The mammalian target of rapamycin (mTOR) is a key signaling node coordinating cell cycle progression and cell growth in response to genetic, epigenetic, and environmental conditions. Pathways involved in mTOR signaling are dysregulated in precancerous human tissues. These findings, together with the intriguing possibility that mTOR suppression may be associated with antitumor actions of caloric restriction, suggest that mTOR signaling may be an important target for chemopreventive drugs. (Cancer Epidemiol Biomarkers Prev 2007;16(7):1330–40)

The National Cancer Institute, Division of Cancer Prevention has recently ublished a series of reviews on mechanism-based targets for cancer-preventive intervention including peroxisome proliferator–activated receptors (1), inducible nitric oxide synthase (2), and epigenetic modulators, primarily histone deacetylases and DNA methyl transferases (3, 4). Here, the potential of mammalian target of rapamycin (mTOR) signaling in chemoprevention strategies is reviewed.

mTOR is the target of rapamycin, a macrolide antibiotic and immunosuppressant of the phosphoinositide kinase family. It is a component of two protein complexes, mTORC1 and mTORC2. mTORC1 consists of mTOR, mLST8, and raptor (regulatory-associated protein of mTOR), and mTORC2 consists of mTOR, mLST8, mSIN1 (mitogen-activated protein kinase–associated protein 1), and rictor (rapamycin insensitive companion of mTOR; refs. 5, 6). Rapamycin bound to FKBP12 inhibits mTORC1, but, with important exceptions described below, not mTORC2 (6). mTOR is a serine-threonine kinase with lipid kinase activity. Its signaling is activated when the genetic and environmental milieu is optimal for cellular growth, and diminishes under stressful conditions including insufficient nutrients, energy, or growth factors, as well as DNA damage (refs. 7-13; Fig. 1). Thus, TOR protein is essential for cell growth and development and is involved in regulating cell cycle progression, cell size, cell migration, and survival; it also negatively governs autophagy, wherein proteins and organelles are degraded during nutrient deprivation (8-13). Disruption of the gene encoding TOR is lethal in all species.

Figure 1.

mTOR functions as a central control protein integrating signals from a host of environmental factors, including amino acids, energy, hormones, growth factors, cytokines, and other stress factors.

Figure 1.

mTOR functions as a central control protein integrating signals from a host of environmental factors, including amino acids, energy, hormones, growth factors, cytokines, and other stress factors.

Close modal

mTOR regulates both transcription of genes relevant to carcinogenesis, including, for example, hypoxia inducible factor (HIF)-1α (14-17), and activity of the procarcinogenic phosphotidylinositol 3-kinase (PI3K)/AKT pathway (18, 19). As described below, mTOR activity mediates AKT-activated cell proliferation and survival; however, one of mTOR effectors, ribosomal protein S6 kinase 1 (S6K1), is a feedback inhibitor of insulin- and insulin-like growth factor (IGF)–induced PI3K activation. S6K1, stimulated by activated mTOR, phosphorylates insulin receptor substrate proteins, inhibiting their function, which in turn diminishes signaling through the PI3K/AKT pathway (reviewed in refs. 19, 20). Thus, cells regulated by this mechanism can become resistant to mTOR inhibition as S6K1 production decreases and AKT rebounds (5, 19, 21). An additional complexity is the finding that mTORC2 may also participate directly in activation of AKT (refs. 5, 6; Fig. 2), and so mTOR inhibitors may reduce AKT activity regardless of losing feedback inhibition by S6K1. Based on studies in cancer cell lines and in leukemia patients treated with rapamycin, Sabatini et al. (5, 6) have suggested that where rapamycin is effective against PI3K/AKT signaling, it interferes with mTORC2 assembly and function.

Figure 2.

Cancer preventive modulation of mTOR signaling pathways and targets for cancer preventive intervention. Upstream of mTOR: Growth factors and hormones signal to mTOR by activating the PI3K/AKT/PTEN axis. Lipid products of PI3K activate AKT via mTORC2 complex. Activated AKT phosphorylates and inactivates TSC2, thereby allowing signaling (which occurs via mTORC1 complex). PTEN dephosphorylates lipid products generated by PI3K, down-regulating PI3K signaling. Rheb functions as a positive regulator of mTORC1; it is inactivated by TSC GTPase activating protein function. Amino acids and stress activate mTORC1 independently of PI3K/AKT. TSC proteins and Rheb are involved, but the mechanisms are largely unknown. In energy-deprived cells, AMPK phosphorylates and increases TSC2 inhibition of mTORC1. LKB1 phosphorylates and activates AMPK. In energy-deprived cells, LKB1 is required for mTORC1 suppression, which is dependent on AMPK and TSC2. Downstream of mTOR: mTORC1 directly or indirectly activates S6K1, which regulates ribosomal protein translation and ribosome biogenesis. S6K1 also is a feedback inhibitor of PI3K/AKT signaling by blocking insulin receptor substrate. mTORC1 also directly phosphorylates and inactivates 4E-BP1, decreasing its binding affinity for eIF-4E, which leads to increased translation of cap-dependent mRNAs. Known tumor suppressors that negatively regulate mTOR pathways appear in yellow boxes. Dashed lines, unknown/uncertain pathways. Targets for cancer preventive intervention (see text and Table 2): In addition to direct inhibition of mTOR by rapamycins and a few other compounds, other targets upstream on mTOR signaling pathways are affected by known chemopreventive agents. Examples shown on this figure in lavender boxes include inhibitors of protein kinase–mediated cell proliferation signaling [ornithine decarboxylase (ODC) and epidermal growth factor (EGF)/epidermal growth factor receptor inhibitors (EGFR)], activators of tumor suppressor (PTEN) gene transcription (DNA methyl transferase, histone deacetylase, and histone acetyltransferase inhibitors), direct inhibitors of PI3K/AKT signaling, and activators of AMPK.

Figure 2.

Cancer preventive modulation of mTOR signaling pathways and targets for cancer preventive intervention. Upstream of mTOR: Growth factors and hormones signal to mTOR by activating the PI3K/AKT/PTEN axis. Lipid products of PI3K activate AKT via mTORC2 complex. Activated AKT phosphorylates and inactivates TSC2, thereby allowing signaling (which occurs via mTORC1 complex). PTEN dephosphorylates lipid products generated by PI3K, down-regulating PI3K signaling. Rheb functions as a positive regulator of mTORC1; it is inactivated by TSC GTPase activating protein function. Amino acids and stress activate mTORC1 independently of PI3K/AKT. TSC proteins and Rheb are involved, but the mechanisms are largely unknown. In energy-deprived cells, AMPK phosphorylates and increases TSC2 inhibition of mTORC1. LKB1 phosphorylates and activates AMPK. In energy-deprived cells, LKB1 is required for mTORC1 suppression, which is dependent on AMPK and TSC2. Downstream of mTOR: mTORC1 directly or indirectly activates S6K1, which regulates ribosomal protein translation and ribosome biogenesis. S6K1 also is a feedback inhibitor of PI3K/AKT signaling by blocking insulin receptor substrate. mTORC1 also directly phosphorylates and inactivates 4E-BP1, decreasing its binding affinity for eIF-4E, which leads to increased translation of cap-dependent mRNAs. Known tumor suppressors that negatively regulate mTOR pathways appear in yellow boxes. Dashed lines, unknown/uncertain pathways. Targets for cancer preventive intervention (see text and Table 2): In addition to direct inhibition of mTOR by rapamycins and a few other compounds, other targets upstream on mTOR signaling pathways are affected by known chemopreventive agents. Examples shown on this figure in lavender boxes include inhibitors of protein kinase–mediated cell proliferation signaling [ornithine decarboxylase (ODC) and epidermal growth factor (EGF)/epidermal growth factor receptor inhibitors (EGFR)], activators of tumor suppressor (PTEN) gene transcription (DNA methyl transferase, histone deacetylase, and histone acetyltransferase inhibitors), direct inhibitors of PI3K/AKT signaling, and activators of AMPK.

Close modal

Many inputs that signal to mTOR converge on the tuberous sclerosis complex (TSC) of tumor suppressor protein hamartin, encoded by the TSC1 gene, and tuberin, encoded by the TSC2 genes. The small GTP-binding protein Ras homologue enriched in brain (Rheb), which can bind directly to and up-regulate mTOR (22), is inactivated by the GTPase activating protein activity of TSC (9-12, 23). Tumor suppressors phosphatase and tensin homologue (PTEN; associated with Cowden disease) and LKB1 (associated with Peutz-Jeghers syndrome; ref. 24) also down-regulate mTOR via mTORC2 and mTORC1, respectively, and their loss is similarly associated with autosomal dominant hamartoma syndromes.

The best characterized targets of mTOR phosphorylation are two families of proteins that control translation, ribosomal protein S6 kinases (S6K1 and S6K2 in mammals) and eukaryotic initiation factor 4E (eIF-4E)–binding protein 1 (4E-BP1). In mTORC1, raptor may act as a scaffolding protein, linking mTOR to S6K1 and 4E-BP1, and has generally been reported as a positive regulator of mTOR (reviewed in ref. 11). S6K1 is activated by phosphorylation and regulates ribosomal protein translation and ribosome biogenesis (9-11). The role of S6K1 in feedback inhibition of AKT was described above, and other studies in S6K-deficient organisms have established its importance in the control of cell and organism growth (25, 26).

Phosphorylation by mTOR inactivates 4E-BP1. In quiescent cells, unphosphorylated 4E-BP1 binds and inhibits eIF-4E, which is present in rate-limiting quantities relative to other components of the translational apparatus and is a key regulatory factor for protein synthesis. eIF-4E binds to the 7-methylguanosine–containing cap of mRNA and participates in the transfer of mRNA to the 40S ribosomal subunit. mTOR phosphorylation decreases the binding affinity of 4E-BP1 for eIF-4E, which leads to increased translation of cap-dependent mRNAs (9-11).

mTOR as a Hormone and Growth Factor Sensor

Although the mechanisms are not yet completely understood, effects of hormones and growth factors seem to be mediated initially by mTORC2 (5). Besides insulin and IGF, epidermal growth factor and platelet-derived growth factor interact with mTORC2 primarily by activating PI3K. PI3K activation is blocked by PTEN as well as by S6K1 (9, 10, 23, 24); PTEN dephosphorylates lipid products generated by PI3K, down-regulating PI3K signaling (Fig. 2). The lipid product of PI3K localizes AKT to the plasma membrane, where it is phosphorylated and activated by 3-phosphoinositide–dependent protein kinase 1 and mTORC2 (5). AKT may then activate mTORC1 signaling, largely by directly phosphorylating and inactivating TSC2. In addition to signaling via PI3K/AKT, growth factors activate mTOR by a pathway involving phosphatidic acid and phospholipase D1 (27-29).

mTOR as a Nutrient and Energy Sensor

mTORC1-mediated activity is positively regulated by the level of intracellular amino acids (8, 10-12, 23). The branched-chain amino acid leucine, principal indicator of amino acid supply in mammals (30), is an effective regulator of mTOR activity in most cell types (8, 31-34); TSC proteins (35, 36) and Rheb (37-39) have also been implicated in the nutrient-sensing branch of the mTOR pathway. However, molecular events involved in nutrient sensing remain largely unknown (10-12, 23).

