Identification of Unique MEK-Dependent Genes in GNAQ … › content › clincanres › 18 › 13...

11
Cancer Therapy: Preclinical Identication of Unique MEK-Dependent Genes in GNAQ Mutant Uveal Melanoma Involved in Cell Growth, Tumor Cell Invasion, and MEK Resistance Grazia Ambrosini 1 , Christine A. Pratilas 2 , Li-Xuan Qin 3 , Madhavi Tadi 2 , Oliver Surriga 1 , Richard D. Carvajal 1 , and Gary K. Schwartz 1 Abstract Purpose: Metastatic uveal melanoma represents the most common intraocular malignancy with very poor prognosis and no effective treatments. Oncogenic mutations in the G-protein a-subunit q and 11 have been described in about 85% of uveal melanomas and confer constitutive activation. Multiple signaling pathways are induced as a consequence of GNAQ/11 activation, which include the MEK/ERK kinase cascade. We analyzed the transcriptional profile of cell lines treated with a mitogen-activated protein (MAP)/ extracellular signal–regulated (ERK) kinase (MEK) inhibitor to identify gene targets of activated GNAQ and to evaluate the biologic importance of these genes in uveal melanoma. Experimental Design: We conducted microarray analysis of uveal melanoma cell lines with GNAQ mutations treated with the MEK inhibitor selumetinib. For comparison, we used cells carrying BRAF V600E and cells without either mutation. Changes in the expression of selected genes were then confirmed by quantitative real-time PCR and immunoblotting. Results: We found that GNAQ mutant cells have a MEK-dependent transcriptional output and identified a unique set of genes that are downregulated by MEK inhibition, including the RNA helicase DDX21 and the cyclin-dependent kinase regulator CDK5R1 whereas Jun was induced. We provide evidence that these genes are involved in cell proliferation, tumor cell invasion, and drug resistance, respectively. Furthermore, we show that selumetinib treatment regulates the expression of these genes in tumor tissues of patients with metastatic GNAQ/11 mutant uveal melanoma. Conclusions: Our findings define a subset of transcriptionally regulated genes by selumetinib in GNAQ mutant cells and provide new insights into understanding the biologic effect of MEK inhibition in this disease. Clin Cancer Res; 18(13); 3552–61. Ó2012 AACR. Introduction The RAS/RAF/MEK/ERK pathway is often activated by genetic alterations in upstream signaling molecules, such as RAS and BRAF. Hyperactivation of this signaling cascade results in dysregulated cell proliferation and malignant transformation of a number of human tumors (1). Recently, the presence of somatic activating mutations in the hetero- trimeric G-protein a-subunit q (GNAQ) and 11 (GNA11) in 46% and 30% of primary uveal melanomas, respectively, has been shown (2–4). These mutations occur in codon 209 in the RAS-like domain, resulting in loss of GTPase activity and constitutive activation. Approximately 85% of ocular melanomas arise from melanocytes within the uveal tract, which consists of the iris, ciliary body, and choroid of the eye (5), and account for a significant rate of deaths. Uveal melanoma is biologically distinct from cutaneous melano- ma, as recurrent chromosomal abnormalities have been detected in chromosomes 3 and 8 (6), as well as a very high tendency to metastasize to the liver (7). NRAS mutations have not been detected in uveal melanoma (8), and BRAF mutations are considered rare (9, 10); however, activation of the mitogen-activated protein kinase (MAPK) pathway by mutant GNAQ/11 appears to be critical for the develop- ment of this disease (2). G-protein a-subunits transduce external signals from 7-transmembrane domain G-protein– coupled receptor (GPCR) to intracellular signaling path- ways. The canonical target of GNAQ/11 stimulation is phospholipase C-b, which, in turn, stimulates inositol lipid signaling [i.e., calcium and protein kinase C (PKC)], in response to mitogenic GPCR agonists (11, 12). The consti- tutively active mutant GNAQ and GNA11 have been reported to activate the extracellular signal–regulated (ERK) Authors' Afliations: 1 Laboratory of New Drug Development and Depart- ment of Medicine, and Departments of 2 Molecular Pharmacology and Chemistry and 3 Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Author: Grazia Ambrosini, Memorial Sloan-Kettering Can- cer Center, 1275 York Avenue, Box 128, New York, NY 10065. Phone: 646- 888-2184; Fax: 646-422-0631; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-11-3086 Ó2012 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 18(13) July 1, 2012 3552 on June 4, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst May 1, 2012; DOI: 10.1158/1078-0432.CCR-11-3086

Transcript of Identification of Unique MEK-Dependent Genes in GNAQ … › content › clincanres › 18 › 13...

  • Cancer Therapy: Preclinical

    Identification of Unique MEK-Dependent Genes in GNAQMutant Uveal Melanoma Involved in Cell Growth, Tumor CellInvasion, and MEK Resistance

    Grazia Ambrosini1, Christine A. Pratilas2, Li-Xuan Qin3, Madhavi Tadi2, Oliver Surriga1,Richard D. Carvajal1, and Gary K. Schwartz1

    AbstractPurpose: Metastatic uveal melanoma represents the most common intraocular malignancy with very

    poor prognosis and no effective treatments. Oncogenicmutations in the G-proteina-subunit q and 11 havebeen described in about 85% of uveal melanomas and confer constitutive activation. Multiple signaling

    pathways are induced as a consequence of GNAQ/11 activation, which include the MEK/ERK kinase

    cascade.Weanalyzed the transcriptional profile of cell lines treatedwith amitogen-activated protein (MAP)/

    extracellular signal–regulated (ERK) kinase (MEK) inhibitor to identify gene targets of activated GNAQ and

    to evaluate the biologic importance of these genes in uveal melanoma.

    Experimental Design: We conducted microarray analysis of uveal melanoma cell lines with GNAQ

    mutations treated with the MEK inhibitor selumetinib. For comparison, we used cells carrying BRAFV600E

    and cells without either mutation. Changes in the expression of selected genes were then confirmed by

    quantitative real-time PCR and immunoblotting.

    Results:We found that GNAQmutant cells have aMEK-dependent transcriptional output and identified

    a unique set of genes that are downregulated byMEK inhibition, including the RNAhelicaseDDX21 and the

    cyclin-dependent kinase regulator CDK5R1whereas Junwas induced.We provide evidence that these genes

    are involved in cell proliferation, tumor cell invasion, and drug resistance, respectively. Furthermore, we

    show that selumetinib treatment regulates the expression of these genes in tumor tissues of patients with

    metastatic GNAQ/11 mutant uveal melanoma.

