Purpose: To determine whether mTORC2 and rapamycin-insensitive (RI)-mTORC1 complexes are present in acute myeloid leukemia (AML) cells and to examine the effects of dual mTORC2/mTORC1 inhibition on primitive AML leukemic progenitors.

Experimental Design: Combinations of different experimental approaches were used, including immunoblotting to detect phosphorylated/activated forms of elements of the mTOR pathway in leukemic cell lines and primary AML blasts; cell-proliferation assays; direct assessment of mRNA translation in polysomal fractions of leukemic cells; and clonogenic assays in methylcellulose to evaluate leukemic progenitor-colony formation.

Results: mTORC2 complexes are active in AML cells and play critical roles in leukemogenesis. RI-mTORC1 complexes are also formed and regulate the activity of the translational repressor 4E-BP1 in AML cells. OSI-027 blocks mTORC1 and mTORC2 activities and suppresses mRNA translation of cyclin D1 and other genes that mediate proliferative responses in AML cells. Moreover, OSI-027 acts as a potent suppressor of primitive leukemic precursors from AML patients and is much more effective than rapamycin in eliciting antileukemic effects in vitro.

Conclusions: Dual targeting of mTORC2 and mTORC1 results in potent suppressive effects on primitive leukemic progenitors from AML patients. Inhibition of the mTOR catalytic site with OSI-027 results in suppression of both mTORC2 and RI-mTORC1 complexes and elicits much more potent antileukemic responses than selective mTORC1 targeting with rapamycin. Clin Cancer Res; 17(13); 4378–88. ©2011 AACR.

Translational Relevance

mTOR provides an attractive cellular target for the development of new therapeutic approaches for hematologic malignancies, but a major limitation of the classic mTOR inhibitor rapamycin and related rapalogues is their selectivity for mTORC1 and their inability to effectively inhibit mTORC2. In the present study, we provide evidence for constitutive formation of mTORC2 and rapamycin-insensitive (RI)-mTORC1 complexes in AML cells and establish that dual mTORC2/mTORC1 inhibition using a novel catalytic inhibitor of mTOR, OSI-027, has greater antileukemic effects than rapamycin on AML progenitors. These findings provide the basis for the development of clinical–translational efforts involving mTORC1/mTORC2 inhibitors, such as OSI-027, or combinations of such agents with chemotherapeutic agents for the treatment of AML.

Despite recent advances in the field, acute myeloid leukemia (AML) remains highly fatal. Although a large number of AML patients achieve clinical remission with current treatment regimens, the majority of patients eventually relapse and die (1, 2). Undoubtedly, there is an urgent need to develop new therapeutic approaches to overcome the resistance that AML cells develop to chemotherapy (1, 2). Several mechanisms underlying the molecular pathogenesis of AML have been uncovered. It is now known that AML results from mutations in classes of genes that affect both proliferation/survival and differentiation/apoptosis (3). Changes in transcriptional control of hematopoietic cells and mutations involving different proto-oncogenes or tumor suppressors account for the deregulation of normal myelopoiesis (3). Such changes ultimately result in the activation of cellular pathways that mediate signals, which promote cell growth and/or maintain survival, leading to leukemogenesis (4, 5).

The phosphoinositide 3-kinase (PI3K)/Akt pathway is a critical network of signals that control key cellular functions, including gene transcription, differentiation and mRNA translation, and survival (6–8). This pathway is frequently dysregulated in various tumors (6–8). Extensive work over the years has established that activation of PI3K/Akt signaling results in a growth advantage for tumor cells by promoting cell proliferation and anti-apoptotic signals (6, 7, 9). There is evidence that PI3K/Akt activation contributes to tumor cell resistance to antineoplastic therapies, and overexpression or constitutive activation of components of this pathway may contribute to a poorer prognosis in different malignancies (reviewed in refs. 10, 11).

mTOR is a kinase activated downstream of PI3K/Akt that plays critical and essential roles in the regulation of mRNA translation, cell-cycle progression, and growth of mammalian cells (6, 12–14). Because mTOR mediates pro-growth signals in malignant cells, its pharmacologic targeting is currently under extensive investigation for the treatment of various malignancies, including leukemias (11, 15). In fact, 2 rapamycin-related drugs, temsirolimus and everolimus, have significant activity in the treatment of renal cell carcinoma (16, 17) and were recently approved by the Food and Drug Administration (FDA) for the treatment of this disease.

mTOR inhibitors that are currently approved for clinical use (e.g., rapamycin, temsirolimus, everolimus) are allosteric inhibitors of mTORC1 but not of mTORC2 (18), however the precise functional roles of mTORC2 and other rapamycin-insensitive (RI) complexes in AML cells is unknown. In the present study, we provided direct evidence for the presence and activity of mTORC2 complexes in AML cell lines and primary AML cells. Furthermore, we established that mTOR-mediated phosphorylation of RI sites on the translational repressor 4E-BP1 plays a key role in mRNA translation in AML cells and regulates cyclin D1 expression. We show that activation of mTORC2 and RI-mTORC1 complexes is selectively inhibited by a novel dual mTORC2/mTORC1 inhibitor OSI-027, a selective ATP-competitive catalytic site inhibitor of mTOR (19), but not by the allosteric inhibitor rapamycin. Our studies establish that OSI-027 induces potent antileukemic effects on a variety of AML cell lines as well as on primary leukemic progenitors [lymphoid colony-forming unit (CFU-L)] from AML patients. In addition, OSI-027 treatment enhances chemotherapy-induced antileukemic responses in vitro. Altogether, our data show critical roles for mTORC2 and RI-mTORC1 complexes in leukemic cell growth and survival, and provide a rationale for the clinical development of dual mTORC2/mTORC1 inhibitors for the treatment of AML.

Cells and reagents

The KG-1, HL-60, KBM-3B, and U937 human leukemia cell lines were cultured in RPMI 1640 supplemented with 10% FBS and 1% l-glutamine. ML-1 cells were grown in Dulbecco's modified Eagle medium (DMEM) supplemented with 10% FBS and 1% l-glutamine. U937, KG-1, and HL60 cells were obtained from the American Type Culture Collection (ATCC). ML-1 and KBM-3B cells were generously provided by E. Harlow, Cold Spring Harbor Laboratories. Rapamycin was purchased from Calbiochem/EMD. Cytarabine (AraC) was purchased from Sigma. All antibodies were purchased from Cell Signaling Technologies except the antibody against PDCD4, which was purchased from Rockland, and the antibody against glyceraldehyde 3-phosphate dehydrogenase (GAPDH), which was purchased from Millipore.

Measurement of proliferation

Inhibition of proliferation was measured using the Cell Titer Glo Assay (Promega Corporation), as noted in figure legends. To generate dose–response curves, cell lines were seeded at a density of 5,000 cells per well in a 96-well plate. After 24 hours of plating, cells were dosed with varying concentrations of either OSI-027 or rapamycin. The signal for Cell Titer Glo Assay was determined 72 hours after dosing and normalized to that of vehicle-treated controls. Inhibition of proliferation, relative to vehicle-treated controls, was expressed as a fraction of 1 and graphed using PRISM software (Graphpad Software).

