Critical Reviews in Oncology/Hematology 56 (2005) 47–60
Rapamycin: An anti-cancer immunosuppressant? Brian K. Law ∗ Department of Pharmacology and Therapeutics and the Shands Cancer Center, University of Florida, P.O. Box 100267, R5-136, ARB, 1600 SW Archer Road, Gainesville, FL 32610, USA Accepted 24 September 2004
Contents 1. 2.
3. 4.
5.
6.
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . mTOR mediates control of the cell cycle by nutrient levels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.1. TOR and mTOR regulate cell proliferation in response to nutrient availability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.2. The mTOR signaling network . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.3. mTOR regulation of the mammalian cell cycle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Rapamycin as an immunosuppressant . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Rapamycin and rapamycin analogs as anti-cancer agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.1. The mTOR signaling pathway is activated in cancers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.2. Rapamycin anti-tumor actions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.3. Rapamycin inhibition of angiogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Future directions: harnessing the anti-cancer potential of mTOR inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.1. The mTOR pathway as a target of multiple existing anti-cancer agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.2. Potential synergy between mTOR inhibitors and other anti-cancer agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Reviewers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Biography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
48 48 48 48 49 50 51 51 52 53 53 53 53 54 54 54 60
Abstract Rapamycin and its derivatives are promising therapeutic agents with both immunosuppressant and anti-tumor properties. These rapamycin actions are mediated through the specific inhibition of the mTOR protein kinase. mTOR serves as part of an evolutionarily conserved signaling pathway that controls the cell cycle in response to changing nutrient levels. The mTOR signaling network contains a number of tumor suppressor genes including PTEN, LKB1, TSC1, and TSC2, and a number of proto-oncogenes including PI3K, Akt, and eIF4E, and mTOR signaling is constitutively activated in many tumor types. These observations point to mTOR as an ideal target for anti-cancer agents and suggest that rapamycin is such an agent. In fact, early preclinical and clinical studies indicate that rapamycin derivatives have efficacy as anti-tumor agents both alone, and when combined with other modes of therapy. Rapamycin appears to inhibit tumor growth by halting tumor cell proliferation, inducing tumor cell apoptosis, and suppressing tumor angiogenesis. Rapamycin immunosuppressant actions result from the inhibition of T and B cell proliferation through the same mechanisms that rapamycin blocks cancer cell proliferation. Therefore, one might think that rapamycin-induced immunosuppression would be detrimental to the use of rapamycin as an anti-cancer agent. To the contrary, rapamycin decreases the frequency of tumor formation that occurs in organ transplant experiments when combined with the widely used immunosuppressant cyclosporine compared with the tumor incidence observed when cyclosporine is used alone. The available evidence indicates that with respect to tumor growth, rapamycin anti-cancer activities are dominant over rapamycin immunosuppressant effects. © 2005 Elsevier Ireland Ltd. All rights reserved. Keywords: Rapamycin; RAD001; CCI-779; mTOR; Cancer; Cell cycle; Cyclin-dependent kinase; Immunosuppressant ∗
Tel.: +1 352 392 3395; fax: +1 352 392 9696. E-mail address:
[email protected].
1040-8428/$ – see front matter © 2005 Elsevier Ireland Ltd. All rights reserved. doi:10.1016/j.critrevonc.2004.09.009
48
B.K. Law / Critical Reviews in Oncology/Hematology 56 (2005) 47–60
1. Introduction
2.2. The mTOR signaling network
Rapamycin is perhaps best known as a potent immunosuppressive agent. It is unclear from this vantage point what utility rapamycin might have as an anti-cancer agent. This perspective however stems from the historical development of rapamycin use rather than the intrinsic nature of rapamycin action. The aim of this review is to rationalize how rapamycin is able to act both as an immunosuppressive agent and an anticancer agent. Rapamycin was first identified as an anti-fungal agent produced by the bacterium Streptomyces hygroscopicus [1,2] and was subsequently demonstrated to be a potent immunosuppressive agent [3–6]. The target of rapamycin (TOR) was identified in a screen of yeast mutants able to proliferate in the presence of rapamycin [7]. A mammalian protein homologous to TOR (mTOR) was isolated later based on its ability to bind the FK506 binding protein FKBP12 [8–10]. TOR bound to FKBP12 in the presence of rapamycin, but not in the presence of the related immunosuppressant bacterial macrolide FK506. mTOR is a 289 kDa protein that is evolutionarily related to lipid kinases, but exhibits protein serine/threonine kinase activity (reviewed in [11]).
It has long been known that FKBP12 is an intracellular receptor for rapamycin [29], that the FKBP12–rapamycin complex binds to TOR/mTOR [7–10], and that the formation of this ternary complex inhibits the intrinsic kinase activity of mTOR toward exogenous substrates [30–33]. It is also well known that p70s6k and 4EBP1 are two critical downstream targets of mTOR signaling and that rapamycin is a dominant inhibitor of p70s6k and 4EBP1 function [34–37]. However, the mechanisms by which mTOR is regulated, and in turn regulates downstream signaling, has largely been a “black box” until quite recently. Raptor was the first protein shown to bind directly to mTOR that is required to mediate mTOR regulation of p70s6k and 4EBP1 activities [38,39]. Strikingly, Raptor forms nutrient-sensitive complexes with mTOR and another protein, GßL, that is required for the nutrient-sensitive interaction between Raptor and mTOR [40]. In addition to participating in a nutrient-sensing complex, Raptor acts as a scaffolding protein that facilitates mTOR-dependent phosphorylation of p70s6k and 4EBP1 by binding the evolutionarily conserved TOR signaling motifs (TOS) present in both p70s6k and 4EBP1 [41,42]. Together, these studies provide the first mechanistic glimpses into the basis for the nutrient sensitivity of the mTOR signaling network and how mTOR is coupled to the downstream effectors p70s6k and 4EBP1. The mTOR signaling network as it is currently understood (Fig. 1) involves a large number of different signaling intermediates. The upstream regulators of mTOR fall into at least two independent signaling cascades. The first involves the lipid kinase phosphatidylinositol 3-kinase (PI3K), the phosphoinositide-dependent protein kinase 1 (PDK1), the proto-oncogenic protein kinase Akt, the tumor suppressive kinase LKB1, and the AMP-dependent protein kinase (AMPK), the tumor suppressive tuberous sclerosis complex proteins 1 and 2 (TSC1/TSC2), and the Ras related Rheb GTPase. The second cascade involves the small GTPase Cdc42 and phospholipase D1 (PLD1).
2. mTOR mediates control of the cell cycle by nutrient levels 2.1. TOR and mTOR regulate cell proliferation in response to nutrient availability Cell proliferation must be regulated such that cell division occurs only when adequate levels of all necessary nutrients are available. However, despite detailed knowledge of how cells respond to growth factors, relatively little is known concerning how cells respond to changes in nutrient levels. In budding yeast, the TOR proteins mediate responses to levels of nutrients such as nitrogen [12,13]. In mammals, mTOR integrates signals and mediates biological responses to growth factors and mitogens [14,15], amino acid levels [16–20], phosphatidic acid levels [21], ATP levels [22], inorganic polyphosphate levels [23], and AMP levels [24,25]. The observation that TOR homologs are responsive to nutrients in both yeast and metazoans suggests that this function of TOR is conserved throughout eukaryotic evolution, and that the TOR proteins play a fundamental role in coupling nutrient availability with cell cycle regulation. In this sense, mTOR can be considered to act as a key component of a nutrient-responsive cell cycle regulatory signaling pathway. Interestingly, all other members of the PIKK family, including ATM and ATR [26–28], act in some capacity as genome/transcriptome surveillance proteins with mTOR being the only direct participant in mitogenic signaling pathways. Since the focus of this review is on rapamycin as an immunosuppressant and anti-cancer agent, subsequent sections will focus on mTOR and its biological functions in cells of the immune system and cancer cells.
Fig. 1. The mTOR cell signaling network.
B.K. Law / Critical Reviews in Oncology/Hematology 56 (2005) 47–60
PI3K is activated by a number of receptor tyrosine kinases and catalyzes the conversion of phosphatidylinositol bisphosphate (PIP2 ) to PIP3 by phosphorylating the 3 position of the inositol ring. Consistent with the notion that PI3K plays a critical role in regulating cell proliferation, the enzyme that catalyzes the dephosphorylation of PIP3 at the 3 position, the phosphatase and Tensin homologous protein (PTEN), is encoded by a well known tumor suppressor gene whose expression is lost in a number of human cancers. PIP3 residing in the plasma membrane docks PDK1 and Akt to the membrane through their respective pleckstrin homology (PH) domains. While colocalized at the membrane, PDK1 phosphorylates and activates Akt. The activity of TSC1/TSC2 is in turn regulated via inhibition by Aktmediated phosphorylation [43,44]. The activity of TSC2 is dominantly inhibited by AMPK-mediated phosphorylation independent of the level of Akt activity [25]. AMPK is activated coordinately by LKB1-dependent phosphorylation and by increased levels of AMP during times of metabolic stress [45–48]. The TSC1 and TSC2 proteins are also referred to as Hamartin and Tuberin, respectively. Several recent reports have elucidated the role that TSC1/TSC2 play in mTOR regulation. Rheb GTPase activity is regulated by the GTPase activating protein (GAP) function of TSC2 as part of the TSC1/TSC2 complex [49–51]. Active GTP-bound Rheb stimulates mTOR kinase activity through mechanisms that are not completely characterized [49,50,52]. In a parallel pathway, GTP-bound Cdc42 binds and activates PLD1 [53]. PLD1 in turn catalyzes the production of phosphatidic acid. Phosphatidic acid stimulates mTOR kinase activity and the phosphorylation of its downstream effectors [21,54–56]. The most well studied downstream effectors of mTOR are eiF4E binding protein 1 (4EBP1) [57,58], which was independently discovered and termed phosphorylated heat and acid stable protein-I (PHAS-I) [59–61], and the protein kinase, p70s6k . 4EBP1 binds and inhibits the activity of the translation factor eiF4E. eiF4E recognizes the 5 -7-Me-GTP cap of mRNAs and is thus required for cap-dependent translation (reviewed in [11]). Growth factor stimulation or stimulation of cells with high amino acid levels induces the multi-site phosphorylation of 4EBP1, resulting in the release of eiF4E and the activation of protein synthesis. Importantly, several 4EBP1 phosphorylation sites are directly phosphorylated by mTOR, and phosphorylation of these sites is inhibited by rapamycin [32,58,62]. p70s6k was one of the earliest characterized protein kinases [63–67] and its activity is strongly stimulated by growth factors including insulin and PDGF [68], and nutrients such as amino acids [16–19]). p70s6k activation in response to all of these stimuli is blocked by rapamycin, implicating p70s6k as a downstream effector of mTOR. It is still a matter of debate as to whether p70s6k is regulated by mTOR directly, indirectly, or both. p70s6k regulation is complex and involves the phosphorylation of a number of different sites by several kinases
49
including PDK1 [69,70], Cdc2 [71], and likely mTOR itself [41,72]. Interestingly, a large number of protein kinases, including p70s6k , require PDK1-dependent phosphorylation for activation. Thus, p70s6k is regulated directly by PDK1-dependent phosphorylation at Thr229 [69] and indirectly through PDK1dependent activation of the Akt–TSC1/TSC2–Rheb–mTOR axis. Several p70s6k phosphorylation sites are rapamycin sensitive and therefore regulated via mTOR, however, it has been argued that the Thr389 phosphorylation site is the primary rapamycin-sensitive phosphorylation site [73]. Once activated, p70s6k phosphorylates several proteins that regulate translation and RNA splicing including the ribosomal S6 protein and the CBP80 cap-binding protein [74]. Since 4EBP1 and p70s6k participate in controlling protein synthesis, and amino acid levels control protein synthesis by regulating the mTOR signaling pathway, the well accepted model has emerged that rapamycin acts primarily by inhibiting mTOR protein kinase activity, resulting in the inhibition of protein synthesis. While this model is accurate, it is likely an oversimplification. Multiple PKC isoforms appear to serve as effectors of mTOR and to participate in the regulation of other mTOR downstream effectors, including p70s6k and 4EBP1. An early report indicated that phorbol ester-sensitive PKC isoforms play a role in p70s6k activation by EGF [75]. More recently, the novel PKC isoforms ␦ and were shown to have regulatory phosphorylation sites whose phosphorylation is inhibited by rapamycin treatment, including Ser662 of PKC␦ [76]. Intriguingly, PKC␦ interacts physically with mTOR and this interaction is required for mTOR-dependent regulation of 4EBP1 phosphorylation [77]. The atypical PKC isoforms PKC and PKC exist in complexes with p70s6k and PDK1, and kinase dead mutants of PKC or PKC antagonize p70s6k activation [78]. PKC also phosphorylates PKC␦ on the rapamycin sensitive site Ser662 [79], suggesting that novel and atypical PKC isoforms collaborate in their participation in the mTOR signaling network. These observations suggest that PKC isoforms play an important, but largely unexplored role in signaling downstream of mTOR. Other recent reports have indicated that the transcriptional activity of STATs 1 [80] and 3 [81,82] are also regulated by mTOR-dependent phosphorylation, and that rapamycin regulates Pol I-dependent transcription [83]. Thus, there is likely still much to learn about the mechanisms of rapamycin action and the mTOR signaling pathway. 2.3. mTOR regulation of the mammalian cell cycle The entry of eukaryotic cells into the cell cycle is controlled primarily through the activation of the cyclindependent kinases (Cdks) Cdk4/Cdk6 and Cdk2 by their respective cyclin partners Cyclins D1, D2, D3, and E and A (Fig. 2). Increased expression of the D-type cyclins is a rapid response to growth factor stimulation and is mediated by several different signaling pathways including the Ras/Map
50
B.K. Law / Critical Reviews in Oncology/Hematology 56 (2005) 47–60
Fig. 2. Rapamycin down-regulates proteins that drive cell proliferation.
