mk-2206, a novel allosteric inhibitor of akt, synergizes...

12
Preclinical Development MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes with Getinib against Malignant Glioma via Modulating Both Autophagy and Apoptosis Yan Cheng 1 , Yi Zhang 1,2 , Li Zhang 2 , Xingcong Ren 1 , Kathryn J. Huber-Keener 1 , Xiaoyuan Liu 2 , Lei Zhou 2 , Jason Liao 1 , Heike Keihack 3 , Li Yan 3 , Eric Rubin 3 , and Jin-Ming Yang 1 Abstract Gefitinib, a small molecule inhibitor of the epidermal growth factor receptor tyrosine kinase, has been shown to induce autophagy as well as apoptosis in tumor cells. Yet, how to use autophagy and apoptosis to improve therapeutic efficacy of this drug against cancer remains to be explored. We reported here that MK-2206, a potent allosteric Akt inhibitor currently in phase I trials in patients with solid tumors, could reinforce the cytocidal effect of gefitinib against glioma. We found that cotreatment with gefitinib and MK-2206 increased the cytotoxicity of this growth factor receptor inhibitor in the glioma cells, and the CompuSyn synergism/ antagonism analysis showed that MK-2206 acted synergistically with gefitinib. The benefit of the combinatorial treatment was also shown in an intracranial glioma mouse model. In the presence of MK-2206, there was a significant increase in apoptosis in glioma cells treated with gefitinib. MK-2206 also augmented the autophagy- inducing effect of gefitinib, as evidenced by increased levels of the autophagy marker, LC3-II. Inhibition of autophagy by silencing of the key autophagy gene, beclin 1 or 3-MA, further increased the cytotoxicity of this combinatorial treatment, suggesting that autophagy induced by these agents plays a cytoprotective role. Notably, at 48 hours following the combinatorial treatment, the level of LC3-II began to decrease but Bim was significantly elevated, suggesting a switch from autophagy to apoptosis. On the basis of the synergistic effect of MK-2206 on gefitinib observed in this study, the combination of these two drugs may be utilized as a new therapeutic regimen for malignant glioma. Mol Cancer Ther; 11(1); 154–64. Ó2011 AACR. Introduction Activation of epidermal growth factor receptor (EGFR), a cell surface protein belonging to the ErbB receptor tyrosine kinase family, exerts stimulatory effects on a number of oncogenic signaling cascades, such as RAS, phosphoinositide 3-kinase (PI3K), and mitogen-activated protein kinase pathways, thereby promoting prolifera- tion, growth, and survival of tumor cells (1). On the basis of the essential role of the EGFR-initiated signaling in tumor development and progression, this receptor tyro- sine kinase has been actively pursued as a therapeutic target for cancer treatment (2). Thus far, EGFR inhibitors such as gefitinib, cetuximab, and erlotinib have already been approved as targeted therapies for treating patients with lung cancer, colon cancer, breast cancer, and some other types of malignancies (3). Malignant gliomas are the most common and aggres- sive type of primary brain tumors in humans. Despite optimal treatment with surgery, chemotherapy, and radi- ation therapy, the prognosis of this disease remains poor. Thus, more effective therapeutic interventions are urgent- ly needed to improve the treatment of this malignancy. Because mutations or aberrant expression of EGFR is frequent in this type of cancer, use of inhibitors of EGFR is believed to be an effective therapeutic intervention for patients with malignant gliomas (4, 5). Indeed, EGFR inhibitors such as lapatinib have been reported to exhibit inhibitory effects on proliferation and migration of glioma cells and to activate apoptosis (6). Gefitinib (Iressa, ZD1839), another selective inhibitor of EGFR tyrosine kinase, has also been shown to possess inhibitory effects on both cell viability and invasion in malignant gliomas that have amplification of EGFR (7). In addition, inhibitors of EGFR such as erlotinib and gefitinib have been reported to inhibit proliferation of tumor-initiating cells in human glioma (8). Nevertheless, in the reported clinical trials, Authors' Afliations: 1 Department of Pharmacology and The Penn State Hershey Cancer Institute, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey; 2 Department of Pharmacology, School of Pharmacy, Soochow University, Jiangsu Province, China; and 3 Merck & Co. Inc., North Wales, Pennsylvania Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). Corresponding Authors: Jin-Ming Yang, Penn State College of Medicine, Department of Pharmacology and The Penn State Hershey Cancer Insti- tute, CH74, 500 University Drive, P.O. Box 850, Hershey, PA 17033. Phone: 717-531-1630; Fax: 717-531-0011; E-mail: [email protected]; and Yan Cheng, E-mail: [email protected] doi: 10.1158/1535-7163.MCT-11-0606 Ó2011 American Association for Cancer Research. Molecular Cancer Therapeutics Mol Cancer Ther; 11(1) January 2012 154 on August 9, 2019. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst November 4, 2011; DOI: 10.1158/1535-7163.MCT-11-0606

Upload: hoangkhuong

Post on 10-Aug-2019

219 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes ...mct.aacrjournals.org/content/molcanther/11/1/154.full.pdf · Preclinical Development MK-2206, a Novel Allosteric Inhibitor

Preclinical Development

MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes withGefitinib against Malignant Glioma via Modulating BothAutophagy and Apoptosis

Yan Cheng1, Yi Zhang1,2, Li Zhang2, Xingcong Ren1, Kathryn J. Huber-Keener1, Xiaoyuan Liu2,Lei Zhou2, Jason Liao1, Heike Keihack3, Li Yan3, Eric Rubin3, and Jin-Ming Yang1

AbstractGefitinib, a smallmolecule inhibitor of the epidermal growth factor receptor tyrosine kinase, has been shown

to induce autophagy as well as apoptosis in tumor cells. Yet, how to use autophagy and apoptosis to improve

therapeutic efficacy of this drugagainst cancer remains to be explored.We reportedhere thatMK-2206, apotent

allosteric Akt inhibitor currently in phase I trials in patients with solid tumors, could reinforce the cytocidal

effect of gefitinib against glioma. We found that cotreatment with gefitinib and MK-2206 increased the

cytotoxicity of this growth factor receptor inhibitor in the glioma cells, and the CompuSyn synergism/

antagonismanalysis showed thatMK-2206 acted synergisticallywith gefitinib. The benefit of the combinatorial

treatment was also shown in an intracranial glioma mouse model. In the presence of MK-2206, there was a

significant increase in apoptosis in glioma cells treatedwith gefitinib.MK-2206 also augmented the autophagy-

inducing effect of gefitinib, as evidenced by increased levels of the autophagy marker, LC3-II. Inhibition of

autophagy by silencing of the key autophagy gene, beclin 1 or 3-MA, further increased the cytotoxicity of this

combinatorial treatment, suggesting that autophagy induced by these agents plays a cytoprotective role.

Notably, at 48 hours following the combinatorial treatment, the level of LC3-II began to decrease but Bim was

significantly elevated, suggesting a switch fromautophagy to apoptosis. On the basis of the synergistic effect of

MK-2206 on gefitinib observed in this study, the combination of these two drugs may be utilized as a new

therapeutic regimen for malignant glioma. Mol Cancer Ther; 11(1); 154–64. �2011 AACR.

Introduction

Activation of epidermal growth factor receptor (EGFR),a cell surface protein belonging to the ErbB receptortyrosine kinase family, exerts stimulatory effects on anumber of oncogenic signaling cascades, such as RAS,phosphoinositide 3-kinase (PI3K), and mitogen-activatedprotein kinase pathways, thereby promoting prolifera-tion, growth, and survival of tumor cells (1). On the basisof the essential role of the EGFR-initiated signaling intumor development and progression, this receptor tyro-

sine kinase has been actively pursued as a therapeutictarget for cancer treatment (2). Thus far, EGFR inhibitorssuch as gefitinib, cetuximab, and erlotinib have alreadybeen approved as targeted therapies for treating patientswith lung cancer, colon cancer, breast cancer, and someother types of malignancies (3).

