13
Companion Diagnostics and Cancer Biomarkers Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus Hung Huynh 1 , Huai-Xiang Hao 2 , Stephen L. Chan 3 , David Chen 4 , Richard Ong 1 , Khee Chee Soo 1 , Panisa Pochanard 2 , David Yang 2 , David Ruddy 2 , Manway Liu 2 , Adnan Derti 2 , Marissa N. Balak 2 , Michael R. Palmer 2 , Yan Wang 2 , Benjamin H. Lee 2 , Dalila Sellami 4 , Andrew X. Zhu 5 , Robert Schlegel 2 , and Alan Huang 2 Abstract Hepatocellular carcinoma (HCC) is the third leading cause of cancer deaths worldwide and hyperactivation of mTOR signaling plays a pivotal role in HCC tumorigenesis. Tuberous sclerosis complex (TSC), a heterodimer of TSC1 and TSC2, functions as a negative regulator of mTOR signaling. In the current study, we discovered that TSC2 loss-of-function is common in HCC. TSC2 loss was found in 4 of 8 HCC cell lines and 8 of 28 (28.6%) patient-derived HCC xenografts. TSC2 mutations and deletions are likely to be the underlying cause of TSC2 loss in HCC cell lines, xenografts, and primary tumors for most cases. We further demonstrated that TSC2-null HCC cell lines and xenografts had elevated mTOR signaling and, more importantly, were signi- cantly more sensitive to RAD001/everolimus, an mTORC1 inhib- itor. These preclinical ndings led to the analysis of TSC2 status in HCC samples collected in the EVOLVE-1 clinical trial of ever- olimus using an optimized immunohistochemistry assay and identied 15 of 139 (10.8%) samples with low to undetectable levels of TSC2. Although the sample size is too small for formal statistical analysis, TSC2-null/low tumor patients who received everolimus tended to have longer overall survival than those who received placebo. Finally, we performed an epidemiology survey of more than 239 Asian HCC tumors and found the frequency of TSC2 loss to be approximately 20% in Asian HBV þ HCC. Taken together, our data strongly argue that TSC2 loss is a predictive biomarker for the response to everolimus in HCC patients. Mol Cancer Ther; 14(5); 122435. Ó2015 AACR. Introduction Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths, accounting for 250,000 to one million deaths globally each year (1). The annual number of incidence and death of HCC are virtually identical, illustrating the high mortality rate of this aggressive disease. Because of the relative paucity of symptoms in the early stages and the rapid doubling time of the tumor, more than 80% of HCC are diagnosed at advanced stages when surgery is no longer an option (2). Two randomized trials of the multikinase inhibitor sorafenib in HCC patients showed an improvement in median overall survival of close to 3 months, establishing sorafenib as the only targeted therapy and the standard of care in advanced HCC (3, 4). However, the efcacy of sorafenib is transient and modest and there are no established predictive biomarkers for sorafenib. Thus, there is an urgent unmet need for more effective novel therapies to combat this deadly disease. mTOR is a serine/threonine kinase that plays a critical role in regulating protein translation, cell-cycle progression, and cellular function (5, 6). Two well-characterized substrates of mTOR are S6K1 and 4E-BP1, and mTOR activity is regulated by the PI3K/ AKT pathway in response to activation of receptor tyrosine kinases. mTOR activation is found in tumors with primary genetic abnormalities activating the PI3K/AKT pathway (7, 8) or in tumors responding to mitogenic signals (9, 10). Hyperactivation of mTOR signaling has been identied in approximately 50%60% of HCC (11). Receptor tyrosine kinase activation rather than somatic mutations of PTEN or PI3KCA, which are rare in HCC, is likely to be responsible for activation of mTOR in HCC. Consis- tent with this notion, activation of AKT has been found in 40%60% of human HCC samples (12) and is a risk factor for early recurrence and poor prognosis (13). More importantly, recent 1 Laboratory of Molecular Endocrinology, Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore. 2 Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachu- setts. 3 State Key Laboratory in Oncology in South China, Department of Clinical Oncology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, PR China. 4 Oncology Global Develop- ment, Novartis Pharmaceuticals Corporation, East Hanover, New Jer- sey. 5 Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). H. Huynh and H.-X. Hao contributed equally to this article. Corresponding Authors: Alan Huang, Novartis Institutes for BioMedical Research, 250 Mass Avenue, Cambridge, MA 02139. Phone: 617-871-7397; Fax: 617-871-7397; E-mail: [email protected]; and H. Huynh, Laboratory of Molecular Endocrinology, Division of Cellular and Molecular Research, Hum- phrey Oei Institute of Cancer Research, National Cancer Centre 11 Hospital Drive, Singapore, 169610, Singapore. E-mail: [email protected] doi: 10.1158/1535-7163.MCT-14-0768 Ó2015 American Association for Cancer Research. Molecular Cancer Therapeutics Mol Cancer Ther; 14(5) May 2015 1224 on June 9, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst February 27, 2015; DOI: 10.1158/1535-7163.MCT-14-0768

Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in ...Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus Hung Huynh1, Huai-Xiang Hao2,

  • Upload
    others

  • View
    6

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in ...Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus Hung Huynh1, Huai-Xiang Hao2,

Companion Diagnostics and Cancer Biomarkers

Loss of Tuberous Sclerosis Complex 2 (TSC2)Is Frequent in Hepatocellular Carcinoma andPredicts Response to mTORC1 InhibitorEverolimusHung Huynh1, Huai-Xiang Hao2, Stephen L. Chan3, David Chen4, Richard Ong1,Khee Chee Soo1, Panisa Pochanard2, David Yang2, David Ruddy2, Manway Liu2,Adnan Derti2, Marissa N. Balak2, Michael R. Palmer2, Yan Wang2, Benjamin H. Lee2,Dalila Sellami4, Andrew X. Zhu5, Robert Schlegel2, and Alan Huang2

Abstract

Hepatocellular carcinoma (HCC) is the third leading cause ofcancer deaths worldwide and hyperactivation of mTOR signalingplays a pivotal role in HCC tumorigenesis. Tuberous sclerosiscomplex (TSC), a heterodimer of TSC1 and TSC2, functions as anegative regulator of mTOR signaling. In the current study, wediscovered that TSC2 loss-of-function is common in HCC. TSC2loss was found in 4 of 8 HCC cell lines and 8 of 28 (28.6%)patient-derived HCC xenografts. TSC2 mutations and deletionsare likely to be the underlying cause of TSC2 loss in HCC celllines, xenografts, and primary tumors for most cases. We furtherdemonstrated that TSC2-null HCC cell lines and xenografts hadelevated mTOR signaling and, more importantly, were signifi-cantly more sensitive to RAD001/everolimus, anmTORC1 inhib-

itor. These preclinical findings led to the analysis of TSC2 statusin HCC samples collected in the EVOLVE-1 clinical trial of ever-olimus using an optimized immunohistochemistry assay andidentified 15 of 139 (10.8%) samples with low to undetectablelevels of TSC2. Although the sample size is too small for formalstatistical analysis, TSC2-null/low tumor patients who receivedeverolimus tended to have longer overall survival than those whoreceived placebo. Finally, we performed an epidemiology surveyof more than 239 Asian HCC tumors and found the frequency ofTSC2 loss to be approximately 20% in Asian HBVþ HCC. Takentogether, our data strongly argue that TSC2 loss is a predictivebiomarker for the response to everolimus in HCC patients. MolCancer Ther; 14(5); 1224–35. �2015 AACR.

IntroductionHepatocellular carcinoma (HCC) is the third leading cause of

cancer-related deaths, accounting for 250,000 to one milliondeaths globally each year (1). The annual number of incidenceand death of HCC are virtually identical, illustrating the high

mortality rate of this aggressive disease. Because of the relativepaucity of symptoms in the early stages and the rapid doublingtime of the tumor, more than 80% of HCC are diagnosed atadvanced stages when surgery is no longer an option (2). Tworandomized trials of the multikinase inhibitor sorafenib in HCCpatients showed an improvement in median overall survival ofclose to 3 months, establishing sorafenib as the only targetedtherapy and the standard of care in advanced HCC (3, 4).However, the efficacy of sorafenib is transient and modest andthere are no established predictive biomarkers for sorafenib. Thus,there is an urgent unmet need formore effective novel therapies tocombat this deadly disease.

mTOR is a serine/threonine kinase that plays a critical role inregulating protein translation, cell-cycle progression, and cellularfunction (5, 6). Two well-characterized substrates of mTOR areS6K1 and 4E-BP1, and mTOR activity is regulated by the PI3K/AKT pathway in response to activation of receptor tyrosinekinases.mTOR activation is found in tumors with primary geneticabnormalities activating the PI3K/AKT pathway (7, 8) or intumors responding to mitogenic signals (9, 10). Hyperactivationof mTOR signaling has been identified in approximately 50%–

60%of HCC (11). Receptor tyrosine kinase activation rather thansomatic mutations of PTEN or PI3KCA, which are rare in HCC, islikely to be responsible for activation of mTOR in HCC. Consis-tent with this notion, activation of AKT has been found in 40%–

60% of human HCC samples (12) and is a risk factor for earlyrecurrence and poor prognosis (13). More importantly, recent

1Laboratory of Molecular Endocrinology, Division of Cellular andMolecular Research, Humphrey Oei Institute of Cancer Research,National CancerCentre, Singapore. 2OncologyTranslationalMedicine,Novartis Institutes for Biomedical Research, Cambridge, Massachu-setts. 3State Key Laboratory in Oncology in South China, Departmentof Clinical Oncology, The Chinese University of Hong Kong, Prince ofWales Hospital, Hong Kong, PR China. 4Oncology Global Develop-ment, Novartis Pharmaceuticals Corporation, East Hanover, New Jer-sey. 5Massachusetts General Hospital Cancer Center, Harvard MedicalSchool, Boston, Massachusetts.

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

H. Huynh and H.-X. Hao contributed equally to this article.

Corresponding Authors: Alan Huang, Novartis Institutes for BioMedicalResearch, 250 Mass Avenue, Cambridge, MA 02139. Phone: 617-871-7397; Fax:617-871-7397; E-mail: [email protected]; and H. Huynh, Laboratory ofMolecular Endocrinology, Division of Cellular and Molecular Research, Hum-phrey Oei Institute of Cancer Research, National Cancer Centre 11 Hospital Drive,Singapore, 169610, Singapore. E-mail: [email protected]

doi: 10.1158/1535-7163.MCT-14-0768

�2015 American Association for Cancer Research.