Cell lysis and immunoblotting

For the immunoblotting experiments, cells were treated with rapamycin (20 nmol/L), OSI-027, or dimethyl sulfoxide (DMSO), used as control for untreated cells, for the indicated times and lysed in phosphorylation lysis buffer. Immunoblotting using an enhanced chemiluminescence (ECL) method was done as in our previous studies (20, 21).

Isolation of polysomal RNA and quantitative reverse transcriptase PCR in polysomal fractions

U937 cells were treated with DMSO, rapamycin, or OSI-027, and polysomal fractionation and quantitative reverse transcriptase PCR (RT-PCR) were carried out with slight modifications of the protocol used in our previous studies (22, 23).

Studies with primary AML samples and hematopoietic progenitor cell assays

Peripheral blood was obtained from patients with AML after obtaining informed consent, following approval by the Institutional Review Board of the Northwestern University. Cells were separated over Ficoll–Hypaque and cultured with the indicated concentrations of rapamycin, OSI-027, and/or AraC, and leukemic progenitor (CFU-L) colony formation was assessed in clonogenic assays in methylcellulose (21, 24–27).

Short hairpin RNA-mediated knockdown

U937 cells were transduced with lentiviral particles (28–30), expressing either control-shRNA or S6K1 (p70 S6 kinase α) short hairpin RNAs (shRNA; Santa Cruz Biotechnology, Inc.) according to the manufacturer's instructions. Briefly, 5 × 104 cells/mL were infected with the abovementioned lentiviruses, and selected in puromycin, as recommended.

In initial studies, we determined whether functional mTORC1 and mTORC2 complexes are present in AML cells and evaluated the effects of OSI-027 or rapamycin on their activation. Using the AML cell line U937 or circulating primary leukemic AML blasts, we examined the phosphorylation of Akt on serine 473 (Ser473), a marker of mTORC2/Akt activity. As shown in Figure 1, Akt phosphorylation was detectable in AML cells, and it was completely inhibited by treatment of cells with OSI-027 (Fig. 1A and B). On the other hand, rapamycin did not inhibit such phosphorylation (Fig. 1A and B) and, in fact, it had a slight stimulatory effect in U937 cells (Fig. 1A), a finding consistent with the previously described rapamycin-mediated inhibition of the S6K–IRS negative feedback loop, which results in enhanced signaling through the mTORC2/Akt node (reviewed in ref. 31). Furthermore, we examined the phosphorylation of mTOR on Ser2448, a marker for mTORC1 activity, as well as the phosphorylation of the protein on Ser2481, a marker for mTORC2 activity (32). Phosphorylation of mTOR on both Ser2448 and Ser2481 was detectable in U937 cells (Fig. 1C and D), reflecting the presence of both mTORC1 and mTORC2 complexes. Ser2448 phosphorylation was inhibited by treatment of cells with either rapamycin or OSI-027 (Fig. 1C), consistent with the expected sensitivity of mTORC1 complexes to both inhibitors. However, only OSI-027 inhibited phosphorylation on Ser2481, showing mTORC2 targeting (Fig. 1D). Similar results were obtained when the AML cell line, KG-1 (Fig. 1E), or primary leukemic blasts from a patient with AML (Fig. 1F) were studied. mTORC1 kinase activity could be also directly showed in in vitro kinase assays on anti-Raptor immunoprecipitates from U937 cell lysates by using S6K as a substrate (Supplementary Fig. S1). These data clearly established the presence of mTORC1 kinase activity and showed decreased phosphorylation of S6K substrate upon treatment with OSI-027 (Supplementary Fig. S1A). Notably, in these studies it was also shown that, in contrast to rapamycin, the suppression of mTOR kinase activity occurs without disruption of the protein–protein interactions in the mTORC1 complex, a finding consistent with ATP-competitive catalytic inhibition of mTOR by OSI-027 (Supplementary Fig. S1B).

Figure 1.

Presence of mTORC2 and mTORC1 complexes in AML cells and differential regulation by rapamycin or OSI-027. A, U937 cells were incubated with or without OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 90 minutes. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against the phosphorylated form of Akt on Ser473. Subsequently, the same blot was re-probed with antibodies against Akt or GAPDH, to control for equal protein loading. B, primary leukemic blasts from a patient with AML were incubated with or without OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 90 minutes. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against the phosphorylated form of Akt on Ser473. Subsequently, the same blot was re-probed with antibodies against Akt or GAPDH, to control for protein loading. C and D, U937 cells were incubated with or without OSI-027 (5 μmol/L) or rapamycin (20 nmol/L) for 90 minutes. Cell lysates were resolved by SDS-PAGE and immunoblotted with antibodies against the phosphorylated form of mTOR on Ser2448 (C) or the phosphorylated form of mTOR on Ser2481 (D). The blots were then re-probed with an antibody against mTOR, to control for protein loading. E, KG-1 cells were incubated with or without OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 24 hours and cell lysates were resolved and immunoblotted with an antibody against the phosphorylated form of mTOR on Ser2481. The same blot was subsequently re-probed with an antibody against mTOR. F, leukemic blasts from a patient with AML were incubated with or without OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 90 minutes. Cell lysates were resolved by SDS-PAGE and then immunoblotted with an antibody against the phosphorylated form of mTOR on Ser2481. The blot was then re-probed with an antibody against mTOR.

Figure 1.

Presence of mTORC2 and mTORC1 complexes in AML cells and differential regulation by rapamycin or OSI-027. A, U937 cells were incubated with or without OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 90 minutes. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against the phosphorylated form of Akt on Ser473. Subsequently, the same blot was re-probed with antibodies against Akt or GAPDH, to control for equal protein loading. B, primary leukemic blasts from a patient with AML were incubated with or without OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 90 minutes. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against the phosphorylated form of Akt on Ser473. Subsequently, the same blot was re-probed with antibodies against Akt or GAPDH, to control for protein loading. C and D, U937 cells were incubated with or without OSI-027 (5 μmol/L) or rapamycin (20 nmol/L) for 90 minutes. Cell lysates were resolved by SDS-PAGE and immunoblotted with antibodies against the phosphorylated form of mTOR on Ser2448 (C) or the phosphorylated form of mTOR on Ser2481 (D). The blots were then re-probed with an antibody against mTOR, to control for protein loading. E, KG-1 cells were incubated with or without OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 24 hours and cell lysates were resolved and immunoblotted with an antibody against the phosphorylated form of mTOR on Ser2481. The same blot was subsequently re-probed with an antibody against mTOR. F, leukemic blasts from a patient with AML were incubated with or without OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 90 minutes. Cell lysates were resolved by SDS-PAGE and then immunoblotted with an antibody against the phosphorylated form of mTOR on Ser2481. The blot was then re-probed with an antibody against mTOR.

