to generate PtdIns(3,4,5)P3. PtdIns(3,4,5)P3 serves as a docking site for effector proteins with pleckstrinhomology (PH) domains, including the AKT and 3-phosphoinositide-dependent protein kinase 1 (PDK1) protein kinases. In addition to PTEN deletions, amplications of both PtdIns3K and AKT occur in several cancers [4,8]. Transforming events not affecting the PtdIns3K, PTEN or AKT genes directly, such as the BCRABL translocation and HER-2/neu or epidermal growth-factor receptor (EGFR) amplication, also activate PtdIns3K signaling [8]. Considering the prevalence of PTEN mutations and aberrantly activated PtdIns3KAKT signaling in cancer, identifying the associated downstream signaling events that are crucial for tumor progression is an area of intense investigation. AKT belongs to the cAMP-dependent, cGMP-dependent protein kinase C (AGC) family of protein kinases that includes ribosomal S6 kinases (S6K1 and S6K2), serum- and

www.sciencedirect.com 1471-4914/$ - see front matter Q 2005 Elsevier Ltd. All rights reserved. doi:10.1016/j.molmed.2005.06.007

DTD 5
2

ARTICLE IN PRESS
TRENDS in Molecular Medicine Vol.xx No.xx Monthxxxx

TRMOME 269

Opinion

Table 1. mTOR signaling in disease


Diseasea Tumor-prone syndromes TSC (tuberous sclerosis complex) LAM (lymphangioleiomyomatosis) Cowdens disease Linked genetic mutation and clinical pathologyb TSC1 or TSC2; harmatomas in multiple organs TSC2; abnormal proliferation of smoothmuscle-like cells in the lung PTEN; harmatomatous tumor syndrome Predicted functional link to mTOR signalingc TSC1 and 2 negatively regulate Rheb TSC1 and 2 negatively regulate Rheb Might promote AKT-dependent inhibition of TSC2 and mTOR phosphorylation Might promote AKT-dependent inhibition of TSC2 and mTOR phosphorylation Might promote AKT-dependent inhibition of TSC2 and mTOR phosphorylation STK11 activates AMPK, a positive regulator TSC2 AMPK promotes TSC2 function Refs

[60,61] [100] [62]

Proteus syndrome

PTEN; harmatomatous tumor syndrome

[62]

LhermitteDuclos disease

PTEN; harmatomatous tumor syndrome

[62,101]

PJS (PeutzJeghers syndrome) HCM (familial hypertrophic cardiomyopathy) Cancer Prostate Breast Lung Bladder Melanoma Renal-cell carcinoma Ovarian Endometrial Thyroid Brain (glioblastoma) CML (chronic myeloid leukemia)
a b c

STK11/LKB1; gastrointestinal harmatoma tumor syndrome AMPK; myocardial hypertrophy

[5254] [102]

PTEN PTEN; PtdIns3K, AKT or Her2/neu amplication or hyperactivation PTEN; HER amplication PTEN PTEN PTEN PTEN; PtdIns3K, AKT or Her2/neu amplication or hyperactivation PTEN PTEN; PtdIns3K, AKT or Her2/neu amplication or hyperactivation PTEN BCRABL translocation

PTEN loss promotes AKT activation PTEN loss or gene amplications promote AKT activation PTEN loss or gene amplications promote AKT activation Promotes AKT activation Promotes AKT activation Promotes AKT activation PTEN loss or gene amplications promote AKT activation Promotes AKT activation PTEN loss or gene amplications promote AKT activation Promotes AKT activation Promotes AKT activation

[103,104] [4,104106] [4,107] [108] [4] [4] [4,105,106,109] [4] [4] [4,104,110] [8]

List is not comprehensive, but rather attempts to indicate diseases commonly linked to genes involved in mTOR signaling. Listed only are the linked mutations that might affect mTOR signaling. Best prediction based on existing data.

glucocorticoid protein kinase (SGK) and protein kinase C (PKC) [9]. Two essential phosphorylation sites characterize AGC kinases: one in the activation loop and one in a C-terminal hydrophobic motif. At the membrane, phosphorylation on Thr308 in the activation loop and Ser473 in the hydrophobic motif activates AKT. In mice and humans, three AKT isoforms exist (Akt1 or PKBa, Akt2 or PKBb and Akt3 or PKBg, respectively, for the mouse and human forms), which are the products of distinct genes. Mouse knockout models indicate that, in spite of their structural similarity, all three have distinct roles in growth and metabolism, whereas overexpression studies implicate Akt1 in cell and organ size control [1017]. Because distinct roles for each AKT isoform are only beginning to be understood, we will refer to all three collectively as AKT. Normally, active AKT regulates cell survival, cell proliferation and metabolism (particularly glucose metabolism) by phosphorylating proteins such as BAD, FOXO, NF- kB, p21Cip1 , p27Kip1 , glycogen synthase kinase (GSK)3b and others [4,9,18]. AKT might also promote cell growth by phosphorylating targets such as TSC2 (a negative regulator of mTOR) and mTOR itself, although the physiological signicance of these interactions is still under investigation [19]. AKT activation probably promotes cellular transformation and resistance to apoptosis
www.sciencedirect.com

by collectively promoting growth, proliferation and survival while inhibiting apoptotic pathways. Elevated AKT signaling might additionally cause cells to adopt a more glycolytic metabolism [8]. Such a glycolytic shift could help to sustain cellular metabolism in the harsh nutrientlimiting environment of a tumor and discourage apoptosis by promoting mitochondrial membrane potential [8,20,21]. Thus, inhibitors of AKT signaling are soughtafter drugs and current strategies target both the kinase and PH-domain functions of AKT directly, in addition to its upstream activators [5,22,23]. mTORraptor, a regulator of growth Because hyperactive AKT signaling is associated with elevated mTOR signaling in some cancers, the mTOR inhibitor rapamycin could have promise as an anti-cancer therapeutic [1,2,8,24,25]. mTOR is a highly conserved protein belonging to the PtdIns3K-related kinase family (PIKK family) of serine/threonine protein kinases that includes ataxia-telangiectasia mutated (ATM), ataxiatelangiectasia and Rad3-related (ATR) and DNA-dependent protein kinase (DNA-PKcs). In cells, mTOR exists in at least two distinct complexes: a rapamycin-sensitive complex dened by its interaction with the accessory protein raptor (regulatory-associated protein of mTOR) and a rapamycin-insensitive complex dened by its

DTD 5

ARTICLE IN PRESS
TRENDS in Molecular Medicine Vol.xx No.xx Monthxxxx

TRMOME 269 3

Opinion

Growth factors Insulin IGF-1

Growth

Transcription Autophagy
RTK IR
P

Protein synthesis eIF4E PI3K S6K


P P

IRS
?