MolecularCancerTherapeutics

Mol Cancer Ther; 14(5) May 20151224

on June 9, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2015; DOI: 10.1158/1535-7163.MCT-14-0768

Page 2: Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in ...Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus Hung Huynh1, Huai-Xiang Hao2,

studies using knockout mice showed that mTOR activation wassufficient to cause HCC in rodent models (14), indicating thepivotal role of mTOR signaling in HCC. The allosteric mTORC1inhibitor RAD001/everolimus has been tested in a randomized,placebo-controlled phase III clinical trial in patients withadvanced HCC who failed sorafenib (EVOLVE-1; ref.15). Overallsurvival (OS), the primary endpoint, showed nonsignificantimprovement with everolimus treatment versus placebo. Themolecular predictors for a small number of patients who benefit-ed from the everolimus treatment are currently being explored.

The tuberous sclerosis complex (TSC) formed by TSC1 andTSC2 functions as a critical negative regulator of mTOR throughthe GAP (GTPase-activating protein) activity of TSC2 towardsthe small GTPase RHEB (16). Phosphorylation of TSC2 by AKTor other kinases inactivates TSC2, which leads to activation ofmTOR (16). Although the GAP activity of TSC2 is of obviousfunctional importance to the TSC complex, TSC1 stabilizesTSC2 by preventing ubiquitin-mediated degradation of TSC2(16). Therefore, inactivation of either TSC1 or TSC2 genedisrupts the TSC1/TSC2 complex and results in unrestrainedkinase activity of mTOR. Everolimus is an approved and effec-tive drug for treating neurologic manifestations of TSC, ahereditary disorder caused by germline mutations in TSC1 orTSC2. Intuitively, TSC lesions in tumors might be potentialpredictive biomarkers for response to everolimus treatment.However, this link has not been rigorously examined, in partbecause the frequency of somatic mutations in TSC1/TSC2 inmost types of tumors, including HCC, is quite low.

In this study, we observed frequent loss of TSC2 in HCC celllines and demonstrated that TSC2-null cell lines were moresensitive to mTOR inhibition by everolimus. We then confirmedTSC2 loss in patient-derived HCC xenografts and observedremarkable antitumor efficacy of everolimus in TSC2-null xeno-grafts. TSC2 mutations and deletions were identified in many ofthe TSC2-null HCC cell lines and xenografts, providing a geneticmechanism for TSC2 loss. We also developed a TSC2 immuno-histochemical (IHC) assay for identifying TSC2 loss in primarytumor samples. Analysis of TSC2 status in EVOLVE-1 clinicalsamples suggested that TSC2-null/low tumor patients whoreceived everolimus tended to have longer OS than those whoreceived placebo. On the basis of our TSC2 IHC profiling of 239tumors, the frequency of TSC2 loss is estimated to be around 20%in Asian HBVþ HCC. Taken together, our data strongly suggestthat TSC2 loss may be used as a predictive biomarker for ever-olimus efficacy in HCC clinical trials.

Materials and MethodsAntibodies and compounds

Rabbit antibodies against TSC1, TSC2, p-S6K1(T389), S6K1, p-4E-BP1(T37/T46), p-4E-BP1(T70), 4E-BP1, p-AKT(S473), AKT, p-CDK2(T14/T15), PTEN and p27 were purchased from Cell Sig-naling Technology. Tubulin antibody (mouse) was purchasedfrom Sigma. RAD001/everolimus, BKM120, MK-2206, andMEK162 were synthesized at Novartis. pcDNA3-Flag-TSC2 plas-mid was a gift from Dr. Pablo Garcia (Novartis Institutes forBiomedical Research, Emeryville, CA). The TSC2 G305V mutantwas generated using the Q5 site-directed mutagenesis kit (NewEngland Biolabs) according to the manufacturer's instructions.The sequences of mutagenic primers used are: forward, 50-gccggtgggctacccagagagcc-30; reverse, 50-ggctctctgggtagcccaccggc-30.

Cell cultureHepatocellular carcinoma cell lines were part of the Novartis

Cancer Cell line Encyclopedia (CCLE) collection (17) and wereauthenticated by single-nucleotide polymorphism (SNP) analysisin 2012.

Reverse phase protein arrayReverse phase protein array (RPPA) was performed at the RPPA

Core Facility at MD Anderson Cancer Center (Houston, TX)according to standard procedures.

ImmunoblottingTotal cell lysates were prepared by lysing cells using RIPA buffer

(50 mmol/L Tris-HCl, pH 7.4, 150 mmol/L NaCl, 1% NP-40,0.5% sodium deoxycholate, 0.1% SDS, 1 mmol/L EDTA) supple-mented with protease inhibitors and phosphatase inhibitors,followed by centrifugation at 14,000 rpm for 10 minutes at 4�C.Equal amounts of proteinwere resolvedby SDS-PAGE, transferredto nitrocellulose membranes, and incubated with primary anti-bodies overnight at 4�C. Secondary antibodies conjugated witheither HRP or infrared dyes were used for signal visualization byECL film method or an LI-COR Odyssey scanner, respectively.

Reverse transcription and TaqMan quantitative RT-PCRTotal RNA was extracted from cells using the RNeasy Plus Mini

Kit (Qiagen) and reverse transcribed with TaqMan Reverse Tran-scription Reagents (Applied Biosystems) according to the man-ufacturers' instructions. Transcript levels were assessed using theABI PRISM7900HT.Quantitative real-timePCRwasperformed in12 mL reactions consisting of 0.6 mL of 20� TaqMan probe andPCR primer mix, 6 mL 2� TaqManUniversal PCRMasterMix, and5.4 mL diluted cDNA template. The thermocycling conditionsutilized were 2 minutes at 50�C, 10 minutes at 95�C, followed by40 cycles of 15 seconds at 95�C and 1 minute at 60�C. Allexperiments were performed in quadruplicate. Gene expressionanalysis was performed using the comparative DDCt method withthe housekeeping gene, GUSB, for normalization. All TaqManprimers/probes were purchased from Applied Biosystems (TSC1,Hs01060648_m1; TSC2, Hs01020387_m1; GUSB, Hs99999908_m1; MZB1, Hs00414907_m1).

CellTiter Glo proliferation assayCells were seeded in 96-well plates and treated with different

doses of everolimus (6 fold serial dilutions starting from1mmol/L)for 3 days. A CellTiter Glo assay was performed according to themanufacturers' protocols. Everolimus dose–response curve andIC50 values were calculated using the XLfit add-in in Excel.

Patient-derived HCC xenograft studyThis study received ethics board approval at the SingHealth. All

miceweremaintained according to theGuide for theCare andUseof Laboratory Animals published by the NIH (Bethesda, MD).

The establishment of our collection of HCC patient-derivedxenografts has been previously described (18) and the indicatedxenograft lines were subcutaneously implanted into male SCIDmice (Animal Resources Centre) aged 9 to 10 weeks. For antitu-mor efficacy experiments, mice bearing the indicated tumors (10mice per group)were orally administratedwith either vehicle (5%dextrose) or varying doses of everolimus (0.25, 0.5, 0.75 or 1.0mg/kg) daily for indicated days. Treatment started when thetumors reached the size of approximately 170 to 600 mm3

Frequent TSC2 Loss in HCC and Everolimus Response

www.aacrjournals.org Mol Cancer Ther; 14(5) May 2015 1225

on June 9, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2015; DOI: 10.1158/1535-7163.MCT-14-0768

Page 3: Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in ...Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus Hung Huynh1, Huai-Xiang Hao2,

(dependent on the lines or experimental purpose). Tumor growthwas monitored and tumor volume was calculated as described(18, 19). At the end of the study, mice were sacrificed with bodyand tumor weights recorded. The efficacy of everolimus wasdetermined by T/C ratio, where T and C are the median weightof everolimus-treated and vehicle-treated tumors, respectively.T/C ratios less than 0.42 were considered active according to thewidely adopted criteria from the Drug Evaluation Branch of theDivision of Cancer Treatment, NCI (Bethesda, MD).

DNA sequencing and copy number analysisHCC cell lines DNA sequencing and copy number analysis

using SNP6.0 array has been previously described (17). GenomicDNA from HCC xenografts and primary tumors were sequencedusing the T5 panel targeted sequencing at Foundation Medicine.Multiplex ligation-dependent probe amplification (MLPA) ofTSC2 duplications and deletions was performed using TSC2SALSA MLPA probemix P046-B2 (MRC-Holland) according tothe manufacturer's instructions and analyzed using GeneMarkersoftware from SoftGenetics LLC.

TSC2 IHC assayHCC cell lines and patient-derived xenografts were collected,

fixed with formalin and processed for paraffin embedding. HCCtissue microarrays were purchased from BioChain (catalog #Z7020056 and Z7020057, unrelated HCC TMAs) and FolioBio(catalog# ARY-HH0225). Formalin-fixed, paraffin-embedded(FFPE) tissue sections (4mm thick) were stained on the VentanaDiscovery XT automated staining platform. Standard antigenretrieval condition in cell conditioning 1 buffer was used, fol-lowed by incubation with rabbit anti-TSC2 antibody (Cell Sig-naling Technology, catalog # 4308) at a dilution of 1:200 for 32minutes at 37�C. Signal was then visualized using Omni Map(OMap) anti-Rabbit HRP and DAB.

TSC2 expression was assessed semiquantitatively by apathologist using a histo-score (H-score) methodology basedon staining intensity and percentage of tumor cells expressingTSC2. Intensity was scored as 0 (none), 1 (weak), 2 (moderate),or 3 (strong). H-score was calculated as follows: H-score ¼[fraction of cells with intensity grade 1 (%)] þ [fraction of cellswith intensity grade 2 (%) � 2] þ [fraction of cells withintensity grade 3 (%) � 3].

Clinical studyThe EVOLVE-1 (clinicaltrial.gov # NCT01035229) clinical trial

design, patient inclusion, and exclusion criteria have been pub-lished (15). Briefly, 546patients aged�18yearswithBCLC stage Bor C HCC whose disease progressed during or after sorafenib orwho were intolerant to sorafenib were randomized 2:1 to ever-olimus 7.5mg/day group or placebo group. Study drug was givencontinuously until disease progression or intolerable toxicity. CT/MRI scans were performed every 6 weeks and primary endpointwas OS.

