mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase ...receptor tyrosine kinase signaling is a...

10
mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase Signaling and Activates Akt Kathryn E. O’Reilly, 1 Fredi Rojo, 2 Qing-Bai She, 1 David Solit, 1 Gordon B. Mills, 3 Debra Smith, 3 Heidi Lane, 4 Francesco Hofmann, 4 Daniel J. Hicklin, 5 Dale L. Ludwig, 5 Jose Baselga, 2 and Neal Rosen 1 1 Memorial Sloan-Kettering Cancer Center New York, New York; 2 Vall d’Hebron University Hospital, Barcelona, Spain; 3 The University of Texas M.D. Anderson Cancer Center, Houston, Texas; 4 Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland; and 5 ImClone Systems Incorporated, New York, New York Abstract Stimulation of the insulin and insulin-like growth factor I (IGF-I) receptor activates the phosphoinositide-3-kinase/Akt/ mTOR pathway causing pleiotropic cellular effects including an mTOR-dependent loss in insulin receptor substrate-1 expression leading to feedback down-regulation of signaling through the pathway. In model systems, tumors exhibiting mutational activation of phosphoinositide-3-kinase/Akt ki- nase, a common event in cancers, are hypersensitive to mTOR inhibitors, including rapamycin. Despite the activity in model systems, in patients, mTOR inhibitors exhibit more modest antitumor activity. We now show that mTOR inhibition induces insulin receptor substrate-1 expression and abrogates feedback inhibition of the pathway, resulting in Akt activation both in cancer cell lines and in patient tumors treated with the rapamycin derivative, RAD001. IGF-I receptor inhibition prevents rapamycin-induced Akt activation and sensitizes tumor cells to inhibition of mTOR. In contrast, IGF-I reverses the antiproliferative effects of rapamycin in serum-free medium. The data suggest that feedback down-regulation of receptor tyrosine kinase signaling is a frequent event in tumor cells with constitutive mTOR activation. Reversal of this feedback loop by rapamycin may attenuate its therapeu- tic effects, whereas combination therapy that ablates mTOR function and prevents Akt activation may have improved antitumor activity. (Cancer Res 2006; 66(3): 1500-8) Introduction Activation of phosphoinositide-3-kinase (PI3K)/Akt/mTOR sig- naling through mutation of pathway components as well as through activation of upstream signaling molecules occurs in a majority of cancers contributing to deregulation of proliferation, resistance to apoptosis, and changes in metabolism characteristic of transformed cells (1, 2). This pathway is normally regulated by upstream receptor tyrosine kinases, especially the insulin and insulin-like growth factor I (IGF-I) receptors (IGF-IR; ref. 3). Physiologic activation of these receptors also results in feedback down-regulation of the pathway, mediated in part by mTOR/S6K- dependent loss of insulin receptor substrate-1 (IRS-1) expression (4–8). IRS-1 is the major substrate of the IGF-I and insulin receptors, responsible for the propagation of the signal induced by ligand binding (2, 9). We speculated that constitutive activa- tion of the Akt/mTOR pathway in cancer cells would induce upstream feedback inhibition of signaling via IGF-I/insulin and perhaps other transmembrane receptors. Feedback inhibition could have marked biological and therapeutic implications. First, feedback inhibition of upstream signaling pathways could cause hypersensitivity to mTOR inhibitors and inhibition of other elements of the activated signaling pathway (so-called ‘‘oncogene addiction’’; refs. 10, 11). Second, inhibition of mTOR could cause the release of feedback inhibition, paradoxically activating IGF-I signaling and reducing the antitumor effects of mTOR inhibitors. We show herein that inhibition of mTOR in cancer cell lines and in patient tumors causes activation of Akt kinase which is associated with induction of IRS-1 and is prevented by IGF-IR inhibition. Furthermore, we show that IGF-I antagonizes the antiproliferative effects of rapamycin in serum-free medium and IGF-IR inhibitors sensitize cancer cell lines to rapamycin’s antiproliferative effects. Materials and Methods Cell lines, antibodies, and reagents. DU-145, MCF-7, and MDA-MB-468 were purchased from the American Type Culture Collection (Manassas, VA). Anti-IRS-1, anti-IRS-2, and anti-FKHRL1 antibodies were from Upstate Biotechnology (Waltham, MA). pAkt(S473), p-p70/S6K (T389), p70/S6K, p-GSK3a/h, GSK3a, GSK3h, Akt, Akt1, Akt 2, Akt 3, p-FKHR(S256)/ p-AFX(S193), FKHR, AFX, and p-FKHR(T24)/p-FKHRL1(T32) antibodies were from Cell Signaling Technology (Beverly, MA). Rapamycin and LY294002 were from Calbiochem (San Diego, CA). NVP-AEW541 was a gift from Novartis Pharma AG (Basel, Switzerland) and A12 was a gift from ImClone Systems, Incorporated (New York, NY). Western blotting. MDA-MB-468, DU-145, and MCF-7 cells were harvested and lysed in mRIPA or NP40 lysis buffer. Protein concentrations were determined with the bicinchoninic acid method (Pierce, Rockford, IL). Samples were subjected to SDS-PAGE. Proteins were detected using the enhanced chemiluminescence kit (Amersham Biosciences, Piscataway, NJ) and bands quantitated with Science Lab 2003 Image Gauge (Fujifilm, Tokyo, Japan). In vitro Akt kinase assay. Cells were treated with either 1 nmol/L rapamycin for 1 or 4 hours, DMSO vehicle (time 0), 10 Amol/L LY294002 pretreatment for 1 hour followed by 1 or 4 hours of 1 nmol/L rapamycin exposure, 800 nmol/L NVP-AEW541 pretreatment for 1 hour followed by 1 or 4 hours of 1 nmol/L rapamycin, or 10 nmol/L A12 pretreatment for 1 hour followed by 4 hours of 1 nmol/L rapamycin. Cells were harvested and the Akt kinase assay from Cell Signaling Technology was used. Akt1 was immunoprecipitated from lysates and used in an in vitro kinase assay to catalyze phosphorylation of a GSK-3 fusion protein (1 Ag) in the presence of 200 Amol/L ATP. The reaction product was subjected to SDS-PAGE and probed with p-GSK3a/h antibody (S21/9). Human solid tumor biopsy and immunohistochemistry. Tumor biopsies from patients treated with RAD001 were analyzed. Patients were Requests for reprints: Neal Rosen, Program in Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Center, Box 271, 1275 York Avenue, New York, NY 10021. Phone: 212-639-2369; Fax: 212-717-3627; E-mail: [email protected]. I2006 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-05-2925 Cancer Res 2006; 66: (3). February 1, 2006 1500 www.aacrjournals.org Research Article Research. on May 8, 2021. © 2006 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Transcript of mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase ...receptor tyrosine kinase signaling is a...