4E-BP
Ric tor

GL

PDK1
pt Ra

or

GL

mTOR

PIP3 PDK1
P P

mTOR

Akt

PTEN

Rheb GTP

FKBP12 Rapamycin

TSC1 TSC2
P

Hypoxia or stress AMPK


?

Survival Proliferation
?

Nutrients
(amino acids and glucose) TRENDS in Molecular Medicine

Figure 1. mTOR signaling in cell growth, proliferation and survival. Growth factors, including insulin and IGFs, bind to their receptors [denoted as the insulin receptor (IR) or receptor tyrosine kinase (RTK) for non-insulin-like growth factors]. This triggers PtdIns3K (PI3K) membrane recruitment and activation directly, or through IRS proteins for insulin or insulin-like growth factors. At the cell membrane, PtdIns3K phosphorylates PtdIns(4,5)P2 to generate PtdIns(3,4,5)P3 (PIP3). The tumor suppressor PTEN, a lipid phosphatase, counteracts PtdIns3K lipid-kinase activity. PIP3 serves as a membrane-docking site for the PH-domain-containing proteins AKT and PDK1. Active PDK1 phosphorylates the activation loop of S6K1 and AKT, two AGC family kinases that are key effectors of cell growth and cell-survival pathways, respectively. Growth-factor signaling, along with nutrient availability (particularly amino acids and glucose), regulates cell growth by controlling the activity of the mTORraptor complex. Most available evidence suggests the TSC complex (TSC1 and TSC2) integrates these diverse inputs along with inputs from energy status, stress response and hypoxia pathways to mTOR raptor through a small GTPase called Rheb. Energy status is communicated to TSC2 through direct phosphorylation by AMPK, whereas the mediators of hypoxia, stress and nutrient availability are unknown. It remains possible that nutrients and growth factors, by unknown mechanisms, could also signal to mTOR independently of the TSC complex. mTORraptor controls cellular growth in part by phosphorylating the hydrophobic motif of S6K1 and activating the kinase. mTORraptor also phosphorylates and inhibits 4E-BPs, which themselves inhibit the eIF4E-dependent translation of capped mRNAs. mTORraptor has less dened but important roles in regulating transcription and autophagy. Rapamycin (when bound to FKBP12) specically inhibits the nutrient-sensitive mTORraptor complex. Compelling evidence suggests that a negativefeedback loop enables the nutrient-sensitive mTORraptor pathway, through S6K1, to desensitize insulin signaling. S6K1 mediates the feedback by phosphorylating and inactivating IRS1. Recent ndings indicate that mTOR, when bound to rictor instead of raptor, activates AKT by phosphorylating its hydrophobic motif in a manner sensitive to growth factors. AKT is a crucial regulator of cell proliferation and survival pathways and is hyperactivated in many cancers. mTOR is also phosphorylated on a C-terminal site (not shown), but which mTOR complex is phosphorylated and why is unknown and thus we have excluded that information from the gure. Evidence also suggests that AKT phosphorylates and inhibits TSC2, thus activating the mTORraptor growth pathway. Other studies have questioned the signicance of this interaction, so although it is an attractive model to explain why some tumors with hyperactive AKT signaling are sensitive to rapamycin, debate on its integrity is ongoing. The main pathways are depicted with black arrows, whereas feedback loops are in red. The physiological relevance of these feedbacks for modulating normal mTOR signaling is still under investigation but, regardless, it appears that considerable cross-talk exists. We have indicated selected direct phosphorylation sites with a P, but only for sites discussed in the main text. Broken lines and question marks indicate poorly dened molecular connections.

interaction with rictor (rapamycin-insensitive companion of mTOR) [2630]. Both complexes additionally contain the small WD-repeat protein Gb-like (GbL) and possibly other unidentied proteins [28,31]. Upon entering cells, rapamycin binds to its intracellular receptor, FKBP12, and the complex binds to and specically inhibits mTOR [19,32]. This small lipophilic macrolide antibiotic is already a clinically valuable drug used as an immunosuppressant and in preventing restenosis after balloon angioplasty [25]. The inhibition of mTOR with rapamycin suppresses the enhanced growth phenotype observed in certain mouse cells that are null for
www.sciencedirect.com

PTEN or expressing constitutively activated AKT [33,34]. Similar effects occur in transformed chicken embryo broblasts, multiple myeloma cells and other transformed cells [35,36]. Although the reason why rapamycin is particularly effective against some transformed cells and not others remains unknown, such studies provide the rationale for exploring mTOR inhibition as a treatment for cancers with PTEN inactivation. Why might rapamycin be an effective drug against tumor cells with PTEN loss of function? One hypothesis is that the loss of PTEN activates a rapamycinsensitive mTOR growth pathway, sensitizing such

DTD 5
4

ARTICLE IN PRESS
TRENDS in Molecular Medicine Vol.xx No.xx Monthxxxx

TRMOME 269

Opinion

transformed cells to the drug. Evaluating this hypothesis requires a brief discussion of current thought as to how the mTOR growth pathway is regulated. The rapamycin-sensitive mTORraptor complex controls cellular growth (the accumulation of cellular mass) by regulating protein synthesis and probably also by suppressing autophagy. In doing so, mTORraptor coordinates growth-promoting signals from nutrient and energy availability, particularly from amino acids, and relays them to downstream targets [19,32]. One target is the translational regulator S6K1, which mTORraptor phosphorylates in coordination with the PDK1 protein kinase (Figure 1). Through its interaction with S6K1, raptor directs mTOR to phosphorylate Thr389 in the hydrophobic motif of S6K1 in a rapamycin-sensitive manner, whereas the PDK1 kinase phosphorylates Thr229 in the activation loop [37]. A second S6 kinase (S6K2), which is O80% identical to S6K1, is also phosphorylated in a rapamycinsensitive manner [38,39]. Considerably less is known about S6K2, but genetic models suggest S6K1 and S6K2 have redundant and distinct functions [38,4042]. Although more attention should be given to S6K2, we will not discuss it further because S6K1 appears to be more crucial for growth control. mTORraptor can also phosphorylate and inhibit the eIF4E-binding proteins (4E-BPs), which are a family of negative regulators of eukaryotic translation-initiation factor 4E (eIF4E)-dependent translation [19]. Studies implicate both S6K1 and eIF4E in cellular transformation [4345]. The normal function of S6K1 and 4E-BP1 in protein synthesis has been extensively described elsewhere [19,32,37]. In brief, S6K1 is believed to drive translation in part by phosphorylating the ribosomal protein S6, but the function of this event is still being studied. Thus, conclusive evidence as to the role of S6K1 (and S6K2) in growth remains elusive. Two recent reports further suggest that S6K1 can phosphorylate mTOR on a C-terminal site previously thought to be an AKT-phosphorylation site, but the functional signicance of this event is also unknown [19,46,47]. 4E-BP1 inhibits eIF4E which, in complex with other translational regulators, binds to the 5 0 m7GpppN cap of nucleartranscribed mRNA. This association recruits the capdependent translational activator eIF4G and the 40S ribosomal subunit. Hypophosphorylated 4E-BP1 competitively inhibits the interaction between eIF4G and eIF4E by tightly binding to the same domain. mTORraptor relieves 4E-BP1 inhibition by promoting its phosphorylation and dissociation. Other less-characterized roles for the rapamycin-sensitive mTOR complex in ribosome biogenesis, transcription, cytoskeletal rearrangements and autophagy have also been described [19,32,37]. The TSC connection The tuberous sclerosis complex (TSC) is a heterodimer of two proteins, hamartin (TSC1) and tuberin (TSC2), which is predicted to integrate signals derived from nutrient availability, cellular energy status and hypoxia into a common growth regulatory signal to the mTORraptor complex (Figure 1) [19]. The uncharacterized nutrientderived growth signal feeds to mTORraptor through
www.sciencedirect.com

