Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by … · Cancer Biology and Signal...

11
Cancer Biology and Signal Transduction Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by Switching Protective Autophagy to Autophagic Cell Death in Hepatocellular Carcinoma Bo Zhai 1 , Fengli Hu 2 , Xian Jiang 1 , Jun Xu 3 , Dali Zhao 1 , Bing Liu 1 , Shangha Pan 1 , Xuesong Dong 1 , Gang Tan 1 , Zheng Wei 1 , Haiquan Qiao 1 , Hongchi Jiang 1 , and Xueying Sun 1 Abstract Sorafenib is the standard first-line systemic drug for advanced hepatocellular carcinoma (HCC), but the acquired resistance to sorafenib results in limited benefits. Activation of Akt is thought to be responsible for mediating the acquired resistance to sorafenib. The present study aims to examine the underlying mechanism and seek potential strategies to reverse this resistance. Two sorafenib-resistant HCC cell lines, which had been established from human HCC HepG2 and Huh7 cells, were refractory to sorafenib-induced growth inhibition and apoptosis in vitro and in vivo. Sustained exposure to sorafenib activated Akt via the feedback loop of mTOR but independent of protein phosphatase 2A in HCC cells. Autophagy participated in the resistance to sorafenib as inhibition of autophagy reduced the sensitivity of sorafenib-resistant HCC cells to sorafenib, whereas activation of autophagy by rapamycin had the opposite effect. However, rapamycin did not show a synergistic effect with sorafenib to inhibit cell proliferation, while it also activated Akt via a feedback mechanism in sorafenib-resistant HCC cells. Inhibition of Akt reversed the acquired resistance to sorafenib by switching autophagy from a cytoprotective role to a death-promoting mechanism in the sorafenib-resistant HCC cells. Akt inhibition by GDC0068 synergized with sorafenib to suppress the growth of sorafenib-resistant HCC tumors that possessed the sorafenib-resistant feature in vivo. The results have provided evidence for clinical investigation of GDC0068, a novel ATP-competitive pan-Akt inhibitor, as the second-line treatment after the failure of sorafenib-medicated molecular targeted therapy for advanced HCC. Mol Cancer Ther; 13(6); 1589–98. Ó2014 AACR. Introduction Hepatocellular carcinoma (HCC), the second most fre- quent cause of cancer death in men worldwide (1), is notoriously resistant to systemic chemotherapy (2). After searching for effective agents to combat HCC for decades, sorafenib has opened a window of hope and presents as the standard first-line systemic drug for advanced HCC (3). However, the promising treatment has demonstrated limited survival benefits with very low response rates (3, 4), and some patients with HCC initially respond to sorafenib but eventually the disease progresses (4), indi- cating that the resistance to sorafenib is common in HCC. As a multitargeted kinase inhibitor, sorafenib targets the Raf/mitogen-activated protein kinase (MAPK)/extra- cellular signaling-regulated kinase (ERK) signaling path- way, and inhibits a number of tyrosine kinase receptors, including VEGF receptor, platelet-derived growth factor receptor, and c-Kit (3). The phosphoinositide 3-kinase (PI3K)/Akt pathway regulates a large number of mole- cules involved in all aspects of cancer progression (5), and is involved in the development and progression of HCC (6). Sorafenib activates Akt and upregulates its down- stream factors such as ribosomal protein S6 kinase (S6K) and eukaryotic translation initiation factor 4E-binding protein 1 (4EBP1) in HCC cells (7, 8). Sorafenib-resistant HCC cells have increased expression of phosphorylated Akt (p-Akt; ref. 9). There exist cross-talks between the PI3K/Akt and MAPK/ERK pathways (10), indicating that the latent compensatory mechanism of the PI3K/Akt pathway may contribute to sorafenib resistance in HCC. However, the mechanisms that underlie the role of Akt activation in this resistance remain unclear. Induction of cell death is the final goal for developing anticancer agents. Autophagy was initially referred as a self-digestion process by which cytoplasmic contents are sequestered in autophagosomes and delivered to lyso- somes for degradation (11), but now autophagy is also Authors' Afliations: 1 The Hepatosplenic Surgery Center, Department of General Surgery, the First Afliated Hospital of Harbin Medical University; and Departments of 2 Gastroenterology and 3 General Surgery, the Fourth Afliated Hospital of Harbin Medical University, Harbin, China Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). Corresponding Author: Xueying Sun, The Hepatosplenic Surgery Center, Department of General Surgery, the First Afliated Hospital of Harbin Medical University, Harbin 150001, China. Phone: 86-451-53643628; Fax: 86-451-53643628; E-mail: [email protected] doi: 10.1158/1535-7163.MCT-13-1043 Ó2014 American Association for Cancer Research. Molecular Cancer Therapeutics www.aacrjournals.org 1589 on April 5, 2020. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst April 4, 2014; DOI: 10.1158/1535-7163.MCT-13-1043

Transcript of Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by … · Cancer Biology and Signal...

Page 1: Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by … · Cancer Biology and Signal Transduction Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by

Cancer Biology and Signal Transduction

Inhibition of Akt Reverses the Acquired Resistance toSorafenib by Switching Protective Autophagy to AutophagicCell Death in Hepatocellular Carcinoma

Bo Zhai1, Fengli Hu2, Xian Jiang1, Jun Xu3, Dali Zhao1, Bing Liu1, Shangha Pan1, Xuesong Dong1,Gang Tan1, Zheng Wei1, Haiquan Qiao1, Hongchi Jiang1, and Xueying Sun1

AbstractSorafenib is the standard first-line systemic drug for advanced hepatocellular carcinoma (HCC), but the

acquired resistance to sorafenib results in limited benefits. Activation of Akt is thought to be responsible for

mediating the acquired resistance to sorafenib. The present study aims to examine the underlying mechanism

and seek potential strategies to reverse this resistance. Two sorafenib-resistant HCC cell lines, which had been

established from humanHCCHepG2 andHuh7 cells, were refractory to sorafenib-induced growth inhibition

and apoptosis in vitro and in vivo. Sustained exposure to sorafenib activatedAkt via the feedback loop ofmTOR

but independent of protein phosphatase 2A inHCC cells. Autophagyparticipated in the resistance to sorafenib

as inhibition of autophagy reduced the sensitivity of sorafenib-resistant HCC cells to sorafenib, whereas

activation of autophagy by rapamycin had the opposite effect. However, rapamycin did not show a synergistic

effect with sorafenib to inhibit cell proliferation, while it also activated Akt via a feedback mechanism in

sorafenib-resistant HCC cells. Inhibition of Akt reversed the acquired resistance to sorafenib by switching

autophagy from a cytoprotective role to a death-promoting mechanism in the sorafenib-resistant HCC cells.