    Conclusions:Our findings define a subset of transcriptionally regulated genes by selumetinib in GNAQ

    mutant cells and provide new insights into understanding the biologic effect of MEK inhibition in this

    disease. Clin Cancer Res; 18(13); 3552–61. �2012 AACR.

    IntroductionThe RAS/RAF/MEK/ERK pathway is often activated by

    genetic alterations in upstream signalingmolecules, such asRAS and BRAF. Hyperactivation of this signaling cascaderesults in dysregulated cell proliferation and malignanttransformationof anumber of human tumors (1). Recently,the presence of somatic activating mutations in the hetero-trimericG-proteina-subunit q (GNAQ) and11 (GNA11) in46% and 30% of primary uveal melanomas, respectively,has been shown (2–4). Thesemutations occur in codon 209

    in the RAS-like domain, resulting in loss of GTPase activityand constitutive activation. Approximately 85% of ocularmelanomas arise from melanocytes within the uveal tract,which consists of the iris, ciliary body, and choroid of theeye (5), and account for a significant rate of deaths. Uvealmelanoma is biologically distinct from cutaneous melano-ma, as recurrent chromosomal abnormalities have beendetected in chromosomes 3 and 8 (6), as well as a very hightendency to metastasize to the liver (7). NRAS mutationshave not been detected in uveal melanoma (8), and BRAFmutations are considered rare (9, 10); however, activationof themitogen-activatedprotein kinase (MAPK)pathwaybymutant GNAQ/11 appears to be critical for the develop-ment of this disease (2). G-protein a-subunits transduceexternal signals from7-transmembrane domainG-protein–coupled receptor (GPCR) to intracellular signaling path-ways. The canonical target of GNAQ/11 stimulation isphospholipase C-b, which, in turn, stimulates inositol lipidsignaling [i.e., calcium and protein kinase C (PKC)], inresponse to mitogenic GPCR agonists (11, 12). The consti-tutively active mutant GNAQ and GNA11 have beenreported to activate the extracellular signal–regulated (ERK)

    Authors' Affiliations: 1Laboratory of New Drug Development and Depart-ment of Medicine, and Departments of 2Molecular Pharmacology andChemistry and 3Epidemiology and Biostatistics, Memorial Sloan-KetteringCancer Center, New York

    Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

    CorrespondingAuthor:Grazia Ambrosini,Memorial Sloan-KetteringCan-cer Center, 1275 York Avenue, Box 128, NewYork, NY 10065. Phone: 646-888-2184; Fax: 646-422-0631; E-mail: [email protected]

    doi: 10.1158/1078-0432.CCR-11-3086

    �2012 American Association for Cancer Research.

    ClinicalCancer

    Research

    Clin Cancer Res; 18(13) July 1, 20123552

    on June 4, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2012; DOI: 10.1158/1078-0432.CCR-11-3086

    http://clincancerres.aacrjournals.org/

  • pathway (2, 4), and knockdown of mutant GNAQ in uvealmelanoma cells resulted in MAPK inhibition, reducedgrowth, and induced apoptosis (2). However, the transcrip-tional output of ERK signaling downstream of mutant G-proteins is not well characterized. Targeting MAP/ERKkinase (MEK) with allosteric small-molecule inhibitors hasbeen reported to be effective in suppressing cell growth and,in some cells, inducing apoptosis. In particular, melanoma,thyroid, and non–small cell lung cancer with mutantBRAFV600E have been shown to be sensitive to MEK inhi-bitors (13–15). In a large number of tumor types treatedwith the MEK inhibitor selumetinib (AZD6244), Dry andcolleagues have reported transcriptional signatures thatpredict MEK addiction or drug resistance (16). In anotherstudy, using microarray analysis of cells treated with a MEKinhibitor, Pratilas and colleagues have found genes, includ-ing members of the dual specificity phosphatase and spro-uty gene families, that were differentially regulated by MEKinhibition in BRAFV600E cells but not in receptor tyrosinekinase–driven tumor cells with similarly elevated levels ofpERK (17). They also showed that BRAFV600E cells haveelevated ERK-dependent transcriptional output and dis-abled feedback inhibition of RAF/MEK signaling. Here, wereport that uvealmelanoma cells withGNAQmutations arehighly sensitive to MEK inhibition with selumetinib.Expressionmicroarray analysis identified aMEK-dependenttranscriptional profile that is, in part, similar to that ofBRAFV600E melanoma cells. In addition, we identified sev-eral genes unique to GNAQ mutant cells, which areinvolved in proliferation and tumor cell invasion. Further-more, pre- and posttreatment tumor biopsies from an

    ongoing clinical trial of selumetinib in patients with uvealmelanoma indicate that these genes are transcriptionallyregulated and may correlate with clinical benefit.

    Materials and MethodsCell culture

    Omm1.3, Mel202, and Mel270 have been kindly pro-vided by Dr. Bruce Ksander (Harvard Medical School,Boston, MA). OCM1A and 92.1 were from Dr. WilliamHarbour (Washington University, St. Louis, MO). OCM3,Mel290, and C918 were from Robert Folberg (University ofIllinois, Chicago, IL). Uveal melanoma cell lines have beensequenced for the presence of activating mutations incodons 209 (exon 5) and 183 (exon 4) of GNAQ andGNA11. Two cell lines had Q209L mutation (92.1,Mel202), whereas Omm1.3 and Mel270 had Q209P muta-tion. None had GNA11 mutations. Cells were cultured inRPMI medium supplemented with 10% FBS, 100 units/mLpenicillin, and 100 mg/mL streptomycin and maintained at37�C in 5% CO2. Cells were treated with selumetinib(AZD6244, graciously supplied by AstraZeneca).

    Cell viability assaysCells were plated in 96-well plates and treated with the

    indicated concentrations of selumetinib, PD0325901, ordimethyl sulfoxide (DMSO) in triplicates. Viability wasassessed after 4 days of treatment using the Cell CountingKit 8 (CCK8) fromDojindoMolecular Technologies accord-ing to the manufacturer’s instructions. Survival is expressedas a percentage of untreated cells. The calculations ofcombination index values were conducted using the Com-puSyn software (ComboSyn; ref. 18).