TSC2, a GAP (GTPase-activating protein)-domain-containing protein that negatively regulates a small GTPase called Rheb [19]. Adding credence to this model are recent biochemical data showing that GTP-bound Rheb can bind to and activate mTOR in an amino-acid-sensitive manner [4850]. By phosphorylating TSC2, hyperactive AKT might promote mTORraptor-dependent cell growth (Figure 1). Evidence indicates that AKT-dependent phosphorylation of TSC2 inactivates the TSC complex, leading to mTOR raptor activation and the hyperphosphorylation of S6K1 and 4E-BP1 [19]. By activating mTORraptor, tumors might gain a growth advantage, suggesting one explanation as to why rapamycin is an effective cytostatic agent against cancer cells with hyperactive AKT signaling. Importantly, the relationship between PTEN inactivation, which presumably hyperactivates AKT, and rapamycin sensitivity is not overly compelling and predicting rapamycin sensitivity based on PTEN status alone is not particularly effective. The downregulation of mTORraptor activity also occurs through the TSC complex. In times of energy crisis, AMP-activated kinase (AMPK) promotes TSC inhibition of mTORraptor by phosphorylating unique sites on TSC2 [51,52]. Because AMPK responds to increasing levels of AMP, it links growth to the cellular ATP:AMP ratio [52]. By phosphorylating AMPK, the LKB1 kinase (also called STK11) contributes to AMPK activation and, accordingly, LKB1-null mouse-embryo broblasts exhibit elevated mTORraptor signaling [5356]. By a mechanism that differs from AMPK, hypoxia promotes the TSC-dependent inhibition of mTOR through the hypoxia-inducible factor (HIF)-1-inducible genes REDD1 (DDIT4) and REDD2 (DDIT4L) [57,58]. HIF-1a is a transcription factor that also promotes angiogenesis in response to low oxygen [59]. Moreover, HIF-1a expression is sensitive to rapamycin [60,61], thus a complex interaction between mTOR and oxygen sensing exists. The clinical value of inhibiting the mTORraptor pathway extends beyond cancer (Table 1). Mutations in the TSC1 or TSC2 genes result in the hyperphosphorylation of mTORraptor effectors and are linked to the harmatomatous syndrome tuberous sclerosis [62,63]. Related hamartoma syndromes such as lymphangioleiomyomatosis (LAM), which is linked to tuberous sclerosis, PeutzJeghers syndrome (PJS), which is caused by mutations in LKB1, and Cowdens disease, Lhermitte Duclos disease and BannayanRileyRuvalcaba syndrome, which are linked to PTEN inactivation, might all be ameliorated by the inhibition of mTORraptor [6264]. Mutations in AMPK are associated with familial cardiac hypertrophy, which also appears to be manifested thorough mTORraptor [62]. Clinical trials with rapamycin are underway in patients with some of these diseases. Feedback inhibition of IRS1 The hamartoma syndromes described above rarely progress to metastatic cancer, raising the obvious question of why not? Perhaps part of the answer lies in the recent conrmation of a negative-feedback loop to the PtdIns3K pathway. Compelling evidence shows that S6K1

DTD 5

ARTICLE IN PRESS
TRENDS in Molecular Medicine Vol.xx No.xx Monthxxxx

TRMOME 269 5

Opinion

phosphorylates and inhibits the insulin-receptor substrate IRS1, an important mediator of insulin-receptordependent activation of PtdIns3K (Figure 1) [6568]. Chronic insulin stimulation leads to the phosphorylation and degradation of IRS1 protein in a rapamycin-sensitive manner. Interestingly, the deletion of S6K1 hypersensitizes mice to insulin, protecting them from age- and dietinduced obesity [41]. These data predict that S6K1 activation promotes growth, but by desensitizing insulinreceptor signaling through IRS1, S6K1 indirectly deactivates PtdIns3KAKT signaling. Whether this negative feedback exclusively targets IRS1 remains to be seen. In fact, AKT phosphorylation in response to platelet-derived growth factor (PDGF) is reduced in some TSC-null cells as a result of reduced expression of the PDGF receptor, which could be analogous to IRS1 downregulation [69]. Biologically, this feedback might be important to control the insulin-dependent inux of nutrients to cells, balancing intake with expenditure. Pathologically, TSCdecient cells, which have elevated mTORraptor activity, fail to activate AKT following insulin stimulation [65]. Amino acids, which also activate mTORraptor activity, can similarly attenuate PtdIns3K signaling following insulin stimulation [70,71]. Thus, activating mTOR raptor could inhibit the AKT-dependent pathways that contribute to cellular transformation, which might explain the benign nature of tuberous sclerosis. Importantly, rapamycin treatment of TSC2-null cells enables the stimulation of AKT phosphorylation in response to insulin and insulin growth factor (IGF)-1; thus, inhibiting the mTORraptor pathway with rapamycin in some tumor cells could promote AKT-dependent survival pathways [66,67]. Unlike a TSC-null deciency, PTEN-null cancers might exhibit cumulative tumorigenic signals that override any potential negative feedback to IRS1; alternatively, PtdIns3K could be activated independently of IRS1. mTORrictor, a missing link to AKT activation To this point, we have focused on targeting the mTOR raptor growth pathway in cancers characterized by hyperactive AKT. However, AKT has key regulatory roles in cell-cycle control, apoptosis and metabolism, in addition to cell growth. In fact, the physiological signicance of AKT-dependent phosphorylation of TSC2 has been challenged in Drosophila in a recent report showing that a nonphosphorylatable version of TSC2 completely rescues the lethality of TSC2-null ies [72]. Other AKT-dependent processes are likely to be crucial for tumor progression. Therefore, inhibitors of AKT or its upstream activators might have broader applications than mTORraptor inhibitors. mTORrictor: the elusive PDK2? AKT activation requires the phosphorylation of Thr308 in the activation loop and Ser473 in the hydrophobic motif [9]. Similar to S6K1, PDK1 phosphorylates AKT in its activation loop. The identity of the Ser473 kinase (often referred to as PDK2) is controversial. In a recent report, it was suggested that the mTORrictor complex fullls the role of PDK2 in vitro and in vivo; importantly, this role is conserved in cultured Drosophila cells [3].
www.sciencedirect.com