The study protocol was reviewed and approved by the Inde-pendent Ethics Committee and the Institutional Review Board foreach center. The study was conducted according to Good ClinicalPractices and according to the ethical principles of theDeclarationof Helsinki. Archival formalin-fixed, paraffin-embedded (FFPE)tissue sections were collected from patients who providedinformed consent for biomarker study.

Statistical analysisFor xenograft growth curves, results are expressed as the means

� SEM from an appropriate number of replicates for each treat-ment group versus time as indicated in the figure legends. PairedStudent t tests were used to calculate the P values. For immuno-blots, relevant phosphorylation signals were quantified by den-sitometry and showed in Supplementary Tables. Statistical sig-nificance was assessed using two-tailed Student t tests anddescribed in the Table footnotes.

ResultsTSC2 loss in hepatocellular carcinoma cell lines conferseverolimus sensitivity

As part of a proteomic effort to comprehensively characterizeNovartis' cancer cell line collection known as the Cancer Cell LineEncyclopedia (CCLE; ref. 17), a RPPA platform was utilized tointerrogate the expression levels or phosphorylation status ofmore than 200 cancer-related proteins in a panel of more than400 cancer cell lines of various lineages. TSC2 protein levelsshowed a clear outlier pattern in HCC compared with all otherlineages and very low expression was observed in five of the nineHCC cell lines (Fig. 1A). The low TSC2 signal in those five HCCcell lines was unlikely to be due to any quality issue of the celllysate spotted onto the protein chip as the levels of other proteinsin these cell lines were normal (Supplementary Fig. S1A). Inaddition, these TSC2 low HCC cell lines tended to have elevatedmTOR signaling, as demonstrated by the trend of elevated phos-phorylation of downstream targets S6K1(T389), 4E-BP1(T37/T46), and S6(S240/S244; ref. 16). This observation is consistentwith the role of TSC2 as a negative regulator of mTOR signaling.

We then performed immunoblots with eight of the nine HCCcell lines used in the RPPA analysis. TSC2 protein in SNU-886,SNU-878, SNU-398, and Li-7 cells were essentially nondetectablewhile TSC2-low status for HuH-6 could not be confirmed (Fig.1B). Those four cell lines were therefore designated as TSC2-nullwhile JHH-7, SNU-761, HuH-7, and HuH-6 were designated asTSC2 wild-type. All eight HCC cell lines had comparable levels ofTSC1, the binding partner for TSC2. The phosphorylation of S6K1(T389) and 4E-BP1(T37/T46) showed a trend of elevation inTSC2-null cell lines compared with the TSC2 wild-type groupwhile the phosphorylation of AKT(S473) tended to be lower, withthe exception of HuH-7 (Fig. 1B; Supplementary Table S1). Asimilar pattern onphosphorylation of S6K1, 4E-BP1, andAKTwasobserved in the RPPA data (Supplementary Fig. S1A). This isconsistent with the previous observation that activation of mTORand S6K1 caused by TSC2 lesions results in the feedback inhibi-tion of the upstream insulin-like growth factor receptor 1(IGF1R)/PI3K signaling pathway (20). As the level of S6K1 and4E-BP1 phosphorylation inHuH-7 is comparable with that of theTSC2-null group, we hypothesized that it is due to activation ofthe PI3Kpathwayupstreamof TSC1/2. Indeed, treatment ofHuH-7 cells with the PI3K inhibitor BKM120 or AKT inhibitor MK-2206, but not the MEK inhibitor MEK162, reduced S6K1 phos-phorylation (Supplementary Fig. S1B). No PIK3CA mutation orPTEN loss was found inHuH-7 cells (data not shown), suggestingthat activation of mTOR in this cell line may be a result ofupstream activation at the level of growth factor receptors suchas IGF1R.

To investigate whether TSC2 loss rendered the HCC cell linessensitive to mTOR inhibition, we studied the antiproliferation

Huynh et al.

Mol Cancer Ther; 14(5) May 2015 Molecular Cancer Therapeutics1226

on June 9, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2015; DOI: 10.1158/1535-7163.MCT-14-0768

Page 4: Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in ...Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus Hung Huynh1, Huai-Xiang Hao2,

effect of everolimus on the eight HCC cell lines using a 3 DayCellTiter Glo (CTG) proliferation assay. While the everolimusIC50 values of three of the four TSC2 wild-type cell lines (exceptHuH-7) weremore than 2,000 nmol/L, the IC50 values of the fourTSC2-null cell lines ranged from 0.7 nmol/L to 25.1 nmol/L (Fig.1C; Supplementary Table S2). The maximal growth inhibition byeverolimus at 1 mmol/L was approximately 60% for the fourTSC2-null cell lines but only about 35% for the three TSC2wild-type cell lines except HuH-7 (Fig. 1C; Supplementary TableS2). TSC2wild-typeHuH-7 cellswere as sensitive as TSC2-null celllines to everolimus (IC50 ¼ 1 nmol/L and maximal growth

inhibition ¼ 66%), possibly due to the activation of mTOR byupstream PI3K signaling (Supplementary Fig. S1B). Consistentwith the IC50 values, we observed complete inhibition of S6K1phosphorylation by 2.5 nmol/L everolimus in all four TSC2-nullcell lines (Supplementary Fig. S1C). Taken together, TSC2 expres-sion level is likely to be the main determinant for mTOR activityand everolimus sensitivity in those HCC cell lines. Although wecannot fully rule out other lesions contributing to everolimussensitivity in the four TSC2-null cell lines, these results stronglysuggest that TSC2 loss is a highly penetrant predictive biomarkerfor everolimus sensitivity in HCC cell lines.

A1

0

−1

−2

−3

snu886

snu878

snu398

li7

huh6

Biliary

trac

t

Bone

Breas

t

Endom

etriu

m

Kidney

Larg

e int

estin

eLiv

erLu

ng

Esoph

agus

Ovary

Pancr

eas

Skin

Soft t

issue

Stom

ach

Thyro

id

Urinar

y tra

ct

Rel

ativ

e T

SC

2 pr

otei

n le

vel

CB

35%

JHH-7

2.0

1.8

1.6

1.4

1.2

1.0

0.8

0.6

0.4

40% 45% 50% 55% 60% 65%

0.2

Everolimus maximal inhibitionat 1 µmol/L (Amax)

SNU-761

HuH-6

HuH-7

SNU-886

Li-7

SNU-878

SNU-398

TSC2 wild-typeTSC2 null

Tubulin

p-S6K1(T389)

TSC2

JHH-7SNU-761

HuH-7HuH-6

SNU-886

SNU-878

SNU-398

Li-7

TSC1

S6K1

p-AKT(S473)

4E-BP1

p-4E-BP1(T37/T46)

IC50 P = 0.02 Amax P = 0.08

Eve

rolim

us IC

50 (

µmol

/L)

Figure 1.TSC2 loss in hepatocellular carcinoma cell lines confers everolimus sensitivity. A, RPPA analysis of TSC2 levels in 275 cancer cell lines of various lineages. Five of9 HCC cell lines (name displayed) showed significantly reduced TSC2 protein levels. B, immunoblot of eight HCC cell lines with indicated antibodies. TSC2was undetectable in four cell lines that expressed low TSC2 protein levels as measured by RPPA. p-S6K1(T389) and p-4E-BP1(T37/T46) levels tend to beelevated in these TSC2-null cell lines and HuH-7 cells. C, mTORC1 inhibitor everolimus IC50 (in mmol/L) and percentage of maximal inhibition by 1 mmol/L everolimus(Amax) in HCC cell lines as determined in a 3-day CellTiter-Glo (CTG) assay. Everolimus was significantly more potent and tended to be more efficacious inTSC2-null cell lines (red color) than in TSC2 wild-type cell lines (blue color), with the exception of HuH-7. P values were calculated by a two-tailed Student t test.Note that both axes do not originate at zero for better visualization of the data. The IC50 and Amax values are provided in Supplementary Table S2.

Frequent TSC2 Loss in HCC and Everolimus Response

www.aacrjournals.org Mol Cancer Ther; 14(5) May 2015 1227

on June 9, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2015; DOI: 10.1158/1535-7163.MCT-14-0768

Page 5: Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in ...Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus Hung Huynh1, Huai-Xiang Hao2,

TSC2 loss in patient-derived HCC xenografts predictseverolimus efficacy

We had previously established a large panel of patient-derivedHCCxenografts for testing the efficacy of various antitumor agents(21, 22). To determine whether TSC2 loss also occurred in thoseHCC xenografts, we performed immunoblots on lysates from 13randomly selected patient-derived HCC xenograft tumors. Asshown in Fig. 2A, four xenografts (HCC05-0411B, HCC19-1010, HCC14-0910, and HCC24-0309) showed no detectablelevels of TSC2 and thus were named as TSC2-null xenografts.Consistent with the immunoblot data from cell lines, basal p-S6K1 (T389) was significantly increased in these four TSC2-nulltumors (Fig. 2A; Supplementary Table S3). A trend of increased p-4E-BP1 (T70, functionally similar to phosphorylation at T37/T46shown in HCC cells lines) levels was also observed in three ofthese four TSC2-null tumors compared with the TSC2 wild-typetumors (Fig. 2A; Supplementary Table S3). Again, p-AKT (S473)levels were relatively high in TSC2 wild-type tumors but signif-icantly reduced in TSC2-null tumors, probably as a result offeedback inhibition of upstream IGF1R signaling by S6K activa-tion. Because p-AKT levels are regulated by PTEN, we thenexamined the PTEN levels and found two models with PTENloss (HCC26-1004 andHCC17-0211). Taken together, these datasuggest that TSC2 loss may occur in HCC tumors and that mTORsignaling is activated in TSC2-null xenografts.