Akt inhibition by GDC0068 synergized with sorafenib to suppress the growth of sorafenib-resistant HCC

tumors that possessed the sorafenib-resistant feature in vivo. The results have provided evidence for clinical

investigation of GDC0068, a novel ATP-competitive pan-Akt inhibitor, as the second-line treatment after the

failure of sorafenib-medicated molecular targeted therapy for advancedHCC.Mol Cancer Ther; 13(6); 1589–98.

�2014 AACR.

IntroductionHepatocellular carcinoma (HCC), the second most fre-

quent cause of cancer death in men worldwide (1), isnotoriously resistant to systemic chemotherapy (2). Aftersearching for effective agents to combat HCC for decades,sorafenib has opened a window of hope and presents asthe standard first-line systemic drug for advanced HCC(3). However, the promising treatment has demonstratedlimited survival benefits with very low response rates (3,4), and some patients with HCC initially respond tosorafenib but eventually the disease progresses (4), indi-cating that the resistance to sorafenib is common in HCC.

As a multitargeted kinase inhibitor, sorafenib targetstheRaf/mitogen-activated protein kinase (MAPK)/extra-cellular signaling-regulated kinase (ERK) signaling path-way, and inhibits a number of tyrosine kinase receptors,including VEGF receptor, platelet-derived growth factorreceptor, and c-Kit (3). The phosphoinositide 3-kinase(PI3K)/Akt pathway regulates a large number of mole-cules involved in all aspects of cancer progression (5), andis involved in the development and progression of HCC(6). Sorafenib activates Akt and upregulates its down-stream factors such as ribosomal protein S6 kinase (S6K)and eukaryotic translation initiation factor 4E-bindingprotein 1 (4EBP1) in HCC cells (7, 8). Sorafenib-resistantHCC cells have increased expression of phosphorylatedAkt (p-Akt; ref. 9). There exist cross-talks between thePI3K/Akt and MAPK/ERK pathways (10), indicatingthat the latent compensatory mechanism of the PI3K/Aktpathway may contribute to sorafenib resistance in HCC.However, the mechanisms that underlie the role of Aktactivation in this resistance remain unclear.

Induction of cell death is the final goal for developinganticancer agents. Autophagy was initially referred as aself-digestion process by which cytoplasmic contents aresequestered in autophagosomes and delivered to lyso-somes for degradation (11), but now autophagy is also

Authors' Affiliations: 1The Hepatosplenic Surgery Center, Department ofGeneral Surgery, the First Affiliated Hospital of Harbin Medical University;and Departments of 2Gastroenterology and 3General Surgery, the FourthAffiliated Hospital of Harbin Medical University, Harbin, China

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

Corresponding Author: Xueying Sun, The Hepatosplenic Surgery Center,Department of General Surgery, the First Affiliated Hospital of HarbinMedical University, Harbin 150001, China. Phone: 86-451-53643628; Fax:86-451-53643628; E-mail: [email protected]

doi: 10.1158/1535-7163.MCT-13-1043

�2014 American Association for Cancer Research.

MolecularCancer

Therapeutics

www.aacrjournals.org 1589

on April 5, 2020. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst April 4, 2014; DOI: 10.1158/1535-7163.MCT-13-1043

Page 2: Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by … · Cancer Biology and Signal Transduction Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by

considered to be type II programmed cell death (PCD;ref. 12).Apoptosis, the type I PCD, is undoubtedly a tumorsuppressing pathway, whereas autophagy is a double-edged sword depending on the cellular context andnature of the stimuli (13). The autophagic pathway cross-talks with apoptotic and other pathways, and the mutiplemolecular nodes in such pathways offer opportunities fortherapeutic intervention (13). Autophagy-related protein(ATG) 6 (also known as Beclin 1) binds to other compo-nents to form a core complex to allow autophagosomenucleation (14), and interacts with apoptotic molecules(13). The PI3K/Akt pathway inhibits autophagy by acti-vating mTOR (15), which exerts an inhibitory effect onautophagy by dysregulating Beclin 1 and ATG8 (micro-tubule-associated protein 1 light chain 3, LC3; ref. 16).These data indicate that activation of Akt may be respon-sible for mediating the acquired resistance to sorafenib byregulating the autophgic pathway.

Sorafenib promotes autophagic death ofHCC cells (17),but contrary results have also been reported (18). Activa-tion of autophagy is involved in the resistance to cisplatin(19), whereas inhibition of autophagy enhances the effectof sorafenib (20, 21) in HCC cells. However, the role ofautophagy and its cross-talk with other pathways inmediating resistance to sorafenib of HCC have not yetbeen reported. Here, we demonstrate, for the first time,that autophagy switches from a cytoprotective function toa death-promoting role and drives the acquired resistanceto sorafenib, while inhibiting Akt reverses the acquiredresistance to sorafenib by activating the autophagic path-way in sorafenib-resistant HCC cells.

Materials and MethodsCell culture, antibodies, and reagents

Human HCC HepG2 cells were obtained from theAmerican Type Culture Collection, and Huh7 cells fromChineseAcademyof SciencesCell Bank (Shanghai,China)in 2009. Cells were immediately expanded, and multiplealiquots were cryopreserved and used within 3 monthsafter resuscitation. No further authenticationwas done bythe authors. Cells were cultured at 37�C in Dulbecco’sModified Eagle Medium (Gibco BRL) supplementedwith 10% FBS. The antibodies (Abs) against Akt, p-Akt(Ser473), ERK, p-ERK (Thr202/Thyr204), glycogensynthase kinase (GSK)-3b, phosphorylated GSK3b (p-GSK3b; Ser9), mTOR, S6K, phosphorylated S6K (p-S6K;Thr389), 4EBP1, phosphorylated 4EBP1 (p-4EBP1; Ser65),PARP, LC3, Beclin1, ATG5, class III PI3K vascular proteinsorting 34 (Vps34), UV radiation resistance-associatedgene (UVRAG), and p62 were purchased from Cell Sig-nalingTechnology. TheAbs against Bad, caspase-3 and -9,p27, cyclin D1, Bcl-2, and b-actin were from Santa CruzBiotechnology. The anti–Ki-67 Ab was from Abcam. Sor-afenib and perifosine were from Jinan Trio PharmatechCo., Ltd. GDC0068 was from ShangHai BiochempartnerCo., Ltd. Rapamycin (RAP), 3-methyladenine (3-MA),bafilomycin A1 (Baf-A1), okadaic acid (OA), and 1,9-

dideoxyforskolin (Forskolin) were from Sigma-Aldrich.Sorafenib, GDC0068, rapamycin, and Baf-A1 were dis-solved in dimethyl sulfoxide to make a stock solution of100 mmol/L, 100 mmol/L, 1 mmol/L, and 1 mmol/Lfor in vitro assays, respectively. Perifosine was dissolvedin PBS to make a stock solution of 30 mmol/L. 3-MAwasdissolved in PBS at a concentration of 200 mmol/L byheating to 60 to 70�C immediately before use. For animalexperiments, sorafenib was suspended in the vehiclesolution containing Cremophor (Sigma-Aldrich), 95%ethanol and water in a ratio of 1:1:6 (22), and GDC0068was dissolved in 0.5% methylcellulose/0.2% Tween-80(23). The PI (propidium iodide)/Annexin V-FITC apo-ptosis detection kit was from BD Biosciences. The CellCounting Kit-8 (CCK-8) kit was from Dojindo Labora-tories. Terminal deoxynucleotidyl transferase–mediat-ed dUTP nick end labeling agent (TUNEL) was fromRoche.