Close modal

A major effector of activated mTORC1 complexes is S6K, which, in turn, phosphorylates and activates several downstream effectors, including the S6 ribosomal protein (rpS6) and the eukaryotic initiation factor 4B (eIF4B; ref. 6, 13). When different AML cell lines or primary AML blasts were treated with rapamycin or OSI-027, there was complete suppression of phosphorylation of S6K on threonine 389 (Thr389), a site whose phosphorylation correlates with activation of its kinase domain (Fig. 2A–C). Consistent with the inhibitory effects of both mTOR inhibitors on S6K activity, phosphorylation of the downstream effector of S6K, rpS6, on both Ser235/236 and Ser240/244 was blocked by treatment of cells with either OSI-027 or rapamycin (Fig. 2D–I). Furthermore, we determined the effects of OSI-027 or rapamycin on PDCD4, a tumor-suppressor protein that acts as an inhibitor of cap-dependent translation by blocking the translation-initiation factor eIF4A (33). This protein was recently identified as a target of the mTOR/S6K cascade, and it was shown that S6K-mediated phosphorylation of PDCD4 results in its degradation by ubiquitin ligases (34), whereas its upregulation in response to BCR-ABL kinase inhibitors appears to mediate generation of antileukemic responses in Ph+ cells (35). Consistent with the suppressive effects of both rapamycin and OSI-027 on S6K activity (Fig. 2A–C), there was upregulation of PDCD4 expression in response to treatment of AML cells with either inhibitor (Fig. 3A and B). Thus, both rapamycin and OSI-027 upregulate PDCD4, suggesting that this S6K target is a mediator of the antileukemic responses elicited by targeting mTORC1 complexes. To define the functional consequences of inhibition of S6K phosphorylation in leukemic progenitors, experiments were conducted in which S6K1 was knocked down using S6K1-shRNA in a lentiviral vector (Fig. 3C). These studies showed substantial inhibition in leukemic CFU-L colony formation by S6K1 knockdown (Fig. 3D), suggesting an important role for S6K1 activity and suppression of PDCD4 in the growth of AML progenitors.

Figure 2.

Phosphorylation/activation of S6K and S6 ribosomal protein in AML cells. A–C, U937 cells (A), KG-1 cells (B), or primary leukemic blasts from an AML patient (C) were incubated with OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for the indicated times. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against the phosphorylated form of S6K on Thr389, or with antibodies against S6K or GAPDH, as indicated. D–I, U937 cells (D, G) KG-1 cells (E, H), or primary AML blasts (F, I) were incubated in the absence or presence of rapamycin (20 nmol/L) or OSI-027 (10 μmol/L) as indicated. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against the phosphorylated form of rpS6 on Ser235/236 (D–F), against the phosphorylated form of rpS6 on Ser240/244 (G–I), or against rpS6, as indicated.

Figure 2.

Phosphorylation/activation of S6K and S6 ribosomal protein in AML cells. A–C, U937 cells (A), KG-1 cells (B), or primary leukemic blasts from an AML patient (C) were incubated with OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for the indicated times. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against the phosphorylated form of S6K on Thr389, or with antibodies against S6K or GAPDH, as indicated. D–I, U937 cells (D, G) KG-1 cells (E, H), or primary AML blasts (F, I) were incubated in the absence or presence of rapamycin (20 nmol/L) or OSI-027 (10 μmol/L) as indicated. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against the phosphorylated form of rpS6 on Ser235/236 (D–F), against the phosphorylated form of rpS6 on Ser240/244 (G–I), or against rpS6, as indicated.

Close modal
Figure 3.

Effects of mTOR inhibition on the expression of the S6K-dependent PDCD4 tumor suppressor in AML cells and functional consequences of S6K knockdown. A and B, U937 (A) or KG-1 (B) cells were incubated in the absence (lane 1) or presence of OSI-027 at concentrations of either 5 μmol/L (lane 2) or 10 μmol/L (lane 3) or rapamycin (20 nmol/L; lane 4) for 24 hours. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against PDCD4. The same blots were re-probed with an antibody against tubulin, to control for protein loading. C, cell lysates from lentivirus transduced control (ctrl)-shRNA or S6K1-shRNA U937cells were resolved by SDS-PAGE and immunoblotted with an anti-S6K1 antibody. The same blot was then re-probed with an anti-GAPDH antibody to control for protein loading. D, leukemic CFU-L colony formation derived from control-shRNA and S6K1-shRNA U937 cells is shown. Data are expressed as percentages of colony formation of untreated control-shRNA transduced cells and represent means ± SE of 5 independent experiments.

Figure 3.

Effects of mTOR inhibition on the expression of the S6K-dependent PDCD4 tumor suppressor in AML cells and functional consequences of S6K knockdown. A and B, U937 (A) or KG-1 (B) cells were incubated in the absence (lane 1) or presence of OSI-027 at concentrations of either 5 μmol/L (lane 2) or 10 μmol/L (lane 3) or rapamycin (20 nmol/L; lane 4) for 24 hours. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against PDCD4. The same blots were re-probed with an antibody against tubulin, to control for protein loading. C, cell lysates from lentivirus transduced control (ctrl)-shRNA or S6K1-shRNA U937cells were resolved by SDS-PAGE and immunoblotted with an anti-S6K1 antibody. The same blot was then re-probed with an anti-GAPDH antibody to control for protein loading. D, leukemic CFU-L colony formation derived from control-shRNA and S6K1-shRNA U937 cells is shown. Data are expressed as percentages of colony formation of untreated control-shRNA transduced cells and represent means ± SE of 5 independent experiments.

Close modal

A key functional cellular response downstream of the mTOR pathway is the regulation of the initiation of cap-dependent mRNA translation, which occurs primarily via regulation of the 4E-BP1 repressor of mRNA translation (6, 13, 14). 4E-BP1 is phosphorylated in a hierarchic manner on Thr37/46, Ser65, and Thr70; and such phosphorylation is essential for the deactivation of the protein and its subsequent dissociation from the eukaryotic initiation factor 4E (eIF4E), so that cap-dependent mRNA translation can proceed (36, 37). Significant baseline levels of phosphorylation of 4E-BP1 on Thr37/46 and Ser65 were detectable in both U937 and KG-1 cells (Fig. 4A and B). This type of phosphorylation was completely blocked by treatment of cells with OSI-027, whereas treatment with rapamycin had no effect (Fig. 4A and B). Similar results were also seen when the phosphorylation of the protein on Thr70 was studied (Fig. 4C). OSI-027 was used at a maximal concentration of 10 μmol/L, which inhibits both mTORC1 and mTORC2 (19) and is readily secreted in plasma in preclinical rodent models (38). Rapamycin was dosed at a maximal concentration of 20 nmol/L, which is consistent with multiple published studies and exceeds the dose achievable in tumors, and the dose that required to inhibit mTORC1 function (38, 39). In dose–response experiments, we found that rapamycin failed to block Thr37/46 4E-BP1 phosphorylation even at very high, supra-normal concentrations (Fig. 4D), thus definitively establishing that 4E-BP1 phosphorylation at Thr37/46 is an RI event. A similar pattern of 4E-BP1 dephosphorylation in response to OSI-027 or rapamycin was seen in studies using primary AML leukemic blasts (Fig. 4E and F; Supplementary Fig. S2). Consistent with the complete suppression of 4E-BP1 phosphorylation, OSI-027 treatment resulted in the formation of 4E–BP1–eIF4E complexes that suppress cap-dependent mRNA translation, while it inhibited the formation of eIF4E–eIF4G complexes that are required for initiation of translation (Fig. 4G). On the other hand, rapamycin had much weaker effects in promoting formation of 4E–BP1–eIF4E complexes and it did not disrupt formation of eIF4E–eIF4G complexes (Fig. 4G). Thus, OSI-27 is a potent inhibitor of mTOR activity in AML cells and suppresses formation of complexes that regulate cap-dependent mRNA translation.