    Microarray data analysisCells were treated in triplicate with 250 nmol/L selu-

    metinib, or 0.01% DMSO as control, for 8 hours. Fol-lowing RNA extraction with TRIzol reagent (Invitrogen),cDNA was synthesized in the presence of oligo(dT)24-T7from Genset Corp. cRNA was prepared using biotinylatedUTP and CTP and was hybridized to Human HT-12oligonucleotide Illumina arrays in triplicates. We useda single array slide for each cell line to minimize the effectof experimental artifact. Differential expression analysiswas conducted to identify genes whose expression isaffected by the treatment by comparing the posttreatmentexpression versus the pretreatment expression. The arraydata were log2-transformed and quantile-normalized. Foreach cell line, gene expression was compared betweentime points using the empirical Bayesian method and theR LIMMA package. An empirical Bayes t test was appliedto each gene (19). Linear models and empirical Bayesmethods were used for assessing differential expression inmicroarray experiments and a P value cutoff of 0.0001was used to select differentially expressed genes. There areabout 47,000 markers on the Illumina array and 5 genesare expected by chance to have a P value

  • ImmunoblottingCells were lysed in radioimmunoprecipitation assay

    (RIPA) buffer supplemented with protease inhibitor cock-tail tablets (Roche Diagnostics) and 1 mmol/L Na3VO4.Equal amounts of protein were loaded on 4% to 12%PAGEgels (Invitrogen). Polyvinylidene difluoride (PVDF) mem-branes were blockedwith 5%nonfat driedmilk and probedwith pERK, ERK, cyclin D1, CDK5R1 (p35), c-Jun, anda-tubulin (Cell Signaling); SPRY2, DUSP6, ETV5, andDDX21 (Abcam).

    Quantitative real-time PCRReverse transcription of 1 mg of RNA was done using the

    SuperScript III First-Strand Synthesis System (Invitrogen).Quantitative real-time PCR (qRT-PCR) assays were done onthe 7300 Real Time PCR System (Applied Biosystems).TaqMangene expression assays,which include gene-specificprobe primer sets (Applied Biosystems), were used to detectthe indicated genes and glyceraldehyde-3-phosphate dehy-drogenase (GAPDH)/hypoxanthine phosphoribosyltrans-ferase (HPRT) mRNA. The relative expression of each genewas calculated by the DDCT method.

    RNA interference–mediated gene knockdownThe list of siRNA is reported in the Supplementary Meth-

    ods. siRNAs were transfected using Lipofectamine RNAi-MAX reagent (Invitrogen). After transfections, cells werecounted using a Nexcelom cell counter and plated in 96-well plates for cell viability assays. The statistical significanceof the experimental results was determined by the 2-sidedt test.

    Tissue sample collection and analysisMatched tumor biopsies were collected from patients

    with metastatic uveal melanoma (clinicaltrials.gov; no.NCT01143402) before and after 14 days of selumetinibtreatment. Flash-frozen specimens were then lysed inRIPA lysis buffer and analyzed by immunoblotting.The protocol was approved by the Institutional ReviewBoard of Memorial Sloan-Kettering Cancer Center (NewYork, NY), and all patients signed informed consentforms.

    ResultsThe MEK inhibitor selumetinib inhibits cell viability ofGNAQ mutant uveal melanoma

    Using a panel of uveal melanoma cell lines expressingGNAQQ209L/P, mutant BRAFV600E or wild-type (WT) forboth, we investigated the effects of MEK inhibition on cellviability with selumetinib. As shown in Fig. 1A, theGNAQQ209L/P cells exhibited dose-dependent decreasein cell viability at nanomolar concentrations (IC50< 0.1 mmol/L). The BRAFV600E cells were the most sensi-tive, with IC50 < 0.05 mmol/L, whereas cells without eitherBRAF or GNAQmutations exhibited little sensitivity, withIC50 > 1 mmol/L. This corresponded to inhibition of pERKwith selumetinib in the sensitive mutant cells (Fig. 1B;

    Supplementary Fig. S1A), whereas induction of pMEK isconsistent with relief of negative feedback (20). The effectof MEK inhibition on GNAQ mutant uveal melanomashowed an accumulation in the G1 phase of the cell cycle(Supplementary Fig. S1B).

    GNAQ downregulation by siRNA induced a decrease inexpression of pERK in the GNAQQ209L/P cells (Supplemen-tary Fig. S2A). This corresponded to a decrease in cellviability which was not appreciably increased by selumeti-nib (Supplementary Fig. S2B). Suppression of GNAQ inWTand BRAFV600E cells did not inhibit pERK (SupplementaryFig. S2A) and did not affect cell viability with or withoutselumetinib (Supplementary Fig. S2B). In addition, over-expression of a GNAQQ209L plasmid in a WT cell line(Supplementary Fig. S2C) sensitized the cells to selumeti-nib, as compared with cells with an empty vector (Supple-mentary Fig. S2D). These results confirm that GNAQQ209L/P

    signals to MEK and specifically renders mutant cells sus-ceptible to MEK inhibition, as reported by Van Raamsdonkand colleagues (2).

    Expression profile of GNAQQ209L/P cells compared withBRAFV600E MEK signature

    To identify the transcriptional profile of MEK inhibitionin GNAQQ209L/P cells and potential novel targets ofGNAQQ209L/P signaling, we used microarray gene expres-sion analysis. Cells from each genetically defined subgroupof uveal melanoma (3 cell lines with GNAQQ209L/P, 1 linewith BRAFV600E, and 1 cell line WT for both) were treatedwith 250 nmol/L selumetinib or vehicle for 8 hours. Geneswhose change upon MEK inhibition exceeded defined sta-tistical thresholds for all 3 GNAQQ209L/P cell lines wereconsidered significant and defined the ERK-dependent tran-scriptional output ofGNAQQ209L/P uvealmelanoma cells. Atotal number of 387 genes met the significance level of P �0.0001 in the GNAQQ209L/P cells. Identical parameters wereused to define the set of genes regulated differentially byMEK inhibitor in the BRAFV600E and WT cell lines, and adirect comparison was conducted to evaluate commongenes among the genetic subgroups. Of the 387 genesdetermined to have changed significantly in response toselumetinib, 308 were differentially expressed only inGNAQQ209L/P cells, whereas 29 overlapped with BRAFV600E

    MEK-dependent genes, 42 overlapped with genes meetingsignificance in WT cells, and 8 genes were common to all 3groups (Fig. 1B; Venn diagram). The top 19 genes withhighest significance and fold change common to allGNAQQ209L/P cells before and after treatment with selume-tinib are displayed in a heatmap (Fig. 1D). A list of P valuesand fold change of representative genes for each group(GNAQQ209L/P-only and overlap groups) are shownin Table 1. Of note, several of the genes that were sharedbetween GNAQQ209L/P and BRAFV600E cells were previouslydescribed ERK targets, such as CCND1, transcription factorsETV5, MYC, and genes involved in the feedback inhibitionof MEK/ERK signaling, that is, dual specificity phosphatase6 (DUSP6), and sprouty family members SPRY2, SPRY4.These genes are key components of "MEK signatures"

    Ambrosini et al.