kinase cascade and the PI3K/PDK1/Akt/GSK3 cascade (reviewed in [84]). Growth factor-induced D-type cyclins form catalytically active kinase complexes with Cdk4 and Cdk6 and initiate phosphorylation and inactivation of the tumor suppressor Rb. Partial phosphorylation of Rb allows some E2F-dependent transcription that results in the expression of Cyclins E and A. Cyclin E forms complexes with Cdk2 causing more complete phosphorylation and inactivation of Rb, leading to irreversible entry into the S-phase of the cell cycle. Given the potent anti-proliferative effects of rapamycin, it is not surprising that the mTOR signaling pathway regulates the levels of several proteins directly involved in controlling cell division. In a number of studies, rapamycin has been shown to inhibit the expression of Cyclins D1 [85–88] and D3 [89,90] and to suppress c-Myc translation [91]. In hepatocytes, rapamycin-induced cell cycle arrest is mediated by Cyclin D1 down-regulation [92]. Likewise, Cyclin A and PCNA expression are suppressed by rapamycin [88,93–97], although the mechanisms involved in this suppression are unclear. Recently, Cyclin E expression was shown to be under the control of p70s6k and therefore under the control of mTOR signaling [98]. It is not known to what extent changes in the levels of these individual proteins contribute to rapamycininduced cell cycle arrest, and which changes are causal to cell cycle arrest or a result of cell cycle arrest. The observation that rapamycin inhibits E2F-dependent transcription [78,99,100] and that several rapamycin-suppressed genes are E2F-dependent, including Cyclins E, A, and PCNA, suggests that rapamycin suppression of the levels of these proteins may be at least partially a result of cell cycle arrest. The cyclin-dependent kinase inhibitor proteins, p21 and p27, are also downstream effectors of rapamycin action and may mediate rapamycin cell cycle effects. Rapamycin inhibition of T cell mitogenesis correlates with decreased p21 expression and increased p27 expression, p27 association with Cdk2, and Cdk2 inhibition [101]. Subsequent studies have demonstrated that rapamycin inhibits increases in p21
levels in response to phorbol esters [102], vascular endothelial cell growth factor (VEGF) [103], and serum [104]. The interpretation of these results is complicated by the fact that p21 and p27 act as cyclin-dependent kinase “inhibitors” by blocking kinase activity [105–111], but also facilitate the assembly and nuclear localization during the maturation of cyclin-dependent kinase complexes and thus can also act as cyclin-dependent kinase “activators” [112–115]. The degree to which p21 or p27 serve as an “activator” or “inhibitor” may depend on the levels of p21 and p27 relative to the levels of the relevant cyclin-dependent kinases. This might explain why phorbol ester-induced increases in p21 is associated with growth arrest in human venous endothelial cells [102], while rapamycin-induced decreases in p21 levels in BP-A31 fibroblasts is associated with growth arrest [104]. Thus, with respect to p21, rapamycin induces p21 downregulation that may have differing effects on proliferation depending on the cellular context. As is the case with p21, the role of p27 in rapamycin action is complex. T cells from p27 null mice have impaired growth inhibitory responses to rapamycin, and rapamycin blocks serum-induced decreases in p27 levels [116]. Consistent with rapamycin down-regulating p27, Tsc2 null fibroblasts, that exhibit constitutively active mTOR signaling (Fig. 1), have very low levels of p27 expression and exhibit p27 mislocalization to the cytoplasm [117]. p27 may also be regulated by rapamycin through indirect mechanisms. It has been proposed that Cyclin D–Cdk complexes act as a “sink” to bind p27 and prevent it from binding and inhibiting Cdk2 [118–120]. Since rapamycin down-regulates Cyclins D1 and D3 [87–90,121,122], it has been suggested that rapamycin also inhibits cell proliferation by removing the p27 sink and allowing p27 to bind and inactivate Cdk2 [87]. Thus, p21 and p27 play important roles in rapamycin cell cycle effects but likely do so in a context-dependent manner. Rapamycin has been assumed to induce cell cycle arrest through the down-regulation of Cyclins D1, E, and A, inactivation of their corresponding cyclin-dependent kinase partners, and by decreasing Rb phosphorylation. Interestingly, rapamycin inhibition of cell proliferation does not always correlate with dramatic decreases in Rb phosphorylation. This lack of decreased Rb phosphorylation might be explained by Cdk-independent functions of the cyclins. Cyclin E, for example, has been proposed to stimulate proliferation through a Cdk-independent mechanism [123], and Cyclin D1 interacts with and regulates a number of different transcription factors including DMP1 and TAF(II)250 [124–126]. Therefore, rapamycin likely inhibits cell proliferation through both Cdk-dependent and Cdk-independent mechanisms.
3. Rapamycin as an immunosuppressant Shortly after it was first described as an anti-fungal antibiotic [1,2] rapamycin was demonstrated to have
B.K. Law / Critical Reviews in Oncology/Hematology 56 (2005) 47–60
immunosuppressant properties as indicated by its ability to inhibit experimental allergic encephalomyelitis, adjuvant arthritis, and the humoral (IgE) immune response [127]. Several years later, rapamycin was shown to inhibit tumor growth in xenograft models [128], an observation seemingly inconsistent with rapamycin acting as an immunosuppressant. The observation that rapamycin shares structural similarities with a known immunosuppressant, FK506, probably led to the early exploration of the immunosuppressive actions of rapamycin. Studies demonstrating that rapamycin inhibits renal rejection in pig allograft models [4] led to cell biological assays showing that rapamycin suppresses T and B cell activation [129,130] and proliferation [131]. A recent report showing that rapamycin induces dendritic cell apoptosis by interfering with GM–CSF signaling suggests that rapamycin not only inhibits T cell activation directly, but may also decrease the population of dendritic cells that present antigen to T cells during activation [132]. Upon investigating the mechanisms of action of FK506 and rapamycin as immunosuppressive agents, it was observed that despite the structural similarity between FK506 and rapamycin, the two drugs antagonize each other’s binding to cells and therefore inhibit each other’s biological function [133]. These results indicate that FK506 and rapamycin bind to the same intracellular receptor(s), but elicit their actions through distinct mechanisms. The intracellular binding protein for FK506 and rapamycin in T cells was termed FK506 binding protein (FKBP). The rapamycin–FKBP12 complex inhibits the activity of the mTOR protein kinase (see Section 2.2), while the FK506–FKBP12 complex inhibits the activity of the protein phosphatase calcineurin [134,135]. mTOR inhibition by rapamycin results in the blockade of interleukin-2 (IL-2)-dependent activation of p70s6k and T cell mitogenesis [136,137], while calcineurin inhibits T cell activation by blocking IL-2 production [138]. Thus, nature has designed two related bacterial macrolides that act at distinct steps to abrogate T cell activation. The effectiveness of rapamycin as an immunosuppressant likely rests on the fact that IL-2-dependent T cell proliferation is selectively dependent on mTOR signaling to drive T cell mitogenesis [136]. Moreover, as discussed in Section 5.2 below, several different oncogenic lesions may render cancer cells dependent on the mTOR signaling for proliferation, cell survival, and angiogenesis. Further, as reviewed in Section 2.1, the mechanistic basis for rapamycin action is conserved from Saccharomyces cerevisiae to man. Thus, rapamycin
51
cannot be viewed simply as an immunosuppressant but must be viewed as an inhibitor of an evolutionarily conserved pathway that couples nutrient sensing and growth factor signaling with cell proliferation. Rapamycin blockade of the mTOR signaling cascade can be viewed as mimicking nutrient deprivation, resulting in cell cycle arrest [139]. Cancer is fundamentally a disease of deregulated cell division suggesting that a cytostatic agent such as rapamycin might have anti-cancer efficacy. A reasonable concern regarding the use of rapamycin as an anti-cancer therapy is that suppression of the immune response might accentuate rather than halt tumor growth. The available empirical evidence argues to the contrary. As described below (Section 4.2), in clinical and preclinical studies rapamycin inhibits the growth of a broad array of tumor types with no reports of rapamycin accentuating tumor growth. In studies using a renal adenocarcinoma model rapamycin inhibits the growth of tumors in both the presence and absence of cyclosporine [140]. This result suggests that combining rapamycin with cyclosporine for immunosuppressant therapy may prevent or suppress the growth of tumors that arise during immunosuppression using cyclosporine alone. Consistent with the anti-proliferative actions of rapamycin on both immune cells and cancer cells, rapamycin and rapamycin derivatives are used as anti-rejection immunosuppressive agents [141–145] and are also in clinical trials as anti-cancer agents (Section 4.2). Although the short-term studies performed to date suggest that the anti-cancer actions of rapamycin derivatives are dominant over the immunosuppressant actions of rapamycin with respect to tumor growth, it is still a concern that the immunosuppressant effects of rapamycin may prove deleterious over the long-term.
