Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Apricoxib,anovelinhibitorofCOX2markedlyimprovesstandardtherapyresponse inmolecularlydefinedmodelsofpancreaticcancer Amanda Kirane1, Jason E. Toombs1, Katherine Ostapoff1, Juliet G. Carbon1, Sara Zaknoen2, Jordan Braunfeld1,RoderichE.Schwarz1,FrancisJ.Burrows2andRolfA.Brekken1 1Division of Surgical Oncology, Department of Surgery, and the Hamon Center for Therapeutic OncologyResearch,UniversityofTexasSouthwesternMedicalCenter,Dallas,TX 2TragaraPharmaceuticals,Inc,SanDiego,CA RunningTitle:Apricoxibenhancestherapyofpancreaticcancer FinancialSupport:ThisworkwassupportedinpartbyasponsoredresearchagreementfromTragara Pharmaceuticals,IncandtheEffieMarieCainScholarshipinAngiogenesisResearch(toRAB)andan institutionaltraininggrantfromtheNCI(T32CA136515,toAK). Addressforcorrespondence: RolfA.Brekken,PhD 6000HarryHinesBlvd HamonCenterforTherapeuticOncologyResearch UTSouthwesternMedicalCenter Dallas,TX753908593 Ph:2146485151 Fax:2146484940 [email protected]

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page2

StatementofTranslationalRelevance

Current standard treatment regimens in pancreatic cancer are minimally effective in prolonging overall survival, highlighting the need for novel treatment strategies. The precise function of COX2 in the tumor microenvironment remains incompletely understood but it has been implicated in tumor angiogenesis and epithelialtomesenchymaltransition.ApricoxibisanovelCOX2inhibitorinPhaseIIclinicaltrialsinpancreatic cancer being investigated as a strategy to augment the efficacy of gemcitabine and erlotinib. Here, in preclinical models of pancreatic cancer, apricoxib significantly enhanced the efficacy of gemcitabine plus erlotinibinreducingprimarytumorburdenandtheoccurrenceofmetastasesinorthotopictumormodelswith elevatedCOX2activity.Strikingly,apricoxibtreatmentalsorobustlypreventedtumorcellsfromadoptinga mesenchymalphenotypeinvivo,regardlessofCOX2expressionlevelsbytheprimarytumor.Enhancingan epithelialphenotypeviaCOX2inhibitionmayimproveefficacyofchemotherapy.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page3

Abstract Purpose:COX2isexpressedhighlyinpancreaticcancerandimplicatedintumorprogression.COX2inhibition can reduce tumor growth and augment therapy. The precise function of COX2 in tumors remains poorly understood, but it is implicated in tumor angiogenesis, evasion of apoptosis and induction of epithelial to mesenchymal transition (EMT). Current therapeutic regimens for pancreatic cancer are minimally effective, highlightingtheneedfornoveltreatmentstrategies.Here,wereportthatapricoxib,anovelCOX2inhibitorin

Phase II clinical trials, significantly enhances the efficacy of gemcitabine/erlotinib in preclinical models of pancreaticcancer.

ExperimentalDesign:Humanpancreaticcelllineswereevaluatedinvitroandinvivoforresponsetoapricoxib

+/standardofcaretherapy(gemcitabine+erlotinib).Tumortissueunderwentposttreatmentanalysisforcell proliferation,viabilityandEMTphenotype.Vascularparameterswerealsodetermined.

Results:COX2inhibitionreducedtheIC50ofgemcitabine+/erlotinibinsixpancreaticcancercelllinestested invitro.Furthermore,apricoxibincreasedtheantitumorefficacyofstandardcombinationtherapyinseveral orthotopic xenograft models. In vivo, apricoxib combination therapy was only effective at reducing tumor growth and metastasis in tumors with elevated COX2 activity. In each model examined, treatment with apricoxibresultedinvascularnormalizationwithoutadecreaseinmicrovesseldensityandpromotionofan epithelialphenotypebytumorcellsregardlessofbasalCOX2expression.

Conclusions:ApricoxibrobustlyreversesEMTandaugmentsstandardtherapywithoutreducingmicrovessel densityandwarrantsfurtherclinicalevaluationinpatientswithpancreaticcancer.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page4

Introduction

Theprogressionofseveralhumancancers,includingpancreatic,hasbeenlinkedtoinflammation,whichcan triggersecretionofgrowthfactors,infiltrationofimmunecells,andDNAdamagebyreactiveoxygenspecies leading to tumor cell proliferation and escape from cell death (1). Epidemiologically, chronic use of anti inflammatorydrugscorrelateswithareductionintheincidenceofcertaincancers,includingpancreaticcancer

(2,3).Upregulationof2(COX2),theratelimitingstepinthesynthesisoffrom arachidonicacid,isanearlyinflammatoryresponseandisregulatedbyseveralgrowthfactorsandcytokines

(4).ElevatedCOX2expressionisfrequentinmanycancers,includingpancreaticcancerwhereitisexpressed highlyinupto75%ofcases(5).IncreasedCOX2inpancreatictumorscorrelateswithincreasedinvasiveness andshorteroverallsurvival(610).ThemostabundantproductofCOX2intumors,E2(PGE2), can affect multiple carcinogenic pathways and participate in proliferation, invasion, angiogenesis, chemoresistanceandmetastasis(5,1114).Inthepancreasundernormalconditions,onlyisletcellsexpress

COX2(10).Aspancreaticintraepitheliallesionsdevelop,COX2expressioniselevatedandtheuseofCOX2 inhibitorscandelaytheprogressionofthediseaseinpreclinicalKRasdrivenmousemodels.Indeed,inhibition ofCOX2canreducetumorgrowthinseveralexperimentalmodelsofpancreaticcancer(15,16).Importantly, prostaglandinssecretedfromstromalfibroblastsfacilitatetumorcellproliferationandsurvivalevenintumors andcelllineswhereCOX2isnotexpressedbytumorcells,suggestingthatCOX2isanimportantsignaling moleculeinthetumormicroenvironment(17).Collectively,thesefindingsmakeCOX2anattractivetargetfor anticancertherapy.

Despiteadvancesindiagnosticsandtreatment,theprognosisforinoperablepancreaticcancerremains poor,largelyduetolocalinvasionandmetastaticprogressionatearlystages(18).Thecurrentstandardofcare forpancreaticcancerisgemcitabinebutitaffordsminimalsurvivalbenefittopatients(19).Additionofthe

COX2inhibitortogemcitabinecontainingregimenshaveyieldedmixedresults;indeed,strategies

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page5 to augment gemcitabine activity have largely failed to improve overall survival in Phase III clinical studies, exceptingcombinationwitherlotinib,aninhibitorofepidermalgrowthfactorreceptor(EGFR).However,while statistically significant, erlotinibbased improvement remains modest (20, 21). Rationale exists for the combinedtargetingofEGFRandCOX2assignificantoverlapandinteractionoccurbetweenthesepathways.

PGE2 can transactivate EGFR, which can subsequently increase expression of COX2. In addition, PGE2, via promotion of epithelial to mesenchymal transition (EMT), can increase resistance to EGFR inhibitors (22).

Furthermore,shortcircuitingbothpathwayssimultaneouslycanhavesynergisticeffectsinpreclinicalcancer models(2325).