Molecular mechanisms involved in mTORC1-mediated energy sensing have been associated with both TSC and LKB1. Although initial studies suggested mTOR as the direct cellular sensor of ATP levels (40), increasing evidence implicates AMP-activated protein kinase A (AMPK) in regulation of mTOR activity. A more sensitive indicator of cellular energy status than ATP, AMP increases when the ATP/ADP ratio decreases, and a high ratio of AMP/ATP signals that the energy status of the cell is compromised (41). AMP-stimulated AMPK activity is sensitive to very small changes in intracellular AMP levels (41). In energy-deprived cells, AMPK directly phosphorylates and enhances the ability of TSC2 to inhibit mTORC1 signaling (42). LKB1 can directly phosphorylate and activate AMPK (43-45), and under conditions of energy stress, LKB1 is required for mTORC1 suppression, which is dependent on AMPK and TSC (46). AMPK can also directly phosphorylate mTORC1 under conditions of energy stress (47). Additional stress-induced mechanisms of mTOR regulation have been described (12, 17, 24, 48-51).

Knowledge about consequences of dysregulated mTOR signaling for tumorigenesis comes mostly from studies in which mTOR has been pharmacologically disrupted by rapamycin and its analogues CCI-779, RAD-001, and AP23573 (5, 13, 48). In therapeutic models, these selective mTOR inhibitors suppress growth of a diverse range of cancer types, with their effectiveness apparently depending on concomitant blockage of PI3K/AKT signaling (48, 52, 53). Although their immediate utility for long-term cancer prevention is not yet established (see Strategies for the Development of mTOR Inhibitors in Clinical Prevention Studies), rapamycins also seem to be efficacious in initial prevention proof-of-principle experiments (discussed in mTOR Inhibition to Prevent Cancer).

eIF-4E seems to be the most crucial downstream effector of mTOR-associated carcinogenesis (54). Overexpression of S6K in rat cells induces morphologic changes but does not lead to oncogenic transformation (55), and S6K1 alterations are rare in human cancers (9). On the other hand, overexpression of eIF-4E alone or in combination with other oncogenes transforms cells in vitro (reviewed in ref. 54). In transgenic mice, overexpression of eIF-4E significantly enhances transformation in cooperation with c-Myc, albeit with a long time lag (56, 57).

mTOR inhibition affects tumorigenesis by slowing or arresting cells in the G1 phase of the cell cycle, promoting apoptosis, and affecting angiogenesis pathways (9, 10, 23, 58). The effects on cell cycle progression are mediated, at least in part, by blocking the ability of eIF-4E to enhance translation of mRNAs encoding positive cell cycle progression regulators, such as cyclin D1 and ornithine decarboxylase, and to inhibit translation of negative regulators, such as cyclin-dependent kinase inhibitors (9, 54). Although the effects of rapamycins are often cytostatic, they can also induce apoptosis in cell lines (59, 60) and in vivo in AKT-dependent precancerous prostatic mouse tissue (17). In AKT-dependent mouse lymphomas, rapamycin restored apoptotic response to cytotoxic agents, which was then reversed by eIF-4E expression; rapamycin alone was minimally effective in this system (56).

Rapamycins also exert antiangiogenic effects in vivo, in conjunction with decreased production of proangiogenic vascular endothelial growth factor (VEGF; refs. 58, 61, 62), which, along with its receptor, is controlled by HIF-1α (63). These findings are consistent with studies showing that PI3K/AKT/mTOR signaling can up-regulate HIF-1α–dependent responses in hypoxic cells (16) and precancerous lesions in vivo (17).

Varying levels of AKT activity may also explain the differences in malignancy potential of tumors arising in subjects with hamartoma syndromes. Unlike cells lacking PTEN, in which AKT is constitutively active, TSC1/2- and LKB1-deficient cells show diminution of AKT activation, which is attributed to higher mTOR activity and S6K feedback inhibition of the PI3K/AKT pathway (reviewed in ref. 64). Malignant tumors are common in Cowden disease (associated with mutated PTEN) but less frequent in TSC and Peutz-Jeghers syndrome (associated with mutations in TSC1/2 and LKB1, respectively). It is unknown whether losing S6K1 feedback inhibition of PI3K/AKT will eventually promote malignant progression in those with benign hamartoma syndromes. However, as rapamycin prevents development of macroscopic renal lesions in a TSC rat model (65), and activation of eIF-4E rather than S6K may be responsible for oncogenic actions of aberrant mTOR signaling, perhaps mTOR inhibition will obstruct signaling pathways promoting tumor growth in TSC despite AKT (64).

Thus far, there is very little published information on clinical studies relevant to inhibition of mTOR signaling in patients with hamartoma syndromes. One very interesting, albeit preliminary, approach is the use of PI3K inhibitors in patients with Cowden syndrome. A phase I study of a new PI3K inhibitor, BEZ235, opened in January 2007 and will include advanced-stage cancer patients with Cowden syndrome (66). In July 2003, the National Institute of Neurological Disorders and Stroke published a research plan for TSC stemming from discussions and results presented at an international symposium on TSC held in September 2002 (67). Recommendations for clinical studies were made in the research plan and at the symposium, including phase I/II studies of mTOR inhibitors in TSC patients. One of the recommended studies, evaluation of the effect of rapamycin on TSC-associated renal angiomyolipoma, is in progress (68). Another phase I/II study is evaluating the effects of RAD-001 on astrocytomas in patients with TSC (69).

Neither mutation nor amplification of mTOR has been found in human cancers (70). However, dysregulation in mTOR signaling pathways in premalignant (Table 1) as well as early malignant (9, 13, 70, 71) human tissues suggests mTOR as a promising target for cancer prevention strategies. For example, loss of PTEN and AKT up-regulation occur during human carcinogenesis generally as relatively late events more prevalent in advanced dysplasia or carcinoma in situ than in early dysplasia. In a study using tissue array analysis, it was found that 11% (13 of 113) of ductal carcinoma in situ human breast lesions had PTEN loss, increasing to 26% (35 of 134) of invasive cancers; AKT up-regulation occurred in 33% (38 of 114) of ductal carcinoma in situ cases (72). In testicular carcinogenesis, PTEN is lost during progression from intratubular germ cell neoplasias to cancers (73). Methylation (silencing) of the PTEN promoter has also been reported in cervical intraepithelial lesions (74). AKT is activated in intraepithelial prostate lesions (75, 76); one study found that transition from histologically normal epithelium to prostatic intraepithelial neoplasia was marked by a surge in AKT activation, concurrent with suppression of apoptotic pathways (76). Other studies observed early onset of high-grade prostatic intraepithelial neoplasia in PTEN mutant mice (77, 78). Endometrial carcinogenesis is one notable exception to late-occurring PTEN loss; PTEN mutations are prevalent in both precancerous and cancerous endometrial lesions (79, 80).

Table 1.

Alterations upstream and downstream of mTOR in precancerous human tissues

TargetPI3K/AKT/mTOR pathway alterationReference
Endometrium PTEN mutation (70, 79) 
Lung PI3K catalytic subunit amplification, AKT activation, eIF-4E overexpression (76-78, 81, 148) 
Head and neck AKT activation, eIF-4E overexpression (91, 149, 150) 
Esophagus (Barrett's) AKT activation (151) 
Oral cavity eIF-4E overexpression (152) 
Colon AKT overexpression; eIF-4E overexpression (86-88) 
Prostate AKT activation and overexpression; PTEN loss (late) (70, 75-78) 
Cervix PTEN promoter Methylation (74) 
Skin (melanoma) AKT activation, HIF-1α expression (89, 90, 153) 
Kidney TSC loss, HIF-1α expression (147) 
Leukemia PTEN loss in hematopoietic stem cells (154) 
Breast AKT activation and overexpression, mTOR activation, S6 activation, PTEN loss (late) (72) 
Testicles PTEN loss (late) (73) 
TargetPI3K/AKT/mTOR pathway alterationReference
Endometrium PTEN mutation (70, 79) 
Lung PI3K catalytic subunit amplification, AKT activation, eIF-4E overexpression (76-78, 81, 148) 
Head and neck AKT activation, eIF-4E overexpression (91, 149, 150) 
Esophagus (Barrett's) AKT activation (151) 
Oral cavity eIF-4E overexpression (152) 
Colon AKT overexpression; eIF-4E overexpression (86-88) 
Prostate AKT activation and overexpression; PTEN loss (late) (70, 75-78) 
Cervix PTEN promoter Methylation (74) 
Skin (melanoma) AKT activation, HIF-1α expression (89, 90, 153) 
Kidney TSC loss, HIF-1α expression (147) 
Leukemia PTEN loss in hematopoietic stem cells (154) 
Breast AKT activation and overexpression, mTOR activation, S6 activation, PTEN loss (late) (72) 
Testicles PTEN loss (late) (73) 

Amplification of the catalytic subunit of PI3K (81) and activation of AKT (81-83) are associated with development of severe dysplasia in the lung; tobacco-specific carcinogens activate AKT in primary human lung epithelial cells in vitro (84). eIF-4E expression also increases during progression from atypical adenomatous hyperplasia to invasive lung cancer (85). In the colon, both AKT (86) and eIF-4E are overexpressed in adenomas (87, 88); the latter is often associated with elevated cyclin D1 levels (87). AKT activation is also seen in precancerous stages of malignant melanoma (89, 90). In head and neck lesions, expression of eIF-4E increases with increasing dysplasia (91).

Five recent studies with rapamycin and its analogues supply compelling data to assess chemopreventive potential of mTOR inhibition. In the first study, a 2-week treatment with the rapamycin analogue RAD-001 (10 mg/kg body weight/d, i.g., as a microemulsion) completely reversed the prostatic intraepithelial neoplasia phenotype in ventral prostates of mice expressing human AKT1 by inducing apoptosis (blocked by coexpression of proapoptotic Bcl-2) and inactivating HIF-1α target genes, including genes encoding most glycolytic enzymes (17). In the second study, another rapamycin analogue, CCI-779 (20 mg/kg body weight/d, i.p., for 8 days), diminished sizes and total number of murine lung lesions (atypical alveoli, hyperplasias, and adenomas) induced by somatic K-RAS activation. Most of this effect was due to decreased progression to adenomas (92).