Malignant gliomas are the most common and aggres-sive type of primary brain tumors in humans. Despiteoptimal treatment with surgery, chemotherapy, and radi-ation therapy, the prognosis of this disease remains poor.Thus,more effective therapeutic interventions are urgent-ly needed to improve the treatment of this malignancy.Because mutations or aberrant expression of EGFR isfrequent in this type of cancer, use of inhibitors of EGFRis believed to be an effective therapeutic intervention forpatients with malignant gliomas (4, 5). Indeed, EGFRinhibitors such as lapatinib have been reported to exhibitinhibitory effects on proliferation andmigration of gliomacells and to activate apoptosis (6). Gefitinib (Iressa,ZD1839), another selective inhibitor of EGFR tyrosinekinase, has also been shown to possess inhibitory effectson both cell viability and invasion in malignant gliomasthat have amplification of EGFR (7). In addition, inhibitorsof EGFR such as erlotinib andgefitinib have been reportedto inhibit proliferation of tumor-initiating cells in humanglioma (8). Nevertheless, in the reported clinical trials,

Authors' Affiliations: 1Department of Pharmacology and The PennState Hershey Cancer Institute, The Pennsylvania State UniversityCollege of Medicine and Milton S. Hershey Medical Center, Hershey;2Department of Pharmacology, School of Pharmacy, SoochowUniversity, Jiangsu Province, China; and 3Merck & Co. Inc., NorthWales, Pennsylvania

Note: Supplementary data for this article are available at Molecular CancerTherapeutics Online (http://mct.aacrjournals.org/).

Corresponding Authors: Jin-Ming Yang, Penn State College of Medicine,Department of Pharmacology and The Penn State Hershey Cancer Insti-tute, CH74, 500University Drive, P.O. Box 850, Hershey, PA 17033. Phone:717-531-1630; Fax: 717-531-0011; E-mail: [email protected]; and YanCheng, E-mail: [email protected]

doi: 10.1158/1535-7163.MCT-11-0606

�2011 American Association for Cancer Research.

MolecularCancer

Therapeutics

Mol Cancer Ther; 11(1) January 2012154

on August 9, 2019. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 4, 2011; DOI: 10.1158/1535-7163.MCT-11-0606

Page 2: MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes ...mct.aacrjournals.org/content/molcanther/11/1/154.full.pdf · Preclinical Development MK-2206, a Novel Allosteric Inhibitor

gefitinib only showed modest antiglioma efficacy (9).More recently, it was shown that, although gefitinib canreach the brain tumors in high concentrations and effi-ciently dephosphorylate EGFR, this drug is not sufficientto repress the activity of the pathway (10). It is likelythat single molecular targeted therapy may not beenough to control the complex pathogenesis of glioblas-toma, and that targeting multiple signaling molecules ofthe EGFR-initiated pathways may produce better antitu-mor effects.TheAkt serine/threonine kinase family, which consists

of Akt1, Akt2, and Akt3, is a central component of the PI3kinase pathway, and aberrant activation ofAkt is found tobe common in malignant glioma (11, 12). The activity ofAkt is closely regulated by the EGFR signaling and hasinhibitory effects on apoptosis. Recently, inhibiting Akthas been shown to possess autophagy-inducing effect inaddition to apoptogenic effect (13), so as do the EGFRinhibitors including gefitinib (14, 15). Autophagy is acatabolic process that degrades cytoplasmic componentsvia the lysosomal machinery. Induction of autophagy canpromote either cell survival or cell death, depending ondifferent circumstances (16). In light of the critical role ofAkt in the EGFR-initiated pathway, we sought to deter-mine whether dual inhibitions of EGFR and Akt by cor-responding inhibitors would reinforce the antigliomaefficacy of EGFR inhibitors, and what roles autophagywould play in determining the antitumor efficacy of thecombinatorial therapy. It has been reported that the novelsmallmolecule allosteric inhibitor ofAkt,MK-2206,whichis being tested both in preclinical settings and clinicaltrials as an anticancer agent, can synergistically enhancethe antitumor efficacy of some conventional chemother-apeutic drugs andmolecular targeted agents in preclinicalmodels of lung cancer, ovarian cancer, and breast cancer(17–19). In this study, we focused on testing whethercombining MK-2206 with gefitinib would improve theantiglioma activity of this EGFR inhibitor, andhow induc-tion of autophagy would affect apoptotic cell deathcaused by dual inhibitions of EGFR and Akt. We foundthat MK-2206 synergistically enhanced the cytocidaleffects of gefitinib on glioma cells, and the mechanism ofthis synergism involved the Akt inhibition–mediatedmodulation of apoptosis and autophagy in tumor cellstreated with the targeted therapies. The results of thisstudy recommend that that combined administration ofgefitinib and MK-2206 should be considered as a newtherapeutic tactic that warrants clinical investigation fortreating patients with malignant brain tumors.

Materials and Methods

Cell lines and cultureThe human glioblastoma cell lines, T98G, LN229, and

U87MG were purchased from American Type CultureCollection. These glioma cell lines all express EGFR. T98Gcells were cultured in Ham’s F-10: DMEM (10:1) medium,and LN229, U251, and U87MG cells were cultured in

Dulbecco’s Modified Eagle’s Medium (DMEM) medium.These media were supplemented with 10% FBS, 100units/mL of penicillin, and 100 mg/mL of streptomycin.Cells were maintained at 37�C in a humidified atmo-sphere containing 5% CO2/95% air. All cell cultures weremonitored routinely and found to be free of contamina-tion byMycoplasma or fungi. All cell lines were discardedafter 3 months and new lines propagated from frozenstocks.

Reagents and antibodiesMK-2206 was a gift from Merck & Co. Inc. Gefitinib

was purchased from LC Laboratories. 3-MA, BafilomycinA1 and MTT were purchased from Sigma. Antibodiesto LC-3, Akt, Bim, survivin, phospho-mTOR, mTOR,phospho-S6 kinase (Ser371), S6 kinase, phospho-EGFR,and EGFR, were purchased from Cell Signaling Techno-logies. Antibodies to Beclin 1 and a-tubulin were pur-chased from Santa Cruz. All cell culture media werepurchased from Invitrogen. Western blot reagents wereobtained from Pierce Biotechnology.

Apoptosis assayApoptosis was determined by flow cytometric analysis

of Annexin V and 7-AAD staining. Briefly, 100 mL ofGuava Nexin reagent (Millipore) was added to 1 � 105

cells (in 100mL), and the cellswere then incubatedwith thereagent for 20minutes at room temperature in the dark.Atthe end of incubation, the samples were analyzed by aGuava EasyCyte Plus FlowCytometry System (Millipore).

Western blotCells were lysed in the M-PER mammalian protein

extraction reagent (Thermo Scientific) supplementedwith a protease inhibitor cocktail (Roche) at room tem-perature for 5 minutes, followed by centrifugation at14,000 � g for 10 minutes. Protein concentrations of celllysates were measured using the Bio-Rad DC assayreagent (Bio-Rad). Proteins (20–40 mg) were resolvedby SDS-PAGE and then transferred to polyvinylidenedifluoride (PVDF) membrane (Bio-Rad). The PVDFmembranes were incubated with the respective antibo-dies in 3% BSA/TBST at 4�C for overnight, followed byincubation with secondary antibody at room tempera-ture for 1 hour. Protein signals were detected byenhanced chemiluminescence method following themanufacturer’s protocol.

Short interfering RNA transfectionShort interfering RNA (siRNA) targeting Beclin 1 was

prepared by Dharmacon Research. Scrambled siRNA wasused as a control. Transfection of siRNAwas done accord-ing to the manufacturer’s protocol. Briefly, cells in expo-nential phase of growthwereplated in 6-well tissue cultureplates at 1 � 105cells per well, grown for 24 hours, thentransfected with siRNA using Oligofectamine and OPTI-MEM I–reduced serum medium. The concentrations ofsiRNAs were chosen based on dose–response studies.