4. Rapamycin and rapamycin analogs as anti-cancer agents 4.1. The mTOR signaling pathway is activated in cancers A hallmark of human cancer is deregulation of the cell cycle [146]. Consistent with the notion that the mTOR pathway is a key regulator of cell proliferation, several upstream activators and downstream effectors of mTOR are deregulated in cancers (Table 1). The two most well studied mTOR effectors are p70s6k and 4EBP1. p70s6k is overexpressed in
Table 1 Constitutive activation of the mTOR signaling network in cancers mTOR pathway signaling intermediates
Biochemical function
Biological function
Alteration in cancers
p70s6k PTEN eiF4E TSC1/TSC2 LKB1
Protein kinase PIP3 phosphatase Translation factor Rheb GAP Protein kinase
Stimulates translation Tumor suppressor Stimulates translation; oncogene Tumor suppressor Tumor suppressor
Overexpression Loss of expression or mutational inactivation Overexpression Loss of expression or mutational inactivation Loss of expression or mutational inactivation
52
B.K. Law / Critical Reviews in Oncology/Hematology 56 (2005) 47–60
breast cancers and is constitutively activated in several types of cancers [121,147,148]. Significantly, p70s6k overexpression correlates with poor prognosis [149,150]. As described above, 4EBP1 binds and inhibits the translation factor eiF4E and in so doing blocks the translation of messages containing a 5 -7-Me-GTP cap. Since 4EBP1 is a titratable inhibitor of eiF4E, it has been proposed [151] that the ratio of eiF4E to 4EBP1 present in cancers should be determined rather than examining the absolute amount of either protein. mTOR-dependent phosphorylation of 4EBP1 releases eiF4E allowing the translation of a number of proteins, including proteins that stimulate proliferation such as Cyclin D1 [152] and c-Myc [153]. Thus, one way by which a cancer might bypass 4EBP1-mediated negative regulation is through the overexpression of eiF4E such that eiF4E is in molar excess of 4EBP1. Consistent with this idea, eiF4E is overexpressed in a number of tumor types including cancers of the colon [154], breast [155,156], and head and neck [157]. Cell culture experiments demonstrate that eiF4E overexpression is sufficient to transform cells [158], and that co-expression of 4EBP1 reverses eiF4E-induced transformation [159]. Additionally, eIF4E is overexpressed in a wide array of cancer cell lines [160]. Thus, the two most well studied mTOR downstream effectors, p70s6k and 4EBP1, are deregulated in cancers and likely contribute to the process of tumorigenesis. At least three tumor suppressors that act as negative upstream regulators of mTOR, LKB1, PTEN, and TSC1/TSC2, are deregulated in cancers and may contribute to tumorigenesis through inappropriate mTOR activation (Fig. 1). TSC1/TSC2 were discovered as genes whose inactivation gives rise to nonmalignant tumors called hamartomas [161–163]. Thus, by definition TSC1 and TSC2 are tumor suppressor genes. PTEN is a potent tumor suppressor gene that is inactivated in tumors of multiple organs including cancers of the prostate, brain (glioblastoma), thyroid, ovaries, lung, kidney, endometrium, and bladder [164–174]. Furthermore, genetic inactivation of PTEN in mice results in the formation of multiple types of cancers [175–181]. Similarly, the tumor suppressor LKB1 is inactivated in the Peutz–Jeghers hereditary cancer syndrome [182–184]. Additionally, inactivation of the LKB1 gene by somatic mutation [185–190], and epigenetic inactivation [191] have been observed. LKB1 inactivation is thought to contribute to the formation of melanoma [186,187], pancreatic and biliary cancer [185], ovarian cancer [188], lung cancer [189,190], and breast cancer [192]. LKB1 likely plays a causal role in driving tumorigenesis because heterozygous inactivation of LKB1 in mice results in hepatocellular carcinoma, and the tumors formed exhibit loss of the remaining LKB1 allele [193]. Together, these observations indicate that the mTOR signaling pathway frequently becomes inappropriately activated during tumor progression and contributes to tumorigenesis by deregulating cancer cell proliferation. Thus, targeting signaling through the mTOR pathway with rapamycin derivatives is a promising approach for inhibiting tumor growth.
4.2. Rapamycin anti-tumor actions Only recently has the anti-tumor potential of rapamycin become widely appreciated. Early reports [128,194] demonstrated rapamycin anti-tumor efficacy in glioma and colorectal carcinoma models. More recently, Wyeth-Ayerst Research and Novartis have developed rapamycin derivatives, CCI-779 and RAD001, respectively, as potential cancer therapeutics. CCI-779 and RAD001 are structurally similar to rapamycin and act through the same mechanisms as rapamycin. Anticancer efficacy of CCI-779, a drug formulated for intravenous use, has been demonstrated in preclinical models of rhabdomyosarcoma [195], medulloblastoma [196], and breast cancer [197]. A flurry of reports indicate that constitutive activation of the PTEN–PI3K–Akt axis renders several tumor types exquisitely sensitive to inhibition by CCI-779 [198–201]. The basis for the sensitizing effect of PTEN inactivation to rapamycin action was suggested in a recent report demonstrating that in cells with high levels of Akt activity rapamycin or CCI-779 potently represses the translation of Cyclin D1 and c-Myc in a very specific manner [202]. This result may also explain the rapamycin sensitivity of cells transformed by oncoproteins and growth factors including v-Src [203], Gli [204], PI3K and Akt [205], and EGF [206] that drive transformation and cell division in part by constitutive activation of the PI3K–Akt–mTOR axis. RAD001 is expected to have similar anti-cancer effects as rapamycin and CCI-779, and in fact potently inhibits the growth of pancreatic cancer in the CA20948 tumor model [207]. However, given the structural differences between RAD001, CCI-779, and rapamycin, along with the fact that RAD001 is formulated for oral administration, the pharmacokinetics of these three agents are likely to differ. The bioactivity of both RAD001 and CCI-779 is prolonged. A single dose of 10 mg/kg CCI-779 inhibits p70s6k activity in peripheral blood mononuclear cells (PBMCs) by greater than 80% 72 h after treatment [208]. CCI-779 effects on PBMC p70s6k activity paralleled effects observed in tumor tissue suggesting that p70s6k activity in PBMCs may be a useful surrogate marker for CCI-779 anti-tumor efficacy. Likewise, a dose of 5 mg/kg RAD001 inhibits p70s6k activity for at least 72 h, and levels of PBMC p70s6k activity serve as a useful surrogate marker for assessing RAD001 treatment schedules [207]. The results of two CCI-779 clinical trials were recently published. The first study [209] was a phase I clinical trial and involved weekly treatments of patients with refractory renal cell carcinoma with 25, 75, or 250 mg of CCI-779 weekly. Seven percent of patients exhibited an objective response and 26% of patients exhibited minor responses. It was observed that CCI-779 dosage did not influence efficacy or toxic side effects that included maculopapular rash, mucositis, asthenia, and nausea. The second study [210] was a randomized phase II clinical trial and involved weekly doses of CCI-779 ranging from 7.5 to 220 mg/m2 . This study reported toxicities similar to those observed in the first study and noted reversible thrombocytopenia as the main dose-limiting toxi-
B.K. Law / Critical Reviews in Oncology/Hematology 56 (2005) 47–60
city. A partial response was observed in a patient with renal clear-cell carcinoma and a patient with breast adenocarcinoma. In summary, the availability of rapamycin derivatives and knowledge concerning their pharmacokinetics and useful surrogate endpoints for biological activity should rapidly accelerate the implementation of mTOR pathway inhibitors as anti-cancer therapeutics. 4.3. Rapamycin inhibition of angiogenesis In addition to the direct effects of rapamycin on tumor cells that are expected to vary from tumor to tumor, rapamycin also inhibits tumor angiogenesis [211]. Given the limited size that tumors can attain in the absence of their own vascular system [212], rapamycin-mediated inhibition of tumor angiogenesis would be expected to inhibit the growth of all solid tumors. Rapamycin inhibits tumor angiogenesis through at least two mechanisms. Rapamycin directly inhibits the proliferation of vascular endothelial cells driven by serum [103] and vascular endothelial growth factor (VEGF) [213]. Rapamycin also suppresses the induction of hypoxia-inducible factor 1␣ (HIF1) by growth factors and oncogenes [214–216], and by hypoxia [217]. HIF1 is a transcriptional regulator of VEGF expression. Thus, rapamycin inhibits vascular endothelial cell proliferation by both decreasing the expression of VEGF, and by inhibiting the mitogenesis of vascular endothelial cells in response to VEGF. Rapamycin also inhibits vascular smooth muscle cell proliferation and migration, likely through the same mechanisms that it inhibits the proliferation of cancer cells and immune cells (summarized in [218]). Coating cardiac stents with rapamycin could potentially revolutionize the use of cardiac stents by overriding their major limitation, restenosis [219,220].
5. Future directions: harnessing the anti-cancer potential of mTOR inhibitors 5.1. The mTOR pathway as a target of multiple existing anti-cancer agents As discussed above the PI3 kinase pathway and mTOR signaling are inextricably intertwined. To complicate matters, presumed PI3K inhibitors such as wortmannin and LY294004 also inhibit mTOR [31]. Thus, the antiproliferative, anti-cancer effects of PI3K inhibitors may be partially attributable to mTOR inactivation. We have shown that the aspirin metabolite salicylate inhibits p70s6k activation and induces many of the same biochemical cell cycle responses as rapamycin [88]. This suggests that some of the chemo-protective effects of salicylates may be due, in part, to suppression of p70s6k activity. We also demonstrated that the ability of the farnesyltransferase inhibitor (FTI) L-744,832 to inhibit p70s6k activity correlates best with its ability to induce cell cycle arrest [221,222].