To determine whether TSC2-null HCC tumors display sen-sitivity to everolimus, we tested everolimus in the four TSC2-null xenografts along with four TSC2 wild-type xenograft(HCC06-0606, HCC25-0809, HCC13-0109, and HCC13-0212) as determined by TSC2 immunoblot in Fig. 2A.Tumor-bearing mice were treated daily with either vehicle or1 mg/kg everolimus. Each treatment group involved 8 to 10independent tumor-bearing mice representing the same xeno-graft line. It is of note that the dose of everolimus used in thisstudy was approximately 2.5 to 5 times lower than doses usedin our previous reports (22) and the toxicity associated withdaily 1 mg/kg everolimus treatment was well tolerated. Asshown in Fig. 2B, everolimus exhibited potent antitumor activ-ities in all four TSC2-null xenografts and substantial tumorgrowth inhibition was observed as early as day 5 after dosing.The growth-inhibitory effects were particularly pronounced intwo of the four TSC2-null xenografts (HCC05-0411B andHCC14-0910). In contrast, the same treatment with everolimushad insignificant antitumor activities in two wild-type xeno-grafts (HCC06-0606 and HCC25-0809) and only modest anti-tumor activities in the other two TSC2 wild-type xenograftsHCC13-0109 and HCC13-0212 (Fig. 2B). These data suggestthat the antitumor activity of everolimus we observed was notdue to general toxicity and that everolimus was selectivelyefficacious in TSC2-null HCC tumors.

To further establish the link between TSC2 loss and everolimusefficacy in vivo, we screened another 15 patient-derived HCCmodels by immunoblot and identified 4 additional TSC2-nullxenografts (data not shown). The eight TSC2-null xenografts weidentified represent 28.6% of the 28HCC xenografts we screened.We next tested the antitumor activity of everolimus at 1 mg/kgdose in the four new TSC2-null models together with additionalTSC2 wild-type models. Efficacy of everolimus was evaluated bycomparing the finalmeanweight of tumors in the treatment arms(T) to those in the control arm (C; T/C ratio), with a value of <0.42considered as an active response (a widely adopted criteria from

the NCI). T/C ratios from the eight models shown in Fig. 2B werealso calculated. Among the 20 xenografts we tested (Fig. 2C),everolimus was considered to be active in 8 (36.4%) xenograftlines, namely HCC05-0411B (T/C ¼ 0.05), HCC01-0207 (T/C ¼0.09), HCC14-0910 (T/C ¼ 0.12), HCC19-1010 (T/C ¼ 0.14),HCC25-0705A (T/C ¼ 0.14), HCC24-0309 (T/C ¼ 0.25),HCC24-1005 (T/C ¼ 0.27), and HCC5-1318(3) (T/C ¼ 0.34).Strikingly, all eight everolimus-sensitive models were TSC2-null.TSC2 wild-type models modestly responded to everolimus withT/C ratios ranging from0.44 to 0.74, indicating the important roleof mTOR signaling to HCC tumor growth even in the TSC2 wild-type setting.With 8 of 8 TSC2-null models and 0 of 12 TSC2wild-type models as everolimus responders, this enrichment is statis-tically significant at P < 0.01 (Fisher exact test value ¼ 8E�06).Interestingly, the twomodelswithPTEN loss only showedmodestresponses to everolimus (T/C¼ 0.40 for HCC26-1004 and T/C¼0.61 for HCC17-0211), suggesting that PTEN loss may not be asgood in predicting everolimus response in HCC models as TSC2loss. This remarkable enrichment of TSC2-null models in ever-olimus responders from a relatively large study (20 models withdistinct mutation profiles as shown in Supplementary Table S4)strongly argues that TSC2 loss in HCC leads to hyperactivation ofthe mTOR signaling pathway and robustly predicts everolimusresponse in xenograft models.

Tumorgrowth inhibitionby everolimuswas through inhibitionof mTOR signaling

As shown in Fig. 2B, the growth-inhibitory effects were partic-ularly pronounced in TSC2-null xenograft HCC14-0910 andHCC05-0411B at the dose of 1 mg/kg everolimus. To determinethe degree and dose-dependent effect of everolimus, we tested itsantitumor activity at the doses of 0.25, 0.5, 0.75, and 1 mg/kg inthose twomodels. As shown in Fig. 3A, tumor stasis was achievedat 0.75mg/kg everolimus and there was a dose-dependent growthinhibition by the four doses of everolimus in HCC14-0910.Similar results were observed in model HCC05-0411B (data notshown).

We then performed pharmacodynamic analysis by immu-noblot at the end of the efficacy study. As shown in Fig. 3B,everolimus caused a dose-dependent decrease in p-S6K1(T389) and p-4E-BP1 (T70), indicating successful mTOR inhi-bition in TSC2-null xenograft HCC14-0910. Similar to previousstudies (22), everolimus treatment led to a dose-dependentelevation of p-AKT (S473), probably as a result of relieving thefeedback inhibition of IGF1R by S6K1 activation. Everolimustreatment also significantly reduced the phosphorylation ofCDK2 (T14/T15) and induced p27 accumulation, both ofwhich are well-established markers of cell-cycle arrest. Ourdata are in agreement with previous studies showing thatinhibition of mTOR leads to G1–S cell cycle arrest throughincreased levels of p27 (23). In addition, as in our previousstudy (22), the antiangiogenic effect of everolimus, measuredby microvessel density, was also observed in HCC14-0910 andother TSC2-null HCC xenografts (data not shown).

TSC2 loss in HCC is caused predominantly by TSC2 deletionsand mutations

To understand the molecular mechanism responsible for TSC2loss in TSC2-null HCC cell lines, we examined the TSC2 mRNAlevels by TaqMan RT-PCR assay. TSC2 mRNA levels in all fourTSC2-null cell lineswere less than5%of that of JHH-7 cells, which

Huynh et al.

Mol Cancer Ther; 14(5) May 2015 Molecular Cancer Therapeutics1228

on June 9, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2015; DOI: 10.1158/1535-7163.MCT-14-0768

Page 6: Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in ...Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus Hung Huynh1, Huai-Xiang Hao2,

A

TSC2

p-S6K1 (T389)

S6K1

p-4E-BP1(T70)

4E-BP1

p-AKT(S473)

AKT

Tubulin

HCC14-0

910

HCC19-1

010

HCC05-0

411B

HCC26-1

004

HCC17-0

211

HCC26-0

808A

HCC25-0

809

HCC24-0

309

HCC06-0

606

HCC21-0

208

HCC13-0

109

HCC13-0

212

HCC01-0

909

PTEN

C

Eve

rolim

us e

ndpo

int T

/C TSC2 nullTSC2 wild-type

Total models=20

HCC06-0606

HCC25-0809

HCC26-0808A

HCC 10-0505

HCC17-0211

HCC01-0909

HCC19-0711

HCC13-0109

HCC 01-0708

HCC02-0310

HCC26-1004

HCC 07-0409

HCC5-1318(3)

HCC24-1005

HCC24-0309

HCC25-0705A

HCC19-1010

HCC14-0910

HCC01-0207

HCC05-0411B0.0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

PTEN null

TSC2 wild-type vs. TSC2 null, P<0.01

B

Tum

or v

olum

e (m

m3 )

Days of treatment

TS

C2

null

12

HCC06-0606

1086420

500

1,000

1,500

2,000

2,500

3,000

Vehicle1 mpk EVE

TS

C2

wild

-typ

e

HCC25-0809

1614121086420

500

1,000

1,500

2,000

2,500

3,000

Vehicle1 mpk EVE

HCC13-0109

121086420

500

1,000

1,500

2,000

2,500

3,000

Vehicle1 mpk EVE

HCC13-0212

121086420

500

1,000

1,500

2,000

2,500

3,000

Vehicle1 mpk EVE

181614

HCC19-1010

121086420

500

1,000

1,500

2,000

2,500

3,000

Vehicle1 mpk EVE

HCC05-0411B

14121086420

500

1,000

1,500

2,000

2,500

3,000

Vehicle1 mpk EVE

HCC24-0309

1614121086421

500

1,000

1,500

2,000

2,500

3,000

Vehicle1 mpk EVE

HCC14-0910

14121086420

500

1,000

1,500

2,000

2,500

3,000

Vehicle1 mpk EVE

Days of treatmentDays of treatmentDays of treatment

Days of treatmentDays of treatmentDays of treatmentDays of treatment

Tum

or v

olum

e (m

m3 )

P<0.01P<0.01P<0.01P<0.01

P<0.01P<0.01P=0.86P=0.51

Figure 2.TSC2 loss in patient-derived HCC xenografts predicts everolimus efficacy. A, levels of TSC2, p-S6K1(T389), p-4E-BP1(T70) and p-AKT(S473) in patient-derivedHCC xenografts. Indicated HCC tumors were subcutaneously implanted in SCID mice. Tumors were collected when they reached 800–1,000 mm3 and processedfor immunoblotting. Note that HCC05-0411B, HCC19-1010, HCC14-0910, and HCC24-0309 xenografts had undetectable levels of TSC2 protein and increasedphosphorylation of mTOR substrate S6K1. B, effects of 1 mg/kg everolimus on growth rates of HCC xenografts. SCIDmice bearing the indicated tumors were treatedwith vehicle or 1 mg/kg everolimus. Each treatment arm involved 8 to 10 independent tumor-bearing mice representing the same xenograft line. Mean tumorvolume is shown and error bars represent SEM. The experiments were repeated at least twice with similar results. P values were calculated by paired Student t testsfor the vehicle group versus the everolimus treatment group. � ,P<0.01 andP values of the lastmeasurement are shown.Note that tumor stasis or regressionwas onlyobserved in TSC2-null xenografts. C, bar graph showing endpoint HCC tumor weight T/C (treated vs. control) at 1 mg/kg everolimus dosing. SCID mice bearingindicated tumors were treated with vehicle or 1 mg/kg everolimus daily for 14–18 days. Each treatment arm involved 8 to 10 independent tumor-bearing micerepresenting the same xenograft line. Efficacy of everolimus was evaluated by comparing the final mean weight of tumors in the treated arms (T) to those in thecontrol arm (C; T/C ratio). Note that the 8 best responders to everolimuswere TSC2-null xenografts and this enrichmentwas statistically significant atP<0.01 (Fisherexact test value ¼ 8E�06). � , models with PTEN loss as determined by immunoblot in A.