Synergistic Antitumor Effect of MK-2206 and Gefitinib

www.aacrjournals.org Mol Cancer Ther; 11(1) January 2012 155

on August 9, 2019. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 4, 2011; DOI: 10.1158/1535-7163.MCT-11-0606

Page 3: MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes ...mct.aacrjournals.org/content/molcanther/11/1/154.full.pdf · Preclinical Development MK-2206, a Novel Allosteric Inhibitor

Cellular viability assayCell viability wasmeasured byMTT assay. Briefly, cells

were plated at 5 � 103 cells per well in 96-well tissueculture plates and subjected to different treatments at37�C for 72 hours in a humidified atmosphere containing5% CO2/95% air. At the end of treatments, MTT wasadded to each well and incubated for another 4 hours.The formazan product was dissolved in dimethyl sulfox-ide (DMSO) and read at 570 nm on a Victor3 Multi Labelplate reader (PerkinElmer).

Animal experimentsSix-week-old male BALB/c nude mice were used for

intracerebral implantation of glioma cells. Briefly, humanglioma cells LN229 (1 � 105 cells in 15 mL of DMEMmedium) were injected into the brains at 4 mm depthunder anesthesiawith chloralic hydras (4%, 2mL/kg, i.p.).Three days after tumor cell implantation, mice wererandomly divided into 4 groups (15 mice per group).Treatments were begun on day 4, according to the follow-ing regimens: gefitinib, 80mg/kg, orally, for 5 consecutivedays; MK-2206, 100 mg/kg, orally, 3 times per week for 2weeks; gefitinib þ MK-2206, given as same as above;control, 10% DMSO in saline, orally, given as same asabove. Animal body weight and physical signs were mon-itoreddaily during the experiments. Themicewerehousedin a temperature-controlled and light-controlled environ-ment in the animal facility. At day 17 postinoculation, themice were euthanized, and the brains were fixed in 10%buffered formalin, embedded in paraffin and then stainedwith hematoxylin-eosin (H&E). The animal experimentswere approved by the Institutional Animal Care and UseCommittee of our university.

Statistical analysisThe differences between treatments were analyzed

using a 2-sample t test. The survival curves of thetumor-bearing mice subjected to different treatmentswere estimated using Kaplan–Meier method and com-pared by log-rank statistic analysis.

Results

Gefitinib simultaneously induces apoptosis andautophagy in human glioma cells

Apoptosis and autophagy are the 2 cellular processeslikely to alter efficacy of a therapeutic agent. In this study,we observed that treatment of the human glioma cell linesLN229 and T98G with gefitinib, which decreased thephosphorylation of p-EGFR (Fig. 1B), triggered both apo-ptosis and autophagy in a dose-dependent manner, asevidenced by increases in the number of cells withAnnexin V staining (Fig. 1C), an indicator of apoptosis,and in the amount of LC3 II (Fig. 1D), a specific marker ofautophagy. We also showed that in the presence of bafi-lomycin A1 (an inhibitor of lysosomal protease), theaccumulation of LC3 II was more abundant, indicatingan increase in autophagic flux (Fig. 1D). Stimulatory effect

of low concentration (0.5 mmol/L) but inhibitory effect ofhigh concentrations (1 and 5mmol/L) of gefitinib onEGFRphosphorylation was also observed by others (20). Treat-ment with gefitinib also caused a dose-dependentdecrease in the phosphorylation of Akt (Fig. 1E), whichindicates an inhibition of Akt activity. These results implythat activation of apoptosis and autophagy in response togefitinib treatment could be a part of cellular processesthat determine the cytocidal efficacy of this EGFRinhibitor.

The Akt inhibitor, MK-2206, synergisticallyincreases the cytocidal effect of gefitinib on humanglioma cells

As gefitinib showed an inhibitory effect on Akt activity(Fig. 1E), and this actionmight account, at least in part, forits proapoptotic effect,wenext tested inLN229, T98G, andU87MG human glioma cell lines whether combining anAkt inhibitor with gefitinib would enhance the cytocidalefficacy of this EGFR inhibitor. Figure 2 shows that thenovel allosteric small molecule Akt inhibitor MK-2206, ata noncytotoxic concentration (0.5 mmol/L), inhibited theactivation of Akt (Fig. 2B) and increased the cytocidalactivity of gefitinib against the glioma cells (Fig. 2C–E). Inthe presence of MK-2206, the IC50 values of gefitinib weredecreased by approximately 2- to 3-fold (Table 1).Although the IC50 values of gefitinib in the presence ofMK-2206 were still in the mmol/L ranges, these concen-trations are achievable in the brain tumor tissues, asgefitinib has a high ability to penetrate into solid tumortissues, including glioma (21–23), and gefitinib concen-tration in tumor tissues can be 10- to 40-fold higher thanthat in blood (21–26). Analysis of the effects of the drugcombination using the CompuSyn synergism/antago-nism analysis software (ComboSyn, Inc.) informed thatMK-2206 acted synergistically with gefitinib in producingcytocidal activity. The combination indexes obtainedwere all below 1.0 (range: 0.16–0.85; Table 2), indicatingthat synergism varies between strong and moderate (27).

MK-2206 augments both apoptogenic andautophagenic effects of gefitinib at early stages butswitches autophagy to apoptosis at late stages of thetreatment

To investigate the mechanisms underlying the syner-gistic cytocidal effect between gefitinib and MK-2206,we compared apoptotic and autophagic activity in cellstreated with gefitinib alone or with combination ofgefitinib and MK-2206. As shown in Fig. 3A, cotreat-ment with gefitinib and MK-2206 for 60 hours increasedthe percentage of cells with Annexin V staining, ascompared with treatment with gefitinib alone, indicat-ing an increase in apoptosis. Augmentation of apoptosisby this combined drug treatment was further evidencedby a downregulation of the antiapoptotic protein, sur-vivin, and an upregulation of the proapoptotic protein,Bim (Fig. 3B). Autophagic activity, as determined byWestern blot of LC3 II and microscopic observation of

Cheng et al.

Mol Cancer Ther; 11(1) January 2012 Molecular Cancer Therapeutics156

on August 9, 2019. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 4, 2011; DOI: 10.1158/1535-7163.MCT-11-0606

Page 4: MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes ...mct.aacrjournals.org/content/molcanther/11/1/154.full.pdf · Preclinical Development MK-2206, a Novel Allosteric Inhibitor

GFP-LC3 localization, was also higher in the cellscotreated with gefitinib andMK-2206, as compared withthose treated with gefitinib alone (Fig. 3C and D),indicating that MK-2206 also augments the gefitinib-induced autophagy.To delineate the relationship between autophagy and

apoptosis activated by gefitinib andMK-2206, we extend-ed our observation of these 2 cellular processes to 96hours.We found that activation of autophagy by gefitinib,as indicated by the levels of LC3 II, lasted up to 96 hours(Fig. 4A); by contrast, in the presence of MK-2206, au-tophagy levels reached a peak at 48 hours, but began todecline thereafter (Fig. 4B). Notably, with the decrease ofautophagy (LC3 II) after 48 hours, the level of the proa-poptotic protein Bim was further increased (Fig. 4B). Incontrast, no detectable increase in the Bim level wasobserved in the cells treated with gefitinib alone

(Fig. 4A). Elevations of Bim in the tumor cells subjectedto combinatory treatment were accompanied by increasesin apoptotic cell death (Fig. 4C). The importance of Bim ininduction of apoptosis was further shown in the experi-ments showing that knockdownofBimby siRNAreducedapoptotic rate in those treated cells (Fig. 4D). These resultssuggested that excessive autophagy promoted by MK-2206 may trigger a switch from autophagy to apoptoticcell death.