ThemechanisticbasisofCOX2inhibitortherapyinpancreaticcancerremainsilldefined,althoughin the HT29 model of colorectal cancer reversion of the EMT phenotype has been reported to be a key mechanismofaction(26).Apricoxibisanovel,selectiveCOX2inhibitorcurrentlyinPhaseIIstudiesforNSCLC andpancreaticcancer.Apricoxibhasdemonstratedsignificantantitumoreffectsinxenograftmodelsoflung and colorectal cancer and appears more potent than previous COX2 inhibitors (23, 27, 28). In the present study, we characterized the baseline expression and activity of EGFR and COX2 in commonlyemployed pancreaticcancercelllinesaswellasfunctionalresponsestoinhibitionofeachpathway.Invivostudieswith

COX2 positive and negative cell lines demonstrated that addition of apricoxib enhanced the antitumor efficacyofgemcitabineanderlotinibinCOX2dependentmodels.Wepresentevidencethatapricoxibadds significantly to the treatment effect of standard therapy in COX2 expressing cell lines and also promotes vascularstabilizationwhilereversingEMTregardlessoftheCOX2statusoftheprimarytumor.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page6

MaterialsandMethods

Cell Lines. Human pancreatic cancer cell lines AsPC1, Su.86.86, HPAFII, PL45, and CFPAC1 were obtained fromATCC(Manassas,VA);Colo357wasobtainedfromMDAndersonCancerCenter.AsPC1,andPL45were growninDMEM,CFPAC1inIMDM,Colo357andHPAFIIinMEM,andSu.86.86inRPMI(Fisher).Allcelllines

were grown in a humidified atmosphere with 5% CO2, at 37 C, and have been DNA fingerprinted for provenanceusingthePowerPlex1.2kit(Promega)andconfirmedtobethesameastheDNAfingerprintlibrary maintainedbyATCCandconfirmedtobefreeofmycoplasmabytheeMycokit(BocaScientific).

Baseline expressionstatus.ForWesternblotanalysis,celllysateswereproducedusing MPER(Pierce)with addedproteaseandphosphataseinhibitors(Pierce)andproteinconcentrationwasdeterminedbyBCAassay

(Pierce). Immunodetection was performed by electrophoretic transfer of SDSPAGE separated proteins to

PVDFmembranes.AntibodiesusedforWesternblotanalysisincludedEGFR(Upstate),Tyr1069phosphoEGFR andCox2(SantaCruz).

For PCR analysis, RNA was prepared using TRIzol (Invitrogen) per manufacturer instructions and concentrationwasdeterminedbyspectrophotometry.ThecDNAusedforsubsequentforPCRwasmadeusing iScript(BioRadLaboratories)andChoiceDNATaqpolymerase(DenvilleScientific).TheexpressionofCOX2 andEGFRwasanalyzedbyquantitativerealtimePCRusingactinasaninternalreferencegene.Eachreaction wasperformedintriplicatewithRNAharvestedfrom3independentcellcultures.ThecomparativeCtmethod wasusedtocomputerelativeexpressionvalues(29).

Invitrocytotoxicityanddrugresponseassay.MTSassayswereperformedin96wellplates;cellswereplated on day 0 and drug was added on day 1 in fourfold dilutions. Drugs were evaluated as single agents with maximumconcentrationof2000nMforgemcitabineand400μMforerlotinibandapricoxib.Forcombination studies gemcitabine was added with a fixed concentration of 1 μM erlotinib, 0.1 μM apricoxib, or 1 μM

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page7 apricoxib, and triple combination with 1 μM erlotinib and 1 μM apricoxib. Relative cell number was determinedbyaddingMTS(Promega,finalconcentration333g/ml),incubatingfor1to3hoursat37°C,and readingabsorbance.DrugsensitivitycurvesandIC50swerecalculatedusinginhousesoftware(30).

PhosphoEGFRELISA(R&DSystems)wasperformedafterincubatingcellsovernightina96wellplate, serumstarvingfor4hoursandstimulatingfor15minuteswith10ng/mlEGF.Theplatewasprocessedper manufacturerinstructions.PGE2ELISA(Cayman)wasusedtoevaluatePGE2levelsinconditionedmedia.Cells wereplatedin24wellplatesovernightandincubatedfor24hourswithincreasingconcentrationsofapricoxib inlowserummedium.Assayswereperformedintriplicateandwereperformedaminimumofthreetimes.

AnimalStudies.Allanimalswerehousedinapathogenfreefacilitywith24houraccesstofoodandwater.

Experimentswereapprovedby,andperformedinaccordancewith,theIACUCatUTSouthwestern(Dallas,TX).

46 weeks old female NOD/SCID mice were obtained from a campus supplier. 1x106 AsPc1, Colo357, and

HPAFIIcellswereinjectedorthotopicallyasdescribed(30)andtumorgrowthmonitoredbyultrasound.Mice withestablishedtumorswererandomizedtoreceiveeithergemcitabine25mg/kgtwiceweeklypluserlotinib

100μgdaily(standardofcare),orstandardofcareplus10mg/kgor30mg/kgapricoxibdailybyoralgavage.

MicebearingColo357andAsPc1tumorsreceived3weeksoftherapypriortosacrifice.AnimalsbearingHPAF

IItumorsweredividedintoanearlysacrificeat3weeksandalatesacrificeat7weeksoftherapy.Primary tumorburdenwasestablishedbyweighingpancreasandtumorenbloc.Metastaticincidencewasdetermined byvisualinspectionoftheliverandabdominalcavityaswellasbyquantitationofH&Eliversections.Tissues werefixedin10%formalinorsnapfrozeninliquidnitrogenforfurtherstudies.

Histologyandtissueanalysis.Formalinfixedtissueswereembeddedinparaffinandcutin10micronsections.

Sections were evaluated by H&E and immunohistochemical analysis using antibodies CD31 (Dianova), NG2

(Millipore),vimentin (Phosphosolutions),endomucin,Zeb1,Ecadherin,PCNA (SantaCruz),phosphohistone

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page8

H3 (Upstate), TUNEL (Promega), cleaved caspase3 (Cell Signaling), VEGF, and COX2 (Abcam). Negative controls included omission of primary antibody and immunofluorescent evaluation was performed as described(31).HumanandmouseVEGFlevelsinplasmaandtumorlysatesweredeterminedbyELISA(R&D

Systems)permanufacturerinstructions.

Statistics.DatawereanalyzedusingGraphPadsoftware(GraphPadPrismversion4.00forWindows;GraphPad

Software,SanDiego,CA,www.graphpad.com).Resultsareexpressedasmean±SEM.Datawasanalyzedbyt testorANOVAandresultsareconsideredsignificantatp<0.05.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page9

Results HumanpancreaticcancercelllinesvaryinexpressionofCOX2andEGFRandshowdifferentialresponseto inhibition.