Rictor was identied both in yeast (AVO3) and mammals as a novel mTOR-interacting protein dening a second, raptor-independent mTOR complex [2830]. As its name implies, the mTORrictor complex, unlike its mTORraptor sibling, cannot bind to FKBP12rapamycin and is insensitive to acute rapamycin treatment [28,29]. This discovery provided solid evidence that rapamycin treatment does not represent a complete inhibition of mTOR function, which had been speculated. The rst reports on the mTORrictor complex suggest that one of its cellular functions is to regulate the cytoskeleton [29,30]. PKCa could mediate this function, because its phosphorylation state is sensitive to RNA-interference (RNAi)-mediated silencing of mTOR and rictor and, in yeast, AVO3p regulates the actin cytoskeleton through PKC1 [28,29]. PKC, interestingly, is an AGC-family kinase and its mTORrictor-dependent phosphorylation site (Ser657) shares homology with the hydrophobic motif sites of S6K1 and AKT [73]. By using cells particularly sensitive to RNAi (such as cultured Drosophila cells) and lentiviral short-hairpin expression systems in mammalian cells, it has been possible to achieve robust RNAi-mediated silencing of mTOR-pathway components. A clue that mTOR has a role in AKT phosphorylation was the observation that silencing raptor, but not mTOR or rictor, activates AKT on Ser473 (Ser505 in Drosophila), presumably by releasing the negative-feedback inhibition to the insulinIGF pathway described above [3]. This suggested that mTOR has a positive role in regulating AKT phosphorylation that is not shared with raptor but possibly is with rictor. The realization that the Ser473 site in AKT is similar to the hydrophobic motif site in S6K1 and PKC prompted the discovery that in both Drosophila and mammalian cells, the phosphorylation of AKT Ser473 decreases following RNAi-mediated silencing of mTOR and rictor, but not raptor [3]. Biochemical experiments indicate that mTOR rictor, but not mTORraptor, phosphorylates AKT Ser473 in vitro and that full mTORrictor activity requires growth factors, conrming the suspicion that mTOR, when bound to rictor, is a Ser473 kinase [3]. Still unknown is the mechanism by which mTORrictor is activated. Although mTORrictor activity is sensitive to growth factors, the fact that constitutively active PtdIns3K alone triggers AKT Ser473 phosphorylation [4,8] suggests that if mTORrictor is the sole Ser473 kinase, its activation lies downstream of PtdIns3K. No known component of either mTOR complex contains a PH domain or other known lipid-binding domain. Thus, membrane recruitment following lipid phosphorylation is not an obvious possibility. Furthermore, a lack of unambiguous cellular localization data for mTOR and its interacting proteins obscures attempts to identify subcellular compartments that might serve as mTOR signaling centers. Understanding how mTORrictor activity is modulated and what signals promote the interaction between mTORrictor and AKT is paramount to understanding its role in cancer. It would be interesting to determine whether elevated mTOR rictor activity or overexpression is a hallmark of any particular cancer cell line.

DTD 5
6

ARTICLE IN PRESS
TRENDS in Molecular Medicine Vol.xx No.xx Monthxxxx

TRMOME 269

Opinion

Considering the importance of AKT signaling in disease, attempts to identify the crucial Ser473 kinase have been hotly pursued. Many other potential candidates have been identied, including PDK1, integrin-linked kinase (ILK), AKT itself and DNA-PKcs, but in each case either in vivo or in vitro data or conservation could not be conrmed [7477]. Because the mTORrictor complex fullls all three criteria, we suggest that it is a Ser473 kinase. One implication of this proposal, considering current models, is that mTORrictor lies upstream and could indirectly regulate mTORraptor through the AKT TSC2Rheb axis. As previously mentioned, the debate concerning the relevance of this interaction is ongoing. However, in the context of a transformed cell, hyperactive AKT could become promiscuous and phosphorylate offtarget proteins. If this were the case for TSC2, then by hijacking the mTORraptor pathway in transformed cells, the mTORrictor sibling (through AKT) would exert its dominance in the pathogenesis of some cancers. Nevertheless, if the role of mTORrictor in AKT activation withstands future experimental challenges, then mTOR rictor will attract considerable attention as a novel drug target. Determining whether mTORrictor is the crucial Ser473 kinase in tumors characterized by hyperactive AKT will be an essential step to this end. With hindsight, there are clues in the literature about the important role of rictor and mTOR in AKT activation. For example, a loss-of-function allele of pianissimo, the Dictostylium orthologue of rictor [78], causes chemotaxis and cell-aggregation defects that are remarkably similar to those caused by loss of the Dictostylium orthologue of AKT [79]. In addition, the orthologues of mTOR in both Drosophila and Caenorhabditis elegans regulate the lifespan of the organism [8082], a well-characterized role of AKT and its downstream effectors. mTOR was previously not considered an AKT kinase because Ser473 phosphorylation is not sensitive to acute rapamycin treatment. But, because the rapamycinFKBP12 complex tightly binds to mTOR in the absence of associated proteins, it remains possible that chronic exposure to rapamycin might compromise the ability of newly synthesized mTOR protein to associate with rictor. Consistent with this, long-term rapamycin treatment decreases AKT phosphorylation in at least one cell type [83]. This idea provides an alternative hypothesis for why rapamycin is particularly effective at inhibiting the proliferation of certain PTEN-decient cancer cells. Such a scenario would necessitate a review of current rapamycin treatment regimens. Considering the structural similarity between S6Ks and AKTs, particularly within the hydrophobic motif, it makes sense in retrospect that mTOR is an AKT Ser473 kinase. Indirectly, this nding strengthens the argument that mTOR directly phosphorylates S6K1 rather than an intermediate. Challenging this assumption was the fact that deleting a C-terminal domain of S6K1 rendered the phosphorylation of its hydrophobic motif insensitive to rapamycin [8486]. Interestingly, deleting this C-terminal domain from S6K1 makes its hydrophobic motif site accessible to phosphorylation by the rapamycin-insensitive mTORrictor complex [86]. In fact, the S6K1
www.sciencedirect.com