Suppression of autophagy enhances the efficacy ofcombined treatment with gefitinib and MK-2206

As autophagy can play either a prosurvival or prodeathrole under various stressful conditions, we nextwanted tounderstand how activation of autophagy affected viabil-ity of glioma cells treated with gefitinib alone or withthe combination of gefitinib and MK-2206. In these

LN229C T98G

D

Baf A1T98G

Gefi (µmol/L) 0 1 5 1 5

LC3 I

LC3 II

Tubulin

LN229

Gefi (µmol/L) 0 1 5 1 5

Baf A1

LC3 I

LC3 II

Tubulin

LC3 II/Tubulin 1 3.8 10.0 8.5 15.8

1 5.0 13.7 15.5 20.3LC3 II/Tubulin

p-AKT(ser 473)

Gefi (µmol/L) 0 1 5 10

Tubulin

LN229

Akt

E

p-AKT/Tubulin 1 0.8 0.8 0.3

p-AKT(ser 473)

Gefi (µmol/L) 0 1 5 10

Tubulin

T98G

Akt

p-AKT/Tubulin 1 1 0.9 0.2

A

p-EGFR(Tyr1068)

Tubulin

LN229

EGFR

p-EGFR/Tubulin 1 2.6 1.2 0.7

Gefi (µmol/L) 0 0.5 1 5

p-EGFR(Tyr1068)

Tubulin

T98G

EGFR

p-EGFR/Tubulin 1 2.9 2.2 0.9

Gefi (µmol/L) 0 0.5 1 5

B

N

NO

NH

F

Cl

N

O

MeO

Con

Gefi 5 µmol/L

1.9%

1.1%

97.1%

0.0%

Con

98.0% 0.5%

1.5%0.0%

Gefi 1 µmol/L

2.9%

4.9%

91.9%

0.3%

12.4%

6.5%

80.9%

0.2%

0

5

10

15

20

25

0 1 5

Ap

op

tosis

(%

)

Gefitinib (µmol/L)

88.9% 7.1%

3.4%0.6%

95.6% 2.6%

1.9%0.0%

0

5

10

15

20

25

510

Ap

op

tosis

(%

)

Gefitinib (µmol/L)

Gefitinib

Gefi 1 µmol/L

Gefi 5 µmol/L

Figure 1. Simultaneous induction of apoptosis and autophagy and inhibition of Akt by gefitinib in human glioma cells. A, chemical structure of gefitinib.B, LN229 and T98G cells were treated with gefitinib for 24 hours; the levels of p-EGFR and EGFR were examined by Western blot. Tubulin was used as aloading control. C, LN229 and T98G cells cultured in medium supplemented with 10% FBS were treated with gefitinib for 60 hours, and apoptosis wasexamined bymeasuring Annexin V staining using flow cytometry. D, LN229 and T98G cells were treated with gefitinib for 24 hours in the absence or presenceof Bafilomycin A1, and the levels of LC3 were examined by Western blot. Tubulin was used as a loading control. E, LN229 and T98G cells were treated withgefitinib for 48 hours, and the levels of Akt and p-Akt were measured by Western blot. Tubulin was used as a loading control.

Synergistic Antitumor Effect of MK-2206 and Gefitinib

www.aacrjournals.org Mol Cancer Ther; 11(1) January 2012 157

on August 9, 2019. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 4, 2011; DOI: 10.1158/1535-7163.MCT-11-0606

Page 5: MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes ...mct.aacrjournals.org/content/molcanther/11/1/154.full.pdf · Preclinical Development MK-2206, a Novel Allosteric Inhibitor

experiments, we first suppressed autophagy by silenc-ing the expression of Beclin 1, a key autophagy-regu-latory gene, and then determined the viability of tumorcells subjected to the treatments. Figure 5A and B showthat inhibition of autophagy via transfection with aBeclin 1–targeted siRNA further decreased viability ofthe cells treated with the combinatorial treatment,as compared with transfection with a nontargetingRNA. The reduced cellular viability seen in Fig. 5A andB seems to result from apoptosis, as suppression of

autophagy by silencing Beclin 1 further increased apo-ptotic cell death rate of the cells subjected to the com-binatorial treatment (Fig. 5C and D). The effect ofautophagy on cytocidal effect of the combinatorial treat-ment was further verified through use of the autophagyinhibitor, 3-MA (1 mmol/L; Supplementary Fig. S1Aand S1B). These results suggest that induction of

LN229

–20

0

20

40

60

80

100

120

201052.510.50C

ell

via

bili

ty (

%)

Gefitinib (µmol/L)

Gefitinib (µmol/L)

Gefitinib (µmol/L)

-MK-2206

+MK-2206

**

****

*

C

0

20

40

60

80

100

120

201052.510.50

Cell

via

bili

ty (

%)

-MK-2206

+MK-2206

T98G

***

**

**

D

A

p-AKT(ser 473)

MK-2206 (µmol/L) 0 0.5

LN229

Tubulin

B

AKT

p-AKT(ser 473)

MK-2206 (µmol/L) 0 0.5

T98G

Tubulin

AKT

N

N

NH2

NH

O

N

0

20

40

60

80

100

120

201052.510.50

Ce

ll via

bili

ty (

%)

-MK-2206

+MK-2206

U87MG

*****

**

**

E

MK-2206

Figure2. Effects of gefitinib onviability of gliomacells in the absenceor presenceofMK-2206. A, chemical structure ofMK-2206.B, LN229andT98GcellsweretreatedwithMK-2206 for 24 hours, and the levels of p-Akt and Akt were examined byWestern blot. Tubulin was used as a loading control. C–E, LN229, T98G,and U87MG human glioma cells cultured in medium supplemented with 10% FBS were treated with a series of concentrations of gefitinib for 72 hours in theabsenceor presenceof 0.5mmol/L ofMK-2206. At the endof treatments, the cell viabilitywasmeasuredbyMTTassay. Eachbar represents themean�SEof 3experiments. �, P < 0.05; ��, P < 0.01.

Table 1. IC50 values of gefitinib in the absenceor presence of 0.5 mmol/L of MK-2206

IC50 (mmol/L)

Cell line Gefitinib GefitinibþMK-2206

LN229 7.9 2.5T98G 12.7 4.9U87MG 17.8 5.9

Table 2. Combination indices for gefitinib andMK-2206 combination therapy, as computed byCompuSyn

Combination index

Cell line ED50 ED75 ED90

LN229 0.46 0.26 0.16T98G 0.46 0.34 0.23U87MG 0.85 0.37 0.18

NOTE: CI < 1.0 suggests synergy.

Cheng et al.

Mol Cancer Ther; 11(1) January 2012 Molecular Cancer Therapeutics158

on August 9, 2019. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 4, 2011; DOI: 10.1158/1535-7163.MCT-11-0606

Page 6: MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes ...mct.aacrjournals.org/content/molcanther/11/1/154.full.pdf · Preclinical Development MK-2206, a Novel Allosteric Inhibitor

autophagy serves as a compensatory mechanism intumor cells in response to the targeted therapies.

Regulation of autophagy in glioma cells cotreatedwith gefitinib and MK-2206 is mediated via themTOR-S6 kinase pathwayTo explore the signaling pathway involved in the

modulation of autophagy by combinatorial treatmentwith gefitinib and MK-2206, we examined the activitiesof mTOR and S6 kinase, 2 components downstream ofthe EGFR–PI3 kinase–Akt cascade with known roles inthe regulation of autophagy in response to cellularstress. As shown in Fig. 5E and F, in comparison withtreatment with gefitinib alone, the combinatorial treat-ment with gefitinib and MK-2206 markedly decreased

the levels of phospho-mTOR and phospho-S6 kinase,indicating a deactivation of the mTOR–S6 kinase path-way. Because inhibition of mTOR and S6 kinase isknown to induce autophagy, the decreases in the activ-ity of this pathway by dual inhibition of EGFR and Aktmay account for the activation of autophagy by gefitiniband MK-2206, as shown in Fig. 3C.