    Clin Cancer Res; 18(13) July 1, 2012 Clinical Cancer Research3554

    on June 4, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2012; DOI: 10.1158/1078-0432.CCR-11-3086

    http://clincancerres.aacrjournals.org/

  • described in other tumor cells (16, 17), and we confirmedtheir decline in expression (mRNA and protein levels) afterselumetinib treatment, using immunoblotting and qRT-PCR (Fig. 2). Following exposure to selumetinib for up to24 hours, pERK levels were determined by immunoblotting(Fig. 2A). In the less sensitiveWT cell line, pERKwas slightlydownregulated at 2 hours and quickly rebounded at latertime points, whereas its inhibition was more complete incells with BRAFV600E and GNAQQ209L/P mutations. Basalexpression levels of ETV5,DUSP6, and SPRY2 proteinswerealmost undetectable in the WT cell line, whereas they werehighly expressed in the mutant cells (both BRAFV600E andGNAQQ209L/P), confirming elevated transcriptional outputof MEK signaling in these cells (17). Cyclin D1 was durablydownregulated in BRAFV600E cells, whereas its expressionrebounded in GNAQQ209L/P and WT cells at later time

    points. The induction of pMEK did not occur in BRAFV600E

    cells, in accordancewith the reported abrogationofnegativefeedback between ERK and RAF proteins by BRAFV600E

    (17, 20). The microarray results of these MEK-dependentgenes were also confirmed by qRT-PCR in 3 GNAQQ209L/P

    cell lines, where these genes were suppressed (Fig. 2B–D).The high basal expression of these transcripts and theirdownregulation by selumetinib confirmed that the MEK/ERK pathway is active in GNAQQ209L/P cells and that thetranscriptional events described in GNAQQ209L/P are atleast, in part, similar to reported MEK functional activa-tion signatures. For example, Pratilas and colleaguesreported a signature of 52 MEK-dependent genes inBRAFV600E cells (17), of which 19 are represented amongour 345 genes (5.5%) and are significantly overenriched(P < 0.0001).

    Figure 1. Selumetinib inhibits cell viability of BRAFV600E and GNAQQ209L/P cell lines. A, cells with wild-type GNAQ/BRAF (C918, Mel290), mutant GNAQ(92.1, Omm1.3, Mel270, Mel202), and BRAFV600E (OCM3, OCM1A) were treated with increasing concentrations of selumetinib (0, 25, 50, 100, 250, 500,1,000 nmol/L) and analyzed for cell viability on day 5, calculated as percentage of untreated controls. Data are representative of 3 independent experiments.Bars, mean � SD. B, immunoblots of pERK and pMEK in response to increasing concentration of selumetinib in cells with different mutational status.Microarray results: C, Venn diagram summarizing differentially expressed genes in selumetinib-treated GNAQQ209L/P cell lines (green circle), BRAFV600E

    (red circle), andWTcells (blue circle), with corresponding overlapping genes as indicated. D, heatmap representation of top 19 genes identified asmeeting thestatistical threshold (see Materials and Methods) for significant change in expression in all the cell lines and with fold change > 2 in GNAQQ209L/P cell lines, inresponse to 250 nmol/L selumetinib (gray bar), or DMSO as control for 8 hours, in triplicate. One replicate of the Mel290 cell line was excluded as itdid not cluster with the other 2 replicates. Cell lines with individual replicates are columns, genes are rows.

    MEK Regulated Genes in Uveal Melanoma with GNAQ Mutation

    www.aacrjournals.org Clin Cancer Res; 18(13) July 1, 2012 3555

    on June 4, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2012; DOI: 10.1158/1078-0432.CCR-11-3086

    http://clincancerres.aacrjournals.org/

  • MEK inhibition regulates unique genes in GNAQQ209L/P

    cellsThe gene profile of MEK inhibition in GNAQQ209L/P cell

    lines also comprised uniquely regulated genes (Table 1).From this list, we selected 3 genes, 2 downregulated(DDX21, CDK5R1) and 1 upregulated (Jun) for validationby immunoblotting and qRT-PCR (Fig. 3). Immunoblotdetection showed that DDX21 and CDK5R1 protein levelswere downregulated in GNAQQ209L/P cells after MEK inhi-bition over time but not in the BRAFV600E and WT cell lines(Fig. 3A). Jun increased exclusively in the GNAQQ209L/P

    cells, whereas it was quickly downregulated in BRAFV600E

    cells. In contrast, Jun protein expression was high inWT cells and did not change upon treatment (Fig. 3A).Selumetinib also reduced DDX21 and CDK5R1 mRNAlevels in GNAQQ209L/P cells, whereas they did not signifi-cantly change in WT cells (Fig. 3B and C). In the BRAFV600E

    cells there was a decrease in mRNA levels but generally to alesser extent than the GNAQQ209L/P cells and this wasassociated with relatively minor changes in the proteinlevels of DDX21 and CDK5R1 (Fig. 3A–C). Also, Jun wasinduced upon treatment in GNAQQ209L/P cells, whereas

    it was profoundly decreased in BRAFV600E (Fig. 3D), inaccordance with the protein levels. There was also a slightdecrease in Jun mRNA in WT cells that did not affect theprotein levels (Fig. 3D).