Establishment of sorafenib-resistant cellsThe IC50 of HCC cells to sorafenib was initially deter-

mined by incubating cells with different concentrations ofsorafenib in 96-well plates, and cell viability was mea-sured 3 days later as described below. The cells werecultured in 6-well plates at 1 � 104 cells/well and incu-bated with sorafenib at a concentration just below theirrespective IC50. The concentration of sorafenibwas slowlyincreased by 0.25 mmol/L per week. After 6 to 7 months,two sorafenib-resistant cell lines were obtained, termedHepG2-SR and Huh7-SR, respectively, and were contin-uously maintained by culturing them in the presence ofsorafenib.

Analysis of autophagy by GFP-LC3 redistributionThe GFP tandem fluorescent-tagged LC3 expression

plasmid was transfected into cells using Lipofectamine2000 (24). LC3 redistribution was detected under a fluo-rescent microscope.

PP2A phosphatase activity assayThe activity of PP2A phosphatase in cell lysates was

measured by using a PP2A immunoprecipitation phos-phatase assay kit (Millipore).

Animal experimentsSix- to 8-week male nude BALB/c-nu/nu mice were

obtained from the Animal Research Center, The FirstAffiliatedHospital ofHarbinMedical University (Harbin,China). The study protocol had been approved by theAnimal Ethic Committee of Harbin Medical University.The animal experiment schedule was designed on thebasis of the preliminary experiments (Fig. 1). Briefly,Huh7-SR cells (5 � 106) were subcutaneously inoculatedinto the back of mice, which received daily oral admin-istration of 15 mg/kg sorafenib. The use of lower dose ofsorafenibwas tomaintain the sorafenib-resistant ability ofHuh7-SR cells, which were kept in the presence of sor-afenib in culture. Fifteen days later, another group ofmice

Zhai et al.

Mol Cancer Ther; 13(6) June 2014 Molecular Cancer Therapeutics1590

on April 5, 2020. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst April 4, 2014; DOI: 10.1158/1535-7163.MCT-13-1043

Page 3: Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by … · Cancer Biology and Signal Transduction Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by

received subcutaneous injections of Huh7 cells (5 � 106).Thirty days later, three Huh7 and three Huh7-SR tumorswere harvested. The remaining mice were assigned todifferent treatments. Sorafenibwas orally administered ata dose of 30mg/kg daily, andGDC0068 orally at a dose of25 mg/kg twice per week. The tumor volumes weremeasured every 3 days. The tumors were harvested atthe end of experiments.

Cell viability analysis, in vitro apoptosis assay,visualization of apoptotic cells by laser scanningconfocal microscopy, assessment of Ki-67proliferation index, in situdetection of apoptotic cell,and immunoblottingAll these methods have been described in details pre-

viously (24–26).

Transfection of Akt siRNAAdouble-strand siRNA targeting humanAkt (50- GUG-

GUCAUGUACGAGAUGATT-30 and 50-UCAUCUC-GUACAUGACCACTT-30) encoding nucleotides 1006–1025 of Akt1 (GenBank: NM_001014431.1), nucleotides1210–1229 of Akt2 (GenBank: XM_005336494.1), andnucleotides 955–974 of Akt3 (GenBank: XM_004691046.1)with two introduced thymidine residues at the 30 endwasproduced by GenePharma Co., Ltd. The nonspecificscrambled siRNA (50-UUCUCCGAACGUGUCACGU-30

and 50-ACGUGACACGUUCGGAGAA-30) served as acontrol. Cells were grown to 60% to 70% confluence, andincubated with siRNAs at a final concentration of 0.1mmol/L by using Lipofectamine 2000 (Invitrogen) in aserum-freemedium for 24 hours and then subjected to theassays.

Electronic microscopyCells or tissues were fixed in 2.5% glutaraldehyde

solution for 1 hour, washed twice with PBS, followed byfurther fixation with 1% Osmic acid for 1 hour, dehy-drated with a graded series of ethanol, embedded, andsectioned. Sections were stained with Uranium Acetateand Lead Citrate, and observed under a transmissionelectronic microscope (JEN-M1220, Toshiba; ref. 24).

Statistical analysisThe data were expressed as mean values � SD. Com-

parisons were made using one-way ANOVA followed byDunnet t test. P < 0.05 was considered significant.

ResultsSorafenib-resistant HCC cells are refractory tosorafenib-induced growth inhibition and apoptosis

After incubation with 10 mmol/L of sorafenib for 48hours, the viability of HepG2-SR and Huh7-SR cells were78.8% and 77.2%, respectively, which were significantlyhigher than that of HepG2 and Huh7 cells (18.4% and31.9%, respectively; Fig. 2A). Evenwhen the concentrationof sorafenib reached 20mmol/L, the viability ofHepG2-SRand Huh7-SR still remained at 38.5% and 46.2%, respec-tively, whereas HepG2 and Huh7 cells were almostcompletely dead (Fig. 2A). The in vitro findings weresupported by the in vivo data (Supplementary Fig. S1).Apoptotic rates of HepG2 cells were 4.1- and 3.9-foldhigher than that of HepG2-SR cells, after exposure to 5and 10 mmol/L of sorafenib, respectively (Fig. 2B and C).Apoptotic rates ofHuh7 cellswere 2.8- and 4.0-fold higherthan that of Huh7-SR cells, after exposure to 5 and

Figure 1. Animal experimental schedule. Two groups of mice (n ¼ 5 and n ¼ 35) underwent subcutaneous injections of Huh7 or Huh7-SR cells (5 � 106),respectively, and received daily oral sorafenib at 15 mg/kg. Fifteen days later, another group of mice (n ¼ 15) underwent subcutaneous injection ofHuh7 cells (5� 106). At day 30, none of the mice (0/5) receiving inoculation of Huh7 cells and sorafenib administration had palpable tumors, whereas 31mice(31/35) receiving inoculation of Huh7-SR cells and sorafenib administration, and 14 mice (14/15) receiving inoculation of Huh7 cells, had tumors ofapproximately 100 mm3 in volume. The mice without tumors were killed. Three Huh7 and 3 Huh7-SR tumors were harvested at day 30. The remaining micewere assigned to different treatments. Sorafenibwas orally administered at a dose of 30mg/kg daily, andGDC-0068 orally administered at a dose of 25mg/kgtwice per week. The frames with solid lines indicate start of the experiment, and frames with dotted lines indicate end of the experiment.