The third study is relevant to prevention in adjuvant settings and to prevention of AKT-induced antiestrogen resistance (93). The mTOR pathway was up-regulated in aromatase expressing breast cancer cells with hormones (estradiol or androstenedione). Up-regulation was reversed by treatment with RAD-001 or the aromatase inhibitor letrozole (94). Both letrozole and RAD-001 inhibited androstenedione-induced proliferation. The agents showed synergistic activity in combination, which is attributed to increased inhibition of G1 progression and increased apoptosis. The combination is now being evaluated in a phase II trial in breast cancer patients before surgery (neoadjuvant setting; ref. 93).

In the fourth study, rapamycin showed chemopreventive activity against mammary gland tumors in transgenic mice bearing activated ErbB2 (HER-2/neu) receptor either alone (NeuYD) or with VEGF expression (NeuYD × VEGF; ref. 58). Low-dose rapamycin (0.75 mg/kg, i.p., every other day) was given to groups of 20 females of both strains, starting ∼2 weeks before the expected appearance of spontaneous mammary gland tumors (i.e., at 92 days of age in NeuYD mice and 38 days of age in NeuYD × VEGF mice) until sacrifice (at 138 days of age). Rapamycin dramatically inhibited tumor formation in NeuYD mice. One third of rapamycin-treated NeuYD mice had no clinically detectable tumors at sacrifice, whereas 100% of controls had multiple tumors; tumor weight and volume per mouse were significantly reduced compared with controls (P < 0.0007). A lesser, yet significant, chemopreventive effect was seen against more aggressive tumors in NeuYD × VEGF mice, and tumor weight and volume per mouse were significantly reduced (P < 0.001). In both strains, rapamycin suppressed growth of established tumors, more profoundly in NeuYD × VEGF mice. Of note, the effect of rapamycin was cytostatic rather than cytotoxic because tumor regrowth was apparent by 10 days after discontinuation of rapamycin. Additional experiments in monolayer and three-dimensional culture suggested that chemopreventive and growth-inhibiting effects of rapamycin in this setting could result from suppression of ErbB3 (thereby inhibiting heterodimerization of ErbB2 and ErbB3 receptors; ref. 95) and of HIF-1α. ErbB2 (HER-2/neu) is overexpressed in approximately one third of human breast tumors, suggesting mTOR inhibition as a possible chemopreventive strategy against metachronous tumors or recurrence in high-risk patients whose primary tumors overexpressed ErbB2 or in patients showing dysregulation of the PI3K/AKT/mTOR signaling pathway.

The fifth study also evaluated chemopreventive effects of rapamycin in a transgenic mouse model of human breast carcinogenesis (96). Mammary intraepithelial neoplasia outgrowths model human ductal carcinoma in situ, carry the oncogene PyV-mT (which activates ErbB2 signaling, including the PI3K/AKT pathway), are transplantable, and develop into invasive mammary gland tumors. Rapamycin (0.75 and 3.0 mg/kg body weight, i.p., every other day for 35 days starting 3 weeks posttransplantation) significantly inhibited growth of mammary intraepithelial neoplasia outgrowths, invasive tumor incidence, and tumor burden.

Other studies suggest the utility of inhibiting mTOR signaling in preventive settings. The area of complex atypical hyperplasia in uterine secretory epithelium of PTEN+/− mice diminished when CCI-779 was administered late in life, after most mice had developed these lesions (97). As noted above, rapamycin prevents development of macroscopic renal lesions in a rat model of TSC (65, 98), although without affecting formation of microscopic precursor lesions [ref. 65; as the role of TSC1/2 loss in sporadic human cancers is unknown (99), it is unclear how these findings affect more general prevention]. Lastly, studies in renal transplant patients treated with rapamycin suggest that mTOR inhibition may prevent development of skin cancer in this population (100-102). Rapamycin and CCI-779 have limited antitumor activity against gliomas (particularly medulloblastomas), often seen in patients with basal cell nevus syndrome, presumably because of an effect on the gli gene (13). These observations and the high frequency of basal cell skin cancers seen in basal cell nevus syndrome patients (103) led the National Cancer Institute, Division of Cancer Prevention to evaluate topical rapamycin in a phase I proof-of-principle and biomarkers study in basal cell nevus syndrome patients.

The mechanistic data and experimental results presented above suggest that mTOR inhibition will have clinical chemopreventive efficacy primarily in preventing progression of precancerous lesions. They also suggest that mTOR inhibition could be efficiently evaluated in clinical trials with subjects at high risk for developing metachronous lesions. Despite these promising activities of the rapamycins, potential up-regulation of PI3K/AKT (18, 21, 53) by mTOR inhibition may temper the use of mTOR-specific inhibitors as single agents. However, blocking effects of mTOR inhibition on procarcinogenic cell proliferation and angiogenesis and identification of other molecular targets associated with carcinogenic activities affected by mTOR signaling (particularly along PI3K/AKT–mediated pathways) suggest additional cancer prevention strategies that do not rely exclusively on directly inhibiting mTOR; these strategies are described below (Table 2; Fig. 2).

Table 2.

Chemopreventive agents with potential to modulate mTOR signaling

AgentChemopreventive activityReferences
DNA methylation inhibitors (activate PTEN gene transcription)   
    5-Aza-2′-deoxycytidine (decitabine) Intestine, lung, prostate (4) 
    Tea polyphenols (epigallocatechin gallate, polyphenon E) Colon, prostate, esophagus, bladder, forestomach, liver, lung, breast, small intestine, skin (155-160) 
    Catechin Intestines, skin (161-163) 
    Soy isoflavones (genistein) Breast, prostate, skin, stomach (164-167) 
    Flavonoids (quercetin, myricetin) Lung, skin (161) 
    Caffeic acid Forestomach (168) 
    Chlorogenic acid Intestines, lung, skin (168) 
    Sodium selenite Colon, esophagus, liver, breast, pancreas, stomach (169) 
    Benzyl selenocyanate Colon, forestomach, breast (169) 
    1,4-Phenylenebis(methylene)selenocyanate Colon, lung, breast (169) 
   
Histone deacetylase inhibitors (activate PTEN gene transcription)   
    Sulforaphane Colon, forestomach, breast (170-173) 
    Sodium butyrate, phenylbutyrate Colon (aberrant crypts) (4, 174) 
    Diallyl sulfide Colon, esophagus, forestomach, liver, lung, breast, skin, thyroid (4, 175, 176) 
    Phenethyl isothiocyanate Esophagus, forestomach, lung, breast, pancreas (177, 178) 
    Suberoylanilide hydroxamic acid Breast, lung (4, 179, 180) 
    Valproic acid Intestine (4, 181) 
   
Histone acetyltransferase inhibitors (activate PTEN gene transcription)   
    Curcumin Colon, duodenum, forestomach, breast, skin, tongue (182, 183) 
   
Ornithine decarboxylase inhibitors (inhibit protein kinase–mediated cell proliferation)   
    2-Difluoromethylornithine Colon, bladder, skin (156, 157) 
    Retinoids (fenretinide) Breast, skin, head and neck, bladder, ovary, colon (156, 157) 
   
PI3K/AKT inhibitors   
    Celecoxib Colon, bladder, skin, lung, esophagus, head and neck, breast, prostate (120-122, 157) 
    Curcumin Colon, duodenum, forestomach, breast, skin, tongue (160, 182-184) 
    Deguelin Lung, breast, colon (119, 185, 186) 
    Indole-3-carbinol, diindolylmethane Breast (116, 117) 
    Resveratrol Colon, prostate, breast (187-192) 
    Rosiglitazone Breast, prostate, colon (1, 125) 
    Soy isoflavones (genistein) Breast, prostate, colon, skin, stomach (164-167) 
    Tea polyphenols (epigallocatechin gallate, polyphenon E) Colon, prostate, esophagus, bladder, head and neck, forestomach, liver, lung, breast, small intestine, skin (155-160) 
   
Epidermal growth factor/epidermal growth factor receptor inhibitors (inhibit protein kinase–mediated cell proliferation)   
    Tyrosine kinase specific inhibitors (erlotinib, gefitinib, EKB569) Lung, colon, head and neck, breast (193-195) 
    Curcumin Colon, duodenum, forestomach, breast, skin, tongue (111, 160, 182, 183) 
    Resveratrol Colon, prostate, breast (160, 187-192, 196) 
    mTOR inhibition   
    Curcumin Colon, duodenum, forestomach, breast, skin, tongue (182, 183) 
AMPK activators (inhibit AKT-driven blockade of TSC1/TSC2 tumor suppressors)   
    Resveratrol Colon, prostate, breast (124, 160, 187-192, 196) 
    Metformin Breast, pancreas (45, 127, 128, 197) 
    Rosiglitazone Breast, prostate, colon (1, 125) 
AgentChemopreventive activityReferences
DNA methylation inhibitors (activate PTEN gene transcription)   
    5-Aza-2′-deoxycytidine (decitabine) Intestine, lung, prostate (4) 
    Tea polyphenols (epigallocatechin gallate, polyphenon E) Colon, prostate, esophagus, bladder, forestomach, liver, lung, breast, small intestine, skin (155-160) 
    Catechin Intestines, skin (161-163) 
    Soy isoflavones (genistein) Breast, prostate, skin, stomach (164-167) 
    Flavonoids (quercetin, myricetin) Lung, skin (161) 
    Caffeic acid Forestomach (168) 
    Chlorogenic acid Intestines, lung, skin (168) 
    Sodium selenite Colon, esophagus, liver, breast, pancreas, stomach (169) 
    Benzyl selenocyanate Colon, forestomach, breast (169) 
    1,4-Phenylenebis(methylene)selenocyanate Colon, lung, breast (169) 
   
Histone deacetylase inhibitors (activate PTEN gene transcription)   
    Sulforaphane Colon, forestomach, breast (170-173) 
    Sodium butyrate, phenylbutyrate Colon (aberrant crypts) (4, 174) 
    Diallyl sulfide Colon, esophagus, forestomach, liver, lung, breast, skin, thyroid (4, 175, 176) 
    Phenethyl isothiocyanate Esophagus, forestomach, lung, breast, pancreas (177, 178) 
    Suberoylanilide hydroxamic acid Breast, lung (4, 179, 180) 
    Valproic acid Intestine (4, 181) 
   
Histone acetyltransferase inhibitors (activate PTEN gene transcription)   
    Curcumin Colon, duodenum, forestomach, breast, skin, tongue (182, 183) 
   
Ornithine decarboxylase inhibitors (inhibit protein kinase–mediated cell proliferation)   
    2-Difluoromethylornithine Colon, bladder, skin (156, 157) 
    Retinoids (fenretinide) Breast, skin, head and neck, bladder, ovary, colon (156, 157) 
   