    To prove specificity of MEK-dependent signaling down-stream of GNAQ and to exclude drug-unrelated effects, theexpression levels of these genes were analyzed in cells afterGNAQ knockdown. Phospho-MEK was markedly down-regulated in the GNAQQ209L/P cells after GNAQ depletion(Fig. 3E). A slight decrease was seen in WT cell line possiblydue to the disruption of WT GNAQ signaling. Consistentwith the effects of selumetinib, CDK5R1 and DDX21 weredownregulated in theGNAQQ209L/P cells, whereas c-Junwasincreased, although less than was observed with drug alone(Fig. 3E). To determinewhether the effects onGNAQQ209L/P

    cells were restricted to selumetinib, we treated uveal mel-anoma cells with a different MEK inhibitor, PD0325901.Both theGNAQQ209L/P andBRAFV600E cellswere sensitive toincreasing concentrations of the drug (Supplementary Fig.S3B). In terms of protein expression, the change in c-Jun(induction inGNAQQ209L/P, suppression in BRAFV600E, andno change in WT) was similar to selumetinib, but

    Table 1. List of differentially expressed genes in response to selumetinib

    Genotype Probe ID Gene name P values Log2 FC

    GANQQ209L/P ILMN_1730928 CDK5R1

  • suppression of CDK5R1 and DDX21 was observed in bothGNAQQ209L/P and BRAFV600E cell lines (Supplementary Fig.S3A). Interestingly, PD0325901 inhibited pERK in the WTcell line but this resulted in essentially no reduction in cellviability, nor a decrease in cyclin D1 expression. Overall,these results suggest that all the effects of selumetinib ontranscriptional expression in uveal melanoma cellsmay notbe shared by other MEK inhibitors. This will require furtherinvestigation.

    MEK-dependent GNAQQ209L/P-specific genes mediatecell proliferation and cell migrationOf the genes uniquely downregulated by selumetinib in

    GNAQQ209L/P cells, we focused on DDX21 and CDKR51.DDX21 is amember of RNAhelicase family, which containsa motif (DExD/H) highly conserved from bacteria tohumans (21). They are multifunctional proteins involvedin RNA unwinding and play a role in transcription regula-tion (22). To test whether DDX21 is involved in the anti-proliferative effects of selumetinib, we carried out geneknocdown experiments. Three GNAQQ209L/P mutant celllines, as well as BRAFV600E and WT cells, were transientlytransfected with 2 different DDX21-specific or control siR-NAs (Fig. 4A, bottom; Supplementary Fig. S4A). Depletionof DDX21 reduced cell viability of all the cell lines, indi-cating a role for DDX21 in cell proliferation that is inde-pendent of the GNAQ status. In fact, the most affected wasthe WT cell line with 60% inhibition of proliferation. TheGNAQQ209L/P cells showed about 40% inhibition relative tocontrol siRNA–transfected cells (Fig. 4A, top; Supplemen-tary Fig. S4B). The addition of selumetinib decreased via-bility of the GNAQQ209L/P cell lines by only an additional10% to20%(Supplementary Fig. S5C–S5E), suggesting that

    DDX21 downregulation contributes to the antiproliferativeeffects of selumetinib in these cells. Furthermore, DDX21downregulation did not affect migration of the cell lines(data not shown).

    CDK5R1 encodes p35, a specific activator of the serine/threonine kinase CDK5,which plays a crucial role in centralnervous system development andmaintenance (23). A rolefor CDK5 and p35 in cell migration has also been reported(24, 25). Seeking to establish a possible functional roleunderlying the observed differences in CDK5R1 expression,we assessed the impact of knockdown of this protein inuveal melanoma cells using 2 different siRNAs. CDK5R1siRNA did not significantly inhibit cell viability (Supple-mentary Fig. 4C). All 3 GNAQQ209L/P cell lines depleted ofCDK5R1 (Fig. 4B, bottom) showed a decrease in cell migra-tion (Fig. 4B, top) compared with cells with control siRNA.The WT cells (Fig. 4B), as well as the BRAFV600E (Supple-mentary Fig. 6), showed no inhibition of migration withsiRNA to CDK5R1, whereas GNAQ siRNA inhibited migra-tion of theGNAQQ209L/P cell lineOmm1.3 (SupplementaryFig. 6). Interestingly, selumetinib inhibited migration ofboth GNAQQ209L/P and BRAFV600E, but not the WT cells(Supplementary Fig. 6), suggesting that selumetinib mayhave effects on the cellmigration of BRAFV600E cells, but thiseffect is independent of CDK5R1.

    Jun expression is related to sensitivity to MEKinhibition

    c-Jun is a component of the AP-1 transcription complexregulated by c-jun-NH2-kinase (JNK), and it is involved ina number of cell responses, such as cell proliferation andcell death (26). c-Jun was upregulated in GNAQQ209L/P

    cells after selumetinib treatment, showing a differential

    Figure 2. Validation of MEK-regulated genes in GNAQQ209L/P

    cells. These genes wererepresentative of those sharedbetween GNAQQ209L/P andBRAFV600E MEK–dependentsignature. A, immunoblot analysis ofcells treated with 250 nmol/Lselumetinib over the time, usingantibodies for the indicatedproteins.Each blot is representative of at least2 experiments showing sameresults. B–D, relativemRNA levels ofselected ERK output genes beforeand after selumetinib in threeGNAQQ209L/P cell lines. Cells weretreated with selumetinib for 8 hoursand RT-PCR was carried out usinggene-specific primers for DUSP6,ETV5, SPRY2, and cyclin D1. Valueswere normalized with GADPH andHPRT as housekeeping genes usingthe DDCT method. Each experimentwas carried out in triplicates. Bars,�SD.

    MEK Regulated Genes in Uveal Melanoma with GNAQ Mutation

    www.aacrjournals.org Clin Cancer Res; 18(13) July 1, 2012 3557

    on June 4, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2012; DOI: 10.1158/1078-0432.CCR-11-3086

    http://clincancerres.aacrjournals.org/

  • regulation compared with cells with BRAFV600E. Knock-down of c-Jun by 2 different siRNAs (Fig. 4C; Supplemen-tary Fig. S7A) significantly increased the antiproliferativeeffects of selumetinib in GNAQQ209L/P cell lines (Fig. 4D;Supplementary Fig. S7B), suggesting that c-Jun inductionmay be involved in mechanisms of resistance to MEKinhibition. In contrast, c-Jun did not seem to play a roleinWTandBRAFV600E cells as its knockdowndidnot alter thesensitivity to the selumetinib when compared with controlsiRNA (Fig. 4D; Supplementary Fig. S7B).