FivehumanpancreaticcancercelllineswerescreenedforbaselineexpressionofCOX2andEGFRbyqPCR.All cell lines expressed detectable levels of mRNA for EGFR and COX2. There was generally close accordance betweenmessageandprotein,exceptCOX2levelsinAsPC1cellsthatweredetectable(albeitbarely)byqPCR

(Fig.1A)butnotattheproteinlevel(Fig.1B).Colo357cellsdemonstratedmoderatetohighexpressionofCOX

2 at the mRNA and protein level (Fig. 1A, B). PL45, HPAFII, and CFPAC1 cells exhibited low to moderate expressionofCOX2atthemRNAandproteinlevel(Fig.1A,B).EGFRexpressionwasdetectableinallcelllines by qPCR. The majority of cells displayed low to moderate expression with the exception of AsPC1, which demonstrated relatively high expression (Fig. 1A). Expression of EGFR at the protein level was assessed by

Western blot and confirmed robust expression of EGFR in AsPC1 cells. However, it was notable that phosphorylatedEGFR(pEGFR)wasweakornotdetectedinthemajorityofcelllinesunderbasalconditions.In contrasttherewasastrongpEGFRsignalinColo357cells(Fig.1B).Theefficacyoferlotinibinhibitionofp

EGFRafterstimulationwithEGFineachcelllinewasdeterminedbyELISA(Fig.1C).AsPC1andHPAFIIcells weremostsensitivewithIC50sof0.1μMand0.2μM,respectively.AllcellsdemonstratedanIC50belowthe pharmacologicrange(2.2μM)withtheexceptionofPL45cells,whichwereresistanttoerlotinibatmaximum dose(500μM,Fig.1C).Todeterminetheeffectofapricoxib,conditionedmediaofcellsincubatedfor24hours withapricoxibwascollectedandPGE2levelsdetermined.BaselinelevelsofPGE2productionbyAsPC1and

PL45 cells was minimal at <10 pg/ml (below detectable range) and remained unchanged by apricoxib treatment(AsPC1shown,Fig.1D).ApricoxibeffectivelypreventedPGE2productionbyColo357andHPAFII cellswithIC50swithinthepharmacologicrange(0.52μM,Colo357,showninFig.1D).CFPAC1displayeda dosedependentreductionbutdidnotachieveanIC50withinthetestedrange(max1μM,datanotshown).

From this we concluded that all cell lines except PL45 demonstrate inhibition of EGFR activity in vitro at

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page10 pharmacologicallyrelevantconcentrationsofdrug;however,whilemostcelllinesexpressCOX2,thisdidnot directlycorrelatetolevelofPGE2production.ForcelllineswithdetectablePGE2expression,apricoxibinhibited itsproductionatnanomolarconcentrationsinvitro.

Erlotinibandapricoxibcombinationtherapyaugmentsefficacyofgemcitabineinvitro.

MTS assays were used to evaluate the antiproliferative effect of each drug as a single agent and in combination. As shown in Table 1, the IC50 for erlotinib and apricoxib as single agents was above pharmacologic range for all cell lines. AsPC1, Su.86.86 and HPAFII demonstrated the least sensitivity to apricoxibasasingleagent,withIC50valuesof7080μM.Celllinesinsensitivetogemcitabine(IC50notachieved with maximum dose 2000 nM) included AsPC1, HPAFII, and Su.86.86. Highly gemcitabine sensitive lines includedPL45andCFPAC1.Colo357demonstratedanintermediateantiproliferativeresponsewithanIC50of

150μM.Additionof0.1μMapricoxibdidnotsignificantlyalterresponsetogemcitabineinanycelllineother than Colo357, which demonstrated 1.3fold reduction in gemcitabine IC50. Apricoxib at 1 μM enhanced the responseofAsPC1,HPAFII,andColo357togemcitabinebuthadlittleeffectongemcitabineactivityinthe remaining lines. Erlotinib augmented response to gemcitabine in all lines evaluated. Triple combination of gemcitabine,1μMerlotinib,and1μMapricoxibshowedmeasurablereductionofgemcitabineIC50inallcell lines(Table1);sensitizationwasparticularlymarkedinHPAFIIcells,wherethegemcitabineIC50wasreduced byoverthreeordersofmagnitudebythecombinationofapricoxibanderlotinib.Colo357andHPAFII,aslines expressing COX2 and demonstrating sensitization to gemcitabine with COX2 inhibition, were selected for furtherinvivostudiesinadditiontoAsPC1,aCOX2negativecellline.

InhibitionofCOX2significantlyreducesprimarytumorgrowthinvivoandmetastaticincidenceincelllines withhighCOX2expression.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page11

Fromtheinvitrodata,weselectedaCOX2negativecellline,AsPC1,andtwoCOX2positivecelllines,one withminimalerlotinibresponse,Colo357,andahighlyerlotinibsensitiveline,HPAFIIforinvivostudies.Mice begantherapy1014dayspostorthotopictumorcellinjection.Therapyconsistedofcontrol(vehiclealone), standardtherapy,i.e.25mg/kggemcitabine+100μgerlotinib(G+E),orG+E+apricoxibat10or30mg/kg.

AsPC1andColo357tumorbearingmiceweresacrificedafter3weeksoftherapy.Tumorandpancreaswere dissectedenbloctodeterminetumorweight.Metastaticburdenwasinitiallysurveyedbyinspectionofthe abdominalcavity,diaphragm,andliverandlaterquantifiedbyH&Estainingoftheleftlobeoftheliver.For

AsPC1tumors,additionofapricoxibresultedinasignificantdecreaseinprimarytumorweightcomparedto controltreatedanimals(p<0.05,twotailedttest,MannWhitney);however,theeffectwasequivalenttothe standardofcare,demonstratingthattheantitumoractivitywasduetogemcitabineanderlotinibinthismodel

(Fig.2A).MetastaticburdeninAsPC1bearingmicewashighlyvariablewithnosignificantdifferencesbetween groups(Fig.2B).MicebearingColo357tumorsdemonstratedasignificantreductioninprimarytumorsizewith the standard of care alone compared to saline (p<0.05, ANOVA) and the effect was further enhanced by apricoxib treatment (p<0.05, ANOVA). Metastases in animals bearing Colo357 were virtually eliminated in apricoxibtreatedgroups,with10mg/kgshowinggreatestresponse(p<0.005comparedtoG+E,DunnMCT,

Fig. 2B). Mice bearing HPAFII tumors received therapy for 3 weeks, at which point saline controls were moribundandrequiredsacrifice.HPAFIItumorswerehighlysensitivetoerlotinibtreatmentandatthistime alltreatmentgroupshadvirtuallynotumorgrowth,thusitwaselectedtosacrificeonly3animals/groupfor comparisontocontrolandallowremainingmicetocontinueontherapy.Therewerenodifferencesintumor burdenamongsttreatmentgroupsatthe3weektimepointwithallfinalweightsbeingequivalenttonormal pancreas weight. The remaining animals continued on therapy for an additional 4 weeks, at which point apricoxibtreated animals showed superior tumor control rate compared to G+E (p<0.001, ANOVA).

MetastasesintheHPAFIImodelweresuppressedsignificantlyinalltreatmentgroupscomparedtocontrol animals,butagainapricoxibenhancedtheactivityofG+E,abolishingmetastaticspreadaltogether(Fig.2A,B).

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page12

Given thesmallprimarytumorburdenofHPAFII,whichlargelyconsisted ofnormalpancreas,only

Colo357andAsPC1tumorswereusedforfurtherevaluation.Paraffinembeddedtumorsectionswereusedto analyzeCOX2expressionbyimmunofluorescence.

COX2expressioncanbeinducedinvivoandinvitrouponEMTinaCOX2negativecellline

Notably, while AsPC1 cells did not express measurable levels of COX2 in vitro, untreated AsPC1 xenograftsstainedpositiveforCOX2,althoughtoafarlesserextentthanColo357tumors.InteractionofEGFR andCOX2signaling(24)issupportedbyaclearreductioninCOX2expressionbyG+Etreatment,althoughthis valuewasnotstatisticallysignificantineithergroup.ApricoxibtreatmentvirtuallyeliminatedCOX2expression intumorsfrombothcelllines.PGE2israpidlymetabolizedinplasmaandthereforedifficulttoassessinvivo.