Frequent TSC2 Loss in HCC and Everolimus Response

www.aacrjournals.org Mol Cancer Ther; 14(5) May 2015 1229

on June 9, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2015; DOI: 10.1158/1535-7163.MCT-14-0768

Page 7: Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in ...Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus Hung Huynh1, Huai-Xiang Hao2,

had the highest level of TSC2 mRNA (Supplementary Fig. S2A).The TSC2 mRNA levels in the other three TSC2 wild-type lines(SNU-761, HuH-7, and HuH-6) were approximately 50%,57%, and 26% of that in JHH-7 cells, respectively. The mRNAlevels of TSC1 were comparable in all cell lines. The extremelylow levels of TSC2 mRNA in TSC2-null cell lines prompted usto examine the status of TSC2 gene in these cell lines. As shownin Table 1, SNU-886 and SNU-878 cells harbored TSC2 non-sense mutations (R1032X and S1514X, respectively) and TSC2wild-type alleles were lost as indicated by the near 100%mutant allele frequency. SNU-398 cells contain a focal deletionin TSC2 from exon 9 to exon 36. No mutation or deletion inTSC2 was detected in Li-7 cells, raising the possibility that theabsence of TSC2 mRNA in this cell line may be due to epige-netic silencing of TSC2. The TSC2 promoter has been found tobe methylated in several cancer types, including breast cancerand oral squamous cell carcinoma (24, 25), and DNA meth-ylation abnormalities have been shown to be a common lesionin HCC cell lines, including Li-7 (26). However, we were unableto observe a robust re-expression of TSC2 transcripts withtreatment of the DNA methylase inhibitor decitabine and/orHDAC inhibitor trichostatin (TSA) while another known meth-ylated geneMZB1 was successfully induced by those treatments(Supplementary Fig. S2B). Although suggestive, this negativeresult does not completely rule out the possibility of TSC2epigenetic silencing in Li-7 cells.

To determine whether TSC2 mutations and deletions are sim-ilarly responsible for TSC2 loss in HCC xenografts as in HCC celllines, we analyzed the exons of both TSC1 and TSC2 (togetherwith 294 other cancer-related genes) using next-generationsequencing. As shown in Table 1, among the eight TSC2-nullHCCmodels we previously identified, TSC2 deletionswere foundin model HCC19-1010 and model HCC24-1005. In addition, aTSC2 splicing mutation c.1444-2A>T was found in modelHCC14-0910 at 100% allele frequency and model HCC24-

0309 contained a G305V missense mutation at 99% allele fre-quency. The impacted glycine residue resides within the TSC1binding domain of TSC2 (amino acid 1–418) and is highlyconserved from mouse to man. To test the functional conse-quence of the G305V mutation, we expressed the TSC2 G305Vmutant by transient transfection of HEK293T cells and evalu-ated its relative expression at both the mRNA level and theprotein level. As shown in Supplementary Fig. S2C, the mRNAlevel of G305V mutant was only about 30% that of wild-typeTSC2 mRNA when equal amount of plasmids were transfected,suggesting that the missense mutation may cause reducedmRNA expression. Importantly, there was no detectableG305V-mutant TSC2 protein by immunoblot regardless of itsmRNA levels (Supplementary Fig. S2D), suggesting that theG305V mutant protein is unstable.

For the four TSC2-null xenografts in which no TSC2 geneticlesions were identified by sequencing, we performed TSC2multiplex ligation-dependent probe amplification (MLPA).This PCR-based assay measures the copy number of each exonof the TSC2 gene and its neighboring regions and provides abetter resolution of TSC2 copy number than the broad sequenc-ing method. Indeed, loss of two upstream regions that possiblycontain the TSC2 promoter and duplication of exon 16–23were identified in HCC5-1318(3) and HCC05-0411B, respec-tively (Table 1). Exon duplication of TSC2 has been foundin TSC patients and the duplicated exons may disrupt thereading frame of TSC2. All together, we have identified geneticmechanisms for TSC2 loss in 6 of the 8 TSC2-null modelsshown in Fig. 2C.

It is known that a xenograft tumor may undergo genetic driftwhen compared with the original tumor and that certain geneticlesions may be enriched for during the model establishment andpassaging process. To validate that TSC2 genetic lesions exist inprimary HCC tumors, we sequenced DNA samples from 13randomly selected Hong Kong HCC biopsies using the same T5

A

Days of treatment

Vehicle

0.25 mpk everolimus

0.50 mpk everolimus

0.75 mpk everolimus

1.00 mpk everolimus

Tum

or v

olum

e (m

m3 )

0151050

500

1,000

1,500

2,000

2,500

BEverolimus

(mg/kg):p-S6K1(T389)

S6K1

p-4E-BP1(T70)

4E-BP1

p-AKT(S473)

AKT

p-CDK2(T14/15)

p27

0.75 0.50.250

Figure 3.Tumor growth inhibition by everolimus was through inhibition of mTOR signaling. A, dose-dependent antitumor effects of everolimus in TSC2-null xenografts.Mice bearing TSC2-null xenograft HCC14-0910 were randomized into 5 groups (8 mice per group) for daily dosing of 0.25, 0.5, 0.75, or 1 mg/kg everolimus orvehicle. The experimental procedure and data are displayed similarly as in Fig. 2B. P values were calculated by paired Student t tests for the vehicle groupversus the everolimus treatment group at 0.25 and 0.50 mg/kg everolimus doses. � , P < 0.01. B, dose-dependent effects of everolimus in TSC2-null xenograftHCC14-0910 on decreasing the phosphorylation of mTOR substrates (S6K1 and 4E-BP1) and inducing cell-cycle arrest (CDK2 phosphorylation and p27 induction).Two hours after the last dose in A mice were sacrificed and lysates from vehicle- and everolimus-treated tumors were subjected to immunoblots with theindicated antibodies. Representative blots of two tumors for each dose are shown and the experiment was repeated at least twice with similar results.

Huynh et al.

Mol Cancer Ther; 14(5) May 2015 Molecular Cancer Therapeutics1230

on June 9, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2015; DOI: 10.1158/1535-7163.MCT-14-0768

Page 8: Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in ...Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus Hung Huynh1, Huai-Xiang Hao2,

panel at Foundation Medicine. As shown in Table 1, a TSC2splicing mutation and a TSC2 deletion were identified in sampleB253 and B266, respectively. Sample B277 contained a pointmutation at 92% allele frequency resulting in an A1141T aminoacid change. This mutation was also found in Taiwanese TSCpatients (27) and therefore it likely inactivates TSC2. In addition,the majority of disease-associated mutations of TSC2 result in asubstantial decrease in the TSC2 protein level (28, 29). In sum-mary,TSC2mutations anddeletions exist in primaryHCC tumors.

TSC2 immunohistochemistry assay identified TSC2 loss inprimary HCC tumors

As not all TSC2-null cell lines and xenografts had mutations ordeletions in theTSC2 gene, it raised thepossibility that nongeneticmechanisms such as epigenetic silencing may be responsible forTSC2 loss in HCC. It might also be challenging to predict thefunctional consequence ofTSC2missensemutations as in the caseof the G305V mutation we identified. Furthermore, for tumorsuppressor genes like TSC2, loss of heterozygosity or extremelyhighmutant allele frequency is needed to predict loss-of-functionat the protein level. To overcome these limitations of the sequenc-ing approach, we developed a TSC2 IHC assay to identify TSC2loss in HCC tumors. We first validated the IHC assay using TSC2wild-type and TSC2-null HCC cell lines. As shown in Supple-mentary Fig. S3A, intense stainingwas observed in TSC2wild-typeHuH-6 cells and little signal was detected in TSC2-null SNU-398cells, suggesting that the assay is robust and specific. We furthervalidated the IHC assay using patient-derived HCC xenografts,which aremore heterogeneous than the cell lines. Again, therewasstrong staining in TSC2wild-type xenograftHCC06-0606but veryfaint staining in TSC2-null xenograft HCC-24-1005 (Supplemen-tary Fig. S3B).

We then used the TSC2 IHC assay to address whether TSC2 lossin our xenograft models occurred in primary tumors or wasacquired during propagation in the mice. As shown in Supple-mentary Fig. S3C, TSC2was not detected in the four primaryHCCtumors from which the TSC2-null xenografts HCC05-0411B,HCC19-1010, HCC14-0910, andHCC24-0309were derived. TheTSC2 staining in primaryHCC sections was from adjacent normalliver tissues and served as positive controls. These data stronglyargue that the TSC2 loss observed in our HCC xenografts was afaithful representation of the primary tumors from which theywere derived and that our IHC assay could robustly identify TSC2loss in HCC clinical samples.

HCC patients with TSC2-null/low tumors benefited fromeverolimus treatment

EVOLVE-1 was a double-blind, randomized, placebo con-trolled (2:1 everolimus vs. placebo), multicenter phase III studyto evaluate the efficacy and safety of everolimus in adult patientswith advanced HCC who had progressed on or after sorafenibtreatment, or were intolerant of sorafenib treatment. The trial didnot meet its primary endpoint of improving OS by everolimusover placebo. The median OS for the everolimus group andplacebo group were 7.56 months and 7.33 months, respectively(HR: 1.05; 95% CI: 0.86-1.27; P¼ 0.675; ref. 15). To test whetherpatientswithnoor lowTSC2expression in their tumors had anOSbenefit from everolimus treatment, we quantified TSC2 expres-sion in tumor samples from the EVOLVE-1 patients using our IHCassay and an H-score scoring system as described in the Materialsand Methods. Evaluable TSC2 IHC results were obtained for 139EVOLVE-1 patients for whose archival formalin-fixed paraffin-embedded tumor samples were available. We defined 15 tumorswith H-scores ranging from 0 to 120 (Table 2) as TSC2-null/low.The 120 H-score cutoff was partially guided by the H-scores ofTSC2-null patient-derived HCC xenografts (ranging from 0 to135, data not shown). These included 8 samples (5.8%) withminimal TSC2 expression (H-score¼ 0-60) and 7 samples (5.0%)with low TSC2 expression (H-score ¼ 61–120). The seven TSC2-low samples had only some or universal weak staining (1þ). Twotumors with H-score of 90 and 120, respectively, but had sub-stantial (40%) 2þ or even some 3þ intensity staining wereannotated as TSC2 wild-type.