53
In retrospect this is not surprising since the small GTPase Rheb that regulates mTOR activity (Fig. 1) is a farnesylated protein, FTIs block Rheb function [223], and a farnesylationdefective Rheb mutant has a diminished ability to activate p70s6k [50]. Additionally, mTOR is activated by growth factors, so anti-cancer agents such as receptor tyrosine kinase inhibitors and receptor-specific neutralizing antibodies also likely act in part by blocking activation of mTOR. Likewise, anticancer agents that induce or mimic amino acid depletion such as asparaginase [224], and l-histidinol or l-leucinol [19] likely work in large part through inactivation of the mTOR pathway. Based on these examples, it is clear that in addition to rapamycin derivatives, several other classes of anti-cancer agents act in part through the inhibition of mTOR signaling. 5.2. Potential synergy between mTOR inhibitors and other anti-cancer agents Cancer is a complex disease resulting from the genetic and epigenetic alteration of multiple oncogenes and tumor suppressor genes. Therefore, inhibiting the growth and progression of tumors will likely require a multipronged approach that targets several cancer-promoting genetic alterations through a combination of mechanism-based signaling inhibitors and classical chemotherapeutic agents and radiotherapy. Along these lines, rapamycin is being tested in combination with conventional modes of cancer therapy. As discussed above, multiple receptor tyrosine kinases (RTKs) activate the mTOR signaling pathway through activation of the PI3 kinase pathway, suggesting that tumors harboring activated RTKs may depend on signaling through the mTOR pathway to drive cell proliferation. Consistent with this idea, rapamycin enhances the ability of the Bcr–Abl kinase inhibitor Imatinib to arrest the proliferation of Ba/F3 cells overexpressing Bcr–Abl, but has no effect on the parental cells [225]. Importantly, rapamycin in combination with Imatinib inhibits the growth of cells expressing Imatinib-resistant mutants of Bcr–Abl [226]. MEK inhibitors cooperate with rapamycin and RTK inhibitors (RTKIs) to halt the growth of tumor cells [226]. This might be expected based on a previous report demonstrating that the MEK inhibitor U0126 is more potent than the MEK inhibitor PD98059 in blocking K-Ras-mediated transformation, and this correlates with the ability of U0126 to block both p70s6k and Erk activation [227]. While PD98059 blocks only Erk activation, it cooperates with rapamycin to reverse K-Ras-mediated transformation. Rapamycin also cooperates with classical anti-cancer therapeutic approaches to inhibit tumor growth and progression. For example, rapamycin cooperates with radiation therapy to inhibit the growth of U87 xenografts [228]. Rapamycin also cooperates with DNA damaging therapeutic agents including cisplatin [196,229] and doxorubicin [200]. Significantly, rapamycin cooperates with doxorubicin to inhibit tumor
B.K. Law / Critical Reviews in Oncology/Hematology 56 (2005) 47–60
54
growth even in cell lines rendered doxorubicin-resistant by PTEN inactivation [200]. As points of cross-talk between the mTOR signaling pathway and other pathways are discovered, and it is determined how these points of cross-talk influence tumor progression, it is likely that additional rationally chosen drug combinations involving rapamycin will be developed. Some signaling pathways/signaling elements where cross-talk with the mTOR signaling pathway has already been identified include: the TGFß signaling pathway [100,230], the p38 Map kinase pathway [221,231], STATs 1 and 3 [80–82], and various PKC isoforms [76,79].
[2]
[3] [4] [5] [6]
[7]
6. Conclusions Rapamycin targets an evolutionarily conserved nutrientresponsive cell cycle regulatory pathway. Thus, the potent cytostatic nature of rapamycin stems from its ability to trigger a nutrient deprivation-like response. The growth inhibitory properties of rapamycin prompted researchers to explore the potential anti-cancer properties of rapamycin. It is clear from the discussion above that rapamycin is both a potent immunosuppressant and a promising anti-cancer agent. The anti-tumor efficacy of rapamycin likely stems from the fact that even though it suppresses the immune system, rapamycin inhibits tumor cell proliferation and angiogenesis and promotes tumor cell apoptosis. Stated another way, the effect of rapamycin on tumor growth and progression outweighs the simultaneous suppression of the immune system. Judging by the reports published to date demonstrating rapamycin inhibition of tumor growth, rapamycin derivatives are poised to become important weapons in the war against cancer both alone and in combination with other treatment modalities.
[8]
[9]
[10]
[11] [12]
[13]
[14] [15]
[16]
Reviewers Dr. Iduna Fichtner, Max-Delbr¨uck-Centrum for Molecular Medicine, Experimental Pharmacology, Robert-R¨ossle-Str. 10, D-13125 Berlin-Buch, Germany. Prof. Jaap Verweij, Department of Oncology, Erasmus University Medical Center, Postbus 5201 (Groene Hilledijk 301), NL-3008AE Rotterdam, The Netherlands. Joon-Ho Sheen, Ph.D., Sabatini Lab, Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142-1479, USA. Robert T. Abraham, Ph.D., Professor and Director, Cancer Research Center, The Burnham Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
[17]
[18]
[19]
[20]
References [21] [1] Vezina C, Kudelski A, Sehgal SN. Rapamycin (AY-22,989), a new antifungal antibiotic. I. Taxonomy of the producing strepto-
mycete and isolation of the active principle. J Antibiot (Tokyo) 1975;28:721–6. Sehgal SN, Baker H, Vezina C. Rapamycin (AY-22,989), a new antifungal antibiotic. II. Fermentation, isolation and characterization. J Antibiot (Tokyo) 1975;28:727–32. Calne RY, Collier DS, Lim S, et al. Rapamycin for immunosuppression in organ allografting. Lancet 1989;2:227. Collier SJ. Immunosuppressive drugs. Curr Opin Immunol 1989;2:854–8. Thomson AW, Woo J. Immunosuppressive properties of FK-506 and rapamycin. Lancet 1989;2:443–4. Tocci MJ, Matkovich DA, Collier KA, et al. The immunosuppressant FK506 selectively inhibits expression of early T cell activation genes. J Immunol 1989;143:718–26. Heitman J, Movva NR, Hall MN. Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast. Science 1991;253:905– 9. Brown EJ, Albers MW, Shin TB, et al. A mammalian protein targeted by G1-arresting rapamycin–receptor complex. Nature 1994;369:756–8. Sabatini DM, Erdjument-Bromage H, Lui M, Tempst P, Snyder SH. RAFT1: a mammalian protein that binds to FKBP12 in a rapamycin-dependent fashion and is homologous to yeast TORs. Cell 1994;78:35–43. Sabers CJ, Martin MM, Brunn GJ, et al. Isolation of a protein target of the FKBP12–rapamycin complex in mammalian cells. J Biol Chem 1995;270:815–22. Gingras AC, Raught B, Sonenberg N. Regulation of translation initiation by FRAP/mTOR. Genes Dev 2001;15:807–26. Schmidt A, Beck T, Koller A, Kunz J, Hall MN. The TOR nutrient signalling pathway phosphorylates NPR1 and inhibits turnover of the tryptophan permease. EMBO J 1998;17:6924–31. Beck T, Hall MN. The TOR signalling pathway controls nuclear localization of nutrient-regulated transcription factors. Nature 1999;402:689–92. Dumont FJ, Su Q. Mechanism of action of the immunosuppressant rapamycin. Life Sci 1996;58:373–95. von Manteuffel SR, Dennis PB, Pullen N, Gingras AC, Sonenberg N, Thomas G. The insulin-induced signalling pathway leading to S6 and initiation factor 4E binding protein 1 phosphorylation bifurcates at a rapamycin-sensitive point immediately upstream of p70s6k. Mol Cell Biol 1997;17:5426–36. Hara K, Yonezawa K, Weng QP, Kozlowski MT, Belham C, Avruch J. Amino acid sufficiency and mTOR regulate p70 S6 kinase and eIF-4E BP1 through a common effector mechanism. J Biol Chem 1998;273:14484–94. Wang X, Campbell LE, Miller CM, Proud CG. Amino acid availability regulates p70 S6 kinase and multiple translation factors. Biochem J 1998;334(Pt 1):261–7. Xu G, Kwon G, Marshall CA, Lin TA, Lawrence Jr JC, McDaniel ML. Branched-chain amino acids are essential in the regulation of PHAS-I and p70 S6 kinase by pancreatic beta-cells. A possible role in protein translation and mitogenic signaling. J Biol Chem 1998;273:28178–84. Iiboshi Y, Papst PJ, Kawasome H, et al. Amino acid-dependent control of p70(s6k). Involvement of tRNA aminoacylation in the regulation. J Biol Chem 1999;274:1092–9. Peyrollier K, Hajduch E, Blair AS, Hyde R, Hundal HS. lLeucine availability regulates phosphatidylinositol 3-kinase, p70 S6 kinase and glycogen synthase kinase-3 activity in L6 muscle cells: evidence for the involvement of the mammalian target of rapamycin (mTOR) pathway in the l-leucine-induced up-regulation of system A amino acid transport. Biochem J 2000;350(Pt 2): 361–8. Fang Y, Vilella-Bach M, Bachmann R, Flanigan A, Chen J. Phosphatidic acid-mediated mitogenic activation of mTOR signaling. Science 2001;294:1942–5.
B.K. Law / Critical Reviews in Oncology/Hematology 56 (2005) 47–60 [22] Dennis PB, Jaeschke A, Saitoh M, Fowler B, Kozma SC, Thomas G. Mammalian TOR: a homeostatic ATP sensor. Science 2001;294:1102–5. [23] Wang L, Fraley CD, Faridi J, Kornberg A, Roth RA. Inorganic polyphosphate stimulates mammalian TOR, a kinase involved in the proliferation of mammary cancer cells. Proc Natl Acad Sci USA 2003;100:11249–54. [24] Corradetti MN, Inoki K, Bardeesy N, DePinho RA, Guan KL. Regulation of the TSC pathway by LKB1: evidence of a molecular link between tuberous sclerosis complex and Peutz–Jeghers syndrome. Genes Dev 2004;18:1533–8. [25] Shaw RJ, Bardeesy N, Manning BD, et al. The LKB1 tumor suppressor negatively regulates mTOR signaling. Cancer Cell 2004;6:91–9. [26] Bentley NJ, Holtzman DA, Flaggs G, et al. The Schizosaccharomyces pombe rad3 checkpoint gene. EMBO J 1996;15:6641–51. [27] Cliby WA, Roberts CJ, Cimprich KA, et al. Overexpression of a kinase-inactive ATR protein causes sensitivity to DNAdamaging agents and defects in cell cycle checkpoints. EMBO J 1998;17:159–69. [28] Wright JA, Keegan KS, Herendeen DR, et al. Protein kinase mutants of human ATR increase sensitivity to UV and ionizing radiation and abrogate cell cycle checkpoint control. Proc Natl Acad Sci USA 1998;95:7445–50. [29] Bierer BE, Mattila PS, Standaert RF, et al. Two distinct signal transmission pathways in T lymphocytes are inhibited by complexes formed between an immunophilin and either FK506 or rapamycin. Proc Natl Acad Sci USA 1990;87:9231–5. [30] Burnett PE, Barrow RK, Cohen NA, Snyder SH, Sabatini DM. RAFT1 phosphorylation of the translational regulators p70 S6 kinase and 4E-BP1. Proc Natl Acad Sci USA 1998;95:1432–7. [31] Brunn GJ, Williams J, Sabers C, Wiederrecht G, Lawrence Jr JC, Abraham RT. Direct inhibition of the signaling functions of the mammalian target of rapamycin by the phosphoinositide 3-kinase inhibitors, wortmannin and LY294002. EMBO J 1996;15:5256–67. [32] Brunn GJ, Hudson CC, Sekulic A, et al. Phosphorylation of the translational repressor PHAS-I by the mammalian target of rapamycin. Science 1997;277:99–101. [33] Withers DJ, Ouwens DM, Nave BT, et al. Expression, enzyme activity, and subcellular localization of mammalian target of rapamycin in insulin-responsive cells. Biochem Biophys Res Commun 1997;241:704–9. [34] Chung J, Kuo CJ, Crabtree GR, Blenis J. Rapamycin-FKBP specifically blocks growth-dependent activation of and signaling by the 70 kd S6 protein kinases. Cell 1992;69:1227–36. [35] Ferrari S, Pearson RB, Siegmann M, Kozma SC, Thomas G. The immunosuppressant rapamycin induces inactivation of p70s6k through dephosphorylation of a novel set of sites. J Biol Chem 1993;268:16091–4. [36] Graves LM, Bornfeldt KE, Argast GM, et al. cAMP- and rapamycin-sensitive regulation of the association of eukaryotic initiation factor 4E and the translational regulator PHAS-I in aortic smooth muscle cells. Proc Natl Acad Sci USA 1995;92:7222–6. [37] Lin TA, Kong X, Saltiel AR, Blackshear PJ, Lawrence Jr JC. Control of PHAS-I by insulin in 3T3-L1 adipocytes. Synthesis, degradation, and phosphorylation by a rapamycin-sensitive and mitogen-activated protein kinase-independent pathway. J Biol Chem 1995;270:18531–8. [38] Hara K, Maruki Y, Long X, et al. Raptor, a binding partner of target of rapamycin (TOR), mediates TOR action. Cell 2002;110:177–89. [39] Kim DH, Sarbassov dos D, Ali SM, et al. mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell 2002;110:163–75. [40] Kim DH, Sarbassov dos D, Ali SM, et al. GbetaL, a positive regulator of the rapamycin-sensitive pathway required for the nutrient-sensitive interaction between raptor and mTOR. Mol Cell 2003;11:895–904.