However,PGE2 stimulatesCOX2expression,formingapositivefeedbackloop,soreductioninoverallCOX2 levels demonstrates that apricoxib effectively inhibits PGE2 production in vivo (32, 33). In COX2 expressing tumors,thisresultedinincreasedefficacyofstandardtherapy(Fig2.A,B).IthasbeenreportedthataCOX2 nonexpressing cell line can upregulate COX2 under conditions of stress. To recapitulate in vivo changes resultingfromtumorhostinteractions,PGE2productionwasevaluatedafterforcedEMT(26).InColo357,PGE2 production was not significantly different between normal and EMT conditions at baseline. At 24 hours followingasingledoseofapricoxib(0.5μM),representingthelowestrangeofinvivolevels(26),PGE2levels had decreased by 50% for both conditions. AsPC1 cells, which produce no measurable amount of PGE2at baseline,demonstratedasignificantincreaseinPGE2levelsfollowinginductionofEMT.IncontrasttoColo357, thiswasnotaffectedbyapricoxib(Fig2.E).

InhibitionofCOX2attenuatesVEGFandpromotesvascularnormalizationinvivo

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page13

COX2 expression correlates with VEGF levels in patient tumor samples and COX2 inhibition can modulate

VEGFproduction(27,34).VEGFlevelsinplasmaandtumorsamplescollectedfrommicetumorbearingmice weremeasuredbyELISA.InAsPC1tumorbearingmice,plasmaVEGFlevelsweredecreasedinalltreatment groups,butthesedifferenceswerenotsignificantlydifferentfromcontroltreatedanimals.InColo357tumor bearingmice,VEGFlevelswereunaffectedbyG+Etreatmentbutadditionofapricoxibresultedindepletionof humanVEGFfromplasmasamples(Fig.3A).Interestingly,thesechangesdidnotcorrelatewithintratumoral levelsofhumanVEGF.Infact,thelowestlevelsoftumorassociatedVEGFwerefoundinG+Etreatedanimals

(NSforAsPC1tumors,p<.005inColo357tumors,Fig.3B).

To assess the ultimate effect of COX2 inhibition on angiogenesis, tumor microvessel density was evaluated by immunofluorescence for endomucin and CD31 (CD31 shown, Fig. 3C, D). Vessel density was decreasedmoderatelybyG+Ealone(NSforColo357buttrendingdownward,p<0.05forAsPC1);however, additionofapricoxibtothetreatmentregimendidnotsignificantlyaltertheeffectofG+Eonvesseldensity

(Fig.3C).Todetermineiftherapyaffectedpericyteendothelialcellinteraction,NG2,apericytemarker,was colocalizedwithendomucinorCD31toassesspericytecoverageofvessels.Pericytecoverageindex(%NG2 positivevessels)increasedsignificantlyinapricoxibtreatedAsPC1andColo357tumors(p<0.001AsPC1,<0.05

Colo357,ANOVA,Fig.3C,D).Insummary,COX2inhibitiondidalterplasmalevelsofVEGFbutthisdidnot translate to a change in levels of tumor associated VEGF or neovascularization, instead COX2 inhibition promotedvascularstabilization,which mayimprovedrugdeliveryaswellascontributetothereductionof metastases(35).

InhibitionofCOX2reducesproliferationandincreasesapoptosis.

InhibitionofCOX2activitywithapricoxibenhancedtheantitumoreffectofG+E.Todetermineifthiswasdue tochangesincellproliferationorsurvivalweevaluatedmarkersofapoptosisandcellproliferationinAsPC1 andColo357tumors.The levelofapoptosisintumorsfromcontrolandtreatedanimalswasdeterminedby

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page14

TUNEL (Fig. 4A, B). Apricoxib significantly increased the number of apoptotic cells in AsPC1 and Colo357 tumors.TheincreaseinTUNELwasmostevidentatthe10mg/kgdose(Fig.4A),butanimalstreatedwith30 mg/kgofapricoxibshowedthehighestlevelsofcleavedcaspase3levels(datanotshown).InAsPC1tumors, proliferative markers (phosphohistone H3 and PCNA) were unchanged (data not shown and Fig. 4C). By contrast,apricoxibstronglyenhancedthemodestantiproliferativeeffectofG+EinColo357tumors,especially atthe30mg/kgdose(p<0.01,Fig.4C,D).Tosubstantiatetheeffectofapricoxibontheefficacyofstandard therapyweevaluatedH2AXlevels,whichhavebeenshowntomarkgemcitabineinducedstalledreplication forks(36).yH2AXexpressionwasevaluatedbyimmunofluorescenceinColo357tumorsectionsandfoundto be significantly increased in apricoxib treated tumors compared to either control or G+E treatment alone

(p<0.05,Fig.4E).

COX2inhibitionreversesEMT

WehaverecentlyfoundthatapricoxibreversesEMTinHT29xenograft(26);thusweassessedtumorsections in the current study for epithelial (Ecadherin) and mesenchymal (vimentin, and Zeb1) markers by immunofluorescence. In Colo357 tumors, vimentin and Zeb1 expression decreased in a dosedependent fashion in apricoxibtreated animals compared to control and G+E groups, although changes in vimentin expression did not reach statistical significance (p<0.05 vs. control, G+E for Zeb1 Fig. 5A). Conversely, E cadherin was strongly induced by apricoxib treatment (p<0.001 vs. control, G+E, Fig. 5A). Representative doublestainedimagesareshownforvimentinandEcadherininColo357tumors(Fig.5B).UntreatedColo357 tumors demonstrated a mesenchymal phenotype with strong vimentin positive staining and negligible E

Cadherinexpression.Bycomparison,Colo357cellsinvitrodemonstrateanepithelialphenotypewhichappears preserved (or reacquired) after apricoxib treatment (Fig. 5C). Treatment of cells in culture with G+E alone actually increased Zeb1 expression, but combination with apricoxib reversed this effect and dramatically upregulatedECadherinexpression,(Fig.5C).

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page15

InhibitionofCOX2resultedinashifttowardamoreepithelialphenotyperegardlessofbaselineCOX2 status of the tumor; although this effect was more dramatic in high COX2 expressing Colo357 tumors and correlatedwiththeoverallreductionofmetastaticincidence.AsPC1cells,whichincreasedCOX2expression invivoandfollowingEMTinvitro(Fig2E),demonstratethisshifttowardepithelialphenotypebyapricoxib treatmentwithdecreasedvimentinandZeb1anddramaticallyincreasedECadherinexpression(Fig.5D).In vitro,Zeb1expressionbyAsPC1cellsisunchangedafterG+Eorcombinationwithapricoxib.Asthesecells haveundetectableCOX2expressionandPGE2productionundernormalcultureconditions,EMTwasinduced inthesecellstorecapitulateinvivochanges.FollowingEMT,apricoxibtreatmenteitherincombinationwith

G+Eorasasingleagentat1μMsignificantlydiminishedZeb1expression,thisshiftdidnotoccurwithG+E alone,suggestingthatCOX2isintimatelyinvolvedwiththeoccurrenceofEMTandthattransformedcellsthat undergoEMTdemonstrateincreasedsensitivitytoapricoxib.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page16

Discussion

Resistancetochemotherapyremainsamajorchallengeinthetreatmentofpancreaticcancer,combination strategiestoaugmentgemcitabinefailedtodemonstrateimprovedoverallsurvivalinphaseIIIclinicaltrials priortoadditionoferlotinib(20).EarlyresultsofsmallphaseIIstudiescombiningtheCOX2inhibitorcelecoxib with gemcitabine or gemcitabine plus irinotecan showed promise with prolonged survival compared to the historicaveragesurvivalof6monthsforgemcitabine(23).ApricoxibiscurrentlyinPhaseIIclinicaltrialsin

NSCLC and pancreatic cancer. Our studies aimed to determine the antitumor activity of apricoxib in molecularly defined preclinical pancreatic cancer models with the hope of identifying subsets of pancreatic cancerpatientswhowouldbenefitfromtheadditionofCOX2inhibitiontocurrenttherapy.