Combinatorial treatment with gefitinib and MK-2206 produces a stronger antitumor activity in anintracranial glioma mouse model

To determine the in vivo relevance of the above obser-vations, we tested the therapeutic benefits of the combi-natorial treatment with gefitinib and MK-2206 in anorthotopic glioma mouse model in which the LN229

LN229

T98G

A LN229

Gefitinib (µmol/L) 0 1 5 0 1 5

Gefitinib (µmol/L) 0 1 5 0 1 5

Gefitinib (µmol/L) 0 1 5 0 1 5

Gefitinib (µmol/L) 0 1 5 0 1 5

MK-2206

Tubulin

Survivin

B

Bim

T98GMK-2206

Tubulin

Survivin

Bim

LN229

MK-2206

LC3 I

LC3 II

Tubulin

C

T98GMK-2206

LC3 I

LC3 II

Tubulin

D

**

LN229

T98G

GefitinibControl Gefitinib+MK-2206

0

10

20

30

40

50

60

70

Cells

with G

FP

-LC

3

puncta

(%

)

LN229

0

10

20

30

40

50

60

70

Cells

with G

FP

-LC

3

puncta

(%

)

T98G

Con Ge Ge+MK Con Ge Ge+MK

**

**

Survivin/Tubulin 1 1 0.8 1 0.8 0.2

Bim/Tubulin 1 1.6 6.6 2.2 9.3 13.5LC3 II/Tubulin 1 1.6 5.7 0.8 4.9 8.3

Survivin/Tubulin 1 1.1 0.9 1 0.7 0.3

Bim/Tubulin 1 1.2 3.1 1 5.1 6.5LC3 II/Tubulin 1 3.0 6.5 1.3 6.5 9.5

0

5

10

15

20

25

30

35

510

Apopto

sis

(%

)

Gefitinib (µmol/L)

Gefitinib (µmol/L)

-MK-2206

+MK-2206

**

*

0

5

10

15

20

25

30

35

510

Apopto

sis

(%

)

-MK-2206

+MK-2206

Figure 3. Effects ofMK-2206 on the gefitinib-induced apoptosis and autophagy. A, LN229 and T98G cells were treatedwith various concentrations of gefitinibfor 60 hours in the absence or presence of 0.5 mmol/L of MK-2206. At the end of treatment, apoptosis was examined by measuring Annexin V staining usingflow cytometry. Each bar represents the mean � SE of 3 experiments. �, P < 0.05; ��, P < 0.01. B, LN229 and T98G cells were treated with variousconcentrations of gefitinib for 24 hours in the absence or presence of 0.5 mmol/L of MK-2206. The levels of survivin and Bimwere examined byWestern blot.Tubulinwasused as a loading control. C, LN229and T98Gcellswere treatedwith gefitinib for 24 hours in the absence or presenceof 0.5mmol/LMK-2206, andthe levels of LC3 were examined by Western blot. Tubulin was used as a loading control. D, LN229 and T98G cells were transfected with a GFP-LC3plasmid and then treated with gefitinib for 24 hours in the absence or presence of 0.5 mmol/L of MK-2206. At the end of treatment, the cells were examinedby fluorescence microscopy. Bars are the quantification of the percentage of cells with 10 or more GFP-LC3 puncta. At least 100 cells were scoredin each treatment. ��, P < 0.01, t test.

Synergistic Antitumor Effect of MK-2206 and Gefitinib

www.aacrjournals.org Mol Cancer Ther; 11(1) January 2012 159

on August 9, 2019. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 4, 2011; DOI: 10.1158/1535-7163.MCT-11-0606

Page 7: MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes ...mct.aacrjournals.org/content/molcanther/11/1/154.full.pdf · Preclinical Development MK-2206, a Novel Allosteric Inhibitor

human glioma cells were implanted intracranially. Inthese experiments, the tumor-bearing mice were eithertreated with vehicle, gefitinib (80 mg/kg) or MK-2206(100 mg/kg) alone, or with combination of the 2drugs. Figure 6A shows that as compared with gefitinibtreatment alone, coadministration of this EGFR inhibi-tor with MK-2206 showed a better therapeutic bene-fit, as indicated by a significant increase in the survivalof the tumor-bearing mice (P ¼ 0.0155). The greaterantitumor effect of this combinatorial treatment thangefitinib alone was also evidenced by histologic exam-inations of the brain tissues on the day 17 followingtumor inoculations. As shown in Fig. 6B, althoughgefitinib and MK-2206 individually showed inhibitoryeffects on tumor growth in comparison with vehicle,the brain tissues of the tumor-bearing mice treated with

the combination of gefitinib and MK-2206 containedeven smaller tumor masses and less glioma cells ascompared with those treated with gefitinib or MK-2206 alone, suggesting that the prolonged survival ofthe mice receiving combination therapy might be attri-buted to the less brain damage caused by glioma.

Discussion

The EGFR-initiated pathway is considered an attractivetherapeutic target in malignant glioma due to its frequentdysregulation in this disease. Thus, small-molecule inhi-bitors of EGFR tyrosine kinase such as gefitinib are beingevaluated as antiglioma therapies (28). Also, it has beennoticed that sensitivity of tumor cells to gefitinib is closelycorrelated to their dependence on Akt activation for

Tubulin

LC3 I

LC3 II

Time (h) 0 24 48 72 96

T98G

Bim

Gefitinib - + + + +

0

2

4

6

8

1 2 3 4 5

LC

3/T

ub

ulin

(r

ela

tive

to

co

ntr

ol)

0

0.5

1

1.5

1 2 3 4 5

Bim

/Tu

bu

lin

(re

lative

to

co

ntr

ol)

1 2 3 4 5

Tubulin

LC3 I

LC3 II

Time (h) 0 24 24 48 72 96

T98G

Bim

1 2 3 4 5 6

0

10

20

30

40

1 2 3 4 5 6

LC

3/T

ub

ulin

(r

ela

tive

to

co

ntr

ol)

0

1

2

3

4

1 2 3 4 5 6

Bim

/Tu

bu

lin

(re

lative

to

co

ntr

ol)

Gefitinib - + + + + +

MK-2206 - - + + + +

LN229

LC3 I

LC3 II

Tubulin

Time (h) 0 24 24 48 72 96

Bim

1 2 3 4 5 6

Gefitinib - + + + + +

MK-2206 - - + + + +

0

10

20

30

40

1 2 3 4 5 6

LC

3/T

ub

ulin

(r

ela

tive

to

co

ntr

ol)

0

1

2

3

4

1 2 3 4 5 6

Bim

/Tu

bu

lin

(re

lative

to

co

ntr

ol)

LN229

LC3 I

LC3 II

Tubulin

Time (h) 0 24 48 72 96

Bim

Gefitinib - + + + +

0

2

4

6

8

1 2 3 4 5

LC

3/T

ub

ulin

(re

lative

to

co

ntr

ol)

0

0.5

1

1.5

1 2 3 4 5B

im/T

ub

ulin

(re

lative

to

co

ntr

ol)

A

1 2 3 4 5

B

0

10

20

30

40

50

60

0 24 48 72 96

Ap

op

tosis

(%

)

Time (h)

LN229

0

10

20

30

40

50

60

0 24 48 72 96

Ap

op

tosis

(%

)

Time (h)

T98GC D T98GLN229

Gefi (µmol/L)

MK (0.5 µmol/L)

Gefi (µmol/L)

MK (0.5 µmol/L)- + +

0

5

10

15

20

25

30

35

0 1 5

Apopto

sis

(%

)

Nontargeting

Bim siRNA

- + +

0

5

10

15

20

25

30

35

0 1 5

Apopto

sis

(%

)

Nontargeting

Bim siRNA

Gefitinib +MK-2206Gefitinib +MK-2206

Figure 4. Prolonged cotreatment with gefitinib andMK-2206 promotes the switch from autophagy to apoptosis. A, LN229 and T98G cells were treated with 5mmol/L of gefitinib for different time periods, and the levels of LC3 and Bim were examined by Western blot. Tubulin was used as a loading control. B,LN229 andT98Gcellswere treatedwith 5mmol/L gefitinib for different timeperiods in the absence or presenceof 0.5mmol/L ofMK-2206, and the levels of LC3and Bim were examined by Western blot. Tubulin was used as a loading control. C, LN229 and T98G cells were treated with 5 mmol/L of gefitinib fordifferent periods of time in the absence or presence of 0.5 mmol/L of MK-2206. Apoptosis was examined by measuring Annexin V staining using flowcytometry. D, LN229 and T98G cells were transfected with a Bim siRNA or a nontargeting siRNA and then treated with gefitinib for 60 hours in the absenceor presence of 0.5 mmol/L of MK-2206. Apoptosis was examined by measuring Annexin V staining using flow cytometry.