PI3K/AKT inhibitors   
    Celecoxib Colon, bladder, skin, lung, esophagus, head and neck, breast, prostate (120-122, 157) 
    Curcumin Colon, duodenum, forestomach, breast, skin, tongue (160, 182-184) 
    Deguelin Lung, breast, colon (119, 185, 186) 
    Indole-3-carbinol, diindolylmethane Breast (116, 117) 
    Resveratrol Colon, prostate, breast (187-192) 
    Rosiglitazone Breast, prostate, colon (1, 125) 
    Soy isoflavones (genistein) Breast, prostate, colon, skin, stomach (164-167) 
    Tea polyphenols (epigallocatechin gallate, polyphenon E) Colon, prostate, esophagus, bladder, head and neck, forestomach, liver, lung, breast, small intestine, skin (155-160) 
   
Epidermal growth factor/epidermal growth factor receptor inhibitors (inhibit protein kinase–mediated cell proliferation)   
    Tyrosine kinase specific inhibitors (erlotinib, gefitinib, EKB569) Lung, colon, head and neck, breast (193-195) 
    Curcumin Colon, duodenum, forestomach, breast, skin, tongue (111, 160, 182, 183) 
    Resveratrol Colon, prostate, breast (160, 187-192, 196) 
    mTOR inhibition   
    Curcumin Colon, duodenum, forestomach, breast, skin, tongue (182, 183) 
AMPK activators (inhibit AKT-driven blockade of TSC1/TSC2 tumor suppressors)   
    Resveratrol Colon, prostate, breast (124, 160, 187-192, 196) 
    Metformin Breast, pancreas (45, 127, 128, 197) 
    Rosiglitazone Breast, prostate, colon (1, 125) 

Direct Inhibition of mTOR

Ease of administration (i.e., availability of oral formulations) and chronic safety risk are critical considerations for clinical development of chemopreventive agents, although, as suggested above, “personalized” medicine may lead to cohort enrichment with high-risk subjects who are more likely to find net benefit in treatment despite inconvenience and some potential toxicity. As potential chemopreventive agents, immunosuppressive activity, cutaneous toxicity (e.g., rashes, nail effects), nephrotoxicity in diseased kidneys, and limited oral bioavailability are concerns with rapamycin and its analogues CCI-779 (rapamycin prodrug), AP23573, and RAD-001, despite their improved solubility and stability (13, 48, 104). RAD-001 may be the most useful of the analogues in chemopreventive settings because of its well-developed oral formulation.

Use of mTOR as a direct target in cancer prevention may also require consideration of other strategies to minimize risk/benefit ratio of using these agents. For example, in preclinical and clinical cancer therapy studies, antitumor effects of mTOR inhibitors are maintained using intermittent dosing schedules, which minimize immunosuppression (13, 52, 70). Using this strategy, partial responses and stable disease have been seen in phase I and II studies in patients with a variety of tumor types, and only mild to moderate toxicities have been observed (13, 48). Topical application to the drug target tissue where feasible (e.g., on skin and lung) may also enhance efficacy and avoid many safety issues (i.e., by decreasing the need for systemic bioavailability and the potential for systemic toxicity). As noted above, topical application of rapamycin is, in fact, being evaluated for prevention of basal cell carcinomas in basal cell nevus syndrome patients. Inhalational exposure to chemopreventive agents also seems to be a promising strategy for chemoprevention of lung cancer (105-107), as evidenced by proof-of-principle studies of budesonide in laboratory animals (105, 106) and in patients with bronchial dysplasia (108). Interestingly, a liposomal formulation of rapamycin for inhalational delivery has been proposed for local immunosuppressive treatment of cytokine-induced inflammation in the lung (109).

A few recent studies found that rapamycin analogues are not the only agents which include direct inhibition of mTOR among their activities. PI-103, a novel dual inhibitor of mTOR and the p110α isoform of PI3K, was recently reported to block phosphorylation of AKT and cause G0-G1 cell cycle arrest in glioma cells (110). Unlike the broad-spectrum PI3K inhibitor LY294002, which suppresses AKT phosphorylation and induces cytotoxicity in the same dose range, the effective dose of PI-103 is much lower than the cytotoxic dose. PI-103 also shows significant activity against established tumors in nude mice (110). The dual blockade of AKT and mTOR at relatively low, relatively nontoxic doses makes PI-103 an appealing candidate for chemoprevention. However, its potential toxicities include inhibition of insulin signaling in mice, suggesting possible diabetogenic activity. PI-103 also inhibits DNA protein kinase, a key enzyme for repairing double-stranded DNA, suggesting interference with DNA repair.

Indirect Inhibition of mTOR

Combinations of mTOR inhibitors with established chemopreventive agents, or use of other targets of these chemopreventive agents as surrogates for mTOR, may be effective cancer prevention strategies (Table 2; Fig. 2). The most attractive potential combinations are those that inhibit both mTORC1- and AKT-driven signaling, while taking advantage of the good safety profiles of established chemopreventive agents (e.g., food-derived products or drugs approved for other indications) to attain low effective doses and minimize the potential for toxicity. An example would be the combination of a rapamycin analogue (e.g., RAD-001) with a food-derived chemopreventive agent with AKT-inhibiting activity [e.g., curcumin (111, 112), resveratrol (113), epigallocatechin gallate (114), soy isoflavones (genistein; ref. 115), indole-3-carbinol (116), or diindoylmethane (116, 117)]. Recent preliminary studies have even suggested that some chemopreventive agents with AKT-inhibiting activity may dampen mTOR signaling partially through direct mTOR inhibition. For example, curcumin has shown chemopreventive activity in animal models of carcinogenesis (118) and is under evaluation in phase I clinical trials. It inhibits phosphorylation of mTOR, S6K1, and 4E-BP1 at physiologic concentrations (2.5 μmol/L) in a panel of cancer cells (111). Whereas this activity may not be specific for mTOR, it is noteworthy that much higher concentrations of curcumin (>40 μmol/L) were required to inhibit AKT (111, 112).

mTOR-specific inhibitors might also be combined with other chemopreventive agents that inhibit PI3K or AKT [e.g., deguelin (119) or celecoxib (120-122)] or affect ancillary pathways, such as 2-difluoromethylornithine, which inhibits ornithine decarboxylase and hence protein kinase C–mediated proliferative activity (123), or DNA methyltransferase inhibitors or histone deacetylase inhibitors, which reactivate the hypermethylated PTEN promoter (4).

Some agents may exert chemopreventive activity, at least in part, via the energy-sensing component of mTOR signaling, possibly by activating AMPK. These chemopreventive agents may prove to be effective as the mTORC1 inhibitor component in agent combinations targeting mTOR signaling. For example, resveratrol increased levels of AMPK in mice fed a high-calorie diet and also increased insulin sensitivity and reduced IGF-1 levels (124). Rosiglitazone, an antidiabetes peroxisome proliferator–activated receptor-γ agonist with chemopreventive activity (reviewed in ref. 1), increased PTEN expression, inhibited AKT phosphorylation, increased AMPK phosphorylation, and decreased S6K1 phosphorylation in non–small-cell lung carcinoma cells (125). Metformin, another type 2 diabetes drug, also activates AMPK (126) and has shown chemopreventive activity, suppressing development of carcinogen-induced pancreatic cancers in hamsters fed a high-fat diet (127) and decreasing size and increasing latency of mammary adenocarcinomas in HER-2/neu transgenic mice (128). Because type 2 diabetes has been associated with increased risk for pancreatic (129), liver (130), and, presumably, other cancers, treatment for type 2 diabetes could also be cancer chemopreventive. Because mTOR inhibition down-regulates expression of genes encoding enzymes of the glycolytic pathway, [18F]fluorodeoxyglucose-positron emission tomography scanning may provide noninvasive readouts of mTOR activity in tissue to help identify likely responders to treatment with mTOR-inhibiting cancer prevention strategies (17).

Human epidemiologic studies of calorie restriction are difficult to conduct and interpret, particularly because of requirements for extensive estimation of food intake and lack of clarity about the appropriate time period or food sources for observation (131-133). Nonetheless, several carefully documented studies in subjects with access to high-quality diets (i.e., where effects would not as likely be confounded by malnutrition) have suggested that calorie restriction can increase longevity and reduce cancer and other disease incidences (reviewed in ref. 132). For example, Kagawa et al. showed that Okinawans ingesting ∼20% fewer calories than the overall Japanese population also had lower incidences of cerebral vascular disease, cancer, and heart disease (132, 134). In addition, cancer incidences in prostate, breast, and colon have been associated with calorie (energy) intake. As one example, Platz and colleagues showed modest associations of energy intake and body size or physical activity with increased risk of metastatic or fatal prostate cancer in younger men (age ≤65 years; refs. 135, 136). As a second example, among women 55 to 74 years old in the screening arm of National Cancer Institute Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial, those in the highest quartile of energy intake (≥2,084 kcal/d) had a significantly increased risk for breast cancer compared with those in the lowest quartile (<1,316 kcal/d) based on data reported in a food frequency questionnaire at baseline (137). Finally, in a study of 2,073 cases of primary colon cancer and 2,466 age- and sex-matched controls, high-energy intake was associated with increased risk of colon cancer in both men and women [OR, 1.74 (95% CI, 1.14-2.67) for men and 1.70 (95% CI, 1.07-2.70) for women; ref. 138].

In experimental studies, caloric restriction has been shown to prevent cancer and increase life span in a wide variety of mammalian and nonmammalian species. In rodent models, caloric restriction inhibits the development of chemically (e.g., with benzo[a]pyrene, N,N-diethylnitrosamine, 7,12-dimethybenz[a]anthracene, or p-cresidine), radiologically, or genetically (e.g., in p53-deficient, WNT-1 transgenic mice) induced tumors (139). Once thought to be a secondary response to decreased cell growth, the inhibitory effects of caloric restriction on tumor development are increasingly recognized as stemming from alterations in specific molecular response pathways (124, 133, 140, 141).

mTOR senses the overall growth milieu of the cell by integrating signals from hormones, growth factors, nutrients, energy, and other environmental factors. During caloric restriction, this checkpoint would not be traversed even in the context of aberrant growth factor stimulation—a hallmark of transformation (142). Cells starved for amino acids (34) or energy (40) do not activate mTOR in response to growth factors. Because mTOR also senses DNA damage (50), decreased signaling through mTOR in caloric restriction may check cell growth subsequent to genetic insult. IGF-1, which is also associated with the antitumor effects of caloric restriction (139, 141), signals through mTOR as well as other downstream effectors.