Page 1: mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase ...receptor tyrosine kinase signaling is a frequent event in tumor cells with constitutive mTOR activation. Reversal of this

mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase

Signaling and Activates Akt

Kathryn E. O’Reilly,1Fredi Rojo,

2Qing-Bai She,

1David Solit,

1Gordon B. Mills,

3

Debra Smith,3Heidi Lane,

4Francesco Hofmann,

4Daniel J. Hicklin,

5

Dale L. Ludwig,5Jose Baselga,

2and Neal Rosen

1

1Memorial Sloan-Kettering Cancer Center New York, New York; 2Vall d’Hebron University Hospital, Barcelona, Spain;3The University of Texas M.D. Anderson Cancer Center, Houston, Texas; 4Novartis Institutes for BioMedical Research,Novartis Pharma AG, Basel, Switzerland; and 5ImClone Systems Incorporated, New York, New York

Abstract

Stimulation of the insulin and insulin-like growth factor I(IGF-I) receptor activates the phosphoinositide-3-kinase/Akt/mTOR pathway causing pleiotropic cellular effects includingan mTOR-dependent loss in insulin receptor substrate-1expression leading to feedback down-regulation of signalingthrough the pathway. In model systems, tumors exhibitingmutational activation of phosphoinositide-3-kinase/Akt ki-nase, a common event in cancers, are hypersensitive to mTORinhibitors, including rapamycin. Despite the activity in modelsystems, in patients, mTOR inhibitors exhibit more modestantitumor activity. We now show that mTOR inhibitioninduces insulin receptor substrate-1 expression and abrogatesfeedback inhibition of the pathway, resulting in Akt activationboth in cancer cell lines and in patient tumors treated withthe rapamycin derivative, RAD001. IGF-I receptor inhibitionprevents rapamycin-induced Akt activation and sensitizestumor cells to inhibition of mTOR. In contrast, IGF-I reversesthe antiproliferative effects of rapamycin in serum-freemedium. The data suggest that feedback down-regulation ofreceptor tyrosine kinase signaling is a frequent event intumor cells with constitutive mTOR activation. Reversal ofthis feedback loop by rapamycin may attenuate its therapeu-tic effects, whereas combination therapy that ablates mTORfunction and prevents Akt activation may have improvedantitumor activity. (Cancer Res 2006; 66(3): 1500-8)

Introduction

Activation of phosphoinositide-3-kinase (PI3K)/Akt/mTOR sig-naling through mutation of pathway components as well asthrough activation of upstream signaling molecules occurs in amajority of cancers contributing to deregulation of proliferation,resistance to apoptosis, and changes in metabolism characteristicof transformed cells (1, 2). This pathway is normally regulated byupstream receptor tyrosine kinases, especially the insulin andinsulin-like growth factor I (IGF-I) receptors (IGF-IR; ref. 3).Physiologic activation of these receptors also results in feedbackdown-regulation of the pathway, mediated in part by mTOR/S6K-dependent loss of insulin receptor substrate-1 (IRS-1) expression(4–8). IRS-1 is the major substrate of the IGF-I and insulinreceptors, responsible for the propagation of the signal induced

by ligand binding (2, 9). We speculated that constitutive activa-tion of the Akt/mTOR pathway in cancer cells would induceupstream feedback inhibition of signaling via IGF-I/insulin andperhaps other transmembrane receptors. Feedback inhibitioncould have marked biological and therapeutic implications. First,feedback inhibition of upstream signaling pathways could causehypersensitivity to mTOR inhibitors and inhibition of otherelements of the activated signaling pathway (so-called ‘‘oncogeneaddiction’’; refs. 10, 11). Second, inhibition of mTOR could causethe release of feedback inhibition, paradoxically activating IGF-Isignaling and reducing the antitumor effects of mTOR inhibitors.We show herein that inhibition of mTOR in cancer cell lines andin patient tumors causes activation of Akt kinase which isassociated with induction of IRS-1 and is prevented by IGF-IRinhibition. Furthermore, we show that IGF-I antagonizes theantiproliferative effects of rapamycin in serum-free medium andIGF-IR inhibitors sensitize cancer cell lines to rapamycin’santiproliferative effects.