The treatment assignment, overall survival (OS), and demo-graphic features of the 15 patients with TSC2-null/low tumors arelisted in Table 2. The patients treatedwith everolimus (n¼ 10) hadlonger OS than those treated with placebo (n ¼ 5). Six of the 10TSC2-null/low patients treated with everolimus (patient 10–15)hadOS ranging from 9.53 to more than 32.72months, comparedwith the 7.56months medianOS of the overall everolimus group.The two patients with very short OS (patient 6 and patient 7) wereon treatment for only2weeks anddiscontinued fromthe studydueto either complications of the underlying cirrhosis or intolerableadverse events. Another patient with a short survival of 4.63months (patient 8) only received treatment for 6 weeks anddiscontinued due to disease progression. In contrast, 4 of the 5TSC2-null/low patients treated with placebo hadOS ranging from1.25 to 5.59 months, much shorter than the 7.33 months medianOS of the overall placebo group. These findings may suggest an

Table 1. TSC2 deletions and mutations identified in TSC2-null HCC cell lines, xenografts, and primary tumors

Sample name Sample type TSC2 mutation Allele frequency Comments

SNU-886 Cell line p.R1032X 97%SNU-878 Cell line p.S1514X 100%SNU-398 Cell line Deletion — Exon 9-36 deletedHCC14-0910 Xenograft Splicing 100% c.1444-2A>T_p.spliceHCC19-1010 Xenograft Deletion —

HCC24-0309 Xenograft p.G305V 99% Within TSC1 binding domainHCC24-1005 Xenograft Deletion — 28 exons deletedHCC5-1318(3) Xenograft p.S211Y 19 2 upstream exon deleted (MLPA)HCC05-0411B Xenograft Deletion — Exon 16-23 duplication (MLPA)B253 Tumor Splicing 54% c.1443þ2_þ13del_p.spliceB266 Tumor Deletion — Exons 41 of 41 deletedB277 Tumor p.A1141T 92% Reported in TSC patients

NOTE: TSC2mutations and deletions in HCC cell lines fromSouth Koreawere identified by targeted sequencing of about 2,000genes and SNP6.0 array copy numberanalysis, respectively. TSC2mutations and deletions in most Singapore HCC xenografts and in all Hong Kong primary HCC tumors were identified by targeted DNAsequencing at Foundation Medicine, Inc. TSC2 deletions in HCC5-1318(3) and HCC05-0411B were identified by Multiplex Ligation-dependent Probe Amplification(MLPA, noted in the table). Mutant vs. wild-type allele frequencies are provided for TSC2 mutations.

Frequent TSC2 Loss in HCC and Everolimus Response

www.aacrjournals.org Mol Cancer Ther; 14(5) May 2015 1231

on June 9, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2015; DOI: 10.1158/1535-7163.MCT-14-0768

Page 9: Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in ...Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus Hung Huynh1, Huai-Xiang Hao2,

association of TSC2 loss/lowwithmore aggressive disease and thatTSC2 status may have prognostic value, consistent with a recentreport (30). The only patient with an OS of more than 18.23months was on placebo for only 6 months and after diseaseprogression received an ALK1 inhibitor (PF-03446962) for 2months, followed by a MET inhibitor. Although the sample sizeof TSC2-null/low patients is too small to perform formal statisticalanalysis, the observedOS benefit from everolimus in patients withTSC2-null/low tumors is consistentwithour hypothesis that TSC2-null tumors are sensitive to mTOR inhibition.

TSC2 loss is more common in Asian HBVþ HCC than inCaucasian HCC

The frequency of TSC2 loss in EVOLVE-1 samples (5.8%–

10.8%, Fig. 4A) is much lower than that in Singapore HCCpatient-derived xenografts (28.6%, 28models) and inHongKongprimary HCC (23.1%, 13 tumors). This is likely due to theheterogeneity of the ethnic makeup of the EVOLVE-1 patients(55% Caucasian, 35% Asian, and 10% others). While the overallprofile of the 139 patients with TSC2 IHC results reflected thisethnic diversity, the percentage of Asian in the 15 TSC2-null/lowtumor patients increased to 53% (Supplementary Fig. S4A). Thisraised the possibility that the prevalence of TSC2 loss may vary byethnicity. To investigate this, we segmented the TSC2 H-scoresinto three groups: 0–60 (likely to be TSC2-null), 61–120 (TSC2-low), and 121–300 (TSC2 wild-type), and compared the H-scoredistribution of each race in the 139 EVOLVE-1 samples. As shownin Fig. 4A, the percentage of TSC2-null/low in the 48 Asian HCCsamples is 18.7%, about 3 times of that of the 78 Caucasian HCCtumors (6.4%). There were only 13 tumors in the group of otherethnicity, which was too few for ameaningful frequency estimate.The racial composition in each of the three TSC2 H-score seg-ments is shown in Supplementary Fig. S4B and the ratio of Asianto Caucasian increased significantly from the high to the low H-score segments. Taken together, these epidemiology data suggestthat TSC2 loss in HCC is more common in Asian than inCaucasian population.

To obtain a more accurate frequency of TSC2 loss in the AsianHCC population, we analyzed additional 322 Asian HCC tumorsof which greater than 90% were HBVþ in the tumor microarray(TMA) format. The H-score range distribution in 239 evaluablecases is shown in Fig. 4B. There were 20.0% samples with an H-score below 60 and 19.7% samples had an H-score from 61 to

120, indicating minimal to low TSC2 expression. Therefore, weestimated TSC2 loss to be at least 20% in Asian HBVþ HCC.Consistentwith theHBV association, sevenof the eight EVOLVE-1TSC2-null/low Asian HCC tumors were HBVþ (Table 2).

DiscussionOnemajor factor limiting the effective treatment of HCC is the

high degree of tumor heterogeneity. The same treatment canproduce remarkably different responses ranging from completeresponse to disease progression in patients with indistinguishabletumors at the histopathologic level (31). Understanding themolecular factors that may predict patient benefits derived fromnovel targeted therapies will be crucial for the successful devel-opment of HCC treatments. Despite the high frequency of mTORactivation in HCC, the EVOLVE-1 phase III trial showed essen-tially no OS benefit by everolimus in unselected HCC patients(15). The fact thatmTOR inhibition was not sufficient to suppresstumor growth in many HCC patients suggests that mTOR depen-dence cannot solely be predicted by general mTOR activation.Rather, specific lesions in mTOR pathway components such asPTEN or TSC1/TSC2 are more likely to be useful in identifyingHCC patients who will most likely benefit from mTORC1 inhi-bitors. In the current study, we demonstrated the remarkableprediction of antigrowth effect of everolimus by TSC2 loss inboth HCC cell lines and patient-derived xenografts. More impor-tantly, the analysis of a subset of EVOLVE-1 patients suggests thatthose with TSC2-null/low tumors indeed benefited from ever-olimus treatment. These clinical data extended our preclinicalcorrelations to human and support the utility of TSC2 loss as aHCC patient selection biomarker for mTORC1 inhibitors, asillustrated in Supplementary Fig. S5.

The molecular mechanisms responsible for TSC2 loss in HCCare likely diverse. The TSC2 deletions andmutations we identifiedinHCC cell lines, xenografts andprimary tumors provided geneticexplanations for themajority of TSC2 loss we observed. However,TSC2-null HCC cell line Li-7 and TSC2-null xenografts (HCC25-0705A and HCC01-0207) do not harbor any genetic lesions inTSC2 gene, suggesting the existence of nongenetic mechanisms ofTSC2 loss in those HCC samples. It has been reported thatpromoter methylation was responsible for silencing of TSC2 genein other cancer types (24, 25). In addition, posttranslationalmechanisms such as phosphorylation and acetylation could lead

Table 2. Patient profile, treatment arm and survival outcome of 15 EVOLVE-1 trial HCC patients with TSC2-null/low tumors by IHC

Patient ID TSC2 IHC H-score Randomized treatment OS (months) Race Gender HBV

1 60 Placebo 1.25 Asian M þ2 100 Placebo 1.35 Caucasian M —

3 120 Placebo 3.61 Other M N.A.4 120 Placebo 5.59 Asian M þ5 90 Placebo 18.23þ Asian M þ6 50 Everolimus 0.76 Other M —

7 50 Everolimus 2.27 Other M —

8 40 Everolimus 4.63 Asian M þ9 110 Everolimus 5.85 Asian M þ10 100 Everolimus 9.53 Caucasian F —

11 10 Everolimus 9.56 Asian M þ12 0 Everolimus 18.96 Caucasian M —

13 40 Everolimus 25.00þ Asian M þ14 0 Everolimus 28.02 Asian M —

15 100 Everolimus 32.72þ Caucasian M —

NOTE: TSC2 status in the tumorswas determined by TSC2 IHC performed at PhenoPath Laboratories. Note that patient 6, 7, and 8were only treated with everolimusfor 2–6 weeks due to various reasons described in the article.

Huynh et al.

Mol Cancer Ther; 14(5) May 2015 Molecular Cancer Therapeutics1232

on June 9, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2015; DOI: 10.1158/1535-7163.MCT-14-0768

Page 10: Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in ...Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus Hung Huynh1, Huai-Xiang Hao2,

to TSC2 loss. TSC2 can be phosphorylated by AKT, ERK, AMPK,and GSK3ß at distinct sites and these phosphorylation eventsinactivate TSC2 (16). Both AKT- and ERK-dependent phosphor-ylation of TSC2 have been shown to disrupt its interaction withTSC1 and destabilize TSC2 (32, 33). Recently, arrest defectiveprotein 1 (ARD1) was shown to acetylate and stabilize TSC2 andloss of heterozygosity (LOH) of ARD1 has been observed inbreast, lung, pancreatic, and ovarian cancer samples (34). It isalso noteworthy to point out that TSC1 inactivation can lead toTSC2 protein degradation. Although no TSC1 mutations wereidentified in any of our HCC samples, a TSC1mutation has beenidentified by high-resolution characterization of one hepatocel-lular carcinoma genome (35). The exact mechanisms of nonge-netic loss of TSC2 in HCC remain to be determined.