Figure 4.

OSI-027, but not rapamycin, inhibits phosphorylation of the 4E-BP1 repressor of mRNA translation and blocks formation of complexes required for cap-dependent mRNA translation. A and B, U937 or KG-1 cells were incubated in the absence or presence of OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 90 minutes. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against the phosphorylated form of 4E-BP1 on Thr37/46 (A) or Ser65 (B). Subsequently, the same blots were re-probed with an antibody against 4E-BP1 or against GAPDH, to control for protein loading. C, KG-1 cells were incubated in the absence or presence of OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 60 minutes. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against 4E-BP1 on Thr70. The same blot was re-probed with an antibody against 4E-BP1 or against GAPDH. D, U937 cells were treated with increasing concentrations of rapamycin or OSI, as indicated for 90 minutes. Cell lysates were resolved by SDS-PAGE and immunoblotted with antibodies against the phosphorylated form of rpS6 at Ser240/244, the phosphorylated form of 4E-BP1 on Thr37/46, or against GAPDH as indicated. E and F. Leukemic blasts from a patient with AML were incubated in the absence or presence of OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 90 minutes. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against 4E-BP1 on Thr37/46 (E) or Ser65 (F). The same blot was re-probed with an antibody against 4E-BP1 and then GAPDH, to assess for equivalent protein loading. G, U937 cells were treated with OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 90 minutes. Cell lysates were immunoprecipitated with an anti-eIF4E antibody and immunoprecipitates were immunoblotted with antibodies against 4E-BP1, eIF4A, eIF4G, or eIF4E, as indicated. A signal seen above the eIF4G band in the anti-eIF4G immunoblot in the OSI-027 treatment lane is an artifact and does not represent a slower migrating form of eIF4G.

Figure 4.

OSI-027, but not rapamycin, inhibits phosphorylation of the 4E-BP1 repressor of mRNA translation and blocks formation of complexes required for cap-dependent mRNA translation. A and B, U937 or KG-1 cells were incubated in the absence or presence of OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 90 minutes. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against the phosphorylated form of 4E-BP1 on Thr37/46 (A) or Ser65 (B). Subsequently, the same blots were re-probed with an antibody against 4E-BP1 or against GAPDH, to control for protein loading. C, KG-1 cells were incubated in the absence or presence of OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 60 minutes. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against 4E-BP1 on Thr70. The same blot was re-probed with an antibody against 4E-BP1 or against GAPDH. D, U937 cells were treated with increasing concentrations of rapamycin or OSI, as indicated for 90 minutes. Cell lysates were resolved by SDS-PAGE and immunoblotted with antibodies against the phosphorylated form of rpS6 at Ser240/244, the phosphorylated form of 4E-BP1 on Thr37/46, or against GAPDH as indicated. E and F. Leukemic blasts from a patient with AML were incubated in the absence or presence of OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 90 minutes. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against 4E-BP1 on Thr37/46 (E) or Ser65 (F). The same blot was re-probed with an antibody against 4E-BP1 and then GAPDH, to assess for equivalent protein loading. G, U937 cells were treated with OSI-027 (10 μmol/L) or rapamycin (20 nmol/L) for 90 minutes. Cell lysates were immunoprecipitated with an anti-eIF4E antibody and immunoprecipitates were immunoblotted with antibodies against 4E-BP1, eIF4A, eIF4G, or eIF4E, as indicated. A signal seen above the eIF4G band in the anti-eIF4G immunoblot in the OSI-027 treatment lane is an artifact and does not represent a slower migrating form of eIF4G.

Close modal

The suppressive effects of OSI-027 on formation of eIF4E–eIF4G complexes prompted us to directly examine the effects of OSI-027 on mRNA recruitment to polysomal fractions in AML cells. OSI-027 treatment resulted in suppression of mRNA recruitment to polysomes, reflecting potent inhibitory effects on translation (Fig. 5A and B). Notably, the effects of OSI-027 were much more pronounced than the effects of rapamycin analyzed in parallel (Fig. 5A and B). In addition, when mRNA translation for cyclin D1 was directly analyzed in polysomal fractions from U937 cells, we found suppression by OSI-027, but not rapamycin (Fig. 5C). Consistent with the effects seen in the suppression of cyclin D1 mRNA translation, time-dependent suppression of cyclin D1 protein expression after OSI-027 treatment was documented (Fig. 5D). Thus, mTORC1-mediated phosphorylation of 4E-BP1 is an RI event that plays a key role in mRNA translation for cyclin D1 and possibly other genes whose products control cell-cycle progression in AML cells. These findings prompted us to directly examine the effects of dual mTORC1/mTORC2 inhibition in AML cells. Treatment of U937 cells with OSI-O27 resulted in G0/G1 arrest (Supplementary Fig. S3), consistent with the suppressive effects of this agent on cyclin D1 mRNA translation and protein expression.

Figure 5.

Effects of OSI-027 on polysomal assembly and mRNA translation of the cyclin D1 gene in AML cells. A, U937 cells were treated with solvent control (DMSO), rapamycin (20 nmol/L), or OSI-027 (10 μmol/L) for 24 hours and the lysates were layered on a 10%–50% sucrose gradient. The gradients were subjected to ultracentrifugation, and fractions were collected by continuous monitoring of optical density (OD) at 254 nm. The OD 254 nm is shown as a function of gradient depth for each treatment and the polysomal peaks are indicated. B, the area under the polysome peaks and polysome (PS) + monosome (MS) peaks was quantified for each treatment using Image J software. The ratio of area under the polysomal and polysomal plus monosomal peaks was calculated for each treatment and is represented as percent control (DMSO). The data represent means ± SE from 2 independent experiments. C, the polysomal fractions were pooled and the total RNA was isolated. Quantitative real-time RT-PCR assay to determine the cyclin D1 mRNA expression in polysomal fractions was carried out using hypoxanthine-guanine phosphoribosyltransferase as a control. Data are expressed as fold change as compared with DMSO-treated samples and represent means ± SE of 2 independent experiments. D, U937 cells were incubated with or without OSI- 027 (1 μmol/L) for the indicated times. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against cyclin D1. Subsequently, the same blot was re-probed with antibodies against GAPDH, to control for equal protein loading.

Figure 5.