C-terminal truncation mutant closely resembles the structure of AKT which, outside of its PH domain, is similar to S6K1 except that it lacks the C-terminal domain. Although the mechanism of inhibition imposed on mTORrictor by this inhibitory domain is unclear, this observation does explain how the S6K1 C-terminal truncation mutant can be rapamycin insensitive if mTORraptor is the physiological S6K1 hydrophobicmotif kinase. The ability of mTOR to phosphorylate similar motifs in different kinases provides an excellent example of how one kinase, through specic interactions with accessory proteins, can function in diverse cellular processes. It will be interesting to determine if mTOR phosphorylates the hydrophobic motifs of other AGC-family kinases and whether different accessory proteins are required. Prospects for drug discovery Three rapamycin analogs, CCI-779 (Wyeth: http://www. wyeth.com/), RAD001 (Novartis: http://www.novartis.com/) and AP23573 (Ariad Pharmaceuticals Inc: http://www. ariad.com/) are in clinical trials for the treatment of cancer [1,24,25]. Early reports indicate that the drugs show promise in several tumor types, such as renal-cell carcinoma, breast carcinomas and non-small-cell lung carcinomas, and have tolerable side effects, including skin reactions, mucositis and myelosuppression [24,25]. Preliminary evidence suggested that rapamycin was particularly effective against tumors with PTEN inactivation, but an important point from the clinical trials is that biomarkers are needed to predict tumor sensitivity to the drug. One study of breast cancer cell lines indicates that AKT phosphorylation and S6K1 overexpression, rather than PTEN status or S6K1 phosphorylation, are the best predictors of rapamycin sensitivity [87]. Moreover, in many instances, the disease remains progressive following rapamycin treatment, even in PTEN-decient tumors. In fact, one of the most favorable responses to rapamycin was observed in mantle-cell lymphoma, which is characterized by cyclin-D1 overexpression [24]. The development and validation of biomarkers is crucial for determining which patients will benet from rapamycin treatment. Interestingly, rapamycin triggers apoptosis in some cell-culture systems, suggesting a role for the mTOR raptor complex in cell survival [8892]. Under serum-free conditions in rhabdomyosarcoma cells lacking p53, rapamycin has been suggested to trigger apoptosis by activating a stress response involving the apoptosissignal-regulating kinase 1 (ASK1) and c-JUN [93]. However, studies implicating rapamycin-dependent activation of apoptosis in general rely on long-term rapamycin treatment, which might, in some cells, compromise AKT signaling by disrupting the mTORrictor complex, as suggested above. Renal tumors bearing TSC2 mutations are also sensitive to rapamycin-induced apoptosis [90]. Perhaps these tumors, by upregulating mTORraptor signaling in the absence of TSC2 function, activate the negative-feedback loop to IRS1, preventing the full activation of AKT-dependent survival mechanisms and rendering the cells more susceptible to apoptosis. Other

DTD 5

ARTICLE IN PRESS
TRENDS in Molecular Medicine Vol.xx No.xx Monthxxxx

TRMOME 269 7

Opinion

data indicate that in B-cell lymphoma, eIF4E overexpression can promote cell survival in a rapamycin-insensitive manner, probably because it lies downstream of mTOR raptor [94,95]. In summary, mTORraptor signaling contributes to a cell-survival mechanism in some instances, but this is through apparently diverse means that are not necessarily sensitive to rapamycin treatment. Clearly an mTORrictor inhibitor might be benecial for treating tumors with elevated AKT phosphorylation. Theoretically, such an inhibitor might downregulate the growth, proliferation and survival effects that are associated with AKT activation and could be used in combination with other chemotherapeutic agents or rapamycin. If mTORrictor is a crucial activator of AKT-dependent survival processes, such a drug might promote apoptosis in tumor cells that have adapted to AKT-dependent regulatory mechanisms. The essential element will be to nd an mTORrictor-specic inhibitor with a promising therapeutic window and acceptable toxicity. However, the lack of mTORrictor structural information might hinder such efforts. Could a hyperactivated mTORraptor pathway, by TSC2 inhibition or another activator, drive cells to deplete energy stores? And, in combination with a compromised AKT-survival pathway, perhaps with an mTORrictor inhibitor, might apoptosis be potently activated? At present, we can only speculate, but with the newly discovered multifunctional roles for mTOR, open debate on new drug discovery and therapeutic approaches should be stimulated. Re-evaluating mTOR signaling Considering these new ndings, we feel the role of mTOR in growth and disease warrants re-evaluation. Growth is dened as an increase in size or mass. With respect to cellular growth, mTOR has been referred to as a master regulator because nutrient sensing through mTOR is an ancient mechanism for determining individual cell mass. Extending the master regulator role of mTOR to organ and organism growth had been complicated by the fact that most organs additionally rely on a complex balance between cell proliferation and cell death to determine nal organ size. The discovery that mTOR is a regulator of AKT implies a direct role for mTOR in these biological processes. Mouse knockout models of mTOR exhibit early embryonic lethality with cell growth and proliferation defects, which supports this theory [9698]. Moreover, although not a major focus of this article, metabolism and aging, which are tightly linked to AKT-dependent processes, are probably more wired to mTOR signaling than previously thought [18,99]. Many mutations that affect insulinIGF-1 signaling extend lifespan, as do some mutations that decrease TOR activity in C. elegans and Drosophila [18,99]. Perhaps the extended life span of TOR mutants reects in part the role of TOR in AKT phosphorylation. Considering that mTOR regulates cell proliferation and cell-survival pathways, in addition to growth, we suggest a more global role for mTOR in controlling organ and organism growth. Rapamycin treatment for cancers with loss of PTEN function is a potential clinical strategy. However, the therapeutic potential of rapamycin has been considered
www.sciencedirect.com

mainly because the loss of PTEN is associated with hyperactive AKT signaling and existing evidence suggests that AKT regulates the TSCmTORRaptorS6K1 growth pathway. Because mTORrictor was recently shown to directly regulate AKT in a manner that was insensitive to acute rapamycin treatment, mTOR probably has a more central role in cancers with hyperactive PtdIns3KAKT signaling. Designing new drugs that specically inhibit mTORrictor might have great therapeutic value. Concluding remarks During the last few years, our understanding of mTOR signaling in many organisms has greatly advanced thanks to excellent reports on many fronts. Although these discoveries have led to considerable interest and evaluation of mTOR signaling, exemplied by the many reviews written on the subject, we eagerly anticipate learning the many secrets that this molecule still holds. In this article, we have addressed the prevailing views on the role of mTOR in cancer and the hype surrounding rapamycin treatment as a promising anti-cancer strategy. However, new evidence suggests that mTOR might have a more central rapamycin-insensitive role in the pathology of some cancers. This discovery begins a new chapter in the mTOR odyssey, importantly opening a new door in the search for promising anti-cancer drug targets.
Acknowledgements
We apologize to those authors whose primary work we did not reference directly in the text. We thank Deanna Stevens and Anne Carpenter for comments on the manuscript. This work was supported by a grant from the NIH (RO1 A147389) to D.M.S. D.A.G. is supported by a grant from the Damon Runyon Cancer Research Foundation.