    ValidationofGNAQQ209L/P-specific ERK transcriptionaloutput in tumor tissues

    To assess the clinical efficacy of MEK inhibition onuveal melanoma, we are conducting a phase II clinicaltrial of selumetinib versus temozolomide in patientswith metastatic uveal melanoma (clinicaltrials.gov; no.NCT01143402). Matched tumor biopsies are collected toexamine target modulation between baseline and day 14from patients with GNAQ/11 mutations receiving selume-tinib. Results from 3 representative biopsy pairs are shown(Fig. 5A). The results of all the patients in the trial will bereported separately. Sustained inhibition of pERK and sup-pression of cyclin D1 were observed in patients A and B onday 14, but not in patient C. This correlated to best clinicalresponse by Response Evaluation Criteria in Solid Tumors

    (RECIST; ref. 27): partial response in liver metastases inpatient A (Fig. 5B), stable disease in patient B, and progres-sion of disease in patient C. Furthermore, DDX21 wasdownregulated by selumetinib in patients A and C,CDK5R1 decreased in patient A and B, whereas they couldnot be detected in patients B and C, respectively. c-Junexpression increased in patient C (Fig. 5A). A similar trendhas been seen in other patients treated on this clinical trial.These preliminary results are consistent with our in vitrostudies indicating the existence of unique subset of genes inGNAQQ209L/P cells that are regulated by selumetinib and theexpression of which could have an impact on clinicaloutcome.

    DiscussionUveal melanoma represents the most common intraoc-

    ular malignancy. However, there are no effective treatmentsfor this aggressive disease. Selumetinib is the only MEKinhibitor in clinical trials currently in the United States forpatients with uveal melanoma. Here, we report that cellswith GNAQQ209L/P mutations are sensitive to MEK inhibi-tion by selumetinib, and sensitizationwas associated with aMEK-dependent gene expression profile. Some features ofthis profile are overlapping with that elicited in BRAFV600E

    uveal melanoma cells and other cell types (16), which

    Figure 3. Confirmation of MEK-dependent genes differentiallyregulated by selumetinib inGNAQQ209L/P cells. A, immunoblotanalysis of DDX21, CDK5R1, andJun expression after treatment with250 nmol/L selumetinib over thetime in cell lines with differentmutational status. Total RNA wasextracted from cells after 8 hours oftreatment and qRT-PCR wascarried out using gene-specificprimers for DDX21 (B), CDK5R1 (C),and Jun (D). Values are relative tomRNA levels of untreated cells.Each experiment was carried out 2or 3 times in triplicates.E, Western blot analysis of uvealmelanoma cells after GNAQknockdown by siRNA using theindicated antibodies.

    Ambrosini et al.

    Clin Cancer Res; 18(13) July 1, 2012 Clinical Cancer Research3558

    on June 4, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2012; DOI: 10.1158/1078-0432.CCR-11-3086

    http://clincancerres.aacrjournals.org/

  • supports the MEK dependence of GNAQQ209L/P cells. Thisgene profile includes the dual-specificity phosphatases(DUSP4/6), the sprouty homologues (SPRY1/2/4), whichare known transcriptional targets of the ERK pathwayinvolved in negative feedback regulation of ERK. The Etsvariant transcription factor ETV5was also regulated byMEKinhibition, along with cell-cycle division–associated pro-tein 7 (CDCA7), the proto-oncogene MYC, and the solutecarrier family 16, member 6 (SLC16A6).Additional features of the MEK profile were identified

    as specific for GNAQQ209L/P cells. A number of genessuppressed in GNAQQ209L/P cells by selumetinib, suchas LYAR, NOP58, GNL3, and PPAT, were reported asnuclear proteins involved in cell growth and tumorigen-esis (28–31). DDX21 was recently identified as a novelbiomarker for colorectal cancer (32), whereas CDK5R1was involved in metastasis (24, 33) and associated withmeningioma progression (34). Interestingly, it has beenreported that mutant K-RAS regulates expression/stability

    of CDK5 and CDK5R1 (p35) to increase malignant pro-gression and invasion of pancreatic cancer cells (35). It isplausible that mutant GNAQ acts similarly to mutant K-RAS, as CDK5R1 expression was in fact elevated in theGNAQQ209L/P cells compared with cells with other geneticbackgrounds. Furthermore, it has been reported thatCDK5 negatively regulates JNK3 activity and its targetc-Jun, to prevent apoptosis in developing neurons (36).This implicates a possible interaction between these pro-teins in promoting survival of uveal melanoma cells.CDK5R1 and DDX21 were also downregulated by selu-metinib in vivo in tissues of patients enrolled in a phase IIclinical trial we are conducting.

    Jun was upregulated after selumetinib treatment in theGNAQQ209L/P cells only. Depending on the cell type anddrug treatment, c-Jun and Jun kinase have been implicatedin both pro- and antiapoptotic responses (37). In cutaneousmelanoma cells, active ERK induces c-Jun expression(38). In contrast, c-Jun was induced by MEK inhibition

    Figure 4. GNAQQ209L/P MEK–dependent genes play a role in cell proliferation and metastasis. A, siRNA-mediated knockdown of DDX21 (þ) in 5 cell lines(bottom); (�), control siRNA. Cell viability wasmeasured after 4 days (top). B, migration assays of WT and GNAQQ209L/P cell lines transfected with CDK5R1 orcontrol siRNA (bottom). Cells migrated through the membrane pores were stained and counted under the microscope. Representative fields of �200magnification are shown. C, uveal melanoma cell lines were transfected with control or c-Jun siRNA and tested in proliferation assays (D) after 4 days of 250nmol/L selumetinib treatment. Experiments were repeated 3 times in triplicates. �, P < 0.0001; ��, P < 0.001; ���, P < 0.005 for comparison of c-Jun versuscontrol siRNA with selumetinib treatment.

    MEK Regulated Genes in Uveal Melanoma with GNAQ Mutation

    www.aacrjournals.org Clin Cancer Res; 18(13) July 1, 2012 3559

    on June 4, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2012; DOI: 10.1158/1078-0432.CCR-11-3086

    http://clincancerres.aacrjournals.org/

  • in GNAQQ209L/P uveal melanoma cells, suggesting a differ-ential regulation of the ERK/JNK pathway. G-protein–mediated signaling is complex and involvesmultiple down-stream binding partners and various regulatory scaffolding/adaptor and effector proteins (12). For example, PKC is atarget of GNAQ activation, and it might be involved infeedback regulation of c-Jun when ERK is inhibited. Theupregulation of c-Jun could represent an alternative route tocell proliferation, which would explain the relative lowersensitivity to selumetinib of GNAQQ209L/P cells as com-pared with BRAFV600E cells. Interestingly, increased expres-sionof c-Junhas also been reported in colorectal cancer cellswith K-RAS or BRAF mutations after acquired resistance toselumetinib (39). We showed that the antiproliferativeeffect of selumetinib can be enhanced by suppressing c-Junin the GNAQQ209L/P cells. This would suggest that targetingc-Jun in the presence of MEK inhibition would result inenhanced antitumor effects and may prevent selumetinibresistance. In conclusion, our findings define a uniquemolecular profile ofMEK inhibition by selumetinib in uvealmelanoma cells with mutant GNAQ and point to a set oftranscriptionally modified genes that could have an impacton the activity of this agent in this disease.