Materials and Methods

Cell lines, antibodies, and reagents. DU-145, MCF-7, andMDA-MB-468

were purchased from the American Type Culture Collection (Manassas, VA).Anti-IRS-1, anti-IRS-2, and anti-FKHRL1 antibodies were from Upstate

Biotechnology (Waltham, MA). pAkt(S473), p-p70/S6K (T389), p70/S6K,

p-GSK3a/h, GSK3a, GSK3h, Akt, Akt1, Akt 2, Akt 3, p-FKHR(S256)/

p-AFX(S193), FKHR, AFX, and p-FKHR(T24)/p-FKHRL1(T32) antibodieswere from Cell Signaling Technology (Beverly, MA). Rapamycin and

LY294002 were from Calbiochem (San Diego, CA). NVP-AEW541 was a gift

from Novartis Pharma AG (Basel, Switzerland) and A12 was a gift fromImClone Systems, Incorporated (New York, NY).

Western blotting. MDA-MB-468, DU-145, and MCF-7 cells were

harvested and lysed in mRIPA or NP40 lysis buffer. Protein concentrations

were determined with the bicinchoninic acid method (Pierce, Rockford, IL).Samples were subjected to SDS-PAGE. Proteins were detected using the

enhanced chemiluminescence kit (Amersham Biosciences, Piscataway, NJ)

and bands quantitated with Science Lab 2003 Image Gauge (Fujifilm,

Tokyo, Japan).In vitro Akt kinase assay. Cells were treated with either 1 nmol/L

rapamycin for 1 or 4 hours, DMSO vehicle (time 0), 10 Amol/L LY294002

pretreatment for 1 hour followed by 1 or 4 hours of 1 nmol/L rapamycinexposure, 800 nmol/L NVP-AEW541 pretreatment for 1 hour followed by

1 or 4 hours of 1 nmol/L rapamycin, or 10 nmol/L A12 pretreatment for

1 hour followed by 4 hours of 1 nmol/L rapamycin. Cells were harvested and

the Akt kinase assay from Cell Signaling Technology was used. Akt1 wasimmunoprecipitated from lysates and used in an in vitro kinase assay to

catalyze phosphorylation of a GSK-3 fusion protein (1 Ag) in the presence of

200 Amol/L ATP. The reaction product was subjected to SDS-PAGE and

probed with p-GSK3a/h antibody (S21/9).Human solid tumor biopsy and immunohistochemistry. Tumor

biopsies from patients treated with RAD001 were analyzed. Patients were

Requests for reprints: Neal Rosen, Program in Molecular Pharmacology andChemistry, Memorial Sloan-Kettering Cancer Center, Box 271, 1275 York Avenue, NewYork, NY 10021. Phone: 212-639-2369; Fax: 212-717-3627; E-mail: [email protected].

I2006 American Association for Cancer Research.doi:10.1158/0008-5472.CAN-05-2925

Cancer Res 2006; 66: (3). February 1, 2006 1500 www.aacrjournals.org

Research Article

Research. on May 8, 2021. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 2: mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase ...receptor tyrosine kinase signaling is a frequent event in tumor cells with constitutive mTOR activation. Reversal of this

treated with RAD001 administered p.o. daily (10 mg, four patients) orweekly (50 mg, four patients). Sequential tumor biopsies at baseline time

point—prior to start treatment—and 28 days after therapy were done in

all patients. All tissue specimens were fixed in 10% buffered neutral

formalin for 24 hours at room temperature, then dehydrated and paraffinembedded. Immunostaining was done on 4 Am tissue sections placed on

charged plus glass slides. After deparaffinization in xylene and graded

alcohols, heat antigen retrieval was done in citrate buffer (pH 6) for 5

minutes in an autoclave. Following epitope retrieval, endogenousperoxidase was blocked by immersing the sections in 0.03% hydrogen

peroxide for 10 minutes. Slides were washed for 5 minutes with TBS.

Incubation with polyclonal anti-S473 pAkt antibody was made at room

temperature for 2 hours at 1:50 dilution on 0.05 mol/L Tris-HCl buffer[DakoCytomation (Carpinteria, CA) Antibody Diluent]. The peroxidase-

labeled polymer conjugated to goat anti-rabbit method was used to detect

antigen-antibody reaction (DakoCytomation EnVision+ System) for 30minutes at room temperature. Sections were then visualized with 3,3V-diaminobenzidine as a chromogen for 5 minutes and counterstained with

Mayer’s hematoxylin. All immunohistochemical stainings were done in a

Dako Autostainer under the same conditions. The same sectionsincubated with rabbit nonimmunized serum were used as negative

controls; for the positive control, sections of a breast carcinoma human

tumor with a known expression of pAkt by immunohistochemistry and

Western blot were stained. Tumor sections were studied on a light

microscope with an ocular magnification of �400. To score a tumor cell

as positive for pAkt, nuclear and cytoplasmic staining was required. The

percentage of stained tumor cells was scored in a whole section and the

average percentage and intensity of tumor cell staining was calculated as

a histoscore as described previously (12). Tumors with >1% of tumor cells

staining were considered positive for such markers. Grading of scoring

ranged from a score of 0 to 300. Scoring was blinded to time point data.

Statistical analysis for Wilcoxon signed ranks test was done between

pretherapy and posttherapy levels of pAkt (SPSS analysis software 10.0).

Cell proliferation studies. 100,000 DU-145 or MCF-7 cells, and 50,000

MDA-MB-468 cells were plated in normal growth medium. The cells

were grown overnight before treatment with DMSO, 1 nmol/L rapa-

mycin, 1 Amol/L NVP-AEW541, or combined 1 nmol/L rapamycin and

1 Amol/L NVP-AEW541. Cells were trypsinized and counted on a Coulter

counter.