Invitro,wedeterminedthemRNAandproteinexpressionofEGFRandCOX2incancercelllinesand measured how these targets contribute to cellular response to gemcitabine. We found in all cell lines that concentrationsofapricoxibrequiredtoexertdirectantitumoractivityfarexceededthoseneededtoeliminate

PGE2 production (0.52 μM). However, apricoxib at pharmacologically achievable concentrations sensitized cells to standard therapy. This suggests that antitumor effects of COX2 inhibition in these models in vivo resulted primarily from sensitization to gemcitabine/erlotinib therapy or modification of tumorhost interactionsasopposedtodirectantitumorcelleffects,althoughitispossiblethatCOX2dependentlines would have responded under anchorageindependent growth conditions (26). This is alsosupported by the increased production of PGE2 in vitro by AsPC1 following forced EMT corresponding to increased COX2 expressionbycellsinvivowhichhaveadoptedanincreasinglymesenchymalphenotype.

In vivo, in mice bearing tumors derived from AsPC1, a COX2 lowexpressing cell line, addition of apricoxibtostandardtherapydidnotimproveantitumoractivitynordramaticallyeffectcellproliferationas measuredbyPCNAexpression.Bycontrast,incelllinesthatexpressmoderatetohighlevelsofCOX2and producehighlevelsofPGE2,significantlyincreasedefficacywasachievedbytheadditionofapricoxib.These results suggest that clinical studies should consider COX2 activity, not simply expression, when evaluating

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page17

apricoxib or other COX2 inhibitors in cancer patients. Assaying the level of PGEM, a PGE2 metabolite, is a potentialstrategyforpatientstratification.

COX2 and PGE2 are strongly linked to angiogenesis via promotion of VEGF and bFGF production stimulating growth, migration, and survival of endothelial cells (27, 34). Reciprocally, these factors form a positivefeedbackloopamplifyingtheproductionofCOX2.Inourstudies,apricoxibdidmodulateVEGFlevels inplasmabutthisdidnotresultinareductioninmicrovesseldensity.Inalltumorgroups,VEGFlevelswere observedtobelowestintumorsthatreceivedstandard(G+E)therapy.HighexpressionofCOX2corresponds to high VEGF levels in tumor specimens; however, studies have reported that while VEGF production is reduced initially by COX2 inhibition, its production is not exclusively COX2 dependent. Recovery and amplificationofVEGFlevelsmayoccurasacompensatoryresponsetolossofCOX2activityandlevelsofVEGF continue to increase with higher doses of COX2 inhibitors (15, 37, 38). This may provide a plausible explanationfortheparadoxicaldoseresponseinourmodels,aswellasotherpreclinicalstudies.Furtherthese datamightalsohelpexplainthefailureofCOX2inhibitorstodemonstratesignificantimprovementinclinical studiesthatpreviouslyemployeddosestwicethatusedforanalgesicandantiinflammatoryeffect(39,40).

Reciprocally, mechanisms of antiVEGF resistance may involve induction of other proangiogenic cytokines, suchasIL1andIL8thatareassociatedwithincreasedCOX2productioninthetumormicroenvironment.

ThiscollateralpathwaymayexplainwhyCOX2inhibitionalonefailedtobesignificantlyantiangiogenicinthis model.

Microvesseldensitywasreducedinalltherapygroupsreceivingstandardtherapyandwasunchanged by the addition of apricoxib. The decrease in microvessel density by treatment with gemcitabine has been reportedpreviously(41)andmaybearesultofthedosingscheduleemployed.However,apricoxibtreatment resultedinincreasedpericytecoverageofbloodvessels.Pericyteattachmentiscriticalforstabilizingvascular structures(35).Absentorloosepericyteattachmentresultsfromimbalancedangiogenicsignalinginthetumor leading to dysfunctional vessels that are hallmarked by hyperpermeability. These vessels display abnormal

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page18 bloodflowthancanincreasetumorhypoxia,reducedeliveryofchemotherapyandfacilitateextravasationand hematogenousspreadofmetastaticcells(35).COX2inhibitionimpededthisprocess,suggestingthatCOX2 participatesinvascularremodelinginthetumormicroenvironment.

TheCOX2productPGE2canimpacttumorprogressionandcancercellproliferationbyactivatingthe

RasMAPK signaling cascade, which subsequently increases expression of PGE2 synthase, forming a positive feedbackloop(42,43).Additionally,exogenousPGE2canstimulateproliferationofCOX2negativecellsinvitro

(17). PGE2 can mediate cell survival by inducing expression of antiapoptotic proteins such as Bcl2 and increasingNFBtranscriptionalactivity(44).ChemotherapycanincreaseCOX2proteinexpressionandPGE2 production,drivingtumorcellsurvivalandresistancetotherapy.Inthepresentstudy,proliferativeactivityin vivo was not affected by COX2 inhibition in AsPC1 tumors; however, apricoxib significantly reduced proliferationinColo357tumors.ApoptosisresultingfromCOX2inhibition,alternatively,wasnotspecificto

COX2 status of the tumor and in each group was most profound in the 10 mg/kg group; indicating that adaptationofAsPC1cellsinvivocanleadtoincreaseddependencyonCOX2.H2AXhasbeenimplicatedasa markerofgemcitabinespecificDNAdamageandwasfoundtobesignificantlyelevatedinapricoxibtreated tumors. This finding correlated with improvement in apoptotic activity and vessel maturity, indicating that improvedchemotherapeuticresponsemayberelatedinparttoimproveddrugdelivery.

COX2andPGE2areimplicatedindrivingEMT(45,46),whichcontributestometastasisandresistance to chemotherapy (47, 48). We found that untreated Colo357 and AsPC1 tumors displayed a robust EMT phenotype. In contrast, tumorbearing mice that received apricoxib had a noticeable shift to an epithelial phenotype. These observations suggest that COX2 inhibition can reverse EMT in pancreatic tumors. PGE2 directlyinducesthetranscriptionfactorZeb1andenhancesitsbindingtotheproximaleboxoftheEcadherin promoter,resultingindownregulationofEcadherin.Ecadherinisessentialforintercellularadhesion,such thatdisruptionfacilitatesmigration,invasion,andmetastasis(5,49).Clinicaltumorsampleshaveshownan inverse relationship between COX2 expression/high grade tumor type and Ecadherin expression (49).