    Disclosure of Potential Conflicts of InterestC.A. Pratilas is a consultant/advisory board member for Roche. No

    potential conflicts of interest were disclosed by the other authors.

    Authors' ContributionsConception and design: G. Ambrosini, C.A. Pratilas, G.K. SchwartzDevelopment of methodology: G. Ambrosini, G.K. SchwartzAcquisitionofdata (provided animals, acquired andmanagedpatients,provided facilities, etc.): O. Surriga, R.D. Carvajal, G.K. SchwartzAnalysis and interpretation of data (e.g., statistical analysis, biosta-tistics, computational analysis): C.A. Pratilas, L.-X. Qin, M. Tadi, G.K.SchwartzWriting, review, and/or revision of the manuscript: G. Ambrosini, C.A.Pratilas, L.-X. Qin, R.D. Carvajal, G.K. SchwartzAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): G. Ambrosini, M. Tadi, G.K.SchwartzStudy supervision: G. Ambrosini, G.K. Schwartz

    Grant SupportThis work was supported by Melanoma Research Alliance and Cycle for

    Survival funds. C.A. Pratilas was supported by the NIH grant # K08-127350.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 December 2, 2011; revised April 18, 2012; accepted April 26,2012; published OnlineFirst May 1, 2012.

    References1. Zebisch A, Czernilofsky AP, Keri G, Smigelskaite J, Sill H, Troppmair J.

    Signaling through RAS-RAF-MEK-ERK: from basics to bedside. CurrMed Chem 2007;14:601–23.

    2. Van Raamsdonk CD, Bezrookove V, Green G, Bauer J, Gaugler L,O'Brien JM, et al. Frequent somatic mutations of GNAQ in uvealmelanoma and blue naevi. Nature 2009;457:599–602.

    3. Onken MD, Worley LA, Long MD, Duan S, Council ML, Bowcock AM,et al. Oncogenic mutations in GNAQ occur early in uveal melanoma.Invest Ophthalmol Vis Sci 2008;49:5230–4.

    4. Van Raamsdonk CD, Griewank KG, Crosby MB, Garrido MC, VemulaS, Wiesner T, et al. Mutations in GNA11 in uveal melanoma. N Engl JMed 2010;363:2191–9.

    5. Strickland D, Lee JA. Melanomas of eye: stability of rates. Am JEpidemiol 1981;113:700–2.

    6. Horsman DE, White VA. Cytogenetic analysis of uveal melanoma.Consistent occurrence of monosomy 3 and trisomy 8q. Cancer1993;71:811–9.

    7. Singh AD, Bergman L, Seregard S. Uveal melanoma: epidemiologicaspects. Ophthalmol Clin North Am 2005;18:75–84.

    8. MooyCM, Van der HelmMJ, Van der Kwast TH, De JongPT, Ruiter DJ,Zwarthoff EC. No N-ras mutations in human uveal melanoma: the roleof ultraviolet light revisited. Br J Cancer 1991;64:411–3.

    9. Zuidervaart W, van Nieuwpoort F, Stark M, Dijkman R, Packer L,Borgstein AM, et al. Activation of the MAPK pathway is a common

    Figure 5. Validation of MEK inhibition and expression of ERK-dependent genes in tumor tissues. A, baseline and day 14 liver tumor biopsies were collected inpatients with mutant GNAQ/11 metastatic uveal melanoma receiving selumetinib in a phase II clinical trial. Matched pair biopsies were analyzed byimmunoblotting for expression levels of pERK, ERK, cyclin D1, CDK5R1, DDX21, c-Jun, and a-tubulin. Clinical efficacy is also shown. PD, progressivedisease; PR, partial response; SD, stable disease. B, liver metastases of patient A with uveal melanoma (pre- and posttreatment with selumetinib). A partialresponse in the liver lesion is assessed by computer-assisted tomographic (CAT) scan (left, arrows) and positron emission tomography (PET) scan (right,arrows).

    Ambrosini et al.

    Clin Cancer Res; 18(13) July 1, 2012 Clinical Cancer Research3560

    on June 4, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2012; DOI: 10.1158/1078-0432.CCR-11-3086

    http://clincancerres.aacrjournals.org/

  • event in uvealmelanomas although it rarely occurs throughmutation ofBRAF or RAS. Br J Cancer 2005;92:2032–8.

    10. Malaponte G, Libra M, Gangemi P, Bevelacqua V, Mangano K,D'Amico F, et al. Detection of BRAF genemutation in primary choroidalmelanoma tissue. Cancer Biol Ther 2006;5:225–7.

    11. Rozengurt E. Mitogenic signaling pathways induced by G protein-coupled receptors. J Cell Physiol 2007;213:589–602.

    12. Hubbard KB, Hepler JR. Cell signalling diversity of the Gqalpha familyof heterotrimeric G proteins. Cell Signal 2006;18:135–50.

    13. Solit DB, Garraway LA, Pratilas CA, Sawai A, Getz G, Basso A, et al.BRAF mutation predicts sensitivity to MEK inhibition. Nature 2006;439:358–62.

    14. Leboeuf R, Baumgartner JE. BRAFV600E mutation is associated withpreferential sensitivity to mitogen-activated protein kinase kinaseinhibition in thyroid cancer cell lines. J Clin Endocrinol Metab2008;93:2194–201

    15. PratilasCA,HanrahanAJ,Halilovic E, PersaudY,SohJ,ChitaleD, et al.Genetic predictors of MEK dependence in non-small cell lung cancer.Cancer Res 2008;68:9375–83.

    16. Dry JR, Pavey S, Pratilas CA, Harbron C, Runswick S, Hodgson D,et al. Transcriptional pathway signatures predict MEK addictionand response to selumetinib (AZD6244). Cancer Res 2010;70:2264–73.

    17. Pratilas CA, Taylor BS, Ye Q, Viale A, Sander C, Solit DB, et al. (V600E)BRAF is associated with disabled feedback inhibition of RAF-MEKsignaling and elevated transcriptional output of the pathway. Proc NatlAcad Sci U S A 2009;106:4519–24.

    18. Chou TC. Drug combination studies and their synergy quantificationusing the Chou-Talalay method. Cancer Res 2010;70:440–6.