Cell cycle analysis. Cells (1 � 106) were plated in 10 cm dishes in normal

growth medium and grown overnight before treatment with DMSO, 1 nmol/L

rapamycin, or a combination of rapamycin (1 nmol/L) and NVP-AEW541

(1 Amol/L). The nuclei were isolated by the Nusse method (13) and

subjected to flow cytometry to determine fraction of cells in sub-G1, G1, S,

and G2.

Results

Inhibition of mTOR induces Akt activity in tumor cells. Wetested whether inhibition of mTOR caused activation of PI3K/Aktsignaling by relieving feedback inhibition of upstream signaling.Prolonged exposure of breast cancer cell lines MCF-7 (PI3Kmutant; ref. 14), and MDA-MB-468 (PTEN mutant), or the prostatecancer cell line DU-145 (PTEN wild-type) to the mTOR inhibitorrapamycin caused a marked increase in S473 phosphorylation andAkt kinase activity (Fig. 1A and B). These results are consistentwith the recent report by Sun et al. who observed that rapamycininduced pAkt(S473) in several human cancer cell lines includingDU-145 and MCF-7 (15). We also observed the induction of pAkt byrapamycin in the breast cancer cell lines, SkBR3 and BT474, cells inwhich PI3K/Akt is driven by overexpression of human epidermalgrowth factor receptor 2, and in the rhabdomyosarcoma line Rh30(Fig. 1C). In DU-145 and MCF-7, induction of S473 phosphorylationoccurred within 20 minutes of rapamycin treatment and followedinhibition of mTOR-dependent S6K phosphorylation (Fig. 1A and B).

In both cell lines, pAkt was increased 4-fold to 5-fold by 24 hoursof rapamycin exposure and remained elevated for at least 48hours after the addition of the drug. As measured by an in vitrokinase assay, Akt activity in DU-145 increased in parallel with theinduction of phosphorylation and was elevated 3-fold comparedwith controls 4 hours after the addition of the drug (Fig. 1D).DU-145 and MCF-7 contain wild-type PTEN and have modest

basal levels of p-Akt. In MDA-MB-468, PTEN is mutationallyinactivated and p-Akt levels are constitutively elevated. In this cellline, it is difficult to discern a further increase in the S473phosphorylation of total Akt after rapamycin exposure. However, a3-fold increase in Akt kinase activity was noted at 1 hour and wasaccompanied by a 6-fold increase in the phosphorylation of Akt1(Fig. 1D). Thus, in both PTEN-wt and PTEN-mutated tumor celllines, inhibition of mTOR function by rapamycin could lead toactivation of Akt kinase.Rapamycin-induced activation of Akt kinase was also associated

with increased phosphorylation of endogenous Akt substrates. InMCF-7 cells treated with rapamycin, the phosphorylation ofFoxO1a, FoxO3a, and FoxO4 transcription factors (Fig. 1E) andGSK3a/h was markedly increased as compared with control cells(Fig. 1A and B). The increase in GSK3a/h phosphorylationoccurred in both DU-145 and MCF-7 cells and began after40 minutes of rapamycin exposure, remaining elevated for at least24 hours after initiation of rapamycin treatment. Phosphorylationof FoxO transcription factors was elevated after 1 hour ofrapamycin exposure in MCF-7 cells and remained elevated for atleast 24 hours. Total FKHR, AFX, and FKHRL1 levels did not changewith treatment (Fig. 1E). Thus, rapamycin-induced Akt phosphor-ylation and kinase activity leads to functional activation of Aktsignaling in tumor cells.Inhibition of mTOR induces S473 Akt phosphorylation

in vivo in human tumors. In model systems, tumors withactivated Akt secondary to PTEN loss or other causes have beenshown to be hypersensitive to rapamycin (16, 17). This data has ledto clinical trials of rapamycin derivatives (2, 18). As indicatedabove, we have shown that mTOR inhibition can activate Aktsignaling in tumor cells in tissue culture. To determine if thisoccurs in patient tumors in vivo as well, we obtained tumor tissuefrom patients with advanced solid tumors who were being treatedon a phase I protocol of the rapamycin derivative RAD001(Everolimus, Novartis Pharma). The levels of S473 phosphorylatedAkt in the tumor biopsies increased after RAD001 treatment(P = 0.018, Wilcoxon signed rank test; Fig. 2A-C). Biopsies of livermetastases or skin lesions were taken from patients with colon orbreast carcinoma before and after 4 weeks of RAD001 treatment.The levels of pAkt, as determined by immunohistochemistry andquantitated as a histoscore ranging from 0 to 300, were elevated inbiopsies from patients receiving daily RAD001 (n = 4), as well as inbiopsies from patients on a weekly dosing schedule (n = 4). Giventhat Akt activation results in cancer cell survival, proliferation, andgrowth, the induction of phosphorylated Akt is an unexpected andpotentially undesirable consequence of mTOR inhibition.Induction of Akt kinase activity is IGF-IR dependent and is

associated with up-regulation of IRS-1 protein levels. In normalcells, IGF-I and insulin induction of PI3K/Akt/mTOR signaling leadsto an mTOR-dependent feedback inhibition of signaling, due in partto mTOR/S6K-dependent loss of IRS-1 expression (4–8). Rapamycinreactivates signaling, reverses insulin resistance, and induces IRS-1expression in these cells (4). We asked whether induction of Aktsignaling by rapamycin in tumor cells is due to loss of negative

mTOR Inhibition Activates Akt

www.aacrjournals.org 1501 Cancer Res 2006; 66: (3). February 1, 2006

Research. on May 8, 2021. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 3: mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase ...receptor tyrosine kinase signaling is a frequent event in tumor cells with constitutive mTOR activation. Reversal of this

regulation of IGF-I signaling and whether this induction is IGF-IR-dependent. To determine whether the induction of Akt activity andsignaling by rapamycin is dependent on IGF-IR, we employed asmall-molecule IGF-IR kinase inhibitor (NVP-AEW541, NovartisPharma; ref. 19), and a human monoclonal antibody to the IGF-IR