References
1 Bjornsti, M.A. and Houghton, P.J. (2004) The TOR pathway: a target for cancer therapy. Nat. Rev. Cancer 4, 335348 2 Chan, S. (2004) Targeting the mammalian target of rapamycin (mTOR): a new approach to treating cancer. Br. J. Cancer 91, 14201424 3 Sarbassov, D.D. et al. (2005) Phosphorylation and regulation of Akt/PKB by the rictormTOR complex. Science 307, 10981101 4 Vivanco, I. and Sawyers, C.L. (2002) The phosphatidylinositol 3-kinase AKT pathway in human cancer. Nat. Rev. Cancer 2, 489501 5 Wymann, M.P. et al. (2003) Phosphoinositide 3-kinase signalling which way to target? Trends Pharmacol. Sci. 24, 366376 6 Stiles, B. et al. (2004) PTENless means more. Dev. Biol. 273, 175184 7 Sansal, I. and Sellers, W.R. (2004) The biology and clinical relevance of the PTEN tumor suppressor pathway. J. Clin. Oncol. 22, 29542963 8 Thompson, J.E. and Thompson, C.B. (2004) Putting the rap on Akt. J. Clin. Oncol. 22, 42174226 9 Brazil, D.P. et al. (2004) Advances in protein kinase B signalling: AKTion on multiple fronts. Trends Biochem. Sci. 29, 233242 10 Easton, R.M. et al. (2005) Role for Akt3/protein kinase Bg in attainment of normal brain size. Mol. Cell. Biol. 25, 18691878 11 Peng, X.D. et al. (2003) Dwarsm, impaired skin development, skeletal muscle atrophy, delayed bone development, and impeded adipogenesis in mice lacking Akt1 and Akt2. Genes Dev. 17, 13521365 12 Garofalo, R.S. et al. (2003) Severe diabetes, age-dependent loss of adipose tissue, and mild growth deciency in mice lacking Akt2/PKB b. J. Clin. Invest. 112, 197208 13 Shioi, T. et al. (2002) Akt/protein kinase B promotes organ growth in transgenic mice. Mol. Cell. Biol. 22, 27992809

DTD 5
8

ARTICLE IN PRESS
TRENDS in Molecular Medicine Vol.xx No.xx Monthxxxx

TRMOME 269

Opinion

14 Tuttle, R.L. et al. (2001) Regulation of pancreatic b-cell growth and survival by the serine/threonine protein kinase Akt1/PKBa. Nat. Med. 7, 11331137 15 Chen, W.S. et al. (2001) Growth retardation and increased apoptosis in mice with homozygous disruption of the Akt1 gene. Genes Dev. 15, 22032208 16 Cho, H. et al. (2001) Akt1/PKBa is required for normal growth but dispensable for maintenance of glucose homeostasis in mice. J. Biol. Chem. 276, 3834938352 17 Cho, H. et al. (2001) Insulin resistance and a diabetes mellitus-like syndrome in mice lacking the protein kinase Akt2 (PKB b). Science 292, 17281731 18 Whiteman, E.L. et al. (2002) Role of Akt/protein kinase B in metabolism. Trends Endocrinol. Metab. 13, 444451 19 Hay, N. and Sonenberg, N. (2004) Upstream and downstream of mTOR. Genes Dev. 18, 19261945 20 Rathmell, J.C. et al. (2003) Akt-directed glucose metabolism can prevent Bax conformation change and promote growth factorindependent survival. Mol. Cell. Biol. 23, 73157328 21 Plas, D.R. et al. (2001) Akt and Bcl-xL promote growth factorindependent survival through distinct effects on mitochondrial physiology. J. Biol. Chem. 276, 1204112048 22 DeFeo-Jones, D. et al. (2005) Tumor cell sensitization to apoptotic stimuli by selective inhibition of specic Akt/PKB family members. Mol. Cancer Ther. 4, 271279 23 Dancey, J.E. (2004) Molecular targeting: PI3 kinase pathway. Ann. Oncol. 15 (Suppl. 4), iv233iv239 24 Rowinsky, E.K. (2004) Targeting the molecular target of rapamycin (mTOR). Curr. Opin. Oncol. 16, 564575 25 Vignot, S. et al. (2005) mTOR-targeted therapy of cancer with rapamycin derivatives. Ann. Oncol. 16, 525537 26 Kim, D-H. et al. (2002) mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell 110, 163175 27 Hara, K. et al. (2002) Raptor, a binding partner of target of rapamycin (TOR), mediates TOR action. Cell 110, 177189 28 Loewith, R. et al. (2002) Two TOR complexes, only one of which is rapamycin sensitive, have distinct roles in cell growth control. Mol. Cell 10, 457468 29 Sarbassov, D.D. et al. (2004) Rictor, a novel binding partner of mTOR, denes a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr. Biol. 14, 12961302 30 Jacinto, E. et al. (2004) Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin insensitive. Nat. Cell Biol. 6, 11221128 31 Kim, D.H. et al. (2003) GbL, a positive regulator of the rapamycinsensitive pathway required for the nutrient-sensitive interaction between raptor and mTOR. Mol. Cell 11, 895904 32 Guertin, D.A. et al. (2004) Growth control through the mTOR network. In Cell Growth: Control of Cell Size (Hall, M.N. et al., eds), pp. 193234, Cold Spring Harbor Laboratory Press 33 Neshat, M.S. et al. (2001) Enhanced sensitivity of PTEN-decient tumors to inhibition of FRAP/mTOR. Proc. Natl. Acad. Sci. U. S. A. 98, 1031410319 34 Podsypanina, K. et al. (2001) An inhibitor of mTOR reduces neoplasia and normalizes p70/S6 kinase activity in PtenC/K mice. Proc. Natl. Acad. Sci. U. S. A. 98, 1032010325 35 Aoki, M. et al. (2001) A role of the kinase mTOR in cellular transformation induced by the oncoproteins P3k and Akt. Proc. Natl. Acad. Sci. U. S. A. 98, 136141 36 Shi, Y. et al. (2002) Enhanced sensitivity of multiple myeloma cells containing PTEN mutations to CCI-779. Cancer Res. 62, 50275034 37 Fingar, D.C. and Blenis, J. (2004) Target of rapamycin (TOR): an integrator of nutrient and growth factor signals and coordinator of cell growth and cell cycle progression. Oncogene 23, 31513171 38 Shima, H. et al. (1998) Disruption of the p70s6k/p85s6k gene reveals a small mouse phenotype and a new functional S6 kinase. EMBO J. 17, 66496659 39 Park, I.H. et al. (2002) Regulation of ribosomal S6 kinase 2 by mammalian target of rapamycin. J. Biol. Chem. 277, 3142331429 40 Pende, M. et al. (2000) Hypoinsulinaemia, glucose intolerance and diminished b-cell size in S6K1-decient mice. Nature 408, 994997
www.sciencedirect.com