Akt/Autophagy for Sorafenib Resistance in HCC

www.aacrjournals.org Mol Cancer Ther; 13(6) June 2014 1591

on April 5, 2020. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst April 4, 2014; DOI: 10.1158/1535-7163.MCT-13-1043

Page 4: Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by … · Cancer Biology and Signal Transduction Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by

10 mmol/L of sorafenib, respectively (Fig. 2B and C). Theapoptotic resultswere supported by the expression of twokey apoptotic proteins, caspase-3 and PARP (Fig. 2D andE), indicating that sorafenib-resistant cells are refractoryto sorafenib-induced apoptosis through caspase-depen-dent and -independent ways.

Autophagy participates in the mechanisms for theresistance to sorafenib

After incubation for 48 hours in the presence andabsence of sorafenib (10 mmol/L), sorafenib-resistantcells expressed higher levels of p-Akt and differentlevels of its downstream factors, including p-GSK3b,mTOR, p-S6K and p-4EBP1, Bad, p27, and cyclin D1,

compared with their respective parental cells (Supple-mentary Fig. S2A and Supplementary Table S1). Theresults indicate that sustained exposure to sorafenibactivates the Akt pathway. In addition, sorafenib-induced phosphorylation of Akt was independent ofprotein phosphatase inactivation (Supplementary Fig.S2B and S2C). Given that mTOR is a gatekeeper ofautophagy by dysregulating ATGs (16), we hypothe-sized that autophagy may participate in the acquiredresistance to sorafenib. Accordingly, sorafenib-resistantcells expressed lower levels of LC3-II, ATG5, Vps34 andBeclin 1, and expressed higher levels of P62 and Bcl-2,than their respective parental cells in either the presenceor absence of sorafenib (Fig. 3A). Sorafenib upregulated

Figure 2. Sorafenib-resistant HCCcells are refractory to sorafenib-induced growth inhibition andapoptosis. A, the sorafenib-resistant cells HepG2-SR andHuh7-SR and parental HepG2 andHuh7 cells were incubated withincreasing concentrations ofsorafenib for 48 hours. Cell viability(%) was compared with thecorresponding untreated cells.B–E, the above cells wereincubated with 0, 5 or 10 mmol/L ofsorafenib for 48 hours. The cellswere analyzed cytometrically todetect apoptosis (B) and therates of apoptosis weredetermined (C), and cell lysateswere immunoblotted (D). E, banddensities were normalized tob-actin. Data represent threeindependent experiments.��, P < 0.001; †, P < 0.05; z, P <0.001 versus the correspondinguntreated cells.

Zhai et al.

Mol Cancer Ther; 13(6) June 2014 Molecular Cancer Therapeutics1592

on April 5, 2020. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst April 4, 2014; DOI: 10.1158/1535-7163.MCT-13-1043

Page 5: Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by … · Cancer Biology and Signal Transduction Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by

the expression of LC3-II, ATG5, Beclin 1, and Vps34,downregulated the expression of P62, and had littleeffects on UVRAG and Bcl-2, in either parental orsorafenib-resistant cells (Fig. 3A). The main in vitrofindings were further supported by the in vivo results(Supplementary Fig. S3).We next showed that inhibition of autophagy by

3-MA and Baf-A1 protected HepG2-SR and Huh7-SRcells against sorafenib-induced reduction in cell viabil-

ity (Fig. 3B and C). In contrast, rapamycin, an inhibitorof mTOR and an inducer of autophagy (27), enhancedsorafenib-induced growth inhibition in a dose-depen-dent manner (Fig. 3D). However, the synergistic effectsof rapamycin and sorafenib were not significant, as thevalues for the coefficient of drug interaction (CDI;refs. 25, 28) for HepG2-SR and Huh7-SR cells treatedwith 1, 10, or 100 nmol/L of rapamycin in the presenceof sorafenib (10 mmol/L) were all more than 0.7.

Figure 3. Autophagy affects thesensitivity of sorafenib-resistantHCC cells to sorafenib. A, lysatesof HepG2, HepG2-SR, Huh7, andHuh7-SR cells incubated with 0, 5or 10 mmol/L of sorafenib for 48hours were immunoblotted todetect expression of autophagy-associated proteins. B–D, theabove cells were incubated for 48hours in the presence or absenceof sorafenib (10 mmol/L in B and Dor 15 or 20 mmol/L in C), and/or 3-MA (3-methyladenine; 10 mmol/L),and/or Baf-A1 (bafilomycin A1, 50nmol/L), or with differentconcentrations of rapamycin (D).Cell viability (%) was comparedwith the corresponding untreatedcells. E, immunoblot analyses oflysates of Huh7-SR cells from B. F,immunoblot analyses of lysates ofHuh7-SR cells from D. The banddensity of LC3-II was normalizedto LC3-I, and that of other proteinswas normalized to b-actin. Datarepresent three independentexperiments. The statisticalcomparison was performed forsorafenib-resistant cells only.�, P < 0.05 and ��, P < 0.001 versusrespective untreated cells; †,P < 0.05 and z, P < 0.001 versussorafenib alone; #, P < 0.05 and ##,P < 0.001 versus rapamycin alone.øø, P < 0.001.

Akt/Autophagy for Sorafenib Resistance in HCC

www.aacrjournals.org Mol Cancer Ther; 13(6) June 2014 1593

on April 5, 2020. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst April 4, 2014; DOI: 10.1158/1535-7163.MCT-13-1043

Page 6: Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by … · Cancer Biology and Signal Transduction Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by

3-MA downregulated the expression of LC3-II andBeclin 1, whereas Baf-A1 upregulated LC3-II and down-regulated Beclin 1, in Huh7-SR cells (Fig. 3E). Both 3-MAand Baf-A1 increased the expression of pro–caspase-3 buthad little effect on cleaved-PARP expression (Fig. 3E).Rapamycinupregulated the expression of LC3-II, Beclin 1,p-Akt, and cleaved-PARP, and increased the activation ofcaspase-3 (Fig. 3F), indicating that induction of autophagyby rapamycin promotes apoptosis, but also results inactivation of Akt, in sorafenib-resistant cells.