Effects of OSI-027 on polysomal assembly and mRNA translation of the cyclin D1 gene in AML cells. A, U937 cells were treated with solvent control (DMSO), rapamycin (20 nmol/L), or OSI-027 (10 μmol/L) for 24 hours and the lysates were layered on a 10%–50% sucrose gradient. The gradients were subjected to ultracentrifugation, and fractions were collected by continuous monitoring of optical density (OD) at 254 nm. The OD 254 nm is shown as a function of gradient depth for each treatment and the polysomal peaks are indicated. B, the area under the polysome peaks and polysome (PS) + monosome (MS) peaks was quantified for each treatment using Image J software. The ratio of area under the polysomal and polysomal plus monosomal peaks was calculated for each treatment and is represented as percent control (DMSO). The data represent means ± SE from 2 independent experiments. C, the polysomal fractions were pooled and the total RNA was isolated. Quantitative real-time RT-PCR assay to determine the cyclin D1 mRNA expression in polysomal fractions was carried out using hypoxanthine-guanine phosphoribosyltransferase as a control. Data are expressed as fold change as compared with DMSO-treated samples and represent means ± SE of 2 independent experiments. D, U937 cells were incubated with or without OSI- 027 (1 μmol/L) for the indicated times. Cell lysates were resolved by SDS-PAGE and immunoblotted with an antibody against cyclin D1. Subsequently, the same blot was re-probed with antibodies against GAPDH, to control for equal protein loading.

Close modal

In subsequent studies, the effects of dual mTORC2/mTORC1 inhibition on cell proliferation of several AML cell lines were examined. As shown in Figure 6A, OSI-027 inhibited the growth of several acute leukemia cell lines of myeloid/megakaryocytic origin in a dose-dependent manner, including U937, KG-1, KBM-3B, ML-1, HL-60, and MEG-01 cells (Fig. 6A). On the other hand, rapamycin treatment of such AML cell lines had minimal effects on proliferation (Fig. 6B).

Figure 6.

Inhibitory effects of OSI-027 on leukemic cell lines and primary leukemic progenitor colony formation from AML patients. A and B, the sensitivity of 5 leukemic cell lines to growth inhibition by OSI-027 (A) or rapamycin (B) is shown. Effects of varying concentrations of each drug at 72 hours are expressed as dose–response curves, and error bars, where visible, show the standard error of the mean for 3 replicates per data point. Results shown are typical of 3 or more independent experiments. C, peripheral blood mononuclear cells from 5 patients with AML were plated in methylcellulose culture assay system with increasing concentrations of rapamycin or OSI-027, as indicated. Data are expressed as percent control of CFU-L colony numbers for untreated cells (UT). Means ± SE of the values from 5 experiments using different patient samples are shown. Paired t-test analysis, comparing the effects of OSI-027 (5 μmol/L) to the effects of rapamycin (20 nmol/L), showed P value = 0.0034. D, peripheral blood mononuclear cells from 3 patients with AML were plated in methylcellulose culture assay system with AraC (0.1 ng/mL) and/or OSI-027 (1 μmol/L), as indicated. Data are expressed as percent control of leukemic colonies for untreated cells and represent means ± SE of 3 experiments. Paired t-test analysis of the combination of AraC plus OSI-027 compared with AraC alone, showed a P = 0.014 and, as compared with OSI-027 alone, P = 0.016.

Figure 6.

Inhibitory effects of OSI-027 on leukemic cell lines and primary leukemic progenitor colony formation from AML patients. A and B, the sensitivity of 5 leukemic cell lines to growth inhibition by OSI-027 (A) or rapamycin (B) is shown. Effects of varying concentrations of each drug at 72 hours are expressed as dose–response curves, and error bars, where visible, show the standard error of the mean for 3 replicates per data point. Results shown are typical of 3 or more independent experiments. C, peripheral blood mononuclear cells from 5 patients with AML were plated in methylcellulose culture assay system with increasing concentrations of rapamycin or OSI-027, as indicated. Data are expressed as percent control of CFU-L colony numbers for untreated cells (UT). Means ± SE of the values from 5 experiments using different patient samples are shown. Paired t-test analysis, comparing the effects of OSI-027 (5 μmol/L) to the effects of rapamycin (20 nmol/L), showed P value = 0.0034. D, peripheral blood mononuclear cells from 3 patients with AML were plated in methylcellulose culture assay system with AraC (0.1 ng/mL) and/or OSI-027 (1 μmol/L), as indicated. Data are expressed as percent control of leukemic colonies for untreated cells and represent means ± SE of 3 experiments. Paired t-test analysis of the combination of AraC plus OSI-027 compared with AraC alone, showed a P = 0.014 and, as compared with OSI-027 alone, P = 0.016.

Close modal

To determine the effects of OSI-027 in a more functionally relevant system, experiments were done in which its effects on primary leukemic progenitor colony formation (CFU-L) from patients with AML were examined in clonogenic survival assays in methylcellulose, using standard approaches (24–27). OSI-027 exhibited very potent dose-dependent suppressive effects on primary leukemic progenitors (Fig. 6C), and such effects were clearly more potent than the effects of rapamycin (Fig. 6C). Furthermore, we assessed whether OSI-027 exhibits enhancing effects on the antileukemic properties of very low concentrations of AraC; OSI-027 was combined with very low concentrations of AraC that have minimal antileukemic effects when used alone. As shown in Figure 6D, combined addition of low-dose AraC and OSI-027 resulted in more suppressive effects than did each agent alone (Fig. 6D), suggesting that combinations of OSI-027 with chemotherapeutic agents may ultimately provide an approach to improve generation of antileukemic responses.

Because of its critical role in regulation of malignant cell growth and survival, the Akt/mTOR pathway has been identified as an attractive target for the treatment of various malignancies. Over the last few years, there has been a significant effort to precisely define the role of this pathway in leukemogenesis and to incorporate its targeting in the development of novel approaches for the treatment of AML. Previous work has shown that there is constitutive phosphorylation and activation of Akt in primary AML blasts, whereas normal human CD34+ cells have only low levels of Akt activation (40, 41). A role for the PI3K in such regulation of Akt activity has been suggested based on the fact that pharmacologic inhibition of PI3K using LY294002 blocks growth of primary leukemic blasts (40). Interestingly, phosphorylation of Akt on the PDK1 phosphorylation site, Thr308, has been associated with high-risk cytogenetics and poor overall survival in AML (42). Other studies have shown that mTOR inhibition using rapamycin increases the cytotoxicity of etoposide chemotherapy against AML blasts in vitro (43). On the other hand, more recent work has shown that mTORC1 targeting using the rapamycin derivative everolimus (RAD001) fails to inhibit eIF4F assembly and c-Myc mRNA translation in AML cells and does not induce apoptosis of leukemic blasts (44). A phase I, dose-escalation study of rapamycin in combination with the chemotherapy regimen MEC (mitoxantrone, etoposide, and AraC) in AML was recently reported (45). The study showed that combination of rapamycin and chemotherapy was a feasible regimen, but failed to show synergistic effects between that mTOR inhibitor and chemotherapy in vivo (45).