41 Um, S.H. et al. (2004) Absence of S6K1 protects against age- and dietinduced obesity while enhancing insulin sensitivity. Nature 431, 200205 42 Pende, M. et al. (2004) S6K1K/K/S6K2K/K mice exhibit perinatal lethality and rapamycin-sensitive 5 0 -terminal oligopyrimidine mRNA translation and reveal a mitogen-activated protein kinasedependent S6 kinase pathway. Mol. Cell. Biol. 24, 31123124 43 Mamane, Y. et al. (2004) eIF4E from translation to transformation. Oncogene 23, 31723179 44 Surace, E.I. et al. (2004) Functional signicance of S6K overexpression in meningioma progression. Ann. Neurol. 56, 295298 45 Barlund, M. et al. (2000) Detecting activation of ribosomal protein S6 kinase by complementary DNA and tissue microarray analysis. J. Natl. Cancer Inst. 92, 12521259 46 Holz, M.K. and Blenis, J. (2005) Identication of S6K1 as a novel mTOR-phosphorylating kinase. J. Biol. Chem. doi: 10.1074/ jbc.M504045200 (http://www.jbc.org/) 47 Chiang, G.G. and Abraham, R.T. (2005) Phosphorylation of mTOR at Ser-2448 is mediated by p70S6 kinase. J. Biol. Chem. doi: 10.1074/jbc.M501707200 (http://www.jbc.org/) 48 Smith, E.M. et al. (2005) The tuberous sclerosis protein TSC2 is not required for the regulation of the mammalian target of rapamycin by amino acids and certain cellular stresses. J. Biol. Chem. 280, 1871718727 49 Long, X. et al. (2005) Rheb binds and regulates the mTOR kinase. Curr. Biol. 15, 702713 50 Long, X. et al. (2005) Rheb binding to mammalian target of rapamycin (mTOR) is regulated by amino acid sufciency. J. Biol. Chem. 280, 2343323436 51 Inoki, K. et al. (2003) TSC2 mediates cellular energy response to control cell growth and survival. Cell 115, 577590 52 Luo, Z. et al. (2005) AMPK, the metabolic syndrome and cancer. Trends Pharmacol. Sci. 26, 6976 53 Lizcano, J.M. et al. (2004) LKB1 is a master kinase that activates 13 kinases of the AMPK subfamily, including MARK/PAR-1. EMBO J. 23, 833843 54 Shaw, R.J. et al. (2004) The LKB1 tumor suppressor negatively regulates mTOR signaling. Cancer Cell 6, 9199 55 Shaw, R.J. et al. (2004) The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc. Natl. Acad. Sci. U. S. A. 101, 33293335 56 Corradetti, M.N. et al. (2004) Regulation of the TSC pathway by LKB1: evidence of a molecular link between tuberous sclerosis complex and PeutzJeghers syndrome. Genes Dev. 18, 15331538 57 Reiling, J.H. and Hafen, E. (2004) The hypoxia-induced paralogs Scylla and Charybdis inhibit growth by down-regulating S6K activity upstream of TSC in Drosophila. Genes Dev. 18, 28792892 58 Brugarolas, J. et al. (2004) Regulation of mTOR function in response to hypoxia by REDD1 and the TSC1/TSC2 tumor suppressor complex. Genes Dev. 18, 28932904 59 Semenza, G.L. (2003) Targeting HIF-1 for cancer therapy. Nat. Rev. Cancer 3, 721732 60 Hudson, C.C. et al. (2002) Regulation of hypoxia-inducible factor 1a expression and function by the mammalian target of rapamycin. Mol. Cell. Biol. 22, 70047014 61 Brugarolas, J.B. et al. (2003) TSC2 regulates VEGF through mTORdependent and -independent pathways. Cancer Cell 4, 147158 62 Inoki, K. et al. (2005) Dysregulation of the TSCmTOR pathway in human disease. Nat. Genet. 37, 1924 63 Tee, A.R. and Blenis, J. (2005) mTOR, translational control and human disease. Semin. Cell Dev. Biol. 16, 2937 64 Eng, C. (2003) PTEN: one gene, many syndromes. Hum. Mutat. 22, 183198 65 Harrington, L.S. et al. (2005) Restraining PI3K: mTOR signalling goes back to the membrane. Trends Biochem. Sci. 30, 3542 66 Harrington, L.S. et al. (2004) The TSC1-2 tumor suppressor controls insulin-PI3K signaling via regulation of IRS proteins. J. Cell Biol. 166, 213223 67 Shah, O.J. et al. (2004) Inappropriate activation of the TSC/Rheb/ mTOR/S6K cassette induces IRS1/2 depletion, insulin resistance, and cell survival deciencies. Curr. Biol. 14, 16501656