(A12, ImClone Systems; ref. 20). NVP-AEW541 shows selectivity forIGF-IR (IC50, 0.086 Amol/L) as compared with the closely relatedinsulin receptor (IC50, 2.3 Amol/L). A12, a human monoclonal anti-body to IGF-IR, blocks IGF-I binding to the receptor with an IC50 of0.6 to 1 nmol/L and does not interfere with insulin binding. One

Figure 1. mTOR inhibition activates Akt in tumorcells. A, 1 nmol/L rapamycin treatment inducedS473 Akt and S21/9 GSK3a/h phosphorylationin vitro in a DU-145 prostate cancer cell line. Akt1,Akt2, and Akt3, total Akt, and total GSK3a/hdid not change. Phosphorylation of p70/S6Kdecreased with rapamycin treatment whereastotal p70/S6K levels did not change. B, 1 nmol/Lrapamycin treatment induced S473 Akt and S21/9GSK3a/h phosphorylation in vitro in a MCF-7cancer cell line. Akt1, Akt2, and Akt3, totalAkt, and total GSK3a/h did not change.Phosphorylation of p70/S6K decreased withrapamycin treatment whereas total p70/S6Klevels did not change. C, 1 nmol/L rapamycintreatment for 24 hours induced S473 Akt in thebreast cancer cell lines BT474, SkBr3, and therhabdomyosarcoma cell line Rh30. Total Aktlevels did not change.

Cancer Research

Cancer Res 2006; 66: (3). February 1, 2006 1502 www.aacrjournals.org

Research. on May 8, 2021. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 4: mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase ...receptor tyrosine kinase signaling is a frequent event in tumor cells with constitutive mTOR activation. Reversal of this

hour of pretreatment with either of the inhibitors abrogatedrapamycin-induced pAkt induction (Fig. 3A and B). The inductionof Akt kinase activity was also abrogated by IGF-IR blockade andPI3K inhibition with LY294002 (Fig. 1D). In addition, FoxO substratephosphorylation was prevented by pretreatment with NVP-AEW541

in MCF-7 cells (Fig. 1E).These data suggest that rapamycin-inducedAkt activation is due to release of mTOR-dependent negativeregulation of IGF-I signaling. Consistent with these data, we foundthat rapamycin increased IRS-1 protein levels, but not IRS-2 in DU-145, MCF-7, and MDA-MB-468 cells (Fig. 3C). We also found that, in

Figure 1 Continued. D, mTOR inhibitionwith 1 nmol/L rapamycin induced AktKinase activity in an IGF-IR-dependentmanner. An Akt kinase assay in DU-145cells confirmed that the increased pAkt(S473) resulted in increased Akt activity,which was abrogated by the inhibition ofPI3K with the small-molecule LY294002(10 Amol/L) and by inhibition of IGF-IR witheither the small-molecule NVP-AEW541(800 nmol/L) or the monoclonal antibodyA12 (10 nmol/L). A Western blot of pAkt(S473) at the same time points in DU-145showed the increased Akt phosphorylationwhich was abrogated by IGF-IR or PI3Kinhibition. Although there is no detectableincrease in pan-phospho-Akt as measuredby Western blot, an Akt kinase assay inMDA-MB-468 cells reveals an inductionof Akt activity and p-Akt1 (S473) that isabrogated by LY294002 as well as theIGF-IR inhibitors. E, treatment of MCF-7cells with rapamycin (1 nmol/L) inducedphosphorylation of the FoxO transcriptionfactors, FKHR (S256 and T24), AFX(S193), and FKHRL1 (T32). Therapamycin-induced FoxO phosphorylationwas abrogated by IGF-IR inhibition with800 nmol/L NVP-AEW541. Total FKHR,AFX, and FKHRL1 levels did notchange with rapamycin or NVP-AEW-541treatment.

mTOR Inhibition Activates Akt

www.aacrjournals.org 1503 Cancer Res 2006; 66: (3). February 1, 2006

Research. on May 8, 2021. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 5: mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase ...receptor tyrosine kinase signaling is a frequent event in tumor cells with constitutive mTOR activation. Reversal of this

serum-free medium, IGF-I stimulated Akt activation in MCF-7 cellsand that cotreatment with rapamycin enhanced this activation >2-fold (Fig. 4A). This further supports the idea that mTOR negativelyregulates IGF-I signaling in these cells.IGF-I prevents and IGF-IR inhibitors enhance the anti-

proliferative effects of rapamycin. Rapamycin analogues haveshown limited antitumor activity in clinical trials (21). IGF-I hasbeen shown to rescue rhabdomyosarcoma cells from rapamycin-induced apoptosis, suggesting that IGF-I may antagonize theeffects of mTOR inhibition (22). If so, our data implies thatinduction of IGF-I-dependent Akt activation by rapamycin couldbe responsible for attenuating its anticancer effects. We testedwhether IGF-I could interfere with rapamycin-induced inhibitionof tumor cell proliferation. MCF-7 cells were serum-starved for 16hours and treated with rapamycin in the presence or absence ofIGF-I (Fig. 4A). Rapamycin treatment dramatically reduced thecell proliferation rate, but cells treated with both rapamycinand IGF-I were not inhibited and proliferated at a rate similar to