Inhibition of Akt synergizes with sorafenib to inhibitcell viability, promote apoptosis, and induceautophagic cell death in sorafenib-resistant cells

Inhibition of Akt by GDC0068 synergized with sorafe-nib to reduce the viability ofHepG2-SR andHuh7-SR cellsin dose- and time-dependent manners (Fig. 4A and Sup-plementary Fig. S4A). The CDIs for HepG2-SR cells trea-ted with 1, 5, or 10 mmol/L of GDC0068 in the presence ofsorafenib (10 mmol/L) were 0.67, 0.63, or 0.65, respective-

ly; and that for Huh7-SR were 0.79, 0.59, or 0.61, respec-tively, indicating that the synergistic effects were signif-icant. The effects of Akt inhibition on cell viability werefurther supported by application of perifosine, anotherAkt inhibitor (Supplementary Fig. S4B). GDC0068 alsoshowed a significantly synergistic effect with sorafenib ininducing cell apoptosis with CDIs of 0.48 and 0.53, inHepG2-SR and Huh7-SR cells, respectively (Fig. 4Band C).

Sorafenib and GDC0068 upregulated, and their combi-nation showed an even stronger effect in increasing theexpression of LC3-II and Beclin 1 in Huh7-SR cells (Fig.4D). Inhibition of Akt by perifosine or depletion of Akt bysiRNAs had a similar effect on the expression of autop-hagic proteins (Supplementary Fig. S4C and S4D). Con-sequently, the number of GFP-LC3–positive dots in cellstreated with sorafenib or GDC0068 were significantlyhigher than untreated cells, and their combinationresulted in the highest number of GFP-LC3–positive dots(Fig. 4E). The CDI for GDC0068 and sorafenib to induce

Figure 4. Inhibition of Akt enhances the sensitivity of sorafenib-resistant HCC cells to sorafenib by inducing autophagy. A, HepG2-SR andHuh7-SR cells wereexposed for 48 hours to different concentrations of GDC0068, or 5 mmol/L of GDC0068 for different periods, in the presence or absence of sorafenib(10 mmol/L). Cell viability (%) was compared with the corresponding untreated cells. B–G, HepG2-SR and Huh7-SR cells were incubated for 48 hourswith sorafenib (10 mmol/L), GDC0068 (5 mmol/L), or in combination. Untreated cells served as controls. B, the apoptosis rates were measured by cytometry.C, representative images were taken from Huh7-SR cells stained with Annexin V/propidium iodide, and viewed by microscopy. D, cell lysates wereimmunoblotted. E and F, the above cells were transfected with GFP-LC3 expression vectors, GFP-LC3–positive dots were counted in 200 randomly selectedcells (E), and viewed under fluorescent microscopy (F). Arrows point to colocalized particles representing autophagosomes. G, electron microscopy;arrows point to autophagosomes (magnification, �10,000). ø, P < 0.05 and øø, P < 0.001. �, P < 0.05 and ��, P < 0.001 versus untreated control; z, P < 0.001versus sorafenib alone; #, P < 0.05 and ##, P < 0.001 versus GDC0068 alone.

Zhai et al.

Mol Cancer Ther; 13(6) June 2014 Molecular Cancer Therapeutics1594

on April 5, 2020. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst April 4, 2014; DOI: 10.1158/1535-7163.MCT-13-1043

Page 7: Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by … · Cancer Biology and Signal Transduction Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by

autophagy of Huh7-SR cells was 0.51, indicating that thesynergistic effectwas significant. The treated cells showedthat GFP-LC3 signals shifted from a diffuse cytoplasmicpattern to a punctate membrane pattern, suggesting theformation of autophagic vacuoles (Fig. 4F). Electronmicroscopy revealed abundant autophagic vacuoles insorafenib or GDC0068-treated cells, but scarcely inuntreated cells; and the combinational therapy resultedin the most autophagosomes (Fig. 4G).

GDC0068 synergizes with sorafenib to suppresssorafenib-resistant tumorsAdministration of sorafenib andGDC0068 into themice

reduced the size of Huh7-SR tumors by 15.8% and 37.5%,respectively, and the combination therapy furtherreduced the size by 71.3%, comparedwith vehicle-treatedtumors at day 15 (Fig. 5A).Immunoblotting analysis of tumor lysates demonstrat-

ed the similar alteration of expression of p-Akt, LC3-II,pro–caspase-3, and PARP (Fig. 5B), as shown in vitro(Supplementary Fig. S5A–S5C). Electron microscopy alsorevealed abundant autophagosomes in cells from tumors

treated by sorafenib plus GDC0068, but scarcely in vehi-cle-treated tumors (Fig. 5C). There were fewer Ki-67–positive cells in tumors treated with sorafenib orGDC0068, compared with vehicle-treated tumors; andthe combination therapy resulted in even fewer Ki-67–positive cells (Fig. 5D and E). Tumors treated with sor-afenib and GDC0068 had a greater number of TUNEL-positive cell than vehicle-treated tumors, and the combi-nation therapy resulted in even more TUNEL-positivecells (Fig. 5D and F).

DiscussionConsidering the dilemma that no effective systemic

therapy for HCC is available so far after failure of sor-afenib therapy (29), sorafenib remains in auniquepositionfor the treatment of HCC. The present study has demon-strated that activation ofAkt induced by chronic exposureof sorafenib regulates the acquired resistance to sorafenibby switching autophagy from a cytoprotective role to adeath-promoting mechanism in HCC cells.

Although it is not a direct target of sorafenib, the PI3K/Akt pathway plays an important role in sorafenib

Figure 5. Inhibition of Akt overcomes sorafenib resistance in vivo. A, subcutaneous tumors were established in mice that received different treatments for 15days as described inMaterials andMethods and Fig. 1. The size (mm3) of tumorswas recorded. B, immunoblot analyses of lysates of tumors harvested fromAat the end of experiments. C, sections of tumors treated with vehicle or sorafenibþGDC0068 were viewed under electron microscopy. Arrows point toautophagosomes (magnification, �10,000). D, representative images from tumor sections stained with an anti–Ki-67 Ab (top) or TUNEL (bottom;magnification,�200). Proliferation index (E) and apoptosis index (F) were quantified. n, the number of samples examined. �, P < 0.05 and ��, P < 0.001 versusvehicle-treated tumors; z, P < 0.001 versus sorafenib alone; ##, P < 0.001 versus GDC0068 alone.