Rapamycin affects expression of many genes involved in nutrient and energy metabolism, protein synthesis and turnover, stress response, immune modulation, and chromatin remodeling. Genes affected by rapamycin in human B lymphoma cells and mouse T lymphocytes (14) significantly overlap with genes affected by calorie starvation in animals (143). Similar antiproliferative effects are observed in mouse T lymphocytes treated with rapamycin, low levels of glutamine, or glucose. Moreover, rapamycin does not increase the degree of growth inhibition when cells are deprived of nutrients, suggesting that the antiproliferative effects of rapamycin result from its ability to induce a starvation-like signal (14). Thus, studies of rapamycin-induced gene expression support the hypothesis that diminished signaling through mTOR contributes to antitumor effects of caloric restriction.

Finally, it is worth noting that increased longevity, which is also associated with caloric restriction (139, 141), may be at least partially dependent on TOR-mediated pathways. TOR deficiency doubles the natural life span of Caenorhabditis elegans; these effects seem to interface with the known negative regulatory effects of the IGF receptor homologue gene on longevity in this organism (144). Inhibition of TOR signaling in Drosophila melanogaster extends life span in a manner similar to dietary restriction (145). In mice fed a high-calorie diet, resveratrol both mimicked the effects of calorie restriction and increased survival of the animals; as noted above, the calorie restriction effects were mediated at least partially via AMPK and IGF-1/mTOR signaling (124).

Although discovered little more than a decade ago (146), mTOR is established as a critical central controller which permits cells to progress through G1 only when all conditions are favorable for growth. Numerous elements of mTOR signaling pathways are dysregulated in precancerous lesions and early evidence shows promising effects of mTOR pathway inhibition in preventive settings. For example, precancerous lesions or early cancers with PTEN loss, or those dependent on PI3K/AKT signaling due to other molecular lesions, may be highly sensitive to mTOR inhibition. Observing clinically normal cells from persons at risk for cancer because of conditions such as TSC suggest that the initial underlying lesion (i.e., TSC2 mutation) seems to be related to cancer development via activation of mTOR pathways (147), potentially leading to identification of the earliest biomarkers and new agents for cancer prevention.

Promising mechanism-based prevention strategies combining rapamycin inhibitors with established chemopreventive agents are suggested by published results. These combination strategies could be directed both to specific cohorts who would likely benefit and to specific cancer settings. For example, dependence of AKT-activated cells on functioning mTOR signaling for growth and proliferation suggests a potential synergy of combinations of mTOR inhibitors with inhibitors of other targets on the hormone/IGF receptor/PI3K/AKT signaling pathways.

Besides IGF receptor/PI3K/AKT, other components of mTOR signaling may also prove to be useful targets for chemoprevention. For example, if eIF-4E is central to mTOR-dependent tumorigenesis, selective targeting of this branch of the mTOR signaling pathway may offer a more favorable therapeutic strategy. An especially promising approach targets the energy sensing component of mTOR signaling by activation of AMPK. Finally, the effect of mTOR inhibition on prevention therapy could be much greater if mTOR pathways are indeed integral to transmitting signals associated with beneficial effects of caloric restriction on tumor development and longevity. This intriguing possibility merits further exploration.

To conclude, despite successful proof-of principle studies with rapamycin and its analogues, use of these drugs alone or in combination with other agents in clinical prevention settings will necessitate development of acceptable chronic toxicity profiles and formulations with improved ease of administration and bioavailability. An alternative approach should involve use of established chemopreventive inhibitors of collateral targets on mTOR pathways (Table 2). Many of these agents are natural food-derived products or drugs developed for chronic oral administration with well-characterized safety profiles. Moreover, it seems that combinations of rapamycin or its analogues together with established chemopreventive agents would most likely be efficacious in clinical cancer prevention trials.