the cells treated with IGF-I alone. When added to rapamycin-treated cells in serum-free medium, IGF-I acted to completelyabrogate the antiproliferative effects of rapamycin. In addition,the induction of pAkt(S473) was >2-fold higher in cells treatedwith both rapamycin and IGF-I as compared with cells treatedwith either agent alone.As IGF-I rescued cells from the antiproliferative effects of

rapamycin, we tested whether IGF-IR inhibition enhanced theantiproliferative effect of rapamycin in cells growing in serum. Cellswere treated with rapamycin, with or without NVP-AEW541, for 3days. Simultaneous administration of NVP-AEW541 and rapamycinto DU-145, MCF-7, and MDA-MB-468 (Fig. 4B) cancer cells resultedin additive antiproliferative effects as compared with either agentalone. Cell cycle analysis revealed additive effects on G1 arrestin the MCF-7 and DU-145 cell lines after 2 days of treatment withthe combination of IGF-IR and mTOR inhibitors as comparedwith cells treated with either single agent or vehicle (Fig. 4C).On comparison with control and single agent–treated cells, an

Figure 2. mTOR inhibition activates Akt in humans. A, livermetastasis of a colorectal carcinoma from a patient treated with adaily administration of RAD001 for 4 weeks. pAkt was expressed innuclei and cytoplasm of tumor cells (1) and levels of expressionincreased after therapy (2). Skin infiltration by ductal carcinoma in apatient treated with a weekly administration of RAD001 for 4 weeks.A similar increment of pAkt was observed before (3) and after therapy(4); (3,3V-diaminobenzidine, pAkt �400). B, evaluation of levels of pAkt inpaired tumor samples from patients treated with daily and weeklyadministration of RAD001 by immunohistochemistry. A significantincrement (P = 0.018) of this protein was observed in eight patients.C, tumor and biopsy characteristics. The characteristics of each tumorbiopsy and the dose schedule of the eight patients in the RAD001clinical trial are described.

Cancer Research

Cancer Res 2006; 66: (3). February 1, 2006 1504 www.aacrjournals.org

Research. on May 8, 2021. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase ...receptor tyrosine kinase signaling is a frequent event in tumor cells with constitutive mTOR activation. Reversal of this

enhanced sub-G1 population was observed in MDA-MB-468 cellsafter 2 days of treatment with combined mTOR and IGF-IRinhibitors (Fig. 4C).

Discussion

Dysregulation of mTOR function via physiologic or mutationalactivation of upstream pathways is a common event in tumorsfrom many lineages (2, 11). We speculated that tumor cells withactivated mTOR display a phenotype functionally equivalent toinsulin-resistant diabetes with an exaggerated down-regulationof upstream signaling molecules like IRS-1 (4–8). We report herethat, in a variety of tumor cell lines, the mTOR inhibitory drugrapamycin up-regulates IRS-1 protein levels and induces Aktphosphorylation, protein kinase activity, and downstream signal-ing. In parallel clinical studies with the rapamycin derivativeRAD001 (Novartis Pharma), the intensity of immunohistochemicalstaining of tumor biopsies for p-Akt was significantly elevated after4 weeks of drug treatment. These data suggest that the induction of

Akt activity observed in tumor cells in tissue culture is biologicallyrelevant and may be important clinically.Rapamycin and rapamycin-like molecules inhibit mTOR effi-

ciently in patients, are useful as immunosuppressants, andsuppress S6 kinase activity in normal and tumor cells in vitroand in vivo (18). Pharmacologic inhibition of mTOR has beenshown to potently inhibit tumor cells with activation of PI3K/Aktsignaling due either to PTEN loss, expression of Akt, or growthfactor activation (16). In this regard, rapamycin derivatives areeffective in in vivo models, as recently shown in myr-Akt-driventransgenic models of prostatic neoplasia (17). The Akt activationinduced by rapamycin in tumor cells, however, is likely to reduce itsantitumor effects, by activating pathways that attenuate its effectson proliferation and apoptosis. In tumors in which Akt activation isinduced, rapamycin will not effectively inhibit PI3K/Akt kinasesignaling except insofar as it is mediated through mTOR. We showhere that IGF-I overcomes the rapamycin-induced inhibition ofMCF-7 proliferation in serum-free medium. This result isconsistent with those of Houghton and coworkers, who showedthat induction of apoptosis by rapamycin via ASK-1 activation

Figure 3. IGF-I signaling mediates AKT activation induced bymTOR inhibition. A and B, the induction of pAkt (S473) byrapamycin was abrogated by the small-molecule inhibitor of IGF-IR,NVP-AEW541, and by the monoclonal antibody against IGF-IR, A12,in both the DU-145 prostate cancer cell line (A ) and the MCF-7breast cancer cell line (B ). Total Akt levels did not change. C, 1nmol/L rapamycin treatment up-regulates IRS-1 levels in MCF-7,DU-145, and MDA-MB-468 cell lines by 1 hour, and this inductionpersists for 24 hours. IRS-2 levels remain unchanged.

mTOR Inhibition Activates Akt

www.aacrjournals.org 1505 Cancer Res 2006; 66: (3). February 1, 2006

Research. on May 8, 2021. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase ...receptor tyrosine kinase signaling is a frequent event in tumor cells with constitutive mTOR activation. Reversal of this

Figure 4. IGF-I prevents andIGF-IR inhibitors enhance theantiproliferative effects ofrapamycin. A, IGF-I (60 ng/mL)rescued MCF-7 cells fromrapamycin’s antiproliferative effectsin serum-free medium and theinduction of pAKT was greater incells cotreated with rapamycin andIGF-I than in cells treated with eithersingle agent. B, the combinationof mTOR inhibition with IGF-IRinhibition resulted in enhancedantitumor effects. In vitro , combinedmTOR and IGF-IR inhibitionwith rapamycin (1 nmol/L) andNVP-AEW541 (1 Amol/L) resulted inadditive inhibition of proliferation inDU-145 cells, MCF-7 cells, andMDA-MB-468 cells as comparedwith either single agent.