55
[41] Nojima H, Tokunaga C, Eguchi S, et al. The mammalian target of rapamycin (mTOR) partner, raptor, binds the mTOR substrates p70 S6 kinase and 4E-BP1 through their TOR signaling (TOS) motif. J Biol Chem 2003;278:15461–4. [42] Schalm SS, Fingar DC, Sabatini DM, Blenis J. TOS motif-mediated raptor binding regulates 4E-BP1 multisite phosphorylation and function. Curr Biol 2003;13:797–806. [43] Inoki K, Li Y, Zhu T, Wu J, Guan KL. TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nat Cell Biol 2002;4:648–57. [44] Dan HC, Sun M, Yang L, et al. Phosphatidylinositol 3-kinase/Akt pathway regulates tuberous sclerosis tumor suppressor complex by phosphorylation of tuberin. J Biol Chem 2002;277:35364–70. [45] Hong SP, Leiper FC, Woods A, Carling D, Carlson M. Activation of yeast Snf1 and mammalian AMP-activated protein kinase by upstream kinases. Proc Natl Acad Sci USA 2003;100:8839– 43. [46] Hawley SA, Boudeau J, Reid JL, et al. Complexes between the LKB1 tumor suppressor, STRADalpha/beta and MO25alpha/beta are upstream kinases in the AMP-activated protein kinase cascade. J Biol 2003;2:28. [47] Woods A, Johnstone SR, Dickerson K, et al. LKB1 is the upstream kinase in the AMP-activated protein kinase cascade. Curr Biol 2003;13:2004–8. [48] Shaw RJ, Kosmatka M, Bardeesy N, et al. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc Natl Acad Sci USA 2004;101:3329–35. [49] Inoki K, Li Y, Xu T, Guan KL. Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling. Genes Dev 2003;17:1829–34. [50] Tee AR, Manning BD, Roux PP, Cantley LC, Blenis J. Tuberous sclerosis complex gene products, Tuberin and Hamartin, control mTOR signaling by acting as a GTPase-activating protein complex toward Rheb. Curr Biol 2003;13:1259–68. [51] Zhang Y, Gao X, Saucedo LJ, Ru B, Edgar BA, Pan D. Rheb is a direct target of the tuberous sclerosis tumour suppressor proteins. Nat Cell Biol 2003;5:578–81. [52] Garami A, Zwartkruis FJ, Nobukuni T, et al. Insulin activation of Rheb, a mediator of mTOR/S6K/4E-BP signaling, is inhibited by TSC1 and 2. Mol Cell 2003;11:1457–66. [53] Fang Y, Park IH, Wu AL, et al. PLD1 regulates mTOR signaling and mediates Cdc42 activation of S6K1. Curr Biol 2003;13:2037–44. [54] Chen J, Fang Y. A novel pathway regulating the mammalian target of rapamycin (mTOR) signaling. Biochem Pharmacol 2002;64:1071. [55] Ballou LM, Jiang YP, Du G, Frohman MA, Lin RZ. Ca(2+)and phospholipase D-dependent and -independent pathways activate mTOR signaling. FEBS Lett 2003;550:51–6. [56] Kam Y, Exton JH. Role of phospholipase D1 in the regulation of mTOR activity by lysophosphatidic acid. Faseb J 2004;18:311–9. [57] Pause A, Belsham GJ, Gingras AC, et al. Insulin-dependent stimulation of protein synthesis by phosphorylation of a regulator of 5 -cap function. Nature 1994;371:762–7. [58] Beretta L, Gingras AC, Svitkin YV, Hall MN, Sonenberg N. Rapamycin blocks the phosphorylation of 4E-BP1 and inhibits capdependent initiation of translation. EMBO J 1996;15:658–64. [59] Haystead TA, Haystead CM, Hu C, Lin TA, Lawrence Jr JC. Phosphorylation of PHAS-I by mitogen-activated protein (MAP) kinase. Identification of a site phosphorylated by MAP kinase in vitro and in response to insulin in rat adipocytes. J Biol Chem 1994;269:23185–91. [60] Hu C, Pang S, Kong X, Velleca M, Lawrence Jr JC. Molecular cloning and tissue distribution of PHAS-I, an intracellular target for insulin and growth factors. Proc Natl Acad Sci USA 1994;91:3730–4.
56
B.K. Law / Critical Reviews in Oncology/Hematology 56 (2005) 47–60
[61] Lin TA, Kong X, Haystead TA, et al. PHAS-I as a link between mitogen-activated protein kinase and translation initiation. Science 1994;266:653–6. [62] Mothe-Satney I, Brunn GJ, McMahon LP, Capaldo CT, Abraham RT, Lawrence Jr JC. Mammalian target of rapamycin-dependent phosphorylation of PHAS-I in four (S/T)P sites detected by phospho-specific antibodies. J Biol Chem 2000;275:33836–43. [63] Price DJ, Nemenoff RA, Avruch J. Purification of a hepatic S6 kinase from cycloheximide-treated Rats. J Biol Chem 1989;264:13825–33. [64] Blenis J, Erikson RL. Regulation of a ribosomal protein S6 kinase activity by the Rous sarcoma virus transforming protein, serum, or phorbol ester. Proc Natl Acad Sci USA 1985;82:7621–5. [65] Blenis J, Erikson RL. Regulation of protein kinase activities in PC12 pheochromocytoma cells. EMBO J 1986;5:3441–7. [66] Blenis J, Kuo CJ, Erikson RL. Identification of a ribosomal protein S6 kinase regulated by transformation and growth-promoting stimuli. J Biol Chem 1987;262:14373–6. [67] Jeno P, Ballou LM, Novak-Hofer I, Thomas G. Identification and characterization of a mitogen-activated S6 kinase. Proc Natl Acad Sci USA 1988;85:406–10. [68] Chung J, Grammer TC, Lemon KP, Kazlauskas A, Blenis J. PDGF- and insulin-dependent pp70S6k activation mediated by phosphatidylinositol-3-OH kinase. Nature 1994;370:71–5. [69] Pullen N, Dennis PB, Andjelkovic M, et al. Phosphorylation and activation of p70s6k by PDK1. Science 1998;279:707–10. [70] Balendran A, Currie R, Armstrong CG, Avruch J, Alessi DR. Evidence that 3-phosphoinositide-dependent protein kinase-1 mediates phosphorylation of p70 S6 kinase in vivo at Thr-412 as well as Thr-252. J Biol Chem 1999;274:37400–6. [71] Shah OJ, Ghosh S, Hunter T. Mitotic regulation of ribosomal S6 kinase 1 involves Ser/Thr, Pro phosphorylation of consensus and non-consensus sites by Cdc2. J Biol Chem 2003;278:16433– 42. [72] Isotani S, Hara K, Tokunaga C, Inoue H, Avruch J, Yonezawa K. Immunopurified mammalian target of rapamycin phosphorylates and activates p70 S6 kinase alpha in vitro. J Biol Chem 1999;274:34493–8. [73] Pearson RB, Dennis PB, Han JW, et al. The principal target of rapamycin-induced p70s6k inactivation is a novel phosphorylation site within a conserved hydrophobic domain. EMBO J 1995;14:5279–87. [74] Wilson KF, Wu WJ, Cerione RA. Cdc42 stimulates RNA splicing via the S6 kinase and a novel S6 kinase target, the nuclear capbinding complex. J Biol Chem 2000;275:37307–10. [75] Susa M, Vulevic D, Lane HA, Thomas G. Inhibition or downregulation of protein kinase C attenuates late phase p70s6k activation induced by epidermal growth factor but not by platelet-derived growth factor or insulin. J Biol Chem 1992;267:6905–9. [76] Parekh D, Ziegler W, Yonezawa K, Hara K, Parker PJ. Mammalian TOR controls one of two kinase pathways acting upon nPKCdelta and nPKCepsilon. J Biol Chem 1999;274:34758–64. [77] Kumar V, Pandey P, Sabatini D, et al. Functional interaction between RAFT1/FRAP/mTOR and protein kinase cdelta in the regulation of cap-dependent initiation of translation. EMBO J 2000;19:1087–97. [78] Akimoto K, Nakaya M, Yamanaka T, et al. Atypical protein kinase Clambda binds and regulates p70 S6 kinase. Biochem J 1998;335:417–24. [79] Ziegler WH, Parekh DB, Le Good JA, et al. Rapamycin-sensitive phosphorylation of PKC on a carboxy-terminal site by an atypical PKC complex. Curr Biol 1999;9:522–9. [80] Kristof AS, Marks-Konczalik J, Billings E, Moss J. Stimulation of signal transducer and activator of transcription-1 (STAT1)dependent gene transcription by lipopolysaccharide and interferongamma is regulated by mammalian target of rapamycin. J Biol Chem 2003;278:33637–44.