1
Kopelovich L, Fay JR, Glazer RI, Crowell JA. Peroxisome proliferator-activated receptor modulators as potential chemopreventive agents.
Mol Cancer Ther
2002
;
1
:
357
–63.
2
Crowell JA, Steele VE, Sigman CC, Fay JR. Is inducible nitric oxide synthase a target for chemoprevention?
Mol Cancer Ther
2003
;
2
:
815
–23.
3
Kopelovich L, Crowell JA, Fay JR. The epigenome as a target for cancer chemoprevention.
J Natl Cancer Inst
2003
;
95
:
1747
–57.
4
Fay JR, Crowell JA, Kopelovich L. Targeting epigenetic regulatory mechanisms in cancer chemoprevention.
Expert Opin Ther Targets
2005
;
9
:
315
–28.
5
Sabatini DM. mTOR and cancer: insights into a complex relationship.
Nat Rev Cancer
2006
;
6
:
729
–34.
6
Sarbassov DD, Ali SM, Sengupta S, et al. Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB.
Mol Cell
2006
;
22
:
159
–68.
7
Corradetti MN, Inoki K, Guan KL. The stress-inducted proteins RTP801 and RTP801L are negative regulators of the mammalian target of rapamycin pathway.
J Biol Chem
2005
;
280
:
9769
–72.
8
Proud CG. The multifaceted role of mTOR in cellular stress responses.
DNA Repair (Amst)
2004
;
3
:
927
–34.
9
Bjornsti MA, Houghton PJ. The TOR pathway: a target for cancer therapy.
Nat Rev Cancer
2004
;
4
:
335
–48.
10
Fingar DC, Blenis J. Target of rapamycin (TOR): an integrator of nutrient and growth factor signals and coordinator of cell growth and cell cycle progression.
Oncogene
2004
;
23
:
3151
–71.
11
Inoki K, Ouyang H, Li Y, Guan KL. Signaling by target of rapamycin proteins in cell growth control.
Microbiol Mol Biol Rev
2005
;
69
:
79
–100.
12
Martin DE, Hall MN. The expanding TOR signaling network.
Curr Opin Cell Biol
2005
;
17
:
158
–66.
13
Faivre S, Kroemer G, Raymond E. Current development of mTOR inhibitors as anticancer agents.
Nat Rev Drug Discov
2006
;
5
:
671
–88.
14
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.
15
Powers T. Ribosome biogenesis: giant steps for a giant problem.
Cell
2004
;
119
:
901
–2.
16
Abraham RT. mTOR as a positive regulator of tumor cell responses to hypoxia.
Curr Top Microbiol Immunol
2004
;
279
:
299
–319.
17
Majumder PK, Febbo PG, Bikoff R, et al. mTOR inhibition reverses Akt-dependent prostate intraepithelial neoplasia through regulation of apoptotic and HIF-1-dependent pathways.
Nat Med
2004
;
10
:
594
–601.
18
Granville CA, Memmott RM, Gills JJ, Dennis PA. Handicapping the race to develop inhibitors of the phosphoinositide 3-kinase/Akt/mammalian target of rapamycin pathway.
Clin Cancer Res
2006
;
12
:
679
–89.
19
Hay N. The Akt-mTOR tango and its relevance to cancer.
Cancer Cell
2005
;
8
:
179
–83.
20
Harrington LS, Findlay GM, Lamb RF. Restraining PI3K: mTOR signalling goes back to the membrane.
Trends Biochem Sci
2005
;
30
:
35
–42.
21
O'Reilly KE, Rojo F, She QB, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt.
Cancer Res
2006
;
66
:
1500
–8.
22
Long X, Lin Y, Ortiz-Vega S, Yonezawa K, Avruch J. Rheb binds and regulates the mTOR kinase.
Curr Biol
2005
;
15
:
702
–13.
23
Avruch J, Lin Y, Long X, Murthy S, Ortiz-Vega S. Recent advances in the regulation of the TOR pathway by insulin and nutrients.
Curr Opin Clin Nutr Metab Care
2005
;
8
:
67
–72.
24
Inoki K, Corradetti MN, Guan KL. Dysregulation of the TSC-mTOR pathway in human disease.
Nat Genet
2005
;
37
:
19
–24.
25
Shima H, Pende M, Chen Y, Fumagalli S, Thomas G, Kozma SC. Disruption of the p70(s6k)/p85(s6k) gene reveals a small mouse phenotype and a new functional S6 kinase.
EMBO J
1998
;
17
:
6649
–59.
26
Gingras AC, Raught B, Sonenberg N. mTOR signaling to translation.
Curr Top Microbiol Immunol
2004
;
279
:
169
–97.
27
Fang Y, Vilella-Bach M, Bachmann R, Flanigan A, Chen J. Phosphatidic acid-mediated mitogenic activation of mTOR signaling.
Science
2001
;
294
:
1942
–5.
28
Fang Y, Park IH, Wu AL, et al. PLD1 regulates mTOR signaling and mediates Cdc42 activation of S6K1.
Curr Biol
2003
;
13
:
2037
–44.
29
Foster DA. Regulation of mTOR by phosphatidic acid?
Cancer Res
2007
;
67
:
1
–4.
30
Patti ME, Brambilla E, Luzi L, Landaker EJ, Kahn CR. Bidirectional modulation of insulin action by amino acids.
J Clin Invest
1998
;
101
:
1519
–29.
31
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.
32
Kim DH, Sarbassov DD, 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.
33
Kim DH, Sarbassov DD, Ali SM, et al. GβL, a positive regulator of the rapamycin-sensitive pathway required for the nutrient-sensitive interaction between raptor and mTOR.
Mol Cell
2003
;
11
:
895
–904.
34
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.
35
Tee AR, Fingar DC, Manning BD, Kwiatkowski DJ, Cantley LC, Blenis J. Tuberous sclerosis complex-1 and -2 gene products function together to inhibit mammalian target of rapamycin (mTOR)-mediated downstream signaling.
Proc Natl Acad Sci U S A
2002
;
99
:
13571
–6.
36
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.
37
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.
38
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.
39
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.
40
Dennis PB, Jaeschke A, Saitoh M, Fowler B, Kozma SC, Thomas G. Mammalian TOR: a homeostatic ATP sensor.
Science
2001
;
294
:
1102
–5.
41
Rutter GA, Da Silva Xavier G, Leclerc I. Roles of 5′-AMP-activated protein kinase (AMPK) in mammalian glucose homoeostasis.
Biochem J
2003
;
375
:
1
–16.
42
Inoki K, Zhu T, Guan KL. TSC2 mediates cellular energy response to control cell growth and survival.
Cell
2003
;
115
:
577
–90.
43
Hawley SA, Boudeau J, Reid JL, et al. Complexes between the LKB1 tumor suppressor, STRADα/β and MO25α/β are upstream kinases in the AMP-activated protein kinase cascade.
J Biol
2003
;
2
:
28
.
44
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.
45
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 U S A
2004
;
101
:
3329
–35.
46
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.
47
Cheng SW, Fryer LG, Carling D, Shepherd PR. Thr2446 is a novel mammalian target of rapamycin (mTOR) phosphorylation site regulated by nutrient status.
J Biol Chem
2004
;
279
:
15719
–22.
48
Rao RD, Buckner JC, Sarkaria JN. Mammalian target of rapamycin (mTOR) inhibitors as anti-cancer agents.
Curr Cancer Drug Targets
2004
;
4
:
621
–35.
49
Feng Z, Zhang H, Levine AJ, Jin S. The coordinate regulation of the p53 and mTOR pathways in cells.
Proc Natl Acad Sci U S A
2005
;
102
:
8204
–9.
50
Tee AR, Proud CG. DNA-damaging agents cause inactivation of translational regulators linked to mTOR signalling.
Oncogene
2000
;
19
:
3021
–31.
51
Schmelzle T, Hall MN. TOR, a central controller of cell growth.
Cell
2000
;
103
:
253
–62.
52
Temsirolimus: CCI 779, CCI-779, cell cycle inhibitor-779.
Drugs R D
2004
;
5
:
363
–7.
53
Sun SY, Rosenberg LM, Wang X, et al. Activation of Akt and eIF4E survival pathways by rapamycin-mediated mammalian target of rapamycin inhibition.
Cancer Res
2005
;
65
:
7052
–8.
54
Mamane Y, Petroulakis E, Rong L, Yoshida K, Ler LW, Sonenberg N. eIF4E-from translation to transformation.
Oncogene
2004
;
23
:
3172
–9.
55
Mahalingam M, Templeton DJ. Constitutive activation of S6 kinase by deletion of amino-terminal autoinhibitory and rapamycin sensitivity domains.
Mol Cell Biol
1996
;
16
:
405
–13.
56
Wendel HG, De Stanchina E, Fridman JS, et al. Survival signalling by Akt and eIF4E in oncogenesis and cancer therapy.
Nature
2004
;
428
:
332
–7.
57
Ruggero D, Montanaro L, Ma L, et al. The translation factor eIF-4E promotes tumor formation and cooperates with c-Myc in lymphomagenesis.
Nat Med
2004
;
10
:
484
–6.
58
Liu M, Howes A, Lesperance J, et al. Antitumor activity of rapamycin in a transgenic mouse model of ErbB2-dependent human breast cancer.
Cancer Res
2005
;
65
:
5325
–36.
59
Huang S, Liu LN, Hosoi H, Dilling MB, Shikata T, Houghton PJ. p53/p21(CIP1) cooperate in enforcing rapamycin-induced G(1) arrest and determine the cellular response to rapamycin.
Cancer Res
2001
;
61
:
3373
–81.
60
Hosoi H, Dilling MB, Shikata T, et al. Rapamycin causes poorly reversible inhibition of mTOR and induces p53-independent apoptosis in human rhabdomyosarcoma cells.
Cancer Res
1999
;
59
:
886
–94.
61
Boffa DJ, Luan F, Thomas D, et al. Rapamycin inhibits the growth and metastatic progression of non-small cell lung cancer.
Clin Cancer Res
2004
;
10
:
293
–300.
62
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.
63
Maxwell PH, Ratcliffe PJ. Oxygen sensors and angiogenesis.
Semin Cell Dev Biol
2002
;
13
:
29
–37.
64
Manning BD. Balancing Akt with S6K: implications for both metabolic diseases and tumorigenesis.
J Cell Biol
2004
;
167
:
399
–403.
65
Kenerson H, Dundon TA, Yeung RS. Effects of rapamycin in the Eker rat model of tuberous sclerosis complex.
Pediatr Res
2005
;
57
:
67
–75.
66
News and events: clinical trial of first-in-class drug therapy begins at Nevada Cancer Institute. Accessed at http://www.nevadacancerinstitute.org/news/02132007.htm on 03/13/2007.
67
National Institute of Neurological Disorders and Stroke. Research plan for tuberous sclerosis. July 2003. Accessed at http://www.ninds.nih.gov/about_ninds/plans/tscler_research_plan.htm on 03/13/2007.
68
Sirolimus in treating patients with angiomyolipoma of the kidney. ClinicalTrials.gov Identifier: NCT00126672. Accessed at http://clinicaltrials.gov/ct/show/NCT00126672?order=1 on 03/13/2007.
69
Everolimus (RAD001) therapy of giant cell astrocytoma in patients with tuberous sclerosis complex. ClinicalTrials.gov Identifier: NCT00411619. Accessed at http://clinicaltrials.gov/ct/show/NCT00411619?order=1 on 03/13/2007.
70
Sawyers CL. Will mTOR inhibitors make it as cancer drugs?
Cancer Cell
2003
;
4
:
343
–8.
71
Vivanco I, Sawyers CL. The phosphatidylinositol 3-kinase AKT pathway in human cancer.
Nat Rev Cancer
2002
;
2
:
489
–501.
72
Bose S, Chandran S, Mirocha JM, Bose N. The Akt pathway in human breast cancer: a tissue-array-based analysis.
Mod Pathol
2006
;
19
:
238
–45.
73
Di Vizio D, Cito L, Boccia A, et al. Loss of the tumor suppressor gene PTEN marks the transition from intratubular germ cell neoplasias (ITGCN) to invasive germ cell tumors.
Oncogene
2005
;
24
:
1882
–94.
74
Cheung TH, Lo KW, Yim SF, et al. Epigenetic and genetic alternation of PTEN in cervical neoplasm.
Gynecol Oncol
2004
;
93
:
621
–7.
75
Liao Y, Grobholz R, Abel U, et al. Increase of AKT/PKB expression correlates with Gleason pattern in human prostate cancer.
Int J Cancer
2003
;
107
:
676
–80.
76
Paweletz CP, Charboneau L, Bichsel VE, et al. Reverse phase protein microarrays which capture disease progression show activation of pro-survival pathways at the cancer invasion front.
Oncogene
2001
;
20
:
1981
–9.
77
Wang S, Gao J, Lei Q, et al. Prostate-specific deletion of the murine PTEN tumor suppressor gene leads to metastatic prostate cancer.
Cancer Cell
2003
;
4
:
209
–21.
78