Cheng et al.

Mol Cancer Ther; 11(1) January 2012 Molecular Cancer Therapeutics160

on August 9, 2019. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 4, 2011; DOI: 10.1158/1535-7163.MCT-11-0606

Page 8: MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes ...mct.aacrjournals.org/content/molcanther/11/1/154.full.pdf · Preclinical Development MK-2206, a Novel Allosteric Inhibitor

survival and proliferation (29), and this suggests thatinhibiting Akt may serve as an approach to improvingthe antitumor efficacy of EGFR inhibitors. In this study,we evaluated whether or not combining the novel allo-steric Akt inhibitor, MK-2206, with gefitinib couldenhance the antiglioma activity of this targeted therapy.Our results showed that gefitinib in combination withMK-2206 produced a synergistic effect against gliomacells (Fig. 2) and enhanced the antitumor activity in anorthotopic glioma mouse model (Fig. 6). It was reportedthat the responsiveness to EGFR inhibitors is associatedwith the coexpressions of EGFRvIII and functionalPTEN in glioma cells (30), and that downstream inhi-bition of the PI3K pathway can be combined with EGFRinhibitors to promote responsiveness in patients withPTEN-deficient tumors (31). Indeed, the synergisticeffect reported here was observed not only in the glioma

cells harboring wild-type PTEN (LN229 cells) but also inthe tumor cells with PTEN mutant (T98G cells) or PTENnull (U87MG cells). PTEN is a tumor suppressor thatacts as a critical negative regulator of PI3K–Akt–mTORsignaling axis. Loss of PTEN is one of the most commongenetic lesions and occurs in 60% to 80% of malignantgliomas. A previous study suggests that glioblastomamultiforme (GBM) patients who have high levels ofEGFR expression and low levels of phosphorylated Akthad better response to EGFR inhibitor erlotinib treat-ment than those with low levels of EGFR expression andhigh levels of phosphorylated Akt (32), but these resultshave not been confirmed in larger studies. Although thesynergistic effect reported here may have potentialimpacts in the treatment of GBM, we are aware thatthis synergism was observed only in 3 glioma cell linesand in a mouse glioma model; whether this synergistic

LN229

T98G

NT Beclin 1 siRNA

Beclin 1

Tubulin

NT

Beclin 1siRNA

Beclin 1

Tubulin

0

20

40

60

80

100

120

1 2.5 5 10

Ce

ll via

bili

ty (

%)

Nontargeting gefitinibNontargeting gefitinib+MK-2206Beclin 1 siRNA gefitinib+MK-2206

0

20

40

60

80

100

120

1 2.5 5 10

Cell

via

bili

ty (

%)

Gefitinib (µmol/L)

Gefitinib (µmol/L)

Nontargeting gefitinib

Nontargeting gefitinib+MK-2206

Beclin 1 siRNA gefitinib+MK-2206

A

B

LN229

0 1 5 1 5Gefitinib (µmol/L)

MK-2206

Tubulin

mTOR

S6K

p-mTOR

(Ser 2448)

p-S6 K

(Ser 371)

E

p-mTOR/Tubulin 1 0.8 0.6 0.5 0.2

p-S6K/Tubulin 1 0.9 0.4 0.3 0.2

Gefitinib (µmol/L) 0 1 5 1 5

MK-2206

Tubulin

p-mTOR

(Ser 2448)

mTOR

p-S6 K

(Ser 371)

S6K

F

p-S6K/Tubulin 1 1 0.9 0.4 0.1

p-mTOR/Tubulin 1 0.9 0.8 0.7 0.6

T98G

0

10

20

30

40

50

0 1 5

Apopto

sis

(%

)

Nontargeting

Beclin 1 siRNA

C

D

0

10

20

30

40

50

0 1 5

Apopto

sis

(%

)

Nontargeting

Beclin 1 siRNA

LN229

Gefi

+ + + MK (0.5 µmol/L)

Gefi

MK (0.5 µmol/L) + + +

T98G

Figure 5. Suppression of autophagy by silencing of Beclin 1 expression sensitizes glioma cells to the combinatorial treatment with gefitinib andMK-2206, andthe effects of gefitinib on activity of mTOR and S6K in the absence or presence of MK-2206. A and B, LN229 and T98G cells were transfected with a Beclin 1siRNA or a nontargeting siRNA and then treated with a series of concentrations of gefitinib for 72 hours in the absence or presence of 0.5 mmol/L ofMK-2206. Cell viability wasmeasured byMTT assay. C andD, LN229 and T98G cells were transfectedwith a Beclin 1 siRNA or a nontargeting siRNA and thentreated with gefitinib (1 and 5 mmol/L) for 60 hours in the absence or presence of 0.5 mmol/L of MK-2206. Apoptosis was examined by measuringAnnexin V staining using flow cytometry. E and F, LN229 and T98G cells were treated with various concentrations of gefitinib for 24 hours in the absenceor presence of 0.5 mmol/L of MK-2206. The levels of phospho-mTOR (Ser2448), mTOR, phospho-S6K (Ser371), and S6K were examined by Westernblot. Tubulin was used as a loading control.

Synergistic Antitumor Effect of MK-2206 and Gefitinib

www.aacrjournals.org Mol Cancer Ther; 11(1) January 2012 161

on August 9, 2019. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 4, 2011; DOI: 10.1158/1535-7163.MCT-11-0606

Page 9: MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes ...mct.aacrjournals.org/content/molcanther/11/1/154.full.pdf · Preclinical Development MK-2206, a Novel Allosteric Inhibitor

action is truly unaffected by PTEN status would needfurther preclinical and clinical investigation. In addi-tion, successful treatment of malignant glioma maydepend on tailoring cocktails of targeted drugs to indi-vidual patients. For instance, combinations of temozo-lomide with inhibitors of mTOR, PI3K, Akt, or withreticulum stress inhibitors have been proposed (33).

The synergismbetweenMK-2206 and gefitinib seems tobe associated with the modulation of autophagy andapoptosis in the cells subjected to the combinatory treat-ment of gefitinib and MK-2206 (Fig. 3 and Fig. 4). Apo-ptosis and autophagy are the 2 cellular processes knownto affect efficacies of numerous cancer therapeutic agents.To explore the mechanism underlying the synergismbetween gefitinib and MK-2206 in killing tumor cells, weexamined the activity of both apoptosis and autophagy inthe glioma cells subjected to combinatorial treatment ofthe 2 agents. We observed that gefitinib simultaneouslyinducedapoptosis andautophagy (Fig. 1CandD) and thatin the presence ofMK-2206, both of apoptosis and autoph-agywere increased in the tumor cells treatedwithgefitinibfor 48 hours. As autophagy may contribute to either cellsurvival or cell death, we then assessed the role of autop-

hagy in determining the efficacy of the treatments. Wefound that suppression of autophagy significantlyenhanced the cytocidal activity of the combinatorial treat-ments, suggesting that induction of autophagy is a com-pensatory response to therapeutic stress. Interestingly, 48hours after the cotreatments, autophagic activity began todecrease but apoptosiswas further activated (Fig. 4B), andthis shift did not occur in the glioma cells treated withgefitinib alone (Fig. 4A). Cotreatment of gefitinib withMK-2206 also caused a reduction of antiapoptotic protein,survivin, but led to an elevation of proapoptotic protein,Bim (Fig. 3B). It has been reported that Bim is required forapoptosis induced by certain targeted therapy in cancercells with oncogenic EGFR (34) and that the downregula-tion of survivin plays a pivotal role in the gefitinib-induced apoptosis (35). Thus, our results are consistentwith those reported observations. More importantly, ourstudy suggests that Akt inhibitors such as MK-2206 mayplay a therapeutically beneficial role in promoting func-tional switch from autophagy to apoptosis, and thisswitch may account for the synergistic effect of MK-2206 on cytocidal activity of gefitinib. Evidence of aswitch from autophagy to apoptosis was previously