[81] Yokogami K, Wakisaka S, Avruch J, Reeves SA. Serine phosphorylation and maximal activation of STAT3 during CNTF signaling is mediated by the rapamycin target mTOR. Curr Biol 2000;10:47–50. [82] Rajan P, Panchision DM, Newell LF, McKay RD. BMPs signal alternately through a SMAD or FRAP-STAT pathway to regulate fate choice in CNS stem cells. J Cell Biol 2003;161:911–21. [83] James MJ, Zomerdijk JC. Phosphatidylinositol-3 kinase and mTOR signaling pathways regulate RNA Polymerase I transcription in response to IGF-1 and nutrients. J Biol Chem 2004;279:8911–8. [84] Schwartz MA, Assoian RK. Integrins and cell proliferation: regulation of cyclin-dependent kinases via cytoplasmic signaling pathways. J Cell Sci 2001;114:2553–60. [85] Albers MW, Williams RT, Brown EJ, Tanaka A, Hall FL, Schreiber SL. FKBP-rapamycin inhibits a cyclin-dependent kinase activity and a cyclin D1-Cdk association in early G1 of an osteosarcoma cell line. J Biol Chem 1993;268:22825–9. [86] Withers DJ, Seufferlein T, Mann D, Garcia B, Jones N, Rozengurt E. Rapamycin dissociates p70(S6K) activation from DNA synthesis stimulated by bombesin and insulin in Swiss 3T3 cells. J Biol Chem 1997;272:2509–14. [87] Hashemolhosseini S, Nagamine Y, Morley SJ, Desrivieres S, Mercep L, Ferrari S. Rapamycin inhibition of the G1 to S transition is mediated by effects on cyclin D1 mRNA and protein stability. J Biol Chem 1998;273:14424–9. [88] Law BK, Waltner-Law ME, Entingh AJ, et al. Salicylate-induced Growth Arrest Is Associated with Inhibition of p70s6k and Downregulation of c-Myc, Cyclin D1, Cyclin A, and Proliferating Cell Nuclear Antigen. J Biol Chem 2000;275:38261–7. [89] Feng LX, Ravindranath N, Dym M. Stem cell factor/c-kit upregulates cyclin D3 and promotes cell cycle progression via the phosphoinositide 3-Kinase/p70 S6 kinase pathway in spermatogonia. J Biol Chem 2000;275:25572–6 [in process citation]. [90] Decker T, Hipp S, Ringshausen I, et al. Rapamycin-induced G1 arrest in cycling B-CLL cells is associated with reduced expression of cyclin D3, cyclin E, cyclin A, and survivin. Blood 2003;101:278–85. [91] West MJ, Stoneley M, Willis AE. Translational induction of the cmyc oncogene via activation of the FRAP/TOR signalling pathway. Oncogene 1998;17:769–80. [92] Nelsen CJ, Rickheim DG, Tucker MM, Hansen LK, Albrecht JH. Evidence that cyclin D1 mediates both growth and proliferation downstream of TOR in hepatocytes. J Biol Chem 2003;278:3656–63. [93] Feuerstein N, Huang D, Prystowsky MB. Rapamycin selectively blocks interleukin-2-induced proliferating cell nuclear antigen gene expression in T lymphocyte. Evidence for inhibition of CREB/ATF binding activities. J Biol Chem 1995;270:9454–8. [94] Javier AF, Bata-Csorgo Z, Ellis CN, Kang S, Voorhees JJ, Cooper KD. Rapamycin (sirolimus) inhibits proliferating cell nuclear antigen expression and blocks cell cycle in the G1 phase in human keratinocyte stem cells. J Clin Invest 1997;99:2094–9. [95] Jayaraman T, Marks AR. Rapamycin-FKBP12 blocks proliferation, induces differentiation, and inhibits cdc2 kinase activity in a myogenic cell line. J Biol Chem 1993;268:25385–8. [96] Morice WG, Wiederrecht G, Brunn GJ, Siekierka JJ, Abraham RT. Rapamycin inhibition of interleukin-2-dependent p33cdk2 and p34cdc2 kinase activation in T lymphocytes. J Biol Chem 1993;268:22737–45. [97] Chen Y, Knudsen ES, Wang JY. The RB/p107/p130 phosphorylation pathway is not inhibited in rapamycin-induced G1prolongation of NIH3T3 cells. Oncogene 1996;13:1765–71. [98] Chou MM, Masuda-Robens JM, Gupta ML. Cdc42 promotes G1 progression through p70 S6 kinase-mediated induction of cyclin E expression. J Biol Chem 2003;278:35241–7. [99] Brennan P, Babbage JW, Thomas G, Cantrell D. p70(s6k) integrates phosphatidylinositol 3-kinase and rapamycin-regulated signals for E2F regulation in T lymphocytes. Mol Cell Biol 1999;19:4729–38.
B.K. Law / Critical Reviews in Oncology/Hematology 56 (2005) 47–60 [100] Law BK, Chytil A, Dumont N, et al. Rapamycin potentiates transforming growth factor beta-induced growth arrest in nontransformed, oncogene-transformed, and human cancer cells. Mol Cell Biol 2002;22:8184–98. [101] Nourse J, Firpo E, Flanagan WM, et al. Interleukin-2-mediated elimination of the p27Kip1 cyclin-dependent kinase inhibitor prevented by rapamycin. Nature 1994;372:570–3. [102] Zezula J, Sexl V, Hutter C, Karel A, Schutz W, Freissmuth M. The cyclin-dependent kinase inhibitor p21cip1 mediates the growth inhibitory effect of phorbol esters in human venous endothelial cells. J Biol Chem 1997;272:29967–74. [103] Vinals F, Chambard JC, Pouyssegur J. p70 S6 kinase-mediated protein synthesis is a critical step for vascular endothelial cell proliferation. J Biol Chem 1999;274:26776–82. [104] Gaben AM, Saucier C, Bedin M, Barbu V, Mester J. Rapamycin inhibits cdk4 activation, p21(WAF1/CIP1) expression and G1phase progression in transformed mouse fibroblasts. Int J Cancer 2004;108:200–6. [105] Harper JW, Adami GR, Wei N, Keyomarsi K, Elledge SJ. The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclindependent kinases. Cell 1993;75:805–16. [106] Xiong Y, Hannon GJ, Zhang H, Casso D, Kobayashi R, Beach D. p21 is a universal inhibitor of cyclin kinases. Nature 1993;366:701–4. [107] Elbendary A, Berchuck A, Davis P, et al. Transforming growth factor beta 1 can induce CIP1/WAF1 expression independent of the p53 pathway in ovarian cancer cells. Cell Growth Differ 1994;5:1301–7. [108] Huppi K, Siwarski D, Dosik J, et al. Molecular cloning, sequencing, chromosomal localization and expression of mouse p21 (Waf1). Oncogene 1994;9:3017–20. [109] Polyak K, Kato JY, Solomon MJ, et al. p27Kip1, a cyclin-Cdk inhibitor, links transforming growth factor-beta and contact inhibition to cell cycle arrest. Genes Dev 1994;8:9–22. [110] Toyoshima H, Hunter T. p27, a novel inhibitor of G1 cyclinCdk protein kinase activity, is related to p21. Cell 1994;78:67– 74. [111] Polyak K, Lee MH, Erdjument-Bromage H, et al. Cloning of p27Kip1, a cyclin-dependent kinase inhibitor and a potential mediator of extracellular antimitogenic signals. Cell 1994;78:59–66. [112] LaBaer J, Garrett MD, Stevenson LF, et al. New functional activities for the p21 family of CDK inhibitors. Genes Dev 1997;11:847–62. [113] Zhang H, Hannon GJ, Beach D. p21-containing cyclin kinases exist in both active and inactive states. Genes Dev 1994;8:1750–8. [114] Cheng M, Olivier P, Diehl JA, et al. The p21(Cip1) and p27(Kip1) CDK ’inhibitors’ are essential activators of cyclin D-dependent kinases in murine fibroblasts. EMBO J 1999;18:1571–83. [115] Muraoka RS, Lenferink AE, Law B, et al. ErbB2/Neu-induced, cyclin D1-dependent transformation is accelerated in p27haploinsufficient mammary epithelial cells but impaired in p27-null cells. Mol Cell Biol 2002;22:2204–19. [116] Luo Y, Marx SO, Kiyokawa H, Koff A, Massague J, Marks AR. Rapamycin resistance tied to defective regulation of p27Kip1. Mol Cell Biol 1996;16:6744–51. [117] Soucek T, Yeung RS, Hengstschlager M. Inactivation of the cyclindependent kinase inhibitor p27 upon loss of the tuberous sclerosis complex gene-2. Proc Natl Acad Sci USA 1998;95:15653–8. [118] Bouchard C, Thieke K, Maier A, et al. Direct induction of cyclin D2 by Myc contributes to cell cycle progression and sequestration of p27. EMBO J 1999;18:5321–33. [119] Perez-Roger I, Kim SH, Griffiths B, Sewing A, Land H. Cyclins D1 and D2 mediate myc-induced proliferation via sequestration of p27(Kip1) and p21(Cip1). EMBO J 1999;18:5310–20. [120] Tsutsui T, Hesabi B, Moons DS, et al. Targeted disruption of CDK4 delays cell cycle entry with enhanced p27(Kip1) activity. Mol Cell Biol 1999;19:7011–9.
57
[121] Grewe M, Gansauge F, Schmid RM, Adler G, Seufferlein T. Regulation of cell growth and cyclin D1 expression by the constitutively active FRAP-p70s6K pathway in human pancreatic cancer cells. Cancer Res 1999;59:3581–7. [122] Takuwa N, Fukui Y, Takuwa Y. Cyclin D1 expression mediated by phosphatidylinositol 3-kinase through mTOR-p70(S6K)independent signaling in growth factor-stimulated NIH 3T3 fibroblasts. Mol Cell Biol 1999;19:1346–58. [123] Roberts JM, Sherr CJ. Bared essentials of CDK2 and cyclin E. Nat Genet 2003;35:9–10. [124] Bodner SM, Naeve CW, Rakestraw KM, et al. Cloning and chromosomal localization of the gene encoding human cyclin D-binding Myb-like protein (hDMP1). Gene 1999;229:223–8. [125] Kato J, Matsushime H, Hiebert SW, Ewen ME, Sherr CJ. Direct binding of cyclin D to the retinoblastoma gene product (pRb) and pRb phosphorylation by the cyclin D-dependent kinase CDK4. Genes Dev 1993;7:331–42. [126] Adnane J, Shao Z, Robbins PD. Cyclin D1 associates with the TBP-associated factor TAF(II)250 to regulate Sp1-mediated transcription. Oncogene 1999;18:239–47. [127] Martel RR, Klicius J, Galet S. Inhibition of the immune response by rapamycin, a new antifungal antibiotic. Can J Physiol Pharmacol 1977;55:48–51. [128] Eng CP, Sehgal SN, Vezina C. Activity of rapamycin (AY-22, 989) against transplanted tumors. J Antibiot (Tokyo) 1984;37:1231–7. [129] Dumont FJ, Staruch MJ, Koprak SL, Melino MR, Sigal NH. Distinct mechanisms of suppression of murine T cell activation by the related macrolides FK-506 and rapamycin. J Immunol 1990;144:251–8. [130] Wicker LS, Boltz Jr RC, Matt V, Nichols EA, Peterson LB, Sigal NH. Suppression of B cell activation by cyclosporin A, FK506 and rapamycin. Eur J Immunol 1990;20:2277–83. [131] Kay JE, Kromwel L, Doe SE, Denyer M. Inhibition of T and B lymphocyte proliferation by rapamycin. Immunology 1991;72:544–9. [132] Woltman AM, van der Kooij SW, Coffer PJ, Offringa R, Daha MR, van Kooten C. Rapamycin specifically interferes with GMCSF signaling in human dendritic cells, leading to apoptosis via increased p27KIP1 expression. Blood 2003;101:1439–45. [133] Dumont FJ, Melino MR, Staruch MJ, Koprak SL, Fischer PA, Sigal NH. The immunosuppressive macrolides FK-506 and rapamycin act as reciprocal antagonists in murine T cells. J Immunol 1990;144:1418–24. [134] Fruman DA, Klee CB, Bierer BE, Burakoff SJ. Calcineurin phosphatase activity in T lymphocytes is inhibited by FK 506 and cyclosporin A. Proc Natl Acad Sci USA 1992;89:3686–90. [135] Liu J, Albers MW, Wandless TJ, et al. Inhibition of T cell signaling by immunophilin–ligand complexes correlates with loss of calcineurin phosphatase activity. Biochemistry 1992;31:3896–901. [136] Kuo CJ, Chung J, Fiorentino DF, Flanagan WM, Blenis J, Crabtree GR. Rapamycin selectively inhibits interleukin-2 activation of p70 S6 kinase. Nature 1992;358:70–3. [137] Calvo V, Crews CM, Vik TA, Bierer BE. Interleukin 2 stimulation of p70 S6 kinase activity is inhibited by the immunosuppressant rapamycin. Proc Natl Acad Sci USA 1992;89:7571–5. [138] O’Keefe SJ, Tamura J, Kincaid RL, Tocci MJ, O’Neill EA. FK506- and CsA-sensitive activation of the interleukin-2 promoter by calcineurin. Nature 1992;357:692–4. [139] Peng T, Golub TR, Sabatini DM. The immunosuppressant rapamycin mimics a starvation-like signal distinct from amino acid and glucose deprivation. Mol Cell Biol 2002;22:5575–84. [140] Luan FL, Hojo M, Maluccio M, Yamaji K, Suthanthiran M. Rapamycin blocks tumor progression: unlinking immunosuppression from antitumor efficacy. Transplantation 2002;73:1565–72. [141] Stegall MD, Larson TS, Prieto M, et al. Kidney transplantation without calcineurin inhibitors using sirolimus. Transplant Proc 2003;35:125S–7S.