Backman SA, Ghazarian D, So K, et al. Early onset of neoplasia in the prostate and skin of mice with tissue-specific deletion of Pten.
Proc Natl Acad Sci U S A
2004
;
101
:
1725
–30.
79
Ali IU, Schriml LM, Dean M. Mutational spectra of PTEN/MMAC1 gene: a tumor suppressor with lipid phosphatase activity.
J Natl Cancer Inst
1999
;
91
:
1922
–32.
80
Mutter GL, Lin MC, Fitzgerald JT, et al. Altered PTEN expression as a diagnostic marker for the earliest endometrial precancers.
J Natl Cancer Inst
2000
;
92
:
924
–30.
81
Massion PP, Taflan PM, Shyr Y, et al. Early involvement of the phosphatidylinositol 3-kinase/Akt pathway in lung cancer progression.
Am J Respir Crit Care Med
2004
;
170
:
1088
–94.
82
Tsao AS, McDonnell T, Lam S, et al. Increased phospho-AKT (Ser(473)) expression in bronchial dysplasia: implications for lung cancer prevention studies.
Cancer Epidemiol Biomarkers Prev
2003
;
12
:
660
–4.
83
Balsara BR, Pei J, Mitsuuchi Y, et al. Frequent activation of AKT in non-small cell lung carcinomas and preneoplastic bronchial lesions.
Carcinogenesis
2004
;
25
:
2053
–9.
84
West KA, Linnoila IR, Belinsky SA, Harris CC, Dennis PA. Tobacco carcinogen-induced cellular transformation increases activation of the phosphatidylinositol 3′-kinase/Akt pathway in vitro and in vivo.
Cancer Res
2004
;
64
:
446
–51.
85
Seki N, Takasu T, Mandai K, et al. Expression of eukaryotic initiation factor 4E in atypical adenomatous hyperplasia and adenocarcinoma of the human peripheral lung.
Clin Cancer Res
2002
;
8
:
3046
–53.
86
Roy HK, Olusola BF, Clemens DL, et al. AKT proto-oncogene overexpression is an early event during sporadic colon carcinogenesis.
Carcinogenesis
2002
;
23
:
201
–5.
87
Rosenwald IB, Chen JJ, Wang S, Savas L, London IM, Pullman J. Up-regulation of protein synthesis initiation factor eIF-4E is an early event during colon carcinogenesis.
Oncogene
1999
;
18
:
2507
–17.
88
Berkel HJ, Turbat-Herrera EA, Shi R, de Benedetti A. Expression of the translation initiation factor eIF4E in the polyp-cancer sequence in the colon.
Cancer Epidemiol Biomarkers Prev
2001
;
10
:
663
–6.
89
Dhawan P, Singh AB, Ellis DL, Richmond A. Constitutive activation of Akt/protein kinase B in melanoma leads to up-regulation of nuclear factor-κB and tumor progression.
Cancer Res
2002
;
62
:
7335
–42.
90
Stahl JM, Sharma A, Cheung M, et al. Deregulated Akt3 activity promotes development of malignant melanoma.
Cancer Res
2004
;
64
:
7002
–10.
91
Nathan CA, Leskov IL, Lin M, et al. COX-2 expression in dysplasia of the head and neck: correlation with elF4E.
Cancer
2001
;
92
:
1888
–95.
92
Wislez M, Spencer ML, Izzo JG, et al. Inhibition of mammalian target of rapamycin reverses alveolar epithelial neoplasia induced by oncogenic K-ras.
Cancer Res
2005
;
65
:
3226
–35.
93
Moy B, Goss PE. Estrogen receptor pathway: resistance to endocrine therapy and new therapeutic approaches.
Clin Cancer Res
2006
;
12
:
4790
–3.
94
Boulay A, Rudloff J, Ye J, et al. Dual inhibition of mTOR and estrogen receptor signaling in vitro induces cell death in models of breast cancer.
Clin Cancer Res
2005
;
11
:
5319
–28.
95
Soltoff SP, Carraway KL III, Prigent SA, Gullick WG, Cantley LC. ErbB3 is involved in activation of phosphatidylinositol 3-kinase by epidermal growth factor.
Mol Cell Biol
1994
;
14
:
3550
–8.
96
Namba R, Young LJ, Abbey CK, et al. Rapamycin inhibits growth of premalignant and malignant mammary lesions in a mouse model of ductal carcinoma in situ.
Clin Cancer Res
2006
;
12
:
2613
–21.
97
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.
98
Liu MY, Poellinger L, Walker CL. Up-regulation of hypoxia-inducible factor 2α in renal cell carcinoma associated with loss of Tsc-2 tumor suppressor gene.
Cancer Res
2003
;
63
:
2675
–80.
99
Mak BC, Yeung RS. The tuberous sclerosis complex genes in tumor development.
Cancer Invest
2004
;
22
:
588
–603.
100
Mathew T, Kreis H, Friend P. Two-year incidence of malignancy in sirolimus-treated renal transplant recipients: results from five multicenter studies.
Clin Transplant
2004
;
18
:
446
–9.
101
Euvrard S, Ulrich C, Lefrancois N. Immunosuppressants and skin cancer in transplant patients: focus on rapamycin.
Dermatol Surg
2004
;
30
:
628
–33.
102
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.
103
Athar M, Tang X, Lee JL, Kopelovich L, Kim AL. Hedgehog signalling in skin development and cancer.
Exp Dermatol
2006
;
15
:
667
–77.
104
Marti HP, Frey FJ. Nephrotoxicity of rapamycin: an emerging problem in clinical medicine.
Nephrol Dial Transplant
2005
;
20
:
13
–5.
105
Wattenberg LW, Wiedmann TS, Estensen RD, et al. Chemoprevention of pulmonary carcinogenesis by brief exposures to aerosolized budesonide or beclomethasone dipropionate and by the combination of aerosolized budesonide and dietary myo-inositol.
Carcinogenesis
2000
;
21
:
179
–82.
106
Wattenberg LW, Wiedmann TS, Estensen RD, Zimmerman CL, Steele VE, Kelloff GJ. Chemoprevention of pulmonary carcinogenesis by aerosolized budesonide in female A/J mice.
Cancer Res
1997
;
57
:
5489
–92.
107
Sharma S, Gao P, Steele VE. The chemopreventive efficacy of inhaled oltipraz particulates in the B[a]P-induced A/J mouse lung adenoma model.
Carcinogenesis
2006
;
27
:
1721
–7.
108
Lam S, leRiche JC, McWilliams A, et al. A randomized phase IIb trial of pulmicort turbuhaler (budesonide) in people with dysplasia of the bronchial epithelium.
Clin Cancer Res
2004
;
10
:
6502
–11.
109
Waldrep JC. New aerosol drug delivery systems for the treatment of immune-mediated pulmonary diseases.
Drugs Today (Barc)
1998
;
34
:
549
–61.
110
Fan QW, Knight ZA, Goldenberg DD, et al. A dual PI3 kinase/mTOR inhibitor reveals emergent efficacy in glioma.
Cancer Cell
2006
;
9
:
341
–9.
111
Beevers CS, Li F, Liu L, Huang S. Curcumin inhibits the mammalian target of rapamycin-mediated signaling pathways in cancer cells.
Int J Cancer
2006
;
119
:
757
–64.
112
Chaudhary LR, Hruska KA. Inhibition of cell survival signal protein kinase B/Akt by curcumin in human prostate cancer cells.
J Cell Biochem
2003
;
89
:
1
–5.
113
Haider UG, Roos TU, Kontaridis MI, et al. Resveratrol inhibits angiotensin II- and epidermal growth factor-mediated Akt activation: role of Gab1 and Shp2.
Mol Pharmacol
2005
;
68
:
41
–8.
114
Tang FY, Nguyen N, Meydani M. Green tea catechins inhibit VEGF-induced angiogenesis in vitro through suppression of VE-cadherin phosphorylation and inactivation of Akt molecule.
Int J Cancer
2003
;
106
:
871
–8.
115
Gong L, Li Y, Nedeljkovic-Kurepa A, Sarkar FH. Inactivation of NF-κB by genistein is mediated via Akt signaling pathway in breast cancer cells.
Oncogene
2003
;
22
:
4702
–9.
116
Chinni SR, Sarkar FH. Akt inactivation is a key event in indole-3-carbinol-induced apoptosis in PC-3 cells.
Clin Cancer Res
2002
;
8
:
1228
–36.
117
Rahman KW, Sarkar FH. Inhibition of nuclear translocation of nuclear factor-κB contributes to 3,3′-diindolylmethane-induced apoptosis in breast cancer cells.
Cancer Res
2005
;
65
:
364
–71.
118
Sharma RA, Gescher AJ, Steward WP. Curcumin: the story so far.
Eur J Cancer
2005
;
41
:
1955
–68.
119
Chun KH, Kosmeder JW II, Sun S, et al. Effects of deguelin on the phosphatidylinositol 3-kinase/Akt pathway and apoptosis in premalignant human bronchial epithelial cells.
J Natl Cancer Inst
2003
;
95
:
291
–302.
120
Leng J, Han C, Demetris AJ, Michalopoulos GK, Wu T. Cyclooxygenase-2 promotes hepatocellular carcinoma cell growth through Akt activation: evidence for Akt inhibition in celecoxib-induced apoptosis.
Hepatology
2003
;
38
:
756
–68.
121
Kulp SK, Yang YT, Hung CC, et al. 3-phosphoinositide-dependent protein kinase-1/Akt signaling represents a major cyclooxygenase-2-independent target for celecoxib in prostate cancer cells.
Cancer Res
2004
;
64
:
1444
–51.
122
Hsu AL, Ching TT, Wang DS, Song X, Rangnekar VM, Chen CS. The cyclooxygenase-2 inhibitor celecoxib induces apoptosis by blocking Akt activation in human prostate cancer cells independently of Bcl-2.
J Biol Chem
2000
;
275
:
11397
–403.
123
Kelloff GJ, Boone CW, Crowell JA, et al. New agents for cancer chemoprevention.
J Cell Biochem Suppl
1996
;
26
:
1
–28.
124
Baur JA, Pearson KJ, Price NL, et al. Resveratrol improves health and survival of mice on a high-calorie diet.
Nature
2006
;
444
:
337
–42.
125
Han S, Roman J. Rosiglitazone suppresses human lung carcinoma cell growth through PPARγ-dependent and PPARγ-independent signal pathways.
Mol Cancer Ther
2006
;
5
:
430
–7.
126
Shaw RJ, Lamia KA, Vasquez D, et al. The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin.
Science
2005
;
310
:
1642
–6.
127
Schneider MB, Matsuzaki H, Haorah J, et al. Prevention of pancreatic cancer induction in hamsters by metformin.
Gastroenterology
2001
;
120
:
1263
–70.
128
Anisimov VN, Egormin PA, Bershtein LM, et al. Metformin decelerates aging and development of mammary tumors in HER-2/neu transgenic mice.
Bull Exp Biol Med
2005
;
139
:
721
–3.
129
Freelove R, Walling AD. Pancreatic cancer: diagnosis and management.
Am Fam Physician
2006
;
73
:
485
–92.
130
El-Serag HB, Hampel H, Javadi F. The association between diabetes and hepatocellular carcinoma: a systematic review of epidemiologic evidence.
Clin Gastroenterol Hepatol
2006
;
4
:
369
–80.
131
Willett WC. Implications of total energy intake for epidemiologic studies of breast and large-bowel cancer.
Am J Clin Nutr
1987
;
45
:
354
–60.
132
Heilbronn LK, Ravussin E. Calorie restriction and aging: review of the literature and implications for studies in humans.
Am J Clin Nutr
2003
;
78
:
361
–9.
133
Heilbronn LK, Ravussin E. Calorie restriction extends life span-but which calories?
PLoS Med
2005
;
2
:
e231
.
134
Kagawa Y. Impact of Westernization on the nutrition of Japanese: changes in physique, cancer, longevity and centenarians.
Prev Med
1978
;
7
:
205
–17.
135
Platz EA. Energy imbalance and prostate cancer.
J Nutr
2002
;
132
:
3471
–81S.
136
Platz EA, Leitzmann MF, Michaud DS, Willett WC, Giovannucci E. Interrelation of energy intake, body size, and physical activity with prostate cancer in a large prospective cohort study.
Cancer Res
2003
;
63
:
8542
–8.
137
Chang SC, Ziegler RG, Dunn B, et al. Association of energy intake and energy balance with postmenopausal breast cancer in the prostate, lung, colorectal, and ovarian cancer screening trial.
Cancer Epidemiol Biomarkers Prev
2006
;
15
:
334
–41.
138
Slattery ML, Potter J, Caan B, et al. Energy balance and colon cancer—beyond physical activity.
Cancer Res
1997
;
57
:
75
–80.
139
Hursting SD, Lavigne JA, Berrigan D, Perkins SN, Barrett JC. Calorie restriction, aging, and cancer prevention: mechanisms of action and applicability to humans.
Annu Rev Med
2003
;
54
:
131
–52.
140
Sharp ZD, Bartke A. Evidence for down-regulation of phosphoinositide 3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR)-dependent translation regulatory signaling pathways in Ames dwarf mice.