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page19

Vimentin, a marker of mesenchymal differentiation, is also highly expressed in pancreatic cancers and correlateswithpoorprognosis(47).Notably,gemcitabineresistancehasbeenlinkedtovimentinexpressionin pancreaticcancercells(50).Invitro,apricoxibdramaticallyincreasedECadherinexpressioninColo357cellsas wellasreversedtheincreasedZeb1productionobservedafterG+Etreatment.Unsurprisingly,noaffectwas seeninAsPC1cellsatbaselineastheyhaveminimalbaselineCOX2activity.However,theincreasedCOX2 production with forced EMT greatly sensitized these cells to apricoxib and demonstrated that apricoxib effectivelyreversedEMT.Thissuggeststhatthemaintenanceofthemesenchymalphenotypeinthesecells reliesinpartontheactivityofCOX2.Invivo,minimalchangewasseeninthemesenchymalphenotypeof

Colo357tumorswithstandardtherapy,whileG+EincreasedZeb1andvimentininAsPC1tumors.Additionof apricoxibsignificantlyshiftedAsPC1tumorstowardsamoreepithelialphenotype,althoughitshouldbenoted thatAsPC1tumorsaretypicallyepithelialatbaseline.Thismayexplainwhytheseshifts,whilepresent,may nothaveanoverwhelmingimpactonoveralltumorresponse.Importantly,Colo357tumorsdemonstrateda dosedependentmesenchymaltoepithelialtransition(MET).Whilethisdidnottranslateintoareductionin metastaticburdenforAsPC1tumors,theeffectwasdramaticinColo357tumorbearingmice,implicatingCOX

2asimportantingoverningEMTandsensitizationtostandardtherapyinpancreaticcancer.

In summary, our findings demonstrate that the clinical COX2 inhibitor, apricoxib, enhances the efficacy of standard chemotherapy in preclinical models of pancreatic cancer. Inhibition of COX2mediated

PGE2productionenhancedtheantiproliferativeandapoptoticeffectofstandardtherapyinCOX2dependent tumorsandaffectedthetumormicroenvironmentbypromotingvascularnormalizationandattenuatingEMT.

Further clinical evaluation of apricoxib in a molecularly selected patient population is warranted in the developmentofstrategiestoimprovetreatmentforpancreaticcancer.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page20

DisclosureofPotentialConflictsofInterest

R.A.Brekken:commercialresearchgrantfromTragaraPharmaceuticals,Inc

F.J.BurrowsandS.Zaknoen:employeesofTragaraPharmaceuticals,Inc

Acknowledgments

WethankSuryaVadrevufortechnicalassistanceandDrs.JoanSchiller,DavidH.JohnsonandJohnMinnaand membersoftheBrekkenlabforadviceandthoughtfuldiscussion.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page21

REFERENCES 1. Sobolewski C, Cerella C, Dicato M, Ghibelli L, Diederich M. The role of cyclooxygenase2 in cell proliferationandcelldeathinhumanmalignancies.IntJCellBiol2010;2010:215158.

2. Harris RE, BeebeDonk J, Doss H, Burr Doss D. , , and other nonsteroidal anti inflammatorydrugsincancerprevention:acriticalreviewofnonselectiveCOX2blockade(review).OncolRep 2005;13:55983.

3. Dannenberg AJ, Subbaramaiah K. Targeting cyclooxygenase2 in human neoplasia: rationale and promise.CancerCell2003;4:4316.

4. Williams CS, Mann M, DuBois RN. The role of in inflammation, cancer, and development.Oncogene1999;18:790816.

5. Dohadwala M, Yang SC, Luo J, Sharma S, Batra RK, Huang M, et al. Cyclooxygenase2dependent regulationofEcadherin:prostaglandinE(2)inducestranscriptionalrepressorsZEB1andsnailinnonsmallcell lungcancer.CancerRes2006;66:533845.

6. DeerEL,GonzalezHernandezJ,CoursenJD,SheaJE,NgatiaJ,ScaifeCL,etal.Phenotypeandgenotype ofpancreaticcancercelllines.Pancreas2010;39:42535.

7. Hasan S, Satake M, Dawson DW, Funahashi H, Angst E, Go VL, et al. Expression analysis of the prostaglandinE2productionpathwayinhumanpancreaticcancers.Pancreas2008;37:1217.

8. Ito H, Duxbury M, Benoit E, Clancy TE, Zinner MJ, Ashley SW, et al. Prostaglandin E2 enhances pancreaticcancerinvasivenessthroughanEts1dependentinductionofmatrixmetalloproteinase2.Cancer Res2004;64:743946.

9. KoshibaT,HosotaniR,MiyamotoY,WadaM,LeeJU,FujimotoK,etal.Immunohistochemicalanalysis ofcyclooxygenase2expressioninpancreatictumors.IntJPancreatol1999;26:6976.

10. Okami J, Yamamoto H, Fujiwara Y, Tsujie M, Kondo M, Noura S, et al. Overexpression of cyclooxygenase2incarcinomaofthepancreas.ClinCancerRes1999;5:201824.

11. GatelyS.Thecontributionsofcyclooxygenase2totumorangiogenesis.CancerMetastasisRev2000; 19:1927.

12. Merati K, said Siadaty M, Andea A, Sarkar F, BenJosef E, Mohammad R, et al. Expression of inflammatory modulator COX2 in pancreatic ductal adenocarcinoma and its relationship to pathologic and clinicalparameters.AmJClinOncol2001;24:44752.

13. TuckerON,DannenbergAJ,YangEK,FaheyTJ,3rd.Bileacidsinducecyclooxygenase2expressionin humanpancreaticcancercelllines.Carcinogenesis2004;25:41923.

14. YipSchneider MT, Barnard DS, Billings SD, Cheng L, Heilman DK, Lin A, et al. Cyclooxygenase2 expressioninhumanpancreaticadenocarcinomas.Carcinogenesis2000;21:13946.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page22

15. EiblG,BruemmerD,OkadaY,DuffyJP,LawRE,ReberHA,etal.PGE(2)isgeneratedbyspecificCOX2 activityandincreasesVEGFproductioninCOX2expressinghumanpancreaticcancercells.BiochemBiophys ResCommun2003;306:88797.

16. TsengWW,DeganuttiA,ChenMN,SaxtonRE,LiuCD.Selectivecyclooxygenase2inhibitor (Vioxx)inducesexpressionofcellcyclearrestgenesandslowstumorgrowthinhumanpancreaticcancer.J GastrointestSurg2002;6:83843;discussion44.

17. OmuraN,GriffithM,VincentA,LiA,HongSM,WalterK,etal.Cyclooxygenasedeficientpancreatic cancercellsuseexogenoussourcesofprostaglandins.MolCancerRes2010;8:82132.

18. BardeesyN,DePinhoRA.Pancreaticcancerbiologyandgenetics.NatRevCancer2002;2:897909.

19. ElRayesBF,ZalupskiMM,ShieldsAF,FerrisAM,VaishampayanU,HeilbrunLK,etal.AphaseIIstudy ofcelecoxib,gemcitabine,andcisplatininadvancedpancreaticcancer.InvestNewDrugs2005;23:58390.

20. Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, et al. Erlotinib plus gemcitabine comparedwithgemcitabinealoneinpatientswithadvancedpancreaticcancer:aphaseIIItrialoftheNational CancerInstituteofCanadaClinicalTrialsGroup.JClinOncol2007;25:19606.

21. ChoeMS,ZhangX,ShinHJ,ShinDM,ChenZG.Interactionbetweenepidermalgrowthfactorreceptor and cyclooxygenase 2mediated pathways and its implications for the chemoprevention of head and neck cancer.MolCancerTher2005;4:144855.