Akt/Autophagy for Sorafenib Resistance in HCC

www.aacrjournals.org Mol Cancer Ther; 13(6) June 2014 1595

on April 5, 2020. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst April 4, 2014; DOI: 10.1158/1535-7163.MCT-13-1043

Page 8: Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by … · Cancer Biology and Signal Transduction Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by

resistance because it crosstalks with the MAPK/ERKpathway (10). Many lines of studies have shown thatsorafenib activates the PI3K/Akt pathway (7, 8, 18, 30),and blockage of this pathway enhances the efficacy ofsorafenib (7, 30), but it has also been reported that sor-afenib enhances proteasome inhibitor-induced cell deathby inactivating Akt in HCC (31), indicating that thecomplicated role of the PI3K/Akt pathway in sorafenibresistance is far from clear. Here, we have demonstratedthat Akt was activated in sorafenib-resistant cells, inaccordance with a previous report (9). Although phos-phorylation ofAkt can be induced byprotein phosphataseinactivation (32), the present study showed that the sor-afenib-induced Akt phosphorylation was independent ofprotein phosphatase 2A (PP2A), amajor serine/threoninephosphatase in eukaryotic cells (33). In accord, the syn-ergistic interaction between sorafenib and bortezomib,but not sorafenib alone, involved PP2A-dependent Aktinactivation in HCC cells (34). Akt regulates the expres-sion and/or phosphorylation of mTOR, GSk3b, Badand p27; mTOR regulates the apoptotic proteins S6Kand 4EBP1, and the autophagic proteins LC3 and Beclin 1;and GSk3b regulates cyclin D1 and caspase-9 (refs.5, 13, 29, 35; Fig. 6). Therefore, specific inhibition of Aktcould downregulate the above downstream factors, thusinducing apoptosis and inhibiting proliferation of HCCcells.

Among the over hundred molecules in the PI3K/Aktpathway, mTOR interacts with theMARK, apoptotic, andautophagic pathways and has been most extensivelystudied, and several mTOR inhibitors are under evalua-tion in clinical trials (36). Inhibition ofmTOR enhances thechemosensitivity (37), yields antiproliferative effects inboth parental and sorafenib-resistant HCC cells (35), andaugments the effects of sorafenib in a syngeneic orthotopicmodel of HCC (32). However, here we found that rapa-mycin did not show a synergistic effect with sorafenib ininhibiting the growth of sorafenib-resistant HCC cellsdespite the higher basal expression of mTOR (8). Theunhampered feedback loop driving PI3K/Akt activationby mTOR inhibition has been recently reported, andinhibition of Akt interrupted this rapamycin-inducedfeedback loop, thereby enhancing the antiproliferativeeffects of mTOR inhibition (38). Accordingly, here wehave shown that chronic exposure of sorafenib inhibitedactivation of ERK (9), sequentially inhibited mTOR inde-pendent of the PI3K/Akt pathway (39), resulting in acti-vation of Akt via the feedback loop in sorafenib-resistantHCC cells (Fig. 6). This may explain that the effect ofmTOR inhibition by rapamycin is not ideal to reversesorafenib resistance.

The role of autophagy as a target in cancer therapyremains controversial (40). It has been reported that sor-afenib induced autophagic death of HCC cells (17).

Figure 6. Proposedmechanisms bywhich activation of AKT contribute to acquired resistance to sorafenib via regulating autophagy, and the actions of agentsin parental and sorafenib-resistant cells. Blank area indicates aparental cell, and shadowed area a sorafenib-resistant cell.!, positive regulation or activation;?, negative regulation or blockade; þ P, regulation by phosphorylation.

Zhai et al.

Mol Cancer Ther; 13(6) June 2014 Molecular Cancer Therapeutics1596

on April 5, 2020. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst April 4, 2014; DOI: 10.1158/1535-7163.MCT-13-1043

Page 9: Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by … · Cancer Biology and Signal Transduction Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by

However, the authors used a high concentration of sor-afenib at 20 mmol/L (18). Even with the same concentra-tion of sorafenib, another study showed that autophagyprotected against sorafenib-induced cell death (20). Here,we have demonstrated that sorafenib at 10 mmol/L acti-vated autophagy, which played a protective role againstcell death of parental HCC cells, and is in agreement witha previous report (21); but exhibited a death-promotingrole in sorafenib-resistant HCC cells. Autophagy inducedbyvorinostat, a histonedeacetylase inhibitor that has beenapproved for treating cutaneousT-cell lymphoma in clinic(41), protected against cell death independent ofmTOR invorinostat-resistant lymphoma cells (42). The autophagicpathway crosstalks with both the caspase-dependent and-independent apoptotic pathways (13, 21, 43). Sustaineddrug exposure could induce imbalanced apoptotic path-ways, leading to the resistance to apoptosis (44).When theapoptotic pathway is suppressed, autophagy as an adap-tive responsewill switch from a protective role to a death-promoting function through the existing cross-talk withthe apoptotic pathway (13, 45). This explanation is sup-ported by a recent study that paclitaxel resistance isassociated with switch from apoptotic to autophagic celldeath in breast cancer cells (46).Themechanisms accounting for the acquired resistance

to sorafenib in HCC remain complex (47). The detailedcellular signaling pathways and their interactions inparental and sorafenib-resistant HCC cells, and the inter-ventions with agents tested in the present study aresummarized in Fig. 6. Briefly, autophagy plays a protec-tive role against sorafenib-induced cell death in the paren-tal cells. In sorafenib-resistant cells, sustained exposure tosorafenib activates Akt directly or via the feedback loop ofmTOR, which is inhibited by sorafenib through the ERKpathway (Fig. 6). Sorafenib induces autophagy indepen-dent of the ERKpathway (20). Activation of autophagy byrapamycin partially reverses the acquired resistance tosorafenib as rapamycin-mediated mTOR inhibition inturn activates Akt via the feedback loop. However, inhi-bition of Akt by GDC0068 significantly enhances the

efficacy of sorafenib to suppress the growth of sorafe-nib-resistant HCC cells by inducing autophagic celldeath. As a novel ATP-competitive pan-Akt inhibitor,GDC0068 binds to the active site of Akt and protects itfrom phosphatases, leading to increased Akt phosphor-ylation (48, 49). It has been reported that GDC0068 hasless dose-limiting toxicity and is more effective in inhibit-ing the function of Akt than allosteric Akt inhibitors (48,49). GDC0068 has displayed anticancer activities againstseveral types of cancers (49). The results presented war-rant clinical investigation of GDC0068 as the second-linetreatment after the failure of sorafenib to treat advancedHCC.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: H. Qiao, H. Jiang, X. SunDevelopment of methodology: B. Zhai, F. Hu, X. SunAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): B. Zhai, X. Jiang, J. Xu, D. Zhao, X. SunAnalysis and interpretation of data (e.g., statistical analysis, biostatis-tics, computational analysis): B. Zhai, S. Pan, H. Qiao, H. Jiang, X. SunWriting, review, and/or revision of the manuscript: B. Zhai, X. Dong,X. SunAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): G. Tan, Z. Wei, H. Jiang, X. SunStudy supervision: J. Xu, B. Liu, H. Qiao, H. Jiang, X. Sun

AcknowledgmentsThe authors thank Dr. Shiva Reddy for reviewing this article.