J Gerontol A Biol Sci Med Sci
2005
;
60
:
293
–300.
141
Koubova J, Guarente L. How does calorie restriction work?
Genes Dev
2003
;
17
:
313
–21.
142
Hahn WC, Weinberg RA. Rules for making human tumor cells.
N Engl J Med
2002
;
347
:
1593
–603.
143
Lee CK, Klopp RG, Weindruch R, Prolla TA. Gene expression profile of aging and its retardation by caloric restriction.
Science
1999
;
285
:
1390
–3.
144
Vellai T, Takacs-Vellai K, Zhang Y, Kovacs AL, Orosz L, Muller F. Genetics: influence of TOR kinase on life span in C. elegans.
Nature
2003
;
426
:
620
.
145
Kapahi P, Zid BM, Harper T, Koslover D, Sapin V, Benzer S. Regulation of life span in Drosophila by modulation of genes in the TOR signaling pathway.
Curr Biol
2004
;
14
:
885
–90.
146
Lorberg A, Hall MN. TOR: the first 10 years.
Curr Top Microbiol Immunol
2004
;
279
:
1
–18.
147
Stoyanova R, Clapper ML, Bellacosa A, et al. Altered gene expression in phenotypically normal renal cells from carriers of tumor suppressor gene mutations.
Cancer Biol Ther
2004
;
3
:
1313
–21.
148
West KA, Brognard J, Clark AS, et al. Rapid Akt activation by nicotine and a tobacco carcinogen modulates the phenotype of normal human airway epithelial cells.
J Clin Invest
2003
;
111
:
81
–90.
149
Amornphimoltham P, Sriuranpong V, Patel V, et al. Persistent activation of the Akt pathway in head and neck squamous cell carcinoma: a potential target for UCN-01.
Clin Cancer Res
2004
;
10
:
4029
–37.
150
Nathan CA, Franklin S, Abreo FW, et al. Expression of eIF4E during head and neck tumorigenesis: possible role in angiogenesis.
Laryngoscope
1999
;
109
:
1253
–8.
151
Harris JC, Clarke PA, Awan A, Jankowski J, Watson SA. An antiapoptotic role for gastrin and the gastrin/CCK-2 receptor in Barrett's esophagus.
Cancer Res
2004
;
64
:
1915
–9.
152
Chandy B, Abreo F, Nassar R, Stucker FJ, Nathan CA. Expression of the proto-oncogene eIF4E in inflammation of the oral cavity.
Otolaryngol Head Neck Surg
2002
;
126
:
290
–5.
153
Michaylira CZ, Nakagawa H. Hypoxic microenvironment as a cradle for melanoma development and progression.
Cancer Biol Ther
2006
;
5
:
476
–9.
154
Yilmaz OH, Valdez R, Theisen BK, et al. PTEN dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells.
Nature
2006
;
441
:
418
–9.
155
Fang MZ, Wang Y, Ai N, et al. Tea polyphenol (−)-epigallocatechin-3-gallate inhibits DNA methyltransferase and reactivates methylation-silenced genes in cancer cell lines.
Cancer Res
2003
;
63
:
7563
–70.
156
Kelloff GJ. Perspectives on cancer chemoprevention research and drug development.
Adv Cancer Res
2000
;
78
:
199
–334.
157
Kelloff GJ, Lippman SM, Dannenberg AJ, et al. Progress in chemoprevention drug development: the promise of molecular biomarkers for prevention of intraepithelial neoplasia and cancer-a plan to move forward.
Clin Cancer Res
2006
;
12
:
3661
–97.
158
Bettuzzi S, Brausi M, Rizzi F, Castagnetti G, Peracchia G, Corti A. Chemoprevention of human prostate cancer by oral administration of green tea catechins in volunteers with high-grade prostate intraepithelial neoplasia: a preliminary report from a one-year proof-of-principle study.
Cancer Res
2006
;
66
:
1234
–40.
159
Scaltriti M, Belloni L, Caporali A, et al. Molecular classification of green tea catechin-sensitive and green tea catechin-resistant prostate cancer in the TRAMP mice model by quantitative real-time PCR gene profiling.
Carcinogenesis
2006
;
27
:
1047
–53.
160
Aggarwal BB, Shishodia S. Molecular targets of dietary agents for prevention and therapy of cancer.
Biochem Pharmacol
2006
;
71
:
1397
–421.
161
Lee WJ, Shim JY, Zhu BT. Mechanisms for the inhibition of DNA methyltransferases by tea catechins and bioflavonoids.
Mol Pharmacol
2005
;
68
:
1018
–30.
162
Weyant MJ, Carothers AM, Dannenberg AJ, Bertagnolli MM. (+)-Catechin inhibits intestinal tumor formation and suppresses focal adhesion kinase activation in the min/+ mouse.
Cancer Res
2001
;
61
:
118
–25.
163
Ebeler SE, Brenneman CA, Kim GS, et al. Dietary catechin delays tumor onset in a transgenic mouse model.
Am J Clin Nutr
2002
;
76
:
865
–72.
164
Fang MZ, Chen D, Sun Y, Jin Z, Christman JK, Yang CS. Reversal of hypermethylation and reactivation of p16INK4a, RARβ, and MGMT genes by genistein and other isoflavones from soy.
Clin Cancer Res
2005
;
11
:
7033
–41.
165
Wang J, Eltoum IE, Lamartiniere CA. Dietary genistein suppresses chemically induced prostate cancer in Lobund-Wistar rats.
Cancer Lett
2002
;
186
:
11
–8.
166
Tatsuta M, Iishi H, Baba M, Yano H, Uehara H, Nakaizumi A. Attenuation by genistein of sodium-chloride-enhanced gastric carcinogenesis induced by N-methyl-N′-nitro-N-nitrosoguanidine in Wistar rats.
Int J Cancer
1999
;
80
:
396
–9.
167
Kelloff GJ, Crowell JA, Hawk ET, et al. Clinical development plans II: genistein.
J Cell Biochem Suppl
1996
;
26
:
114
–26.
168
Lee WJ, Zhu BT. Inhibition of DNA methylation by caffeic acid and chlorogenic acid, two common catechol-containing coffee polyphenols.
Carcinogenesis
2006
;
27
:
269
–77.
169
Fiala ES, Staretz ME, Pandya GA, El-Bayoumy K, Hamilton SR. Inhibition of DNA cytosine methyltransferase by chemopreventive selenium compounds, determined by an improved assay for DNA cytosine methyltransferase and DNA cytosine methylation.
Carcinogenesis
1998
;
19
:
597
–604.
170
Myzak MC, Dashwood WM, Orner GA, Ho E, Dashwood RH. Sulforaphane inhibits histone deacetylase in vivo and suppresses tumorigenesis in Apc-minus mice.
FASEB J
2006
;
20
:
506
–8.
171
Myzak MC, Hardin K, Wang R, Dashwood RH, Ho E. Sulforaphane inhibits histone deacetylase activity in BPH-1, LnCaP and PC-3 prostate epithelial cells.
Carcinogenesis
2006
;
27
:
811
–9.
172
Myzak MC, Karplus PA, Chung FL, Dashwood RH. A novel mechanism of chemoprotection by sulforaphane: inhibition of histone deacetylase.
Cancer Res
2004
;
64
:
5767
–74.
173
Fahey JW, Haristoy X, Dolan PM, et al. Sulforaphane inhibits extracellular, intracellular, and antibiotic-resistant strains of Helicobacter pylori and prevents benzo[a]pyrene-induced stomach tumors.
Proc Natl Acad Sci U S A
2002
;
99
:
7610
–5.
174
Wargovich MJ, Chen CD, Jimenez A, et al. Aberrant crypts as a biomarker for colon cancer: evaluation of potential chemopreventive agents in the rat.
Cancer Epidemiol Biomarkers Prev
1996
;
5
:
355
–60.
175
Pereira MA. Chemoprevention of diethylnitrosamine-induced liver foci and hepatocellular adenomas in C3H mice.
Anticancer Res
1995
;
15
:
1953
–6.
176
el-Bayoumy K, Chae YH, Upadhyaya P, Ip C. Chemoprevention of mammary cancer by diallyl selenide, a novel organoselenium compound.
Anticancer Res
1996
;
16
:
2911
–5.
177
Hecht SS. Chemoprevention of cancer by isothiocyanates, modifiers of carcinogen metabolism.
J Nutr
1999
;
129
:
768
–74S.
178
Nishikawa A, Furukawa F, Lee IS, Tanaka T, Hirose M. Potent chemopreventive agents against pancreatic cancer.
Curr Cancer Drug Targets
2004
;
4
:
373
–84.
179
Cohen LA, Amin S, Marks PA, Rifkind RA, Desai D, Richon VM. Chemoprevention of carcinogen-induced mammary tumorigenesis by the hybrid polar cytodifferentiation agent, suberanilohydroxamic acid (SAHA).
Anticancer Res
1999
;
19
:
4999
–5005.
180
Desai D, Das A, Cohen L, el-Bayoumy K, Amin S. Chemopreventive efficacy of suberoylanilide hydroxamic acid (SAHA) against 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced lung tumorigenesis in female A/J mice.
Anticancer Res
2003
;
23
:
499
–503.
181
Zhu P, Martin E, Mengwasser J, Schlag P, Janssen KP, Gottlicher M. Induction of HDAC2 expression upon loss of APC in colorectal tumorigenesis.
Cancer Cell
2004
;
5
:
455
–63.
182
Balasubramanyam K, Varier RA, Altaf M, et al. Curcumin, a novel p300/CREB-binding protein-specific inhibitor of acetyltransferase, represses the acetylation of histone/nonhistone proteins and histone acetyltransferase-dependent chromatin transcription.
J Biol Chem
2004
;
279
:
51163
–71.
183
Kelloff GJ, Crowell JA, Hawk ET, et al. Clinical development plans II: curcumin.
J Cell Biochem Suppl
1996
;
26
:
72
–85.
184
Bava SV, Puliappadamba VT, Deepti A, Nair A, Karunagaran D, Anto RJ. Sensitization of Taxol-induced apoptosis by curcumin involves down-regulation of nuclear factor-κB and the serine/threonine kinase Akt and is independent of tubulin polymerization.
J Biol Chem
2005
;
280
:
6301
–8.
185
Ito C, Itoigawa M, Kojima N, et al. Cancer chemopreventive activity of rotenoids from Derris trifoliata.
Planta Med
2004
;
70
:
8
–11.
186
Murillo G, Kosmeder JW II, Pezzuto JM, Mehta RG. Deguelin suppresses the formation of carcinogen-induced aberrant crypt foci in the colon of CF-1 mice.
Int J Cancer
2003
;
104
:
7
–11.
187
Carbo N, Costelli P, Baccino FM, Lopez-Soriano FJ, Argiles JM. Resveratrol, a natural product present in wine, decreases tumour growth in a rat tumour model.
Biochem Biophys Res Commun
1999
;
254
:
739
–43.
188
Li ZG, Hong T, Shimada Y, et al. Suppression of N-nitrosomethylbenzylamine (NMBA)-induced esophageal tumorigenesis in F344 rats by resveratrol.
Carcinogenesis
2002
;
23
:
1531
–6.
189
Schneider Y, Duranton B, Gosse F, Schleiffer R, Seiler N, Raul F. Resveratrol inhibits intestinal tumorigenesis and modulates host-defense-related gene expression in an animal model of human familial adenomatous polyposis.
Nutr Cancer
2001
;
39
:
102
–7.
190
Whitsett TG, Jr., Carpenter DM, Lamartiniere CA. Resveratrol, but not EGCG, in the diet suppresses DMBA-induced mammary cancer in rats.
J Carcinog
2006
;
5
:
15
.
191
Banerjee S, Bueso-Ramos C, Aggarwal BB. Suppression of 7,12-dimethylbenz(a)anthracene-induced mammary carcinogenesis in rats by resveratrol: role of nuclear factor-κB, cyclooxygenase 2, and matrix metalloprotease 9.
Cancer Res
2002
;
62
:
4945
–54.
192
Provinciali M, Re F, Donnini A, et al. Effect of resveratrol on the development of spontaneous mammary tumors in HER-2/neu transgenic mice.
Int J Cancer
2005
;
115
:
36
–45.
193
Khalil MY, Grandis JR, Shin DM. Targeting epidermal growth factor receptor: novel therapeutics in the management of cancer.
Expert Rev Anticancer Ther
2003
;
3
:
367
–80.
194
Saba NF, Khuri FR, Shin DM. Targeting the epidermal growth factor receptor. Trials in head and neck and lung cancer.
Oncology (Huntingt)
2006
;
20
:
153
–61;discussion 162, 166, 169 passim.
195
Dannenberg AJ, Lippman SM, Mann JR, Subbaramaiah K, DuBois RN. Cyclooxygenase-2 and epidermal growth factor receptor: pharmacologic targets for chemoprevention.
J Clin Oncol
2005
;
23
:
254
–66.
196
Kaneuchi M, Sasaki M, Tanaka Y, Yamamoto R, Sakuragi N, Dahiya R. Resveratrol suppresses growth of Ishikawa cells through down-regulation of EGF.
Int J Oncol
2003
;
23
:
1167
–72.
197
Spindler SR. Use of microarray biomarkers to identify longevity therapeutics.
Aging Cell
2006
;
5
:
39
–50.