In agreement with our findings, mutations in TSC1/2 as puta-tive biomarkers that predict efficacy of mTORC1 inhibitors haveemerged recently from whole-genome or -exome sequencinganalyses of tumor samples from patients who had experiencedextraordinary benefit from mTORC1 inhibitor treatments. Solitand colleagues showed that TSC1 loss-of-function mutationswere present in one metastatic bladder cancer patient with adurable (>2 years) complete response and four with partialresponse or stable disease after receiving everolimus, but in only1 of 9 patients with disease progression (36). Similarly, Voss andcolleagues found inactivating mutations in TSC1 or TSC2, oractivating mutations in MTOR, in 4 of 6 renal cell carcinoma

patients who had durable response to either everolimus or tem-sirolimus, but not in patients who progressed rapidly on the sametreatments (37). Recently,TSC2 (Q1178X) loss-of-functionmuta-tion was identified in one thyroid cancer patient who achieved acomplete response that lasted for 18 months when treated witheverolimus (38). Despite lacking placebo arms for comparison inthose studies, the remarkable clinical efficacy of mTORC1 inhi-bitors in distinct types of TSC1/TSC2-mutated tumors (bladder,kidney, thyroid, and liver cancer) suggests a strong correlationbetween TSC1/TSC2 loss-of-function and clinical efficacy ofmTORC1 inhibitors such as everolimus. This link is consistentwith the preclinical observation that the loss of TSC complex gaverise to high levels of constitutive mTOR activation and is notsurprising given the large number of signaling inputs regulatingmTOR through the TSC1/TSC2 complex (16). Interestingly, theprevalence of mutations in TSC1 versus TSC2 seems to varyamong tumor types.

In this study, we also developed a TSC2 IHC assay to be usedwith primary HCC samples in the clinic. The IHC approach issuperior to the sequencing assay for the cases of missense TSC2mutationswith unknown functional consequences and it can alsocapture TSC2 loss by all types of mechanisms. However, as for allIHC assays, it was challenging to define the H-score cutoff forTSC2-null HCC tumors. We used H-score of 60 and 120 as thecutoff for TSC2-null and TSC2-low tumors, respectively. The H-score cutoff for TSC2-null or low need to be better defined by

B

A

Per

cent

age

of H

-sco

re r

ange

0

10

20

30

40

50

60

70

80

90

100

Caucasian

(n = 78)

Asian

(n = 48)

Other

(n = 13)

1.3%5.1%

93.6%

15.4%

81.3%

8.3%10.4%

76.9%

7.7%

All samples

(n = 139)

5.8%

87.7%

6.5%

0–60

61–120

121–300

Per

cent

age

in A

sian

HC

C (

n =

239

)

TSC2 IHC H-score range

0

10

20

30

40

50

60

70

80

90

100

121–30061–1200–60

20.1% 19.7%

60.2%

Figure 4.TSC2 loss is more common in AsianHBVþ HCC than in Caucasian HCC.A, TSC2 IHC H-score range distributionin 139 HCC tumors from the EVOLVE-1phase III clinical trial. H-scores(0–300) were divided into threegroups: 0–60 (likely to be TSC2-null),61–120 (TSC2 low), and 121–300 (TSC2wild-type). The percentage of eachrange is marked at the top of the bars.The H-score distribution of each ethnicgroup in the 139 samples is alsodisplayed. B, TSC2 IHC H-score rangedistribution in 239 Asian HCC caseswith evaluable IHC staining usingduplicate-core TMA. The percentageof each range is marked at the top ofthe bars. More than 90% of thesetumors are HBVþ.

Frequent TSC2 Loss in HCC and Everolimus Response

www.aacrjournals.org Mol Cancer Ther; 14(5) May 2015 1233

on June 9, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2015; DOI: 10.1158/1535-7163.MCT-14-0768

Page 11: Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in ...Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus Hung Huynh1, Huai-Xiang Hao2,

correlating with TSC2mutations, TSC2 levels in immunoblot andmore importantly, everolimus clinical response in more patients.

In this study, we identified 15 TSC2-null/low tumors from139 EVOLVE-1 samples. By analyzing the patient ethnicity ofthose samples, we found that TSC2 loss is more common inAsian than in Caucasian HCC. The cause of such ethnic differ-ence in TSC2 loss is unknown but may be related to the distinctetiologies of Caucasian HCC (HCV and alcohol) and AsianHCC (HBV). Additional epidemiology studies in non-Asianpopulations using our IHC assay are warranted and may betterinform the broad utility of TSC2 loss as a patient selectionbiomarker in HCC.

TSC2 mutations were not reported in recent next-generationsequencing studies of HCC (39–42) and there is only one reportfinding a TSC1 mutation in HCC (35). The different patientethnicity (Caucasian HCC) and HCC etiology (HCVþ for JapanHCC) in those studies may account for the differences betweenthose and our results. In fact, the HCC mutation spectrums inthose papers significantly differed from each other. For thosestudies with Asian HBVþ HCC samples, only a small discoveryset was subjected to whole-genome or -exome sequencing andTSC2mutations may not be represented in their initial discoveryset. Finally, the targeted exon sequencing assay used inour study ismore sensitive than the whole-genome or -exome sequencingmethod for a particular gene. The sequencing depth at TSC2 locusin our assay (median exon coverage ranging from 496 to 1043) ismuch higher than that expected from the whole-genome or-exome sequencing.

Previously, the challenge of using TSC1/TSC2 lesions as patientselectionbiomarkerswas that somaticTSC1/2mutations in cancerare generally rare. TSC1/2mutations are estimated to be less than2% in several major cancer types, including kidney cancer, basedonTCGA(TheCancerGenomeAtlas) studies andother large-scaledeep sequencing efforts (36, 43). Only recently, bladder cancerwas found to have a high frequency of PI3K–mTOR pathwayalterations with approximately 8% TSC1 mutations and approx-imately 2% TSC2 mutations (44). Identifying tumor types withreasonable frequency of TSC1/2mutations will enable TSC statusto be used as a biomarker in the clinic for practical reasons.On thebasis of our TSC2 IHC survey of 239 Asian HCC tumors, thefrequency of TSC2 loss is estimated to be around 20% in AsianHBVþ HCC. China alone accounts for more than 50% of theworld's HCC cases and about 80% of HCC cases in China areHBVþ (45). Therefore, Asian HBVþ HCC represents nearly half ofthe total cases of HCCworldwide. This epidemiology data strong-ly support TSC2 loss as a potential patient selection biomarker forAsian HBVþ HCC likely to benefit from everolimus therapy andfurther exploration of the prognostic value of TSC2 loss.

If the high frequency of TSC2 loss can be confirmed and itsprognostic significance can be established in larger and morediverse Asian HCC populations, an everolimus clinical trial

focused on the Asian patients will be justified andwe recommendTSC2 loss defined by IHC as the patient enrollment criterion. Likeother predictive biomarkers, it will always be certain that not allTSC2-null tumors will respond to everolimus treatment. It will beimportant to examine the additional genetic lesions or clinicalfeatures of TSC2-null tumors to enhance the predicating speci-ficity of this biomarker and to enable drug combinations witheverolimus to achieve better antitumor efficacy, such as PI3Kinhibitors as suggested by a previous report (46). In addition,TSC lesion is unlikely to explain all cases of everolimus responsein HCC patients and other predictive biomarkers for everolimusresponders, such as activating mutations in MTOR or RHEB,remain to be identified.

Disclosure of Potential Conflicts of InterestS.L. Chan is a consultant/advisory board member for Novartis. D. Sellami

has ownership interest in Novartis stocks. R. Schlegel and A. Huang haveownership interest in the Novartis Institutes for BioMedical Research stocks.No potential conflicts of interest were disclosed by the other authors.

Authors' ContributionsConception and design: H. Huynh, H.-X. Hao, D. Chen, D. Sellami, A. HuangDevelopment of methodology: D. Chen, D. Yang, M.R. Palmer, B.H. LeeAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): H. Huynh, H.-X. Hao, S.L. Chan, D. Chen, R. Ong,D.A. Ruddy, M.N. Balak, B.H. Lee, D. Sellami, A.X. ZhuAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis):H.Huynh,H.-X.Hao,D. Chen, R.Ong, P. Pochanard,D. Yang, D.A. Ruddy, M. Liu, A. Derti, M.N. Balak, M.R. Palmer, B.H. Lee,A.X. Zhu, A. HuangWriting, review, and/or revision of the manuscript:H. Huynh, H.-X. Hao, S.L.Chan, D. Chen, D.A. Ruddy, A. Derti, M.R. Palmer, B.H. Lee, D. Sellami,A.X. Zhu, R. Schlegel, A. HuangAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): H.-X. Hao, K.C. Soo, P. Pochanard, M. LiuStudy supervision: H. Huynh, H.-X. Hao, D. Sellami, A.X. Zhu, R. Schlegel,A. Huang

AcknowledgmentsThe authors thank Jinsheng Liang, Xiaoyan Li, Yan Chen, and Angela Tam for

technical assistance. The authors also thank Hui-Qin Wang, Fang Li, QingSheng, Ensar Halilovic, JohnMonahan, and Joseph Lehar for advice and helpfuldiscussions.

Grant SupportThis work was supported by Novartis Pharmaceutical Company to both

Novartis authors and authors from other institutions.The costs of publication of this article were defrayed in part by the

payment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received October 27, 2014; revised February 4, 2015; accepted February 17,2015; published OnlineFirst February 27, 2015.

References1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, et al. Cancer Stat 2008;

58:71–96.2. Nagasue N, Kohno H, Chang YC, Taniura H, Yamanoi A, Uchida M, et al.

Liver resection for hepatocellular carcinoma. Results of 229 consecutivepatients during 11 years. Ann Surg 1993;217:375–84.

3. Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S, Kim JS, et al. Efficacy and safetyof sorafenib in patients in the Asia-Pacific region with advanced hepato-

cellular carcinoma: a phase III randomised, double-blind, placebo-con-trolled trial. Lancet Oncol 2009;10:25–34.

4. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al.Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 2008;359:378–90.

5. Schmelzle T, Hall MN. TOR, a central controller of cell growth. Cell 2000;103:253–62.

Mol Cancer Ther; 14(5) May 2015 Molecular Cancer Therapeutics1234

Huynh et al.

on June 9, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2015; DOI: 10.1158/1535-7163.MCT-14-0768

Page 12: Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in ...Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus Hung Huynh1, Huai-Xiang Hao2,

6. Gingras AC, Raught B, SonenbergN. Regulation of translation initiation byFRAP/mTOR. Genes Dev 2001;15:807–26.

7. Neshat MS, Mellinghoff IK, Tran C, Stiles B, Thomas G, Petersen R, et al.Enhanced sensitivity of PTEN-deficient tumors to inhibition of FRAP/mTOR. Proc Natl Acad Sci U S A 2001;98:10314–9.

8. Podsypanina K, Lee RT, Politis C, Hennessy I, Crane A, Puc J, et al. Aninhibitor of mTOR reduces neoplasia and normalizes p70/S6 kinaseactivity in Ptenþ/- mice. Proc Natl Acad Sci U S A 2001;98:10320–5.