Grant SupportThisworkwas supported by theNationalNatural Scientific Foundation

of China (30973474, to X. Sun and H. Qiao; 81172331, to X. Sun), theHeilongjiang Provincial Scientific Foundation (GB07C32405, to B. Liu;H201307, to G. Tan), and The Youth Scientific Fund of HeilongjiangProvince (QC2013C103, to B. Zhai; QC2013C098, to X. Jiang), China.

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.

ReceivedDecember 17, 2013; revisedMarch 18, 2014; acceptedMarch 25,2014; published OnlineFirst April 4, 2014.

References1. JemalA,BrayF,CenterMM,Ferlay J,WardE, FormanD.Global cancer

statistics. CA Cancer J Clin 2011;61:69–90.2. Zhu AX. Systemic treatment of hepatocellular carcinoma: dawn of a

new era? Ann Surg Oncol 2010;17:1247–56.3. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al.

Sorafenib in advanced hepatocellular carcinoma. N Engl J Med2008;359:378–90.

4. ChengAL, KangYK,ChenZ, TsaoCJ,Qin S, KimJS, et al. Efficacy andsafety of sorafenib in patients in the Asia-Pacific region with advancedhepatocellular carcinoma: a phase III randomised, double-blind, pla-cebo-controlled trial. Lancet Oncol 2009;10:25–34.

5. Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov2005;4:988–1004.

6. Zhou L, Huang Y, Li J, Wang Z. The mTOR pathway is associated withthe poor prognosis of human hepatocellular carcinoma. Med Oncol2010;27:255–61.

7. Gedaly R, Angulo P, Hundley J, DailyMF, ChenC, Koch A, et al. PI-103and sorafenib inhibit hepatocellular carcinoma cell proliferation byblocking Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. AnticancerRes 2010;30:4951–8.

8. Huynh H, Ngo VC, Koong HN, Poon D, Choo SP, Thng CH, et al.Sorafenib and rapamycin induce growth suppression in mousemodels of hepatocellular carcinoma. J Cell Mol Med 2009;13:2673–83.

9. Chen KF, Chen HL, Tai WT, Feng WC, Hsu CH, Chen PJ, et al.Activation of phosphatidylinositol 3-kinase/Akt signaling pathwaymediates acquired resistance to sorafenib in hepatocellular carcinomacells. J Pharmacol Exp Ther 2011;337:155–61.

10. Mendoza MC, Er EE, Blenis J. The Ras-ERK and PI3K-mTOR path-ways: cross-talk and compensation. Trends Biochem Sci 2011;36:320–8.

11. DavidR.Metabolism:Keeping fit with autophagy. NatRevMolCell Biol2012;13:136.

Akt/Autophagy for Sorafenib Resistance in HCC

www.aacrjournals.org Mol Cancer Ther; 13(6) June 2014 1597

on April 5, 2020. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst April 4, 2014; DOI: 10.1158/1535-7163.MCT-13-1043

Page 10: Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by … · Cancer Biology and Signal Transduction Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by

12. Codogno P, Mehrpour M, Proikas-Cezanne T. Canonical and non-canonical autophagy: variations on a common theme of self-eating?Nat Rev Mol Cell Biol 2012;13:7–12.

13. Su M, Mei Y, Sinha S. Role of the crosstalk between autophagy andapoptosis in cancer. J Oncol 2013;2013:102735.

14. Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Packer M, et al. Bcl-2antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell2005;122:927–39.

15. Funderburk SF, Wang QJ, Yue Z. The Beclin 1-VPS34 complex–at thecrossroads of autophagy and beyond. Trends Cell Biol 2010;20:355–62.

16. Sini P, James D, Chresta C, Guichard S. Simultaneous inhibition ofmTORC1 and mTORC2 by mTOR kinase inhibitor AZD8055 inducesautophagy and cell death in cancer cells. Autophagy 2010;6:553–4.

17. Tai WT, Shiau CW, Chen HL, Liu CY, Lin CS, Cheng AL, et al. Mcl-1-dependent activation of Beclin 1 mediates autophagic cell deathinduced by sorafenib and SC-59 in hepatocellular carcinoma cells.Cell Death Dis 2013;4:e485.

18. Manov I, Pollak Y, Broneshter R, Iancu TC. Inhibition of doxorubicin-induced autophagy in hepatocellular carcinoma Hep3B cells by sor-afenib–the role of extracellular signal-regulated kinase counteraction.FEBS J 2011;278:3494–507.

19. Xu N, Zhang J, Shen C, Luo Y, Xia L, Xue F, et al. Cisplatin-induceddownregulation of miR-199a-5p increases drug resistance by activat-ing autophagy in HCC cell. Biochem Biophys Res Commun2012;423:826–31.

20. Shi YH, Ding ZB, Zhou J, Hui B, Shi GM, Ke AW, et al. Targetingautophagy enhances sorafenib lethality for hepatocellular carcinomavia ER stress-related apoptosis. Autophagy 2011;7:1159–72.

21. Shimizu S, Takehara T, Hikita H, Kodama T, Tsunematsu H, Miyagi T,et al. Inhibition of autophagy potentiates the antitumor effect of themultikinase inhibitor sorafenib in hepatocellular carcinoma. Int J Can-cer 2012;131:548–57.

22. Zhang QB, Sun HC, Zhang KZ, Jia QA, Bu Y, Wang M, et al. Sup-pression of natural killer cells by sorafenib contributes to prometastaticeffects in hepatocellular carcinoma. PLoS ONE 2013;8:e55945.

23. Lin J, Sampath D, Nannini MA, Lee BB, Degtyarev M, Oeh J, et al.Targeting activated Akt with GDC-0068, a novel selective Akt inhibitorthat is efficacious in multiple tumor models. Clin Cancer Res2013;19:1760–72.

24. Wang D, Ma Y, Li Z, Kang K, Sun X, Pan S, et al. The role of AKT1 andautophagy in the protective effect of hydrogen sulphide against hepat-ic ischemia/reperfusion injury in mice. Autophagy 2012;8:954–62.

25. WangJ,MaY, JiangH, ZhuH,Liu L,SunB, et al. Overexpressionof vonHippel-Lindau protein synergizes with doxorubicin to suppress hepa-tocellular carcinoma in mice. J Hepatol 2011;55:359–68.