CTRL+

10%DMSO

Gefitinib

MK-2206

MK-2206+

Gefitinib

BA20X 400X

17 days

1.0

0.8

0.6

0.4

0.2

0.0

0 10

Group

CTRL+10%DMSO Gef Gef+MK-2206 MK-2206

20 30

Su

rviv

al p

rob

abili

ty

Product-limit survival estimates

Days posttumor implantation

Figure 6. Effect of the combinatorial treatment with gefitinib and MK-2206 on tumor growth in an intracranial glioma model. The human glioma cells LN229(1 � 105 cells in 15 mL of DMEM medium) were injected into the brains of 6-week-old male BALB/c nude mice at 4 mm depth under anesthesia withchloralic hydras (4%, 2mL/kg, i.p.). Three days after tumor cell implantation, mice were randomly divided into 4 groups (15 mice per group). Treatments werebegunonday4, according to the following regimens: gefitinib, 80mg/kg, orally, 5 consecutive days;MK-2206, 100mg/kg, orally, 3 timesperweek for 2weeks;gefitinibþMK-2206, same as above; control, 10%DMSO in saline, orally. A, the Kaplan–Meier survival curves, n¼ 10; gefitinibþMK-2206 versus gefitinib,P ¼ 0.0155, log-rank statistic analysis; B, at day 17 after tumor cell implantation, the mice were euthanized, and the brains were fixed in 10% bufferedformalin, embedded in paraffin, and then stained with H&E. The images shown are representative of 5 mice from each group.

Cheng et al.

Mol Cancer Ther; 11(1) January 2012 Molecular Cancer Therapeutics162

on August 9, 2019. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 4, 2011; DOI: 10.1158/1535-7163.MCT-11-0606

Page 10: MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes ...mct.aacrjournals.org/content/molcanther/11/1/154.full.pdf · Preclinical Development MK-2206, a Novel Allosteric Inhibitor

reported for temozolomide (36), a chemotherapeuticdrug commonly used in treatment of GBM, and target-ing autophagy has been proposed as a new approach totreating apoptosis-resistant GBM (37). In addition, wefound that mTOR–S6K pathway, which locates down-stream of Akt and normally plays a role in suppressingautophagy (38), was involved in the induction of autop-hagy in the cells cotreated with gefitinib and MK-2206,as evidenced by a decrease in the levels of p-mTOR andp-S6K (Fig. 5E and F). These observations were consis-tent with our previous results showing the involvementof mTOR–S6K pathway in altering autophagic responsein tumor cells treated with MK-2206 (39). As mTOR andS6K are downstream effectors of Akt, and their inhibi-tion might cause deinhibition of a negative feedbackloop, how precisely this pathway affects the targetedtherapy might be worth further study.The need and importance for combining gefitinib with

othermolecular targeted agents in treating glioblastoma isalso underscored by a recent phase II study showing thatgefitinib exerts little effects on the activity of the down-stream pathways, despite its inhibitory effect on EGFRphosphorylation (10). Therefore, multitarget based newtherapeutic strategies may be necessary for better sup-pression of the EGFR-initiated oncogenic pathways. It hasnow become increasingly appreciated that manipulatingautophagy and apoptosis may achieve better therapeuticoutcomes in cancer treatment; hence, the molecularmechanisms and pathways involved in the regulation ofthe cross-talk between autophagy and apoptosis, and theapproaches to exploiting these cellular process for kill-

ing cancer cells, are undergoing extensive exploration.The results reported here show that the novel allostericAkt inhibitor, MK-2206, can synergize with gefitinib ininhibiting malignant glioma by promoting a switchfrom autophagy to apoptosis, suggesting that this dualtargeted therapy may make these drugs more usefuland valuable in the treatment of malignant glioma orother types of cancer. Considering that the majority ofhuman malignant gliomas harbor mutations that resultin activation of PI3K/Akt pathway, and that aberrantEGFR signaling is a primary contributor to the patho-genesis of gliomas, our results provide a rationale andbasis for further clinical investigation of the combina-torial use of MK-2206 and gefitinib in the treatment ofthis disease, and thus may have potential impact incancer therapy.

Disclosure of Potential Conflicts of Interest

J-M. Yang received a commercial research grant.

Grant Support

The work was supported by grants from the US Public Health ServiceR01CA135038 and from Merck & Co. Inc.

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received August 5, 2011; revised September 29, 2011; acceptedNovember 1, 2011; published OnlineFirst November 4, 2011.

References1. Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell

2000;103:211–25.2. Ritter CA, Arteaga CL. The epidermal growth factor receptor-tyrosine

kinase: a promising therapeutic target in solid tumors. Semin Oncol2003;30:3–11.

3. Ciardiello F, TortoraG. EGFR antagonists in cancer treatment. NEngl JMed 2008;358:1160–74.

4. Sathornsumetee S, Desjardins A, Vredenburgh JJ, McLendon RE,Marcello J,HerndonJE, et al. Phase II trial of bevacizumabanderlotinibin patients with recurrent malignant glioma. Neuro Oncol 2010;12:1300–10.

5. Thiessen B, Stewart C, Tsao M, Kamel-Reid S, Schaiquevich P,MasonW, et al. A phase I/II trial of GW572016 (lapatinib) in recurrentglioblastoma multiforme: clinical outcomes, pharmacokinetics andmolecular correlation. Cancer Chemother Pharmacol 2010;65:353–61.

6. Giannopoulou E, Dimitropoulos K, Argyriou AA, Koutras AK, Dimitra-kopoulos F, Kalofonos HP. An in vitro study, evaluating the effect ofsunitinib and/or lapatinib on two glioma cell lines. Invest New Drugs2010;28:554–60.

7. Guillamo JS, de Bouard S, Valable S, Marteau L, Leuraud P, Marie Y,et al. Molecular mechanisms underlying effects of epidermal growthfactor receptor inhibition on invasion, proliferation, and angiogenesisin experimental glioma. Clin Cancer Res 2009;15:3697–704.

8. Griffero F, Daga A, Marubbi D, Capra MC, Melotti A, Pattarozzi A, et al.Different response of human glioma tumor-initiating cells to epidermalgrowth factor receptor kinase inhibitors. J Biol Chem 2009;284:7138–48.

9. Thaker NG, Pollack IF. Molecularly targeted therapies for malignantglioma: rationale for combinatorial strategies. Expert Rev Neurother2009;9:1815–36.

10. Hegi ME, Diserens AC, Bady P, Kamoshima Y, Kouwenhoven MC,Delorenzi M, et al. Pathway analysis of glioblastoma tissue afterpreoperative treatment with the EGFR tyrosine kinase inhibitor gefiti-nib–a phase II trial. Mol Cancer Ther 2011;10:1102–12.

11. ManningBD, Cantley LC. AKT/PKB signaling: navigating downstream.Cell 2007;129:1261–74.

12. Riemenschneider MJ, Betensky RA, Pasedag SM, Louis DN. AKTactivation in human glioblastomas enhances proliferation via TSC2and S6 kinase signaling. Cancer Res 2006;66:5618–23.

13. Degtyarev M, De Maziere A, Orr C, Lin J, Lee BB, Tien JY, et al. Aktinhibition promotes autophagy and sensitizes PTEN-null tumors tolysosomotropic agents. J Cell Biol 2008;183:101–16.

14. Cheng Y, Li H, Ren X, Niu T, Hait WN, Yang J. Cytoprotective effect ofthe elongation factor-2 kinase-mediated autophagy in breast cancercells subjected to growth factor inhibition. PLoS One 2010;5:e9715.