Although the role of mTORC1-mediated signaling in AML is well established, the precise functional relevance of mTORC2 complexes has been unknown. As rapalogues have only limited antileukemic effects, this has suggested that mTORC2 or other RI-mTOR signals may be of importance in the regulation of leukemic cell growth and survival. mTORC1 and mTORC2 are distinct protein complexes, both of which have mTOR as their central catalytic subunit (46). In the case of mTORC1, mTOR is associated with Raptor and mLST8, whereas in mTORC2 it is associated with Rictor, mLST8, and SIN1 (45, 47). There have been some reports that rapamycin or related rapalogues are capable of inhibiting mTORC2 activity under certain conditions in vitro and in vivo (48, 49). However, it appears that such effects occur in a cell type-, time-, and dose-dependent manner (48), although, in general, rapamycin exhibits specificity toward mTORC1 (46, 47). Similarly, other FDA-approved rapalogues (e.g., temsirolimus, everolimus) selectively block mTORC1, but have minimal activity against mTORC2 (46, 47). This is highly relevant, as engagement of mTORC2 during inhibition of mTORC1 leads to Akt activation and generation of anti-apoptotic signals (46, 47). In addition, other negative feedback regulatory mechanisms are induced during treatment of AML cells with classic mTOR inhibitors, resulting in Akt activation. For instance, inhibition of mTOR in AML cells by RAD001 also upregulates expression of IRS2 (50), providing an alternative mechanism for activation of Akt and induction of anti-apoptotic responses.

In the present study, we provided evidence for the existence of mTORC2 complexes in AML cells, and showed that such mTORC2 activity is selectively blocked by the novel dual mTORC2/mTORC1 inhibitor OSI-027 (19, 38). In addition, our studies establish the presence of RI-mTORC1 complexes that regulate phosphorylation of the translational repressor 4E-BP1 at sites required for its deactivation and dissociation from the eIF4E. In contrast to rapamycin, OSI-027 acts as a potent inhibitor of phosphorylation of 4E-BP1 on Thr37/46, Ser65, and Thr70, and this results in dissociation of eIF4E–eIF4G complexes as well as enhanced formation of eIF4E–4E-BP1 complexes that negatively control mRNA translation. Consistent with this, OSI-027 treatment results in potent suppression of polysomal assembly and mRNA translation and this correlates with generation of much more potent antileukemic responses than treatment with rapamycin. It should be noted that a previous study had suggested that rapalogues can inhibit phosphorylation of 4E-BP1 in AML cells and also reduce mTORC2 signaling (49). However, in that study (49), the concentrations of CCI-779 that were used for the in vitro experiments showing inhibition of 4E-BP1 phosphorylation were very high, when taken in context with previous pharmacokinetic studies of CCI-779 in patients with cancer (51).

The precise relationship of pathways targeted by dual mTORC1/mTORC2 inhibition with other cellular events that may contribute to cap-dependent mRNA translation in AML cells remains to be precisely defined. A recently published study (44) showed that phosphorylation of 4E-BP1 on Ser65 and assembly of eIF4F complexes is resistant to RAD001 in AML cells. In this study, it was also shown that Pim-2 regulates phosphorylation of 4E-BP1 on Ser65 and may ultimately control cap-dependent mRNA translation in an mTORC1-independent manner (44). In the study, siRNA knockdown of Pim-2 blocked phosphorylation of 4E-BP1 on Ser65, but not Thr37/46 (44). It is possible that OSI-027 also affects the function of Pim-2 in AML cells, and this should be examined in future studies. However, as OSI-027 blocks phosphorylation of all key phosphorylation sites of 4E-BP1 (e.g., Thr37/46, Ser65, Thr70), its major effects on cap-dependent translation are consistent with inhibition of RI-mTORC1 complexes (52).

Our data establish that dual targeting of mTORC2 and mTORC1 with a catalytic site inhibitor is much more effective than treatment with rapamycin in blocking the growth of AML cell lines and primitive leukemic progenitors from AML patients in vitro. In addition, we show that OSI-027 enhances the suppressive effects of AraC on leukemic precursors, raising the possibility of future clinical–translational efforts involving OSI-027, or combinations of chemotherapy with OSI-027, for the treatment of AML. Notably, recently published studies showed potent antileukemic effects of other dual mTORC2/mTORC1 inhibitors, PP242 (53) or OSI-027 (54), on BCR-ABL–expressing leukemia cells including Ph+ acute lymphoblastic leukemia (ALL) cells. This suggests that, beyond AML, dual mTORC2/mTORC1 inhibition may provide an approach for the treatment of other leukemias as well. Future studies should examine the antileukemic potential of combined AML cell targeting using OSI-027 together with other agents that target cap-dependent translation or with inhibitors of pathways activated in a negative feedback regulatory manner to counteract the antileukemic effects of mTOR inhibition. For instance, it would be interesting to test combinations of OSI-027 with the 4E-BP1 mimetic 4EGI-1 that blocks mRNA translation in AML cells by competing with 4E-BP1 binding (44). There is also evidence that, in certain solid tumor cells, mTORC1 inhibition results in activation of Mek/Erk via an S6K–PI3K–Ras feedback loop (55, 56). Based on this, studies to determine whether the Mek/Erk pathway is activated in AML cells and whether it can be exploited to further enhance the antileukemic effects of mTORC2/mTORC1 inhibition are warranted.