22. ReckampK,GitlitzB,ChenLC,PatelR,MilneG,SytoM,etal.BiomarkerbasedphaseIdoseescalation, pharmacokinetic, and pharmacodynamic study of oral apricoxib in combination with erlotinib in advanced nonsmallcelllungcancer.Cancer2011;117:80918.

23. Lipton A, CampbellBaird C, Witters L, Harvey H, Ali S. Phase II trial of gemcitabine, irinotecan, and celecoxibinpatientswithadvancedpancreaticcancer.JClinGastroenterol2010;44:2868.

24. GadgeelSM,AliS,PhilipPA,AhmedF,WozniakA,SarkarFH.Responsetodualblockadeofepidermal growth factor receptor (EGFR) and cycloxygenase2 in nonsmall cell lung cancer may be dependent on the EGFRmutationalstatusofthetumor.Cancer2007;110:277584.

25. ZhangX,ChenZG,ChoeMS,LinY,SunSY,WieandHS,etal.Tumorgrowthinhibitionbysimultaneously blockingepidermalgrowthfactorreceptorandcyclooxygenase2inaxenograftmodel.ClinCancerRes2005; 11:62619.

26. KiraneAR,ToombsJE,LarsenJE,OstapoffKT,MeshawKR,ZaknoenS,etal.Epithelialmesenchymal transitionincreasestumorsensitivitytoCOX2inhibitionbyapricoxib.Carcinogenesis2012.

27. Senzaki M, Ishida S, Yada A, Hanai M, Fujiwara K, Inoue S, et al. CS706, a novel cyclooxygenase2 selectiveinhibitor,prolongedthesurvivaloftumorbearingmicewhentreatedaloneorincombinationwith antitumorchemotherapeuticagents.IntJCancer2008;122:138490.

28. UshiyamaS, YamadaT, MurakamiY,Kumakura S,InoueS,SuzukiK,etal.Preclinicalpharmacology profile of CS706, a novel cyclooxygenase2 selective inhibitor, with potent antinociceptive and anti inflammatoryeffects.EurJPharmacol2008;578:7686.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page23

29. Schmittgen TD, Livak KJ. Analyzing realtime PCR data by the comparative C(T) method. Nat Protoc 2008;3:11018.

30. Dineen SP, Roland CL, Greer R, Carbon JG, Toombs JE, Gupta P, et al. Smac mimetic increases chemotherapyresponseandimprovessurvivalinmicewithpancreaticcancer.CancerRes2010;70:285261.

31. Arnold SA, Rivera LB, Miller AF, Carbon JG, Dineen SP, Xie Y, et al. Lack of host SPARC enhances vascularfunctionandtumorspreadinanorthotopicmurinemodelofpancreaticcarcinoma.DisModelMech 2010;3:5772.

32. WangD,DuboisRN.Eicosanoidsandcancer.NatRevCancer2010;10:18193.

33. WangD,DuboisRN.Prostaglandinsandcancer.Gut2006;55:11522.

34. deGrootDJ,deVriesEG,GroenHJ,deJongS.Nonsteroidalantiinflammatorydrugstopotentiate chemotherapyeffects:fromlabtoclinic.CritRevOncolHematol2007;61:5269.

35. Carmeliet P, Jain RK. Principles and mechanisms of vessel normalization for cancer and other angiogenicdiseases.NatRevDrugDiscov2011;10:41727.

36. EwaldB,SampathD,PlunkettW.H2AXphosphorylationmarksgemcitabineinducedstalledreplication forksandtheircollapseuponSphasecheckpointabrogation.MolCancerTher2007;6:123948.

37. XuK,GaoH,ShuHK.Celecoxibcaninducevascularendothelialgrowthfactorexpressionandtumor angiogenesis.MolCancerTher2011;10:13847.

38. ZhuYM,AzahriNS,YuDC,WollPJ.EffectsofCOX2inhibitiononexpressionofvascularendothelial growthfactorandinterleukin8inlungcancercells.BMCCancer2008;8:218.

39. Edelman MJ, Watson D, Wang X, Morrison C, Kratzke RA, Jewell S, et al. Eicosanoid modulation in advanced lung cancer: cyclooxygenase2 expression is a positive predictive factor for celecoxib + chemotherapyCancerandLeukemiaGroupBTrial30203.JClinOncol2008;26:84855.

40. Ferrari V, Valcamonico F, Amoroso V, Simoncini E, Vassalli L, Marpicati P, et al. Gemcitabine plus celecoxib(GECO)inadvancedpancreaticcancer:aphaseIItrial.CancerChemotherPharmacol2006;57:185 90.

41. Korpanty G, Carbon JG, Grayburn PA, Fleming JB, Brekken RA. Monitoring response to anticancer therapybytargetingmicrobubblestotumorvasculature.ClinCancerRes2007;13:32330.

42. KatohH,HosonoK,ItoY,SuzukiT,OgawaY,KuboH,etal.COX2andprostaglandinEP3/EP4signaling regulate the tumor stromal proangiogenic microenvironment via CXCL12CXCR4 chemokine systems. Am J Pathol2010;176:146983.

43. WangMT,HonnKV,NieD.Cyclooxygenases,,andtumorprogression.CancerMetastasis Rev2007;26:52534.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page24

44. Yao M, Lam EC, Kelly CR, Zhou W, Wolfe MM. Cyclooxygenase2 selective inhibition with NS398 suppressesproliferationandinvasivenessanddelayslivermetastasisincolorectalcancer.BrJCancer2004; 90:7129.

45. Jang TJ, Jeon KH, Jung KH. Cyclooxygenase2 expression is related to the epithelialtomesenchymal transitioninhumancoloncancers.YonseiMedJ2009;50:81824.

46. Neil JR, Johnson KM, Nemenoff RA, Schiemann WP. Cox2 inactivates Smad signaling and enhances EMTstimulatedbyTGFbetathroughaPGE2dependentmechanisms.Carcinogenesis2008;29:222735.

47. HandraLuca A, Hong SM, Walter K, Wolfgang C, Hruban R, Goggins M. Tumour epithelial vimentin expressionandoutcomeofpancreaticductaladenocarcinomas.BrJCancer2011;104:1296302.

48. Arumugam T, Ramachandran V, Fournier KF, Wang H, Marquis L, Abbruzzese JL, et al. Epithelial to mesenchymaltransitioncontributestodrugresistanceinpancreaticcancer.CancerRes2009;69:58208.

49. TsujiiM,DuBoisRN.Alterationsincellularadhesionandapoptosisinepithelialcellsoverexpressing prostaglandinendoperoxidesynthase2.Cell1995;83:493501.