58
B.K. Law / Critical Reviews in Oncology/Hematology 56 (2005) 47–60
[142] Nair S, Eason J, Loss G. Sirolimus monotherapy in nephrotoxicity due to calcineurin inhibitors in liver transplant recipients. Liver Transplant 2003;9:126–9. [143] Flechner SM. Minimizing calcineurin inhibitor drugs in renal transplantation. Transplant Proc 2003;35:118S–21S. [144] Flechner SM, Goldfarb D, Modlin C, et al. Kidney transplantation without calcineurin inhibitor drugs: a prospective, randomized trial of sirolimus versus cyclosporine. Transplantation 2002;74: 1070–6. [145] Reis A, Megahed M, Reinhard T, Godehardt E, Braunstein C, Sundmacher R. Synergism of RAD and cyclosporin A in prevention of acute rat corneal allograft rejection. Cornea 2002;21:81–4. [146] Sherr CJ. Cancer cell cycles. Science 1996;274:1672–7. [147] Seufferlein T, Rozengurt E. Rapamycin inhibits constitutive p70s6k phosphorylation, cell proliferation, and colony formation in small cell lung cancer cells. Cancer Res 1996;56:3895–7. [148] Moore SM, Rintoul RC, Walker TR, Chilvers ER, Haslett C, Sethi T. The presence of a constitutively active phosphoinositide 3-kinase in small cell lung cancer cells mediates anchorage-independent proliferation via a protein kinase B and p70s6k-dependent pathway. Cancer Res 1998;58:5239–47. [149] Couch FJ, Wang XY, Wu GJ, Qian J, Jenkins RB, James CD. Localization of PS6K to chromosomal region 17q23 and determination of its amplification in breast cancer. Cancer Res 1999;59:1408–11. [150] Barlund M, Forozan F, Kononen J, et al. Detecting activation of ribosomal protein S6 kinase by complementary DNA and tissue microarray analysis. J Natl Cancer Inst 2000;92:1252–9. [151] Dilling MB, Germain GS, Dudkin L, et al. 4E-binding proteins, the suppressors of eukaryotic initiation factor 4E, are down-regulated in cells with acquired or intrinsic resistance to rapamycin. J Biol Chem 2002;277:13907–17. [152] Rosenwald IB, Lazaris-Karatzas A, Sonenberg N, Schmidt EV. Elevated levels of cyclin D1 protein in response to increased expression of eukaryotic initiation factor 4E. Mol Cell Biol 1993;13:7358–63. [153] De Benedetti A, Harris AL. eIF4E expression in tumors: its possible role in progression of malignancies. Int J Biochem Cell Biol 1999;31:59–72. [154] Rosenwald IB, Chen JJ, Wang S, Savas L, London IM, Pullman J. Upregulation of protein synthesis initiation factor eIF-4E is an early event during colon carcinogenesis. Oncogene 1999;18:2507–17. [155] Li BD, Liu L, Dawson M, De Benedetti A. Overexpression of eukaryotic initiation factor 4E (eIF4E) in breast carcinoma. Cancer 1997;79:2385–90. [156] Li BD, McDonald JC, Nassar R, De Benedetti A. Clinical outcome in stage I to III breast carcinoma and eIF4E overexpression. Ann Surg 1998;227:756–61 [61–3; discussion]. [157] Nathan CA, Liu L, Li BD, Abreo FW, Nandy I, De Benedetti A. Detection of the proto-oncogene eIF4E in surgical margins may predict recurrence in head and neck cancer. Oncogene 1997;15:579–84. [158] Smith MR, Jaramillo M, Liu YL, et al. Translation initiation factors induce DNA synthesis and transform NIH 3T3 cells. New Biol 1990;2:648–54. [159] Rousseau D, Gingras AC, Pause A, Sonenberg N. The eIF4Ebinding proteins 1 and 2 are negative regulators of cell growth. Oncogene 1996;13:2415–20. [160] Miyagi Y, Sugiyama A, Asai A, Okazaki T, Kuchino Y, Kerr SJ. Elevated levels of eukaryotic translation initiation factor eIF-4E, mRNA in a broad spectrum of transformed cell lines. Cancer Lett 1995;91:247–52. [161] Green AJ, Smith M, Yates JR. Loss of heterozygosity on chromosome 16p13.3 in hamartomas from tuberous sclerosis patients. Nat Genet 1994;6:193–6. [162] Green AJ, Johnson PH, Yates JR. The tuberous sclerosis gene on chromosome 9q34 acts as a growth suppressor. Hum Mol Genet 1994;3:1833–4.
[163] Sampson JR, Harris PC. The molecular genetics of tuberous sclerosis. Hum Mol Genet 1994;3(Spec No):1477–80. [164] Cairns P, Okami K, Halachmi S, et al. Frequent inactivation of PTEN/MMAC1 in primary prostate cancer. Cancer Res 1997;57:4997–5000. [165] Dahia PL, Marsh DJ, Zheng Z, et al. Somatic deletions and mutations in the Cowden disease gene, PTEN, in sporadic thyroid tumors. Cancer Res 1997;57:4710–3. [166] Wang SI, Puc J, Li J, et al. Somatic mutations of PTEN in glioblastoma multiforme. Cancer Res 1997;57:4183–6. [167] Liu W, James CD, Frederick L, Alderete BE, Jenkins RB. PTEN/MMAC1 mutations and EGFR amplification in glioblastomas. Cancer Res 1997;57:5254–7. [168] Cairns P, Evron E, Okami K, et al. Point mutation and homozygous deletion of PTEN/MMAC1 in primary bladder cancers. Oncogene 1998;16:3215–8. [169] Kim SK, Su LK, Oh Y, Kemp BL, Hong WK, Mao L. Alterations of PTEN/MMAC1, a candidate tumor suppressor gene, and its homologue, PTH2, in small cell lung cancer cell lines. Oncogene 1998;16:89–93. [170] Kohno T, Takahashi M, Manda R, Yokota J. Inactivation of the PTEN/MMAC1/TEP1 gene in human lung cancers. Genes Chromosomes Cancer 1998;22:152–6. [171] Levine RL, Cargile CB, Blazes MS, van Rees B, Kurman RJ, Ellenson LH. PTEN mutations and microsatellite instability in complex atypical hyperplasia, a precursor lesion to uterine endometrioid carcinoma. Cancer Res 1998;58:3254–8. [172] Obata K, Morland SJ, Watson RH, et al. Frequent PTEN/MMAC mutations in endometrioid but not serous or mucinous epithelial ovarian tumors. Cancer Res 1998;58:2095–7. [173] Aveyard JS, Skilleter A, Habuchi T, Knowles MA. Somatic mutation of PTEN in bladder carcinoma. Br J Cancer 1999;80:904–8. [174] Alimov A, Li C, Gizatullin R, et al. Somatic mutation and homozygous deletion of PTEN/MMAC1 gene of 10q23 in renal cell carcinoma. Anticancer Res 1999;19:3841–6. [175] Di Cristofano A, Pesce B, Cordon-Cardo C, Pandolfi PP. Pten is essential for embryonic development and tumour suppression. Nat Genet 1998;19:348–55. [176] Suzuki A, de la Pompa JL, Stambolic V, et al. High cancer susceptibility and embryonic lethality associated with mutation of the PTEN tumor suppressor gene in mice. Curr Biol 1998;8:1169– 78. [177] Podsypanina K, Ellenson LH, Nemes A, et al. Mutation of Pten/Mmac1 in mice causes neoplasia in multiple organ systems. Proc Natl Acad Sci USA 1999;96:1563–8. [178] Stambolic V, Tsao MS, Macpherson D, Suzuki A, Chapman WB, Mak TW. High incidence of breast and endometrial neoplasia resembling human Cowden syndrome in pten+/− mice. Cancer Res 2000;60:3605–11. [179] Kwabi-Addo B, Giri D, Schmidt K, et al. Haploinsufficiency of the Pten tumor suppressor gene promotes prostate cancer progression. Proc Natl Acad Sci USA 2001;98:11563–8. [180] Li G, Robinson GW, Lesche R, et al. Conditional loss of PTEN leads to precocious development and neoplasia in the mammary gland. Development 2002;129:4159–70. [181] Trotman LC, Niki M, Dotan ZA, et al. Pten dose dictates cancer progression in the prostate. PLoS Biol 2003;1:59. [182] Hemminki A, Tomlinson I, Markie D, et al. Localization of a susceptibility locus for Peutz–Jeghers syndrome to 19p using comparative genomic hybridization and targeted linkage analysis. Nat Genet 1997;15:87–90. [183] Hemminki A, Markie D, Tomlinson I, et al. A serine/threonine kinase gene defective in Peutz–Jeghers syndrome. Nature 1998;391:184–7. [184] Wang ZJ, Churchman M, Avizienyte E, et al. Germline mutations of the LKB1 (STK11) gene in Peutz–Jeghers patients. J Med Genet 1999;36:365–8.