    19. Smyth GK. Linear models and empirical Bayes methods for assessingdifferential expression in microarray experiments. Stat Appl GenetMolBiol 2004;3:Article3.

    20. Friday BB, YuC, DyGK, Smith PD,Wang L, Thibodeau SN, et al. BRAFV600E disrupts AZD6244-induced abrogation of negative feedbackpathways between extracellular signal-regulated kinase and Raf pro-teins. Cancer Res 2008;68:6145–53.

    21. Fuller-Pace FV. DExD/H box RNA helicases: multifunctional proteinswith important roles in transcriptional regulation. Nucleic Acids Res2006;34:4206–15.

    22. Linder P, Jankowsky E. From unwinding to clamping - the DEAD boxRNA helicase family. Nat Rev Mol Cell Biol 2011;12:505–16.

    23. Lopes JP, Agostinho P. Cdk5:multitasking between physiological andpathological conditions. Prog Neurobiol 2011;94:49–63.

    24. Strock CJ, Park JI, Nakakura EK, Bova GS, Isaacs JT, Ball DW, et al.Cyclin-dependent kinase 5 activity controls cell motility and met-astatic potential of prostate cancer cells. Cancer Res 2006;66:7509–15.

    25. Contreras-Vallejos E, Utreras E, Gonzalez-Billault C. Going out of thebrain: non-nervous system physiological and pathological functions ofCdk5. Cell Signal 2011;24:44–52.

    26. Vogt PK. Jun, the oncoprotein. Oncogene 2001;20:2365–77.27. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS,

    Rubinstein L, et al. New guidelines to evaluate the response to treat-ment in solid tumors. European Organization for Research and Treat-ment ofCancer,NationalCancer Instituteof theUnitedStates,NationalCancer Institute of Canada. J Natl Cancer Inst 2000;92:205–16.

    28. Su L, Hershberger RJ, Weissman IL. LYAR, a novel nucleolar proteinwith zinc finger DNA-binding motifs, is involved in cell growth regu-lation. Genes Dev 1993;7:735–48.

    29. ReichowSL,HammaT, Ferre-D'AmareAR,VaraniG. The structure andfunction of small nucleolar ribonucleoproteins. Nucleic Acids Res2007;35:1452–64.

    30. Yoshida R, Fujimoto T, Kudoh S, Nagata M, Nakayama H, ShinoharaM, et al. Nucleostemin affects the proliferation but not differentiation oforal squamous cell carcinoma cells. Cancer Sci 2011;102:1418–23.

    31. GassmannMG, Stanzel A, Werner S. Growth factor-regulated expres-sion of enzymes involved in nucleotide biosynthesis: a novel mech-anism of growth factor action. Oncogene 1999;18:6667–76.

    32. Jung Y, Lee S, Choi HS, Kim SN, Lee E, Shin Y, et al. Clinical validationof colorectal cancer biomarkers identified frombioinformatics analysisof public expression data. Clin Cancer Res 2011;17:700–9.

    33. Moncini S, Salvi A, Zuccotti P, VieroG,Quattrone A, Barlati S, et al. Therole of miR-103 and miR-107 in regulation of CDK5R1 expression andin cellular migration. PLoS One 2011;6:e20038.

    34. Wrobel G, Roerig P, Kokocinski F, Neben K, Hahn M, Reifenberger G,et al. Microarray-based gene expression profiling of benign, atypicaland anaplastic meningiomas identifies novel genes associated withmeningioma progression. Int J Cancer 2005;114:249–56.

    35. Eggers JP, Grandgenett PM, Collisson EC, Lewallen ME, Tremayne J,Singh PK, et al. Cyclin-dependent kinase 5 is amplified and over-expressed in pancreatic cancer and activated by mutant K-Ras. ClinCancer Res 2011;17:6140–50.

    36. Li BS, Zhang L, Takahashi S, MaW, Jaffe H, Kulkarni AB, et al. Cyclin-dependent kinase 5 prevents neuronal apoptosis by negative regula-tion of c-Jun N-terminal kinase 3. EMBO J 2002;21:324–33.

    37. Vasilevskaya I, O'Dwyer PJ. Role of Jun and Jun kinase in resistance ofcancer cells to therapy. Drug Resist Updat 2003;6:147–56.

    38. Lopez-Bergami P, Huang C, Goydos JS, Yip D, Bar-Eli M, Herlyn M,et al. Rewired ERK-JNK signaling pathways in melanoma. Cancer Cell2007;11:447–60.

    39. Little AS, Balmanno K, SaleMJ, Newman S, Dry JR, HampsonM, et al.Amplification of the driving oncogene, KRAS or BRAF, underpinsacquired resistance to MEK1/2 inhibitors in colorectal cancer cells.Sci Signal 2011;4:ra17.

    MEK Regulated Genes in Uveal Melanoma with GNAQ Mutation

    www.aacrjournals.org Clin Cancer Res; 18(13) July 1, 2012 3561

    on June 4, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2012; DOI: 10.1158/1078-0432.CCR-11-3086

    http://clincancerres.aacrjournals.org/

  • 2012;18:3552-3561. Published OnlineFirst May 1, 2012.Clin Cancer Res Grazia Ambrosini, Christine A. Pratilas, Li-Xuan Qin, et al. MEK ResistanceUveal Melanoma Involved in Cell Growth, Tumor Cell Invasion, and Identification of Unique MEK-Dependent Genes in GNAQ Mutant

    Updated version

    10.1158/1078-0432.CCR-11-3086doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://clincancerres.aacrjournals.org/content/suppl/2012/05/01/1078-0432.CCR-11-3086.DC1

    Access the most recent supplemental material at:

    Cited articles

    http://clincancerres.aacrjournals.org/content/18/13/3552.full#ref-list-1

    This article cites 38 articles, 12 of which you can access for free at:

    Citing articles

    http://clincancerres.aacrjournals.org/content/18/13/3552.full#related-urls

    This article has been cited by 5 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://clincancerres.aacrjournals.org/content/18/13/3552To request permission to re-use all or part of this article, use this link

    on June 4, 2021. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2012; DOI: 10.1158/1078-0432.CCR-11-3086

    http://clincancerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-11-3086http://clincancerres.aacrjournals.org/content/suppl/2012/05/01/1078-0432.CCR-11-3086.DC1http://clincancerres.aacrjournals.org/content/18/13/3552.full#ref-list-1http://clincancerres.aacrjournals.org/content/18/13/3552.full#related-urlshttp://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/18/13/3552http://clincancerres.aacrjournals.org/