50. TraubP,PeridesG,ScherbarthA,TraubU.Tenaciousbindingoflipidstovimentinduringitsisolation andpurificationfromEhrlichascitestumorcells.FEBSLett1985;193:21721.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page25

Table1.Apricoxibenhancesinvitrosensitivitytogemcitabine Cellline Apricoxib Erlotinib Gemcitabine Gemcitabine(nM) Fold (μM) (μM) (nM) 1μME 0.1μMA 1μMA 1μMA/E change 1630 830 ASPC1 70(16) 375(15) 2000(0) 2000(0) 537(200) 3.7 (319) (210) Colo357 30(14) 57(10) 158(51) 92(7) 116(31) 98(16) 52(8) 2.9 172 400 HPAFII 80(1) 400(0) 2000(0) 2000(0) 13(2) 13.2 (32) (23) CFPAC1 19(1) 4.3(1.3) 2.7(1) ND 2.1(0.1) 2(0.3) 0.72(0.5) 3.75 9.1 PL45 26(15) 400(0) 12.8(6) ND 9.6(1) 7(0.6) 1.8 (0.5) Su.86.86 77(8) 400(0) 2000(0) ND ND ND 630(1) 3.17

ThemeanIC50(SD)forgemcitabine,erlotinib,andapricoxibwasdeterminedbyMTSassayassingleagentsor in combination. Means represent a minimum of 3 independent assays with conditions performed in octuplicateperassay.ThefoldchangeintheIC50forgemcitabineinthepresenceofapricoxib(A)anderlotinib

(E) each at 1 μM is displayed. SD = 0 indicates that an IC50 value was not achieved and the maximum concentrationofdrugusedisdisplayed.ND,notdetermined

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page26

FigureLegends

Figure1.BaselineexpressionandfunctionalactivityofEGFRandCOX2inhumanpancreaticcancer celllines.A,RNAwasharvestedfromhumanpancreaticcancercelllinesandevaluatedbyqPCRfor expression of COX2 and EGFR, levels were normalized to actin internal loading control. B, The expression of EGFR, pEGFR at tyr1068, and COX2 was determined by Western blot analysis.

Expression of actin was used as a loading control. C, The IC50 of erlotinib for inhibition of phosphorylationofEGFRafterstimulationwith10ng/mlEGFwasdeterminedbyELISA.D,PGE2levels inconditionedmediameasuredbyELISAafter24hourincubationwithapricoxib.

Figure2.COX2inhibitionresultsindecreasedprimarytumorgrowthandmetastaticburden.A&B,

AsPC1(n=68animals/group),Colo357(n=78animals/group),andHPAFII(n=710animals/group) humanpancreaticcancercells(1x106)wereinjectedorthotopicallyintothepancreasofSCIDmice.

Treatment began when established tumor was visible by ultrasound (~10 mm3) and consisted of control (vehicle alone, delivered ip), standard therapy of gemcitabine 25 mg/kg twice weekly plus erlotinib100gdaily(G+E)orstandardtherapyplusapricoxibat10mg/kg(10A)or30mg/kg(30A) dailyandcontinuedfor3weeksor7weeks(HPAFIItumorbearingmiceonly).Uponsacrificemean tumorweight(A)andmetastaticburden(B)werecompared.C,D,Paraffinembeddedtumorsections wereanalyzedforCOX2expressionbyimmunofluorescence.Dataaredisplayedasmean±SEMand represents5imagespertumorwithalltumorsevaluated.*p<0.05vs.control,#p<0.05vs.G+E.D,

Representativeimages(COX2,red;DAPI,blue)areshown.Totalmagnification,200X;scalebar,100

μM(C).E,AsPC1andColo357cellswereplatedeitherundernormalconditionsorunderconditions

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page27

offorcedEMT.PGE2levelsweremeasuredbyELISAinconditionedmediaatbaselineand24hours followingonetimeapricoxibdosingat0.5μM.

Figure 3. COX2 inhibition does not alter intratumoral levels of VEGF or microvessel density but facilitates vascular normalization. A & B, Levels of human VEGF in plasma (A) and tumors (B) collectedattimeofsacrificeweredeterminedbyELISAorimmunofluorescence.Meanplasmalevels ofVEGFareexpressedaspg/ml+/SEMandwereassayedintriplicatefromeachanimalunderstudy.

The level of VEGF in tumor tissue was determined by ELISA (AsPC1, pg/50 μg protein) or immunofluorescence (Colo357, % area fraction), the mean +/ SEM are displayed. C & D, Paraffin embedded sections of AsPC1 and Colo357 tumors were analyzed by immunofluorescence for endomucin,CD31andNG2expression.C,Dataaredisplayedasmean±SEMandrepresent5images pertumorwithfivetumorspergroupevaluated.D,RepresentativeimagesofCD31(green)andNG2

(red) are shown for Colo357 tumors. Total magnification, 200X; scale bar, 100 μM. Images were analyzedusingElementssoftware.*p<0.05,**p<0.01vs.control,#p<0.05,##p<0.005,#p<0.0005vs.

G+EbyonewayANOVA.

Figure 4. COX2 inhibition enhances therapyinduced effects on cell survival and proliferation in pancreatic cancer xenografts. A, Apoptosis was evaluated through TUNEL analysis in sections of

AsPC1 and Colo357 tumors. Data are displayed as mean % area fraction ± SEM and represents 5 imagespertumorwithfivetumorspergroupevaluated.B,RepresentativeimagesofTUNEL(green) reactivity in Colo357 tumors are shown. Total magnification, 200X; scale bar, 100 μM. C, Paraffin embedded sections of AsPC1 and Colo357 tumors were analyzed for cell proliferation (PCNA) by

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Kiraneetal,page28 immunofluorescence.Dataaredisplayedasmean%areafraction±SEMandrepresents5imagesper tumor with five tumors per group evaluated. D, Representative images of PCNA (green) immunofluorescence in sections of Colo357 tumors are displayed. Total magnification, 400X; scale bar,50μM.E,Colo357tumorswereanalyzedforH2AXbyimmunofluorescence.Dataisdisplayedas mean % area fraction and represents 5 images per tumor with five tumors per group evaluated, representative images are shown (H2AX, red; DAPI, blue). Total magnification 400x. *p<0.05,

**p<0.005vs.control,#p<0.05,##p<0.005vs.G+E.

Figure 5: COX2 inhibition results in reversal of EMT. A, Paraffin embedded sections of Colo357 tumorswereanalyzedforEcadherin,vimentin,andZeb1expressionbyimmunofluorescence.Data aredisplayedasmean%areafractionperhighpowerfield±SEMandrepresents5imagespertumor with five tumors per group evaluated. Images were analyzed using Elements software. B,

Representative images of vimentin (red) and Ecadherin (green) levels in treated Colo357 tumors.

Totalmagnification,200X;scalebar,100μM.C,Colo357cellsinvitroweretreatedwitheither100 nM gemcitabine (G) plus 1 M erlotinib (E) or G+E+ 1 M apricoxib and Zeb1 and ECadherin expression was evaluated by western blot. D, Paraffin embedded sections of AsPC1 tumors were analyzedforEcadherin,vimentin,Zeb1expressionbyimmunofluorescence.E,Westernblotanalysis ofZeb1expressionbyAsPC1cellsundernormalcultureconditionsorfollowinginductionofEMTand treatment with G+E, G+E+apricoxib, or apricoxib single agent. *p<0.05, **p<0.005 vs. control,

#p<0.05,##p<0.005,###p<0.0005vs.G+E.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 24, 2012; DOI: 10.1158/1078-0432.CCR-12-0453 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Apricoxib, a novel inhibitor of COX-2 markedly improves standard therapy response in molecularly defined models of pancreatic cancer

Amanda Kirane, Jason E Toombs, Katherine T Ostapoff, et al.

Clin Cancer Res Published OnlineFirst July 24, 2012.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-12-0453

Author Author manuscripts have been peer reviewed and accepted for publication but have not yet been Manuscript edited.

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

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2012/07/24/1078-0432.CCR-12-0453. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research.