S. Barr and S. Russo are employees of OSI Pharmaceuticals.

This work was supported in part by NIH grants R01CA121192, R01CA77816, and KL2RR025740-01, a VA Merit review grant, and the Leukemia and Lymphoma Society grant LLS-6166-09.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Lowenberg
B
,
Downing
JR
,
Burnett
A
. 
Acute myeloid leukemia
.
N Engl J Med
1999
;
341
:
1051
62
.
2.
Tallman
MS
,
Gilliland
DG
,
Rowe
JM
. 
Drug therapy for acute myeloid leukemia
.
Blood
2005
;
106
:
1154
63
.
3.
Gilliland
DG
,
Tallman
MS
. 
Focus on acute leukemias
.
Cancer Cell
2002
;
1
:
417
20
.
4.
Cammenga
J
. 
Gatekeeper pathways and cellular background in the pathogenesis and therapy of AML
.
Leukemia
2005
;
19
:
1719
28
.
5.
Fathi
AT
,
Grant
S
,
Karp
JE
. 
Exploiting cellular pathways to develop new treatment strategies for AML
.
Cancer Treat Rev
2010
;
36
:
142
50
.
6.
Hay
N
. 
The Akt-mTOR tango and its relevance to cancer
.
Cancer Cell
2005
;
8
:
179
83
.
7.
Martelli
AM
,
Nyåkern
M
,
Tabellini
G
,
Bortul
R
,
Tazzari
PL
,
Evangelisti
C
, et al
Phosphoinositide 3-kinase/Akt signaling pathway and its therapeutical implications for human acute myeloid leukemia
.
Leukemia
2006
;
20
:
911
28
.
8.
Engelman
JA
,
Luo
J
,
Cantley
LC
. 
The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism
.
Nat Rev Genet
2006
;
7
:
606
19
.
9.
Bellacosa
A
,
Kumar
CC
,
Di Cristofano
A
,
Testa
JR
. 
Activation of AKT kinases in cancer: implications for therapeutic targeting
.
Adv Cancer Res
2005
;
94
:
29
86
.
10.
Kim
D
,
Dan
HC
,
Park
S
,
Yang
L
,
Liu
Q
,
Kaneko
S
, et al
AKT/PKB signaling mechanisms in cancer and chemoresistance
.
Front Biosci
2005
;
10
:
975
87
.
11.
Martelli
AM
,
Evangelisti
C
,
Chiarini
F
,
Grimaldi
C
,
Manzoli
L
,
McCubrey
JA
. 
Targeting the PI3K/AKT/mTOR signaling network in acute myelogenous leukemia
.
Expert Opin Investig Drugs
2009
;
18
:
1333
49
.
12.
Guertin
DA
,
Sabatini
DM
. 
Defining the role of mTOR in cancer
.
Cancer Cell
2007
;
12
:
9
22
.
13.
Hay
N
,
Sonenberg
N
. 
Upstream and downstream of mTOR
.
Genes Dev
2004
;
18
:
1926
45
.
14.
Platanias
LC
. 
Mechanisms of type-I- and type-II-interferon-mediated signaling
.
Nat Rev Immunol
2005
;
5
:
375
86
.
15.
Altman
JK
,
Platanias
LC
. 
Exploiting the mammalian target of rapamycin (mTOR) pathway in hematologic malignancies
.
Curr Opin Hematol
2008
;
15
:
88
94
.
16.
Hudes
G
,
Carducci
M
,
Tomczak
P
,
Dutcher
J
,
Figlin
R
,
Kapoor
A
, et al
Global ARCC Trial
. 
Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma
.
N Engl J Med
2007
;
356
:
2271
81
.
17.
Oudard
S
,
Medioni
J
,
Aylllon
J
,
Barrascourt
E
,
Elaidi
RT
,
Balcaceres
J
, et al
Everolimus (RAD001): an mTOR inhibitor for the treatment of metastatic renal cell carcinoma
.
Expert Rev Anticancer Ther
2009
;
9
:
705
17
.
18.
Shor
B
,
Gibbons
JJ
,
Abraham
RT
,
Yu
K
. 
Targeting mTOR globally in cancer: thinking beyond rapamycin
.
Cell Cycle
2009
;
8
:
3831
7
.
19.
Falcon
BL
,
Barr
S
,
Gokhale
PC
,
Cho
J
,
Fogarty
J
,
Depeille
P
, et al
Dual mTORC1/mTORC2 inhibitors reduce tumor growth, VEGF production, angiogenesis, and vascular re-growth
.
Cancer Res
2011
;
71
:
1573
83
.
20.
Alsayed
Y
,
Uddin
S
,
Mahmud
N
,
Lekmine
F
,
Kalvakolanu
DV
,
Minucci
S
, et al
Activation of Rac1 and the p38 mitogen-activated protein kinase pathway in response to all-trans-retinoic acid
.
J Biol Chem
2001
;
276
:
4012
9
.
21.
Altman
JK
,
Yoon
P
,
Katsoulidis
E
,
Kroczynska
B
,
Sassano
A
,
Redig
AJ
, et al
Regulatory effects of mammalian target of rapamycin-mediated signals in the generation of arsenic trioxide responses
.
J Biol Chem
2008
;
283
:
1992
2001
.
22.
Kaur
S
,
Sassano
A
,
Dolniak
B
,
Joshi
S
,
Majchrzak-Kita
B
,
Baker
DP
, et al
Role of the Akt pathway in mRNA translation of interferon-stimulated genes
.
Proc Natl Acad Sci U S A
2008
;
105
:
4808
13
.
23.
Joshi
S
,
Kaur
S
,
Redig
AJ
,
Goldsborough
K
,
David
K
,
Ueda
T
, et al
Type I Interferon (IFN)-dependent activation of Mnk1 and its role in the generation of growth inhibitory responses
.
Proc Natl Acad Sci U S A
2009
;
106
:
12097
102
.
24.
Parmar
S
,
Smith
J
,
Sassano
A
,
Uddin
S
,
Katsoulidis
E
,
Majchrzak
B
, et al
Differential regulation of the p70 S6 kinase pathway by interferon alpha (IFNalpha) and imatinib mesylate (STI571) in chronic myelogenous leukemia cells
.
Blood
2005
;
106
:
2436
43
.
25.
Blair
A
,
Hogge
DE
,
Ailles
LE
,
Lansdorp
PM
,
Sutherland
HJ
. 
Lack of expression of Thy-1 (CD90) on acute myeloid leukemia cells with long-term proliferative ability in vitro and in vivo
.
Blood
1997
;
89
:
3104
12
.
26.
Bhatia
R
,
Holtz
M
,
Niu
N
,
Gray
R
,
Snyder
DS
,
Sawyers
CL
, et al
Persistence of malignant hematopoietic progenitors in chronic myelogenous leukemia patients in complete cytogenetic remission following imatinib mesylate treatment
.
Blood
2003
;
101
:
4701
7
.
27.
La Rosée
P
,
Johnson
K
,
Corbin
AS
,
Stoffregen
EP
,
Moseson
EM
,
Willis
S
, et al
In vitro efficacy of combined treatment depends on the underlying mechanism of resistance in imatinib-resistant Bcr-Abl-positive cell lines
.
Blood
2004
;
103
:
208
15
.
28.
Liu
L
,
Li
F
,
Cardelli
JA
,
Martin
KA
,
Blenis
J
,
Huang
S
. 
Rapamycin inhibits cell motility by suppression of mTOR-mediated S6K1 and 4E-BP1 pathways
.
Oncogene
2006
;
25
:
7029
40
.
29.
Armour
SM
,
Baur
JA
,
Hsieh
SN
,
Land-Bracha
A
,
Thomas
SM
,
Sinclair
DA
. 
Inhibition of mammalian S6 kinase by resveratrol suppresses autophagy
.
Aging
2009
;
1
:
515
28
.
30.
Fritz
RD
,
Varga
Z
,
Radziwill
G
. 
CNK1 is a novel Akt interaction partner that promotes cell proliferation through the Akt-FoxO signalling axis