9. Scott PH, BrunnGJ, Kohn AD, Roth RA, Lawrence JC Jr. Evidence of insulin-stimulated phosphorylation and activation of the mammalian target ofrapamycin mediated by a protein kinase B signaling pathway. Proc NatlAcad Sci U S A 1998;95:7772–7.

10. Fang Y, Vilella-Bach M, Bachmann R, Flanigan A, Chen J. Phosphatidicacid-mediated mitogenic activation of mTOR signaling. Science 2001;294:1942–5.

11. Villanueva A, Chiang DY, Newell P, Peix J, Thung S, Alsinet C, et al. Pivotalrole of mTOR signaling in hepatocellular carcinoma. Gastroenterology2008;135:1972–83, 83 e1–11.

12. Wang L,WangWL, Zhang Y, Guo SP, Zhang J, Li QL. Epigenetic and geneticalterations of PTEN in hepatocellular carcinoma. Hepatol Res 2007;37:389–96.

13. Hu TH, Huang CC, Lin PR, Chang HW, Ger LP, Lin YW, et al. Expressionand prognostic role of tumor suppressor gene PTEN/MMAC1/TEP1 inhepatocellular carcinoma. Cancer 2003;97:1929–40.

14. Menon S, Yecies JL, ZhangHH, Howell JJ, Nicholatos J, Harputlugil E, et al.Chronic activation ofmTOR complex 1 is sufficient to cause hepatocellularcarcinoma in mice. Sci Signaling 2012;5:ra24.

15. Zhu AX, Kudo M, Assenat E, Cattan S, Kang YK, Lim HY, et al. Effect ofeverolimus on survival in advanced hepatocellular carcinoma after failureof sorafenib: the EVOLVE-1 randomized clinical trial. JAMA 2014;312:57–67.

16. Huang J, Manning BD. The TSC1-TSC2 complex: a molecular switchboardcontrolling cell growth. Biochem J 2008;412:179–90.

17. Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S,et al. The Cancer Cell Line Encyclopedia enables predictive modelling ofanticancer drug sensitivity. Nature 2012;483:603–7.

18. Huynh H, Soo KC, Chow PK, Panasci L, Tran E. Xenografts of humanhepatocellular carcinoma: a usefulmodel for testing drugs. Clin Cancer Res2006;12:4306–14.

19. HuynhH, Ngo VC, Koong HN, Poon D, Choo SP, Toh HC, et al. AZD6244enhances the anti-tumor activity of sorafenib in ectopic and orthotopicmodels of human hepatocellular carcinoma (HCC). J Hepatol 2010;52:79–87.

20. O'Reilly KE, Rojo F, She QB, Solit D, Mills GB, Smith D, et al. mTORinhibition induces upstream receptor tyrosine kinase signaling and acti-vates Akt. Cancer Res 2006;66:1500–8.

21. HuynhH,Chow PK, TaiWM,Choo SP, Chung AY,OngHS, et al. Dovitinibdemonstrates antitumor and antimetastatic activities in xenograft modelsof hepatocellular carcinoma. J Hepatol 2012;56:595–601.

22. Huynh H, Chow KH, Soo KC, Toh HC, Choo SP, Foo KF, et al. RAD001(everolimus) inhibits tumour growth in xenograft models of humanhepatocellular carcinoma. J Cell Mol Med 2009;13:1371–80.

23. Law BK, Chytil A, Dumont N, Hamilton EG, Waltner-Law ME, Aakre ME,et al. Rapamycin potentiates transforming growth factor beta-inducedgrowth arrest in nontransformed, oncogene-transformed, and humancancer cells. Mol Cell Biol 2002;22:8184–98.

24. JiangWG, Sampson J, Martin TA, Lee-Jones L,Watkins G, Douglas-Jones A,et al. Tuberin and hamartin are aberrantly expressed and linked to clinicaloutcome in human breast cancer: the role of promoter methylation of TSCgenes. Eur J Cancer 2005;41:1628–36.

25. Chakraborty S, Mohiyuddin SM, Gopinath KS, Kumar A. Involvement ofTSC genes and differential expression of other members of the mTORsignaling pathway in oral squamous cell carcinoma. BMC Cancer 2008;8:163.

26. Matsumura S, Imoto I, Kozaki K, Matsui T, Muramatsu T, Furuta M, et al.Integrative array-based approach identifies MZB1 as a frequently methyl-

ated putative tumor suppressor in hepatocellular carcinoma. Clin CancerRes 2012;18:3541–51.

27. Hung CC, Su YN, Chien SC, Liou HH, Chen CC, Chen PC, et al. Molecularand clinical analyses of 84 patients with tuberous sclerosis complex. BMCMed Genet 2006;7:72.

28. Nellist M, Sancak O, Goedbloed MA, Rohe C, van Netten D, Mayer K, et al.Distinct effects of single amino-acid changes to tuberin on the function ofthe tuberin-hamartin complex. Eur J Hum Genet 2005;13:59–68.

29. Hu J, Zacharek S,He YJ, LeeH, Shumway S,Duronio RJ, et al.WD40proteinFBW5 promotes ubiquitination of tumor suppressor TSC2 by DDB1-CUL4-ROC1 ligase. Genes Dev 2008;22:866–71.

30. Huang KT, Huang YH, Li P, He B, Chen ZK, Yu X, et al. Correlation betweentuberous sclerosis complex 2 and glycogen synthase kinase 3 beta levels,and outcomes of patients with hepatocellular carcinoma treated by hep-atectomy. Hepatol Res 2014;44:1142–50.

31. Hoshida Y, Toffanin S, Lachenmayer A, Villanueva A,Minguez B, Llovet JM.Molecular classification and novel targets in hepatocellular carcinoma:recent advancements. Semin Liver Dis 2010;30:35–51.

32. Inoki K, Li Y, Zhu T, Wu J, Guan KL. TSC2 is phosphorylated andinhibited by Akt and suppresses mTOR signalling. Nat Cell Biol 2002;4:648–57.

33. Ma L, Chen Z, Erdjument-Bromage H, Tempst P, Pandolfi PP. Phosphor-ylation and functional inactivation of TSC2 by Erk implications fortuberous sclerosis and cancer pathogenesis. Cell 2005;121:179–93.

34. KuoHP, LeeDF,ChenCT, LiuM,ChouCK, LeeHJ, et al. ARD1 stabilizationof TSC2 suppresses tumorigenesis through the mTOR signaling pathway.Sci Signaling 2010;3:ra9.

35. Totoki Y, TatsunoK, Yamamoto S, Arai Y,Hosoda F, Ishikawa S, et al. High-resolution characterization of a hepatocellular carcinoma genome. NatGenet 2011;43:464–9.

36. Iyer G, Hanrahan AJ, Milowsky MI, Al-Ahmadie H, Scott SN, JanakiramanM, et al. Genome sequencing identifies a basis for everolimus sensitivity.Science 2012;338:221.

37. Voss MH, Hakimi AA, Pham CG, Brannon AR, Chen YB, Cunha LF, et al.Tumor genetic analyses of patientswithmetastatic renal cell carcinoma andextended benefit from mTOR inhibitor therapy. Clin Cancer Res 2014;20:1955–64.

38. Wagle N, Grabiner BC, Van Allen EM, Amin-Mansour A, Taylor-Weiner A,Rosenberg M, et al. Response and acquired resistance to everolimus inanaplastic thyroid cancer. N Engl J Med 2014;371:1426–33.

39. Guichard C, Amaddeo G, Imbeaud S, Ladeiro Y, Pelletier L, Maad IB, et al.Integrated analysis of somatic mutations and focal copy-number changesidentifies key genes and pathways in hepatocellular carcinoma. Nat Genet2012;44:694–8.

40. Fujimoto A, Totoki Y, Abe T, Boroevich KA, Hosoda F, Nguyen HH, et al.Whole-genome sequencing of liver cancers identifies etiological influencesonmutation patterns and recurrentmutations in chromatin regulators. NatGenet 2012;44:760–4.

41. Kan Z, Zheng H, Liu X, Li S, Barber TD, Gong Z, et al. Whole-genomesequencing identifies recurrent mutations in hepatocellular carcinoma.Genome Res 2013;23:1422–33.

42. Huang J, Deng Q, Wang Q, Li KY, Dai JH, Li N, et al. Exome sequencing ofhepatitis B virus-associated hepatocellular carcinoma. Nat Genet 2012;44:1117–21.

43. TCGA. Comprehensive molecular characterization of clear cell renal cellcarcinoma. Nature 2013;499:43–9.

44. Weinstein JN, Akbani R, Broom BM, Wang W, Verhaak RG, McConkey D,et al. Comprehensive molecular characterization of urothelial bladdercarcinoma. Nature 2014;507:315–22.

45. Venook AP, Papandreou C, Furuse J, de Guevara LL. The incidence andepidemiology of hepatocellular carcinoma: a global and regional perspec-tive. Oncologist 2010;15:5–13.

46. Thomas HE, Mercer CA, Carnevalli LS, Park J, Andersen JB, Conner EA,et al. mTOR inhibitors synergize on regression, reversal of gene expres-sion, and autophagy in hepatocellular carcinoma. Sci Transl Med 2012;4:139ra84.

www.aacrjournals.org Mol Cancer Ther; 14(5) May 2015 1235

Frequent TSC2 Loss in HCC and Everolimus Response

on June 9, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2015; DOI: 10.1158/1535-7163.MCT-14-0768

Page 13: Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in ...Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus Hung Huynh1, Huai-Xiang Hao2,

2015;14:1224-1235. Published OnlineFirst February 27, 2015.Mol Cancer Ther   Hung Huynh, Huai-Xiang Hao, Stephen L. Chan, et al.   Inhibitor EverolimusHepatocellular Carcinoma and Predicts Response to mTORC1 Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in

  Updated version

  10.1158/1535-7163.MCT-14-0768doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2015/02/26/1535-7163.MCT-14-0768.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/14/5/1224.full#ref-list-1

This article cites 46 articles, 16 of which you can access for free at:

  Citing articles

  http://mct.aacrjournals.org/content/14/5/1224.full#related-urls

This article has been cited by 18 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/14/5/1224To request permission to re-use all or part of this article, use this link

on June 9, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst February 27, 2015; DOI: 10.1158/1535-7163.MCT-14-0768