26. Wei Z, JiangX,QiaoH, Zhai B, ZhangL, ZhangQ, et al. STAT3 interactswith Skp2/p27/p21 pathway to regulate the motility and invasion ofgastric cancer cells. Cell Signal 2013;25:931–8.

27. Jung CH, Ro SH, Cao J, Otto NM, Kim DH. mTOR regulation ofautophagy. FEBS Lett 2010;584:1287–95.

28. He C, Sun XP, Qiao H, Jiang X, Wang D, Jin X, et al. Downregulatinghypoxia-inducible factor-2alpha improves the efficacy of doxorubicinin the treatment of hepatocellular carcinoma. Cancer Sci 2012;103:528–34.

29. Zhai B, Sun XY. Mechanisms of resistance to sorafenib and thecorresponding strategies in hepatocellular carcinoma.World JHepatol2013;5:345–52.

30. Piguet AC, Saar B, Hlushchuk R, St-Pierre MV, McSheehy PM, Rado-jevic V, et al. Everolimus augments the effects of sorafenib in asyngeneic orthotopic model of hepatocellular carcinoma. Mol CancerTher 2011;10:1007–17.

31. Honma Y, Shimizu S, Takehara T, Harada M. Sorafenib enhancesproteasome inhibitor-induced cell death via inactivation of Aktand stress-activated protein kinases. J Gastroenterol 2014;49:517–26.

32. Cheung M, Testa JR. Diverse mechanisms of AKT pathway activationin human malignancy. Curr Cancer Drug targets. 2013;13:234–44.

33. Perrotti D, Neviani P. Protein phosphatase 2A: a target for anticancertherapy. Lancet Oncol 2013;14:e229–38.

34. Chen KF, Yu HC, Liu TH, Lee SS, Chen PJ, Cheng AL. Synergisticinteractions between sorafenib and bortezomib in hepatocellular car-cinoma involve PP2A-dependent Akt inactivation. J Hepatol 2010;52:88–95.

35. Serova M, de Gramont A, Tijeras-Raballand A, Dos Santos C, RiveiroME, Slimane K, et al. Benchmarking effects of mTOR, PI3K, and dualPI3K/mTOR inhibitors in hepatocellular and renal cell carcinomamod-els developing resistance to sunitinib and sorafenib. Cancer Che-mother Pharmacol 2013;71:1297–307.

36. Burris HA III. Overcoming acquired resistance to anticancer therapy:focus on the PI3K/AKT/mTOR pathway. Cancer Chemother Pharma-col 2013;71:829–42.

37. Tam KH, Yang ZF, Lau CK, Lam CT, Pang RW, Poon RT. Inhibition ofmTOR enhances chemosensitivity in hepatocellular carcinoma.Cancer Lett 2009;273:201–9.

38. Seront E, Pinto A, Bouzin C, Bertrand L, Machiels JP, Feron O. PTENdeficiency is associated with reduced sensitivity to mTOR inhibitor inhuman bladder cancer through the unhampered feedback loop drivingPI3K/Akt activation. Br J Cancer 2013;109:1586–92.

39. Miyazaki M, McCarthy JJ, Fedele MJ, Esser KA. Early activation ofmTORC1 signalling in response to mechanical overload is indepen-dent of phosphoinositide 3-kinase/Akt signalling. J Physiol 2011;589:1831–46.

40. Carew JS, Kelly KR, Nawrocki ST. Autophagy as a target for cancertherapy: new developments. Cancer Manage Res 2012;4:357–65.

41. Hideshima T, Richardson PG, Anderson KC. Mechanism of action ofproteasome inhibitors and deacetylase inhibitors and the biologicalbasis of synergy in multiple myeloma. Mol Cancer Ther 2011;10:2034–42.

42. Dupere-Richer D, Kinal M, Menasche V, Nielsen TH, Del Rincon S,Pettersson F, et al. Vorinostat-induced autophagy switches from adeath-promoting to a cytoprotective signal to drive acquired resis-tance. Cell Death Dis 2013;4:e486.

43. Mu~noz-G�amez JA, Rodríguez-Vargas JM, Quiles-P�erez R, Aguilar-Quesada R, Martín-Oliva D, de Murcia G, et al. PARP-1 is involved inautophagy induced by DNA damage. Autophagy 2009;5:61–74.

44. Lackner MR, Wilson TR, Settleman J. Mechanisms of acquiredresistance to targeted cancer therapies. Future Oncol 2012;8:999–1014.

45. Hu YL, Jahangiri A, Delay M, Aghi MK. Tumor cell autophagy as anadaptive response mediating resistance to treatments such as anti-angiogenic therapy. Cancer Res 2012;72:4294–9.

46. Ajabnoor GM, Crook T, Coley HM. Paclitaxel resistance is associatedwith switch from apoptotic to autophagic cell death in MCF-7 breastcancer cells. Cell Death Dis 2012;3:e260.

47. Berasain C. Hepatocellular carcinoma and sorafenib: too many resis-tance mechanisms? Gut 2013;62:1674–5.

48. LinK, Lin J,WuWI,Ballard J, LeeBB,GloorSL, et al. AnATP-site on-offswitch that restricts phosphatase accessibility of Akt. Sci Signal2012;5:ra37.

49. Lin J, Sampath D, Nannini MA, Lee BB, Degtyarev M, Oeh J, et al.Targeting activated Akt with GDC-0068, a novel selective Akt inhibitorthat is efficacious in multiple tumor models. Clin Cancer Res2013;19:1760–72.

Mol Cancer Ther; 13(6) June 2014 Molecular Cancer Therapeutics1598

Zhai et al.

on April 5, 2020. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst April 4, 2014; DOI: 10.1158/1535-7163.MCT-13-1043

Page 11: Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by … · Cancer Biology and Signal Transduction Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by

2014;13:1589-1598. Published OnlineFirst April 4, 2014.Mol Cancer Ther   Bo Zhai, Fengli Hu, Xian Jiang, et al.   Hepatocellular CarcinomaSwitching Protective Autophagy to Autophagic Cell Death in Inhibition of Akt Reverses the Acquired Resistance to Sorafenib by

  Updated version

  10.1158/1535-7163.MCT-13-1043doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2014/04/04/1535-7163.MCT-13-1043.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/13/6/1589.full#ref-list-1

This article cites 49 articles, 8 of which you can access for free at:

  Citing articles

  http://mct.aacrjournals.org/content/13/6/1589.full#related-urls

This article has been cited by 3 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/13/6/1589To request permission to re-use all or part of this article, use this link

on April 5, 2020. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst April 4, 2014; DOI: 10.1158/1535-7163.MCT-13-1043