.
Oncogene
2010
;
29
:
3575
82
.
31.
Manning
BD
. 
Balancing Akt with S6K: implications for both metabolic diseases and tumorigenesis
.
J Cell Biol
2004
;
167
:
399
403
.
32.
Copp
J
,
Manning
G
,
Hunter
T
. 
TORC-specific phosphorylation of mammalian target of rapamycin (mTOR): phospho-Ser 2481 is a marker for intact mTOR signaling complex 2
.
Cancer Res
2009
;
69
:
1821
7
.
33.
Yang
HS
,
Jansen
AP
,
Komar
AA
,
Zheng
X
,
Merrick
WC
,
Costes
S
, et al
The transformation suppressor Pdcd4 is a novel eukaryotic translation initiation factor 4A binding protein that inhibits translation
.
Mol Cell Biol
2003
;
23
:
26
37
.
34.
Dorrello
NV
,
Peschiaroli
A
,
Guardavaccaro
D
,
Colburn
NH
,
Sherman
NE
,
Pagano
M
. 
S6K1- and betaTRCP-mediated degradation of PDCD4 promotes protein translation and cell growth
.
Science
2006
;
314
:
467
71
.
35.
Carayol
N
,
Katsoulidis
E
,
Sassano
A
,
Altman
JK
,
Druker
BJ
,
Platanias
LC
. 
Suppression of programmed cell death 4 (PDCD4) protein expression by BCR-ABL-regulated engagement of the mTOR/p70 S6 kinase pathway
.
J Biol Chem
2008
;
283
:
8601
10
.
36.
Gingras
AC
,
Kennedy
SG
,
O'Leary
MA
,
Sonenberg
N
,
Hay
N
. 
4E-BP1, a repressor of mRNA translation, is phosphorylated and inactivated by the Akt (PKB) signaling pathway
.
Genes Dev
1998
;
12
:
502
13
.
37.
Gingras
AC
,
Raught
B
,
Gygi
SP
,
Niedzwiecka
A
,
Miron
M
,
Burley
SK
, et al
Hierarchical phosphorylation of the translation inhibitor 4E-BP1
.
Genes Dev
2001
;
15
:
2852
64
.
38.
Gokhale
PC
,
Bhagwat
SV
,
Crew
AP
,
Cooke
A
,
Mantis
C
,
Workman
JE
, et al
OSI-027, a selective dual mTORC1/TORC2 kinase inhibitor displays broad spectrum anti-tumor activity in preclinical models of human cancer
.
Proc AACR 2010
(
abstr 4486
).
39.
Kuhn
JG
,
Chang
SM
,
Wen
PY
,
Cloughesy
TF
,
Greenberg
H
,
Schiff
D
, et al
North American Brain Tumor Consortium and the National Cancer Institute. Pharmacokinetic and tumor distribution characteristics of temsirolimus in patients with recurrent malignant glioma
.
Clin Cancer Res
2007
;
13
:
7401
6
.
40.
Xu
Q
,
Simpson
S
,
Scialla
TJ
,
Bagg
A
,
Carroll
M
. 
Survival of acute myeloid leukemia requires PI3 kinase activation
.
Blood
2003
;
102
:
972
80
.
41.
Grandage
VL
,
Gale
RE
,
Linch
DC
,
Khwaja
A
. 
PI3-kinase/Akt is constitutively active in primary acute myeloid leukaemia cells and regulates survival and chemoresistance via NF-κB, MAP kinase and p53 pathways
.
Leukemia
2005
;
19
:
586
94
.
42.
Gallay
N
,
Dos Santos
C
,
Cuzin
L
,
Bousquet
M
,
Simmonet
Gouy V
,
Chaussade
C
, et al
The level of Akt phosphorylation on threonine 308 but not on serine 473 is associated with high-risk cytogenetics and predicts poor overall survival in acute myeloid leukaemia
.
Leukemia
2009
;
23
:
1029
38
.
43.
Xu
Q
,
Thompson
JE
,
Carroll
M
. 
mTOR regulates cell survival after etoposide treatment in primary AML cells
.
Blood
2005
;
106
:
4261
8
.
44.
Tamburini
J
,
Green
AS
,
Bardet
V
,
Chapuis
N
,
Park
S
,
Willems
L
, et al
Protein synthesis is resistant to rapamycin and constitutes a promising therapeutic target in acute myeloid leukemia
.
Blood
2009
;
114
:
1618
27
.
45.
Perl
AE
,
Kasner
MT
,
Tsai
DE
,
Vogl
DT
,
Loren
AW
,
Schuster
SJ
, et al
A phase I study of the mammalian target of rapamycin inhibitor sirolimus and MEC chemotherapy in relapsed and refractory acute myelogenous leukemia
.
Clin Cancer Res
2009
;
15
:
6732
9
.
46.
Bhaskar
PT
,
Hay
N
. 
The two TORCs and Akt
.
Dev Cell
2007
;
12
:
487
502
.
47.
Cybulski
N
,
Hall
MN
. 
TOR complex 2: a signaling pathway of its own
.
Trends Biochem Sci
2009
;
34
:
620
7
.
48.
Sarbassov
DD
,
Ali
SM
,
Sengupta
S
,
Sheen
JH
,
Hsu
PP
,
Bagley
AF
, et al
Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB
.
Mol Cell
2006
;
22
:
159
68
.
49.
Zeng
Z
,
Sarbassov
dos D
,
Samudio
IJ
,
Yee
KW
,
Munsell
MF
,
Ellen
Jackson C
, et al
Rapamycin derivatives reduce mTORC2 signaling and inhibit Akt activation in AML
.
Blood
2007
;
109
:
3509
12
.
50.
Tamburini
J
,
Chapuis
N
,
Bardet
V
,
Park
S
,
Sujobert
P
,
Willems
L
, et al
Mammalian target of rapamycin (mTOR) inhibition activates phosphatidylinositol 3-kinase/Akt by up-regulating insulin-like growth factor-1 receptor signaling in acute myeloid leukemia: rationale for therapeutic inhibition of both pathways
.
Blood
2008
;
111
:
379
82
.
51.
Raymond
E
,
Alexandre
J
,
Faivre
S
,
Vera
K
,
Materman
E
,
Boni
J
, 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
.
52.
Thoreen
CC
,
Kang
SA
,
Chang
JW
,
Liu
Q
,
Zhang
J
,
Gao
Y
, et al
An ATP-competitive mammalian target of rapamycin inhibitor reveals rapamycin-resistant functions of mTORC1
.
J Biol Chem
2009
;
284
:
8023
32
.
53.
Janes
MR
,
Limon
JJ
,
So
L
,
Chen
J
,
Lim
RJ
,
Chavez
MA
, et al
Effective and selective targeting of leukemia cells using a TORC1/2 kinase inhibitor
.
Nat Med
2010
;
16
:
205
13
.
54.
Carayol
N
,
Vakana
E
,
Sassano
A
,
Kaur
S
,
Goussetis
DJ
,
Glaser
H
, et al
Critical roles for mTORC2- and rapamycin-insensitive mTORC1-complexes in growth and survival of BCR-ABL-expressing leukemic cells
.
Proc Natl Acad Sci U S A
2010
;
107
;
12469
74
.
55.
Carracedo
A
,
Ma
L
,
Teruya-Feldstein
J
,
Rojo
F
,
Salmena
L
,
Alimonti
A
, et al
Inhibition of mTORC1 leads to MAPK pathway activation through a PI3′K-dependent feedback loop in human cancer
.
J Clin Invest
2008
;
118
:
3065
74
.
56.
Kinkade
CW
,
Castillo-Martin
M
,
Puzio-Kuter
A
,
Yan
J
,
Foster
TH
,
Gao
H
, et al
Targeting Akt/mTOR and ERK MAPK signaling inhibits hormone-refractory prostate cancer in a preclinical mouse model
.
J Clin Invest
2008
;
18
:
3051
64
.