B.K. Law / Critical Reviews in Oncology/Hematology 56 (2005) 47–60 [185] Su GH, Hruban RH, Bansal RK, et al. Germline and somatic mutations of the STK11/LKB1 Peutz–Jeghers gene in pancreatic and biliary cancers. Am J Pathol 1999;154:1835–40. [186] Guldberg P, thor Straten P, Ahrenkiel V, Seremet T, Kirkin AF, Zeuthen J. Somatic mutation of the Peutz–Jeghers syndrome gene, LKB1/STK11, in malignant melanoma. Oncogene 1999;18:1777–80. [187] Rowan A, Bataille V, MacKie R, et al. Somatic mutations in the Peutz–Jeghers (LKB1/STKII) gene in sporadic malignant melanomas. J Invest Dermatol 1999;112:509–11. [188] Wang ZJ, Churchman M, Campbell IG, et al. Allele loss and mutation screen at the Peutz–Jeghers (LKB1) locus (19p13.3) in sporadic ovarian tumours. Br J Cancer 1999;80:70–2. [189] Sanchez-Cespedes M, Parrella P, Esteller M, et al. Inactivation of LKB1/STK11 is a common event in adenocarcinomas of the lung. Cancer Res 2002;62:3659–62. [190] Carretero J, Medina PP, Pio R, Montuenga LM, Sanchez-Cespedes M. Novel and natural knockout lung cancer cell lines for the LKB1/STK11 tumor suppressor gene. Oncogene 2004;23:4037– 40. [191] Esteller M, Avizienyte E, Corn PG, et al. Epigenetic inactivation of LKB1 in primary tumors associated with the Peutz–Jeghers syndrome. Oncogene 2000;19:164–8. [192] Shen Z, Wen XF, Lan F, Shen ZZ, Shao ZM. The tumor suppressor gene LKB1 is associated with prognosis in human breast carcinoma. Clin Cancer Res 2002;8:2085–90. [193] Nakau M, Miyoshi H, Seldin MF, Imamura M, Oshima M, Taketo MM. Hepatocellular carcinoma caused by loss of heterozygosity in Lkb1 gene knockout mice. Cancer Res 2002;62:4549–53. [194] Houchens DP, Ovejera AA, Riblet SM, Slagel DE. Human brain tumor xenografts in nude mice as a chemotherapy model. Eur J Cancer Clin Oncol 1983;19:799–805. [195] Dudkin L, Dilling MB, Cheshire PJ, et al. Biochemical correlates of mTOR inhibition by the rapamycin ester CCI-779 and tumor growth inhibition. Clin Cancer Res 2001;7:1758–64. [196] Geoerger B, Kerr K, Tang CB, et al. Antitumor activity of the rapamycin analog CCI-779 in human primitive neuroectodermal tumor/medulloblastoma models as single agent and in combination chemotherapy. Cancer Res 2001;61:1527–32. [197] Yu K, Toral-Barza L, Discafani C, et al. mTOR, a novel target in breast cancer: the effect of CCI-779, an mTOR inhibitor, in preclinical models of breast cancer. Endocr Relat Cancer 2001;8:249– 58. [198] Neshat MS, Mellinghoff IK, Tran C, et al. Enhanced sensitivity of PTEN-deficient tumors to inhibition of FRAP/mTOR. Proc Natl Acad Sci USA 2001;98:10314–9. [199] Podsypanina K, Lee RT, Politis C, et al. An inhibitor of mTOR reduces neoplasia and normalizes p70/S6 kinase activity in Pten+/− mice. Proc Natl Acad Sci U S A 2001;98:10320–5. [200] Grunwald V, DeGraffenried L, Russel D, Friedrichs WE, Ray RB, Hidalgo M. Inhibitors of mTOR reverse doxorubicin resistance conferred by PTEN status in prostate cancer cells. Cancer Res 2002;62:6141–5. [201] Shi Y, Gera J, Hu L, et al. Enhanced sensitivity of multiple myeloma cells containing PTEN mutations to CCI-779. Cancer Res 2002;62:5027–34. [202] Gera JF, Mellinghoff IK, Shi Y, et al. AKT activity determines sensitivity to mammalian target of rapamycin (mTOR) inhibitors by regulating cyclin D1 and c-myc expression. J Biol Chem 2004;279:2737–46. [203] Penuel E, Martin GS. Transformation by v-Src: Ras-MAPK and PI3K-mTOR mediate parallel pathways. Mol Biol Cell 1999;10:1693–703. [204] Louro ID, McKie-Bell P, Gosnell H, Brindley BC, Bucy RP, Ruppert JM. The zinc finger protein GLI induces cellular sensitivity to the mTOR inhibitor rapamycin. Cell Growth Differ 1999;10:503–16.
59
[205] Aoki M, Blazek E, Vogt PK. A role of the kinase mTOR in cellular transformation induced by the oncoproteins P3k and Akt. Proc Natl Acad Sci USA 2001;98:136–41. [206] Nomura M, He Z, Koyama I, Ma WY, Miyamoto K, Dong Z. Involvement of the Akt/mTOR pathway on EGF-induced cell transformation. Mol Carcinog 2003;38:25–32. [207] Boulay A, Zumstein-Mecker S, Stephan C, et al. Antitumor efficacy of intermittent treatment schedules with the rapamycin derivative RAD001 correlates with prolonged inactivation of ribosomal protein S6 kinase 1 in peripheral blood mononuclear cells. Cancer Res 2004;64:252–61. [208] Peralba JM, DeGraffenried L, Friedrichs W, et al. Pharmacodynamic Evaluation of CCI-779, an Inhibitor of mTOR, in Cancer Patients. Clin Cancer Res 2003;9:2887–92. [209] Raymond E, Alexandre J, Faivre S, et al. Safety and pharmacokinetics of escalated doses of weekly intravenous infusion of CCI-779, a novel mTOR inhibitor, in patients with cancer. J Clin Oncol 2004;22:2336–47. [210] Atkins MB, Hidalgo M, Stadler WM, et al. Randomized phase II study of multiple dose levels of CCI-779, a novel mammalian target of rapamycin kinase inhibitor, in patients with advanced refractory renal cell carcinoma. J Clin Oncol 2004;22:909–18. [211] Guba M, von Breitenbuch P, Steinbauer M, et al. Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor. Nat Med 2002;8:128–35. [212] Knighton D, Ausprunk D, Tapper D, Folkman J. Avascular and vascular phases of tumour growth in the chick embryo. Br J Cancer 1977;35:347–56. [213] Yu Y, Sato JD. MAP kinases, phosphatidylinositol 3-kinase, and p70 S6 kinase mediate the mitogenic response of human endothelial cells to vascular endothelial growth factor. J Cell Physiol 1999;178:235–46. [214] Zhong H, Chiles K, Feldser D, et al. Modulation of hypoxiainducible factor 1alpha expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics. Cancer Res 2000;60:1541–5. [215] Treins C, Giorgetti-Peraldi S, Murdaca J, Semenza GL, Van E. Obberghen Insulin stimulates hypoxia-inducible factor 1 through a phosphatidylinositol 3-kinase/target of rapamycin-dependent signaling pathway. J Biol Chem 2002;277:27975–81. [216] Mayerhofer M, Valent P, Sperr WR, Griffin JD, Sillaber C. BCR/ABL induces expression of vascular endothelial growth factor and its transcriptional activator, hypoxia inducible factor1alpha, through a pathway involving phosphoinositide 3-kinase and the mammalian target of rapamycin. Blood 2002;100:3767– 75. [217] Hudson CC, Liu M, Chiang GG, et al. Regulation of hypoxiainducible factor 1alpha expression and function by the mammalian target of rapamycin. Mol Cell Biol 2002;22:7004–14. [218] Marx SO, Marks AR. Bench to bedside: the development of rapamycin and its application to stent restenosis. Circulation 2001;104:852–5. [219] Sousa JE, Costa MA, Abizaid A, et al. Lack of neointimal proliferation after implantation of sirolimus-coated stents in human coronary arteries: a quantitative coronary angiography and threedimensional intravascular ultrasound study. Circulation 2001;103: 192–5. [220] Grube E, Sonoda S, Ikeno F, et al. Six- and twelve-month results from first human experience using everolimus-eluting stents with bioabsorbable polymer. Circulation 2004;109:2168–71. [221] Law BK, Norgaard P, Moses HL. Farnesyltransferase inhibitor induces rapid growth arrest and blocks p70s6k activation by multiple stimuli. J Biol Chem 2000;275:10796–801. [222] Law BK, Norgaard P, Gnudi L, Kahn BB, Poulson HS, Moses HL. Inhibition of DNA synthesis by a farnesyltransferase inhibitor
60
[223]
[224]
[225]
[226]
[227]
[228]
B.K. Law / Critical Reviews in Oncology/Hematology 56 (2005) 47–60 involves inhibition of the p70(s6k) pathway. J Biol Chem 1999;274:4743–8. Castro AF, Rebhun JF, Clark GJ, Quilliam LA. Rheb binds tuberous sclerosis complex 2 (TSC2) and promotes S6 kinase activation in a rapamycin- and farnesylation-dependent manner. J Biol Chem 2003;278:32493–6. Iiboshi Y, Papst PJ, Hunger SP, Terada N. l-Asparaginase inhibits the rapamycin-targeted signaling pathway. Biochem Biophys Res Commun 1999;260:534–9. Ly C, Arechiga AF, Melo JV, Walsh CM, Ong ST. Bcr–Abl kinase modulates the translation regulators ribosomal protein S6 and 4E-BP1 in chronic myelogenous leukemia cells via the mammalian target of rapamycin. Cancer Res 2003;63:5716– 22. Mohi MG, Boulton C, Gu TL, et al. Combination of rapamycin and protein tyrosine kinase (PTK) inhibitors for the treatment of leukemias caused by oncogenic PTKs. Proc Natl Acad Sci USA 2004;101:3130–5. Fukazawa H, Uehara Y. U0126 reverses Ki-ras-mediated transformation by blocking both mitogen-activated protein kinase and p70 S6 kinase pathways. Cancer Res 2000;60:2104–7. Eshleman JS, Carlson BL, Mladek AC, Kastner BD, Shide KL, Sarkaria JN. Inhibition of the mammalian target of rapamycin sen-
sitizes U87 xenografts to fractionated radiation therapy. Cancer Res 2002;62:7291–7. [229] Shi Y, Frankel A, Radvanyi LG, Penn LZ, Miller RG, Mills GB. Rapamycin enhances apoptosis and increases sensitivity to cisplatin in vitro. Cancer Res 1995;55:1982–8. [230] van der Poel HG, Hanrahan C, Zhong H, Simons JW. Rapamycin induces Smad activity in prostate cancer cell lines. Urol Res 2003;30:380–6. [231] Li Y, Inoki K, Vacratsis P, Guan KL. The p38 and MK2 kinase cascade phosphorylates tuberin, the tuberous sclerosis 2 gene product, and enhances its interaction with 14-3-3. J Biol Chem 2003;278:13663–71.
Biography Brian Law obtained his Ph.D. in biochemistry in 1996 from Purdue University, West Lafayette, IN, USA. He is currently an assistant professor in the Department of Pharmacology and Therapeutics at the University of Florida, Gainesville, FL, USA.