15. HanW, PanH, Chen Y, Sun J,Wang Y, Li J, et al. EGFR tyrosine kinaseinhibitors activate autophagy as a cytoprotective response in humanlung cancer cells. PLoS One 2011;6:e18691.

16. Eskelinen EL. Doctor Jekyll and Mister Hyde: autophagy can promoteboth cell survival and cell death. Cell Death Differ 2005;12:1468–72.

17. Balasis ME, Forinash KD, Chen YA, FulpWJ, Coppola D, Hamilton AD,et al. Combination of farnesyltransferase and Akt inhibitors is syner-gistic in breast cancer cells and causes significant breast tumorregression in ErbB2 transgenic mice. Clin Cancer Res 2011;17:2852–62.

Synergistic Antitumor Effect of MK-2206 and Gefitinib

www.aacrjournals.org Mol Cancer Ther; 11(1) January 2012 163

on August 9, 2019. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 4, 2011; DOI: 10.1158/1535-7163.MCT-11-0606

Page 11: MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes ...mct.aacrjournals.org/content/molcanther/11/1/154.full.pdf · Preclinical Development MK-2206, a Novel Allosteric Inhibitor

18. Meng J, Dai B, Fang B, Bekele BN, Bornmann WG, Sun D, et al.Combination treatment with MEK and AKT inhibitors is more effectivethan eachdrug alone in humannon-small cell lung cancer in vitro and invivo. PLoS One 2010;5:e14124.

19. Hirai H, Sootome H, Nakatsuru Y, Miyama K, Taguchi S, Tsujioka K,et al. MK-2206, an allosteric Akt inhibitor, enhances antitumor efficacyby standard chemotherapeutic agents or molecular targeted drugs invitro and in vivo. Mol Cancer Ther 2010;9:1956–67.

20. Premkumar DR, Arnold B, Pollack IF. Cooperative inhibitory effect ofZD1839 (Iressa) in combination with 17-AAG on glioma cell growth.Mol Carcinog 2006;45:288–301.

21. Hofer S, Frei K. Gefitinib concentrations in human glioblastoma tissue.J Neurooncol 2007;82:175–6.

22. Hofer S, Frei K, Rutz HP. Gefitinib accumulation in glioblastoma tissue.Cancer Biol Ther 2006;5:483–4.

23. McKillopD, Partridge EA, Kemp JV, SpenceMP, Kendrew J, Barnett S,et al. Tumor penetration of gefitinib (Iressa), an epidermal growth factorreceptor tyrosine kinase inhibitor. Mol Cancer Ther 2005;4:641–9.

24. Fury MG, Solit DB, Su YB, Rosen N, Sirotnak FM, Smith RP, et al. Aphase I trial of intermittent high-dose gefitinib and fixed-dose doc-etaxel in patients with advanced solid tumors. Cancer ChemotherPharmacol 2007;59:467–75.

25. Wang S, Guo P, Wang X, Zhou Q, Gallo JM. Preclinical pharmacoki-netic/pharmacodynamic models of gefitinib and the design of equiv-alent dosing regimens in EGFR wild-type and mutant tumor models.Mol Cancer Ther 2008;7:407–17.

26. Wang S, Zhou Q, Gallo JM. Demonstration of the equivalent pharma-cokinetic/pharmacodynamic dosing strategy in a multiple-dose studyof gefitinib. Mol Cancer Ther 2009;8:1438–47.

27. Chou TC. Theoretical basis, experimental design, and computerizedsimulation of synergism and antagonism in drug combination studies.Pharmacol Rev 2006;58:621–81.

28. Halatsch ME, Schmidt U, Behnke-Mursch J, Unterberg A, Wirtz CR.Epidermal growth factor receptor inhibition for the treatment of glio-blastomamultiforme and other malignant brain tumours. Cancer TreatRev 2006;32:74–89.

29. OnoM, Hirata A, Kometani T, Miyagawa M, Ueda S, Kinoshita H, et al.Sensitivity to gefitinib (Iressa, ZD1839) in non-small cell lung cancer

cell lines correlates with dependence on the epidermal growth factor(EGF) receptor/extracellular signal-regulated kinase 1/2 and EGFreceptor/Akt pathway for proliferation. Mol Cancer Ther 2004;3:465–72.

30. Mellinghoff IK, Wang MY, Vivanco I, Haas-Kogan DA, Zhu S, Dia EQ,et al. Molecular determinants of the response of glioblastomas toEGFR kinase inhibitors. N Engl J Med 2005;353:2012–24.

31. Gonzalez J, de Groot J. Combination therapy for malignant gliomabased on PTEN status. Expert Rev Anticancer Ther 2008;8:1767–79.

32. Haas-Kogan DA, PradosMD, Tihan T, Eberhard DA, Jelluma N, ArvoldND, et al. Epidermal growth factor receptor, protein kinase B/Akt, andglioma response to erlotinib. J Natl Cancer Inst 2005;97:880–7.

33. Lefranc F, Brotchi J, Kiss R. Possible future issues in the treatment ofglioblastomas: special emphasis on cell migration and the resistanceof migrating glioblastoma cells to apoptosis. J Clin Oncol 2005;23:2411–22.

34. Gong Y, Somwar R, Politi K, Balak M, Chmielecki J, Jiang X, et al.Induction of BIM is essential for apoptosis triggered by EGFR kinaseinhibitors in mutant EGFR-dependent lung adenocarcinomas. PLoSMed 2007;4:e294.

35. Okamoto K, Okamoto I, Okamoto W, Tanaka K, Takezawa K, KuwataK, et al. Role of survivin in EGFR inhibitor-induced apoptosis in non-small cell lung cancers positive for EGFR mutations. Cancer Res2010;70:10402–10.

36. Roos WP, Batista LF, Naumann SC, Wick W, Weller M, Menck CF,et al. Apoptosis in malignant glioma cells triggered by the temo-zolomide-induced DNA lesion O6-methylguanine. Oncogene 2007;26:186–97.

37. Lefranc F, Facchini V, Kiss R. Proautophagic drugs: a novel means tocombat apoptosis-resistant cancers, with a special emphasis onglioblastomas. Oncologist 2007;12:1395–403.

38. Pan T, Rawal P, Wu Y, Xie W, Jankovic J, Le W. Rapamycin protectsagainst rotenone-induced apoptosis through autophagy induction.Neuroscience 2009;164:541–51.

39. Cheng Y, Ren X, Zhang Y, Patel R, Sharma A,WuH, et al. eEF-2 kinasedictates cross-talk between autophagy and apoptosis induced by AktInhibition, thereby modulating cytotoxicity of novel Akt inhibitor MK-2206. Cancer Res 2011;71:2654–63.

Mol Cancer Ther; 11(1) January 2012 Molecular Cancer Therapeutics164

Cheng et al.

on August 9, 2019. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 4, 2011; DOI: 10.1158/1535-7163.MCT-11-0606

Page 12: MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes ...mct.aacrjournals.org/content/molcanther/11/1/154.full.pdf · Preclinical Development MK-2206, a Novel Allosteric Inhibitor

2012;11:154-164. Published OnlineFirst November 4, 2011.Mol Cancer Ther   Yan Cheng, Yi Zhang, Li Zhang, et al.   and ApoptosisGefitinib against Malignant Glioma via Modulating Both Autophagy MK-2206, a Novel Allosteric Inhibitor of Akt, Synergizes with

  Updated version

  10.1158/1535-7163.MCT-11-0606doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2011/11/04/1535-7163.MCT-11-0606.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/11/1/154.full#ref-list-1

This article cites 39 articles, 16 of which you can access for free at:

  Citing articles

  http://mct.aacrjournals.org/content/11/1/154.full#related-urls

This article has been cited by 10 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]

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

  Permissions

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

.http://mct.aacrjournals.org/content/11/1/154To request permission to re-use all or part of this article, use this link

on August 9, 2019. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 4, 2011; DOI: 10.1158/1535-7163.MCT-11-0606