DTD 5

ARTICLE IN PRESS
TRENDS in Molecular Medicine Vol.xx No.xx Monthxxxx

TRMOME 269 9

Opinion

68 Radimerski, T. et al. (2002) Lethality of Drosophila lacking TSC tumor suppressor function rescued by reducing dS6K signaling. Genes Dev. 16, 26272632 69 Zhang, H. et al. (2003) Loss of Tsc1/Tsc2 activates mTOR and disrupts PI3KAkt signaling through downregulation of PDGFR. J. Clin. Invest. 112, 12231233 70 Takano, A. et al. (2001) Mammalian target of rapamycin pathway regulates insulin signaling via subcellular redistribution of insulin receptor substrate 1 and integrates nutritional signals and metabolic signals of insulin. Mol. Cell. Biol. 21, 50505062 71 Tremblay, F. and Marette, A. (2001) Amino acid and insulin signaling via the mTOR/p70 S6 kinase pathway. A negative feedback mechanism leading to insulin resistance in skeletal muscle cells. J. Biol. Chem. 276, 3805238060 72 Dong, J. and Pan, D. (2004) Tsc2 is not a critical target of Akt during normal Drosophila development. Genes Dev. 18, 24792484 73 Mora, A. et al. (2004) PDK1, the master regulator of AGC kinase signal transduction. Semin. Cell Dev. Biol. 15, 161170 74 Balendran, A. et al. (1999) PDK1 acquires PDK2 activity in the presence of a synthetic peptide derived from the carboxyl terminus of PRK2. Curr. Biol. 9, 393404 75 Persad, S. et al. (2001) Regulation of protein kinase B/Akt-serine 473 phosphorylation by integrin-linked kinase: critical roles for kinase activity and amino acids arginine 211 and serine 343. J. Biol. Chem. 276, 2746227469 76 Toker, A. and Newton, A.C. (2000) Akt/protein kinase B is regulated by autophosphorylation at the hypothetical PDK-2 site. J. Biol. Chem. 275, 82718274 77 Feng, J. et al. (2004) Identication of a PKB/Akt hydrophobic motif Ser-473 kinase as DNA-dependent protein kinase. J. Biol. Chem. 279, 4118941196 78 Chen, M.Y. et al. (1997) A novel cytosolic regulator, Pianissimo, is required for chemoattractant receptor and G protein-mediated activation of the 12 transmembrane domain adenylyl cyclase in Dictyostelium. Genes Dev. 11, 32183231 79 Meili, R. et al. (1999) Chemoattractant-mediated transient activation and membrane localization of Akt/PKB is required for efcient chemotaxis to cAMP in Dictyostelium. EMBO J. 18, 20922105 80 Kapahi, P. et al. (2004) Regulation of lifespan in Drosophila by modulation of genes in the TOR signaling pathway. Curr. Biol. 14, 885890 81 Jia, K. et al. (2004) The TOR pathway interacts with the insulin signaling pathway to regulate C. elegans larval development, metabolism and life span. Development 131, 38973906 82 Vellai, T. et al. (2003) Genetics: inuence of TOR kinase on lifespan in C. elegans. Nature 426, 620 83 Edinger, A.L. et al. (2003) Differential effects of rapamycin on mammalian target of rapamycin signaling functions in mammalian cells. Cancer Res. 63, 84518460 84 Cheatham, L. et al. (1995) Structural and functional analysis of pp70S6k. Proc. Natl. Acad. Sci. U. S. A. 92, 1169611700 85 Weng, Q.P. et al. (1995) Multiple independent inputs are required for activation of the p70 S6 kinase. Mol. Cell. Biol. 15, 23332340 86 Ali, S.M. and Sabatini, D.M. (2005) Structure of S6K1 determines if raptormTOR or rictormTOR phosphorylates its hydrophobic motif site. J. Biol. Chem. 280, 1944519448 87 Noh, W.C. et al. (2004) Determinants of rapamycin sensitivity in breast cancer cells. Clin. Cancer Res. 10, 10131023 88 Huang, S. et al. (2001) p53/p21CIP1 cooperate in enforcing rapamycininduced G1 arrest and determine the cellular response to rapamycin. Cancer Res. 61, 33733381 89 Lieberthal, W. et al. (2001) Rapamycin impairs recovery from acute renal failure: role of cell-cycle arrest and apoptosis of tubular cells. Am. J. Physiol. Renal Physiol. 281, F693F706

90 Kenerson, H.L. et al. (2002) Activated mammalian target of rapamycin pathway in the pathogenesis of tuberous sclerosis complex renal tumors. Cancer Res. 62, 56455650 91 Thimmaiah, K.N. et al. (2003) Insulin-like growth factor I-mediated protection from rapamycin-induced apoptosis is independent of RasErk1Erk2 and phosphatidylinositol 3 0 -kinase-Akt signaling pathways. Cancer Res. 63, 364374 92 Woltman, A.M. et al. (2003) Rapamycin specically interferes with GM-CSF signaling in human dendritic cells, leading to apoptosis via increased p27KIP1 expression. Blood 101, 14391445 93 Huang, S. et al. (2003) Sustained activation of the JNK cascade and rapamycin-induced apoptosis are suppressed by p53/p21Cip1. Mol. Cell 11, 14911501 94 Ruggero, D. et al. (2004) The translation factor eIF-4E promotes tumor formation and cooperates with c-Myc in lymphomagenesis. Nat. Med. 10, 484486 95 Wendel, H.G. et al. (2004) Survival signalling by Akt and eIF4E in oncogenesis and cancer therapy. Nature 428, 332337 96 Hentges, K.E. et al. (2001) FRAP/mTOR is required for proliferation and patterning during embryonic development in the mouse. Proc. Natl. Acad. Sci. U. S. A. 98, 1379613801 97 Murakami, M. et al. (2004) mTOR is essential for growth and proliferation in early mouse embryos and embryonic stem cells. Mol. Cell. Biol. 24, 67106718 98 Gangloff, Y.G. et al. (2004) Disruption of the mouse mTOR gene leads to early postimplantation lethality and prohibits embryonic stem cell development. Mol. Cell. Biol. 24, 95089516 99 Kenyon, C. (2005) The plasticity of aging: insights from long-lived mutants. Cell 120, 449460 100 Krymskaya, V.P. and Shipley, J.M. (2003) Lymphangioleiomyomatosis: a complex tale of serum response factor-mediated tissue inhibitor of metalloproteinase-3 regulation. Am. J. Respir. Cell Mol. Biol. 28, 546550 101 Abel, T.W. et al. (2005) LhermitteDuclos disease: a report of 31 cases with immunohistochemical analysis of the PTEN/AKT/mTOR pathway. J. Neuropathol. Exp. Neurol. 64, 341349 102 Blair, E. et al. (2001) Mutations in the g2 subunit of AMP-activated protein kinase cause familial hypertrophic cardiomyopathy: evidence for the central role of energy compromise in disease pathogenesis. Hum. Mol. Genet. 10, 12151220 103 Dreher, T. et al. (2004) Reduction of PTEN and p27kip1 expression correlates with tumor grade in prostate cancer. Analysis in radical prostatectomy specimens and needle biopsies. Virchows Arch. 444, 509517 104 Li, J. et al. (1997) PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science 275, 19431947 105 Bellacosa, A. et al. (1995) Molecular alterations of the AKT2 oncogene in ovarian and breast carcinomas. Int. J. Cancer 64, 280285 106 Zhou, B.P. and Hung, M.C. (2003) Dysregulation of cellular signaling by HER2/neu in breast cancer. Semin. Oncol. 30 (Suppl. 16), 3848 107 Barnes, C.J. and Kumar, R. (2003) Epidermal growth factor receptor family tyrosine kinases as signal integrators and therapeutic targets. Cancer Metastasis Rev. 22, 301307 108 Aveyard, J.S. et al. (1999) Somatic mutation of PTEN in bladder carcinoma. Br. J. Cancer 80, 904908 109 Shayesteh, L. et al. (1999) PIK3CA is implicated as an oncogene in ovarian cancer. Nat. Genet. 21, 99102 110 Rasheed, B.K. et al. (1999) Molecular pathogenesis of malignant gliomas. Curr. Opin. Oncol. 11, 162167

www.sciencedirect.com

You might also like