Cancer Research

Cancer Res 2006; 66: (3). February 1, 2006 1506 www.aacrjournals.org

Research. on May 8, 2021. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase ...receptor tyrosine kinase signaling is a frequent event in tumor cells with constitutive mTOR activation. Reversal of this

occurs in serum-free but not serum-containing medium (22). Wefind that inhibition of induction of Akt activation with agents thatblock IGF-I signaling enhances cell cycle arrest and apoptosisinduction by rapamycin.Despite the results from model systems, the clinical antitumor

activity of mTOR inhibitor analogues has been modest at best.Our demonstration that rapamycin can induce Akt phosphory-lation in tumors implies that its potential antitumor activity isattenuated by release of feedback inhibition of growth signalingpathways. The results also suggest a new model for thedevelopment of effective combinatorial anticancer therapy.Combined inhibition of constitutively activated oncoproteinsand of normal pathways that are down-regulated by oncopro-tein-inhibition (and thus up-regulated by oncoprotein-targeteddrugs) may be much more effective than either alone. For thespecific case discussed in this article, the work provides arationale for tailored combination therapy with an mTORinhibitor and an inhibitor of the growth factor receptor, suchas IGF-IR, that normally drives PI3K activity in that tumor.Considering our results in which LY294002 abrogates rapamycin-induced Akt kinase activity, and the report by Sun et al. detailingthe combined efficacy of LY294002 and rapamycin in non–smallcell lung cancer cell lines, mTOR inhibitors and PI3K inhibitorsmight also be a promising combination therapy (15).We have shown that rapamycin induces Akt activity and that

abrogating this induction could enhance the antitumor effects ofrapamycin in vitro , but the mechanism of rapamycin-induced Akt

activity remains unclear. Our finding that rapamycin treatmentinduces IRS-1 expression suggests that rapamycin’s inhibition ofp70/S6K results in increased IGF-IR/IRS-1/PI3K signaling to Akt.Previous reports have shown that p70/S6K mediates phosphor-ylation of IRS-1 inhibitory serine sites (S312 and/or S636/639)which lead to IRS-1 degradation (8, 23, 24). Thus, suppression ofp70 activity by rapamycin may prevent inhibitory IRS-1 phos-phorylation, thereby stabilizing IRS-1. An increase in IRS-1adapter protein levels may induce Akt activity by augmentingIGF-IR signaling to PI3K/Akt. The inhibition of pAkt inductionwith LY294002 implies that the phenomenon is PI3K-dependentand supports this mechanism. However, LY294002 inhibits severalPI3K-like kinases, including mTOR, and has been reported toinhibit the activity of rictor-mTOR, recently described as the AktS473 kinase/PDK2 (25–27). It is possible that rapamycin, by someunknown mechanism, induces rictor-mTOR activity, resulting inincreased S473 Akt levels. These possibilities are currently underinvestigation.

Acknowledgments

Received 8/17/2005; revised 11/22/2005; accepted 11/30/2005.Grant support: NIH grant PO1-CA094060 and the generous support of the Taub

Foundation. K.E. O’Reilly was supported by the Medical Scientist Training ProgramGrant from the NIH.

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

Figure 4 Continued. C, in the breast cancer cell line, MCF-7, and the prostate cancer cell line, DU-145, 2 days of combined mTOR and IGF-IR inhibition withrapamycin (1 nmol/L) and NVP-AEW541 (1 Amol/L) resulted in enhanced G1 arrest. In the breast cancer cell line, MDA-MB-468, 2 days of combination treatmentresulted in enhanced apoptosis compared with single agent treatment groups.

mTOR Inhibition Activates Akt

www.aacrjournals.org 1507 Cancer Res 2006; 66: (3). February 1, 2006

Research. on May 8, 2021. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 9: mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase ...receptor tyrosine kinase signaling is a frequent event in tumor cells with constitutive mTOR activation. Reversal of this

References

1. Vivanco I, Sawyers CL. The phosphatidylinositol3-kinase AKT pathway in human cancer. Nat RevCancer 2002;2:489–501.

2. Bjornsti MA, Houghton PJ. The TOR pathway: a targetfor cancer therapy. Nat Rev Cancer 2004;4:335–48.

3. Oldham S, Hafen E. Insulin/IGF and target ofrapamycin signaling: a TOR de force in growth control.Trends Cell Biol 2003;13:79–85.

4. Haruta T, Uno T, Kawahara J, et al. A rapamycin-sensitive pathway down-regulates insulin signaling viaphosphorylation and proteasomal degradation ofinsulin receptor substrate-1. Mol Endocrinol 2000;14:783–94.

5. Pederson TM, Kramer DL, Rondinone CM. Serine/threonine phosphorylation of IRS-1 triggers its degra-dation: possible regulation by tyrosine phosphorylation.Diabetes 2001;50:24–31.

6. Sun XJ, Goldberg JL, Qiao LY, Mitchell JJ. Insulin-induced insulin receptor substrate-1 degradation ismediated by the proteasome degradation pathway.Diabetes 1999;48:1359–64.

7. Shah OJ, Wang Z, Hunter T. Inappropriate activationof the TSC/Rheb/mTOR/S6K cassette induces IRS1/2depletion, insulin resistance, and cell survival deficien-cies. Curr Biol 2004;14:1650–6.

8. Um SH, Frigerio F, Watanabe M, et al. Absence of S6K1protects against age- and diet-induced obesity whileenhancing insulin sensitivity. Nature 2004;431:200–5.

9. White MF, Yenush L. The IRS-signaling system: anetwork of docking proteins that mediate insulin andcytokine action. Curr Top Microbiol Immunol 1998;228:179–208.

10. Weinstein IB. Cancer. Addiction to oncogenes—theAchilles heal of cancer. Science 2002;297:63–4.

11. Mills GB, Lu Y, Kohn EC. Linking moleculartherapeutics to molecular diagnostics: inhibition of theFRAP/RAFT/TOR component of the PI3K pathwaypreferentially blocks PTEN mutant cells in vitro andin vivo . Proc Natl Acad Sci U S A 2001;98:10031–3.

12. Goulding H, Pinder S, Cannon P, et al. A newimmunohistochemical antibody for the assessment ofestrogen receptor status on routine formalin-fixed tissuesamples. Hum Pathol 1995;26:291–4.

13. Giaretti W, Nusse M. Light scatter of isolated cellnuclei as a parameter discriminating the cell-cyclesubcompartments. Methods Cell Biol 1994;41:389–400.

14. Bachman KE, Argani P, Samuels Y, et al. The PIK3CAgene is mutated with high frequency in human breastcancers. Cancer Biol Ther 2004;3:772–5.

15. Sun SY, Rosenberg LM, Wang X, et al. Activation of Aktand eIF4E survival pathways by rapamycin-mediatedmammalian target of rapamycin inhibition. Cancer Res2005;65:7052–8.

16. Neshat MS, Mellinghoff IK, Tran C, et al. Enhancedsensitivity of PTEN-deficient tumors to inhibition ofFRAP/mTOR. Proc Natl Acad Sci U S A 2001;98:10314–9.

17. Majumder PK, Febbo PG, Bikoff R, et al. mTORinhibition reverses Akt-dependent prostate intraepithe-lial neoplasia through regulation of apoptotic and HIF-1-dependent pathways. Nat Med 2004;10:594–601.

18. Boulay A, Zumstein-Mecker S, Stephan C, et al.Antitumor efficacy of intermittent treatment scheduleswith the rapamycin derivative RAD001 correlates withprolonged inactivation of ribosomal protein S6 kinase 1in peripheral blood mononuclear cells. Cancer Res 2004;64:252–61.

19. Garcia-Echeverria C, Pearson MA, Marti A, et al.In vivo antitumor activity of NVP-AEW541-A novel,potent, and selective inhibitor of the IGF-IR kinase.Cancer Cell 2004;5:231–9.

20. Burtrum D, Zhu Z, Lu D, et al. A fully humanmonoclonal antibody to the insulin-like growth factor Ireceptor blocks ligand-dependent signaling and inhib-its human tumor growth in vivo . Cancer Res 2003;63:8912–21.

21. Sawyers CL. Will mTOR inhibitors make it as cancerdrugs? Cancer Cell 2003;4:343–8.

22. Galvan V, Logvinova A, Sperandio S, Ichijo H,Bredesen DE. Type 1 insulin-like growth factorreceptor (IGF-IR) signaling inhibits apoptosis signal-regulating kinase 1 (ASK1). J Biol Chem 2003;278:13325–32.

23. Harrington LS, Findlay GM, Gray A, et al. TheTSC1-2 tumor suppressor controls insulin-PI3K sig-naling via regulation of IRS proteins. J Cell Biol 2004;166:213–23.

24. Manning BD. Balancing Akt with S6K: implicationsfor both metabolic diseases and tumorigenesis. J CellBiol 2004;167:399–403.

25. Sarbassov DD, Ali SM, Kim DH, et al. Rictor, anovel binding partner of mTOR, defines a rapa-mycin-insensitive and raptor-independent pathwaythat regulates the cytoskeleton. Curr Biol 2004;14:1296–302.

26. Hresko RC, Mueckler M. mTOR/RICTOR is theSer473 kinase for Akt/PKB in 3T3-L1 adipocytes. J BiolChem 2005;280:40406–16.

27. Sarbassov DD, Guertin DA, Ali SM, Sabatini DM.Phosphorylation and regulation of Akt/PKB by therictor-mTOR complex. Science 2005;307:1098–101.

Cancer Research

Cancer Res 2006; 66: (3). February 1, 2006 1508 www.aacrjournals.org

Research. on May 8, 2021. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 10: mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase ...receptor tyrosine kinase signaling is a frequent event in tumor cells with constitutive mTOR activation. Reversal of this

2006;66:1500-1508. Cancer Res   Kathryn E. O'Reilly, Fredi Rojo, Qing-Bai She, et al.   Kinase Signaling and Activates AktmTOR Inhibition Induces Upstream Receptor Tyrosine

  Updated version

  http://cancerres.aacrjournals.org/content/66/3/1500

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/66/3/1500.full#ref-list-1

This article cites 27 articles, 13 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/66/3/1500.full#related-urls

This article has been cited by 100 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. (CCC)Click on "Request Permissions" which will take you to the Copyright Clearance Center's

.http://cancerres.aacrjournals.org/content/66/3/1500To request permission to re-use all or part of this article, use this link

Research. on May 8, 2021. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from